CN114645023B - System and method for reprogramming peripheral blood mononuclear cells to induced pluripotent stem cells - Google Patents
System and method for reprogramming peripheral blood mononuclear cells to induced pluripotent stem cells Download PDFInfo
- Publication number
- CN114645023B CN114645023B CN202210541037.XA CN202210541037A CN114645023B CN 114645023 B CN114645023 B CN 114645023B CN 202210541037 A CN202210541037 A CN 202210541037A CN 114645023 B CN114645023 B CN 114645023B
- Authority
- CN
- China
- Prior art keywords
- inhibitor
- reprogramming
- concentration
- cells
- pluripotent stem
- Prior art date
- Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
- Active
Links
- 230000008672 reprogramming Effects 0.000 title claims abstract description 125
- 210000003819 peripheral blood mononuclear cell Anatomy 0.000 title claims abstract description 75
- 210000004263 induced pluripotent stem cell Anatomy 0.000 title claims abstract description 67
- 238000000034 method Methods 0.000 title claims description 52
- 210000004027 cell Anatomy 0.000 claims abstract description 80
- 239000003112 inhibitor Substances 0.000 claims abstract description 78
- 230000006698 induction Effects 0.000 claims abstract description 60
- 239000013598 vector Substances 0.000 claims abstract description 52
- 108010045512 cohesins Proteins 0.000 claims abstract description 36
- 229940122498 Gene expression inhibitor Drugs 0.000 claims abstract description 30
- 108010014905 Glycogen Synthase Kinase 3 Proteins 0.000 claims abstract description 27
- 108700025694 p53 Genes Proteins 0.000 claims abstract description 26
- 102000002254 Glycogen Synthase Kinase 3 Human genes 0.000 claims abstract description 25
- 239000002829 mitogen activated protein kinase inhibitor Substances 0.000 claims abstract description 24
- 229940124647 MEK inhibitor Drugs 0.000 claims abstract description 23
- 229940122964 Deacetylase inhibitor Drugs 0.000 claims abstract description 22
- 239000007640 basal medium Substances 0.000 claims abstract description 21
- DBLXOVFQHHSKRC-UHFFFAOYSA-N ethanesulfonic acid;2-piperazin-1-ylethanol Chemical compound CCS(O)(=O)=O.OCCN1CCNCC1 DBLXOVFQHHSKRC-UHFFFAOYSA-N 0.000 claims abstract description 16
- 108010068425 Octamer Transcription Factor-3 Proteins 0.000 claims abstract description 13
- 102000002584 Octamer Transcription Factor-3 Human genes 0.000 claims abstract description 13
- 239000012531 culture fluid Substances 0.000 claims abstract description 12
- 239000011435 rock Substances 0.000 claims abstract 9
- 229910002091 carbon monoxide Inorganic materials 0.000 claims description 31
- 239000002609 medium Substances 0.000 claims description 31
- 230000014509 gene expression Effects 0.000 claims description 29
- 230000002401 inhibitory effect Effects 0.000 claims description 23
- 239000004055 small Interfering RNA Substances 0.000 claims description 19
- 108091032973 (ribonucleotides)n+m Proteins 0.000 claims description 18
- 108091027967 Small hairpin RNA Proteins 0.000 claims description 18
- 238000012258 culturing Methods 0.000 claims description 18
- 102000008100 Human Serum Albumin Human genes 0.000 claims description 17
- 108091006905 Human Serum Albumin Proteins 0.000 claims description 17
- CIWBSHSKHKDKBQ-JLAZNSOCSA-N Ascorbic acid Chemical compound OC[C@H](O)[C@H]1OC(=O)C(O)=C1O CIWBSHSKHKDKBQ-JLAZNSOCSA-N 0.000 claims description 16
- 239000013612 plasmid Substances 0.000 claims description 16
- 238000001890 transfection Methods 0.000 claims description 16
- 239000006144 Dulbecco’s modified Eagle's medium Substances 0.000 claims description 11
- 102000046299 Transforming Growth Factor beta1 Human genes 0.000 claims description 11
- 101800002279 Transforming growth factor beta-1 Proteins 0.000 claims description 11
- 210000000601 blood cell Anatomy 0.000 claims description 10
- 108090000379 Fibroblast growth factor 2 Proteins 0.000 claims description 9
- 102000003974 Fibroblast growth factor 2 Human genes 0.000 claims description 9
- 108090000723 Insulin-Like Growth Factor I Proteins 0.000 claims description 9
- 102000004218 Insulin-Like Growth Factor I Human genes 0.000 claims description 9
- 108090000556 Neuregulin-1 Proteins 0.000 claims description 9
- 102000048238 Neuregulin-1 Human genes 0.000 claims description 9
- 210000001778 pluripotent stem cell Anatomy 0.000 claims description 9
- 210000000130 stem cell Anatomy 0.000 claims description 9
- AQGNHMOJWBZFQQ-UHFFFAOYSA-N CT 99021 Chemical group CC1=CNC(C=2C(=NC(NCCNC=3N=CC(=CC=3)C#N)=NC=2)C=2C(=CC(Cl)=CC=2)Cl)=N1 AQGNHMOJWBZFQQ-UHFFFAOYSA-N 0.000 claims description 8
- 102000016938 Catalase Human genes 0.000 claims description 8
- 108010053835 Catalase Proteins 0.000 claims description 8
- 108020004459 Small interfering RNA Proteins 0.000 claims description 8
- 102000019197 Superoxide Dismutase Human genes 0.000 claims description 8
- 108010012715 Superoxide dismutase Proteins 0.000 claims description 8
- 102000004338 Transferrin Human genes 0.000 claims description 8
- 235000010323 ascorbic acid Nutrition 0.000 claims description 8
- 229960005070 ascorbic acid Drugs 0.000 claims description 8
- 239000011668 ascorbic acid Substances 0.000 claims description 8
- BVTBRVFYZUCAKH-UHFFFAOYSA-L disodium selenite Chemical compound [Na+].[Na+].[O-][Se]([O-])=O BVTBRVFYZUCAKH-UHFFFAOYSA-L 0.000 claims description 8
- 239000011781 sodium selenite Substances 0.000 claims description 8
- 235000015921 sodium selenite Nutrition 0.000 claims description 8
- 229960001471 sodium selenite Drugs 0.000 claims description 8
- 239000012581 transferrin Substances 0.000 claims description 8
- 108020005544 Antisense RNA Proteins 0.000 claims description 7
- 102000040945 Transcription factor Human genes 0.000 claims description 7
- 108091023040 Transcription factor Proteins 0.000 claims description 7
- 239000003184 complementary RNA Substances 0.000 claims description 7
- 230000001939 inductive effect Effects 0.000 claims description 7
- 108091070501 miRNA Proteins 0.000 claims description 6
- 239000002679 microRNA Substances 0.000 claims description 6
- DOBKQCZBPPCLEG-UHFFFAOYSA-N n-benzyl-2-(pyrimidin-4-ylamino)-1,3-thiazole-4-carboxamide Chemical group C=1SC(NC=2N=CN=CC=2)=NC=1C(=O)NCC1=CC=CC=C1 DOBKQCZBPPCLEG-UHFFFAOYSA-N 0.000 claims description 6
- 239000002924 silencing RNA Substances 0.000 claims description 6
- 108090000901 Transferrin Proteins 0.000 claims description 5
- 239000000969 carrier Substances 0.000 claims description 4
- 238000011278 co-treatment Methods 0.000 claims description 2
- 230000001976 improved effect Effects 0.000 abstract description 5
- 238000002360 preparation method Methods 0.000 abstract description 5
- 238000012136 culture method Methods 0.000 abstract 2
- 108090000623 proteins and genes Proteins 0.000 description 27
- 239000000243 solution Substances 0.000 description 22
- 239000011573 trace mineral Substances 0.000 description 17
- 235000013619 trace mineral Nutrition 0.000 description 17
- 239000001963 growth medium Substances 0.000 description 16
- NIJJYAXOARWZEE-UHFFFAOYSA-N Valproic acid Chemical group CCCC(C(O)=O)CCC NIJJYAXOARWZEE-UHFFFAOYSA-N 0.000 description 14
- 108020004999 messenger RNA Proteins 0.000 description 12
- 102100035423 POU domain, class 5, transcription factor 1 Human genes 0.000 description 10
- 230000008569 process Effects 0.000 description 8
- 210000001519 tissue Anatomy 0.000 description 8
- 101710126211 POU domain, class 5, transcription factor 1 Proteins 0.000 description 7
- 210000004369 blood Anatomy 0.000 description 7
- 239000008280 blood Substances 0.000 description 7
- 229960000604 valproic acid Drugs 0.000 description 7
- 108020004414 DNA Proteins 0.000 description 6
- OKKJLVBELUTLKV-UHFFFAOYSA-N Methanol Chemical compound OC OKKJLVBELUTLKV-UHFFFAOYSA-N 0.000 description 6
- 238000012228 RNA interference-mediated gene silencing Methods 0.000 description 5
- 230000010261 cell growth Effects 0.000 description 5
- 230000004069 differentiation Effects 0.000 description 5
- 238000004520 electroporation Methods 0.000 description 5
- 210000001671 embryonic stem cell Anatomy 0.000 description 5
- 230000009368 gene silencing by RNA Effects 0.000 description 5
- 102000004169 proteins and genes Human genes 0.000 description 5
- 230000004083 survival effect Effects 0.000 description 5
- 241000711408 Murine respirovirus Species 0.000 description 4
- 230000004663 cell proliferation Effects 0.000 description 4
- 238000001514 detection method Methods 0.000 description 4
- 230000000694 effects Effects 0.000 description 4
- 238000002474 experimental method Methods 0.000 description 4
- 238000010166 immunofluorescence Methods 0.000 description 4
- 239000003550 marker Substances 0.000 description 4
- 239000000203 mixture Substances 0.000 description 4
- 210000005087 mononuclear cell Anatomy 0.000 description 4
- 238000001179 sorption measurement Methods 0.000 description 4
- 239000006228 supernatant Substances 0.000 description 4
- QTBSBXVTEAMEQO-UHFFFAOYSA-N Acetic acid Chemical compound CC(O)=O QTBSBXVTEAMEQO-UHFFFAOYSA-N 0.000 description 3
- 108010077544 Chromatin Proteins 0.000 description 3
- 108091026890 Coding region Proteins 0.000 description 3
- 101001094700 Homo sapiens POU domain, class 5, transcription factor 1 Proteins 0.000 description 3
- 101000766306 Homo sapiens Serotransferrin Proteins 0.000 description 3
- 101000713275 Homo sapiens Solute carrier family 22 member 3 Proteins 0.000 description 3
- 101100247004 Rattus norvegicus Qsox1 gene Proteins 0.000 description 3
- 238000003556 assay Methods 0.000 description 3
- 239000006285 cell suspension Substances 0.000 description 3
- 210000003483 chromatin Anatomy 0.000 description 3
- 238000010790 dilution Methods 0.000 description 3
- 239000012895 dilution Substances 0.000 description 3
- 201000010099 disease Diseases 0.000 description 3
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 description 3
- 230000012010 growth Effects 0.000 description 3
- 102000039446 nucleic acids Human genes 0.000 description 3
- 108020004707 nucleic acids Proteins 0.000 description 3
- 150000007523 nucleic acids Chemical class 0.000 description 3
- 210000005259 peripheral blood Anatomy 0.000 description 3
- 239000011886 peripheral blood Substances 0.000 description 3
- NLKNQRATVPKPDG-UHFFFAOYSA-M potassium iodide Chemical compound [K+].[I-] NLKNQRATVPKPDG-UHFFFAOYSA-M 0.000 description 3
- 230000001737 promoting effect Effects 0.000 description 3
- 230000001105 regulatory effect Effects 0.000 description 3
- OZFAFGSSMRRTDW-UHFFFAOYSA-N (2,4-dichlorophenyl) benzenesulfonate Chemical compound ClC1=CC(Cl)=CC=C1OS(=O)(=O)C1=CC=CC=C1 OZFAFGSSMRRTDW-UHFFFAOYSA-N 0.000 description 2
- IAKHMKGGTNLKSZ-INIZCTEOSA-N (S)-colchicine Chemical compound C1([C@@H](NC(C)=O)CC2)=CC(=O)C(OC)=CC=C1C1=C2C=C(OC)C(OC)=C1OC IAKHMKGGTNLKSZ-INIZCTEOSA-N 0.000 description 2
- GKHIVNAUVKXIIY-UHFFFAOYSA-N 2-[3-[4-(1h-indazol-5-ylamino)quinazolin-2-yl]phenoxy]-n-propan-2-ylacetamide Chemical compound CC(C)NC(=O)COC1=CC=CC(C=2N=C3C=CC=CC3=C(NC=3C=C4C=NNC4=CC=3)N=2)=C1 GKHIVNAUVKXIIY-UHFFFAOYSA-N 0.000 description 2
- JKMHFZQWWAIEOD-UHFFFAOYSA-N 2-[4-(2-hydroxyethyl)piperazin-1-yl]ethanesulfonic acid Chemical compound OCC[NH+]1CCN(CCS([O-])(=O)=O)CC1 JKMHFZQWWAIEOD-UHFFFAOYSA-N 0.000 description 2
- 102100025064 Cellular tumor antigen p53 Human genes 0.000 description 2
- 239000012591 Dulbecco’s Phosphate Buffered Saline Substances 0.000 description 2
- 102000018233 Fibroblast Growth Factor Human genes 0.000 description 2
- 108050007372 Fibroblast Growth Factor Proteins 0.000 description 2
- 102000001267 GSK3 Human genes 0.000 description 2
- 102000003964 Histone deacetylase Human genes 0.000 description 2
- 108090000353 Histone deacetylase Proteins 0.000 description 2
- 101000599951 Homo sapiens Insulin-like growth factor I Proteins 0.000 description 2
- VEXZGXHMUGYJMC-UHFFFAOYSA-N Hydrochloric acid Chemical compound Cl VEXZGXHMUGYJMC-UHFFFAOYSA-N 0.000 description 2
- 102100037852 Insulin-like growth factor I Human genes 0.000 description 2
- 102000043136 MAP kinase family Human genes 0.000 description 2
- 108091054455 MAP kinase family Proteins 0.000 description 2
- 102000004232 Mitogen-Activated Protein Kinase Kinases Human genes 0.000 description 2
- 108091028043 Nucleic acid sequence Proteins 0.000 description 2
- SUDAHWBOROXANE-VIFPVBQESA-N PD 0325901-Cl Chemical compound OC[C@H](O)CONC(=O)C1=CC=C(F)C(F)=C1NC1=CC=C(I)C=C1F SUDAHWBOROXANE-VIFPVBQESA-N 0.000 description 2
- 108010048992 Transcription Factor 4 Proteins 0.000 description 2
- 102100023489 Transcription factor 4 Human genes 0.000 description 2
- RTKIYFITIVXBLE-UHFFFAOYSA-N Trichostatin A Natural products ONC(=O)C=CC(C)=CC(C)C(=O)C1=CC=C(N(C)C)C=C1 RTKIYFITIVXBLE-UHFFFAOYSA-N 0.000 description 2
- 108010076089 accutase Proteins 0.000 description 2
- 101150063416 add gene Proteins 0.000 description 2
- VSCWAEJMTAWNJL-UHFFFAOYSA-K aluminium trichloride Chemical compound Cl[Al](Cl)Cl VSCWAEJMTAWNJL-UHFFFAOYSA-K 0.000 description 2
- 238000004458 analytical method Methods 0.000 description 2
- 230000006907 apoptotic process Effects 0.000 description 2
- 239000002585 base Substances 0.000 description 2
- 210000002459 blastocyst Anatomy 0.000 description 2
- YKYOUMDCQGMQQO-UHFFFAOYSA-L cadmium dichloride Chemical compound Cl[Cd]Cl YKYOUMDCQGMQQO-UHFFFAOYSA-L 0.000 description 2
- 238000004113 cell culture Methods 0.000 description 2
- 239000006143 cell culture medium Substances 0.000 description 2
- 238000005119 centrifugation Methods 0.000 description 2
- 238000010367 cloning Methods 0.000 description 2
- 230000000295 complement effect Effects 0.000 description 2
- 239000003814 drug Substances 0.000 description 2
- 238000005516 engineering process Methods 0.000 description 2
- 239000013604 expression vector Substances 0.000 description 2
- 229940126864 fibroblast growth factor Drugs 0.000 description 2
- 239000012997 ficoll-paque Substances 0.000 description 2
- 210000001654 germ layer Anatomy 0.000 description 2
- YBMRDBCBODYGJE-UHFFFAOYSA-N germanium dioxide Chemical compound O=[Ge]=O YBMRDBCBODYGJE-UHFFFAOYSA-N 0.000 description 2
- 229940121372 histone deacetylase inhibitor Drugs 0.000 description 2
- 239000003276 histone deacetylase inhibitor Substances 0.000 description 2
- 238000003780 insertion Methods 0.000 description 2
- 230000037431 insertion Effects 0.000 description 2
- 210000004962 mammalian cell Anatomy 0.000 description 2
- 238000004519 manufacturing process Methods 0.000 description 2
- 108010082117 matrigel Proteins 0.000 description 2
- 230000007246 mechanism Effects 0.000 description 2
- 238000002156 mixing Methods 0.000 description 2
- 239000002773 nucleotide Substances 0.000 description 2
- 125000003729 nucleotide group Chemical group 0.000 description 2
- 235000016709 nutrition Nutrition 0.000 description 2
- 230000035764 nutrition Effects 0.000 description 2
- 210000000056 organ Anatomy 0.000 description 2
- IOLCXVTUBQKXJR-UHFFFAOYSA-M potassium bromide Chemical compound [K+].[Br-] IOLCXVTUBQKXJR-UHFFFAOYSA-M 0.000 description 2
- 239000000047 product Substances 0.000 description 2
- 230000035755 proliferation Effects 0.000 description 2
- 230000001172 regenerating effect Effects 0.000 description 2
- FGDZQCVHDSGLHJ-UHFFFAOYSA-M rubidium chloride Chemical compound [Cl-].[Rb+] FGDZQCVHDSGLHJ-UHFFFAOYSA-M 0.000 description 2
- SQGYOTSLMSWVJD-UHFFFAOYSA-N silver(1+) nitrate Chemical compound [Ag+].[O-]N(=O)=O SQGYOTSLMSWVJD-UHFFFAOYSA-N 0.000 description 2
- PUZPDOWCWNUUKD-UHFFFAOYSA-M sodium fluoride Chemical compound [F-].[Na+] PUZPDOWCWNUUKD-UHFFFAOYSA-M 0.000 description 2
- 210000001082 somatic cell Anatomy 0.000 description 2
- 229940099456 transforming growth factor beta 1 Drugs 0.000 description 2
- RTKIYFITIVXBLE-QEQCGCAPSA-N trichostatin A Chemical compound ONC(=O)/C=C/C(/C)=C/[C@@H](C)C(=O)C1=CC=C(N(C)C)C=C1 RTKIYFITIVXBLE-QEQCGCAPSA-N 0.000 description 2
- 239000013603 viral vector Substances 0.000 description 2
- WAEXFXRVDQXREF-UHFFFAOYSA-N vorinostat Chemical compound ONC(=O)CCCCCCC(=O)NC1=CC=CC=C1 WAEXFXRVDQXREF-UHFFFAOYSA-N 0.000 description 2
- 229960000237 vorinostat Drugs 0.000 description 2
- XLYOFNOQVPJJNP-UHFFFAOYSA-N water Substances O XLYOFNOQVPJJNP-UHFFFAOYSA-N 0.000 description 2
- TXUICONDJPYNPY-UHFFFAOYSA-N (1,10,13-trimethyl-3-oxo-4,5,6,7,8,9,11,12,14,15,16,17-dodecahydrocyclopenta[a]phenanthren-17-yl) heptanoate Chemical compound C1CC2CC(=O)C=C(C)C2(C)C2C1C1CCC(OC(=O)CCCCCC)C1(C)CC2 TXUICONDJPYNPY-UHFFFAOYSA-N 0.000 description 1
- JCSGFHVFHSKIJH-UHFFFAOYSA-N 3-(2,4-dichlorophenyl)-4-(1-methyl-3-indolyl)pyrrole-2,5-dione Chemical compound C12=CC=CC=C2N(C)C=C1C(C(NC1=O)=O)=C1C1=CC=C(Cl)C=C1Cl JCSGFHVFHSKIJH-UHFFFAOYSA-N 0.000 description 1
- VPVLEBIVXZSOMQ-UHFFFAOYSA-N 3-[[6-(3-aminophenyl)-7H-pyrrolo[2,3-d]pyrimidin-4-yl]oxy]phenol Chemical compound NC1=CC=CC(C=2NC3=NC=NC(OC=4C=C(O)C=CC=4)=C3C=2)=C1 VPVLEBIVXZSOMQ-UHFFFAOYSA-N 0.000 description 1
- FWMNVWWHGCHHJJ-SKKKGAJSSA-N 4-amino-1-[(2r)-6-amino-2-[[(2r)-2-[[(2r)-2-[[(2r)-2-amino-3-phenylpropanoyl]amino]-3-phenylpropanoyl]amino]-4-methylpentanoyl]amino]hexanoyl]piperidine-4-carboxylic acid Chemical compound C([C@H](C(=O)N[C@H](CC(C)C)C(=O)N[C@H](CCCCN)C(=O)N1CCC(N)(CC1)C(O)=O)NC(=O)[C@H](N)CC=1C=CC=CC=1)C1=CC=CC=C1 FWMNVWWHGCHHJJ-SKKKGAJSSA-N 0.000 description 1
- 101150111660 53 gene Proteins 0.000 description 1
- 108010013043 Acetylesterase Proteins 0.000 description 1
- 241000894006 Bacteria Species 0.000 description 1
- 229910021555 Chromium Chloride Inorganic materials 0.000 description 1
- 108010051219 Cre recombinase Proteins 0.000 description 1
- 102000004127 Cytokines Human genes 0.000 description 1
- 108090000695 Cytokines Proteins 0.000 description 1
- 241000282326 Felis catus Species 0.000 description 1
- 241000233866 Fungi Species 0.000 description 1
- 108010051975 Glycogen Synthase Kinase 3 beta Proteins 0.000 description 1
- 102100022975 Glycogen synthase kinase-3 alpha Human genes 0.000 description 1
- 102100038104 Glycogen synthase kinase-3 beta Human genes 0.000 description 1
- 101100137155 Homo sapiens POU5F1 gene Proteins 0.000 description 1
- 101100094846 Homo sapiens SLC22A3 gene Proteins 0.000 description 1
- 108091092195 Intron Proteins 0.000 description 1
- 108700021430 Kruppel-Like Factor 4 Proteins 0.000 description 1
- HRJWTAWVFDCTGO-UHFFFAOYSA-N LY-2090314 Chemical compound C1CN(C=23)C=C(C=4C(NC(=O)C=4C=4N5C=CC=CC5=NC=4)=O)C3=CC(F)=CC=2CN1C(=O)N1CCCCC1 HRJWTAWVFDCTGO-UHFFFAOYSA-N 0.000 description 1
- 108010085895 Laminin Proteins 0.000 description 1
- 241000713666 Lentivirus Species 0.000 description 1
- 241001465754 Metazoa Species 0.000 description 1
- 102100024193 Mitogen-activated protein kinase 1 Human genes 0.000 description 1
- 102100038895 Myc proto-oncogene protein Human genes 0.000 description 1
- 101710135898 Myc proto-oncogene protein Proteins 0.000 description 1
- 206010028980 Neoplasm Diseases 0.000 description 1
- 102000001253 Protein Kinase Human genes 0.000 description 1
- 101150086694 SLC22A3 gene Proteins 0.000 description 1
- 239000004115 Sodium Silicate Substances 0.000 description 1
- 229910021626 Tin(II) chloride Inorganic materials 0.000 description 1
- 101710150448 Transcriptional regulator Myc Proteins 0.000 description 1
- 102000004243 Tubulin Human genes 0.000 description 1
- 108090000704 Tubulin Proteins 0.000 description 1
- 108700025716 Tumor Suppressor Genes Proteins 0.000 description 1
- 102000044209 Tumor Suppressor Genes Human genes 0.000 description 1
- 241000700605 Viruses Species 0.000 description 1
- IYOZTVGMEWJPKR-IJLUTSLNSA-N Y-27632 Chemical compound C1C[C@@H]([C@H](N)C)CC[C@@H]1C(=O)NC1=CC=NC=C1 IYOZTVGMEWJPKR-IJLUTSLNSA-N 0.000 description 1
- 230000009471 action Effects 0.000 description 1
- 230000004913 activation Effects 0.000 description 1
- APUPEJJSWDHEBO-UHFFFAOYSA-P ammonium molybdate Chemical compound [NH4+].[NH4+].[O-][Mo]([O-])(=O)=O APUPEJJSWDHEBO-UHFFFAOYSA-P 0.000 description 1
- 239000011609 ammonium molybdate Substances 0.000 description 1
- 235000018660 ammonium molybdate Nutrition 0.000 description 1
- 229940010552 ammonium molybdate Drugs 0.000 description 1
- 230000000692 anti-sense effect Effects 0.000 description 1
- 239000000427 antigen Substances 0.000 description 1
- 108091007433 antigens Proteins 0.000 description 1
- 102000036639 antigens Human genes 0.000 description 1
- UNTBPXHCXVWYOI-UHFFFAOYSA-O azanium;oxido(dioxo)vanadium Chemical compound [NH4+].[O-][V](=O)=O UNTBPXHCXVWYOI-UHFFFAOYSA-O 0.000 description 1
- ITHZDDVSAWDQPZ-UHFFFAOYSA-L barium acetate Chemical compound [Ba+2].CC([O-])=O.CC([O-])=O ITHZDDVSAWDQPZ-UHFFFAOYSA-L 0.000 description 1
- 230000008901 benefit Effects 0.000 description 1
- 230000031018 biological processes and functions Effects 0.000 description 1
- 230000015572 biosynthetic process Effects 0.000 description 1
- 238000002659 cell therapy Methods 0.000 description 1
- 230000008859 change Effects 0.000 description 1
- SAQUSDSPQYQNBG-UHFFFAOYSA-N chembl355496 Chemical compound N1C2=CC=CC=C2C(N=O)=C1C1=C(O)NC2=CC(Br)=CC=C21 SAQUSDSPQYQNBG-UHFFFAOYSA-N 0.000 description 1
- 239000003153 chemical reaction reagent Substances 0.000 description 1
- 239000003795 chemical substances by application Substances 0.000 description 1
- QSWDMMVNRMROPK-UHFFFAOYSA-K chromium(3+) trichloride Chemical compound [Cl-].[Cl-].[Cl-].[Cr+3] QSWDMMVNRMROPK-UHFFFAOYSA-K 0.000 description 1
- 230000002759 chromosomal effect Effects 0.000 description 1
- 210000000349 chromosome Anatomy 0.000 description 1
- GVPFVAHMJGGAJG-UHFFFAOYSA-L cobalt dichloride Chemical compound [Cl-].[Cl-].[Co+2] GVPFVAHMJGGAJG-UHFFFAOYSA-L 0.000 description 1
- 229960001338 colchicine Drugs 0.000 description 1
- 230000000593 degrading effect Effects 0.000 description 1
- 238000000432 density-gradient centrifugation Methods 0.000 description 1
- 238000013461 design Methods 0.000 description 1
- 230000029087 digestion Effects 0.000 description 1
- 238000007877 drug screening Methods 0.000 description 1
- 230000013020 embryo development Effects 0.000 description 1
- 239000003623 enhancer Substances 0.000 description 1
- 230000002708 enhancing effect Effects 0.000 description 1
- 210000003527 eukaryotic cell Anatomy 0.000 description 1
- 230000003203 everyday effect Effects 0.000 description 1
- 210000002950 fibroblast Anatomy 0.000 description 1
- 230000030279 gene silencing Effects 0.000 description 1
- 238000012226 gene silencing method Methods 0.000 description 1
- 229940119177 germanium dioxide Drugs 0.000 description 1
- 108010049611 glycogen synthase kinase 3 alpha Proteins 0.000 description 1
- 210000003958 hematopoietic stem cell Anatomy 0.000 description 1
- 230000006195 histone acetylation Effects 0.000 description 1
- 238000003125 immunofluorescent labeling Methods 0.000 description 1
- 238000000338 in vitro Methods 0.000 description 1
- 238000011534 incubation Methods 0.000 description 1
- 239000000411 inducer Substances 0.000 description 1
- 230000005764 inhibitory process Effects 0.000 description 1
- 230000002452 interceptive effect Effects 0.000 description 1
- 239000002502 liposome Substances 0.000 description 1
- 210000004698 lymphocyte Anatomy 0.000 description 1
- 229940099596 manganese sulfate Drugs 0.000 description 1
- 239000011702 manganese sulphate Substances 0.000 description 1
- 235000007079 manganese sulphate Nutrition 0.000 description 1
- SQQMAOCOWKFBNP-UHFFFAOYSA-L manganese(II) sulfate Chemical compound [Mn+2].[O-]S([O-])(=O)=O SQQMAOCOWKFBNP-UHFFFAOYSA-L 0.000 description 1
- 239000000463 material Substances 0.000 description 1
- 238000005259 measurement Methods 0.000 description 1
- 230000001404 mediated effect Effects 0.000 description 1
- 230000004060 metabolic process Effects 0.000 description 1
- 238000001000 micrograph Methods 0.000 description 1
- 238000000520 microinjection Methods 0.000 description 1
- 210000001616 monocyte Anatomy 0.000 description 1
- 230000000877 morphologic effect Effects 0.000 description 1
- 239000002547 new drug Substances 0.000 description 1
- LGQLOGILCSXPEA-UHFFFAOYSA-L nickel sulfate Chemical compound [Ni+2].[O-]S([O-])(=O)=O LGQLOGILCSXPEA-UHFFFAOYSA-L 0.000 description 1
- 229910000363 nickel(II) sulfate Inorganic materials 0.000 description 1
- 235000015097 nutrients Nutrition 0.000 description 1
- 239000002245 particle Substances 0.000 description 1
- 230000037361 pathway Effects 0.000 description 1
- 230000002093 peripheral effect Effects 0.000 description 1
- 239000013600 plasmid vector Substances 0.000 description 1
- 210000004180 plasmocyte Anatomy 0.000 description 1
- 230000008092 positive effect Effects 0.000 description 1
- 230000001124 posttranscriptional effect Effects 0.000 description 1
- 238000011533 pre-incubation Methods 0.000 description 1
- 238000012545 processing Methods 0.000 description 1
- 108060006633 protein kinase Proteins 0.000 description 1
- 238000009256 replacement therapy Methods 0.000 description 1
- 238000011160 research Methods 0.000 description 1
- 108091008146 restriction endonucleases Proteins 0.000 description 1
- 229940102127 rubidium chloride Drugs 0.000 description 1
- 238000000926 separation method Methods 0.000 description 1
- 238000004904 shortening Methods 0.000 description 1
- 230000019491 signal transduction Effects 0.000 description 1
- 229910001961 silver nitrate Inorganic materials 0.000 description 1
- -1 small molecule compound Chemical class 0.000 description 1
- 150000003384 small molecules Chemical class 0.000 description 1
- MFBOGIVSZKQAPD-UHFFFAOYSA-M sodium butyrate Chemical compound [Na+].CCCC([O-])=O MFBOGIVSZKQAPD-UHFFFAOYSA-M 0.000 description 1
- 239000011775 sodium fluoride Substances 0.000 description 1
- 235000013024 sodium fluoride Nutrition 0.000 description 1
- NTHWMYGWWRZVTN-UHFFFAOYSA-N sodium silicate Chemical compound [Na+].[Na+].[O-][Si]([O-])=O NTHWMYGWWRZVTN-UHFFFAOYSA-N 0.000 description 1
- 229910052911 sodium silicate Inorganic materials 0.000 description 1
- 235000011150 stannous chloride Nutrition 0.000 description 1
- 239000001119 stannous chloride Substances 0.000 description 1
- 230000002195 synergetic effect Effects 0.000 description 1
- 238000003786 synthesis reaction Methods 0.000 description 1
- PMJIHLSCWIDGMD-UHFFFAOYSA-N tideglusib Chemical compound O=C1SN(C=2C3=CC=CC=C3C=CC=2)C(=O)N1CC1=CC=CC=C1 PMJIHLSCWIDGMD-UHFFFAOYSA-N 0.000 description 1
- 229950005284 tideglusib Drugs 0.000 description 1
- 238000012546 transfer Methods 0.000 description 1
- 238000013519 translation Methods 0.000 description 1
- 238000002054 transplantation Methods 0.000 description 1
- 230000003612 virological effect Effects 0.000 description 1
- IPCAPQRVQMIMAN-UHFFFAOYSA-L zirconyl chloride Chemical compound Cl[Zr](Cl)=O IPCAPQRVQMIMAN-UHFFFAOYSA-L 0.000 description 1
Images
Classifications
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N5/00—Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
- C12N5/06—Animal cells or tissues; Human cells or tissues
- C12N5/0602—Vertebrate cells
- C12N5/0696—Artificially induced pluripotent stem cells, e.g. iPS
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2500/00—Specific components of cell culture medium
- C12N2500/05—Inorganic components
- C12N2500/10—Metals; Metal chelators
- C12N2500/20—Transition metals
- C12N2500/24—Iron; Fe chelators; Transferrin
- C12N2500/25—Insulin-transferrin; Insulin-transferrin-selenium
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2500/00—Specific components of cell culture medium
- C12N2500/30—Organic components
- C12N2500/38—Vitamins
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2501/00—Active agents used in cell culture processes, e.g. differentation
- C12N2501/065—Modulators of histone acetylation
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2501/00—Active agents used in cell culture processes, e.g. differentation
- C12N2501/10—Growth factors
- C12N2501/105—Insulin-like growth factors [IGF]
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2501/00—Active agents used in cell culture processes, e.g. differentation
- C12N2501/10—Growth factors
- C12N2501/115—Basic fibroblast growth factor (bFGF, FGF-2)
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2501/00—Active agents used in cell culture processes, e.g. differentation
- C12N2501/10—Growth factors
- C12N2501/13—Nerve growth factor [NGF]; Brain-derived neurotrophic factor [BDNF]; Cilliary neurotrophic factor [CNTF]; Glial-derived neurotrophic factor [GDNF]; Neurotrophins [NT]; Neuregulins
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2501/00—Active agents used in cell culture processes, e.g. differentation
- C12N2501/10—Growth factors
- C12N2501/15—Transforming growth factor beta (TGF-β)
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2501/00—Active agents used in cell culture processes, e.g. differentation
- C12N2501/40—Regulators of development
- C12N2501/405—Cell cycle regulated proteins, e.g. cyclins, cyclin-dependant kinases
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2501/00—Active agents used in cell culture processes, e.g. differentation
- C12N2501/60—Transcription factors
- C12N2501/603—Oct-3/4
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2501/00—Active agents used in cell culture processes, e.g. differentation
- C12N2501/70—Enzymes
- C12N2501/71—Oxidoreductases (EC 1.)
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2501/00—Active agents used in cell culture processes, e.g. differentation
- C12N2501/70—Enzymes
- C12N2501/72—Transferases [EC 2.]
- C12N2501/727—Kinases (EC 2.7.)
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2501/00—Active agents used in cell culture processes, e.g. differentation
- C12N2501/998—Proteins not provided for elsewhere
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2501/00—Active agents used in cell culture processes, e.g. differentation
- C12N2501/999—Small molecules not provided for elsewhere
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2506/00—Differentiation of animal cells from one lineage to another; Differentiation of pluripotent cells
- C12N2506/11—Differentiation of animal cells from one lineage to another; Differentiation of pluripotent cells from blood or immune system cells
Landscapes
- Health & Medical Sciences (AREA)
- Engineering & Computer Science (AREA)
- Life Sciences & Earth Sciences (AREA)
- Biomedical Technology (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Wood Science & Technology (AREA)
- Biotechnology (AREA)
- Organic Chemistry (AREA)
- Chemical & Material Sciences (AREA)
- Genetics & Genomics (AREA)
- Zoology (AREA)
- Developmental Biology & Embryology (AREA)
- Microbiology (AREA)
- Transplantation (AREA)
- Biochemistry (AREA)
- General Engineering & Computer Science (AREA)
- General Health & Medical Sciences (AREA)
- Cell Biology (AREA)
- Micro-Organisms Or Cultivation Processes Thereof (AREA)
Abstract
The invention discloses a reprogramming system and a culture method for reprogramming Peripheral Blood Mononuclear Cells (PBMCs) into human induced pluripotent stem cells. The reprogramming system comprises a reprogramming induction culture solution and one or more vectors capable of expressing human transcription factor OCT4, a p53 gene expression inhibitor and a Cohesin gene expression inhibitor; wherein the reprogramming induction culture fluid comprises IX-PSC basal medium, GSK-3 inhibitor, deacetylase inhibitor, MEK inhibitor, ROCK inhibitor and 4-hydroxyethyl piperazine ethanesulfonic acid. By using the system and the culture method, the reprogramming efficiency of the cells can be improved, the preparation period of the human induced pluripotent stem cells is shortened, and the preparation cost of the human induced pluripotent stem cells is saved.
Description
Technical Field
The invention belongs to the technical field of induced pluripotent stem cell culture, and particularly relates to a system and a method for reprogramming peripheral blood mononuclear cells into induced pluripotent stem cells.
Background
The embryonic stem cell is a pluripotent stem cell, has the potential of differentiating into various types of cells of various tissues and organs, can be applied to organ tissue transplantation and cell therapy in clinic, and can also be used for providing a cell model for researching diseases. However, since the inner cell mass of the blastocyst in the early stage of embryonic development is derived, the operation of the egg cells is inevitable during the establishment of the blastocyst, so that a plurality of ethical problems are involved and are always controversial.
The Yamanaka laboratory of Kyoto university in Japan in 2006 utilizes four transcription factors, Oct3/4, Sox2, Klf4 and c-Myc 4 to reprogram mouse fibroblasts into induced multifunctional stem cells, and marks the emergence of a novel embryonic stem cell. Induced Pluripotent Stem Cells (iPSCs) can be prepared from almost all adult tissue cells, so that the preparation of autologous pluripotent stem cells of patients becomes possible, and the limitation of clinical application of embryonic stem cells can be effectively avoided. Therefore, the iPS cell has wide application prospect in the aspects of new drug screening, in-vitro disease model establishment, cell replacement therapy and regenerative medicine.
At present, a plurality of iPSC reprogramming methods are developed internationally, but all the methods have certain limitations. If Sendai virus or lentivirus is used to carry different exogenous transcription factors to express in cells so as to start cell reprogramming, the method has the disadvantages that the immediate insertion of genome may bring potential risks and the virus induction experiment cost is high. The reprogramming component is deleted by transiently expressing Cre recombinase after induction is finished, the system can delete exogenous genes after reprogramming is finished, activation of the reprogramming factor can be avoided, and therefore the risk of tumor generation is reduced.
In addition, in order to provide sufficient nutrition for cell growth, a large amount of animal-derived components are added into a culture medium used in the existing reprogramming process of induced pluripotent stem cells, so that the clinical application of the iPSC is limited.
In a word, the existing technical system for inducing human body cells to be iPS cells generally faces the problems of low efficiency, long induction time, high preparation cost and the like. In order to widely apply human iPS cells in regenerative medicine and disease occurrence mechanism research, an efficient induction strategy, namely a reprogramming culture medium and a reprogramming method capable of shortening induction time of the iPS cells, needs to be explored.
Disclosure of Invention
The invention provides a reprogramming system and a reprogramming method for reprogramming Peripheral Blood Mononuclear Cells (PBMCs) into human induced pluripotent stem cells, aiming at the technical problems of low reprogramming efficiency and long induction time of the existing iPS cells.
One aspect of the present invention provides a reprogramming system for reprogramming Peripheral Blood Mononuclear Cells (PBMCs) into human induced pluripotent stem cells, characterized in that the reprogramming system comprises a reprogramming induction culture solution and one or more vectors capable of expressing human transcription factor OCT4, a p53 gene expression inhibitor, and a Cohesin gene expression inhibitor; wherein the reprogramming induction culture fluid comprises IX-PSC basal medium, GSK-3 inhibitor, deacetylase inhibitor, MEK inhibitor, ROCK inhibitor and 4-hydroxyethyl piperazine ethanesulfonic acid; wherein the IX-PSC basic culture medium is DMEM/F12 as basic culture medium, and is added with ascorbic acid, FGF2, TGF beta 1, neuregulin 1, IGF-1, superoxide dismutase, catalase, human serum albumin, transferrin and sodium selenite.
In some embodiments, the inhibitor of p53 gene expression and/or inhibitor of Cohesin gene expression is an inhibitory RNA molecule; preferably, the inhibitory RNA molecule is an antisense RNA, miRNA, siRNA or shRNA; more preferably, the inhibitory RNA molecule is a shRNA.
In some embodiments, the one or more vectors are non-genomically integrated vectors; more preferably, the one or more vectors are plasmids.
In some embodiments, the GSK-3 inhibitor is CHIR99021, the deacetylase inhibitor is VPA, the MEK inhibitor is PD0325901, and/or the ROCK inhibitor is Thiazovivin.
In some embodiments, in the reprogramming induction culture fluid, the human serum albumin concentration is 1-10mg/mL, the GSK-3 inhibitor concentration is 0.5-3 μ Μ, the deacetylase inhibitor concentration is 0.3-3 μ Μ, the MEK inhibitor concentration is 10-100nM, the ROCK inhibitor concentration is 0.5-5 μ Μ, and/or the 4-hydroxyethylpiperazine ethanesulfonic acid concentration is 1-10 mM;
preferably, in the reprogramming induction culture fluid, the human serum albumin concentration is 5mg/mL, the GSK-3 inhibitor concentration is 2 μ M, the deacetylase inhibitor concentration is 2 μ M, the MEK inhibitor concentration is 50nM, the ROCK inhibitor concentration is 2.5 μ M, and/or the 4-hydroxyethylpiperazine ethanesulfonic acid concentration is 5 mM.
Another aspect of the invention provides a method of reprogramming Peripheral Blood Mononuclear Cells (PBMCs) to induced pluripotent stem cells, the method comprising:
i) transfecting peripheral blood mononuclear cells with one or more vectors, wherein the one or more vectors can express human transcription factors OCT4, p53 gene expression inhibitors and Cohesin gene expression inhibitors, and obtaining peripheral blood mononuclear cells expressing the human transcription factors OCT4, p53 gene expression inhibitors and the Cohesin gene expression inhibitors;
ii) culturing the peripheral blood mononuclear cells in a reprogramming induction culture solution to obtain induced pluripotent stem cells;
wherein the reprogramming induction culture fluid comprises IX-PSC basal medium, GSK-3 inhibitor, deacetylase inhibitor, MEK inhibitor, ROCK inhibitor and 4-hydroxyethyl piperazine ethanesulfonic acid; wherein the IX-PSC basal medium is prepared by taking DMEM/F12 as a basal medium and adding ascorbic acid, FGF2, TGF beta 1, neuregulin 1, IGF-1, superoxide dismutase, catalase, human serum albumin, transferrin and sodium selenite.
In some embodiments, the inhibitor of p53 gene expression and/or inhibitor of Cohesin gene expression is an inhibitory RNA molecule; preferably, the inhibitory RNA molecule is an antisense RNA, miRNA, siRNA or shRNA; more preferably, the inhibitory RNA molecule is a shRNA.
In some embodiments, the one or more vectors are non-genomically integrated vectors; more preferably, the one or more vectors are plasmids.
In some embodiments, the total concentration of the one or more carriers is 1-3 μ g carrier/1.0 × 10 5 Blood cells, preferably 2. mu.g of carrier/1.0X 10 5 Blood cells.
In some embodiments, the GSK-3 inhibitor is CHIR99021, the deacetylase inhibitor is VPA, the MEK inhibitor is PD0325901 and/or the ROCK inhibitor is Thiazovivin.
In some embodiments, in the reprogramming induction culture fluid, the human serum albumin concentration is 1-10mg/mL, the GSK-3 inhibitor concentration is 0.5-3 μ Μ/mL, the deacetylase inhibitor concentration is 0.3-3 μ Μ/mL, the MEK inhibitor concentration is 10-100nM, the ROCK inhibitor concentration is 0.5-5 μ Μ, and/or the 4-hydroxyethylpiperazineethanesulfonic acid concentration is 1-10 mM;
preferably, in the reprogramming induction culture fluid, the human serum albumin concentration is 5mg/mL, the GSK-3 inhibitor concentration is 2 μ M, the deacetylase inhibitor concentration is 2 μ M, the MEK inhibitor concentration is 50nM, the ROCK inhibitor concentration is 2.5 μ M, and/or the 4-hydroxyethylpiperazine ethanesulfonic acid concentration is 5 mM.
In some embodiments, the method comprises the steps of:
step 1): peripheral Blood Mononuclear Cells (PBMC) are treated at 35-39 ℃ with 3-7% CO 2 And 0.5-2X 10 6 Culturing the cells/well in a well plate for 1-3 days;
step 2): according to 1-3. mu.g/1.0X 10 of each carrier 5 Blood cells the one or more vectors are transfected into the peripheral blood mononuclear cells obtained in step 1), and the cells are inoculated onto a tissue culture plate and cultured in StemPro-34 medium at 35-39 ℃ with 3-7% CO 2 And 3 to 7% of O 2 Culturing for 1-3 days;
and step 3): day 2-3 after transfection, fresh StemPro-34 medium was used instead; then replacing the reprogramming induction culture solution every 1-3 days, and carrying out 3-7% CO treatment at the temperature of 35-39 DEG C 2 And 3 to 7% of O 2 Until the induced pluripotent stem cell clone appears;
preferably, the method comprises the steps of:
step 1): peripheral Blood Mononuclear Cells (PBMC) were incubated at 37 ℃ with 5% CO 2 And 1X 10 6 Culturing the cells/well in a well plate for 2 days;
step 2): according to a total concentration of 2. mu.g carrier/1.0X 10 5 Blood cells the vector or vectors are transfected into peripheral blood mononuclear cells, which are plated onto tissue culture plates using StemPro-34 medium at 37 ℃ with 5% CO 2 And 5% of O 2 Culturing for 2 days under the conditions of (1);
step 3): day 2 post-transfection, fresh StemPro-chambers-34 were replaced; thereafter, the reprogramming induction medium was replaced every 2 days at 37 ℃ with 5% CO 2 And 5% of O 2 Until the induced pluripotent stem cell clone appears;
optionally, the method further comprises: step 4): replacement of the reprogramming induction culture after the occurrence of induced pluripotent stem cell cloningNutrient solution with 3-7% CO at 35-39 deg.C 2 The culture was continued with replacement of the stem cell medium every 1 to 2 days during the culture.
Yet another aspect of the present invention provides a kit comprising any one of the reprogramming systems described above.
A further aspect of the invention provides the use of any one of the reprogramming systems described above or any one of the kits described above for inducing reprogramming of Peripheral Blood Mononuclear Cells (PBMCs) into human induced pluripotent stem cells.
The invention has the positive effects that: according to the invention, a GSK-3 inhibitor, a deacetylase inhibitor, a MEK inhibitor and a ROCK inhibitor are added into a reprogramming induction culture medium during cell reprogramming, and a human transcription factor OCT4, a p53 gene expression inhibitor and a Cohesin gene expression inhibitor are used for promoting cell reprogramming, so that in the process of reprogramming PBMC (peripheral component interconnect) to iPS (induced pluripotent stem cells), all components act synergistically, the cell growth and proliferation are maintained, and the cell induced reprogramming efficiency is improved. In a preferred embodiment of the invention, non-genomically integrated vectors are used, and the risk of random gene insertion encountered in conventional viral transfection is also avoided.
Drawings
FIG. 1 is a microphotograph of Peripheral Blood Mononuclear Cells (PBMC) obtained by separation;
FIG. 2 shows the number of clones obtained by different reprogramming methods;
FIG. 3 is a photograph showing the EB differentiation assay of the obtained cell line;
FIG. 4 is a photograph showing the identification of the pluripotent immunofluorescence of the obtained cell line;
FIG. 5 is a photograph showing the flow identification of the obtained cell line;
FIG. 6 is a photograph showing the karyotype identification of the obtained cell line.
Detailed description of the preferred embodiments
The present invention provides a reprogramming system for inducing reprogramming of PBMCs into induced pluripotent stem cells. The reprogramming system is a combination of multiple components, wherein each component can synergistically induce the reprogramming of the cells, so that the reprogramming efficiency of the cells is improved, and the time for inducing the reprogramming is shortened. The IX-PSC basal medium containing human serum albumin can provide abundant nutrition and necessary cytokines for cell growth and proliferation. The GSK-3 inhibitor CHIR99021 can inhibit the activity of GSK-3 alpha and GSK-3 beta in a mode of competing ATP binding sites, so that the apoptosis is inhibited, the survival rate of cells in the reprogramming process is improved, a WNT signal channel is enhanced, and the reprogramming efficiency of somatic cells into iPSCs is enhanced. The deacetylase inhibitor VPA can promote histone acetylation, so that chromatin adopts a loose structure, thereby promoting the combination of transcription factors and improving the reprogramming efficiency. The MEK inhibitor PD0325901 can achieve the effect of inhibiting MEK by changing conformation of ATP binding sites through binding with MEK, and remarkably improves survival, proliferation and reprogramming efficiency of iPSC by inhibiting MEK-ERK which regulates signal pathways related to cell proliferation and apoptosis. The ROCK inhibitor Thiazovivin can enhance the survival of embryonic stem cells, improve the generation efficiency of iPSC and shorten the reprogramming induction time of iPSC cells. 4-hydroxyethyl piperazine ethanesulfonic acid (HEPES) can maintain a stable pH value during cell culture, thereby promoting the growth and proliferation of cells. The p53 gene expression inhibitor and the Cohesin gene expression inhibitor can promote cell survival in the reprogramming process, and simultaneously can remarkably change the three-dimensional structure of chromatin to make the chromatin loose and open, thereby enhancing the action frequency of OCT4 and added induced small molecules (such as VPA, CHIR99021 and PD 0325901) and remarkably accelerating the reprogramming process.
The term "Peripheral Blood Mononuclear Cells (PBMC)" as used herein means mononuclear cells isolated from peripheral blood, and includes mainly lymphocytes, and also includes a small number of monocytes, plasma cells, hematopoietic stem cells and other progenitor cells. PBMCs for use in the present invention may be PBMCs isolated from blood or cryopreserved. PBMCs isolated from blood can generally be isolated from a cryopreserved or freshly collected blood sample by Ficoll-Paque density gradient centrifugation, e.g., in some embodiments, a cryopreserved or freshly collected blood sample can be isolated from a Ficoll-Paque density gradient centrifugationPeripheral Blood Mononuclear Cells (PBMC) with 3-7% CO at 35-39 deg.C preferably at 37 deg.C 2 Preferably 5% CO 2 、0.5-2×10 6 The cell density is preferably 1X 10 6 The cells/well are inoculated into an ultra-low adsorption 24-well plate for culture for 1-3 days, preferably 2 days, and the obtained PBMCs can be used for transfection of the vector. In other embodiments, cryopreserved PBMCs are used, e.g., cryopreserved and then reconstituted for transfection of vectors, e.g., in some embodiments, cryopreserved PBMCs are taken and rapidly thawed in a 37 ℃ water bath. Cells were gently pipetted 1-2 times using a 1mL pipette and transferred to a 15mL centrifuge tube. Rinsing the frozen tube with 1mL of room temperature preheated StemPro-34 basal medium for 1 time, transferring the rinsed cell suspension into the centrifuge tube, and adding 9mL of StemPro-34 basal medium into the centrifuge tube drop by drop; centrifuge at 200g for 3 min. Discarding supernatant, suspending cells by StemPro-34 complete culture medium, inoculating into ultra-low adsorption 24-well plate, and selecting 3-7% CO at 35-39 deg.C preferably 37 deg.C 2 Preferably 5% CO 2 Culturing in an incubator for 1-3 days, preferably 2 days. In some embodiments, the PBMCs may be human PBMCs.
"reprogramming" as used herein refers to the process of dedifferentiating a somatic cell into a pluripotent stem cell.
The reprogramming induction culture solution is prepared by adding a GSK-3 inhibitor, a deacetylase inhibitor, a MEK inhibitor, a ROCK inhibitor and 4-hydroxyethyl piperazine ethanesulfonic acid into an IX-PSC basic culture medium. The IX-PSC basic culture medium is obtained by taking DMEM/F12 as a basic culture medium and adding ascorbic acid, FGF2, TGF beta 1, neuregulin 1, IGF-1, superoxide dismutase, catalase, human serum albumin, transferrin and sodium selenite. In a preferred embodiment, the IX-PSC basal medium may be supplemented with 10-30. mu.g/mL ascorbic acid, 30-50 ng/mL FGF2, 0.5-2 ng/mL TGF β 1, 3-9 ng/mL neuregulin 1, 10-20 ng/mL IGF-1, 2-3. mu.g/mL superoxide dismutase, 2-3. mu.g/mL catalase, 1-10mg/mL human serum albumin, 10-30. mu.g/mL transferrin, and 10-30. mu.g/mL sodium selenite, based on DMEM/F12. In a more preferred embodiment, the IX-PSC basal medium may be DMEM/F12 supplemented with 20. mu.g/mL ascorbic acid, 40ng/mL FGF2, 1ng/mL TGF β 1, 6ng/mL neuregulin 1, 15ng/mL IGF-1, 2.5. mu.g/mL superoxide dismutase, 2.5. mu.g/mL catalase, 5mg/mL human serum albumin, 20. mu.g/mL transferrin, and 20. mu.g/mL sodium selenite.
"DMEM/F12 (Dulbecco's Modified Eagle Medium/Nutrient Mixture F-12)" as described herein is a 1:1 Mixture of DMEM and Ham's F-12, a widely used basal Medium that can be used to support the growth of a variety of mammalian cells, and is commercially available from a variety of suppliers (e.g., Thermo Fisher Scientific).
The FGF2 of the present invention is Fibroblast Growth Factor (Fibroblast Growth Factor) 2.
"TGF β 1" according to the present invention refers to Transforming Growth Factor β 1 (Transforming Growth Factor β 1).
The "Neuregulin 1" of the present invention may also be referred to as "NGR 1 (Neuredulin-1").
The term "IGF-1" as used herein refers to Insulin-like growth factor-1 (Insulin-like growth factor-1).
The reprogramming induction medium of the present invention contains 4-hydroxyethylpiperazineethanesulfonic acid (HEPES) at a concentration of 1 to 10mM, preferably 5 mM.
The term "ROCK inhibitor" as used herein refers to an inhibitor of Rho-associated protein kinase (ROCK), including but not limited to Thiazovivin, Y-27632, and KD-025 (also referred to as SLx-2119). One or more ROCK inhibitors may be included in the culture broth of the invention. The ROCK inhibitor may be contained in the culture solution of the present invention in an amount of 0.5 to 5. mu.M, preferably 2 to 2.5. mu.M, more preferably 2.5. mu.M.
The "MEK inhibitor" of the present invention refers to an inhibitor of mitogen-activated protein kinase (MAPK/ERK kinase), including but not limited to PD 0325901. One or more MEK inhibitors may be included in the culture fluids of the present invention. The MEK inhibitor may be present in the culture medium of the invention in an amount of 10-100nM, preferably 25-50nM, more preferably 50 nM.
The "deacetylase inhibitor" according to the present invention may also be referred to as Histone deacetylase inhibitor (HDACI), which is an agent capable of inhibiting Histone Deacetylase (HDAC) activity, and includes, but is not limited to, VPA (Valproic acid), NaBu (sodium butyrate), TSA (Trichostatin a), and SAHA (suberoylanilide hydroxamic acid). One or more deacetylase inhibitors can be included in the culture broth of the invention. The content of the deacetylase inhibitor in the culture solution of the present invention may be 0.3 to 3. mu.M, preferably 1 to 2. mu.M, more preferably 1.5 to 2. mu.M, and still more preferably 2. mu.M.
The "GSK-3 inhibitor" refers to an inhibitor of Glycogen synthase kinase-3 (Glycogen synthase kinase-3), and includes but is not limited to CHIR99021, SB 216763, TWS119, LY2090314, Tideglusib and a small molecule compound BIO (6-bromoindirubin-3-oxime, 6-bromoindirubin-3-oxide). One or more GSK-3 inhibitors may be included in the culture broth of the invention. The ROCK inhibitors, AHR inhibitors, deacetylase inhibitors, and GSK-3 inhibitors of the present invention are all commercially available from a variety of suppliers. The content of the GSK-3 inhibitor in the culture solution of the present invention may be 0.5 to 3. mu.M, preferably 1 to 2. mu.M, and more preferably 2. mu.M.
The reprogramming induction culture solution can be further added with trace elements B and C, and the trace elements B and C can participate in the synthesis of protein and the metabolism of nucleic acid and can also promote the survival of cells.
The trace element B is a composition of multiple trace elements, and preferably comprises ammonium molybdate, ammonium vanadate, manganese sulfate, nickel sulfate, sodium silicate, stannous chloride and hydrochloric acid. The Trace element B may be purchased from a supplier, such as Trace Elements B from Corning, cat # 25-022.
The trace element C is a composition of multiple trace elements, and preferably comprises aluminum chloride, barium acetate, cadmium chloride, chromium chloride, cobalt dichloride, germanium dioxide, potassium bromide, potassium iodide, rubidium chloride, silver nitrate, sodium fluoride and zirconyl chloride. The Trace element C may be purchased from a supplier, such as Trace Elements C from Corning, cat # 25-023.
The dilution concentrations of the trace elements B and C are 300-1000, preferably 500. Wherein the dilution concentration refers to the dilution factor when added to the medium, e.g., "300-" 1000 × "means 300-" 1000 × "when added to the medium, and similarly" 500 × "means 500 ×" when added to the medium.
The reprogramming system and method of the present invention include delivering human transcription factor OCT4, a p53 gene expression inhibitor, and a Cohesin gene expression inhibitor into a cell through a vector. Therefore, the reprogramming system of the present invention comprises one or more vectors for expressing human transcription factor OCT4, a p53 gene expression inhibitor, and a Cohesin gene expression inhibitor. When the vector or vectors are transferred into cells (e.g., PBMCs), the human transcription factor OCT4, the inhibitor of p53 gene expression, and the inhibitor of Cohesin gene expression can be expressed in PBMCs.
The term "OCT 4" refers to an octamer-binding transcription factor 4 (OCT-binding transcription factor 4), also known as OCT3 or OCT3/4, is a protein that contains a POU domain. See A K Ryan and M G Rosenfeld, Genes Dev. 1997, 11, 1207-. In some embodiments, the OCT3/4 is human OCT3/4 (hOCT 3/4). In some embodiments, the coding sequence of hhct 3/4 is:
ATGGCGGGACACCTGGCTTCGGATTTCGCCTTCTCGCCCCCTCCAGGTGGTGGAGGTGATGGGCCAGGGGGGCCGGAGCCGGGCTGGGTTGATCCTCGGACCTGGCTAAGCTTCCAAGGCCCTCCTGGAGGGCCAGGAATCGGGCCGGGGGTTGGGCCAGGCTCTGAGGTGTGGGGGATTCCCCCATGCCCCCCGCCGTATGAGTTCTGTGGGGGGATGGCGTACTGTGGGCCCCAGGTTGGAGTGGGGCTAGTGCCCCAAGGCGGCTTGGAGACCTCTCAGCCTGAGGGCGAAGCAGGAGTCGGGGTGGAGAGCAACTCCGATGGGGCCTCCCCGGAGCCCTGCACCGTCACCCCTGGTGCCGTGAAGCTGGAGAAGGAGAAGCTGGAGCAAAACCCGGAGGAGTCCCAGGACATCAAAGCTCTGCAGAAAGAACTCGAGCAATTTGCCAAGCTCCTGAAGCAGAAGAGGATCACCCTGGGATATACACAGGCCGATGTGGGGCTCACCCTGGGGGTTCTATTTGGGAAGGTATTCAGCCAAACGACCATCTGCCGCTTTGAGGCTCTGCAGCTTAGCTTCAAGAACATGTGTAAGCTGCGGCCCTTGCTGCAGAAGTGGGTGGAGGAAGCTGACAACAATGAAAATCTTCAGGAGATATGCAAAGCAGAAACCCTCGTGCAGGCCCGAAAGAGAAAGCGAACCAGTATCGAGAACCGAGTGAGAGGCAACCTGGAGAATTTGTTCCTGCAGTGCCCGAAACCCACACTGCAGCAGATCAGCCACATCGCCCAGCAGCTTGGGCTCGAGAAGGATGTGGTCCGAGTGTGGTTCTGTAACCGGCGCCAGAAGGGCAAGCGATCAAGCAGCGACTATGCACAACGAGAGGATTTTGAGGCTGCTGGGTCTCCTTTCTCAGGGGGACCAGTGTCCTTTCCTCTGGCCCCAGGGCCCCATTTTGGTACCCCAGGCTATGGGAGCCCTCACTTCACTGCACTGTACTCCTCGGTCCCTTTCCCTGAGGGGGAAGCCTTTCCCCCTGTCTCCGTCACCACTCTGGGCTCTCCCATGCATTCAAACTGATGATGA(SEQ ID NO:1)
the term "p 53 gene" is a tumor suppressor gene, encoding the p53 protein, p53 protein and p53 gene are well known to those skilled in the art. In some embodiments, the sequence of the shRNA that inhibits expression of the p53 gene is CCGGGTCCAGATGAAGCTCCCAGAACTCGAGTTCTGGGAGCTTCATCTGGACTTTTT (SEQ ID NO: 2)
The term "Cohesin gene" refers to the gene encoding the chromosomal adhesion protein (Cohesin) that is commonly used to link the circular structural backbone of chromosomes, and Cohesin proteins and their encoding genes are also well known to those skilled in the art. In some embodiments, the mRNA sequence of Cohesin can be found in GenBank Accession number NC-000008. In some embodiments, the sequence of shRNA that inhibits Cohesin gene expression CACCGCCTTCATGTTGTTTGCATTCCGAAGAATGCAAACAACATGAAGGC (SEQ ID NO: 3)
In the present invention, the "gene expression inhibitor" refers to an inhibitory molecule capable of inhibiting the expression of a target gene, and this molecule can inhibit the production of mRNA or degrade mRNA by a mechanism such as, for example, inhibiting the production of mRNA or degrading mRNA, so that mRNA or protein encoded by the gene cannot be expressed by the gene, or so that the amount of expressed mRNA or protein is reduced, for example, by 10% or more, 20% or more, 30% or more, 40% or more, 50% or more, 60% or more, 70% or more, 80% or more, or 90% or more, relative to the case where the inhibitor is not used. The "gene expression inhibitor" may be a molecule that inhibits the expression of a target gene by antisense inhibition or RNAi, for example, an inhibitory RNA molecule, and examples thereof include antisense RNA, siRNA, shRNA, miRNA, and the like. It will be appreciated that the inhibitor of gene expression may also be in other forms, such as a dominant negative mutant of the gene of interest, and the like. In the present invention, the gene expression inhibitor may have a specific inhibitory effect on a target gene. By "specifically inhibits" is generally meant that the inhibitor inhibits the target gene without inhibiting or substantially inhibiting other genes.
As known to those skilled in the art, the term "RNAi" refers to RNA interference, a biological process mediated by a nucleic acid molecule that inhibits or down-regulates gene expression in a cell.
As known to those skilled in the art, the term "shRNA" refers to a double-stranded structure formed from single strands of RNA that are self-complementary. shRNA constructs containing a nucleotide sequence identical to a portion of the coding or non-coding sequence of a target gene can be used to inhibit expression of the target gene. When the shRNA is expressed in a cell, it is processed through a series of steps into small interfering rna (sirna) for directing gene silencing. The shRNA can have a length of about 50-100 bp. The duplex forming portion of the shRNA may be at least 20, 21 or 22 nucleotides in length.
As known to those skilled in the art, the term "siRNA" refers to small inhibitory RNA duplexes (typically between 17-30 base pairs, but also longer base pairs such as 31-50 bp) that induce the RNA interference (RNAi) pathway.
As known to those skilled in the art, the term "antisense RNA" refers to an RNA transcript complementary to all or part of a target mRNA that can block the expression of a target gene by interfering with the processing, transport and/or translation of the mRNA. The antisense RNA can be complementary to any portion of the target mRNA, including 5 'non-coding sequences, 3' non-coding sequences, introns, and coding sequences.
As known to those skilled in the art, the term "miRNA" refers to a single-stranded RNA molecule (or synthetic derivative thereof) that is capable of binding to a target gene (mRNA or DNA) and regulating the expression of that gene.
The term "vector" in the present invention generally refers to an expression vector, which can be used to express a target gene. In a preferred embodiment, the vector is a non-integrating vector, i.e., does not integrate into the genome of the cell into which it is transfected. Non-integrative vectors are well known to those skilled in the art, such as, but not limited to, plasmids, non-integrative recombinant viral vectors (e.g., non-integrative adenoviral vectors or adeno-associated viral vectors, etc.). Expression vectors typically include a gene of interest to be expressed and regulatory sequences, such as promoters, enhancers, post-transcriptional regulatory sequences, and the like, operably linked to the gene of interest.
In the invention, the human transcription factor OCT4, the p53 gene expression inhibitor and the Cohesin gene expression inhibitor can be expressed in one vector, two vectors or three vectors respectively. For example, any two of the human transcription factor OCT4, the p53 gene expression inhibitor, and the Cohesin gene expression inhibitor (e.g., human transcription factor OCT4 and p53 gene expression inhibitor) may be expressed using one vector, and the other vector may be expressed using the other vector. For another example, three vectors each expressing one of human transcription factor OCT4, a p53 gene expression inhibitor, and a Cohesin gene expression inhibitor may be used.
The term "transfection" refers to the introduction of a nucleic acid molecule, such as a DNA or RNA molecule, into a cell, preferably a eukaryotic cell, for example into a mammalian cell, such as into a PBMC cell. "transfection" can be carried out by any method known to those skilled in the art for transferring DNA (e.g., plasmid vector) into cells, for example, by a competent cell method, an artificial liposome method, a microinjection method, a particle gun method, or an electroporation method (also referred to as electroporation, electrotransformation, or electrotransfection in the present invention), and the like.
After the one or more vectors are used for transfecting the peripheral blood mononuclear cells, the transfected mononuclear cells can express human transcription factor OCT4, a p53 gene expression inhibitor and a Cohesin gene expression inhibitor, wherein the expression of the p53 gene expression inhibitor and the Cohesin gene expression inhibitor in the peripheral blood mononuclear cells can inhibit the expression of a p53 gene and a Cohesin gene in the peripheral blood mononuclear cells.
The peripheral blood mononuclear cells may be cultured in advance for an appropriate period of time to activate the cells, for example, for 1 to 3 days, preferably 2 days, before transfection. The pre-culture conditions areAs is well known to those skilled in the art, for example, 3-7% CO at 35-39 deg.C, preferably 37 deg.C 2 Preferably 5% CO 2 、0.5-2×10 6 The cells/well are preferably 1X 10 6 The density of cells/well is cultured. The pre-incubation may be performed using any suitable vessel, for example, the incubation may be performed in an ultra-low adsorption well plate. The preculture may be carried out using any suitable medium, for example, StemPro-34 medium.
When the vector or vectors are used to transfect peripheral blood mononuclear cells, the amount of the vector to be used may be determined in accordance with the actual situation, and may be, for example, 1 to 3. mu.g of the total amount of the vector per 1.0X 10 5 Peripheral blood mononuclear cells, preferably 2. mu.g of total vector/1.0X 10 5 Individual cells of peripheral blood. When more than one carrier is used, the ratio between the carriers may be determined as the case may be, and may be, for example, an equal mass ratio, i.e., a mass ratio of 1: 1.
After transfection, the peripheral blood mononuclear cells transfected by the vector may be cultured directly using the reprogramming induction culture solution of the present invention, or may be cultured in another suitable medium (e.g., StemPro-34 medium) for a certain period of time, and then replaced with the reprogramming induction culture solution of the present invention. For example, in some embodiments, transfected peripheral blood mononuclear cells may be cultured for 1-3 days, preferably 2 days, using other suitable media, and then replaced with the reprogramming induction medium of the present invention. The culture conditions may be those commonly used in the art for reprogramming PBMC, and may be, for example, 35 to 39 ℃ preferably 37 ℃ and 3 to 7% CO 2 Preferably 5% CO 2 、3~7% O 2 Preferably 5% O 2 The culturing may be carried out using any suitable vessel, e.g.on a laminin-coated plate, e.g.1. mu.g/cm coated 2 The method of Lamnin521 on a plate.
The conditions for culturing peripheral blood mononuclear cells in the reprogramming induction culture solution of the present invention may be conditions generally used in the art for reprogramming PBMC, and may be, for example, 35 to 39 ℃, preferably 37 ℃ and 3 to 7% CO 2 Preferably 5% CO 2 、3~7% O 2 Preferably 5% O 2 Under the conditions of (1). During the culture, the fresh reprogramming induction culture solution may be replaced at an appropriate time according to circumstances, and for example, the fresh reprogramming induction culture solution may be replaced every 1 to 3 days, preferably every 2 days.
The skilled person knows how to judge the occurrence of Induced Pluripotent Stem Cell (iPSC) clones, for example, morphological judgment can be performed according to unique characteristics of human embryonic stem cells (ES cells), for example, cell clones with flat, homogeneous and distinct boundary contour, i.e., induced pluripotent stem cell clones, occur.
After the induced pluripotent stem cell clone has appeared, the culture may be continued in the reprogramming inducer of the present invention in order to promote the growth thereof, and the culture conditions are well known to those skilled in the art, and may be, for example, 35 to 39 ℃ preferably 37 ℃ with 3 to 7% CO 2 Preferably 5% CO 2 For example, the stem cell culture medium may be replaced every 1 to 2 days, preferably 1 day, during the culture.
After the induced pluripotent stem cell clones have grown, individual clones may be picked and plated for further culture, and optionally subcultured to purify the induced pluripotent stem cells and expand their numbers for subsequent manipulation (e.g., detection or further use). The conditions for culturing and subculturing of induced pluripotent stem cells are well known to those skilled in the art, and for example, the induced pluripotent stem cell clones may be picked and seeded into a new matrigel-coated plate at 35-39 deg.C, preferably 37 deg.C, with 3-7% CO 2 Preferably 5% CO 2 The subculture is continued under the condition that the medium can be replaced every 1-2 days, preferably every day, and every 3-5 days, preferably every 3 days, wherein the medium can be the reprogramming induction culture solution or other stem cell culture medium of the present invention.
Examples
The technical solution of the present invention will be described in further detail below by way of examples with reference to the accompanying drawings, but the present invention is not limited to the following examples.
Example 1 cloning by different reprogramming methods
Experimental groups 1-3 PBMC cells were transfected (i.e., electroporated) using reprogramming plasmid electroporation, comprising the steps of:
step 1): the frozen PBMC is taken and rapidly thawed in a water bath at 37 ℃. Cells were gently pipetted 1-2 times using a 1mL pipette and transferred to a 15mL centrifuge tube. Rinsing the frozen tube with 1mL of room temperature preheated StemPro-34 basal medium for 1 time, transferring the rinsed cell suspension into the centrifuge tube, and adding 9mL of StemPro-34 basal medium into the centrifuge tube drop by drop; centrifuge at 200g for 3 min. The supernatant was discarded, cells were resuspended in StemPro ™ full-medium resuspension medium, counted and plated onto ultra-low adsorption 24-well plates. PBMC at 37 ℃ with 5% CO 2 Culturing in an incubator for 2 days.
Step 2): PBMC were electrotransfected, approximately 1 hour prior to adding 2mL StemPro ™ complete medium to 1 well of a 6-well plate, and the plate was equilibrated in a 37 ℃ incubator. Collecting PBMC in a 24-well plate, rinsing with 1mL of StemPro-34 basal medium, centrifuging at 200g for 3 min; the supernatant was discarded, washed with DPBS resuspension, and counted. Centrifuging at 200g for 3 min; during centrifugation, 5mL of electrotransfer buffer E was added to the electrotransfer cup. Setting the electric transfer parameters as follows: 1650V, 10ms, 3 pulses; after centrifugation, the DPBS was aspirated and the cells were resuspended in a ratio of two plasmids equal (mass ratio) and in a total amount of 2. mu.g plasmid/1.0X 10 5 Adding reprogramming plasmids into blood cells, uniformly mixing, completing electrotransfer according to electrotransfer parameters, and recording as day 0; the cells after electroporation were inoculated to 1. mu.g/cm 2 The Lamnin 521-coated 6-well tissue culture plate containing StemPro-34 Medium was cultured at 37 ℃ in 5% CO 2 、5%O 2 Culturing for 2 days under the condition;
step 3): on day 2 after electroporation, fresh StemPro ™ chamber-34 medium was replaced; the reprogramming induction medium was replaced every 2 days thereafter and 5% CO at 37 deg.C 2 、5%O 2 Culturing under the condition until cloning occurs;
step 4): induced pluripotent stem cell clone appears 10-12 days after electrotransfer, and reprogramming induction culture is replaced based on 37 ℃ and 5% CO 2 Continuously culturing under the condition, replacing the culture medium every 1 day during the culture, and calculating the number of the appeared induced pluripotent stem cell clones;
step 5): when the reprogrammed pluripotent stem cell clone grows to be suitable for picking, picking the induced pluripotent stem cell clone and inoculating the induced pluripotent stem cell clone into a new culture plate coated with matrigel at 37 ℃ and 5% CO 2 And continuously subculturing under the condition, wherein during the culture, the reprogramming induction culture medium is replaced once every 1 day, and the subculturing is performed once every 3 days.
The reprogramming plasmids and reprogramming induction media used in experimental groups 1-3 are shown in Table 1, wherein the plasmid pCXLE-hOCT3/4-shP53 is from Addgene, #27077), and the coding sequence of hOCT3/4 contained in the plasmid is:
ATGGCGGGACACCTGGCTTCGGATTTCGCCTTCTCGCCCCCTCCAGGTGGTGGAGGTGATGGGCCAGGGGGGCCGGAGCCGGGCTGGGTTGATCCTCGGACCTGGCTAAGCTTCCAAGGCCCTCCTGGAGGGCCAGGAATCGGGCCGGGGGTTGGGCCAGGCTCTGAGGTGTGGGGGATTCCCCCATGCCCCCCGCCGTATGAGTTCTGTGGGGGGATGGCGTACTGTGGGCCCCAGGTTGGAGTGGGGCTAGTGCCCCAAGGCGGCTTGGAGACCTCTCAGCCTGAGGGCGAAGCAGGAGTCGGGGTGGAGAGCAACTCCGATGGGGCCTCCCCGGAGCCCTGCACCGTCACCCCTGGTGCCGTGAAGCTGGAGAAGGAGAAGCTGGAGCAAAACCCGGAGGAGTCCCAGGACATCAAAGCTCTGCAGAAAGAACTCGAGCAATTTGCCAAGCTCCTGAAGCAGAAGAGGATCACCCTGGGATATACACAGGCCGATGTGGGGCTCACCCTGGGGGTTCTATTTGGGAAGGTATTCAGCCAAACGACCATCTGCCGCTTTGAGGCTCTGCAGCTTAGCTTCAAGAACATGTGTAAGCTGCGGCCCTTGCTGCAGAAGTGGGTGGAGGAAGCTGACAACAATGAAAATCTTCAGGAGATATGCAAAGCAGAAACCCTCGTGCAGGCCCGAAAGAGAAAGCGAACCAGTATCGAGAACCGAGTGAGAGGCAACCTGGAGAATTTGTTCCTGCAGTGCCCGAAACCCACACTGCAGCAGATCAGCCACATCGCCCAGCAGCTTGGGCTCGAGAAGGATGTGGTCCGAGTGTGGTTCTGTAACCGGCGCCAGAAGGGCAAGCGATCAAGCAGCGACTATGCACAACGAGAGGATTTTGAGGCTGCTGGGTCTCCTTTCTCAGGGGGACCAGTGTCCTTTCCTCTGGCCCCAGGGCCCCATTTTGGTACCCCAGGCTATGGGAGCCCTCACTTCACTGCACTGTACTCCTCGGTCCCTTTCCCTGAGGGGGAAGCCTTTCCCCCTGTCTCCGTCACCACTCTGGGCTCTCCCATGCATTCAAACTGATGATGA(SEQ ID NO:1)。
the sequence of shP53 contained in this plasmid is:
CCGGGTCCAGATGAAGCTCCCAGAACTCGAGTTCTGGGAGCTTCATCTGGACTTTTT(SEQ ID NO:2)。
plasmid pCXLE-sh Cohesin is obtained by integrating the DNA sequence of shRNA of Cohesin into pCXLE (Addgene, # 37626) vector by restriction endonuclease BamHI, the mRNA sequence of Cohesin can be referred to GenBank Accession number XM-011512331.2, and the DNA sequence of shRNA of Cohesin is used as follows:
CACCGCCTTCATGTTGTTTGCATTCCGAAGAATGCAAACAACATGAAGGC(SEQ ID NO:3)。
the components of the experimental reprogramming induction culture solution are as follows: IX-PSC basal medium (containing DMEM/F12, 20. mu.g/mL ascorbic acid, 40ng/mL FGF2, 1ng/mL TGF β 1, 6ng/mL neuregulin 1, 15ng/mL IGF-1, 2.5. mu.g/mL superoxide dismutase, 2.5. mu.g/mL catalase, 5mg/mL human serum albumin, 20. mu.g/mL transferrin, and 20. mu.g/mL sodium selenite), and 2. mu.M CHIR99021, 2. mu.M VPA, 50nM PD0325901, 2.5. mu.M Thiazovin, and 500. times Trace element B, 500. times Trace element C, and 2mM 4-hydroxyethylpiperazine ethanesulfonic acid. Wherein the Trace element B is Trace elements B, manufactured by Corning, with a product number of 25-022; trace elements C is Trace elements C, Corning, product number 25-023. The electrotransformation instrument system is purchased from Saimei Feishale science and technology (China) Co., Ltd. (MPK 5000, MPK 10096), and the electrotransformation conditions are 1650V and 10 ms.
The reprogramming method of Experimental group 4 was that Sendai virus (Gibco, A16517, A16518) was transfected into PBMC cells, and the reprogramming induction medium was a commercially available induced pluripotent Stem cell Medium mTeSR TM 1 (manufacturer stemcel Technologies inc., cat 85850), see table 1.
TABLE 1 design of different reprogramming methods
Experimental group | Reprogramming mode | Reprogramming induction medium |
Experimental group 1 | pCXLE-hOCT3/4-shP53 and pCXLE-sh Cohesin transfections | mTeSR TM 1 |
Experimental group 2 | pCXLE-hOCT3/4-shP53 transfection | Experimental reprogramming induction culture solution |
Experimental group 3 | pCXLE-hOCT3/4-shP53 and pCXLE-sh Cohesin transfection | Experimental reprogramming induction culture solution |
Experimental group 4 | Sendai virus transfection | mTeSR TM 1 |
In the reprogramming process, when cell clones with flat and homogeneous shapes and obvious boundary outlines appear, namely induced pluripotent stem cell clones appear, the counting of the appeared induced pluripotent stem cell clones (hereinafter referred to as clones) of each experimental group is counted, and the effectiveness of the selected plasmids and reagents on reprogramming efficiency is researched.
FIG. 2 shows the results of different experimental groups per 1X 10 6 Clones were generated after reprogramming of individual PBMCs. As can be seen from FIG. 2, the induced pluripotent stem cells obtained from the experimental groups 1-3 were all greater than those obtained from the experimental group 4, whereas the induced pluripotent stem cells obtained from the experimental group 3 were the most when the experimental group 3 was compared with the experimental groups 1 and 2, which indicates that the above-mentioned experimental reprogramming induction culture solution and pCXLE-sh cohesin exert a critical effect on stem cell induction. Comparing the experimental group 3 with the experimental group 4, it can be seen that the experimental group 3 has an average of 15 more clones than the experimental group 4. Currently, the accepted method for reprogramming blood cells is Sendai virus transfection and at mTeSR TM 1 to further obtain stem cell clone, mTeSR TM 1 is a classical inducerThe experiments prove that the combination of the pCXLE-sh cohesin plasmid and the experimental reprogramming induction culture solution can obtain higher pluripotent stem cell clone yield.
Example 2 identification of the potential of induced pluripotent Stem cells to differentiate into three germ layer tissue
EB differentiation function identification was performed on the induced pluripotent stem cells obtained from step 1) -5) of experiment group 3 in example 1, and the specific steps were as follows: inducing the obtained cells to differentiate into EB by using KOSR EB differentiation complete culture medium, and detecting ectodermal marker protein beta-Tubulin III, mesodermal marker protein SMA and endodermal marker protein AFP by carrying out immunofluorescence staining on the EB.
FIG. 3 shows the results of differentiation identification of the obtained cell line EB, which illustrates the potential of differentiation into tissues of each of the three germ layers of the cell line obtained by the reprogramming method.
Example 3 induced pluripotent Stem cell pluripotent immunofluorescence assay
Cloned cells obtained from induced pluripotent stem cells obtained in experimental group 3 of example 1 through steps 1) -5) were plated with mTeSR in a 4-well plate TM 1 complete culture medium culture, observing the cell density and the cell growth state under a 4-fold lens and a 10-fold lens, observing whether the cell is polluted by bacteria and fungi under a 20-fold lens, observing the cell density, if the cell density reaches 30%, carrying out non-pluripotent immunofluorescence detection, and detecting stem cell characteristic factors of Nanog, Oct4, Sox2 and SSEA 4.
FIG. 4 shows the results of the measurement of pluripotent immunofluorescence of the obtained cells, which revealed that the obtained cells expressed the pluripotent genes Nanog, Oct4, Sox2 and SSEA4, and were pluripotent stem cells.
Example 4 flow assay for induced pluripotent Stem cell pluripotency
Using mTeSR TM 1 complete culture medium culture of cells obtained from induced pluripotent stem cells obtained from step 1) -5) of the experimental group 3 of example 1, when the cells grow to about 90%, using Accutase to digest the cells, gently transferring the cell suspension into a 15ml centrifuge tube, mixing the cells uniformly, counting the cells, and ensuring that 2 × 10 cells are obtained for each 1 Marker flow 6 And (4) cells. Centrifuging to remove supernatant, adding 90% cold into centrifuge tubeThe cells were fixed in methanol and treated for flow detection of surface antigens Nanog, Oct4 and SSEA4 on a flow cytometer.
FIG. 5 shows the flow-through identification results of the obtained cell lines, which indicate that the cell purity of the cells expressing pluripotency genes Nanog, Oct4 and SSEA4 is more than 97%, and thus, the purity of the obtained induced pluripotent stem cells is high.
Example 5 induced pluripotent Stem cell karyotype identification
The method comprises the following specific steps: using mTeSR TM 1 complete medium culture of induced pluripotent stem cells obtained from the induced pluripotent stem cells obtained through steps 1) to 5) of experiment group 3 in example 1. The cell number is measured by taking cells in one hole of a 6-hole plate, karyotype analysis can be carried out when the cell density reaches about 70%, 0.2 mu g/ml colchicine is added into each hole, the treatment is carried out for 2 hours, after Accutase digestion, KCL hypotonic is carried out for 30 minutes, and karyotype detection is carried out after the treatment by adding a fixing solution (glacial methanol: acetic acid = 3: 1).
FIG. 6 shows the results of karyotype examination of the obtained cell lines, which revealed that the karyotype analysis of the obtained cell lines was normal.
The present invention can reprogram human induced pluripotent stem cells from peripheral blood mononuclear cells taken from the blood of a patient. The method for inducing the pluripotent stem cells has the advantages that: 1) the materials are easy to obtain, the mononuclear cells of the peripheral blood can be taken from the blood of a patient, the number is not limited, and moral disputes can be avoided; 2) human transcription factor OCT4, p53 gene expression inhibitor and Cohesin gene expression inhibitor are adopted during cell reprogramming, and the needed factors are few; 3) the electrotransfer reprogramming induction culture medium is added with a GSK-3 inhibitor, an acetylase inhibitor, an MEK inhibitor, a ROCK inhibitor and 4-hydroxyethyl piperazine ethanesulfonic acid, all components play a role in a synergistic manner, and the cell induction reprogramming efficiency is improved while cell growth and proliferation are maintained in the iPS cell reprogramming process; compared with the prior art, the method greatly shortens the induction time, only needs 10 to 12 days, and can be used for large-scale industrial preparation.
Sequence listing
<110> Shanghai Isale Biotech Co., Ltd
<120> System and method for reprogramming peripheral blood mononuclear cells to induced pluripotent stem cells
<130> DPC200751ASE
<160> 3
<170> SIPOSequenceListing 1.0
<210> 1
<211> 1089
<212> DNA
<213> Artificial Sequence
<400> 1
atggcgggac acctggcttc ggatttcgcc ttctcgcccc ctccaggtgg tggaggtgat 60
gggccagggg ggccggagcc gggctgggtt gatcctcgga cctggctaag cttccaaggc 120
cctcctggag ggccaggaat cgggccgggg gttgggccag gctctgaggt gtgggggatt 180
cccccatgcc ccccgccgta tgagttctgt ggggggatgg cgtactgtgg gccccaggtt 240
ggagtggggc tagtgcccca aggcggcttg gagacctctc agcctgaggg cgaagcagga 300
gtcggggtgg agagcaactc cgatggggcc tccccggagc cctgcaccgt cacccctggt 360
gccgtgaagc tggagaagga gaagctggag caaaacccgg aggagtccca ggacatcaaa 420
gctctgcaga aagaactcga gcaatttgcc aagctcctga agcagaagag gatcaccctg 480
ggatatacac aggccgatgt ggggctcacc ctgggggttc tatttgggaa ggtattcagc 540
caaacgacca tctgccgctt tgaggctctg cagcttagct tcaagaacat gtgtaagctg 600
cggcccttgc tgcagaagtg ggtggaggaa gctgacaaca atgaaaatct tcaggagata 660
tgcaaagcag aaaccctcgt gcaggcccga aagagaaagc gaaccagtat cgagaaccga 720
gtgagaggca acctggagaa tttgttcctg cagtgcccga aacccacact gcagcagatc 780
agccacatcg cccagcagct tgggctcgag aaggatgtgg tccgagtgtg gttctgtaac 840
cggcgccaga agggcaagcg atcaagcagc gactatgcac aacgagagga ttttgaggct 900
gctgggtctc ctttctcagg gggaccagtg tcctttcctc tggccccagg gccccatttt 960
ggtaccccag gctatgggag ccctcacttc actgcactgt actcctcggt ccctttccct 1020
gagggggaag cctttccccc tgtctccgtc accactctgg gctctcccat gcattcaaac 1080
tgatgatga 1089
<210> 2
<211> 57
<212> DNA
<213> Artificial Sequence
<400> 2
ccgggtccag atgaagctcc cagaactcga gttctgggag cttcatctgg acttttt 57
<210> 3
<211> 50
<212> DNA
<213> Artificial Sequence
<400> 3
caccgccttc atgttgtttg cattccgaag aatgcaaaca acatgaaggc 50
Claims (19)
1. A reprogramming system for reprogramming Peripheral Blood Mononuclear Cells (PBMCs) into human induced pluripotent stem cells, comprising a reprogramming induction culture solution and one or more vectors capable of expressing human transcription factor OCT4, a p53 gene expression inhibitor and a Cohesin gene expression inhibitor; wherein the reprogramming induction culture fluid comprises IX-PSC basal medium, GSK-3 inhibitor, deacetylase inhibitor, MEK inhibitor, ROCK inhibitor and 4-hydroxyethyl piperazine ethanesulfonic acid; wherein the IX-PSC basal medium is prepared by taking DMEM/F12 as a basal medium and adding 10-30 mug/mL ascorbic acid, 30-50 ng/mL FGF2, 0.5-2 ng/mL TGF beta 1, 3-9 ng/mL neuregulin 1, 10-20 ng/mL IGF-1, 2-3 mug/mL superoxide dismutase, 2-3 mug/mL catalase, 1-10mg/mL human serum albumin, 10-30 mug/mL transferrin and 10-30 mug/mL sodium selenite;
wherein the inhibitor of p53 gene expression and inhibitor of Cohesin gene expression are inhibitory RNA molecules;
wherein the GSK-3 inhibitor is CHIR99021, the deacetylase inhibitor is VPA, the MEK inhibitor is PD0325901, and the ROCK inhibitor is Thiazovivin;
wherein in the reprogramming induction culture solution, the concentration of the human serum albumin is 1-10mg/mL, the concentration of the GSK-3 inhibitor is 0.5-3 mu M, the concentration of the deacetylase inhibitor is 0.3-3 mu M, the concentration of the MEK inhibitor is 10-100nM, the concentration of the ROCK inhibitor is 0.5-5 mu M, and the concentration of the 4-hydroxyethyl piperazine ethanesulfonic acid is 1-10 mM.
2. The reprogramming system of claim 1, wherein the inhibitory RNA molecule is an antisense RNA, miRNA, siRNA or shRNA.
3. The reprogramming system of claim 2, wherein the inhibitory RNA molecule is an shRNA.
4. The reprogramming system of any one of claims 1-3, wherein said one or more vectors are non-genomically integrated vectors.
5. The reprogramming system of claim 4, wherein the one or more vectors are plasmids.
6. The reprogramming system of any one of claims 1 to 3, wherein in the reprogramming induction culture fluid, the human serum albumin concentration is 5mg/mL, the GSK-3 inhibitor concentration is 2 μ M, the deacetylase inhibitor concentration is 2 μ M, the MEK inhibitor concentration is 50nM, the ROCK inhibitor concentration is 2.5 μ M, and the 4-hydroxyethylpiperazine ethanesulfonic acid concentration is 5 mM.
7. A method of reprogramming Peripheral Blood Mononuclear Cells (PBMCs) to induced pluripotent stem cells, the method comprising:
i) transfecting peripheral blood mononuclear cells with one or more vectors, wherein the one or more vectors can express human transcription factors OCT4, p53 gene expression inhibitors and Cohesin gene expression inhibitors, and obtaining peripheral blood mononuclear cells expressing the human transcription factors OCT4, p53 gene expression inhibitors and the Cohesin gene expression inhibitors;
ii) culturing the peripheral blood mononuclear cells in a reprogramming induction culture solution to obtain induced pluripotent stem cells;
wherein the reprogramming induction culture fluid comprises IX-PSC basal medium, GSK-3 inhibitor, deacetylase inhibitor, MEK inhibitor, ROCK inhibitor and 4-hydroxyethyl piperazine ethanesulfonic acid; wherein the IX-PSC basal medium is prepared by taking DMEM/F12 as a basal medium and adding 10-30 mug/mL ascorbic acid, 30-50 ng/mL FGF2, 0.5-2 ng/mL TGF beta 1, 3-9 ng/mL neuregulin 1, 10-20 ng/mL IGF-1, 2-3 mug/mL superoxide dismutase, 2-3 mug/mL catalase, 1-10mg/mL human serum albumin, 10-30 mug/mL transferrin and 10-30 mug/mL sodium selenite;
wherein the inhibitor of p53 gene expression and inhibitor of Cohesin gene expression are inhibitory RNA molecules;
wherein the GSK-3 inhibitor is CHIR99021, the deacetylase inhibitor is VPA, the MEK inhibitor is PD0325901, and the ROCK inhibitor is Thiazovivin;
wherein in the reprogramming induction culture solution, the concentration of the human serum albumin is 1-10mg/mL, the concentration of the GSK-3 inhibitor is 0.5-3 mu M, the concentration of the deacetylase inhibitor is 0.3-3 mu M, the concentration of the MEK inhibitor is 10-100nM, the concentration of the ROCK inhibitor is 0.5-5 mu M, and the concentration of the 4-hydroxyethyl piperazine ethanesulfonic acid is 1-10 mM.
8. The method of claim 7, wherein the inhibitory RNA molecule is an antisense RNA, miRNA, siRNA, or shRNA.
9. The method of claim 8, wherein the inhibitory RNA molecule is an shRNA.
10. The method of any one of claims 7-9, wherein the one or more vectors are non-genomically integrated vectors.
11. The method of claim 10, wherein the one or more vectors are plasmids.
12. The method of any one of claims 7-9, wherein the total concentration of the one or more carriers is 1-3 μ g carrier/1.0 x 10 5 Blood cells.
13. The method of claim 12, wherein the total concentration of the one or more carriers is 2 μ g carrier/1.0 x 10 5 Blood cells.
14. The method of any one of claims 7-9, wherein in the reprogramming induction culture fluid, the human serum albumin concentration is 5mg/mL, the GSK-3 inhibitor concentration is 2 μ Μ, the deacetylase inhibitor concentration is 2 μ Μ, the MEK inhibitor concentration is 50nM, the ROCK inhibitor concentration is 2.5 μ Μ, and the 4-hydroxyethylpiperazine ethanesulfonic acid concentration is 5 mM.
15. The method according to any one of claims 7-9, comprising the steps of:
step 1): peripheral Blood Mononuclear Cells (PBMC) are treated at 35-39 ℃ with 3-7% CO 2 And 0.5 to 2X 10 6 Culturing the cells/well in a well plate for 1-3 days;
step 2): according to 1-3. mu.g/1.0X 10 of each carrier 5 Blood cells the one or more vectors are transfected into the peripheral blood mononuclear cells obtained in step 1), and the cells are inoculated onto a tissue culture plate and cultured in StemPro-34 medium at 35-39 ℃ with 3-7% CO 2 And 3 to 7% of O 2 Culturing for 1-3 days;
step 3): day 2-3 after transfection, fresh StemPro-34 medium was used instead; then replacing the reprogramming induction culture solution every 1-3 days, and carrying out 3-7% CO treatment at the temperature of 35-39 DEG C 2 And 3 to 7% of O 2 Until the induction of pluripotent stem cell clone appears.
16. The method of claim 15, comprising the steps of:
step 1): peripheral Blood Mononuclear Cells (PBMC) were incubated at 37 ℃ with 5% CO 2 And 1X 10 6 Culturing the cells/well in a well plate for 2 days;
step 2): according to a total concentration of 2. mu.g carrier/1.0X 10 5 Blood cells the vector or vectors are transfected into peripheral blood mononuclear cells, which are plated onto tissue culture plates using StemPro-34 medium at 37 ℃ with 5% CO 2 And 5% of O 2 Culturing for 2 days;
step 3): day 2 post-transfection, fresh StemPro-chambers-34 were replaced; thereafter, the reprogramming induction medium was replaced every 2 days at 37 ℃ with 5% CO 2 And 5% of O 2 Until the induction of pluripotent stem cell clone appears.
17. The method of claim 16, the method further comprising: step 4): replacing the reprogramming induction culture solution after the induced pluripotent stem cell clone appears, and carrying out the reprogramming induction culture solution at 35-39 ℃ and 3-7% CO 2 The culture was continued with replacement of the stem cell medium every 1 to 2 days during the culture.
18. A kit comprising the reprogramming system of any one of claims 1 to 6.
19. Use of the reprogramming system of any one of claims 1 to 6 or the kit of claim 18 for inducing reprogramming of Peripheral Blood Mononuclear Cells (PBMCs) into human induced pluripotent stem cells.
Priority Applications (1)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
CN202210541037.XA CN114645023B (en) | 2022-05-18 | 2022-05-18 | System and method for reprogramming peripheral blood mononuclear cells to induced pluripotent stem cells |
Applications Claiming Priority (1)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
CN202210541037.XA CN114645023B (en) | 2022-05-18 | 2022-05-18 | System and method for reprogramming peripheral blood mononuclear cells to induced pluripotent stem cells |
Publications (2)
Publication Number | Publication Date |
---|---|
CN114645023A CN114645023A (en) | 2022-06-21 |
CN114645023B true CN114645023B (en) | 2022-09-20 |
Family
ID=81996500
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
CN202210541037.XA Active CN114645023B (en) | 2022-05-18 | 2022-05-18 | System and method for reprogramming peripheral blood mononuclear cells to induced pluripotent stem cells |
Country Status (1)
Country | Link |
---|---|
CN (1) | CN114645023B (en) |
Families Citing this family (5)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
CN113862221B (en) * | 2021-10-27 | 2024-04-30 | 上海爱萨尔生物科技有限公司 | Culture solution and culture method of peripheral blood mononuclear cells |
CN115772505B (en) * | 2023-02-13 | 2023-05-19 | 淇嘉科技(天津)有限公司 | Culture medium and method for promoting reprogramming of somatic cells into induced pluripotent stem cells |
CN117511847A (en) * | 2023-11-06 | 2024-02-06 | 西安添易秦悦生物科技有限公司 | A method for preparing pancreatic islet cell clusters from autologous induced pluripotent stem cells in vitro |
CN118272437B (en) * | 2024-04-11 | 2024-12-17 | 广州瑞铂茵健康科技有限公司 | Induced pluripotent stem cell, and preparation method and application thereof |
CN118325823B (en) * | 2024-05-17 | 2024-10-25 | 广东康盾高新技术产业集团股份公司 | Inducer suitable for somatic reprogramming and application method |
Family Cites Families (5)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO2013177228A1 (en) * | 2012-05-22 | 2013-11-28 | Loma Linda University | Generation of integration/transgene-free stem cells |
WO2013176233A1 (en) * | 2012-05-23 | 2013-11-28 | 国立大学法人京都大学 | Highly efficient method for establishing artificial pluripotent stem cell |
CN108265025A (en) * | 2017-12-28 | 2018-07-10 | 安徽中盛溯源生物科技有限公司 | A kind of reprogramming culture medium of mankind's induced multi-potent stem cell |
CN108642083B (en) * | 2018-04-28 | 2022-03-22 | 安徽中盛溯源生物科技有限公司 | Reprogramming method for efficiently inducing T cells into pluripotent stem cells |
CN113862221B (en) * | 2021-10-27 | 2024-04-30 | 上海爱萨尔生物科技有限公司 | Culture solution and culture method of peripheral blood mononuclear cells |
-
2022
- 2022-05-18 CN CN202210541037.XA patent/CN114645023B/en active Active
Also Published As
Publication number | Publication date |
---|---|
CN114645023A (en) | 2022-06-21 |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
CN114645023B (en) | System and method for reprogramming peripheral blood mononuclear cells to induced pluripotent stem cells | |
Drozd et al. | Generation of human iPSCs from cells of fibroblastic and epithelial origin by means of the oriP/EBNA-1 episomal reprogramming system | |
JP2021035399A (en) | Generation of induced pluripotent stem cells from a small amount of peripheral blood | |
US8962331B2 (en) | Method of making induced pluripotent stem cell from adipose stem cells using minicircle DNA vectors | |
EP3152295B1 (en) | A novel and efficient method for reprogramming immortalized lymphoblastoid cell lines to induced pluripotent stem cells | |
JP2012509072A (en) | Reprogramming cells to a pluripotent state | |
JP2017525351A (en) | Culture medium for pluripotent stem cells | |
CN103555661B (en) | A kind of serum-free, multipotential stem cell cultural method without feeder layer | |
CN110713973B (en) | A medium combination and method for inducing pluripotent stem cells to differentiate into mesenchymal stem cells | |
KR102784455B1 (en) | Reprogramming vector | |
CN115975914A (en) | Method for inducing pluripotent stem cells by reprogramming of chemical small molecule drugs | |
KR102143320B1 (en) | METHODS FOR DIRECT CONVERTION OF HUMAN URINE CELLS INTO NEURAL STEM CELLS USING A SYNTHETIC mRNA | |
CN101684455A (en) | Application of vitamin C in preparation of induced multi-potent stem cells and culture of embryonic stem cells | |
US20130217125A1 (en) | Culture method | |
JP2015534830A (en) | MicroRNA and cell reprogramming | |
CN111100842A (en) | Neural stem cells carrying tumor-related genes and preparation method and application thereof | |
CN106085951B (en) | Method for establishing tree shrew spermatogonial stem cell line capable of being continuously passaged | |
WO2022097716A1 (en) | Method for producing reversibly immortalized cell | |
CN109825468B (en) | Method for efficiently inducing fish pluripotent stem cells and induction medium used for method | |
Alvarez et al. | Nanog overexpression allows human mesenchymal stem cells to differentiate into neural cells——Nanog transdifferentiates mesenchymal stem cells | |
US20130273655A1 (en) | ENGRAFTMENT POTENTIAL OF EMBRYONIC STEM CELLS AND PLURIPOTENT STEM CELLS USING MICRO RNAs | |
JP7536245B2 (en) | Method for generating oligodendrocytes | |
CN117487743B (en) | Chemical inducer for inducing chick embryo fibroblast to be chick pluripotent stem cell and induction method | |
US20210010030A1 (en) | Method for reprogramming somatic cells | |
JP2023090959A (en) | Method for producing stem cell, and method of reducing risk of transforming into cancer cell |
Legal Events
Date | Code | Title | Description |
---|---|---|---|
PB01 | Publication | ||
PB01 | Publication | ||
SE01 | Entry into force of request for substantive examination | ||
SE01 | Entry into force of request for substantive examination | ||
GR01 | Patent grant | ||
GR01 | Patent grant |