WO2020030822A1 - Hepatoblast organoids - Google Patents
Hepatoblast organoids Download PDFInfo
- Publication number
- WO2020030822A1 WO2020030822A1 PCT/EP2019/071587 EP2019071587W WO2020030822A1 WO 2020030822 A1 WO2020030822 A1 WO 2020030822A1 EP 2019071587 W EP2019071587 W EP 2019071587W WO 2020030822 A1 WO2020030822 A1 WO 2020030822A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- hepatoblasts
- population
- cells
- medium
- hepatoblast
- Prior art date
Links
- 210000003999 epithelial cell of bile duct Anatomy 0.000 title claims abstract description 235
- 210000002220 organoid Anatomy 0.000 title claims abstract description 116
- 210000004027 cell Anatomy 0.000 claims abstract description 152
- 210000003494 hepatocyte Anatomy 0.000 claims abstract description 109
- 238000000034 method Methods 0.000 claims abstract description 62
- 102000013814 Wnt Human genes 0.000 claims abstract description 60
- 108050003627 Wnt Proteins 0.000 claims abstract description 60
- 239000003112 inhibitor Substances 0.000 claims abstract description 41
- 230000011664 signaling Effects 0.000 claims abstract description 26
- 210000005229 liver cell Anatomy 0.000 claims abstract description 23
- 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 abstract description 21
- 238000012258 culturing Methods 0.000 claims abstract description 12
- 239000011435 rock Substances 0.000 claims abstract 7
- 239000002609 medium Substances 0.000 claims description 78
- 230000004069 differentiation Effects 0.000 claims description 38
- 150000001875 compounds Chemical class 0.000 claims description 31
- 238000000338 in vitro Methods 0.000 claims description 28
- 238000012360 testing method Methods 0.000 claims description 22
- 208000019423 liver disease Diseases 0.000 claims description 19
- 230000000694 effects Effects 0.000 claims description 18
- 239000001963 growth medium Substances 0.000 claims description 18
- 238000004519 manufacturing process Methods 0.000 claims description 15
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 claims description 12
- 239000007640 basal medium Substances 0.000 claims description 7
- 208000031220 Hemophilia Diseases 0.000 claims description 5
- 208000009292 Hemophilia A Diseases 0.000 claims description 5
- 238000012216 screening Methods 0.000 claims description 5
- 102000009618 Transforming Growth Factors Human genes 0.000 claims 8
- 230000007774 longterm Effects 0.000 abstract description 7
- 238000012136 culture method Methods 0.000 abstract description 3
- 230000014509 gene expression Effects 0.000 description 62
- 210000004185 liver Anatomy 0.000 description 56
- 108090000623 proteins and genes Proteins 0.000 description 56
- 108010026331 alpha-Fetoproteins Proteins 0.000 description 34
- 102000013529 alpha-Fetoproteins Human genes 0.000 description 34
- 108010088751 Albumins Proteins 0.000 description 32
- 102000009027 Albumins Human genes 0.000 description 32
- 102000004169 proteins and genes Human genes 0.000 description 28
- 235000018102 proteins Nutrition 0.000 description 27
- 210000001519 tissue Anatomy 0.000 description 26
- 238000004458 analytical method Methods 0.000 description 23
- 239000000243 solution Substances 0.000 description 20
- 102100022712 Alpha-1-antitrypsin Human genes 0.000 description 19
- 108010066687 Epithelial Cell Adhesion Molecule Proteins 0.000 description 19
- 102000018651 Epithelial Cell Adhesion Molecule Human genes 0.000 description 19
- 102100033420 Keratin, type I cytoskeletal 19 Human genes 0.000 description 19
- 101000998011 Homo sapiens Keratin, type I cytoskeletal 19 Proteins 0.000 description 18
- 239000000017 hydrogel Substances 0.000 description 18
- 239000003814 drug Substances 0.000 description 16
- 239000000203 mixture Substances 0.000 description 15
- 238000011282 treatment Methods 0.000 description 15
- 230000002440 hepatic effect Effects 0.000 description 14
- 101000823116 Homo sapiens Alpha-1-antitrypsin Proteins 0.000 description 13
- 238000001727 in vivo Methods 0.000 description 13
- 210000005228 liver tissue Anatomy 0.000 description 13
- 210000000130 stem cell Anatomy 0.000 description 13
- 108010081668 Cytochrome P-450 CYP3A Proteins 0.000 description 12
- 238000011161 development Methods 0.000 description 12
- 230000018109 developmental process Effects 0.000 description 12
- 230000006870 function Effects 0.000 description 12
- 238000003556 assay Methods 0.000 description 11
- 210000004500 stellate cell Anatomy 0.000 description 11
- 108010035532 Collagen Proteins 0.000 description 10
- 102000008186 Collagen Human genes 0.000 description 10
- 102100039205 Cytochrome P450 3A4 Human genes 0.000 description 10
- 102100039203 Cytochrome P450 3A7 Human genes 0.000 description 10
- 101710104801 Cytochrome P450 3A7 Proteins 0.000 description 10
- -1 GlutamaxTM) Chemical compound 0.000 description 10
- 210000000013 bile duct Anatomy 0.000 description 10
- 229920001436 collagen Polymers 0.000 description 10
- 230000001605 fetal effect Effects 0.000 description 10
- 102100040202 Apolipoprotein B-100 Human genes 0.000 description 9
- 238000004113 cell culture Methods 0.000 description 9
- 230000001413 cellular effect Effects 0.000 description 9
- 230000001172 regenerating effect Effects 0.000 description 9
- 210000002966 serum Anatomy 0.000 description 9
- 102000004190 Enzymes Human genes 0.000 description 8
- 108090000790 Enzymes Proteins 0.000 description 8
- 229940088598 enzyme Drugs 0.000 description 8
- 239000011159 matrix material Substances 0.000 description 8
- 230000002062 proliferating effect Effects 0.000 description 8
- 239000000523 sample Substances 0.000 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 description 8
- 102100026292 Asialoglycoprotein receptor 1 Human genes 0.000 description 7
- 239000006144 Dulbecco’s modified Eagle's medium Substances 0.000 description 7
- 101800003838 Epidermal growth factor Proteins 0.000 description 7
- 101000889953 Homo sapiens Apolipoprotein B-100 Proteins 0.000 description 7
- 101000998020 Homo sapiens Keratin, type I cytoskeletal 18 Proteins 0.000 description 7
- 102100033421 Keratin, type I cytoskeletal 18 Human genes 0.000 description 7
- 102100033237 Pro-epidermal growth factor Human genes 0.000 description 7
- 231100000839 Vanishing bile duct syndrome Toxicity 0.000 description 7
- 239000006285 cell suspension Substances 0.000 description 7
- 230000006378 damage Effects 0.000 description 7
- 208000035475 disorder Diseases 0.000 description 7
- 229940079593 drug Drugs 0.000 description 7
- 229940116977 epidermal growth factor Drugs 0.000 description 7
- 239000003102 growth factor Substances 0.000 description 7
- 239000003550 marker Substances 0.000 description 7
- 235000015097 nutrients Nutrition 0.000 description 7
- 230000035755 proliferation Effects 0.000 description 7
- 101000785944 Homo sapiens Asialoglycoprotein receptor 1 Proteins 0.000 description 6
- 241000699666 Mus <mouse, genus> Species 0.000 description 6
- 241000699670 Mus sp. Species 0.000 description 6
- 238000011529 RT qPCR Methods 0.000 description 6
- 108010050122 alpha 1-Antitrypsin Proteins 0.000 description 6
- 229940024142 alpha 1-antitrypsin Drugs 0.000 description 6
- 238000000429 assembly Methods 0.000 description 6
- 230000000712 assembly Effects 0.000 description 6
- 239000000463 material Substances 0.000 description 6
- 230000002503 metabolic effect Effects 0.000 description 6
- 150000007523 nucleic acids Chemical group 0.000 description 6
- 230000005305 organ development Effects 0.000 description 6
- 238000000513 principal component analysis Methods 0.000 description 6
- 230000019491 signal transduction Effects 0.000 description 6
- 238000012174 single-cell RNA sequencing Methods 0.000 description 6
- 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 6
- 208000015163 Biliary Tract disease Diseases 0.000 description 5
- 102000004506 Blood Proteins Human genes 0.000 description 5
- 108010017384 Blood Proteins Proteins 0.000 description 5
- 238000002965 ELISA Methods 0.000 description 5
- 241000283074 Equus asinus Species 0.000 description 5
- 108091028043 Nucleic acid sequence Proteins 0.000 description 5
- FAPWRFPIFSIZLT-UHFFFAOYSA-M Sodium chloride Chemical compound [Na+].[Cl-] FAPWRFPIFSIZLT-UHFFFAOYSA-M 0.000 description 5
- 108090001012 Transforming Growth Factor beta Proteins 0.000 description 5
- 150000001413 amino acids Chemical group 0.000 description 5
- 238000013459 approach Methods 0.000 description 5
- 239000000872 buffer Substances 0.000 description 5
- 230000007423 decrease Effects 0.000 description 5
- 238000009795 derivation Methods 0.000 description 5
- 238000010494 dissociation reaction Methods 0.000 description 5
- 230000005593 dissociations Effects 0.000 description 5
- 238000007877 drug screening Methods 0.000 description 5
- 238000005516 engineering process Methods 0.000 description 5
- 210000000981 epithelium Anatomy 0.000 description 5
- 238000009472 formulation Methods 0.000 description 5
- 239000012595 freezing medium Substances 0.000 description 5
- 230000012010 growth Effects 0.000 description 5
- 230000003908 liver function Effects 0.000 description 5
- 108010082117 matrigel Proteins 0.000 description 5
- 210000000056 organ Anatomy 0.000 description 5
- 230000037361 pathway Effects 0.000 description 5
- 239000008194 pharmaceutical composition Substances 0.000 description 5
- 108091032973 (ribonucleotides)n+m Proteins 0.000 description 4
- JKMHFZQWWAIEOD-UHFFFAOYSA-N 2-[4-(2-hydroxyethyl)piperazin-1-yl]ethanesulfonic acid Chemical compound OCC[NH+]1CCN(CCS([O-])(=O)=O)CC1 JKMHFZQWWAIEOD-UHFFFAOYSA-N 0.000 description 4
- HJCMDXDYPOUFDY-WHFBIAKZSA-N Ala-Gln Chemical compound C[C@H](N)C(=O)N[C@H](C(O)=O)CCC(N)=O HJCMDXDYPOUFDY-WHFBIAKZSA-N 0.000 description 4
- 102100033715 Apolipoprotein A-I Human genes 0.000 description 4
- 102100036264 Glucose-6-phosphatase catalytic subunit 1 Human genes 0.000 description 4
- 239000007995 HEPES buffer Substances 0.000 description 4
- 101000930910 Homo sapiens Glucose-6-phosphatase catalytic subunit 1 Proteins 0.000 description 4
- 101000587539 Homo sapiens Metallothionein-1A Proteins 0.000 description 4
- 101001027938 Homo sapiens Metallothionein-1G Proteins 0.000 description 4
- 101001013799 Homo sapiens Metallothionein-1X Proteins 0.000 description 4
- PWKSKIMOESPYIA-BYPYZUCNSA-N L-N-acetyl-Cysteine Chemical compound CC(=O)N[C@@H](CS)C(O)=O PWKSKIMOESPYIA-BYPYZUCNSA-N 0.000 description 4
- 102100029698 Metallothionein-1A Human genes 0.000 description 4
- 102000004140 Oncostatin M Human genes 0.000 description 4
- 108090000630 Oncostatin M Proteins 0.000 description 4
- 229930182555 Penicillin Natural products 0.000 description 4
- 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 4
- 229920000954 Polyglycolide Polymers 0.000 description 4
- 102000004887 Transforming Growth Factor beta Human genes 0.000 description 4
- 230000005856 abnormality Effects 0.000 description 4
- 229960004308 acetylcysteine Drugs 0.000 description 4
- 239000003114 blood coagulation factor Substances 0.000 description 4
- 230000008859 change Effects 0.000 description 4
- 239000003153 chemical reaction reagent Substances 0.000 description 4
- 201000010099 disease Diseases 0.000 description 4
- 230000002068 genetic effect Effects 0.000 description 4
- 239000004615 ingredient Substances 0.000 description 4
- 230000031142 liver development Effects 0.000 description 4
- 210000004962 mammalian cell Anatomy 0.000 description 4
- 230000007246 mechanism Effects 0.000 description 4
- 239000002207 metabolite Substances 0.000 description 4
- 229940049954 penicillin Drugs 0.000 description 4
- 239000000546 pharmaceutical excipient Substances 0.000 description 4
- 239000004633 polyglycolic acid Substances 0.000 description 4
- 229960005322 streptomycin Drugs 0.000 description 4
- 238000002054 transplantation Methods 0.000 description 4
- XLYOFNOQVPJJNP-UHFFFAOYSA-N water Substances O XLYOFNOQVPJJNP-UHFFFAOYSA-N 0.000 description 4
- 102100040881 60S acidic ribosomal protein P0 Human genes 0.000 description 3
- 102100021723 Arginase-1 Human genes 0.000 description 3
- 102100030802 Beta-2-glycoprotein 1 Human genes 0.000 description 3
- 208000008439 Biliary Liver Cirrhosis Diseases 0.000 description 3
- 208000033222 Biliary cirrhosis primary Diseases 0.000 description 3
- 102000015081 Blood Coagulation Factors Human genes 0.000 description 3
- 108010039209 Blood Coagulation Factors Proteins 0.000 description 3
- 102000002004 Cytochrome P-450 Enzyme System Human genes 0.000 description 3
- 108010015742 Cytochrome P-450 Enzyme System Proteins 0.000 description 3
- IGXWBGJHJZYPQS-SSDOTTSWSA-N D-Luciferin Chemical compound OC(=O)[C@H]1CSC(C=2SC3=CC=C(O)C=C3N=2)=N1 IGXWBGJHJZYPQS-SSDOTTSWSA-N 0.000 description 3
- CYCGRDQQIOGCKX-UHFFFAOYSA-N Dehydro-luciferin Natural products OC(=O)C1=CSC(C=2SC3=CC(O)=CC=C3N=2)=N1 CYCGRDQQIOGCKX-UHFFFAOYSA-N 0.000 description 3
- LYCAIKOWRPUZTN-UHFFFAOYSA-N Ethylene glycol Chemical compound OCCO LYCAIKOWRPUZTN-UHFFFAOYSA-N 0.000 description 3
- BJGNCJDXODQBOB-UHFFFAOYSA-N Fivefly Luciferin Natural products OC(=O)C1CSC(C=2SC3=CC(O)=CC=C3N=2)=N1 BJGNCJDXODQBOB-UHFFFAOYSA-N 0.000 description 3
- WZUVPPKBWHMQCE-UHFFFAOYSA-N Haematoxylin Natural products C12=CC(O)=C(O)C=C2CC2(O)C1C1=CC=C(O)C(O)=C1OC2 WZUVPPKBWHMQCE-UHFFFAOYSA-N 0.000 description 3
- 101000673456 Homo sapiens 60S acidic ribosomal protein P0 Proteins 0.000 description 3
- 101000733802 Homo sapiens Apolipoprotein A-I Proteins 0.000 description 3
- 101000752037 Homo sapiens Arginase-1 Proteins 0.000 description 3
- 101000793425 Homo sapiens Beta-2-glycoprotein 1 Proteins 0.000 description 3
- 101000975502 Homo sapiens Keratin, type II cytoskeletal 7 Proteins 0.000 description 3
- 101001027956 Homo sapiens Metallothionein-1B Proteins 0.000 description 3
- 101000800287 Homo sapiens Tubulointerstitial nephritis antigen-like Proteins 0.000 description 3
- 108010003272 Hyaluronate lyase Proteins 0.000 description 3
- 102000001974 Hyaluronidases Human genes 0.000 description 3
- 102100023974 Keratin, type II cytoskeletal 7 Human genes 0.000 description 3
- 108010085895 Laminin Proteins 0.000 description 3
- DDWFXDSYGUXRAY-UHFFFAOYSA-N Luciferin Natural products CCc1c(C)c(CC2NC(=O)C(=C2C=C)C)[nH]c1Cc3[nH]c4C(=C5/NC(CC(=O)O)C(C)C5CC(=O)O)CC(=O)c4c3C DDWFXDSYGUXRAY-UHFFFAOYSA-N 0.000 description 3
- 239000004793 Polystyrene Substances 0.000 description 3
- 208000012654 Primary biliary cholangitis Diseases 0.000 description 3
- DNIAPMSPPWPWGF-UHFFFAOYSA-N Propylene glycol Chemical compound CC(O)CO DNIAPMSPPWPWGF-UHFFFAOYSA-N 0.000 description 3
- 208000027418 Wounds and injury Diseases 0.000 description 3
- DFPAKSUCGFBDDF-ZQBYOMGUSA-N [14c]-nicotinamide Chemical compound N[14C](=O)C1=CC=CN=C1 DFPAKSUCGFBDDF-ZQBYOMGUSA-N 0.000 description 3
- 229960002648 alanylglutamine Drugs 0.000 description 3
- 102000012005 alpha-2-HS-Glycoprotein Human genes 0.000 description 3
- 108010075843 alpha-2-HS-Glycoprotein Proteins 0.000 description 3
- 230000000890 antigenic effect Effects 0.000 description 3
- 210000003445 biliary tract Anatomy 0.000 description 3
- 239000006143 cell culture medium Substances 0.000 description 3
- 238000011109 contamination Methods 0.000 description 3
- 230000001419 dependent effect Effects 0.000 description 3
- 238000001784 detoxification Methods 0.000 description 3
- 238000010790 dilution Methods 0.000 description 3
- 239000012895 dilution Substances 0.000 description 3
- 238000002474 experimental method Methods 0.000 description 3
- 238000000684 flow cytometry Methods 0.000 description 3
- 238000001943 fluorescence-activated cell sorting Methods 0.000 description 3
- 239000012737 fresh medium Substances 0.000 description 3
- 230000005714 functional activity Effects 0.000 description 3
- 229960002773 hyaluronidase Drugs 0.000 description 3
- 238000003125 immunofluorescent labeling Methods 0.000 description 3
- 238000003364 immunohistochemistry Methods 0.000 description 3
- 208000014674 injury Diseases 0.000 description 3
- 239000007788 liquid Substances 0.000 description 3
- OUBCNLGXQFSTLU-UHFFFAOYSA-N nitisinone Chemical compound [O-][N+](=O)C1=CC(C(F)(F)F)=CC=C1C(=O)C1C(=O)CCCC1=O OUBCNLGXQFSTLU-UHFFFAOYSA-N 0.000 description 3
- 229960001721 nitisinone Drugs 0.000 description 3
- 229920002223 polystyrene Polymers 0.000 description 3
- 230000008569 process Effects 0.000 description 3
- 238000012545 processing Methods 0.000 description 3
- 230000002829 reductive effect Effects 0.000 description 3
- 238000012163 sequencing technique Methods 0.000 description 3
- 238000003860 storage Methods 0.000 description 3
- 239000000126 substance Substances 0.000 description 3
- 230000001225 therapeutic effect Effects 0.000 description 3
- 238000002560 therapeutic procedure Methods 0.000 description 3
- 230000007704 transition Effects 0.000 description 3
- 230000035899 viability Effects 0.000 description 3
- BTUOOXPZOVNPMF-UHFFFAOYSA-N 2-[4-(1,3-benzodioxol-5-yl)-2-tert-butyl-1h-imidazol-5-yl]-6-methylpyridine;hydrochloride Chemical compound Cl.CC1=CC=CC(C2=C(N=C(N2)C(C)(C)C)C=2C=C3OCOC3=CC=2)=N1 BTUOOXPZOVNPMF-UHFFFAOYSA-N 0.000 description 2
- JCSGFHVFHSKIJH-UHFFFAOYSA-N 3-(2,4-dichlorophenyl)-4-(1-methyl-3-indolyl)pyrrole-2,5-dione Chemical compound C12=CC=CC=C2N(C)C=C1C(C(NC1=O)=O)=C1C1=CC=C(Cl)C=C1Cl JCSGFHVFHSKIJH-UHFFFAOYSA-N 0.000 description 2
- FHVDTGUDJYJELY-UHFFFAOYSA-N 6-{[2-carboxy-4,5-dihydroxy-6-(phosphanyloxy)oxan-3-yl]oxy}-4,5-dihydroxy-3-phosphanyloxane-2-carboxylic acid Chemical compound O1C(C(O)=O)C(P)C(O)C(O)C1OC1C(C(O)=O)OC(OP)C(O)C1O FHVDTGUDJYJELY-UHFFFAOYSA-N 0.000 description 2
- 101150072844 APOM gene Proteins 0.000 description 2
- 102100022089 Acyl-[acyl-carrier-protein] hydrolase Human genes 0.000 description 2
- 229920000936 Agarose Polymers 0.000 description 2
- 201000011374 Alagille syndrome Diseases 0.000 description 2
- 101710095342 Apolipoprotein B Proteins 0.000 description 2
- 102100037324 Apolipoprotein M Human genes 0.000 description 2
- IJGRMHOSHXDMSA-UHFFFAOYSA-N Atomic nitrogen Chemical compound N#N IJGRMHOSHXDMSA-UHFFFAOYSA-N 0.000 description 2
- KXDAEFPNCMNJSK-UHFFFAOYSA-N Benzamide Chemical compound NC(=O)C1=CC=CC=C1 KXDAEFPNCMNJSK-UHFFFAOYSA-N 0.000 description 2
- 108091003079 Bovine Serum Albumin Proteins 0.000 description 2
- AQGNHMOJWBZFQQ-UHFFFAOYSA-N CT 99021 Chemical compound CC1=CNC(C=2C(=NC(NCCNC=3N=CC(=CC=3)C#N)=NC=2)C=2C(=CC(Cl)=CC=2)Cl)=N1 AQGNHMOJWBZFQQ-UHFFFAOYSA-N 0.000 description 2
- 101100497948 Caenorhabditis elegans cyn-1 gene Proteins 0.000 description 2
- CURLTUGMZLYLDI-UHFFFAOYSA-N Carbon dioxide Chemical compound O=C=O CURLTUGMZLYLDI-UHFFFAOYSA-N 0.000 description 2
- 206010057573 Chronic hepatic failure Diseases 0.000 description 2
- 206010010317 Congenital absence of bile ducts Diseases 0.000 description 2
- 108010001202 Cytochrome P-450 CYP2E1 Proteins 0.000 description 2
- 102000004328 Cytochrome P-450 CYP3A Human genes 0.000 description 2
- 102100024889 Cytochrome P450 2E1 Human genes 0.000 description 2
- 108020004414 DNA Proteins 0.000 description 2
- 229920002307 Dextran Polymers 0.000 description 2
- 102000001301 EGF receptor Human genes 0.000 description 2
- 108060006698 EGF receptor Proteins 0.000 description 2
- 208000010334 End Stage Liver Disease Diseases 0.000 description 2
- 108700039887 Essential Genes Proteins 0.000 description 2
- 102000010834 Extracellular Matrix Proteins Human genes 0.000 description 2
- 108010037362 Extracellular Matrix Proteins Proteins 0.000 description 2
- 108010014173 Factor X Proteins 0.000 description 2
- 108010039731 Fatty Acid Synthases Proteins 0.000 description 2
- 102000009123 Fibrin Human genes 0.000 description 2
- 108010073385 Fibrin Proteins 0.000 description 2
- BWGVNKXGVNDBDI-UHFFFAOYSA-N Fibrin monomer Chemical compound CNC(=O)CNC(=O)CN BWGVNKXGVNDBDI-UHFFFAOYSA-N 0.000 description 2
- 102100031752 Fibrinogen alpha chain Human genes 0.000 description 2
- 108010067306 Fibronectins Proteins 0.000 description 2
- 102000016359 Fibronectins Human genes 0.000 description 2
- 102100022272 Fructose-bisphosphate aldolase B Human genes 0.000 description 2
- 102000038624 GSKs Human genes 0.000 description 2
- 108091007911 GSKs Proteins 0.000 description 2
- 208000031448 Genomic Instability Diseases 0.000 description 2
- 206010019663 Hepatic failure Diseases 0.000 description 2
- 101000755933 Homo sapiens Fructose-bisphosphate aldolase B Proteins 0.000 description 2
- 101000976697 Homo sapiens Inter-alpha-trypsin inhibitor heavy chain H1 Proteins 0.000 description 2
- 101001133091 Homo sapiens Mucin-20 Proteins 0.000 description 2
- 101000665882 Homo sapiens Retinol-binding protein 4 Proteins 0.000 description 2
- 101000711846 Homo sapiens Transcription factor SOX-9 Proteins 0.000 description 2
- 101000803403 Homo sapiens Vimentin Proteins 0.000 description 2
- 102100023490 Inter-alpha-trypsin inhibitor heavy chain H1 Human genes 0.000 description 2
- XEEYBQQBJWHFJM-UHFFFAOYSA-N Iron Chemical compound [Fe] XEEYBQQBJWHFJM-UHFFFAOYSA-N 0.000 description 2
- 108010093811 Kazal Pancreatic Trypsin Inhibitor Proteins 0.000 description 2
- 102000001626 Kazal Pancreatic Trypsin Inhibitor Human genes 0.000 description 2
- 108010052014 Liberase Proteins 0.000 description 2
- 102100034242 Mucin-20 Human genes 0.000 description 2
- DFPAKSUCGFBDDF-UHFFFAOYSA-N Nicotinamide Chemical compound NC(=O)C1=CC=CN=C1 DFPAKSUCGFBDDF-UHFFFAOYSA-N 0.000 description 2
- 101150038994 PDGFRA gene Proteins 0.000 description 2
- 208000031481 Pathologic Constriction Diseases 0.000 description 2
- 108010094028 Prothrombin Proteins 0.000 description 2
- 102100038246 Retinol-binding protein 4 Human genes 0.000 description 2
- 239000008156 Ringer's lactate solution Substances 0.000 description 2
- PQCXVIPXISBFPN-UHFFFAOYSA-N SB 415286 Chemical compound C1=C(Cl)C(O)=CC=C1NC1=C(C=2C(=CC=CC=2)[N+]([O-])=O)C(=O)NC1=O PQCXVIPXISBFPN-UHFFFAOYSA-N 0.000 description 2
- 108010071390 Serum Albumin Proteins 0.000 description 2
- 102000007562 Serum Albumin Human genes 0.000 description 2
- 102100034204 Transcription factor SOX-9 Human genes 0.000 description 2
- 230000001594 aberrant effect Effects 0.000 description 2
- 239000012190 activator Substances 0.000 description 2
- 229940072056 alginate Drugs 0.000 description 2
- 229920000615 alginic acid Polymers 0.000 description 2
- 235000010443 alginic acid Nutrition 0.000 description 2
- 239000003242 anti bacterial agent Substances 0.000 description 2
- 229940088710 antibiotic agent Drugs 0.000 description 2
- 210000000941 bile Anatomy 0.000 description 2
- 201000005271 biliary atresia Diseases 0.000 description 2
- 230000015572 biosynthetic process Effects 0.000 description 2
- 230000000903 blocking effect Effects 0.000 description 2
- 229940098773 bovine serum albumin Drugs 0.000 description 2
- 239000007975 buffered saline Substances 0.000 description 2
- 230000015556 catabolic process Effects 0.000 description 2
- 230000010261 cell growth Effects 0.000 description 2
- 230000004663 cell proliferation Effects 0.000 description 2
- 238000005119 centrifugation Methods 0.000 description 2
- 238000012512 characterization method Methods 0.000 description 2
- 208000011444 chronic liver failure Diseases 0.000 description 2
- 230000000052 comparative effect Effects 0.000 description 2
- 239000000470 constituent Substances 0.000 description 2
- 238000005138 cryopreservation Methods 0.000 description 2
- 230000003247 decreasing effect Effects 0.000 description 2
- 230000002950 deficient Effects 0.000 description 2
- 210000001671 embryonic stem cell Anatomy 0.000 description 2
- 238000007459 endoscopic retrograde cholangiopancreatography Methods 0.000 description 2
- 230000002255 enzymatic effect Effects 0.000 description 2
- YQGOJNYOYNNSMM-UHFFFAOYSA-N eosin Chemical compound [Na+].OC(=O)C1=CC=CC=C1C1=C2C=C(Br)C(=O)C(Br)=C2OC2=C(Br)C(O)=C(Br)C=C21 YQGOJNYOYNNSMM-UHFFFAOYSA-N 0.000 description 2
- 210000002744 extracellular matrix Anatomy 0.000 description 2
- 239000000284 extract Substances 0.000 description 2
- 229950003499 fibrin Drugs 0.000 description 2
- 238000001914 filtration Methods 0.000 description 2
- 239000012909 foetal bovine serum Substances 0.000 description 2
- 238000007710 freezing Methods 0.000 description 2
- 230000008014 freezing Effects 0.000 description 2
- 210000000232 gallbladder Anatomy 0.000 description 2
- 210000001035 gastrointestinal tract Anatomy 0.000 description 2
- 239000000499 gel Substances 0.000 description 2
- 238000009396 hybridization Methods 0.000 description 2
- 210000002865 immune cell Anatomy 0.000 description 2
- 238000012744 immunostaining Methods 0.000 description 2
- 239000007943 implant Substances 0.000 description 2
- 238000011065 in-situ storage Methods 0.000 description 2
- 230000006698 induction Effects 0.000 description 2
- 230000000302 ischemic effect Effects 0.000 description 2
- 210000001865 kupffer cell Anatomy 0.000 description 2
- 239000003446 ligand Substances 0.000 description 2
- 150000002632 lipids Chemical class 0.000 description 2
- 208000007903 liver failure Diseases 0.000 description 2
- 231100000835 liver failure Toxicity 0.000 description 2
- 238000004020 luminiscence type Methods 0.000 description 2
- 210000004072 lung Anatomy 0.000 description 2
- 210000004698 lymphocyte Anatomy 0.000 description 2
- 230000035800 maturation Effects 0.000 description 2
- 210000003593 megakaryocyte Anatomy 0.000 description 2
- 210000004379 membrane Anatomy 0.000 description 2
- 239000012528 membrane Substances 0.000 description 2
- 230000004060 metabolic process Effects 0.000 description 2
- 239000011325 microbead Substances 0.000 description 2
- 238000012986 modification Methods 0.000 description 2
- 230000004048 modification Effects 0.000 description 2
- 208000008338 non-alcoholic fatty liver disease Diseases 0.000 description 2
- 239000003755 preservative agent Substances 0.000 description 2
- 238000000746 purification Methods 0.000 description 2
- XSCHRSMBECNVNS-UHFFFAOYSA-N quinoxaline Chemical compound N1=CC=NC2=CC=CC=C21 XSCHRSMBECNVNS-UHFFFAOYSA-N 0.000 description 2
- 238000011084 recovery Methods 0.000 description 2
- 230000003014 reinforcing effect Effects 0.000 description 2
- 238000011160 research Methods 0.000 description 2
- 238000003757 reverse transcription PCR Methods 0.000 description 2
- 208000010157 sclerosing cholangitis Diseases 0.000 description 2
- 238000010561 standard procedure Methods 0.000 description 2
- 238000007619 statistical method Methods 0.000 description 2
- 229920001059 synthetic polymer Polymers 0.000 description 2
- 231100000331 toxic Toxicity 0.000 description 2
- 230000002588 toxic effect Effects 0.000 description 2
- 230000002103 transcriptional effect Effects 0.000 description 2
- 230000014616 translation Effects 0.000 description 2
- 230000000472 traumatic effect Effects 0.000 description 2
- WZUVPPKBWHMQCE-XJKSGUPXSA-N (+)-haematoxylin Chemical compound C12=CC(O)=C(O)C=C2C[C@]2(O)[C@H]1C1=CC=C(O)C(O)=C1OC2 WZUVPPKBWHMQCE-XJKSGUPXSA-N 0.000 description 1
- UZOVYGYOLBIAJR-UHFFFAOYSA-N 4-isocyanato-4'-methyldiphenylmethane Chemical compound C1=CC(C)=CC=C1CC1=CC=C(N=C=O)C=C1 UZOVYGYOLBIAJR-UHFFFAOYSA-N 0.000 description 1
- DDLZLOKCJHBUHD-WAVHTBQISA-N 6-bromoindirubin-3'-oxime Chemical compound O=C/1NC2=CC(Br)=CC=C2C\1=C\1/C(=N/O)/C2=CC=CC=C2N/1 DDLZLOKCJHBUHD-WAVHTBQISA-N 0.000 description 1
- 101150092476 ABCA1 gene Proteins 0.000 description 1
- 208000030507 AIDS Diseases 0.000 description 1
- 101150005096 AKR1 gene Proteins 0.000 description 1
- 101150037123 APOE gene Proteins 0.000 description 1
- 102000055510 ATP Binding Cassette Transporter 1 Human genes 0.000 description 1
- 108700005241 ATP Binding Cassette Transporter 1 Proteins 0.000 description 1
- 102100036732 Actin, aortic smooth muscle Human genes 0.000 description 1
- 108010059616 Activins Proteins 0.000 description 1
- 102000005606 Activins Human genes 0.000 description 1
- 102100031831 Adipogenesis regulatory factor Human genes 0.000 description 1
- 208000022309 Alcoholic Liver disease Diseases 0.000 description 1
- 108010059886 Apolipoprotein A-I Proteins 0.000 description 1
- 102100030942 Apolipoprotein A-II Human genes 0.000 description 1
- 102100036451 Apolipoprotein C-I Human genes 0.000 description 1
- 102100030970 Apolipoprotein C-III Human genes 0.000 description 1
- 102100029470 Apolipoprotein E Human genes 0.000 description 1
- 101710200897 Asialoglycoprotein receptor 1 Proteins 0.000 description 1
- 102100027950 Bile acid-CoA:amino acid N-acyltransferase Human genes 0.000 description 1
- 102100023109 Bile acyl-CoA synthetase Human genes 0.000 description 1
- 206010004593 Bile duct cancer Diseases 0.000 description 1
- 102100024217 CAMPATH-1 antigen Human genes 0.000 description 1
- 108010017009 CD11b Antigen Proteins 0.000 description 1
- 102100036008 CD48 antigen Human genes 0.000 description 1
- 108010065524 CD52 Antigen Proteins 0.000 description 1
- MDZCSIDIPDZWKL-UHFFFAOYSA-N CHIR-98014 Chemical compound C1=C([N+]([O-])=O)C(N)=NC(NCCNC=2N=C(C(=CN=2)N2C=NC=C2)C=2C(=CC(Cl)=CC=2)Cl)=C1 MDZCSIDIPDZWKL-UHFFFAOYSA-N 0.000 description 1
- 102100025473 Carcinoembryonic antigen-related cell adhesion molecule 6 Human genes 0.000 description 1
- 102000016362 Catenins Human genes 0.000 description 1
- 108010067316 Catenins Proteins 0.000 description 1
- 101710010675 Cerberus Proteins 0.000 description 1
- 241000202252 Cerberus Species 0.000 description 1
- 208000008964 Chemical and Drug Induced Liver Injury Diseases 0.000 description 1
- 229920002101 Chitin Polymers 0.000 description 1
- 229920001661 Chitosan Polymers 0.000 description 1
- 206010008609 Cholangitis sclerosing Diseases 0.000 description 1
- VYZAMTAEIAYCRO-UHFFFAOYSA-N Chromium Chemical compound [Cr] VYZAMTAEIAYCRO-UHFFFAOYSA-N 0.000 description 1
- 208000005595 Chronic Idiopathic Jaundice Diseases 0.000 description 1
- 102100038449 Claudin-6 Human genes 0.000 description 1
- 102100029117 Coagulation factor X Human genes 0.000 description 1
- 206010053567 Coagulopathies Diseases 0.000 description 1
- 102000029816 Collagenase Human genes 0.000 description 1
- 108060005980 Collagenase Proteins 0.000 description 1
- 206010010356 Congenital anomaly Diseases 0.000 description 1
- 208000001819 Crigler-Najjar Syndrome Diseases 0.000 description 1
- 102100032857 Cyclin-dependent kinase 1 Human genes 0.000 description 1
- 101710106279 Cyclin-dependent kinase 1 Proteins 0.000 description 1
- 201000003883 Cystic fibrosis Diseases 0.000 description 1
- 102100036727 Deformed epidermal autoregulatory factor 1 homolog Human genes 0.000 description 1
- 101000802964 Dendroaspis angusticeps Muscarinic toxin 1 Proteins 0.000 description 1
- 206010072268 Drug-induced liver injury Diseases 0.000 description 1
- 201000004943 Dubin-Johnson syndrome Diseases 0.000 description 1
- 108010014258 Elastin Proteins 0.000 description 1
- 102000016942 Elastin Human genes 0.000 description 1
- 101000823089 Equus caballus Alpha-1-antiproteinase 1 Proteins 0.000 description 1
- LFQSCWFLJHTTHZ-UHFFFAOYSA-N Ethanol Chemical compound CCO LFQSCWFLJHTTHZ-UHFFFAOYSA-N 0.000 description 1
- 206010016077 Factor IX deficiency Diseases 0.000 description 1
- 206010016202 Familial Amyloidosis Diseases 0.000 description 1
- 102000009114 Fatty acid desaturases Human genes 0.000 description 1
- 108010087894 Fatty acid desaturases Proteins 0.000 description 1
- 102100026745 Fatty acid-binding protein, liver Human genes 0.000 description 1
- 102100024783 Fibrinogen gamma chain Human genes 0.000 description 1
- 102100028412 Fibroblast growth factor 10 Human genes 0.000 description 1
- 206010016654 Fibrosis Diseases 0.000 description 1
- 102000016970 Follistatin Human genes 0.000 description 1
- 108010014612 Follistatin Proteins 0.000 description 1
- 102000001267 GSK3 Human genes 0.000 description 1
- 108060006662 GSK3 Proteins 0.000 description 1
- 102100023448 GTP-binding protein 1 Human genes 0.000 description 1
- 101710102121 GTP-binding protein 1 Proteins 0.000 description 1
- 108020004206 Gamma-glutamyltransferase Proteins 0.000 description 1
- 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 1
- PEDCQBHIVMGVHV-UHFFFAOYSA-N Glycerine Chemical compound OCC(O)CO PEDCQBHIVMGVHV-UHFFFAOYSA-N 0.000 description 1
- 229920002527 Glycogen Polymers 0.000 description 1
- 102000003886 Glycoproteins Human genes 0.000 description 1
- 108090000288 Glycoproteins Proteins 0.000 description 1
- 229920002683 Glycosaminoglycan Polymers 0.000 description 1
- 208000018565 Hemochromatosis Diseases 0.000 description 1
- HTTJABKRGRZYRN-UHFFFAOYSA-N Heparin Chemical compound OC1C(NC(=O)C)C(O)OC(COS(O)(=O)=O)C1OC1C(OS(O)(=O)=O)C(O)C(OC2C(C(OS(O)(=O)=O)C(OC3C(C(O)C(O)C(O3)C(O)=O)OS(O)(=O)=O)C(CO)O2)NS(O)(=O)=O)C(C(O)=O)O1 HTTJABKRGRZYRN-UHFFFAOYSA-N 0.000 description 1
- 208000027761 Hepatic autoimmune disease Diseases 0.000 description 1
- 208000005176 Hepatitis C Diseases 0.000 description 1
- 208000005331 Hepatitis D Diseases 0.000 description 1
- 206010019773 Hepatitis G Diseases 0.000 description 1
- 102100022057 Hepatocyte nuclear factor 1-alpha Human genes 0.000 description 1
- 208000003591 Hepatoerythropoietic Porphyria Diseases 0.000 description 1
- 208000002972 Hepatolenticular Degeneration Diseases 0.000 description 1
- 241000282412 Homo Species 0.000 description 1
- 101000929319 Homo sapiens Actin, aortic smooth muscle Proteins 0.000 description 1
- 101000775473 Homo sapiens Adipogenesis regulatory factor Proteins 0.000 description 1
- 101000693913 Homo sapiens Albumin Proteins 0.000 description 1
- 101000793406 Homo sapiens Apolipoprotein A-II Proteins 0.000 description 1
- 101000928628 Homo sapiens Apolipoprotein C-I Proteins 0.000 description 1
- 101000793223 Homo sapiens Apolipoprotein C-III Proteins 0.000 description 1
- 101000697858 Homo sapiens Bile acid-CoA:amino acid N-acyltransferase Proteins 0.000 description 1
- 101000716130 Homo sapiens CD48 antigen Proteins 0.000 description 1
- 101000914326 Homo sapiens Carcinoembryonic antigen-related cell adhesion molecule 6 Proteins 0.000 description 1
- 101000914195 Homo sapiens Cerberus Proteins 0.000 description 1
- 101000882898 Homo sapiens Claudin-6 Proteins 0.000 description 1
- 101000929421 Homo sapiens Deformed epidermal autoregulatory factor 1 homolog Proteins 0.000 description 1
- 101000911317 Homo sapiens Fatty acid-binding protein, liver Proteins 0.000 description 1
- 101000846244 Homo sapiens Fibrinogen alpha chain Proteins 0.000 description 1
- 101001052043 Homo sapiens Fibrinogen gamma chain Proteins 0.000 description 1
- 101000917237 Homo sapiens Fibroblast growth factor 10 Proteins 0.000 description 1
- 101001045751 Homo sapiens Hepatocyte nuclear factor 1-alpha Proteins 0.000 description 1
- 101001078143 Homo sapiens Integrin alpha-IIb Proteins 0.000 description 1
- 101000975496 Homo sapiens Keratin, type II cytoskeletal 8 Proteins 0.000 description 1
- 101001091590 Homo sapiens Kininogen-1 Proteins 0.000 description 1
- 101001046587 Homo sapiens Krueppel-like factor 1 Proteins 0.000 description 1
- 101000777628 Homo sapiens Leukocyte antigen CD37 Proteins 0.000 description 1
- 101000608935 Homo sapiens Leukosialin Proteins 0.000 description 1
- 101000917858 Homo sapiens Low affinity immunoglobulin gamma Fc region receptor III-A Proteins 0.000 description 1
- 101000984710 Homo sapiens Lymphocyte-specific protein 1 Proteins 0.000 description 1
- 101000934372 Homo sapiens Macrosialin Proteins 0.000 description 1
- 101001027945 Homo sapiens Metallothionein-1E Proteins 0.000 description 1
- 101001027943 Homo sapiens Metallothionein-1F Proteins 0.000 description 1
- 101001013794 Homo sapiens Metallothionein-1H Proteins 0.000 description 1
- 101001013797 Homo sapiens Metallothionein-1L Proteins 0.000 description 1
- 101001013796 Homo sapiens Metallothionein-1M Proteins 0.000 description 1
- 101001014059 Homo sapiens Metallothionein-2 Proteins 0.000 description 1
- 101000934338 Homo sapiens Myeloid cell surface antigen CD33 Proteins 0.000 description 1
- 101001111320 Homo sapiens Nestin Proteins 0.000 description 1
- 101000973259 Homo sapiens Neurogranin Proteins 0.000 description 1
- 101000947178 Homo sapiens Platelet basic protein Proteins 0.000 description 1
- 101000928535 Homo sapiens Protein delta homolog 1 Proteins 0.000 description 1
- 101000738771 Homo sapiens Receptor-type tyrosine-protein phosphatase C Proteins 0.000 description 1
- 101000742932 Homo sapiens Retinol dehydrogenase 16 Proteins 0.000 description 1
- 101000884271 Homo sapiens Signal transducer CD24 Proteins 0.000 description 1
- 101000651211 Homo sapiens Transcription factor PU.1 Proteins 0.000 description 1
- 101000636213 Homo sapiens Transcriptional activator Myb Proteins 0.000 description 1
- 101000835093 Homo sapiens Transferrin receptor protein 1 Proteins 0.000 description 1
- 108091006905 Human Serum Albumin Proteins 0.000 description 1
- 102000008100 Human Serum Albumin Human genes 0.000 description 1
- 208000035150 Hypercholesterolemia Diseases 0.000 description 1
- 208000015229 Idiopathic ductopenia Diseases 0.000 description 1
- 102100025306 Integrin alpha-IIb Human genes 0.000 description 1
- 102100022338 Integrin alpha-M Human genes 0.000 description 1
- 102100023972 Keratin, type II cytoskeletal 8 Human genes 0.000 description 1
- 108010066302 Keratin-19 Proteins 0.000 description 1
- 102100035792 Kininogen-1 Human genes 0.000 description 1
- 102100022248 Krueppel-like factor 1 Human genes 0.000 description 1
- ZDXPYRJPNDTMRX-VKHMYHEASA-N L-glutamine Chemical compound OC(=O)[C@@H](N)CCC(N)=O ZDXPYRJPNDTMRX-VKHMYHEASA-N 0.000 description 1
- 229930182816 L-glutamine Natural products 0.000 description 1
- 102100031586 Leukocyte antigen CD37 Human genes 0.000 description 1
- 108090001030 Lipoproteins Proteins 0.000 description 1
- 102000004895 Lipoproteins Human genes 0.000 description 1
- 102100029193 Low affinity immunoglobulin gamma Fc region receptor III-A Human genes 0.000 description 1
- 102100027105 Lymphocyte-specific protein 1 Human genes 0.000 description 1
- 102100025136 Macrosialin Human genes 0.000 description 1
- 206010064912 Malignant transformation Diseases 0.000 description 1
- 102100037512 Metallothionein-1G Human genes 0.000 description 1
- 102100031781 Metallothionein-1X Human genes 0.000 description 1
- 102100031347 Metallothionein-2 Human genes 0.000 description 1
- 241001465754 Metazoa Species 0.000 description 1
- 102100025243 Myeloid cell surface antigen CD33 Human genes 0.000 description 1
- FABQUVYDAXWUQP-UHFFFAOYSA-N N4-(1,3-benzodioxol-5-ylmethyl)-6-(3-methoxyphenyl)pyrimidine-2,4-diamine Chemical compound COC1=CC=CC(C=2N=C(N)N=C(NCC=3C=C4OCOC4=CC=3)C=2)=C1 FABQUVYDAXWUQP-UHFFFAOYSA-N 0.000 description 1
- 206010028980 Neoplasm Diseases 0.000 description 1
- 102100022159 Neurogranin Human genes 0.000 description 1
- 101100215778 Neurospora crassa (strain ATCC 24698 / 74-OR23-1A / CBS 708.71 / DSM 1257 / FGSC 987) ptr-1 gene Proteins 0.000 description 1
- 208000008589 Obesity Diseases 0.000 description 1
- 229930040373 Paraformaldehyde Natural products 0.000 description 1
- ISWSIDIOOBJBQZ-UHFFFAOYSA-N Phenol Chemical compound OC1=CC=CC=C1 ISWSIDIOOBJBQZ-UHFFFAOYSA-N 0.000 description 1
- 102100038223 Phenylalanine-4-hydroxylase Human genes 0.000 description 1
- 101710125939 Phenylalanine-4-hydroxylase Proteins 0.000 description 1
- 102100036154 Platelet basic protein Human genes 0.000 description 1
- 102000018967 Platelet-Derived Growth Factor beta Receptor Human genes 0.000 description 1
- 108010051742 Platelet-Derived Growth Factor beta Receptor Proteins 0.000 description 1
- 239000002202 Polyethylene glycol Substances 0.000 description 1
- 102100036467 Protein delta homolog 1 Human genes 0.000 description 1
- 108010067787 Proteoglycans Proteins 0.000 description 1
- 102000016611 Proteoglycans Human genes 0.000 description 1
- 102100027378 Prothrombin Human genes 0.000 description 1
- CZPWVGJYEJSRLH-UHFFFAOYSA-N Pyrimidine Chemical compound C1=CN=CN=C1 CZPWVGJYEJSRLH-UHFFFAOYSA-N 0.000 description 1
- 238000003559 RNA-seq method Methods 0.000 description 1
- 239000012980 RPMI-1640 medium Substances 0.000 description 1
- 102100037422 Receptor-type tyrosine-protein phosphatase C Human genes 0.000 description 1
- 208000005587 Refsum Disease Diseases 0.000 description 1
- 102100038057 Retinol dehydrogenase 16 Human genes 0.000 description 1
- 108091006532 SLC27A5 Proteins 0.000 description 1
- 101150059736 SRY gene Proteins 0.000 description 1
- 206010039491 Sarcoma Diseases 0.000 description 1
- 102100038081 Signal transducer CD24 Human genes 0.000 description 1
- 238000000692 Student's t-test Methods 0.000 description 1
- QAOWNCQODCNURD-UHFFFAOYSA-L Sulfate Chemical compound [O-]S([O-])(=O)=O QAOWNCQODCNURD-UHFFFAOYSA-L 0.000 description 1
- 108060008245 Thrombospondin Proteins 0.000 description 1
- 102000002938 Thrombospondin Human genes 0.000 description 1
- 102100027654 Transcription factor PU.1 Human genes 0.000 description 1
- 102100030780 Transcriptional activator Myb Human genes 0.000 description 1
- 102100026144 Transferrin receptor protein 1 Human genes 0.000 description 1
- 102100030742 Transforming growth factor beta-1 proprotein Human genes 0.000 description 1
- 229920004890 Triton X-100 Polymers 0.000 description 1
- 239000013504 Triton X-100 Substances 0.000 description 1
- 206010054753 Vanishing bile duct syndrome Diseases 0.000 description 1
- 241000251539 Vertebrata <Metazoa> Species 0.000 description 1
- ITBGJNVZJBVPLJ-UHFFFAOYSA-N WAY-316606 Chemical compound FC(F)(F)C1=CC=C(S(=O)(=O)C=2C=CC=CC=2)C=C1S(=O)(=O)NC1CCNCC1 ITBGJNVZJBVPLJ-UHFFFAOYSA-N 0.000 description 1
- 208000018839 Wilson disease Diseases 0.000 description 1
- 108091035715 XIST (gene) Proteins 0.000 description 1
- 210000000683 abdominal cavity Anatomy 0.000 description 1
- 108091006088 activator proteins Proteins 0.000 description 1
- 239000000488 activin Substances 0.000 description 1
- 210000000577 adipose tissue Anatomy 0.000 description 1
- 208000030597 adult Refsum disease Diseases 0.000 description 1
- 210000004504 adult stem cell Anatomy 0.000 description 1
- 239000000556 agonist Substances 0.000 description 1
- 208000006682 alpha 1-Antitrypsin Deficiency Diseases 0.000 description 1
- OLUKILHGKRVDCT-UHFFFAOYSA-N alsterpaullone Chemical compound C1C(=O)NC2=CC=CC=C2C2=C1C1=CC([N+](=O)[O-])=CC=C1N2 OLUKILHGKRVDCT-UHFFFAOYSA-N 0.000 description 1
- 210000004102 animal cell Anatomy 0.000 description 1
- 239000010775 animal oil Substances 0.000 description 1
- 239000000427 antigen Substances 0.000 description 1
- 108091007433 antigens Proteins 0.000 description 1
- 102000036639 antigens Human genes 0.000 description 1
- 239000003963 antioxidant agent Substances 0.000 description 1
- 230000003078 antioxidant effect Effects 0.000 description 1
- 235000006708 antioxidants Nutrition 0.000 description 1
- 230000006907 apoptotic process Effects 0.000 description 1
- 239000012062 aqueous buffer Substances 0.000 description 1
- 239000007864 aqueous solution Substances 0.000 description 1
- QVGXLLKOCUKJST-UHFFFAOYSA-N atomic oxygen Chemical compound [O] QVGXLLKOCUKJST-UHFFFAOYSA-N 0.000 description 1
- 230000001363 autoimmune Effects 0.000 description 1
- 208000021033 autosomal dominant polycystic liver disease Diseases 0.000 description 1
- 239000011324 bead Substances 0.000 description 1
- 230000008901 benefit Effects 0.000 description 1
- KMGARVOVYXNAOF-UHFFFAOYSA-N benzpiperylone Chemical compound C1CN(C)CCC1N1C(=O)C(CC=2C=CC=CC=2)=C(C=2C=CC=CC=2)N1 KMGARVOVYXNAOF-UHFFFAOYSA-N 0.000 description 1
- 125000000440 benzylamino group Chemical group [H]N(*)C([H])([H])C1=C([H])C([H])=C([H])C([H])=C1[H] 0.000 description 1
- 239000003613 bile acid Substances 0.000 description 1
- 239000012472 biological sample Substances 0.000 description 1
- 210000000988 bone and bone Anatomy 0.000 description 1
- 210000004556 brain Anatomy 0.000 description 1
- 238000009107 bridge therapy Methods 0.000 description 1
- BPKIGYQJPYCAOW-FFJTTWKXSA-I calcium;potassium;disodium;(2s)-2-hydroxypropanoate;dichloride;dihydroxide;hydrate Chemical compound O.[OH-].[OH-].[Na+].[Na+].[Cl-].[Cl-].[K+].[Ca+2].C[C@H](O)C([O-])=O BPKIGYQJPYCAOW-FFJTTWKXSA-I 0.000 description 1
- BMLSTPRTEKLIPM-UHFFFAOYSA-I calcium;potassium;disodium;hydrogen carbonate;dichloride;dihydroxide;hydrate Chemical compound O.[OH-].[OH-].[Na+].[Na+].[Cl-].[Cl-].[K+].[Ca+2].OC([O-])=O BMLSTPRTEKLIPM-UHFFFAOYSA-I 0.000 description 1
- 201000011510 cancer Diseases 0.000 description 1
- 150000001720 carbohydrates Chemical class 0.000 description 1
- 229910002092 carbon dioxide Inorganic materials 0.000 description 1
- 239000001569 carbon dioxide Substances 0.000 description 1
- 239000000969 carrier Substances 0.000 description 1
- 108700021031 cdc Genes Proteins 0.000 description 1
- 238000012832 cell culture technique Methods 0.000 description 1
- 230000022131 cell cycle Effects 0.000 description 1
- 230000011712 cell development Effects 0.000 description 1
- 230000024245 cell differentiation Effects 0.000 description 1
- 239000002771 cell marker Substances 0.000 description 1
- 238000002659 cell therapy Methods 0.000 description 1
- 229920002678 cellulose Polymers 0.000 description 1
- 239000001913 cellulose Substances 0.000 description 1
- 210000001175 cerebrospinal fluid Anatomy 0.000 description 1
- 238000013189 cholangiography Methods 0.000 description 1
- 229910052804 chromium Inorganic materials 0.000 description 1
- 239000011651 chromium Substances 0.000 description 1
- 230000007882 cirrhosis Effects 0.000 description 1
- 208000019425 cirrhosis of liver Diseases 0.000 description 1
- 230000035602 clotting Effects 0.000 description 1
- 238000003501 co-culture Methods 0.000 description 1
- 230000004186 co-expression Effects 0.000 description 1
- AGVAZMGAQJOSFJ-WZHZPDAFSA-M cobalt(2+);[(2r,3s,4r,5s)-5-(5,6-dimethylbenzimidazol-1-yl)-4-hydroxy-2-(hydroxymethyl)oxolan-3-yl] [(2r)-1-[3-[(1r,2r,3r,4z,7s,9z,12s,13s,14z,17s,18s,19r)-2,13,18-tris(2-amino-2-oxoethyl)-7,12,17-tris(3-amino-3-oxopropyl)-3,5,8,8,13,15,18,19-octamethyl-2 Chemical compound [Co+2].N#[C-].[N-]([C@@H]1[C@H](CC(N)=O)[C@@]2(C)CCC(=O)NC[C@@H](C)OP(O)(=O)O[C@H]3[C@H]([C@H](O[C@@H]3CO)N3C4=CC(C)=C(C)C=C4N=C3)O)\C2=C(C)/C([C@H](C\2(C)C)CCC(N)=O)=N/C/2=C\C([C@H]([C@@]/2(CC(N)=O)C)CCC(N)=O)=N\C\2=C(C)/C2=N[C@]1(C)[C@@](C)(CC(N)=O)[C@@H]2CCC(N)=O AGVAZMGAQJOSFJ-WZHZPDAFSA-M 0.000 description 1
- 229960002424 collagenase Drugs 0.000 description 1
- 230000000295 complement effect Effects 0.000 description 1
- 239000002299 complementary DNA Substances 0.000 description 1
- 239000003636 conditioned culture medium Substances 0.000 description 1
- 230000008094 contradictory effect Effects 0.000 description 1
- 238000012937 correction Methods 0.000 description 1
- 210000004748 cultured cell Anatomy 0.000 description 1
- 235000018417 cysteine Nutrition 0.000 description 1
- XUJNEKJLAYXESH-UHFFFAOYSA-N cysteine Natural products SCC(N)C(O)=O XUJNEKJLAYXESH-UHFFFAOYSA-N 0.000 description 1
- 230000007812 deficiency Effects 0.000 description 1
- 238000006731 degradation reaction Methods 0.000 description 1
- 210000004443 dendritic cell Anatomy 0.000 description 1
- 238000001514 detection method Methods 0.000 description 1
- 239000008121 dextrose Substances 0.000 description 1
- 230000029087 digestion Effects 0.000 description 1
- 239000003085 diluting agent Substances 0.000 description 1
- 239000012153 distilled water Substances 0.000 description 1
- 238000009826 distribution Methods 0.000 description 1
- 230000003828 downregulation Effects 0.000 description 1
- 238000007876 drug discovery Methods 0.000 description 1
- 101150036810 eco gene Proteins 0.000 description 1
- 229920002549 elastin Polymers 0.000 description 1
- 210000002257 embryonic structure Anatomy 0.000 description 1
- 210000002889 endothelial cell Anatomy 0.000 description 1
- 229920005558 epichlorohydrin rubber Polymers 0.000 description 1
- 210000002919 epithelial cell Anatomy 0.000 description 1
- 125000001495 ethyl group Chemical group [H]C([H])([H])C([H])([H])* 0.000 description 1
- NGOGFTYYXHNFQH-UHFFFAOYSA-N fasudil Chemical compound C=1C=CC2=CN=CC=C2C=1S(=O)(=O)N1CCCNCC1 NGOGFTYYXHNFQH-UHFFFAOYSA-N 0.000 description 1
- 229960002435 fasudil Drugs 0.000 description 1
- 238000013467 fragmentation Methods 0.000 description 1
- 238000006062 fragmentation reaction Methods 0.000 description 1
- 238000004108 freeze drying Methods 0.000 description 1
- 102000006640 gamma-Glutamyltransferase Human genes 0.000 description 1
- 239000003365 glass fiber Substances 0.000 description 1
- 150000004676 glycans Chemical class 0.000 description 1
- 229940096919 glycogen Drugs 0.000 description 1
- 208000007345 glycogen storage disease Diseases 0.000 description 1
- 208000011460 glycogen storage disease due to glucose-6-phosphatase deficiency type IA Diseases 0.000 description 1
- 150000002334 glycols Chemical class 0.000 description 1
- 230000005484 gravity Effects 0.000 description 1
- 230000003394 haemopoietic effect Effects 0.000 description 1
- 238000003306 harvesting Methods 0.000 description 1
- 238000010438 heat treatment Methods 0.000 description 1
- 210000003958 hematopoietic stem cell Anatomy 0.000 description 1
- 208000009429 hemophilia B Diseases 0.000 description 1
- 229920000669 heparin Polymers 0.000 description 1
- 229960002897 heparin Drugs 0.000 description 1
- 208000006454 hepatitis Diseases 0.000 description 1
- 231100000283 hepatitis Toxicity 0.000 description 1
- 208000005252 hepatitis A Diseases 0.000 description 1
- 208000002672 hepatitis B Diseases 0.000 description 1
- 201000010284 hepatitis E Diseases 0.000 description 1
- 206010073071 hepatocellular carcinoma Diseases 0.000 description 1
- 231100000844 hepatocellular carcinoma Toxicity 0.000 description 1
- 125000005842 heteroatom Chemical group 0.000 description 1
- 230000005745 host immune response Effects 0.000 description 1
- 102000044814 human ALB Human genes 0.000 description 1
- 102000056844 human CER1 Human genes 0.000 description 1
- 210000005260 human cell Anatomy 0.000 description 1
- 230000000642 iatrogenic effect Effects 0.000 description 1
- 238000003384 imaging method Methods 0.000 description 1
- 230000001900 immune effect Effects 0.000 description 1
- 230000036039 immunity Effects 0.000 description 1
- 238000003119 immunoblot Methods 0.000 description 1
- 238000003365 immunocytochemistry Methods 0.000 description 1
- 238000010166 immunofluorescence Methods 0.000 description 1
- 230000005847 immunogenicity Effects 0.000 description 1
- 238000002513 implantation Methods 0.000 description 1
- 238000010874 in vitro model Methods 0.000 description 1
- 238000010921 in-depth analysis Methods 0.000 description 1
- 208000016245 inborn errors of metabolism Diseases 0.000 description 1
- 238000011534 incubation Methods 0.000 description 1
- 208000015978 inherited metabolic disease Diseases 0.000 description 1
- 230000010354 integration Effects 0.000 description 1
- 230000003993 interaction Effects 0.000 description 1
- 210000004966 intestinal stem cell Anatomy 0.000 description 1
- 229910052742 iron Inorganic materials 0.000 description 1
- 238000002955 isolation Methods 0.000 description 1
- QQUXFYAWXPMDOE-UHFFFAOYSA-N kenpaullone Chemical compound C1C(=O)NC2=CC=CC=C2C2=C1C1=CC(Br)=CC=C1N2 QQUXFYAWXPMDOE-UHFFFAOYSA-N 0.000 description 1
- 210000003734 kidney Anatomy 0.000 description 1
- 230000000670 limiting effect Effects 0.000 description 1
- 210000002540 macrophage Anatomy 0.000 description 1
- 238000012423 maintenance Methods 0.000 description 1
- 230000036212 malign transformation Effects 0.000 description 1
- 210000001161 mammalian embryo Anatomy 0.000 description 1
- 210000005075 mammary gland Anatomy 0.000 description 1
- 238000004949 mass spectrometry Methods 0.000 description 1
- 239000012577 media supplement Substances 0.000 description 1
- 230000007102 metabolic function Effects 0.000 description 1
- 230000037353 metabolic pathway Effects 0.000 description 1
- 125000002496 methyl group Chemical group [H]C([H])([H])* 0.000 description 1
- 230000003278 mimic effect Effects 0.000 description 1
- 230000004079 mineral homeostasis Effects 0.000 description 1
- 239000002480 mineral oil Substances 0.000 description 1
- 235000010446 mineral oil Nutrition 0.000 description 1
- 238000002156 mixing Methods 0.000 description 1
- 230000001002 morphogenetic effect Effects 0.000 description 1
- 238000010172 mouse model Methods 0.000 description 1
- NFVJNJQRWPQVOA-UHFFFAOYSA-N n-[2-chloro-5-(trifluoromethyl)phenyl]-2-[3-(4-ethyl-5-ethylsulfanyl-1,2,4-triazol-3-yl)piperidin-1-yl]acetamide Chemical compound CCN1C(SCC)=NN=C1C1CN(CC(=O)NC=2C(=CC=C(C=2)C(F)(F)F)Cl)CCC1 NFVJNJQRWPQVOA-UHFFFAOYSA-N 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
- 229910052757 nitrogen Inorganic materials 0.000 description 1
- 102000045246 noggin Human genes 0.000 description 1
- 108700007229 noggin Proteins 0.000 description 1
- 231100000252 nontoxic Toxicity 0.000 description 1
- 230000003000 nontoxic effect Effects 0.000 description 1
- 238000012758 nuclear staining Methods 0.000 description 1
- 108020004707 nucleic acids Proteins 0.000 description 1
- 102000039446 nucleic acids Human genes 0.000 description 1
- 230000000050 nutritive effect Effects 0.000 description 1
- 235000020824 obesity Nutrition 0.000 description 1
- 239000003921 oil Substances 0.000 description 1
- 235000019198 oils Nutrition 0.000 description 1
- 239000001301 oxygen Substances 0.000 description 1
- 229910052760 oxygen Inorganic materials 0.000 description 1
- 210000000496 pancreas Anatomy 0.000 description 1
- 101150093826 par1 gene Proteins 0.000 description 1
- 229920002866 paraformaldehyde Polymers 0.000 description 1
- 210000004738 parenchymal cell Anatomy 0.000 description 1
- 230000008506 pathogenesis Effects 0.000 description 1
- 230000001575 pathological effect Effects 0.000 description 1
- 210000003200 peritoneal cavity Anatomy 0.000 description 1
- 239000003208 petroleum Substances 0.000 description 1
- 230000035479 physiological effects, processes and functions Effects 0.000 description 1
- 230000035790 physiological processes and functions Effects 0.000 description 1
- 239000002504 physiological saline solution Substances 0.000 description 1
- INAAIJLSXJJHOZ-UHFFFAOYSA-N pibenzimol Chemical compound C1CN(C)CCN1C1=CC=C(N=C(N2)C=3C=C4NC(=NC4=CC=3)C=3C=CC(O)=CC=3)C2=C1 INAAIJLSXJJHOZ-UHFFFAOYSA-N 0.000 description 1
- 210000001778 pluripotent stem cell Anatomy 0.000 description 1
- 208000028589 polycystic liver disease Diseases 0.000 description 1
- 229920000728 polyester Polymers 0.000 description 1
- 229920001223 polyethylene glycol Polymers 0.000 description 1
- 239000004626 polylactic acid Substances 0.000 description 1
- 229920001282 polysaccharide Polymers 0.000 description 1
- 239000005017 polysaccharide Substances 0.000 description 1
- 210000003240 portal vein Anatomy 0.000 description 1
- 230000002980 postoperative effect Effects 0.000 description 1
- 238000001556 precipitation Methods 0.000 description 1
- 230000002335 preservative effect Effects 0.000 description 1
- 125000002924 primary amino group Chemical group [H]N([H])* 0.000 description 1
- 201000000742 primary sclerosing cholangitis Diseases 0.000 description 1
- 230000000750 progressive effect Effects 0.000 description 1
- 230000002035 prolonged effect Effects 0.000 description 1
- 238000011321 prophylaxis Methods 0.000 description 1
- 210000002307 prostate Anatomy 0.000 description 1
- 229940039716 prothrombin Drugs 0.000 description 1
- 238000011002 quantification Methods 0.000 description 1
- 102000005962 receptors Human genes 0.000 description 1
- 108020003175 receptors Proteins 0.000 description 1
- 238000010188 recombinant method Methods 0.000 description 1
- 108010054624 red fluorescent protein Proteins 0.000 description 1
- 230000008929 regeneration Effects 0.000 description 1
- 238000011069 regeneration method Methods 0.000 description 1
- 230000014493 regulation of gene expression Effects 0.000 description 1
- 230000001105 regulatory effect Effects 0.000 description 1
- 230000003938 response to stress Effects 0.000 description 1
- 230000002441 reversible effect Effects 0.000 description 1
- 102000000568 rho-Associated Kinases Human genes 0.000 description 1
- 108010041788 rho-Associated Kinases Proteins 0.000 description 1
- 150000003839 salts Chemical class 0.000 description 1
- 238000007790 scraping Methods 0.000 description 1
- 238000000926 separation method Methods 0.000 description 1
- 239000011780 sodium chloride Substances 0.000 description 1
- NTHWMYGWWRZVTN-UHFFFAOYSA-N sodium silicate Chemical compound [Na+].[Na+].[O-][Si]([O-])=O NTHWMYGWWRZVTN-UHFFFAOYSA-N 0.000 description 1
- 239000007787 solid Substances 0.000 description 1
- 238000001228 spectrum Methods 0.000 description 1
- 210000000952 spleen Anatomy 0.000 description 1
- 239000003381 stabilizer Substances 0.000 description 1
- 238000010186 staining Methods 0.000 description 1
- 210000002536 stromal cell Anatomy 0.000 description 1
- 229910021653 sulphate ion Inorganic materials 0.000 description 1
- 239000013589 supplement Substances 0.000 description 1
- 230000004083 survival effect Effects 0.000 description 1
- 239000000725 suspension Substances 0.000 description 1
- 208000011580 syndromic disease Diseases 0.000 description 1
- 238000003786 synthesis reaction Methods 0.000 description 1
- 238000012353 t test Methods 0.000 description 1
- 230000002123 temporal effect Effects 0.000 description 1
- 238000010257 thawing Methods 0.000 description 1
- 210000001578 tight junction Anatomy 0.000 description 1
- 239000003104 tissue culture media Substances 0.000 description 1
- 208000037816 tissue injury Diseases 0.000 description 1
- 231100000419 toxicity Toxicity 0.000 description 1
- 230000001988 toxicity Effects 0.000 description 1
- 231100000041 toxicology testing Toxicity 0.000 description 1
- 230000026683 transduction Effects 0.000 description 1
- 238000010361 transduction Methods 0.000 description 1
- 239000010981 turquoise Substances 0.000 description 1
- 201000011296 tyrosinemia Diseases 0.000 description 1
- 210000003606 umbilical vein Anatomy 0.000 description 1
- 210000005166 vasculature Anatomy 0.000 description 1
- 235000015112 vegetable and seed oil Nutrition 0.000 description 1
- 239000008158 vegetable oil Substances 0.000 description 1
- 238000012800 visualization Methods 0.000 description 1
- 235000013343 vitamin Nutrition 0.000 description 1
- 239000011782 vitamin Substances 0.000 description 1
- 229940088594 vitamin Drugs 0.000 description 1
- 229930003231 vitamin Natural products 0.000 description 1
- 150000003722 vitamin derivatives Chemical class 0.000 description 1
- 239000002699 waste material Substances 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
- C12N5/067—Hepatocytes
- C12N5/0671—Three-dimensional culture, tissue culture or organ culture; Encapsulated 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
- C12N5/06—Animal cells or tissues; Human cells or tissues
- C12N5/0602—Vertebrate cells
- C12N5/067—Hepatocytes
-
- 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
- C12N2501/11—Epidermal growth factor [EGF]
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2501/00—Active agents used in cell culture processes, e.g. differentation
- C12N2501/10—Growth factors
- C12N2501/15—Transforming growth factor beta (TGF-β)
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2501/00—Active agents used in cell culture processes, e.g. differentation
- C12N2501/40—Regulators of development
- C12N2501/415—Wnt; Frizzeled
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2501/00—Active agents used in cell culture processes, e.g. differentation
- C12N2501/70—Enzymes
- C12N2501/72—Transferases [EC 2.]
- C12N2501/727—Kinases (EC 2.7.)
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2501/00—Active agents used in cell culture processes, e.g. differentation
- C12N2501/999—Small molecules not provided for elsewhere
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2513/00—3D culture
-
- 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
- C12N2533/90—Substrates of biological origin, e.g. extracellular matrix, decellularised tissue
Definitions
- a sixth aspect provides a method of treating a liver disease comprising administering an isolated population of the fourth aspect or a scaffold of the fifth aspect to an individual in need thereof.
- a non-canonical Wnt signalling potentiator is a compound that stimulates, promotes or increases the activity of the non-canonical Wnt signalling pathway.
- the non- canonical Wnt signalling pathway is a b-catenin- independent pathway involved in tissue polarity and morphogenetic processes in vertebrates (Komiya, Y. & Habas, R. Organogenesis 4, 68-75 (2008); Patel, V. et ai . Hum. Mol. Genet. 17, 1578-1590 (2008);
- Hepatoblasts expanded as described herein may be cultured or maintained using standard mammalian cell culture techniques or subjected to further manipulation or processing.
- the cell populations produced as described herein may be stored, for example by lyophilisation and/or
- Hepatoblasts, hepatocytes or cholangiocytes for use in modelling and screening may be in the form of organoids (hepatoblast organoids, HOs), sub-organoid clusters or individual cells (hepatoblasts) produced, for example by disruption of organoids.
- organoids hepatoblast organoids, HOs
- sub-organoid clusters or individual cells (hepatoblasts) produced, for example by disruption of organoids.
- the one or more media may be a 1x formulation or a more concentrated formulation, e.g. a 2x to 250x concentrated medium formulation.
- a 1x formulation each ingredient in the medium is at the concentration intended for cell culture, for example a concentration set out above.
- a concentrated formulation one or more of the ingredients is present at a higher concentration than intended for cell culture.
- Concentrated culture media are well known in the art. Culture media can be concentrated using known methods e.g. salt precipitation or selective filtration.
- a concentrated medium may be diluted for use with water (preferably deionized and distilled) or any appropriate solution, e.g. an aqueous saline solution, an aqueous buffer or a culture medium.
- the single cell suspension was resuspended in the hepatoblast organoid media (HO-M) for liver samples.
- HO-M hepatoblast organoid media
Landscapes
- Health & Medical Sciences (AREA)
- Engineering & Computer Science (AREA)
- Biomedical Technology (AREA)
- Life Sciences & Earth Sciences (AREA)
- Biotechnology (AREA)
- Genetics & Genomics (AREA)
- Zoology (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Organic Chemistry (AREA)
- Chemical & Material Sciences (AREA)
- Wood Science & Technology (AREA)
- Microbiology (AREA)
- Cell Biology (AREA)
- Biochemistry (AREA)
- General Engineering & Computer Science (AREA)
- General Health & Medical Sciences (AREA)
- Gastroenterology & Hepatology (AREA)
- Micro-Organisms Or Cultivation Processes Thereof (AREA)
- Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
Abstract
This invention relates to culture methods that allow the efficient long-term expansion of human hepatoblasts in the form of organoids. The methods comprise culturing primary immature human liver cells in a hepatoblast expansion medium comprising epidermal growth factor (EGF), a ΤΟΓβ inhibitor, a non-canonical Wnt signalling potentiator, a canonical Wnt potentiator and a ROCK inhibitor, to produce an expanded population of human hepatoblasts. The human hepatoblasts are bipotent and may be further differentiated into hepatocytes and cholangiocytes. Culture methods, cell populations and uses thereof are provided.
Description
Hepatoblast Organoids
Field
The present invention relates to the in vitro generation of organoids comprising hepatoblasts, for example for use in modelling liver disease or development, drug screening and regenerative medicine.
Background
The liver is unique as an organ in the broad spectrum of its functions during adult life (Gille et al., 2010). These functions include iron, vitamin and mineral homeostasis, and the detoxification of alcohol, drugs and other chemicals circulating in the bloodstream. The liver also synthesises bile for the digestion of fat and secretes blood clotting factors and serum proteins such as albumin (Alb) that represents the most abundant protein in the plasma. Finally, the liver plays an essential metabolic activity by storing glycogen and lipids (Gille et al., 2010). Most of these activities are managed by hepatocytes which constitute over 80% of the liver mass (Blouin, Bolender and Weibel, 1977). The cholangiocytes from the biliary tree also have an essential role in collecting waste, processing bile acids and possibly in hepatic regeneration after injury (Deng et al., 2018). Importantly, disorders affecting functions of these two cell types can be life-threatening and organ transplantation remains the only treatment for end stage liver diseases. However, the number of organ donors has remained constant for the past 10 years while the demand for liver transplantation has more than doubled in the meantime (Hopkinson and Allen, 2017). This situation is likely to worsen in the foreseeable future due to the pandemic of liver disease associated with obesity and non-alcoholic fatty liver disease (Younossi et al., 2016). Importantly, understanding liver organogenesis, especially the mechanisms by which hepatocytes and cholangiocytes are generated during development, could help to develop alternative regenerative approach such as cell-based therapies and also in vitro systems for disease modelling and drug screening.
Studies in the mouse and human have clearly established that hepatocytes and cholangiocytes originate from bi-potential progenitors named hepatoblasts (Yang et al., 2017)(Shiojiri et al., 2001 ). More precisely, hepatoblasts represent a proliferative population first detected in the liver bud at 5 weeks post conception in humans. These progenitors persist until 20 weeks of development playing a crucial role in liver
organogenesis. They are characterised by their capacity to express markers specific for both hepatocytes and cholangiocytes such as Epithelial Cell Adhesion Molecule (EPCAM), Albumin and Alpha-Fetoprotein (AFP). Importantly, the signalling pathways involved in hepatoblast self-renewal and differentiation remain to be fully uncovered as contradicting reports have suggested that Wnt signalling could block or promote differentiation of hepatoblasts toward hepatocytes and cholangiocytes in the mouse embryo (Decaens et al., 2008; Tan et al., 2008). On the other hand, TGF has been shown to direct differentiation of hepatoblasts toward the biliary lineage. However, such mechanisms have not been confirmed in human and other pathways may be involved (Clotman et al., 2005). Of note, transplantation of isolated human hepatoblasts have shown that these cells can colonise the rat adult liver in hepatic failure models thereby demonstrating their interest for regenerative medicine applications (Oertel et al., 2003). Thus, derivation of human hepatoblasts may provide a unique platform not only to study human liver development but also to produce cells with clinical interests. However, the development of robust protocols to grow human hepatoblasts in vitro have remained elusive, thereby limiting the study and utilisation of these progenitors.
Organoid technology consists of growing stem cells, and more broadly a combination of progenitor and epithelial cells, in three-dimensional culture conditions. This technology was first developed using intestinal stem cells (Sato et al., 201 1 ) and then was rapidly applied to a diversity of adult organs including the liver, pancreas, prostate, mammary gland, biliary three and lungs (Huch and Koo, 2015). In addition, similar culture conditions have been combined with differentiation of human Pluripotent Stem Cells (hPSCs) to generate brain, kidney, gut and liver organoids which mimic the cellular complexity and architecture of native organs. The interest of organoids has been broadly demonstrated for basic studies, disease modelling (Schwank et al., 2013), drug screening (van de Wetering et al., 2015), and also regenerative medicine applications (Cruz-Acuha et al., 2017). Nonetheless, this technology has been more rarely applied to foetal tissues in the context of developmental studies. Indeed, primary organoids have only been derived so far from the foetal gut and lung tips. Interestingly, the resulting cells display capacity of differentiation in vitro (Kraiczy et al., 2017) and in vivo after co-culture with mesenchymal cells (Yui et al., 2018).
The existence of liver stem cells in the adult liver has been reported (Huch et al 2015, Hu et al 2018).
However, the resulting organoids maintain a strong biliary identity. More recently, human fetal hepatocyte organoids have been derived from livers of the second trimester. However, these cells are committed to a hepatocytic fate and no longer have the capacity to differentiate into cholangiocytes.
Summary
The present inventors have developed culture methods that allow the efficient long-term expansion of human hepatoblasts in the form of organoids. Populations of hepatoblasts expanded as described herein may be differentiated into hepatocytes and cholangiocytes and may be useful for example in regenerative medicine.
A first aspect of the invention provides a method for producing an expanded population of human hepatoblasts in vitro comprising:
(i) providing a population of primary immature human liver cells and;
(ii) culturing the population in a hepatoblast expansion medium comprising epidermal growth factor (EGF), a TGF inhibitor, a non-canonical Wnt signalling potentiator, a canonical Wnt potentiator and a ROCK inhibitor, to produce an expanded population of human hepatoblasts.
Preferably, the immature human liver cells are cultured in three-dimensional culture in the expansion medium. The expanded populations of hepatoblasts may form organoids in the expansion medium.
In some embodiments, the method may further comprise disrupting the organoids to produce a population of isolated hepatoblasts. The isolated hepatoblasts may be further cultured in the expansion medium to expand or propagate the population.
The human hepatoblasts produced by the method of the first aspect are bipotent and are capable of differentiation into cholangiocytes and hepatocytes.
A second aspect of the invention provides a method for producing a population of human cholangiocytes in vitro comprising
(i) producing an expanded population of human hepatoblasts by a method according to the first aspect; and,
(ii) culturing the expanded population in a differentiation medium comprising TGF to produce a population of human cholangiocytes.
A third aspect of the invention provides a method for producing a population of human hepatocytes in vitro comprising
(i) producing an expanded population of hepatoblasts by a method according to the first aspect; and,
(ii) culturing the expanded population in a hepatocyte culture medium to produce a population of human hepatocytes.
A fourth aspect of the invention provides an isolated population of human hepatoblasts, hepatocytes or cholangiocytes produced by a method according to any one of the first to third aspects.
A fifth aspect of the invention provides a biocompatible scaffold comprising an isolated population of the fourth aspect.
A sixth aspect provides a method of treating a liver disease comprising administering an isolated population of the fourth aspect or a scaffold of the fifth aspect to an individual in need thereof.
A seventh aspect of the invention provides a method of screening comprising;
contacting an isolated population of the fourth aspect or a scaffold of the fifth aspect with a test compound, and;
determining the effect of the test compound on the population or scaffold and/or the effect of the population or scaffold on the test compound.
Preferably, the population contacted with the test compound is in the form of organoids.
An eighth aspect of the invention provides a kit for the production of an expanded population of hepatoblasts comprising a hepatoblast expansion medium that comprises epidermal growth factor (EGF), a TGF inhibitor, non-canonical Wnt signalling potentiator, a canonical Wnt potentiator and a ROCK inhibitor.
The kit may further comprise a hepatocyte culture medium and/or a differentiation medium comprising TGF .
Other aspects and embodiments of the invention are described in more detail below.
Brief Description of the Figures
Figure 1 shows the basic characterisation of Hepatoblast Organoid (HO). A) qPCR gene expression data comparing HO (n=25, p3-15, 9 donors) FBO (n=9, p3-15, 2 donors), PFL (n=3 donors) and PAH (n = 6, 3 donors, 6 plates) for the genes listed above. Expression is relative to housekeeping gene RPLP-0, mean+/- SEM. B) Representative brightfield images of HO (scale bar = 400uM, top, 200uM, bottom). C)
Immunofluorescent staining of HO for HNF4a (green, top left), AFP (red, top right), ALB (yellow, bottom left),
and overlay image (bottom right). Nuclear staining (blue) was performed using Hoechst dye. D)
Concentration of secreted proteins in the culture medium of HO (n=16, 1 1 donors, p2-15) and FBO (n=4, p11-15, 2 donors) after 48 hours of freshly applied medium as detected by ELISA and normalised to cell number. E) CYP3A5/7 and CYP3A4 activity measured by chemiluminescent assay (relative luminescence units, RLU) with luciferin PFBE (top) and IPA (bottom), normalised to cell number for HO (n=3, p14), FBO, (n=2, p14), and PAH (n=6).
Figure 2 shows the characterisation of Primary Foetal Liver (PFL). Sections of an 8 post-conceptional week (pew) primary foetal liver, stained using haematoxylin and eosin (H&E, far left), immunohistochemistry for AFP (center left), KRT19 (center right), and hepatocyte marker Hep Par-1 (HEPPAR1 ) (far right). B) tSNE plot derived from whole primary foetal liver scRNAseq data (n=2, 6pcw, 1406 cells) demonstrating independent clustering into six groups, labelled according to cellular identity (stellate, hepatoblast, haematopoietic stem/progenitor cells (HSPC), megakaryocytes, lymphocytes, and Kupffer cells). C)
Heatmap comparing the top 10 differentially expressed genes differentiating each cluster as identified on the tSNE from B. D) Violin plots displaying markers distinct to each cluster of primary foetal liver cells. E) Feature plots displaying cellular gene expression for each gene listed, based upon the tSNE in E, with darker points representing higher gene expression in that cell, and lighter points demonstrating lower gene expression.
Figure 3 shows a comparison of HO to primary foetal liver. tSNE analysis based on scRNAseq data for HO (n=2 lines, p8, 1973 cells) and PFL hepatoblast (n=2 lines, 308 cells) demonstrating independent clustering based on original cellular identity (B) Feature plots displaying cellular gene expression for each gene listed, based upon the tSNE in A, with darker colored points representing higher gene expression in that cell, and lighter points demonstrating lower gene expression. C) Violin plots demonstrating the expression of key hepatic markers across each cellular population of PFL-hepatoblasts and HO-hepatoblasts. D) Principal component analysis of scRNAseq data from PFL-hepatoblasts (red), HO (green), CoP (turquoise) and Co (purple). E) Violin plots of scRNAseq data from PFL hepatoblasts, HO, CoP and Co, demonstrating the respective hepatoblast and cholangiocytic profiles. F) Heatmap comparing the top genes for each cluster identity for PFL hepatoblast, HO, CoP and Co. Yellow = higher expression, purple = lower expression.
Figure 4 shows In vitro differentiation of HO to cholangiocyte fate. 4A shows representative brightfield images of untreated HO (top), and TGF-beta (TGF-b) treated HO (bottom). B) Change in gene expression before and after treatment with TGF-b for HO (n=9, 7 lines, p4-p9) and FBO (n=4, 2 lines, p4&p9), measured by qPCR. C) Immunofluorescent staining for KRT19 expression (green, upper panel) and ALB expression (yellow, lower panel) in HO and HO treated with TGF-b (HO-TGF, center), with FBO as comparison (right).
D) scRNA-seq derived violin plots demonstrating distribution of gene expression in cells from different groups, comparing PFL hepatoblasts (PFL, n=308), HO (n=1976), HO-TGF (n=461 ), CoP (n=2238) and Co (n=913). E) Pseudotime analysis plot of sc-RNA sequencing data demonstrating a progression of cellular identity from a common time point (darker), and transition through two more differentiated branches. F) Identical plot from E, labelled according to cellular identity PFL hepatoblasts (orange), HO (yellow), HO-TGF (green), CoP (blue), and Co (purple), demonstrating two independent branches of CoP and Co, with HO- TGF aligning with Co. G) Plots of relative gene expression for each cell ordered according to pseudotime from left to right, demonstrating the trends in gene expression during pseudotime.
Figure 5A shows representative brightfield images of HO (left) and HO treated with Hepatozyme (HZ, right). B) Immunofluorescent staining demonstrating ALB (yellow) and alpha-fetoprotein (AFP, red) in untreated HO (left) and HO treated with HZ (HO-HZ, right) C) Relative gene expression of selected hepatic markers measured by qPCR of HO (n=3 lines, p4) and HO after differentiation with WNT withdrawal and hepatozyme (n=3 lines, p4). Maturation markers SERPINA1 and G6PC were significantly upregulated, whilst AFP was downregulated, with no significant change in ALB. E) Violin plots demonstrating gene expression profiles across the scRNAseq data sets from PFL-hepatoblasts (n=308), HO (n=1986), and hepatozyme-treated HO (HO-HZ, n=4006). F) Pseudotime plots demonstrating trend of cells from earlier developmental time point (top, darker blue), to a later time point (light blue) (upper panel), with the same transition labelled according to cellular identity (lower panel, PFL (red), HO (green) and HO-HZ (blue). G) Plots of gene expression for each cell ordered according to pseudotime from left to right, demonstrating the trends in gene expression.
Figure 6 shows violin plots of normalised gene expression comparing PFL, HBO, BO and DBO.
Figure 7 shows a comparison of three human liver organoid systems. (A) Principal component analysis of differentiated biliary organoids (DBO, n =2), fetal hepatocyte organoids (FHO n = 6), hepatoblast organoids (HBO, n = 4), primary adult hepatocytes (PAH, n=2), and primary fetal hepatocytes (PFH, n=2). (B) Jitter plots comparing scaled expression values for DBO, FHO, HBO, PAH, and PAH for selected marker genes.
Figure 8 shows hepatoblast organoid differentiation into hepatocytes and cholangiocytes in vivo. (A) Human ALB in the serum of HBO recipient mice at the time of sacrifice (27 days after engraftment). (B)
Quantification (percentage) of KRT19-positive cells within KRT18-positive nodules following immunostaining for KRT19 (green) and KRT18 (red) identified nodules with no KRT19 (NEGATIVE), nodules that contained some KRT19-positive cells (MIX), and nodules in which cells self-assembled to resemble bile ducts and were nearly entirely positive for KRT19 (POSITIVE), (scale bars = 20uM).
Detailed Description
This invention relates to the expansion of primary hepatoblasts in vitro using a hepatoblast expansion medium comprising epidermal growth factor (EGF), a TGF inhibitor, a non-canonical Wnt signalling potentiator, a canonical Wnt potentiator and a ROCK inhibitor. Optionally, the hepatoblasts may be further differentiated into hepatocytes or cholangiocytes. This may be useful for example in modelling liver development, drug screening and regenerative medicine, for example in the treatment of liver disease,
The hepatoblasts described herein may be expanded from immature liver cells. Immature liver cells may include neonatal and prenatal liver cells.
Immature liver cells may be primary cells isolated from a sample of immature liver tissue. Hepatoblasts are not present in mature liver tissue, for example in individuals of more than 2 years old. Hepatoblasts are present in immature liver tissue. In some embodiments, a sample of immature liver tissue may be obtained from an individual of 2 years or less, 1 year or less 6 months or less or 1 month or less (e.g. a neonate (Tolosa et al (2014) Cell Transplant. 23 (10) 1229-1242)). For example, neonatal liver cells may be
employed. In other embodiments, immature liver cells may be obtained from a sample of foetal liver tissue pre-birth, for example a sample of tissue having a gestational age of 5 weeks or more, 6 weeks or more, 8 weeks or more or 12 weeks or more, for example to 5 to 20 weeks, or 6 to 12 weeks. Suitable samples of foetal liver tissue may, for example, be obtained from patients following elective terminations. Immature liver cells may express hepatic markers, such as ALB, AFP and EpCAM.
The immature liver cells may be isolated from a sample of immature liver tissue using any convenient technique. For example, a sample of immature liver tissue may be dissociated into a single cell suspension by enzymatic treatment, for example with collagenase and hyaluronidase, and the suspension sorted for EpCAM positive cells using anti-EpCAM coated microbeads.
Immature liver cells for use as described herein may be mammalian cells, for example human, mouse or rat cells. Preferably, the immature liver cells are human.
Populations of hepatoblasts may be expanded from the immature liver cells. Hepatoblasts are proliferative bipotent hepatic progenitor cells that are capable of differentiation into hepatocytes or cholangiocytes.
Hepatoblasts are involved in liver organogenesis in the developing mammalian foetus and express markers specific for both hepatocytes and cholangiocytes, such as Epithelial Cell Adhesion Molecule (EpCAM), Albumin and Alpha-Fetoprotein (AFP).
Preferably the hepatoblasts are human hepatoblasts.
The hepatoblasts are expanded in a hepatoblast expansion medium. This is a cell culture medium that supports the proliferation of hepatoblasts in the form of organoids (hepatoblast organoids or HOs).
The hepatoblast expansion medium is a nutrient medium which comprises epidermal growth factor (EGF), a TGF inhibitor, a non-canonical Wnt signalling potentiator, a canonical Wnt potentiator and a ROCK inhibitor. In preferred embodiments, the hepatoblast expansion medium is a chemically defined medium comprising epidermal growth factor (EGF), a TGF inhibitor, a non-canonical Wnt signalling potentiator, a canonical Wnt potentiator and a ROCK inhibitor.
Epidermal Growth Factor (EGF; NCBI GenelD: 1950, nucleic acid sequence NM_001178130.1 Gl:
296011012; amino acid sequence NP_001 171601.1 Gl: 296011013) is a protein factor which stimulates cellular growth, proliferation and cellular differentiation by binding to an epidermal growth factor receptor (EGFR). EGF may be produced using routine recombinant techniques or obtained from commercial suppliers (e.g. R&D Systems, Minneapolis, MN; Stemgent Inc, USA). Suitable concentrations of EGF for expanding cholangiocyte organoids as described herein may be readily determined using standard techniques. For example, the expansion medium may comprise 2 to 500ng/ml EGF, preferably about 20ng/ml.
A TGF inhibitor is a compound that reduces, blocks or inhibits TGF signalling through the TGF RI and TGF RII receptors. Suitable TGF inhibitors include A83-01 3-(6-Methyl-2-pyridinyl)-N-phenyl-4-(4-
quinoiinyi)-1 H-pyrazoie-1-carbothioamide), D4476 (4-[4-(2,3-Dihydro-1 ,4-benzodioxin-6-yl)-5-(2-pyridinyl)- 1 H-imidazoI-2-yl]benzamide), GW788388 (4-[4-[3-(2-Pyrid inyl)- 1 H-pyrazol-4-yl]-2-pyrid i nyl]-A/-(tetrahyd ro- 2H-pyran-4-yl)-benzamide), IN 1 130 (3-[[5-(6-Methyl-2-pyridinyl)-4-(6-quinoxalinyl)-1 /-/-imidazol-2- yl]methyl]benzamide), LY364947 (4-[3-(2-Pyridinyl)-1 tf-pyrazol-4-yl]-quinoline), SB525334 (6-[2-(1 , 1- DimethyIethyl)-5-(6-methyl-2-pyridinyl)-1 H-imidazoI-4-yl]quinoxaline), SB431542 (4-(5-Benzol[1 ,3]dioxol-5-yl- 4-pyrldin-2-yl-1 H-imidazol-2-yl)-benzannide hydrate; Sigma, Tocris Bioscience, Bristol UK), SB-505124 (2-(5- benzo[1 ,3]dioxol-5-yl-2-tert-butyl-3H-imidazol-4-yl)-6-methylpyridine hydrochloride) and soluble protein factors, such as lefty (e.g. human lefty 2: NP_003231.2 Gl:27436881 ), cerberus (e.g. human Cerberus 1 : NP_005445.1 Gl:4885135) and follistatin (e.g. human foistatin: NP_006341.1 Gl:5453652). Suitable TGF inhibitors are available from commercial suppliers. In some embodiments, the TGF inhibitor may be A8301 , for example at 1 to 1000mM, preferably about 50mM.
A non-canonical Wnt signalling potentiator is a compound that stimulates, promotes or increases the activity of the non-canonical Wnt signalling pathway. The non- canonical Wnt signalling pathway is a b-catenin- independent pathway involved in tissue polarity and morphogenetic processes in vertebrates (Komiya, Y. & Habas, R. Organogenesis 4, 68-75 (2008); Patel, V. et ai . Hum. Mol. Genet. 17, 1578-1590 (2008);
Strazzabosco, M. & Somlo, S. Gastroenterology 140, (201 1 ).) Components of the non- canonical Wnt signalling pathway include Wnt4, Wnt5a, Wnt1 1 , LRP5/6, Dsh, Fz, Daaml , Rho, Rac, Prickle and
Strabismus. Suitable methods for determining the activity of the non- canonical Wnt/PCP signalling pathway are well known in the art and include ATF-2-based reporter assays (Ohkawara et al (201 1 ) Dev Dyn 240 (1 ) 188-194) and Rho-associated protein kinase (ROCK)-based assays. A non- canonical Wnt signalling potentiator may selectively potentiate non-canonical Wnt signalling or more preferably, may potentiate both the non-canonical Wnt signalling and the canonical Wnt signalling pathway (i.e. a Wnt signalling agonist).
Preferred non- canonical Wnt signalling potentiators include the Wnt signalling agonist R-spondin. R-spondin is a secreted activator protein with two cysteine-rich, furin-like domains and one thrombospondin type 1 domain that positively regulates Wnt signalling pathways. Preferably, R-spondin is human R-spondin.
R-spondin may include RSP01 (GenelD 284654 nucleic acid sequence reference NM_001038633.3, amino acid sequence reference NP_001033722.1 ), RSP02 (GenelD 340419 nucleic acid sequence reference NM_001282863.1 , amino acid sequence reference NP_001269792.1 ), RSP03 (GenelD 84870, nucleic acid sequence reference NM_032784.4, amino acid sequence reference NP_1 16173.2) or RSP04 (GenelD 343637, nucleic acid sequence reference NM_001029871.3, amino acid sequence reference
NP_001025042.2). R-spondin is readily available from commercial sources (e.g. R&D Systems,
Minneapolis, MN). Suitable concentrations of R-spondin for expanding heptoblasts as described herein may be readily determined using standard techniques. For example, the expansion medium may comprise 50ng/ml to 5pg/ml R-spondin, preferably about 500ng/ml.
A canonical Wnt potentiator is compound that stimulates, promotes or increases the activity of the canonical Wnt/ b-catenin-sig nailing pathway. The canonical Wnt signalling pathway is a b-catenin-dependent pathway involved in the regulation of gene expression (Klaus et al Nat. Rev. Cancer (2008) 8 387-398; Moon et al (2004) Nat. Rev. Genet. 5 691-701 ; Niehrs et al Nat Rev Mol. Cell Biol. (2012) 13 763-779). Suitable methods for determining the activity of the canonical Wnt signalling pathway are well known in the art and
include the TOP-flash assay (Molenaar et al Cell. 1996 Aug 9; 86(3):391-9) and assays for b-catenin.
Suitable Wnt signalling activators include Wnt ligands, glycogen synthase kinase 3b (63K3b) inhibitors; b- catenin and activators of b-catenin. ObK3b inhibitors inhibit the activity of glycogen synthase kinase 3b (Gene ID 2932: EC2.7.11.26).
Preferred canonical Wnt potentiators include (hetero)arylpyrinnidines (Gilbert et al Bioorg Med Chem Lett. 2010 Jan 1 ;20(1 ):366-70), WAY-316606 (Bodine et al Bone. 2009 Jun;44(6): 1063-8), IQ1 (Miyabayashi et al PNAS USA 2007 104(13) 5668-5673), QS11 (Zhang et al (2007) PNAS USA 104(18) 7444-7448), and 2- amino-4-[3,4-(nnethylenedioxy)benzyl-annino]-6-(3-nnethoxyphenyl)pyhnnidine (Liu et al Angew Chem Int Ed Engl. 2005 Mar 18;44(13): 1987-90) 2-amino-4-[3,4-(methlyenedioxy)benzylamino]-6-(3- methoxyphenyl)pyrimidine (Wnt agonist), WAS P-1 , Wnt, for example Wnt3a, and GSK3 inhibitors, such as CHIR99021 (6-[2-[[4-(2,4-dichlorophenyl)-5-(5-methyl-1H-imidazol-2-yl)pyrimidin-2- yl]amino]ethylamino]pyridine-3-carbonitrile), CHIR98014 (A/-6-[2-[[4-(2,4-Dichlorophenyl)-5-(1/-/-imidazol-1- yl)-2-pyrimidinyl]amino]ethyl]-3-nitro-2,6-pyridinediamine) alsterpaullone, kenpaullone, BIO(6-bromoindirubin- 3'-oxime (Sato et al Nat Med. 2004 Jan;10(1 ):55-63), SB216763 (3-(2,4-dichlorophenyl)-4-(1 -methyl-1 H- indol-3-yl)-1H-pyrrole-2,5-dione), and SB415286 ( 3-[(3-chloro-4-hydroxyphenyl)amino]-4-(2-nitrophenyl)-1H - pyrrole-2, 5-dione; Coghlan et al Chem Biol. 2000 Oct;7(10):793-803). Suitable canonical Wnt potentiators are available from commercial suppliers.
A ROCK inhibitor is a compound that reduces, blocks or inhibits Rho kinase (ROCK) (Liao et al (2007) J. Cardiovasc Pharmacol. 50 (1 ) 17-24). Suitable ROCK inhibitors include fasudil, Y39983 (4-[(1R)-1- Aminoethyl]-A/-1/-/-pyrrolo[2,3-b]pyridin-4-ylbenzamide dihydrochloride), azabenzimidazole-aminofurazans and Y-27632 (trans-4-[(1 R)-1-Aminoethyl]-N-4-pyridinylcyclohexanecarboxamide) and are available from commercial suppliers. For example, the hepatoblast expansion medium may comprise 1 to 100pM Y-27632, preferably about 10 mM.
The hepatoblast expansion medium may be devoid of growth factors other than the epidermal growth factor (EGF), TQRb inhibitor, non-canonical Wnt signalling potentiator, canonical Wnt potentiator and ROCK inhibitor.
Suitable hepatoblast expansion media may include HO-M (Table 1 ).
Preferably, the immature liver cells are cultured in the hepatocyte expansion medium in three-dimensional culture in the methods described above. For three-dimensional culture, the expansion medium further comprises a scaffold matrix which supports the growth and proliferation of cells in 3-dimensions and allows the hepatoblasts to assemble into organoids. For example, the expansion medium may comprise or consist of the scaffold matrix and the nutrient medium.
Suitable scaffold matrices are well-known in the art and include hydrogels, such as collagen,
collagen/laminin, compressed collagen (e.g. RAFT™, TAP Biosystems), alginate, agarose, complex protein hydrogels, such as Base Membrane Extracts, and synthetic polymer hydrogels (Gjorevski et al Nature (2016) 539 560-564), such as polyglycolic acid (PGA) hydrogels and crosslinked dextran and PVA hydrogels (e.g.
Cellendes Gmbh, Reutlingen DE), inert matrices, such as porous polystyrene, and isolated natural ECM scaffolds (Engitix Ltd, London UK).
The scaffold matrix may be chemically defined, for example a collagen or densified collagen hydrogel, or non-chemical ly defined, for example a complex protein hydrogel. Preferably, the scaffold matrix in the expansion medium is a complex protein hydrogel. Suitable complex protein hydrogels may comprise extracellular matrix components, such as laminin, collagen IV, enactin and heparin sulphate proteoglycans. Complex protein hydrogels may also include hydrogels of extracellular matrix proteins from Engelbreth- Holm-Swarm (EHS) mouse sarcoma cells. Suitable complex protein hydrogels are available from commercial sources and include Matrigel™ (Corning Life Sciences) or Cultrex™ BME 2 RGF (Amsbio™ Inc). For example, the expansion medium may comprise 55% Matrigel™.
The hepatoblast expansion medium may comprise or consist of a scaffold matrix and a nutrient medium supplemented with epidermal growth factor (EGF), a TGF inhibitor, a non- canonical Wnt signalling potentiator, such as R-spondin, a canonical Wnt potentiator and a ROCK inhibitor.
A nutrient medium may comprise a basal medium. Suitable basal media include Iscove’s Modified
Dulbecco’s Medium (IMDM), Ham’s F12, Advanced Dulbecco’s modified eagle medium (DMEM) or
DMEM/F12 (Price et al Focus (2003), 25 3-6), Williams E (Williams, G.M. et al Exp. Cell Research, 89, 139- 142 (1974)), and RPMI-1640 (Moore, G.E. and Woods L.K., (1976) Tissue Culture Association Manual. 3, 503-508.
In some embodiments, DMEM/F12 medium may be preferred.
The basal medium may be supplemented with a media supplement, such as B27 (ThermoFisher Scientific) and/or one or more additional components, for example L-glutamine or substitutes, such as L-alanyl-L- glutamine (e.g. Glutamax™), nicotinamide, N-acetylcysteine, buffers, such as HEPES, and antibiotics such as penicillin and streptomycin. For example, the basal medium may be supplemented with 2% (v/v) B27, 20mM nicotinamide, 2mM N=acetylcysteine, 1 % L-alanyl-L-glutamine, 1 % HEPES, 1 % penicillin and 1 % streptomycin.
The nutrient medium may be a chemically defined basal nutrient medium. A chemically defined medium is a nutritive solution for culturing cells which contains only specified components, preferably components of known chemical structure. A chemically defined medium is devoid of undefined components or constituents which include undefined components, such as feeder cells, stromal cells, serum, serum albumin and complex extracellular matrices, such as Matrigel™. A chemically defined medium may be humanised. A humanised chemically defined medium is devoid of components or supplements derived or isolated from non-human animals, such as Foetal Bovine Serum (FBS) and Bovine Serum Albumin (BSA), and mouse feeder cells. Conditioned medium includes undefined components from cultured cells and is not chemically defined.
Suitable chemically defined nutrient media are well-known in the art and include DMEM/F12 supplemented with B27, nicotinamide, N-acetylcysteine, L-alanyl-L-glutamine, HEPES, penicillin and streptomycin.
The hepatoblasts described herein do not require the presence of HGF or FGF, such as FGF10 for long-term culture.
In some preferred embodiments, the primary immature liver cells are not subjected to any pre-culture before being expanded in the hepatoblast expansion medium.
The hepatoblasts may form organoids in the expansion medium. The hepatoblast organoids may be disrupted as required, such that the expanded population comprises individual cells.
The present methods allow the long-term proliferation of hepatoblasts. The hepatoblasts may be cultured in the expansion medium for multiple passages. For example, the hepatoblasts may be cultured for 25 or more, 30 or more, 40 or more or 50 or more passages. A passage may take 7-14 days, preferably about 10 days.
The hepatoblasts may be passaged by digesting the scaffold matrix, harvesting organoids by centrifugation and disrupting the organoids into individual hepatoblasts. The hepatoblasts may be re-suspended and cultured as described above in the expansion medium where they reform into organoids.
Suitable techniques for cell culture are well-known in the art (see, for example, Basic Cell Culture Protocols, C. Helgason, Humana Press Inc. U.S. (15 Oct 2004) ISBN: 1588295451 ; Human Cell Culture Protocols (Methods in Molecular Medicine S.) Humana Press Inc., U.S. (9 Dec 2004) ISBN: 1588292223; Culture of Animal Cells: A Manual of Basic Technique, R. Freshney, John Wiley & Sons Inc (2 Aug 2005) ISBN:
0471453293, Ho WY et al J Immunol Methods. (2006) 310:40-52, Handbook of Stem Cells (ed. R. Lanza) ISBN: 0124366430) Basic Cell Culture Protocols’ by J. Pollard and J. M. Walker (1997),‘Mammalian Cell Culture: Essential Techniques’ by A. Doyle and J. B. Griffiths (1997),‘Human Embryonic Stem Cells’ by A. Chiu and M. Rao (2003), Stem Cells: From Bench to Bedside’ by A. Bongso (2005), Peterson & Loring (2012)Human Stem Cell Manual: A Laboratory Guide Academic Press and‘Human Embryonic Stem Cell Protocols’ by K. Turksen (2006). Media and ingredients thereof may be obtained from commercial sources (e.g. Gibco, Roche, Sigma, Europa bioproducts, R&D Systems). Standard mammalian cell culture conditions may be employed for the above culture steps, for example 37°C, 21 % Oxygen, 5% Carbon Dioxide. Media is preferably changed every two days and cells allowed to settle by gravity
The population of hepatoblasts may be expanded 1010 fold or more, 1020 fold or more, 1030 fold or more, 104° fold or more or 105° fold or more as organoids in the expansion medium as described herein.
The population of hepatoblasts proliferates in the expansion medium and assembles into organoids.
Organoids are three-dimensional multicellular assemblies that comprise hepatoblasts linked by tight junctions. The organoids may display the morphology or physical characteristics of hepatoblasts and may for example form dense, branching or spherical structures. The morphology and physical characteristics of organoids may be determined by standard microscopic procedures.
The hepatoblasts in the expanded population are bipotent progenitor cells and are capable of differentiation into either hepatocytes or cholangiocytes. The hepatoblasts may express genes important for hepatic function, including detoxification enzymes, such as cytochrome p450 3A7 (CYP3A7), and cytochrome p450 3A4 (CYP3A4), serum proteins, such as albumin, APOB, alpha-fetoprotein and TF and clotting factors, such as SERPINA1. The hepatoblasts may show very high expression of ALB, moderate expression of MUC20 and low or no expression of Factor II (prothrombin) and Factor X (Stuart-Prower factor) (see Table 2).
The hepatoblasts may express both hepatocytic and cholangiocytic markers. Hepatocytic markers may include ASGR1 , CYP3A4, albumin (ALB), alpha-1 -antitrypsin (SERPINA1 ), APOH, APOM, APOC1 ,
CYP1 A1 , RBP4, HP, AHSG and TTR. In some embodiments, the hepatoblasts may express similar levels of hepatocytic markers to primary adult hepatocytes. Cholangiocytic markers may include KRT19, EPCAM and SOX9. In some embodiments, the hepatoblasts may express lower levels of cholangiocytic markers than primary cholangiocytes.
The hepatoblasts in the expanded population may express one or more foetal markers. Foetal markers include alpha-fetoprotein (AFP), DLK1 , CYP2E1 and CYP3A7. The hepatoblasts in the expanded population may also express serum proteins, such as APOB and TF.
In some preferred embodiments, the hepatoblasts in the expanded population may express the markers ALB, AFP, SERPINA1 , APOE, APOC3, APOA1 , APOA2, TTR, AHSG, ARG1 , ASGR1 and ITIH1 and may not express the markers KRT7 and CTFR.
The expression of cell markers may be determined by any suitable technique, including
immunocytochemistry, immunofluorescence, RT-PCR, immunoblotting, fluorescence activated cell sorting (FACS), and enzymatic analysis. A cell may be considered to express a cell marker if the expression of the marker in the cell is detectable by one or more of the above techniques.
The hepatoblasts in the expanded population may display long term stability. For example, the hepatoblasts may be maintained in culture for at least 2 months without DNA copy number or other genetic abnormalities and with stable, preferably homogeneous, expression of hepatic markers, such as EpCAM, ASGR1 , AFP, A1AT, ALB, CYP3A4, CYP3A7, HNF4a, HNF1 b and APOB. The presence of genetic abnormalities may be determined for example by comparative genomic hybridisation (CGH).
The hepatoblasts in the expanded population may display high proliferative potential. For example, the hepatoblasts may display a doubling time of 2-4 days, for example about 3 days.
The expanded population of hepatoblasts, whether in the form of organoids or individual cells, may be free or substantially free from other cell types i.e. the population of hepatoblasts may be homogeneous or substantially homogeneous. For example, the population may contain, 80% or more, 90% or more, 95% or more, 98% or more or 99% or more hepatoblasts, following culture in the medium. Preferably, the population
of hepatoblasts is sufficiently free of other cell types that no purification is required. Alternatively, the hepatoblasts may be further purified, for example by magnetic bead sorting.
Hepatoblasts expanded as described herein may be cultured or maintained using standard mammalian cell culture techniques or subjected to further manipulation or processing. In some embodiments, the cell populations produced as described herein may be stored, for example by lyophilisation and/or
cryopreservation. The hepatoblasts may be stored as organoids, sub-organoid assemblies or individual cells. Suitable storage methods are well known in the art. For example, the hepatoblasts may be suspended in a cryopreservation medium (for example, Cellbanker™ (AMS Biotechnology Ltd, UK)) and frozen, for example at -70°C or below.
In some embodiments, the hepatoblasts in the expanded population may be further purified and/or isolated. For example, the hepatoblasts in the expansion medium may be separated from stellate cells. Conveniently, hepatoblasts may be distinguished from stellate cells by the expression of EpCAM and separated using routine techniques, such as flow cytometry.
In some embodiments, hepatoblasts expanded by the methods described above may be used directly, for example in regenerative medicine applications. In other embodiments, the hepatoblasts may be further differentiated in vitro.
The hepatoblasts may be differentiated into cholangiocytes by culturing in a differentiation medium that supports TGF signalling. For example, the differentiation medium may comprise TGF or a TGF ligand such as activin. In some embodiments, the hepatoblasts may be cultured in the absence of canonical Wnt signalling. For example, the differentiation medium may lack canonical Wnt potentiators, such as Wnt3a. Suitable differentiation media include CO-M that lacks Wnt3a and A8301 and is supplemented with TGF . The constituents of CO-M are shown in Table 1.
Culturing in the differentiation medium may increase the expression of cholangiocytic markers, such as KRT19, and reduce the expression of hepatocytic markers, such as ALB and AFP. For example, cholangiocytes produced as described herein may express cholangiocytic markers and not hepatocytic markers.
The hepatoblasts may be differentiated into hepatocytes by culturing in hepatocyte culture medium. The hepatoblasts may be cultured in the absence of canonical Wnt potentiators. The hepatoblasts may be cultured in the absence of canonical Wnt signalling. For example, the hepatocyte culture medium may lack canonical Wnt potentiators, such as Wnt3a. A hepatocyte culture medium is a medium that supports the culture of hepatocytes. In some embodiments, hepatocyte culture medium may be a chemically defined medium devoid of growth factors other than oncostatin-M. Suitable hepatocyte culture media are available from commercial suppliers and may include hepatocyte cell culture medium (#H1777 Sigma Aldrich), primary HEP medium (Cellartis) or Hepatozyme™ (Life Technologies), with supplementary oncostatin-M.
Culturing in the hepatocyte culture medium may reduce the expression of cholangiocytic and foetal markers, such as AFP, and increase the expression of hepatocytic and mature markers, such as G6PC. For example, hepatocytes produced by differentiation of hepatoblasts as described herein may express hepatocytic markers and not cholangiocytic or foetal markers.
The hepatocytes and cholangiocytes produced by differentiation of hepatoblasts may be fully differentiated and may lack proliferative capacity in vitro. The hepatocytes and cholangiocytes produced by differentiation of hepatoblasts may display the functional activity of mature hepatocytes and cholangiocytes.
The population of hepatoblasts, cholangiocytes or hepatocytes produced as described herein may be admixed with other reagents, such as buffers, carriers, diluents, preservatives, and/or pharmaceutically acceptable excipients. Suitable reagents are described in more detail below. A method described herein may comprise admixing the population with a therapeutically acceptable excipient to produce a therapeutic composition. The admixed hepatoblasts may be in the form of organoids, sub-organoid assemblies or individual cells.
In some embodiments, the hepatoblasts, cholangiocytes or hepatocytes produced as described herein may be useful in therapy. For therapeutic applications, the hepatoblasts, cholangiocytes or hepatocytes are preferably clinical grade cells. Populations of hepatoblasts, cholangiocytes or hepatocytes for use in treatment are preferably produced from immature liver cells as described herein using chemically defined media. The hepatoblasts may be in the form of organoids, sub-organoid assemblies or individual cells, depending on the specific application.
The population of hepatoblasts, cholangiocytes or hepatocytes may be transplanted, infused or otherwise administered into the individual. Suitable techniques are well known in the art.
In some preferred embodiments, the population of hepatoblasts, cholangiocytes or hepatocytes produced as described herein may be admixed with a biocompatible scaffold.
A biocompatible scaffold may be seeded with hepatoblasts, cholangiocytes or hepatocytes expanded as described above. For example, individual hepatoblasts or sub-organoid assemblies of hepatoblasts may be injected on or into a scaffold or mixing into the scaffold during the manufacturing process. The scaffold containing the hepatoblasts may then be cultured in expansion medium, such that the hepatoblasts populate the scaffold. The hepatoblasts may proliferate within the scaffold and assemble into organoids.
Suitable biocompatible scaffolds may include hydrogels, such as fibrin, chitosan, glycosaminoglycans, silk, fibrin, fibronectin, elastin, collagen, glycoproteins such as fibronectin, or polysaccharides such as chitin, or cellulose collagen, collagen/laminin, densified collagen, alginate, agarose, complex protein hydrogels, such as Base Membrane Extracts, bio-organic gels, and synthetic polymer hydrogels, such as polylactic acid (PLA) polyglycolic acid (PGA), polycapryolactone (PCL) hydrogels, crosslinked dextran and PVA hydrogels (e.g. Cellendes Gmbh, Reutlingen DE), inert matrices, such as porous polystyrene, polyester, soluble glass
fibres porous polystyrene, and isolated natural ECM scaffolds, for example decellularized gall bladder and bile duct scaffolds (Engitix Ltd, London UK). The scaffold may be biodegradable.
The size or shape of the scaffold is dependent on the intended application. Suitable scaffold shapes may for example include patches, sheets and tubes, including straight and branched tubes, with diameters up to for example 10-12 mm.
Hepatoblasts, cholangiocytes or hepatocytes cultured within a biocompatible scaffold organize into a functional hepatic tissue, for example functional hepatocytic or biliary tissue. For example, cholangiocytes cultured within a biocompatible scaffold may organize into a functional biliary epithelium that displays one or more properties of the biliary epithelium. Hepatoblasts or hepatocytes cultured within a biocompatible scaffold may organize into a functional hepatocytic tissue that displays one or more properties of hepatocytic tissue.
An aspect of the invention provides an isolated population of hepatoblasts, cholangiocytes or hepatocytes produced by a method described above. The hepatoblasts may be in the form of organoids, sub-organoid clusters or individual cells.
A population of hepatoblasts, cholangiocytes or hepatocytes generated as described herein may be substantially free from other cell types. For example, the population may contain 70% or more, 80% or more, 85% or more, 90% or more, or 95% or more hepatoblasts, following culture in the expansion medium. The presence or proportion of hepatoblasts, cholangiocytes or hepatocytes in the population may be determined through the expression of biliary and/or hepatic markers as described above.
Preferably, the population of hepatoblasts, cholangiocytes or hepatocytes is sufficiently free of other cell types that no purification is required. If required, the population of cells or organoids may be purified by any convenient technique, including FACS.
In some embodiments, the hepatoblasts, cholangiocytes or hepatocytes may be engineered to express a heterologous protein, for example a marker protein, such as GFP, or an enzyme and/or to reduce or prevent expression of one or more endogenous protein, for example proteins associated with immunogenicity, such as HLA antigens.
The isolated population of hepatoblasts, cholangiocytes or hepatocytes may be within a biocompatible scaffold. Another aspect of the invention provides a biocompatible scaffold comprising an isolated population of hepatoblasts produced by a method described herein. Suitable scaffolds are described above.
Another aspect of the invention provides a collection of isolated populations of hepatoblasts as described herein, wherein each population in the collection comprise a different set of tissue markers. For example, each population in the collection may have a different antigenic profile or HLA type. Populations of hepatoblasts with different antigenic profiles may be generated from immature liver cells that display different
antigenic profiles. A population in the collection may be matched to a recipient individual in which the hepatoblasts do not elicit a host immune response. This may be useful in allogenic cell therapy.
Aspects of the invention also extend to a pharmaceutical composition, medicament, drug or other composition comprising hepatoblasts, hepatocytes or cholangiocytes produced as described herein in solution or in a biocompatible scaffold, and a method of making a pharmaceutical composition comprising admixing such hepatoblasts with a pharmaceutically acceptable excipient, vehicle, carrier or biodegradable scaffold, and optionally one or more other ingredients.
A pharmaceutical composition containing hepatoblasts expanded in accordance with the invention or hepatocytes or cholangiocytes differentiated therefrom, may comprise one or more additional components. Pharmaceutical compositions may comprise, in addition to the hepatoblasts, hepatocytes or cholangiocytes, a pharmaceutically acceptable excipient, carrier, buffer, preservative, stabiliser, anti-oxidant, or other material well known to those skilled in the art. Such materials should be non-toxic and should not interfere with the activity of the cholangiocytes. The precise nature of the carrier or other material will depend on the route of administration.
Liquid pharmaceutical compositions generally include a liquid carrier such as water, petroleum, animal or vegetable oils, mineral oil or synthetic oil. Physiological saline solution, tissue or cell culture media, dextrose or other saccharide solution or glycols such as ethylene glycol, propylene glycol or polyethylene glycol may be included.
The composition may be in the form of a parenterally acceptable aqueous solution, which is pyrogen-free and has suitable pH, isotonicity and stability. Those of relevant skill in the art are well able to prepare suitable solutions using, for example, isotonic vehicles such as Sodium Chloride, Ringer's Injection, or Lactated Ringer's Injection. A composition may be prepared using artificial cerebrospinal fluid.
Another aspect of the invention provides a method of treatment of a liver disease comprising administering a population of hepatoblasts, hepatocytes or cholangiocytes produced as described herein to an individual in need thereof.
Another aspect of the invention provides a population of hepatoblasts, hepatocytes or cholangiocytes produced as described herein for use in a method of treatment of a liver disease in an individual in need thereof comprising administering the population to the individual.
Another aspect of the invention provides the use of a population of hepatoblasts, hepatocytes or cholangiocytes produced as described herein in the manufacture of a medicament for use in the treatment of a liver disease.
The hepatoblasts may be in the form of organoids, sub-organoid assemblies or clusters or individual cells.
A liver disease is a condition in which liver tissue, such as hepatocytic tissue, in an individual is damaged, defective or otherwise dysfunctional, for example, disorders characterised by damage to or destruction of liver tissue, or aberrant liver tissue. Liver disease may include hepatitis (e.g. hepatitis A, B, C, D, E, G or K), cirrhosis, hepatocellular carcinoma, non-alcoholic fatty liver disease, drug induced liver injury, alcoholic liver disease, autoimmune liver disease or an inherited metabolic disorder such as Alpha 1 Antitrypsin deficiency, a Glycogen Storage Disease, for example Glycogen Storage Disease Type 1a, Familial
Hypercholesterolemia, Hereditary Tyrosinaemia, Crigler Najjar syndrome, ornithtine transcarbamylase deficiency, or factor IX deficiency or other haemophilia, haemochromatosis, Wilson's disease, Dubin- Johnson syndrome, familial amyloidosis, and Refsum’s disease.
In some preferred embodiments, the liver disease is haemophilia. A population of hepatoblasts, hepatocytes or cholangiocytes as described herein may express clotting factors, such as SERPINA1 , sufficient to rescue a haemophilia phenotype in an individual.
In some embodiments, the liver disease may be a biliary disorder. A biliary disorder is a condition in which the biliary tissue in an individual is damaged, defective or otherwise dysfunctional, for example, disorders characterised by damage to or destruction of bile ducts, aberrant bile ducts or the absence of bile ducts. Biliary disorders may include biliary tissue injury, ischaemic strictures, traumatic bile duct injury and cholangiopathies, for example inherited, developmental, autoimmune and environment-induced
cholangiopathies, such as Cystic Fibrosis associated cholangiopathy, drug induced cholangiopathy, Alagille Syndrome, polycystic liver disease, primary biliary cirrhosis (PBC), primary sclerosing cholangitis (PSC), AIDS associated cholangiopathy, disappearing bile duct syndrome, biliary cancer, ductopenias such as adult idiopathic ductopenia, post-operative biliary complications, and biliary atresia.
In some embodiments, a population of hepatoblasts, hepatocytes or cholangiocytes may be administered to the individual in solution. The administration of a population of hepatoblasts or cholangiocytes in solution may be useful for example in the treatment of ductopenias, for example ischaemic ductopenia, congenital ductopenia, such as alagille syndrome, metabolic ductopenia, complex diseases, such as intrahepatic PSC and PBC, drug induced ductopenia, vanishing bile duct syndrome and conditions affecting the intrahepatic biliary tree
In other embodiments, a population of hepatoblasts, hepatocytes or cholangiocytes may be administered to the individual within a biocompatible scaffold. For example, a scaffold populated with hepatoblasts may be administered to the individual. For example, the administration of a population of hepatoblasts, in a scaffold may be useful for example in the treatment of biliary atresia, biliary strictures, traumatic or iatrogenic biliary injury and conditions affecting the extrahepatic biliary tree
Hepatoblasts, hepatocytes or cholangiocytes in solution or in scaffolds may be implanted into a patient by any technique known in the art (e.g. Lindvall, O. (1998) Mov. Disord. 13 Suppl. 1 :83-7; Freed, C.R., et al., (1997) Cell Transplant, 6, 201-202; Kordower, et al., (1995) New England Journal of Medicine, 332 1118- 1124; Freed, C.R.,(1992) New England Journal of Medicine, 327, 1549-1555, Le Blanc et al, Lancet 2004 May 1 ;363(9419):1439-41 ). In particular, cell suspensions may be injected or infused into the bile duct,
gallbladder, portal vein, liver parenchyma, peritoneal cavity or spleen of a patient. A hepatic cell suspension may be administered intravenously, intraperitoneally or via an endoscopic retrograde cholangiopancreatography (ERCP) or percutaneous cholangiography (PTC). A scaffold populated with hepatoblasts may be administered to the individual by surgical implantation.
Administration of a composition in accordance with the present invention is preferably in a "prophylactically effective amount" or a "therapeutically effective amount" (as the case may be, although prophylaxis may be considered therapy), this being sufficient to show benefit to the individual. The actual amount administered, and rate and time-course of administration, will depend on the nature and severity of what is being treated. Prescription of treatment, e.g. decisions on dosage etc., is within the responsibility of general practitioners and other medical doctors.
A composition comprising hepatoblasts, hepatocytes or cholangiocytes may be administered alone or in combination with other treatments, either simultaneously or sequentially dependent upon the condition to be treated.
Populations of isolated hepatoblasts, hepatocytes or cholangiocytes produced as described above, may be useful in modelling the interaction of test compounds with hepatoblasts, for example in toxicity screening, modelling liver disorders or screening for compounds with potential therapeutic effects.
Another aspect of the invention provides the use of a population of hepatoblasts, hepatocytes or cholangiocytes produced as described herein for disease modelling and study of pathogenesis of liver diseases, such as biliary disorders.
Hepatoblasts, hepatocytes or cholangiocytes for use in modelling and screening may be in the form of organoids (hepatoblast organoids, HOs), sub-organoid clusters or individual cells (hepatoblasts) produced, for example by disruption of organoids.
A method of screening a compound may comprise;
contacting a population of hepatoblasts, hepatocytes or cholangiocytes produced by a method described herein with a test compound, and;
determining the effect of the test compound on said the hepatoblasts, hepatocytes or
cholangiocytes and/or the effect of said the hepatoblasts, hepatocytes or cholangiocytes on the test compound.
The proliferation, growth, apoptosis or viability of hepatoblasts, hepatocytes or cholangiocytes, protein production, metabolic activity of key enzymes, expression of stress response genes, or the ability of the hepatoblasts to perform one or more cell or organoid functions may be determined in the presence relative to the absence of the test compound.
A decrease in proliferation, growth, viability or ability to perform one or more cell or organoid functions is indicative that the compound has a toxic effect and an increase in growth, viability or ability to perform one or
more cell or organoid functions is indicative that the compound has an ameliorative effect on the
hepatoblasts.
Gene expression may be determined in the presence relative to the absence of the test compound. For example, the expression of one or more hepatocytic or biliary marker genes may be determined. Suitable genes include genes encoding plasma proteins, such as albumin, APOB, APOA1 , FGG, C2, KNG1 , and FGA, enzymes, such as HA01 , RDH16, and ALDOB, bile proteins, such as AKR1 C4, SLC27A5, and BAAT, clotting factors, such as SERPINA1 and metabolic proteins. Combined decrease in expression is indicative that the compound has a toxic effect or can modify the functional state of the hepatoblasts. Gene expression may be determined at the nucleic acid level, for example by RT-PCR, or at the protein level, for example, by immunological techniques, such as ELISA, or by activity assays. Cytochrome p450 assays, for example, luminescent, fluorescent or chromogenic assays are well known in the art and available from commercial suppliers. The metabolism, degradation, or breakdown of the test compound by the hepatoblasts, hepatocytes or cholangiocytes may be determined. In some embodiments, changes in the amount or concentration of test compound and/or a metabolite of said test compound may be determined or measured over time, either continuously or at one or more time points. For example, decreases in the amount or concentration of test compound and/or increases in the amount or concentration of a metabolite of said test compound may be determined or measured. In some embodiments, the rate of change in the amount or concentration of test compound and/or metabolite may be determined. Suitable techniques for measuring the amount of test compound or metabolite include mass spectrometry.
This may be useful in determining the in vivo half-life, toxicity, efficacy or other in vivo properties of the test compound. Other aspects of the invention relate to kits and their use for production of expanded populations as described herein. A kit for the production of an expanded population of hepatoblasts may comprise a hepatoblast expansion medium comprising epidermal growth factor (EGF), a TGF inhibitor, a non- canonical Wnt signalling potentiator, Wnt and a ROCK inhibitor.
Suitable hepatoblast expansion media are described in more detail above.
A kit may further comprise a scaffold matrix, such as Matrigel™. The scaffold matrix may be provided as part of the expansion medium or may be provided separately.
The kit may further comprise a differentiation medium and/or a hepatocyte culture medium, as described above.
The expansion medium may be formulated in deionized, distilled water. The expansion medium will typically be sterilized prior to use to prevent contamination, e.g. by ultraviolet light, heating, irradiation or filtration. The one or more media may be frozen (e.g. at -20°C or -80°C) for storage or transport. The one or more media may contain one or more antibiotics to prevent contamination.
The kit may further comprise a dissociation buffer to dissociate immature liver cells from sample tissue. Suitable buffers include Hanks Buffered Saline Solution (HBSS) supplemented with Liberase DH (Roche Applied Science) and Hyaluronidase (Sigma-Aldrich).
The kit may further comprise cryopreservation solution. Suitable cryopreservation media are described above.
The one or more media may be a 1x formulation or a more concentrated formulation, e.g. a 2x to 250x concentrated medium formulation. In a 1x formulation each ingredient in the medium is at the concentration intended for cell culture, for example a concentration set out above. In a concentrated formulation one or more of the ingredients is present at a higher concentration than intended for cell culture. Concentrated culture media are well known in the art. Culture media can be concentrated using known methods e.g. salt precipitation or selective filtration. A concentrated medium may be diluted for use with water (preferably deionized and distilled) or any appropriate solution, e.g. an aqueous saline solution, an aqueous buffer or a culture medium.
The one or more media in the kit may be contained in hermetically-sealed vessels. Hermetically-sealed vessels may be preferred for transport or storage of the culture media, to prevent contamination. The vessel may be any suitable vessel, such as a flask, a plate, a bottle, a jar, a vial or a bag.
Another aspect of the invention provides the use of a hepatoblast expansion medium for the production of an expanded population of hepatoblasts.
Other aspects and embodiments of the invention provide the aspects and embodiments described above with the term“comprising” replaced by the term“consisting of and the aspects and embodiments described above with the term“comprising” replaced by the term’’consisting essentially of.
It is to be understood that the application discloses all combinations of any of the above aspects and embodiments described above with each other, unless the context demands otherwise. Similarly, the application discloses all combinations of the preferred and/or optional features either singly or together with any of the other aspects, unless the context demands otherwise.
Modifications of the above embodiments, further embodiments and modifications thereof will be apparent to the skilled person on reading this disclosure, and as such, these are within the scope of the present invention.
All documents and sequence database entries mentioned in this specification are incorporated herein by reference in their entirety for all purposes.
“and/or” where used herein is to be taken as specific disclosure of each of the two specified features or components with or without the other. For example“A and/or B” is to be taken as specific disclosure of each of (i) A, (ii) B and (iii) A and B, just as if each is set out individually herein.
Experimental
Materials and Methods
Processing of human foetal liver tissue
Primary human foetal tissue was obtained from patients undergoing elective terminations (REC-96/085). The liver was dissected from the abdominal cavity and placed into a solution containing Hanks Buffered Saline Solution (HBSS) supplemented with 1.07 Wdnsch units/ml Liberase DH (Roche Applied Science) and 70U/ml Hyaluronidase (Sigma-Aldrich), and placed on a microplate shaker at 37°C, 750rpm, for 15 minutes. The sample was subsequently washed three times in HBSS using centrifugation at 400g for five minutes each time. The single cell suspension could then be sorted for EPCAM/CD326 positive cells using CD326 microbeads (Miltenyi), according to the manufacturer’s guidelines.
Establishment of hepatoblast organoids
The single cell suspension was resuspended in the hepatoblast organoid media (HO-M) for liver samples.
To the resuspended cells was added an equal volume of Growth Factor Reduced Phenol Free Matrigel (Corning), and the mixture pipetted into 48 well plates (20uL per well). The plates were placed at 37°C for fifteen minutes to allow the mixture to set, and subsequently 200uL of fresh HO-media applied to each well.
Maintenance of cell lines
The respective culture medium was changed every 48-72 hours, and organoids mechanically passaged every 7-10 days. Organoids were passaged by scraping the gel away from the plate, pipetting the resulting solution into 1.5ml tubes, and pipetting the solution up and down to break individual organoids into pieces. If a precise cell number was required, the organoids can alternatively be processed to a single cell solution as described below, and then re-plated at the required dilution in the 55% Matrigel-medium solution.
Dissociation of HO
Organoids established in culture were dissociated to single cell suspensions for splitting/ single cell sequencing/ cell counting by first removing the media from each well and replacing with Cell Recovery Solution (Corning). The organoids in the Matrigel could then be scraped off the plate and placed on ice for 30 minutes to remove the Matrigel. The organoids should then be washed with PBS, and placed in TrypLE (ThermoFisher Scientific) for 15 mins at 37°C. Cells should finally be washed three times in PBS or basal media.
Differentiation to choloangiocytes
CO-M lacking A8301 and supplemented with 2ug/ml TGF (Thermo Fisher Scientific) was applied to HO for 7d, applying fresh media every 48 hours.
Differentiation to hepatocytes
Hepatozyme medium (Gibco) supplemented with oncostatin-M (Sigma Aldrich) was applied to HO for 7d, applying fresh media every 48 hours.
Imaging of organoids
The medium was removed and replaced with 4% Paraformaldehyde solution for 20 minutes at room temperature to fix the organoids in situ, followed by three washes in PBS (each for five minutes). Fixed organoids could then be stored in PBS for later Immunohistochemistry. Immunohistochemistry was performed by permeabilizing and blocking with 0.3% Triton X-100 (Sigma-Aldrich), and 10% donkey serum (Biorad) for three hours at room temperature, and then incubated overnight at 4°C, with the primary antibody in 1 % donkey serum. Samples were then washed with three one-hour PBS washes, followed by incubation with the secondary antibody for one hour at room temperature. The samples were finally washed three times in PBS, with 1 :10000 dilution Hoechst 33258 (ThermoFisher Scientific) added to the first wash. Images can then be obtained using an appropriate microscope. qPCR analysis
RNA was extracted from cells using GenElute Mammalian Total RNA Miniprep Kit (Sigma-Aldrich) according to the manufacturer’s instruction. cDNA was synthesised from RNA using Superscript II (Invitrogen) according to the manufacturer’s instructions. qPCR was performed using SensiMix SYBR low-ROX (Bioline) and 150 nM forward and reverse primers (Sigma-Aldrich; primers listed in supplementary materials). All samples were run in technical duplicates and gene expression calculated relative to housekeeping gene Ribosomal Protein Lateral Stalk Subunit P0 (RPLP-0).
Single Cell RNA-sequencing
Primary Foetal Liver or organoids were dissociated to a single cell solution by the respective methods described above. The single cell solution from each was then processed using the Chromium single cell controller and reagents according to the manufacturer’s guidelines (10X genomics). Sequencing was performed on the HiSeq4000 sequencing system, and the subsequent raw data was aligned and used to generate matrices using the Cell Ranger platform (version 2.1.0). Basic analysis of the data was performed in R using the Seurat, ggplot2, and Monocle packages.
Assessment of genomic stability
Genomic stability of the organoids was assessed using the CytoScan 750k array, performed by the Medical Genetics Service at Cambridge University Hospitals.
Determination of cell line gender
Cell line gender was assessed by assessing RNA expression of the SRY gene (male), or Xist gene (female) based upon qPCR and/or single cell RNA sequencing analysis
Flow cytometry
Organoids were dissociated to single cell solution as described above. The single cells were then fixed and permeabilised in FixPerm (ThermoFisher Scientific) for 20 minutes, then washed three times in PBS before resuspension in PermWash (ThermoFisher Scientific). Blocking was performed using 10% donkey serum (Biorad) in PBS for 30 minutes. Primary antibodies were diluted 1 : 100 in 1 % donkey serum in PBS and incubated with the cell suspension at room temperature for 1 hour. After three washes in PBS, cells were incubated with secondary antibodies diluted 1 :1000 in 1 % donkey serum in PBS. Data were collected using Cyan ADP flow cytometer, and analysed using FlowJo X.
Assessment of cell proliferation rates
Organoids were extracted and dissociated to single cells as described above. The cells were then counted using the countess II automated cell counter (Biorad), and the final number adjusted according to the number of wells used and the volume of resuspension.
Cytochrome P-450 assays
P450 Glo (Promega) assays were used according to manufacturer’s guidelines. Luciferin IPA (CYP3A4 specific), and Luciferin PFBE (CYP3A7 & CYP3A5 > CYP3A4), were added to media and incubated at 37°C for four hours, before using detection assay as per the manufacturer’s instruction. Luminescence RLU was then corrected for cell number.
Serum protein analysis
Albumin, alpha-fetoprotein, apolipoprotein-B, and alpha-1 -antitrypsin were detected in media by enzyme linked immunosorbent assay (performed by core biomedical assay laboratory, Cambridge University Hospitals). Concentrations were normalised to cell number.
Bodipy assay
BODIPY 493/503 (ThermoFisher Scientific) was diluted 1 :1000 in the organoid culture media, and applied to organoids for 30 minutes. After this time the organoids were washed with fresh media, and imaged in situ using a fluorescent microscope.
Freezing/ thawing of HO
Organoids were extracted from Matrigel using cell recovery solution as described above. The organoids were then washed in PBS and resuspended in Cell Culture Freezing Medium (ThermoFisher Scientific), placed in cryogenic vials (ThermoFisher Scientific), and cooled to -80oC using a“Mr. Frosty” Freezing Container (ThermoFisher Scientific). Organoids could then be kept long term in liquid nitrogen. To thaw, cryovials were kept at 4oC until the freezing medium had melted. Organoids were then washed three times in PBS to remove any residual freezing medium, and then replated as above.
Statistical analyses
Unless otherwise stated, statistical analyses of direct comparisons of two groups were performed using t-test with Welch correction. Those comparisons labelled * represent P<0.05, ** p <0.01 , *** P<0.001 , and **** p<0.0001.
Results
Combination of WNT and reduced density ECM allows derivation of hepatobiast organoids from human hepatic foetal tissue
Organoids have been previously derived from the adult liver using two approaches. The first method consists of deriving adult stem cells from the intrahepatic biliary epithelium using WNT, Noggin and Rock-inhibitor for the first three days of in vitro culture (Huch et al., 2015). The resulting liver stem cell organoids (LSCO) mainly express biliary markers while displaying a limited capacity to differentiate into hepatocytes (Huch et al., 2015). The second method derived similar cells from the extra-hepatic biliary epithelium (Sampaziotis et al., 2017). These Extra-hepatic Cholangiocytes Organoids or ECOs closely resemble cholangiocytes and cannot differentiate into any other hepatic lineage.
We hypothesized that hepatobiast derivation would require the development of culture conditions different to those currently available to grow organoids from adult liver. Thus, we screened additional growth factors and basal media to notice that foetal liver tissue dissociated into single cells and grown in the presence of Wnt, R-Spondin, EGF, A83-01 , and Y-27632 (i.e. the same conditions as for FBO, but the addition of Wnt) gave raise to organoids with a markedly different morphology characterised by solid branching structures (Fig 1 C). These organoids were capable of regenerating after fragmentation or dissociation into single cells (Materials and Methods). Furthermore, a 55% dilution of Matrigel formed a permissive microenvironment for the organoids to expand. These“branching” organoids could be split every 10-14 days to a ratio of 1 :4-1 :6, thus allowing rapid expansion of individual cell lines. Crucially, these organoids express key
hepatoblast/hepatocyte markers such as the proteins albumin and alpha-1 -antitrypsin at similar levels to both the primary foetal liver and primary adult hepatocytes (Fig 2A). Genes associated with a foetal stage of development such as the serum protein alpha-fetoprotein or the p450 enzyme CYP3A7, were at comparable levels in organoid and primary cells, whilst being at low level in adult hepatocytes. Importantly, biliary markers such as KRT19, EPCAM and SOX9 were also detected albeit at low levels thereby suggesting that these cells could have the capacity to differentiate into both hepatocytes and cholangiocytes. Considered together, these data suggest that our culture conditions allow the derivation of hepatobiast organoid (HO). Importantly, this method was used to generate 12 HO lines derived from the liver of 5.5 to 10.2 pew embryos of different gender thereby demonstrating the robustness of our approach. The resulting HO lines have been maintained in culture for at least nine months, and all lines derived so far continue to proliferate and expand after each split while maintaining a homogenous expression of EPCAM, ASGR1 , AFP, and A1 AT as shown by flow cytometry. Dissociation and counting of individual cells in culture demonstrate a doubling time of approximately three days, and thus confirm the capacity of these cell line for large scale expansion. Analysis of genetic stability using comparative genomic hybridization in three lines, confirm the absence of DNA copy number abnormalities or any other clinically relevant abnormality. Of note, HO could be frozen and stored for several months while maintaining their characteristics (data not shown). Finally, ELISA analyses confirmed that HO secrete high levels of key cell type specific proteins such as albumin, alpha-fetoprotein, the enzyme alpha-1 -antitrypsin, and the lipoprotein apolipoprotein B (Fig 2C). Similarly, assessment of the function of key detoxification enzymes such as cytochrome p450 3A7 (CYP3A7; expressed in the immature liver), and cytochrome p450 3A4 (CYP3A4, expressed in the mature liver), demonstrated physiological function of both enzymes in the HO, whilst CYP3A7 is not highly active in the primary adult hepatocytes, reinforcing the difference in developmental state of the two cell types. Taken together these results show that HO can be
derived from human foetal liver and that the resulting cells can be grown in vitro for prolonged period of time while maintaining hepatic functions in vitro.
Single cell analyses confirm the presence of hepatoblasts in the early human foetal liver
To fully validate the identity of HO, we decided to define the gene expression profile of their in vivo counterparts using scRNA-seq. For that, we isolated 2 foetal livers at 6 pew, performed single cell dissociation, and analysed the transcriptome of 1406 cells using 10X technology. tSNE analysis demonstrated independent clustering of non-parenchymal cell types including Kupffer Cells (CD68,
FCGR3A, CD33, ITGAM, SPI1 ) Stellate Cells (PDGFRa, ACTA2, VIM, NES), hematopoietic stem/progenitor cells (PTPRC, SPN, MYB, KLF1 , TFRC), lymphocytes (LSP1 , CD52, CD37, CD48) and megakaryocytes (NRGN, PPBP, ITGA2B, GP1 BA, GP6), as well as a homogeneous population of hepatoblasts (ALB, AFP, KRT8, KRT18, EPCAM, SERPINA1 , TTR) (Figure 3B).
Interestingly, these analyses could not identify cells with a biliary expression profile suggesting that these cells are extremely rare at this stage of development and that large scale scRNA-seq analyses might be required to establish a complete atlas of the human foetal liver at different stages of development.
Nonetheless, our data provides substantial novel information into the landscape of the liver at this developmental stage by demonstrating the early colonization of the liver by a diversity of cell types including immune cells, such as tissue resident macrophages and dendritic cells. Furthermore, IHC of an 8 pew liver demonstrates KRT19 positive cells near primitive ducts at this later stage (Fig 3A). Our data also confirm the production of MK cells by HSPC in the liver thereby confirming previous reports. Finally, the presence of stellate cells is also intriguing and suggests that these cells could colonise the liver earlier than initially thought and thus could have a function in early organogenesis. The population of hepatoblast cells showed great similarity between the biological replicates and showed a homogeneity in their gene expression signature, demonstrating the global ability of the liver parenchyma at this stage to expand the hepatoblast population. Importantly, this population of hepatoblasts did express a spectrum of known hepatic markers including albumin, alpha-fetoprotein, TTR and SERPINA1 and also new genes which were not identified previously such as RBP4, AHSG, APOH, APOM or FABP1. Interestingly, a majority of these genes are expressed at the protein level in adult hepatocytes based on the protein atlas with the exception of SPINK1 and FGB (Table S1 ). Based on this information, hepatoblast could be defined by the combined expression of ALB, SPINK1 , AFP, SERPINA1 , CYP3A7. Taken together, these data show that the human foetal liver contains a diversity of cell types including a homogeneous population of hepatoblast cells.
Hepatoblast organoids form a homogeneous population that closely resembles the in vivo counterpart In order to further characterise our novel foetal liver organoid culture we next applied scRNA-seq to our HO (n=2) and compared the generated profiles to those from primary tissue. Interestingly, we observed that HO culture contain two separate populations of cells. The first cluster consists of the majority of cells and includes cells homogeneously expressing hepatoblast markers contrary to the second small cluster which only express VIM, PDGFRa, and PDGFRb. Further analysis of the genes differentially expressed between these two populations confirmed that the first cluster contains a homogeneous population of hepatoblasts whilst the second cluster contains cells expressing markers specific to stellate cells. To confirm this observation, stellate cells grown in vitro were compared to their primary counterpart using single cell data
generated on primary foetal liver. This analysis confirmed that HO culture contains a population of stellate cells (Fig S3B). To further understand the importance of these stellate cells, we isolated hepatoblast cells using EPCAM sorting and also derived new HO lines after EPCAM sorting of single cell from foetal tissue. In both cases, the isolation of EPCAM expressing cells resulted in the absence of stellate cells and the resulting HO maintain the same capacity to grow and to differentiate. Interestingly, stellate cells isolated by negative selection using EPCAM sorting were not able to grow independently more than a couple of passages thereby suggesting that these cells may require the presence of hepatoblasts for their
proliferation/survival while the reciprocity does not apply. Thus, these observations suggest that stellate cells development could rely on hepatoblast during liver organogenesis.
We next compared the single cell expression profiles of HO hepatoblast cells to PFL hepatoblast cells and the FBO. The Co and CoP populations maintain their distinct clustering, to each other and to the
hepatoblasts, with the most differentially expressed genes confirming the different identities of the hepatoblasts (SERPINA1 , APOB, ALB, ASGR1 , TF, ITIH1 ), CoP (CA4, CD24, CLDN6) and Co (KRT7, ADIRF, CEACAM6). Thus, HO and CoP clearly represent different types of progenitors despite sharing the expression of key genes such as AFP. Further detailed tSNE analyses refines these observations by showing that HO and primary hepatoblast can be distributed in separate clusters. Nonetheless, heat map visualisation reveals that HO and primary hepatoblasts broadly express the same markers but at different levels (fig 4F). Principal component analysis (PCA) demonstrated that HO and PH are highly similar (Fig 4D), expressing similar levels of genes such as apolipoprotein A1 (APOA1 ), asialoglycoprotein receptor 1 (ASGR1 ) and albumin (ALB). The genes driving the differences between the two populations included higher expression of genes from the Methallothionein sub-family of genes (MT1 H, MT1A, MT1G, MT1 E, MT1 F, MT2A, MT1X) and also higher expression of some markers of immaturity such as AFP. In comparison, the HO had higher levels of expression of metabolic enzymes such as CYP1 A1 and CYP2E1 , and metabolic proteins such as fatty acid synthase (FASN) and fatty acid desaturase (FADS1 ). GO term analyses show that these genes mainly correspond to hepatic function. Considered together these results show that HOs strongly resemble their in vivo counterparts and that expansion in vitro affects only a limited number of metabolic functions.
Hepatoblast Organoids can differentiate into cells expressing cholangiocyte markers after addition of TGFb in the absence of Wnt3a.
We then decided to explore the capacity of HO to differentiate into cholangiocytes. For that we studied the effect of growth factors that have been shown to drive differentiation of hepatoblasts into biliary cells in vivo especially TGFb. These analyses uncovered that withdrawal of TGFb inhibitor A8301 and Wnt3a, and the simultaneous addition of TGFb, resulted in a significant increase in the biliary marker KRT19. More precisely, HO grown for seven days in the presence of TGFb without WNT express biliary markers at a level similar to FBO while losing hepatoblast markers including ALB and AFP (Fig 4B). These results were further supported by the changes in protein expression shown by immunostaining and in secreted protein detected by ELISA (Fig 4C). Taken together, these data suggest that TGFbeta could drive differentiation of HO toward the biliary lineages. To further confirm this possibility, we compared the single cell transcriptomic profile of HO grown in the presence of TGFb with that of HO and FBO (Fig 5D). scRNAseq analysis demonstrated TGFb treated caused the cells to cluster separated from HO, and to adopt a profile similar to the FBO.
Furthermore, HO, HO+TGFb and FBO cells ordered in pseudotime, with PFL and HO transitioning to CoP to Co and HO-TGF, confirming TGF beta treatment drives HO differentiation toward the biliary lineage following a precise temporal sequence of gene expression events (Fig 4E-G). Taken together, these results suggest that HO display the capacity to differentiate into cells expressing biliary markers in vitro. This process occurs in the absence of WNT signalling and in the presence of TGFb which promotes a biliary specification.
Absence of Wnt signalling drives differentiation of hepatoblast toward foetal hepatocytes.
In parallel to the experiments outlined above, we also explored culture conditions that could promote the differentiation of hepatoblast towards hepatocytes. To this end, we found that removing Wnt3a to promote differentiation and transferring into a hepatocyte specific medium (Hepatozyme with oncostatin-M), resulted in hepatoblast organoids undergoing a phenotypic shift (Fig 5A and B) and increased the expression of more mature hepatocytic markers such as G6PC, and in functional proteins such as alpha-1 -antitrypsin Figure 5C). We referred to these organoids as HO-HZ. Additionally, hepatoblast markers of the cholangiocyte lineage, such as cytokeratin-19 or gamma-glutamyl transferase, were further down regulated, demonstrating a robust change in cell fate from hepatoblast to a more foetal/mature hepatocyte (Fig 5C-G). Analysis of medium protein content also demonstrated a significant increase in proteins such as alpha-1 -antitrypsin, and down regulation in proteins associated with immaturity, such as alpha fetoprotein (Fig 5D). Of note, differentiated cell types lost proliferative capacity in vitro after treatment, even if treatment is withdrawn and original culture conditions are restored (data not shown). This further indicates a terminal differentiation of the cells in this system and mirrors the behaviour of primary cells cultured in vitro. Finally, we also observed that HO-HZ acquire functional activity absence in HO such as lipid uptake or CYP3A4 activity. To validate these results, HO-HZ were analysed at the single cell level and compared with HO and the PFL. PCA demonstrated that indeed absence of WNT induces differentiation of HO while heat map representation of the cells differentially expressed between each cluster suggest that the identity of HO-HZ differ from HO and PFL. Interestingly, genes specifically expressed in HO-HZ are in majority expressed at the protein level in adult hepatocytes (See table S1 ). GO analyses show that these genes are involved in metabolic pathways, serum and immune protein production, which all correspond to function fulfilled by the adult liver (Fig S6F). Markedly reduced expression of cell cycle genes such as CDK1 and Mi67 indicate that HO-HZ have stopped proliferating. Finally, pseudotime ordering revealed the precise gene expression signature corresponding to the transition between PFL/HO to foetal hepatocytes confirming dynamic decrease of key hepatoblast markers such as AFP and an induction of more mature markers such as G6PC and SERPINA1 (Fig 5F and G). Taken together, these results show that absence of WNT drives differentiation of HO towards a hepatocyte phenotype associated with the induction of specific marker, decrease in proliferation capacity and acquisition of specific function.
Hepatoblast organoids have a distinct identity from fetal hepatocyte and adult liver organoids.
Two previous reports have described culture systems to grow organoids from the liver. One approach consists of deriving organoids from the intrahepatic biliary epithelium of the adult liver (BO) (Huch M, et al. Cell. 2015;160(1-2):299-312) while the other method described organoids from the human fetal liver of the second trimester (fetal hepatocyte organoids or FHO; Hu H et al. Cell. 2018;175(6):1591-1606.e19).
Importantly, these organoids have different requirement for growth factors (See Table 2) while BO maintain a strong biliary identity and thus have a limited capacity to generate hepatocytes. To confirm this aspect, we
derived two BO lines and then differentiated them toward hepatocyte-like cells them as described previously (Hu H et al supra, Huch et al supra). The resulting differentiated BO (DBO) secrete very low levels of ALB, AFP, APOB, and A1AT when compared to HBO (Fig. 6) thereby reinforcing recent reports (Hu H et al supra, Huch et al supra). Comparison at a single cell level demonstrated that BO and DBO overlap while forming distinct clusters from HBO or primary fetal liver hepatoblasts (PFL). The main difference between these populations was the high level of cholangiocytic markers such as KRT19 and KRT7 in BO/DBO in contrast to the high level of hepatoblast markers such as ALB and ARG1 in PFL/HBO. Next we compared the transcriptional profile of DBO and FHO, and HBO (Hu et al supra). PCA demonstrated separation into three categories; biliary (DBO), hepatocytic (primary adult hepatocytes, primary fetal hepatocytes and fetal hepatocyte organoids), and hepatoblast (HBO) according to genes associated with either biliary ( KRT19 , MUC20) or hepatocyte {ALB, AFP) identity. Interestingly, HBO occupied an intermediate position by coexpressing both categories of markers (Fig. 7A, 7B) while primary hepatocytes displayed adult markers involved in clotting (F2, F10, PROC, FGA), immunity (C2, C3), and metabolic processes (APOB, APOH, ALDOB, ABCA1 , PAH). Taken together these observations (summarized in Table 2) show that HBO have a unique transcriptional profile when compared to previously derived liver organoids and thus they provide a novel complementary model system for human fetal liver development
Hepatoblast Organoids can differentiate into biliary cells and hepatocytes in vivo
Capacity to differentiate into hepatocytes and cholangiocytes represents a hallmark of hepatoblast function. To confirm this key characteristic, we transplanted HBO into immunodeficient mice. We first generated HBO expressing a tdTomato reporter using lentiviral transduction. These fluorescent cells were transplanted into mice using an approach recently developed for primary hepatocytes (Stevens et al Sci Transl Med.
2017;9(399)). In sum,“human organoid tissues” were assembled by embedding HBO and human umbilical vein endothelial cells (HUVECs) in a fibrin-matrigel hydrogel (20:1 fibrimmatrigel mixture). The resulting organoid tissues were sutured into the inguinal fat pad of FRG mice which experience progressive liver failure unless administered nitisinone (NTBC) (Wilson et al Stem Cell Res. 2014;13(3 Pt A):404-412). The transplanted mice were cycled on and off NTBC over the course of the experiment to induce chronic liver disease, which has been shown to promote hepatic engraftment and growth (Stevens et al supra, Azuma et al Nat Biotechnol. 2007;25(8):903-910). After 27 days, implants were recovered and red fluorescent cells could be observed in all the grafts, indicating that HBO had engrafted efficiently. Hematoxylin & eosin staining of explanted tissue sections revealed the presence of numerous nodules resembling densely packed hepatocytes, as well as biliary epithelial-like cells assembled into structures reminiscent of bile ducts.
To assess the maturation state of grafted organoids, we then stained tissue sections for AFP and KRT18. Organoids in engineered tissues stained positively for both KRT18 and AFP at the time of implant, but AFP was either not identified or markedly decreased in KRT18-positive grafted organoids suggesting
differentiation of hepatoblasts into hepatocytes. Accordingly, numerous cells in hepatic nodule stained positively for ARG1 , A1 AT, and ALB while human albumin was identified in mouse blood serum suggesting functional activity and successful integration with the host vasculature (Fig. 8A). Finally, KRT18-positive nodules also contained cells expressing KRT19 (11 % had some cells that expressed KRT19 and 18% were nearly entirely positive for KRT19 with characteristic bile duct morphology; Fig. 8B). Together, our results
demonstrate that HOs have the capacity to differentiate into cells characterized by hepatocyte and biliary markers after transplantation into mouse model for liver disease.
We studied the effect of growth factors that have been shown to drive differentiation of hepatoblasts into biliary cells in vivo, particularly TGF which is known to control formation of the ductal plate in vivo (Clotman et al Genes Dev. 2005;19(16):1849-1854; Seth et al Development. 2014;141 (3):538-54; Ober et al J Hepatol. 2018;68(5):1049-1062. These analyses demonstrated that the withdrawal of TGF inhibitor A83-01 and the simultaneous addition of TGF increased biliary markers such as KRT19 and decreased hepatoblast markers including ALB.
The above experiments demonstrate the derivation of the first human hepatoblast organoid model. This model offers an exciting opportunity to study human foetal liver development in vitro long term and in three dimensions, allowing assessment of the behaviour and characteristics of previously poorly understood cell types that have been difficult to investigate until now.
Hepatoblast organoids crucially exhibit high levels of function in vitro, continuing to express the key genes involved in hepatic function found in vivo and demonstrating their robustness as a representative in vitro model. Thus, this new model gives insight and opportunity to study human hepatoblast physiology, cell cycle characteristics, and potential insight to pathological mechanisms such as malignant transformation. A key concept of hepatoblast cells is the co-expression of hepatocytic and cholangiocytic markers. An interesting finding is that whilst the hepatocytic markers (ALB/A1 AT) are expressed at similar levels to in vivo, the cholangiocytic markers are more lowly expressed compared to cholangiocytes. In addition, human hepatoblast organoids transplanted into mice demonstrated the capacity to differentiate toward either hepatocytic or biliary fates while maintaining functional capacity.
The genetic and transcriptomic stability along with the high proliferative capacity of the organoids also allows for large scale expansion of a single biological sample, thus enabling in depth analysis of the system and allowing complex research questions to be investigated in the same system over long periods. Similarly, the homogeneity and high potential for cell expansion enables the potential of generating large scale biobanks of tissue that can be selectively utilised.
Single cell assessment of the primary foetal liver has allowed us to demonstrate the homogeneous character of the early hepatoblast, and also the early colonization of the liver by immune cells. This finding gives insight into how the early liver is populated, with a large population of bipotent dividing hepatoblast cells. Comparison with organoid demonstrated a similarly homogeneous population of self-organizing proliferative cells, underlining the broad nature of this cell type. The ability to differentiate these cells in vitro will allow future detailed analysis of how this process occurs, and greater interrogation of the pathways involved.
The ability to generate cells that could be expanded and transplanted into tissue matched recipients could represent an exciting opportunity to manage patients requiring liver transplant. The efficiency of organoid generation alongside a ready supply of tissue offers the potential of rapidly generating a large-scale biobank of hepatoblast organoids that could cover the majority of variations in tissue markers such as HLA. The ability of the organoids to produce serum albumin and differentiate to daughter cell types present an exciting
prospect of developing a future bridge therapy for end stage liver disease, restoring some level of hepatic function through tissue matched donor organoids.
The controlled differentiation of hepatoblasts into either of the two specific daughter cell types offers a potentially unique insight into these mechanisms that can be further explored. Furthermore, the only current possibility to maintain hepatocytes in vitro is to await donor samples removed from deceased donors. The ability to generate large numbers of either cholangiocytes or hepatocytes on demand in vitro from multiple biological backgrounds, represents the first self-replenishing source of such cells. This again opens exciting possibilities to interrogate the development of these cells and opportunities for drug discovery/ screening. Overall, this new organoid system represents an exciting system to model development, cell fate decisions, and opportunities for regenerative medicine.
Reagent HO-M CO-M Stock concentration Final concentration
Basal medium:
DMEM/F12 500ml 500ml
Glutamax 5ml 5ml 100% 1 % (by vol) HEPES 5ml 5ml 100% 1 % (by vol)
Penicillin/streptomycin 5ml 5ml 100% 1 % (by vol)
B27 10ml 10ml 100% 2% (by vol)
Nicotinamide 10ml 10ml 1 M 20mM
N-acetylcysteine 2ml 2ml 0.5M 2mM
Complete medium:
Basal medium 19ml 19ml
DMEM 25ml
Wnt3a 25ml - 100% 50% (by vol.)
R-spondin 5ml 5ml 100% 10% (by vol.)
EGF 25uL 25ul 100ug/ml 50ng/ml
A8301 50uL 50uL 50mM 50uM
Y27632 50ul 50ul 10mM 10uM
Table 1
Key: - = low expression, + moderate expression, ++ high expression, +++ = very high expression
Table 2: Summary of human hepatic organoids
References
Oertel, M. et ai. (2003) Hepatology (Baltimore, Md.), 37(5), pp. 994-1005. doi: 10.1053/jhep.2003.50183 Shiojiri, N. et al. (2001 ) Laboratory Investigation. The United States and Canadian Academy of Pathology, Inc., 81 , p. 17. Available at: http://dx.doi.org/10.1038/labinvest.3780208.
Clotman, F. et ai. (2005) Genes & development, 19(16), pp. 1849-54. doi: 10.1 101/gad.340305.
Yang, L. et ai. (2017) Hepatology (Baltimore, Md.), 66(5), pp. 1387-1401. doi: 10.1002/hep.29353.
Cruz-Acuha, R. et ai. (2017) Nature Cell Biology. Nature Publishing Group, 19, p. 1326. Available at:
http://dx.doi.Org/10.1038/ncb3632.
van de Wetering, M. et ai. (2015) Cell, 161 (4), pp. 933-945. https://doi.Org/10.1016/j.cell.2015.03.053. Schwank, G. et ai. (2013) Cell Stem Cell, 13(6), pp. 653-658. doi:
https://doi.Org/10.1016/j.stem.2013.1 1.002.
Huch, M. et ai. (2015) Cell. Cell Press, 160(1-2), pp. 299-312. doi: 10.1016/j.cell.2014.1 1 .050.
Yui, S. et ai. (2018) Cell Stem Cell. Cell Press, 22(1 ), p. 35-49. e7. doi: 10.1016/j.stem.2017.1 1.001.
Kraiczy, J. et ai. (2017) Gut. Available at: http://gut.bmj.com/content/early/2017/1 1/15/gutjnl-2017- 314817. abstract.
Huch, M. and Koo, B.-K. (2015) Development, 142(18), p. 31 13 LP-3125. Available at:
http://dev.biologists.org/content/142/18/31 13. abstract.
Decaens, T. et ai. (2008) Hepatology (Baltimore, MdJ 47(1 ), pp. 247-58. doi: 10.1002/hep.21952.
Tan, X. et ai. (2008) Hepatology (Baltimore, Md.), 47(5), pp. 1667-1679. doi: 10.1002/hep.22225.
Younossi, Z. M. et ai. (2016) Hepatology, 64(1 ), pp. 73-84. doi: 10.1002/hep.28431.
Deng, X. et ai. (2018) Cell Stem Cell, 23(1 ), p. 1 14-122.e3. doi: https://doi.Org/10.1016/j.stem.2018.05.022. Hopkinson, C. and Allen, E. (2017) Annual Report on Liver Transplantation 2016/2017. NHS Blood and Transplant. Cambridge, UK.
Katsura, N. et ai. (2002) Journal of Surgical Research, 106(1 ), pp. 1 15-123. doi:
https://doi.Org/10.1006/jsre.2002.6446.
Blouin, A., Bolender, R. P. and Weibel, E. R. (1977) The Journal of Cell Biology, 72(2), p. 441 LP-455. Available at: http://jcb.rupress.Org/content/72/2/441.abstract.
Gille, C. et al. (2010) Molecular Systems Biology, 6(1 ). Available at:
http://msb.embopress.Org/content/6/1/41 1.abstract.
Haruna, Y. et ai. (1996) Hepatology. Wiley Online Library, 23(3), pp. 476-481. doi: 10.1002/hep.510230312. Huch M et al Cell. (2015) 160(1-2):299-312. doi: 10.1016/j.cell.2014.1 1.050.
Hu H et ai. Cell. 2018; 175(6):1591-1606.e19. doi:https://doi.org/10.1016/j.cell.2018.1 1.013.
Claims
1. A method for producing an expanded population of human hepatoblasts in vitro comprising:
(i) providing a population of isolated immature human liver cells and;
(ii) culturing the population in a hepatoblast expansion medium comprising epidermal growth factor (EGF), a TGF inhibitor, a non-canonical Wnt signalling potentiator, a canonical Wnt potentiator and a ROCK inhibitor, to produce an expanded population of human hepatoblasts.
2. A method for producing a population of human cholangiocytes in vitro comprising
(i) producing an expanded population of human hepatoblasts by a method according to claim 1 ; and,
(ii) culturing the expanded population in a differentiation medium comprising TGF to produce a population of cholangiocytes.
3. A method for producing a population of human hepatocytes in vitro comprising
(i) producing an expanded population of hepatoblasts by a method according to claim 1 ; and,
(ii) culturing the expanded population in a hepatocyte culture medium to produce a population of human hepatocytes.
4. A method according to any one of the preceding claims wherein the expanded population forms organoids in the hepatoblast expansion medium.
5. A method according to any one of the preceding claims wherein immature human liver cells are foetal or neonatal liver cells.
6. A method according to any one of the preceding claims wherein the hepatoblast expansion medium is a chemically defined medium consisting of a basal medium, epidermal growth factor (EGF), a TGF inhibitor, a non-canonical Wnt signalling potentiator, a canonical Wnt potentiator and a ROCK inhibitor,
7. A method according to any one of the preceding claims wherein the differentiation medium is a chemically defined medium consisting of a basal medium, epidermal growth factor (EGF), TGF , a non- canonical Wnt signalling potentiator, and a ROCK inhibitor.
8. A method according to any one of the preceding claims wherein the TGF inhibitor is A8301.
9. A method according to any one of the preceding claims wherein the non-canonical Wnt signalling potentiator is R-spondin.
10. A method according to according to any one of the preceding claims wherein the ROCK inhibitor is Y-27632.
1 1. A method according to any one of the preceding claims wherein the canonical Wnt potentiator is Wnt3a.
12. An isolated population of hepatoblasts, biliary cells, cholangiocyte progenitors or cholangiocytes produced by a method according to any one of claims 1 to 1 1.
13. A biocompatible scaffold comprising an isolated population according to claim 12.
14. A method of treating a liver disease, for example a hepatocytic or biliary disorder, comprising administering an isolated population according to claim 12 or a scaffold according to claim 13 to an individual in need thereof.
15. An isolated population according to claim 12 or a scaffold according to claim 13 for use in a method of treating a liver disease in an individual in need thereof.
16. A method according to claim 14 wherein the liver disease is haemophilia.
17. An isolated population or a scaffold for use according to claim 15 wherein the liver disease is haemophilia.
18. A method of screening comprising;
contacting an isolated population according to claim 12 or a scaffold according to claim 13 with a test compound, and;
determining the effect of the test compound on the population or scaffold and/or the effect of the population or scaffold on the test compound.
19. A method according to claim 18 wherein the population is contacted with the test compound is in the form of organoids.
20. A kit for the production of an expanded population of hepatoblasts comprising a hepatoblast expansion medium that comprises epidermal growth factor (EGF), a TGF inhibitor, non-canonical Wnt signalling potentiator, a canonical Wnt potentiator and a ROCK inhibitor.
21. A kit according to claim 20 further comprising a hepatocyte culture medium and/or a differentiation medium comprising TGF .
22. Use of a hepatoblast expansion medium for the in vitro expansion of hepatoblasts, wherein the hepatoblast expansion medium comprises epidermal growth factor (EGF), a TGF inhibitor, non-canonical Wnt signalling potentiator, a canonical Wnt potentiator and a ROCK inhibitor.
Applications Claiming Priority (2)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
GBGB1813127.6A GB201813127D0 (en) | 2018-08-10 | 2018-08-10 | Hepatoblast Organoids |
GB1813127.6 | 2018-08-10 |
Publications (1)
Publication Number | Publication Date |
---|---|
WO2020030822A1 true WO2020030822A1 (en) | 2020-02-13 |
Family
ID=63667213
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/EP2019/071587 WO2020030822A1 (en) | 2018-08-10 | 2019-08-12 | Hepatoblast organoids |
Country Status (2)
Country | Link |
---|---|
GB (1) | GB201813127D0 (en) |
WO (1) | WO2020030822A1 (en) |
Cited By (1)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO2023093808A1 (en) * | 2021-11-24 | 2023-06-01 | 南京昕瑞再生医药科技有限公司 | Compound and method for hepatocyte amplification |
Citations (2)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
EP2412800A1 (en) * | 2010-07-29 | 2012-02-01 | Koninklijke Nederlandse Akademie van Wetenschappen | Liver organoid, uses thereof and culture method for obtaining them |
WO2016207621A1 (en) * | 2015-06-22 | 2016-12-29 | Cambridge Enterprise Limited | In vitro production of cholangiocytes |
-
2018
- 2018-08-10 GB GBGB1813127.6A patent/GB201813127D0/en not_active Ceased
-
2019
- 2019-08-12 WO PCT/EP2019/071587 patent/WO2020030822A1/en active Application Filing
Patent Citations (2)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
EP2412800A1 (en) * | 2010-07-29 | 2012-02-01 | Koninklijke Nederlandse Akademie van Wetenschappen | Liver organoid, uses thereof and culture method for obtaining them |
WO2016207621A1 (en) * | 2015-06-22 | 2016-12-29 | Cambridge Enterprise Limited | In vitro production of cholangiocytes |
Non-Patent Citations (65)
Title |
---|
"Human Cell Culture Protocols (Methods in Molecular Medicine S.", 9 December 2004, HUMANA PRESS INC. |
"NCBI", Database accession no. NM_001178130.1 |
AZUMA ET AL., NAT BIOTECHNOL., vol. 25, no. 8, 2007, pages 903 - 910 |
BLOUIN, A.BOLENDER, R. P.WEIBEL, E. R., THE JOURNAL OF CELL BIOLOGY, vol. 72, no. 2, 1977, pages 441 LP - 455, Retrieved from the Internet <URL:http://jcb.rupress.org/content/72/2/441.abstract> |
BODINE ET AL., BONE, vol. 44, no. 6, June 2009 (2009-06-01), pages 1063 - 8 |
CLOTMAN ET AL., GENES DEV., vol. 19, no. 16, 2005, pages 1849 - 1854 |
CLOTMAN, F. ET AL., GENES & DEVELOPMENT, vol. 19, no. 16, 2005, pages 1849 - 54 |
COGHLAN ET AL., CHEM BIOL., vol. 7, no. 10, October 2000 (2000-10-01), pages 793 - 803 |
COLIN H. BECKWITT ET AL: "Liver 'organ on a chip'", EXPERIMENTAL CELL RESEARCH, vol. 363, no. 1, 1 February 2018 (2018-02-01), AMSTERDAM, NL, pages 15 - 25, XP055534749, ISSN: 0014-4827, DOI: 10.1016/j.yexcr.2017.12.023 * |
DECAENS, T. ET AL., HEPATOLOGY (BALTIMORE, MD.), vol. 47, no. 5, 2008, pages 1667 - 1679 |
DENG, X. ET AL., CELL STEM CELL, vol. 23, no. 1, 2018, pages 114 - 122.e3, Retrieved from the Internet <URL:https://doi.org/10.1016/j.stem.2018.05.022> |
FOTIOS SAMPAZIOTIS ET AL: "Directed differentiation of human induced pluripotent stem cells into functional cholangiocyte-like cells", NATURE PROTOCOLS, vol. 12, no. 4, 23 March 2017 (2017-03-23), GB, pages 814 - 827, XP055534582, ISSN: 1754-2189, DOI: 10.1038/nprot.2017.011 * |
FREED, C.R. ET AL., CELL TRANSPLANT, vol. 6, 1997, pages 201 - 202 |
FREED, C.R., NEW ENGLAND JOURNAL OF MEDICINE, vol. 327, 1992, pages 1549 - 1555 |
GILBERT ET AL., BIOORG MED CHEM LETT., vol. 20, no. 1, 1 January 2010 (2010-01-01), pages 366 - 70 |
GILLE, C. ET AL., MOLECULAR SYSTEMS BIOLOGY, vol. 6, no. 1, 2010, Retrieved from the Internet <URL:http://msb.embopress.org/content/6/1/411.abstract> |
GJOREVSKI ET AL., NATURE, vol. 539, 2016, pages 560 - 564 |
HARUNA, Y. ET AL.: "Hepatology", vol. 23, 1996, WILEY ONLINE LIBRARY, pages: 476 - 481 |
HEDWIG S. KRUITWAGEN ET AL: "Long-Term Adult Feline Liver Organoid Cultures for Disease Modeling of Hepatic Steatosis", STEM CELL REPORTS, vol. 8, no. 4, 1 April 2017 (2017-04-01), United States, pages 822 - 830, XP055534697, ISSN: 2213-6711, DOI: 10.1016/j.stemcr.2017.02.015 * |
HO WY ET AL., J IMMUNOL METHODS, vol. 310, 2006, pages 40 - 52 |
HOPKINSON, C.ALLEN, E.: "Annual Report on Liver Transplantation 2016/2017", 2017, NHS BLOOD AND TRANSPLANT |
HU H ET AL., CELL, vol. 175, no. 6, 2018, pages 1591 - 1606.e19, Retrieved from the Internet <URL:https://doi.org/10.1016/j.cell.2018.11.013> |
HUCH, M.KOO, B.-K., DEVELOPMENT, vol. 142, no. 18, 2015, pages 3113 LP - 3125, Retrieved from the Internet <URL:http://dev.biologists.org/content/142/18/3113.abstract> |
K. TURKSEN: "Human Embryonic Stem Cell Protocols", 2006 |
KATSURA, N. ET AL., JOURNAL OF SURGICAL RESEARCH, vol. 106, no. 1, 2002, pages 115 - 123 |
KAZUYA ANZAI ET AL: "Foetal hepatic progenitor cells assume a cholangiocytic cell phenotype during two-dimensional pre-culture", SCIENTIFIC REPORTS, vol. 6, no. 1, 23 June 2016 (2016-06-23), XP055534175, DOI: 10.1038/srep28283 * |
KLAUS ET AL., NAT. REV. CANCER, vol. 8, 2008, pages 387 - 398 |
KOMIYA, Y.HABAS, R., ORGANOGENESIS, vol. 4, 2008, pages 68 - 75 |
KORDOWER ET AL., NEW ENGLAND JOURNAL OF MEDICINE, vol. 332, 1995, pages 1118 - 1124 |
KRAICZY, J. ET AL., GUT, vol. 19, 2017, pages 1326, Retrieved from the Internet <URL:http://gut.bmj.com/content/early/2017/11/15/gutjnl-2017-314817.abstract> |
LAURA BROUTIER ET AL: "Culture and establishment of self-renewing human and mouse adult liver and pancreas 3D organoids and their genetic manipulation", NATURE PROTOCOLS, vol. 11, no. 9, 25 August 2016 (2016-08-25), GB, pages 1724 - 1743, XP055534706, ISSN: 1754-2189, DOI: 10.1038/nprot.2016.097 * |
LAURA BROUTIER ET AL: "Human primary liver cancer-derived organoid cultures for disease modeling and drug screening", NATURE MEDICINE, vol. 23, no. 12, 13 November 2017 (2017-11-13), New York, pages 1424 - 1435, XP055534757, ISSN: 1078-8956, DOI: 10.1038/nm.4438 * |
LE BLANC ET AL., LANCET, vol. 363, no. 9419, 1 May 2004 (2004-05-01), pages 1439 - 41 |
LIAO ET AL., J. CARDIOVASC PHARMACOL., vol. 50, no. 1, 2007, pages 17 - 24 |
LINDVALL, O., MOV. DISORD., vol. 13, no. 1, 1998, pages 83 - 7 |
LIU ET AL., ANGEW CHEM INT ED ENGL., vol. 44, no. 13, 18 March 2005 (2005-03-18), pages 1987 - 90 |
MATHAPATI SANTOSH ET AL: "Small-Molecule-Directed Hepatocyte-Like Cell Differentiation of Human Pluripotent Stem Cells", CURRENT PROTOCOLS IN STEM CELL BIOLOGY 2012,, vol. 38, 17 August 2016 (2016-08-17), pages 1.6.1 - 1.6.18, XP009509972, ISSN: 1938-8969, DOI: 10.1002/CPSC.13 * |
MERITXELL HUCH ET AL: "In vitro expansion of single Lgr5+ liver stem cells induced by Wnt-driven regeneration", NATURE, vol. 494, no. 7436, 1 January 2013 (2013-01-01), pages 247 - 250, XP055058163, ISSN: 0028-0836, DOI: 10.1038/nature11826 * |
MINA OGAWA ET AL: "Directed differentiation of cholangiocytes from human pluripotent stem cells. Supplementary Text and Figures.", NATURE BIOTECHNOLOGY, 13 July 2015 (2015-07-13), pages 853 - 861, XP055282317, Retrieved from the Internet <URL:http://www.nature.com/nbt/journal/v33/n8/extref/nbt.3294-S1.pdf> [retrieved on 20160621], DOI: 10.1038/nbt.3294 * |
MIYABAYASHI ET AL., PNAS USA, vol. 104, no. 18, 2007, pages 7444 - 7448 |
MOLENAAR ET AL., CELL, vol. 86, no. 3, 9 August 1996 (1996-08-09), pages 391 - 9 |
MOON ET AL., NAT. REV. GENET., vol. 5, 2004, pages 691 - 701 |
MOORE, G.E.WOODS L.K., TISSUE CULTURE ASSOCIATION MANUAL, vol. 3, 1976, pages 503 - 508 |
NIEHRS ET AL., NAT REV MOL. CELL BIOL., vol. 13, 2012, pages 763 - 779 |
OBER ET AL., J HEPATOL., vol. 68, no. 5, 2018, pages 1049 - 1062 |
OERTEL, M. ET AL., HEPATOLOGY (BALTIMORE, MD.), vol. 37, no. 5, 2003, pages 994 - 1005 |
OHKAWARA ET AL., DEV DYN, vol. 240, no. 1, 2011, pages 188 - 194 |
PATEL, V. ET AL., HUM. MOL. GENET., vol. 17, 2008, pages 1578 - 1590 |
PETERSONLORING: "Human Stem Cell Manual: A Laboratory Guide", 2012, ACADEMIC PRESS |
PRICE ET AL., FOCUS, vol. 25, 2003, pages 3 - 6 |
R. FRESHNEY: "Culture of Animal Cells: A Manual of Basic Technique", 2 August 2005, JOHN WILEY & SONS INC |
SATO ET AL., NAT MED., vol. 10, no. 1, January 2004 (2004-01-01), pages 55 - 63 |
SCHWANK, G. ET AL., CELL STEM CELL, vol. 13, no. 6, 2013, pages 653 - 658, Retrieved from the Internet <URL:https://doi.org/10.1016/j.stem.2013.11.002> |
SETH ET AL., DEVELOPMENT, vol. 141, no. 3, 2014, pages 538 - 54 |
SHIOJIRI, N. ET AL.: "Laboratory Investigation", vol. 81, 2001, THE UNITED STATES AND CANADIAN ACADEMY OF PATHOLOGY, INC., pages: 17 |
STEVENS ET AL., SCI TRANSL MED., vol. 9, 2017, pages 399 |
STRAZZABOSCO, M.SOMLO, S., GASTROENTEROLOGY, vol. 140, 2011 |
TOLOSA ET AL., CELL TRANSPLANT., vol. 23, no. 10, 2014, pages 1229 - 1242 |
TOUBOUL THOMAS ET AL: "Stage-specific regulation of the WNT/[beta]-catenin pathway enhances differentiation of hESCs into hepatocytes", JOURNAL OF HEPATOLOGY, ELSEVIER, AMSTERDAM, NL, vol. 64, no. 6, 26 February 2016 (2016-02-26), pages 1315 - 1326, XP029538552, ISSN: 0168-8278, DOI: 10.1016/J.JHEP.2016.02.028 * |
VAN DE WETERING, M. ET AL., CELL, vol. 161, no. 1-2, 2015, pages 933 - 945, Retrieved from the Internet <URL:https://doi.org/10.1016/j.cell.2015.03.053> |
WILLIAMS, G.M. ET AL., EXP. CELL RESEARCH, vol. 89, 1974, pages 139 - 142 |
WILSON ET AL., STEM CELL RES., vol. 13, no. 3, 2014, pages 404 - 412 |
XINPING TAN ET AL: "[beta]-Catenin deletion in hepatoblasts disrupts hepatic morphogenesis and survival during mouse development", HEPATOLOGY, vol. 47, no. 5, 7 April 2008 (2008-04-07), US, pages 1667 - 1679, XP055534724, ISSN: 0270-9139, DOI: 10.1002/hep.22225 * |
YANG, L. ET AL., HEPATOLOGY (BALTIMORE, MD.), vol. 66, no. 5, 2017, pages 1387 - 1401 |
YOUNOSSI, Z. M. ET AL., HEPATOLOGY, vol. 64, no. 1, 2016, pages 73 - 84 |
Cited By (1)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO2023093808A1 (en) * | 2021-11-24 | 2023-06-01 | 南京昕瑞再生医药科技有限公司 | Compound and method for hepatocyte amplification |
Also Published As
Publication number | Publication date |
---|---|
GB201813127D0 (en) | 2018-09-26 |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
Hu et al. | Long-term expansion of functional mouse and human hepatocytes as 3D organoids | |
US20200172861A1 (en) | Culture Method for Organoids | |
US20200354683A1 (en) | Derivation of hepatic stem cells and mature liver cell types and uses thereof | |
Lees et al. | Transplantation of 3D scaffolds seeded with human embryonic stem cells: biological features of surrogate tissue and teratoma-forming potential | |
AU2019390669B2 (en) | Hepatocyte expansion methods | |
JP2023085514A (en) | Compositions and methods for obtaining organoids | |
JP7063624B2 (en) | Method for producing liver stem / progenitor cells from mature hepatocytes using small molecule compounds | |
US20220233605A1 (en) | Methods of making and using liver cells | |
US20210403874A1 (en) | Method for Producing Stem/Precursor Cells, By Using Low Molecular Weight Compound, From Cells Derived From Endodermal Tissue or Organ | |
JP2022534555A (en) | Method for producing hepatocyte | |
Huang et al. | Bioengineering of a CLiP‐derived tubular biliary‐duct‐like structure for bile transport in vitro | |
Jones et al. | Paediatric gastric organoids as a tool for disease modelling and clinical translation | |
WO2020030821A1 (en) | Biliary organoids | |
WO2020030822A1 (en) | Hepatoblast organoids | |
Pettinato et al. | Development of a scalable three-dimensional culture of human induced pluripotent stem cells-derived liver organoids | |
JP2024506414A (en) | How to proliferate cholangiocytes | |
WO2020203753A1 (en) | Method for manufacturing cell population including liver precursor cells | |
WO2020155909A1 (en) | Culture medium for screening and expanding liver stem cells and use thereof | |
Hayashi et al. | A basic study on self-reconstitution of alveolar epithelium-like cells by tissue stem cells in mouse lung | |
Tsang | In Vitro 3D Liver Model Utilizing iPSC-Induced Hepatocytes for Drug Testing | |
Banadka et al. | Organoids: inception and utilization of 3D organ models | |
Feuerborn | Functional improvement of stem cell derived hepatocyte-like cells after targeted FXR gene regulatory network manipulation | |
Ang et al. | Accelerated human liver progenitor generation from pluripotent stem cells by inhibiting formation of unwanted lineages | |
Rawlins et al. | Human lung development: recent progress and new challenges |
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: 19758639 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: 19758639 Country of ref document: EP Kind code of ref document: A1 |