WO2011130217A1 - Induced pluripotent stem cells and uses thereof - Google Patents
Induced pluripotent stem cells and uses thereof Download PDFInfo
- Publication number
- WO2011130217A1 WO2011130217A1 PCT/US2011/032044 US2011032044W WO2011130217A1 WO 2011130217 A1 WO2011130217 A1 WO 2011130217A1 US 2011032044 W US2011032044 W US 2011032044W WO 2011130217 A1 WO2011130217 A1 WO 2011130217A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- cell
- mutation
- agent
- stem cell
- ptpnl
- Prior art date
Links
- 210000004263 induced pluripotent stem cell Anatomy 0.000 title description 59
- 210000004027 cell Anatomy 0.000 claims abstract description 202
- 230000035772 mutation Effects 0.000 claims abstract description 189
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 claims abstract description 105
- 201000010099 disease Diseases 0.000 claims abstract description 92
- 210000000130 stem cell Anatomy 0.000 claims abstract description 92
- 210000001778 pluripotent stem cell Anatomy 0.000 claims abstract description 85
- 210000001082 somatic cell Anatomy 0.000 claims abstract description 55
- 101150084750 1 gene Proteins 0.000 claims abstract description 41
- 238000011282 treatment Methods 0.000 claims abstract description 34
- 108090000623 proteins and genes Proteins 0.000 claims description 135
- 102000004169 proteins and genes Human genes 0.000 claims description 103
- 235000018102 proteins Nutrition 0.000 claims description 100
- 210000004413 cardiac myocyte Anatomy 0.000 claims description 98
- 239000003795 chemical substances by application Substances 0.000 claims description 95
- 208000010708 Noonan syndrome with multiple lentigines Diseases 0.000 claims description 68
- 208000005101 LEOPARD Syndrome Diseases 0.000 claims description 65
- 206010062901 Multiple lentigines syndrome Diseases 0.000 claims description 64
- 102100033019 Tyrosine-protein phosphatase non-receptor type 11 Human genes 0.000 claims description 57
- 238000000034 method Methods 0.000 claims description 56
- 206010029748 Noonan syndrome Diseases 0.000 claims description 53
- 101001087416 Homo sapiens Tyrosine-protein phosphatase non-receptor type 11 Proteins 0.000 claims description 49
- 238000006467 substitution reaction Methods 0.000 claims description 49
- 101150048674 PTPN11 gene Proteins 0.000 claims description 46
- 210000002950 fibroblast Anatomy 0.000 claims description 45
- 125000000539 amino acid group Chemical group 0.000 claims description 39
- 230000000694 effects Effects 0.000 claims description 38
- 230000001939 inductive effect Effects 0.000 claims description 38
- 230000004069 differentiation Effects 0.000 claims description 34
- 241000282414 Homo sapiens Species 0.000 claims description 27
- 101710126211 POU domain, class 5, transcription factor 1 Proteins 0.000 claims description 27
- OUYCCCASQSFEME-QMMMGPOBSA-N L-tyrosine Chemical compound OC(=O)[C@@H](N)CC1=CC=C(O)C=C1 OUYCCCASQSFEME-QMMMGPOBSA-N 0.000 claims description 25
- OUYCCCASQSFEME-UHFFFAOYSA-N tyrosine Natural products OC(=O)C(N)CC1=CC=C(O)C=C1 OUYCCCASQSFEME-UHFFFAOYSA-N 0.000 claims description 25
- 210000003958 hematopoietic stem cell Anatomy 0.000 claims description 24
- 102000003974 Fibroblast growth factor 2 Human genes 0.000 claims description 22
- 108090000379 Fibroblast growth factor 2 Proteins 0.000 claims description 22
- 230000000747 cardiac effect Effects 0.000 claims description 22
- 102000016914 ras Proteins Human genes 0.000 claims description 20
- 230000026731 phosphorylation Effects 0.000 claims description 19
- 238000006366 phosphorylation reaction Methods 0.000 claims description 19
- 230000019491 signal transduction Effects 0.000 claims description 19
- 239000003112 inhibitor Substances 0.000 claims description 18
- 230000001969 hypertrophic effect Effects 0.000 claims description 17
- 108010014186 ras Proteins Proteins 0.000 claims description 17
- 101100247004 Rattus norvegicus Qsox1 gene Proteins 0.000 claims description 16
- 230000035755 proliferation Effects 0.000 claims description 16
- 230000007423 decrease Effects 0.000 claims description 15
- 102100031480 Dual specificity mitogen-activated protein kinase kinase 1 Human genes 0.000 claims description 14
- 208000032839 leukemia Diseases 0.000 claims description 14
- 230000004083 survival effect Effects 0.000 claims description 14
- AYFVYJQAPQTCCC-UHFFFAOYSA-N Threonine Natural products CC(O)C(N)C(O)=O AYFVYJQAPQTCCC-UHFFFAOYSA-N 0.000 claims description 13
- 239000004473 Threonine Substances 0.000 claims description 13
- 206010007572 Cardiac hypertrophy Diseases 0.000 claims description 12
- 208000006029 Cardiomegaly Diseases 0.000 claims description 12
- 102100038895 Myc proto-oncogene protein Human genes 0.000 claims description 12
- -1 TERT Proteins 0.000 claims description 12
- 150000007523 nucleic acids Chemical class 0.000 claims description 12
- 101710135898 Myc proto-oncogene protein Proteins 0.000 claims description 10
- 241000288906 Primates Species 0.000 claims description 10
- 101150086694 SLC22A3 gene Proteins 0.000 claims description 10
- 101710150448 Transcriptional regulator Myc Proteins 0.000 claims description 10
- 238000004113 cell culture Methods 0.000 claims description 10
- 229930182817 methionine Natural products 0.000 claims description 10
- 125000001360 methionine group Chemical group N[C@@H](CCSC)C(=O)* 0.000 claims description 10
- 108010049955 Bone Morphogenetic Protein 4 Proteins 0.000 claims description 9
- 102100024505 Bone morphogenetic protein 4 Human genes 0.000 claims description 9
- 235000018417 cysteine Nutrition 0.000 claims description 9
- XUJNEKJLAYXESH-UHFFFAOYSA-N cysteine Natural products SCC(N)C(O)=O XUJNEKJLAYXESH-UHFFFAOYSA-N 0.000 claims description 9
- 125000000151 cysteine group Chemical group N[C@@H](CS)C(=O)* 0.000 claims description 9
- 206010020871 hypertrophic cardiomyopathy Diseases 0.000 claims description 9
- 102000039446 nucleic acids Human genes 0.000 claims description 9
- 108020004707 nucleic acids Proteins 0.000 claims description 9
- 229920001184 polypeptide Polymers 0.000 claims description 9
- 102000004196 processed proteins & peptides Human genes 0.000 claims description 9
- 108090000765 processed proteins & peptides Proteins 0.000 claims description 9
- 108700021430 Kruppel-Like Factor 4 Proteins 0.000 claims description 8
- 206010043276 Teratoma Diseases 0.000 claims description 8
- 108010073929 Vascular Endothelial Growth Factor A Proteins 0.000 claims description 8
- 102000005789 Vascular Endothelial Growth Factors Human genes 0.000 claims description 8
- 108010019530 Vascular Endothelial Growth Factors Proteins 0.000 claims description 8
- ZDXPYRJPNDTMRX-UHFFFAOYSA-N glutamine Natural products OC(=O)C(N)CCC(N)=O ZDXPYRJPNDTMRX-UHFFFAOYSA-N 0.000 claims description 8
- 102000002260 Alkaline Phosphatase Human genes 0.000 claims description 7
- 108020004774 Alkaline Phosphatase Proteins 0.000 claims description 7
- DHMQDGOQFOQNFH-UHFFFAOYSA-N Glycine Chemical compound NCC(O)=O DHMQDGOQFOQNFH-UHFFFAOYSA-N 0.000 claims description 6
- ONIBWKKTOPOVIA-UHFFFAOYSA-N Proline Natural products OC(=O)C1CCCN1 ONIBWKKTOPOVIA-UHFFFAOYSA-N 0.000 claims description 6
- 238000012217 deletion Methods 0.000 claims description 6
- 230000037430 deletion Effects 0.000 claims description 6
- 125000001500 prolyl group Chemical group [H]N1C([H])(C(=O)[*])C([H])([H])C([H])([H])C1([H])[H] 0.000 claims description 6
- 239000004475 Arginine Substances 0.000 claims description 5
- 102100035364 Growth/differentiation factor 3 Human genes 0.000 claims description 5
- 101001023986 Homo sapiens Growth/differentiation factor 3 Proteins 0.000 claims description 5
- 108010017842 Telomerase Proteins 0.000 claims description 5
- ODKSFYDXXFIFQN-UHFFFAOYSA-N arginine Natural products OC(=O)C(N)CCCNC(N)=N ODKSFYDXXFIFQN-UHFFFAOYSA-N 0.000 claims description 5
- 108091026890 Coding region Proteins 0.000 claims description 4
- 102100037126 Developmental pluripotency-associated protein 4 Human genes 0.000 claims description 4
- 239000004471 Glycine Substances 0.000 claims description 4
- 101000881868 Homo sapiens Developmental pluripotency-associated protein 4 Proteins 0.000 claims description 4
- 206010061598 Immunodeficiency Diseases 0.000 claims description 4
- 108010023082 activin A Proteins 0.000 claims description 4
- 235000004279 alanine Nutrition 0.000 claims description 4
- 230000002062 proliferating effect Effects 0.000 claims description 4
- 239000012679 serum free medium Substances 0.000 claims description 4
- 101000851376 Homo sapiens Tumor necrosis factor receptor superfamily member 8 Proteins 0.000 claims description 3
- 101000976622 Homo sapiens Zinc finger protein 42 homolog Proteins 0.000 claims description 3
- ROHFNLRQFUQHCH-UHFFFAOYSA-N Leucine Natural products CC(C)CC(N)C(O)=O ROHFNLRQFUQHCH-UHFFFAOYSA-N 0.000 claims description 3
- 229940124647 MEK inhibitor Drugs 0.000 claims description 3
- 102100036857 Tumor necrosis factor receptor superfamily member 8 Human genes 0.000 claims description 3
- 102100023550 Zinc finger protein 42 homolog Human genes 0.000 claims description 3
- 125000003295 alanine group Chemical group N[C@@H](C)C(=O)* 0.000 claims description 3
- 238000012258 culturing Methods 0.000 claims description 3
- 125000001909 leucine group Chemical group [H]N(*)C(C(*)=O)C([H])([H])C(C([H])([H])[H])C([H])([H])[H] 0.000 claims description 3
- 101150111214 lin-28 gene Proteins 0.000 claims description 3
- WHUUTDBJXJRKMK-UHFFFAOYSA-N Glutamic acid Natural products OC(=O)C(N)CCC(O)=O WHUUTDBJXJRKMK-UHFFFAOYSA-N 0.000 claims description 2
- QNAYBMKLOCPYGJ-REOHCLBHSA-N L-alanine Chemical compound C[C@H](N)C(O)=O QNAYBMKLOCPYGJ-REOHCLBHSA-N 0.000 claims description 2
- MTCFGRXMJLQNBG-UHFFFAOYSA-N Serine Natural products OCC(N)C(O)=O MTCFGRXMJLQNBG-UHFFFAOYSA-N 0.000 claims description 2
- QIVBCDIJIAJPQS-UHFFFAOYSA-N Tryptophan Natural products C1=CC=C2C(CC(N)C(O)=O)=CNC2=C1 QIVBCDIJIAJPQS-UHFFFAOYSA-N 0.000 claims description 2
- 235000013922 glutamic acid Nutrition 0.000 claims description 2
- 239000004220 glutamic acid Substances 0.000 claims description 2
- 125000000291 glutamic acid group Chemical group N[C@@H](CCC(O)=O)C(=O)* 0.000 claims description 2
- 125000003630 glycyl group Chemical group [H]N([H])C([H])([H])C(*)=O 0.000 claims description 2
- 125000003607 serino group Chemical group [H]N([H])[C@]([H])(C(=O)[*])C(O[H])([H])[H] 0.000 claims description 2
- 125000000341 threoninyl group Chemical group [H]OC([H])(C([H])([H])[H])C([H])(N([H])[H])C(*)=O 0.000 claims description 2
- 125000000430 tryptophan group Chemical group [H]N([H])C(C(=O)O*)C([H])([H])C1=C([H])N([H])C2=C([H])C([H])=C([H])C([H])=C12 0.000 claims description 2
- 102000052116 epidermal growth factor receptor activity proteins Human genes 0.000 claims 4
- 108700015053 epidermal growth factor receptor activity proteins Proteins 0.000 claims 4
- YOHYSYJDKVYCJI-UHFFFAOYSA-N n-[3-[[6-[3-(trifluoromethyl)anilino]pyrimidin-4-yl]amino]phenyl]cyclopropanecarboxamide Chemical compound FC(F)(F)C1=CC=CC(NC=2N=CN=C(NC=3C=C(NC(=O)C4CC4)C=CC=3)C=2)=C1 YOHYSYJDKVYCJI-UHFFFAOYSA-N 0.000 claims 4
- 101710146526 Dual specificity mitogen-activated protein kinase kinase 1 Proteins 0.000 claims 2
- 239000003814 drug Substances 0.000 abstract description 12
- 229940124597 therapeutic agent Drugs 0.000 abstract description 6
- 238000003745 diagnosis Methods 0.000 abstract description 4
- 230000014509 gene expression Effects 0.000 description 42
- 239000013598 vector Substances 0.000 description 37
- 101000843572 Homo sapiens Transcription factor HES-2 Proteins 0.000 description 35
- 102100030772 Transcription factor HES-2 Human genes 0.000 description 35
- 102100035423 POU domain, class 5, transcription factor 1 Human genes 0.000 description 26
- 238000004458 analytical method Methods 0.000 description 21
- 235000001014 amino acid Nutrition 0.000 description 17
- 102000002727 Protein Tyrosine Phosphatase Human genes 0.000 description 16
- 229940024606 amino acid Drugs 0.000 description 16
- 150000001413 amino acids Chemical class 0.000 description 16
- 210000002242 embryoid body Anatomy 0.000 description 16
- 108020000494 protein-tyrosine phosphatase Proteins 0.000 description 16
- 238000000338 in vitro Methods 0.000 description 15
- 230000008672 reprogramming Effects 0.000 description 15
- 208000037538 Myelomonocytic Juvenile Leukemia Diseases 0.000 description 13
- 210000004369 blood Anatomy 0.000 description 13
- 239000008280 blood Substances 0.000 description 13
- 208000035475 disorder Diseases 0.000 description 13
- 201000005992 juvenile myelomonocytic leukemia Diseases 0.000 description 13
- 108020004414 DNA Proteins 0.000 description 12
- 108010068342 MAP Kinase Kinase 1 Proteins 0.000 description 12
- 241000699666 Mus <mouse, genus> Species 0.000 description 12
- 108700019146 Transgenes Proteins 0.000 description 12
- 239000003550 marker Substances 0.000 description 12
- 101000687905 Homo sapiens Transcription factor SOX-2 Proteins 0.000 description 11
- 102100024270 Transcription factor SOX-2 Human genes 0.000 description 11
- 102000001301 EGF receptor Human genes 0.000 description 10
- 108060006698 EGF receptor Proteins 0.000 description 10
- 230000001177 retroviral effect Effects 0.000 description 10
- 102220004670 rs121918457 Human genes 0.000 description 10
- 238000011529 RT qPCR Methods 0.000 description 9
- 108091023040 Transcription factor Proteins 0.000 description 9
- 102000040945 Transcription factor Human genes 0.000 description 9
- 101710116241 Tyrosine-protein phosphatase non-receptor type 11 Proteins 0.000 description 9
- 102000040430 polynucleotide Human genes 0.000 description 9
- 108091033319 polynucleotide Proteins 0.000 description 9
- 239000002157 polynucleotide Substances 0.000 description 9
- 210000001519 tissue Anatomy 0.000 description 9
- 102000007469 Actins Human genes 0.000 description 8
- 108010085238 Actins Proteins 0.000 description 8
- ZDXPYRJPNDTMRX-VKHMYHEASA-N L-glutamine Chemical compound OC(=O)[C@@H](N)CCC(N)=O ZDXPYRJPNDTMRX-VKHMYHEASA-N 0.000 description 8
- 230000004913 activation Effects 0.000 description 8
- 210000001654 germ layer Anatomy 0.000 description 8
- 239000003102 growth factor Substances 0.000 description 8
- 230000003394 haemopoietic effect Effects 0.000 description 8
- 239000002609 medium Substances 0.000 description 8
- 230000008520 organization Effects 0.000 description 8
- 239000000523 sample Substances 0.000 description 8
- 239000006144 Dulbecco’s modified Eagle's medium Substances 0.000 description 7
- 206010028980 Neoplasm Diseases 0.000 description 7
- 230000008859 change Effects 0.000 description 7
- 230000007547 defect Effects 0.000 description 7
- 238000011161 development Methods 0.000 description 7
- 230000018109 developmental process Effects 0.000 description 7
- 210000001671 embryonic stem cell Anatomy 0.000 description 7
- 230000006870 function Effects 0.000 description 7
- 230000002068 genetic effect Effects 0.000 description 7
- 230000012010 growth Effects 0.000 description 7
- 210000003716 mesoderm Anatomy 0.000 description 7
- 239000002773 nucleotide Substances 0.000 description 7
- 125000003729 nucleotide group Chemical group 0.000 description 7
- 230000001225 therapeutic effect Effects 0.000 description 7
- 241000283074 Equus asinus Species 0.000 description 6
- 102100020677 Krueppel-like factor 4 Human genes 0.000 description 6
- 108040008097 MAP kinase activity proteins Proteins 0.000 description 6
- 102000019149 MAP kinase activity proteins Human genes 0.000 description 6
- 102000043136 MAP kinase family Human genes 0.000 description 6
- 108091054455 MAP kinase family Proteins 0.000 description 6
- 102100034398 Nuclear factor of activated T-cells, cytoplasmic 4 Human genes 0.000 description 6
- 101710151215 Nuclear factor of activated T-cells, cytoplasmic 4 Proteins 0.000 description 6
- 125000003275 alpha amino acid group Chemical group 0.000 description 6
- 229940079593 drug Drugs 0.000 description 6
- 210000004940 nucleus Anatomy 0.000 description 6
- 230000008506 pathogenesis Effects 0.000 description 6
- 208000011580 syndromic disease Diseases 0.000 description 6
- 238000001262 western blot Methods 0.000 description 6
- 101100039010 Caenorhabditis elegans dis-3 gene Proteins 0.000 description 5
- 108010010803 Gelatin Proteins 0.000 description 5
- 101710193519 Glial fibrillary acidic protein Proteins 0.000 description 5
- 101001139134 Homo sapiens Krueppel-like factor 4 Proteins 0.000 description 5
- 108010032107 Non-Receptor Type 11 Protein Tyrosine Phosphatase Proteins 0.000 description 5
- 230000005856 abnormality Effects 0.000 description 5
- 230000015572 biosynthetic process Effects 0.000 description 5
- 230000001413 cellular effect Effects 0.000 description 5
- 230000005754 cellular signaling Effects 0.000 description 5
- 238000006243 chemical reaction Methods 0.000 description 5
- 230000003247 decreasing effect Effects 0.000 description 5
- 210000003981 ectoderm Anatomy 0.000 description 5
- 108010048367 enhanced green fluorescent protein Proteins 0.000 description 5
- 239000008273 gelatin Substances 0.000 description 5
- 229920000159 gelatin Polymers 0.000 description 5
- 235000019322 gelatine Nutrition 0.000 description 5
- 235000011852 gelatine desserts Nutrition 0.000 description 5
- 210000005046 glial fibrillary acidic protein Anatomy 0.000 description 5
- 238000003365 immunocytochemistry Methods 0.000 description 5
- 238000001727 in vivo Methods 0.000 description 5
- 238000002493 microarray Methods 0.000 description 5
- 230000037361 pathway Effects 0.000 description 5
- 208000009138 pulmonary valve stenosis Diseases 0.000 description 5
- 230000011664 signaling Effects 0.000 description 5
- 238000010186 staining Methods 0.000 description 5
- 230000000638 stimulation Effects 0.000 description 5
- 238000010361 transduction Methods 0.000 description 5
- 230000026683 transduction Effects 0.000 description 5
- 206010069754 Acquired gene mutation Diseases 0.000 description 4
- 201000002927 Cardiofaciocutaneous syndrome Diseases 0.000 description 4
- 208000032170 Congenital Abnormalities Diseases 0.000 description 4
- 108091008794 FGF receptors Proteins 0.000 description 4
- 102000044168 Fibroblast Growth Factor Receptor Human genes 0.000 description 4
- 102100039289 Glial fibrillary acidic protein Human genes 0.000 description 4
- WZUVPPKBWHMQCE-UHFFFAOYSA-N Haematoxylin Chemical compound C12=CC(O)=C(O)C=C2CC2(O)C1C1=CC=C(O)C(O)=C1OC2 WZUVPPKBWHMQCE-UHFFFAOYSA-N 0.000 description 4
- 229930182816 L-glutamine Natural products 0.000 description 4
- 201000003793 Myelodysplastic syndrome Diseases 0.000 description 4
- 102000004160 Phosphoric Monoester Hydrolases Human genes 0.000 description 4
- 108090000608 Phosphoric Monoester Hydrolases Proteins 0.000 description 4
- 102000004987 Troponin T Human genes 0.000 description 4
- 108090001108 Troponin T Proteins 0.000 description 4
- 102100035071 Vimentin Human genes 0.000 description 4
- 108010065472 Vimentin Proteins 0.000 description 4
- 230000033228 biological regulation Effects 0.000 description 4
- 230000024245 cell differentiation Effects 0.000 description 4
- 239000002299 complementary DNA Substances 0.000 description 4
- 210000001900 endoderm Anatomy 0.000 description 4
- 239000000284 extract Substances 0.000 description 4
- 230000030279 gene silencing Effects 0.000 description 4
- 239000001963 growth medium Substances 0.000 description 4
- 230000006698 induction Effects 0.000 description 4
- 208000015181 infectious disease Diseases 0.000 description 4
- 206010024217 lentigo Diseases 0.000 description 4
- 230000003902 lesion Effects 0.000 description 4
- 230000004807 localization Effects 0.000 description 4
- 239000000463 material Substances 0.000 description 4
- 210000003205 muscle Anatomy 0.000 description 4
- 210000000066 myeloid cell Anatomy 0.000 description 4
- 230000008569 process Effects 0.000 description 4
- 230000000306 recurrent effect Effects 0.000 description 4
- 238000003757 reverse transcription PCR Methods 0.000 description 4
- 102200155655 rs121918456 Human genes 0.000 description 4
- 210000002966 serum Anatomy 0.000 description 4
- 230000037439 somatic mutation Effects 0.000 description 4
- 241000894007 species Species 0.000 description 4
- 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 4
- 208000024891 symptom Diseases 0.000 description 4
- 238000012360 testing method Methods 0.000 description 4
- 238000001890 transfection Methods 0.000 description 4
- 210000005048 vimentin Anatomy 0.000 description 4
- 108700028369 Alleles Proteins 0.000 description 3
- 108020004705 Codon Proteins 0.000 description 3
- 102000004127 Cytokines Human genes 0.000 description 3
- 108090000695 Cytokines Proteins 0.000 description 3
- 102100036912 Desmin Human genes 0.000 description 3
- 108010044052 Desmin Proteins 0.000 description 3
- 108700024394 Exon Proteins 0.000 description 3
- 108010007457 Extracellular Signal-Regulated MAP Kinases Proteins 0.000 description 3
- 102000007665 Extracellular Signal-Regulated MAP Kinases Human genes 0.000 description 3
- 206010053759 Growth retardation Diseases 0.000 description 3
- 101000984042 Homo sapiens Protein lin-28 homolog A Proteins 0.000 description 3
- OKKJLVBELUTLKV-UHFFFAOYSA-N Methanol Chemical compound OC OKKJLVBELUTLKV-UHFFFAOYSA-N 0.000 description 3
- 238000011789 NOD SCID mouse Methods 0.000 description 3
- 241000282373 Panthera pardus Species 0.000 description 3
- 102100025460 Protein lin-28 homolog A Human genes 0.000 description 3
- 102000004022 Protein-Tyrosine Kinases Human genes 0.000 description 3
- 108090000412 Protein-Tyrosine Kinases Proteins 0.000 description 3
- 238000002105 Southern blotting Methods 0.000 description 3
- 230000002159 abnormal effect Effects 0.000 description 3
- 238000010009 beating Methods 0.000 description 3
- 238000001369 bisulfite sequencing Methods 0.000 description 3
- 238000012512 characterization method Methods 0.000 description 3
- 210000000349 chromosome Anatomy 0.000 description 3
- 239000003636 conditioned culture medium Substances 0.000 description 3
- 201000000160 cryptorchidism Diseases 0.000 description 3
- 210000005045 desmin Anatomy 0.000 description 3
- 230000002500 effect on skin Effects 0.000 description 3
- 238000010195 expression analysis Methods 0.000 description 3
- 230000001815 facial effect Effects 0.000 description 3
- 238000000684 flow cytometry Methods 0.000 description 3
- 102000044858 human PTPN11 Human genes 0.000 description 3
- 210000005260 human cell Anatomy 0.000 description 3
- 230000001404 mediated effect Effects 0.000 description 3
- 230000011987 methylation Effects 0.000 description 3
- 238000007069 methylation reaction Methods 0.000 description 3
- 230000001575 pathological effect Effects 0.000 description 3
- 239000000243 solution Substances 0.000 description 3
- 238000002560 therapeutic procedure Methods 0.000 description 3
- 230000009772 tissue formation Effects 0.000 description 3
- 238000011144 upstream manufacturing Methods 0.000 description 3
- LAQPKDLYOBZWBT-NYLDSJSYSA-N (2s,4s,5r,6r)-5-acetamido-2-{[(2s,3r,4s,5s,6r)-2-{[(2r,3r,4r,5r)-5-acetamido-1,2-dihydroxy-6-oxo-4-{[(2s,3s,4r,5s,6s)-3,4,5-trihydroxy-6-methyloxan-2-yl]oxy}hexan-3-yl]oxy}-3,5-dihydroxy-6-(hydroxymethyl)oxan-4-yl]oxy}-4-hydroxy-6-[(1r,2r)-1,2,3-trihydrox Chemical compound O[C@H]1[C@H](O)[C@H](O)[C@H](C)O[C@H]1O[C@H]([C@@H](NC(C)=O)C=O)[C@@H]([C@H](O)CO)O[C@H]1[C@H](O)[C@@H](O[C@]2(O[C@H]([C@H](NC(C)=O)[C@@H](O)C2)[C@H](O)[C@H](O)CO)C(O)=O)[C@@H](O)[C@@H](CO)O1 LAQPKDLYOBZWBT-NYLDSJSYSA-N 0.000 description 2
- 108091032973 (ribonucleotides)n+m Proteins 0.000 description 2
- FWBHETKCLVMNFS-UHFFFAOYSA-N 4',6-Diamino-2-phenylindol Chemical compound C1=CC(C(=N)N)=CC=C1C1=CC2=CC=C(C(N)=N)C=C2N1 FWBHETKCLVMNFS-UHFFFAOYSA-N 0.000 description 2
- QTBSBXVTEAMEQO-UHFFFAOYSA-N Acetic acid Chemical compound CC(O)=O QTBSBXVTEAMEQO-UHFFFAOYSA-N 0.000 description 2
- 208000031261 Acute myeloid leukaemia Diseases 0.000 description 2
- CIWBSHSKHKDKBQ-JLAZNSOCSA-N Ascorbic acid Chemical compound OC[C@H](O)[C@H]1OC(=O)C(O)=C1O CIWBSHSKHKDKBQ-JLAZNSOCSA-N 0.000 description 2
- 206010010356 Congenital anomaly Diseases 0.000 description 2
- 206010011498 Cryptorchism Diseases 0.000 description 2
- 206010011703 Cyanosis Diseases 0.000 description 2
- 238000001712 DNA sequencing Methods 0.000 description 2
- 206010011878 Deafness Diseases 0.000 description 2
- 206010061818 Disease progression Diseases 0.000 description 2
- 241000255581 Drosophila <fruit fly, genus> Species 0.000 description 2
- 102000004190 Enzymes Human genes 0.000 description 2
- 108090000790 Enzymes Proteins 0.000 description 2
- WSFSSNUMVMOOMR-UHFFFAOYSA-N Formaldehyde Chemical compound O=C WSFSSNUMVMOOMR-UHFFFAOYSA-N 0.000 description 2
- 102100030708 GTPase KRas Human genes 0.000 description 2
- DHCLVCXQIBBOPH-UHFFFAOYSA-N Glycerol 2-phosphate Chemical compound OCC(CO)OP(O)(O)=O DHCLVCXQIBBOPH-UHFFFAOYSA-N 0.000 description 2
- 102100035716 Glycophorin-A Human genes 0.000 description 2
- 108010043121 Green Fluorescent Proteins Proteins 0.000 description 2
- 102000004144 Green Fluorescent Proteins Human genes 0.000 description 2
- 108010033040 Histones Proteins 0.000 description 2
- 241000282412 Homo Species 0.000 description 2
- 101000584612 Homo sapiens GTPase KRas Proteins 0.000 description 2
- 101001074244 Homo sapiens Glycophorin-A Proteins 0.000 description 2
- 101000599951 Homo sapiens Insulin-like growth factor I Proteins 0.000 description 2
- 101001078143 Homo sapiens Integrin alpha-IIb Proteins 0.000 description 2
- 101000835093 Homo sapiens Transferrin receptor protein 1 Proteins 0.000 description 2
- 241000701044 Human gammaherpesvirus 4 Species 0.000 description 2
- 208000010086 Hypertelorism Diseases 0.000 description 2
- 206010020771 Hypertelorism of orbit Diseases 0.000 description 2
- DGAQECJNVWCQMB-PUAWFVPOSA-M Ilexoside XXIX Chemical compound C[C@@H]1CC[C@@]2(CC[C@@]3(C(=CC[C@H]4[C@]3(CC[C@@H]5[C@@]4(CC[C@@H](C5(C)C)OS(=O)(=O)[O-])C)C)[C@@H]2[C@]1(C)O)C)C(=O)O[C@H]6[C@@H]([C@H]([C@@H]([C@H](O6)CO)O)O)O.[Na+] DGAQECJNVWCQMB-PUAWFVPOSA-M 0.000 description 2
- 102100037852 Insulin-like growth factor I Human genes 0.000 description 2
- 102100025306 Integrin alpha-IIb Human genes 0.000 description 2
- 241001465754 Metazoa Species 0.000 description 2
- 102000007607 Non-Receptor Type 11 Protein Tyrosine Phosphatase Human genes 0.000 description 2
- 108091028043 Nucleic acid sequence Proteins 0.000 description 2
- 229930040373 Paraformaldehyde Natural products 0.000 description 2
- 229930182555 Penicillin Natural products 0.000 description 2
- 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 2
- 108091000080 Phosphotransferase Proteins 0.000 description 2
- 101150036867 SYP gene Proteins 0.000 description 2
- 238000000692 Student's t-test Methods 0.000 description 2
- 102100026144 Transferrin receptor protein 1 Human genes 0.000 description 2
- 229920004890 Triton X-100 Polymers 0.000 description 2
- 239000013504 Triton X-100 Substances 0.000 description 2
- 241000700605 Viruses Species 0.000 description 2
- 230000003213 activating effect Effects 0.000 description 2
- 230000003321 amplification Effects 0.000 description 2
- 239000000427 antigen Substances 0.000 description 2
- 108091007433 antigens Proteins 0.000 description 2
- 102000036639 antigens Human genes 0.000 description 2
- 238000013459 approach Methods 0.000 description 2
- 238000003556 assay Methods 0.000 description 2
- QVGXLLKOCUKJST-UHFFFAOYSA-N atomic oxygen Chemical compound [O] QVGXLLKOCUKJST-UHFFFAOYSA-N 0.000 description 2
- 230000003115 biocidal effect Effects 0.000 description 2
- 230000007698 birth defect Effects 0.000 description 2
- 210000002459 blastocyst Anatomy 0.000 description 2
- 230000000903 blocking effect Effects 0.000 description 2
- 210000000988 bone and bone Anatomy 0.000 description 2
- 201000011510 cancer Diseases 0.000 description 2
- 230000003197 catalytic effect Effects 0.000 description 2
- 230000032823 cell division Effects 0.000 description 2
- 230000010261 cell growth Effects 0.000 description 2
- 230000004663 cell proliferation Effects 0.000 description 2
- 208000028831 congenital heart disease Diseases 0.000 description 2
- 238000010276 construction Methods 0.000 description 2
- 230000001086 cytosolic effect Effects 0.000 description 2
- 231100000895 deafness Toxicity 0.000 description 2
- 230000002950 deficient Effects 0.000 description 2
- 230000017858 demethylation Effects 0.000 description 2
- 238000010520 demethylation reaction Methods 0.000 description 2
- 230000005750 disease progression Effects 0.000 description 2
- 238000007877 drug screening Methods 0.000 description 2
- 230000013020 embryo development Effects 0.000 description 2
- 238000005516 engineering process Methods 0.000 description 2
- 229940088598 enzyme Drugs 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
- 230000000925 erythroid effect Effects 0.000 description 2
- 239000003797 essential amino acid Substances 0.000 description 2
- 235000020776 essential amino acid Nutrition 0.000 description 2
- 239000013604 expression vector Substances 0.000 description 2
- 239000012091 fetal bovine serum Substances 0.000 description 2
- 230000001605 fetal effect Effects 0.000 description 2
- 239000000834 fixative Substances 0.000 description 2
- 238000007667 floating Methods 0.000 description 2
- 239000012634 fragment Substances 0.000 description 2
- 210000004392 genitalia Anatomy 0.000 description 2
- 239000005090 green fluorescent protein Substances 0.000 description 2
- 231100000001 growth retardation Toxicity 0.000 description 2
- 230000036541 health Effects 0.000 description 2
- 208000016354 hearing loss disease Diseases 0.000 description 2
- 229940088597 hormone Drugs 0.000 description 2
- 230000002209 hydrophobic effect Effects 0.000 description 2
- 238000012744 immunostaining Methods 0.000 description 2
- 238000010874 in vitro model Methods 0.000 description 2
- 238000011534 incubation Methods 0.000 description 2
- 238000002347 injection Methods 0.000 description 2
- 239000007924 injection Substances 0.000 description 2
- 238000003780 insertion Methods 0.000 description 2
- 230000037431 insertion Effects 0.000 description 2
- 230000010354 integration Effects 0.000 description 2
- 230000003993 interaction Effects 0.000 description 2
- 238000011835 investigation Methods 0.000 description 2
- 210000003141 lower extremity Anatomy 0.000 description 2
- 210000004698 lymphocyte Anatomy 0.000 description 2
- 239000012139 lysis buffer Substances 0.000 description 2
- 230000007246 mechanism Effects 0.000 description 2
- 239000012528 membrane Substances 0.000 description 2
- 108020004999 messenger RNA Proteins 0.000 description 2
- 238000010208 microarray analysis Methods 0.000 description 2
- 230000004048 modification Effects 0.000 description 2
- 238000012986 modification Methods 0.000 description 2
- 230000009456 molecular mechanism Effects 0.000 description 2
- PJUIMOJAAPLTRJ-UHFFFAOYSA-N monothioglycerol Chemical compound OCC(O)CS PJUIMOJAAPLTRJ-UHFFFAOYSA-N 0.000 description 2
- 210000002569 neuron Anatomy 0.000 description 2
- 231100001160 nonlethal Toxicity 0.000 description 2
- 238000003199 nucleic acid amplification method Methods 0.000 description 2
- 210000000287 oocyte Anatomy 0.000 description 2
- 229910052760 oxygen Inorganic materials 0.000 description 2
- 239000001301 oxygen Substances 0.000 description 2
- 229920002866 paraformaldehyde Polymers 0.000 description 2
- 229940049954 penicillin Drugs 0.000 description 2
- YBYRMVIVWMBXKQ-UHFFFAOYSA-N phenylmethanesulfonyl fluoride Chemical compound FS(=O)(=O)CC1=CC=CC=C1 YBYRMVIVWMBXKQ-UHFFFAOYSA-N 0.000 description 2
- 102000020233 phosphotransferase Human genes 0.000 description 2
- 239000013612 plasmid Substances 0.000 description 2
- 239000013641 positive control Substances 0.000 description 2
- 239000000047 product Substances 0.000 description 2
- KAQKFAOMNZTLHT-OZUDYXHBSA-N prostaglandin I2 Chemical compound O1\C(=C/CCCC(O)=O)C[C@@H]2[C@@H](/C=C/[C@@H](O)CCCCC)[C@H](O)C[C@@H]21 KAQKFAOMNZTLHT-OZUDYXHBSA-N 0.000 description 2
- 150000003180 prostaglandins Chemical class 0.000 description 2
- 208000030390 pulmonic stenosis Diseases 0.000 description 2
- 238000003762 quantitative reverse transcription PCR Methods 0.000 description 2
- 230000004044 response Effects 0.000 description 2
- 102200155478 rs121918459 Human genes 0.000 description 2
- 102220004653 rs121918467 Human genes 0.000 description 2
- 102220004672 rs121918468 Human genes 0.000 description 2
- 102220004673 rs121918469 Human genes 0.000 description 2
- 102200155670 rs28933386 Human genes 0.000 description 2
- 210000002235 sarcomere Anatomy 0.000 description 2
- 238000012163 sequencing technique Methods 0.000 description 2
- 239000011734 sodium Substances 0.000 description 2
- 229910052708 sodium Inorganic materials 0.000 description 2
- PUZPDOWCWNUUKD-UHFFFAOYSA-M sodium fluoride Chemical compound [F-].[Na+] PUZPDOWCWNUUKD-UHFFFAOYSA-M 0.000 description 2
- 229960005322 streptomycin Drugs 0.000 description 2
- 239000000758 substrate Substances 0.000 description 2
- 230000017423 tissue regeneration Effects 0.000 description 2
- 238000012546 transfer Methods 0.000 description 2
- 241001430294 unidentified retrovirus Species 0.000 description 2
- NNJPGOLRFBJNIW-HNNXBMFYSA-N (-)-demecolcine Chemical compound C1=C(OC)C(=O)C=C2[C@@H](NC)CCC3=CC(OC)=C(OC)C(OC)=C3C2=C1 NNJPGOLRFBJNIW-HNNXBMFYSA-N 0.000 description 1
- KKVYYGGCHJGEFJ-UHFFFAOYSA-N 1-n-(4-chlorophenyl)-6-methyl-5-n-[3-(7h-purin-6-yl)pyridin-2-yl]isoquinoline-1,5-diamine Chemical compound N=1C=CC2=C(NC=3C(=CC=CN=3)C=3C=4N=CNC=4N=CN=3)C(C)=CC=C2C=1NC1=CC=C(Cl)C=C1 KKVYYGGCHJGEFJ-UHFFFAOYSA-N 0.000 description 1
- FWMNVWWHGCHHJJ-SKKKGAJSSA-N 4-amino-1-[(2r)-6-amino-2-[[(2r)-2-[[(2r)-2-[[(2r)-2-amino-3-phenylpropanoyl]amino]-3-phenylpropanoyl]amino]-4-methylpentanoyl]amino]hexanoyl]piperidine-4-carboxylic acid Chemical compound C([C@H](C(=O)N[C@H](CC(C)C)C(=O)N[C@H](CCCCN)C(=O)N1CCC(N)(CC1)C(O)=O)NC(=O)[C@H](N)CC=1C=CC=CC=1)C1=CC=CC=C1 FWMNVWWHGCHHJJ-SKKKGAJSSA-N 0.000 description 1
- 229920000936 Agarose Polymers 0.000 description 1
- 239000012099 Alexa Fluor family Substances 0.000 description 1
- 108010039627 Aprotinin Proteins 0.000 description 1
- 101100404726 Arabidopsis thaliana NHX7 gene Proteins 0.000 description 1
- LSNNMFCWUKXFEE-UHFFFAOYSA-M Bisulfite Chemical compound OS([O-])=O LSNNMFCWUKXFEE-UHFFFAOYSA-M 0.000 description 1
- 102100024506 Bone morphogenetic protein 2 Human genes 0.000 description 1
- 102100022525 Bone morphogenetic protein 6 Human genes 0.000 description 1
- 108091003079 Bovine Serum Albumin Proteins 0.000 description 1
- 238000009010 Bradford assay Methods 0.000 description 1
- 241001656894 Buxus microphylla var. japonica Species 0.000 description 1
- 208000000380 Cafe-au-Lait Spots Diseases 0.000 description 1
- 102000004631 Calcineurin Human genes 0.000 description 1
- 108010042955 Calcineurin Proteins 0.000 description 1
- 241000283707 Capra Species 0.000 description 1
- XZFRIPGNUQRGPI-WBQKLGIQSA-N Carbaprostacyclin Chemical compound C1\C(=C/CCCC(O)=O)C[C@@H]2[C@@H](/C=C/[C@@H](O)CCCCC)[C@H](O)C[C@@H]21 XZFRIPGNUQRGPI-WBQKLGIQSA-N 0.000 description 1
- 208000005623 Carcinogenesis Diseases 0.000 description 1
- 201000009030 Carcinoma Diseases 0.000 description 1
- 208000031229 Cardiomyopathies Diseases 0.000 description 1
- 108010077544 Chromatin Proteins 0.000 description 1
- 102000029816 Collagenase Human genes 0.000 description 1
- 108060005980 Collagenase Proteins 0.000 description 1
- 208000002330 Congenital Heart Defects Diseases 0.000 description 1
- 102000002585 Contractile Proteins Human genes 0.000 description 1
- 108010068426 Contractile Proteins Proteins 0.000 description 1
- 239000003298 DNA probe Substances 0.000 description 1
- 241000252212 Danio rerio Species 0.000 description 1
- 206010061619 Deformity Diseases 0.000 description 1
- NNJPGOLRFBJNIW-UHFFFAOYSA-N Demecolcine Natural products C1=C(OC)C(=O)C=C2C(NC)CCC3=CC(OC)=C(OC)C(OC)=C3C2=C1 NNJPGOLRFBJNIW-UHFFFAOYSA-N 0.000 description 1
- 102000016911 Deoxyribonucleases Human genes 0.000 description 1
- 108010053770 Deoxyribonucleases Proteins 0.000 description 1
- 206010012559 Developmental delay Diseases 0.000 description 1
- 208000012239 Developmental disease Diseases 0.000 description 1
- 102100036949 Developmental pluripotency-associated protein 2 Human genes 0.000 description 1
- 101000876610 Dictyostelium discoideum Extracellular signal-regulated kinase 2 Proteins 0.000 description 1
- 102100023266 Dual specificity mitogen-activated protein kinase kinase 2 Human genes 0.000 description 1
- 101710146529 Dual specificity mitogen-activated protein kinase kinase 2 Proteins 0.000 description 1
- 208000000059 Dyspnea Diseases 0.000 description 1
- 206010013975 Dyspnoeas Diseases 0.000 description 1
- KCXVZYZYPLLWCC-UHFFFAOYSA-N EDTA Chemical compound OC(=O)CN(CC(O)=O)CCN(CC(O)=O)CC(O)=O KCXVZYZYPLLWCC-UHFFFAOYSA-N 0.000 description 1
- 101150047683 ESC1 gene Proteins 0.000 description 1
- 102100030013 Endoribonuclease Human genes 0.000 description 1
- 101710199605 Endoribonuclease Proteins 0.000 description 1
- 102000010834 Extracellular Matrix Proteins Human genes 0.000 description 1
- 108010037362 Extracellular Matrix Proteins Proteins 0.000 description 1
- 238000012413 Fluorescence activated cell sorting analysis Methods 0.000 description 1
- 101150111020 GLUL gene Proteins 0.000 description 1
- 102100029974 GTPase HRas Human genes 0.000 description 1
- 102000053171 Glial Fibrillary Acidic Human genes 0.000 description 1
- 108010017213 Granulocyte-Macrophage Colony-Stimulating Factor Proteins 0.000 description 1
- 102100039620 Granulocyte-macrophage colony-stimulating factor Human genes 0.000 description 1
- 101150017137 Haspin gene Proteins 0.000 description 1
- 208000032843 Hemorrhage Diseases 0.000 description 1
- 229920000209 Hexadimethrine bromide Polymers 0.000 description 1
- 102000006947 Histones Human genes 0.000 description 1
- 102000009331 Homeodomain Proteins Human genes 0.000 description 1
- 108010048671 Homeodomain Proteins Proteins 0.000 description 1
- 101000762366 Homo sapiens Bone morphogenetic protein 2 Proteins 0.000 description 1
- 101000899390 Homo sapiens Bone morphogenetic protein 6 Proteins 0.000 description 1
- 101000804948 Homo sapiens Developmental pluripotency-associated protein 2 Proteins 0.000 description 1
- 101000881866 Homo sapiens Developmental pluripotency-associated protein 3 Proteins 0.000 description 1
- 101000584633 Homo sapiens GTPase HRas Proteins 0.000 description 1
- 101100510266 Homo sapiens KLF4 gene Proteins 0.000 description 1
- 101001052493 Homo sapiens Mitogen-activated protein kinase 1 Proteins 0.000 description 1
- 101100137155 Homo sapiens POU5F1 gene Proteins 0.000 description 1
- 101000712530 Homo sapiens RAF proto-oncogene serine/threonine-protein kinase Proteins 0.000 description 1
- 101000738771 Homo sapiens Receptor-type tyrosine-protein phosphatase C Proteins 0.000 description 1
- 101000984753 Homo sapiens Serine/threonine-protein kinase B-raf Proteins 0.000 description 1
- 101000868152 Homo sapiens Son of sevenless homolog 1 Proteins 0.000 description 1
- 101000617738 Homo sapiens Survival motor neuron protein Proteins 0.000 description 1
- 101000980827 Homo sapiens T-cell surface glycoprotein CD1a Proteins 0.000 description 1
- 101000716149 Homo sapiens T-cell surface glycoprotein CD1b Proteins 0.000 description 1
- 101000716124 Homo sapiens T-cell surface glycoprotein CD1c Proteins 0.000 description 1
- 101001050288 Homo sapiens Transcription factor Jun Proteins 0.000 description 1
- 101000777245 Homo sapiens Undifferentiated embryonic cell transcription factor 1 Proteins 0.000 description 1
- 101000784538 Homo sapiens Zinc finger and SCAN domain-containing protein 10 Proteins 0.000 description 1
- 206010021143 Hypoxia Diseases 0.000 description 1
- 108700002232 Immediate-Early Genes Proteins 0.000 description 1
- 102000001706 Immunoglobulin Fab Fragments Human genes 0.000 description 1
- 108010054477 Immunoglobulin Fab Fragments Proteins 0.000 description 1
- 208000026350 Inborn Genetic disease Diseases 0.000 description 1
- 102100034343 Integrase Human genes 0.000 description 1
- 108091092195 Intron Proteins 0.000 description 1
- ODKSFYDXXFIFQN-BYPYZUCNSA-P L-argininium(2+) Chemical compound NC(=[NH2+])NCCC[C@H]([NH3+])C(O)=O ODKSFYDXXFIFQN-BYPYZUCNSA-P 0.000 description 1
- HNDVDQJCIGZPNO-YFKPBYRVSA-N L-histidine Chemical compound OC(=O)[C@@H](N)CC1=CN=CN1 HNDVDQJCIGZPNO-YFKPBYRVSA-N 0.000 description 1
- AGPKZVBTJJNPAG-WHFBIAKZSA-N L-isoleucine Chemical compound CC[C@H](C)[C@H](N)C(O)=O AGPKZVBTJJNPAG-WHFBIAKZSA-N 0.000 description 1
- KDXKERNSBIXSRK-YFKPBYRVSA-N L-lysine Chemical compound NCCCC[C@H](N)C(O)=O KDXKERNSBIXSRK-YFKPBYRVSA-N 0.000 description 1
- FFEARJCKVFRZRR-BYPYZUCNSA-N L-methionine Chemical group CSCC[C@H](N)C(O)=O FFEARJCKVFRZRR-BYPYZUCNSA-N 0.000 description 1
- KZSNJWFQEVHDMF-BYPYZUCNSA-N L-valine Chemical group CC(C)[C@H](N)C(O)=O KZSNJWFQEVHDMF-BYPYZUCNSA-N 0.000 description 1
- GDBQQVLCIARPGH-UHFFFAOYSA-N Leupeptin Natural products CC(C)CC(NC(C)=O)C(=O)NC(CC(C)C)C(=O)NC(C=O)CCCN=C(N)N GDBQQVLCIARPGH-UHFFFAOYSA-N 0.000 description 1
- 208000035752 Live birth Diseases 0.000 description 1
- 108060001084 Luciferase Proteins 0.000 description 1
- 239000005089 Luciferase Substances 0.000 description 1
- KDXKERNSBIXSRK-UHFFFAOYSA-N Lysine Natural products NCCCCC(N)C(O)=O KDXKERNSBIXSRK-UHFFFAOYSA-N 0.000 description 1
- 239000004472 Lysine Substances 0.000 description 1
- 229940126560 MAPK inhibitor Drugs 0.000 description 1
- 239000007993 MOPS buffer Substances 0.000 description 1
- 208000036626 Mental retardation Diseases 0.000 description 1
- 102100024193 Mitogen-activated protein kinase 1 Human genes 0.000 description 1
- 208000035489 Monocytic Acute Leukemia Diseases 0.000 description 1
- 208000016850 Moynahan syndrome Diseases 0.000 description 1
- 241001529936 Murinae Species 0.000 description 1
- 241000699660 Mus musculus Species 0.000 description 1
- 101100381978 Mus musculus Braf gene Proteins 0.000 description 1
- 241000699670 Mus sp. Species 0.000 description 1
- 208000033776 Myeloid Acute Leukemia Diseases 0.000 description 1
- 208000014767 Myeloproliferative disease Diseases 0.000 description 1
- 201000007224 Myeloproliferative neoplasm Diseases 0.000 description 1
- 108010018525 NFATC Transcription Factors Proteins 0.000 description 1
- 102000002673 NFATC Transcription Factors Human genes 0.000 description 1
- 208000018251 Neurofibromatosis type 6 Diseases 0.000 description 1
- 108010085793 Neurofibromin 1 Proteins 0.000 description 1
- 102000007530 Neurofibromin 1 Human genes 0.000 description 1
- 239000004677 Nylon Substances 0.000 description 1
- 101150092239 OTX2 gene Proteins 0.000 description 1
- 102000002584 Octamer Transcription Factor-3 Human genes 0.000 description 1
- 108010068425 Octamer Transcription Factor-3 Proteins 0.000 description 1
- 108700020796 Oncogene Proteins 0.000 description 1
- 102000043276 Oncogene Human genes 0.000 description 1
- 208000018737 Parkinson disease Diseases 0.000 description 1
- 241001494479 Pecora Species 0.000 description 1
- 108091005804 Peptidases Proteins 0.000 description 1
- 102000045595 Phosphoprotein Phosphatases Human genes 0.000 description 1
- 108700019535 Phosphoprotein Phosphatases Proteins 0.000 description 1
- 108010089430 Phosphoproteins Proteins 0.000 description 1
- 102000007982 Phosphoproteins Human genes 0.000 description 1
- 208000020565 Proportionate short stature Diseases 0.000 description 1
- 239000004365 Protease Substances 0.000 description 1
- 102000001253 Protein Kinase Human genes 0.000 description 1
- 108700020978 Proto-Oncogene Proteins 0.000 description 1
- 102000052575 Proto-Oncogene Human genes 0.000 description 1
- 102000007568 Proto-Oncogene Proteins c-fos Human genes 0.000 description 1
- 108010071563 Proto-Oncogene Proteins c-fos Proteins 0.000 description 1
- 108091008109 Pseudogenes Proteins 0.000 description 1
- 102000057361 Pseudogenes Human genes 0.000 description 1
- 238000012181 QIAquick gel extraction kit Methods 0.000 description 1
- 102100033479 RAF proto-oncogene serine/threonine-protein kinase Human genes 0.000 description 1
- 108010092799 RNA-directed DNA polymerase Proteins 0.000 description 1
- 229940078123 Ras inhibitor Drugs 0.000 description 1
- 241000700159 Rattus Species 0.000 description 1
- 102100037422 Receptor-type tyrosine-protein phosphatase C Human genes 0.000 description 1
- 206010057190 Respiratory tract infections Diseases 0.000 description 1
- 206010038997 Retroviral infections Diseases 0.000 description 1
- 102100037486 Reverse transcriptase/ribonuclease H Human genes 0.000 description 1
- 238000011579 SCID mouse model Methods 0.000 description 1
- 102000011859 SH2 Domain-Containing Protein Tyrosine Phosphatases Human genes 0.000 description 1
- 108010061033 SH2 Domain-Containing Protein Tyrosine Phosphatases Proteins 0.000 description 1
- 102000014400 SH2 domains Human genes 0.000 description 1
- 108050003452 SH2 domains Proteins 0.000 description 1
- 102000057028 SOS1 Human genes 0.000 description 1
- 108700022176 SOS1 Proteins 0.000 description 1
- 101100197320 Saccharomyces cerevisiae (strain ATCC 204508 / S288c) RPL35A gene Proteins 0.000 description 1
- 208000034189 Sclerosis Diseases 0.000 description 1
- BUGBHKTXTAQXES-UHFFFAOYSA-N Selenium Chemical compound [Se] BUGBHKTXTAQXES-UHFFFAOYSA-N 0.000 description 1
- 238000012300 Sequence Analysis Methods 0.000 description 1
- 101710113029 Serine/threonine-protein kinase Proteins 0.000 description 1
- 102100027103 Serine/threonine-protein kinase B-raf Human genes 0.000 description 1
- 208000020221 Short stature Diseases 0.000 description 1
- DWAQJAXMDSEUJJ-UHFFFAOYSA-M Sodium bisulfite Chemical compound [Na+].OS([O-])=O DWAQJAXMDSEUJJ-UHFFFAOYSA-M 0.000 description 1
- 102100032929 Son of sevenless homolog 1 Human genes 0.000 description 1
- 101150100839 Sos1 gene Proteins 0.000 description 1
- 241000862969 Stella Species 0.000 description 1
- 101000930762 Sulfolobus acidocaldarius (strain ATCC 33909 / DSM 639 / JCM 8929 / NBRC 15157 / NCIMB 11770) Signal recognition particle receptor FtsY Proteins 0.000 description 1
- 102100021947 Survival motor neuron protein Human genes 0.000 description 1
- 241000282898 Sus scrofa Species 0.000 description 1
- 102100024219 T-cell surface glycoprotein CD1a Human genes 0.000 description 1
- 208000001871 Tachycardia Diseases 0.000 description 1
- 108010006785 Taq Polymerase Proteins 0.000 description 1
- 101710136739 Teichoic acid poly(glycerol phosphate) polymerase Proteins 0.000 description 1
- 108020005038 Terminator Codon Proteins 0.000 description 1
- 102100023132 Transcription factor Jun Human genes 0.000 description 1
- 101710101305 Transducin-like enhancer protein 1 Proteins 0.000 description 1
- 102100039362 Transducin-like enhancer protein 1 Human genes 0.000 description 1
- 102000004338 Transferrin Human genes 0.000 description 1
- 108090000901 Transferrin Proteins 0.000 description 1
- 102000004903 Troponin Human genes 0.000 description 1
- 108090001027 Troponin Proteins 0.000 description 1
- 102100031278 Undifferentiated embryonic cell transcription factor 1 Human genes 0.000 description 1
- KZSNJWFQEVHDMF-UHFFFAOYSA-N Valine Chemical group CC(C)C(N)C(O)=O KZSNJWFQEVHDMF-UHFFFAOYSA-N 0.000 description 1
- 101001029301 Xenopus tropicalis Forkhead box protein D3 Proteins 0.000 description 1
- 102100020919 Zinc finger and SCAN domain-containing protein 10 Human genes 0.000 description 1
- 229960000583 acetic acid Drugs 0.000 description 1
- 230000002378 acidificating effect Effects 0.000 description 1
- 230000001154 acute effect Effects 0.000 description 1
- 210000000577 adipose tissue Anatomy 0.000 description 1
- 239000011543 agarose gel Substances 0.000 description 1
- 230000004075 alteration Effects 0.000 description 1
- 210000004102 animal cell Anatomy 0.000 description 1
- 238000010171 animal model Methods 0.000 description 1
- 239000003242 anti bacterial agent Substances 0.000 description 1
- 229940088710 antibiotic agent Drugs 0.000 description 1
- 230000006907 apoptotic process Effects 0.000 description 1
- 229960004405 aprotinin Drugs 0.000 description 1
- 125000003118 aryl group Chemical group 0.000 description 1
- 235000010323 ascorbic acid Nutrition 0.000 description 1
- 229960005070 ascorbic acid Drugs 0.000 description 1
- 239000011668 ascorbic acid Substances 0.000 description 1
- 210000004103 basophilic normoblast Anatomy 0.000 description 1
- 230000009286 beneficial effect Effects 0.000 description 1
- 230000008901 benefit Effects 0.000 description 1
- PXXJHWLDUBFPOL-UHFFFAOYSA-N benzamidine Chemical compound NC(=N)C1=CC=CC=C1 PXXJHWLDUBFPOL-UHFFFAOYSA-N 0.000 description 1
- 230000005540 biological transmission Effects 0.000 description 1
- 208000034158 bleeding Diseases 0.000 description 1
- 230000000740 bleeding effect Effects 0.000 description 1
- 210000002798 bone marrow cell Anatomy 0.000 description 1
- 210000004556 brain Anatomy 0.000 description 1
- 210000000133 brain stem Anatomy 0.000 description 1
- 239000000872 buffer Substances 0.000 description 1
- 230000036952 cancer formation Effects 0.000 description 1
- 150000001720 carbohydrates Chemical class 0.000 description 1
- 231100000504 carcinogenesis Toxicity 0.000 description 1
- 208000015685 cardiocutaneous syndrome Diseases 0.000 description 1
- 210000000748 cardiovascular system Anatomy 0.000 description 1
- 210000000845 cartilage Anatomy 0.000 description 1
- 230000015556 catabolic process Effects 0.000 description 1
- 230000000453 cell autonomous effect Effects 0.000 description 1
- 230000022131 cell cycle Effects 0.000 description 1
- 230000007910 cell fusion Effects 0.000 description 1
- 239000002771 cell marker Substances 0.000 description 1
- 239000006285 cell suspension Substances 0.000 description 1
- 230000033077 cellular process Effects 0.000 description 1
- 230000004715 cellular signal transduction Effects 0.000 description 1
- 239000003153 chemical reaction reagent Substances 0.000 description 1
- 210000003483 chromatin Anatomy 0.000 description 1
- 230000002759 chromosomal effect Effects 0.000 description 1
- 230000008711 chromosomal rearrangement Effects 0.000 description 1
- ARUGKOZUKWAXDS-SEWALLKFSA-N cicaprost Chemical compound C1\C(=C/COCC(O)=O)C[C@@H]2[C@@H](C#C[C@@H](O)[C@@H](C)CC#CCC)[C@H](O)C[C@@H]21 ARUGKOZUKWAXDS-SEWALLKFSA-N 0.000 description 1
- 229950000634 cicaprost Drugs 0.000 description 1
- 238000003759 clinical diagnosis Methods 0.000 description 1
- 229960002424 collagenase Drugs 0.000 description 1
- 230000000295 complement effect Effects 0.000 description 1
- 239000013068 control sample Substances 0.000 description 1
- 210000000805 cytoplasm Anatomy 0.000 description 1
- 230000002074 deregulated effect Effects 0.000 description 1
- 230000001627 detrimental effect Effects 0.000 description 1
- 206010012601 diabetes mellitus Diseases 0.000 description 1
- 239000012895 dilution Substances 0.000 description 1
- 238000010790 dilution Methods 0.000 description 1
- VHJLVAABSRFDPM-QWWZWVQMSA-N dithiothreitol Chemical compound SC[C@@H](O)[C@H](O)CS VHJLVAABSRFDPM-QWWZWVQMSA-N 0.000 description 1
- 238000009509 drug development Methods 0.000 description 1
- 231100001047 early fetal death Toxicity 0.000 description 1
- 230000005014 ectopic expression Effects 0.000 description 1
- 230000002526 effect on cardiovascular system Effects 0.000 description 1
- 238000004520 electroporation Methods 0.000 description 1
- 210000002308 embryonic cell Anatomy 0.000 description 1
- 230000002255 enzymatic effect Effects 0.000 description 1
- 210000002919 epithelial cell Anatomy 0.000 description 1
- 229960001123 epoprostenol Drugs 0.000 description 1
- 210000003743 erythrocyte Anatomy 0.000 description 1
- 210000000267 erythroid cell Anatomy 0.000 description 1
- DEFVIWRASFVYLL-UHFFFAOYSA-N ethylene glycol bis(2-aminoethyl)tetraacetic acid Chemical compound OC(=O)CN(CC(O)=O)CCOCCOCCN(CC(O)=O)CC(O)=O DEFVIWRASFVYLL-UHFFFAOYSA-N 0.000 description 1
- 238000011156 evaluation Methods 0.000 description 1
- 230000000763 evoking effect Effects 0.000 description 1
- 230000001747 exhibiting effect Effects 0.000 description 1
- 238000002474 experimental method Methods 0.000 description 1
- 210000002744 extracellular matrix Anatomy 0.000 description 1
- 238000001943 fluorescence-activated cell sorting Methods 0.000 description 1
- 230000004927 fusion Effects 0.000 description 1
- 108010027225 gag-pol Fusion Proteins Proteins 0.000 description 1
- 238000012239 gene modification Methods 0.000 description 1
- 208000016361 genetic disease Diseases 0.000 description 1
- 230000005017 genetic modification Effects 0.000 description 1
- 235000013617 genetically modified food Nutrition 0.000 description 1
- 210000004602 germ cell Anatomy 0.000 description 1
- 239000012362 glacial acetic acid Substances 0.000 description 1
- 239000011521 glass Substances 0.000 description 1
- 239000000122 growth hormone Substances 0.000 description 1
- 238000003306 harvesting Methods 0.000 description 1
- 230000009067 heart development Effects 0.000 description 1
- 208000019622 heart disease Diseases 0.000 description 1
- 230000002489 hematologic effect Effects 0.000 description 1
- 230000011132 hemopoiesis Effects 0.000 description 1
- 210000003494 hepatocyte Anatomy 0.000 description 1
- 238000004128 high performance liquid chromatography Methods 0.000 description 1
- HNDVDQJCIGZPNO-UHFFFAOYSA-N histidine Natural products OC(=O)C(N)CC1=CN=CN1 HNDVDQJCIGZPNO-UHFFFAOYSA-N 0.000 description 1
- 239000005556 hormone Substances 0.000 description 1
- 238000009396 hybridization Methods 0.000 description 1
- 229910052739 hydrogen Inorganic materials 0.000 description 1
- 239000001257 hydrogen Substances 0.000 description 1
- 206010021093 hypospadias Diseases 0.000 description 1
- 239000000815 hypotonic solution Substances 0.000 description 1
- 230000007954 hypoxia Effects 0.000 description 1
- 229960002240 iloprost Drugs 0.000 description 1
- HIFJCPQKFCZDDL-ACWOEMLNSA-N iloprost Chemical compound C1\C(=C/CCCC(O)=O)C[C@@H]2[C@@H](/C=C/[C@@H](O)C(C)CC#CC)[C@H](O)C[C@@H]21 HIFJCPQKFCZDDL-ACWOEMLNSA-N 0.000 description 1
- 238000003384 imaging method Methods 0.000 description 1
- 230000036039 immunity Effects 0.000 description 1
- 238000003119 immunoblot Methods 0.000 description 1
- 230000001771 impaired effect Effects 0.000 description 1
- 238000011065 in-situ storage Methods 0.000 description 1
- 230000002401 inhibitory effect Effects 0.000 description 1
- ZPNFWUPYTFPOJU-LPYSRVMUSA-N iniprol Chemical compound C([C@H]1C(=O)NCC(=O)NCC(=O)N[C@H]2CSSC[C@H]3C(=O)N[C@@H](CCCCN)C(=O)N[C@@H](C)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@H](C(N[C@H](C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CC=4C=CC(O)=CC=4)C(=O)N[C@@H](CC=4C=CC=CC=4)C(=O)N[C@@H](CC=4C=CC(O)=CC=4)C(=O)N[C@@H](CC(N)=O)C(=O)N[C@@H](C)C(=O)N[C@@H](CCCCN)C(=O)N[C@@H](C)C(=O)NCC(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CSSC[C@H](NC(=O)[C@H](CC(O)=O)NC(=O)[C@H](CCC(O)=O)NC(=O)[C@H](C)NC(=O)[C@H](CO)NC(=O)[C@H](CCCCN)NC(=O)[C@H](CC=4C=CC=CC=4)NC(=O)[C@H](CC(N)=O)NC(=O)[C@H](CC(N)=O)NC(=O)[C@H](CCCNC(N)=N)NC(=O)[C@H](CCCCN)NC(=O)[C@H](C)NC(=O)[C@H](CCCNC(N)=N)NC2=O)C(=O)N[C@@H](CCSC)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H]([C@@H](C)O)C(=O)N[C@@H](CSSC[C@H](NC(=O)[C@H](CC=2C=CC=CC=2)NC(=O)[C@H](CC(O)=O)NC(=O)[C@H]2N(CCC2)C(=O)[C@@H](N)CCCNC(N)=N)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CCC(O)=O)C(=O)N2[C@@H](CCC2)C(=O)N2[C@@H](CCC2)C(=O)N[C@@H](CC=2C=CC(O)=CC=2)C(=O)N[C@@H]([C@@H](C)O)C(=O)NCC(=O)N2[C@@H](CCC2)C(=O)N3)C(=O)NCC(=O)NCC(=O)N[C@@H](C)C(O)=O)C(=O)N[C@@H](CCC(N)=O)C(=O)N[C@H](C(=O)N[C@@H](CC=2C=CC=CC=2)C(=O)N[C@H](C(=O)N1)C(C)C)[C@@H](C)O)[C@@H](C)CC)=O)[C@@H](C)CC)C1=CC=C(O)C=C1 ZPNFWUPYTFPOJU-LPYSRVMUSA-N 0.000 description 1
- 229910052500 inorganic mineral Inorganic materials 0.000 description 1
- 230000000968 intestinal effect Effects 0.000 description 1
- 230000003834 intracellular effect Effects 0.000 description 1
- 230000004068 intracellular signaling Effects 0.000 description 1
- 238000010212 intracellular staining Methods 0.000 description 1
- 208000028867 ischemia Diseases 0.000 description 1
- AGPKZVBTJJNPAG-UHFFFAOYSA-N isoleucine Natural products CCC(C)C(N)C(O)=O AGPKZVBTJJNPAG-UHFFFAOYSA-N 0.000 description 1
- 229960000310 isoleucine Drugs 0.000 description 1
- 230000000366 juvenile effect Effects 0.000 description 1
- 229940043355 kinase inhibitor Drugs 0.000 description 1
- GDBQQVLCIARPGH-ULQDDVLXSA-N leupeptin Chemical compound CC(C)C[C@H](NC(C)=O)C(=O)N[C@@H](CC(C)C)C(=O)N[C@H](C=O)CCCN=C(N)N GDBQQVLCIARPGH-ULQDDVLXSA-N 0.000 description 1
- 108010052968 leupeptin Proteins 0.000 description 1
- 230000006517 limb development Effects 0.000 description 1
- 210000004185 liver Anatomy 0.000 description 1
- 210000004072 lung Anatomy 0.000 description 1
- 239000006166 lysate Substances 0.000 description 1
- 210000002540 macrophage Anatomy 0.000 description 1
- 238000012423 maintenance Methods 0.000 description 1
- 230000036244 malformation Effects 0.000 description 1
- 230000036210 malignancy Effects 0.000 description 1
- 230000003211 malignant effect Effects 0.000 description 1
- 210000001161 mammalian embryo Anatomy 0.000 description 1
- 108010082117 matrigel Proteins 0.000 description 1
- 238000005259 measurement Methods 0.000 description 1
- 230000031864 metaphase Effects 0.000 description 1
- 238000013508 migration Methods 0.000 description 1
- 239000011707 mineral Substances 0.000 description 1
- 235000010755 mineral Nutrition 0.000 description 1
- 239000000203 mixture Substances 0.000 description 1
- 230000001343 mnemonic effect Effects 0.000 description 1
- 238000010369 molecular cloning Methods 0.000 description 1
- 238000000302 molecular modelling Methods 0.000 description 1
- 230000004784 molecular pathogenesis Effects 0.000 description 1
- 230000003562 morphometric effect Effects 0.000 description 1
- 238000013425 morphometry Methods 0.000 description 1
- 238000010172 mouse model Methods 0.000 description 1
- 208000000908 multiple cafe au lait spots Diseases 0.000 description 1
- 239000013642 negative control Substances 0.000 description 1
- 230000009826 neoplastic cell growth Effects 0.000 description 1
- 230000005937 nuclear translocation Effects 0.000 description 1
- 230000030648 nucleus localization Effects 0.000 description 1
- 238000011580 nude mouse model Methods 0.000 description 1
- 229920001778 nylon Polymers 0.000 description 1
- 210000000056 organ Anatomy 0.000 description 1
- 210000004789 organ system Anatomy 0.000 description 1
- 238000012261 overproduction Methods 0.000 description 1
- 210000004681 ovum Anatomy 0.000 description 1
- 108010068338 p38 Mitogen-Activated Protein Kinases Proteins 0.000 description 1
- 102000002574 p38 Mitogen-Activated Protein Kinases Human genes 0.000 description 1
- 238000004806 packaging method and process Methods 0.000 description 1
- 239000012188 paraffin wax Substances 0.000 description 1
- 230000008775 paternal effect Effects 0.000 description 1
- 230000008756 pathogenetic mechanism Effects 0.000 description 1
- 230000007170 pathology Effects 0.000 description 1
- 230000008068 pathophysiological alteration Effects 0.000 description 1
- 238000000059 patterning Methods 0.000 description 1
- 239000008188 pellet Substances 0.000 description 1
- 229950000964 pepstatin Drugs 0.000 description 1
- 108010091212 pepstatin Proteins 0.000 description 1
- FAXGPCHRFPCXOO-LXTPJMTPSA-N pepstatin A Chemical compound OC(=O)C[C@H](O)[C@H](CC(C)C)NC(=O)[C@H](C)NC(=O)C[C@H](O)[C@H](CC(C)C)NC(=O)[C@H](C(C)C)NC(=O)[C@H](C(C)C)NC(=O)CC(C)C FAXGPCHRFPCXOO-LXTPJMTPSA-N 0.000 description 1
- 239000003757 phosphotransferase inhibitor Substances 0.000 description 1
- 238000007747 plating Methods 0.000 description 1
- 230000008488 polyadenylation Effects 0.000 description 1
- 108010011110 polyarginine Proteins 0.000 description 1
- 238000003752 polymerase chain reaction Methods 0.000 description 1
- 230000008092 positive effect Effects 0.000 description 1
- 239000002243 precursor Substances 0.000 description 1
- 125000002924 primary amino group Chemical group [H]N([H])* 0.000 description 1
- 238000012545 processing Methods 0.000 description 1
- 230000000750 progressive effect Effects 0.000 description 1
- 230000000644 propagated effect Effects 0.000 description 1
- 239000003531 protein hydrolysate Substances 0.000 description 1
- 108060006633 protein kinase Proteins 0.000 description 1
- 230000036593 pulmonary vascular resistance Effects 0.000 description 1
- 238000011536 re-plating Methods 0.000 description 1
- 238000003753 real-time PCR Methods 0.000 description 1
- 102000027426 receptor tyrosine kinases Human genes 0.000 description 1
- 108091008598 receptor tyrosine kinases Proteins 0.000 description 1
- 102000005962 receptors Human genes 0.000 description 1
- 108020003175 receptors Proteins 0.000 description 1
- 230000002829 reductive effect Effects 0.000 description 1
- 238000011069 regeneration method Methods 0.000 description 1
- 230000009703 regulation of cell differentiation Effects 0.000 description 1
- 230000012760 regulation of cell migration Effects 0.000 description 1
- 230000025053 regulation of cell proliferation Effects 0.000 description 1
- 230000001105 regulatory effect Effects 0.000 description 1
- 230000010076 replication Effects 0.000 description 1
- 238000011160 research Methods 0.000 description 1
- 230000000241 respiratory effect Effects 0.000 description 1
- 238000007894 restriction fragment length polymorphism technique Methods 0.000 description 1
- 230000002441 reversible effect Effects 0.000 description 1
- 238000012552 review Methods 0.000 description 1
- 102200155479 rs121918460 Human genes 0.000 description 1
- 102200155473 rs121918461 Human genes 0.000 description 1
- 102200155728 rs121918462 Human genes 0.000 description 1
- 102200155671 rs121918463 Human genes 0.000 description 1
- 102200155721 rs121918464 Human genes 0.000 description 1
- 102200155746 rs121918466 Human genes 0.000 description 1
- 102200155580 rs121918470 Human genes 0.000 description 1
- 102200067300 rs281865270 Human genes 0.000 description 1
- 102220011047 rs397507541 Human genes 0.000 description 1
- 238000007790 scraping Methods 0.000 description 1
- 238000012216 screening Methods 0.000 description 1
- 229910052711 selenium Inorganic materials 0.000 description 1
- 239000011669 selenium Substances 0.000 description 1
- 238000002864 sequence alignment Methods 0.000 description 1
- 230000012488 skeletal system development Effects 0.000 description 1
- 238000007390 skin biopsy Methods 0.000 description 1
- 102000030938 small GTPase Human genes 0.000 description 1
- 108060007624 small GTPase Proteins 0.000 description 1
- 150000003384 small molecules Chemical class 0.000 description 1
- 239000001509 sodium citrate Substances 0.000 description 1
- NLJMYIDDQXHKNR-UHFFFAOYSA-K sodium citrate Chemical compound O.O.[Na+].[Na+].[Na+].[O-]C(=O)CC(O)(CC([O-])=O)C([O-])=O NLJMYIDDQXHKNR-UHFFFAOYSA-K 0.000 description 1
- 239000011775 sodium fluoride Substances 0.000 description 1
- 235000013024 sodium fluoride Nutrition 0.000 description 1
- 235000010267 sodium hydrogen sulphite Nutrition 0.000 description 1
- 208000020431 spinal cord injury Diseases 0.000 description 1
- 210000004989 spleen cell Anatomy 0.000 description 1
- 238000010561 standard procedure Methods 0.000 description 1
- 230000004936 stimulating effect Effects 0.000 description 1
- 238000007920 subcutaneous administration Methods 0.000 description 1
- 239000000126 substance Substances 0.000 description 1
- 239000006228 supernatant Substances 0.000 description 1
- 238000001356 surgical procedure Methods 0.000 description 1
- 230000006794 tachycardia Effects 0.000 description 1
- 208000008203 tachypnea Diseases 0.000 description 1
- 206010043089 tachypnoea Diseases 0.000 description 1
- 238000013518 transcription Methods 0.000 description 1
- 230000035897 transcription Effects 0.000 description 1
- 230000002103 transcriptional effect Effects 0.000 description 1
- 239000012096 transfection reagent Substances 0.000 description 1
- 239000012581 transferrin Substances 0.000 description 1
- 230000009261 transgenic effect Effects 0.000 description 1
- 230000007704 transition Effects 0.000 description 1
- 238000002054 transplantation Methods 0.000 description 1
- 230000032258 transport Effects 0.000 description 1
- 238000011269 treatment regimen Methods 0.000 description 1
- GPRLSGONYQIRFK-MNYXATJNSA-N triton Chemical compound [3H+] GPRLSGONYQIRFK-MNYXATJNSA-N 0.000 description 1
- 241000701161 unidentified adenovirus Species 0.000 description 1
- 239000004474 valine Chemical group 0.000 description 1
- LSGOVYNHVSXFFJ-UHFFFAOYSA-N vanadate(3-) Chemical compound [O-][V]([O-])([O-])=O LSGOVYNHVSXFFJ-UHFFFAOYSA-N 0.000 description 1
- 210000002845 virion Anatomy 0.000 description 1
- 230000003612 virological effect Effects 0.000 description 1
- 239000011782 vitamin Substances 0.000 description 1
- 235000013343 vitamin Nutrition 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
- DGVVWUTYPXICAM-UHFFFAOYSA-N β‐Mercaptoethanol Chemical compound OCCS DGVVWUTYPXICAM-UHFFFAOYSA-N 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
- C12N5/0696—Artificially induced pluripotent stem cells, e.g. iPS
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N5/00—Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
- C12N5/06—Animal cells or tissues; Human cells or tissues
- C12N5/0602—Vertebrate cells
- C12N5/0652—Cells of skeletal and connective tissues; Mesenchyme
- C12N5/0657—Cardiomyocytes; Heart cells
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2501/00—Active agents used in cell culture processes, e.g. differentation
- C12N2501/60—Transcription factors
- C12N2501/602—Sox-2
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2501/00—Active agents used in cell culture processes, e.g. differentation
- C12N2501/60—Transcription factors
- C12N2501/603—Oct-3/4
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2501/00—Active agents used in cell culture processes, e.g. differentation
- C12N2501/60—Transcription factors
- C12N2501/604—Klf-4
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2501/00—Active agents used in cell culture processes, e.g. differentation
- C12N2501/60—Transcription factors
- C12N2501/606—Transcription factors c-Myc
-
- 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
- C12N2503/00—Use of cells in diagnostics
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2506/00—Differentiation of animal cells from one lineage to another; Differentiation of pluripotent cells
- C12N2506/45—Differentiation of animal cells from one lineage to another; Differentiation of pluripotent cells from artificially induced pluripotent stem cells
Definitions
- Pluripotent stem cells can grow indefinitely while maintaining pluripotency, and can differentiate into cells of all three germ layers (Evans & Kaufman, Nature 292: 154-156 (1981)). Human pluripotent stem cells have promise for treating diseases such as Parkinson's disease, spinal cord injury and diabetes (Thomson et al, Science 282:1145-1147 (1998)).
- Somatic cells can be reprogrammed into pluripotent stem cells by transferring their nuclear contents into oocytes (Wilmut et al, Nature 385:810-813(1997)) or by fusion with embryonic stem (ES) cells (Cowan et al, Science 309: 1369-1373 (2005)), indicating that unfertilized eggs and ES cells contain factors that confer totipotency or pluripotency in somatic cells.
- Yu et al. showed that cells derived by in vitro differentiation from an Oct4 knock-in human ES cell line did not express EGFP, but that EGFP expression was restored upon cell-cell fusion with human ES cells (Yu et al, Stem Cells 24: 168-176 (2006)).
- iPS cells are pluripotent stem cell derived from non-pluripotent cells (e.g., adult somatic cells). iPS cells are typically produced by introducing into somatic cells certain transcription factors that are involved in maintaining ES cell pluripotency. Although the transcriptional
- Takahashi & Yamanaka introduced four reprogramming factors (Oct3/4, Sox2, c-Myc and Klf ) into mouse adult fibroblasts to obtain iPSCs. These iPSCs exhibited mouse ES cell morphology and growth properties, and expressed mouse ES cell marker genes (Takahashi & Yamanaka, Cell 126:663-676 (2006)). Notably, exogenous Oct-4 introduced into the mouse fibroblasts resulted in only marginal Oct-4 expression. Subcutaneous transplantation of iPS cells into nude mice resulted in tumors containing a variety of tissues from all three germ layers. Following injection into blastocysts, iPS cells contributed to mouse embryonic
- Yu et al. Science, 318:1917-20
- WO2008/0233610 disclose the reprogramming of human fibroblast cells into pluripotent stem cells using a lentiviral system and a different set of factors: OCT4, SOX2, NANOG, and LIN28.
- WO2008/0233610 notes that ES cells from mice and humans require distinct sets of factors to remain undifferentiated.
- LEOPARD syndrome (LS; also known as “Cardiocutaneous syndrome,” “Gorlin syndrome II,” “Lentiginosis profusa syndrome,” “Progressive cardiomyopathic lentiginosis,” “Capute-Rimoin-Konigsmark-Esterly-Richardson syndrome,” or “Moynahan syndrome”) is a rare autosomal dominant, multisystem disease having a complex of features, mostly involving the skin, skeletal and cardiovascular systems.
- Noonan syndrome is an autosomal dominant disorder characterized by dysmorphic facial features, proportionate short stature, and heart disease, i.e., pulmonic stenosis and hypertrophic cardiomyopathy most commonly (Noonan, Am. J. Dis. Child. 1968, 116:373- 380; Allanson, J. Med. Genet.
- NS is a relatively common syndrome with an estimated incidence of 1 : 1000 to 1 :2500 live births.
- PTPNl 1 non-receptor type 11
- SHP2, Syp, SHPTP2, PTP2C, PTP1D or BPTP3 wild-type PTPNl 1 protein
- PTP2C protein tyrosine phosphatase oncogenes
- BPTP3 protein tyrosine kinase oncogenes
- Somatic mutations in the PTPNl 1 gene also contribute to leukemogenesis in children (e.g., juvenile myelomonocytic leukemia (JMML), or acute myelogenous leukemia).
- JMML is a progressive myelodysplastic/myeloproliferative disorder characterized by
- Tyrosine phosphorylation is involved in the regulation of human cellular processes from cell differentiation and growth to apoptosis. The process of tyrosine
- phosphorylation is regulated by protein-tyrosine phosphatases (PTP) and protein-tyrosine kinases (PTK).
- PTP protein-tyrosine phosphatases
- PTK protein-tyrosine kinases
- activated PTPNl 1 protein interacts with the Gab family of docking proteins. This interaction activates a pathway leading to cell proliferation and tumorigenesis. Therefore, the PTPNl 1 protein signaling pathway can be an attractive therapeutic target for treating, for example, cancer, Noonan syndrome, and LEOPARD syndrome.
- Noonan syndrome e.g., heart defects, undescended testicles, or excessively short stature
- LEOPARD syndrome e.g., cryptorchidism, hypospadias, or severe skeletal deformity
- no therapeutic treatment of the underlying disorder has been determined so far.
- This invention generally relates to disease-specific pluripotent stem cells that are derived from somatic cells that carry a mutation in the PTPNl 1 gene.
- the stem cells are capable of differentiating into cells or tissues affected by the PTPNl 1 mutation, thereby providing a powerful tool to study various diseases that are associated with mutations in the PTPNl 1 gene, and to identify novel therapeutic agents for diagnosis and treatment of diseases that are associated with mutations in the PTPNl 1 gene.
- the invention provides an isolated primate pluripotent stem cell derived from a somatic cell, wherein the stem cell comprises a mutation in the protein tyrosine phosphatase non-receptor type 11 (PTPNl 1) gene.
- PTPNl 1 protein tyrosine phosphatase non-receptor type 11
- the PTPNl 1 gene comprises a mutation associated with a cardiac anomaly.
- the cardiac anomaly is hypertrophic cardiomyopathy.
- the PTPNl 1 gene comprises a mutation associated with LEOPARD Syndrome, Noonan Syndrome or leukemia.
- the mutation in the PTPNl 1 gene is a deletion mutation, a substitution mutation, an addition mutation, or a combination thereof, in the coding region of the PTPNl 1 gene. In certain embodiments, the mutation in the PTPNl 1 gene results in a mutation in the encoded PTPNl 1 protein.
- the mutation in the PTPNl 1 gene results in a threonine to methionine substitution in the encoded PTPNl 1 protein at an amino acid residue position corresponding to position 468 of SEQ ID NO: l, or a tyrosine to cysteine substitution in the encoded PTPNl 1 protein at an amino acid residue position corresponding to position 279 of SEQ ID NO: l .
- the mutation in the PTPNl 1 gene results in a tyrosine to cysteine substitution in the encoded PTPNl 1 protein at an amino acid residue position corresponding to position 279 of SEQ ID NO: 1, a tyrosine to serine substitution in the encoded PTPNl 1 protein at an amino acid residue position corresponding to position 279 of SEQ ID NO: 1, an alanine to threonine substitution in the encoded PTPNl 1 protein at an amino acid residue position corresponding to position 461 of SEQ ID NO: 1, a glycine to alanine substitution in the encoded PTPNl 1 protein at an amino acid residue position corresponding to position 464 of SEQ ID NO: 1, a threonine to methionine substitution in the encoded PTPNl 1 protein at an amino acid residue position corresponding to position 468 of SEQ ID NO: 1, a threonine to proline substitution in the encoded PTPNl 1 protein at an amino acid residue position corresponding
- the PTPNl 1 gene encodes a polypeptide comprising SEQ ID NO: 1, and the mutation in the PTPNl 1 gene results in a threonine to methionine substitution at amino acid residue position 468 of SEQ ID NO: 1 , or a tyrosine to cysteine substitution at amino acid residue position 279 of SEQ ID NO: 1.
- the pluripotent stem cell forms a teratoma in an immunocompromised mouse.
- the pluripotent stem cell expresses SSEA-3, SSEA-4, TRA-1-60, TRA-1-81, Nanog, OCT-4, Sox2, GDF3, DPPA4, REX1, TERT, Alkaline
- Phosphatase Phosphatase, Telomerase, or CD30, or a combination thereof.
- the pluripotent stem cell is capable of differentiating into a cardiomyocyte.
- stem cell culture of the stem cells described herein exemplary stem cell lines were deposited on , 2010 with American Type Culture Collection
- the invention provides a cardiomyocyte comprising a mutation in the PTPNl 1 gene, wherein the cardiomyocyte is produced by differentiating a disease-specific pluripotent stem cell as described herein.
- the invention provides a cell culture of a
- cardiomyocyte as described herein.
- Exemplary cardiomyocyte cell lines were deposited on
- the primate is human.
- the somatic cell is a fibroblast.
- the invention provides a method of identifying an agent for use in treatment of a disease that is associated with a mutation in the PTPNl 1 gene, comprising (1) contacting a disease-specific pluripotent stem cell as described herein with the agent; and (2) determining the effect of the agent on survival, proliferation, phenotype, or differentiation of the pluripotent stem cell.
- the invention provides a method of identifying an agent for use in treatment of a disease that is associated with a mutation in the PTPNl 1 gene, comprising (1) contacting a disease-specific cardiomyocyte as described herein with the agent; and (2) determining the effect of the agent on survival, proliferation, or phenotype of the cardiomyocyte.
- the invention provides a method of identifying an agent for use in treatment of the cardiac hypertrophy that is associated with a mutation in the PTPNl 1 gene, comprising: (1) contacting a cardiomyocyte as described herein with the agent; and (2) determining the effect of the agent on the hypertrophic state of the cardiomyocyte.
- a decrease in the hypertrophic state of the cardiomyocyte, as compared to that of a control cell, is indicative that the agent is an agent for use in treatment of a disease that is associated with a mutation in the PTPNl 1 gene.
- the invention provides a method of producing a pluripotent stem cell as described herein, comprising: (a) obtaining a somatic cell from a donor whose genome comprises a mutation in the PTPNl 1 gene; (b) introducing into the somatic cell: (i) a nucleic acid molecule that encodes a pluripotency-inducing protein; or (ii) a polypeptide that comprises a pluripotency-inducing protein; wherein said pluripotency-inducing protein is Oct3/4, Sox2, Klf4, Nanog, Lin28, c-Myc, or a combination thereof.
- one or more nucleic acid molecules that encode Oct3/4, Sox2, Klf4, and c-Myc are introduced into the somatic cell.
- the somatic cell is a fibroblast.
- the invention provides a method of differentiating a pluripotent stem cell as described herein into a cardiomyocyte, comprising: culturing a pluripotent stem cell as described herein in a serum- free medium that comprises: activin A, bone morphogenetic protein 4 (BMP4), basic fibroblast growth factor (bFGF), vascular endothelial growth factor (VEGF) and dickkopf homo log 1 (DK 1).
- BMP4 bone morphogenetic protein 4
- bFGF basic fibroblast growth factor
- VEGF vascular endothelial growth factor
- DK dickkopf homo log 1
- the invention provides a method of identifying an agent as a candidate Ras-signaling pathway inhibitor, comprising: (1) contacting a pluripotent stem cell as described herein with the agent; and (2) determining the effect of the agent on tyrosine phosphorylation of EGFR or MEK1. A decrease of tyrosine phosphorylation of EGFR or MEK1, as compared to that of a control cell, is indicative that the agent is a Ras-signaling pathway inhibitor.
- the invention provides a method of identifying an agent as a candidate Ras-signaling pathway inhibitor, comprising: (1) contacting a cardiomyocyte as described herein with the agent; and (2) determining the effect of the agent on tyrosine phosphorylation of EGFR or MEK1. A decrease of tyrosine phosphorylation of EGFR or MEK1, as compared to that of a control cell, is indicative that the agent is a Ras-signaling pathway inhibitor.
- the invention provides a method of identifying an agent for use in treatment of a disease that is associated with a mutation in the PTPNl 1 gene, comprising: (1) contacting a pluripotent stem cell as described herein with the agent; (2) differentiating the stem cell into a cardiomyocyte; and (3) determining the hypertrophic state of the cardiomyocyte. A decrease in the hypertrophic state of the cardiomyocyte, as compared to that of a control cell, is indicative that the agent is an agent for use in treatment of a disease that is associated with a mutation in the PTPNl 1 gene.
- the stem cells of the invention are capable of differentiating into a hematopoietic cell.
- hematopoietic cells may comprise a mutation in the PTPNl 1 gene, wherein the hematopoietic cell is produced by differentiating the stem cells of the invention.
- methods of identifying an agent for use in treatment of a disease that is associated with a mutation in the PTPN11 gene comprising (1) contacting the pluripotent stem cells with the agent; (2) differentiating the stem cell into a hematopoietic cell; and (3) determining the effect of the agent on survival, proliferation, phenotype, or differentiation of the hematopoietic cell.
- the disease is leukemia.
- methods of identifying an agent for use in treatment of leukemia comprising contacting the pluripotent stem cell with the agent; and differentiating the stem cell into a hematopoietic cell; and determining the proliferative state of the hematopoietic cell, wherein a decrease in the proliferative state of the hematopoietic cell, as compared to that of a control cell, is indicative that the agent is an agent for use in treatment of leukemia.
- Figs, la-lc demonstrate that the gene expression profile of LS-iPSC is similar to that of HESC.
- la quantitative real-time PCR assay for the expression of endogenous OCT4, NANOG and SOX2 in iPSC and parental fibroblasts (Fib).
- PCR reactions were normalized against ⁇ -ACTIN and plotted relative to expression levels in HES2. Error bars indicate ⁇ s.d. of triplicates, lb, bisulfite sequencing analyses of the OCT4 and NANOG promoters.
- the cell line and the percentage of methylation is indicated to the left of each cluster, lc, heat map showing hierarchical clustering of 3657 genes with at least two-fold expression change between the average of the three fibroblast cell lines versus all the iPSC lines/HES samples. Expression levels are represented by color; red indicates lower and yellow higher expression.
- Figs. 2a-2b demonstrate that LS-iPSC can differentiate in vitro and in vivo into all three germ layers.
- L2-iPS6 cells were differentiated as floating EBs for eight days and then plated onto gelatin-coated dishes and allowed to differentiate for another eight days. Immunocytochemistry showed cell types positively stained for differentiation markers including Desmin/SMA (mesoderm), AFP (endoderm), vimentin (mesoderm), and GFAP/piII-Tubulin (ectoderm). The arrow indicates a ⁇ -tubulin-positive cell. Scale bar, 100 ⁇ .
- HES2 HES2, Ll- iPSC and L2-iPSC were injected subcutaneously into the right hindleg of immuno-compromised NOD-SCID mice.
- the resulting teratomas were stained with hematoxylin and eosin and tissues representative of all three germ layers were observed.
- Figs. 3a-3d show that cardiomyocytes derived from LS-iPSC had hypertrophic features.
- 3a HES2, HI, wt S3-iPS4 and three LS-iPS clones were differentiated into cardiac lineage.
- Cell areas of 50 random cTNT-positive cardiomyocytes of each cell line were measured using Image J. Boxes show the span from the median (50th percentile) to the first and third quartiles. The lines represent the largest/smallest sizes that are no more than 1.5 times the median to quartile distance. Additional points drawn represent extreme values.
- 3b Sarcomeric organization was assessed in 50 cTNT positive (red) cardiomyocytes. Data are presented as mean ⁇ s.d.
- n 3; ** ⁇ 0.01 (Student's t-test).
- 3d Nuclear localization of NFATc protein in a cTNT -positive cell from L2-iPS10 is shown.
- Figs 4a-4c show the results of phosphoproteomic and MAPK activation analyses.
- 4a Protein extracts of two iPSC from each LS patient (LI and L2), wt iPSC (BJ- iPSB5) and HES2 were hybridized to an antibody microarray. The heat map represents the most significant protein changes preserved in all the comparison groups.
- 4b pMEKl and pEGFR expression was confirmed by Western blot using phospho-specific antibodies. Band density was measured (ImageJ software), and normalized to ⁇ -Actin.
- HES2, wt S3-iPS4, and LS-iPSC were serum- and bFGF-starved for 6 hours and then treated with bFGF (20 ng/ml) for the indicated time.
- Phosphorylated ERK1/2 (p-ER l/2) and total ER were assessed by
- p-ERKl/2 levels were compared to the untreated p-ERKl/2 level in each sample, normalized to the total ERK1/2 and represented graphically at the right of each panel.
- FIGs 5a-5b illustrate the formation of LS-iPSC. 5a, schematic representation of iPSC generation. 5b, typical image of a TRA-1-81 positive colony growing in the plate three weeks after infection.
- Figs. 6a-6b demonstrate that LS-iPSC lines were derived from their parental fibroblasts and maintained normal karyotypes.
- 6a the inventors PCR-amp lifted across three discrete genomic loci containing highly variable numbers of tandem repeats with different primer sets (D10S1214, D17S1290, and D21S2055). The resulting amplification patterns confirmed that each iPSC line was derived from its indicated parental fibroblast.
- 6b G-banding of HES2 cell line, wild-type iPS clone BJ-iPSB5, and LS-iPSC lines (one clone of each patient) demonstrates normal diploid chromosomal contents.
- Figs. 7a-7b show the result ⁇ T468M mutation analysis in LS-iPSC and fibroblasts.
- 7a the T468M point mutation in exon 12 of one allele of PTPN11 gene was verified by DNA sequencing.
- 7b the inventors amplified by RT-PCR a 1.2 Kb region containing the mutation sequence and the DNA was digested with BsmFI, an enzyme that cuts the 5'-GGGAC(N)io-3' sequence of the wild type allele but does not cut the mutant allele. In all the LS samples, an undigested 1.2 Kb band was observed.
- Figs. 8a-8b show the integration of retroviral transgenes in iPSC.
- 8a transgene-specific primers were used to amplify OCT4, SOX2, KLF4 and c-MYC.
- Three LS- derived iPSC lines from each patient were analyzed, and HES2 cells and parental fibroblasts were used as negative controls.
- 8b Southern blot analyses of Bgl-II digested gDNA extracted from HES2 cells, parental fibroblasts and iPSC, using DIG-labeled DNA probes against OCT4, SOX2, KLF4 and c-MYC.
- Retrovirally-inserted transgenic copies of these genes are indicated by asterisks and the number of detected bands is shown at the bottom.
- the parental fibroblasts and HES2 cells share bands in common with all the iPSC lines (arrowheads), which reflect the endogenous loci (including potential pseudogenes).
- Figs. 9a-9b demonstrate the silencing of retroviral transgenes in iPSC lines.
- 9a transgene-specific primers were used to determine OCT4, SOX2, KLF4 and c-MYC expression.
- 9b qPCR analysis of retroviral transgene (Tg) expression. Results are normalized against ⁇ -ACTIN and plotted relative to the expression levels in transfected GP2 cells.
- Figs. 10a- 10b demonstrate that LS-iPSC expressed cell markers that are common to pluripotent cells.
- 10a SSEA4 expression was determined by flow cytometry in two iPSC lines derived from two LEOPARD syndrome (LS) patients (Ll-iPSl, Ll-iPS13, L2-iPS6 and L2-iPS16), and a wt-iPSC line derived from BJ fibroblasts (BJ-iPSB5).
- HES2 cell line was used as positive control.
- HES2 and iPSC were grown on MEFs, and were fixed and stained for the following pluripotency markers: alkaline phosphatase (AP), TRA-1-81, TRA-1-60, NANOG and OCT4. Nuclei were stained with DAPI.
- Figs. 1 la-1 lb show the result of gene expression analysis of the cells.
- 11a qPCR was used to evaluate the expression of GDF3, REXl and TERT in two LS- iPSC lines from each patient and compared to their parental fibroblasts.
- HES2 cells were used as positive control of pluripotency gene expression.
- the position of SOX2, LIN28 and OCT4 transcription factors is indicated.
- Figs. 12a-12b illustrate in vitro differentiation of iPSC lines. 12a, day 8 EBs of HES2, BJ-iPSB5, Ll-iPS13 and L2-iPS6 cells. 12b, after 16 days of differentiation, cells were stained with SMA, GFAP and Vimentin antibodies. Scale bar, 100 ⁇ .
- Figs. 13a- 13b show that LS-iPSC differentiated into hematopoietic cells.
- 13a red EBs, an indication of erythrocyte development, were observed in Ll-iPSC plates 14 days after differentiation.
- Scale bar 100 ⁇ . 13b, CD41, CD45, CD1 lb, CD71 and CD235a hematopoietic markers were analyzed by flow cytometry in EBs derived from HES2 and Ll- iPSC, 18 days after induction of hematopoietic differentiation.
- Figs. 14a- 14c show the characterization of wt S3-iPS4 and L2-iPSC. 14a, the iPSC had the four transgenes integrated in their genome. 14b, Tg silencing analysis. 14c,
- Figs. 15a-15c show the pluripotency of S3-iPS4 and L2-iPSC.
- 15a and 15b S3-iPS4, L2-iPS10 and L2-iPS20 expressed pluripotency markers.
- 15c the iPSC differentiated into derivatives of the three germ layers.
- Figs. 16a- 16c show the disruption of MAPK activation upon bFGF
- 16a FGF receptors expression analysis by RT-qPCR in LS-iPSC and fibroblasts compared to HES2.
- 16b Ll-iPSl, Ll-iPS6 and HES2 cells were serum- and bFGF- starved overnight. The following day, the cells were treated with bFGF for 10 minutes (+) or not (-).
- Total ER 1/2 and p-ER l/2 expression was analyzed by immunobloting of total lysates.
- 16c basal p-ERKl/2 was quantified in each sample and compared to HES2C (top left panel). The relative increase of p-ERKl/2 level upon stimulation was quantified in each sample (top right and lower panels). All the p-ERKl/2 values were normalized to their corresponding total ERK1/2 values.
- Fig. 17 is a table showing the primer sets that were used for PCRs.
- Fig. 18 is a schematic drawing showing PTPN11 gene organization and domain structure.
- the numbered, filled boxes at the top indicate the coding exons; the positions of the ATG and TGA codons are shown.
- the functional domains of the PTPN11 protein consisting of two tandemly arranged src-homology 2(SH2) domains at the N-terminus (N-SH2 and C-SH2) followed by a protein tyrosine phosphatase (PTP) domain, are shown below.
- the numbers below that cartoon indicate the amino acid boundaries of those domains.
- Fig. 19 shows the distribution of PTPN11 (SHP-2) mutations and their relative prevalence in Noonan syndrome.
- Fig. 20 shows the cDNA sequence of human PTPN11 (SEQ ID NO: 2).
- Fig. 21 shows the amino acid sequence of human PTPN11 (SEQ ID NO: 1). DETAILED DESCRIPTION OF THE INVENTION 1. OVERVIEW
- This invention generally relates to disease-specific pluripotent stem cells that are derived from somatic cells that carry a mutation in the PTPN11 gene.
- the stem cells are capable of differentiating into cells or tissues (e.g., cardiomyocytes) affected by the PTPN11 mutation, thereby recapitulating both normal and pathologic tissue formation in vitro.
- the stem cells and cardiomyocytes provided herein can facilitate disease investigation and drug development - they may be used to study various diseases that are associated with mutations in the PTPN11 gene, and to identify novel therapeutic agents for diagnosis and treatment of diseases that are associated with mutations in PTPN11.
- the present invention is based on the discovery that somatic cells from
- LEOPARD syndrome (LS) patients which carry a mutation in the PTPN11 gene, can be reprogrammed into pluripotent stem cells. Further, the stem cells may be cultured under appropriate differentiation conditions to generate differentiated, disease-specific cardiomyocytes. Such disease-specific stem cells and cardiomyocytes are useful for studying the genetic basis of disease progression and pathogenesis, e.g., by comparing the survival, proliferation, phenotype, or differentiation of the disease-specific cells to that of cells that do not carry the mutation in the PTPN11 gene.
- LS-iPSC LS-specific induced pluripotent stem cell
- vz ' tro-derived cardiomyocytes from LS-iPSC were larger, had a higher degree of sarcomeric organization and showed preferential localization of NFATc4 in the nucleus, when compared to cardiomyocytes derived from human embryonic stem cells (HESC) or iPSCs derived from an unaffected brother (i.e., not having the PTPN11 mutation) of one of the LS patients.
- HESC human embryonic stem cells
- iPSCs derived from an unaffected brother i.e., not having the PTPN11 mutation
- the disease-specific iPSCs and cardiomyocytes provided herein offer several advantages for disease study and drug screening as compared to existing cell lines. For example, most of the human cell lines in wide use today are derived either from malignant tissues or are genetically modified to drive immortal growth. On the other hand, primary human cells have a limited life span in culture, a constraint that thwarts inquiry into the regulation of tissue formation, regeneration, and repair. Therefore, the immortal pluripotent stem cells and the in vitro differentiated cardiomyocytes provided herein are particularly valuable as disease models for studying mutations in the PTPN11 gene.
- pluripotency refers to the nature of a cell, i.e., an ability to differentiate into a variety of tissues or organs. Typically, the pluripotency of a differentiated cell is limited.
- reprogramming refers to a genetic process whereby differentiated somatic cells are converted into de-differentiated, pluripotent cells, and thus having a greater pluripotency potential than the cells from which they were derived.
- the reprogrammed cells may express at least one of the following pluripotent cell- specific markers: SSEA-3, SSEA-4, TRA-1-60 or TRA 1-81.
- the reprogrammed cells express all these markers.
- the invention provides an isolated primate pluripotent stem cell derived from a somatic cell, wherein the stem cell comprises a mutation in the protein tyrosine phosphatase non-receptor type 11 (PTPN11) gene. Mutations in the PTPN11 gene have been implicated in several diseases, such as LEOPARD syndrome, Noonan syndrome, and leukemia. Therefore, the stem cells provided herein are useful for studying the pathologic tissue formation in vitro, and for screening novel therapeutic agents for treating or diagnosing diseases that are associated with PTPN11 mutations.
- PTPN11 protein also known as SHP2, Syp, SHPTP2, PTP2C, PTP1D or BPTP3
- SHP2, Syp SHPTP2, PTP2C, PTP1D or BPTP3
- SH2-domain-containing protein tyrosine phosphatases that control cellular proliferation and differentiation
- Human PTPN11 is ubiquitously expressed in all tissues examined, with higher levels of expression in the heart and the brain (Ahmad et al, Proc Natl Acad Sci USA 1993;90:2197-2201; Bastien et al, Biochem Biophys Res Commun 1993 ; 196: 124-133; Freeman et al, Proc Natl Acad Sci USA, 1992;89: 11239-11243).
- PTPN11 is a key molecule in intracellular signaling and is necessary for activation of the RAS/MAPK cascade in response to a variety of growth factors, hormones and cytokines (Maroun et al, Mol Cell Biol 2000;20:8513-25; Shi Z-Q, et al, Mol Cell Biol 2000; 20: 1526-1536; Cunnick et al, J Biol Chem 2002;277:9498-504).
- PTPN11 is required during embryogenesis for mesodermal patterning (Tang et al, Cell 1995;80:473-483), semilunar valvuogenesis (Chen et al, Nat Genet 2000;24:296-9) and skeletal and limb development (Qu et al, Mol Cell Biol 1998; 18:6075-82; Saxon et al, Nat Genet 2000;24:420-3).
- PTPN11 also controls cell differentiation at later stages of hematopoiesis, and has a role in the function of differentiated erythroid, myeloid and lymphoid cells (Pazdrak et al, J Exp Med 1997; 186:561-8; Edmead et al, FEBS Lett 1999;459:27-32; Ohtani et al, Immunity 2000;12:95-105; Tamir et al, Curr Opin Immunol 2000;12:307-15; Bordin et al, Blood 2002; 100:276-82).
- the human PTPNl 1 gene organization and intron boundary sequence can be established using cDNA (GENBANK Accession Nos. NM_002834; amino acid and nucleotide sequences represented herein as SEQ ID Nos.: 1 and 2, respectively) and genomic sequences (GENBANK Accession Nos. NG 007459).
- Figure 18 shows the organization of the PTPNl 1 gene and the functional domains of the PTPNl 1 protein.
- the PTPNl 1 protein comprises two SH2 (src-homology 2) domains, one from amino acid 3 to amino acid 104, the other from amino acid 112 to amino acid 216, and one PTP (protein tyrosine phosphatase) domain, from amino acid 221 to amino acid 524.
- the PTPNl 1 gene of the invention also encompasses nucleic acid sequences comprising a coding sequence as set forth in SEQ ID NO:2, homo logs (including allelic variants and orthologs), sequence-conservative variants, or function-conservative variants thereof.
- the PTPNl 1 protein of the invention also encompasses amino acid sequences comprising the amino acid sequence as set forth in SEQ ID NO: l, homo logs (including orthologs), or function- conservative variants thereof.
- homologous in all its grammatical forms and spelling variations refers to the relationship between polynucleotides or proteins that possess a "common evolutionary origin,” including polynucleotides or proteins from superfamilies and homologous polynucleotides or proteins from different species (Reeck et al, Cell 50:667, 1987). Such polynucleotides or proteins have sequence homology, as reflected by their sequence similarity, whether in terms of percent identity or the presence of specific amino acids or motifs at conserved positions.
- sequence-conservative variants of a polynucleotide sequence are those in which a change of one or more nucleotides in a given codon position results in no alteration in the amino acid encoded at that position.
- “Function-conservative variants” are those in which one or more nucleotides of a polynucleotide sequence have been changed without altering the overall conformation and function of the polypeptide encoded by the polynucleotide; or one or more amino acid residues of a polypeptide sequence have been changed without altering the overall conformation and function of the polypeptide.
- Function-conservative variants includes, e.g., replacement of an amino acid with one having similar properties (such as, for example, polarity, hydrogen bonding potential, acidic, basic, hydrophobic, aromatic, and the like). Amino acids with similar properties are well known in the art.
- arginine, histidine and lysine are hydrophilic- basic amino acids and may be interchangeable.
- isoleucine a hydrophobic amino acid, may be replaced with leucine, methionine or valine. Such changes are expected to have little or no effect on the apparent molecular weight or isoelectric point of the protein or polypeptide.
- amino acid position "corresponding to" a position in another sequence is the position that lines up to the reference position when the amino acid sequence is aligned with the reference sequence. Alignments can be done by hand or by using well-known sequence alignment programs such as ClustalW2.
- variants of PTPN11 gene may include those polynucleotide sequences comprising a coding sequence that is at least about 70%, about 75%, about 80%>, about 85%, about 90%, about 95%, or about 99% identical to SEQ ID NO:2.
- the variant gene encodes a protein that has the same or substantially similar properties or functions as the native or parent PTPN11 protein.
- variants of PTPN11 protein may include those polypeptide sequences comprising an amino acid sequence that is at least about 70%>, about 75%, about 80%, about 85%, about 90%, about 95%, or about 99% identical to SEQ ID NO: 1.
- the variant protein has the same or substantially similar properties or functions as the native or parent PTPN11 protein.
- a disease is "associated with" a mutation in the PTPN11 gene when the disease is directly or indirectly caused by, or correlates with a mutation in the PTPN11 gene.
- Such mutation of PTPN11 may lead to a non- functioning or defective PTPN11 protein, or altered expression level of the PTPN11 gene.
- Such mutation may be a substitution mutation (i.e., a mutation in which one or more bases within the nucleic acid sequence have been replaced by a different base), an insertion mutation (i.e, a mutation in which the total length of the gene of interest has been increased by the insertion of one or more bases), a deletion mutation (a mutation in which the total length of the gene of interest has been decreased by removal of one or more bases) or an inversion mutation (a mutation in which a region of two or more bases has been rotated 180 degrees), or a combination thereof.
- the mutation may be directly, or indirectly, fully or partly responsible for the disease, or alternatively, the mutation may not be responsible for the disease but correlates with the diseased state in the sense that it is diagnostic/indicative of the diseased state.
- the mutation in the PTPN11 gene results in a mutation in the encoded PTPN11 protein.
- the stem cells described herein comprise a mutation in the PTPN11 gene that is associated with Noonan Syndrome.
- NS Noonan syndrome
- Such disorders include, but are not limited to, the Watson (MIM 193520) and LEOPARD (MIM 151100) Syndromes (Mendez and Opitz, Am J Med Genet 1985;21 :493-506); male Turner and female pseudo-Turner Syndrome, as well as Turner phenotype with normal karyotype (see MIM 163950); Noonan syndrome with multiple giant-cell lesions (MIM 163955; Tartaglia et al, Am J Hum Genet 2002;70: 1555-1563) and/or Noonan syndrome with multiple caf-au-lait spots (also known as LEOPARD syndrome, MIM 151100; Digilio et al, Am J Hum Genet 2002;71 :389-394; Legius et al, J Med Genet 2002;39:571-574); valvular sclerosis (Snellen et al, Circulation 1968;38(1 Suppl):93-101); and idiopathic short stature (Attie K
- NS encompasses familial or sporadic forms, including NS1, whose locus has been identified on chromosome 12.
- the present invention takes into consideration, however, that NS and its related disorders are genetically heterogeneous, but share phenotypical features.
- the features of NS have been well described and a clinical scoring system devised. See, Mendez and Opitz, Am J Med Genet 1985;21 :493-506; Noonan, Clin Pediatr (Phila) 1994;33:548-555; Sharland et al, Arch Dis Child 1992;67: 178-183; Duncan et al, Am J Med Genet 1981;10:37- 50).
- phosphotyrosine phosphatase (PTP) domain About one-third of the patients who had mutations in the PTPN11 gene had the N308D mutation, which was by far the most common. That codon 308 is a hotspot for Noonan syndrome is further supported by the finding of an Asn308-to-Ser missense mutation in 2 families that had typical features of Noonan syndrome associated with multiple giant cell lesions in bone (Tartaglia et al., Am. J. Hum. Genet. 70: 1555-1563, 2002).
- Maheshwari et al. (Hum. Mutat. 20: 298-304, 2002) surveyed 16 subjects with the clinical diagnosis of Noonan syndrome from 12 families and their relevant family members for mutations in the PTPN11/SHP2 gene, and found 3 different mutations among 5 families.
- Two unrelated subjects shared a Ser502-to-Thr (S502T) substitution in exon 13; 2 additional unrelated families had a Tyr63-to-Cys (Y63C) mutation in exon 3; and 1 subject had a Tyr62-to-Asp (Y62D) substitution, also in exon 3.
- S502T Ser502-to-Thr
- Y63C Tyr63-to-Cys
- Y62D Tyr62-to-Asp
- Tartaglia et al. investigated the parental origin of de novo PTPN11 lesions and explored the effect of paternal age in Noonan syndrome. By analyzing intronic positions that flank the exonic PTPN11 lesions in 49 sporadic Noonan syndrome cases, they traced the parental origin of mutations in 14 families. All mutations were inherited from the father.
- A-to-G mutation at nucleotide 182 in exon 3 Asp61-to-Gly, D61G
- 218C-T mutation in exon 3 Thr73-to-Ile, T73I
- Fig. 19 shows the distribution of PTPNl 1 (SHP-2) mutations and their relative prevalence in Noonan syndrome
- the stem cells described herein comprise a mutation in the PTPNl 1 gene that is associated with LEOPARD Syndrome.
- LEOPARD syndrome is an autosomal dominant disorder characterized by lentigines and cafe-au-lait spots, facial anomalies, and cardiac defects, sharing several clinical features with Noonan syndrome. Digilio et al. (Am. J. Hum. Genet. 71 : 389-394, 2002) screened 9 patients with LEOPARD syndrome (including a mother-daughter pair), and 2 children with Noonan syndrome who had multiple cafe-au-lait spots, for mutations in the PTPNl 1 gene.
- LEOPARD syndrome mutants are catalytically defective and act as dominant-negative mutations that interfere with growth factor/ERK-MAPK-mediated signaling.
- Molecular modeling and biochemical studies suggested that LEOPARD syndrome mutations control the SHP2 catalytic domain and result in open, inactive forms of SHP2.
- Kontaridis et al. concluded that the pathogenesis of LEOPARD syndrome is distinct from that of Noonan syndrome and suggested that these disorders should be distinguished by mutation analysis rather than clinical
- PTPNl 1 Other diseases that are known to be associated with a mutation in the PTPNl 1 gene include, e.g., cardiofaciocutaneous syndrome (MIM 163955), juvenile myelomonocytic leukemia (NIM 607785), and certain other malignancies.
- MIM 163955 cardiofaciocutaneous syndrome
- NIM 607785 juvenile myelomonocytic leukemia
- certain other malignancies include, e.g., cardiofaciocutaneous syndrome (MIM 163955), juvenile myelomonocytic leukemia (NIM 607785), and certain other malignancies.
- the stem cells described herein comprise a mutation in the PTPNl 1 gene that is associated with leukemia.
- JMML Juvenile myelomonocytic leukemia
- MDS myelodysplasia syndrome
- JMML is observed occasionally in patients with Noonan syndrome.
- Tartaglia et al. found heterozygosity with respect to a mutation in exon 3 of PTPNl 1.
- Four of the children shared the same mutation (218C-T).
- missense defects in PTPNl 1 the 218C-T transition, and a defect in exon 13 affecting the protein tyrosine phosphatase domain.
- Tartaglia et al. also identified missense mutations in PTPNl 1 in 21 of 62 individuals with JMML but without Noonan syndrome, with 9 different molecular defects in exon 3 and 1 in exon 13.
- Nonhemato logic DNAs were available for 9 individuals with a mutation in PTPNl 1 in their leukemic cells, and none harbored the defect.
- PTPNl 1 mutations that have been associated with JMML include, e.g., 226G-A mutation (Glu76-to-Lys, E76K), Glu76-to-Val, Glu76-to-Gly, Glu76-to-Ala (Tartaglia et al).
- Tartaglia et al. also investigated the prevalence of somatic mutations in PTPNl 1 among 50 children with myelodysplasia syndrome. They identified missense mutations in exon 3 in 5 of 27 children with an excess of blasts. Three of these mutations were also associated with JMML in other patients. Among 24 children with de novo AML (MIM 601626), they identified a novel trinucleotide substitution in an infant with acute monoblastic leukemia.
- the stem cells described herein comprise a mutation in the PTPNl 1 gene that is associated with a cardiac anomaly.
- a cardiac anomaly refers to any structural or functional abnormality or defect of the heart or great vessels.
- the general effects of cardiac malformations on cardiovascular functioning are increased cardiac workload, increased pulmonary vascular resistance, inadequate cardiac output, and, in the case of cyanotic anomalies, decreased oxygen saturation.
- the general physical symptoms of these pathophysiologic alterations are growth retardation, decreased exercise tolerance, recurrent respiratory infections, dyspnea, tachypnea, tachycardia, cyanosis, tissue hypoxia, and murmurs, all of which vary in severity, depending on the type and degree of the defect.
- CFC cardiofaciocutaneous syndrome
- Noonan syndrome namely, congenital heart defects, cutaneous abnormalities, Noonan-like facial features, and severe psychomotor developmental delay.
- T411M Thr411 -to-Met
- the stem cells described herein comprise a mutation in the PTPNl 1 gene that is associated with cardiac hypertrophy (also known as hypertrophic cardiomyopathy) .
- Cardiac hypertrophy refers to the process in which cardiac myocytes respond to stress through hypertrophic growth. Such growth is characterized by cell size increases without cell division, assembling of additional sarcomeres within the cell to maximize force generation, and an activation of a fetal cardiac gene program. Pathologic cardiac hypertrophy is often associated with increased risk of morbidity and mortality, and thus studies aimed at understanding the molecular mechanisms of cardiac hypertrophy could have a significant impact on human health.
- the stem cells described herein comprise a threonine to methionine substitution in PTPNl 1 protein at an amino acid residue position corresponding to position 468 of SEQ ID NO: 1 , or a tyrosine to cysteine substitution in PTPNl 1 protein at an amino acid residue position corresponding to position 279 of SEQ ID NO: 1.
- the PTPNl 1 protein comprises SEQ ID NO: 1, and the mutation is a threonine to methionine substitution at amino acid residue position 468 of SEQ ID NO: 1 , or a tyrosine to cysteine substitution at amino acid residue position 279 of SEQ ID NO: 1.
- the disease-specific pluripotent stem cells provided herein show two important characteristics that distinguish them from other types of cells. First, they are non- lineage committed cells that are capable of maintaining their pluripotent state and of renewing themselves for long periods through cell division. Second, under appropriate conditions, they can be induced to differentiate into cells with specialized functions.
- the disease-specific pluripotent stem cells may be characterized by any of several criteria of pluripotency known in the art.
- the stem cells are capable of continuous indefinite replication in vitro.
- Continued proliferation for a long period of time (e.g., at least about 1 month, at least about 2 months, at least about 4 months, at least about 6 months, at least about 9 months, at least about one year) of a culture is sufficient evidence of immortality, as primary cell cultures without this property fail to continuously divide for such a length of time (Freshney, Culture of animal cells. New York: Wiley-Liss, 1994).
- the disease-specific pluripotent stem cells may be characterized by the expression of certain markers, including but not limited to cell surface markers.
- Stem cells from different species may exhibit species-specific markers on their cell surfaces.
- Thomson U.S. Pat. Nos. 5,843,780 and 6,200,806 discloses certain cell surface markers that may be used to identify pluripotent stem cells derived from primates.
- Stage Specific Embryonic Antigens are unique carbohydrate epitopes that may be used to characterize pluripotent stem cells. Stem cells derived from different species exhibit different patterns of SSEAs. For example, undifferentiated primate pluripotent stem cells (including human pluripotent stem cells) express SSEA-3 and SSEA-4, but not SSEA-1.
- Undifferentiated mouse pluripotent stem cells express SSEA-1, but not SSEA-3 or SSEA-4.
- markers that are not exhibited on the surface of a cell may be used to characterize a pluripotent stem cell.
- the homeodomain transcription factor Oct 4 also termed Oct-3 or Oct3/4 is frequently used as a marker for pluripotent stem cells.
- the pluripotent stem cells provided herein express SSEA-3, SSEA-4, TRA-1-60, TRA-1-81, Nanog, OCT-4, Sox2, GDF3, DPPA4, REX1, TERT, Alkaline Phosphatase, Telomerase, or CD30, or a combination thereof.
- the disease-specific pluripotent stem cells described herein are capable of differentiating into all three embryonic germ layers (endoderm, mesoderm, and ectoderm) in a fashion similar to embryonic stem cells. Such potency and differentiability can be demonstrated by, e.g., teratoma formation (e.g., injecting the stem cells into an immunodeficient mouse such as a SCID mouse, and then histologically examining the resulting tumors). Additionally or alternatively, the disease-specific pluripotent stem cells may be characterized by the capacity to develop into fully differentiated somatic cell lineages (e.g., neurons, cardiomyocytes, etc) and/or the germ line.
- somatic cell lineages e.g., neurons, cardiomyocytes, etc
- the disease-specific pluripotent stem cells may be characterized by the capacity to participate in normal development when transplanted into a preimplantation embryo to generate a chimeric embryo.
- the disease-specific pluripotent stem cells may be characterized by their morphology (a stem cell has round shape, large nucleolus and scant cytoplasm; human iPSCs form sharp-edged, flat, tightly-packed colonies similar to human embryonic stem cells), telomerase activity (pluripotent stem cells express high telomerase activity to sustain self-renewal and proliferation), promoter demethylation (promoters of pluripotency-inducing genes, such as Oct-3/4, Rexl, and Nanog, are generally demethylated in iPSCs), or histone demethylation (H3 histones associated with Oct-3/4, Sox2, and Nanog genes are generally demethylated).
- ATCC Type Culture Collection
- P.O. Box 1549 Manassas, VA 20108, USA, under ATCC Accession No. and .
- the pluripotent stem cell of the present invention is a primate stem cell.
- the primate is human.
- the pluripotent stem cell is a non-human stem cell (e.g., a mouse stem cell, a rat stem cell, a pig stem cell, a sheep stem cell, a goat stem cell, or a non-human primate stem cell).
- pluripotent stem cells to self renew in culture, while retaining their pluripotent potential, provides the opportunity to produce virtually unlimited numbers of undifferentiated and differentiated cell types for in vitro and in vivo investigation of disease mechanisms (Lerou, P. H. & Daley, G. Q. Therapeutic potential of embryonic stem cells. Blood Rev 19, 321-31, 2005; Ben-Nun, I. F. & Benvenisty, N. Human embryonic stem cells as a cellular model for human disorders. Mol Cell Endocrinol 252, 154-9, 2006).
- Pluripotent stem cells carrying the genes responsible for a particular disease e.g., a mutation in the PTPN11 gene
- Studies of the differentiated cells in culture could provide important information regarding the molecular and cellular nature of events leading to pathology.
- this approach is used to develop an in vitro model of LEOPARD syndrome and cardiac hypertrophy.
- induced pluripotent stem cell lines were derived from two LEOPARD syndrome patients, and an unaffected brother not bearing the T468M mutation of one of the LS patients. These three pluripotent stem cell lines were differentiated into cardiomyocytes in culture. These in vitro differentiated cardiomyocytes could be maintained in culture, providing the opportunity to detect differences between the cardiomyocytes derived from stem cells carrying a mutant PTPN11 and those derived from stem cells that do not carry the mutation.
- the disease-specific pluripotent stem cells described herein may be induced in vitro to differentiate into a variety of somatic cell lineages (e.g., neurons, cardiomyocytes, etc).
- somatic cell lineages e.g., neurons, cardiomyocytes, etc.
- the invention provides cardiomyocytes comprising a mutation in the PTPN11 gene, wherein the cardiomyocyte is produced by differentiating the disease-specific pluripotent stem cell described herein.
- the disease-specific pluripotent stem cells may be subjected to any conditions that induce the stem cells to differentiate into cardiomyocytes.
- the disease-specific pluripotent stem cells may be suspended into a single-cell suspension, allowed to spontaneously aggregate into embryoid bodies over a first period of time (e.g. 48 hours, although such a period of time may be increased or decreased depending on other conditions to which the embryonic stem cells are subjected), and then treated with a suitable differentiation factor or factors for a second period of time such that the stem cells differentiate into cardiomyocytes.
- such differentiation factors may include activin A, bone morphogenetic protein 4 (BMP4), basic fibroblast growth factor (bFGF), vascular endothelial growth factor (VEGF) dickkopf homo log 1 (DK 1), or a combination thereof.
- BMP4 bone morphogenetic protein 4
- bFGF basic fibroblast growth factor
- VEGF vascular endothelial growth factor
- DK 1 dickkopf homo log 1
- the invention provides a method of differentiating a pluripotent stem cell as described herein into a cardiomyocyte, comprising culturing the stem cell in a serum- free medium that comprises: activin A, bone morphogenetic protein 4 (BMP4), basic fibroblast growth factor (bFGF), vascular endothelial growth factor (VEGF) and dickkopf homolog 1 (DKK1).
- BMP4 bone morphogenetic protein 4
- bFGF basic fibroblast growth factor
- VEGF vascular endothelial growth factor
- DKK1 dickkopf homolog 1
- the stem cells may be induced to differentiate into
- cardiomyocytes in the presence of a prostaglandin, analogue or functional equivalent thereof, alone or in combination with other factors (such as minerals (e.g., selenium), small molecules (e.g., a p38 MAPK inhibitor such as SB203580), transferring, protein growth factors of the FGF, IGF and BMP families (e.g., IGF1, FGF2, BMP2, BMP4, BMP6, or a homologue or functional equivalent thereof), ⁇ - ⁇ , non-essential amino acids, L-glutamine, etc.), as described in U.S. Application Publication No. 20070204351.
- factors such as minerals (e.g., selenium), small molecules (e.g., a p38 MAPK inhibitor such as SB203580), transferring, protein growth factors of the FGF, IGF and BMP families (e.g., IGF1, FGF2, BMP2, BMP4, BMP6, or a homologue or functional equivalent thereof), ⁇ - ⁇ , non-essential amino acids, L-
- the prostaglandin, analogue or functional equivalent thereof may be prostacyclin (PGI2), an analogue or functional equivalent thereof, including its naturally breakdown form of 6-keto-Prostaglandin Fla (6k-PGFla) and 2,3-dinor-6-keto- Prostaglandin Fla (2,3d-6k-PGFla); its synthetic analogs, such as iloprost, cicaprost, and carbarprostacyclin (cPGI) and stable chemical structures (Whittle, 1980, Town, 1982,
- cardiomyocyte progenitors that are differentiated from the pluripotent stem cells described herein.
- a cardiomyocyte progenitor, or cardiac progenitor cell refers to a cell that is resident in the heart, or that comes into the heart from elsewhere after acute ischemia, is smaller than a mature cardiomyocyte, expresses a- sarcomeric actin but is negative for troponin, is normally quiescent but can be induced to go into the cell cycle as defined by positive Ki67 staining.
- the cardiomyocytes and cardiomyocyte progenitors described herein may be beating.
- the cardiomyocytes may be fixed and stained with a-actinin antibodies to confirm muscle phenotype.
- a-troponin, a-tropomysin and a-MHC antibodies also give characteristic muscle staining.
- a cardiomyocyte differentiated from a stem cell comprises a mutation in the PTPN11 gene.
- the mutation is associated with a cardiac anomaly (such as hypertrophic cardiomyopathy).
- the mutation is associated with LEOPARD Syndrome, Noonan Syndrome or leukemia.
- a threonine to methionine substitution at amino acid residue position 468 of PTPN11 protein (SEQ ID NO: 1), or a tyrosine to cysteine substitution at amino acid residue position 279 of PTPN11 protein (SEQ ID NO: 1) is associated with LEOPARD syndrome/ hypertrophic cardiomyopathy.
- the invention provides a cell culture of the cardiomyocytes described herein. Exemplary cardiomyocyte cell lines were deposited on
- the invention provides methods of producing the disease-specific pluripotent stem cells as described herein.
- the invention provides methods of producing the disease-specific pluripotent stem cells comprising (1) obtaining a somatic cell from a donor whose genome comprises a mutation in the PTPN11 gene, and (2) reprogramming the somatic cell into a pluripotent stem cell.
- the reprogramming comprises introducing into the somatic cell: (i) a nucleic acid molecule that encodes a pluripotency- inducing protein; or (ii) a polypeptide that comprises a pluripotency-inducing protein.
- a somatic cell nucleus can be reprogrammed by treating a differentiated cell with an undifferentiated human carcinoma cell extract (Taranger et al. Mol. Biol. Cell 16:5719-35, 2005).
- Induced pluripotent stem cells typically can be produced by introducing into somatic cells certain proteins (in particular, transcription factors) that are involved in maintaining ES cell pluripotency. These proteins are referred herein as "pluripotency-inducing proteins.” Exemplary pluripotency-inducing proteins and genes encoding pluripotency-inducing proteins have been disclosed by Yamanaka et al. (EP Application Publication No. 1970446; US
- the pluripotency-inducing proteins are referred to as "nuclear reprogramming factors" by Yamanaka, and “potency-determining factors” by Thomson.
- Exemplary pluripotency-inducing protein include, but are not limited to, Oct- 4, Sox2, FoxD3, UTF1, Stella, Rexl, ZNF206, Soxl5, Mybl2, Lin28, Nanog, DPPA2, ESG1, Otx2, Klf4, c-Myc, or combinations thereof.
- the pluripotency-inducing protein is a protein from the Oct family (e.g., Oct3/4), Klf family (e.g., Klf4), Sox family (e.g., Sox2), or a combination thereof.
- a set of two or more pluripotency-inducing proteins are introduced into the somatic cells.
- the set include Oct-4 and Sox2.
- the set include Oct3/4, Sox2, Klf4, and c-Myc.
- the set include Oct3/4, Klf and c-Myc.
- the set include OCT4, SOX2, NANOG, and LIN28.
- the set of pluripotency-inducing proteins sufficient to reprogram somatic cells can vary with the cell type of the somatic cells.
- Suitable somatic cells can be any somatic cell, although higher
- Somatic cells useful in the invention may be non-embryonic cells obtained from a fetal, newborn, juvenile or adult primate, including a human.
- somatic cells that can be used with the methods described herein include, but are not limited to, bone marrow cells, epithelial cells, fibroblast cells, hematopoietic cells, hepatic cells, intestinal cells, mesenchymal cells, myeloid precursor cells and spleen cells.
- Another type of somatic cell is a mesenchymal cell that attaches to a substrate.
- the somatic cells can be cells that can themselves proliferate and differentiate into other types of cells, including blood stem cells (multipotent hematopoietic cells), muscle/bone stem cells, brain stem cells, liver stem cells, etc.
- blood stem cells multipotent hematopoietic cells
- muscle/bone stem cells muscle/bone stem cells
- brain stem cells brain stem cells
- liver stem cells etc.
- the somatic cell is a fibroblast.
- Suitable somatic cells are receptive, or can be made receptive using methods generally known in the art, to uptake pluripotency-inducing proteins or nucleic acid sequences that encode the pluripotency-inducing proteins. Uptake-enhancing methods can vary depending on the cell type and expression system. Exemplary conditions used to prepare receptive somatic cells having suitable transduction efficiency are known in the art (such as electroporation).
- a nucleic acid encoding a pluripotency-inducing protein can be introduced by transfection or transduction into the somatic cells using a vector, such as an integrating- or non- integrating vector (e.g., a retroviral vector, an adenoviral vector).
- a retroviral vectors e.g., a lentiviral vector
- the DNA segment(s) encoding the pluripotency-inducing protein can be located extra-chromosomally (e.g., on an episomal plasmid) or stably integrated into cellular chromosome(s).
- a viral-based gene transfer and expression vector is a genetic construct that enables efficient and robust delivery of genetic material to most cell types, including non- dividing and hard-to-transfect cells (primary, blood, stem cells) in vitro or in vivo. Often, viral- based constructs that are integrated into genomic DNA result in high expression levels.
- the vectors may include a transcription promoter and a polyadenylation signal operatively linked, upstream and downstream, respectively, to the DNA segment.
- nucleic acid encoding a pluripotency-inducing protein may be operably linked to a heterologous promoter, which may become inactive after somatic cells are reprogrammed.
- the heterologous promoter is any promoter sequence that can drive expression of a polynucleotide sequence encoding the pluripotency-inducing protein in the somatic cell, such as an Oct4 promoter.
- the vector can include a single DNA segment encoding a single pluripotency- inducing protein factor or encoding a plurality of pluripotency-inducing proteins.
- a plurality of vectors can be introduced into a single somatic cell.
- the vector can optionally encode a selectable marker to identify cells that have taken up and express the vector. As an example, when the vector confers antibiotic resistance on the cells, antibiotic can be added to the culture medium to identify successful introduction of the vector into the cells.
- Either integrating vectors or non-integrating vectors may be used. Suitable integrating vectors include retroviral (e.g., lentiviral) vectors.
- Suitable non-integrating vectors include Epstein-Barr virus (EBV) vectors (Ren C, et al, Acta. Biochim. Biophys. Sin. 37:68-73 (2005); and Ren C, et al, Stem Cells 24: 1338-1347 (2006)).
- EBV Epstein-Barr virus
- Adenoviral vectors may also be used to transport the nucleic acid into the somatic cell. Since the adenovirus does not integrated into cellular chromosome(s), the risk of creating tumors is reduced. Stadtfeld et al, Science, 332: 945 - 949 (2008). If desired, non- integrating vectors can be lost from cells by dilution after reprogramming.
- a non-lethal marker such as Green Fluorescent Protein (GFP), Enhanced Green Fluorescent Protein (EGFP) or luciferase
- GFP Green Fluorescent Protein
- EGFP Enhanced Green Fluorescent Protein
- luciferase luciferase
- a selectable marker gene may be used to identify the reprogrammed cells expressing the marker through visible cell selection techniques, such as fluorescent cell sorting techniques.
- the reprogrammed cells can be produced without a selectable marker.
- a marker may be integrated into the genome of the somatic cells downstream of the promoter that regulates Oct4 expression. The endogenous Oct4 promoter is active in undifferentiated, pluripotent stem cells.
- the non-lethal marker can be constructed to enable its subsequent removal using any of a variety of art-recognized techniques, such as removal via Cre -mediated, site- specific gene excision. For example, it may become desirable to delete the marker gene after the pluripotent cell population is obtained, to avoid interference by the marker gene product in the experiment or process to be performed with the cells. Targeted deletions can be accomplished by providing structure(s) near the marker gene that permits its ready excision. For example, a Cre/Lox genetic element can be used. The Lox sites can be built into the cells. If it is desirable to remove the marker from the pluripotent cells, the Cre agent can be added to the cells. Other similar systems also can be used.
- Cre/Lox excision can introduce chromosomal rearrangements and can leave residual genetic material after excision, it may also be desirable to introduce the pluripotency-inducing proteins into the somatic cells using non-integrating, episomal vectors and obtaining cells from which the episomal vectors are lost (e.g., at a rate of about 5% per generation) by subsequently withdrawing the drug selection used to maintain the vectors during the reprogramming step.
- the vectors described herein can be constructed and engineered using art- recognized techniques. Standard techniques for the construction of expression vectors suitable for use are well-known in the art and can be found in publications such as Sambrook J, et al., "Molecular cloning: a laboratory manual,” (3rd ed. Cold Spring Harbor Press, Cold Spring Harbor, N.Y. 2001).
- pluripotency-inducing proteins it is also possible to generate iPS cells by direct delivery of pluripotency- inducing proteins, thus eliminating the need for viruses or genetic modification of the somatic cells. For example, it has been reported that repeated treatment of the cells with certain proteins channeled into the cells via poly-arginine anchors was sufficient to induce pluripotency (Zhou et al, Cell Stem Cell, Volume 4, 381-384 (2009)). The expression of pluripotency-inducing genes can also be increased by treating somatic cells with FGF2 under low oxygen conditions.
- the relative ratio of pluripotency-inducing proteins may be adjusted to increase reprogramming efficiency. For example, it has been reported that linking Oct-4 and Sox2 in a 1 : 1 ratio on a single vector increased reprogramming efficiency in cells by a factor of four, compared to reprogramming efficiency wherein the pluripotency-inducing genes were provided to cells in separate constructs and vectors, where the uptake ratio of the respective pluripotency-inducing genes into single cells was uncontrolled. Although the ratio of pluripotency-inducing proteins may differ depending upon the set of pluripotency-inducing proteins used, one of ordinary skill can readily determine optimal ratios of pluripotency-inducing proteins.
- Pluripotent stem cells can be cultured in any medium used to support growth of pluripotent cells.
- Typical culture medium includes, but is not limited to, a defined medium, such as TeSRTM (StemCell Technologies, Inc.; Vancouver, Canada), mTeSRTM (StemCell Technologies, Inc.) and StemLine® serum-free medium (Sigma; St. Louis, Mo.), as well as a conditioned medium, such as mouse embryonic fibroblast (MEF)-conditioned medium.
- cells can be maintained on MEFs in culture medium. 5. DISEASE MODELING AND DRUG SCREENING
- the invention provides a method of identifying an agent for use in treatment of a disease that is associated with a mutation in the PTPN11 gene, comprising (1) contacting a disease-specific pluripotent stem cell as described herein with the agent; and (2) determining the effect of the agent on survival, proliferation, phenotype, or differentiation of the pluripotent stem cell.
- the invention provides a method of identifying an agent for use in treatment of a disease that is associated with a mutation in the PTPN11 gene, comprising (1) contacting a cardiomyocyte as described herein with the agent; and (2) determining the effect of the agent on survival, proliferation, or phenotype of the cardiomyocyte.
- the invention provides a method of identifying an agent for use in treatment of the cardiac hypertrophy that is associated with a mutation in the PTPN11 gene, comprising: (1) contacting a cardiomyocyte as described herein with the agent; and (2) determining the effect of the agent on the hypertrophic state of the cardiomyocyte, wherein a decrease in the hypertrophic state of the cardiomyocyte, as compared to that of a control cell, is indicative that the agent is an agent for use in treatment of a disease that is associated with a mutation in the PTPN11 gene.
- the disease-specific pluripotent stem cells and cardiomyocytes described herein may be used to screen for test agents that affect survival, proliferation, phenotype, or differentiation of the cells.
- the disease-specific stem cells may be induced to differentiate into cardiomyocytes by placing it under appropriate differentiation conditions. Before, during and/or after differentiation, the cell may be subjected to a test agent in order to determine whether that agent has an effect on survival, proliferation, phenotype, or
- the agents can be used for treatment, preventing or ameliorating the symptoms of the diseases.
- the disease-specific stem cells and cardiomyocytes described herein may also be used to develop personalized treatment regimens. For example, certain patients may respond better to a given therapy or drug regimen than other patients. Additionally or alternatively, certain patients may experience fewer and/or less severe side effects after being administered a given therapy or drug regimen than other patients.
- stem cells that contain the genetic complement of a patient suffering from and/or predicted to suffer from a disease of interest, and permitting such cells to differentiate into a cell type associated with that disease, it will be possible to better predict which therapy or drug regimen will be most beneficial and/or result in the least detrimental side effects.
- a control cell may be a parallel sample cell that has not been treated with the agent (e.g., recipient cells mock-treated with buffers), or a cell that has been treated with an agent having a known effect (e.g., a positive effect, a negative effect, or no effect).
- the agent e.g., recipient cells mock-treated with buffers
- an agent having a known effect e.g., a positive effect, a negative effect, or no effect.
- a control cell may be a cell having known or pre-determined properties (e.g., a characterized cell line from a database or a cell not carrying the PTPNl 1 mutation of the disease- specific cell).
- a disease of interest may be modeled and/or studied by creating a disease-specific pluripotent stem cell line, and inducing the cell line to differentiate under appropriate differentiation conditions.
- a disease-specific pluripotent stem cell line may be generated in which the stem cells carry a mutation in the PTPNl 1 gene that is associated with LEOPARD syndrome, or a cardiac anomaly.
- the cells are particularly useful in studying and/or modeling diseases that have not been amenable to such study and/or modeling.
- the test agent may be any molecule that increases or decreases the expression level or the activity level of PTPNl 1 protein.
- the agent may be a small molecule compound, an antibody, a hormone, a vitamin, a nucleic acid molecule, an enzyme, an amino acid, or a virus.
- the invention also demonstrates for the first time that mutations in the PTPNl 1 gene perturbs RAS-MAPK signal transduction as early as the pluripotent stem cell stage.
- RAS is a family of genes encoding small GTPases that are involved in cellular signal transduction. Activation of Ras signalling causes cell growth, differentiation and survival.
- This pathway includes, but is not limited to, components such as RAFl (V-Raf-1 Murine Leukemia Viral Oncogene Homo log 1), SOS1 (Son of Sevenless Homolog 1 (Drosophila)), ERK2, KRAS (V-KI-RAS2 Kirsten Rat Sarcoma 2 Viral Oncogene Homo log), PTPN11, BRAF (B-Raf proto-oncogene serine/threonine -protein kinase), MEK1 (mitogen-activated protein kinase kinase 1), MEK2, and HRAS. Additional components of this pathway have been identified and described (see, e.g., Lee and McCubrey, Leukemia 16:486- 507, 2002).
- the invention provides a method of identifying an agent as a candidate Ras-signaling pathway inhibitor, comprising: (1) contacting a pluripotent stem cell as described herein with the agent; and (2) determining the effect of the agent on tyrosine phosphorylation of EGFR (Epidermal growth factor receptor) or MEK1. A decrease of tyrosine phosphorylation of EGFR or MEK1, as compared to that of a control cell, is indicative that the agent is a Ras-signaling pathway inhibitor.
- the invention provides a method of identifying an agent as a candidate Ras-signaling pathway inhibitor, comprising: (1) contacting a cardiomyocyte as described herein with the agent; and (2) determining the effect of the agent on tyrosine phosphorylation of EGFR or MEK1. A decrease of tyrosine phosphorylation of EGFR or MEK1, as compared to that of a control cell, is indicative that the agent is a Ras-signaling pathway inhibitor.
- the invention provides a method of identifying an agent for use in treatment of a disease that is associated with a mutation in the PTPN11 gene, comprising: (1) contacting a pluripotent stem cell as described herein with the agent; (2) differentiating the stem cell into a cardiomyocyte; and (3) determining the hypertrophic state of the cardiomyocyte.
- a decrease in the hypertrophic state of the cardiomyocyte, as compared to that of a control cell, is indicative that the agent is an agent for use in treatment of a disease that is associated with a mutation in the PTPN11 gene.
- the pluripotent stem cells and cardiomyocytes described herein may also be used to test or validate the therapeutic activities of known Ras kinase inhibitors or PTPN11 inhibitors.
- the pluripotent stem cells or cardiomyocytes described herein may be cultured in the presence of a Ras inhibitor or PTPN11 inhibitor, and effect of the inhibitor on survival, proliferation, phenotype, or differentiation of the cells may be determined.
- PTPN11 inhibitors such as CDL 4340-0580 (Hellmuth et al, P roc Natl Acad Sci USA 105,7275-7280, 2008) and NAT6-297775 (Noren-Muller et al, Proc Natl Acad Sci USA . , 103 : 10606- 11 2006).
- iPSC reprogrammed induced pluripotent stem cells
- the patient-derived cells have a mutation in the PTPN11 gene, which encodes the SHP2 phosphatase
- the iPSC have been extensively characterized and produce multiple differentiated cell lineages.
- a major disease phenotype in patients with LEOPARD syndrome is hypertrophic
- vzYro-derived cardiomyocytes from LS-iPSC are larger, have a higher degree of sarcomeric organization and preferential localization of NFATc4 in the nucleus when compared to cardiomyocytes derived from human embryonic stem cells (HESC) or wild type (wt) iPSC derived from an unaffected, non-PTPNl lbrother of one of the LS patients.
- HESC human embryonic stem cells
- wt wild type iPSC derived from an unaffected, non-PTPNl lbrother of one of the LS patients.
- DMEM Dulbecco's modified Eagle medium
- HES2 and iPSC were maintained on irradiated Swiss Webster mouse embryonic feeder cells (MEFs), in a serum free HESC medium composed of DMEM/F12 (Cellgro, Mediatech) containing 20 ng/ml basic fibroblast growth factor (bFGF, R&D systems), 20% (vol/vol) KSR (Invitrogen), 5% (vol/vol) MEF-conditioned medium, Penicillin/Streptomycin, L-glutamine (L-Gln), non-essential amino acids (Invitrogen), and ⁇ -mercaptoethanol ( ⁇ - ⁇ , Sigma-Aldrich).
- MEFs irradiated Swiss Webster mouse embryonic feeder cells
- Plasmid construction Full-length sequences of human OCT4, SOX2, KLF4 and c-MYC transcription factors were obtained from Open Biosystems. The coding sequences were PCR amplified using Pfu Turbo (Stratagene) and cloned into pMXs vector and verified by sequencing. pMXs-EGFP vector was constructed by introducing the BamHI/NotI EGFP fragment from FUGW (kindly provided by Dr. Lois, MIT, Massachusetts) into pMXs vector. The latter vector was used to monitor the transfection and infection efficiency.
- Retroviral infection and LEOPARD syndrome iPSC generation OCT4, SOX2, KLF4 and c-MYC transcription factors were introduced in dermal fibroblasts derived from two patients with LEOPARD syndrome via the pMXs retroviral vector.
- pMXs-EGFP vector was used to estimate the infection efficiency (data not shown).
- GP-2 cells were plated at 8xl0 6 cells per 10-cm dish and transfected with pMXs, VSV-G and Gag-Pol vectors using SuperFect transfection reagent on the following day.
- human fibroblasts were seeded at 8xl0 5 cells per 10-cm dish.
- the four retroviruses hOCT4, hSOX2, hKLF-4 and hC- MYC
- Retroviruses containing medium was added to the fibroblasts plates. The following day, forty- eight hours post-transfection, the fibroblasts were reinfected following the same procedure as the day before. Six days after transduction, fibroblasts were transferred into four dishes coated with MEFs, at 50,000 fibroblasts per plate.
- Genomic DNA was isolated with Easy-DNATM kit (Invitrogen). Fifty nanograms of genomic DNA was used per reaction. Primers are summarized in Figure 17 (Table 1).
- HES2 and iPSC were grown on Matrigel-coated glass coverslip dishes (MatTek, Ma). The day of culture harvest, 20 ⁇ of colcemid (5 ⁇ g/ml) was added to the in situ ESC1 culture which was 30-50% confluent. The culture was re-incubated for 15 min at 37°C. A robotic harvester (Tecan) was utilized, which included automatic addition of 2cc of hypotonic solution (sodium citrate solution 0.8%) with incubation for 20 min at room temperature, prefixation with addition of 2cc of fixative (methanol: glacial acetic acid; 3: 1), followed by addition of 4cc of fixative, twice.
- 2cc of hypotonic solution sodium citrate solution 0.86%
- fixative methanol: glacial acetic acid; 3: 1
- the coverslip was dried completely at 37 °C with 45-50%) humidity and mounted on a microscope slide and GTG-banded according to standard protocols. Metaphases were captured and karyotypes were prepared using the Cyto Vision software program (Version 3.92 Build 7, Applied Imaging).
- qPCR and transgenes integration were performed using Trizol® Reagent (Invitrogen) and subsequently column-purified with RNeasy kit (Qiagen) and treated with RNase-free DNase (Qiagen). One microgram of total RNA was reverse transcribed into cDNA using random primers and Superscript II Reverse Transcriptase (Invitrogen). PCR for transgene silencing was performed with Expand High Fidelity Enzyme Tag Polymerase (Roche). Real-time qPCR was performed on a StepOne Plus Real-Time PCR System (Applied Biosystems) with Fast
- the primary antibodies used for intracellular immunostaining were OCT4 (1 : 100, Bio Vision), NANOG (1 : 100, R&D systems), desmin (1 : 100, Lab Vision), a-SMA (pre-diluted, DAKO), vimentin (1 : 100, Chemicon), AFP (1 :500, DAKO), GFAP (1 :1000, DAKO), ⁇ -Tubulin (1 : 100, Chemicon), or NFATc4 (1 : 100, Santa Cruz Biotechnology). All the secondary antibodies Alexa 488 Donkey anti-Rabbit (1 : 100), Alexa 546 donkey anti-goat (1 : 100) and Alexa 546 donkey anti-mouse (1 : 100) were obtained from Invitrogen.
- SSEA-4, troponin T, and hematopoietic markers expression were evaluated on a BD Biosciences LSRII FACS machine analyzer.
- Primary antibodies SSEA-4-PE (R&D systems), cardiac troponin T (Lab Vision), CD1 lb-APC (Caltag), and CD45-APC, CD45- PE, CD71-PE, CD41-PE and CD235a-APC were purchased from BD Biosciences.
- EBs were grown in ultra low-binding plates (Costar) and medium was changed every three days. After eight days of differentiation, EBs were collected, resuspended in DMEM 10% and transferred to gelatin-coated dishes to allow them to attach and differentiate for eight additional days before processing for immunocytochemistry analyses.
- MMG monothioglycerol
- DMEM dimethyl methacrylate
- EBs were collected, resuspended in DMEM 10% and transferred to gelatin-coated dishes to allow them to attach and differentiate for eight additional days before processing for immunocytochemistry analyses.
- For hematopoietic differentiation the inventors used a described protocol (Kennedy, M., et al, Blood 109, 2679-2687 (2007)) with certain modifications (unpublished data, Kennedy M. and Keller G. et al.).
- cardiomyocyte induction the inventors used a well-established protocol (Yang, L., et al, Nature 453, 524-528 (2008)).
- Microarray analysis Gene level mRNA abundance measures were extracted using the Affymetrix GeneChip Exon 1.0 ST array according to the manufacturer's protocols by the Genomics Core Lab in The Institute for Personalized Medicine at Mount Sinai Medical Center. Microarrays were scanned and data were Robust Multi- Array (RMA)-normalized using the Affymetrix Expression Console software. Subsequently, these genes were clustered and a heat map was generated against a background subset of genes showing at least two-fold change between sample averages of iPSC/HES2 cells and fibroblast samples.
- RMA Robust Multi- Array
- Cytology/cardiomyocyte size determination On D18 of differentiation, beating EBs were plated on gelatin coated dishes. Three days after plating, EBs outgrowths were trypsinized, filtered through a 40 ⁇ size pore-size filter, and single cells were replated at low density on gelatin coated dishes. The following day, cells were fixed with 4% paraformaldehyde, permeablized, blocked in PBS/1% BSA/0.1%> Triton/10%) donkey serum, and Stained for cardiac Troponin T (1 :200, Lab Vision), overnight at 4°C.
- HES2 and iPSC were incubated overnight in HESC culture medium deprived of bFGF and knockout serum replacement (KSR). Protein lysates were quantified by Bradford assay and sent to Kinexus Bioinformatics Corporation (Vancouver, Canada) for antibody microarray screening. The proteins with at least 1.5 fold change between the LS-iPSC samples and control sample (either HES2 or wt-iPSC), and conserved in the majority of the comparison groups were represented in a heat map.
- a lysis buffer (pH 7.2) containing 20 mM MOPS pH 7.0, 2 mM EGTA, 5 mM EDTA, 30 mM sodium fluoride, 60 mM ⁇ - glycerophosphate, 20 mM sodium phyrophospate and 1% Triton X-100.
- Protease and phosphatase inhibitors (1 mM phenylmethylsulfonyl fluoride, 3 mM benzamidine, 10 ⁇ g/ml aprotinin, 10 ⁇ leupeptin, 5 ⁇ pepstatin, ImM dithiothreitol and 1 mM sodium
- PTPN11 protein tyrosine phosphatase SHP2.
- PTPN11 is ubiquitously expressed, essential for normal development, and somatic mutations in this gene contribute to leukemogenesis in children (Loh, M.L., et al, Blood 103, 2325-2331 (2004), Tartaglia, M., et al, Blood 104, 307-313 (2004)).
- T468M and Y279C are most recurrent (Tartaglia, M., et al., Am J Hum Genet 78, 279-290 (2006)).
- Hypertrophic cardiomyopathy is the most common life-threatening cardiac anomaly in LS (Sarkozy, A., et al., Orphanet J Rare Dis 3, 13 (2008)). Animal models of LS have been generated in Drosophila and zebrafish (Jopling, C, et al, PLoS Genet 3, e225 (2007); Oishi, K., et al, Hum Mol Genet (2008)), but the molecular pathogenesis of LS remains obscure.
- Ectopic expression of four transcription factors (OCT4, SOX2, KLF4 and c- MYC) in adult human dermal fibroblasts can generate pluripotent iPSC (Lowry, W.E., et al., Proc Natl Acad Sci U S A 105, 2883-2888 (2008), Park, I.H., et al, Cell 134, 877-886 (2008), Takahashi, K., et al., Cell 131, 861-872 (2007)).
- the inventors have established iPSC lines from two LS patients, a 25-year-old female (LI), and a 34-year-old male (L2).
- a heterozygous T468M substitution mutation in PTPN11 is present in both.
- iPSC originated from patient-derived fibroblasts
- the inventors performed DNA fingerprinting analysis (Fig. 6a). All iPSC had normal karyotypes of 46,XX (LI) and 46,XY (L2) (Fig. 6b and data not shown). In addition, they carried the expected T468M mutation (Fig. 7a). Restriction fragment length polymorphism analysis of an RT-PCR amplimer containing the mutation with BsmFI showed biallelic expression of PTPN11 (Fig. 7b). PCR and Southern blots indicated the presence of all four transgene proviruses in the LS-iPSC (Fig. 8) and quantitative RT-PCR (qRT-PCR) results confirmed efficient transgene silencing (Fig. 9).
- pluripotency markers including surface antigens TRA- 1-81, TRA- 1-60, and SSEA-4, as well as the nuclear transcription factors OCT4 and NANOG (Fig. 10).
- Quantitative RT-PCR confirmed the activation of a series of endogenous sternness genes (OCT4, NANOG, SOX2, GDF3, DPPA4, REXl and TERT) in iPSC (Fig. la and Fig. 11a).
- bisulfite sequencing analyses determined that the great majority of the CpG dinucleotides analyzed in the OCT4 and NANOG promoters were demethylated in iPSC when compared to their parental fibroblasts (Fig. lb).
- the inventors next examined genome-wide mR A expression profiles of two LS-iPSC lines from each patient, the BJ-iPSB5 cell line, parental fibroblasts and HES2 cells.
- Pluripotent HESC can differentiate into cell types representative of all three germ layers.
- the inventors tested the differentiation abilities of the iPSC using an in vitro 8-day floating embryoid body (EBs) system, followed by replating on gelatin-coated dishes for another 8 days (Takahashi, K., et al, Cell 131, 861-872 (2007); Dimos, J.T., et al, Science 321, 1218- 1221 (2008)).
- EBs floating embryoid body
- a-smooth muscle actin a- SMA, mesoderm), desmin (mesoderm), a-fetoprotein (AFP, endoderm), vimentin (mesoderm), glial fibrillary acidic protein (GFAP, ectoderm) and ⁇ -tubulin (ectoderm) markers (Fig. 2a and Fig. 12).
- a- SMA smooth muscle actin
- desmin mesoderm
- AFP a-fetoprotein
- vimentin mesoderm
- GFAP glial fibrillary acidic protein
- ectoderm glial fibrillary acidic protein
- ⁇ -tubulin ectoderm
- mesenchyme mesenchyme, adipose tissue and cartilage (mesoderm) (Fig. 2b and data not shown).
- hypertrophic cardiomyopathy is one of the major features of LS, affecting 80% of the patients.
- affected individuals occasionally manifest hematologic complications such as myelodysplasia and leukemia (Laux, D. et al., J Pediatr Hematol Oncol 30, 602-604 (2008); Ucar, C, et al, J Pediatr Hematol Oncol 28, 123-125 (2006)). Therefore, the inventors decided to explore if LS-iPSC were able to differentiate into hematopoietic and cardiac lineages.
- LS-iPSC from both patients differentiated into a variety of hematopoietic cell types including early hematopoietic progenitors (CD41 + ) (Mikkola, H.K., et al, Blood 101, 508-516 (2003)), early erythroblasts (CD71 + /CD235a + ) (Wu, C.J., et al, Blood 106, 3639-3645 (2005)), and macrophages (CD1 lb + ) (Fan, S.T., et al, J Clin Invest 87, 50-57 (1991)) (Fig. 13 and data not shown).
- CD41 + early hematopoietic progenitors
- CD71 + /CD235a + early erythroblasts
- macrophages CD1 lb +
- the cardiac hypertrophic response includes induction of immediate-early genes (such as c-jun, c-fos and c-myc), an increase in cell size, and organization of contractile proteins into sarcomeric units (Aoki, H., et al, Nat Med 6, 183-188 (2000);
- the inventors also observed increased sarcomere assembly in Ll-iPS6 and L2-iPS10 cells when compared to wt S3-iPS4 cells (Fig. 3b). Recently, the calcineurin-NFAT pathway has been shown to be an important regulator of cardiac hypertrophy. Active calcineurin dephosphorylates NFAT transcription factors, resulting in their nuclear translocation (Buitrago, M., et al, Nat Med 11, 837-844 (2005); Molkentin, J.D.,
- the inventors analyzed the localization of NFAT c4 using immunocytochemistry in 50 cTNT -positive cardiomyocytes derived from the L2-iPS10 cell line, which produced the largest cardiomyocytes, and wt S3-iPS4. The inventors observed a significantly higher proportion of LS cardiomyocytes with nuclear NFATc4, (-80% versus -30%, respectively) (Fig. 3c and 3d).
- Some of the proteins were more abundantly present in LS-iPS when compared to either HES2 (Tyro 10, Tyk2 and Haspin) or wt- iPSC (p-MARCKs, p-Synapsinl, p-NMDAR2B, p-MSKl, p-RSKl/3 and p-p53).
- HES2 Tyro 10, Tyk2 and Haspin
- wt- iPSC p-MARCKs, p-Synapsinl, p-NMDAR2B, p-MSKl, p-RSKl/3 and p-p53.
- the phosphorylation of other proteins was increased (p-Caveolin2, p-MEKl, p-EGFR and p-FAK) or decreased (p-Vinculin, p-S6 and p-Lck) in LS-iPSC when compared to control cell lines.
- WB Western blot
- RAS-MAPK represents the major signaling pathway deregulated by SHP2 mutants.
- NS mutants increase basal and stimulated phosphatase activity, whereas LS mutants are catalytically impaired and have dominant-negative effects, inhibiting growth factor-evoked ERK1/2 activation (Kontaridis, M.I., J Biol Chem 281, 6785-6792 (2006)).
- the inventors analyzed the ability of LS-iPSC to respond to external growth factors.
- the inventors used bFGF (basic Fibroblast Growth Factor), the main growth factor in the maintenance of HESC, to induce the stimulation of the MAPK signaling pathway.
- bFGF basic Fibroblast Growth Factor
- bFGF treatment increased the phosphorylation of ERK1/2 (p-ERK) levels over time in HES2 and wt S3-iPS4 cells (Fig. 4c).
- p-ERK ERK1/2
- FGFR FGF receptor family members
- bFGF stimulation did not cause any substantial change in p-ERK levels (Fig. 4c and Figs. 16b- 16c).
- the LS- iPSC lines had higher basal p-ERK levels compared to HES2 and S3-iPS4 cells (Figs. 16b- 16c), in accordance with the increased pMEKl, ERK upstream kinase, levels found in LS-iPSC samples in phosphoproteomic array results.
- the inventors have generated and characterized LS patient- specific iPSC, providing a new system for the study of disease pathogenesis.
- the inventors observed increases in cell size, sarcomeric organization and nuclear NFATc4 localization in LS- iPSC-derived cardiomyocytes, when compared to HESC and wt-iPSC-derived cardiomyocytes. These results are consistent with cardiac hypertrophy, a condition commonly found in LS patients (Sarkozy, A. et al., Orphanet J Rare Dis 3, 13 (2008)), and indicate that this abnormality occurs through a cell autonomous mechanism due to the PTPN11 mutation. Since many human cell types, such as cardiomyocytes, cannot be propagated readily in cell culture, iPS-derived cells exhibiting disease-relevant phenotypes provide the requisite resource for precisely elucidating pathogenesis and pursuing novel therapeutic strategies.
- the inventors provided insights into the molecular events that are affected by the PTPN11 mutation in the pluripotent iPSC using antibody microarrays.
- the inventors found that the phosphorylation of certain proteins was increased in LS-iPSC when compared to wild-type HESC and iPSC.
- one of the more upregulated phosphoproteins was MEK1, the upstream kinase of ERK1/2, whose gene is sometimes mutated in the related disorder, cardiofaciocutaneous syndrome.
- PTPN11 mutations underlie 45% and 90% of NS and LS, respectively.
- CD 1 lb/CD 18 to functional enhancement of effector macrophage tissue factor response. J Clin Invest 87, 50-57 (1991).
Landscapes
- Health & Medical Sciences (AREA)
- Engineering & Computer Science (AREA)
- Life Sciences & Earth Sciences (AREA)
- Biomedical Technology (AREA)
- Wood Science & Technology (AREA)
- Organic Chemistry (AREA)
- Chemical & Material Sciences (AREA)
- Biotechnology (AREA)
- Zoology (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Genetics & Genomics (AREA)
- Microbiology (AREA)
- Cell Biology (AREA)
- Biochemistry (AREA)
- General Engineering & Computer Science (AREA)
- General Health & Medical Sciences (AREA)
- Rheumatology (AREA)
- Cardiology (AREA)
- Developmental Biology & Embryology (AREA)
- Transplantation (AREA)
- Micro-Organisms Or Cultivation Processes Thereof (AREA)
- Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
Abstract
This invention generally relates to disease-specific pluripotent stem cells that are derived from somatic cells that carry a mutation in the PTPNl 1 gene. The stem cells are capable of differentiating into cells or tissues affected by the PTPNl 1 mutation. The stem cells and differentiated cells can be used to study various diseases that are associated with mutations in the PTPNl 1 gene, and to identify novel therapeutic agents for diagnosis and treatment of diseases that are associated with mutations in PTPNl 1.
Description
INDUCED PLURIPOTENT STEM CELLS AND USES THEREOF
CROSS-REFERENCE TO RELATED APPLICATIONS
[0001] The present application relates to U.S. Provisional Application Serial
No. 61/324,150, filed April 14, 2010, which is incorporated by reference in its entirety.
GOVERNMENT SUPPORT
[0002] The subject matter of this application has been supported by a research grant from the National Institutes of Health under grant number 5R01GM078465 and a grant from NYSTEM. The United States government has a paid-up license in this invention and the right in limited circumstances to require the patent owner to license others on reasonable terms as provided for by the terms of this grant.
BACKGROUND OF THE INVENTION
[0003] Pluripotent stem cells can grow indefinitely while maintaining pluripotency, and can differentiate into cells of all three germ layers (Evans & Kaufman, Nature 292: 154-156 (1981)). Human pluripotent stem cells have promise for treating diseases such as Parkinson's disease, spinal cord injury and diabetes (Thomson et al, Science 282:1145-1147 (1998)).
[0004] Somatic cells can be reprogrammed into pluripotent stem cells by transferring their nuclear contents into oocytes (Wilmut et al, Nature 385:810-813(1997)) or by fusion with embryonic stem (ES) cells (Cowan et al, Science 309: 1369-1373 (2005)), indicating that unfertilized eggs and ES cells contain factors that confer totipotency or pluripotency in somatic cells. For example, Yu et al. showed that cells derived by in vitro differentiation from an Oct4 knock-in human ES cell line did not express EGFP, but that EGFP expression was restored upon cell-cell fusion with human ES cells (Yu et al, Stem Cells 24: 168-176 (2006)).
[0005] Induced pluripotent stem cells, commonly abbreviated as "iPS cells" or "iPSCs," are pluripotent stem cell derived from non-pluripotent cells (e.g., adult somatic cells). iPS cells are typically produced by introducing into somatic cells certain transcription factors that are involved in maintaining ES cell pluripotency. Although the transcriptional
determination of pluripotency is not fully understood, several transcription factors, including Oct
3/4 (Nichols et al, Cell 95:379-391(1998)), Sox2 (Avilion et al, Genes Dev. 17: 126-140 (2003)) and Nanog (Chambers et al., Cell 113:643-655(2003)) were found to be involved in maintaining ES cell pluripotency.
[0006] Takahashi & Yamanaka introduced four reprogramming factors (Oct3/4, Sox2, c-Myc and Klf ) into mouse adult fibroblasts to obtain iPSCs. These iPSCs exhibited mouse ES cell morphology and growth properties, and expressed mouse ES cell marker genes (Takahashi & Yamanaka, Cell 126:663-676 (2006)). Notably, exogenous Oct-4 introduced into the mouse fibroblasts resulted in only marginal Oct-4 expression. Subcutaneous transplantation of iPS cells into nude mice resulted in tumors containing a variety of tissues from all three germ layers. Following injection into blastocysts, iPS cells contributed to mouse embryonic
development. These data demonstrate that pluripotent cells can be directly generated from mouse fibroblast cultures by adding only a few defined factors using retroviral transduction. One year later, using the same principle developed in mouse models, Takahashi et al.
successfully transformed human fibroblasts into pluripotent stem cells using the same four genes: Oct3/4, Sox2, Klf4, and c-Myc with a retroviral system (Cell, 131 :861-72, 2007).
[0007] Yu et al. (Science, 318:1917-20) and WO2008/0233610 (Thomson and Yu) disclose the reprogramming of human fibroblast cells into pluripotent stem cells using a lentiviral system and a different set of factors: OCT4, SOX2, NANOG, and LIN28.
WO2008/0233610 notes that ES cells from mice and humans require distinct sets of factors to remain undifferentiated.
[0008] LEOPARD syndrome (LS; also known as "Cardiocutaneous syndrome," "Gorlin syndrome II," "Lentiginosis profusa syndrome," "Progressive cardiomyopathic lentiginosis," "Capute-Rimoin-Konigsmark-Esterly-Richardson syndrome," or "Moynahan syndrome") is a rare autosomal dominant, multisystem disease having a complex of features, mostly involving the skin, skeletal and cardiovascular systems. The name of the condition is a mnemonic, as the condition is characterized by the following seven conditions, the first letters of which spell LEOPARD: Lentigines, Electrocardiographic abnormalities, Ocular hypertelorism, Pulmonary valve stenosis, Abnormal genitalia, Retardation of growth and Deafness. These conditions may not be present in all patients.
[0009] Noonan syndrome (NS) is an autosomal dominant disorder characterized by dysmorphic facial features, proportionate short stature, and heart disease, i.e., pulmonic stenosis and hypertrophic cardiomyopathy most commonly (Noonan, Am. J. Dis. Child. 1968, 116:373- 380; Allanson, J. Med. Genet. 1987, 24:9-13). Webbed neck, chest deformity, cryptorchidism, mental retardation, and bleeding diatheses constitute other frequently associated findings. NS is a relatively common syndrome with an estimated incidence of 1 : 1000 to 1 :2500 live births.
[0010] Approximately 90% of LS cases, and 45% of NS, are caused by missense mutations in the protein tyrosine phosphatase, non-receptor type 11 (PTPNl 1) gene, which encodes a protein tyrosine phosphatase. PTPNl 1 is ubiquitously expressed and essential for normal development. Further, wild-type PTPNl 1 protein (also known as SHP2, Syp, SHPTP2, PTP2C, PTP1D or BPTP3) mediates cell signaling of many protein tyrosine kinase oncogenes, such as ErbB and Met. PTPNl 1 protein is necessary for embryonic development and for growth factor, cytokine, and extra-cellular matrix signaling, and is involved in the regulation of cell proliferation, differentiation, and migration.
[0011] Somatic mutations in the PTPNl 1 gene also contribute to leukemogenesis in children (e.g., juvenile myelomonocytic leukemia (JMML), or acute myelogenous leukemia). JMML is a progressive myelodysplastic/myeloproliferative disorder characterized by
overproduction of tissue-infiltrating myeloid cells. Somatic mutations in PTPNl 1 account for about 35% of JMML patients that do not have Ras or neurofibromin mutations (Tartaglia et al., Nature Genetics, Jun;34(2), 2003: 148-50; Kratz et al, Blood 106(6):2183-2185, 2005).
[0012] Tyrosine phosphorylation is involved in the regulation of human cellular processes from cell differentiation and growth to apoptosis. The process of tyrosine
phosphorylation is regulated by protein-tyrosine phosphatases (PTP) and protein-tyrosine kinases (PTK). When this regulation is disrupted, diseases such as cancer can arise. For example, activated PTPNl 1 protein interacts with the Gab family of docking proteins. This interaction activates a pathway leading to cell proliferation and tumorigenesis. Therefore, the PTPNl 1 protein signaling pathway can be an attractive therapeutic target for treating, for example, cancer, Noonan syndrome, and LEOPARD syndrome.
[0013] A number of therapeutic or surgical treatments are available to alleviate various symptoms of Noonan syndrome (e.g., heart defects, undescended testicles, or excessively short stature) or LEOPARD syndrome (e.g., cryptorchidism, hypospadias, or severe skeletal deformity). However, no therapeutic treatment of the underlying disorder has been determined so far.
[0014] Therefore, there is a need to identify novel therapeutic agents for diagnosis and treatment of diseases that are associated with mutations in the PTPNl 1 gene and/or abnormal PTPNl 1 signaling. There is also a need to provide novel tools that would facilitate the identification of such therapeutic agents.
SUMMARY OF THE INVENTION
[0015] This invention generally relates to disease-specific pluripotent stem cells that are derived from somatic cells that carry a mutation in the PTPNl 1 gene. The stem cells are capable of differentiating into cells or tissues affected by the PTPNl 1 mutation, thereby providing a powerful tool to study various diseases that are associated with mutations in the PTPNl 1 gene, and to identify novel therapeutic agents for diagnosis and treatment of diseases that are associated with mutations in the PTPNl 1 gene.
[0016] In one aspect, the invention provides an isolated primate pluripotent stem cell derived from a somatic cell, wherein the stem cell comprises a mutation in the protein tyrosine phosphatase non-receptor type 11 (PTPNl 1) gene.
[0017] In certain embodiments, the PTPNl 1 gene comprises a mutation associated with a cardiac anomaly. In certain embodiments, the cardiac anomaly is hypertrophic cardiomyopathy.
[0018] In certain embodiments, the PTPNl 1 gene comprises a mutation associated with LEOPARD Syndrome, Noonan Syndrome or leukemia.
[0019] In certain embodiments, the mutation in the PTPNl 1 gene is a deletion mutation, a substitution mutation, an addition mutation, or a combination thereof, in the coding
region of the PTPNl 1 gene. In certain embodiments, the mutation in the PTPNl 1 gene results in a mutation in the encoded PTPNl 1 protein.
[0020] In certain embodiments, the mutation in the PTPNl 1 gene results in a threonine to methionine substitution in the encoded PTPNl 1 protein at an amino acid residue position corresponding to position 468 of SEQ ID NO: l, or a tyrosine to cysteine substitution in the encoded PTPNl 1 protein at an amino acid residue position corresponding to position 279 of SEQ ID NO: l .
[0021] In certain embodiments, the mutation in the PTPNl 1 gene results in a tyrosine to cysteine substitution in the encoded PTPNl 1 protein at an amino acid residue position corresponding to position 279 of SEQ ID NO: 1, a tyrosine to serine substitution in the encoded PTPNl 1 protein at an amino acid residue position corresponding to position 279 of SEQ ID NO: 1, an alanine to threonine substitution in the encoded PTPNl 1 protein at an amino acid residue position corresponding to position 461 of SEQ ID NO: 1, a glycine to alanine substitution in the encoded PTPNl 1 protein at an amino acid residue position corresponding to position 464 of SEQ ID NO: 1, a threonine to methionine substitution in the encoded PTPNl 1 protein at an amino acid residue position corresponding to position 468 of SEQ ID NO: 1, a threonine to proline substitution in the encoded PTPNl 1 protein at an amino acid residue position corresponding to position 468 of SEQ ID NO: 1, an arginine to tryptophan substitution in the encoded PTPNl 1 protein at an amino acid residue position corresponding to position 498 of SEQ ID NO: 1, an arginine to leucine substitution in the encoded PTPNl 1 protein at an amino acid residue position corresponding to position 498 of SEQ ID NO: 1, a glutamine to proline substitution in the encoded PTPNl 1 protein at an amino acid residue position corresponding to position 506 of SEQ ID NO: 1, a glutamine to proline substitution in the encoded PTPNl 1 protein at an amino acid residue position corresponding to position 510 of SEQ ID NO: l, a glutamine to glutamic acid substitution in the encoded PTPNl 1 protein at an amino acid residue position corresponding to position 510 of SEQ ID NO : 1 , or a glutamine to glycine substitution in the encoded PTPN 11 protein at an amino acid residue position corresponding to position 510 of SEQ ID NO: 1
[0022] In certain embodiments, the PTPNl 1 gene encodes a polypeptide comprising SEQ ID NO: 1, and the mutation in the PTPNl 1 gene results in a threonine to methionine
substitution at amino acid residue position 468 of SEQ ID NO: 1 , or a tyrosine to cysteine substitution at amino acid residue position 279 of SEQ ID NO: 1.
[0023] In certain embodiments, the pluripotent stem cell forms a teratoma in an immunocompromised mouse.
[0024] In certain embodiments, the pluripotent stem cell expresses SSEA-3, SSEA-4, TRA-1-60, TRA-1-81, Nanog, OCT-4, Sox2, GDF3, DPPA4, REX1, TERT, Alkaline
Phosphatase, Telomerase, or CD30, or a combination thereof.
[0025] In certain embodiments, the pluripotent stem cell is capable of differentiating into a cardiomyocyte.
[0026] Also provided is a cell culture of the stem cells described herein. Exemplary stem cell lines were deposited on , 2010 with American Type Culture Collection
(ATCC), P.O. Box 1549, Manassas, VA 20108, USA, under ATCC Accession Nos. , and .
[0027] In another aspect, the invention provides a cardiomyocyte comprising a mutation in the PTPNl 1 gene, wherein the cardiomyocyte is produced by differentiating a disease-specific pluripotent stem cell as described herein.
[0028] In certain embodiments, the invention provides a cell culture of a
cardiomyocyte as described herein. Exemplary cardiomyocyte cell lines were deposited on
, 2010 with American Type Culture Collection (ATCC), P.O. Box 1549, Manassas, VA
20108, USA, under ATCC Accession Nos. , and .
[0029] In certain embodiments, the primate is human.
[0030] In certain embodiments, the somatic cell is a fibroblast.
[0031] In another aspect, the invention provides a method of identifying an agent for use in treatment of a disease that is associated with a mutation in the PTPNl 1 gene, comprising (1) contacting a disease-specific pluripotent stem cell as described herein with the agent; and (2)
determining the effect of the agent on survival, proliferation, phenotype, or differentiation of the pluripotent stem cell.
[0032] In another aspect, the invention provides a method of identifying an agent for use in treatment of a disease that is associated with a mutation in the PTPNl 1 gene, comprising (1) contacting a disease-specific cardiomyocyte as described herein with the agent; and (2) determining the effect of the agent on survival, proliferation, or phenotype of the cardiomyocyte.
[0033] In another aspect, the invention provides a method of identifying an agent for use in treatment of the cardiac hypertrophy that is associated with a mutation in the PTPNl 1 gene, comprising: (1) contacting a cardiomyocyte as described herein with the agent; and (2) determining the effect of the agent on the hypertrophic state of the cardiomyocyte. A decrease in the hypertrophic state of the cardiomyocyte, as compared to that of a control cell, is indicative that the agent is an agent for use in treatment of a disease that is associated with a mutation in the PTPNl 1 gene.
[0034] In another aspect, the invention provides a method of producing a pluripotent stem cell as described herein, comprising: (a) obtaining a somatic cell from a donor whose genome comprises a mutation in the PTPNl 1 gene; (b) introducing into the somatic cell: (i) a nucleic acid molecule that encodes a pluripotency-inducing protein; or (ii) a polypeptide that comprises a pluripotency-inducing protein; wherein said pluripotency-inducing protein is Oct3/4, Sox2, Klf4, Nanog, Lin28, c-Myc, or a combination thereof.
[0035] In certain embodiments, one or more nucleic acid molecules that encode Oct3/4, Sox2, Klf4, and c-Myc are introduced into the somatic cell.
[0036] In certain embodiments, the somatic cell is a fibroblast.
[0037] In another aspect, the invention provides a method of differentiating a pluripotent stem cell as described herein into a cardiomyocyte, comprising: culturing a pluripotent stem cell as described herein in a serum- free medium that comprises: activin A, bone morphogenetic protein 4 (BMP4), basic fibroblast growth factor (bFGF), vascular endothelial growth factor (VEGF) and dickkopf homo log 1 (DK 1).
[0038] In another aspect, the invention provides a method of identifying an agent as a candidate Ras-signaling pathway inhibitor, comprising: (1) contacting a pluripotent stem cell as described herein with the agent; and (2) determining the effect of the agent on tyrosine phosphorylation of EGFR or MEK1. A decrease of tyrosine phosphorylation of EGFR or MEK1, as compared to that of a control cell, is indicative that the agent is a Ras-signaling pathway inhibitor.
[0039] In another aspect, the invention provides a method of identifying an agent as a candidate Ras-signaling pathway inhibitor, comprising: (1) contacting a cardiomyocyte as described herein with the agent; and (2) determining the effect of the agent on tyrosine phosphorylation of EGFR or MEK1. A decrease of tyrosine phosphorylation of EGFR or MEK1, as compared to that of a control cell, is indicative that the agent is a Ras-signaling pathway inhibitor.
[0040] In another aspect, the invention provides a method of identifying an agent for use in treatment of a disease that is associated with a mutation in the PTPNl 1 gene, comprising: (1) contacting a pluripotent stem cell as described herein with the agent; (2) differentiating the stem cell into a cardiomyocyte; and (3) determining the hypertrophic state of the cardiomyocyte. A decrease in the hypertrophic state of the cardiomyocyte, as compared to that of a control cell, is indicative that the agent is an agent for use in treatment of a disease that is associated with a mutation in the PTPNl 1 gene.
[0041] In another aspect of the invention, the stem cells of the invention are capable of differentiating into a hematopoietic cell. Such hematopoietic cells may comprise a mutation in the PTPNl 1 gene, wherein the hematopoietic cell is produced by differentiating the stem cells of the invention.
[0042] Cell cultures of these hematopoietic cells are also provided. Exemplary hematopoietic cell lines were deposited on , 2010 with American Type Culture
Collection (ATCC), P.O. Box 1549, Manassas, VA 20108, USA, under ATCC Accession Nos.
, and .
[0043] In another aspect, methods of identifying an agent for use in treatment of a disease that is associated with a mutation in the PTPN11 gene are provided, comprising (1) contacting the pluripotent stem cells with the agent; (2) differentiating the stem cell into a hematopoietic cell; and (3) determining the effect of the agent on survival, proliferation, phenotype, or differentiation of the hematopoietic cell. In one embodiment, the disease is leukemia.
[0044] In yet another aspect, methods of identifying an agent for use in treatment of leukemia are provided, comprising contacting the pluripotent stem cell with the agent; and differentiating the stem cell into a hematopoietic cell; and determining the proliferative state of the hematopoietic cell, wherein a decrease in the proliferative state of the hematopoietic cell, as compared to that of a control cell, is indicative that the agent is an agent for use in treatment of leukemia.
BRIEF DESCRIPTION OF THE DRAWINGS
[0045] Figs, la-lc demonstrate that the gene expression profile of LS-iPSC is similar to that of HESC. la, quantitative real-time PCR assay for the expression of endogenous OCT4, NANOG and SOX2 in iPSC and parental fibroblasts (Fib). PCR reactions were normalized against β-ACTIN and plotted relative to expression levels in HES2. Error bars indicate ± s.d. of triplicates, lb, bisulfite sequencing analyses of the OCT4 and NANOG promoters. The cell line and the percentage of methylation is indicated to the left of each cluster, lc, heat map showing hierarchical clustering of 3657 genes with at least two-fold expression change between the average of the three fibroblast cell lines versus all the iPSC lines/HES samples. Expression levels are represented by color; red indicates lower and yellow higher expression.
[0046] Figs. 2a-2b demonstrate that LS-iPSC can differentiate in vitro and in vivo into all three germ layers. 2a, L2-iPS6 cells were differentiated as floating EBs for eight days and then plated onto gelatin-coated dishes and allowed to differentiate for another eight days. Immunocytochemistry showed cell types positively stained for differentiation markers including Desmin/SMA (mesoderm), AFP (endoderm), vimentin (mesoderm), and GFAP/piII-Tubulin (ectoderm). The arrow indicates a βΙΙΙ-tubulin-positive cell. Scale bar, 100 μιη. 2b, HES2, Ll- iPSC and L2-iPSC were injected subcutaneously into the right hindleg of immuno-compromised
NOD-SCID mice. The resulting teratomas were stained with hematoxylin and eosin and tissues representative of all three germ layers were observed.
[0047] Figs. 3a-3d show that cardiomyocytes derived from LS-iPSC had hypertrophic features. 3a, HES2, HI, wt S3-iPS4 and three LS-iPS clones were differentiated into cardiac lineage. Cell areas of 50 random cTNT-positive cardiomyocytes of each cell line were measured using Image J. Boxes show the span from the median (50th percentile) to the first and third quartiles. The lines represent the largest/smallest sizes that are no more than 1.5 times the median to quartile distance. Additional points drawn represent extreme values. 3b, Sarcomeric organization was assessed in 50 cTNT positive (red) cardiomyocytes. Data are presented as mean ± s.d. n = 3; ** < 0.01 (Student's t-test). 3c, S3-iPS4 and L2-iPS10 cells-derived cardiomyocytes were restained with NFATc4 antibody, and the nuclear versus cytosolic expression was analyzed, n = 3; **P < 0.01 (Student's t-test). 3d, Nuclear localization of NFATc protein in a cTNT -positive cell from L2-iPS10 is shown.
[0048] Figs 4a-4c show the results of phosphoproteomic and MAPK activation analyses. 4a, Protein extracts of two iPSC from each LS patient (LI and L2), wt iPSC (BJ- iPSB5) and HES2 were hybridized to an antibody microarray. The heat map represents the most significant protein changes preserved in all the comparison groups. 4b, pMEKl and pEGFR expression was confirmed by Western blot using phospho-specific antibodies. Band density was measured (ImageJ software), and normalized to β-Actin. 4c, HES2, wt S3-iPS4, and LS-iPSC were serum- and bFGF-starved for 6 hours and then treated with bFGF (20 ng/ml) for the indicated time. Phosphorylated ERK1/2 (p-ER l/2) and total ER were assessed by
immunoblotting and quantitated. p-ERKl/2 levels were compared to the untreated p-ERKl/2 level in each sample, normalized to the total ERK1/2 and represented graphically at the right of each panel.
[0049] Figs 5a-5b illustrate the formation of LS-iPSC. 5a, schematic representation of iPSC generation. 5b, typical image of a TRA-1-81 positive colony growing in the plate three weeks after infection.
[0050] Figs. 6a-6b demonstrate that LS-iPSC lines were derived from their parental fibroblasts and maintained normal karyotypes. 6a, the inventors PCR-amp lifted across three
discrete genomic loci containing highly variable numbers of tandem repeats with different primer sets (D10S1214, D17S1290, and D21S2055). The resulting amplification patterns confirmed that each iPSC line was derived from its indicated parental fibroblast. 6b, G-banding of HES2 cell line, wild-type iPS clone BJ-iPSB5, and LS-iPSC lines (one clone of each patient) demonstrates normal diploid chromosomal contents.
[0051] Figs. 7a-7b show the result οΐΡΤΡΝΙΙ T468M mutation analysis in LS-iPSC and fibroblasts. 7a, the T468M point mutation in exon 12 of one allele of PTPN11 gene was verified by DNA sequencing. 7b, the inventors amplified by RT-PCR a 1.2 Kb region containing the mutation sequence and the DNA was digested with BsmFI, an enzyme that cuts the 5'-GGGAC(N)io-3' sequence of the wild type allele but does not cut the mutant allele. In all the LS samples, an undigested 1.2 Kb band was observed.
[0052] Figs. 8a-8b show the integration of retroviral transgenes in iPSC. 8a, transgene-specific primers were used to amplify OCT4, SOX2, KLF4 and c-MYC. Three LS- derived iPSC lines from each patient were analyzed, and HES2 cells and parental fibroblasts were used as negative controls. 8b, Southern blot analyses of Bgl-II digested gDNA extracted from HES2 cells, parental fibroblasts and iPSC, using DIG-labeled DNA probes against OCT4, SOX2, KLF4 and c-MYC. Retrovirally-inserted transgenic copies of these genes are indicated by asterisks and the number of detected bands is shown at the bottom. The parental fibroblasts and HES2 cells share bands in common with all the iPSC lines (arrowheads), which reflect the endogenous loci (including potential pseudogenes).
[0053] Figs. 9a-9b demonstrate the silencing of retroviral transgenes in iPSC lines. 9a, transgene-specific primers were used to determine OCT4, SOX2, KLF4 and c-MYC expression. 9b, qPCR analysis of retroviral transgene (Tg) expression. Results are normalized against β-ACTIN and plotted relative to the expression levels in transfected GP2 cells.
[0054] Figs. 10a- 10b demonstrate that LS-iPSC expressed cell markers that are common to pluripotent cells. 10a, SSEA4 expression was determined by flow cytometry in two iPSC lines derived from two LEOPARD syndrome (LS) patients (Ll-iPSl, Ll-iPS13, L2-iPS6 and L2-iPS16), and a wt-iPSC line derived from BJ fibroblasts (BJ-iPSB5). HES2 cell line was used as positive control. 10b, HES2 and iPSC were grown on MEFs, and were fixed and stained
for the following pluripotency markers: alkaline phosphatase (AP), TRA-1-81, TRA-1-60, NANOG and OCT4. Nuclei were stained with DAPI.
[0055] Figs. 1 la-1 lb show the result of gene expression analysis of the cells. 11a, qPCR was used to evaluate the expression of GDF3,
REXl and TERT in two LS- iPSC lines from each patient and compared to their parental fibroblasts. HES2 cells were used as positive control of pluripotency gene expression. PCR reactions were normalized against β- ACTIN and represented relative to the expression in HES2 cells, lib, two sample comparison plots of log 2 expression values, with the line Y=X as a basis for comparison along with Y = X +/- 1 or 2 lines, representing 2-fold and 4-fold expression changes, respectively. In each scatter- plot the position of SOX2, LIN28 and OCT4 transcription factors is indicated.
[0056] Figs. 12a-12b illustrate in vitro differentiation of iPSC lines. 12a, day 8 EBs of HES2, BJ-iPSB5, Ll-iPS13 and L2-iPS6 cells. 12b, after 16 days of differentiation, cells were stained with SMA, GFAP and Vimentin antibodies. Scale bar, 100 μιη.
[0057] Figs. 13a- 13b show that LS-iPSC differentiated into hematopoietic cells. 13a, red EBs, an indication of erythrocyte development, were observed in Ll-iPSC plates 14 days after differentiation. Scale bar, 100 μπι. 13b, CD41, CD45, CD1 lb, CD71 and CD235a hematopoietic markers were analyzed by flow cytometry in EBs derived from HES2 and Ll- iPSC, 18 days after induction of hematopoietic differentiation.
[0058] Figs. 14a- 14c show the characterization of wt S3-iPS4 and L2-iPSC. 14a, the iPSC had the four transgenes integrated in their genome. 14b, Tg silencing analysis. 14c,
Fingerprinting analysis of the iPSC lines using their parental fibroblasts and HES2 cells as controls.
[0059] Figs. 15a-15c show the pluripotency of S3-iPS4 and L2-iPSC. 15a and 15b, S3-iPS4, L2-iPS10 and L2-iPS20 expressed pluripotency markers. 15c, the iPSC differentiated into derivatives of the three germ layers.
[0060] Figs. 16a- 16c show the disruption of MAPK activation upon bFGF
stimulation in LS-iPSC. 16a, FGF receptors expression analysis by RT-qPCR in LS-iPSC and fibroblasts compared to HES2. 16b, Ll-iPSl, Ll-iPS6 and HES2 cells were serum- and bFGF-
starved overnight. The following day, the cells were treated with bFGF for 10 minutes (+) or not (-). Total ER 1/2 and p-ER l/2 expression was analyzed by immunobloting of total lysates. 16c, basal p-ERKl/2 was quantified in each sample and compared to HES2C (top left panel). The relative increase of p-ERKl/2 level upon stimulation was quantified in each sample (top right and lower panels). All the p-ERKl/2 values were normalized to their corresponding total ERK1/2 values.
[0061] Fig. 17 is a table showing the primer sets that were used for PCRs.
[0062] Fig. 18 is a schematic drawing showing PTPN11 gene organization and domain structure. The numbered, filled boxes at the top indicate the coding exons; the positions of the ATG and TGA codons are shown. The functional domains of the PTPN11 protein, consisting of two tandemly arranged src-homology 2(SH2) domains at the N-terminus (N-SH2 and C-SH2) followed by a protein tyrosine phosphatase (PTP) domain, are shown below. The numbers below that cartoon indicate the amino acid boundaries of those domains.
[0063] Fig. 19 shows the distribution of PTPN11 (SHP-2) mutations and their relative prevalence in Noonan syndrome.
[0064] Fig. 20 shows the cDNA sequence of human PTPN11 (SEQ ID NO: 2).
[0065] Fig. 21 shows the amino acid sequence of human PTPN11 (SEQ ID NO: 1). DETAILED DESCRIPTION OF THE INVENTION 1. OVERVIEW
[0066] This invention generally relates to disease-specific pluripotent stem cells that are derived from somatic cells that carry a mutation in the PTPN11 gene. The stem cells are capable of differentiating into cells or tissues (e.g., cardiomyocytes) affected by the PTPN11 mutation, thereby recapitulating both normal and pathologic tissue formation in vitro. The stem cells and cardiomyocytes provided herein can facilitate disease investigation and drug development - they may be used to study various diseases that are associated with mutations in the PTPN11 gene, and to identify novel therapeutic agents for diagnosis and treatment of diseases that are associated with mutations in PTPN11.
[0067] The present invention is based on the discovery that somatic cells from
LEOPARD syndrome (LS) patients, which carry a mutation in the PTPN11 gene, can be reprogrammed into pluripotent stem cells. Further, the stem cells may be cultured under appropriate differentiation conditions to generate differentiated, disease-specific cardiomyocytes. Such disease-specific stem cells and cardiomyocytes are useful for studying the genetic basis of disease progression and pathogenesis, e.g., by comparing the survival, proliferation, phenotype, or differentiation of the disease-specific cells to that of cells that do not carry the mutation in the PTPN11 gene.
[0068] For example, as described and exemplified herein, this approach is used to develop an in vitro model of LEOPARD syndrome and cardiac hypertrophy. The inventors generated LS-specific induced pluripotent stem cell (LS-iPSC) lines from fibroblasts of LS patients. The pluripotency of the induced LS-specific stem cells were confirmed by the expression of certain stem cell markers, chromatin methylation patterns, genome-wide mRNA expression profiles, teratoma formation, and differentiability. Then the LS-specific stem cells were differentiated into cardiomyocytes in vitro. Consistent with the observation that a major disease phenotype in patients with LEOPARD syndrome is hypertrophic cardiomyopathy, in vz'tro-derived cardiomyocytes from LS-iPSC were larger, had a higher degree of sarcomeric organization and showed preferential localization of NFATc4 in the nucleus, when compared to cardiomyocytes derived from human embryonic stem cells (HESC) or iPSCs derived from an unaffected brother (i.e., not having the PTPN11 mutation) of one of the LS patients.
[0069] The disease-specific iPSCs and cardiomyocytes provided herein offer several advantages for disease study and drug screening as compared to existing cell lines. For example, most of the human cell lines in wide use today are derived either from malignant tissues or are genetically modified to drive immortal growth. On the other hand, primary human cells have a limited life span in culture, a constraint that thwarts inquiry into the regulation of tissue formation, regeneration, and repair. Therefore, the immortal pluripotent stem cells and the in vitro differentiated cardiomyocytes provided herein are particularly valuable as disease models for studying mutations in the PTPN11 gene.
[0070] Also provided herein are methods of identifying an agent for use in treatment of a disease that is associated with a mutation in the PTPN11 gene; methods of identifying an agent for use in treatment of the cardiac hypertrophy that is associated with a mutation in the PTPN11 gene; methods of producing an LS-specific pluripotent stem cell; methods of differentiating an LS-specific pluripotent stem cell into a cardiomyocyte; and methods of identifying an agent as a candidate Ras-signaling pathway inhibitor.
2. DEFINITIONS
[0071] As used herein, the singular forms "a," "an" and "the" include plural references unless the content clearly dictates otherwise.
[0072] The term "about", as used here, refers to +/- 10% of a value.
[0073] As used herein, the term "pluripotency" refers to the nature of a cell, i.e., an ability to differentiate into a variety of tissues or organs. Typically, the pluripotency of a differentiated cell is limited.
[0074] As used herein, "reprogramming" refers to a genetic process whereby differentiated somatic cells are converted into de-differentiated, pluripotent cells, and thus having a greater pluripotency potential than the cells from which they were derived. For example, the reprogrammed cells may express at least one of the following pluripotent cell- specific markers: SSEA-3, SSEA-4, TRA-1-60 or TRA 1-81. Preferably, the reprogrammed cells express all these markers.
3. DISEASE-SPECIFIC PLURIPOTENT STEM CELLS
[0075] In one aspect, the invention provides an isolated primate pluripotent stem cell derived from a somatic cell, wherein the stem cell comprises a mutation in the protein tyrosine phosphatase non-receptor type 11 (PTPN11) gene. Mutations in the PTPN11 gene have been implicated in several diseases, such as LEOPARD syndrome, Noonan syndrome, and leukemia. Therefore, the stem cells provided herein are useful for studying the pathologic tissue formation in vitro, and for screening novel therapeutic agents for treating or diagnosing diseases that are associated with PTPN11 mutations.
A. PTPN11 Gene and PTPN11 Protein
[0076] PTPN11 protein (also known as SHP2, Syp, SHPTP2, PTP2C, PTP1D or BPTP3) is a member of a small subfamily of cytoplasmic, SH2-domain-containing protein tyrosine phosphatases that control cellular proliferation and differentiation (Feng, Exp Cell Res 1999;253:47-54; Tartaglia et al, Nat genet 2001;29:465-68; Tartaglia et al, Am J Hum Genet 2002;70: 1555-63). Human PTPN11 is ubiquitously expressed in all tissues examined, with higher levels of expression in the heart and the brain (Ahmad et al, Proc Natl Acad Sci USA 1993;90:2197-2201; Bastien et al, Biochem Biophys Res Commun 1993 ; 196: 124-133; Freeman et al, Proc Natl Acad Sci USA, 1992;89: 11239-11243).
[0077] PTPN11 is a key molecule in intracellular signaling and is necessary for activation of the RAS/MAPK cascade in response to a variety of growth factors, hormones and cytokines (Maroun et al, Mol Cell Biol 2000;20:8513-25; Shi Z-Q, et al, Mol Cell Biol 2000; 20: 1526-1536; Cunnick et al, J Biol Chem 2002;277:9498-504). PTPN11 is required during embryogenesis for mesodermal patterning (Tang et al, Cell 1995;80:473-483), semilunar valvuogenesis (Chen et al, Nat Genet 2000;24:296-9) and skeletal and limb development (Qu et al, Mol Cell Biol 1998; 18:6075-82; Saxon et al, Nat Genet 2000;24:420-3). Loss of murine SHP2 severely suppresses development of erythroid/myeloid and lymphoid cell progenitors, (Qu et al, Mol Cell Biol 1998; 18:6075-82; Qu et al, Mol Cell Biol 1997; 17:5499-507; Qu et al, Blood 2001;97:911-4), suggesting that it participates in early events during hematopoetic stem/progenitor cell commitment and differentiation. PTPN11 also controls cell differentiation at later stages of hematopoiesis, and has a role in the function of differentiated erythroid, myeloid and lymphoid cells (Pazdrak et al, J Exp Med 1997; 186:561-8; Edmead et al, FEBS Lett 1999;459:27-32; Ohtani et al, Immunity 2000;12:95-105; Tamir et al, Curr Opin Immunol 2000;12:307-15; Bordin et al, Blood 2002; 100:276-82). These effects appear to be mediated through a signal relay downstream of receptors for a number of hematopoietic growth factors and cytokines, including GM-CSF (Pazdrak et al, J Exp Med 1997;186:561-8; Welham et al, J Biol Chem 1994;269:23764-8; Tauchi et al, J Biol Chem 1995;270:5631-5; Tauchi et al, J Biol Chem 1994;269:25206-1138; Ward et al, Biochem Biophys Acta 1998;1448:70-6; Lecoq-Lafon et al, Blood 1999;93:2578-85; You et al, J exp Med 2001;193: 101-10).
[0078] The human PTPNl 1 gene organization and intron boundary sequence can be established using cDNA (GENBANK Accession Nos. NM_002834; amino acid and nucleotide sequences represented herein as SEQ ID Nos.: 1 and 2, respectively) and genomic sequences (GENBANK Accession Nos. NG 007459).
[0079] Figure 18 shows the organization of the PTPNl 1 gene and the functional domains of the PTPNl 1 protein. The PTPNl 1 protein comprises two SH2 (src-homology 2) domains, one from amino acid 3 to amino acid 104, the other from amino acid 112 to amino acid 216, and one PTP (protein tyrosine phosphatase) domain, from amino acid 221 to amino acid 524.
[0080] The PTPNl 1 gene of the invention also encompasses nucleic acid sequences comprising a coding sequence as set forth in SEQ ID NO:2, homo logs (including allelic variants and orthologs), sequence-conservative variants, or function-conservative variants thereof. The PTPNl 1 protein of the invention also encompasses amino acid sequences comprising the amino acid sequence as set forth in SEQ ID NO: l, homo logs (including orthologs), or function- conservative variants thereof.
[0081] As used herein, the term "homologous" in all its grammatical forms and spelling variations refers to the relationship between polynucleotides or proteins that possess a "common evolutionary origin," including polynucleotides or proteins from superfamilies and homologous polynucleotides or proteins from different species (Reeck et al, Cell 50:667, 1987). Such polynucleotides or proteins have sequence homology, as reflected by their sequence similarity, whether in terms of percent identity or the presence of specific amino acids or motifs at conserved positions.
[0082] "Sequence-conservative variants" of a polynucleotide sequence are those in which a change of one or more nucleotides in a given codon position results in no alteration in the amino acid encoded at that position.
[0083] "Function-conservative variants" are those in which one or more nucleotides of a polynucleotide sequence have been changed without altering the overall conformation and function of the polypeptide encoded by the polynucleotide; or one or more amino acid residues
of a polypeptide sequence have been changed without altering the overall conformation and function of the polypeptide. Function-conservative variants includes, e.g., replacement of an amino acid with one having similar properties (such as, for example, polarity, hydrogen bonding potential, acidic, basic, hydrophobic, aromatic, and the like). Amino acids with similar properties are well known in the art. For example, arginine, histidine and lysine are hydrophilic- basic amino acids and may be interchangeable. Similarly, isoleucine, a hydrophobic amino acid, may be replaced with leucine, methionine or valine. Such changes are expected to have little or no effect on the apparent molecular weight or isoelectric point of the protein or polypeptide.
[0084] An amino acid position "corresponding to" a position in another sequence is the position that lines up to the reference position when the amino acid sequence is aligned with the reference sequence. Alignments can be done by hand or by using well-known sequence alignment programs such as ClustalW2.
[0085] Other variants of PTPN11 gene may include those polynucleotide sequences comprising a coding sequence that is at least about 70%, about 75%, about 80%>, about 85%, about 90%, about 95%, or about 99% identical to SEQ ID NO:2. Preferably the variant gene encodes a protein that has the same or substantially similar properties or functions as the native or parent PTPN11 protein.
[0086] Other variants of PTPN11 protein may include those polypeptide sequences comprising an amino acid sequence that is at least about 70%>, about 75%, about 80%, about 85%, about 90%, about 95%, or about 99% identical to SEQ ID NO: 1. Preferably the variant protein has the same or substantially similar properties or functions as the native or parent PTPN11 protein.
B. Diseases Associated with Mutations in the PTPN11 Gene
[0087] A disease is "associated with" a mutation in the PTPN11 gene when the disease is directly or indirectly caused by, or correlates with a mutation in the PTPN11 gene. Such mutation of PTPN11 may lead to a non- functioning or defective PTPN11 protein, or altered expression level of the PTPN11 gene. Such mutation may be a substitution mutation (i.e., a mutation in which one or more bases within the nucleic acid sequence have been replaced by a
different base), an insertion mutation (i.e, a mutation in which the total length of the gene of interest has been increased by the insertion of one or more bases), a deletion mutation (a mutation in which the total length of the gene of interest has been decreased by removal of one or more bases) or an inversion mutation (a mutation in which a region of two or more bases has been rotated 180 degrees), or a combination thereof. The mutation may be directly, or indirectly, fully or partly responsible for the disease, or alternatively, the mutation may not be responsible for the disease but correlates with the diseased state in the sense that it is diagnostic/indicative of the diseased state.
[0088] In certain embodiments, the mutation in the PTPN11 gene results in a mutation in the encoded PTPN11 protein.
[0089] In certain embodiments, the stem cells described herein comprise a mutation in the PTPN11 gene that is associated with Noonan Syndrome.
[0090] "Noonan syndrome" (NS) encompasses all forms of the disorder as described under the accession No. MIM 163950 in the Online Mendelian Inheritance in Man (OMIM) at World-Wide Web Address www.ncbi.nlm.nih.gov/Omim, (as accessed in April 2010), as well as disorders similar or related to NS. Such disorders include, but are not limited to, the Watson (MIM 193520) and LEOPARD (MIM 151100) Syndromes (Mendez and Opitz, Am J Med Genet 1985;21 :493-506); male Turner and female pseudo-Turner Syndrome, as well as Turner phenotype with normal karyotype (see MIM 163950); Noonan syndrome with multiple giant-cell lesions (MIM 163955; Tartaglia et al, Am J Hum Genet 2002;70: 1555-1563) and/or Noonan syndrome with multiple caf-au-lait spots (also known as LEOPARD syndrome, MIM 151100; Digilio et al, Am J Hum Genet 2002;71 :389-394; Legius et al, J Med Genet 2002;39:571-574); valvular sclerosis (Snellen et al, Circulation 1968;38(1 Suppl):93-101); and idiopathic short stature (Attie K M, Curr Opin Pediatr 2000;12:400-404).
[0091] NS encompasses familial or sporadic forms, including NS1, whose locus has been identified on chromosome 12. The present invention takes into consideration, however, that NS and its related disorders are genetically heterogeneous, but share phenotypical features. The features of NS have been well described and a clinical scoring system devised. See, Mendez and Opitz, Am J Med Genet 1985;21 :493-506; Noonan, Clin Pediatr (Phila) 1994;33:548-555;
Sharland et al, Arch Dis Child 1992;67: 178-183; Duncan et al, Am J Med Genet 1981;10:37- 50).
[0092] Approximately 45% of NS are caused by missense mutations in the PTPN11 gene. For example, in more than 50% of patients with Noonan syndrome, Tartaglia et al.
(Nature Genet. 29: 465-468, 2001; Errata: Nature Genet. 29: 491, 2001; Nature Genet. 30: 123, 2002) identified several missense mutations in the PTPN11 gene: a G-to-T mutation at position 214 in exon 3, resulting in an Ala72-to-Ser (A72S) substitution in the N-SH2 domain; a C-to-G mutation at nucleotide 215 in exon 3, resulting in an Ala72-to-Gly (A72G) substitution; and an A-G mutation in exon 8, resulting in an Asn308-to-Asp (N308D) substitution in the
phosphotyrosine phosphatase (PTP) domain. About one-third of the patients who had mutations in the PTPN11 gene had the N308D mutation, which was by far the most common. That codon 308 is a hotspot for Noonan syndrome is further supported by the finding of an Asn308-to-Ser missense mutation in 2 families that had typical features of Noonan syndrome associated with multiple giant cell lesions in bone (Tartaglia et al., Am. J. Hum. Genet. 70: 1555-1563, 2002). All the PTPN11 missense mutations identified are clustered in the interacting portions of the amino N-SH2 domain and the phosphotyrosine phosphatase (PTP) domains, which are involved in switching the protein between its inactive and active conformations. The findings suggest that gain-of-function changes resulting in excessive SHP-2 activity underlie the pathogenesis of Noonan syndrome.
[0093] Using direct DNA sequencing, Maheshwari et al. (Hum. Mutat. 20: 298-304, 2002) surveyed 16 subjects with the clinical diagnosis of Noonan syndrome from 12 families and their relevant family members for mutations in the PTPN11/SHP2 gene, and found 3 different mutations among 5 families. Two unrelated subjects shared a Ser502-to-Thr (S502T) substitution in exon 13; 2 additional unrelated families had a Tyr63-to-Cys (Y63C) mutation in exon 3; and 1 subject had a Tyr62-to-Asp (Y62D) substitution, also in exon 3. In the mature protein model, the exon 3 mutants and the exon 13 mutant amino acids cluster at the interface between the N-terminal SH2 domain and the phosphatase catalytic domain. Six of 8 subjects with mutations had pulmonary valve stenosis. These results confirm that mutations in PTPN11 underlie a common form of Noonan syndrome, and that the disease exhibits both allelic and
locus heterogeneity. The observation of recurrent mutations supports the hypothesis that a special class of gain-of-function mutations in SHP2 gives rise to Noonan syndrome.
[0094] Kosaki et al. (J. Clin. Endocr. Metab. 87: 3529-3533, 2002) analyzed the PTPN11 gene in 21 Japanese patients with Noonan syndrome. Mutation analysis of the 15 coding exons and their flanking introns by denaturing HPLC and direct sequencing revealed 6 different heterozygous missense mutations in 7 cases. The mutations clustered either in the N- Src homology 2 domain or in the protein-tyrosine phosphatase domain.
[0095] Musante et al. {Europ. J. Hum. Genet. 11 : 201-206, 2003; Erratum: Europ. J. Hum. Genet. 11 : 551, 2003) screened the PTPN11 gene for mutations in 96 familial or sporadic Noonan syndrome patients. They identified 15 mutations, all of which were missense mutations; 11 of them were located in exon 3, which encodes the N-SH2 domain.
[0096] Tartaglia et al. (Am. J. Hum. Genet. 75: 492-497, 2004) investigated the parental origin of de novo PTPN11 lesions and explored the effect of paternal age in Noonan syndrome. By analyzing intronic positions that flank the exonic PTPN11 lesions in 49 sporadic Noonan syndrome cases, they traced the parental origin of mutations in 14 families. All mutations were inherited from the father.
[0097] Yoshida et al. (J. Clin. Endocr. Metab. 89: 3359-3364, 2004) reported PTPN11 mutation analysis and clinical assessment in 45 Japanese patients with Noonan syndrome. Sequence analysis of the coding exons 1 through 15 of PTPN11 revealed a novel 3- bp deletion (181delGTG, resulting in deletion of the Gly60 codon in the N-SH2 domain of the protein) and 10 recurrent missense mutations in 18 patients. Because Gly60 is directly involved in the N-SH2/PTP interaction, loss of this residue was predicted to disrupt N-SH2/PTP binding, activating the phosphatase function.
[0098] Becker et al. (Am. J. Med. Genet. 143A: 1249-1252, 2007) reported what they stated was the first known case of compound heterozygosity for NS-causing mutations in the PTPN11 gene (compound heterozygosity for N308S and Y63C mutations), resulting in early fetal death.
[0099] Other PTPNl 1 mutations that have been associated with Noonan syndrome include, e.g., A-to-G mutation at nucleotide 182 in exon 3 (Asp61-to-Gly, D61G), 218C-T mutation in exon 3 (Thr73-to-Ile, T73I), T-to-C mutation at nucleotide 854 in exon 8 (Phe285-to- Ser, F285S), 1909A-G mutation (Gln510-to-Arg); 236A-G mutation in exon 3 (Gln79-to-Arg, Q79R); and T-to-C mutation in exon 11 (Thr411 -to-Met, T411M). See, Kosaki et al. (J. Clin. Endocr. Metab. 87: 3529-3533, 2002); Bertola et al. (Am. J. Med. Genet. 136A: 242-245, 2005); Schollen et al. (Europ. J. Hum. Genet. 11 : 85-88, 2003).
[00100] Fig. 19 shows the distribution of PTPNl 1 (SHP-2) mutations and their relative prevalence in Noonan syndrome
[00101] In certain embodiments, the stem cells described herein comprise a mutation in the PTPNl 1 gene that is associated with LEOPARD Syndrome.
[00102] LEOPARD syndrome is an autosomal dominant disorder characterized by lentigines and cafe-au-lait spots, facial anomalies, and cardiac defects, sharing several clinical features with Noonan syndrome. Digilio et al. (Am. J. Hum. Genet. 71 : 389-394, 2002) screened 9 patients with LEOPARD syndrome (including a mother-daughter pair), and 2 children with Noonan syndrome who had multiple cafe-au-lait spots, for mutations in the PTPNl 1 gene. They found two missense mutations: an A-to-G mutation at nucleotide 836 in exon 7, resulting in a Tyr279-to-Cys (Y279C) mutation; and a Thr468-to-Met (T468M) mutation resulting from a C- to-T mutation at nucleotide 1403 in exon 12, respectively. Both mutations affected the PTPNl 1 phosphotyrosine phosphatase domain, which is involved in less than 30% of the Noonan syndrome PTPNl 1 mutations. This study demonstrated that LEOPARD syndrome and Noonan syndrome are allelic disorders. The detected mutations suggested that distinct molecular and pathogenetic mechanisms cause the peculiar cutaneous manifestations of the LEOPARD syndrome subtype of Noonan syndrome.
[00103] In 4 of 6 Japanese patients with LEOPARD syndrome, Yoshida et al. (Am. J. Med. Genet. 130A: 432-434, 2004) identified 3 heterozygous missense mutations: Tyr279 to Cys (Y279C), Ala461 to Thr (A461T, resulting from a 1381G-A mutation in exon 12), or Gly464 to Ala (G464A, resulting from a 1391G-C mutation in exon 12).
[00104] Kontaridis et al. (J. Biol. Chem. 281 : 6785-6792, 2006) examined the enzymatic properties of mutations in PTPNl 1 causing LEOPARD syndrome and found that, in contrast to the activating mutations that cause Noonan syndrome and neoplasia, LEOPARD syndrome mutants are catalytically defective and act as dominant-negative mutations that interfere with growth factor/ERK-MAPK-mediated signaling. Molecular modeling and biochemical studies suggested that LEOPARD syndrome mutations control the SHP2 catalytic domain and result in open, inactive forms of SHP2. Kontaridis et al. concluded that the pathogenesis of LEOPARD syndrome is distinct from that of Noonan syndrome and suggested that these disorders should be distinguished by mutation analysis rather than clinical
presentation.
[00105] Other PTPN 11 mutations that have been associated with LEOPARD syndrome include, e.g., 1529A-C mutation in exon 13 (Gln510-to-Pro, Q510P) (Kalidas et al. (J. Hum. Genet. 50: 21-25, 2005) and the following missense PTPNl I mutations, in exon 7, 12 and 13: Tyr279Cys/Ser, Ala461Thr, Gly464Ala, Thr468Met/Pro, Arg498Trp/Leu, Gin506Pro, and GlnS lOGlu/Giy (Sarkozy A., et al, Orphanet J Rare Dis 3, 13 (2008)).
[00106] Other diseases that are known to be associated with a mutation in the PTPNl 1 gene include, e.g., cardiofaciocutaneous syndrome (MIM 163955), juvenile myelomonocytic leukemia (NIM 607785), and certain other malignancies.
[00107] In certain embodiments, the stem cells described herein comprise a mutation in the PTPNl 1 gene that is associated with leukemia.
[00108] Juvenile myelomonocytic leukemia (JMML), a disorder with excessive proliferation of myelomonocytic cells, constitutes approximately 30% of childhood cases of myelodysplasia syndrome (MDS) and 2% of leukemia. JMML is observed occasionally in patients with Noonan syndrome. In 5 unrelated children with Noonan syndrome and JMML, Tartaglia et al. (Nature Genet. 34: 148-150, 2003) found heterozygosity with respect to a mutation in exon 3 of PTPNl 1. Four of the children shared the same mutation (218C-T). In 2 unrelated individuals with growth retardation, pulmonic stenosis, and JMML, they found missense defects in PTPNl 1 : the 218C-T transition, and a defect in exon 13 affecting the protein tyrosine phosphatase domain.
[00109] Tartaglia et al. also identified missense mutations in PTPNl 1 in 21 of 62 individuals with JMML but without Noonan syndrome, with 9 different molecular defects in exon 3 and 1 in exon 13. Nonhemato logic DNAs were available for 9 individuals with a mutation in PTPNl 1 in their leukemic cells, and none harbored the defect.
[00110] Other PTPNl 1 mutations that have been associated with JMML include, e.g., 226G-A mutation (Glu76-to-Lys, E76K), Glu76-to-Val, Glu76-to-Gly, Glu76-to-Ala (Tartaglia et al).
[00111] Tartaglia et al. also investigated the prevalence of somatic mutations in PTPNl 1 among 50 children with myelodysplasia syndrome. They identified missense mutations in exon 3 in 5 of 27 children with an excess of blasts. Three of these mutations were also associated with JMML in other patients. Among 24 children with de novo AML (MIM 601626), they identified a novel trinucleotide substitution in an infant with acute monoblastic leukemia.
[00112] In certain embodiments, the stem cells described herein comprise a mutation in the PTPNl 1 gene that is associated with a cardiac anomaly.
[00113] A cardiac anomaly refers to any structural or functional abnormality or defect of the heart or great vessels. The general effects of cardiac malformations on cardiovascular functioning are increased cardiac workload, increased pulmonary vascular resistance, inadequate cardiac output, and, in the case of cyanotic anomalies, decreased oxygen saturation. The general physical symptoms of these pathophysiologic alterations are growth retardation, decreased exercise tolerance, recurrent respiratory infections, dyspnea, tachypnea, tachycardia, cyanosis, tissue hypoxia, and murmurs, all of which vary in severity, depending on the type and degree of the defect.
[00114] Patients affected with cardiofaciocutaneous syndrome (CFC) present with symptoms that some considered to represent a more severe expression of Noonan syndrome, namely, congenital heart defects, cutaneous abnormalities, Noonan-like facial features, and severe psychomotor developmental delay. Bertola et al. {Am. J. Med. Genet. 130A: 378-383, 2004) identified a T-to-C mutation in exon 11 of the PTPNl 1 gene, resulting in a Thr411 -to-Met (T411M) substitution.
[00115] In certain embodiments, the stem cells described herein comprise a mutation in the PTPNl 1 gene that is associated with cardiac hypertrophy (also known as hypertrophic cardiomyopathy) .
[00116] Cardiac hypertrophy refers to the process in which cardiac myocytes respond to stress through hypertrophic growth. Such growth is characterized by cell size increases without cell division, assembling of additional sarcomeres within the cell to maximize force generation, and an activation of a fetal cardiac gene program. Pathologic cardiac hypertrophy is often associated with increased risk of morbidity and mortality, and thus studies aimed at understanding the molecular mechanisms of cardiac hypertrophy could have a significant impact on human health.
[00117] In certain embodiments, the stem cells described herein comprise a threonine to methionine substitution in PTPNl 1 protein at an amino acid residue position corresponding to position 468 of SEQ ID NO: 1 , or a tyrosine to cysteine substitution in PTPNl 1 protein at an amino acid residue position corresponding to position 279 of SEQ ID NO: 1. In certain embodiments, the PTPNl 1 protein comprises SEQ ID NO: 1, and the mutation is a threonine to methionine substitution at amino acid residue position 468 of SEQ ID NO: 1 , or a tyrosine to cysteine substitution at amino acid residue position 279 of SEQ ID NO: 1.
C. Characterization of the Diseases-Specific Pluripotent Stem Cells
[00118] The disease-specific pluripotent stem cells provided herein show two important characteristics that distinguish them from other types of cells. First, they are non- lineage committed cells that are capable of maintaining their pluripotent state and of renewing themselves for long periods through cell division. Second, under appropriate conditions, they can be induced to differentiate into cells with specialized functions.
[00119] The disease-specific pluripotent stem cells may be characterized by any of several criteria of pluripotency known in the art. For example, the stem cells are capable of continuous indefinite replication in vitro. Continued proliferation for a long period of time (e.g., at least about 1 month, at least about 2 months, at least about 4 months, at least about 6 months, at least about 9 months, at least about one year) of a culture is sufficient evidence of immortality,
as primary cell cultures without this property fail to continuously divide for such a length of time (Freshney, Culture of animal cells. New York: Wiley-Liss, 1994).
[00120] Additionally or alternatively, the disease-specific pluripotent stem cells may be characterized by the expression of certain markers, including but not limited to cell surface markers. Stem cells from different species may exhibit species-specific markers on their cell surfaces. For example, Thomson (U.S. Pat. Nos. 5,843,780 and 6,200,806) discloses certain cell surface markers that may be used to identify pluripotent stem cells derived from primates.
Furthermore, Stage Specific Embryonic Antigens (SSEAs) are unique carbohydrate epitopes that may be used to characterize pluripotent stem cells. Stem cells derived from different species exhibit different patterns of SSEAs. For example, undifferentiated primate pluripotent stem cells (including human pluripotent stem cells) express SSEA-3 and SSEA-4, but not SSEA-1.
Undifferentiated mouse pluripotent stem cells express SSEA-1, but not SSEA-3 or SSEA-4.
[00121] Additionally or alternatively, markers that are not exhibited on the surface of a cell may be used to characterize a pluripotent stem cell. For example, the homeodomain transcription factor Oct 4 (also termed Oct-3 or Oct3/4) is frequently used as a marker for pluripotent stem cells.
[00122] In certain embodiments, the pluripotent stem cells provided herein express SSEA-3, SSEA-4, TRA-1-60, TRA-1-81, Nanog, OCT-4, Sox2, GDF3, DPPA4, REX1, TERT, Alkaline Phosphatase, Telomerase, or CD30, or a combination thereof.
[00123] The disease-specific pluripotent stem cells described herein are capable of differentiating into all three embryonic germ layers (endoderm, mesoderm, and ectoderm) in a fashion similar to embryonic stem cells. Such potency and differentiability can be demonstrated by, e.g., teratoma formation (e.g., injecting the stem cells into an immunodeficient mouse such as a SCID mouse, and then histologically examining the resulting tumors). Additionally or alternatively, the disease-specific pluripotent stem cells may be characterized by the capacity to develop into fully differentiated somatic cell lineages (e.g., neurons, cardiomyocytes, etc) and/or the germ line. Additionally or alternatively, the disease-specific pluripotent stem cells may be characterized by the capacity to participate in normal development when transplanted into a preimplantation embryo to generate a chimeric embryo.
[00124] Additionally or alternatively, the disease-specific pluripotent stem cells may be characterized by their morphology (a stem cell has round shape, large nucleolus and scant cytoplasm; human iPSCs form sharp-edged, flat, tightly-packed colonies similar to human embryonic stem cells), telomerase activity (pluripotent stem cells express high telomerase activity to sustain self-renewal and proliferation), promoter demethylation (promoters of pluripotency-inducing genes, such as Oct-3/4, Rexl, and Nanog, are generally demethylated in iPSCs), or histone demethylation (H3 histones associated with Oct-3/4, Sox2, and Nanog genes are generally demethylated).
[00125] Exemplary stem cell lines were deposited on , 2010 with American
Type Culture Collection (ATCC), P.O. Box 1549, Manassas, VA 20108, USA, under ATCC Accession No. and .
[00126] In certain embodiments, the pluripotent stem cell of the present invention is a primate stem cell. In certain embodiments, the primate is human. In other embodiments, the pluripotent stem cell is a non-human stem cell (e.g., a mouse stem cell, a rat stem cell, a pig stem cell, a sheep stem cell, a goat stem cell, or a non-human primate stem cell).
[00127] The capacity of pluripotent stem cells to self renew in culture, while retaining their pluripotent potential, provides the opportunity to produce virtually unlimited numbers of undifferentiated and differentiated cell types for in vitro and in vivo investigation of disease mechanisms (Lerou, P. H. & Daley, G. Q. Therapeutic potential of embryonic stem cells. Blood Rev 19, 321-31, 2005; Ben-Nun, I. F. & Benvenisty, N. Human embryonic stem cells as a cellular model for human disorders. Mol Cell Endocrinol 252, 154-9, 2006). Pluripotent stem cells carrying the genes responsible for a particular disease (e.g., a mutation in the PTPN11 gene) can be induced to differentiate into the cell types affected in that disease. Studies of the differentiated cells in culture could provide important information regarding the molecular and cellular nature of events leading to pathology.
[00128] In certain embodiments, this approach is used to develop an in vitro model of LEOPARD syndrome and cardiac hypertrophy. As described more fully below in the Examples section, induced pluripotent stem cell lines were derived from two LEOPARD syndrome patients, and an unaffected brother not bearing the T468M mutation of one of the LS patients.
These three pluripotent stem cell lines were differentiated into cardiomyocytes in culture. These in vitro differentiated cardiomyocytes could be maintained in culture, providing the opportunity to detect differences between the cardiomyocytes derived from stem cells carrying a mutant PTPN11 and those derived from stem cells that do not carry the mutation.
D. Differentiation of the Disease-Specific Pluripotent Stem Cells
[00129] The disease-specific pluripotent stem cells described herein may be induced in vitro to differentiate into a variety of somatic cell lineages (e.g., neurons, cardiomyocytes, etc). In one aspect, the invention provides cardiomyocytes comprising a mutation in the PTPN11 gene, wherein the cardiomyocyte is produced by differentiating the disease-specific pluripotent stem cell described herein.
[00130] The disease-specific pluripotent stem cells may be subjected to any conditions that induce the stem cells to differentiate into cardiomyocytes. For example, the disease-specific pluripotent stem cells may be suspended into a single-cell suspension, allowed to spontaneously aggregate into embryoid bodies over a first period of time (e.g. 48 hours, although such a period of time may be increased or decreased depending on other conditions to which the embryonic stem cells are subjected), and then treated with a suitable differentiation factor or factors for a second period of time such that the stem cells differentiate into cardiomyocytes. By way of example, such differentiation factors may include activin A, bone morphogenetic protein 4 (BMP4), basic fibroblast growth factor (bFGF), vascular endothelial growth factor (VEGF) dickkopf homo log 1 (DK 1), or a combination thereof.
[00131] In certain embodiments, the invention provides a method of differentiating a pluripotent stem cell as described herein into a cardiomyocyte, comprising culturing the stem cell in a serum- free medium that comprises: activin A, bone morphogenetic protein 4 (BMP4), basic fibroblast growth factor (bFGF), vascular endothelial growth factor (VEGF) and dickkopf homolog 1 (DKK1).
[00132] Alternatively, the stem cells may be induced to differentiate into
cardiomyocytes in the presence of a prostaglandin, analogue or functional equivalent thereof, alone or in combination with other factors (such as minerals (e.g., selenium), small molecules
(e.g., a p38 MAPK inhibitor such as SB203580), transferring, protein growth factors of the FGF, IGF and BMP families (e.g., IGF1, FGF2, BMP2, BMP4, BMP6, or a homologue or functional equivalent thereof), β-ΜΕ, non-essential amino acids, L-glutamine, etc.), as described in U.S. Application Publication No. 20070204351. The prostaglandin, analogue or functional equivalent thereof may be prostacyclin (PGI2), an analogue or functional equivalent thereof, including its naturally breakdown form of 6-keto-Prostaglandin Fla (6k-PGFla) and 2,3-dinor-6-keto- Prostaglandin Fla (2,3d-6k-PGFla); its synthetic analogs, such as iloprost, cicaprost, and carbarprostacyclin (cPGI) and stable chemical structures (Whittle, 1980, Town, 1982,
Sturzebecher, 1986); and its derivatives, as described in U.S. Application Publication No.
20070204351.
[00133] Also encompassed within the invention are cardiomyocyte progenitors that are differentiated from the pluripotent stem cells described herein. A cardiomyocyte progenitor, or cardiac progenitor cell, refers to a cell that is resident in the heart, or that comes into the heart from elsewhere after acute ischemia, is smaller than a mature cardiomyocyte, expresses a- sarcomeric actin but is negative for troponin, is normally quiescent but can be induced to go into the cell cycle as defined by positive Ki67 staining.
[00134] The cardiomyocytes and cardiomyocyte progenitors described herein may be beating. The cardiomyocytes may be fixed and stained with a-actinin antibodies to confirm muscle phenotype. a-troponin, a-tropomysin and a-MHC antibodies also give characteristic muscle staining.
[00135] In certain embodiments, a cardiomyocyte differentiated from a stem cell provided herein comprises a mutation in the PTPN11 gene. In certain embodiments, the mutation is associated with a cardiac anomaly (such as hypertrophic cardiomyopathy). In certain embodiments, the mutation is associated with LEOPARD Syndrome, Noonan Syndrome or leukemia. For example, in humans, a threonine to methionine substitution at amino acid residue position 468 of PTPN11 protein (SEQ ID NO: 1), or a tyrosine to cysteine substitution at amino acid residue position 279 of PTPN11 protein (SEQ ID NO: 1) is associated with LEOPARD syndrome/ hypertrophic cardiomyopathy.
[00136] In certain embodiments, the invention provides a cell culture of the cardiomyocytes described herein. Exemplary cardiomyocyte cell lines were deposited on
, 2010 with American Type Culture Collection (ATCC), P.O. Box 1549, Manassas, VA
20108, USA, under ATCC Accession No. and .
4. METHODS OF MAKING DISEASE-SPECIFIC PLURIPOTENT STEM CELLS
[00137] In another aspect, the invention provides methods of producing the disease- specific pluripotent stem cells as described herein.
[00138] In certain embodiment, the invention provides methods of producing the disease-specific pluripotent stem cells comprising (1) obtaining a somatic cell from a donor whose genome comprises a mutation in the PTPN11 gene, and (2) reprogramming the somatic cell into a pluripotent stem cell. In certain embodiments, the reprogramming comprises introducing into the somatic cell: (i) a nucleic acid molecule that encodes a pluripotency- inducing protein; or (ii) a polypeptide that comprises a pluripotency-inducing protein.
[00139] Methods for nuclear reprogramming of a somatic cell nucleus have been reported. One technique for nuclear reprogramming involves nuclear transfer into oocytes (Wakayama et al. Nature 394:369-374, 1998; Wilmut et al. Nature 385:810-813, 1997). Another method of reprogramming a somatic cell nucleus involves fusing a somatic cell and an ES cell (Tada et al. Curr. Biol. 11 : 1553-1558, 2001; Cowan et al. Science 309: 1369-73, 2005).
Alternatively, a somatic cell nucleus can be reprogrammed by treating a differentiated cell with an undifferentiated human carcinoma cell extract (Taranger et al. Mol. Biol. Cell 16:5719-35, 2005).
[00140] Induced pluripotent stem cells typically can be produced by introducing into somatic cells certain proteins (in particular, transcription factors) that are involved in maintaining ES cell pluripotency. These proteins are referred herein as "pluripotency-inducing proteins." Exemplary pluripotency-inducing proteins and genes encoding pluripotency-inducing proteins have been disclosed by Yamanaka et al. (EP Application Publication No. 1970446; US
Application Publication Nos. 2009/0068742 and 2009/0047263), and Thomson et al. (US Application Publication No. 2008/0233610). The pluripotency-inducing proteins are referred to
as "nuclear reprogramming factors" by Yamanaka, and "potency-determining factors" by Thomson.
[00141] Exemplary pluripotency-inducing protein include, but are not limited to, Oct- 4, Sox2, FoxD3, UTF1, Stella, Rexl, ZNF206, Soxl5, Mybl2, Lin28, Nanog, DPPA2, ESG1, Otx2, Klf4, c-Myc, or combinations thereof.
[00142] In certain embodiments, the pluripotency-inducing protein is a protein from the Oct family (e.g., Oct3/4), Klf family (e.g., Klf4), Sox family (e.g., Sox2), or a combination thereof.
[00143] In certain embodiments, a set of two or more pluripotency-inducing proteins are introduced into the somatic cells. In embodiments, the set include Oct-4 and Sox2. In certain embodiments, the set include Oct3/4, Sox2, Klf4, and c-Myc. In other embodiments, the set include Oct3/4, Klf and c-Myc. Yet in another embodiment, the set include OCT4, SOX2, NANOG, and LIN28.
[00144] It is specifically envisioned that the set of pluripotency-inducing proteins sufficient to reprogram somatic cells can vary with the cell type of the somatic cells. One can identify a set of pluripotency-inducing proteins sufficient to reprogram other cells using different combinations of pluripotency-inducing proteins.
[00145] Suitable somatic cells can be any somatic cell, although higher
reprogramming frequencies are observed when the starting somatic cells have a doubling time of about twenty-four hours. Somatic cells useful in the invention may be non-embryonic cells obtained from a fetal, newborn, juvenile or adult primate, including a human. Examples of somatic cells that can be used with the methods described herein include, but are not limited to, bone marrow cells, epithelial cells, fibroblast cells, hematopoietic cells, hepatic cells, intestinal cells, mesenchymal cells, myeloid precursor cells and spleen cells. Another type of somatic cell is a mesenchymal cell that attaches to a substrate. Alternatively, the somatic cells can be cells that can themselves proliferate and differentiate into other types of cells, including blood stem cells (multipotent hematopoietic cells), muscle/bone stem cells, brain stem cells, liver stem cells, etc.
[00146] In certain embodiments, the somatic cell is a fibroblast.
[00147] Suitable somatic cells are receptive, or can be made receptive using methods generally known in the art, to uptake pluripotency-inducing proteins or nucleic acid sequences that encode the pluripotency-inducing proteins. Uptake-enhancing methods can vary depending on the cell type and expression system. Exemplary conditions used to prepare receptive somatic cells having suitable transduction efficiency are known in the art (such as electroporation).
[00148] A nucleic acid encoding a pluripotency-inducing protein can be introduced by transfection or transduction into the somatic cells using a vector, such as an integrating- or non- integrating vector (e.g., a retroviral vector, an adenoviral vector). For example, a retroviral vectors (e.g., a lentiviral vector) may be transduced by packaging the vector into virions prior to contact with a cell. After introduction, the DNA segment(s) encoding the pluripotency-inducing protein can be located extra-chromosomally (e.g., on an episomal plasmid) or stably integrated into cellular chromosome(s).
[00149] A viral-based gene transfer and expression vector is a genetic construct that enables efficient and robust delivery of genetic material to most cell types, including non- dividing and hard-to-transfect cells (primary, blood, stem cells) in vitro or in vivo. Often, viral- based constructs that are integrated into genomic DNA result in high expression levels. In addition to a DNA segment that encodes a pluripotency-inducing protein of interest, the vectors may include a transcription promoter and a polyadenylation signal operatively linked, upstream and downstream, respectively, to the DNA segment. For example, nucleic acid encoding a pluripotency-inducing protein may be operably linked to a heterologous promoter, which may become inactive after somatic cells are reprogrammed. The heterologous promoter is any promoter sequence that can drive expression of a polynucleotide sequence encoding the pluripotency-inducing protein in the somatic cell, such as an Oct4 promoter.
[00150] The vector can include a single DNA segment encoding a single pluripotency- inducing protein factor or encoding a plurality of pluripotency-inducing proteins. A plurality of vectors can be introduced into a single somatic cell. The vector can optionally encode a selectable marker to identify cells that have taken up and express the vector. As an example, when the vector confers antibiotic resistance on the cells, antibiotic can be added to the culture
medium to identify successful introduction of the vector into the cells. Either integrating vectors or non-integrating vectors may be used. Suitable integrating vectors include retroviral (e.g., lentiviral) vectors. Suitable non-integrating vectors include Epstein-Barr virus (EBV) vectors (Ren C, et al, Acta. Biochim. Biophys. Sin. 37:68-73 (2005); and Ren C, et al, Stem Cells 24: 1338-1347 (2006)). Adenoviral vectors may also be used to transport the nucleic acid into the somatic cell. Since the adenovirus does not integrated into cellular chromosome(s), the risk of creating tumors is reduced. Stadtfeld et al, Science, 332: 945 - 949 (2008). If desired, non- integrating vectors can be lost from cells by dilution after reprogramming.
[00151] To facilitate the identification of induced pluripotent stem cells, a non-lethal marker, such as Green Fluorescent Protein (GFP), Enhanced Green Fluorescent Protein (EGFP) or luciferase, under the control of a promoter active only after the somatic cell has been converted to a pluripotent state, may be used. A selectable marker gene may be used to identify the reprogrammed cells expressing the marker through visible cell selection techniques, such as fluorescent cell sorting techniques. Alternatively, the reprogrammed cells can be produced without a selectable marker. For example, a marker may be integrated into the genome of the somatic cells downstream of the promoter that regulates Oct4 expression. The endogenous Oct4 promoter is active in undifferentiated, pluripotent stem cells.
[00152] The non-lethal marker can be constructed to enable its subsequent removal using any of a variety of art-recognized techniques, such as removal via Cre -mediated, site- specific gene excision. For example, it may become desirable to delete the marker gene after the pluripotent cell population is obtained, to avoid interference by the marker gene product in the experiment or process to be performed with the cells. Targeted deletions can be accomplished by providing structure(s) near the marker gene that permits its ready excision. For example, a Cre/Lox genetic element can be used. The Lox sites can be built into the cells. If it is desirable to remove the marker from the pluripotent cells, the Cre agent can be added to the cells. Other similar systems also can be used. Because Cre/Lox excision can introduce chromosomal rearrangements and can leave residual genetic material after excision, it may also be desirable to introduce the pluripotency-inducing proteins into the somatic cells using non-integrating, episomal vectors and obtaining cells from which the episomal vectors are lost (e.g., at a rate of
about 5% per generation) by subsequently withdrawing the drug selection used to maintain the vectors during the reprogramming step.
[00153] The vectors described herein can be constructed and engineered using art- recognized techniques. Standard techniques for the construction of expression vectors suitable for use are well-known in the art and can be found in publications such as Sambrook J, et al., "Molecular cloning: a laboratory manual," (3rd ed. Cold Spring Harbor Press, Cold Spring Harbor, N.Y. 2001).
[00154] It is also possible to generate iPS cells by direct delivery of pluripotency- inducing proteins, thus eliminating the need for viruses or genetic modification of the somatic cells. For example, it has been reported that repeated treatment of the cells with certain proteins channeled into the cells via poly-arginine anchors was sufficient to induce pluripotency (Zhou et al, Cell Stem Cell, Volume 4, 381-384 (2009)). The expression of pluripotency-inducing genes can also be increased by treating somatic cells with FGF2 under low oxygen conditions.
[00155] The relative ratio of pluripotency-inducing proteins may be adjusted to increase reprogramming efficiency. For example, it has been reported that linking Oct-4 and Sox2 in a 1 : 1 ratio on a single vector increased reprogramming efficiency in cells by a factor of four, compared to reprogramming efficiency wherein the pluripotency-inducing genes were provided to cells in separate constructs and vectors, where the uptake ratio of the respective pluripotency-inducing genes into single cells was uncontrolled. Although the ratio of pluripotency-inducing proteins may differ depending upon the set of pluripotency-inducing proteins used, one of ordinary skill can readily determine optimal ratios of pluripotency-inducing proteins.
[00156] Pluripotent stem cells can be cultured in any medium used to support growth of pluripotent cells. Typical culture medium includes, but is not limited to, a defined medium, such as TeSR™ (StemCell Technologies, Inc.; Vancouver, Canada), mTeSR™ (StemCell Technologies, Inc.) and StemLine® serum-free medium (Sigma; St. Louis, Mo.), as well as a conditioned medium, such as mouse embryonic fibroblast (MEF)-conditioned medium.
Alternatively, cells can be maintained on MEFs in culture medium.
5. DISEASE MODELING AND DRUG SCREENING
[00157] In another aspect, the invention provides a method of identifying an agent for use in treatment of a disease that is associated with a mutation in the PTPN11 gene, comprising (1) contacting a disease-specific pluripotent stem cell as described herein with the agent; and (2) determining the effect of the agent on survival, proliferation, phenotype, or differentiation of the pluripotent stem cell.
[00158] In another aspect, the invention provides a method of identifying an agent for use in treatment of a disease that is associated with a mutation in the PTPN11 gene, comprising (1) contacting a cardiomyocyte as described herein with the agent; and (2) determining the effect of the agent on survival, proliferation, or phenotype of the cardiomyocyte.
[00159] In another aspect, the invention provides a method of identifying an agent for use in treatment of the cardiac hypertrophy that is associated with a mutation in the PTPN11 gene, comprising: (1) contacting a cardiomyocyte as described herein with the agent; and (2) determining the effect of the agent on the hypertrophic state of the cardiomyocyte, wherein a decrease in the hypertrophic state of the cardiomyocyte, as compared to that of a control cell, is indicative that the agent is an agent for use in treatment of a disease that is associated with a mutation in the PTPN11 gene.
[00160] The disease-specific pluripotent stem cells and cardiomyocytes described herein may be used to screen for test agents that affect survival, proliferation, phenotype, or differentiation of the cells. For example, the disease-specific stem cells may be induced to differentiate into cardiomyocytes by placing it under appropriate differentiation conditions. Before, during and/or after differentiation, the cell may be subjected to a test agent in order to determine whether that agent has an effect on survival, proliferation, phenotype, or
differentiation of the cells. The agents can be used for treatment, preventing or ameliorating the symptoms of the diseases.
[00161] The disease-specific stem cells and cardiomyocytes described herein may also be used to develop personalized treatment regimens. For example, certain patients may respond better to a given therapy or drug regimen than other patients. Additionally or alternatively,
certain patients may experience fewer and/or less severe side effects after being administered a given therapy or drug regimen than other patients. By utilizing stem cells that contain the genetic complement of a patient suffering from and/or predicted to suffer from a disease of interest, and permitting such cells to differentiate into a cell type associated with that disease, it will be possible to better predict which therapy or drug regimen will be most beneficial and/or result in the least detrimental side effects.
[00162] A control cell may be a parallel sample cell that has not been treated with the agent (e.g., recipient cells mock-treated with buffers), or a cell that has been treated with an agent having a known effect (e.g., a positive effect, a negative effect, or no effect).
Alternatively, a control cell may be a cell having known or pre-determined properties (e.g., a characterized cell line from a database or a cell not carrying the PTPNl 1 mutation of the disease- specific cell).
[00163] In certain embodiments, a disease of interest may be modeled and/or studied by creating a disease-specific pluripotent stem cell line, and inducing the cell line to differentiate under appropriate differentiation conditions. For example, a disease-specific pluripotent stem cell line may be generated in which the stem cells carry a mutation in the PTPNl 1 gene that is associated with LEOPARD syndrome, or a cardiac anomaly. By observing survival,
proliferation, phenotype, or differentiation of the stem cells and the in vitro differentiated cardiomyocytes, and comparing them to stem cells or cardiomyocytes that do not carry the mutation, one can better understand the genetic basis of disease progression and pathogenesis. The cells are particularly useful in studying and/or modeling diseases that have not been amenable to such study and/or modeling.
[00164] The test agent may be any molecule that increases or decreases the expression level or the activity level of PTPNl 1 protein. For example, the agent may be a small molecule compound, an antibody, a hormone, a vitamin, a nucleic acid molecule, an enzyme, an amino acid, or a virus.
[00165] The invention also demonstrates for the first time that mutations in the PTPNl 1 gene perturbs RAS-MAPK signal transduction as early as the pluripotent stem cell stage. RAS is a family of genes encoding small GTPases that are involved in cellular signal
transduction. Activation of Ras signalling causes cell growth, differentiation and survival.
Transmission of stimulatory signals from Ras to nuclear targets involves regulation of the family of kinases known as MAPKs ("mitogen-activated protein kinases") or ER s ("extracellular signal regulated kinases"). This pathway includes, but is not limited to, components such as RAFl (V-Raf-1 Murine Leukemia Viral Oncogene Homo log 1), SOS1 (Son of Sevenless Homolog 1 (Drosophila)), ERK2, KRAS (V-KI-RAS2 Kirsten Rat Sarcoma 2 Viral Oncogene Homo log), PTPN11, BRAF (B-Raf proto-oncogene serine/threonine -protein kinase), MEK1 (mitogen-activated protein kinase kinase 1), MEK2, and HRAS. Additional components of this pathway have been identified and described (see, e.g., Lee and McCubrey, Leukemia 16:486- 507, 2002).
[00166] In another aspect, the invention provides a method of identifying an agent as a candidate Ras-signaling pathway inhibitor, comprising: (1) contacting a pluripotent stem cell as described herein with the agent; and (2) determining the effect of the agent on tyrosine phosphorylation of EGFR (Epidermal growth factor receptor) or MEK1. A decrease of tyrosine phosphorylation of EGFR or MEK1, as compared to that of a control cell, is indicative that the agent is a Ras-signaling pathway inhibitor.
[00167] In another aspect, the invention provides a method of identifying an agent as a candidate Ras-signaling pathway inhibitor, comprising: (1) contacting a cardiomyocyte as described herein with the agent; and (2) determining the effect of the agent on tyrosine phosphorylation of EGFR or MEK1. A decrease of tyrosine phosphorylation of EGFR or MEK1, as compared to that of a control cell, is indicative that the agent is a Ras-signaling pathway inhibitor.
[00168] In another aspect, the invention provides a method of identifying an agent for use in treatment of a disease that is associated with a mutation in the PTPN11 gene, comprising: (1) contacting a pluripotent stem cell as described herein with the agent; (2) differentiating the stem cell into a cardiomyocyte; and (3) determining the hypertrophic state of the cardiomyocyte. A decrease in the hypertrophic state of the cardiomyocyte, as compared to that of a control cell, is indicative that the agent is an agent for use in treatment of a disease that is associated with a mutation in the PTPN11 gene.
[00169] The pluripotent stem cells and cardiomyocytes described herein may also be used to test or validate the therapeutic activities of known Ras kinase inhibitors or PTPN11 inhibitors. For example, the pluripotent stem cells or cardiomyocytes described herein may be cultured in the presence of a Ras inhibitor or PTPN11 inhibitor, and effect of the inhibitor on survival, proliferation, phenotype, or differentiation of the cells may be determined. Currently, there are a few known PTPN11 inhibitors, such as CDL 4340-0580 (Hellmuth et al, P roc Natl Acad Sci USA 105,7275-7280, 2008) and NAT6-297775 (Noren-Muller et al, Proc Natl Acad Sci USA . , 103 : 10606- 11 2006).
EXAMPLES
[0170] The invention now being generally described, it will be more readily understood by reference to the following examples, which are included merely for purposes of illustration of certain aspects and embodiments of the present invention, and are not intended to limit the invention.
[0171] The generation of reprogrammed induced pluripotent stem cells (iPSC) from patients with defined genetic disorders promises important avenues to understand the etiologies of complex diseases, and the development of novel therapeutic interventions. As shown by the Examples described below, the inventors have generated iPSCs from patients with LEOPARD syndrome (LS; acronym of its main features: Lentigines, Electrocardiographic abnormalities, Ocular hypertelorism, Pulmonary valve stenosis, Abnormal genitalia, Retardation of growth and Deafness), an autosomal dominant developmental disorder belonging to a relatively prevalent class of inherited RAS-MAPK signaling diseases, which also includes Noonan syndrome (NS), with pleomorphic effects on several tissues and organ systems (Gorlin, R.J., et al, Birth Defects Orig Artie Ser 07, 110-115 (1971); Sarkozy, A., et al, Orphanet J Rare Dis 3, 13 (2008)). The patient-derived cells have a mutation in the PTPN11 gene, which encodes the SHP2 phosphatase The iPSC have been extensively characterized and produce multiple differentiated cell lineages. A major disease phenotype in patients with LEOPARD syndrome is hypertrophic
cardiomyopathy. The inventors show that in vzYro-derived cardiomyocytes from LS-iPSC are larger, have a higher degree of sarcomeric organization and preferential localization of NFATc4 in the nucleus when compared to cardiomyocytes derived from human embryonic stem cells
(HESC) or wild type (wt) iPSC derived from an unaffected, non-PTPNl lbrother of one of the LS patients. These features indicate a hypertrophic state. The inventors also provide molecular insights into signaling pathways that may promote the disease phenotype.
EXAMPLE 1: MATERIALS AND METHODS
[00172] Cell culture. Dermal fibroblast lines were obtained from skin biopsies, collected under an Institutional Review Board-approved protocol and with informed consent. LEOPARD syndrome patients derived fibroblasts, BJ fibroblasts (American Type Culture Collection) and GP2 cells were maintained in Dulbecco's modified Eagle medium (DMEM) containing 10% fetal bovine serum and penicillin/streptomycin (Invitrogen). HES2 and iPSC were maintained on irradiated Swiss Webster mouse embryonic feeder cells (MEFs), in a serum free HESC medium composed of DMEM/F12 (Cellgro, Mediatech) containing 20 ng/ml basic fibroblast growth factor (bFGF, R&D systems), 20% (vol/vol) KSR (Invitrogen), 5% (vol/vol) MEF-conditioned medium, Penicillin/Streptomycin, L-glutamine (L-Gln), non-essential amino acids (Invitrogen), and β-mercaptoethanol (β-ΜΕ, Sigma-Aldrich).
[00173] Plasmid construction. Full-length sequences of human OCT4, SOX2, KLF4 and c-MYC transcription factors were obtained from Open Biosystems. The coding sequences were PCR amplified using Pfu Turbo (Stratagene) and cloned into pMXs vector and verified by sequencing. pMXs-EGFP vector was constructed by introducing the BamHI/NotI EGFP fragment from FUGW (kindly provided by Dr. Lois, MIT, Massachusetts) into pMXs vector. The latter vector was used to monitor the transfection and infection efficiency.
[00174] Retroviral infection and LEOPARD syndrome iPSC generation. OCT4, SOX2, KLF4 and c-MYC transcription factors were introduced in dermal fibroblasts derived from two patients with LEOPARD syndrome via the pMXs retroviral vector. In parallel, pMXs-EGFP vector was used to estimate the infection efficiency (data not shown). Six days after infection, fibroblasts were seeded onto MEFs. The following day the medium was replaced with the HESC medium and bFGF.
[00175] Specifically, GP-2 cells were plated at 8xl06 cells per 10-cm dish and transfected with pMXs, VSV-G and Gag-Pol vectors using SuperFect transfection reagent on the
following day. The same day, human fibroblasts were seeded at 8xl05 cells per 10-cm dish. Twenty-four hours after transfection, the four retroviruses (hOCT4, hSOX2, hKLF-4 and hC- MYC) containing supernatants were collected, and equal amounts of each were mixed and filtered through a 0.45 μιη pore-size filter, and supplemented with 4 μg/mL polybrene.
Retroviruses containing medium was added to the fibroblasts plates. The following day, forty- eight hours post-transfection, the fibroblasts were reinfected following the same procedure as the day before. Six days after transduction, fibroblasts were transferred into four dishes coated with MEFs, at 50,000 fibroblasts per plate.
[00176] DNA Fingerprinting analysis. In order to verify the genetic relatedness of the iPSC to their parental fibroblasts, the inventors PCR amplified across three discrete genomic loci containing highly variable numbers of tandem repeats. Genomic DNA (gDNA) was isolated with Easy-DNA™ kit (Invitrogen). Fifty nanograms of genomic DNA was used per reaction. Primers are summarized in Figure 17 (Table 1).
[00177] Karyotype analysis. HES2 and iPSC were grown on Matrigel-coated glass coverslip dishes (MatTek, Ma). The day of culture harvest, 20 μΐ of colcemid (5 μg/ml) was added to the in situ ESC1 culture which was 30-50% confluent. The culture was re-incubated for 15 min at 37°C. A robotic harvester (Tecan) was utilized, which included automatic addition of 2cc of hypotonic solution (sodium citrate solution 0.8%) with incubation for 20 min at room temperature, prefixation with addition of 2cc of fixative (methanol: glacial acetic acid; 3: 1), followed by addition of 4cc of fixative, twice. The coverslip was dried completely at 37 °C with 45-50%) humidity and mounted on a microscope slide and GTG-banded according to standard protocols. Metaphases were captured and karyotypes were prepared using the Cyto Vision software program (Version 3.92 Build 7, Applied Imaging).
[00178] qPCR and transgenes integration. For quantitative real-time PCR (qPCR) analyses, total RNA was extracted from cells using Trizol® Reagent (Invitrogen) and subsequently column-purified with RNeasy kit (Qiagen) and treated with RNase-free DNase (Qiagen). One microgram of total RNA was reverse transcribed into cDNA using random primers and Superscript II Reverse Transcriptase (Invitrogen). PCR for transgene silencing was performed with Expand High Fidelity Enzyme Tag Polymerase (Roche). Real-time qPCR was
performed on a StepOne Plus Real-Time PCR System (Applied Biosystems) with Fast
SYBR® Green Master Mix (Applied Biosystems). The results were analyzed with the StepOne Software v2.0, normalized to β-Actin gene expression, and compared to HES2 cell expression levels. To examine the presence of transgenes in the iPSC lines, gDNA was isolated with Easy- DNA Kit (Invitrogen). PCR reactions were carried out with the Expand High Fidelity Enzyme Taq Polymerase (Roche). Primer sequences are described in Table 1 (Figure 17). Primers for FGF receptors expression analysis have been previously described (Dvorak, P., et al, Stem Cells 23, 1200-1211 (2005)).
[00179] Southern blot analyses. gDNA (2μ ) was completely digested with Bgl II, separated on a 0.8% agarose gel, transferred to a positively charged nylon membrane, and hybridized with DIG-labeled hOCT4, hSOX2, hKLF-4 and hC-MYC cDNA probes. After hybridization, membranes were washed, blocked with DIG blocking solution, and incubated with anti-DIG-AP Fab fragments (Roche). Probe-target hybrids were then incubated with
chemiluminescent CDF-Star substrates (Roche) and detected via exposure to X-ray film.
[00180] Bisulfite sequencing. 500 ng of purified gDNA were treated with sodium bisulfite using the Zymo EZ-DNA Methylation Kit, following the manufacturer's instructions. The sequences of primers used for amplification of genomic fragments were previously published (Freberg, C.T., et al, Mol Biol Cell 18, 1543-1553 (2007)). PCR products were then size fractionated in 1% TAE-agarose, extracted using the Qiaquick gel extraction kit (Qiagen) and cloned into the pGEM-T Easy Vector system (Promega). Blue-white selection was applied to eliminate false positives, and twelve random clones were picked and sequenced. Bisulfite conversion efficiency of non-CpG cytosines was >90% for all individual clones for each sample.
[00181] Immunocytochemistry, AP staining and FACS analysis. For in vivo immunostaining, HES2 and iPSC were washed once with DMEM medium supplemented with 10% FBS and antibiotics (DMEM 10%), and incubated with biotin-TRA-1-81 antibody (1 : 100, eBiosciences) for 2 h. Cells were washed three times with DMEM 10% and they were incubated with the secondary antibody streptavidin-FITC (1 : 100, eBiosciences) and the phycoeritrin TRA- 1-60 (PE-TRA-1-60) antibody (1 : 100, eBiosciences) where indicated. All the incubations were performed in a humidified incubator at 37 °C with 5% C02. For intracellular staining, cells were
fixed in 2% paraformaldehyde for 30 min, and blocked and permeabilized in PBS containing 10% donkey serum, 1% BSA and 0.1% Triton X-100 for 45 min. Cells were incubated with primary antibody in blocking solution overnight at 4 °C, washed and incubated with the corresponding Alexa donkey secondary antibody for 1 h at room temperature (RT). Then cells were washed and stained with DAPI (1 μg/ml) for 20 min. The primary antibodies used for intracellular immunostaining were OCT4 (1 : 100, Bio Vision), NANOG (1 : 100, R&D systems), desmin (1 : 100, Lab Vision), a-SMA (pre-diluted, DAKO), vimentin (1 : 100, Chemicon), AFP (1 :500, DAKO), GFAP (1 :1000, DAKO), βΙΙΙ-Tubulin (1 : 100, Chemicon), or NFATc4 (1 : 100, Santa Cruz Biotechnology). All the secondary antibodies Alexa 488 Donkey anti-Rabbit (1 : 100), Alexa 546 donkey anti-goat (1 : 100) and Alexa 546 donkey anti-mouse (1 : 100) were obtained from Invitrogen. Alkaline phosphatase staining was detected following manufacturer's recommendations (Millipore). SSEA-4, troponin T, and hematopoietic markers expression were evaluated on a BD Biosciences LSRII FACS machine analyzer. Primary antibodies SSEA-4-PE (R&D systems), cardiac troponin T (Lab Vision), CD1 lb-APC (Caltag), and CD45-APC, CD45- PE, CD71-PE, CD41-PE and CD235a-APC were purchased from BD Biosciences.
[00182] Teratoma formation. All animal procedures were performed in accordance with the Mount Sinai Medical Center's Institutional Animal Care and Use Committee.
Approximately l-2xl06 cells were injected subcutaneously into the right hindleg of immunocompromised NOD-SCID mice (The Jackson Laboratory). Teratomas were excised 6-10 weeks post-injection, fixed overnight in formalin, embedded in paraffin, sectioned and stained with hematoxylin and eosin by the Morphology and Assessment Core of the Department of Gene and Cell Medicine. Histological evaluation was performed using a Nikon TE2000-U microscope and ACT-1 software.
[00183] In vitro differentiation. For non-lineage specific and hematopoietic differentiation the inventors used previously described protocols, with certain modifications (Takahashi, K., et al, Cell 131, 861-872 (2007); Dimos, J.T., et al, Science 321, 1218-1221 (2008), unpublished data, Kennedy M. and Keller G. et al). For cardiomyocytes induction, the inventors used a well-established assay (Yang, L., et al, Nature 453, 524-528 (2008)). For embryoid body (EB) formation, HES2 and iPSC were treated with collagenase B (Roche) for 10 min, and collected by scraping. After centrifuging, cell pellets were resuspended in basic
differentiation media, StemPro 34 (Invitrogen) containing 2 mM L-Gln, 4x10"
monothioglycerol (MTG), 50 μg/ml ascorbic acid (Sigma) and 150 μg/ml transferrin (Sigma). EBs were grown in ultra low-binding plates (Costar) and medium was changed every three days. After eight days of differentiation, EBs were collected, resuspended in DMEM 10% and transferred to gelatin-coated dishes to allow them to attach and differentiate for eight additional days before processing for immunocytochemistry analyses. For hematopoietic differentiation the inventors used a described protocol (Kennedy, M., et al, Blood 109, 2679-2687 (2007)) with certain modifications (unpublished data, Kennedy M. and Keller G. et al.). For cardiomyocyte induction, the inventors used a well-established protocol (Yang, L., et al, Nature 453, 524-528 (2008)).
[00184] Microarray analysis. Gene level mRNA abundance measures were extracted using the Affymetrix GeneChip Exon 1.0 ST array according to the manufacturer's protocols by the Genomics Core Lab in The Institute for Personalized Medicine at Mount Sinai Medical Center. Microarrays were scanned and data were Robust Multi- Array (RMA)-normalized using the Affymetrix Expression Console software. Subsequently, these genes were clustered and a heat map was generated against a background subset of genes showing at least two-fold change between sample averages of iPSC/HES2 cells and fibroblast samples.
[00185] Cytology/cardiomyocyte size determination. On D18 of differentiation, beating EBs were plated on gelatin coated dishes. Three days after plating, EBs outgrowths were trypsinized, filtered through a 40 μιη size pore-size filter, and single cells were replated at low density on gelatin coated dishes. The following day, cells were fixed with 4% paraformaldehyde, permeablized, blocked in PBS/1% BSA/0.1%> Triton/10%) donkey serum, and Stained for cardiac Troponin T (1 :200, Lab Vision), overnight at 4°C. Stained cells were washed three times with PBS, and then incubated with the Alexa Fluor 547 donkey-anti-mouse antibody (Invitrogen) for 1 h. The areas of HESC- and LS-iPSC-derived cardiomyocytes were analyzed using ImageJ software (NIH).
[00186] Phosphoproteomics and Western blotting. HES2 and iPSC were incubated overnight in HESC culture medium deprived of bFGF and knockout serum replacement (KSR). Protein lysates were quantified by Bradford assay and sent to Kinexus Bioinformatics
Corporation (Vancouver, Canada) for antibody microarray screening. The proteins with at least 1.5 fold change between the LS-iPSC samples and control sample (either HES2 or wt-iPSC), and conserved in the majority of the comparison groups were represented in a heat map.
[00187] Specifically, the inventors prepared a lysis buffer (pH 7.2) containing 20 mM MOPS pH 7.0, 2 mM EGTA, 5 mM EDTA, 30 mM sodium fluoride, 60 mM β- glycerophosphate, 20 mM sodium phyrophospate and 1% Triton X-100. Protease and phosphatase inhibitors (1 mM phenylmethylsulfonyl fluoride, 3 mM benzamidine, 10 μg/ml aprotinin, 10 μΜ leupeptin, 5 μΜ pepstatin, ImM dithiothreitol and 1 mM sodium
ortho vanadate) were added to the lysis buffer immediately before use. Protein extracts were sent to Kinexus Bioinformatics Corporation (Vancouver, Canada). The antibody microarray results were processed following the company recommendations. Western blot was carried out as previously described (Carvajal-Vergara, X., et al, Blood 105, 4492-4499 (2005)). The primary antibodies used were: pS6 S235/236 (1 : 1000, Cell Signaling), pEGFR Y1086 (1 :1000, Cell Signaling), pMEKl S298 (1 : 1000, Cell Signaling), β-Actin (1 :5000, Abeam), p-ER l/2
T202/Y204 (1 :2000 Cell Signaling) and ER 1 (1 :2500, Santa Cruz Biotechnology).
EXAMPLE 2: PATIENT-SPECIFIC INDUCED PLURIPOTENT STEM CELL DERIVED FROM MODELS OF LEOPARD SYNDROME
[00188] Approximately 90% of LS cases, and 45% of NS, are caused by missense mutations in the PTPN11 gene that encodes the protein tyrosine phosphatase SHP2. PTPN11 is ubiquitously expressed, essential for normal development, and somatic mutations in this gene contribute to leukemogenesis in children (Loh, M.L., et al, Blood 103, 2325-2331 (2004), Tartaglia, M., et al, Blood 104, 307-313 (2004)). For LS, two mutations, T468M and Y279C, are most recurrent (Tartaglia, M., et al., Am J Hum Genet 78, 279-290 (2006)). Hypertrophic cardiomyopathy is the most common life-threatening cardiac anomaly in LS (Sarkozy, A., et al., Orphanet J Rare Dis 3, 13 (2008)). Animal models of LS have been generated in Drosophila and zebrafish (Jopling, C, et al, PLoS Genet 3, e225 (2007); Oishi, K., et al, Hum Mol Genet (2008)), but the molecular pathogenesis of LS remains obscure.
[00189] Ectopic expression of four transcription factors (OCT4, SOX2, KLF4 and c- MYC) in adult human dermal fibroblasts can generate pluripotent iPSC (Lowry, W.E., et al., Proc
Natl Acad Sci U S A 105, 2883-2888 (2008), Park, I.H., et al, Cell 134, 877-886 (2008), Takahashi, K., et al., Cell 131, 861-872 (2007)). The inventors have established iPSC lines from two LS patients, a 25-year-old female (LI), and a 34-year-old male (L2). A heterozygous T468M substitution mutation in PTPN11 is present in both.
[00190] Patient-derived fibroblasts were transduced with OCT4-, SOX2-, KLF4- and c-MYC-encoding VSV-pseudotyped Moloney-based retroviral vectors. Granular morphology and compact ESC-like colonies emerged during the ensuing two weeks, distinguishable by TRA- 1-81 staining (Fig. 5), as described previously (Lowry, W.E., et al, Proc Natl Acad Sci U S A 105, 2883-2888 (2008)). TRA- 1-81 -positive colonies were clonally expanded to create stable LS-iPSC lines. Three iPSC lines per patient were used for preliminary characterization: Ll- iPSl, Ll-iPS6, Ll-iPS13, L2-iPS6, L2-iPS16 and L2-iPS18.
[00191] To verify that the iPSC originated from patient-derived fibroblasts, the inventors performed DNA fingerprinting analysis (Fig. 6a). All iPSC had normal karyotypes of 46,XX (LI) and 46,XY (L2) (Fig. 6b and data not shown). In addition, they carried the expected T468M mutation (Fig. 7a). Restriction fragment length polymorphism analysis of an RT-PCR amplimer containing the mutation with BsmFI showed biallelic expression of PTPN11 (Fig. 7b). PCR and Southern blots indicated the presence of all four transgene proviruses in the LS-iPSC (Fig. 8) and quantitative RT-PCR (qRT-PCR) results confirmed efficient transgene silencing (Fig. 9).
[00192] To further characterize the LS-iPSC clones, expression of several HESC markers in two LS-iPSC lines from each patient (Ll-iPSl, Ll-iPS13, L2-iPS6 and L2-iPS16) was analyzed and compared to the HES2 HESC and a wt-iPSC line, BJ-iPSB5, derived in the lab from a normal human fibroblast line (BJ). The BJ-iPSB5 cell line was also karyotypically normal (46,XY), contained all four transgene proviruses, which were silenced (Fig. 6b, 8b and 9b). All LS and control iPSC lines exhibited high alkaline phosphatase activity, and expressed
pluripotency markers, including surface antigens TRA- 1-81, TRA- 1-60, and SSEA-4, as well as the nuclear transcription factors OCT4 and NANOG (Fig. 10). Quantitative RT-PCR (RT-qPCR) confirmed the activation of a series of endogenous sternness genes (OCT4, NANOG, SOX2, GDF3, DPPA4, REXl and TERT) in iPSC (Fig. la and Fig. 11a). Moreover, bisulfite
sequencing analyses determined that the great majority of the CpG dinucleotides analyzed in the OCT4 and NANOG promoters were demethylated in iPSC when compared to their parental fibroblasts (Fig. lb).
[00193] The inventors next examined genome-wide mR A expression profiles of two LS-iPSC lines from each patient, the BJ-iPSB5 cell line, parental fibroblasts and HES2 cells. The resulting heat map and scatter-plot analyses indicated that iPSC lines shared a higher degree of similarity with HES2 cells than with their parental fibroblast cell lines (Fig. lc and Fig. 1 lb).
[00194] Pluripotent HESC can differentiate into cell types representative of all three germ layers. The inventors tested the differentiation abilities of the iPSC using an in vitro 8-day floating embryoid body (EBs) system, followed by replating on gelatin-coated dishes for another 8 days (Takahashi, K., et al, Cell 131, 861-872 (2007); Dimos, J.T., et al, Science 321, 1218- 1221 (2008)). Immunocytochemistry analyses detected expression of a-smooth muscle actin (a- SMA, mesoderm), desmin (mesoderm), a-fetoprotein (AFP, endoderm), vimentin (mesoderm), glial fibrillary acidic protein (GFAP, ectoderm) and βΙΙΙ-tubulin (ectoderm) markers (Fig. 2a and Fig. 12). In order to determine pluripotency in vivo, the inventors injected LS-iPS, BJ-iPSB5 and HES2 cells into immune-compromised NOD-SCID mice. Histological analyses of the resulting teratomas showed cell types representative of the three germ layers, including pigmented cells (ectoderm), lung, respiratory and gut-like epithelia (endoderm), and
mesenchyme, adipose tissue and cartilage (mesoderm) (Fig. 2b and data not shown).
[00195] As mentioned previously, hypertrophic cardiomyopathy is one of the major features of LS, affecting 80% of the patients. In addition, affected individuals occasionally manifest hematologic complications such as myelodysplasia and leukemia (Laux, D. et al., J Pediatr Hematol Oncol 30, 602-604 (2008); Ucar, C, et al, J Pediatr Hematol Oncol 28, 123-125 (2006)). Therefore, the inventors decided to explore if LS-iPSC were able to differentiate into hematopoietic and cardiac lineages. LS-iPSC from both patients differentiated into a variety of hematopoietic cell types including early hematopoietic progenitors (CD41+) (Mikkola, H.K., et al, Blood 101, 508-516 (2003)), early erythroblasts (CD71+/CD235a+) (Wu, C.J., et al, Blood 106, 3639-3645 (2005)), and macrophages (CD1 lb+) (Fan, S.T., et al, J Clin Invest 87, 50-57 (1991)) (Fig. 13 and data not shown). The cardiac hypertrophic response includes induction of
immediate-early genes (such as c-jun, c-fos and c-myc), an increase in cell size, and organization of contractile proteins into sarcomeric units (Aoki, H., et al, Nat Med 6, 183-188 (2000);
Buitrago, M., et al., Nat Med 11, 837-844 (2005)). To have an appropriate control cell line to analyze some of these parameters, besides HESC, the inventors generated a wt-iPSC line (S3- iPS4) from fibroblasts obtained from an unaffected brother of LI without the T468M mutation (Fig. 14 and Fig. 15). Using a well-established cardiac differentiation protocol (Yang, L., et al, Nature 453, 524-528 (2008)), the inventors observed contracting embryoid bodies (EBs) emerging around day 11 of differentiation. In order to monitor cardiac development, the inventors analyzed cardiac troponin T (cTNT^ expression on day 18 of differentiation by flow cytometry (data not shown). Replated cells from beating EBs were processed as described in Material and Methods. Briefly, cells were fixed, immunostained for cTNT (Fig. 3b), and 50 cardiomyocytes were randomly chosen from each sample for surface area measurement using a computerized morphometric system (ImageJ software, NIH). Cardiomyocytes derived from LS- iPSC lines: Ll-iPS13, Ll-iPS6 and L2-iPS10 (Fig. 14 and Fig. 15), had a significantly increased median surface area compared to wt-iPSC cardiomyocytes; 1.8 times, 2.5 times and 4.8 times larger, respectively, whereas the area median of the cardiomyocytes obtained from HESC was similar to wt-iPSC cardiomyocytes (Fig. 3a). The inventors also observed increased sarcomere assembly in Ll-iPS6 and L2-iPS10 cells when compared to wt S3-iPS4 cells (Fig. 3b). Recently, the calcineurin-NFAT pathway has been shown to be an important regulator of cardiac hypertrophy. Active calcineurin dephosphorylates NFAT transcription factors, resulting in their nuclear translocation (Buitrago, M., et al, Nat Med 11, 837-844 (2005); Molkentin, J.D.,
Cardiovasc Res 63, 467-475 (2004)). The inventors analyzed the localization of NFAT c4 using immunocytochemistry in 50 cTNT -positive cardiomyocytes derived from the L2-iPS10 cell line, which produced the largest cardiomyocytes, and wt S3-iPS4. The inventors observed a significantly higher proportion of LS cardiomyocytes with nuclear NFATc4, (-80% versus -30%, respectively) (Fig. 3c and 3d).
EXAMPLE 3: MUTATIONS IN THE PTPN11 GENE PERTURBS RAS-MAPK SIGNAL
TRANSDUCTION IN PLURIPOTENT STEM CELLS.
[00196] In order to identify potential molecular targets that could be affected by the T468M PTPN11 mutation, protein extracts from LS-iPS, wt BJ-iPSB5 and HES2 cells were
analyzed using a phosphoproteomic microarray chip containing approximately 600 pan and phospho-specific antibodies (Kinexus Bioinformatics Corporation). The inventors established eight groups for comparison, each of the LS-iPSC lines versus one control cell line, either HES2 or wt-iPSC. Proteins with a 1.5 fold change were filtered, and those that were conserved in most of the groups were represented in a heat map (Fig. 4a). Some of the proteins were more abundantly present in LS-iPS when compared to either HES2 (Tyro 10, Tyk2 and Haspin) or wt- iPSC (p-MARCKs, p-Synapsinl, p-NMDAR2B, p-MSKl, p-RSKl/3 and p-p53). The phosphorylation of other proteins was increased (p-Caveolin2, p-MEKl, p-EGFR and p-FAK) or decreased (p-Vinculin, p-S6 and p-Lck) in LS-iPSC when compared to control cell lines. In order to eliminate false positives, the inventors verified the phosphoproteomic results by Western blot (WB) for three of the most altered proteins (p-S6, p-EGFR and p-MEKl) in four LS-iPSC lines, in comparison to wt-iPSC. While the inventors did not confirm a major change in the phosphorylation status of S6 protein (data not shown), WB confirmed that the phosphorylation of EGFR and MEK1 proteins was considerably increased in the LS-iPSC samples (Fig. 4b).
[00197] RAS-MAPK represents the major signaling pathway deregulated by SHP2 mutants. NS mutants increase basal and stimulated phosphatase activity, whereas LS mutants are catalytically impaired and have dominant-negative effects, inhibiting growth factor-evoked ERK1/2 activation (Kontaridis, M.I., J Biol Chem 281, 6785-6792 (2006)). The inventors analyzed the ability of LS-iPSC to respond to external growth factors. The inventors used bFGF (basic Fibroblast Growth Factor), the main growth factor in the maintenance of HESC, to induce the stimulation of the MAPK signaling pathway. bFGF treatment increased the phosphorylation of ERK1/2 (p-ERK) levels over time in HES2 and wt S3-iPS4 cells (Fig. 4c). Although the LS- iPSC expressed the four FGF receptor (FGFR) family members (Fig. 16a), bFGF stimulation did not cause any substantial change in p-ERK levels (Fig. 4c and Figs. 16b- 16c). However, the LS- iPSC lines had higher basal p-ERK levels compared to HES2 and S3-iPS4 cells (Figs. 16b- 16c), in accordance with the increased pMEKl, ERK upstream kinase, levels found in LS-iPSC samples in phosphoproteomic array results.
[00198] In summary, the inventors have generated and characterized LS patient- specific iPSC, providing a new system for the study of disease pathogenesis. The inventors observed increases in cell size, sarcomeric organization and nuclear NFATc4 localization in LS-
iPSC-derived cardiomyocytes, when compared to HESC and wt-iPSC-derived cardiomyocytes. These results are consistent with cardiac hypertrophy, a condition commonly found in LS patients (Sarkozy, A. et al., Orphanet J Rare Dis 3, 13 (2008)), and indicate that this abnormality occurs through a cell autonomous mechanism due to the PTPN11 mutation. Since many human cell types, such as cardiomyocytes, cannot be propagated readily in cell culture, iPS-derived cells exhibiting disease-relevant phenotypes provide the requisite resource for precisely elucidating pathogenesis and pursuing novel therapeutic strategies.
[00199] In the studies described herein, the inventors provided insights into the molecular events that are affected by the PTPN11 mutation in the pluripotent iPSC using antibody microarrays. The inventors found that the phosphorylation of certain proteins was increased in LS-iPSC when compared to wild-type HESC and iPSC. Interestingly, one of the more upregulated phosphoproteins was MEK1, the upstream kinase of ERK1/2, whose gene is sometimes mutated in the related disorder, cardiofaciocutaneous syndrome. PTPN11 mutations underlie 45% and 90% of NS and LS, respectively. It is not well understood how mutations that provoke opposite effects on SHP2 phosphatase activity cause syndromes with similar features (Edouard, T., et al, Cell Mol Life Sci 64, 1585-1590 (2007)). In concordance with observations in the Drosophila LS model (Oishi, K., et al, Hum Mol Genet (2008)), basal p-ERK levels were increased in LS-iPSC. Of note, receptor tyrosine kinase stimulation with bFGF in LS-iPSC failed to elicit further activation of ERK, as previously observed in a different cellular model
(Kontaridis, ML, et al, J Biol Chem 281, 6785-6792 (2006)). Interestingly, this result demonstrates for the first time that RAS-MAPK signal transduction is perturbed in LS as early as the pluripotent stem cell stage.
[00200] Taken together, this is the first described human model of an inherited RAS pathway disorder.
REFERENCES
[00201] 1. Gorlin, R. J., Anderson, R.C. & Moller, J.H. The Leopard (multiple lentigines) syndrome revisited. Birth Defects Orig Artie Ser 07, 110-115 (1971).
[00202] 2. Sarkozy, A., Digilio, M.C. & Dallapiccola, B. Leopard syndrome.
Orphanet J Rare Dis 3, 13 (2008).
[00203] 3. Loh, M.L., et al. Mutations in PTPNl 1 implicate the SHP-2 phosphatase in leukemogenesis. Blood 103, 2325-2331 (2004).
[00204] 4. Tartaglia, M., et al. Genetic evidence for lineage-related and
differentiation stage-related contribution of somatic PTPNl 1 mutations to leukemogenesis in childhood acute leukemia. Blood 104, 307-313 (2004).
[00205] 5. Tartaglia, M., et al. Diversity and functional consequences of germline and somatic PTPNl 1 mutations in human disease. Am J Hum Genet 78, 279-290 (2006).
[00206] 6. Jopling, C, van Geemen, D. & den Hertog, J. Shp2 knockdown and Noonan/LEOPARD mutant Shp2-induced gastrulation defects. PLoS Genet 3, e225 (2007).
[00207] 7. Oishi, K., et al. Phosphatase-defective LEOPARD syndrome mutations in PTPNl 1 have gain-of- function effects during Drosophila development. Hum Mol Genet (2008).
[00208] 8. Lowry, W.E., et al. Generation of human induced pluripotent stem cells from dermal fibroblasts. Proc Natl Acad Sci USA 105, 2883-2888 (2008).
[00209] 9. Park, I.H., et al. Disease-specific induced pluripotent stem cells. Cell 134, 877-886 (2008).
[00210] 10. Takahashi, K., et al. Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell 131, 861-872 (2007).
[00211] 11. Ebert, A.D., et al. Induced pluripotent stem cells from a spinal muscular atrophy patient. Nature (2008).
[00212] 12. Lee, G., et al. Modelling pathogenesis and treatment of familial dysautonomia using patient-specific iPSCs. Nature 461, 402-406 (2009).
[00213] 13. Raya, A et al. Disease-corrected haematopoietic progenitors from Fanconi anaemia induced pluripotent stem cells. Nature 460, 53-59 (2009).
[00214] 14. Ye, Z et al. Human induced pluripotent stem cells from blood cells of healthy donors and patients with acquired blood disorders. Blood (2009).
[00215] 15. Dimos, J.T., et al. Induced pluripotent stem cells generated from patients with ALS can be differentiated into motor neurons. Science 321, 1218-1221 (2008).
[00216] 16. Laux, D., Kratz, C. & Sauerbrey, A. Common acute lymphoblastic leukemia in a girl with genetically confirmed LEOPARD syndrome. J Pediatr Hematol Oncol 30, 602-604 (2008).
[00217] 17. Ucar, C, Calyskan, U., Martini, S. & Heinritz, W. Acute myelomonocytic leukemia in a boy with LEOPARD syndrome (PTPN11 gene mutation positive). J Pediatr Hematol Oncol 28, 123-125 (2006).
[00218] 18. Mikkola, H.K., Fujiwara, Y., Schlaeger, T.M., Traver, D. & Orkin, S.H. Expression of CD41 marks the initiation of definitive hematopoiesis in the mouse embryo. Blood 101, 508-516 (2003).
[00219] 19. Wu, C.J., et al. Evidence for ineffective erythropoiesis in severe sickle cell disease. Blood 106, 3639-3645 (2005).
[00220] 20. Fan, S.T. & Edgington, T.S. Coupling of the adhesive receptor
CD 1 lb/CD 18 to functional enhancement of effector macrophage tissue factor response. J Clin Invest 87, 50-57 (1991).
[00221] 21. Aoki, FL, Sadoshima, J. & Izumo, S. Myosin light chain kinase mediates sarcomere organization during cardiac hypertrophy in vitro. Nat Med 6, 183-188 (2000).
[00222] 22. Buitrago, M., et al. The transcriptional repressor Nabl is a specific regulator of pathological cardiac hypertrophy. Nat Med 11, 837-844 (2005).
[00223] 23. Yang, L., et al. Human cardiovascular progenitor cells develop from a KDR+ embryonic-stem-cell-derived population. Nature 453, 524-528 (2008).
[00224] 24. Molkentin, J.D. Calcineurin-NFAT signaling regulates the cardiac hypertrophic response in coordination with the MAPKs. Cardiovasc Res 63, 467-475 (2004).
[00225] 25. Kontaridis, ML, Swanson, K.D., David, F.S., Barford, D. & Neel, B.G. PTPN11 (Shp2) mutations in LEOPARD syndrome have dominant negative, not activating, effects. J Biol Chem 281, 6785-6792 (2006).
[00226] 26. Edouard, T., et al. How do Shp2 mutations that oppositely influence its biochemical activity result in syndromes with overlapping symptoms? Cell Mol Life Sci 64, 1585-1590 (2007).
[00227] 27. Dvorak, P., et al. Expression and potential role of fibroblast growth factor 2 and its receptors in human embryonic stem cells. Stem Cells 23, 1200-1211 (2005).
[00228] 28. Freberg, C.T., Dahl, J.A., Timoskainen, S. & Collas, P. Epigenetic reprogramming of OCT4 and NANOG regulatory regions by embryonal carcinoma cell extract. Mol Biol Cell 18, 1543-1553 (2007).
[00229] 29. Kennedy, M., D'Souza, S.L., Lynch-Kattman, M., Schwantz, S. & Keller, G. Development of the hemangioblast defines the onset of hematopoiesis in human ES cell differentiation cultures. Blood 109, 2679-2687 (2007).
[00230] 30. Carvajal-Vergara, X., et al. Multifunctional role of Erk5 in multiple myeloma. Blood 105, 4492-4499 (2005).
[00231] 31. Carvajal-Vergara, X., et al., International Society for Stem Cell Research 7th Annual Meeting, Thursday Poster Session Abstract, p 150, Poster Section I. (July 9, 2009).
[00232] 32. Tartaglia et al, Nature Genetics, Jun;34(2), 2003: 148-50
[00233] The specification is most thoroughly understood in light of the teachings of the references cited within the specification. The embodiments within the specification provide an illustration of embodiments of the invention and should not be construed to limit the scope of the invention. The skilled artisan readily recognizes that many other embodiments are encompassed by the invention. All publications and patents cited in this disclosure are
incorporated by reference in their entirety. To the extent the material incorporated by reference contradicts or is inconsistent with this specification, the specification will supersede any such material. The citation of any references herein is not an admission that such references are prior art to the present invention.
[00234] Those skilled in the art will recognize, or be able to ascertain using no more than routine experimentation, many equivalents to the specific embodiments of the invention described herein. Such equivalents are intended to be encompassed by the following embodiments.
Claims
An isolated primate pluripotent stem cell derived from a somatic cell, wherein said stem cell comprises a mutation in the protein tyrosine phosphatase non-receptor type 11 (PTPNl 1) gene.
The stem cell of claim 1, wherein said PTPNl 1 gene comprises a mutation associated with a cardiac anomaly.
The stem cell of claim 2, wherein said cardiac anomaly is hypertrophic cardiomyopathy.
The stem cell of claim 1, wherein said PTPNl 1 gene comprises a mutation associated with LEOPARD Syndrome, Noonan Syndrome or leukemia.
The stem cell of any one of claims 1-4, wherein the mutation is a deletion mutation, a substitution mutation, an addition mutation, or a combination thereof, in the coding region of said PTPNl 1 gene.
The stem cell of any one of claims 1-5, wherein the mutation in said PTPNl 1 gene results in a mutation in the encoded PTPNl 1 protein.
The stem cell of claim 6, wherein the mutation is a threonine to methionine substitution in the encoded PTPNl 1 protein at an amino acid residue position corresponding to position 468 of SEQ ID NO: 1, or a tyrosine to cysteine substitution in the encoded PTPNl 1 protein at an amino acid residue position corresponding to position 279 of SEQ ID NO: 1.
The stem cell of claim 7, wherein the encoded protein comprises SEQ ID NO: 1 and the mutation is a threonine to methionine substitution at amino acid residue position 468 of SEQ ID NO: l, or a tyrosine to cysteine substitution at amino acid residue position 279 of SEQ ID NO: l .
The stem cell of claim 6, wherein the mutation is selected from the group consisting of a tyrosine to cysteine substitution in the encoded PTPNl 1 protein at an amino acid residue position corresponding to position 279 of SEQ ID NO: 1, a tyrosine to serine substitution
in the encoded PTPNl 1 protein at an amino acid residue position corresponding to position 279 of SEQ ID NO: 1, an alanine to threonine substitution in the encoded PTPNl 1 protein at an amino acid residue position corresponding to position 461 of SEQ ID NO: 1, a glycine to alanine substitution in the encoded PTPNl 1 protein at an amino acid residue position corresponding to position 464 of SEQ ID NO: 1, a threonine to methionine substitution in the encoded PTPNl 1 protein at an amino acid residue position corresponding to position 468 of SEQ ID NO: 1, a threonine to proline substitution in the encoded PTPNl 1 protein at an amino acid residue position corresponding to position 468 of SEQ ID NO: 1, an arginine to tryptophan substitution in the encoded PTPNl 1 protein at an amino acid residue position corresponding to position 498 of SEQ ID NO: 1, an arginine to leucine substitution in the encoded PTPNl 1 protein at an amino acid residue position corresponding to position 498 of SEQ ID NO: 1, a glutamine to proline substitution in the encoded PTPNl 1 protein at an amino acid residue position
corresponding to position 506 of SEQ ID NO: 1, a glutamine to proline substitution in the encoded PTPNl 1 protein at an amino acid residue position corresponding to position 510 of SEQ ID NO:l, a glutamine to glutamic acid substitution in the encoded PTPNl 1 protein at an amino acid residue position corresponding to position 510 of SEQ ID NO: l, and a glutamine to glycine substitution in the encoded PTPNl 1 protein at an amino acid residue position corresponding to position 510 of SEQ ID NO: 1.
10. The stem cell of any one of claims 1-9, wherein the cell forms a teratoma in an
immunocompromised mouse.
11. The stem cell of any one of claims 1-10, wherein the cell expresses SSEA-3, SSEA-4, TRA-1-60, TRA-1-81, Nanog, OCT-4, Sox2, GDF3, DPPA4, REX1, TERT, Alkaline Phosphatase, Telomerase, or CD30, or a combination thereof.
12. The stem cell of any one of claims 1-11, wherein the stem cell is capable of
differentiating into a cardiomyocyte.
13. A cell culture of the stem cell of any one of claims 1-12.
14. The stem cell of claim 1 deposited at the American Type Culture Collection having ATCC Accession No. .
15. A cardiomyocyte comprising a mutation in the PTPN11 gene, wherein the cardiomyocyte is produced by differentiating the stem cell of any one of claims 1-12.
16. A cell culture of the cardiomyocyte of claim 15.
17. The cardiomyocyte of claim 15 deposited at the American Type Culture Collection
having ATCC Accession No. .
18. The stem cell of any one of claims 1-12, wherein the primate is human.
19. The stem cell of any one of claims 1-12, wherein the somatic cell is a fibroblast.
20. A method of identifying an agent for use in treatment of a disease that is associated with a mutation in the PTPN11 gene, comprising: contacting the pluripotent stem cell of any one of claims 1-12 with the agent; and determining the effect of the agent on survival, proliferation, phenotype, or differentiation of the pluripotent stem cell.
21. A method of identifying an agent for use in treatment of a disease that is associated with a mutation in the PTPN11 gene, comprising: contacting the cardiomyocyte of claim 15 with the agent; and determining the effect of the agent on survival, proliferation, or phenotype of the cardiomyocyte.
22. A method of identifying an agent for use in treatment of the cardiac hypertrophy that is associated with a mutation in the PTPN11 gene, comprising: contacting the cardiomyocyte of claim 15 with the agent; and
determining the effect of the agent on the hypertrophic state of the cardiomyocyte, wherein a decrease in the hypertrophic state of the cardiomyocyte, as compared to that of a control cell, is indicative that the agent is an agent for use in treatment of a disease that is associated with a mutation in the PTPN11 gene.
23. A method of producing a pluripotent stem cell of any one of claims 1-12, comprising:
(a) obtaining a somatic cell from a donor whose genome comprises a mutation in the PTPN11 gene;
(b) introducing into the somatic cell: (i) a nucleic acid molecule that encodes a pluripotency-inducing protein; or (ii) a polypeptide that comprises a pluripotency- inducing protein; wherein said pluripotency-inducing protein is Oct3/4, Sox2, Klf4, Nanog, Lin28, c-Myc, or a combination thereof.
24. The method of claim 23, wherein one or more nucleic acid molecules that encode Oct3/4, Sox2, Klf4, and c-Myc are introduced into the somatic cell.
25. The methods of claims 23 or 24, wherein the somatic cell is a fibroblast.
26. A method of differentiating a pluripotent stem cell of any one of claims 1-12 into a
cardiomyocyte, comprising: culturing the stem cell in a serum- free medium that comprises: activin A, bone morphogenetic protein 4 (BMP4), basic fibroblast growth factor (bFGF), vascular endothelial growth factor (VEGF) and dickkopf homo log 1 (DK 1).
27. A method of identifying an agent as a candidate Ras-signaling pathway inhibitor,
comprising: contacting the pluripotent stem cell of any one of claims 1-12 with the agent; and determining the effect of the agent on tyrosine phosphorylation of EGFR or MEK1; wherein a decrease of tyrosine phosphorylation of EGFR or MEK1, as compared to that of a control cell, is indicative that the agent is a Ras-signaling pathway inhibitor.
28. A method of identifying an agent as a candidate Ras-signaling pathway inhibitor, comprising: contacting the cardiomyocyte of claim 15 with the agent; and determining the effect of the agent on tyrosine phosphorylation of EGFR or MEKl; wherein a decrease of tyrosine phosphorylation of EGFR or MEKl, as compared to that of a control cell, is indicative that the agent is a Ras-signaling pathway inhibitor.
29. A method of identifying an agent for use in treatment of a disease that is associated with a mutation in the PTPN11 gene, comprising: contacting the pluripotent stem cell of any one of claims 1-12 with the agent; differentiating the stem cell into a cardiomyocyte; and determining the hypertrophic state of the cardiomyocyte, wherein a decrease in the hypertrophic state of the cardiomyocyte, as compared to that of a control cell, is indicative that the agent is an agent for use in treatment of a disease that is associated with a mutation in the PTPN11 gene.
30. The stem cell of any one of claims 1-11, wherein the stem cell is capable of
differentiating into a hematopoietic cell.
31. A hematopoietic cell comprising a mutation in the PTPN11 gene, wherein the
hematopoietic cell is produced by differentiating the stem cell of any one of claims 1-11 or 30.
32. A cell culture of the hematopoietic cell of claim 31.
33. The hematopoietic cell of claim 31 deposited at the American Type Culture Collection having ATCC Accession No. .
34. A method of identifying an agent for use in treatment of a disease that is associated with a mutation in the PTPN11 gene, comprising:
contacting the pluripotent stem cell of any one of claims 1-11 or 30 with the agent; differentiating the stem cell into a hematopoietic cell; and determining the effect of the agent on survival, proliferation, phenotype, or differentiation of the hematopoietic cell.
35. The method of claim 34, wherein the disease is leukemia.
36. A method of identifying an agent for use in treatment of leukemia, comprising: contacting the pluripotent stem cell of any one of claims 1-11 or 30 with the agent; differentiating the stem cell into a hematopoietic cell; and determining the proliferative state of the hematopoietic cell, wherein a decrease in the proliferative state of the hematopoietic cell, as compared to that of a control cell, is indicative that the agent is an agent for use in treatment of leukemia.
Applications Claiming Priority (4)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
US32415010P | 2010-04-14 | 2010-04-14 | |
US61/324,150 | 2010-04-14 | ||
US32981310P | 2010-04-30 | 2010-04-30 | |
US61/329,813 | 2010-04-30 |
Publications (1)
Publication Number | Publication Date |
---|---|
WO2011130217A1 true WO2011130217A1 (en) | 2011-10-20 |
Family
ID=44798983
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/US2011/032044 WO2011130217A1 (en) | 2010-04-14 | 2011-04-12 | Induced pluripotent stem cells and uses thereof |
Country Status (1)
Country | Link |
---|---|
WO (1) | WO2011130217A1 (en) |
Cited By (3)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
KR101490867B1 (en) * | 2013-12-11 | 2015-02-09 | 한국과학기술원 | cardiofaciocutaneous syndrome induced pluripotent stem cell model and use thereof |
WO2015115690A1 (en) * | 2014-01-28 | 2015-08-06 | 한국과학기술원 | Induced pluripotent stem cell model of noonan syndrome and use thereof |
WO2021213479A1 (en) * | 2020-04-22 | 2021-10-28 | 复星凯特生物科技有限公司 | Shp2 specific inactivating mutant protein and application thereof in car-t therapy |
Citations (1)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO2009061442A1 (en) * | 2007-11-06 | 2009-05-14 | Children's Medical Center Corporation | Method to produce induced pluripotent stem (ips) cells form non-embryonic human cells |
-
2011
- 2011-04-12 WO PCT/US2011/032044 patent/WO2011130217A1/en active Application Filing
Patent Citations (1)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO2009061442A1 (en) * | 2007-11-06 | 2009-05-14 | Children's Medical Center Corporation | Method to produce induced pluripotent stem (ips) cells form non-embryonic human cells |
Non-Patent Citations (2)
Title |
---|
EBERT ET AL.: "Induced pluripotent stem cells from a spinal muscular atrophy patient.", NATURE, vol. 457, no. 7227, 15 January 2009 (2009-01-15), pages 277 - 280 * |
EDOUARD ET AL.: "Functional effects of PTPN11 (SHP2) mutations causing LEOPARD syndrome on epidermal growth factor-induced phosphoinositide 3-kinase/AKT/glycogen synthase kinase 3beta signaling.", MOL CELL BIOL, vol. 30, no. 10, May 2010 (2010-05-01), pages 2498 - 2507 * |
Cited By (6)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
KR101490867B1 (en) * | 2013-12-11 | 2015-02-09 | 한국과학기술원 | cardiofaciocutaneous syndrome induced pluripotent stem cell model and use thereof |
WO2015088073A1 (en) * | 2013-12-11 | 2015-06-18 | 한국과학기술원 | Induced pluripotent stem cell model for cardiofaciocutaneous syndrome and uses thereof |
US20160355788A1 (en) * | 2013-12-11 | 2016-12-08 | Korea Advanced Institute Of Science And Technology | Induced Pluripotent Stem Cell Model for Cardiofaciocutaneous Syndrome and Uses Thereof |
US10174288B2 (en) | 2013-12-11 | 2019-01-08 | Korea Advanced Institute Of Science And Technology | Induced pluripotent stem cell model for cardiofaciocutaneous syndrome and uses thereof |
WO2015115690A1 (en) * | 2014-01-28 | 2015-08-06 | 한국과학기술원 | Induced pluripotent stem cell model of noonan syndrome and use thereof |
WO2021213479A1 (en) * | 2020-04-22 | 2021-10-28 | 复星凯特生物科技有限公司 | Shp2 specific inactivating mutant protein and application thereof in car-t therapy |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
US20230282445A1 (en) | Method of nuclear reprogramming | |
JP5553289B2 (en) | New nuclear initialization material | |
JP5547064B2 (en) | Multipotent / pluripotent cells and methods | |
AU2014224119B2 (en) | Wnt pathway stimulation in reprogramming somatic cells | |
JP7089298B2 (en) | How to induce differentiation from pluripotent stem cells to germ cells | |
CN102782122B (en) | Method of efficiently establishing induced pluripotent stem cells | |
JP5936134B2 (en) | Selection method of human induced pluripotent stem cells | |
JP6029137B2 (en) | Efficient method for establishing induced pluripotent stem cells | |
JP5888753B2 (en) | Efficient method for establishing induced pluripotent stem cells | |
EP2980207B1 (en) | Cell sorting method | |
US10077429B2 (en) | Method of efficiently establishing induced pluripotent stem cells | |
WO2019107576A1 (en) | Maintenance-and-amplification method and differentiation induction method for primordial germ cells/primordial germ cell—like cells | |
US20110033936A1 (en) | CANINE iPS CELLS AND METHOD OF PRODUCING SAME | |
US20100303775A1 (en) | Generation of Genetically Corrected Disease-free Induced Pluripotent Stem Cells | |
JP2011522520A (en) | Methods for cell dedifferentiation | |
EP3481944A1 (en) | Means and methods for the generation of oligodendrocytes | |
US20140024119A1 (en) | Cell reprogramming composition comprising rex1 and an induced pluripotent stem cell production method using the same | |
JP5773393B2 (en) | Efficient method for establishing induced pluripotent stem cells | |
WO2011130217A1 (en) | Induced pluripotent stem cells and uses thereof | |
WO2015178496A1 (en) | Method for producing lung progenitor cell | |
WO2017126616A1 (en) | Cell model for ewing's sarcoma family of tumors, and antitumor agent screening method using same | |
WO2012141181A1 (en) | Nuclear reprogramming substance | |
Salazar-Roa et al. | A novel microRNA-based strategy to expand the differentiation potency of stem cells | |
US9145547B2 (en) | Nuclear reprogrammed cells generated by introduction of a histone H2aa or TH2A gene, a histone H2ba or TH2B gene, or a phosphorylation-mimic of histone chaperon Npm2 gene, an Oct family gene and a klf family gene into a mammalian somatic cell |
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: 11769408 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: 11769408 Country of ref document: EP Kind code of ref document: A1 |