WO2010116665A1 - 単核球由来の新規血管再生細胞群及びその分化誘導法 - Google Patents
単核球由来の新規血管再生細胞群及びその分化誘導法 Download PDFInfo
- Publication number
- WO2010116665A1 WO2010116665A1 PCT/JP2010/002254 JP2010002254W WO2010116665A1 WO 2010116665 A1 WO2010116665 A1 WO 2010116665A1 JP 2010002254 W JP2010002254 W JP 2010002254W WO 2010116665 A1 WO2010116665 A1 WO 2010116665A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- cells
- cell
- cd11b
- cell group
- medium
- Prior art date
Links
- 210000004027 cell Anatomy 0.000 title claims abstract description 309
- 210000005087 mononuclear cell Anatomy 0.000 title claims abstract description 87
- 238000000034 method Methods 0.000 title claims abstract description 31
- 230000000250 revascularization Effects 0.000 title claims description 18
- 230000004069 differentiation Effects 0.000 title abstract description 100
- 230000001939 inductive effect Effects 0.000 title abstract description 31
- 108010073929 Vascular Endothelial Growth Factor A Proteins 0.000 claims abstract description 75
- 108010019530 Vascular Endothelial Growth Factors Proteins 0.000 claims abstract description 73
- 210000004204 blood vessel Anatomy 0.000 claims abstract description 61
- 102000003974 Fibroblast growth factor 2 Human genes 0.000 claims abstract description 54
- 108090000379 Fibroblast growth factor 2 Proteins 0.000 claims abstract description 54
- 102000036693 Thrombopoietin Human genes 0.000 claims abstract description 48
- 108010041111 Thrombopoietin Proteins 0.000 claims abstract description 48
- 108010017080 Granulocyte Colony-Stimulating Factor Proteins 0.000 claims abstract description 30
- 102000004269 Granulocyte Colony-Stimulating Factor Human genes 0.000 claims abstract description 30
- 108700014844 flt3 ligand Proteins 0.000 claims abstract description 22
- 239000012679 serum free medium Substances 0.000 claims abstract description 22
- 230000035800 maturation Effects 0.000 claims abstract description 18
- 230000006641 stabilisation Effects 0.000 claims abstract description 18
- 238000011105 stabilization Methods 0.000 claims abstract description 18
- 238000012258 culturing Methods 0.000 claims abstract description 16
- 102000005789 Vascular Endothelial Growth Factors Human genes 0.000 claims abstract 8
- 101001046686 Homo sapiens Integrin alpha-M Proteins 0.000 claims abstract 3
- 102100022338 Integrin alpha-M Human genes 0.000 claims abstract 3
- 239000002609 medium Substances 0.000 claims description 108
- 206010028980 Neoplasm Diseases 0.000 claims description 45
- 210000001185 bone marrow Anatomy 0.000 claims description 29
- 238000002360 preparation method Methods 0.000 claims description 28
- 201000011510 cancer Diseases 0.000 claims description 27
- 229910052760 oxygen Inorganic materials 0.000 claims description 26
- 102100024616 Platelet endothelial cell adhesion molecule Human genes 0.000 claims description 22
- 230000000302 ischemic effect Effects 0.000 claims description 22
- 210000005259 peripheral blood Anatomy 0.000 claims description 22
- 239000011886 peripheral blood Substances 0.000 claims description 22
- QVGXLLKOCUKJST-UHFFFAOYSA-N atomic oxygen Chemical compound [O] QVGXLLKOCUKJST-UHFFFAOYSA-N 0.000 claims description 21
- 239000001301 oxygen Substances 0.000 claims description 21
- 102100031650 C-X-C chemokine receptor type 4 Human genes 0.000 claims description 20
- 101000922348 Homo sapiens C-X-C chemokine receptor type 4 Proteins 0.000 claims description 17
- 206010021143 Hypoxia Diseases 0.000 claims description 17
- 230000001146 hypoxic effect Effects 0.000 claims description 17
- 230000000694 effects Effects 0.000 claims description 16
- 230000008929 regeneration Effects 0.000 claims description 11
- 238000011069 regeneration method Methods 0.000 claims description 11
- 210000003714 granulocyte Anatomy 0.000 claims description 7
- 230000004807 localization Effects 0.000 claims description 6
- 210000004700 fetal blood Anatomy 0.000 claims description 5
- 230000006872 improvement Effects 0.000 claims description 4
- 239000000032 diagnostic agent Substances 0.000 claims description 3
- 229940039227 diagnostic agent Drugs 0.000 claims description 3
- 230000000638 stimulation Effects 0.000 claims 1
- 208000028867 ischemia Diseases 0.000 abstract description 14
- 230000008439 repair process Effects 0.000 abstract description 4
- 239000001963 growth medium Substances 0.000 abstract description 2
- 102100039037 Vascular endothelial growth factor A Human genes 0.000 description 67
- 230000006698 induction Effects 0.000 description 63
- 210000003819 peripheral blood mononuclear cell Anatomy 0.000 description 55
- 230000014509 gene expression Effects 0.000 description 37
- 230000001464 adherent effect Effects 0.000 description 32
- 210000002966 serum Anatomy 0.000 description 25
- 101000946889 Homo sapiens Monocyte differentiation antigen CD14 Proteins 0.000 description 22
- 102100035877 Monocyte differentiation antigen CD14 Human genes 0.000 description 22
- 239000012091 fetal bovine serum Substances 0.000 description 21
- 210000000130 stem cell Anatomy 0.000 description 19
- 210000003556 vascular endothelial cell Anatomy 0.000 description 19
- 241000699666 Mus <mouse, genus> Species 0.000 description 18
- 230000033115 angiogenesis Effects 0.000 description 16
- 239000011324 bead Substances 0.000 description 16
- 238000002054 transplantation Methods 0.000 description 15
- 230000002792 vascular Effects 0.000 description 15
- 210000003141 lower extremity Anatomy 0.000 description 14
- 210000001616 monocyte Anatomy 0.000 description 14
- 230000001965 increasing effect Effects 0.000 description 13
- 102000004127 Cytokines Human genes 0.000 description 12
- 108090000695 Cytokines Proteins 0.000 description 12
- 201000002818 limb ischemia Diseases 0.000 description 12
- 210000004698 lymphocyte Anatomy 0.000 description 12
- 210000001519 tissue Anatomy 0.000 description 12
- 239000000427 antigen Substances 0.000 description 11
- 102000036639 antigens Human genes 0.000 description 11
- 108091007433 antigens Proteins 0.000 description 11
- 238000000684 flow cytometry Methods 0.000 description 11
- 210000003958 hematopoietic stem cell Anatomy 0.000 description 10
- 208000034578 Multiple myelomas Diseases 0.000 description 9
- 206010035226 Plasma cell myeloma Diseases 0.000 description 9
- 230000015572 biosynthetic process Effects 0.000 description 9
- 230000006870 function Effects 0.000 description 9
- 239000002523 lectin Substances 0.000 description 9
- 230000001737 promoting effect Effects 0.000 description 9
- 102000005962 receptors Human genes 0.000 description 9
- 108020003175 receptors Proteins 0.000 description 9
- 108010022164 acetyl-LDL Proteins 0.000 description 8
- 210000003414 extremity Anatomy 0.000 description 8
- 239000003550 marker Substances 0.000 description 8
- 108010082117 matrigel Proteins 0.000 description 8
- 102100037241 Endoglin Human genes 0.000 description 7
- 102100031573 Hematopoietic progenitor cell antigen CD34 Human genes 0.000 description 7
- 101000881679 Homo sapiens Endoglin Proteins 0.000 description 7
- 101001027128 Homo sapiens Fibronectin Proteins 0.000 description 7
- 101000777663 Homo sapiens Hematopoietic progenitor cell antigen CD34 Proteins 0.000 description 7
- 102000008790 VE-cadherin Human genes 0.000 description 7
- 108010018828 cadherin 5 Proteins 0.000 description 7
- 239000003814 drug Substances 0.000 description 7
- 230000001976 improved effect Effects 0.000 description 7
- 238000003757 reverse transcription PCR Methods 0.000 description 7
- 108010054017 Granulocyte Colony-Stimulating Factor Receptors Proteins 0.000 description 6
- 102100039622 Granulocyte colony-stimulating factor receptor Human genes 0.000 description 6
- 241001465754 Metazoa Species 0.000 description 6
- 241000699670 Mus sp. Species 0.000 description 6
- DNIAPMSPPWPWGF-UHFFFAOYSA-N Propylene glycol Chemical compound CC(O)CO DNIAPMSPPWPWGF-UHFFFAOYSA-N 0.000 description 6
- 102000016971 Proto-Oncogene Proteins c-kit Human genes 0.000 description 6
- 108010014608 Proto-Oncogene Proteins c-kit Proteins 0.000 description 6
- 108091008605 VEGF receptors Proteins 0.000 description 6
- 102000009484 Vascular Endothelial Growth Factor Receptors Human genes 0.000 description 6
- 238000002512 chemotherapy Methods 0.000 description 6
- 201000010099 disease Diseases 0.000 description 6
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 description 6
- 238000001727 in vivo Methods 0.000 description 6
- 238000011580 nude mouse model Methods 0.000 description 6
- 230000035755 proliferation Effects 0.000 description 6
- 101100481408 Danio rerio tie2 gene Proteins 0.000 description 5
- 101100481410 Mus musculus Tek gene Proteins 0.000 description 5
- -1 PlGF-1 Proteins 0.000 description 5
- 230000024245 cell differentiation Effects 0.000 description 5
- 238000010195 expression analysis Methods 0.000 description 5
- 239000003102 growth factor Substances 0.000 description 5
- 210000002540 macrophage Anatomy 0.000 description 5
- 210000003668 pericyte Anatomy 0.000 description 5
- 108010053099 Vascular Endothelial Growth Factor Receptor-2 Proteins 0.000 description 4
- 230000002491 angiogenic effect Effects 0.000 description 4
- 230000017531 blood circulation Effects 0.000 description 4
- 210000002798 bone marrow cell Anatomy 0.000 description 4
- 230000003394 haemopoietic effect Effects 0.000 description 4
- 238000012744 immunostaining Methods 0.000 description 4
- 208000015181 infectious disease Diseases 0.000 description 4
- 238000001990 intravenous administration Methods 0.000 description 4
- 239000003446 ligand Substances 0.000 description 4
- 229920000729 poly(L-lysine) polymer Polymers 0.000 description 4
- 238000011476 stem cell transplantation Methods 0.000 description 4
- 230000004083 survival effect Effects 0.000 description 4
- 238000012360 testing method Methods 0.000 description 4
- 238000002560 therapeutic procedure Methods 0.000 description 4
- 230000017423 tissue regeneration Effects 0.000 description 4
- 206010003210 Arteriosclerosis Diseases 0.000 description 3
- 101710082513 C-X-C chemokine receptor type 4 Proteins 0.000 description 3
- FBPFZTCFMRRESA-KVTDHHQDSA-N D-Mannitol Chemical compound OC[C@@H](O)[C@@H](O)[C@H](O)[C@H](O)CO FBPFZTCFMRRESA-KVTDHHQDSA-N 0.000 description 3
- LFQSCWFLJHTTHZ-UHFFFAOYSA-N Ethanol Chemical compound CCO LFQSCWFLJHTTHZ-UHFFFAOYSA-N 0.000 description 3
- 101000808011 Homo sapiens Vascular endothelial growth factor A Proteins 0.000 description 3
- 241000699660 Mus musculus Species 0.000 description 3
- 206010028851 Necrosis Diseases 0.000 description 3
- 206010061902 Pancreatic neoplasm Diseases 0.000 description 3
- 102000016549 Vascular Endothelial Growth Factor Receptor-2 Human genes 0.000 description 3
- 230000002159 abnormal effect Effects 0.000 description 3
- 239000002870 angiogenesis inducing agent Substances 0.000 description 3
- 208000011775 arteriosclerosis disease Diseases 0.000 description 3
- 210000004369 blood Anatomy 0.000 description 3
- 239000008280 blood Substances 0.000 description 3
- 230000021164 cell adhesion Effects 0.000 description 3
- 238000004113 cell culture Methods 0.000 description 3
- 230000004087 circulation Effects 0.000 description 3
- 238000007796 conventional method Methods 0.000 description 3
- 230000003247 decreasing effect Effects 0.000 description 3
- 210000004443 dendritic cell Anatomy 0.000 description 3
- 229940079593 drug Drugs 0.000 description 3
- 210000002889 endothelial cell Anatomy 0.000 description 3
- 230000003511 endothelial effect Effects 0.000 description 3
- 102000058223 human VEGFA Human genes 0.000 description 3
- 238000000338 in vitro Methods 0.000 description 3
- 208000015486 malignant pancreatic neoplasm Diseases 0.000 description 3
- 108020004999 messenger RNA Proteins 0.000 description 3
- 210000004088 microvessel Anatomy 0.000 description 3
- 210000000822 natural killer cell Anatomy 0.000 description 3
- 230000017074 necrotic cell death Effects 0.000 description 3
- 238000012758 nuclear staining Methods 0.000 description 3
- 201000002528 pancreatic cancer Diseases 0.000 description 3
- 208000008443 pancreatic carcinoma Diseases 0.000 description 3
- 230000008569 process Effects 0.000 description 3
- 230000001172 regenerating effect Effects 0.000 description 3
- 238000000926 separation method Methods 0.000 description 3
- 230000000087 stabilizing effect Effects 0.000 description 3
- 108091032973 (ribonucleotides)n+m Proteins 0.000 description 2
- 102000007469 Actins Human genes 0.000 description 2
- 108010085238 Actins Proteins 0.000 description 2
- 102100022014 Angiopoietin-1 receptor Human genes 0.000 description 2
- 101710131689 Angiopoietin-1 receptor Proteins 0.000 description 2
- IJGRMHOSHXDMSA-UHFFFAOYSA-N Atomic nitrogen Chemical compound N#N IJGRMHOSHXDMSA-UHFFFAOYSA-N 0.000 description 2
- 102100022595 Broad substrate specificity ATP-binding cassette transporter ABCG2 Human genes 0.000 description 2
- 102000004354 CD11b Antigen Human genes 0.000 description 2
- 108010017009 CD11b Antigen Proteins 0.000 description 2
- 102100028757 Chondroitin sulfate proteoglycan 4 Human genes 0.000 description 2
- FBPFZTCFMRRESA-FSIIMWSLSA-N D-Glucitol Natural products OC[C@H](O)[C@H](O)[C@@H](O)[C@H](O)CO FBPFZTCFMRRESA-FSIIMWSLSA-N 0.000 description 2
- FBPFZTCFMRRESA-JGWLITMVSA-N D-glucitol Chemical compound OC[C@H](O)[C@@H](O)[C@H](O)[C@H](O)CO FBPFZTCFMRRESA-JGWLITMVSA-N 0.000 description 2
- WQZGKKKJIJFFOK-GASJEMHNSA-N Glucose Natural products OC[C@H]1OC(O)[C@H](O)[C@@H](O)[C@@H]1O WQZGKKKJIJFFOK-GASJEMHNSA-N 0.000 description 2
- PEDCQBHIVMGVHV-UHFFFAOYSA-N Glycerine Chemical compound OCC(O)CO PEDCQBHIVMGVHV-UHFFFAOYSA-N 0.000 description 2
- 102000007651 Macrophage Colony-Stimulating Factor Human genes 0.000 description 2
- 108010046938 Macrophage Colony-Stimulating Factor Proteins 0.000 description 2
- 108010090306 Member 2 Subfamily G ATP Binding Cassette Transporter Proteins 0.000 description 2
- 206010027476 Metastases Diseases 0.000 description 2
- 206010029113 Neovascularisation Diseases 0.000 description 2
- 229930040373 Paraformaldehyde Natural products 0.000 description 2
- 108010069381 Platelet Endothelial Cell Adhesion Molecule-1 Proteins 0.000 description 2
- 108010038512 Platelet-Derived Growth Factor Proteins 0.000 description 2
- 102000010780 Platelet-Derived Growth Factor Human genes 0.000 description 2
- 239000002202 Polyethylene glycol Substances 0.000 description 2
- FAPWRFPIFSIZLT-UHFFFAOYSA-M Sodium chloride Chemical compound [Na+].[Cl-] FAPWRFPIFSIZLT-UHFFFAOYSA-M 0.000 description 2
- 108010073919 Vascular Endothelial Growth Factor D Proteins 0.000 description 2
- 102100038234 Vascular endothelial growth factor D Human genes 0.000 description 2
- 108010031318 Vitronectin Proteins 0.000 description 2
- 102100035140 Vitronectin Human genes 0.000 description 2
- 230000004913 activation Effects 0.000 description 2
- 210000000601 blood cell Anatomy 0.000 description 2
- 239000000872 buffer Substances 0.000 description 2
- 210000000882 cd133-positive hematopoietic stem cell Anatomy 0.000 description 2
- 239000002771 cell marker Substances 0.000 description 2
- 210000000170 cell membrane Anatomy 0.000 description 2
- 230000003833 cell viability Effects 0.000 description 2
- 230000001684 chronic effect Effects 0.000 description 2
- 230000005757 colony formation Effects 0.000 description 2
- 238000000432 density-gradient centrifugation Methods 0.000 description 2
- 206010012601 diabetes mellitus Diseases 0.000 description 2
- 230000002708 enhancing effect Effects 0.000 description 2
- 238000011156 evaluation Methods 0.000 description 2
- 239000012530 fluid Substances 0.000 description 2
- 238000002073 fluorescence micrograph Methods 0.000 description 2
- 239000007850 fluorescent dye Substances 0.000 description 2
- 239000008103 glucose Substances 0.000 description 2
- 230000001771 impaired effect Effects 0.000 description 2
- 239000007924 injection Substances 0.000 description 2
- 238000002347 injection Methods 0.000 description 2
- 238000002372 labelling Methods 0.000 description 2
- 239000007788 liquid Substances 0.000 description 2
- 238000012423 maintenance Methods 0.000 description 2
- 235000010355 mannitol Nutrition 0.000 description 2
- 238000004519 manufacturing process Methods 0.000 description 2
- 239000000463 material Substances 0.000 description 2
- 210000003593 megakaryocyte Anatomy 0.000 description 2
- 230000009401 metastasis Effects 0.000 description 2
- 239000000203 mixture Substances 0.000 description 2
- 210000002464 muscle smooth vascular Anatomy 0.000 description 2
- 208000010125 myocardial infarction Diseases 0.000 description 2
- 208000031225 myocardial ischemia Diseases 0.000 description 2
- 210000004967 non-hematopoietic stem cell Anatomy 0.000 description 2
- 238000010899 nucleation Methods 0.000 description 2
- GLDOVTGHNKAZLK-UHFFFAOYSA-N octadecan-1-ol Chemical compound CCCCCCCCCCCCCCCCCCO GLDOVTGHNKAZLK-UHFFFAOYSA-N 0.000 description 2
- 239000012188 paraffin wax Substances 0.000 description 2
- 229920002866 paraformaldehyde Polymers 0.000 description 2
- 230000002093 peripheral effect Effects 0.000 description 2
- 239000002504 physiological saline solution Substances 0.000 description 2
- 229920001223 polyethylene glycol Polymers 0.000 description 2
- 235000010482 polyoxyethylene sorbitan monooleate Nutrition 0.000 description 2
- 229920000053 polysorbate 80 Polymers 0.000 description 2
- 108090000765 processed proteins & peptides Proteins 0.000 description 2
- 230000002062 proliferating effect Effects 0.000 description 2
- 235000018102 proteins Nutrition 0.000 description 2
- 102000004169 proteins and genes Human genes 0.000 description 2
- 108090000623 proteins and genes Proteins 0.000 description 2
- 238000011084 recovery Methods 0.000 description 2
- 239000000523 sample Substances 0.000 description 2
- 229960002920 sorbitol Drugs 0.000 description 2
- 239000000725 suspension Substances 0.000 description 2
- 230000008961 swelling Effects 0.000 description 2
- KIUKXJAPPMFGSW-DNGZLQJQSA-N (2S,3S,4S,5R,6R)-6-[(2S,3R,4R,5S,6R)-3-Acetamido-2-[(2S,3S,4R,5R,6R)-6-[(2R,3R,4R,5S,6R)-3-acetamido-2,5-dihydroxy-6-(hydroxymethyl)oxan-4-yl]oxy-2-carboxy-4,5-dihydroxyoxan-3-yl]oxy-5-hydroxy-6-(hydroxymethyl)oxan-4-yl]oxy-3,4,5-trihydroxyoxane-2-carboxylic acid Chemical compound CC(=O)N[C@H]1[C@H](O)O[C@H](CO)[C@@H](O)[C@@H]1O[C@H]1[C@H](O)[C@@H](O)[C@H](O[C@H]2[C@@H]([C@@H](O[C@H]3[C@@H]([C@@H](O)[C@H](O)[C@H](O3)C(O)=O)O)[C@H](O)[C@@H](CO)O2)NC(C)=O)[C@@H](C(O)=O)O1 KIUKXJAPPMFGSW-DNGZLQJQSA-N 0.000 description 1
- IXPNQXFRVYWDDI-UHFFFAOYSA-N 1-methyl-2,4-dioxo-1,3-diazinane-5-carboximidamide Chemical compound CN1CC(C(N)=N)C(=O)NC1=O IXPNQXFRVYWDDI-UHFFFAOYSA-N 0.000 description 1
- 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 1
- 102100033350 ATP-dependent translocase ABCB1 Human genes 0.000 description 1
- 244000215068 Acacia senegal Species 0.000 description 1
- 229920001817 Agar Polymers 0.000 description 1
- GUBGYTABKSRVRQ-XLOQQCSPSA-N Alpha-Lactose Chemical compound O[C@@H]1[C@@H](O)[C@@H](O)[C@@H](CO)O[C@H]1O[C@@H]1[C@@H](CO)O[C@H](O)[C@H](O)[C@H]1O GUBGYTABKSRVRQ-XLOQQCSPSA-N 0.000 description 1
- 206010002383 Angina Pectoris Diseases 0.000 description 1
- 102000009088 Angiopoietin-1 Human genes 0.000 description 1
- 108010048154 Angiopoietin-1 Proteins 0.000 description 1
- 102000009075 Angiopoietin-2 Human genes 0.000 description 1
- 108010048036 Angiopoietin-2 Proteins 0.000 description 1
- 108090000672 Annexin A5 Proteins 0.000 description 1
- 102000004121 Annexin A5 Human genes 0.000 description 1
- 208000032467 Aplastic anaemia Diseases 0.000 description 1
- 238000011729 BALB/c nude mouse Methods 0.000 description 1
- 108091003079 Bovine Serum Albumin Proteins 0.000 description 1
- OKTJSMMVPCPJKN-UHFFFAOYSA-N Carbon Chemical compound [C] OKTJSMMVPCPJKN-UHFFFAOYSA-N 0.000 description 1
- 102100023126 Cell surface glycoprotein MUC18 Human genes 0.000 description 1
- 206010009900 Colitis ulcerative Diseases 0.000 description 1
- 108010035532 Collagen Proteins 0.000 description 1
- 102000008186 Collagen Human genes 0.000 description 1
- 102000007644 Colony-Stimulating Factors Human genes 0.000 description 1
- 108010071942 Colony-Stimulating Factors Proteins 0.000 description 1
- 235000005956 Cosmos caudatus Nutrition 0.000 description 1
- 208000011231 Crohn disease Diseases 0.000 description 1
- WQZGKKKJIJFFOK-QTVWNMPRSA-N D-mannopyranose Chemical compound OC[C@H]1OC(O)[C@@H](O)[C@@H](O)[C@@H]1O WQZGKKKJIJFFOK-QTVWNMPRSA-N 0.000 description 1
- 229920002307 Dextran Polymers 0.000 description 1
- 208000032131 Diabetic Neuropathies Diseases 0.000 description 1
- 206010059866 Drug resistance 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
- 108010041308 Endothelial Growth Factors Proteins 0.000 description 1
- 102000009024 Epidermal Growth Factor Human genes 0.000 description 1
- 239000001856 Ethyl cellulose Substances 0.000 description 1
- ZZSNKZQZMQGXPY-UHFFFAOYSA-N Ethyl cellulose Chemical compound CCOCC1OC(OC)C(OCC)C(OCC)C1OC1C(O)C(O)C(OC)C(CO)O1 ZZSNKZQZMQGXPY-UHFFFAOYSA-N 0.000 description 1
- 108010037362 Extracellular Matrix Proteins Proteins 0.000 description 1
- 102000010834 Extracellular Matrix Proteins Human genes 0.000 description 1
- 102100020715 Fms-related tyrosine kinase 3 ligand protein Human genes 0.000 description 1
- 101710162577 Fms-related tyrosine kinase 3 ligand protein Proteins 0.000 description 1
- 108010010803 Gelatin Proteins 0.000 description 1
- 102000003886 Glycoproteins Human genes 0.000 description 1
- 108090000288 Glycoproteins Proteins 0.000 description 1
- 244000060234 Gmelina philippensis Species 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
- 229920000084 Gum arabic Polymers 0.000 description 1
- 206010019280 Heart failures Diseases 0.000 description 1
- HTTJABKRGRZYRN-UHFFFAOYSA-N Heparin Chemical compound OC1C(NC(=O)C)C(O)OC(COS(O)(=O)=O)C1OC1C(OS(O)(=O)=O)C(O)C(OC2C(C(OS(O)(=O)=O)C(OC3C(C(O)C(O)C(O3)C(O)=O)OS(O)(=O)=O)C(CO)O2)NS(O)(=O)=O)C(C(O)=O)O1 HTTJABKRGRZYRN-UHFFFAOYSA-N 0.000 description 1
- 241000282412 Homo Species 0.000 description 1
- 101000746367 Homo sapiens Granulocyte colony-stimulating factor Proteins 0.000 description 1
- 101000599951 Homo sapiens Insulin-like growth factor I Proteins 0.000 description 1
- 101000595923 Homo sapiens Placenta growth factor Proteins 0.000 description 1
- 101000738771 Homo sapiens Receptor-type tyrosine-protein phosphatase C Proteins 0.000 description 1
- 101001052849 Homo sapiens Tyrosine-protein kinase Fer Proteins 0.000 description 1
- 208000031226 Hyperlipidaemia Diseases 0.000 description 1
- 206010020772 Hypertension Diseases 0.000 description 1
- 208000022559 Inflammatory bowel disease Diseases 0.000 description 1
- 102100037852 Insulin-like growth factor I Human genes 0.000 description 1
- GUBGYTABKSRVRQ-QKKXKWKRSA-N Lactose Natural products OC[C@H]1O[C@@H](O[C@H]2[C@H](O)[C@@H](O)C(O)O[C@@H]2CO)[C@H](O)[C@@H](O)[C@H]1O GUBGYTABKSRVRQ-QKKXKWKRSA-N 0.000 description 1
- 102000004856 Lectins Human genes 0.000 description 1
- 108090001090 Lectins Proteins 0.000 description 1
- 229930195725 Mannitol Natural products 0.000 description 1
- 108010047230 Member 1 Subfamily B ATP Binding Cassette Transporter Proteins 0.000 description 1
- 101100335081 Mus musculus Flt3 gene Proteins 0.000 description 1
- 101001055320 Myxine glutinosa Insulin-like growth factor Proteins 0.000 description 1
- 208000008469 Peptic Ulcer Diseases 0.000 description 1
- 239000004264 Petrolatum Substances 0.000 description 1
- 102100035194 Placenta growth factor Human genes 0.000 description 1
- 102100040681 Platelet-derived growth factor C Human genes 0.000 description 1
- 229920001213 Polysorbate 20 Polymers 0.000 description 1
- 239000004372 Polyvinyl alcohol Substances 0.000 description 1
- 208000004210 Pressure Ulcer Diseases 0.000 description 1
- 101710098940 Pro-epidermal growth factor Proteins 0.000 description 1
- 239000012979 RPMI medium Substances 0.000 description 1
- 238000010240 RT-PCR analysis Methods 0.000 description 1
- 102100037422 Receptor-type tyrosine-protein phosphatase C Human genes 0.000 description 1
- 208000017442 Retinal disease Diseases 0.000 description 1
- 206010038923 Retinopathy Diseases 0.000 description 1
- 206010039580 Scar Diseases 0.000 description 1
- 206010040943 Skin Ulcer Diseases 0.000 description 1
- 229920002125 Sokalan® Polymers 0.000 description 1
- 229920002472 Starch Polymers 0.000 description 1
- 235000021355 Stearic acid Nutrition 0.000 description 1
- 102100021669 Stromal cell-derived factor 1 Human genes 0.000 description 1
- 101710088580 Stromal cell-derived factor 1 Proteins 0.000 description 1
- 210000001744 T-lymphocyte Anatomy 0.000 description 1
- 206010043540 Thromboangiitis obliterans Diseases 0.000 description 1
- 102100024537 Tyrosine-protein kinase Fer Human genes 0.000 description 1
- 201000006704 Ulcerative Colitis Diseases 0.000 description 1
- 108010073925 Vascular Endothelial Growth Factor B Proteins 0.000 description 1
- 108010073923 Vascular Endothelial Growth Factor C Proteins 0.000 description 1
- 102100038217 Vascular endothelial growth factor B Human genes 0.000 description 1
- 102100038232 Vascular endothelial growth factor C Human genes 0.000 description 1
- 206010052428 Wound Diseases 0.000 description 1
- 208000027418 Wounds and injury Diseases 0.000 description 1
- 230000005856 abnormality Effects 0.000 description 1
- 235000010489 acacia gum Nutrition 0.000 description 1
- 239000000205 acacia gum Substances 0.000 description 1
- 230000001133 acceleration Effects 0.000 description 1
- DPXJVFZANSGRMM-UHFFFAOYSA-N acetic acid;2,3,4,5,6-pentahydroxyhexanal;sodium Chemical compound [Na].CC(O)=O.OCC(O)C(O)C(O)C(O)C=O DPXJVFZANSGRMM-UHFFFAOYSA-N 0.000 description 1
- 230000009471 action Effects 0.000 description 1
- 239000000654 additive Substances 0.000 description 1
- 239000002671 adjuvant Substances 0.000 description 1
- 239000008272 agar Substances 0.000 description 1
- 235000010419 agar Nutrition 0.000 description 1
- 230000032683 aging Effects 0.000 description 1
- 238000004378 air conditioning Methods 0.000 description 1
- 238000004458 analytical method Methods 0.000 description 1
- 239000002246 antineoplastic agent Substances 0.000 description 1
- 229940041181 antineoplastic drug Drugs 0.000 description 1
- 230000003078 antioxidant effect Effects 0.000 description 1
- 239000007864 aqueous solution Substances 0.000 description 1
- 230000027746 artery morphogenesis Effects 0.000 description 1
- 239000012752 auxiliary agent Substances 0.000 description 1
- 229920002988 biodegradable polymer Polymers 0.000 description 1
- 239000004621 biodegradable polymer Substances 0.000 description 1
- 230000023555 blood coagulation Effects 0.000 description 1
- 239000007853 buffer solution Substances 0.000 description 1
- 229910052799 carbon Inorganic materials 0.000 description 1
- 239000001768 carboxy methyl cellulose Substances 0.000 description 1
- 239000005018 casein Substances 0.000 description 1
- BECPQYXYKAMYBN-UHFFFAOYSA-N casein, tech. Chemical compound NCCCCC(C(O)=O)N=C(O)C(CC(O)=O)N=C(O)C(CCC(O)=N)N=C(O)C(CC(C)C)N=C(O)C(CCC(O)=O)N=C(O)C(CC(O)=O)N=C(O)C(CCC(O)=O)N=C(O)C(C(C)O)N=C(O)C(CCC(O)=N)N=C(O)C(CCC(O)=N)N=C(O)C(CCC(O)=N)N=C(O)C(CCC(O)=O)N=C(O)C(CCC(O)=O)N=C(O)C(COP(O)(O)=O)N=C(O)C(CCC(O)=N)N=C(O)C(N)CC1=CC=CC=C1 BECPQYXYKAMYBN-UHFFFAOYSA-N 0.000 description 1
- 235000021240 caseins Nutrition 0.000 description 1
- 230000032677 cell aging Effects 0.000 description 1
- 230000032823 cell division Effects 0.000 description 1
- 230000003915 cell function Effects 0.000 description 1
- 230000012292 cell migration Effects 0.000 description 1
- 239000006285 cell suspension Substances 0.000 description 1
- 238000002659 cell therapy Methods 0.000 description 1
- 239000001913 cellulose Substances 0.000 description 1
- 229920002678 cellulose Polymers 0.000 description 1
- 206010008118 cerebral infarction Diseases 0.000 description 1
- 208000026106 cerebrovascular disease Diseases 0.000 description 1
- 239000003153 chemical reaction reagent Substances 0.000 description 1
- 239000003795 chemical substances by application Substances 0.000 description 1
- 208000019425 cirrhosis of liver Diseases 0.000 description 1
- 230000003749 cleanliness Effects 0.000 description 1
- 229920001436 collagen Polymers 0.000 description 1
- 230000001332 colony forming effect Effects 0.000 description 1
- 210000002808 connective tissue Anatomy 0.000 description 1
- 230000016396 cytokine production Effects 0.000 description 1
- 230000003013 cytotoxicity Effects 0.000 description 1
- 231100000135 cytotoxicity Toxicity 0.000 description 1
- 230000001419 dependent effect Effects 0.000 description 1
- 238000001514 detection method Methods 0.000 description 1
- 238000011161 development Methods 0.000 description 1
- 230000018109 developmental process Effects 0.000 description 1
- 238000003745 diagnosis Methods 0.000 description 1
- 229940105990 diglycerin Drugs 0.000 description 1
- GPLRAVKSCUXZTP-UHFFFAOYSA-N diglycerol Chemical compound OCC(O)COCC(O)CO GPLRAVKSCUXZTP-UHFFFAOYSA-N 0.000 description 1
- 239000002552 dosage form Substances 0.000 description 1
- 239000003937 drug carrier Substances 0.000 description 1
- 230000000042 effect on ischemia Effects 0.000 description 1
- 210000001671 embryonic stem cell Anatomy 0.000 description 1
- 239000003995 emulsifying agent Substances 0.000 description 1
- 210000003989 endothelium vascular Anatomy 0.000 description 1
- 235000019325 ethyl cellulose Nutrition 0.000 description 1
- 229920001249 ethyl cellulose Polymers 0.000 description 1
- 238000002474 experimental method Methods 0.000 description 1
- 210000002744 extracellular matrix Anatomy 0.000 description 1
- 210000001105 femoral artery Anatomy 0.000 description 1
- 230000001605 fetal effect Effects 0.000 description 1
- 238000001215 fluorescent labelling Methods 0.000 description 1
- 229920000159 gelatin Polymers 0.000 description 1
- 239000008273 gelatin Substances 0.000 description 1
- 235000019322 gelatine Nutrition 0.000 description 1
- 235000011852 gelatine desserts Nutrition 0.000 description 1
- SDUQYLNIPVEERB-QPPQHZFASA-N gemcitabine Chemical compound O=C1N=C(N)C=CN1[C@H]1C(F)(F)[C@H](O)[C@@H](CO)O1 SDUQYLNIPVEERB-QPPQHZFASA-N 0.000 description 1
- 229960005144 gemcitabine hydrochloride Drugs 0.000 description 1
- 235000011187 glycerol Nutrition 0.000 description 1
- 230000005484 gravity Effects 0.000 description 1
- 230000004217 heart function Effects 0.000 description 1
- 229920000669 heparin Polymers 0.000 description 1
- 229960002897 heparin Drugs 0.000 description 1
- 229920002674 hyaluronan Polymers 0.000 description 1
- 229960003160 hyaluronic acid Drugs 0.000 description 1
- 210000002865 immune cell Anatomy 0.000 description 1
- 230000028993 immune response Effects 0.000 description 1
- 230000037451 immune surveillance Effects 0.000 description 1
- 238000010348 incorporation Methods 0.000 description 1
- 210000004263 induced pluripotent stem cell Anatomy 0.000 description 1
- 230000008595 infiltration Effects 0.000 description 1
- 238000001764 infiltration Methods 0.000 description 1
- 210000004969 inflammatory cell Anatomy 0.000 description 1
- 208000027866 inflammatory disease Diseases 0.000 description 1
- 239000003112 inhibitor Substances 0.000 description 1
- 229910052500 inorganic mineral Inorganic materials 0.000 description 1
- 238000007918 intramuscular administration Methods 0.000 description 1
- 238000007912 intraperitoneal administration Methods 0.000 description 1
- 238000007914 intraventricular administration Methods 0.000 description 1
- 208000023589 ischemic disease Diseases 0.000 description 1
- 238000002955 isolation Methods 0.000 description 1
- 239000000644 isotonic solution Substances 0.000 description 1
- 239000008101 lactose Substances 0.000 description 1
- 210000002414 leg Anatomy 0.000 description 1
- 210000000265 leukocyte Anatomy 0.000 description 1
- 230000007774 longterm Effects 0.000 description 1
- 238000009593 lumbar puncture Methods 0.000 description 1
- 230000035168 lymphangiogenesis Effects 0.000 description 1
- 210000003563 lymphoid tissue Anatomy 0.000 description 1
- 230000005389 magnetism Effects 0.000 description 1
- 230000005741 malignant process Effects 0.000 description 1
- 239000000594 mannitol Substances 0.000 description 1
- 230000007246 mechanism Effects 0.000 description 1
- 230000010534 mechanism of action Effects 0.000 description 1
- 239000012092 media component Substances 0.000 description 1
- 239000013028 medium composition Substances 0.000 description 1
- 239000012528 membrane Substances 0.000 description 1
- 229920000609 methyl cellulose Polymers 0.000 description 1
- 239000001923 methylcellulose Substances 0.000 description 1
- 235000010981 methylcellulose Nutrition 0.000 description 1
- 210000004925 microvascular endothelial cell Anatomy 0.000 description 1
- 238000013508 migration Methods 0.000 description 1
- 230000001617 migratory effect Effects 0.000 description 1
- 239000011707 mineral Substances 0.000 description 1
- 230000002297 mitogenic effect Effects 0.000 description 1
- 210000002433 mononuclear leukocyte Anatomy 0.000 description 1
- 239000004570 mortar (masonry) Substances 0.000 description 1
- 210000004165 myocardium Anatomy 0.000 description 1
- 210000001087 myotubule Anatomy 0.000 description 1
- GOQYKNQRPGWPLP-UHFFFAOYSA-N n-heptadecyl alcohol Natural products CCCCCCCCCCCCCCCCCO GOQYKNQRPGWPLP-UHFFFAOYSA-N 0.000 description 1
- 239000002105 nanoparticle Substances 0.000 description 1
- 210000005036 nerve Anatomy 0.000 description 1
- 208000004235 neutropenia Diseases 0.000 description 1
- 210000000440 neutrophil Anatomy 0.000 description 1
- 229910052757 nitrogen Inorganic materials 0.000 description 1
- 239000002736 nonionic surfactant Substances 0.000 description 1
- 230000000414 obstructive effect Effects 0.000 description 1
- QIQXTHQIDYTFRH-UHFFFAOYSA-N octadecanoic acid Chemical compound CCCCCCCCCCCCCCCCCC(O)=O QIQXTHQIDYTFRH-UHFFFAOYSA-N 0.000 description 1
- OQCDKBAXFALNLD-UHFFFAOYSA-N octadecanoic acid Natural products CCCCCCCC(C)CCCCCCCCC(O)=O OQCDKBAXFALNLD-UHFFFAOYSA-N 0.000 description 1
- 239000003960 organic solvent Substances 0.000 description 1
- 230000011164 ossification Effects 0.000 description 1
- 230000003076 paracrine Effects 0.000 description 1
- 239000002245 particle Substances 0.000 description 1
- 239000001814 pectin Substances 0.000 description 1
- 235000010987 pectin Nutrition 0.000 description 1
- 229920001277 pectin Polymers 0.000 description 1
- 210000004976 peripheral blood cell Anatomy 0.000 description 1
- 210000001322 periplasm Anatomy 0.000 description 1
- 235000019271 petrolatum Nutrition 0.000 description 1
- 229940066842 petrolatum Drugs 0.000 description 1
- 210000001539 phagocyte Anatomy 0.000 description 1
- 239000000546 pharmaceutical excipient Substances 0.000 description 1
- 108010017992 platelet-derived growth factor C Proteins 0.000 description 1
- 229920000747 poly(lactic acid) Polymers 0.000 description 1
- 229920001495 poly(sodium acrylate) polymer Polymers 0.000 description 1
- 239000004626 polylactic acid Substances 0.000 description 1
- 235000010486 polyoxyethylene sorbitan monolaurate Nutrition 0.000 description 1
- 239000000256 polyoxyethylene sorbitan monolaurate Substances 0.000 description 1
- 239000000244 polyoxyethylene sorbitan monooleate Substances 0.000 description 1
- 229940068968 polysorbate 80 Drugs 0.000 description 1
- 229920002451 polyvinyl alcohol Polymers 0.000 description 1
- 229920000036 polyvinylpyrrolidone Polymers 0.000 description 1
- 239000001267 polyvinylpyrrolidone Substances 0.000 description 1
- 235000013855 polyvinylpyrrolidone Nutrition 0.000 description 1
- 239000002243 precursor Substances 0.000 description 1
- 235000013772 propylene glycol Nutrition 0.000 description 1
- 238000000746 purification Methods 0.000 description 1
- 238000011002 quantification Methods 0.000 description 1
- 238000004445 quantitative analysis Methods 0.000 description 1
- 108091008598 receptor tyrosine kinases Proteins 0.000 description 1
- 102000027426 receptor tyrosine kinases Human genes 0.000 description 1
- 150000003839 salts Chemical class 0.000 description 1
- 230000009758 senescence Effects 0.000 description 1
- 239000004017 serum-free culture medium Substances 0.000 description 1
- 238000004904 shortening Methods 0.000 description 1
- 230000019491 signal transduction Effects 0.000 description 1
- 231100000019 skin ulcer Toxicity 0.000 description 1
- 239000011734 sodium Substances 0.000 description 1
- 229910052708 sodium Inorganic materials 0.000 description 1
- 235000010413 sodium alginate Nutrition 0.000 description 1
- 239000000661 sodium alginate Substances 0.000 description 1
- 229940005550 sodium alginate Drugs 0.000 description 1
- 235000019812 sodium carboxymethyl cellulose Nutrition 0.000 description 1
- 229920001027 sodium carboxymethylcellulose Polymers 0.000 description 1
- 239000011780 sodium chloride Substances 0.000 description 1
- NNMHYFLPFNGQFZ-UHFFFAOYSA-M sodium polyacrylate Chemical compound [Na+].[O-]C(=O)C=C NNMHYFLPFNGQFZ-UHFFFAOYSA-M 0.000 description 1
- 239000000243 solution Substances 0.000 description 1
- 239000002904 solvent Substances 0.000 description 1
- 239000000600 sorbitol Substances 0.000 description 1
- 235000010356 sorbitol Nutrition 0.000 description 1
- 238000001179 sorption measurement Methods 0.000 description 1
- 241000894007 species Species 0.000 description 1
- 239000003381 stabilizer Substances 0.000 description 1
- 238000011272 standard treatment Methods 0.000 description 1
- 239000008107 starch Substances 0.000 description 1
- 235000019698 starch Nutrition 0.000 description 1
- 239000008117 stearic acid Substances 0.000 description 1
- 239000004575 stone Substances 0.000 description 1
- 238000007920 subcutaneous administration Methods 0.000 description 1
- 239000000126 substance Substances 0.000 description 1
- 150000005846 sugar alcohols Polymers 0.000 description 1
- 239000004094 surface-active agent Substances 0.000 description 1
- 238000007910 systemic administration Methods 0.000 description 1
- 229940124597 therapeutic agent Drugs 0.000 description 1
- 230000001225 therapeutic effect Effects 0.000 description 1
- 230000009466 transformation Effects 0.000 description 1
- 230000005747 tumor angiogenesis Effects 0.000 description 1
- 210000004881 tumor cell Anatomy 0.000 description 1
- 210000000689 upper leg Anatomy 0.000 description 1
- 238000010200 validation analysis Methods 0.000 description 1
- 208000019553 vascular disease Diseases 0.000 description 1
- 230000008728 vascular permeability Effects 0.000 description 1
- 239000011782 vitamin Substances 0.000 description 1
- 229940088594 vitamin Drugs 0.000 description 1
- 229930003231 vitamin Natural products 0.000 description 1
- 235000013343 vitamin Nutrition 0.000 description 1
- 239000000230 xanthan gum Substances 0.000 description 1
- 235000010493 xanthan gum Nutrition 0.000 description 1
- 229920001285 xanthan gum Polymers 0.000 description 1
- 229940082509 xanthan gum Drugs 0.000 description 1
Images
Classifications
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P43/00—Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P9/00—Drugs for disorders of the cardiovascular system
- A61P9/10—Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P9/00—Drugs for disorders of the cardiovascular system
- A61P9/14—Vasoprotectives; Antihaemorrhoidals; Drugs for varicose therapy; Capillary stabilisers
-
- 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
- C12N5/0634—Cells from the blood or the immune system
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K35/00—Medicinal preparations containing materials or reaction products thereof with undetermined constitution
- A61K35/12—Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
- A61K2035/124—Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells the cells being hematopoietic, bone marrow derived or blood cells
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2501/00—Active agents used in cell culture processes, e.g. differentation
- C12N2501/10—Growth factors
- C12N2501/105—Insulin-like growth factors [IGF]
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2501/00—Active agents used in cell culture processes, e.g. differentation
- C12N2501/10—Growth factors
- C12N2501/11—Epidermal growth factor [EGF]
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2501/00—Active agents used in cell culture processes, e.g. differentation
- C12N2501/10—Growth factors
- C12N2501/115—Basic fibroblast growth factor (bFGF, FGF-2)
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2501/00—Active agents used in cell culture processes, e.g. differentation
- C12N2501/10—Growth factors
- C12N2501/145—Thrombopoietin [TPO]
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2501/00—Active agents used in cell culture processes, e.g. differentation
- C12N2501/10—Growth factors
- C12N2501/165—Vascular endothelial growth factor [VEGF]
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2501/00—Active agents used in cell culture processes, e.g. differentation
- C12N2501/20—Cytokines; Chemokines
- C12N2501/22—Colony stimulating factors (G-CSF, GM-CSF)
-
- 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
- C12N2501/20—Cytokines; Chemokines
- C12N2501/26—Flt-3 ligand (CD135L, flk-2 ligand)
Definitions
- the present invention relates to a novel vascular regenerative cell group derived from a mononuclear cell group and a method for inducing differentiation thereof. More specifically, the present invention relates to a novel mononuclear cell-derived revascularized cell group that promotes stabilization and maturation of new blood vessels and leads to ischemia and tissue repair, and a safe and simple differentiation induction method thereof.
- G-CSF granulocyte colony-stimulating factor
- Non-Patent Documents 1 and 2 In the case of patients and elderly people who have basic diseases such as arteriosclerosis and diabetes, it is feared that in addition to difficulty in collecting bone marrow, the function of cells itself is reduced (see Non-Patent Documents 1 and 2). . As a new attempt, a method has been reported in which progenitor cells contained in umbilical cord blood are amplified in vivo ex vivo (see Patent Document 2). In the future, embryonic stem cells (ES cells) and induced pluripotent stem cells (iPS cells) are also expected to be used as a source of such undifferentiated cells (stem cells). However, the road to realization is still far away.
- ES cells embryonic stem cells
- iPS cells induced pluripotent stem cells
- EPCs endothelial progenitor cells
- mononuclear cells were cultured in a medium such as EBM2 that contains cytokines such as vascular endothelial growth factor (VEGF) and optimized for vascular endothelial cell culture.
- VEGF vascular endothelial growth factor
- Adherent cells obtained by inducing differentiation culture of mouse peripheral blood or bone marrow mononuclear cells in EGM2-MV medium supplemented with 10% FBS in a culture dish treated with rat vitronectin are acetylated LDL (acetylated LDL).
- acetylated LDL acetylated LDL
- cells having a spindle shape and cells having a circular shape are mixed.
- the ratio of the former is large, but when cultured for a longer period of time, cells having a paving stone-like morphology presumed to be derived from the latter grow densely.
- the adherent cells obtained by the above-mentioned method include different cell groups, and that EPC is included in the latter fraction.
- the frequency of appearance of cells that can be passaged from the formation of paving stone-like colonies is extremely low, and spindle-shaped cells that are often found in the early stage of differentiation-inducing culture expand and expand, and the survival rate thereof is significantly reduced.
- Improving ischemia can be obtained by transplanting cells obtained by culturing mouse mononuclear cells for a short period of about 1 week into small animals such as mice whose myocardium or lower limbs are ischemic (local and systemic administration). It is known. That is, it is known that cells (group) induced to differentiate from mononuclear cells have an effect of promoting angiogenesis. On the other hand, cells induced to differentiate from mononuclear cells have an effect of reducing the hypoxic region of tumor (cancer) tissue (see Patent Document 3), an effect of suppressing liver fibrosis (see Patent Document 4), and the like. It is also known.
- EPC markers such as CD34 and VEGF receptor 2 (VEGFR2 / Flk-1 / KDR) (see Non-Patent Document 4) on the cell membrane. It is unclear whether these are cells that lost stem cell antigens during the differentiation process of hematopoietic stem cells or cells derived from non-hematopoietic stem cells that do not express stem cell antigens from the beginning.
- adherent cells obtained by differentiation induction culture in an EGM2-MV medium supplemented with 10% FBS in a culture dish obtained by treating human peripheral blood mononuclear cells with human fibronectin are circular in the culture period of about 1 week. It has the ability to maintain the spindle-shaped morphology well, promote the luminal formation of human vascular endothelial cells such as HUVEC (see Non-Patent Document 5), and inhibit necrosis associated with lower limb ischemia in nude mice (See Non-Patent Document 6). Clinical trial results have shown that cardiac function after myocardial infarction has been improved by autologous transplantation of human peripheral blood mononuclear cells that have undergone similar differentiation-inducing treatment.
- CD11b is one of the blood cell differentiation antigens mainly expressed on monocytes and lymphocytes.
- CD11b-positive cells include cells and lymphocytes that play a role in immune surveillance, such as macrophages, dendritic cells, natural killer cells (NK cells), and the like.
- VEGFR1 VEGF receptor 1
- CXCR4 SDF-1 receptor
- Tie-2 angiopoietin-1 receptor
- CD11b positive cells there are relatively undifferentiated fractions among CD11b positive cells.
- Some of the CD11b-positive cells in the bone marrow are CD31 antigen-positive in the presence of angiogenic factors such as vascular endothelial growth factor (VEGF) and platelet-derived growth factor (PDGF).
- VEGF vascular endothelial growth factor
- PDGF platelet-derived growth factor
- VEGF vascular endothelial growth factor
- SMA smooth muscle actin
- relatively young cells such as vascular progenitor cells; VPC (see Non-Patent Document 12) obtained by inducing differentiation of ES cells are considered to have the same properties, and their final differentiation direction. May depend on the environment.
- CD11b positive cells directly become components of new blood vessels, or are indirectly involved in the promotion of neovascularization or stabilization of new blood vessels through cytokine production. Suggest.
- non-hematopoietic stem cells that express monocyte differentiation markers such as CD11b, which are relatively abundant in mononuclear cells, as a source for differentiation induction of cells that have functions such as vascular regeneration, vascular repair, and vascular stabilization
- FBS fetal bovine serum
- Non-Patent Document 13 cells expressing a monocyte marker such as CD11b differentiate into endothelial cells in tumor blood vessels in a living body having a tumor (cancer)
- a monocyte marker such as CD11b
- CD11b positive cell co-expressing Tie2 or the like see Non-Patent Document 14
- it is difficult to distinguish the characteristics and roles strictly for convenience see Non-Patent Document 15).
- the inventors have intensively studied to solve the above-mentioned problems, and have found that cells (groups), mainly a monocyte / lymphocyte fraction present in a relatively large amount in peripheral blood, specifically, a CD11b-positive cell. From the department, we succeeded in inducing differentiation of the target cells.
- the obtained cells did not directly differentiate into vascular endothelial cells, but promoted the stabilization and maturation of new blood vessels, thereby promoting vascular regeneration and causing ischemia and tissue repair. That is, when systemically administered to a living body having an ischemic region such as cancer, it was distributed around new blood vessels and promoted stabilization and maturation of blood vessels. Moreover, this cell expressed CD11b in addition to CD31 and CXCR4, and slightly expressed the surface antigen of c-Kit. From the above characteristics, it was suggested that the cells induced to differentiate from mononuclear cells are cells (groups) belonging to a class different from those conventionally defined as EPC.
- the mononuclear cell group is divided into vascular endothelial growth factor (VEGF), basic fibroblast growth factor (bFGF), thrombopoietin (TPO). ), Differentiation induction by culturing using a medium containing one or more selected from granulocyte-colony stimulating factor (G-CSF) and FMS-like tyrosine kinase 3 ligand (FLT3L),
- G-CSF granulocyte-colony stimulating factor
- FLT3L FMS-like tyrosine kinase 3 ligand
- the present invention relates to a cell group characterized by expressing CD11b.
- the cell group of the present invention is preferably cultured using a serum-free medium.
- the cell group of the present invention is further characterized by expressing CD31 and CXCR4. CD105 is also expressed.
- the mononuclear cells used include peripheral blood, bone marrow, or umbilical cord blood-derived mononu
- Cultivation is preferably performed under hypoxic conditions.
- the low oxygen condition means an oxygen concentration condition of 1% to 10%.
- the mononuclear cell group is cultured using a medium containing VEGF, bFGF, and TPO.
- the cell group of the present invention is characterized by having blood vessel regeneration ability.
- the cell group of the present invention has the ability to regenerate blood vessels through stabilization of new blood vessels or promotion of maturation.
- the present invention also provides a cell preparation for revascularization treatment comprising the above-described cell group of the present invention.
- the cell preparation of the present invention is characterized by having an ischemic improvement and / or blood vessel maturation effect.
- the present invention also provides a cancer localization diagnostic agent comprising the above-described cell group of the present invention.
- this invention provides the preparation method of the cell group which has the blood vessel regeneration ability including the following processes: 1) The mononuclear cell group is divided into vascular endothelial growth factor (VEGF), basic fibroblast growth factor (bFGF), thrombopoietin (TPO), granulocyte Culturing using a medium containing at least one selected from granulocyte-colony stimulating factor (G-CSF) and FMS-like tyrosine kinase 3 ligand (FLT3L) 2) From the cell mass obtained by the culture A group of cells expressing CD11b is collected.
- VEGF vascular endothelial growth factor
- bFGF basic fibroblast growth factor
- TPO thrombopoietin
- G-CSF granulocyte-colony stimulating factor
- FLT3L FMS-like tyrosine kinase 3 ligand
- the medium is preferably a serum-free medium, and the culture is preferably performed under hypoxic conditions.
- the low oxygen condition means a condition with an oxygen concentration of 1% to 10%.
- the mononuclear cell group is cultured using a medium containing VEGF, bFGF, and TPO.
- the cells according to the present invention have functions of stabilizing new blood vessels, maturation, and protection, promote lumen formation by mature vascular endothelial cells, and functionally normalize tumor blood vessels. Since the present invention uses monocyte cells that can be collected relatively easily from peripheral blood as a source, the present invention is useful as an alternative method for conventional revascularization treatment using rare (hematopoietic) stem cells as a source. In addition, since the cells according to the present invention are induced to differentiate from monocytic cells under conditions where animal serum is not used, there is no risk of infection and a safe cell preparation that can be clinically applied can be provided.
- FIG. 1 shows the promotion of MS-1 lumen formation by adherent cells obtained by inducing differentiation of mouse bone marrow mononuclear cells in EGM2-MV medium supplemented with 10% FBS.
- FIG. 2-1 shows vascular maturation by adherent cells obtained by differentiation induction culture of mouse bone marrow mononuclear cells in EGM2-MV medium supplemented with 10% FBS.
- FIG. 2-2 shows the results of transplantation of adherent cells obtained by inducing differentiation culture of mouse bone marrow mononuclear cells in EGM2-MV medium supplemented with 10% FBS to cancer-bearing nude mice.
- FIG. 1 shows the promotion of MS-1 lumen formation by adherent cells obtained by inducing differentiation of mouse bone marrow mononuclear cells in EGM2-MV medium supplemented with 10% FBS.
- FIG. 2-1 shows vascular maturation by adherent cells obtained by differentiation induction culture of mouse bone marrow mononuclear cells in EGM2-MV medium supplemented with 10% FBS
- FIG. 3 shows the results of gene expression analysis by quantitative RT-PCR using a TaqMan probe (upper left: CA9, upper middle: hENT1, upper right: dCK, lower left: Oct4, lower middle: MDR1, lower right: ABCG2).
- FIG. 4 shows the morphology of mouse bone marrow mononuclear cell CD11b positive fraction and CD11b negative fraction.
- FIG. 5 shows the results of differentiation induction culture of mouse bone marrow mononuclear cell CD11b positive fraction in EGM2-MV medium supplemented with 10% FBS.
- FIG. 6 shows the results of differentiation induction culture of human peripheral blood mononuclear cells in EGM2-MV medium supplemented with 10% FBS.
- FIG. 7 shows the results of differentiation induction culture of human peripheral blood mononuclear cells in an EGM2-MV medium supplemented with 5% or 1% FBS in a hypoxic environment.
- FIG. 8 shows the results of differentiation induction culture of human peripheral blood mononuclear cells in 20% autologous serum-supplemented X-VIVO 15 medium (1 ng / mL, human VEGF also added) and 20% autologous serum-added EGM2-MV medium.
- FIG. 9 shows the differentiation induction culture results of human peripheral blood mononuclear cells in 5% autologous serum, 50 ng / mL VEGF, 50 ng / mL bFGF added X-VIVO 15 medium.
- FIG. 10 shows the expression of surface markers on adherent cells obtained by differentiation induction culture of human peripheral blood mononuclear cells in 5% autologous serum, 50 ng / mL VEGF, 50 ng / mL bFGF-added X-VIVO 15 medium.
- FIG. 11 shows the results of differentiation induction culture of human peripheral blood mononuclear cells in X-VIVO 15 medium supplemented with 50 ng / mL VEGF and 50 ng / mL bFGF in the presence of 0, 1, 5, 10% autologous serum. Show.
- FIG. 11 shows the results of differentiation induction culture of human peripheral blood mononuclear cells in X-VIVO 15 medium supplemented with 50 ng / mL VEGF and 50 ng / mL bFGF in the presence of 0, 1, 5, 10% autologous serum. Show.
- FIG. 11 shows the results of differentiation induction culture of human peripheral blood mononuclear cells in X-VIVO 15 medium supplemente
- FIG. 12 shows the results of differentiation induction culture of human peripheral blood mononuclear cells in 50-ng / mL VEGF, 50-ng / mL bFGF-added X-VIVO 15 medium in the presence or absence of 10% autologous serum.
- FIG. 13 shows the FLT3L concentration dependence of differentiation-inducing culture of human peripheral blood mononuclear cells in 50 ng / mL VEGF, 50 ng / mL bFGF-added X-VIVO 15 medium.
- FIG. 13 shows the FLT3L concentration dependence of differentiation-inducing culture of human peripheral blood mononuclear cells in 50 ng / mL VEGF, 50 ng / mL bFGF-added X-VIVO 15 medium.
- FIG. 14 shows the results of differentiation induction culture of human peripheral blood mononuclear cells in 50 ng / mL VEGF, 50 ng / mL bFGF, 0-100 ng / mL G-CSF supplemented X-VIVO 15 medium.
- FIG. 15-1 shows the results of differentiation induction culture of human peripheral blood mononuclear cells in X-VIVO 15 medium supplemented with 50 ng / mL VEGF, 50 ng / mL bFGF, 0-100 ng / mL TPO.
- FIG. 15-1 shows the results of differentiation induction culture of human peripheral blood mononuclear cells in X-VIVO 15 medium supplemented with 50 ng / mL VEGF, 50 ng / mL bFGF, 0-100 ng / mL TPO.
- FIG. 15-2 shows a cell aggregate obtained by inducing differentiation of human peripheral blood mononuclear cells in 50 ng / mL VEGF, 50 ng / mL bFGF, 0-100 ng / mL TPO-added X-VIVO 15 medium. The quantitative analysis results are shown.
- FIG. 16 shows the results of differentiation induction culture of human peripheral blood mononuclear cells in X-VIVO 15 medium supplemented with 50 ng / mL VEGF, 50 ng / mL bFGF, 0-100 ng / mL TPO.
- FIG. 16 shows the results of differentiation induction culture of human peripheral blood mononuclear cells in X-VIVO 15 medium supplemented with 50 ng / mL VEGF, 50 ng / mL bFGF, 0-100 ng / mL TPO.
- FIG. 17 shows a cell cluster obtained by inducing differentiation of human peripheral blood mononuclear cells in X-VIVO 15 medium supplemented with 50 ng / mL VEGF, 50 ng / mL bFGF, and 100 ng / mL TPO. The result of further culturing in 20% FBS-added EGM2-MV medium is shown.
- FIG. 18 shows the results of differentiation induction culture of peripheral blood mononuclear cells (CD11b positive cells / CD11b negative cells) from multiple myeloma patients in 50-ng / mL VEGF, 50-ng / mL bFGF-added X-VIVO 15 medium. .
- FIG. 19-1 shows the results of 2-color flow cytometry analysis of human peripheral blood mononuclear cell CD11b positive fraction.
- FIG. 19-2 shows a CD11b weak positive fraction (CD11b) of a cell group obtained by inducing differentiation of human peripheral blood mononuclear cells in X-VIVO 15 medium supplemented with 50 ng / mL VEGF and 50 ng / mL bFGF for 4 days.
- (dim ) shows the result of 2 color flow cytometry analysis.
- FIG. 19-2 shows a CD11b weak positive fraction (CD11b) of a cell group obtained by inducing differentiation of human peripheral blood mononuclear cells in X-VIVO 15 medium supplemented with 50 ng / mL VEGF and 50 ng / mL bFGF for 4 days.
- (dim ) shows the result of 2 color flow cytometry analysis.
- FIG. 19-3 shows a CD11b strong positive fraction (CD11b) of a cell group obtained by differentiation induction culture of human peripheral blood mononuclear cells in X-VIVO 15 medium supplemented with 50 ng / mL VEGF and 50 ng / mL bFGF for 4 days. The result of 2 color flow cytometry analysis of bright ) is shown.
- Fig. 19-4 shows cells obtained by inducing differentiation of human peripheral blood mononuclear cells, their CD11b positive fraction and CD14 positive fraction in 50-ng / mL VEGF, 50-ng / mL bFGF-added X-VIVO 15 medium. Show groups.
- Fig. 19-3 shows a CD11b strong positive fraction (CD11b) of a cell group obtained by differentiation induction culture of human peripheral blood mononuclear cells in X-VIVO 15 medium supplemented with 50 ng / mL VEGF and 50 ng / mL bFGF for 4 days.
- FIG. 19-5 shows magnetic beads of the CD11b positive fraction of the cell group obtained by differentiation induction culture of human peripheral blood mononuclear cells in 50 ng / mL VEGF, 50 ng / mL bFGF added X-VIVO 15 medium for 4 days 2 shows the result of 2 color flow cytometry analysis in which was separated by labeled antibody.
- FIG. 20 shows the expression analysis results of G-CSF receptor, TPO receptor, and VE-cadherin mRNA in human peripheral blood mononuclear cells (CD11b positive fraction / CD11b negative fraction / total mononuclear cells).
- FIG. 20 shows the expression analysis results of G-CSF receptor, TPO receptor, and VE-cadherin mRNA in human peripheral blood mononuclear cells (CD11b positive fraction / CD11b negative fraction / total mononuclear cells).
- 21-1 shows CD11b-positive cells among peripheral blood mononuclear cells of a patient with multiple myeloma and cells obtained by differentiation induction culture in 50 ng / mL VEGF, 50 ng / mL bFGF-added X-VIVO 15 medium.
- 2 shows the results of color flow cytometry analysis (CD14, UEA-lectin affinity).
- FIG. 21-2 shows TPO reception in cells obtained by differentiation induction culture of peripheral blood mononuclear CD11b positive cells of multiple myeloma patients in 50-ng / mL VEGF, 50-ng / mL bFGF-added X-VIVO 15 medium.
- FIG. 22 shows CD11b obtained by inducing differentiation of human peripheral blood mononuclear cells in a culture dish using X-VIVO 15 medium supplemented with 50 ng / mL VEGF, 50 ng / mL bFGF, and 20 ng / mL TPO.
- the luminal formation of the positive fraction on Matrigel (cultured in EGM2-MV medium supplemented with 10% FBS) is shown.
- FIG. 23-1 shows the results of the lower limb ischemia model test.
- FIG. 23-2 shows a fluorescence micrograph (20x objective) immunohistologically detected with an anti-BS1-lectin antibody (nude mouse lower limb ischemia model test).
- FIG. 23-3 shows the results of quantification of functional blood vessels in the vicinity of the ischemic area visualized by BS1-lectin using ImageJ software (nude mouse lower limb ischemia model test).
- FIG. 24 shows the results of blood flow evaluation by laser Doppler (improved ratio of ischemic limb / healthy limb ratio immediately after treatment) (nude mouse lower limb ischemia model test).
- FIG. 25 shows the results of gene expression analysis by quantitative RT-PCR in peripheral mononuclear cell-derived CD11b-positive cells of multiple myeloma patients who received G-CSF after chemotherapy.
- CD11b positive cells purified with magnetic beads (fre CD11b), the CD11b positive cells in 20% oxygen (cul CD11b) in X-VIVO 15 medium supplemented with 50 ng / mL VEGF and 50 ng / mLbFGF in 20% O 2 ), cells cultured under 5% oxygen (cul CD11b in 20% O 2 ).
- Cell population of the present invention is a CD11b-positive cell population having the ability to regenerate blood vessels, which is induced to differentiate from mammalian mononuclear cells.
- the cell group of the present invention is derived from “mononuclear cells”.
- “Mononuclear cells” are mononuclear mesenchymal cells that are widely distributed in connective tissue, lymphoid tissue, and bloodstream, and are present in migratory mononuclear leukocytes and tissues such as monocytes and lymphocytes. It is classified into a mononuclear phagocytic cell group represented by macrophages.
- mononuclear cells (white blood cells) derived from peripheral blood, bone marrow, or umbilical cord blood belonging to the former are preferable. In particular, mononuclear cells derived from peripheral blood are preferable because they are abundant and easy to obtain.
- the mononuclear cells used are derived from the patient to whom they are administered, so that a safe regenerative medical cell group (cell preparation) avoiding rejection can be prepared.
- the cell group of the present invention is prepared by subjecting the mononuclear cell group prepared as described above to differentiation induction culture using a serum-free medium containing an appropriate “cytokine”. By adding a cytokine, the mononuclear cells proliferate suitably even in a serum-free medium, and are induced to differentiate into cells having the intended ability to regenerate blood vessels. In addition, since the medium used does not contain serum, there is no risk of infection and the prepared cell group (cell preparation) can be directly used for clinical application.
- the cell group of the present invention can be obtained as a semi-floating (spheroid) cell cluster with weak adhesion by culturing using a serum-free medium.
- serum such as the patient's autologous serum
- CD11b is one of the blood cell differentiation antigens expressed mainly in monocytes and lymphocytes.
- CD11b positive cells include macrophages, dendritic cells, immune cells such as NK cells and lymphocytes, abnormal neovascular cells found in cancer, CD31 antigen positive vascular endothelial cells or smooth muscle actin (SMA) Relatively undifferentiated cells that differentiate into antigen-positive mural cells are also included.
- SMA smooth muscle actin
- the cell group of the present invention expresses CD31 and CXCR4, in addition to CD11b, and a slight amount of c-Kit is observed. CD105 expression is also observed.
- the inventors have found that a CD11b positive cell group that is induced to differentiate from a mononuclear cell by a specific method using a serum-free medium has excellent blood vessel regeneration ability in vivo and in vitro.
- the cell group of the present invention is considered to be derived from the CD11b dim / CD31 dim / CD14 ⁇ cell group (mainly lymphocytes) or the CD11b bright / CD31 bright / CD14 bright cell group (mainly monocytes). .
- CD11b dim / CD14 - / CD8 - and populations expressing / CD31 dim / CXCR4 + a population that express CD11b bright / CD14 + / CD105 + / CXCR4 + present .
- “Dim” indicates that immunostaining is weak and the expression level of the marker is relatively small
- “bright” indicates that immunostaining is strong and the expression level of the marker is relatively large.
- the in vivo revascularization ability of the mononuclear cell group and the CD11b positive cell group originally contained in the mononuclear cell group is low.
- the cell group conventionally called EPC is induced to differentiate from the CD11b-negative cell group of the mononuclear cell group, and the expression of the surface marker is CD45 ⁇ / CD11b ⁇ / CD34 + / CD133 + / Flk-1 + In this respect, it is clearly distinguished from the cell group of the present invention.
- the cell group of the present invention does not differentiate directly into vascular endothelial cells, but promotes the regeneration and maturation of new blood vessels, thereby promoting vascular regeneration and causing ischemia and tissue repair.
- the blood vessels are distributed around the new blood vessels, enhancing the lining of the new blood vessels (micro blood vessels) endothelial cells by the pericytes, etc. Promote transformation and maturation.
- Revascularization ability means a function that promotes or assists the mechanism by which a new blood vessel is formed in a tissue.
- Angiogenesis in which existing blood vessel endothelial cells proliferate and migrate to create a new blood vessel, ischemic blood vessel Is reconstructed (becomes thicker), forms collateral blood vessels that form a conduit that replenishes blood flow to new blood vessels, and bone marrow-derived cells reach the ischemic site via the blood flow, and the vascular endothelium and periplasm Includes angiogenesis that differentiates into any contribution at any stage.
- EPCs endothelial progenitor cells
- the cell group of the present invention does not directly differentiate into vascular endothelial cells, but contributes to revascularization treatment by promoting the stabilization and maturation of new blood vessels.
- the cell group of the present invention is prepared from the mononuclear cell group by the following steps. 1) The mononuclear cell group is divided into vascular endothelial growth factor (VEGF), basic fibroblast growth factor (bFGF), thrombopoietin (TPO), granulocyte Culturing in serum-free medium containing one or more selected from colony stimulating factor (G-CSF) and FMS-like tyrosine kinase 3 ligand (FLT3L) 2) Expressing CD11b from cell clusters Collecting cell populations.
- VEGF vascular endothelial growth factor
- bFGF basic fibroblast growth factor
- TPO thrombopoietin
- G-CSF colony stimulating factor
- FLT3L FMS-like tyrosine kinase 3 ligand
- the medium used in the present invention is not particularly limited as long as it is a medium suitable for mononuclear cell culture.
- Standard media include MEM medium, BME medium, DME medium, ⁇ -MEM medium, IMEM medium, ES medium, DM-160 medium, Fisher medium, F12 medium, WE medium, RPMI medium, StemSpan medium, StemPro medium and Mention may be made of these mixtures.
- vascular endothelial cell medium for example, EGM-2 medium, EBM-2 medium and the like.
- the medium is preferably a “serum-free medium” that does not contain animal serum such as FBS and FCS.
- the “serum-free medium” is not particularly limited as long as it is a medium suitable for mononuclear cell culture, and a commercially available serum-free medium may be used or may be appropriately prepared.
- the present inventors have established a simple differentiation induction method for the cell group of the present invention using a “serum-free medium”. A serum-free medium containing no animal serum is free from infection and the prepared cell group (cell preparation) can be directly used for clinical application.
- Cytokines “Differentiation induction” from a mononuclear cell group is performed by adding an appropriate “cytokine” to the aforementioned serum-free medium and culturing. Due to cytokines, mononuclear cells proliferate favorably even in serum-free media and are induced to differentiate into cells having the desired ability to regenerate blood vessels.
- cytokine used in the present invention, vascular endothelial growth factor (VEGF), basic fibroblast growth factor (bFGF), thrombopoietin (TPO), Granulocyte-colony stimulating factor (G-CSF), FMS-like tyrosine kinase 3 ligand (FLT3L), Macrophage-colony stimulating factor (M-CSF), hedgehog ligand, CEACAM (cancer) Fetal antigen-related cell adhesion factor) and the like, but are not limited thereto as long as they meet the purpose and effect of the present invention.
- VEGF vascular endothelial growth factor
- bFGF basic fibroblast growth factor
- TPO thrombopoietin
- G-CSF Granulocyte-colony stimulating factor
- FMS-like tyrosine kinase 3 ligand FMS-like tyrosine kinase 3 ligand
- M-CSF Macrophage-
- VEGF vascular endothelial growth factor
- VEGFR VEGF receptor
- VEGF-A Growth factors involved in angiogenesis, angiogenesis, and lymphangiogenesis are VEGF-A, VEGF-B, VEGF-C, VEGF-D, VEGF-D, VEGF-E, PlGF-1, and PlGF-2. These are collectively referred to as the “VEGF family”, and only VEGF-A is sometimes simply referred to as VEGF. There are also several subtypes of some VEGF family members.
- the “vascular endothelial growth factor (VEGF)” used in the present invention includes these VEGF families and their subtypes as long as the object and effect of the present invention are not impaired.
- Base fibroblast growth factor is a heparin-binding mitogenic protein, and has a function of promoting angiogenesis and arteriogenesis as a powerful angiogenic factor (peptide), and is a nerve. Also involved in bone formation. It is known to have an effect of increasing proliferation of various types of cells under serum-free culture or culture conditions with a small amount of serum.
- TPO Thrombopoietin
- Platelets differentiate from hematopoietic stem cells via megakaryocytes, play an important role in blood coagulation, and are also involved in various immune responses.
- TPO was reported as a factor having the activity of promoting platelet formation and was first cloned in 1994. Later, it was elucidated that TPO is a ligand of c-mpl that has the function of suppressing the formation of megakaryocyte colonies, and is thought to be an important factor in the production of hematopoietic cells.
- G-CSF Granulocyte-colony stimulating factor
- FMS-like tyrosine kinase 3 ligand is a tyrosine kinase 3 ligand, and the proliferation and differentiation of hematopoietic progenitor cells and stem cells through signal transduction through the Flt3 receptor (CD135), a type of receptor tyrosine kinase. It is known to control.
- Flt3 ligand is known to have proliferative activity against monocytic cells such as CD34 or CD133 positive hematopoietic stem cells and dendritic cells, and these can be amplified in vivo or in vitro.
- the cytokines such as VEGF, bFGF, TPO and FLT3L used in the present invention may be natural or recombinant. These cytokines are preferably derived from the same species as the mononuclear cells used. Therefore, human VEGF is preferred when human mononuclear cells are used. As VEGF, commercially available products (reagents or pharmaceuticals) may be used, or recombinantly produced based on known sequence information may be used.
- a serum-free medium containing at least one of the above cytokines is used.
- the serum-free medium contains VEGF, bFGF, and TPO.
- the expression of TPO receptor is low in CD11b-positive cells, which may be due to the paracrine effect via CD11b-negative cells, or in the case of young CD11b-positive cells from bone marrow with G-CSF, the process of differentiation induction Although a direct effect is also expected due to the increased expression of TPO receptor, the mechanism of action is unknown.
- the amount of cytokine in the medium is appropriately set according to the cells to be used, but is generally about 1 to 100 ⁇ g / ml.
- Culture is performed under conditions normally used for lymphocyte culture, using a surface-treated culture dish or the like. That is, the temperature is 37 ° C. and the oxygen concentration is 20%.
- Cultivation is preferably performed under hypoxic conditions.
- “low oxygen condition” means an oxygen concentration at least lower than the oxygen content in air (about 21%), and specifically means an oxygen concentration of 1% to 10%. .
- Cultivation of cells is preferably carried out in a cell preparation facility “CPC (Cell Processing Center)” based on GMP.
- CPC Cell Processing Center
- Preparation of “clinical grade cells” to be administered to a subject is a facility specially designed to manipulate cells in a sterile condition, more specifically air conditioning control, room pressure control, temperature / humidity control, particle counter, HEPA It is preferable to use CPC with cleanliness guaranteed by a filter or the like.
- all devices used in the CPC are guaranteed to have performance through validation, and it is preferable to monitor and record their functions as needed. It is desirable to strictly manage and record according to the “procedure manual”.
- CD11b-positive cells By culturing using a serum-free medium, it is obtained as a semi-floating (spheroid) cell cluster with weak adhesion. Cells expressing CD11b are recovered from this cell cluster. Recovery of cells expressing CD11b can be easily performed using a CD11b antibody according to a conventional method.
- CD11b positive cells may be separated using a magnetic bead labeled with a CD11b antibody, a cell sorter using a fluorescently labeled CD11b antibody, or a column on which a CD11b antibody is immobilized.
- the CD11b antibody a commercially available one may be used, or it may be prepared using CD11b or a partial peptide thereof according to a conventional method.
- Cell preparation 3.1 Cell preparation for revascularization The cell group of the present invention does not directly differentiate into vascular endothelial cells, but promotes the regeneration of blood vessels by promoting the stabilization and maturation of new blood vessels to promote ischemia and tissue. Bring repair.
- the cell group of the present invention can be used as a “cell preparation for revascularization treatment” that promotes the stabilization and maturation of blood vessels when administered to a patient having an ischemic region such as cancer.
- the cell preparation of the present invention does not directly differentiate itself into vascular endothelial cells, but exhibits the ability to regenerate blood vessels through the stabilization of new blood vessels or the promotion of maturation. It is clearly distinguished from cell preparations for revascularization treatment.
- the administration method of the cell preparation of the present invention is not particularly limited, and depending on the application site, local transplantation by surgical means, intravenous administration, lumbar puncture administration, local injection administration, subcutaneous administration, intradermal administration, intraperitoneal administration, Intramuscular administration, intracerebral administration, intraventricular administration, intravenous administration, and the like are possible.
- local administration or intravenous administration is preferable as an administration method to ischemic sites including cancer.
- Angiogenesis such as VE-cadherin, VEGF receptor 1 (VEGFR1), SDF-1 receptor (CXCR4), angiopoietin-1 receptor (Tie-2) together with CD11b antigen It is known that receptor expression for factors is observed (see above).
- the cell group of the present invention has selective directivity to tumor tissues and ischemic regions, and is administered intravenously or locally to localize the tumor and to provide structural and functional blood vessels of the tumor blood vessels. There is a possibility of repair.
- the angiogenic ability of mononuclear cells obtained from peripheral blood of patients with lifestyle-related diseases such as hypertension, diabetes and hyperlipidemia, or elderly patients may be impaired. Even in patients having such various complications, regenerative treatment using their own cells becomes possible by preparing the cell preparation of the present invention using mononuclear cells obtained from the peripheral blood.
- the cell preparation of the present invention may contain scaffold materials and components for assisting cell maintenance / proliferation and administration to the affected area, and other pharmaceutically acceptable carriers.
- Components necessary for cell maintenance / proliferation include media components such as carbon sources, nitrogen sources, vitamins, minerals, salts, various cytokines, and extracellular matrix preparations such as Matrigel TM .
- scaffold materials and components that assist administration to the affected area include biodegradable polymers; for example, collagen, polylactic acid, hyaluronic acid, cellulose, and derivatives thereof, and a complex composed of two or more thereof, an aqueous solution for injection;
- aqueous solution for injection For example, physiological saline, medium, physiological buffer such as PBS, isotonic solution (eg D-sorbitol, D-mannose, D-mannitol, sodium chloride) containing glucose and other adjuvants, etc.
- An auxiliary agent such as alcohol, specifically ethanol, polyalcohol such as propylene glycol, polyethylene glycol, nonionic surfactant such as polysorbate 80, HCO-50, etc. may be used in combination.
- organic solvents polyvinyl alcohol, polyvinyl pyrrolidone, carboxyvinyl polymer, sodium carboxymethylcellulose, sodium polyacrylate, sodium alginate, water-soluble dextran, sodium carboxymethyl starch, pectin, methylcellulose as necessary , Ethyl cellulose, xanthan gum, gum arabic, casein, agar, polyethylene glycol, diglycerin, glycerin, propylene glycol, petrolatum, paraffin, stearyl alcohol, stearic acid, mannitol, sorbitol, lactose, surfactants acceptable as pharmaceutical additives, It may contain a buffer, an emulsifier, a suspension, a soothing agent, a stabilizer and the like.
- a purified antibody is dissolved in a solvent such as physiological saline, buffer solution, glucose solution, etc., and an adsorption inhibitor such as Tween 80, Tween 20, gelatin or the like is added thereto.
- a solvent such as physiological saline, buffer solution, glucose solution, etc.
- an adsorption inhibitor such as Tween 80, Tween 20, gelatin or the like is added thereto.
- Tween 80, Tween 20, gelatin or the like is added thereto.
- Diseases that can be the subject of the cell preparation of the present invention include, for example, bed sores / skin ulcers, surgical scars, wounds including refractory peptic ulcers, inflammatory diseases including chronic inflammatory bowel diseases such as ulcerative colitis and Crohn's disease All diseases requiring revascularization are included, such as diseases, severe limb ischemia, ischemic heart disease including myocardial infarction / angina pectoris / heart failure, cerebral infarction, diabetic neuropathy, cancer with severe ischemia.
- diabetics including severe chronic lower limb ischemia (obstructive arteriosclerosis, Buerger's disease), treatment-refractory ischemic heart disease, cancer with severe ischemia, and retinopathy that are difficult to treat with conventional medicine Vascular disorders and the like are preferred as target diseases.
- severe chronic lower limb ischemia obstructive arteriosclerosis, Buerger's disease
- treatment-refractory ischemic heart disease cancer with severe ischemia
- cancer with severe ischemia cancer with severe ischemia
- retinopathy that are difficult to treat with conventional medicine Vascular disorders and the like are preferred as target diseases.
- the cell group of the present invention has selective directivity to tumor tissues and ischemic regions. Therefore, if the cell group of the present invention is labeled with nanoparticles or the like, it can be applied to an image diagnosis in which the localization of cancer including ischemia and metastasis is observed. Cell labeling can be easily performed by labeling with a magnetic substance or a fluorescent dye according to a conventional method.
- the cell group of the present invention has selective directivity to tumor tissue, it may be used as a carrier for proteins and drugs having cytotoxicity against anticancer drugs and tumor cells. .
- Example 1 Induction of differentiation of mouse mononuclear cells in EGM2-MV medium Mononuclear cells were prepared from mouse bone marrow as follows. The femur of mice was crushed using a mortar and DPBSE (PBS containing EDTA at a concentration of 5 mM), and bone marrow fluid was collected. The collected bone marrow fluid is filtered using a membrane filter with a diameter of 70 ⁇ m to collect a bone marrow cell suspension, suspended in 10 ml of DPBSE, and this suspension is put into a 15 ml centrifuge tube containing 4 ml of Histopaque 1083 (Sigma). Layered quietly. After this mixture was subjected to density gradient centrifugation (400 g, 20 minutes, room temperature), the cells layered in the middle were collected with a pipette to isolate bone marrow mononuclear cells (BM-MNC).
- BM-MNC bone marrow mononuclear cells
- EGM2-MV supplemented with 10% FBS in a temperature-sensitive culture dish obtained by treating rat mouse bone marrow mononuclear cells with rat vitronctin Differentiation induction culture was performed in a medium for 1 week to obtain adherent cells.
- -EGM2-MV medium Contains EGF, VEGF, IGF, bFGF (Growth factor concentrations are not disclosed; manufactured by Lonza)
- Culture conditions 20% oxygen, 5% CO 2 , cultured at 37 ° C for 4 days, re-seeded in a new UpCell culture dish that has been suspended at room temperature and not coated, and adheres after 3 days was resuspended and collected.
- adherent cells derived from mouse bone marrow mononuclear cells on angiogenesis.
- CD31 antibody vascular endothelial cells; red, manufactured by BD
- NG2 antibody pericytes; green, manufactured by Millipore
- nuclear staining blue
- pericyte lining of the lumen area and microvessels The proportion of accompanying mature blood vessels was determined. As a result, it was confirmed that the vascular area of tumor blood vessels and the lining (blood vessel maturation) by NG2-positive pericyte were improved by transplantation of cultured mononuclear cells (FIG. 2-1).
- adherent cells obtained by inducing differentiation of mononuclear cells using EGM2-MV medium supplemented with serum can promote the stabilization of new blood vessels and correct structural abnormalities of tumor blood vessels It was confirmed.
- Example 2 Induction of differentiation of mouse mononuclear cell CD11b positive fraction Mononuclear cells were prepared from mouse bone marrow in the same manner as in Example 1. Next, a CD11b positive fraction was prepared from the obtained mouse bone marrow mononuclear cells using immunomagnetic beads (Miltenyi Biotech) with a CD11b antibody immobilized thereon.
- CD11b positive fraction When cells of CD11b positive fraction were cultured in an EGM2-MV medium supplemented with 10% FBS in a culture dish treated with rat vitronectin for 3 weeks, the cells expanded and developed, but showed no tendency to proliferate. That is, CD11b-positive cells are observed as spindle-shaped adherent cells from the early stage of culture, but colony formation is weak and proliferation ability is poor (upper part of FIG. 5).
- the Lineage negative c-Kit positive fraction (fraction containing hematopoietic stem cells) was similarly subjected to differentiation induction culture for 3 weeks or more in EGM2-MV medium supplemented with 10% FBS.
- EGM2-MV medium supplemented with 10% FBS.
- FBS 10% FBS
- the appearance of cells that grew from colony formation and had a paving stone shape was observed (FIG. 5). Since this cell can be passaged, it can be said that it is close to EPC having proliferative ability and is different from a cell derived from CD11b positive cells (lower row in FIG. 5).
- EPC vascular endothelial progenitor cells
- Example 3 Differentiation induction of human mononuclear cells under various conditions 20 mL of DPBSE was added to 30 mL of peripheral blood obtained from healthy volunteers, and centrifuged at 400 xg for 35 minutes at 20 ° C to collect the buffy coat. After resuspending in 20 mL of DPBSE, density gradient centrifugation (400 g, 20 minutes, room temperature) using Histopaque 1077 (manufactured by Sigma), and the layered cells in the middle were collected with a pipette. Mononuclear cells were isolated and cultured on a plate coated with human fibronectin for 4-7 days using EBM-2 supplemented with a medium for microvascular endothelial cells EGM2-MV medium kit (Lonza). Adherent cells Got.
- Human peripheral blood mononuclear cells were cultured for differentiation induction in an EGM2-MV medium supplemented with 10% FBS in a culture dish treated with human fibronectin. In the culture period of about 1 week, the circular shape and the spindle shape are well maintained, but when the culture is performed for about 2 weeks, aging phenomenon such as swelling and extension is shown and the survival rate is remarkably reduced (FIG. 6). ).
- the EGM2-MV medium composition and culture conditions are the same as in Example 1.
- DiI-acLDL labeled with acetylated LDL was added to EGM2-MV medium supplemented with 5% or 1% FBS, and human peripheral blood mononuclear cells were subjected to differentiation induction culture in a culture dish treated with human fibronectin as in the previous section.
- Adherent cells visualized by acetylated LDL uptake are cultured in a 5% hypoxic environment to reduce senescence phenomena such as swelling and extension, improve cell viability, and reduce serum dependence. (Fig. 7).
- Human peripheral blood mononuclear cells were cultured for differentiation induction in a culture dish treated with human fibronectin using X-VIVO 15 medium supplemented with 20% autologous serum (1 ng / mL human VEGF also added).
- X-VIVO 15 medium is suitable for culturing human monocytes, macrophages and various cell lines, granulocytes, and natural killer (NK) cells in addition to purified CD3 + lymphocytes isolated from peripheral blood and human tumors. Culturing was performed at 20% oxygen, 5% CO 2 and 37 ° C. The obtained adherent cells were confirmed to have a low level of cell aging observed 2 weeks after the start of culture, compared to cells obtained with EGM2-MV medium supplemented with 20% autologous serum. (FIG. 8).
- Peripheral blood mononuclear cells were cultured for 7 days in a culture dish treated with human fibronectin. The resulting adherent cells showed a spindle-shaped morphology (FIG. 9).
- Di-acLDL labeled with acetylated LDL was added to X-VIVO 15 medium supplemented with 0, 1, 5, 10% autologous serum, 10 ng / mL VEGF, and 10 ng / mL bFGF, and human peripheral blood Differentiation induction culture was performed in a culture dish in which mononuclear cells were treated with poly-L-lysine (PLL) for 14 days. As a result, it was confirmed that there were viable cells that were visualized by the uptake of DiI-labeled acetylated LDL even under serum-free conditions (FIG. 11).
- Human peripheral blood mononuclear cells are obtained by inducing differentiation induction culture in a culture dish in X-VIVO 15 medium supplemented with 50 ng / mL VEGF, 50 ng / mL bFGF, and 0-100 ng / mL TPO for 1 week.
- the semi-floating cell conglomerate was collected, replated in an EGM2-MV medium supplemented with 20% FBS in a culture dish treated with PLL, and cultured for 3 days.
- adherent cells with spindle-shaped morphology were obtained, and that the number of adherent cells after re-seeding increased most when differentiation induction culture was performed in the presence of 100 ng / mL TPO.
- FIG. 16 Human peripheral blood mononuclear cells are obtained by inducing differentiation induction culture in a culture dish in X-VIVO 15 medium supplemented with 50 ng / mL VEGF, 50 ng / mL bFGF, and 0-100
- the X-VIVO 15 medium is more suitable than the EGM2-MV medium for inducing differentiation of cells having the desired angiogenic ability. It was also suggested that cell viability was improved by culture under hypoxic conditions and addition of bFGF. In addition, it was confirmed that the addition of FLT3L, G-CSF, and TPO (during serum-free culture) improved the recovery rate of the target angiogenic ability in a concentration-dependent manner. Furthermore, differentiation-induced culture under hypoxic conditions dramatically increases the adhesion in the presence of blood, so that it is predicted that the ability to localize in the ischemic site of interest will be enhanced even in vivo. . In addition, pre-administration of G-CSF at the time of collection of peripheral blood mononuclear cells is expected to increase the ability to form spheroids and obtain target vascular stabilized cells more effectively.
- Example 4 Flow cytometry and quantitative RT-PCR analysis of human peripheral blood mononuclear cell CD11b positive fraction
- Human peripheral blood mononuclear cells were analyzed by 2 color flow cytometry.
- the CD11b positive fraction of human peripheral blood mononuclear cells consists of CD11b dim and CD11b bright , the former being CD31 dim / CD14 ⁇ (mainly lymphocytes) and the latter being CD31 bright / CD14 + (mainly monocytes) ( Fig. 19-1).
- “Dim” indicates that immunostaining is weak and the expression level of the marker is relatively small
- “bright” indicates that immunostaining is strong and the expression level of the marker is relatively large.
- CD11b dim among the CD11b positive fractions of human peripheral blood mononuclear cells / CD31 dim / CD14 ⁇ became the main cell source of cell clumps with vascular stabilization by differentiation-inducing culture, or the presence of CD11b dim / CD31 dim / CD14 ⁇ is a CD11b bright / CD31 bright / CD14 + blood vessel It was predicted to be necessary for differentiation into cells having a stabilizing action (FIG. 19-4).
- CD11b positive fractions were purified from human peripheral blood mononuclear cells using magnetic beads, and the expression of G-CSF receptor, TPO receptor and VE-cadherin mRNA in these was compared with CD11b negative fractions and mononuclear cells as a whole.
- the expression of G-CSF receptor and VE-cadherin was high in the CD11b positive fraction, whereas the expression of TPO receptor was high in the CD11b negative fraction (FIG. 20).
- G-CSF receptor in CD11b-positive cells was significantly higher than that of mononuclear cells before isolation, but no increase in expression was observed by differentiation-inducing culture.
- CXCR4 in CD11b positive cells was enhanced about 3-fold (FIG. 21-2).
- CD11b dim / CD31 dim / CD14 - / CXCR4 + or CD11b bright / CD14 + / CD105 presence of + can be important to the induction of differentiation of cell clusters having a vascular stabilizing effect Sex was suggested.
- the CD11b positive fraction of peripheral blood mononuclear cells mobilized from the bone marrow by administration of G-CSF has high colony-forming ability in serum-free medium supplemented with VEGF, etc., thus inducing stabilization of new blood vessels It was considered more preferable as a source of cells.
- CXCR4 expression is enhanced, it is expected that the ability to localize to ischemic tissue is enhanced. Further study is needed on the significance of the decrease in CD14 expression in the process of differentiation induction and the enhancement of affinity for UEA-lectin.
- Example 5 In vitro angiogenesis ability and mouse lower limb ischemia model transplantation experiment (1) Half of human peripheral blood mononuclear cells obtained by inducing differentiation induction culture for 4 days in X-VIVO 15 medium supplemented with 50 ng / mL VEGF, 50 ng / mL bFGF, and 20 ng / mL TPO in a culture dish The floating cell clumps were collected, and the CD11b positive fraction was purified using magnetic beads on which the CD11b antibody was immobilized. This CD11b positive fraction was incorporated with DiI-labeled acetylated LDL and cultured on Matrigel in EGM2-MV medium supplemented with 10% FBS for 7 days. As a result, it was confirmed that the visualized cells formed a lumen (FIG. 22).
- BS1-lectin was injected into the heart, allowed to stand for 5 minutes, and then perfused with 4% paraformaldehyde to collect ischemic limbs. Post-fixation with 4% paraformaldehyde was performed (3 hours, 4 ° C.) and embedded in paraffin to prepare a tissue section. Fluorescence micrograph showing immunohistological detection of reactive angiogenesis (functional blood vessels) in the vicinity of ischemia (in the vicinity of the area where inflammatory cell infiltration accompanying ischemia is observed; border ischemic zone) with anti-BS1-lectin antibody ( (Object 20 ⁇ ) (FIG. 23-2: The lower part is a merge image with nuclear staining by DAPI. Scale is 100 ⁇ m).
- CD11b positive cells CD11b + from cultured MNC purified by magnetic beads in lower limb ischemic mice, CD11b positive cells without differentiation induction culture (fresh CD11b + ), mononuclear cells with differentiation induction culture Balls (cultured MNC) and Matrigel (control) were transplanted, and blood flow evaluation (improvement ratio of ischemic limb / healthy limb ratio immediately after treatment) was performed 14 days after ligation of the lower limbs.
- Example 6 Multiple myeloma patient Gene expression analysis by quantitative RT-PCR in peripheral mononuclear cell-derived CD11b-positive cells G-CSF was administered after chemotherapy and collected at the time of peripheral blood stem cell transplantation CD11b-positive fractions from peripheral blood mononuclear cells from patients with multiple myeloma (fre CD11b), and these CD11b-positive cells supplemented with 50 ng / mL VEGF and 50 ng / mLbFGF X-VIVO Differentiation induction culture was performed for 4 days in 15 medium under 20% oxygen (cul CD11b in 20% O 2 ) and 5% oxygen (cul CD11b in 20% O 2 ).
- the cell group according to the present invention is induced to differentiate under conditions in which animal serum is not used, using a mononuclear cell that can be collected relatively easily from the peripheral blood of a patient as a source. Therefore, the cell group according to the present invention has no risk of infection and is useful as a safe cell preparation that can be clinically applied.
- the present invention is useful as an alternative method of conventional revascularization treatment using rare (hematopoietic) stem cells.
Landscapes
- Health & Medical Sciences (AREA)
- Engineering & Computer Science (AREA)
- Life Sciences & Earth Sciences (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Organic Chemistry (AREA)
- Chemical & Material Sciences (AREA)
- Biomedical Technology (AREA)
- General Health & Medical Sciences (AREA)
- Genetics & Genomics (AREA)
- Biotechnology (AREA)
- Zoology (AREA)
- Wood Science & Technology (AREA)
- Chemical Kinetics & Catalysis (AREA)
- General Chemical & Material Sciences (AREA)
- Veterinary Medicine (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Animal Behavior & Ethology (AREA)
- Public Health (AREA)
- Pharmacology & Pharmacy (AREA)
- Medicinal Chemistry (AREA)
- Microbiology (AREA)
- Immunology (AREA)
- Hematology (AREA)
- Cell Biology (AREA)
- Biochemistry (AREA)
- General Engineering & Computer Science (AREA)
- Heart & Thoracic Surgery (AREA)
- Cardiology (AREA)
- Vascular Medicine (AREA)
- Urology & Nephrology (AREA)
- Medicines Containing Material From Animals Or Micro-Organisms (AREA)
- Micro-Organisms Or Cultivation Processes Thereof (AREA)
Abstract
Description
本発明の細胞群は、無血清培地を用いて培養されることが好ましい。
本発明の細胞群は、さらに、CD31及びCXCR4を発現していることを特徴とする。また、CD105も発現していることを特徴とする。
用いられる単核球としては、末梢血、骨髄、又は臍帯血由来の単核球が挙げられる。
本発明の細胞製剤は、虚血改善及び/又は血管成熟効果を有することを特徴とする。
本発明はまた、上記した本発明の細胞群を含む、がんの局在診断剤も提供する。
1)単核球細胞群を、血管内皮細胞増殖因子(vascular endothelial growth factor; VEGF)、塩基性線維芽細胞増殖因子(basic fibroblast growth factor: bFGF)、トロンボポイエチン(thrombopoietin; TPO)、顆粒球コロニー刺激因子(granulocyte-colony stimulating factor; G-CSF)、及びFMS-like tyrosine kinase 3 ligand(FLT3L)から選ばれる1以上を含む培地を用いて培養する
2)前記培養によって得られる細胞集塊からCD11bを発現している細胞群を回収する。
本発明の細胞群(Cell Population)は、哺乳動物の単核球から分化誘導される、血管再生能を有するCD11b陽性の細胞集団である。
本発明の細胞群は「単核球」に由来する。「単核球」とは、結合組織、リンパ組織、血流中に広く分布する単核の間葉系細胞群で、単球やリンパ球に代表される遊走単核白血球と組織中に存在するマクロファージに代表される単核貪食系の細胞群に分類される。本発明で用いられる単核球としては、前者に属する末梢血、骨髄、又は臍帯血由来の単核球(白血球)が好ましい。特に、豊富に存在し、取得が容易である点において、末梢血由来の単核球が好ましい。
本発明の細胞群は、上記のようにして調製した単核球細胞群を、適切な「サイトカイン」を含む無血清培地を用いて分化誘導培養することにより調製される。サイトカインを添加することにより、単核球は無血清培地においても好適に増殖し、目的とする血管再生能を有する細胞へと分化誘導される。また、用いられる培地は血清を含まないため、感染等の恐れがなく、調製された細胞群(細胞製剤)は、そのまま臨床応用に供することができる。
本発明の細胞群は、無血清培地を用いた培養により、接着性の弱い、半浮遊(スフェロイド)状の細胞集塊として得られる。この細胞群は、再播種して血清(患者の自己血清等)存在下で培養すると接着性の強い紡錘形をした付着性細胞となる。
本発明の細胞群は、CD11bを発現していることを特徴とする。
「CD11b」は、主として単球及びリンパ球に発現している血球分化抗原のひとつである。CD11b陽性細胞には、マクロファージや樹状細胞、NK細胞等のような免疫細胞やリンパ球のほか、がん等において見られる異常な新生血管の細胞、CD31抗原陽性の血管内皮細胞あるいはsmooth muscle actin(SMA)抗原陽性の壁細胞へと分化する比較的未分化な細胞も含まれる。これまでの報告は、CD11b発現細胞が血管新生促進効果を有する細胞に分化するなど、血管新生において重要な役割を担っている可能性は示唆するものの(前掲)、これが最終的に血管内皮細胞へと分化する場合とそうでない場合の両方がありうることを示す。
本発明の細胞群は、血管内皮細胞には直接分化しないが、新生血管の安定化と成熟化を促進することで、血管再生を促し虚血や組織修復をもたらす。すなわち、がんなどの虚血領域を有する生体に全身あるいは局所投与すると、新生血管の周辺に分布し、周被細胞による新生血管(微少血管)内皮細胞の裏打ちを増強すること等によって血管の安定化や成熟化を促進する。
本発明の細胞群は、単核球細胞群から以下の工程により調製される。
1)単核球細胞群を、血管内皮細胞増殖因子(vascular endothelial growth factor; VEGF)、塩基性線維芽細胞増殖因子(basic fibroblast growth factor: bFGF)、トロンボポイエチン(thrombopoietin; TPO)、顆粒球コロニー刺激因子(granulocyte-colony stimulating factor; G-CSF)、及びFMS-like tyrosine kinase 3 ligand(FLT3L)から選ばれる1以上を含む無血清培地を用いて培養する
2)細胞集塊からCD11bを発現している細胞群を回収する。
各組織からの単核球の分離は、市販のキット等を用いて、周知の方法により容易に実施できる。たとえば、採取した血液を適宜希釈し、あらかじめ分離液が入った遠心管に入れ、1500rpm程度で遠心して、比重の違いにより分離する。リンパ球・単球から成る末梢血単核細胞は、血漿(黄色味を帯びる)と分離液(透明)の中間に、白い帯状の層として回収される。
本発明で用いられる培地は、単核球の培養に適した培地である限り、特に限定されない。標準的な培地としては、MEM培地、BME培地、DME培地、α-MEM培地、IMEM培地、ES培地、DM-160培地、Fisher培地、F12培地、WE培地、RPMI培地、StemSpan培地、StemPro培地及びこれらの混合物を挙げることができる。あるいは市販のリンパ球培養用培地:たとえばGT-T培地(タカラバイオ)、AIM V培地(インビトロジェン)、Tリンパ球培養用培養液(コスモバイオ)、X-VIVO培地(Lonza社製)、市販の血管内皮細胞用培地:たとえばEGM-2培地やEBM-2培地等を挙げることができる。
単核球細胞群からの「分化誘導」は、適切な「サイトカイン」を、前記した無血清培地に添加して培養することにより行われる。サイトカインにより、単核球は無血清培地においても好適に増殖し、目的とする血管再生能を有する細胞へと分化誘導される。
培養は、表面処理した培養皿等を用いて、通常リンパ球の培養に用いられる条件において行われる。すなわち、温度37℃、酸素濃度20%である。
無血清培地を用いた培養により、接着性の弱い、半浮遊(スフェロイド)状の細胞集塊として得られる。この細胞集塊からCD11bを発現している細胞を回収する。CD11bを発現している細胞の回収は、常法にしたがいCD11b抗体を用いて容易に実施できる。たとえば、CD11b抗体で標識された磁気ビーズ、蛍光標識されたCD11b抗体を用いたセルソーターによる分離、あるいはCD11b抗体を固相化したカラムなどを用いて、CD11b陽性細胞を分離すればよい。CD11b抗体は、市販のものを利用してもよいし、常法にしたがいCD11bあるいはその部分ペプチドを用いて作製してもよい。
3.1 血管再生治療用細胞製剤
本発明の細胞群は、血管内皮細胞には直接分化しないが、新生血管の安定化と成熟化を促進することで、血管再生を促し虚血や組織修復をもたらす。
本発明の細胞群は腫瘍組織や虚血領域への選択的指向性を有する。それゆえ、本発明の細胞群をナノ粒子等で標識すれば、虚血、転移巣を含むがんの局在をみる画像診断に応用することができる。細胞の標識は、常法にしたがい、磁性体や蛍光色素等で標識することにより簡単に行うことができる。
本発明の細胞群は腫瘍組織への選択的指向性があるため、抗がん剤や腫瘍細胞に対して細胞障害性を有するタンパク質や薬剤などのキャリアーとして利用できる可能性がある。
マウス骨髄から、以下のようにして単核球を調製した。マウスの大腿骨などを乳鉢とDPBSE(5mMの濃度でEDTAを含有したPBS)を用いて破砕し、骨髄液を採取した。採取した骨髄液から径70μmのメンブレンフィルターを用いてろ過して骨髄細胞懸濁液を集め、DPBSE10mlに懸濁し、この懸濁液を4mlのHistopaque 1083(Sigma社)が入った15mlの遠心チューブに静かに重層した。この混合物を用いて密度勾配遠心分離(400g、20分間、室温)した後、中間に層状になった細胞をピペットにより採取して骨髄単核球(BM-MNC)を単離した。
・EGM2-MV培地:EGF, VEGF, IGF, bFGFを含む(増殖因子の濃度は非公開;Lonza社製)
・培養条件:20 %酸素、5 % CO2、37℃にて4日間培養し、付着細胞を室温にて浮遊させコーティング処理を施さない新しいUpCell培養皿に再播種して、3日後に付着細胞を再度浮遊させて回収した。
マウス骨髄単核球から分化誘導された付着細胞群の血管形成に対する影響について検討した。
前項で得られた付着細胞をGFPで標識し、増殖因子の含有量の少ないマトリゲル上でマウス血管内皮細胞株MS-1と同様の培地を用いて共培養した。その結果、培養単核球の添加により、MS-1の管腔形成が促進されることが確認された(図1)。
前項で得られた付着細胞105個を4日の間隔で合計3回、ヒト膵癌細胞株KP-1Nを皮下移植した担がんヌードマウスに対して経静脈的に移植した。すなわち、腫瘍径が8 mm以上に達した時点で、培養単核球5 x 105個を経静脈的に移植し、1週間後に腫瘍組織を回収して、腫瘍血管を免疫組織学的に解析した。
また、CD31抗体(血管内皮細胞;赤)、核染色(青)を行い、GFPで蛍光標識(緑)して移植単核球の局在をみた。その結果、移植細胞は腫瘍血管の周辺(perivascular area)に分布するだけで、血管内皮細胞には分化しないことが確認された(図2-2)。
上記(2)で得られた腫瘍よりRNAを抽出し、TaqManプローブを用いた定量的RT-PCRによって癌細胞由来(ヒト膵癌細胞KP-1N)の遺伝子発現を解析した(図3)。その結果、CA9の発現が顕著に減少する一方、hENTの発現は上昇し、幹細胞マーカーであるOct4、薬剤耐性に関わるMDR-1、ABCG2などの発現が低下することが確認された。
以上の結果から、血清を添加したEGM2-MV培地を用いて単核球を分化誘導して得られる付着細胞は、新生血管の安定化を促進させ、腫瘍血管の構造的異常性を修正しうることが確認された。
実施例1と同様にして、マウス骨髄から単核球を調製した。次いで、取得したマウス骨髄単核球からCD11b抗体を固定化した免疫磁気ビーズ(ミルテニーバイオテク社製)を用いてCD11b陽性分画を調製した。
CD11b陽性分画の細胞では、全単核球と同様に紡錘形の付着細胞が観察された。一方、同様の形態を有する細胞は、CD11b陰性分画を用いた場合には極めて少なかった(図4)。このことから、培養初期に見られる紡錘形細胞は単球由来であると考えられた。
CD11b陽性分画の細胞を、ラットvitronectin処理した培養皿で10 % FBSを添加したEGM2-MV培地において3週間培養したところ、細胞の膨化と進展がみられたが、増殖傾向は示さなかった。すなわち、CD11b陽性細胞は培養の早期段階より紡錘形の付着細胞として観察されるがコロニー形成が弱く、増殖能に乏しい(図5上段)。
以上の結果から、従来血管内皮前駆細胞(EPC)と呼ばれてきた細胞は、CD11b陰性分画のうち特にLineage陰性c-Kit陽性分画に由来し、一方初期付着細胞の多くは、CD11b発現細胞であると思われた。
健常ボランティアから得た末梢血30mLにDPBSEを20mLを添加して、20℃、400×g で 35分間遠心し、バフィーコートを回収して20mLのDPBSEに再懸濁した後に、Histopaque 1077(Sigma社製)を用いて密度勾配遠心分離(400g、20分間、室温)した後、中間に層状になった細胞をピペットにより採取して、単核球を単離し、ヒトフィブロネクチンをコーティングしたプレート上で、微小血管内皮細胞用培地EGM2-MV培地キット(Lonza社製)で補完したEBM-2を用いて4~7日間培養し、付着細胞を得た。
ヒト末梢血単核球をヒトfibronectin処理した培養皿で10 % FBSを添加したEGM2-MV培地にて分化誘導培養した。1週間程度の培養期間においては類円形から紡錘形の形態を良く維持するが、2週間程度の培養を行うと膨化・伸展といった老化現象を示し生存率は著しく低下することが確認された(図6)。EGM2-MV培地組成と培養条件は実施例1と同じである。
5 %又は1 % FBSを添加したEGM2-MV培地にアセチル化LDLでラベルしたDiI-acLDLを加え、前項と同様に、ヒト末梢血単核球をヒトfibronectin処理した培養皿で分化誘導培養した。アセチル化LDLの取り込みによって可視化した付着細胞は、5 %の低酸素環境で培養することによって、膨化や伸展などの老化現象が軽減され、細胞の生存率が向上すること、また血清依存性が低下することが確認された(図7)。
20%自家血清を添加したX-VIVO 15培地(1 ng/mL ヒトVEGFも添加)を用いて、ヒト末梢血単核球をヒトfibronectin処理した培養皿で分化誘導培養した。X-VIVO 15培地は末梢血及びヒト腫瘍から分離された精製CD3+リンパ球の他、ヒト単球、マクロファージ及び各種細胞株、顆粒球、及びナチュラルキラー(NK)細胞の培養に適している。培養は、20 %酸素、5 % CO2、37℃にて実施した。得られた付着細胞は、20 %自家血清を添加したEGM2-MV培地によって得た細胞に比較して、培養開始から2週間後に観察される細胞の老化現象が低いレベルに抑えられることが確認された(図8)。
5 %自家血清と10 ng/mLのVEGF、10 ng/mLの塩基性線維芽細胞増殖因子(basic fibroblast growth factor: bFGF)を添加したX-VIVO 15培地を用いて、前項と同様に、ヒト末梢血単核球をヒトfibronectin処理した培養皿で7日間分化誘導培養した。得られた付着細胞は、紡錘形の形態を示した(図9)。
5 %自家血清と10 ng/mLのVEGF、10 ng/mLのbFGFを添加したX-VIVO 15培地を用いて、前項と同様に、ヒト末梢血単核球をヒトfibronectin処理した培養皿で7日間分化誘導培養した。得られた付着細胞について、フローサイトメトリーを用いて表面マーカーの発現を確認した。その結果、付着細胞は細胞膜上にCD11b、CD14、CD31、CD105、CD146、VEGF受容体2(VEGFR2)、SDF-1受容体(CXCR4),G-CSF受容体を発現することが確認された(図10)。
0, 1, 5, 10 %自家血清と10 ng/mLのVEGF、10 ng/mLのbFGFを添加したX-VIVO 15培地に、アセチル化LDLでラベルしたDiI-acLDLを添加し、ヒト末梢血単核球をpoly-L-lysine(PLL)処理した培養皿で14日間分化誘導培養を行った。その結果、無血清条件下においても接着は弱いがDiIラベルしたアセチル化LDLの取り込みによって可視化される生細胞が存在することが確認された(図11)。
ヒト末梢血単核球を培養皿で50 ng/mLのVEGF、50 ng/mLのbFGFを添加したX-VIVO 15培地にて1週間分化誘導培養した。その結果、無血清状態では半浮遊状(スフェロイド状)の細胞集塊が出現することが確認された。これらの細胞集塊は細胞接着性が弱いのに対し、10 %自家血清の存在下では、類円形から紡錘形の形態を示す細胞集塊が得られ、細胞の接着性が増強し、細胞面積は増大することが確認された(図12)。これら、血清含有培地で見られる接着細胞はacLDLの取り込み能を有するものの、CD11b陽性かつ、単球系マーカーであるCD14などを発現するためEPCとは言えないと思われた。
ヒト末梢血単核球を培養皿で50 ng/mLのVEGF、50 ng/mLのbFGF、0-100 ng/mL FMS-like tyrosine kinase 3 ligand(FLT3L)を添加したX-VIVO 15培地にて1週間分化誘導培養した。その結果、半浮遊状の細胞集塊の数はFLT3Lの濃度依存性に増加することが確認された(図13)。
ヒト末梢血単核球を培養皿で50 ng/mLのVEGF、50 ng/mLのbFGF、0-100 ng/mL 顆粒球増殖因子(granulocyte-colony stimulating factor; G-CSF)を添加したX-VIVO 15培地にて1週間分化誘導培養した。その結果、半浮遊状の細胞集塊の数がG-CSFの濃度依存性に増加することが確認された(図14)。
ヒト末梢血単核球を培養皿で50 ng/mLのVEGF、50 ng/mLのbFGF、0-100 ng/mL TPOを添加したX-VIVO 15培地にて1週間分化誘導培養した。その結果、半浮遊状の細胞集塊の数がthrombopoietin(TPO)の濃度依存性に増加することが確認された(図15-1)。また、培養4日目における細胞集塊の数をサイズごとに計測し、定量的に解析した結果、TPOの添加によって、サイズの大きな細胞集塊の数が増大し、特に10‐100 ng/mLを添加した際にサイズが大きな細胞集塊の出現頻度が増加する傾向が得られた(図15-2)。この実験結果から、培地に添加するTPOの至適濃度は10‐100 ng/mLと考えられた。
ヒト末梢血単核球を培養皿で50 ng/mLのVEGF、50 ng/mLのbFGF、0-100 ng/mL TPOを添加したX-VIVO 15培地にて1週間分化誘導培養することで得た半浮遊状の細胞集塊を回収し、PLL処理した培養皿で20 % FBSを添加したEGM2-MV培地に再播種して3日間培養した。その結果、紡錘形の形態を呈する付着細胞が得られること、さらに100 ng/mL TPOの存在下で分化誘導培養を行った場合に、再播種後の付着細胞数が最も増加することが確認された(図16)。
ヒト末梢血単核球を培養皿で50 ng/mLのVEGF、50 ng/mLのbFGF、100 ng/mL TPOを添加したX-VIVO 15培地にて、20%あるいは5%酸素濃度のもとで1週間分化誘導培養して得た半浮遊状の細胞集塊を回収し、PLL処理した培養皿で20 % FBSを添加したEGM2-MV培地に再播種して3日間培養した。その結果、紡錘形の形態を呈する付着細胞が得られること、さらにこれらがDiIをラベルしたアセチル化LDLの取り込みによって可視化されることが確認された。また、初期培養を5 %の酸素濃度下(低酸素培養)で行った場合に、再播種後の付着細胞数が増加することが確認された(図17)。
自家末梢血幹細胞移植のための細胞採取時に、化学療法後にG-CSFの投与を受けて、骨髄からの前駆細胞の動員を行った多発性骨髄腫の患者より得た末梢血単核球、磁気ビーズによって純化したCD11b陽性細胞及びCD11b陰性細胞を50 ng/mLのVEGF、50 ng/mLのbFGFを添加したX-VIVO 15培地にて4日間分化誘導培養した。その結果、CD11b陽性細胞においてスフェロイド状の細胞集塊の形成が高頻度にみられた(図18)。また、一部に紡錘形の付着細胞も観察された。
以上の結果から、目的とする血管新生能力を有する細胞の分化誘導には、EGM2-MV培地よりもX-VIVO 15培地のほうが適していること。また、低酸素条件下での培養やbFGFの添加によって、細胞の生存率が向上することが示唆された。
また、FLT3L、G-CSF、TPOの添加(無血清培養時)によって濃度依存的に目的とする血管新生能力の回収率が向上することが確認された。さらに低酸素条件下での分化誘導培養によって、血性存在下での接着性が飛躍的に高まることから、生体内においても目的とする虚血部位などにおける局在能が増強することが予測される。また、末梢血単核球の採取に際して、G-CSFの前投与が行われることで、スフェロイドの形成能が高まり、より効果的に目的とする血管安定化細胞が得られることが期待できる。
さらに、単核球から分化誘導される細胞群のうち、CD11b陽性分画が初期付着細胞の主要なソースであることが示唆された。
(1)
ヒト末梢血単核球を2 color フローサイトメトリーで解析した。ヒト末梢血単核球のCD11b陽性分画は、CD11bdim及びCD11bbrightからなり、前者はCD31dim/CD14-(主としてリンパ球)、後者はCD31bright/CD14+(主として単球)であった(図19-1)。なお、「dim」は、免疫染色が弱く、マーカーの発現量が比較的少ないこと、「bright」は免疫染色が強く、マーカーの発現量が比較的多いことを示す。
ヒト末梢血単核球を培養皿で50 ng/mLのVEGF、50 ng/mLのbFGFを添加したX-VIVO 15培地にて4日間分化誘導培養し、2 color フローサイトメトリーで解析した。CD11bdim分画はCXCR4、CD31を発現しており、c‐Kit陽性細胞もわずかながら検出された(図19-2)。また、CD11bbright分画はCD14、CD31、CD105、CXCR4を発現しており、c‐Kit、Flk‐1陽性細胞もわずかながら検出された。(図19-3)。
ヒト末梢血単核球を血清を加えないX-VIVO 15(50 ng/mL VEGF、50 ng/mL bFGFを添加)で4日間分化誘導培養を行い、CD11b陽性細胞を磁気ビーズにてソーティングした。
2 colorフローサイトメトリー法によってCD11b陽性分画の表面マーカーの発現を解析したところ、CD14陽性細胞が主体であった(図19-5)
ヒト末梢血単核球から磁気ビーズによってCD11b陽性分画を純化し、これらにおけるG-CSF受容体、TPO受容体ならびにVE-cadherin mRNAの発現をCD11b陰性分画及び単核球全体と比較解析を行った結果、G-CSF受容体、VE-cadherinの発現がCD11b陽性分画に高かったが、逆にTPO受容体の発現はCD11b陰性分画で高かった(図20)。
化学療法後にG-CSFの投与を受け、末梢血幹細胞移植の細胞採取時に回収された多発性骨髄腫患者の末梢血単核球、及びこれらを血清を加えないX-VIVO 15(50 ng/mL VEGF、50 ng/mL bFGFを添加)で4日間分化誘導培養を行った細胞を、それぞれCD11b陽性細胞を磁気ビーズにてソーティングして、2 colorフローサイトメトリー法によってCD11b陽性分画のCD14の発現及びUEA-lectinに対する親和性を解析した(図21-1)。
自家末梢血幹細胞移植のための細胞採取時に、化学療法後にG-CSFの投与を受けて、骨髄からの前駆細胞の動員を行った多発性骨髄腫の患者より得た末梢血単核球から磁気ビーズによってCD11b陽性細胞に分離した細胞(fresh CD11b+)と、これらを50 ng/mLのVEGF、50 ng/mLのbFGFを添加したX-VIVO 15培地にて4日間分化誘導培養して得た細胞よりそれぞれRNAを抽出し、TPO受容体mRNAを定量的RT-PCRによって解析した。その結果、TPO受容体の発現量は培養前に比較して100倍以上となった。また、CD11b陽性細胞におけるG-CSF受容体の発現は分離前の単核球に比較して著しく高かったが、分化誘導培養による発現の増強はみられなかった。一方で、CD11b陽性細胞におけるCXCR4の発現は約3倍に増強した(図21-2)。
以上より、CD11b陽性分画のうち、CD11bdim/CD31dim/CD14-/CXCR4+又はCD11bbright/CD14+/CD105+の存在が血管安定化作用を有する細胞集塊の分化誘導に重要である可能性が示唆された。
また、G-CSFの投与によって骨髄より動員された末梢血単核球のCD11b陽性分画はVEGF等を添加した無血清培地において高いコロニー形成能を有することから、新生血管の安定化を誘導する細胞のソースとしてより好ましいと考えられた。さらにCXCR4の発現増強がみられることから、虚血組織への局在能が増強されることが期待できる。分化誘導の過程においてCD14の発現が低下し、UEA-lectinに対する親和性が増強する意義については、さらなる検討が必要である。
(1)
ヒト末梢血単核球を培養皿で50 ng/mLのVEGF、50 ng/mLのbFGF、20 ng/mL TPOを添加したX-VIVO 15培地にて4日間分化誘導培養することで得た半浮遊状の細胞集塊を回収して、CD11b抗体を固定化した磁気ビーズを用いてCD11b陽性分画を純化した。このCD11b陽性分画にDiIをラベルしたアセチル化LDLを取り込ませ、マトリゲル上にて10 % FBSを添加したEGM2-MV培地で7日間培養した。その結果、可視化された細胞が管腔を形成することが確認された(図22)。
ヒト末梢血単核球をX‐VIVO 15(50 ng/mL VEGF、50 ng/mL bFGFを添加)に20 ng/mL TPOを添加して、4日間培養したときに得られるスフェロイド状の細胞集塊を回収してAnnexin‐Vを標識した磁気ビーズを用いて死細胞を除去した後に、CD11b陽性細胞を磁気ビーズによって純化した(CD11b+ cultured MNC)。PBS、分化誘導培養を行わないCD11b陽性細胞(fresh CD11b+)、分化誘導培養を行った単核球(cultured MNC)をコントロールとして用いた。8週齢、メスのBalb/c nude mouseを用いて右大腿動脈を2カ所結紮して作成した下肢虚血モデルを作成して、3日後に各群3 x105個を増殖因子濃度の低いマトリゲル(growth factor reduced matrigel; BD354230)に懸濁して、5カ所に分けて虚血肢に局注した。2週間後の虚血肢及び健側肢の写真を示す(図23-1)。
上記した方法にしたがい、下肢虚血マウスに磁気ビーズによって純化したCD11b陽性細胞(CD11b+ from cultured MNC)、分化誘導培養を行わないCD11b陽性細胞(fresh CD11b+)、分化誘導培養を行った単核球(cultured MNC)、マトリゲル(コントロール)を移植し、下肢結紮14日後にレーザードップラーによる血流評価(治療直後の虚血肢/健側肢比の改善割合)を実施した。
以上の結果から、ヒト末梢血単核球自体(CD11b陽性細胞群も含めて)が有する血管新生能に比較して、この細胞から分化誘導されるCD11b陽性細胞群は新生血管の構造的・機能的な安定化を促進する能力を有し、下肢虚血を著名に改善しうることが確認された。
化学療法後にG-CSFの投与を受け、末梢血幹細胞移植の細胞採取時に回収された多発性骨髄腫患者の末梢血単核球よりCD11b陽性細胞を磁気ビーズによって純化した分画(fre CD11b)、及びこれらのCD11b陽性細胞を50 ng/mL VEGF、50 ng/mLbFGFを添加したX-VIVO 15培地にて20% 酸素下(cul CD11b in 20% O2)、5% 酸素下(cul CD11b in 20% O2)で4日間の分化誘導培養を行った。
このことから、骨髄より動員された末梢血CD11b陽性単核球は特に低酸素環境下での分化誘導培養によって新生血管の安定化に関わる周皮細胞(pericyte)への分化が促進されることが示唆された。また、培養によってCXCR4やTie2の発現が高くなることから、SDF-1やAngiopoietin-1およびAngiopoietin-2が産生されるがん微少環境への集積能が高まること、さらに同部位に局在した後のこれらのリガンドによる活性化を受けやすい状態になると考えられる。
Claims (17)
- 単核球細胞群を、血管内皮細胞増殖因子(vascular endothelial growth factor; VEGF)、塩基性線維芽細胞増殖因子(basic fibroblast growth factor: bFGF)、トロンボポイエチン(thrombopoietin; TPO)、顆粒球コロニー刺激因子(granulocyte-colony stimulating factor; G-CSF)、及びFMS-like tyrosine kinase 3 ligand(FLT3L)から選ばれる1以上を含む培地を用いて培養することにより分化誘導される、CD11bを発現していることを特徴とする細胞群。
- 培地が無血清培地であることを特徴とする、請求項1記載の細胞群。
- さらに、CD31及びCXCR4を発現していることを特徴とする、請求項1又は2記載の細胞群。
- 単核球が末梢血、骨髄、又は臍帯血由来であることを特徴とする、請求項1~3のいずれか1項に記載の細胞群。
- 培養が低酸素条件下で行われることを特徴とする、請求項1~4のいずれか1項に記載の細胞群。
- 低酸素条件が1%~10%の酸素濃度の条件であることを特徴とする、請求項5に記載の細胞群。
- VEGF、bFGF、及びTPOを含む培地を用いて培養することを特徴とする、請求項1~6のいずれか1項に記載の細胞群。
- 血管再生能を有することを特徴とする、請求項1~7のいずれか1項に記載の細胞群。
- 新生血管の安定化あるいは成熟化の促進を介して血管再生能を有することを特徴とする、請求項8記載の細胞群。
- 請求項1~9のいずれか1項に記載の細胞群を含む、血管再生治療用細胞製剤。
- 虚血改善及び/又は血管成熟効果を有することを特徴とする、請求項10記載の細胞製剤。
- 請求項1~9のいずれか1項に記載の細胞群を含む、がんの局在診断剤。
- 以下の工程を含む、血管再生能を有する細胞群の調製方法:
1)単核球細胞群を、血管内皮細胞増殖因子(vascular endothelial growth factor; VEGF)、塩基性線維芽細胞増殖因子(basic fibroblast growth factor: bFGF)、トロンボポイエチン(thrombopoietin; TPO)、顆粒球コロニー刺激因子(granulocyte-colony stimulating factor; G-CSF)、及びFMS-like tyrosine kinase 3 ligand(FLT3L)から選ばれる1以上を含む培地を用いて培養する
2)前記培養によって得られる細胞集塊からCD11bを発現している細胞群を回収する。 - 培地が無血清培地であることを特徴とする、請求項13記載の方法。
- 培養が低酸素条件下で行われることを特徴とする、請求項14記載の方法。
- 低酸素条件が1%~10%の酸素濃度の条件であることを特徴とする、請求項15記載の方法。
- VEGF、bFGF、及びTPOを含む培地を用いて培養することを特徴とする、請求項13~16のいずれか1項に記載の方法。
Priority Applications (3)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
JP2011508224A JP5725509B2 (ja) | 2009-04-07 | 2010-03-29 | 単核球由来の新規血管再生細胞群及びその分化誘導法 |
EP10761371A EP2418272A4 (en) | 2009-04-07 | 2010-03-29 | NOVEL REVASCULARIZATION CELLS DERIVED FROM MONOCYTES, AND METHOD FOR INDUCING THE DIFFERENTIATION THEREOF |
US13/138,835 US8951795B2 (en) | 2009-04-07 | 2010-03-29 | Revascularization cells derived from mononuclear cells, and method of inducing differentiation thereof |
Applications Claiming Priority (2)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
JP2009-093459 | 2009-04-07 | ||
JP2009093459 | 2009-04-07 |
Publications (1)
Publication Number | Publication Date |
---|---|
WO2010116665A1 true WO2010116665A1 (ja) | 2010-10-14 |
Family
ID=42935964
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/JP2010/002254 WO2010116665A1 (ja) | 2009-04-07 | 2010-03-29 | 単核球由来の新規血管再生細胞群及びその分化誘導法 |
Country Status (4)
Country | Link |
---|---|
US (1) | US8951795B2 (ja) |
EP (1) | EP2418272A4 (ja) |
JP (1) | JP5725509B2 (ja) |
WO (1) | WO2010116665A1 (ja) |
Cited By (4)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO2013175759A1 (ja) * | 2012-05-22 | 2013-11-28 | 国立大学法人旭川医科大学 | ヒト単核球由来の新規血管再生細胞群及びその分化誘導法 |
JPWO2014051154A1 (ja) * | 2012-09-28 | 2016-08-25 | 公益財団法人先端医療振興財団 | 虚血性疾患治療に適した細胞を含む細胞群の生体外増幅方法 |
WO2018043596A1 (ja) * | 2016-08-30 | 2018-03-08 | 国立大学法人 新潟大学 | 細胞製剤および細胞製剤の製造方法 |
KR20180123106A (ko) * | 2016-03-24 | 2018-11-14 | 스템랩, 에스에이 | 조직 회복을 위한 제대혈 파생 엑소좀의 이용 |
Families Citing this family (1)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US9809798B2 (en) | 2011-03-11 | 2017-11-07 | National University Of Singapore | Pericyte progenitors from peripheral blood |
Citations (3)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
JP2002020298A (ja) * | 2000-07-10 | 2002-01-23 | Junkanki Kenkyusho:Kk | 血管新生促進剤 |
WO2004063365A1 (ja) * | 2003-01-15 | 2004-07-29 | Toudai Tlo, Ltd. | TGFβ阻害活性を有する化合物の新規用途 |
WO2008142862A1 (ja) * | 2007-05-18 | 2008-11-27 | National University Corporation Asahikawa Medical College | 血管内皮前駆細胞の移植による抗がん療法 |
Family Cites Families (6)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US5980887A (en) | 1996-11-08 | 1999-11-09 | St. Elizabeth's Medical Center Of Boston | Methods for enhancing angiogenesis with endothelial progenitor cells |
US8492148B2 (en) | 2005-02-23 | 2013-07-23 | Foundation For Biomedical Research & Innovation | Method for amplification of endothelial progenitor cell in vitro |
CA2644508A1 (en) * | 2006-03-01 | 2007-09-07 | The Regenerative Medicine Institute | Compostions and populations of cells obtained from the umbilical cord and methods of producing the same |
US20080070830A1 (en) * | 2006-07-28 | 2008-03-20 | Dzau Victor J | Homing of cells to myocardium |
JP2008266220A (ja) | 2007-04-20 | 2008-11-06 | Univ Of Tsukuba | 肝線維化抑制剤 |
JP2009055817A (ja) | 2007-08-30 | 2009-03-19 | Tokai Univ | 細胞運命の解析方法 |
-
2010
- 2010-03-29 WO PCT/JP2010/002254 patent/WO2010116665A1/ja active Application Filing
- 2010-03-29 JP JP2011508224A patent/JP5725509B2/ja not_active Expired - Fee Related
- 2010-03-29 US US13/138,835 patent/US8951795B2/en not_active Expired - Fee Related
- 2010-03-29 EP EP10761371A patent/EP2418272A4/en not_active Withdrawn
Patent Citations (3)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
JP2002020298A (ja) * | 2000-07-10 | 2002-01-23 | Junkanki Kenkyusho:Kk | 血管新生促進剤 |
WO2004063365A1 (ja) * | 2003-01-15 | 2004-07-29 | Toudai Tlo, Ltd. | TGFβ阻害活性を有する化合物の新規用途 |
WO2008142862A1 (ja) * | 2007-05-18 | 2008-11-27 | National University Corporation Asahikawa Medical College | 血管内皮前駆細胞の移植による抗がん療法 |
Non-Patent Citations (1)
Title |
---|
See also references of EP2418272A4 * |
Cited By (9)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO2013175759A1 (ja) * | 2012-05-22 | 2013-11-28 | 国立大学法人旭川医科大学 | ヒト単核球由来の新規血管再生細胞群及びその分化誘導法 |
JPWO2014051154A1 (ja) * | 2012-09-28 | 2016-08-25 | 公益財団法人先端医療振興財団 | 虚血性疾患治療に適した細胞を含む細胞群の生体外増幅方法 |
KR20180123106A (ko) * | 2016-03-24 | 2018-11-14 | 스템랩, 에스에이 | 조직 회복을 위한 제대혈 파생 엑소좀의 이용 |
JP2019513414A (ja) * | 2016-03-24 | 2019-05-30 | ステムラボ,エスエイ | 組織修復のための臍帯血由来エキソソームの使用 |
KR102469326B1 (ko) * | 2016-03-24 | 2022-11-23 | 스템랩, 에스에이 | 조직 회복을 위한 제대혈 파생 엑소좀의 이용 |
WO2018043596A1 (ja) * | 2016-08-30 | 2018-03-08 | 国立大学法人 新潟大学 | 細胞製剤および細胞製剤の製造方法 |
JPWO2018043596A1 (ja) * | 2016-08-30 | 2019-08-08 | 国立大学法人 新潟大学 | 細胞製剤および細胞製剤の製造方法 |
US11311579B2 (en) | 2016-08-30 | 2022-04-26 | Niigata University | Cell preparation and method for producing cell preparation |
JP7089283B2 (ja) | 2016-08-30 | 2022-06-22 | 国立大学法人 新潟大学 | 細胞製剤および細胞製剤の製造方法 |
Also Published As
Publication number | Publication date |
---|---|
EP2418272A4 (en) | 2013-02-27 |
JPWO2010116665A1 (ja) | 2012-10-18 |
JP5725509B2 (ja) | 2015-05-27 |
EP2418272A1 (en) | 2012-02-15 |
US8951795B2 (en) | 2015-02-10 |
US20120100610A1 (en) | 2012-04-26 |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
US7470538B2 (en) | Cell-based therapies for ischemia | |
JP4672370B2 (ja) | 虚血の細胞ベースの治療 | |
Tasso et al. | The recruitment of two consecutive and different waves of host stem/progenitor cells during the development of tissue-engineered bone in a murine model | |
US20060165667A1 (en) | Novel methods, compositions and devices for inducing neovascularization | |
JP6105846B2 (ja) | 虚血組織の細胞療法 | |
JP6571537B2 (ja) | 療法および予防のための細胞を単離する方法 | |
JP7549355B2 (ja) | 間葉系間質細胞を拡大するための方法 | |
WO2008085229A2 (en) | Cell-based therapies for treating liver disease | |
JP2009535035A (ja) | 肝臓再生における脂肪細胞由来成体幹細胞 | |
WO2010056341A2 (en) | Compositions and methods for tissue repair | |
JP5725509B2 (ja) | 単核球由来の新規血管再生細胞群及びその分化誘導法 | |
WO2014046417A1 (en) | Method for preparing mesenchymal stem cell aggregates | |
JP5339533B2 (ja) | 血管内皮前駆細胞の移植による抗がん療法 | |
Al-Rifai et al. | In vivo efficacy of endothelial growth medium stimulated mesenchymal stem cells derived from patients with critical limb ischemia | |
JP2024128116A (ja) | 細胞群及びその取得方法 | |
EP2643028B1 (en) | Composition and method to improve the therapeutic effect of stem cells | |
WO2013175759A1 (ja) | ヒト単核球由来の新規血管再生細胞群及びその分化誘導法 | |
KR101659846B1 (ko) | Har-nds 유래 조혈줄기세포, 그 분리방법 및 용도 | |
KR20240094983A (ko) | vMSC의 분비체 및 이의 용도 | |
JP2022017977A (ja) | 細胞、組成物及び治療用組成物 | |
JP2017139975A (ja) | 単核球培養用無血清培地 |
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: 10761371 Country of ref document: EP Kind code of ref document: A1 |
|
WWE | Wipo information: entry into national phase |
Ref document number: 2011508224 Country of ref document: JP |
|
NENP | Non-entry into the national phase |
Ref country code: DE |
|
REEP | Request for entry into the european phase |
Ref document number: 2010761371 Country of ref document: EP |
|
WWE | Wipo information: entry into national phase |
Ref document number: 2010761371 Country of ref document: EP |
|
WWE | Wipo information: entry into national phase |
Ref document number: 13138835 Country of ref document: US |