EP1370642A2 - Cellules souches adultes pluripotentes provenant de tissu regeneratif - Google Patents
Cellules souches adultes pluripotentes provenant de tissu regeneratifInfo
- Publication number
- EP1370642A2 EP1370642A2 EP02712824A EP02712824A EP1370642A2 EP 1370642 A2 EP1370642 A2 EP 1370642A2 EP 02712824 A EP02712824 A EP 02712824A EP 02712824 A EP02712824 A EP 02712824A EP 1370642 A2 EP1370642 A2 EP 1370642A2
- Authority
- EP
- European Patent Office
- Prior art keywords
- cells
- adult stem
- stem cell
- composition
- pluripotent
- Prior art date
- Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
- Withdrawn
Links
- 210000004504 adult stem cell Anatomy 0.000 title claims abstract description 76
- 230000001172 regenerating effect Effects 0.000 title claims abstract description 29
- 230000000694 effects Effects 0.000 claims abstract description 20
- 102000002260 Alkaline Phosphatase Human genes 0.000 claims abstract description 11
- 108020004774 Alkaline Phosphatase Proteins 0.000 claims abstract description 11
- 108010017842 Telomerase Proteins 0.000 claims abstract description 11
- 210000002242 embryoid body Anatomy 0.000 claims abstract description 11
- 210000001654 germ layer Anatomy 0.000 claims abstract description 9
- 210000004027 cell Anatomy 0.000 claims description 236
- 238000000034 method Methods 0.000 claims description 57
- 210000000130 stem cell Anatomy 0.000 claims description 49
- 230000004069 differentiation Effects 0.000 claims description 45
- 210000002950 fibroblast Anatomy 0.000 claims description 43
- 239000000203 mixture Substances 0.000 claims description 40
- 210000001519 tissue Anatomy 0.000 claims description 39
- 108090000623 proteins and genes Proteins 0.000 claims description 36
- 238000012258 culturing Methods 0.000 claims description 22
- 239000003550 marker Substances 0.000 claims description 20
- 201000010099 disease Diseases 0.000 claims description 19
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 claims description 19
- 230000001537 neural effect Effects 0.000 claims description 18
- 230000014509 gene expression Effects 0.000 claims description 16
- 230000000392 somatic effect Effects 0.000 claims description 15
- 238000000338 in vitro Methods 0.000 claims description 13
- 238000007747 plating Methods 0.000 claims description 13
- 210000004966 intestinal stem cell Anatomy 0.000 claims description 12
- 238000011536 re-plating Methods 0.000 claims description 11
- 206010012601 diabetes mellitus Diseases 0.000 claims description 10
- 210000005260 human cell Anatomy 0.000 claims description 9
- 210000000981 epithelium Anatomy 0.000 claims description 8
- 210000004153 islets of langerhan Anatomy 0.000 claims description 8
- 208000023275 Autoimmune disease Diseases 0.000 claims description 7
- 208000019622 heart disease Diseases 0.000 claims description 7
- 230000002440 hepatic effect Effects 0.000 claims description 7
- 208000019423 liver disease Diseases 0.000 claims description 7
- 210000001185 bone marrow Anatomy 0.000 claims description 6
- 210000004556 brain Anatomy 0.000 claims description 6
- 229940126534 drug product Drugs 0.000 claims description 6
- 210000004185 liver Anatomy 0.000 claims description 6
- 230000002107 myocardial effect Effects 0.000 claims description 6
- 239000000825 pharmaceutical preparation Substances 0.000 claims description 6
- 239000000725 suspension Substances 0.000 claims description 6
- 230000002500 effect on skin Effects 0.000 claims description 5
- 210000003205 muscle Anatomy 0.000 claims description 5
- 210000000496 pancreas Anatomy 0.000 claims description 5
- 230000008569 process Effects 0.000 claims description 5
- 210000004700 fetal blood Anatomy 0.000 claims description 4
- 210000004209 hair Anatomy 0.000 claims description 4
- 210000000936 intestine Anatomy 0.000 claims description 4
- 210000005259 peripheral blood Anatomy 0.000 claims description 4
- 239000011886 peripheral blood Substances 0.000 claims description 4
- 210000001525 retina Anatomy 0.000 claims description 4
- 210000003491 skin Anatomy 0.000 claims description 4
- 238000002360 preparation method Methods 0.000 claims description 3
- 239000002609 medium Substances 0.000 description 19
- 239000001963 growth medium Substances 0.000 description 13
- 108091003079 Bovine Serum Albumin Proteins 0.000 description 10
- 210000004413 cardiac myocyte Anatomy 0.000 description 10
- 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 description 10
- 230000000968 intestinal effect Effects 0.000 description 10
- 108020004414 DNA Proteins 0.000 description 7
- 238000013459 approach Methods 0.000 description 7
- 238000002955 isolation Methods 0.000 description 7
- 210000001178 neural stem cell Anatomy 0.000 description 7
- 230000002103 transcriptional effect Effects 0.000 description 7
- 239000000872 buffer Substances 0.000 description 6
- 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 description 6
- DGVVWUTYPXICAM-UHFFFAOYSA-N β‐Mercaptoethanol Chemical compound OCCS DGVVWUTYPXICAM-UHFFFAOYSA-N 0.000 description 6
- APKFDSVGJQXUKY-KKGHZKTASA-N Amphotericin-B Natural products O[C@H]1[C@@H](N)[C@H](O)[C@@H](C)O[C@H]1O[C@H]1C=CC=CC=CC=CC=CC=CC=C[C@H](C)[C@@H](O)[C@@H](C)[C@H](C)OC(=O)C[C@H](O)C[C@H](O)CC[C@@H](O)[C@H](O)C[C@H](O)C[C@](O)(C[C@H](O)[C@H]2C(O)=O)O[C@H]2C1 APKFDSVGJQXUKY-KKGHZKTASA-N 0.000 description 5
- YQYJSBFKSSDGFO-UHFFFAOYSA-N Epihygromycin Natural products OC1C(O)C(C(=O)C)OC1OC(C(=C1)O)=CC=C1C=C(C)C(=O)NC1C(O)C(O)C2OCOC2C1O YQYJSBFKSSDGFO-UHFFFAOYSA-N 0.000 description 5
- 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 description 5
- 102000004877 Insulin Human genes 0.000 description 5
- 108090001061 Insulin Proteins 0.000 description 5
- 241000699666 Mus <mouse, genus> Species 0.000 description 5
- APKFDSVGJQXUKY-INPOYWNPSA-N amphotericin B Chemical compound O[C@H]1[C@@H](N)[C@H](O)[C@@H](C)O[C@H]1O[C@H]1/C=C/C=C/C=C/C=C/C=C/C=C/C=C/[C@H](C)[C@@H](O)[C@@H](C)[C@H](C)OC(=O)C[C@H](O)C[C@H](O)CC[C@@H](O)[C@H](O)C[C@H](O)C[C@](O)(C[C@H](O)[C@H]2C(O)=O)O[C@H]2C1 APKFDSVGJQXUKY-INPOYWNPSA-N 0.000 description 5
- 229960003942 amphotericin b Drugs 0.000 description 5
- 229940098773 bovine serum albumin Drugs 0.000 description 5
- LOKCTEFSRHRXRJ-UHFFFAOYSA-I dipotassium trisodium dihydrogen phosphate hydrogen phosphate dichloride Chemical compound P(=O)(O)(O)[O-].[K+].P(=O)(O)([O-])[O-].[Na+].[Na+].[Cl-].[K+].[Cl-].[Na+] LOKCTEFSRHRXRJ-UHFFFAOYSA-I 0.000 description 5
- 239000012091 fetal bovine serum Substances 0.000 description 5
- 239000008103 glucose Substances 0.000 description 5
- 229940125396 insulin Drugs 0.000 description 5
- 210000004347 intestinal mucosa Anatomy 0.000 description 5
- 210000003716 mesoderm Anatomy 0.000 description 5
- 210000000056 organ Anatomy 0.000 description 5
- 239000002953 phosphate buffered saline Substances 0.000 description 5
- 210000001778 pluripotent stem cell Anatomy 0.000 description 5
- 102000004169 proteins and genes Human genes 0.000 description 5
- 210000000813 small intestine Anatomy 0.000 description 5
- 101000976075 Homo sapiens Insulin Proteins 0.000 description 4
- 241000699670 Mus sp. Species 0.000 description 4
- 230000003115 biocidal effect Effects 0.000 description 4
- 238000004113 cell culture Methods 0.000 description 4
- 210000003981 ectoderm Anatomy 0.000 description 4
- 210000001671 embryonic stem cell Anatomy 0.000 description 4
- 210000002919 epithelial cell Anatomy 0.000 description 4
- 230000002068 genetic effect Effects 0.000 description 4
- ZDXPYRJPNDTMRX-UHFFFAOYSA-N glutamine Natural products OC(=O)C(N)CCC(N)=O ZDXPYRJPNDTMRX-UHFFFAOYSA-N 0.000 description 4
- 230000003394 haemopoietic effect Effects 0.000 description 4
- 238000003365 immunocytochemistry Methods 0.000 description 4
- PBGKTOXHQIOBKM-FHFVDXKLSA-N insulin (human) Chemical compound C([C@@H](C(=O)N[C@@H](CC(C)C)C(=O)N[C@H]1CSSC[C@H]2C(=O)N[C@H](C(=O)N[C@@H](CO)C(=O)N[C@H](C(=O)N[C@H](C(N[C@@H](CO)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CC=3C=CC(O)=CC=3)C(=O)N[C@@H](CCC(N)=O)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](CC(N)=O)C(=O)N[C@@H](CC=3C=CC(O)=CC=3)C(=O)N[C@@H](CSSC[C@H](NC(=O)[C@H](C(C)C)NC(=O)[C@H](CC(C)C)NC(=O)[C@H](CC=3C=CC(O)=CC=3)NC(=O)[C@H](CC(C)C)NC(=O)[C@H](C)NC(=O)[C@H](CCC(O)=O)NC(=O)[C@H](C(C)C)NC(=O)[C@H](CC(C)C)NC(=O)[C@H](CC=3NC=NC=3)NC(=O)[C@H](CO)NC(=O)CNC1=O)C(=O)NCC(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](CCCNC(N)=N)C(=O)NCC(=O)N[C@@H](CC=1C=CC=CC=1)C(=O)N[C@@H](CC=1C=CC=CC=1)C(=O)N[C@@H](CC=1C=CC(O)=CC=1)C(=O)N[C@@H]([C@@H](C)O)C(=O)N1[C@@H](CCC1)C(=O)N[C@@H](CCCCN)C(=O)N[C@@H]([C@@H](C)O)C(O)=O)C(=O)N[C@@H](CC(N)=O)C(O)=O)=O)CSSC[C@@H](C(N2)=O)NC(=O)[C@H](CCC(N)=O)NC(=O)[C@H](CCC(O)=O)NC(=O)[C@H](C(C)C)NC(=O)[C@@H](NC(=O)CN)[C@@H](C)CC)[C@@H](C)CC)[C@@H](C)O)NC(=O)[C@H](CCC(N)=O)NC(=O)[C@H](CC(N)=O)NC(=O)[C@@H](NC(=O)[C@@H](N)CC=1C=CC=CC=1)C(C)C)C1=CN=CN1 PBGKTOXHQIOBKM-FHFVDXKLSA-N 0.000 description 4
- 230000003914 insulin secretion Effects 0.000 description 4
- 210000002660 insulin-secreting cell Anatomy 0.000 description 4
- 210000002901 mesenchymal stem cell Anatomy 0.000 description 4
- 210000002569 neuron Anatomy 0.000 description 4
- 230000003248 secreting effect Effects 0.000 description 4
- 238000010186 staining Methods 0.000 description 4
- QTBSBXVTEAMEQO-UHFFFAOYSA-N Acetic acid Chemical compound CC(O)=O QTBSBXVTEAMEQO-UHFFFAOYSA-N 0.000 description 3
- 102000007469 Actins Human genes 0.000 description 3
- 108010085238 Actins Proteins 0.000 description 3
- 102000003974 Fibroblast growth factor 2 Human genes 0.000 description 3
- 108090000379 Fibroblast growth factor 2 Proteins 0.000 description 3
- 108700005087 Homeobox Genes Proteins 0.000 description 3
- 108090000581 Leukemia inhibitory factor Proteins 0.000 description 3
- 101100519293 Neurospora crassa (strain ATCC 24698 / 74-OR23-1A / CBS 708.71 / DSM 1257 / FGSC 987) pdx-1 gene Proteins 0.000 description 3
- 229930182555 Penicillin Natural products 0.000 description 3
- 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 description 3
- 102000004142 Trypsin Human genes 0.000 description 3
- 108090000631 Trypsin Proteins 0.000 description 3
- 208000035896 Twin-reversed arterial perfusion sequence Diseases 0.000 description 3
- 150000001413 amino acids Chemical class 0.000 description 3
- 238000004458 analytical method Methods 0.000 description 3
- 238000003556 assay Methods 0.000 description 3
- 230000001580 bacterial effect Effects 0.000 description 3
- 210000000227 basophil cell of anterior lobe of hypophysis Anatomy 0.000 description 3
- 210000002798 bone marrow cell Anatomy 0.000 description 3
- 230000001413 cellular effect Effects 0.000 description 3
- 238000004624 confocal microscopy Methods 0.000 description 3
- 230000037020 contractile activity Effects 0.000 description 3
- 210000001198 duodenum Anatomy 0.000 description 3
- 238000002474 experimental method Methods 0.000 description 3
- 239000012634 fragment Substances 0.000 description 3
- 210000001035 gastrointestinal tract Anatomy 0.000 description 3
- 210000004602 germ cell Anatomy 0.000 description 3
- 210000002490 intestinal epithelial cell Anatomy 0.000 description 3
- 230000001114 myogenic effect Effects 0.000 description 3
- 229940049954 penicillin Drugs 0.000 description 3
- 239000004033 plastic Substances 0.000 description 3
- 230000002062 proliferating effect Effects 0.000 description 3
- 230000035755 proliferation Effects 0.000 description 3
- 238000003127 radioimmunoassay Methods 0.000 description 3
- 238000003757 reverse transcription PCR Methods 0.000 description 3
- 230000002269 spontaneous effect Effects 0.000 description 3
- 229960005322 streptomycin Drugs 0.000 description 3
- JGVWCANSWKRBCS-UHFFFAOYSA-N tetramethylrhodamine thiocyanate Chemical compound [Cl-].C=12C=CC(N(C)C)=CC2=[O+]C2=CC(N(C)C)=CC=C2C=1C1=CC=C(SC#N)C=C1C(O)=O JGVWCANSWKRBCS-UHFFFAOYSA-N 0.000 description 3
- 238000002560 therapeutic procedure Methods 0.000 description 3
- 238000011282 treatment Methods 0.000 description 3
- 239000012588 trypsin Substances 0.000 description 3
- 210000002237 B-cell of pancreatic islet Anatomy 0.000 description 2
- OYPRJOBELJOOCE-UHFFFAOYSA-N Calcium Chemical compound [Ca] OYPRJOBELJOOCE-UHFFFAOYSA-N 0.000 description 2
- 241000701022 Cytomegalovirus Species 0.000 description 2
- 102100036912 Desmin Human genes 0.000 description 2
- 108010044052 Desmin Proteins 0.000 description 2
- 108010010803 Gelatin Proteins 0.000 description 2
- 102000053171 Glial Fibrillary Acidic Human genes 0.000 description 2
- 101710193519 Glial fibrillary acidic protein Proteins 0.000 description 2
- 241000282412 Homo Species 0.000 description 2
- 101500027325 Homo sapiens Atrial natriuretic peptide Proteins 0.000 description 2
- 108010025815 Kanamycin Kinase Proteins 0.000 description 2
- 102100033421 Keratin, type I cytoskeletal 18 Human genes 0.000 description 2
- 108010066327 Keratin-18 Proteins 0.000 description 2
- 102000004058 Leukemia inhibitory factor Human genes 0.000 description 2
- 241001529936 Murinae Species 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
- 206010028980 Neoplasm Diseases 0.000 description 2
- DFPAKSUCGFBDDF-UHFFFAOYSA-N Nicotinamide Chemical compound NC(=O)C1=CC=CN=C1 DFPAKSUCGFBDDF-UHFFFAOYSA-N 0.000 description 2
- 101710126211 POU domain, class 5, transcription factor 1 Proteins 0.000 description 2
- 108010010469 Qa-SNARE Proteins Proteins 0.000 description 2
- 102000044126 RNA-Binding Proteins Human genes 0.000 description 2
- 102000050389 Syntaxin Human genes 0.000 description 2
- 102100035071 Vimentin Human genes 0.000 description 2
- 108010065472 Vimentin Proteins 0.000 description 2
- 102000006646 aminoglycoside phosphotransferase Human genes 0.000 description 2
- 239000003242 anti bacterial agent Substances 0.000 description 2
- 239000000427 antigen Substances 0.000 description 2
- 102000036639 antigens Human genes 0.000 description 2
- 108091007433 antigens Proteins 0.000 description 2
- 230000008901 benefit Effects 0.000 description 2
- 230000015572 biosynthetic process Effects 0.000 description 2
- 210000002459 blastocyst Anatomy 0.000 description 2
- 210000000988 bone and bone Anatomy 0.000 description 2
- 210000004271 bone marrow stromal cell Anatomy 0.000 description 2
- 229910052791 calcium Inorganic materials 0.000 description 2
- 239000011575 calcium Substances 0.000 description 2
- 201000011510 cancer Diseases 0.000 description 2
- 210000005056 cell body Anatomy 0.000 description 2
- 210000003169 central nervous system Anatomy 0.000 description 2
- 238000012512 characterization method Methods 0.000 description 2
- 238000006243 chemical reaction Methods 0.000 description 2
- 239000002299 complementary DNA Substances 0.000 description 2
- 210000005045 desmin Anatomy 0.000 description 2
- 238000010790 dilution Methods 0.000 description 2
- 239000012895 dilution Substances 0.000 description 2
- 238000004520 electroporation Methods 0.000 description 2
- 210000001900 endoderm Anatomy 0.000 description 2
- 239000008273 gelatin Substances 0.000 description 2
- 229920000159 gelatin Polymers 0.000 description 2
- 235000019322 gelatine Nutrition 0.000 description 2
- 235000011852 gelatine desserts Nutrition 0.000 description 2
- 210000005046 glial fibrillary acidic protein Anatomy 0.000 description 2
- 230000005484 gravity Effects 0.000 description 2
- 230000012010 growth Effects 0.000 description 2
- 239000003102 growth factor Substances 0.000 description 2
- 238000001727 in vivo Methods 0.000 description 2
- 238000011534 incubation Methods 0.000 description 2
- 230000002147 killing effect Effects 0.000 description 2
- 210000001161 mammalian embryo Anatomy 0.000 description 2
- 238000010172 mouse model Methods 0.000 description 2
- 108010091047 neurofilament protein H Proteins 0.000 description 2
- 210000002027 skeletal muscle Anatomy 0.000 description 2
- 108091035539 telomere Proteins 0.000 description 2
- 102000055501 telomere Human genes 0.000 description 2
- 210000003411 telomere Anatomy 0.000 description 2
- 230000001225 therapeutic effect Effects 0.000 description 2
- 238000001890 transfection Methods 0.000 description 2
- 238000002054 transplantation Methods 0.000 description 2
- LAQPKDLYOBZWBT-NYLDSJSYSA-N (2s,4s,5r,6r)-5-acetamido-2-{[(2s,3r,4s,5s,6r)-2-{[(2r,3r,4r,5r)-5-acetamido-1,2-dihydroxy-6-oxo-4-{[(2s,3s,4r,5s,6s)-3,4,5-trihydroxy-6-methyloxan-2-yl]oxy}hexan-3-yl]oxy}-3,5-dihydroxy-6-(hydroxymethyl)oxan-4-yl]oxy}-4-hydroxy-6-[(1r,2r)-1,2,3-trihydrox Chemical compound O[C@H]1[C@H](O)[C@H](O)[C@H](C)O[C@H]1O[C@H]([C@@H](NC(C)=O)C=O)[C@@H]([C@H](O)CO)O[C@H]1[C@H](O)[C@@H](O[C@]2(O[C@H]([C@H](NC(C)=O)[C@@H](O)C2)[C@H](O)[C@H](O)CO)C(O)=O)[C@@H](O)[C@@H](CO)O1 LAQPKDLYOBZWBT-NYLDSJSYSA-N 0.000 description 1
- FWMNVWWHGCHHJJ-SKKKGAJSSA-N 4-amino-1-[(2r)-6-amino-2-[[(2r)-2-[[(2r)-2-[[(2r)-2-amino-3-phenylpropanoyl]amino]-3-phenylpropanoyl]amino]-4-methylpentanoyl]amino]hexanoyl]piperidine-4-carboxylic acid Chemical compound C([C@H](C(=O)N[C@H](CC(C)C)C(=O)N[C@H](CCCCN)C(=O)N1CCC(N)(CC1)C(O)=O)NC(=O)[C@H](N)CC=1C=CC=CC=1)C1=CC=CC=C1 FWMNVWWHGCHHJJ-SKKKGAJSSA-N 0.000 description 1
- 101710159080 Aconitate hydratase A Proteins 0.000 description 1
- 101710159078 Aconitate hydratase B Proteins 0.000 description 1
- 241000972773 Aulopiformes Species 0.000 description 1
- 102000015735 Beta-catenin Human genes 0.000 description 1
- 108060000903 Beta-catenin Proteins 0.000 description 1
- 206010006187 Breast cancer Diseases 0.000 description 1
- 208000026310 Breast neoplasm Diseases 0.000 description 1
- 102100022002 CD59 glycoprotein Human genes 0.000 description 1
- 108010083123 CDX2 Transcription Factor Proteins 0.000 description 1
- 102000006277 CDX2 Transcription Factor Human genes 0.000 description 1
- 208000005623 Carcinogenesis Diseases 0.000 description 1
- 238000000018 DNA microarray Methods 0.000 description 1
- 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 1
- 102100031573 Hematopoietic progenitor cell antigen CD34 Human genes 0.000 description 1
- 102100031672 Homeobox protein CDX-1 Human genes 0.000 description 1
- 101000897400 Homo sapiens CD59 glycoprotein Proteins 0.000 description 1
- 101000777663 Homo sapiens Hematopoietic progenitor cell antigen CD34 Proteins 0.000 description 1
- 101000777808 Homo sapiens Homeobox protein CDX-1 Proteins 0.000 description 1
- 101100519290 Homo sapiens PDX1 gene Proteins 0.000 description 1
- 101000976959 Homo sapiens Transcription factor 4 Proteins 0.000 description 1
- 101000596771 Homo sapiens Transcription factor 7-like 2 Proteins 0.000 description 1
- GRRNUXAQVGOGFE-UHFFFAOYSA-N Hygromycin-B Natural products OC1C(NC)CC(N)C(O)C1OC1C2OC3(C(C(O)C(O)C(C(N)CO)O3)O)OC2C(O)C(CO)O1 GRRNUXAQVGOGFE-UHFFFAOYSA-N 0.000 description 1
- 206010061598 Immunodeficiency Diseases 0.000 description 1
- 101710113773 Intestine-specific homeobox Proteins 0.000 description 1
- 102100040443 Keratin, type I cytoskeletal 15 Human genes 0.000 description 1
- 108010066330 Keratin-15 Proteins 0.000 description 1
- 102100032352 Leukemia inhibitory factor Human genes 0.000 description 1
- 241000699660 Mus musculus Species 0.000 description 1
- 101000591117 Mus musculus RNA-binding protein Musashi homolog 1 Proteins 0.000 description 1
- 102000005604 Myosin Heavy Chains Human genes 0.000 description 1
- 108010084498 Myosin Heavy Chains Proteins 0.000 description 1
- 239000004677 Nylon Substances 0.000 description 1
- 102000011755 Phosphoglycerate Kinase Human genes 0.000 description 1
- 208000002151 Pleural effusion Diseases 0.000 description 1
- 241000288906 Primates Species 0.000 description 1
- 101710183548 Pyridoxal 5'-phosphate synthase subunit PdxS Proteins 0.000 description 1
- 108700020471 RNA-Binding Proteins Proteins 0.000 description 1
- 101710105008 RNA-binding protein Proteins 0.000 description 1
- 239000012980 RPMI-1640 medium Substances 0.000 description 1
- 101100247004 Rattus norvegicus Qsox1 gene Proteins 0.000 description 1
- 229920005654 Sephadex Polymers 0.000 description 1
- 239000012507 Sephadex™ Substances 0.000 description 1
- FAPWRFPIFSIZLT-UHFFFAOYSA-M Sodium chloride Chemical compound [Na+].[Cl-] FAPWRFPIFSIZLT-UHFFFAOYSA-M 0.000 description 1
- 238000002105 Southern blotting Methods 0.000 description 1
- 102000015215 Stem Cell Factor Human genes 0.000 description 1
- 108010039445 Stem Cell Factor Proteins 0.000 description 1
- 206010043276 Teratoma Diseases 0.000 description 1
- 101001099217 Thermotoga maritima (strain ATCC 43589 / DSM 3109 / JCM 10099 / NBRC 100826 / MSB8) Triosephosphate isomerase Proteins 0.000 description 1
- 108091023040 Transcription factor Proteins 0.000 description 1
- 102000040945 Transcription factor Human genes 0.000 description 1
- 102100035101 Transcription factor 7-like 2 Human genes 0.000 description 1
- DFPAKSUCGFBDDF-ZQBYOMGUSA-N [14c]-nicotinamide Chemical compound N[14C](=O)C1=CC=CN=C1 DFPAKSUCGFBDDF-ZQBYOMGUSA-N 0.000 description 1
- 239000000654 additive Substances 0.000 description 1
- 239000002671 adjuvant Substances 0.000 description 1
- 230000001746 atrial effect Effects 0.000 description 1
- 239000011324 bead Substances 0.000 description 1
- 230000009286 beneficial effect Effects 0.000 description 1
- 230000027455 binding Effects 0.000 description 1
- 230000004071 biological effect Effects 0.000 description 1
- 210000004369 blood Anatomy 0.000 description 1
- 239000008280 blood Substances 0.000 description 1
- 210000000601 blood cell Anatomy 0.000 description 1
- 201000008275 breast carcinoma Diseases 0.000 description 1
- 230000003139 buffering effect Effects 0.000 description 1
- 230000036952 cancer formation Effects 0.000 description 1
- 231100000504 carcinogenesis Toxicity 0.000 description 1
- 230000011712 cell development Effects 0.000 description 1
- 230000024245 cell differentiation Effects 0.000 description 1
- 230000032823 cell division Effects 0.000 description 1
- 230000003915 cell function Effects 0.000 description 1
- 239000013592 cell lysate Substances 0.000 description 1
- 239000002771 cell marker Substances 0.000 description 1
- 238000002659 cell therapy Methods 0.000 description 1
- 239000003153 chemical reaction reagent Substances 0.000 description 1
- 239000003795 chemical substances by application Substances 0.000 description 1
- 230000002648 chondrogenic effect Effects 0.000 description 1
- 238000003501 co-culture Methods 0.000 description 1
- 210000001072 colon Anatomy 0.000 description 1
- 230000002596 correlated effect Effects 0.000 description 1
- 238000005138 cryopreservation Methods 0.000 description 1
- 210000000805 cytoplasm Anatomy 0.000 description 1
- 230000003247 decreasing effect Effects 0.000 description 1
- 238000009795 derivation Methods 0.000 description 1
- 238000011161 development Methods 0.000 description 1
- 230000018109 developmental process Effects 0.000 description 1
- VHJLVAABSRFDPM-QWWZWVQMSA-N dithiothreitol Chemical compound SC[C@@H](O)[C@H](O)CS VHJLVAABSRFDPM-QWWZWVQMSA-N 0.000 description 1
- 238000001962 electrophoresis Methods 0.000 description 1
- 210000002257 embryonic structure Anatomy 0.000 description 1
- 238000005516 engineering process Methods 0.000 description 1
- 210000001842 enterocyte Anatomy 0.000 description 1
- 210000003158 enteroendocrine cell Anatomy 0.000 description 1
- 210000005175 epidermal keratinocyte Anatomy 0.000 description 1
- 210000002514 epidermal stem cell Anatomy 0.000 description 1
- 230000009786 epithelial differentiation Effects 0.000 description 1
- DEFVIWRASFVYLL-UHFFFAOYSA-N ethylene glycol bis(2-aminoethyl)tetraacetic acid Chemical compound OC(=O)CN(CC(O)=O)CCOCCOCCN(CC(O)=O)CC(O)=O DEFVIWRASFVYLL-UHFFFAOYSA-N 0.000 description 1
- 230000001747 exhibiting effect Effects 0.000 description 1
- 239000012530 fluid Substances 0.000 description 1
- 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 1
- 230000006870 function Effects 0.000 description 1
- 239000000499 gel Substances 0.000 description 1
- 238000003209 gene knockout Methods 0.000 description 1
- 238000001415 gene therapy Methods 0.000 description 1
- 230000000762 glandular Effects 0.000 description 1
- 239000011521 glass Substances 0.000 description 1
- 210000003780 hair follicle Anatomy 0.000 description 1
- 210000003958 hematopoietic stem cell Anatomy 0.000 description 1
- 210000003494 hepatocyte Anatomy 0.000 description 1
- 230000001744 histochemical effect Effects 0.000 description 1
- 210000004276 hyalin Anatomy 0.000 description 1
- 210000004408 hybridoma Anatomy 0.000 description 1
- GRRNUXAQVGOGFE-NZSRVPFOSA-N hygromycin B Chemical compound O[C@@H]1[C@@H](NC)C[C@@H](N)[C@H](O)[C@H]1O[C@H]1[C@H]2O[C@@]3([C@@H]([C@@H](O)[C@@H](O)[C@@H](C(N)CO)O3)O)O[C@H]2[C@@H](O)[C@@H](CO)O1 GRRNUXAQVGOGFE-NZSRVPFOSA-N 0.000 description 1
- 229940097277 hygromycin b Drugs 0.000 description 1
- 230000001900 immune effect Effects 0.000 description 1
- 238000013394 immunophenotyping Methods 0.000 description 1
- 238000002513 implantation Methods 0.000 description 1
- 230000002401 inhibitory effect Effects 0.000 description 1
- 230000005732 intercellular adhesion Effects 0.000 description 1
- 210000002510 keratinocyte Anatomy 0.000 description 1
- 208000032839 leukemia Diseases 0.000 description 1
- 210000005229 liver cell Anatomy 0.000 description 1
- 230000007774 longterm Effects 0.000 description 1
- 238000012423 maintenance Methods 0.000 description 1
- 210000004962 mammalian cell Anatomy 0.000 description 1
- 230000035800 maturation Effects 0.000 description 1
- 238000005259 measurement Methods 0.000 description 1
- 230000001404 mediated effect Effects 0.000 description 1
- 229960004857 mitomycin Drugs 0.000 description 1
- 238000012986 modification Methods 0.000 description 1
- 230000004048 modification Effects 0.000 description 1
- 230000000877 morphologic effect Effects 0.000 description 1
- 210000004877 mucosa Anatomy 0.000 description 1
- 230000009756 muscle regeneration Effects 0.000 description 1
- 231100000219 mutagenic Toxicity 0.000 description 1
- 230000003505 mutagenic effect Effects 0.000 description 1
- 210000000107 myocyte Anatomy 0.000 description 1
- 239000013642 negative control Substances 0.000 description 1
- 230000001613 neoplastic effect Effects 0.000 description 1
- 210000004498 neuroglial cell Anatomy 0.000 description 1
- 229960003966 nicotinamide Drugs 0.000 description 1
- 235000005152 nicotinamide Nutrition 0.000 description 1
- 239000011570 nicotinamide Substances 0.000 description 1
- 229920001778 nylon Polymers 0.000 description 1
- 230000002188 osteogenic effect Effects 0.000 description 1
- 230000002018 overexpression Effects 0.000 description 1
- 101710135378 pH 6 antigen Proteins 0.000 description 1
- 238000000059 patterning Methods 0.000 description 1
- 239000008188 pellet Substances 0.000 description 1
- 230000010412 perfusion Effects 0.000 description 1
- 150000008300 phosphoramidites Chemical class 0.000 description 1
- 229920000136 polysorbate Polymers 0.000 description 1
- 239000011546 protein dye Substances 0.000 description 1
- 239000011541 reaction mixture Substances 0.000 description 1
- 230000008929 regeneration Effects 0.000 description 1
- 238000011069 regeneration method Methods 0.000 description 1
- 230000008439 repair process Effects 0.000 description 1
- 230000004044 response Effects 0.000 description 1
- 230000008458 response to injury Effects 0.000 description 1
- 235000019515 salmon Nutrition 0.000 description 1
- 238000012216 screening Methods 0.000 description 1
- 230000000580 secretagogue effect Effects 0.000 description 1
- 230000028327 secretion Effects 0.000 description 1
- 230000011664 signaling Effects 0.000 description 1
- 210000004927 skin cell Anatomy 0.000 description 1
- 239000000243 solution Substances 0.000 description 1
- 210000001988 somatic stem cell Anatomy 0.000 description 1
- 230000003068 static effect Effects 0.000 description 1
- 230000024642 stem cell division Effects 0.000 description 1
- 238000002660 stem cell treatment Methods 0.000 description 1
- 238000013337 sub-cultivation Methods 0.000 description 1
- 239000000758 substrate Substances 0.000 description 1
- 230000000153 supplemental effect Effects 0.000 description 1
- 230000004083 survival effect Effects 0.000 description 1
- 238000004114 suspension culture Methods 0.000 description 1
- 210000004876 tela submucosa Anatomy 0.000 description 1
- 238000012360 testing method Methods 0.000 description 1
- 210000001550 testis 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
- 238000012546 transfer Methods 0.000 description 1
- 238000011830 transgenic mouse model Methods 0.000 description 1
- 230000007306 turnover Effects 0.000 description 1
- 230000002792 vascular Effects 0.000 description 1
- 230000002861 ventricular Effects 0.000 description 1
- 210000005048 vimentin Anatomy 0.000 description 1
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
- C12N5/0679—Cells of the gastro-intestinal tract
- C12N5/068—Stem cells; Progenitors
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P37/00—Drugs for immunological or allergic disorders
-
- 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
- C12N5/0607—Non-embryonic pluripotent stem cells, e.g. MASC
-
- 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
- C12N5/0676—Pancreatic cells
-
- G—PHYSICS
- G01—MEASURING; TESTING
- G01N—INVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
- G01N33/00—Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
- G01N33/48—Biological material, e.g. blood, urine; Haemocytometers
- G01N33/50—Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
- G01N33/5005—Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
-
- 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
- A61K35/12—Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified 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
- C12N2501/00—Active agents used in cell culture processes, e.g. differentation
- C12N2501/10—Growth factors
- C12N2501/115—Basic fibroblast growth factor (bFGF, FGF-2)
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2502/00—Coculture with; Conditioned medium produced by
- C12N2502/13—Coculture with; Conditioned medium produced by connective tissue cells; generic mesenchyme cells, e.g. so-called "embryonic fibroblasts"
- C12N2502/1323—Adult fibroblasts
-
- 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
- C12N2506/03—Differentiation of animal cells from one lineage to another; Differentiation of pluripotent cells from non-embryonic pluripotent 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
- C12N2506/00—Differentiation of animal cells from one lineage to another; Differentiation of pluripotent cells
- C12N2506/23—Differentiation of animal cells from one lineage to another; Differentiation of pluripotent cells from cells of the gastro-intestinal tract
-
- 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
- C12N2510/00—Genetically modified cells
Definitions
- Pluripotent adult stem cells derived from regenerative tissue Pluripotent adult stem cells derived from regenerative tissue
- the present invention relates generally to isolated, enriched, mammalian, preferably human, adult stem cell and progenitor cell populations derived from regenerative tissue, having pluripotent stem cell-like characteristics, their differentiation into any cell type, and therapeutic use thereof.
- Regenerative stem cell populations constitute only a small percentage of the total number of cells, but are of immense interest because of their ability to repopulate the body. There is significant commercial interest in these methods because stem cells have a number of clinical uses. Using stem cells for generation of organs and tissues for transplantation provides a promising alternative therapy for diabetes, neuronal disease, liver disease, heart disease and autoimmune disorders, to name a few. Diabetes, for instance is an excellent candidate for stem cell treatment (Soria et al. (2001) Diabetalogia 44: 407-415; Soria et al. (2000) Eur. J. Physiol. 440: 1-18; and Berna et al. (2001) Biomed. Pharmacother. 55: 206-212).
- Regenerative stem cells can be found in many adult tissues, participating in normal replacement due to physiological turnover or in response to injury.
- a mouse model of intestinal stem cells has been developed (Roth et al. (1991) Cell Biol. 88: 9407-9411; and Slorach et al. (1999) J. Cell Sci. 11 : 3029-3038).
- neural stem cells from the adult mouse brain produced hematopoietic progeny and incorporated into an embryo, contributing to all the tissues of the resulting chimeric mouse (Bjomson et al. (1999) Science 283: 534; and Clarke et al. (2000) Science 288: 1666).
- clonally derived neural stem cells from mice and humans can produce skeletal myotubes in vitro and in vivo (Galli et al. (2000) Nat. Neurosci. 3: 986).
- bone marrow cells can differentiate into myogenic progenitors, myocytes, vascular structures, neurons, or liver cells (Ferrari et al. (1998) Science 279: 1528; Orlic et al. (2001) Proc. Natl. Acad. Sci. USA 98: 10344; Woodbury et al. (2000) Neurosci. Res. 61 : 364; and Alison et al. (2000) Nature 404: 257).
- stem cells isolated from skeletal muscle have shown hematopoietic potential (Jackson et al. (1999) Proc. Natl. Acad. Sci. USA 96: 14482).
- An object of the present invention is to provide isolated pluripotent adult stem cell and progenitor cell populations, derived from regenerative tissue, which can differentiate into any cell type, and methods for isolating and enriching pluripotent adult stem cell and progenitor cell populations.
- the object is achieved by means of providing an isolated pluripotent adult stem cell or stem cell population and a composition comprising said pluripotent adult stem cell or stem cell population, derived from regenerative tissue, and having alkaline phosphatase activity, high levels of telomerase activity and the ability to form derivatives of all three embryonic germ layers and/or the ability to form an embryoid body.
- the object is further achieved by a method for isolating and enriching the pluripotent adult stem cell population, comprising the steps of: a) isolating a population derived from regenerative tissue containing somatic adult stem cells; b) plating and culturing the cells of a) on mitotically inactivated fibroblast feeder layer; c) selecting and replating colonies which have a pluripotent morphology; wherein the cells of the selected colonies are preferably expanded over 2-100 passages, very particularly over 2-15 passages.
- Another object of the present invention is to provide a differentiated cell population and methods for selecting and differentiating the pluripotent adult stem cell population.
- the object is achieved by a method for selection and differentiation of a pluripotent adult stem cell population to a desired differentiated cell population, comprising the steps of: a) isolating a population derived from regenerative tissue containing somatic adult stem cells; b) plating and culturing the cells of a) on mitotically inactivated fibroblast feeder layer; c) selecting and replating colonies which have a pluripotent morphology; d) causing differentiation by deleting the fibroblast feeder layer; and e) selecting the cells of said desired cell lineage.
- the object is further achieved by a method for selection and differentiation of a pluripotent adult stem cell population to a desired differentiated cell population, comprising the steps of: a) isolating a population derived from regenerative tissue containing somatic adult stem cells; b) plating and culturing the cells of a) on mitotically inactivated fibroblast feeder layer; c) selecting and replating colonies which have a pluripotent morphology; d) transfecting cells in selected colonies to introduce a marker gene that enables the selection of one desired cell lineage from the other cell lineages that result from differentiation of the stem cells; e) causing differentiation by deleting the fibroblast feeder layer and culturing at high density in suspension; and f) selecting the cells of said desired cell lineage based on the marker gene.
- the object is further achieved by a method for selection and differentiation of a pluripotent adult stem cell population to a desired differentiated cell population, comprising the steps of: a) isolating a population derived from regenerative tissue containing somatic adult stem cells; b) plating and culturing the cells of a) on mitotically inactivated fibroblast feeder layer; c) selecting and replating colonies which have a pluripotent morphology; d) transfecting cells in selected colonies to introduce a lineage- specific gene for the purpose of driving differentiation toward a chosen cell lineage, and a marker gene for the purpose of selection; e) causing differentiation by deleting the fibroblast feeder layer and culturing at high density in suspension; and f) selecting the enriched chosen cell lineage based on the marker gene.
- Another objects of the invention are to provide a method of treating a disease, comprising administering said pluripotent adult stem cell population or said differentiated cell population to a patient in need thereof, and to provide a therapeutic and/or diagnostic composition comprising said pluripotent or said differentiated cell population.
- the present invention relates to the isolation of pluripotent adult stem cell and progenitor cell populations, derived from regenerative tissue, which can differentiate into any cell type, and methods for the isolation and enrichment of pluripotent adult stem cell and progenitor cell populations.
- the isolated pluripotent adult stem cell or stem cell population is derived from regenerative tissue, and has alkaline phosphatase activity, high levels of telomerase activity and the ability to form derivatives of all three embryonic germ layers.
- the isolated pluripotent adult stem cell population also has the ability to form an embryoid body.
- the method for isolating and enriching the pluripotent adult stem cell population comprises the steps of: a) isolating a population derived from regenerative tissue containing somatic adult stem cells, preferably human; b) plating and culturing the cells of a) on mitotically inactivated fibroblast feeder layer, preferably human; c) selecting and replating colonies which have a pluripotent morphology; wherein the cells of the selected colonies are preferably expanded over 2-100 passages, more preferably over 2-15 passages and most preferably over 10-15 passages. Specific methods for pursuing each step are provided for instance, in the Examples herein.
- the undifferentiated cells are mammalian cells, and even more preferably, human cells, which have the potential to differentiate into any cell lineage when subjected to differentiating conditions.
- the undifferentiated cells preferably can proliferate and propagate in vitro, and can maintain an undifferentiated state when cultured up to 15 passages on a fibroblast feeder layer.
- the undifferentiated cells preferably have a total expansion capacity of more than 1x10 10 , more preferably of more than 1x10 11 .
- the undifferentiated cells express SSEA-3, and preferably can express any other characteristic stem cell markers known in the art.
- the regenerative tissue can be selected from the group consisting of peripheral blood, cord blood, bone marrow, brain, skin, retina, hair papilla, muscle, liver, pancreas, epithelium.
- the regenerative tissue are epithelia, preferably mammalian intestinal epithelia, most preferably human intestinal epithelia derived from the small intestine, in particular, derived from crypts.
- HISC adult human intestinal stem cells
- Epithelia are usually composed of several distinct cell types, and the ability to form all of them is an aspect of stem cell behavior.
- the mammalian intestinal epithelium is a rapidly proliferating tissue, with stem cells residing at the crypt base and giving rise to all cell types found within the crypt (Bach et al. (2000) Carcinogenesis 21 : 469).
- stem cell is meant, in accordance with the present invention, a cell that can undergo self-renewing cell division, for an indefinite time, to give rise to phenotypically and genotypically identical daughter cells, and can ultimately differentiate into at least one final cell type.
- the quintessential stem cell is the embryonic stem (ES) cell, as it has unlimited self- renewal and multipotent and/or pluripotent differentiation potential, thus possessing the capability of developing into any organ, tissue, or cell type.
- ES embryonic stem
- EG embryonic germ
- ES and EG cells have been derived from mice and, more recently, from non-human primates and humans (Evans et al. (1981) Nature 292: 154-6; Matsui et al. (1991) Nature 353: 750-2; Thomson et al. (1995) Proc. Natl. Acad. Sci. USA 92: 7844-8; Thomson et al. (1998) Science 282: 1145-7; and Shamblott et al. (1998) Proc. Natl. Acad. Sci. USA 95: 13726-31).
- Stem cells have been identified in most organs and tissues. The best-characterized is the hematopoietic stem cell, which can give rise to any of the different types of terminally differentiated blood cells. This is a mesoderm-derived cell, purified based on cell surface markers and functional characteristics (Hill et al. (1996) Exp. Hematol. 24: 936-43). Also well-characterized are the neural stem cell and a number of mesenchymal stem cells derived from multiple sources (Flax et al. (1998) Nature Biotechnol. 16: 1033-1039; Clarke et al. (2000) Science 288: 1666; Bruder et al. (1997) J. Cell. Biochem.
- adult stem cells and progenitor cell populations are to be understood, in accordance with the present invention, as meaning cells that can be derived from any source of adult tissue or organ, can replicate as undifferentiated cells, and have the potential to differentiate into at least one, preferably multiple, cell lineages.
- “In vitro” means, for the purposes of the present invention, a conversion, i.e. a reaction, which proceeds outside a live organism.
- Regenerative tissue is a mammalian, preferably human, fluid, tissue or organ, which is preferably selected from the group consisting of peripheral blood, cord blood, bone marrow, brain, skin, retina, hair papilla, muscle, liver, pancreas and intestine.
- human intestinal epithelia derived from the small intestine are most preferred. Such intestinal stem cells reside just above the crypt base in the small intestine and/or at the crypt base in the colon.
- HISC can overcome their ectodermal commitment and show a very broad plasticity that is not restricted to ectodermal derivatives.
- the preferred HISC in spite of being adult stem cells, have an enormous capacity to proliferate. These cells preferably can divide at least 50-80 times, more preferably 60- 70 times, without losing their ability to proliferate and differentiate. After about 15 passages, the cells may become senescent and telomerase activity may decrease. Changes in the environment could have an influence on loss of potency; it has been calculated that during a human life-time, approximately 5,000 intestinal stem-cell divisions occur (Booth and Potten (2000) J. Clin. Invest. 105: 1493). Furthermore, the invention relates to a pluripotent adult stem cell or pluripotent adult stem cell population, wherein the cell can be induced to differentiate to form at least one differentiated cell type of mesodermal, ectodermal and endodermal origin.
- the pluripotent adult stem cell or stem cell population can be used to generate differentiated cell lineages.
- the differentiated cell types can be identified or selected by any method known in the art.
- lineage specific markers can be used for sorting methods such as fluorescent activated cell sorting (FACS) or other surface antigen based enrichment.
- FACS fluorescent activated cell sorting
- Other examples include DNA chip technology, which can be used to detect lineage-specific genes, or reverse transcription polymerase chain reaction (RT-PCR), which can be used to analyze lineage-specific gene expression.
- RT-PCR reverse transcription polymerase chain reaction
- the object is preferably achieved by a method for selection and differentiation of a pluripotent adult stem cell population to a desired differentiated cell population, comprising the steps of: a) isolating a population derived from regenerative tissue containing somatic adult stem cells; b) plating and culturing the cells of a) on mitotically inactivated fibroblast feeder layer; c) selecting and replating colonies which have a pluripotent morphology; d) causing differentiation by deleting the fibroblast feeder layer; and e) selecting the cells of said desired cell lineage.
- such a differentiated cell lineage can be obtained by selecting and differentiating a pluripotent adult stem cell population to a desired differentiated cell population, by: a) isolating a population derived from regenerative tissue containing somatic adult stem cells, preferably human; b) plating and culturing the cells of a) on mitotically inactivated fibroblast feeder layer, preferably human; c) selecting and replating colonies which have a pluripotent morphology; d) transfecting cells in selected colonies to introduce a marker gene that enables the selection of one desired cell lineage from the other cell lineages that result from differentiation of the stem cells; e) causing differentiation by deleting the fibroblast feeder layer and culturing at high density in suspension; and f) selecting the cells of said desired cell lineage based on the marker gene.
- the desired cell lineage for example can be neuronal, myocardial, hepatic, pancreatic, pancreatic islet, renal, or dermal.
- the marker gene is under the control of a promoter that causes expression of the marker gene to enable selection of one desired cell lineage from other cell lineages. This is achieved by using a promoter which is activated only in the desired cell lineage. Furthermore, the marker gene can confer antibiotic resistance, wherein selection includes contacting the mixed differentiated cellular population with an antibiotic to which cells of the desired cell lineage have been conferred resistance by the expression of the first marker gene, thereby killing cells of other cell lineages, but not cells of the desired one cell lineage that express the marker gene.
- WO 95/14079 The cell preparation and process for the differentiation and selection thereof is disclosed in WO 95/14079, which is encompassed in the disclosure of the present invention. Therefore a further aspect of the invention relates to a method for the selection and differentiation of pluripotent adult stem cells to a desired differentiated cell lineage comprising the steps a) - f), by using the indicated promoter (as in Table 1) for obtaining the desired differentiated cell lineage.
- a differentiated cell lineage can be obtained by selecting and differentiating a pluripotent adult stem cell population to a desired differentiated cell population by performing steps a) - c) as above, and then, as step (d), transfecting cells in selected colonies to introduce a lineage-specific gene for the purpose of driving differentiation toward a chosen cell lineage, and a marker gene for the purpose of selection.
- the cells or cell population can then be differentiated and selected according to steps e) and f).
- the marker gene can confer antibiotic resistance, wherein selection includes contacting the mixed differentiated cellular population with an antibiotic to which cells of the desired cell lineage have been conferred resistance by the expression of said lineage-specific gene, thereby killing cells of the other cell lineages, but not cells of the desired, one cell lineage that express the lineage-specific gene.
- the following constructs can be used to drive undifferentiated stem cells toward a pancreatic, insulin-secreting fate: CMV-pdx, L32-pdx, ⁇ -actin-pdx, CMV-cdx, hlns-pGK-hygro, where CMV (cytomegalovirus), L32 and ⁇ -actin are constitutive promoters, and pdx (pancreas/duodenum homeobox) and hlns (human insulin) are pancreatic cell-specific genes. Suitable genes for these constructs are described for instance in the literature and are known to those of skill in the art. See e.g., Sakakibara et al. (1996) Dev. Biol.
- Pluripotent adult stem cells can also be differentiated by any method known in the art. For instance, co-culture with differentiated cells, gene knockout, and the addition of growth factors or other modification of culture conditions can be used in the differentiation of stem cells.
- Another subject matter of the invention is a drug product which comprises pluripotent adult stem cells as mentioned before and/or differentiated cells as mentioned before wherein the differentiated cells are selected from the group consisting of neuronal, myocardial, hepatic, pancreatic (including islet cells), renal and/or dermal cells.
- the drug product preferably further comprises suitable additives, adjuvants and/or a pharmaceutically acceptable vehicle.
- the drug product can be used for treatment of diseases such as diabetes, neuronal diseases, liver diseases, heart diseases and/or autoimmune disorders.
- Subject matter of the invention is further a process for the preparation of such a drug product.
- Another subject matter of the present invention is a method of treating a disease, wherein pluripotent cells as mentioned before or differentiated cells as mentioned before or a composition containing these cells are administered to a patient in need thereof.
- the differentiated cell or cell population is here preferably a neuronal, myocardial, hepatic, pancreatic (including islet cells), renal, or dermal differentiated cell or cell population.
- the disease can be diabetes, neuronal disease, liver disease, heart disease or autoimmune disorders.
- the therapy can be human cell or gene therapy, preferably in transplantation therapy.
- the invention additionally relates to providing a diagnostic composition comprising the pluripotent cell or cell population or the differentiated cell or cell population or a composition comprising at least one of these cells or cell populations.
- the composition may contain a pharmaceutically acceptable vehicle.
- vehicle is meant a carrier for the administration of differentiated cells, preferably sterile isotonic saline.
- the composition may also contain reagents for determining the physiological or morphological response of the differentiated cells or cell line to mutagenic, toxic or beneficial agents or treatments.
- A Phase contrast image of human intestinal epithelial cells isolated from fresh small intestine specimens obtained from cadaveric donors.
- B Phase contrast image of an HISC-2 colony showing pluripotent-like morphology after 23 days of culture.
- C HISC-2 colony (indicated by arrow) showing histochemical staining for alkaline phosphatase. The fibroblast feeder layer (indicated by triangle) was always negative for alkaline phosphatase staining.
- Figure 2 shows some adult human stem cells stained with an antibody recognizing the SSEA-4 epitope (A) and a phase contrast view of the same field of cells (B).
- Figure 3 shows alkaline phosphatase staining of a human intestinal stem cell EB (A) and a negative control (B).
- Adult human stem cells after 6 days formed EBs and were subsequently differentiated by plating at high density, in the absence of fibroblasts, on gelatin-treated tissue culture plates. After 1-3 weeks of culture in these conditions a variety of differentiated cells appeared.
- the structures identified were (A) hyaline-like structures (indicated by arrow), (B) neural ganglionic-like structures with elongated processes that extended out from their cell bodies forming networks (indicated by arrow), and (C) secretory glandular-like structures.
- HISC-2 cells strongly reacted in immunocytochemistry assays with monoclonal antibodies (mAb) against ectoderm proteins, such as neurofilament-200 (A), syntaxin (B), and glial fibrillary acidic protein (C), and against mesoderm proteins, such as vimentin (C), actin (D), and desmin (E).
- mAb monoclonal antibodies
- ectoderm proteins such as neurofilament-200 (A), syntaxin (B), and glial fibrillary acidic protein (C)
- mesoderm proteins such as vimentin (C), actin (D), and desmin (E).
- F -fetoprotein
- Figure 6 shows G418 selection of human intestinal stem cells using a human atrial natriuretic factor (hANF) promoter (A) and a human insulin (hlns) promoter (B).
- hANF human atrial natriuretic factor
- hlns human insulin
- the mucosa was dissected from the submucosa, cut into strips that were rinsed again in 100 ml of sterilized PBS, supplemented with 250 ng/ml amphotericin B (Sigma), at 37 9 C, and then incubated in 20 ml of PBS, supplemented with 250 ng/ml amphotericin B and containing 10 mM dithiothreitol (Sigma), for 30 minutes at room temperature.
- the strips were rinsed again in 100 ml of sterilized PBS supplemented with 250 ng/ml amphotericin B, at 37 e C and then incubated for 45 minutes in 1 mM EDTA (Sigma) plus 0.25% trypsin (Gibco-BRL) in Dulbecco's modified essential medium (DMEM) (Gibco-BRL) at 37 9 C.
- DMEM Dulbecco's modified essential medium
- a line of human fibroblasts was isolated from the piece of duodenum of a donor. For isolating this cell line, 10 7 cells obtained from the previous protocol, were plated onto 100-mm tissue culture dishes (25020-100, Corning) and cultured in DMEM supplemented with 10% fetal bovine serum (Gibco-BRL), 2 mM glutamine (Gibco-BRL), 100 lU/ml penicillin (Gibco-BRL), 0.1 mg/ml streptomycin (Gibco-BRL) and 250 ng/ml of amphotericin B (Sigma). Every 48 hours, the culture medium was changed. After ten days of culture, fibroblast growth was evident.
- human fibroblasts were selected, transferred to new dishes and expanded by culturing in the fibroblast culture medium.
- 5 X 10 6 fibroblasts were plated onto 100-mm tissue culture dishes, cultured in the same fibroblast culture medium, and grown to confluency (3-5 days). The medium was changed every 24 hours. When confluent, the fibroblasts were trypsinized and split.
- isolated intestinal epithelial cells died rapidly, but after 3 days of culture, 27% of the cells remained as monolayers. To assess the purity of these cells, they were stained for the epithelial cell marker, cytokeratin-18, with specific primary mAb (Sigma) and localized with fluorescent (TRITC) secondary antibody (Sigma) by confocal microscopy. The percentage of cells positive for cytokeratin-18 was determined by immunocytochemistry to be 95%.
- epithelial cells continued proliferating. After this time, small colonies with a morphology resembling pluripotent stem cells were occasionally observed growing from the intestinal epithelial cell cultures. These cells were selected, transferred to new dishes and expanded by culturing in the same culture medium. For each passage, 2 x 10 6 cells were cultured in tissue culture flasks (Falcon 75 cm 2 , 353084, Becton Dickinson), on new inactivated human fibroblast feeder layer (75.000 cells/cm 2 ), in the same culture medium, and grown to confluence (5-7 days). The medium was changed every 48 hours. When confluent, the cells were trypsinized and split. Using this methodology, four clones were established (clones: HISC 1 to 4; Table 3). Although the four clones showed similar characteristics, only data from clone HISC-2 are shown.
- Alkaline phosphatase was detected with Vector Blue substrate (Vector Labs) and SSEA-3 was detected by immunocytochemistry with specific primary mAb (Hybridoma Bank) and localized with fluorescent (TRITC) secondary antibody (Sigma) by confocal microscopy.
- SSEA-1 , SSEA-2, GCTM-2, TRA-1-60 and TRA-1-81 can also be used to characterize pluripotent stem cells. Shamblott et al. (1998) Proc. Natl. Acad. Sci. USA 95:13726-31.
- telomerase activity was measured with the TRAP assay.
- Cells were lysed as previously described.
- TRAP analysis the reaction mixture was performed in 0.2 ml thin-walled tubes, in a 10 ⁇ l volume containing 50 ⁇ M dNTPs, TRAP buffer (final concentrations of 20 mM Tris-HCI, pH 8.3, 1.5 mM MgCls, 68 mM KCl, 0.05% Tween ® , 20.1 mM EGTA), 100 ng TS primer (AATCCGTCGAGCAGAGTT) (SEQ ID NO: 1), 100 ng CX primer (CCCTTACCCTTACCCTTACCCTAA) (SEQ ID NO: 2) labeled with HEX phosphoramidites (Perkin-Elmer Applied Biosystems), 2.5 U AmpliTaq Golg (Perkin
- the tubes were incubated at room temperature for 20 min, then heated to 95 C for 10 min, followed by 35 cycles at 94 Q C for 30 sec, 50 9 C for 30 sec and 72 Q C for 45 sec, with a final extension step at 72 9 C for 1 min in a Techne Thermal Cycler.
- an electrophoresis was carried out in an ABI PRISM 377 DNA sequencer (Perkin-Elmer). The samples were run in the GS 36C- 2400 module for 3 h. DNA fragment analysis was performed using GeneScan 2.1 software (Applied Biosystems). Other methods for characterization of isolated HISC can also be employed. Telomere length can be determined by Southern blotting for telomere restriction fragment.
- Quantitative RT-PCR can be used to confirm the origin and character of isolated stem cells by detecting expression of embryonic transcription factors such as TCF4, Sox2 and Oct-4.
- embryonic transcription factors such as TCF4, Sox2 and Oct-4.
- teratoma studies can be performed to demonstrate the pluripotency of cells. This data can be used to distinguish HISC from intestinal epithelial non-stem cells
- HISC-2 cells When allowed to differentiate in vitro by culturing at high density, in plastic Petri dishes, in the absence of fibroblasts and rhLIF and in the same medium as that used for initial culture, HISC-2 cells (passages 2-12) formed EBs ( Figure 1 D). Subsequent in vitro differentiation, by plating trypsinized EBs at high density, in the absence of fibroblasts, on gelatin-treated tissue culture plates, in the last mentioned medium, for three to six days, revealed a variety of differentiated cells. Thus, in vitro differentiation occurred when HISC-2 cells were cultured in the absence of embryonic fibroblasts and at a high density, both in the presence and absence of rhLIF.
- Hyaline structures (Figure 4A), neural ganglionic-like structures with elongated processes that extended out from cell bodies forming networks (Figure 4B), secretory glandular structures (Figure 4C) and contracting muscle, pacing at frequencies of 30-70 pulses/min., were observed.
- the approach relies on the use of two transcriptional units, introduced on a common vector backbone into undifferentiated intestinally derived pluripotent cells.
- the first transcriptional unit comprises a promoter expressed in undifferentiated stem cells, linked to a marker gene suitable for enrichment of cells carrying the DNA.
- a marker gene suitable for enrichment of cells carrying the DNA In this example, the phosphoglycerate kinase promoter and a cDNA encoding resistance to hygromycin were used; the transcriptional unit is designated pGK-hygror.
- the second transcriptional unit comprises a cell lineage-restricted promoter, linked to a marker gene suitable for enrichment of the desired cells.
- the cardiomyocyte- restricted ⁇ -cardiac myosin heavy chain promoter and a cDNA encoding aminoglycoside phosphotransferase were used; the transcriptional unit is designated MHC-neor.
- Undifferentiated, intestinally derived pluripotent cells are transfected with the MHC- neor/pGK-hygror construct. Cells incorporating the DNA are enriched based on their resistance to hygromycin. Differentiation is then induced, and once evidence of cardiomyogenesis is observed (i.e. spontaneous contractile activity), the cultures are treated with G418. Since the ⁇ -MHC promoter is only active in cardiomyocytes, only these cells express aminoglycoside phosphotransferase and survive G418 treatment.
- the genetic enrichment approach has the advantage that very long-term cultures of terminally differentiated cardiomyocytes can be generated, since all non- cardiomyocytes are eliminated from the culture. Moreover, the approach is easily amenable to gene transfer, either prior to differentiation or after the generation of terminally differentiated cells. In addition, the genetic enrichment approach is applicable to all pluripotent stem cell systems.
- the Methods are divided into sub-sections describing: (1) the transfection and selection of undifferentiated intestinally derived pluripotent cells; (2) the "en mass” differentiation of the selected cells; and (3) the generation of highly enriched cardiomyocyte cultures.
- the selection cassette Prior to transfecting intestinally derived pluripotent cells, the selection cassette (pMHC-neor/PGK-hygror) was digested with Xho I/Hind III and the fragment containing the entire MHC-neor/PGK-hygror sequence was isolated using a gene clean kit.
- Intestinally derived pluripotent cells are maintained in an undifferentiated state by culturing on mitotically inactivated human fibroblast feeder layer in Growth Medium: DMEM (Gibco-BRL) supplemented with 15% fetal bovine serum (Gibco-BRL), 0.1 mM nonessential amino acids (Gibco-BRL), 0.1 mM 2-mercaptoethanol (Gibco-BRL), 2 mM glutamine (Gibco-BRL), 500 ng/ml human recombinant fibroblast growth factor-2 (rhFGF-2) (Upstate Biotech) and 1 ,000 units/ml of human recombinant leukemia inhibitory factor (rhLIF) (Sigma).
- DMEM Gibco-BRL
- fetal bovine serum Gibco-BRL
- 0.1 mM nonessential amino acids Gibco-BRL
- 0.1 mM 2-mercaptoethanol Gibco-BRL
- MHC-neor/PGK-hygror DNA (1 ⁇ g) were mixed with salmon testes DNA (25 ⁇ g) in a total volume of 70 ⁇ l and sonicated. This mixture was added to the cells and left in the electroporation chamber on ice for 15 minutes.
- DMEM fetal bovine serum
- 0.1 mM nonessential amino acids Gibco-BRL
- 0.1 mM 2-mercaptoethanol Gibco-BRL
- 2 mM glutamine Gibco-BRL
- 500 ng/ml human recombinant fibroblast growth factor-2 rhFGF-2 (Upstate Biotech). Cells grow in suspension under these conditions.
- the cells were supplemented with 5 ml of Differentiation Medium on the next day to facilitate EB formation.
- EBs were collected on the fifth day by gravity and resuspended in 10 ml of Differentiation Medium. EBs were plated in 100 mm Corning cell culture dishes at different dilutions (1 :2, 1 :5, 1 :10 etc).
- pancreatic ⁇ cells from differentiating intestinally derived pluripotent cells.
- Example 4 is modified such that a second expression cassette is introduced to produce a gene product coding for a transcription /growth factor involved in beta-cell differentation (for example pdx-1).
- the first transcriptional unit comprises hlNSp-neo-pGK-hygro where hlNSp stands for the human insulin promoter and the second transcriptional unit comprises pGK-pdx-1 where pdx-1 represents the human pdx-1 gene.
- Undifferentiated, intestinally derived pluripotent cells are transfected with the hlNSp-neo-pGK-hygro / pGK-pdx-1 constructs as described in Example 4. Cells which have incorporated the DNA are enriched based on their resistance to hygromycin, and are directed toward an insulin-secreting cell fate by their expression of the pdx-1 gene.
- hygromycin-resistant HISC cells obtained by Method 1 of Example 4, are plated onto a 100 mm bacterial Petri dish and cultured in DMEM lacking supplemental LIF. After 8-10 days in suspension culture, the resulting EBs are plated onto plastic 100 mm cell culture dishes and allowed to attach for 5-8 days.
- the differentiated cultures are grown in the same medium in the presence of 200 ⁇ g/ml G418 (GIBCO/BRL). Single G418 EB clones are selected, isolated into 24 well plates, and then expanded. This approach avoids the dilution of robust insulin-secreting clones by clones that are weaker insulin producers.
- the resulting clones are trypsinized and plated in a 100 mm bacterial Petri dish and gown for 14 days in DMEM supplemented with 200 ⁇ g/ml G418 and 10 mM nicotinamide (Sigma, Madrid). Finally, the resulting clusters (pseudo-islet structures) are cultured for 1-5 days in RPMI 1640 supplemented with 10% fetal bovine serum, 10 mM nicotinamide, 200 ⁇ g/ml G418, 100 lU/ml penicillin, 0.1 mg/ml streptomycin, and 5.6 mM glucose.
- Pancreatic ⁇ cells produced by the methods of Example 5 can be screened for insulin secretion. For static incubations, cells are plated in 12-well dishes (Corning Glass
- the buffer is removed form the vials. Each condition is assayed in triplicate.
- Perfusion studies are performed as described in Perrusa et al. (1999) J. Physiol. 520:470-80. Briefly, 1 x 10 5 HISC-derived insulin-secreting cells (as free floating clusters) are packed into a 1 cm diameter column and sandwiched between two layers of swollen Sephadex G-200 microcarrier beads (Sigma, St. Louis, MO). The column is perfused at a flow rate of 1 ml/min at 37 ° C with fresh modified Krebs buffer with 1% BSA plus glucose (3 or 22 mM).
- the HISC-derived insulin-secreting cells are first perfused in 3 mM glucose for 30 min to reach a state of stable insulin release.
- the solutions are prewarmed at 37"C and continuously gassed.
- Insulin is assayed by radio- immunoassay (RIA) using the Coat-a-Count kit (DPC, Los Angeles, CA) that detects both rat and human insulin.
- RIA radio- immunoassay
- cell pellets are sonicated in 1 mM acetic acid containing 1% BSA and cellular extract is also determined by RIA. Secretion is normalized for cell number by measuring total protein in each experiment by the method of Bradford. Bradford (1976) Anal. Biochem. 72:248-54.
- CD34 + bone marrow cells highly enriched for pluripotent stem cells Exp. Hematol.
- Sakakibara et al. (1996) “Mouse-Musashi-1 , a neural RNA-binding protein highly enriched in the mammalian CNS stem cell” Dev. Biol. 176:230-242. Sakakibara et al. (1997) "Expression of neural RNA-binding proteins in the postnatal CNS: Implications of their roles in neuronal and glial cell development” J. Neurosci.
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Engineering & Computer Science (AREA)
- Biomedical Technology (AREA)
- Chemical & Material Sciences (AREA)
- Biotechnology (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Organic Chemistry (AREA)
- Genetics & Genomics (AREA)
- Wood Science & Technology (AREA)
- Zoology (AREA)
- General Health & Medical Sciences (AREA)
- Cell Biology (AREA)
- Microbiology (AREA)
- Immunology (AREA)
- Biochemistry (AREA)
- General Engineering & Computer Science (AREA)
- Hematology (AREA)
- Developmental Biology & Embryology (AREA)
- Urology & Nephrology (AREA)
- Medicinal Chemistry (AREA)
- Molecular Biology (AREA)
- Tropical Medicine & Parasitology (AREA)
- General Chemical & Material Sciences (AREA)
- Public Health (AREA)
- Veterinary Medicine (AREA)
- Chemical Kinetics & Catalysis (AREA)
- Pharmacology & Pharmacy (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Animal Behavior & Ethology (AREA)
- Food Science & Technology (AREA)
- Physics & Mathematics (AREA)
- Analytical Chemistry (AREA)
- General Physics & Mathematics (AREA)
- Pathology (AREA)
- Gastroenterology & Hepatology (AREA)
- Micro-Organisms Or Cultivation Processes Thereof (AREA)
Abstract
Priority Applications (1)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
EP02712824A EP1370642A2 (fr) | 2001-01-20 | 2002-01-18 | Cellules souches adultes pluripotentes provenant de tissu regeneratif |
Applications Claiming Priority (8)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
EP01101333 | 2001-01-20 | ||
EP01101333 | 2001-01-20 | ||
US28710501P | 2001-04-27 | 2001-04-27 | |
US287105P | 2001-04-27 | ||
US32400801P | 2001-09-21 | 2001-09-21 | |
US324008P | 2001-09-21 | ||
PCT/EP2002/000475 WO2002057430A2 (fr) | 2001-01-20 | 2002-01-18 | Cellules souches adultes pluripotentes provenant de tissu regeneratif |
EP02712824A EP1370642A2 (fr) | 2001-01-20 | 2002-01-18 | Cellules souches adultes pluripotentes provenant de tissu regeneratif |
Publications (1)
Publication Number | Publication Date |
---|---|
EP1370642A2 true EP1370642A2 (fr) | 2003-12-17 |
Family
ID=56290241
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
EP02712824A Withdrawn EP1370642A2 (fr) | 2001-01-20 | 2002-01-18 | Cellules souches adultes pluripotentes provenant de tissu regeneratif |
Country Status (3)
Country | Link |
---|---|
EP (1) | EP1370642A2 (fr) |
CA (1) | CA2434362A1 (fr) |
WO (1) | WO2002057430A2 (fr) |
Families Citing this family (29)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
CA2461859A1 (fr) * | 2001-09-28 | 2003-04-10 | Es Cell International Pte Ltd | Procedes de derivation et de propagation de cellules souches embryonnaires humaines (hes) non differenciees sur des matrices sans cellules nourricieres et sur des couches nourricieres humaines |
US7422736B2 (en) * | 2002-07-26 | 2008-09-09 | Food Industry Research And Development Institute | Somatic pluripotent cells |
DE10261125A1 (de) * | 2002-12-20 | 2004-07-01 | Cardion Ag | Verfahren zur Herstellung von Stammzellen aus Pankreas |
DE10362002B4 (de) * | 2003-06-23 | 2006-10-12 | Fraunhofer-Gesellschaft zur Förderung der angewandten Forschung e.V. | Adulte pluripotente Stammzellen |
US20050186182A1 (en) * | 2003-11-10 | 2005-08-25 | Theresa Deisher | Methods of using G-CSF mobilized C-Kit+ cells in the production of embryoid body-like cell clusters for tissue repair and in the treatment of cardiac myopathy |
AU2005210581B2 (en) * | 2004-02-04 | 2010-06-24 | Am-Pharma B.V. | Use of alkaline phosphatase for the detoxification of LPS present at mucosal barriers |
AU2005331559B2 (en) * | 2005-05-05 | 2012-04-19 | Regents Of The University Of Minnesota | Use of NK cell inhibition to facilitate persistence of engrafted MHC-I negative cells |
JP2009509911A (ja) * | 2005-05-05 | 2009-03-12 | リージェンツ オブ ザ ユニバーシティ オブ ミネソタ | リンパ造血組織を定着させるためのmapcまたはそれらの子孫の使用 |
AU2005331534B2 (en) * | 2005-05-05 | 2011-12-08 | Regents Of The University Of Minnesota | Use of MAPC or progeny therefrom to populate lymphohematopoietic tissues |
DE102006003996A1 (de) | 2006-01-27 | 2007-08-02 | Fraunhofer-Gesellschaft zur Förderung der angewandten Forschung e.V. | Verfahren zur Herstellung autonom kontrahierender Herzmuskelzellen aus adulten Stammzellen, insbesondere humanen adulten Stammzellen |
DE102007034679A1 (de) | 2007-07-25 | 2009-01-29 | Fraunhofer-Gesellschaft zur Förderung der angewandten Forschung e.V. | Materialzusammensetzungen, welche aus exokrinem Drüsengewebe erhaltene adulte Stammzellen enthalten, insbesondere zur Verwendung in der Regenerationsmedizin, z.B. zur Wiederherstellung von verletztem oder geschädigtem Myokardgewebe |
US20090269769A1 (en) * | 2008-04-23 | 2009-10-29 | Asit Panja | Drug Discovery Methods Involving A Preclinical, In Vitro Isolated Gastrointestinal Epithelial Stem Cell-Like Progenitor Cell System |
US20100093552A1 (en) | 2008-10-09 | 2010-04-15 | Asit Panja | Use and identification of biomarkers for gastrointestinal diseases |
US9081008B2 (en) | 2009-12-04 | 2015-07-14 | James Sherley | Detecting and counting tissue—specific stem cells and uses thereof |
WO2012048298A2 (fr) | 2010-10-08 | 2012-04-12 | Caridianbct, Inc. | Procédés et systèmes de culture et de récolte de cellules dans un système de bioréacteur à fibres creuses avec conditions de régulation |
WO2014027474A1 (fr) | 2012-08-17 | 2014-02-20 | 株式会社Clio | Cellule souche pluripotente induisant une réparation et une régénération après un infarctus du myocarde |
EP3068867B1 (fr) | 2013-11-16 | 2018-04-18 | Terumo BCT, Inc. | Expansion de cellules dans un bioréacteur |
JP6940949B2 (ja) | 2014-01-24 | 2021-09-29 | アーエム−ファルマ ベー.フェー.AM−Pharma B.V. | キメラアルカリホスファターゼ様タンパク質 |
EP3097188B1 (fr) | 2014-01-24 | 2018-08-29 | AM-Pharma B.V. | Traitement en aval d'une phosphatase alcaline |
WO2015148704A1 (fr) | 2014-03-25 | 2015-10-01 | Terumo Bct, Inc. | Remplacement passif de milieu |
CN106715676A (zh) | 2014-09-26 | 2017-05-24 | 泰尔茂比司特公司 | 按计划供养 |
CN104862272B (zh) * | 2015-05-29 | 2018-08-10 | 吉林大学 | Y-27632抑制剂在cd44阳性肠干细胞分选中的应用 |
WO2017004592A1 (fr) | 2015-07-02 | 2017-01-05 | Terumo Bct, Inc. | Croissance cellulaire à l'aide de stimuli mécaniques |
US11965175B2 (en) | 2016-05-25 | 2024-04-23 | Terumo Bct, Inc. | Cell expansion |
US11685883B2 (en) | 2016-06-07 | 2023-06-27 | Terumo Bct, Inc. | Methods and systems for coating a cell growth surface |
US11104874B2 (en) | 2016-06-07 | 2021-08-31 | Terumo Bct, Inc. | Coating a bioreactor |
US11624046B2 (en) | 2017-03-31 | 2023-04-11 | Terumo Bct, Inc. | Cell expansion |
WO2018184028A2 (fr) | 2017-03-31 | 2018-10-04 | Terumo Bct, Inc. | Expansion cellulaire |
JP2024511064A (ja) | 2021-03-23 | 2024-03-12 | テルモ ビーシーティー、インコーポレーテッド | 細胞捕獲及び増殖 |
Family Cites Families (2)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US5968546A (en) * | 1997-05-16 | 1999-10-19 | Baur; Marcus | Keratinocyte culture from precursor cells |
IL144654A0 (en) * | 1999-02-10 | 2002-05-23 | Curis Inc | Pancreatic progenitor cells, methods and uses related thereto |
-
2002
- 2002-01-18 WO PCT/EP2002/000475 patent/WO2002057430A2/fr not_active Application Discontinuation
- 2002-01-18 EP EP02712824A patent/EP1370642A2/fr not_active Withdrawn
- 2002-01-18 CA CA002434362A patent/CA2434362A1/fr not_active Abandoned
Non-Patent Citations (1)
Title |
---|
See references of WO02057430A2 * |
Also Published As
Publication number | Publication date |
---|---|
WO2002057430A2 (fr) | 2002-07-25 |
WO2002057430A3 (fr) | 2003-10-16 |
CA2434362A1 (fr) | 2002-07-25 |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
WO2002057430A2 (fr) | Cellules souches adultes pluripotentes provenant de tissu regeneratif | |
US20240309330A1 (en) | Differentiation and enrichment of islet-like cells from human pluripotent stem cells | |
JP6527487B2 (ja) | 多能性幹細胞から誘導した細胞を精製するための方法 | |
US9005897B2 (en) | Method of deriving progenitor cell line | |
van der WEE et al. | Immunomagnetic isolation and long‐term culture of mouse type A spermatogonia | |
Thomas et al. | Expression of endoderm stem cell markers: evidence for the presence of adult stem cells in human thyroid glands | |
US7432104B2 (en) | Methods for the culture of human embryonic stem cells on human feeder cells | |
US20050095703A1 (en) | Method for the establishment of a pluripotent human blastocyst - derived stem cell line | |
US20050032207A1 (en) | Method for isolating, culturing and differentiating intestinal stem cells for therapeutic use | |
US20100129906A1 (en) | Method for Obtaining Xeno-Free Hbs Cell line | |
US20090104158A1 (en) | Non-Embryonic Totipotent Blastomere-Like Stem Cells And Methods Therefor | |
US10093895B2 (en) | Hepatic stellate cell precursors and methods of isolating same | |
WO2001053465A9 (fr) | Cellules issues de corps embryoides humains | |
WO2009116893A1 (fr) | Procédé de fabrication de cellules endothéliales (et variantes) | |
US20050239201A1 (en) | Methods of inducing differentiation of stem cells into a specific cell lineage | |
WO1997047734A1 (fr) | Differenciation et culture in vitro de cellules souches multipotentes de primates et usages therapeutiques | |
US20090253151A1 (en) | Self-Renewing Master Adult Pluripotent Stem Cells | |
US20100136598A1 (en) | Novel mesenchymal progenitor cells derived from human blastocyst-derived stem cells | |
AU2002244652A1 (en) | Pluripotent adult stem cells derived from regenerative tissue | |
Pall et al. | Establishment of an embryonic stem cell line from blastocyst stage mouse embryos | |
KR20240056604A (ko) | 수임 심장 전구세포의 제조 방법 | |
Mehta et al. | Germinal Stem Cells: Culture and Replication | |
KR20040071259A (ko) | 다능성의 인간포배로부터 파생된 간세포주를 확립하기위한 방법 | |
AU2003209830A1 (en) | Methods of inducing differentiation of stem cells into a specific cell lineage |
Legal Events
Date | Code | Title | Description |
---|---|---|---|
PUAI | Public reference made under article 153(3) epc to a published international application that has entered the european phase |
Free format text: ORIGINAL CODE: 0009012 |
|
17P | Request for examination filed |
Effective date: 20030807 |
|
AK | Designated contracting states |
Kind code of ref document: A2 Designated state(s): AT BE CH CY DE DK ES FI FR GB GR IE IT LI LU MC NL PT SE TR |
|
AX | Request for extension of the european patent |
Extension state: AL LT LV MK RO SI |
|
17Q | First examination report despatched |
Effective date: 20040319 |
|
RAP1 | Party data changed (applicant data changed or rights of an application transferred) |
Owner name: SORIA ESCOMS, BERNAT |
|
RTI1 | Title (correction) |
Free format text: PLURIPOTENT ADULT STEM CELLS DERIVED FROM HUMAN INTESTINE CRYPT TISSUE |
|
GRAP | Despatch of communication of intention to grant a patent |
Free format text: ORIGINAL CODE: EPIDOSNIGR1 |
|
STAA | Information on the status of an ep patent application or granted ep patent |
Free format text: STATUS: THE APPLICATION IS DEEMED TO BE WITHDRAWN |
|
18D | Application deemed to be withdrawn |
Effective date: 20060801 |