WO2013108949A1 - Procédé de préparation de cellules progénitrices périvasculaires dérivées de cellules souches embryonnaires et composition pour thérapie cellulaire les contenant - Google Patents
Procédé de préparation de cellules progénitrices périvasculaires dérivées de cellules souches embryonnaires et composition pour thérapie cellulaire les contenant Download PDFInfo
- Publication number
- WO2013108949A1 WO2013108949A1 PCT/KR2012/000769 KR2012000769W WO2013108949A1 WO 2013108949 A1 WO2013108949 A1 WO 2013108949A1 KR 2012000769 W KR2012000769 W KR 2012000769W WO 2013108949 A1 WO2013108949 A1 WO 2013108949A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- cells
- progenitor cells
- perivascular
- cell
- disease
- Prior art date
Links
- 210000000130 stem cell Anatomy 0.000 title claims abstract description 141
- 238000000034 method Methods 0.000 title claims abstract description 68
- 239000000203 mixture Substances 0.000 title claims abstract description 25
- 210000001671 embryonic stem cell Anatomy 0.000 title claims description 19
- 238000002659 cell therapy Methods 0.000 title abstract description 12
- 210000004027 cell Anatomy 0.000 claims abstract description 153
- 208000024172 Cardiovascular disease Diseases 0.000 claims abstract description 34
- 208000017442 Retinal disease Diseases 0.000 claims abstract description 33
- 210000002242 embryoid body Anatomy 0.000 claims abstract description 26
- 102100024505 Bone morphogenetic protein 4 Human genes 0.000 claims abstract description 23
- 239000003102 growth factor Substances 0.000 claims abstract description 23
- 239000000758 substrate Substances 0.000 claims abstract description 19
- 239000004480 active ingredient Substances 0.000 claims abstract description 9
- 238000012258 culturing Methods 0.000 claims abstract description 8
- 239000002609 medium Substances 0.000 claims description 32
- 238000011282 treatment Methods 0.000 claims description 24
- 102000008186 Collagen Human genes 0.000 claims description 21
- 108010035532 Collagen Proteins 0.000 claims description 21
- 229920001436 collagen Polymers 0.000 claims description 21
- 239000008194 pharmaceutical composition Substances 0.000 claims description 19
- 239000006144 Dulbecco’s modified Eagle's medium Substances 0.000 claims description 18
- 230000002265 prevention Effects 0.000 claims description 16
- 210000000107 myocyte Anatomy 0.000 claims description 14
- 210000004786 perivascular cell Anatomy 0.000 claims description 13
- 210000000329 smooth muscle myocyte Anatomy 0.000 claims description 13
- 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 11
- WQZGKKKJIJFFOK-GASJEMHNSA-N Glucose Natural products OC[C@H]1OC(O)[C@H](O)[C@@H](O)[C@@H]1O WQZGKKKJIJFFOK-GASJEMHNSA-N 0.000 claims description 11
- 239000008103 glucose Substances 0.000 claims description 11
- 230000002792 vascular Effects 0.000 claims description 11
- 210000001789 adipocyte Anatomy 0.000 claims description 10
- 230000002093 peripheral effect Effects 0.000 claims description 9
- DHCLVCXQIBBOPH-UHFFFAOYSA-N Glycerol 2-phosphate Chemical compound OCC(CO)OP(O)(O)=O DHCLVCXQIBBOPH-UHFFFAOYSA-N 0.000 claims description 8
- UREBDLICKHMUKA-CXSFZGCWSA-N dexamethasone Chemical compound C1CC2=CC(=O)C=C[C@]2(C)[C@]2(F)[C@@H]1[C@@H]1C[C@@H](C)[C@@](C(=O)CO)(O)[C@@]1(C)C[C@@H]2O UREBDLICKHMUKA-CXSFZGCWSA-N 0.000 claims description 8
- 229960003957 dexamethasone Drugs 0.000 claims description 8
- CGIGDMFJXJATDK-UHFFFAOYSA-N indomethacin Chemical compound CC1=C(CC(O)=O)C2=CC(OC)=CC=C2N1C(=O)C1=CC=C(Cl)C=C1 CGIGDMFJXJATDK-UHFFFAOYSA-N 0.000 claims description 8
- NOESYZHRGYRDHS-UHFFFAOYSA-N insulin Chemical compound N1C(=O)C(NC(=O)C(CCC(N)=O)NC(=O)C(CCC(O)=O)NC(=O)C(C(C)C)NC(=O)C(NC(=O)CN)C(C)CC)CSSCC(C(NC(CO)C(=O)NC(CC(C)C)C(=O)NC(CC=2C=CC(O)=CC=2)C(=O)NC(CCC(N)=O)C(=O)NC(CC(C)C)C(=O)NC(CCC(O)=O)C(=O)NC(CC(N)=O)C(=O)NC(CC=2C=CC(O)=CC=2)C(=O)NC(CSSCC(NC(=O)C(C(C)C)NC(=O)C(CC(C)C)NC(=O)C(CC=2C=CC(O)=CC=2)NC(=O)C(CC(C)C)NC(=O)C(C)NC(=O)C(CCC(O)=O)NC(=O)C(C(C)C)NC(=O)C(CC(C)C)NC(=O)C(CC=2NC=NC=2)NC(=O)C(CO)NC(=O)CNC2=O)C(=O)NCC(=O)NC(CCC(O)=O)C(=O)NC(CCCNC(N)=N)C(=O)NCC(=O)NC(CC=3C=CC=CC=3)C(=O)NC(CC=3C=CC=CC=3)C(=O)NC(CC=3C=CC(O)=CC=3)C(=O)NC(C(C)O)C(=O)N3C(CCC3)C(=O)NC(CCCCN)C(=O)NC(C)C(O)=O)C(=O)NC(CC(N)=O)C(O)=O)=O)NC(=O)C(C(C)CC)NC(=O)C(CO)NC(=O)C(C(C)O)NC(=O)C1CSSCC2NC(=O)C(CC(C)C)NC(=O)C(NC(=O)C(CCC(N)=O)NC(=O)C(CC(N)=O)NC(=O)C(NC(=O)C(N)CC=1C=CC=CC=1)C(C)C)CC1=CN=CN1 NOESYZHRGYRDHS-UHFFFAOYSA-N 0.000 claims description 8
- UCSJYZPVAKXKNQ-HZYVHMACSA-N streptomycin Chemical compound CN[C@H]1[C@H](O)[C@@H](O)[C@H](CO)O[C@H]1O[C@@H]1[C@](C=O)(O)[C@H](C)O[C@H]1O[C@@H]1[C@@H](NC(N)=N)[C@H](O)[C@@H](NC(N)=N)[C@H](O)[C@H]1O UCSJYZPVAKXKNQ-HZYVHMACSA-N 0.000 claims description 8
- 102000018233 Fibroblast Growth Factor Human genes 0.000 claims description 7
- 108050007372 Fibroblast Growth Factor Proteins 0.000 claims description 7
- 210000002449 bone cell Anatomy 0.000 claims description 7
- 229940126864 fibroblast growth factor Drugs 0.000 claims description 7
- DGVVWUTYPXICAM-UHFFFAOYSA-N β‐Mercaptoethanol Chemical compound OCCS DGVVWUTYPXICAM-UHFFFAOYSA-N 0.000 claims description 7
- 108010073929 Vascular Endothelial Growth Factor A Proteins 0.000 claims description 6
- 102000005789 Vascular Endothelial Growth Factors Human genes 0.000 claims description 6
- 108010019530 Vascular Endothelial Growth Factors Proteins 0.000 claims description 6
- 230000002491 angiogenic effect Effects 0.000 claims description 6
- 238000004519 manufacturing process Methods 0.000 claims description 6
- 229930182555 Penicillin Natural products 0.000 claims description 5
- JGSARLDLIJGVTE-MBNYWOFBSA-N Penicillin G Chemical compound N([C@H]1[C@H]2SC([C@@H](N2C1=O)C(O)=O)(C)C)C(=O)CC1=CC=CC=C1 JGSARLDLIJGVTE-MBNYWOFBSA-N 0.000 claims description 5
- 210000004263 induced pluripotent stem cell Anatomy 0.000 claims description 5
- 229940049954 penicillin Drugs 0.000 claims description 5
- VBEQCZHXXJYVRD-GACYYNSASA-N uroanthelone Chemical compound C([C@@H](C(=O)N[C@H](C(=O)N[C@@H](CS)C(=O)N[C@@H](CC(N)=O)C(=O)N[C@@H](CS)C(=O)N[C@H](C(=O)N[C@@H]([C@@H](C)CC)C(=O)NCC(=O)N[C@@H](CC=1C=CC(O)=CC=1)C(=O)N[C@@H](CO)C(=O)NCC(=O)N[C@@H](CC(O)=O)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CS)C(=O)N[C@@H](CCC(N)=O)C(=O)N[C@@H]([C@@H](C)O)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CC(O)=O)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CC=1C2=CC=CC=C2NC=1)C(=O)N[C@@H](CC=1C2=CC=CC=C2NC=1)C(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CCCNC(N)=N)C(O)=O)C(C)C)[C@@H](C)O)NC(=O)[C@H](CO)NC(=O)[C@H](CC(O)=O)NC(=O)[C@H](CC(C)C)NC(=O)[C@H](CO)NC(=O)[C@H](CCC(O)=O)NC(=O)[C@@H](NC(=O)[C@H](CC=1NC=NC=1)NC(=O)[C@H](CCSC)NC(=O)[C@H](CS)NC(=O)[C@@H](NC(=O)CNC(=O)CNC(=O)[C@H](CC(N)=O)NC(=O)[C@H](CC(C)C)NC(=O)[C@H](CS)NC(=O)[C@H](CC=1C=CC(O)=CC=1)NC(=O)CNC(=O)[C@H](CC(O)=O)NC(=O)[C@H](CC=1C=CC(O)=CC=1)NC(=O)[C@H](CO)NC(=O)[C@H](CO)NC(=O)[C@H]1N(CCC1)C(=O)[C@H](CS)NC(=O)CNC(=O)[C@H]1N(CCC1)C(=O)[C@H](CC=1C=CC(O)=CC=1)NC(=O)[C@H](CO)NC(=O)[C@@H](N)CC(N)=O)C(C)C)[C@@H](C)CC)C1=CC=C(O)C=C1 VBEQCZHXXJYVRD-GACYYNSASA-N 0.000 claims description 5
- APIXJSLKIYYUKG-UHFFFAOYSA-N 3 Isobutyl 1 methylxanthine Chemical compound O=C1N(C)C(=O)N(CC(C)C)C2=C1N=CN2 APIXJSLKIYYUKG-UHFFFAOYSA-N 0.000 claims description 4
- 208000031229 Cardiomyopathies Diseases 0.000 claims description 4
- 206010012689 Diabetic retinopathy Diseases 0.000 claims description 4
- 102000004877 Insulin Human genes 0.000 claims description 4
- 108090001061 Insulin Proteins 0.000 claims description 4
- MIJPAVRNWPDMOR-ZAFYKAAXSA-N L-ascorbic acid 2-phosphate Chemical compound OC[C@H](O)[C@H]1OC(=O)C(OP(O)(O)=O)=C1O MIJPAVRNWPDMOR-ZAFYKAAXSA-N 0.000 claims description 4
- 206010038923 Retinopathy Diseases 0.000 claims description 4
- 239000003797 essential amino acid Substances 0.000 claims description 4
- 235000020776 essential amino acid Nutrition 0.000 claims description 4
- 229960000905 indomethacin Drugs 0.000 claims description 4
- 229940125396 insulin Drugs 0.000 claims description 4
- 238000000926 separation method Methods 0.000 claims description 4
- 210000002966 serum Anatomy 0.000 claims description 4
- 238000010374 somatic cell nuclear transfer Methods 0.000 claims description 4
- 229960005322 streptomycin Drugs 0.000 claims description 4
- 206010003210 Arteriosclerosis Diseases 0.000 claims description 3
- 206010020772 Hypertension Diseases 0.000 claims description 3
- 102100026547 Platelet-derived growth factor receptor beta Human genes 0.000 claims description 3
- 208000011775 arteriosclerosis disease Diseases 0.000 claims description 3
- 208000029078 coronary artery disease Diseases 0.000 claims description 3
- 206010002329 Aneurysm Diseases 0.000 claims description 2
- 208000002330 Congenital Heart Defects Diseases 0.000 claims description 2
- 208000010412 Glaucoma Diseases 0.000 claims description 2
- 206010019280 Heart failures Diseases 0.000 claims description 2
- 208000025584 Pericardial disease Diseases 0.000 claims description 2
- 208000018262 Peripheral vascular disease Diseases 0.000 claims description 2
- 101710164680 Platelet-derived growth factor receptor beta Proteins 0.000 claims description 2
- 206010038933 Retinopathy of prematurity Diseases 0.000 claims description 2
- 208000006011 Stroke Diseases 0.000 claims description 2
- 102000004142 Trypsin Human genes 0.000 claims description 2
- 108090000631 Trypsin Proteins 0.000 claims description 2
- 208000001910 Ventricular Heart Septal Defects Diseases 0.000 claims description 2
- 208000000208 Wet Macular Degeneration Diseases 0.000 claims description 2
- 206010064930 age-related macular degeneration Diseases 0.000 claims description 2
- 206010003119 arrhythmia Diseases 0.000 claims description 2
- 230000006793 arrhythmia Effects 0.000 claims description 2
- 208000028831 congenital heart disease Diseases 0.000 claims description 2
- 208000018578 heart valve disease Diseases 0.000 claims description 2
- 208000015210 hypertensive heart disease Diseases 0.000 claims description 2
- 201000003130 ventricular septal defect Diseases 0.000 claims description 2
- 108010049955 Bone Morphogenetic Protein 4 Proteins 0.000 claims 2
- 108090000723 Insulin-Like Growth Factor I Proteins 0.000 claims 2
- 102000013275 Somatomedins Human genes 0.000 claims 2
- 241000219357 Cactaceae Species 0.000 claims 1
- 239000002253 acid Substances 0.000 claims 1
- 230000001902 propagating effect Effects 0.000 claims 1
- 230000004069 differentiation Effects 0.000 abstract description 31
- 101000762379 Homo sapiens Bone morphogenetic protein 4 Proteins 0.000 abstract description 21
- 239000004698 Polyethylene Substances 0.000 description 17
- 210000004204 blood vessel Anatomy 0.000 description 15
- 238000000684 flow cytometry Methods 0.000 description 14
- 230000000302 ischemic effect Effects 0.000 description 14
- -1 CD31 Proteins 0.000 description 13
- 108091003079 Bovine Serum Albumin Proteins 0.000 description 12
- 239000012091 fetal bovine serum Substances 0.000 description 12
- 210000001525 retina Anatomy 0.000 description 12
- 210000001519 tissue Anatomy 0.000 description 12
- 238000002054 transplantation Methods 0.000 description 12
- 230000015572 biosynthetic process Effects 0.000 description 11
- 206010012601 diabetes mellitus Diseases 0.000 description 11
- 210000002901 mesenchymal stem cell Anatomy 0.000 description 11
- 210000004165 myocardium Anatomy 0.000 description 11
- 238000010586 diagram Methods 0.000 description 9
- 108010053099 Vascular Endothelial Growth Factor Receptor-2 Proteins 0.000 description 8
- 102100033177 Vascular endothelial growth factor receptor 2 Human genes 0.000 description 8
- 210000002469 basement membrane Anatomy 0.000 description 8
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 description 8
- 239000001963 growth medium Substances 0.000 description 8
- 241000282412 Homo Species 0.000 description 7
- 206010061216 Infarction Diseases 0.000 description 7
- 201000010099 disease Diseases 0.000 description 7
- 230000007574 infarction Effects 0.000 description 7
- 230000008569 process Effects 0.000 description 7
- 239000000243 solution Substances 0.000 description 7
- 238000010186 staining Methods 0.000 description 7
- ZSJLQEPLLKMAKR-GKHCUFPYSA-N streptozocin Chemical compound O=NN(C)C(=O)N[C@H]1[C@@H](O)O[C@H](CO)[C@@H](O)[C@@H]1O ZSJLQEPLLKMAKR-GKHCUFPYSA-N 0.000 description 7
- LFQSCWFLJHTTHZ-UHFFFAOYSA-N Ethanol Chemical compound CCO LFQSCWFLJHTTHZ-UHFFFAOYSA-N 0.000 description 6
- DNIAPMSPPWPWGF-UHFFFAOYSA-N Propylene glycol Chemical compound CC(O)CO DNIAPMSPPWPWGF-UHFFFAOYSA-N 0.000 description 6
- ZSJLQEPLLKMAKR-UHFFFAOYSA-N Streptozotocin Natural products O=NN(C)C(=O)NC1C(O)OC(CO)C(O)C1O ZSJLQEPLLKMAKR-UHFFFAOYSA-N 0.000 description 6
- 230000017531 blood circulation Effects 0.000 description 6
- 238000004113 cell culture Methods 0.000 description 6
- 239000011248 coating agent Substances 0.000 description 6
- 238000000576 coating method Methods 0.000 description 6
- 210000002216 heart Anatomy 0.000 description 6
- 239000007924 injection Substances 0.000 description 6
- 238000002347 injection Methods 0.000 description 6
- 239000008055 phosphate buffer solution Substances 0.000 description 6
- 102000005962 receptors Human genes 0.000 description 6
- 108020003175 receptors Proteins 0.000 description 6
- 210000002460 smooth muscle Anatomy 0.000 description 6
- 229960001052 streptozocin Drugs 0.000 description 6
- 210000003556 vascular endothelial cell Anatomy 0.000 description 6
- 102000004190 Enzymes Human genes 0.000 description 5
- 108090000790 Enzymes Proteins 0.000 description 5
- 101000916489 Homo sapiens Chondroitin sulfate proteoglycan 4 Proteins 0.000 description 5
- 101001107084 Homo sapiens E3 ubiquitin-protein ligase RNF5 Proteins 0.000 description 5
- 241000699670 Mus sp. Species 0.000 description 5
- 235000010323 ascorbic acid Nutrition 0.000 description 5
- 229960005070 ascorbic acid Drugs 0.000 description 5
- 239000011668 ascorbic acid Substances 0.000 description 5
- 230000000694 effects Effects 0.000 description 5
- 229940088598 enzyme Drugs 0.000 description 5
- 210000003414 extremity Anatomy 0.000 description 5
- 230000006870 function Effects 0.000 description 5
- 230000006698 induction Effects 0.000 description 5
- 239000003550 marker Substances 0.000 description 5
- 210000002826 placenta Anatomy 0.000 description 5
- 108090000623 proteins and genes Proteins 0.000 description 5
- 230000002207 retinal effect Effects 0.000 description 5
- 238000012546 transfer Methods 0.000 description 5
- CURLTUGMZLYLDI-UHFFFAOYSA-N Carbon dioxide Chemical compound O=C=O CURLTUGMZLYLDI-UHFFFAOYSA-N 0.000 description 4
- PEDCQBHIVMGVHV-UHFFFAOYSA-N Glycerine Chemical compound OCC(O)CO PEDCQBHIVMGVHV-UHFFFAOYSA-N 0.000 description 4
- 102100031573 Hematopoietic progenitor cell antigen CD34 Human genes 0.000 description 4
- 101000777663 Homo sapiens Hematopoietic progenitor cell antigen CD34 Proteins 0.000 description 4
- 101000800116 Homo sapiens Thy-1 membrane glycoprotein Proteins 0.000 description 4
- 241001465754 Metazoa Species 0.000 description 4
- 241000699666 Mus <mouse, genus> Species 0.000 description 4
- ISWSIDIOOBJBQZ-UHFFFAOYSA-N Phenol Chemical compound OC1=CC=CC=C1 ISWSIDIOOBJBQZ-UHFFFAOYSA-N 0.000 description 4
- 102100024616 Platelet endothelial cell adhesion molecule Human genes 0.000 description 4
- 102100033523 Thy-1 membrane glycoprotein Human genes 0.000 description 4
- 230000009471 action Effects 0.000 description 4
- 238000004220 aggregation Methods 0.000 description 4
- 239000003125 aqueous solvent Substances 0.000 description 4
- 210000004369 blood Anatomy 0.000 description 4
- 239000008280 blood Substances 0.000 description 4
- 210000001185 bone marrow Anatomy 0.000 description 4
- 230000001419 dependent effect Effects 0.000 description 4
- 239000003814 drug Substances 0.000 description 4
- 230000002526 effect on cardiovascular system Effects 0.000 description 4
- 238000005516 engineering process Methods 0.000 description 4
- 210000003976 gap junction Anatomy 0.000 description 4
- 238000007898 magnetic cell sorting Methods 0.000 description 4
- 210000000663 muscle cell Anatomy 0.000 description 4
- 208000010125 myocardial infarction Diseases 0.000 description 4
- 210000000963 osteoblast Anatomy 0.000 description 4
- WVDDGKGOMKODPV-ZQBYOMGUSA-N phenyl(114C)methanol Chemical compound O[14CH2]C1=CC=CC=C1 WVDDGKGOMKODPV-ZQBYOMGUSA-N 0.000 description 4
- 239000003755 preservative agent Substances 0.000 description 4
- 238000011084 recovery Methods 0.000 description 4
- 208000024891 symptom Diseases 0.000 description 4
- 210000004509 vascular smooth muscle cell Anatomy 0.000 description 4
- FWBHETKCLVMNFS-UHFFFAOYSA-N 4',6-Diamino-2-phenylindol Chemical compound C1=CC(C(=N)N)=CC=C1C1=CC2=CC=C(C(N)=N)C=C2N1 FWBHETKCLVMNFS-UHFFFAOYSA-N 0.000 description 3
- 102100022464 5'-nucleotidase Human genes 0.000 description 3
- 102100032912 CD44 antigen Human genes 0.000 description 3
- 102100023126 Cell surface glycoprotein MUC18 Human genes 0.000 description 3
- 102000010970 Connexin Human genes 0.000 description 3
- 108050001175 Connexin Proteins 0.000 description 3
- 102100037241 Endoglin Human genes 0.000 description 3
- 102000009024 Epidermal Growth Factor Human genes 0.000 description 3
- 101800003838 Epidermal growth factor Proteins 0.000 description 3
- 101000678236 Homo sapiens 5'-nucleotidase Proteins 0.000 description 3
- 101000868273 Homo sapiens CD44 antigen Proteins 0.000 description 3
- 101000623903 Homo sapiens Cell surface glycoprotein MUC18 Proteins 0.000 description 3
- 101000881679 Homo sapiens Endoglin Proteins 0.000 description 3
- 101000738771 Homo sapiens Receptor-type tyrosine-protein phosphatase C Proteins 0.000 description 3
- 208000031481 Pathologic Constriction Diseases 0.000 description 3
- 102000001393 Platelet-Derived Growth Factor alpha Receptor Human genes 0.000 description 3
- 108010068588 Platelet-Derived Growth Factor alpha Receptor Proteins 0.000 description 3
- 102100037422 Receptor-type tyrosine-protein phosphatase C Human genes 0.000 description 3
- 206010038848 Retinal detachment Diseases 0.000 description 3
- 241000316887 Saissetia oleae Species 0.000 description 3
- 235000010724 Wisteria floribunda Nutrition 0.000 description 3
- 230000002776 aggregation Effects 0.000 description 3
- 238000004458 analytical method Methods 0.000 description 3
- 238000010171 animal model Methods 0.000 description 3
- 210000001367 artery Anatomy 0.000 description 3
- 239000000872 buffer Substances 0.000 description 3
- 210000004413 cardiac myocyte Anatomy 0.000 description 3
- 230000024245 cell differentiation Effects 0.000 description 3
- 210000003169 central nervous system Anatomy 0.000 description 3
- 239000003795 chemical substances by application Substances 0.000 description 3
- 230000008602 contraction Effects 0.000 description 3
- 230000034994 death Effects 0.000 description 3
- 210000002257 embryonic structure Anatomy 0.000 description 3
- 230000003511 endothelial effect Effects 0.000 description 3
- 210000003989 endothelium vascular Anatomy 0.000 description 3
- 229940116977 epidermal growth factor Drugs 0.000 description 3
- 235000019441 ethanol Nutrition 0.000 description 3
- 210000002950 fibroblast Anatomy 0.000 description 3
- 238000007667 floating Methods 0.000 description 3
- 230000000774 hypoallergenic effect Effects 0.000 description 3
- 238000003365 immunocytochemistry Methods 0.000 description 3
- 238000007918 intramuscular administration Methods 0.000 description 3
- 238000001990 intravenous administration Methods 0.000 description 3
- 210000001161 mammalian embryo Anatomy 0.000 description 3
- 239000011159 matrix material Substances 0.000 description 3
- 238000007431 microscopic evaluation Methods 0.000 description 3
- 210000000056 organ Anatomy 0.000 description 3
- 210000003668 pericyte Anatomy 0.000 description 3
- 238000002360 preparation method Methods 0.000 description 3
- 102000004169 proteins and genes Human genes 0.000 description 3
- 230000004264 retinal detachment Effects 0.000 description 3
- 210000001210 retinal vessel Anatomy 0.000 description 3
- 239000003381 stabilizer Substances 0.000 description 3
- 230000036262 stenosis Effects 0.000 description 3
- 208000037804 stenosis Diseases 0.000 description 3
- 238000007920 subcutaneous administration Methods 0.000 description 3
- 229940124597 therapeutic agent Drugs 0.000 description 3
- 230000001225 therapeutic effect Effects 0.000 description 3
- 208000019553 vascular disease Diseases 0.000 description 3
- QTWJRLJHJPIABL-UHFFFAOYSA-N 2-methylphenol;3-methylphenol;4-methylphenol Chemical compound CC1=CC=C(O)C=C1.CC1=CC=CC(O)=C1.CC1=CC=CC=C1O QTWJRLJHJPIABL-UHFFFAOYSA-N 0.000 description 2
- 102100031650 C-X-C chemokine receptor type 4 Human genes 0.000 description 2
- 102100029761 Cadherin-5 Human genes 0.000 description 2
- VTYYLEPIZMXCLO-UHFFFAOYSA-L Calcium carbonate Chemical compound [Ca+2].[O-]C([O-])=O VTYYLEPIZMXCLO-UHFFFAOYSA-L 0.000 description 2
- 101100481408 Danio rerio tie2 gene Proteins 0.000 description 2
- 229920002307 Dextran Polymers 0.000 description 2
- 102100021811 E3 ubiquitin-protein ligase RNF5 Human genes 0.000 description 2
- KCXVZYZYPLLWCC-UHFFFAOYSA-N EDTA Chemical compound OC(=O)CN(CC(O)=O)CCN(CC(O)=O)CC(O)=O KCXVZYZYPLLWCC-UHFFFAOYSA-N 0.000 description 2
- 206010016654 Fibrosis Diseases 0.000 description 2
- 239000012981 Hank's balanced salt solution Substances 0.000 description 2
- 101000922348 Homo sapiens C-X-C chemokine receptor type 4 Proteins 0.000 description 2
- 101000794587 Homo sapiens Cadherin-5 Proteins 0.000 description 2
- 101000610551 Homo sapiens Prominin-1 Proteins 0.000 description 2
- 208000031226 Hyperlipidaemia Diseases 0.000 description 2
- ZDXPYRJPNDTMRX-VKHMYHEASA-N L-glutamine Chemical compound OC(=O)[C@@H](N)CCC(N)=O ZDXPYRJPNDTMRX-VKHMYHEASA-N 0.000 description 2
- 229930182816 L-glutamine Natural products 0.000 description 2
- 241000124008 Mammalia Species 0.000 description 2
- 101100481410 Mus musculus Tek gene Proteins 0.000 description 2
- NWIBSHFKIJFRCO-WUDYKRTCSA-N Mytomycin Chemical compound C1N2C(C(C(C)=C(N)C3=O)=O)=C3[C@@H](COC(N)=O)[C@@]2(OC)[C@@H]2[C@H]1N2 NWIBSHFKIJFRCO-WUDYKRTCSA-N 0.000 description 2
- 239000004677 Nylon Substances 0.000 description 2
- 102100040120 Prominin-1 Human genes 0.000 description 2
- 241000700159 Rattus Species 0.000 description 2
- 102000007374 Smad Proteins Human genes 0.000 description 2
- 108010007945 Smad Proteins Proteins 0.000 description 2
- DWAQJAXMDSEUJJ-UHFFFAOYSA-M Sodium bisulfite Chemical compound [Na+].OS([O-])=O DWAQJAXMDSEUJJ-UHFFFAOYSA-M 0.000 description 2
- 102000004887 Transforming Growth Factor beta Human genes 0.000 description 2
- 108090001012 Transforming Growth Factor beta Proteins 0.000 description 2
- 239000013543 active substance Substances 0.000 description 2
- 210000000577 adipose tissue Anatomy 0.000 description 2
- RGCKGOZRHPZPFP-UHFFFAOYSA-N alizarin Chemical compound C1=CC=C2C(=O)C3=C(O)C(O)=CC=C3C(=O)C2=C1 RGCKGOZRHPZPFP-UHFFFAOYSA-N 0.000 description 2
- 230000008901 benefit Effects 0.000 description 2
- 229960000686 benzalkonium chloride Drugs 0.000 description 2
- CADWTSSKOVRVJC-UHFFFAOYSA-N benzyl(dimethyl)azanium;chloride Chemical compound [Cl-].C[NH+](C)CC1=CC=CC=C1 CADWTSSKOVRVJC-UHFFFAOYSA-N 0.000 description 2
- 230000033228 biological regulation Effects 0.000 description 2
- 230000008499 blood brain barrier function Effects 0.000 description 2
- 230000008081 blood perfusion Effects 0.000 description 2
- 210000001218 blood-brain barrier Anatomy 0.000 description 2
- 101150067309 bmp4 gene Proteins 0.000 description 2
- 235000019282 butylated hydroxyanisole Nutrition 0.000 description 2
- 102000006783 calponin Human genes 0.000 description 2
- 108010086826 calponin Proteins 0.000 description 2
- 239000001569 carbon dioxide Substances 0.000 description 2
- 229910002092 carbon dioxide Inorganic materials 0.000 description 2
- 230000008614 cellular interaction Effects 0.000 description 2
- 210000001175 cerebrospinal fluid Anatomy 0.000 description 2
- HVYWMOMLDIMFJA-DPAQBDIFSA-N cholesterol Chemical compound C1C=C2C[C@@H](O)CC[C@]2(C)[C@@H]2[C@@H]1[C@@H]1CC[C@H]([C@H](C)CCCC(C)C)[C@@]1(C)CC2 HVYWMOMLDIMFJA-DPAQBDIFSA-N 0.000 description 2
- 210000001612 chondrocyte Anatomy 0.000 description 2
- 210000001608 connective tissue cell Anatomy 0.000 description 2
- 229930003836 cresol Natural products 0.000 description 2
- 210000000805 cytoplasm Anatomy 0.000 description 2
- 230000001086 cytosolic effect Effects 0.000 description 2
- GVJHHUAWPYXKBD-UHFFFAOYSA-N d-alpha-tocopherol Natural products OC1=C(C)C(C)=C2OC(CCCC(C)CCCC(C)CCCC(C)C)(C)CCC2=C1C GVJHHUAWPYXKBD-UHFFFAOYSA-N 0.000 description 2
- 230000006378 damage Effects 0.000 description 2
- 230000001934 delay Effects 0.000 description 2
- 238000011161 development Methods 0.000 description 2
- 230000018109 developmental process Effects 0.000 description 2
- 235000014113 dietary fatty acids Nutrition 0.000 description 2
- 239000003085 diluting agent Substances 0.000 description 2
- 238000010790 dilution Methods 0.000 description 2
- 239000012895 dilution Substances 0.000 description 2
- 239000003937 drug carrier Substances 0.000 description 2
- 239000003995 emulsifying agent Substances 0.000 description 2
- 238000000605 extraction Methods 0.000 description 2
- 229930195729 fatty acid Natural products 0.000 description 2
- 239000000194 fatty acid Substances 0.000 description 2
- 230000004761 fibrosis Effects 0.000 description 2
- MHMNJMPURVTYEJ-UHFFFAOYSA-N fluorescein-5-isothiocyanate Chemical compound O1C(=O)C2=CC(N=C=S)=CC=C2C21C1=CC=C(O)C=C1OC1=CC(O)=CC=C21 MHMNJMPURVTYEJ-UHFFFAOYSA-N 0.000 description 2
- 239000000499 gel Substances 0.000 description 2
- 235000011187 glycerol Nutrition 0.000 description 2
- 230000012010 growth Effects 0.000 description 2
- 230000006872 improvement Effects 0.000 description 2
- 238000000338 in vitro Methods 0.000 description 2
- 238000002372 labelling Methods 0.000 description 2
- 239000002523 lectin Substances 0.000 description 2
- 210000003141 lower extremity Anatomy 0.000 description 2
- 238000012423 maintenance Methods 0.000 description 2
- 210000003716 mesoderm Anatomy 0.000 description 2
- 238000010172 mouse model Methods 0.000 description 2
- 210000002894 multi-fate stem cell Anatomy 0.000 description 2
- 210000003205 muscle Anatomy 0.000 description 2
- 210000003098 myoblast Anatomy 0.000 description 2
- 210000004940 nucleus Anatomy 0.000 description 2
- 229920001778 nylon Polymers 0.000 description 2
- 239000003921 oil Substances 0.000 description 2
- 235000019198 oils Nutrition 0.000 description 2
- 210000004409 osteocyte Anatomy 0.000 description 2
- 238000010979 pH adjustment Methods 0.000 description 2
- 239000002504 physiological saline solution Substances 0.000 description 2
- 239000000049 pigment Substances 0.000 description 2
- 239000004033 plastic Substances 0.000 description 2
- 229920003023 plastic Polymers 0.000 description 2
- 210000001778 pluripotent stem cell Anatomy 0.000 description 2
- 230000035755 proliferation Effects 0.000 description 2
- 238000003753 real-time PCR Methods 0.000 description 2
- 230000008929 regeneration Effects 0.000 description 2
- 238000011069 regeneration method Methods 0.000 description 2
- 230000001105 regulatory effect Effects 0.000 description 2
- 238000007634 remodeling Methods 0.000 description 2
- 231100000241 scar Toxicity 0.000 description 2
- 235000010267 sodium hydrogen sulphite Nutrition 0.000 description 2
- ZRKFYGHZFMAOKI-QMGMOQQFSA-N tgfbeta Chemical compound C([C@H](NC(=O)[C@H](C(C)C)NC(=O)CNC(=O)[C@H](CCC(O)=O)NC(=O)[C@H](CCCNC(N)=N)NC(=O)[C@H](CC(N)=O)NC(=O)[C@H](CC(C)C)NC(=O)[C@H]([C@@H](C)O)NC(=O)[C@H](CCC(O)=O)NC(=O)[C@H]([C@@H](C)O)NC(=O)[C@H](CC(C)C)NC(=O)CNC(=O)[C@H](C)NC(=O)[C@H](CO)NC(=O)[C@H](CCC(N)=O)NC(=O)[C@@H](NC(=O)[C@H](C)NC(=O)[C@H](C)NC(=O)[C@@H](NC(=O)[C@H](CC(C)C)NC(=O)[C@@H](N)CCSC)C(C)C)[C@@H](C)CC)C(=O)N[C@@H]([C@@H](C)O)C(=O)N[C@@H](C(C)C)C(=O)N[C@@H](CC=1C=CC=CC=1)C(=O)N[C@@H](C)C(=O)N1[C@@H](CCC1)C(=O)N[C@@H]([C@@H](C)O)C(=O)N[C@@H](CC(N)=O)C(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](C)C(=O)N[C@@H](CC=1C=CC=CC=1)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](C)C(=O)N[C@@H](CC(C)C)C(=O)N1[C@@H](CCC1)C(=O)N1[C@@H](CCC1)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CO)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CC(C)C)C(O)=O)C1=CC=C(O)C=C1 ZRKFYGHZFMAOKI-QMGMOQQFSA-N 0.000 description 2
- 230000030968 tissue homeostasis Effects 0.000 description 2
- 235000010384 tocopherol Nutrition 0.000 description 2
- 229960001295 tocopherol Drugs 0.000 description 2
- 229930003799 tocopherol Natural products 0.000 description 2
- 239000011732 tocopherol Substances 0.000 description 2
- 210000004291 uterus Anatomy 0.000 description 2
- 230000003966 vascular damage Effects 0.000 description 2
- 230000037197 vascular physiology Effects 0.000 description 2
- 235000015112 vegetable and seed oil Nutrition 0.000 description 2
- 239000008158 vegetable oil Substances 0.000 description 2
- 210000003462 vein Anatomy 0.000 description 2
- 210000000636 white adipocyte Anatomy 0.000 description 2
- GVJHHUAWPYXKBD-IEOSBIPESA-N α-tocopherol Chemical compound OC1=C(C)C(C)=C2O[C@@](CCC[C@H](C)CCC[C@H](C)CCCC(C)C)(C)CCC2=C1C GVJHHUAWPYXKBD-IEOSBIPESA-N 0.000 description 2
- WRIDQFICGBMAFQ-UHFFFAOYSA-N (E)-8-Octadecenoic acid Natural products CCCCCCCCCC=CCCCCCCC(O)=O WRIDQFICGBMAFQ-UHFFFAOYSA-N 0.000 description 1
- DXOFMKNITCKTIS-QXMHVHEDSA-N (z)-2-ethyloctadec-9-enoic acid Chemical compound CCCCCCCC\C=C/CCCCCCC(CC)C(O)=O DXOFMKNITCKTIS-QXMHVHEDSA-N 0.000 description 1
- WPRAXAOJIODQJR-UHFFFAOYSA-N 1-(3,4-dimethylphenyl)ethanone Chemical compound CC(=O)C1=CC=C(C)C(C)=C1 WPRAXAOJIODQJR-UHFFFAOYSA-N 0.000 description 1
- DQECFVGMGBQCPA-GLCLSGQWSA-N 2,2-dimethylpropanoyloxymethyl (2s,5r,6r)-6-[[(2r)-2-amino-2-phenylacetyl]amino]-3,3-dimethyl-7-oxo-4-thia-1-azabicyclo[3.2.0]heptane-2-carboxylate;hydron;chloride Chemical compound Cl.C1([C@@H](N)C(=O)N[C@H]2[C@@H]3N(C2=O)[C@H](C(S3)(C)C)C(=O)OCOC(=O)C(C)(C)C)=CC=CC=C1 DQECFVGMGBQCPA-GLCLSGQWSA-N 0.000 description 1
- LQJBNNIYVWPHFW-UHFFFAOYSA-N 20:1omega9c fatty acid Natural products CCCCCCCCCCC=CCCCCCCCC(O)=O LQJBNNIYVWPHFW-UHFFFAOYSA-N 0.000 description 1
- RYKKQQUKJJGFMN-HVDRVSQOSA-N 4,5-bis(hydroxymethyl)-2-methylpyridin-3-ol;(2s)-5-oxopyrrolidine-2-carboxylic acid Chemical compound OC(=O)[C@@H]1CCC(=O)N1.CC1=NC=C(CO)C(CO)=C1O RYKKQQUKJJGFMN-HVDRVSQOSA-N 0.000 description 1
- QSBYPNXLFMSGKH-UHFFFAOYSA-N 9-Heptadecensaeure Natural products CCCCCCCC=CCCCCCCCC(O)=O QSBYPNXLFMSGKH-UHFFFAOYSA-N 0.000 description 1
- 102000014777 Adipokines Human genes 0.000 description 1
- 108010078606 Adipokines Proteins 0.000 description 1
- 102000011690 Adiponectin Human genes 0.000 description 1
- 108010076365 Adiponectin Proteins 0.000 description 1
- 206010002383 Angina Pectoris Diseases 0.000 description 1
- 241000283690 Bos taurus Species 0.000 description 1
- 101100381481 Caenorhabditis elegans baz-2 gene Proteins 0.000 description 1
- 241000282472 Canis lupus familiaris Species 0.000 description 1
- 102000029816 Collagenase Human genes 0.000 description 1
- 108060005980 Collagenase Proteins 0.000 description 1
- 102100036912 Desmin Human genes 0.000 description 1
- 108010044052 Desmin Proteins 0.000 description 1
- 241000283086 Equidae Species 0.000 description 1
- 102000010834 Extracellular Matrix Proteins Human genes 0.000 description 1
- 108010037362 Extracellular Matrix Proteins Proteins 0.000 description 1
- 241000282326 Felis catus Species 0.000 description 1
- 101150048336 Flt1 gene Proteins 0.000 description 1
- 241000287828 Gallus gallus Species 0.000 description 1
- 108010010803 Gelatin Proteins 0.000 description 1
- 101000935043 Homo sapiens Integrin beta-1 Proteins 0.000 description 1
- 101000692455 Homo sapiens Platelet-derived growth factor receptor beta Proteins 0.000 description 1
- 101001059454 Homo sapiens Serine/threonine-protein kinase MARK2 Proteins 0.000 description 1
- 208000001953 Hypotension Diseases 0.000 description 1
- 206010061218 Inflammation Diseases 0.000 description 1
- 102100025304 Integrin beta-1 Human genes 0.000 description 1
- 102000016267 Leptin Human genes 0.000 description 1
- 108010092277 Leptin Proteins 0.000 description 1
- 102000018697 Membrane Proteins Human genes 0.000 description 1
- 108010052285 Membrane Proteins Proteins 0.000 description 1
- 102000029749 Microtubule Human genes 0.000 description 1
- 108091022875 Microtubule Proteins 0.000 description 1
- 101710167839 Morphogenetic protein Proteins 0.000 description 1
- 239000005642 Oleic acid Substances 0.000 description 1
- ZQPPMHVWECSIRJ-UHFFFAOYSA-N Oleic acid Natural products CCCCCCCCC=CCCCCCCCC(O)=O ZQPPMHVWECSIRJ-UHFFFAOYSA-N 0.000 description 1
- 101150093908 PDGFRB gene Proteins 0.000 description 1
- 241001494479 Pecora Species 0.000 description 1
- 239000002202 Polyethylene glycol Substances 0.000 description 1
- 241000288906 Primates Species 0.000 description 1
- 102000016971 Proto-Oncogene Proteins c-kit Human genes 0.000 description 1
- 108010014608 Proto-Oncogene Proteins c-kit Proteins 0.000 description 1
- 241000669298 Pseudaulacaspis pentagona Species 0.000 description 1
- 238000010240 RT-PCR analysis Methods 0.000 description 1
- 101100372762 Rattus norvegicus Flt1 gene Proteins 0.000 description 1
- 108090000873 Receptor Protein-Tyrosine Kinases Proteins 0.000 description 1
- 102000004278 Receptor Protein-Tyrosine Kinases Human genes 0.000 description 1
- 102000007156 Resistin Human genes 0.000 description 1
- 108010047909 Resistin Proteins 0.000 description 1
- 201000007737 Retinal degeneration Diseases 0.000 description 1
- 102100028904 Serine/threonine-protein kinase MARK2 Human genes 0.000 description 1
- FAPWRFPIFSIZLT-UHFFFAOYSA-M Sodium chloride Chemical compound [Na+].[Cl-] FAPWRFPIFSIZLT-UHFFFAOYSA-M 0.000 description 1
- 241000282887 Suidae Species 0.000 description 1
- 102000040945 Transcription factor Human genes 0.000 description 1
- 108091023040 Transcription factor Proteins 0.000 description 1
- 241000251539 Vertebrata <Metazoa> Species 0.000 description 1
- 230000002159 abnormal effect Effects 0.000 description 1
- 230000005856 abnormality Effects 0.000 description 1
- 230000001464 adherent effect Effects 0.000 description 1
- 239000000478 adipokine Substances 0.000 description 1
- 210000004504 adult stem cell Anatomy 0.000 description 1
- 210000002945 adventitial reticular cell Anatomy 0.000 description 1
- 150000001298 alcohols Chemical class 0.000 description 1
- 230000004075 alteration Effects 0.000 description 1
- 150000001413 amino acids Chemical class 0.000 description 1
- 229940035676 analgesics Drugs 0.000 description 1
- 230000033115 angiogenesis Effects 0.000 description 1
- 239000000730 antalgic agent Substances 0.000 description 1
- 210000000709 aorta Anatomy 0.000 description 1
- 239000007864 aqueous solution Substances 0.000 description 1
- 230000004888 barrier function Effects 0.000 description 1
- 239000002585 base Substances 0.000 description 1
- 230000004071 biological effect Effects 0.000 description 1
- 210000002459 blastocyst Anatomy 0.000 description 1
- 210000000601 blood cell Anatomy 0.000 description 1
- 239000012503 blood component Substances 0.000 description 1
- 210000001124 body fluid Anatomy 0.000 description 1
- 239000010839 body fluid Substances 0.000 description 1
- 210000000988 bone and bone Anatomy 0.000 description 1
- 210000001593 brown adipocyte Anatomy 0.000 description 1
- 239000007853 buffer solution Substances 0.000 description 1
- 229910000019 calcium carbonate Inorganic materials 0.000 description 1
- 239000001506 calcium phosphate Substances 0.000 description 1
- 229910000389 calcium phosphate Inorganic materials 0.000 description 1
- 235000011010 calcium phosphates Nutrition 0.000 description 1
- 210000000845 cartilage Anatomy 0.000 description 1
- 210000000170 cell membrane Anatomy 0.000 description 1
- 210000003855 cell nucleus Anatomy 0.000 description 1
- 239000003153 chemical reaction reagent Substances 0.000 description 1
- 238000001553 co-assembly Methods 0.000 description 1
- 238000003501 co-culture Methods 0.000 description 1
- 239000000512 collagen gel Substances 0.000 description 1
- 229960002424 collagenase Drugs 0.000 description 1
- 230000000052 comparative effect Effects 0.000 description 1
- 239000000306 component Substances 0.000 description 1
- 210000000555 contractile cell Anatomy 0.000 description 1
- 230000000593 degrading effect Effects 0.000 description 1
- 210000005045 desmin Anatomy 0.000 description 1
- 238000003745 diagnosis Methods 0.000 description 1
- 108010007093 dispase Proteins 0.000 description 1
- 239000012153 distilled water Substances 0.000 description 1
- 238000009826 distribution Methods 0.000 description 1
- 229940079593 drug Drugs 0.000 description 1
- 230000004064 dysfunction Effects 0.000 description 1
- 210000003981 ectoderm Anatomy 0.000 description 1
- 210000001900 endoderm Anatomy 0.000 description 1
- 210000002889 endothelial cell Anatomy 0.000 description 1
- 230000002255 enzymatic effect Effects 0.000 description 1
- 210000002919 epithelial cell Anatomy 0.000 description 1
- DNJIEGIFACGWOD-UHFFFAOYSA-N ethyl mercaptane Natural products CCS DNJIEGIFACGWOD-UHFFFAOYSA-N 0.000 description 1
- 230000003203 everyday effect Effects 0.000 description 1
- 238000002474 experimental method Methods 0.000 description 1
- 210000002744 extracellular matrix Anatomy 0.000 description 1
- 230000001605 fetal effect Effects 0.000 description 1
- 210000003754 fetus Anatomy 0.000 description 1
- 238000001943 fluorescence-activated cell sorting Methods 0.000 description 1
- 238000009472 formulation Methods 0.000 description 1
- 239000008273 gelatin Substances 0.000 description 1
- 229920000159 gelatin Polymers 0.000 description 1
- 235000019322 gelatine Nutrition 0.000 description 1
- 235000011852 gelatine desserts Nutrition 0.000 description 1
- 230000001434 glomerular Effects 0.000 description 1
- 208000019622 heart disease Diseases 0.000 description 1
- 210000003958 hematopoietic stem cell Anatomy 0.000 description 1
- 229910052588 hydroxylapatite Inorganic materials 0.000 description 1
- 230000036543 hypotension Effects 0.000 description 1
- 238000010191 image analysis Methods 0.000 description 1
- 210000002865 immune cell Anatomy 0.000 description 1
- 230000000984 immunochemical effect Effects 0.000 description 1
- 238000003125 immunofluorescent labeling Methods 0.000 description 1
- 230000001771 impaired effect Effects 0.000 description 1
- 238000002513 implantation Methods 0.000 description 1
- 238000011534 incubation Methods 0.000 description 1
- 230000004054 inflammatory process Effects 0.000 description 1
- 230000028709 inflammatory response Effects 0.000 description 1
- 230000002608 insulinlike Effects 0.000 description 1
- 230000003993 interaction Effects 0.000 description 1
- 210000000936 intestine Anatomy 0.000 description 1
- 238000001361 intraarterial administration Methods 0.000 description 1
- 230000004068 intracellular signaling Effects 0.000 description 1
- 238000007912 intraperitoneal administration Methods 0.000 description 1
- 208000023589 ischemic disease Diseases 0.000 description 1
- 238000002955 isolation Methods 0.000 description 1
- QXJSBBXBKPUZAA-UHFFFAOYSA-N isooleic acid Natural products CCCCCCCC=CCCCCCCCCC(O)=O QXJSBBXBKPUZAA-UHFFFAOYSA-N 0.000 description 1
- 239000007951 isotonicity adjuster Substances 0.000 description 1
- 210000003734 kidney Anatomy 0.000 description 1
- NRYBAZVQPHGZNS-ZSOCWYAHSA-N leptin Chemical compound O=C([C@H](CO)NC(=O)[C@H](CC(C)C)NC(=O)[C@H](CC(O)=O)NC(=O)[C@H](CC(C)C)NC(=O)[C@H](CCC(N)=O)NC(=O)[C@H](CC=1C2=CC=CC=C2NC=1)NC(=O)[C@H](CC(C)C)NC(=O)[C@@H](NC(=O)[C@H](CC(O)=O)NC(=O)[C@H](CCC(N)=O)NC(=O)[C@H](CC(C)C)NC(=O)[C@H](CO)NC(=O)CNC(=O)[C@H](CCC(N)=O)NC(=O)[C@@H](N)CC(C)C)CCSC)N1CCC[C@H]1C(=O)NCC(=O)N[C@@H](CS)C(O)=O NRYBAZVQPHGZNS-ZSOCWYAHSA-N 0.000 description 1
- 229940039781 leptin Drugs 0.000 description 1
- 230000003902 lesion Effects 0.000 description 1
- 230000004132 lipogenesis Effects 0.000 description 1
- 230000003520 lipogenic effect Effects 0.000 description 1
- 238000011068 loading method Methods 0.000 description 1
- 230000007774 longterm Effects 0.000 description 1
- 239000000314 lubricant Substances 0.000 description 1
- 238000004020 luminiscence type Methods 0.000 description 1
- 210000002540 macrophage Anatomy 0.000 description 1
- 230000035800 maturation Effects 0.000 description 1
- 229910001987 mercury nitrate Inorganic materials 0.000 description 1
- 210000001704 mesoblast Anatomy 0.000 description 1
- 230000000813 microbial effect Effects 0.000 description 1
- 244000005700 microbiome Species 0.000 description 1
- 238000001000 micrograph Methods 0.000 description 1
- 210000004688 microtubule Anatomy 0.000 description 1
- 230000005012 migration Effects 0.000 description 1
- 238000013508 migration Methods 0.000 description 1
- 239000007758 minimum essential medium Substances 0.000 description 1
- 229960004857 mitomycin Drugs 0.000 description 1
- 230000011278 mitosis Effects 0.000 description 1
- 230000004001 molecular interaction Effects 0.000 description 1
- 230000000877 morphologic effect Effects 0.000 description 1
- 230000004660 morphological change Effects 0.000 description 1
- 230000037257 muscle growth Effects 0.000 description 1
- 210000002464 muscle smooth vascular Anatomy 0.000 description 1
- 210000003365 myofibril Anatomy 0.000 description 1
- 230000004070 myogenic differentiation Effects 0.000 description 1
- 210000001087 myotubule Anatomy 0.000 description 1
- 210000001178 neural stem cell Anatomy 0.000 description 1
- 210000004498 neuroglial cell Anatomy 0.000 description 1
- DRXYRSRECMWYAV-UHFFFAOYSA-N nitrooxymercury Chemical compound [Hg+].[O-][N+]([O-])=O DRXYRSRECMWYAV-UHFFFAOYSA-N 0.000 description 1
- 235000015097 nutrients Nutrition 0.000 description 1
- ZQPPMHVWECSIRJ-KTKRTIGZSA-N oleic acid Chemical compound CCCCCCCC\C=C/CCCCCCCC(O)=O ZQPPMHVWECSIRJ-KTKRTIGZSA-N 0.000 description 1
- 235000008390 olive oil Nutrition 0.000 description 1
- 239000004006 olive oil Substances 0.000 description 1
- 230000011164 ossification Effects 0.000 description 1
- 230000004072 osteoblast differentiation Effects 0.000 description 1
- 230000037361 pathway Effects 0.000 description 1
- XYJRXVWERLGGKC-UHFFFAOYSA-D pentacalcium;hydroxide;triphosphate Chemical compound [OH-].[Ca+2].[Ca+2].[Ca+2].[Ca+2].[Ca+2].[O-]P([O-])([O-])=O.[O-]P([O-])([O-])=O.[O-]P([O-])([O-])=O XYJRXVWERLGGKC-UHFFFAOYSA-D 0.000 description 1
- 239000000546 pharmaceutical excipient Substances 0.000 description 1
- 230000003169 placental effect Effects 0.000 description 1
- 229920001223 polyethylene glycol Polymers 0.000 description 1
- 239000002243 precursor Substances 0.000 description 1
- 230000002335 preservative effect Effects 0.000 description 1
- 230000002062 proliferating effect Effects 0.000 description 1
- 230000000644 propagated effect Effects 0.000 description 1
- 238000011321 prophylaxis Methods 0.000 description 1
- 235000013772 propylene glycol Nutrition 0.000 description 1
- 230000002685 pulmonary effect Effects 0.000 description 1
- 230000002285 radioactive effect Effects 0.000 description 1
- 230000004258 retinal degeneration Effects 0.000 description 1
- 238000003757 reverse transcription PCR Methods 0.000 description 1
- 210000002235 sarcomere Anatomy 0.000 description 1
- 238000010187 selection method Methods 0.000 description 1
- 102000034285 signal transducing proteins Human genes 0.000 description 1
- 108091006024 signal transducing proteins Proteins 0.000 description 1
- 230000011664 signaling Effects 0.000 description 1
- 210000002027 skeletal muscle Anatomy 0.000 description 1
- 210000002356 skeleton Anatomy 0.000 description 1
- 210000003491 skin Anatomy 0.000 description 1
- 230000000391 smoking effect Effects 0.000 description 1
- HRZFUMHJMZEROT-UHFFFAOYSA-L sodium disulfite Chemical compound [Na+].[Na+].[O-]S(=O)S([O-])(=O)=O HRZFUMHJMZEROT-UHFFFAOYSA-L 0.000 description 1
- 229940079827 sodium hydrogen sulfite Drugs 0.000 description 1
- 235000010262 sodium metabisulphite Nutrition 0.000 description 1
- 239000002904 solvent Substances 0.000 description 1
- 230000002269 spontaneous effect Effects 0.000 description 1
- 230000006641 stabilisation Effects 0.000 description 1
- 238000011105 stabilization Methods 0.000 description 1
- 210000002784 stomach Anatomy 0.000 description 1
- 239000000126 substance Substances 0.000 description 1
- 239000000725 suspension Substances 0.000 description 1
- 230000009897 systematic effect Effects 0.000 description 1
- 230000008685 targeting Effects 0.000 description 1
- 230000009772 tissue formation Effects 0.000 description 1
- 238000011200 topical administration Methods 0.000 description 1
- 210000003014 totipotent stem cell Anatomy 0.000 description 1
- 231100000331 toxic Toxicity 0.000 description 1
- 230000002588 toxic effect Effects 0.000 description 1
- 238000013518 transcription Methods 0.000 description 1
- 230000035897 transcription Effects 0.000 description 1
- QORWJWZARLRLPR-UHFFFAOYSA-H tricalcium bis(phosphate) Chemical compound [Ca+2].[Ca+2].[Ca+2].[O-]P([O-])([O-])=O.[O-]P([O-])([O-])=O QORWJWZARLRLPR-UHFFFAOYSA-H 0.000 description 1
- 239000012588 trypsin Substances 0.000 description 1
- 230000006444 vascular growth Effects 0.000 description 1
- 239000003981 vehicle Substances 0.000 description 1
- 210000001835 viscera Anatomy 0.000 description 1
- 238000012800 visualization Methods 0.000 description 1
- 239000002699 waste material Substances 0.000 description 1
- XLYOFNOQVPJJNP-UHFFFAOYSA-N water Chemical compound O XLYOFNOQVPJJNP-UHFFFAOYSA-N 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
- C12N5/069—Vascular Endothelial cells
- C12N5/0692—Stem cells; Progenitor cells; Precursor cells
-
- 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
- C12N5/0068—General culture methods using substrates
-
- 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
- C12N5/0652—Cells of skeletal and connective tissues; Mesenchyme
- C12N5/0662—Stem cells
-
- 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
- C12N9/00—Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
- C12N9/14—Hydrolases (3)
- C12N9/48—Hydrolases (3) acting on peptide bonds (3.4)
- C12N9/50—Proteinases, e.g. Endopeptidases (3.4.21-3.4.25)
- C12N9/64—Proteinases, e.g. Endopeptidases (3.4.21-3.4.25) derived from animal tissue
- C12N9/6421—Proteinases, e.g. Endopeptidases (3.4.21-3.4.25) derived from animal tissue from mammals
- C12N9/6424—Serine endopeptidases (3.4.21)
- C12N9/6427—Chymotrypsins (3.4.21.1; 3.4.21.2); Trypsin (3.4.21.4)
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K35/00—Medicinal preparations containing materials or reaction products thereof with undetermined constitution
-
- 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
- C12N2501/155—Bone morphogenic proteins [BMP]; Osteogenins; Osteogenic factor; Bone inducing factor
-
- 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
- C12N2501/165—Vascular endothelial growth factor [VEGF]
-
- 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
- C12N2506/02—Differentiation of animal cells from one lineage to another; Differentiation of pluripotent cells from embryonic cells
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2533/00—Supports or coatings for cell culture, characterised by material
- C12N2533/50—Proteins
- C12N2533/52—Fibronectin; Laminin
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2533/00—Supports or coatings for cell culture, characterised by material
- C12N2533/50—Proteins
- C12N2533/54—Collagen; Gelatin
Definitions
- the present invention is a method for producing perivascular progenitor cells from stem cells derived embryonic body, perivascular progenitor cells prepared by the above method, cardiovascular disease comprising the same as an active ingredient Or it relates to a cell therapy composition for the prevention or treatment of retinal disease, and a method for preventing or treating cardiovascular disease or retinal disease using the composition. More specifically, the stem cell-derived embryonic bodies are treated with BMP4 growth factors to induce differentiation into mesodermal cells, and after separating the embryoid bodies containing the differentiated mesodermal cells into single cells, the substrate is coated.
- perivascular progenitor cells from stem cells that naturally grow only cells attached and cultured in a culture dish, perivascular progenitor cells prepared by the above method, prevention or treatment of cardiovascular disease or retinal disease comprising the same as an active ingredient And a method for treating cardiovascular disease or retinal disease using the composition.
- BM microvascular basement membrane
- vascular endothelial cells In the vascular system, various types of cells come into contact with the microvascular basement membrane (BM), and these cells interact with vascular endothelial cells to participate in maintaining and determining the characteristics of the microvascular vessels.
- the cells that interact and specifically distribute between vascular endothelial cells and BM are perivascular progenitor cells.
- Perivascular progenitor cells are a kind of mesenchymal stem cells and are known to exist in capillaries, precapillaries, posterior capillaries, or collecting ducts of various human tissues such as the placenta, bone marrow, and adipose tissue.
- perivascular progenitor cells are similar to vascular smooth muscle cells, they are distinguished by markers, shape, and location in contact with the vascular endothelium. In the aorta or vein, vascular smooth muscle cells have a special structure and can be distinguished from perivascular cells because they are separated from BM where extracellular matrix is present. Perivascular progenitor cells, on the other hand, are present in the vascular endothelial basement membrane.
- Perivascular progenitor cells are known to surround the outer portion of the vascular endothelial by about 10 to 50%. These differences indicate that the degree of enveloping blood vessels varies according to the type and proportion of perivascular progenitor cells and endothelial cells. In skeletal muscle, their ratio is about 1: 100 (perivascular progenitor cells: vascular endothelial cells) and in the retina, the ratio is 1: 1. Perivascular progenitor cells surround the blood vessels most often in the central nervous system microvascular. The reason why the distribution of perivascular progenitor cells in the central nervous system is higher than in other organs is not yet clear, but it is likely that it contributes to the formation and maintenance of the blood-brain barrier.
- perivascular progenitor cells show morphological diversity depending on the organs present. In the central nervous system, the shape of perivascular progenitor cells is flat or elongated. It appears in a variety of forms, from radioactive perivascular cells that are in contact with the outside of blood vessels to rounded or glomerular mesial cells, or to localized contact with BM.
- Perivascular progenitor cells can differentiate into vascular smooth muscle cells during vascular growth or remodeling during development, whereas vascular smooth muscle cells can also differentiate into perivascular progenitor cells.
- Perivascular progenitor cells are also characterized by mesenchymal stem cells with the ability to differentiate into fibroblasts, osteoblasts, chondrocytes and adipocytes. Recently, it has been reported that perivascular progenitor cells identified in the human body have differentiation ability into cardiomyocytes.
- perivascular progenitor cells Although the existence and role of perivascular progenitor cells have been underestimated for a long time, the perivascular progenitor cells have recently attracted attention as essential elements for the formation of microvascular system by acting as progenitor cells through various differentiation ability. , A key regulator in stabilization, maturation and remodeling. It is also considered an important target in understanding vascular physiology and studying the causes and treatment of vascular diseases. Peripheral progenitor cell studies recently identified in human tissues have identified their new function and emphasize the fact that perivascular progenitor cells have great biological significance. The demand for mass supply of perivascular progenitor cells is increasing. However, there is a lack of typical markers for perivascular progenitor cells, and it is practically impossible to identify enough cells to meet demand in human tissue. In addition, the amount of perivascular progenitor cells in limited tissues does not meet the demand for perivascular progenitor cells.
- the present inventors have made diligent efforts to find a method for mass production of perivascular progenitor cells. As a result, the present inventors have developed a differentiation technique for obtaining perivascular progenitor cells by applying coating substrate-dependent single cell attachment technology from embryonic bodies derived from human embryonic stem cells. It was. In this process, it was found that high-purity perivascular progenitor cells can be isolated and obtained, thus completing the present invention.
- An object of the present invention is to treat a BMP4 growth factor in an embryoid body derived from stem cells to differentiate into mesodermal cells, separating the differentiated mesodermal cells into single cells, and coating the substrate thereon. It is to provide a method for producing perivascular progenitor cells from stem cells, comprising the step of culturing in a cultured plate attached only to the cells attached naturally.
- Another object of the present invention to provide a perivascular progenitor cell prepared by the above method.
- Another object of the present invention is to provide a method for differentiating the perivascular progenitor cells into perivascular cells, smooth muscle cells, muscle cells, adipocytes or bone cells.
- Another object of the present invention to provide a pharmaceutical composition for the prevention or treatment of cardiovascular diseases comprising the perivascular progenitor cells as an active ingredient.
- Another object of the present invention to provide a pharmaceutical composition for the prevention or treatment of retinal diseases, including the perivascular progenitor cells as an active ingredient.
- perivascular progenitor cells derived from stem cells characterized by natural selection by substrate-specific adhesion culture of the present invention are purely separated without additional processes such as flow cytometry or magnetic cell sorting. It was confirmed that it can be grown by. In addition, it was confirmed that it has a characteristic multipotent ability of perivascular progenitor cells. Therefore, stem cell-derived perivascular progenitor cells prepared by the method of the present invention can be usefully used as a cell therapy composition for the treatment or prevention of cardiovascular diseases or retinal diseases.
- FIG. 1 is a diagram showing mesoderm specific induced differentiation of BMP4.
- A shows the induced differentiation of H9 hESC embryoid bodies by BMP4 added from the start of culture (EB day 0) to EB day 8.
- B shows the percentage of viable cells positive for KDR and PDGFR ⁇ determined by flow cytometry on EB day 6. Bars represent standard error for the mean of three independent experiments compared to the NT group; ** p ⁇ 0.01, *** p ⁇ 0.001.
- FIG. 2 is a diagram showing substrate dependent adhesion of single cells derived from BMP4-EB.
- A single cells from undifferentiated H9 hESCs dispensed into cell culture dishes
- B single cells from EB on day 6 of BMP4 induced differentiation dispensed into uncoated plastic Petri dishes
- C, D coated with collagen Single cells from naturally-differentiated EBs dispensed into the dish
- E, F single cells from EBs on day 6 of BMP4 induced differentiation dispensed into collagen coated dishes.
- Black scale bar 20 ⁇ .
- Figure 3 is a diagram showing substrate dependent natural selection of PDGFR ⁇ -positive cell populations in BMP4-EB.
- A shows each cell dispensed in collagen coated dishes after separation of distinct PDGFR ⁇ -positive and negative populations using FACS sorter
- B shows single cell collagen matrix derived from BMP4-EB without separation
- Figure shows dependent attachment.
- Black scale bar 20 ⁇ .
- Figure 4 is a diagram showing the flow cytometry results of hESC-PVPC.
- A shows flow cytometry and
- B shows the overlap of each histogram for 8 independent hESC-PVPCs.
- Figure 5 shows the generalized perivascular potential of hESC-PVPC.
- A, B are the results of microscopic analysis of long-term cultured H9 hESC-PVPC and human placenta-induced perivascular cells. Positive for blue DAPI, green NG2, negative for red SMA.
- E Introduce DiO-labeled hESC-PVPCs to the ablumenal surface of DiI-labeled HUVECs cell lines in vascular induction tunnels using EC-perivascular tube coassembly in three-dimensional collagen matrix.
- F Bilayer ellipsoid formation of DiO-labeled HUVECs (internal) and DiI-labeled hESC-PVPCs (outer). White scale bar, 20 mm.
- FIG. 6 is a diagram showing the characteristics of smooth muscle derived from hESC-PVPC.
- A RT-PCR analysis of ⁇ SMA expression in hESC, hESC-PVPC, PVPC-induced day 2-4 SMLC and mature SMLC
- Immunocytochemistry shows the expression of ⁇ SMA in hESC (C), SMLC (D) and contractile SMLC (E and F).
- ⁇ SMA red
- DAPI blue
- SM-MHC green, E
- calponin green, F.
- Scale bar is 100 ⁇ m.
- FIG. 7 is a diagram showing the characteristics of myocytes derived from hESC-PVPC. Morphological changes in hESC-PVPC under myocyte differentiation conditions.
- A, D is hESC-PVPC
- B, E is myocyte differentiation medium
- C, D is typical cell morphology in myocyte growth medium, respectively.
- Black scale bars in (A-C) show 20 ⁇ m (D-F) immunocytochemistry showing myocyte specific Desmin expression. Desmine is red and DAPI is blue.
- FIG. 9 is a diagram showing the pigment transfer analysis between hESC-PVPC and other vascular systems.
- DiI-labeled hESC-PVPC top panel: red
- calcine-labeled HUVEC center panel, left: green
- UASMC center panel, center: green
- cardiomyocytes center panel, right: green
- Calcine's green-fluorescence was detected in hESC-PVPC (bottom panel) with red-fluorescence of DiI (bottom panel: red green, asterisk).
- FIG. 10 is a diagram showing the improvement of ischemic limb rescue / blood flow in ischemic Fuji after hESC-PVPC transplantation.
- A shows image analysis showing a series of lower limb recovery on days 0 and 28 after treatment
- B shows blood perfusion rate of ischemic limbs measured by series of laser Doppler images on days 0 and 28 after treatment. Indicates.
- FIG. 11 shows capillary density in ischemic myocardium increased by hESC-PVPC transplantation.
- a to C are representative photographs of the Masson tricolor-stained heart cross-section, (A) sham group injected with medium, (B) group injected with hESC-PVPC and (C) injected CB-EPC Photo for a group.
- D represents the fibrosis area of the infarct myocardium
- E the scar length of the infarct myocardium
- F the capillary density in the infarct myocardium
- G the infarct wall thickness of the infarct myocardium.
- H, I represents GS-lectin (green) staining capillaries of the cardiomyopathy region.
- FIG. 12 shows engraftment in the perivascular region of damaged blood vessels of hESC-PVPC transplanted in diabetic retinal disease model.
- DiI red
- STZ streptozotocin
- Labeled hESC-PVPC was injected into the retina to confirm engraftment after 4 weeks.
- the animal models used were mice with blood glucose levels of 250 mg / dl or more after the first 5 injections of diabetes (once a day), and were used after 8 weeks of diabetes in consideration of the time required to induce retinopathy. .
- FITC-labeled dextran FITC-labeled dextran (dextran-FITC, green) was injected into the tail blood vessel before retinal extraction. It was confirmed that the transplanted hESC-PVPC is located in the retina, especially around the vessels of the damaged blood vessels.
- Figure 13 is a diagram showing the transplantation of hESC-PVPC, human bone marrow-derived mesenchymal stem cells (BM-MSC) and human placenta-derived mesenchymal stem cells (placenta-MSC) in diabetic retinal disease model, and confirmed the engraftment for one month after transplantation to be.
- BM-MSC human bone marrow-derived mesenchymal stem cells
- placenta-MSC human placenta-derived mesenchymal stem cells
- hESC-PVPC is engrafted in the perivascular area and does not cause additional retinal vascular damage.
- the transplanted cells do not engraft and retinal detachment and lens-retinal stenosis are induced. It was confirmed to lead to death.
- the present invention comprises the first step of differentiating into mesodermal cells by treating BMP4 growth factor in the stem cell-derived embryonic body (embryoid body); Separating a embryoid body comprising differentiated mesodermal cells into single cells; And a third step of culturing the isolated single cells in a substrate coated with a substrate, thereby proliferating only the attached cells by natural selection.
- the first step is a step of differentiating mesodermal cells by treating BMP 4 growth factors in embryoid bodies derived from stem cells.
- the embryoid bodies derived from stem cells differentiated into mesodermal cells are cultured under appropriate differentiation conditions, thereby increasing the cell population in which the expression of mesodermal markers is increased by at least one or more.
- the expression of mesodermal markers can be detected by biochemical or immunochemical methods, and methods capable of detecting mesodermal markers can be used without limitation.
- specific polyclonal antibodies or monoclonal antibodies that bind to mesodermal markers can be used.
- Antibodies targeting individual specific markers can be used commercially and without limitation, those prepared by known methods.
- Representative mesodermal markers include, but are not limited to, KDR, PDGFR ⁇ / ⁇ CD29, CD31, CD34, CD73, CD90, and CD105.
- the first step may be performed for 6 to 8 days, preferably for 6 days.
- KDR and PDGFR ⁇ expression of mesodermal cell markers increased more than 30% and 50%, respectively, in the embryoid body (FIG. 1B).
- stem cell refers to a pluripotency capable of differentiating into cells derived from endoderm, mesoderm, and ectoderm of an animal, or a cell closely related to tissue or function. It means a cell having a multipotency that can be differentiated into. Means cells that have the ability to self-replicate and differentiate into two or more new cells, and include totipotent stem cells, pluripotent stem cells, and pluripotent stem cells ( multipotent stem cells).
- embryonic stem cell refers to an embryo that is capable of differentiating into all cells of an animal by extracting an inner cell mass from an blastocyst embryo immediately before implantation into an uterus of a mother. Means a cell having a pluripotency, but is not limited thereto, such as an embryonic body derived from embryonic stem cells or induced pluripotent stem cells (iPS cells) Similar stem cells are also included.
- iPS cells induced pluripotent stem cells
- adult stem cell refers to a cell having a multipotency by separating stem cells existing in each tissue and culturing in vitro, and including bone marrow stem cells, retinal stem cells, and M. glial cells in the retina. Cells, neural stem cells, and the like.
- the stem cells may be derived from mammals including humans and primates, as well as domestic animals such as cattle, pigs, sheep, horses, dogs, mice, rats, and cats, preferably humans.
- Stem cells of the present invention may be human embryonic stem cells (embryonic stem cells), induced pluripotent stem cells (iPSC) and somatic cell nuclear transfer cells (SCNT), preferably May be an embryonic stem cell, more preferably a human embryonic stem cell.
- the stem cells may be derived from mammals including humans, and most preferably may be human derived cells.
- BMP4 refers to a growth factor as a protein encoded by the BMP4 gene in humans.
- Bond morphogenetic protein (BMP) is a signaling protein belonging to the transforming growth factor- ⁇ (TGF- ⁇ ) superfamily that regulates early fetal differentiation, fetal tissue formation, and maintenance of adult tissue homeostasis.
- TGF- ⁇ transforming growth factor- ⁇
- the extracellular secreted BMP binds to type I and type II serine / threonine kinase receptors on the cell membrane to initiate BMP signaling.
- the type 2 receptor phosphorylates the type 1 receptor, and the phosphorylated type 1 receptor phosphorylates the Smad protein, a substrate in the cell, for intracellular signaling.
- Smad proteins regulated by receptors are called receptor regulated Smads (R-Smads), and Smad-1, 2, 3, 5 and 8 belong to R-Smad. They bind to Smad-4, a common partner Smad (Co-Smad) in the cell, move into the cell nucleus and bind to transcription factors to regulate transcription of target genes (Yamamoto & Oelgeschlager, Naturwissenschaften, 91: 519-34, 2004 ).
- embryoid body refers to cell aggregates derived from embryonic stem cells.
- Cell aggregation occurs by drop culture, untreated cell culture dish culture or stirred culture, which prevents cells from adhering to the surface to form typical colony proliferation.
- Differentiation begins with aggregation, and cells begin to repeat a limited degree of embryogenicity.
- the goblet is derived from pluripotent cells and thus can consist of a wide variety of differentiated cell types.
- differentiation in embryoid bodies despite occurring in a three-dimensional manner, is not very systematic compared to the case of carefully organized normal embryology. Embryos, however, are still good model systems for studying cellular and molecular interactions in the early stages of development, which are still difficult to study. Therefore, in the present invention, the differentiation was induced by treating BMP4 growth factor from the first day of the formation of embryoid bodies to differentiate them into mesodermal cells.
- meodermal cell refers to cells capable of differentiating into muscle cells, bone cells, chondrocytes, adipocytes, reticular cells or other connective tissue cells, including blood cells, vascular endothelial cells, smooth muscle and myocardium, and the like. it means.
- the second step is the step of separating the embryoid body containing the differentiated mesodermal cells into single cells, in the present invention, the enzyme was treated for single cell formation of the embryoid body.
- the enzyme may be used without limitation as long as it can inhibit intercellular binding by degrading cell surface proteins involved in intercellular binding.
- trypsin, trypsin-EDTA, collagenase, etc. may be used, but is not limited thereto.
- it may be a hypoallergenic enzyme, more preferably TrypLE (Invitrogen).
- the isolated single cells are cultured in a substrate coated with a substrate, and only the attached cells are propagated by natural selection.
- natural selection of the present invention does not use flow cell sorting or magnetic cell sorting techniques, which are used in the art to isolate highly differentiated specific cells. It means to separate naturally by using the inherent characteristics of the cell.
- flow cytometry or magnetic cell sorting technology there is an advantage in that purity can be confirmed through cell-specific markers.
- the cost is increased by the use of a variety of reagents, machines, and the like.
- the method of the present invention does not require a machine or the like, and has the advantage of differentiating without the problem of cell deformation or high cost by using simple natural selection.
- the natural selection technique of the present invention focuses on the functional characteristics of the cells that can be distinguished, rather than focusing on cell specific markers. Examples are selective cultures due to media suitability or specific surface adhesion.
- the "substrate" of the present invention can be used without limitation so long as it can help the selective attachment of cells to the culture dish, for example collagen or laminin may be used, preferably collagen may be used.
- the third step may be performed for 16 to 24 hours, preferably 18 hours.
- the naturally selected high purity perivascular progenitor cells can continue to proliferate, differentiate into various tissue cells, or be recovered and cryopreserved.
- 5 mg / ml collagen was coated on a culture plate as a specific substrate.
- the BMP4 growth factor-induced mesodermal cell embryoid body was subjected to enzymatic treatment and single cell culture, followed by incubation for 18 hours in a collagen-coated culture dish. Thereafter, only the cells specifically attached to collagen were naturally selected through medium exchange.
- PDGFR ⁇ platelet-derived growth factor receptor ⁇
- PDGFR ⁇ is a protein that is encoded by the PDGFRB gene in humans.
- the gene encodes a cell surface tyrosine kinase receptor belonging to the PDGF family.
- the growth factor is one of specific markers of mesodermal cells as a proliferation agent for mesenchymal-derived cells.
- the cells before and after the culture by the natural selection technology it is confirmed that the cells proliferated after the culture by the natural selection technology are cultured and grown in high purity only the cells positive for PDGFR ⁇ . This showed similar levels of purity as the selective culture of only cells positive for PDGFR ⁇ by flow cytometry (FIG. 3).
- the present invention provides a perivascular progenitor cell prepared by the above method.
- Peripheral progenitor cells prepared by the present invention showed positive expression patterns in the progenitor cell-specific PDGFR, CD44, CD146 and NG2 and mesenchymal stem cell-specific markers CD73 and CD90, etc.
- the markers CD31 and CD144 and the hematopoietic cell specific markers CD34 and CD45 and the like are not expressed (FIG. 4).
- the perivascular progenitor cells have differentiation ability into mesenchymal cell-derived differentiated bodies such as cartilage, osteoblasts and adipocytes, smooth muscle cells and myocytes (FIGS. 5 to 8), and are lost when applied as a cell therapy composition in a cardiovascular disease model. Has the potential to recover (FIGS. 10 and 11).
- progenitor cell refers to a cell capable of asymmetric division and is called a precursor, and because of asymmetric division, even if each cell has the same number of passages, Some are differentiated and some are multiplied, and may differ in age and nature.
- the perivascular progenitor cells produced by the above method is angiogenic growth factors (angiogenic growth factors), fibroblast growth factor (FGF), vascular endothelial growth factor (VEGF), IGF (insulin-like)
- angiogenic growth factors angiogenic growth factors
- FGF fibroblast growth factor
- VEGF vascular endothelial growth factor
- IGF insulin-like
- fetal bovine serum 10 ng / ml angiogenic growth factor, 10 ng / ml FGF, 20 ng / ml VEGF, 10 ng / ml IGF, 10 ng / in EGM-2MV medium. It may be cultured in perivascular cell differentiation medium containing ml EGF, ascorbic acid, GA-1000, but is not limited thereto.
- pericyte refers to vascular wall cells buried in the basement membrane of the microvascular system, which forms a specific local contact with the vascular endothelium.
- Connective tissue cells that form around small vessels also called Rouget cells, adventitial cells, or wall cells, are known to surround about 10-50% of the outer area of vascular endothelium. It is a long, narrow contractile cell that surrounds the precarotid artery outside the basement membrane. It is a relatively undifferentiated multipotent cell and functions to support blood vessels. If necessary, they can also be differentiated into fibroblasts, smooth muscle or macrophages. In addition to angiogenesis, it plays an important role in the stability of the blood-brain barrier.
- Adhesion to cells in the blood vessels has emerged as a blood flow regulator in the microvascular system, and in fact it is possible to regulate blood flow at the capillary level. Therefore, the study of perivascular cells is considered to be an important target for understanding vascular physiology and studying the causes and treatment of vascular diseases.
- the perivascular progenitor cells produced by the above method is a minimum essential medium non-essential amino acids (NEAA, Invitrogen), penicillin / streptomycin (penicillin / streptomycin; PS, Invitrogen), ⁇ -mercaptoethanol (Invitrogen) provides a method for differentiating perivascular progenitor cells into smooth muscle cells, characterized in that the culture medium.
- NEAA Invitrogen
- penicillin / streptomycin penicillin / streptomycin
- PS Invitrogen
- ⁇ -mercaptoethanol provides a method for differentiating perivascular progenitor cells into smooth muscle cells, characterized in that the culture medium.
- it may be cultured in a smooth muscle cell differentiation medium containing 5% FBS, 1% NEAA, 1% PS, 0.1 mM ⁇ -mercaptoethanol in DMEM (Dulbecco's modified eagle medium) medium, but is not limited thereto.
- smooth muscle cell refers to a muscle without a horizontal pattern in muscle, and all internal organs other than the heart muscle of a vertebrate are smooth muscle. Although contraction rate is slow, it is involuntary muscle that does not feel fatigue easily. It is long and fusiform, rarely multinucleus, but usually has one elliptical nucleus in the center. It is found in various parts of the body, such as the walls of blood vessels and walls of hollow organs such as the stomach, intestines, and uterus. Because it plays a pivotal role in the regulation of various human functions, a number of human diseases are associated with the dysfunction of smooth muscle.
- the perivascular progenitor cells prepared by the above method is prepared by horse serum (HS, Invitrogen), chicken embryo extract (CEE, Sera Lab, Wales, UK), PS It provides a method for differentiating perivascular progenitor cells into myocytes, characterized in that it is cultured in a culture medium comprising.
- the medium may be cultured in myocyte differentiation medium containing 10% FBS, 10% HS, 1% CEE, 1% PS in DMEM high glucose (DMEM) medium, but is not limited thereto.
- myocyte also known as muscle cells (muscle cells) arise from myoblasts (myoblast). Each myocyte contains a long myofibril, a long chain of sarcomere, and a contraction unit of the cell. Myocytes have a variety of differentiated forms of varying properties, including heart, skeleton, or smooth muscle cells.
- the perivascular progenitor cells prepared by the above method is insulin (Sigma-Aldrich), dexamethasone (Sigma-Aldrich), isobutylmethylxanthine (Sigma-Aldrich), India It provides a method for differentiating perivascular progenitor cells into adipocytes, which is characterized by culturing in a culture medium containing metasin (indomethacin, Sigma-Aldrich).
- DMEM medium Preferably in low glucose DMEM medium can be cultured in adipocyte differentiation medium containing 10% FBS, 5 ⁇ g / ml insulin, 1 ⁇ M dexamethasone, 0.5 mM isobutylmethyl xanthine, 60 ⁇ M indomethacin, but is not limited thereto. no.
- adipocyte refers to the major cells that make up adipose tissue that stores energy in the form of fat. It exists in two forms: white fat cells, which contain large fat droplets surrounded by a cytoplasmic layer, and polygonal brown fat cells, which contain a significant amount of cytoplasm, evenly dispersed fat droplets. White adipocytes secrete proteins that act as adipokine, such as resistin, adiponectin and leptin.
- the perivascular progenitor cells produced by the above method is cultured in a culture medium containing dexamethasone, ⁇ -glycerophosphate, ascorbic acid-2-phosphate It provides a method for differentiating into bone cells.
- the present invention may be cultured in low concentration glucose DMEM medium in osteoblast differentiation medium containing 10% FBS, 1 ⁇ M dexamethasone, 10 mM ⁇ -glycerophosphate, 60 ⁇ M ascorbic acid-2-phosphate, but not limited thereto. .
- osteocyte of the present invention is a star-shaped cell that is most abundant in dense bone tissue and includes a thin ring of nucleus and cytoplasm. Osteoblasts are trapped in the secreted matrix and form bone cells. Bone cells connect to each other through long cytoplasmic kidneys that fill small ducts called microtubules that are used to exchange nutrients and waste through gap junctions. On the other hand, osteocytes are reduced in their ability to synthesize, do not mitosis, are produced in the mesenchyme, and hydroxyapatite, calcium carbonate, and calcium phosphate accumulate around the cells.
- the present invention provides a pharmaceutical composition for the prevention or treatment of cardiovascular diseases comprising the prepared perivascular progenitor cells as an active ingredient.
- prevention of the present invention means any action that inhibits or delays the onset of cardiovascular disease by the administration of the pharmaceutical composition according to the present invention
- treatment refers to cardiovascular disease by the administration of the pharmaceutical composition It means any action that improves or beneficially changes the symptoms caused by it.
- the composition of the present invention can be used without limitation in the prevention or treatment of cardiovascular diseases.
- cardiovascular disease of the present invention includes heart disease and vascular disease, and refers to a disease that occurs in the arteries and veins including the heart and affects the heart and blood vessels. Risk factors such as hyperlipidemia, diabetes and smoking are all causes of blood vessel damage and can lead to cardiovascular disease. However, there are usually no clear symptoms, usually called silent disease. Most of the conditions are already severe when symptoms begin to develop asymptomatically and worsen without awareness. Many of these cardiovascular diseases are also associated with adult diseases.
- Examples include hyperlipidemia, in which blood cholesterol and fat components are accumulated in blood vessels due to poor blood circulation, arteriosclerosis, which hardens as the arteries age, and high blood pressure caused by high pressure during the contraction and relaxation of the heart.
- Representatives include hypotension, angina caused by pericardial coronary artery disease, and myocardial infarction caused by heart muscle abnormalities.
- cardiovascular diseases treatable by the present invention include, for example, heart failure, hypertensive heart disease, arrhythmia, heart valve disease, ventricular septal defect, congenital heart disease, cardiomyopathy, pericardial disease, stroke, peripheral vascular disease, aneurysm, arteriosclerosis, Coronary artery disease or high blood pressure may be included, but is not limited thereto.
- the perivascular progenitor cells of the present invention implanted in the mouse has an effect of improving ischemic limb relief / blood flow in ischemic Fuji (FIG. 10). It was also confirmed that there is an effect of increasing the capillary density in the ischemic myocardium (FIG. 11). Therefore, it was confirmed that perivascular progenitor cells derived from stem cells of the present invention have the potential to treat cardiovascular diseases as cell therapy compositions.
- composition of the present invention may be used interchangeably with "cell therapy composition”, which is a cell and tissue prepared through isolation, culture and special manipulation from humans, which is used for the purpose of treatment, diagnosis and prophylaxis.
- cell therapy composition which is a cell and tissue prepared through isolation, culture and special manipulation from humans, which is used for the purpose of treatment, diagnosis and prophylaxis.
- U.S. FDA Regulations treats, diagnoses, and treats live, autologous, or heterologous cells in vitro or otherwise alters the biological properties of cells to restore the function of cells or tissues. Means a drug used for the purpose of prevention.
- the cell therapy composition of the present invention may further comprise a pharmaceutically acceptable carrier.
- “Pharmaceutically acceptable” means that the cells or humans exposed to the composition are not toxic.
- the carrier can be used without limitation so long as it is known in the art such as buffers, preservatives, analgesics, solubilizers, isotonic agents, stabilizers, bases, excipients, lubricants, preservatives and the like.
- the pharmaceutical compositions of the present invention can be prepared according to techniques commonly used in the form of various formulations.
- the cell therapy agent of the composition of the present invention can be administered by any route as long as it can induce migration to the disease site. In some cases, one may consider loading the vehicle with a means for directing stem cells to the lesion.
- compositions of the present invention may be used topically (including buccal, sublingual, skin and intraocular administration), parenteral (including subcutaneous, intradermal, intramuscular, instillation, intravenous, intraarterial, intraarticular and cerebrospinal fluid) or transdermal administration. It can be administered through several routes, including parenteral, most preferably directly to the affected area.
- the stem cells can be administered to a subject by suspending it in a suitable diluent at a concentration of about 1 ⁇ 10 3 to 5 ⁇ 10 6 cells / ml, which dilution protects and maintains the cells and upon injection into the desired tissue. Used for ease of use.
- the diluent may include a saline solution, a phosphate buffer solution, a buffer solution such as HBSS, plasma, cerebrospinal fluid, or a blood component.
- the pharmaceutical composition can be administered by any device such that the active substance can migrate to the target cell. Preferred modes of administration and preparations are injections.
- Injections include aqueous solvents such as physiological saline solution, ring gel solution, Hank's solution or sterilized aqueous solution, vegetable oils such as olive oil, higher fatty acid esters such as ethyl oleic acid, and non-aqueous solvents such as ethanol, benzyl alcohol, propylene glycol, polyethylene glycol or glycerin
- aqueous solvents such as physiological saline solution, ring gel solution, Hank's solution or sterilized aqueous solution
- vegetable oils such as olive oil
- higher fatty acid esters such as ethyl oleic acid
- non-aqueous solvents such as ethanol, benzyl alcohol, propylene glycol, polyethylene glycol or glycerin
- non-invasive agents known in the art, suitable for the barrier to pass through, for mucosal permeation, and ascorbic acid, sodium hydrogen sulfite, BHA, tocopherol, EDTA as
- an emulsifier a buffer for pH adjustment, phenyl mercury nitrate, chimerosal, benzalkonium chloride, phenol, cresol, benzyl alcohol, and the like, and a pharmaceutical carrier such as a preservative for preventing the growth of microorganisms.
- the present invention provides a pharmaceutical composition for the prevention or treatment of retinal diseases, including the prepared perivascular progenitor cells as an active ingredient.
- prevention of the present invention means any action that inhibits or delays the onset of retinal disease by the administration of the pharmaceutical composition according to the present invention
- treatment refers to retinal disease by the administration of the pharmaceutical composition It means any action that improves or beneficially changes the symptoms caused by it.
- the composition of the present invention can be used for the prevention or treatment of retinal diseases.
- retinal disease refers to a disease caused by abnormal regeneration of vascular endothelial cells due to impaired pericyte or retinal degeneration due to leakage of blood or body fluids. Thus, it can be treated by implanting perivascular or differentiateable cells into the retina.
- retinal diseases treatable by the present invention include wet age related macular degeneration (neovascular or exudative AMD), glaucoma, diabetic retinopathy, wet age related retinopathy, Retinopathy of prematurity, but is not limited thereto.
- the perivascular progenitor cells derived from the stem cells of the present invention transplanted into the retina of the diabetic retinal disease mouse model have an effect of engulfing the perivascular vessels of the damaged blood vessels to alleviate the damage.
- FIG. 12 when compared with the transplantation of adult-derived stem cells, the transplanted adult-derived stem cells did not engraft in the desired location, it was confirmed that causing retinal detachment, lens-retinal stenosis excessive inflammatory reactions leading to death (Fig. 13). ). Therefore, it was confirmed that the perivascular progenitor cells derived from the stem cells of the present invention have the potential to treat retinal diseases as cell therapy compositions.
- the present invention provides a method for preventing or treating a cardiovascular disease of a subject, comprising administering the pharmaceutical composition for preventing or treating the cardiovascular disease to a subject in need thereof.
- the term "individual" of the present invention means any animal including a human having or may develop a cardiovascular disease as described above, and can effectively prevent or treat the diseases by administering the pharmaceutical composition of the present invention to an individual.
- the pharmaceutical composition of the present invention can be administered in parallel with existing therapeutic agents.
- administration means introducing a predetermined substance into a patient in an appropriate manner, and the route of administration of the composition may be administered via any general route as long as it can reach the target tissue.
- Intraperitoneal administration intravenous administration, intramuscular administration, subcutaneous administration, intradermal administration, oral administration, topical administration, nasal administration, pulmonary administration, rectal administration, but is not limited thereto.
- compositions of the present invention may be administered by any device in which the active substance may migrate to target cells.
- Preferred modes of administration and preparations are intravenous, subcutaneous, intradermal, intramuscular, injectable and the like.
- Injections include non-aqueous solvents such as aqueous solvents such as physiological saline solution and ring gel solution, vegetable oils, higher fatty acid esters (e.g., oleic acid, etc.), and alcohols (e.g., ethanol, benzyl alcohol, propylene glycol, glycerin, etc.).
- Stabilizers e.g.
- Preservatives eg, mercury nitrate, chimerosal, benzalkonium chloride, phenol, cresol, benzyl alcohol, etc. may be included.
- the present invention provides a method for preventing or treating a cardiovascular disease of an individual, comprising administering the pharmaceutical composition for preventing or treating the retinal disease to an individual in need thereof.
- the present invention provides a use of the stem cell-derived perivascular progenitor cells for the prevention or treatment of cardiovascular diseases.
- the present invention provides a use for the preparation of a cardiovascular disease therapeutic agent of the stem cell-derived perivascular progenitor cells.
- the present invention provides a use for the prevention or treatment of retinal disease of the stem cell-derived perivascular progenitor cells.
- the present invention provides a use for the manufacture of a therapeutic agent for retinal disease of the stem cell-derived perivascular progenitor cells.
- a culture dish (Nunc, Roskide, Denmark) coated with undifferentiated human embryonic stem cells (H9, CHA11-hESC) with 0.1% gelatin, mitomycin C (Sigma, St. Louis, MO, USA) was cocultured with mouse embryonic fibroblasts (MEFs) prepared by treatment at a concentration of 10 ⁇ g / ml for 90 minutes as support cells.
- H9 human embryonic stem cells
- CHA11-hESC undifferentiated human embryonic stem cells
- mitomycin C Sigma, St. Louis, MO, USA
- hESC is DMEM / F12 (50:50, Invitrogen, Carlsbad, CA, USA), 20% (v / v) serum replacement (SR), 1% non-essential amino acid (Invitrogen), 1% penicillin strepto A culture consisting of mycin (Invitrogen), 0.1% beta-mercaptoethanol (Invitrogen), 4 ng / ml bFGF (basix fibroblast growth factor; Invitrogen) was used. The culture was replaced every day, subcultured to new support cells every 5-7 days, cell culture was incubated while maintaining 37 °C, 5% carbon dioxide, 95% humidity.
- human embryonic stem cells were separated from support cells with dispase, transferred to a new culture dish (Falcon / BD Biosciences, San Jose, CA, USA), washed once, and washed with DMEM / Aggregation was induced by suspension for 45 minutes in a culture medium in which 2% (v / v) serum replacement (SR; Invitrogen) was added to F12 (50:50, Invitrogen).
- SR v / v
- F12 50:50, Invitrogen
- Aggregated embryoid bodies were 10% (v / v) SR, 1 mM L-glutamine (Invitrogen), 1% nonessential amino acid (NEAA; Invitrogen), 1% penicillin streptomycin (Invitrogen), 0.1% beta Transfer to culture medium composed of mercaptoethanol (Invitrogen) was suspended in culture. Cultures were replaced every other day and maintained for 5-6 days.
- Example 3 Increase of Mesodermal Cell Population in Embryos by BMP4 Growth Factor Treatment
- DMEM DMEM with 1 mM L-glutamine, 1% non-essential amino acid, 0.1% beta-mercaptoethanol and 10% SR added on the first day of embryoid formation to differentiate embryoid bodies prepared by the above method into mesodermal cells
- the / F12 culture was treated with 20 ng / ml BMP4 growth factor (Peprotech Inc., Rocky Hill, NJ, USA), and was replaced with the cultured BMP4 growth factor once every two days from the next day.
- Example 4 Single Cellization of Mesodermal Embryos by Hypoallergenic Enzyme Treatment
- the average number of embryoid bodies obtained from human embryonic stem cells maintained in one 60 mm culture dish is 80 to 100, and one embryonic body is composed of 2,500 cells on average. Therefore, in one step of the present invention, embryonic bodies obtained from human embryonic stem cells of six 60 mm culture dishes on average are 500 to 600, and single cells obtained from them are 1.5 to 2 ⁇ 10 6 .
- FIG. 2A shows undifferentiated H9 hESC single cells dispensed in a cell culture dish
- FIG. 2B shows spontaneous differentiation of single cells from EB on day 6 of BMP4 induction dispensed in an uncoated plastic Petri dish
- 2C and 2D in collagen coated dishes
- Single cells from EBs, and 2E and 2F are micrographs of single cells from EB on day 6 of BMP4 induction dispensed into collagen coated dishes.
- 2C and 2D show a non-uniform cell population in which epithelial cells and fibrous cells are observed together
- FIGS. 2E and 2F show a uniform cell population.
- Single cells prepared in Example 4 were incubated overnight in a cell culture apparatus at 37 ° C., 5% carbon dioxide conditions with EGM2-MV (Lonza) medium in a pre-prepared collagen coated culture dish.
- Single cells cultured in the above process is divided into floating and adherent population, the floating population was removed by natural selection through phosphate buffer solution and medium exchange process.
- cells attached through natural selection were set to passage 0. It is passaged every 2 to 3 days and propagates from passage 0 to passage 3 within 10 days of culture.
- 1.5 to 2 ⁇ 10 6 single cells are attached to 50 to 80% in a 35 mm collagen-coated culture dish, and in passage 3 to 3 to 5 ⁇ 10 7 in five T-75 flasks. Multiplies.
- Collagen used in the coating was a distilled water dilution solution of a collagen solution (Stem Cell Tech.) Of 3 mg / ml concentration.
- the collagen coating for passage 0 used a 5-fold diluted collagen solution, and subsequent passages used a 20-fold diluted collagen solution.
- the coating was performed by exposing the culture dish to the collagen solution of the corresponding concentration for at least 1 hour at a sterile work table at room temperature.
- FIG. 3A flow cytometry is shown in FIG. 3A for the types of cells cultured by categorizing positive and negative cell populations according to the expression level of PDGFR ⁇ with a flow cytometer.
- FIG. 3B cells cultured according to the natural selection method of the present invention is shown in Figure 3B. From the flow cytometry results after several days of culture, substrate specific natural selection, which is a characteristic step of the culturing method of the present invention, is capable of pure separation of perivascular progenitor cells without additional processes such as flow cytometry or magnetic cell sorting. Confirmed.
- Example 6 Search for marker markers of perivascular progenitor cells
- Cells prepared through the procedure of Examples 1 to 5 were analyzed by a flow cytometer (FACSCalibur flow cytometer; BD Bioscience, San Jose, CA, USA) using the markers of perivascular progenitor cells.
- Cells cultured in passage 3 were suspended using Tryp-LE, a hypoallergenic enzyme, transferred to an Eppendorf tube, washed with phosphate buffer solution and filtered using a 35 ⁇ m nylon mesh cell filter to 2% fetal bovine serum.
- Flow cytometry was performed after floating with phosphate buffer solution containing (Terracell, Webtec Inc., Charltte, NC, USA).
- Control markers include IgG isotype (PE, APC; BD Biosciences), other PDGFR ⁇ (PE; R & D systems), PDGFR ⁇ (PE; R & D systems), CD44 (PE, APC; R & D systems), KDR (PE; R & D systems) ), Tie-2 (PE; R & D systems), Flt-1 (PE; R & D systems), VE-CAD (PE; R & D systems), CD31 (PE; R & D systems), CD146 (PE; R & D systems), NG2 ( PE; R & D systems), CD34 (PE; R & D systems), CD45 (PE; R & D systems), CD105 (PE; R & D systems), CD90 (PE; R & D systems), CXCR4 (PE; R & D systems), SSEA4 (PE; R & D systems) and CD133 (APC; Miltenyi Biotec Inc.) were used.
- FIG. 4a The flow cytometry results are shown in FIG. 4.
- the perivascular progenitor cells of the present invention are positive for PDGFR ⁇ , PDGFR ⁇ , CD44, CD73, CD90, CD105, CD146, NG2, KDR, Tie2 and Flt1, and CD31, CD34, CD45, CD133, CD144 , CXCR4 and c-kit showed a negative immune phenotype.
- Peripheral progenitor cells identified by the method of Example 6 were continuously treated with 5% FBS, 10 ng / ml angiogenic growth factors, 10 ng / ml FGF, 20 ng / ml VEGF, 10 ng / ml IGF, 10 ng / ml
- EGM-2MV SingleQuots, Cambrex Bio Science
- FIG. 5A H9 hESC-PVPC
- FIG. 5B human placenta-induced perivascular cells
- FIG. 5C and FIG. 5D NG2 (Green, a specific marker of perivascular cells).
- SMA adult tissue specific marker SMA
- Peripheral vascular progenitor cells formed by co-culture of vascular endothelial cells and human embryonic stem cell-derived vascular endothelial progenitor cells in the three-dimensional collagen gel of FIG. 5E and the low adhesion culture dish of FIG. 5F (FIG. In 5E, the vascular morphology and in FIG. 5F were located outside of the elliptic inner cell mass, that is, in the perivascular region.
- Perivascular progenitor cells identified by the method of Example 6 were differentiated in DMEM medium containing smooth muscle cell differentiation medium, that is, 5% FBS, 1% NEAA, 1% PS, 0.1 mM ⁇ -mercaptoethanol, It was shown that the perivascular progenitor cells of the invention are capable of differentiating into smooth muscle cells.
- the expression of ⁇ SMA, a smooth muscle cell specific marker, was confirmed by RT-PCR, real-time PCR and immunocytochemistry by comparing with mature SMLC.
- FIGS. 6A and 6B show that the smooth muscle differentiation medium gradually increases with the days of induction of differentiation.
- Immunofluorescence staining in perivascular progenitor cells prior to differentiation ⁇ SMA was not expressed, but was expressed as it was differentiated into smooth muscle cells (FIGS. 6C and 6D).
- 6E and 6F showed that the mature smooth muscle cell specific markers SM-MHC (FIG. 6E, green) and calponin (FIG. 6F, green) were simultaneously expressed with ⁇ SMA.
- Peripheral progenitor cells identified by the method of Example 6 were cultured in high glucose DMEM medium containing 10% FBS, 10% HS, 1% CEE and 1% PS to differentiate into myocytes.
- 7A and 7D show the perivascular progenitor cells prior to differentiation
- FIGS. 7B and 7E show the cells that differentiate into myocytes in the myogenic differentiation medium
- FIGS. 7C and 7F finally show the morphology of muscle fibers under muscle growth medium.
- myocyte-specific marker desmine (red) was used.
- Peripheral progenitor cells identified by the method of Example 6 were 10% FBS, 5 ⁇ g / ml insulin, 1 ⁇ M dexamethasone, 0.5 mM isobutylmethylxanthine, and 60 ⁇ M indomethacin and 10% of low glucose DMEM medium.
- Adipocytes generated by differentiation under the above conditions were identified by oil red staining, and bone cells by alizarin red staining, which are shown in FIG. 8.
- hESC-PVPC human embryonic stem cell-derived vascular periphery progenitor cells
- Calcine is a fluorescence that is only transferred through gap-junction, which is part of cellular interactions. Thus, intercellular gap-junction formation is necessary for the transfer of calcine labeled in other cardiovascular cells to hESC-PVPC.
- DiI-labeled hESC-PVPC (FIG. 9, top panel, red) was calcine-labeled HUVEC (FIG. 9, middle panel left, green), UASMC (FIG. 9, middle panel center, green) or cardiomyocytes (FIG. 9, center).
- HESC-PVPC Human embryonic stem cell-derived perivascular progenitor cells
- FIG. 11 shows increased capillary density in ischemic myocardium with hESC-PVPC transplantation.
- Heart sections of the Siamese group injected with media FIG. 11A
- the experimental group implanted with hESC-PVPC FIG. 11B
- the comparative group injected with CB-EPC FIG. 11C
- 11D-11G show a decrease in infarct myocardial fibrosis area and infarct myocardial scar length and increase in capillary density and infarct wall thickness in infarct myocardium due to hESC-PVPC transplantation.
- 11H and 11I show staining of cardiomyopathy site capillary with GS-lectin (green). From the series of results, it was confirmed that the recovery effect of ischemic myocardial infarction through capillary regeneration in the hESC-PVPC transplantation experimental group.
- perivascular progenitor cells of the present invention can treat cardiovascular disease.
- hESC-PVPC was transplanted into mice induced with diabetic retinopathy to confirm the therapeutic effect.
- streptozotocin streptozotocin, STZ
- STZ streptozotocin
- Diabetes-induced model was used to measure the blood glucose level after the first five times (once a day) STZ was used to select mice with a value of 250 mg / dl or more, in consideration of the time required to induce retinopathy Those after 8 weeks were used.
- hESC-PVPC labeled with DiI red
- engraftment was confirmed 4 weeks after injection into the retina.
- FITC-labeled dextran (dextran-FITC, green) was injected into the tail blood vessel before retinal extraction.
- the transplanted hESC-PVPC was engrafted into the retina, and was found to be located particularly around the vessel of the damaged blood vessel.
- BM-MSCs Human bone marrow-derived mesenchymal stem cells
- placenta-derived mesenchymal stem cells placenta-derived mesenchymal stem cells
- hESC-PVPC is engrafted in the perivascular area and does not cause additional retinal vascular damage.
- the transplanted cells do not engraft and retinal detachment and lens-retinal stenosis are induced. It was confirmed to lead to death. This is shown in FIG. 13.
- the red label in the adult-derived mesenchymal stem cell transplant group in the figure is due to the autofluorescence of immune cells by inflammatory responses, not by DiI. It is clearly green-green luminescence that exists inside the retinal vessels regardless of cell transplantation pathway.
- perivascular progenitor cells of the present invention have excellent engraftment ability in the retina, and thus can treat retinal diseases that are not regenerated after perivascular cells are damaged.
Landscapes
- Health & Medical Sciences (AREA)
- Engineering & Computer Science (AREA)
- Life Sciences & Earth Sciences (AREA)
- Biomedical Technology (AREA)
- Chemical & Material Sciences (AREA)
- Wood Science & Technology (AREA)
- Organic Chemistry (AREA)
- Zoology (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Genetics & Genomics (AREA)
- Biotechnology (AREA)
- Microbiology (AREA)
- Biochemistry (AREA)
- General Engineering & Computer Science (AREA)
- General Health & Medical Sciences (AREA)
- Cell Biology (AREA)
- Developmental Biology & Embryology (AREA)
- Vascular Medicine (AREA)
- Rheumatology (AREA)
- Medicines Containing Material From Animals Or Micro-Organisms (AREA)
- Medicinal Chemistry (AREA)
- Molecular Biology (AREA)
- Micro-Organisms Or Cultivation Processes Thereof (AREA)
Abstract
La présente invention concerne un procédé de préparation de cellules progénitrices périvasculaires à partir de corps embryoïdes de cellules souches, une composition pour thérapie cellulaire visant à prévenir ou traiter des maladies cardiovasculaires ou des maladies de la rétine contenant lesdites cellules comme principe actif, et un procédé de traitement de maladies cardiovasculaires ou de maladies de la rétine utilisant la composition, et plus spécifiquement, un procédé de préparation de cellules progénitrices périvasculaires comprenant les étapes consistant à : traiter les corps embryoïdes de cellules souches avec le facteur de croissance BMP4 pour induire leur différentiation en cellules mésodermiques ; séparer les corps embryoïdes contenant les cellules mésodermiques différenciées en cellules uniques ; et les cultiver dans une boîte de Pietri revêtue d'un substrat pour ne faire proliférer que les cellules adhérentes par sélection naturelle, et les cellules progénitrices périvasculaires préparées par le procédé, une composition pour thérapie cellulaire pour prévenir ou traiter des maladies cardiovasculaires ou des maladies de la rétine contenant lesdites cellules comme principe actif, et un procédé de traitement de maladies cardiovasculaires ou de maladies de la rétine utilisant la composition.
Applications Claiming Priority (2)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
KR10-2012-0006371 | 2012-01-19 | ||
KR1020120006371A KR101477016B1 (ko) | 2012-01-19 | 2012-01-19 | 인간 배아줄기세포 유래 혈관주위 전구세포의 제조방법 및 이를 포함하는 세포치료 조성물 |
Publications (1)
Publication Number | Publication Date |
---|---|
WO2013108949A1 true WO2013108949A1 (fr) | 2013-07-25 |
Family
ID=48799364
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/KR2012/000769 WO2013108949A1 (fr) | 2012-01-19 | 2012-01-31 | Procédé de préparation de cellules progénitrices périvasculaires dérivées de cellules souches embryonnaires et composition pour thérapie cellulaire les contenant |
Country Status (2)
Country | Link |
---|---|
KR (1) | KR101477016B1 (fr) |
WO (1) | WO2013108949A1 (fr) |
Cited By (3)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US11566228B2 (en) | 2006-04-14 | 2023-01-31 | Astellas Institute For Regenerative Medicine | Hemangio-colony forming cells |
US12097223B2 (en) | 2011-11-30 | 2024-09-24 | Astellas Institute For Regenerative Medicine | Mesenchymal stromal cells and uses related thereto |
US12209255B2 (en) | 2012-07-12 | 2025-01-28 | Astellas Institute For Regenerative Medicine | Mesenchymal-like stem cells derived from human embryonic stem cells, methods and uses thereof |
Families Citing this family (1)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
KR20160002247A (ko) | 2014-06-30 | 2016-01-07 | 건국대학교 산학협력단 | 배아줄기세포 유래 다기능성 혈관 생성 주피 세포의 제조방법 및 이를 포함하는 세포치료 조성물 |
Citations (3)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
KR20090089206A (ko) * | 2008-02-18 | 2009-08-21 | 한국과학기술원 | 배아줄기세포를 혈관모세포로 분화 유도하는 방법 |
KR20090129230A (ko) * | 2008-06-12 | 2009-12-16 | (주)차바이오앤디오스텍 | 인간배아줄기세포 유래 혈관전구세포를 이용한 창상 치료방법 |
KR20110129842A (ko) * | 2009-03-24 | 2011-12-02 | 한국과학기술연구원 | 줄기세포를 혈관세포로 분화시키는 방법 및 이를 이용한 생체 내 혈관신생 유도 |
-
2012
- 2012-01-19 KR KR1020120006371A patent/KR101477016B1/ko active Active
- 2012-01-31 WO PCT/KR2012/000769 patent/WO2013108949A1/fr active Application Filing
Patent Citations (3)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
KR20090089206A (ko) * | 2008-02-18 | 2009-08-21 | 한국과학기술원 | 배아줄기세포를 혈관모세포로 분화 유도하는 방법 |
KR20090129230A (ko) * | 2008-06-12 | 2009-12-16 | (주)차바이오앤디오스텍 | 인간배아줄기세포 유래 혈관전구세포를 이용한 창상 치료방법 |
KR20110129842A (ko) * | 2009-03-24 | 2011-12-02 | 한국과학기술연구원 | 줄기세포를 혈관세포로 분화시키는 방법 및 이를 이용한 생체 내 혈관신생 유도 |
Non-Patent Citations (2)
Title |
---|
DAR ET AL.: "Multipotent vasculogenic pericytes from human pluripotent stem cells promote recovery of murine ischemic limb", CIRCULATION, vol. 125, 17 November 2011 (2011-11-17), pages 87 - 99, XP055229620, DOI: doi:10.1161/CIRCULATIONAHA.111.048264 * |
MANCUSO ET AL.: "Circulating perivascular progenitors: a target of PDGFR inhibition", INTERNATIONAL JOURNAL OF CANCER, vol. 129, 2 December 2010 (2010-12-02), pages 1344 - 1350 * |
Cited By (3)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US11566228B2 (en) | 2006-04-14 | 2023-01-31 | Astellas Institute For Regenerative Medicine | Hemangio-colony forming cells |
US12097223B2 (en) | 2011-11-30 | 2024-09-24 | Astellas Institute For Regenerative Medicine | Mesenchymal stromal cells and uses related thereto |
US12209255B2 (en) | 2012-07-12 | 2025-01-28 | Astellas Institute For Regenerative Medicine | Mesenchymal-like stem cells derived from human embryonic stem cells, methods and uses thereof |
Also Published As
Publication number | Publication date |
---|---|
KR20130085308A (ko) | 2013-07-29 |
KR101477016B1 (ko) | 2014-12-29 |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
Huang et al. | Efficient generation of lung and airway epithelial cells from human pluripotent stem cells | |
US10570369B2 (en) | Pluripotent autologous stem cells from oral mucosa and methods of use | |
US8192348B2 (en) | Engineered blood vessels | |
US20180171290A1 (en) | Method of producing progenitor cells from differentiated cells | |
US9867854B2 (en) | Therapeutic method using cardiac tissue-derived pluripotent stem cells | |
US20210079351A1 (en) | Tissue-specific differentiation matrices and uses thereof | |
JP6832049B2 (ja) | 心臓神経堤細胞、及びその使用方法 | |
JP6981751B2 (ja) | 心外膜細胞を形成するための方法及び組成物 | |
Ribatti et al. | Epithelial-mesenchymal interactions: a fundamental developmental biology mechanism | |
US20190153385A1 (en) | Method of producing progenitor cells from differentiated cells | |
US20030031651A1 (en) | Methods and reagents for cell transplantation | |
US20080145860A1 (en) | Encapsulated cell indicator system | |
US20120100110A1 (en) | Physiological methods for isolation of high purity cell populations | |
JP2019510758A (ja) | 間葉系マーカーおよびニューロンマーカーを発現する歯髄幹細胞の使用、ならびに神経学的疾患を治療するためのその組成物 | |
US9422522B2 (en) | Method of producing adipocytes from fibroblast cells | |
WO2013108949A1 (fr) | Procédé de préparation de cellules progénitrices périvasculaires dérivées de cellules souches embryonnaires et composition pour thérapie cellulaire les contenant | |
WO2012008733A2 (fr) | Cellules souches issues de tissu de placenta primaire et agent thérapeutique contenant celles-ci | |
AU2013287307A1 (en) | Derivation and self-renewal of IsI1+ cells and uses thereof | |
JP2008500821A (ja) | 心筋細胞への分化の改善 | |
WO2021096218A1 (fr) | Procédé d'isolement de culture pure de cellules endothéliales vasculaires, milieu de maintien des caractéristiques de cellules endothéliales vasculaires, et procédé de culture le comprenant | |
Asashima et al. | Elucidation of the role of activin in organogenesis using a multiple organ induction system with amphibian and mouse undifferentiated cells in vitro | |
Klemmt et al. | Murine amniotic fluid stem cells contribute mesenchymal but not epithelial components to reconstituted mammary ducts | |
KR20160002247A (ko) | 배아줄기세포 유래 다기능성 혈관 생성 주피 세포의 제조방법 및 이를 포함하는 세포치료 조성물 | |
D’Ippolito et al. | Isolation and characterization of swine MIAMI cells: a valuable animal model for adult stem cell therapy | |
JP2005287478A (ja) | ヒト脂肪前駆細胞株及びその利用方法 |
Legal Events
Date | Code | Title | Description |
---|---|---|---|
121 | Ep: the epo has been informed by wipo that ep was designated in this application |
Ref document number: 12866224 Country of ref document: EP Kind code of ref document: A1 |
|
NENP | Non-entry into the national phase |
Ref country code: DE |
|
122 | Ep: pct application non-entry in european phase |
Ref document number: 12866224 Country of ref document: EP Kind code of ref document: A1 |