US20030199464A1 - Regeneration of endogenous myocardial tissue by induction of neovascularization - Google Patents
Regeneration of endogenous myocardial tissue by induction of neovascularization Download PDFInfo
- Publication number
- US20030199464A1 US20030199464A1 US10/128,738 US12873802A US2003199464A1 US 20030199464 A1 US20030199464 A1 US 20030199464A1 US 12873802 A US12873802 A US 12873802A US 2003199464 A1 US2003199464 A1 US 2003199464A1
- Authority
- US
- United States
- Prior art keywords
- agent
- subject
- progenitor cells
- cardiomyocyte
- endothelial progenitor
- Prior art date
- Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
- Abandoned
Links
- 230000008929 regeneration Effects 0.000 title description 22
- 238000011069 regeneration method Methods 0.000 title description 22
- 230000002107 myocardial effect Effects 0.000 title description 20
- 206010029113 Neovascularisation Diseases 0.000 title description 15
- 230000006698 induction Effects 0.000 title description 11
- 230000003511 endothelial effect Effects 0.000 claims abstract description 132
- 210000000130 stem cell Anatomy 0.000 claims abstract description 132
- 210000004413 cardiac myocyte Anatomy 0.000 claims abstract description 97
- 238000000034 method Methods 0.000 claims abstract description 78
- 241000282414 Homo sapiens Species 0.000 claims abstract description 67
- 230000006907 apoptotic process Effects 0.000 claims abstract description 58
- 210000002216 heart Anatomy 0.000 claims abstract description 42
- 230000035755 proliferation Effects 0.000 claims abstract description 30
- 108020004999 messenger RNA Proteins 0.000 claims description 74
- 239000003795 chemical substances by application Substances 0.000 claims description 62
- 230000014509 gene expression Effects 0.000 claims description 57
- 102100031344 Thioredoxin-interacting protein Human genes 0.000 claims description 53
- 101710114149 Thioredoxin-interacting protein Proteins 0.000 claims description 53
- 150000007523 nucleic acids Chemical class 0.000 claims description 37
- 102000039446 nucleic acids Human genes 0.000 claims description 36
- 108020004707 nucleic acids Proteins 0.000 claims description 36
- 230000000302 ischemic effect Effects 0.000 claims description 34
- 230000003197 catalytic effect Effects 0.000 claims description 33
- 102100031701 Nuclear factor erythroid 2-related factor 2 Human genes 0.000 claims description 30
- 210000001519 tissue Anatomy 0.000 claims description 30
- 230000000694 effects Effects 0.000 claims description 27
- 101000588302 Homo sapiens Nuclear factor erythroid 2-related factor 2 Proteins 0.000 claims description 26
- 210000004165 myocardium Anatomy 0.000 claims description 26
- 102100031573 Hematopoietic progenitor cell antigen CD34 Human genes 0.000 claims description 23
- 101000777663 Homo sapiens Hematopoietic progenitor cell antigen CD34 Proteins 0.000 claims description 23
- 102000002933 Thioredoxin Human genes 0.000 claims description 23
- 108060008226 thioredoxin Proteins 0.000 claims description 23
- 229940094937 thioredoxin Drugs 0.000 claims description 23
- 108091034117 Oligonucleotide Proteins 0.000 claims description 22
- 101000922348 Homo sapiens C-X-C chemokine receptor type 4 Proteins 0.000 claims description 19
- 102000004890 Interleukin-8 Human genes 0.000 claims description 19
- 108090001007 Interleukin-8 Proteins 0.000 claims description 19
- 239000000074 antisense oligonucleotide Substances 0.000 claims description 19
- 238000012230 antisense oligonucleotides Methods 0.000 claims description 19
- XKTZWUACRZHVAN-VADRZIEHSA-N interleukin-8 Chemical group C([C@H](NC(=O)[C@H](CC(O)=O)NC(=O)[C@H](CC=1C2=CC=CC=C2NC=1)NC(=O)[C@@H](NC(C)=O)CCSC)C(=O)N[C@@H](CC(O)=O)C(=O)N[C@@H](CC(O)=O)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CC(N)=O)C(=O)N[C@@H](CC=1C=CC=CC=1)C(=O)N[C@@H]([C@@H](C)O)C(=O)NCC(=O)N[C@@H](CCSC)C(=O)N1[C@H](CCC1)C(=O)N1[C@H](CCC1)C(=O)N[C@@H](C)C(=O)N[C@H](CC(O)=O)C(=O)N[C@H](CCC(O)=O)C(=O)N[C@H](CC(O)=O)C(=O)N[C@H](CC=1C=CC(O)=CC=1)C(=O)N[C@H](CO)C(=O)N1[C@H](CCC1)C(N)=O)C1=CC=CC=C1 XKTZWUACRZHVAN-VADRZIEHSA-N 0.000 claims description 19
- 229940096397 interleukin-8 Drugs 0.000 claims description 19
- 102000007456 Peroxiredoxin Human genes 0.000 claims description 18
- 108030002458 peroxiredoxin Proteins 0.000 claims description 18
- 230000032258 transport Effects 0.000 claims description 18
- 102100031650 C-X-C chemokine receptor type 4 Human genes 0.000 claims description 16
- 238000013519 translation Methods 0.000 claims description 16
- 108050006947 CXC Chemokine Proteins 0.000 claims description 12
- 102000019388 CXC chemokine Human genes 0.000 claims description 12
- 102000016950 Chemokine CXCL1 Human genes 0.000 claims description 12
- 208000010125 myocardial infarction Diseases 0.000 claims description 11
- 108010014419 Chemokine CXCL1 Proteins 0.000 claims description 10
- 208000031225 myocardial ischemia Diseases 0.000 claims description 9
- 230000001023 pro-angiogenic effect Effects 0.000 claims description 9
- 230000004913 activation Effects 0.000 claims description 8
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 claims description 8
- 102000019034 Chemokines Human genes 0.000 claims description 7
- 108010012236 Chemokines Proteins 0.000 claims description 7
- 206010019280 Heart failures Diseases 0.000 claims description 7
- 241000124008 Mammalia Species 0.000 claims description 7
- 210000004351 coronary vessel Anatomy 0.000 claims description 7
- 108091028664 Ribonucleotide Proteins 0.000 claims description 6
- 208000035475 disorder Diseases 0.000 claims description 6
- 238000010494 dissociation reaction Methods 0.000 claims description 6
- 230000005593 dissociations Effects 0.000 claims description 6
- 239000002336 ribonucleotide Substances 0.000 claims description 6
- 125000002652 ribonucleotide group Chemical group 0.000 claims description 6
- 208000024172 Cardiovascular disease Diseases 0.000 claims description 5
- 102000004316 Oxidoreductases Human genes 0.000 claims description 5
- 108090000854 Oxidoreductases Proteins 0.000 claims description 5
- 108010019530 Vascular Endothelial Growth Factors Proteins 0.000 claims description 5
- 102100021943 C-C motif chemokine 2 Human genes 0.000 claims description 4
- 101710155857 C-C motif chemokine 2 Proteins 0.000 claims description 4
- 102000001902 CC Chemokines Human genes 0.000 claims description 4
- 108010040471 CC Chemokines Proteins 0.000 claims description 4
- 101710082961 GATA-binding factor 2 Proteins 0.000 claims description 4
- 102100027754 Mast/stem cell growth factor receptor Kit Human genes 0.000 claims description 4
- 108010071382 NF-E2-Related Factor 2 Proteins 0.000 claims description 4
- 102000012335 Plasminogen Activator Inhibitor 1 Human genes 0.000 claims description 4
- 108010022233 Plasminogen Activator Inhibitor 1 Proteins 0.000 claims description 4
- 108090000412 Protein-Tyrosine Kinases Proteins 0.000 claims description 4
- 102000004022 Protein-Tyrosine Kinases Human genes 0.000 claims description 4
- 150000003573 thiols Chemical class 0.000 claims description 4
- 102000018233 Fibroblast Growth Factor Human genes 0.000 claims description 3
- 108050007372 Fibroblast Growth Factor Proteins 0.000 claims description 3
- 102100040120 Prominin-1 Human genes 0.000 claims description 3
- 108010073929 Vascular Endothelial Growth Factor A Proteins 0.000 claims description 3
- 230000000735 allogeneic effect Effects 0.000 claims description 3
- 229940126864 fibroblast growth factor Drugs 0.000 claims description 3
- KTUFNOKKBVMGRW-UHFFFAOYSA-N imatinib Chemical group C1CN(C)CCN1CC1=CC=C(C(=O)NC=2C=C(NC=3N=C(C=CN=3)C=3C=NC=CC=3)C(C)=CC=2)C=C1 KTUFNOKKBVMGRW-UHFFFAOYSA-N 0.000 claims description 3
- 229960002411 imatinib Drugs 0.000 claims description 3
- 239000003112 inhibitor Substances 0.000 claims description 3
- 210000005240 left ventricle Anatomy 0.000 claims description 3
- 206010002383 Angina Pectoris Diseases 0.000 claims description 2
- 102000009840 Angiopoietins Human genes 0.000 claims description 2
- 108010009906 Angiopoietins Proteins 0.000 claims description 2
- 102100032367 C-C motif chemokine 5 Human genes 0.000 claims description 2
- 102100032366 C-C motif chemokine 7 Human genes 0.000 claims description 2
- 101710155834 C-C motif chemokine 7 Proteins 0.000 claims description 2
- 102100034871 C-C motif chemokine 8 Human genes 0.000 claims description 2
- 101710155833 C-C motif chemokine 8 Proteins 0.000 claims description 2
- 206010007559 Cardiac failure congestive Diseases 0.000 claims description 2
- 208000031229 Cardiomyopathies Diseases 0.000 claims description 2
- 108010055166 Chemokine CCL5 Proteins 0.000 claims description 2
- 102100023688 Eotaxin Human genes 0.000 claims description 2
- 101710139422 Eotaxin Proteins 0.000 claims description 2
- 101710121996 Hexon protein p72 Proteins 0.000 claims description 2
- 102000016878 Hypoxia-Inducible Factor 1 Human genes 0.000 claims description 2
- 108010028501 Hypoxia-Inducible Factor 1 Proteins 0.000 claims description 2
- 102100024573 Macrophage-capping protein Human genes 0.000 claims description 2
- 101710125418 Major capsid protein Proteins 0.000 claims description 2
- 239000002870 angiogenesis inducing agent Substances 0.000 claims description 2
- 210000001175 cerebrospinal fluid Anatomy 0.000 claims description 2
- 239000005547 deoxyribonucleotide Substances 0.000 claims description 2
- 125000002637 deoxyribonucleotide group Chemical group 0.000 claims description 2
- 210000003754 fetus Anatomy 0.000 claims description 2
- 230000003834 intracellular effect Effects 0.000 claims description 2
- 210000001161 mammalian embryo Anatomy 0.000 claims description 2
- AEUKDPKXTPNBNY-XEYRWQBLSA-N mcp 2 Chemical compound C([C@@H](C(=O)N[C@@H](CS)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H]([C@@H](C)CC)C(=O)N[C@@H](CCCNC(N)=N)C(=O)NCC(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H]([C@@H](C)CC)C(=O)N[C@@H](CC=1NC=NC=1)C(=O)N1[C@@H](CCC1)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CS)C(=O)N[C@@H](CS)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CCCNC(N)=N)C(O)=O)NC(=O)CNC(=O)[C@H](C)NC(=O)[C@H](CCCNC(N)=N)NC(=O)[C@H](CCCNC(N)=N)NC(=O)[C@H](CCC(O)=O)NC(=O)[C@H](CC(C)C)NC(=O)[C@H]1N(CCC1)C(=O)[C@H](CC(C)C)NC(=O)[C@H](CS)NC(=O)[C@H](CC(C)C)NC(=O)[C@H](C)NC(=O)[C@H](CCCNC(N)=N)NC(=O)[C@H](CCCNC(N)=N)NC(=O)[C@H](CS)NC(=O)[C@H](C)NC(=O)[C@H](CS)NC(=O)[C@@H](NC(=O)[C@@H](N)C(C)C)C(C)C)C1=CC=CC=C1 AEUKDPKXTPNBNY-XEYRWQBLSA-N 0.000 claims description 2
- 230000001537 neural effect Effects 0.000 claims description 2
- 230000003836 peripheral circulation Effects 0.000 claims description 2
- 210000005241 right ventricle Anatomy 0.000 claims description 2
- 210000002027 skeletal muscle Anatomy 0.000 claims description 2
- 102100039037 Vascular endothelial growth factor A Human genes 0.000 claims 1
- 241000700159 Rattus Species 0.000 description 98
- 210000004027 cell Anatomy 0.000 description 52
- 206010061216 Infarction Diseases 0.000 description 48
- 108090000623 proteins and genes Proteins 0.000 description 41
- 108020004414 DNA Proteins 0.000 description 31
- 210000001185 bone marrow Anatomy 0.000 description 31
- 230000007574 infarction Effects 0.000 description 30
- 210000000648 angioblast Anatomy 0.000 description 27
- 102100021669 Stromal cell-derived factor 1 Human genes 0.000 description 25
- 125000003729 nucleotide group Chemical group 0.000 description 25
- FAPWRFPIFSIZLT-UHFFFAOYSA-M Sodium chloride Chemical compound [Na+].[Cl-] FAPWRFPIFSIZLT-UHFFFAOYSA-M 0.000 description 24
- 101710088580 Stromal cell-derived factor 1 Proteins 0.000 description 24
- 210000000107 myocyte Anatomy 0.000 description 23
- 239000011780 sodium chloride Substances 0.000 description 23
- 239000002773 nucleotide Substances 0.000 description 22
- 241001465754 Metazoa Species 0.000 description 20
- 230000001965 increasing effect Effects 0.000 description 19
- 230000009467 reduction Effects 0.000 description 17
- 238000006722 reduction reaction Methods 0.000 description 17
- 230000014616 translation Effects 0.000 description 17
- 238000003776 cleavage reaction Methods 0.000 description 16
- 230000006872 improvement Effects 0.000 description 16
- 235000000346 sugar Nutrition 0.000 description 16
- 108091032973 (ribonucleotides)n+m Proteins 0.000 description 15
- 108010075639 MAP Kinase Kinase Kinase 5 Proteins 0.000 description 15
- 102100033127 Mitogen-activated protein kinase kinase kinase 5 Human genes 0.000 description 15
- 230000001419 dependent effect Effects 0.000 description 15
- 238000002347 injection Methods 0.000 description 15
- 239000007924 injection Substances 0.000 description 15
- 230000007017 scission Effects 0.000 description 15
- 230000004862 vasculogenesis Effects 0.000 description 15
- 238000010186 staining Methods 0.000 description 14
- 108020000948 Antisense Oligonucleotides Proteins 0.000 description 13
- 102000004169 proteins and genes Human genes 0.000 description 13
- 230000014621 translational initiation Effects 0.000 description 13
- 230000000875 corresponding effect Effects 0.000 description 12
- 230000006870 function Effects 0.000 description 12
- 210000005260 human cell Anatomy 0.000 description 12
- 208000028867 ischemia Diseases 0.000 description 12
- 230000036542 oxidative stress Effects 0.000 description 12
- 230000002829 reductive effect Effects 0.000 description 12
- 230000002861 ventricular Effects 0.000 description 12
- 101100220044 Homo sapiens CD34 gene Proteins 0.000 description 11
- 108091081024 Start codon Proteins 0.000 description 11
- 238000011260 co-administration Methods 0.000 description 11
- 238000009396 hybridization Methods 0.000 description 11
- 230000003993 interaction Effects 0.000 description 11
- YBJHBAHKTGYVGT-ZKWXMUAHSA-N (+)-Biotin Chemical compound N1C(=O)N[C@@H]2[C@H](CCCCC(=O)O)SC[C@@H]21 YBJHBAHKTGYVGT-ZKWXMUAHSA-N 0.000 description 10
- 238000002474 experimental method Methods 0.000 description 10
- 238000010253 intravenous injection Methods 0.000 description 10
- 230000001404 mediated effect Effects 0.000 description 10
- 230000005012 migration Effects 0.000 description 10
- 238000013508 migration Methods 0.000 description 10
- 239000003963 antioxidant agent Substances 0.000 description 9
- 230000003078 antioxidant effect Effects 0.000 description 9
- 230000027455 binding Effects 0.000 description 9
- 230000001413 cellular effect Effects 0.000 description 9
- 229910052739 hydrogen Inorganic materials 0.000 description 9
- 230000002401 inhibitory effect Effects 0.000 description 9
- 235000018102 proteins Nutrition 0.000 description 9
- 102000004269 Granulocyte Colony-Stimulating Factor Human genes 0.000 description 8
- 108010017080 Granulocyte Colony-Stimulating Factor Proteins 0.000 description 8
- 108020005038 Terminator Codon Proteins 0.000 description 8
- 235000006708 antioxidants Nutrition 0.000 description 8
- 230000022131 cell cycle Effects 0.000 description 8
- 239000001257 hydrogen Substances 0.000 description 8
- 238000001727 in vivo Methods 0.000 description 8
- 239000007925 intracardiac injection Substances 0.000 description 8
- 230000004224 protection Effects 0.000 description 8
- 101150116862 KEAP1 gene Proteins 0.000 description 7
- 102000013394 Troponin I Human genes 0.000 description 7
- 108010065729 Troponin I Proteins 0.000 description 7
- 230000001640 apoptogenic effect Effects 0.000 description 7
- LOKCTEFSRHRXRJ-UHFFFAOYSA-I dipotassium trisodium dihydrogen phosphate hydrogen phosphate dichloride Chemical compound P(=O)(O)(O)[O-].[K+].P(=O)(O)([O-])[O-].[Na+].[Na+].[Cl-].[K+].[Cl-].[Na+] LOKCTEFSRHRXRJ-UHFFFAOYSA-I 0.000 description 7
- 230000007246 mechanism Effects 0.000 description 7
- 239000002953 phosphate buffered saline Substances 0.000 description 7
- 231100000241 scar Toxicity 0.000 description 7
- WEVYAHXRMPXWCK-UHFFFAOYSA-N Acetonitrile Chemical compound CC#N WEVYAHXRMPXWCK-UHFFFAOYSA-N 0.000 description 6
- 102000008186 Collagen Human genes 0.000 description 6
- 108010035532 Collagen Proteins 0.000 description 6
- LFQSCWFLJHTTHZ-UHFFFAOYSA-N Ethanol Chemical compound CCO LFQSCWFLJHTTHZ-UHFFFAOYSA-N 0.000 description 6
- 101001124867 Homo sapiens Peroxiredoxin-1 Proteins 0.000 description 6
- 102100029139 Peroxiredoxin-1 Human genes 0.000 description 6
- 101710172405 Thiol peroxidase Proteins 0.000 description 6
- 239000011616 biotin Substances 0.000 description 6
- 229960002685 biotin Drugs 0.000 description 6
- 230000000747 cardiac effect Effects 0.000 description 6
- 229920001436 collagen Polymers 0.000 description 6
- 230000000295 complement effect Effects 0.000 description 6
- 230000001086 cytosolic effect Effects 0.000 description 6
- 230000010016 myocardial function Effects 0.000 description 6
- 230000008569 process Effects 0.000 description 6
- 238000011084 recovery Methods 0.000 description 6
- 230000001105 regulatory effect Effects 0.000 description 6
- 230000004044 response Effects 0.000 description 6
- 241000894007 species Species 0.000 description 6
- RWQNBRDOKXIBIV-UHFFFAOYSA-N thymine Chemical compound CC1=CNC(=O)NC1=O RWQNBRDOKXIBIV-UHFFFAOYSA-N 0.000 description 6
- -1 without limitation Chemical class 0.000 description 6
- 102000053602 DNA Human genes 0.000 description 5
- 108091027757 Deoxyribozyme Proteins 0.000 description 5
- 101000692259 Homo sapiens Phosphoprotein associated with glycosphingolipid-enriched microdomains 1 Proteins 0.000 description 5
- HCHKCACWOHOZIP-UHFFFAOYSA-N Zinc Chemical compound [Zn] HCHKCACWOHOZIP-UHFFFAOYSA-N 0.000 description 5
- JLCPHMBAVCMARE-UHFFFAOYSA-N [3-[[3-[[3-[[3-[[3-[[3-[[3-[[3-[[3-[[3-[[3-[[5-(2-amino-6-oxo-1H-purin-9-yl)-3-[[3-[[3-[[3-[[3-[[3-[[5-(2-amino-6-oxo-1H-purin-9-yl)-3-[[5-(2-amino-6-oxo-1H-purin-9-yl)-3-hydroxyoxolan-2-yl]methoxy-hydroxyphosphoryl]oxyoxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(5-methyl-2,4-dioxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxyoxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(5-methyl-2,4-dioxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(5-methyl-2,4-dioxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(5-methyl-2,4-dioxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methyl [5-(6-aminopurin-9-yl)-2-(hydroxymethyl)oxolan-3-yl] hydrogen phosphate Polymers Cc1cn(C2CC(OP(O)(=O)OCC3OC(CC3OP(O)(=O)OCC3OC(CC3O)n3cnc4c3nc(N)[nH]c4=O)n3cnc4c3nc(N)[nH]c4=O)C(COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3CO)n3cnc4c(N)ncnc34)n3ccc(N)nc3=O)n3cnc4c(N)ncnc34)n3ccc(N)nc3=O)n3ccc(N)nc3=O)n3ccc(N)nc3=O)n3cnc4c(N)ncnc34)n3cnc4c(N)ncnc34)n3cc(C)c(=O)[nH]c3=O)n3cc(C)c(=O)[nH]c3=O)n3ccc(N)nc3=O)n3cc(C)c(=O)[nH]c3=O)n3cnc4c3nc(N)[nH]c4=O)n3cnc4c(N)ncnc34)n3cnc4c(N)ncnc34)n3cnc4c(N)ncnc34)n3cnc4c(N)ncnc34)O2)c(=O)[nH]c1=O JLCPHMBAVCMARE-UHFFFAOYSA-N 0.000 description 5
- 235000020958 biotin Nutrition 0.000 description 5
- 230000035605 chemotaxis Effects 0.000 description 5
- 230000008021 deposition Effects 0.000 description 5
- 238000010790 dilution Methods 0.000 description 5
- 239000012895 dilution Substances 0.000 description 5
- 210000002966 serum Anatomy 0.000 description 5
- 239000000126 substance Substances 0.000 description 5
- 150000008163 sugars Chemical class 0.000 description 5
- KDCGOANMDULRCW-UHFFFAOYSA-N 7H-purine Chemical compound N1=CNC2=NC=NC2=C1 KDCGOANMDULRCW-UHFFFAOYSA-N 0.000 description 4
- 208000035657 Abasia Diseases 0.000 description 4
- 102000002260 Alkaline Phosphatase Human genes 0.000 description 4
- 108020004774 Alkaline Phosphatase Proteins 0.000 description 4
- 102100036166 C-X-C chemokine receptor type 1 Human genes 0.000 description 4
- 108020004705 Codon Proteins 0.000 description 4
- 108050006400 Cyclin Proteins 0.000 description 4
- 102100033270 Cyclin-dependent kinase inhibitor 1 Human genes 0.000 description 4
- 102100033215 DNA nucleotidylexotransferase Human genes 0.000 description 4
- 101000947174 Homo sapiens C-X-C chemokine receptor type 1 Proteins 0.000 description 4
- 101000944380 Homo sapiens Cyclin-dependent kinase inhibitor 1 Proteins 0.000 description 4
- 108091026898 Leader sequence (mRNA) Proteins 0.000 description 4
- 108091054437 MHC class I family Proteins 0.000 description 4
- 108091028043 Nucleic acid sequence Proteins 0.000 description 4
- RYYWUUFWQRZTIU-UHFFFAOYSA-N Thiophosphoric acid Chemical group OP(O)(S)=O RYYWUUFWQRZTIU-UHFFFAOYSA-N 0.000 description 4
- 108091036066 Three prime untranslated region Proteins 0.000 description 4
- 102000005789 Vascular Endothelial Growth Factors Human genes 0.000 description 4
- 235000001014 amino acid Nutrition 0.000 description 4
- 150000001413 amino acids Chemical class 0.000 description 4
- 230000015572 biosynthetic process Effects 0.000 description 4
- 230000000903 blocking effect Effects 0.000 description 4
- 230000006369 cell cycle progression Effects 0.000 description 4
- 230000001447 compensatory effect Effects 0.000 description 4
- 239000002299 complementary DNA Substances 0.000 description 4
- OPTASPLRGRRNAP-UHFFFAOYSA-N cytosine Chemical compound NC=1C=CNC(=O)N=1 OPTASPLRGRRNAP-UHFFFAOYSA-N 0.000 description 4
- 230000006378 damage Effects 0.000 description 4
- 238000011161 development Methods 0.000 description 4
- 230000018109 developmental process Effects 0.000 description 4
- GNBHRKFJIUUOQI-UHFFFAOYSA-N fluorescein Chemical compound O1C(=O)C2=CC=CC=C2C21C1=CC=C(O)C=C1OC1=CC(O)=CC=C21 GNBHRKFJIUUOQI-UHFFFAOYSA-N 0.000 description 4
- UYTPUPDQBNUYGX-UHFFFAOYSA-N guanine Chemical compound O=C1NC(N)=NC2=C1N=CN2 UYTPUPDQBNUYGX-UHFFFAOYSA-N 0.000 description 4
- 230000004217 heart function Effects 0.000 description 4
- 238000002372 labelling Methods 0.000 description 4
- 238000005259 measurement Methods 0.000 description 4
- 238000003752 polymerase chain reaction Methods 0.000 description 4
- 239000000523 sample Substances 0.000 description 4
- 150000003384 small molecules Chemical class 0.000 description 4
- 239000000243 solution Substances 0.000 description 4
- RYYWUUFWQRZTIU-UHFFFAOYSA-K thiophosphate Chemical compound [O-]P([O-])([O-])=S RYYWUUFWQRZTIU-UHFFFAOYSA-K 0.000 description 4
- 238000011282 treatment Methods 0.000 description 4
- 125000003903 2-propenyl group Chemical group [H]C([*])([H])C([H])=C([H])[H] 0.000 description 3
- QTBSBXVTEAMEQO-UHFFFAOYSA-N Acetic acid Chemical compound CC(O)=O QTBSBXVTEAMEQO-UHFFFAOYSA-N 0.000 description 3
- CSCPPACGZOOCGX-UHFFFAOYSA-N Acetone Chemical compound CC(C)=O CSCPPACGZOOCGX-UHFFFAOYSA-N 0.000 description 3
- 108090001008 Avidin Proteins 0.000 description 3
- 108090000994 Catalytic RNA Proteins 0.000 description 3
- 102000053642 Catalytic RNA Human genes 0.000 description 3
- 108091026890 Coding region Proteins 0.000 description 3
- 108091035707 Consensus sequence Proteins 0.000 description 3
- 102100036912 Desmin Human genes 0.000 description 3
- 108010044052 Desmin Proteins 0.000 description 3
- 108010070675 Glutathione transferase Proteins 0.000 description 3
- 102100031181 Glyceraldehyde-3-phosphate dehydrogenase Human genes 0.000 description 3
- 102100029100 Hematopoietic prostaglandin D synthase Human genes 0.000 description 3
- 101001055222 Homo sapiens Interleukin-8 Proteins 0.000 description 3
- MHAJPDPJQMAIIY-UHFFFAOYSA-N Hydrogen peroxide Chemical compound OO MHAJPDPJQMAIIY-UHFFFAOYSA-N 0.000 description 3
- 206010020880 Hypertrophy Diseases 0.000 description 3
- 108010018951 Interleukin-8B Receptors Proteins 0.000 description 3
- 102000002791 Interleukin-8B Receptors Human genes 0.000 description 3
- OKKJLVBELUTLKV-UHFFFAOYSA-N Methanol Chemical compound OC OKKJLVBELUTLKV-UHFFFAOYSA-N 0.000 description 3
- 101100298488 Mus musculus Prdx1 gene Proteins 0.000 description 3
- 102000003992 Peroxidases Human genes 0.000 description 3
- 108090000430 Phosphatidylinositol 3-kinases Proteins 0.000 description 3
- 102000003993 Phosphatidylinositol 3-kinases Human genes 0.000 description 3
- 108010004729 Phycoerythrin Proteins 0.000 description 3
- 102100036691 Proliferating cell nuclear antigen Human genes 0.000 description 3
- 241000251131 Sphyrna Species 0.000 description 3
- 238000013459 approach Methods 0.000 description 3
- 150000001720 carbohydrates Chemical class 0.000 description 3
- 235000014633 carbohydrates Nutrition 0.000 description 3
- 230000025084 cell cycle arrest Effects 0.000 description 3
- 230000008859 change Effects 0.000 description 3
- 238000006243 chemical reaction Methods 0.000 description 3
- 230000001351 cycling effect Effects 0.000 description 3
- 235000018417 cysteine Nutrition 0.000 description 3
- 210000005045 desmin Anatomy 0.000 description 3
- 230000009977 dual effect Effects 0.000 description 3
- 108020004445 glyceraldehyde-3-phosphate dehydrogenase Proteins 0.000 description 3
- 230000012010 growth Effects 0.000 description 3
- 229940025294 hemin Drugs 0.000 description 3
- BTIJJDXEELBZFS-QDUVMHSLSA-K hemin Chemical compound CC1=C(CCC(O)=O)C(C=C2C(CCC(O)=O)=C(C)\C(N2[Fe](Cl)N23)=C\4)=N\C1=C/C2=C(C)C(C=C)=C3\C=C/1C(C)=C(C=C)C/4=N\1 BTIJJDXEELBZFS-QDUVMHSLSA-K 0.000 description 3
- 102000053523 human CXCR4 Human genes 0.000 description 3
- 230000008595 infiltration Effects 0.000 description 3
- 238000001764 infiltration Methods 0.000 description 3
- 230000005764 inhibitory process Effects 0.000 description 3
- 238000001990 intravenous administration Methods 0.000 description 3
- 230000007774 longterm Effects 0.000 description 3
- 239000011159 matrix material Substances 0.000 description 3
- 210000003205 muscle Anatomy 0.000 description 3
- 239000013642 negative control Substances 0.000 description 3
- 229910052760 oxygen Inorganic materials 0.000 description 3
- 230000008092 positive effect Effects 0.000 description 3
- 230000002265 prevention Effects 0.000 description 3
- 230000001681 protective effect Effects 0.000 description 3
- 108700043101 rat CXCL12 Proteins 0.000 description 3
- 102000005962 receptors Human genes 0.000 description 3
- 108020003175 receptors Proteins 0.000 description 3
- 230000002441 reversible effect Effects 0.000 description 3
- 108091092562 ribozyme Proteins 0.000 description 3
- 239000000758 substrate Substances 0.000 description 3
- 230000001225 therapeutic effect Effects 0.000 description 3
- 229940113082 thymine Drugs 0.000 description 3
- 238000013518 transcription Methods 0.000 description 3
- 230000035897 transcription Effects 0.000 description 3
- 210000003462 vein Anatomy 0.000 description 3
- QRXMUCSWCMTJGU-UHFFFAOYSA-N 5-bromo-4-chloro-3-indolyl phosphate Chemical compound C1=C(Br)C(Cl)=C2C(OP(O)(=O)O)=CNC2=C1 QRXMUCSWCMTJGU-UHFFFAOYSA-N 0.000 description 2
- 229930024421 Adenine Natural products 0.000 description 2
- GFFGJBXGBJISGV-UHFFFAOYSA-N Adenine Chemical compound NC1=NC=NC2=C1N=CN2 GFFGJBXGBJISGV-UHFFFAOYSA-N 0.000 description 2
- 102000003903 Cyclin-dependent kinases Human genes 0.000 description 2
- 108090000266 Cyclin-dependent kinases Proteins 0.000 description 2
- HMFHBZSHGGEWLO-SOOFDHNKSA-N D-ribofuranose Chemical compound OC[C@H]1OC(O)[C@H](O)[C@@H]1O HMFHBZSHGGEWLO-SOOFDHNKSA-N 0.000 description 2
- 230000005778 DNA damage Effects 0.000 description 2
- 231100000277 DNA damage Toxicity 0.000 description 2
- 230000006820 DNA synthesis Effects 0.000 description 2
- 108010014303 DNA-directed DNA polymerase Proteins 0.000 description 2
- 102000016928 DNA-directed DNA polymerase Human genes 0.000 description 2
- 102000004190 Enzymes Human genes 0.000 description 2
- 108090000790 Enzymes Proteins 0.000 description 2
- WSFSSNUMVMOOMR-UHFFFAOYSA-N Formaldehyde Chemical compound O=C WSFSSNUMVMOOMR-UHFFFAOYSA-N 0.000 description 2
- 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
- 108010017213 Granulocyte-Macrophage Colony-Stimulating Factor Proteins 0.000 description 2
- 102100039620 Granulocyte-macrophage colony-stimulating factor Human genes 0.000 description 2
- 108090001102 Hammerhead ribozyme Proteins 0.000 description 2
- 102000008949 Histocompatibility Antigens Class I Human genes 0.000 description 2
- 241000282412 Homo Species 0.000 description 2
- 101100441536 Homo sapiens CXCR1 gene Proteins 0.000 description 2
- 101001069921 Homo sapiens Growth-regulated alpha protein Proteins 0.000 description 2
- 101000891649 Homo sapiens Transcription elongation factor A protein-like 1 Proteins 0.000 description 2
- 206010021143 Hypoxia Diseases 0.000 description 2
- 229930010555 Inosine Natural products 0.000 description 2
- UGQMRVRMYYASKQ-KQYNXXCUSA-N Inosine Chemical compound O[C@@H]1[C@H](O)[C@@H](CO)O[C@H]1N1C2=NC=NC(O)=C2N=C1 UGQMRVRMYYASKQ-KQYNXXCUSA-N 0.000 description 2
- 102000043129 MHC class I family Human genes 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
- 241000699666 Mus <mouse, genus> Species 0.000 description 2
- 101710163270 Nuclease Proteins 0.000 description 2
- 229910004679 ONO2 Inorganic materials 0.000 description 2
- 108700026244 Open Reading Frames Proteins 0.000 description 2
- PYMYPHUHKUWMLA-LMVFSUKVSA-N Ribose Natural products OC[C@@H](O)[C@@H](O)[C@@H](O)C=O PYMYPHUHKUWMLA-LMVFSUKVSA-N 0.000 description 2
- 102000000395 SH3 domains Human genes 0.000 description 2
- 108050008861 SH3 domains Proteins 0.000 description 2
- 108700009124 Transcription Initiation Site Proteins 0.000 description 2
- ISAKRJDGNUQOIC-UHFFFAOYSA-N Uracil Chemical compound O=C1C=CNC(=O)N1 ISAKRJDGNUQOIC-UHFFFAOYSA-N 0.000 description 2
- 229910052770 Uranium Inorganic materials 0.000 description 2
- 108010053099 Vascular Endothelial Growth Factor Receptor-2 Proteins 0.000 description 2
- 102000016549 Vascular Endothelial Growth Factor Receptor-2 Human genes 0.000 description 2
- QYSXJUFSXHHAJI-XFEUOLMDSA-N Vitamin D3 Natural products C1(/[C@@H]2CC[C@@H]([C@]2(CCC1)C)[C@H](C)CCCC(C)C)=C/C=C1\C[C@@H](O)CCC1=C QYSXJUFSXHHAJI-XFEUOLMDSA-N 0.000 description 2
- 230000001594 aberrant effect Effects 0.000 description 2
- 239000000654 additive Substances 0.000 description 2
- 230000000996 additive effect Effects 0.000 description 2
- 229960000643 adenine Drugs 0.000 description 2
- 125000000217 alkyl group Chemical group 0.000 description 2
- HMFHBZSHGGEWLO-UHFFFAOYSA-N alpha-D-Furanose-Ribose Natural products OCC1OC(O)C(O)C1O HMFHBZSHGGEWLO-UHFFFAOYSA-N 0.000 description 2
- WQZGKKKJIJFFOK-PHYPRBDBSA-N alpha-D-galactose Chemical compound OC[C@H]1O[C@H](O)[C@H](O)[C@@H](O)[C@H]1O WQZGKKKJIJFFOK-PHYPRBDBSA-N 0.000 description 2
- 210000002565 arteriole Anatomy 0.000 description 2
- QVGXLLKOCUKJST-UHFFFAOYSA-N atomic oxygen Chemical compound [O] QVGXLLKOCUKJST-UHFFFAOYSA-N 0.000 description 2
- 230000003190 augmentative effect Effects 0.000 description 2
- WQZGKKKJIJFFOK-VFUOTHLCSA-N beta-D-glucose Chemical compound OC[C@H]1O[C@@H](O)[C@H](O)[C@@H](O)[C@@H]1O WQZGKKKJIJFFOK-VFUOTHLCSA-N 0.000 description 2
- 210000004899 c-terminal region Anatomy 0.000 description 2
- 230000015556 catabolic process Effects 0.000 description 2
- 101150073031 cdk2 gene Proteins 0.000 description 2
- 230000011712 cell development Effects 0.000 description 2
- 238000000205 computational method Methods 0.000 description 2
- 239000000562 conjugate Substances 0.000 description 2
- 230000001186 cumulative effect Effects 0.000 description 2
- 229940043378 cyclin-dependent kinase inhibitor Drugs 0.000 description 2
- XUJNEKJLAYXESH-UHFFFAOYSA-N cysteine Natural products SCC(N)C(O)=O XUJNEKJLAYXESH-UHFFFAOYSA-N 0.000 description 2
- 229940104302 cytosine Drugs 0.000 description 2
- 210000000172 cytosol Anatomy 0.000 description 2
- 238000006731 degradation reaction Methods 0.000 description 2
- 230000004069 differentiation Effects 0.000 description 2
- 201000010099 disease Diseases 0.000 description 2
- BFMYDTVEBKDAKJ-UHFFFAOYSA-L disodium;(2',7'-dibromo-3',6'-dioxido-3-oxospiro[2-benzofuran-1,9'-xanthene]-4'-yl)mercury;hydrate Chemical compound O.[Na+].[Na+].O1C(=O)C2=CC=CC=C2C21C1=CC(Br)=C([O-])C([Hg])=C1OC1=C2C=C(Br)C([O-])=C1 BFMYDTVEBKDAKJ-UHFFFAOYSA-L 0.000 description 2
- 238000009826 distribution Methods 0.000 description 2
- AUZONCFQVSMFAP-UHFFFAOYSA-N disulfiram Chemical compound CCN(CC)C(=S)SSC(=S)N(CC)CC AUZONCFQVSMFAP-UHFFFAOYSA-N 0.000 description 2
- 239000000975 dye Substances 0.000 description 2
- 238000002592 echocardiography Methods 0.000 description 2
- 229960000301 factor viii Drugs 0.000 description 2
- 230000001605 fetal effect Effects 0.000 description 2
- 238000000684 flow cytometry Methods 0.000 description 2
- 229930182830 galactose Natural products 0.000 description 2
- 239000008103 glucose Substances 0.000 description 2
- RWSXRVCMGQZWBV-WDSKDSINSA-N glutathione Chemical compound OC(=O)[C@@H](N)CCC(=O)N[C@@H](CS)C(=O)NCC(O)=O RWSXRVCMGQZWBV-WDSKDSINSA-N 0.000 description 2
- 230000003394 haemopoietic effect Effects 0.000 description 2
- 102000028546 heme binding Human genes 0.000 description 2
- 108091022907 heme binding Proteins 0.000 description 2
- FDGQSTZJBFJUBT-UHFFFAOYSA-N hypoxanthine Chemical compound O=C1NC=NC2=C1NC=N2 FDGQSTZJBFJUBT-UHFFFAOYSA-N 0.000 description 2
- 230000007954 hypoxia Effects 0.000 description 2
- 239000000411 inducer Substances 0.000 description 2
- 229960003786 inosine Drugs 0.000 description 2
- 238000004519 manufacturing process Methods 0.000 description 2
- 210000004379 membrane Anatomy 0.000 description 2
- 239000012528 membrane Substances 0.000 description 2
- 229910052751 metal Inorganic materials 0.000 description 2
- 239000002184 metal Substances 0.000 description 2
- 125000004170 methylsulfonyl group Chemical group [H]C([H])([H])S(*)(=O)=O 0.000 description 2
- 125000001893 nitrooxy group Chemical group [O-][N+](=O)O* 0.000 description 2
- 230000005937 nuclear translocation Effects 0.000 description 2
- 230000002018 overexpression Effects 0.000 description 2
- 239000001301 oxygen Substances 0.000 description 2
- 239000012188 paraffin wax Substances 0.000 description 2
- 108040007629 peroxidase activity proteins Proteins 0.000 description 2
- 229920000768 polyamine Polymers 0.000 description 2
- 230000029279 positive regulation of transcription, DNA-dependent Effects 0.000 description 2
- 238000002360 preparation method Methods 0.000 description 2
- 125000002924 primary amino group Chemical group [H]N([H])* 0.000 description 2
- 230000002250 progressing effect Effects 0.000 description 2
- 239000011541 reaction mixture Substances 0.000 description 2
- 238000003757 reverse transcription PCR Methods 0.000 description 2
- 229920002477 rna polymer Polymers 0.000 description 2
- 150000003839 salts Chemical class 0.000 description 2
- 238000010561 standard procedure Methods 0.000 description 2
- 230000035882 stress Effects 0.000 description 2
- 125000001424 substituent group Chemical group 0.000 description 2
- 238000006467 substitution reaction Methods 0.000 description 2
- 238000001356 surgical procedure Methods 0.000 description 2
- 238000003786 synthesis reaction Methods 0.000 description 2
- 238000002560 therapeutic procedure Methods 0.000 description 2
- 230000005945 translocation Effects 0.000 description 2
- 125000000876 trifluoromethoxy group Chemical group FC(F)(F)O* 0.000 description 2
- 235000005282 vitamin D3 Nutrition 0.000 description 2
- QYSXJUFSXHHAJI-YRZJJWOYSA-N vitamin D3 Chemical compound C1(/[C@@H]2CC[C@@H]([C@]2(CCC1)C)[C@H](C)CCCC(C)C)=C\C=C1\C[C@@H](O)CCC1=C QYSXJUFSXHHAJI-YRZJJWOYSA-N 0.000 description 2
- 239000011647 vitamin D3 Substances 0.000 description 2
- 229940021056 vitamin d3 Drugs 0.000 description 2
- 238000005406 washing Methods 0.000 description 2
- XLYOFNOQVPJJNP-UHFFFAOYSA-N water Chemical compound O XLYOFNOQVPJJNP-UHFFFAOYSA-N 0.000 description 2
- BHQCQFFYRZLCQQ-UHFFFAOYSA-N (3alpha,5alpha,7alpha,12alpha)-3,7,12-trihydroxy-cholan-24-oic acid Natural products OC1CC2CC(O)CCC2(C)C2C1C1CCC(C(CCC(O)=O)C)C1(C)C(O)C2 BHQCQFFYRZLCQQ-UHFFFAOYSA-N 0.000 description 1
- BJHCYTJNPVGSBZ-YXSASFKJSA-N 1-[4-[6-amino-5-[(Z)-methoxyiminomethyl]pyrimidin-4-yl]oxy-2-chlorophenyl]-3-ethylurea Chemical compound CCNC(=O)Nc1ccc(Oc2ncnc(N)c2\C=N/OC)cc1Cl BJHCYTJNPVGSBZ-YXSASFKJSA-N 0.000 description 1
- VGONTNSXDCQUGY-RRKCRQDMSA-N 2'-deoxyinosine Chemical compound C1[C@H](O)[C@@H](CO)O[C@H]1N1C(N=CNC2=O)=C2N=C1 VGONTNSXDCQUGY-RRKCRQDMSA-N 0.000 description 1
- WJBNIBFTNGZFBW-DJLDLDEBSA-N 2'-deoxynebularine Chemical compound C1[C@H](O)[C@@H](CO)O[C@H]1N1C2=NC=NC=C2N=C1 WJBNIBFTNGZFBW-DJLDLDEBSA-N 0.000 description 1
- HDZZVAMISRMYHH-UHFFFAOYSA-N 2-(4-aminopyrrolo[2,3-d]pyrimidin-7-yl)-5-(hydroxymethyl)oxolane-3,4-diol Chemical compound C1=CC=2C(N)=NC=NC=2N1C1OC(CO)C(O)C1O HDZZVAMISRMYHH-UHFFFAOYSA-N 0.000 description 1
- JRYMOPZHXMVHTA-DAGMQNCNSA-N 2-amino-7-[(2r,3r,4s,5r)-3,4-dihydroxy-5-(hydroxymethyl)oxolan-2-yl]-1h-pyrrolo[2,3-d]pyrimidin-4-one Chemical compound C1=CC=2C(=O)NC(N)=NC=2N1[C@@H]1O[C@H](CO)[C@@H](O)[C@H]1O JRYMOPZHXMVHTA-DAGMQNCNSA-N 0.000 description 1
- 108020005345 3' Untranslated Regions Proteins 0.000 description 1
- 108020003589 5' Untranslated Regions Proteins 0.000 description 1
- IHHSSHCBRVYGJX-UHFFFAOYSA-N 6-chloro-2-methoxyacridin-9-amine Chemical compound C1=C(Cl)C=CC2=C(N)C3=CC(OC)=CC=C3N=C21 IHHSSHCBRVYGJX-UHFFFAOYSA-N 0.000 description 1
- VKKXEIQIGGPMHT-UHFFFAOYSA-N 7h-purine-2,8-diamine Chemical class NC1=NC=C2NC(N)=NC2=N1 VKKXEIQIGGPMHT-UHFFFAOYSA-N 0.000 description 1
- NQAZHXBSLFDVKM-KVQBGUIXSA-N 9-[(2r,4s,5r)-4-hydroxy-5-(hydroxymethyl)oxolan-2-yl]-3h-purine-2,6-dione Chemical compound C1[C@H](O)[C@@H](CO)O[C@H]1N1C(NC(=O)NC2=O)=C2N=C1 NQAZHXBSLFDVKM-KVQBGUIXSA-N 0.000 description 1
- VHUUQVKOLVNVRT-UHFFFAOYSA-N Ammonium hydroxide Chemical compound [NH4+].[OH-] VHUUQVKOLVNVRT-UHFFFAOYSA-N 0.000 description 1
- 206010002091 Anaesthesia Diseases 0.000 description 1
- 108091007914 CDKs Proteins 0.000 description 1
- 241000283707 Capra Species 0.000 description 1
- 102000011727 Caspases Human genes 0.000 description 1
- 108010076667 Caspases Proteins 0.000 description 1
- 239000004380 Cholic acid Substances 0.000 description 1
- 208000032544 Cicatrix Diseases 0.000 description 1
- 102000012422 Collagen Type I Human genes 0.000 description 1
- 108010022452 Collagen Type I Proteins 0.000 description 1
- 239000004971 Cross linker Substances 0.000 description 1
- 102000016736 Cyclin Human genes 0.000 description 1
- 108010068192 Cyclin A Proteins 0.000 description 1
- 102000002554 Cyclin A Human genes 0.000 description 1
- 102100030497 Cytochrome c Human genes 0.000 description 1
- 108010075031 Cytochromes c Proteins 0.000 description 1
- 108010008286 DNA nucleotidylexotransferase Proteins 0.000 description 1
- 230000004568 DNA-binding Effects 0.000 description 1
- 101100481408 Danio rerio tie2 gene Proteins 0.000 description 1
- AHCYMLUZIRLXAA-SHYZEUOFSA-N Deoxyuridine 5'-triphosphate Chemical compound O1[C@H](COP(O)(=O)OP(O)(=O)OP(O)(O)=O)[C@@H](O)C[C@@H]1N1C(=O)NC(=O)C=C1 AHCYMLUZIRLXAA-SHYZEUOFSA-N 0.000 description 1
- 102100023471 E-selectin Human genes 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
- 238000002965 ELISA Methods 0.000 description 1
- 102000004533 Endonucleases Human genes 0.000 description 1
- 108010042407 Endonucleases Proteins 0.000 description 1
- 108010067770 Endopeptidase K Proteins 0.000 description 1
- 102100030013 Endoribonuclease Human genes 0.000 description 1
- 101710199605 Endoribonuclease Proteins 0.000 description 1
- 241000206602 Eukaryota Species 0.000 description 1
- 206010063560 Excessive granulation tissue Diseases 0.000 description 1
- 108060002716 Exonuclease Proteins 0.000 description 1
- 102000001690 Factor VIII Human genes 0.000 description 1
- 108010054218 Factor VIII Proteins 0.000 description 1
- 102000003974 Fibroblast growth factor 2 Human genes 0.000 description 1
- 108090000379 Fibroblast growth factor 2 Proteins 0.000 description 1
- 206010016654 Fibrosis Diseases 0.000 description 1
- 108010024636 Glutathione Proteins 0.000 description 1
- 108091027874 Group I catalytic intron Proteins 0.000 description 1
- 102000002812 Heat-Shock Proteins Human genes 0.000 description 1
- 108010004889 Heat-Shock Proteins Proteins 0.000 description 1
- 102000002737 Heme Oxygenase-1 Human genes 0.000 description 1
- 108010018924 Heme Oxygenase-1 Proteins 0.000 description 1
- 241000724709 Hepatitis delta virus Species 0.000 description 1
- 101000916059 Homo sapiens C-X-C chemokine receptor type 2 Proteins 0.000 description 1
- 101000622123 Homo sapiens E-selectin Proteins 0.000 description 1
- 101000599852 Homo sapiens Intercellular adhesion molecule 1 Proteins 0.000 description 1
- 101000716729 Homo sapiens Kit ligand Proteins 0.000 description 1
- 101000617130 Homo sapiens Stromal cell-derived factor 1 Proteins 0.000 description 1
- UFHFLCQGNIYNRP-UHFFFAOYSA-N Hydrogen Chemical compound [H][H] UFHFLCQGNIYNRP-UHFFFAOYSA-N 0.000 description 1
- UGQMRVRMYYASKQ-UHFFFAOYSA-N Hypoxanthine nucleoside Natural products OC1C(O)C(CO)OC1N1C(NC=NC2=O)=C2N=C1 UGQMRVRMYYASKQ-UHFFFAOYSA-N 0.000 description 1
- 108020005350 Initiator Codon Proteins 0.000 description 1
- 108090000723 Insulin-Like Growth Factor I Proteins 0.000 description 1
- 102000014429 Insulin-like growth factor Human genes 0.000 description 1
- 102100034343 Integrase Human genes 0.000 description 1
- 102100037877 Intercellular adhesion molecule 1 Human genes 0.000 description 1
- FFEARJCKVFRZRR-BYPYZUCNSA-N L-methionine Chemical compound CSCC[C@H](N)C(O)=O FFEARJCKVFRZRR-BYPYZUCNSA-N 0.000 description 1
- OUYCCCASQSFEME-QMMMGPOBSA-N L-tyrosine Chemical compound OC(=O)[C@@H](N)CC1=CC=C(O)C=C1 OUYCCCASQSFEME-QMMMGPOBSA-N 0.000 description 1
- 102000014962 Monocyte Chemoattractant Proteins Human genes 0.000 description 1
- 108010064136 Monocyte Chemoattractant Proteins Proteins 0.000 description 1
- 241001529936 Murinae Species 0.000 description 1
- 101100481410 Mus musculus Tek gene Proteins 0.000 description 1
- 241000699670 Mus sp. Species 0.000 description 1
- 241000221960 Neurospora Species 0.000 description 1
- 101001068640 Nicotiana tabacum Basic form of pathogenesis-related protein 1 Proteins 0.000 description 1
- 102100028452 Nitric oxide synthase, endothelial Human genes 0.000 description 1
- 101710090055 Nitric oxide synthase, endothelial Proteins 0.000 description 1
- JCXJVPUVTGWSNB-UHFFFAOYSA-N Nitrogen dioxide Chemical compound O=[N]=O JCXJVPUVTGWSNB-UHFFFAOYSA-N 0.000 description 1
- 238000000636 Northern blotting Methods 0.000 description 1
- 108091007494 Nucleic acid- binding domains Proteins 0.000 description 1
- CTQNGGLPUBDAKN-UHFFFAOYSA-N O-Xylene Chemical compound CC1=CC=CC=C1C CTQNGGLPUBDAKN-UHFFFAOYSA-N 0.000 description 1
- 101150089457 PAG gene Proteins 0.000 description 1
- 108091007960 PI3Ks Proteins 0.000 description 1
- 229910019142 PO4 Inorganic materials 0.000 description 1
- 108700020962 Peroxidase Proteins 0.000 description 1
- ABLZXFCXXLZCGV-UHFFFAOYSA-N Phosphorous acid Chemical class OP(O)=O ABLZXFCXXLZCGV-UHFFFAOYSA-N 0.000 description 1
- OAICVXFJPJFONN-UHFFFAOYSA-N Phosphorus Chemical compound [P] OAICVXFJPJFONN-UHFFFAOYSA-N 0.000 description 1
- 108091000080 Phosphotransferase Proteins 0.000 description 1
- 102100024616 Platelet endothelial cell adhesion molecule Human genes 0.000 description 1
- 239000004952 Polyamide Substances 0.000 description 1
- 108010039918 Polylysine Proteins 0.000 description 1
- 239000004721 Polyphenylene oxide Substances 0.000 description 1
- WDVSHHCDHLJJJR-UHFFFAOYSA-N Proflavine Chemical compound C1=CC(N)=CC2=NC3=CC(N)=CC=C3C=C21 WDVSHHCDHLJJJR-UHFFFAOYSA-N 0.000 description 1
- 101150035060 Prx gene Proteins 0.000 description 1
- 108091034057 RNA (poly(A)) Proteins 0.000 description 1
- 108010092799 RNA-directed DNA polymerase Proteins 0.000 description 1
- 238000010240 RT-PCR analysis Methods 0.000 description 1
- 101100383168 Rattus norvegicus Cdkn2b gene Proteins 0.000 description 1
- 230000018199 S phase Effects 0.000 description 1
- 101710113029 Serine/threonine-protein kinase Proteins 0.000 description 1
- 229930006000 Sucrose Natural products 0.000 description 1
- CZMRCDWAGMRECN-UGDNZRGBSA-N Sucrose Chemical compound O[C@H]1[C@H](O)[C@@H](CO)O[C@@]1(CO)O[C@@H]1[C@H](O)[C@@H](O)[C@H](O)[C@@H](CO)O1 CZMRCDWAGMRECN-UGDNZRGBSA-N 0.000 description 1
- 102000019197 Superoxide Dismutase Human genes 0.000 description 1
- 108010012715 Superoxide dismutase Proteins 0.000 description 1
- 108010006785 Taq Polymerase Proteins 0.000 description 1
- AUYYCJSJGJYCDS-LBPRGKRZSA-N Thyrolar Chemical class IC1=CC(C[C@H](N)C(O)=O)=CC(I)=C1OC1=CC=C(O)C(I)=C1 AUYYCJSJGJYCDS-LBPRGKRZSA-N 0.000 description 1
- 102000004357 Transferases Human genes 0.000 description 1
- 108090000992 Transferases Proteins 0.000 description 1
- 108090001012 Transforming Growth Factor beta Proteins 0.000 description 1
- 102000004887 Transforming Growth Factor beta Human genes 0.000 description 1
- 239000007983 Tris buffer Substances 0.000 description 1
- 108010091356 Tumor Protein p73 Proteins 0.000 description 1
- 102000018252 Tumor Protein p73 Human genes 0.000 description 1
- 108010040002 Tumor Suppressor Proteins Proteins 0.000 description 1
- 102000001742 Tumor Suppressor Proteins Human genes 0.000 description 1
- 108091008605 VEGF receptors Proteins 0.000 description 1
- 102000009484 Vascular Endothelial Growth Factor Receptors Human genes 0.000 description 1
- 241000700605 Viruses Species 0.000 description 1
- 208000027418 Wounds and injury Diseases 0.000 description 1
- XJLXINKUBYWONI-DQQFMEOOSA-N [[(2r,3r,4r,5r)-5-(6-aminopurin-9-yl)-3-hydroxy-4-phosphonooxyoxolan-2-yl]methoxy-hydroxyphosphoryl] [(2s,3r,4s,5s)-5-(3-carbamoylpyridin-1-ium-1-yl)-3,4-dihydroxyoxolan-2-yl]methyl phosphate Chemical compound NC(=O)C1=CC=C[N+]([C@@H]2[C@H]([C@@H](O)[C@H](COP([O-])(=O)OP(O)(=O)OC[C@@H]3[C@H]([C@@H](OP(O)(O)=O)[C@@H](O3)N3C4=NC=NC(N)=C4N=C3)O)O2)O)=C1 XJLXINKUBYWONI-DQQFMEOOSA-N 0.000 description 1
- 230000002159 abnormal effect Effects 0.000 description 1
- 239000002253 acid Substances 0.000 description 1
- 150000001251 acridines Chemical class 0.000 description 1
- 239000011543 agarose gel Substances 0.000 description 1
- 150000001298 alcohols Chemical class 0.000 description 1
- 125000003282 alkyl amino group Chemical group 0.000 description 1
- 125000005600 alkyl phosphonate group Chemical group 0.000 description 1
- 230000002152 alkylating effect Effects 0.000 description 1
- 125000003277 amino group Chemical group 0.000 description 1
- 125000002431 aminoalkoxy group Chemical group 0.000 description 1
- 125000005122 aminoalkylamino group Chemical group 0.000 description 1
- 239000000908 ammonium hydroxide Substances 0.000 description 1
- 230000037005 anaesthesia Effects 0.000 description 1
- 238000004458 analytical method Methods 0.000 description 1
- 230000033115 angiogenesis Effects 0.000 description 1
- 238000010171 animal model Methods 0.000 description 1
- 230000002424 anti-apoptotic effect Effects 0.000 description 1
- 230000000692 anti-sense effect Effects 0.000 description 1
- 229930185229 antidesmin Natural products 0.000 description 1
- 239000000427 antigen Substances 0.000 description 1
- 108091007433 antigens Proteins 0.000 description 1
- 102000036639 antigens Human genes 0.000 description 1
- 229940027998 antiseptic and disinfectant acridine derivative Drugs 0.000 description 1
- 210000000709 aorta Anatomy 0.000 description 1
- 230000005775 apoptotic pathway Effects 0.000 description 1
- 238000003556 assay Methods 0.000 description 1
- 239000012298 atmosphere Substances 0.000 description 1
- 230000003416 augmentation Effects 0.000 description 1
- 239000011324 bead Substances 0.000 description 1
- 230000008901 benefit Effects 0.000 description 1
- 230000033228 biological regulation Effects 0.000 description 1
- 210000004369 blood Anatomy 0.000 description 1
- 239000008280 blood Substances 0.000 description 1
- 210000004271 bone marrow stromal cell Anatomy 0.000 description 1
- 210000004556 brain Anatomy 0.000 description 1
- 239000000872 buffer Substances 0.000 description 1
- 238000010804 cDNA synthesis Methods 0.000 description 1
- 229910052793 cadmium Inorganic materials 0.000 description 1
- BDOSMKKIYDKNTQ-UHFFFAOYSA-N cadmium atom Chemical compound [Cd] BDOSMKKIYDKNTQ-UHFFFAOYSA-N 0.000 description 1
- 238000004364 calculation method Methods 0.000 description 1
- 125000001369 canonical nucleoside group Chemical group 0.000 description 1
- 239000000298 carbocyanine Substances 0.000 description 1
- 210000001054 cardiac fibroblast Anatomy 0.000 description 1
- 230000005961 cardioprotection Effects 0.000 description 1
- 230000003293 cardioprotective effect Effects 0.000 description 1
- 239000000969 carrier Substances 0.000 description 1
- 239000003054 catalyst Substances 0.000 description 1
- 108020001778 catalytic domains Proteins 0.000 description 1
- 230000012292 cell migration Effects 0.000 description 1
- 238000002659 cell therapy Methods 0.000 description 1
- 230000007248 cellular mechanism Effects 0.000 description 1
- 230000004700 cellular uptake Effects 0.000 description 1
- 238000012512 characterization method Methods 0.000 description 1
- 125000003636 chemical group Chemical group 0.000 description 1
- 239000003153 chemical reaction reagent Substances 0.000 description 1
- HVYWMOMLDIMFJA-DPAQBDIFSA-N cholesterol group Chemical group [C@@H]1(CC[C@H]2[C@@H]3CC=C4C[C@@H](O)CC[C@]4(C)[C@H]3CC[C@]12C)[C@H](C)CCCC(C)C HVYWMOMLDIMFJA-DPAQBDIFSA-N 0.000 description 1
- BHQCQFFYRZLCQQ-OELDTZBJSA-N cholic acid Chemical compound C([C@H]1C[C@H]2O)[C@H](O)CC[C@]1(C)[C@@H]1[C@@H]2[C@@H]2CC[C@H]([C@@H](CCC(O)=O)C)[C@@]2(C)[C@@H](O)C1 BHQCQFFYRZLCQQ-OELDTZBJSA-N 0.000 description 1
- 229960002471 cholic acid Drugs 0.000 description 1
- 235000019416 cholic acid Nutrition 0.000 description 1
- 230000004087 circulation Effects 0.000 description 1
- 229910017052 cobalt Inorganic materials 0.000 description 1
- 239000010941 cobalt Substances 0.000 description 1
- GUTLYIVDDKVIGB-UHFFFAOYSA-N cobalt atom Chemical compound [Co] GUTLYIVDDKVIGB-UHFFFAOYSA-N 0.000 description 1
- 229940096422 collagen type i Drugs 0.000 description 1
- 230000009918 complex formation Effects 0.000 description 1
- 150000001875 compounds Chemical class 0.000 description 1
- 239000005289 controlled pore glass Substances 0.000 description 1
- 230000002596 correlated effect Effects 0.000 description 1
- 125000000151 cysteine group Chemical group N[C@@H](CS)C(=O)* 0.000 description 1
- 150000001945 cysteines Chemical class 0.000 description 1
- 210000000805 cytoplasm Anatomy 0.000 description 1
- 239000000824 cytostatic agent Substances 0.000 description 1
- 230000001085 cytostatic effect Effects 0.000 description 1
- 230000034994 death Effects 0.000 description 1
- 230000003247 decreasing effect Effects 0.000 description 1
- 230000006735 deficit Effects 0.000 description 1
- 238000012217 deletion Methods 0.000 description 1
- 230000037430 deletion Effects 0.000 description 1
- KXGVEGMKQFWNSR-UHFFFAOYSA-N deoxycholic acid Natural products C1CC2CC(O)CCC2(C)C2C1C1CCC(C(CCC(O)=O)C)C1(C)C(O)C2 KXGVEGMKQFWNSR-UHFFFAOYSA-N 0.000 description 1
- VGONTNSXDCQUGY-UHFFFAOYSA-N desoxyinosine Natural products C1C(O)C(CO)OC1N1C(NC=NC2=O)=C2N=C1 VGONTNSXDCQUGY-UHFFFAOYSA-N 0.000 description 1
- 238000001514 detection method Methods 0.000 description 1
- 230000001627 detrimental effect Effects 0.000 description 1
- 239000005546 dideoxynucleotide Substances 0.000 description 1
- 235000005911 diet Nutrition 0.000 description 1
- 230000037213 diet Effects 0.000 description 1
- 230000029087 digestion Effects 0.000 description 1
- 150000004662 dithiols Chemical class 0.000 description 1
- NAGJZTKCGNOGPW-UHFFFAOYSA-N dithiophosphoric acid Chemical class OP(O)(S)=S NAGJZTKCGNOGPW-UHFFFAOYSA-N 0.000 description 1
- 230000003828 downregulation Effects 0.000 description 1
- 238000001962 electrophoresis Methods 0.000 description 1
- 238000004520 electroporation Methods 0.000 description 1
- 210000001671 embryonic stem cell Anatomy 0.000 description 1
- 210000002889 endothelial cell Anatomy 0.000 description 1
- 210000003038 endothelium Anatomy 0.000 description 1
- 230000002708 enhancing effect Effects 0.000 description 1
- 102000013165 exonuclease Human genes 0.000 description 1
- 210000004700 fetal blood Anatomy 0.000 description 1
- 210000002950 fibroblast Anatomy 0.000 description 1
- 230000004761 fibrosis Effects 0.000 description 1
- 229960002143 fluorescein Drugs 0.000 description 1
- 238000000799 fluorescence microscopy Methods 0.000 description 1
- 239000012634 fragment Substances 0.000 description 1
- 238000013467 fragmentation Methods 0.000 description 1
- 238000006062 fragmentation reaction Methods 0.000 description 1
- 230000004927 fusion Effects 0.000 description 1
- 230000002068 genetic effect Effects 0.000 description 1
- 229960003180 glutathione Drugs 0.000 description 1
- 210000001126 granulation tissue Anatomy 0.000 description 1
- 244000144993 groups of animals Species 0.000 description 1
- 239000003102 growth factor Substances 0.000 description 1
- 150000003278 haem Chemical class 0.000 description 1
- 238000003306 harvesting Methods 0.000 description 1
- 230000035876 healing Effects 0.000 description 1
- 230000036541 health Effects 0.000 description 1
- 125000004404 heteroalkyl group Chemical group 0.000 description 1
- 125000000592 heterocycloalkyl group Chemical group 0.000 description 1
- 150000002402 hexoses Chemical class 0.000 description 1
- 230000002962 histologic effect Effects 0.000 description 1
- 230000009097 homeostatic mechanism Effects 0.000 description 1
- 102000055357 human CXCR2 Human genes 0.000 description 1
- 102000048429 human PAG1 Human genes 0.000 description 1
- 102000046661 human PECAM1 Human genes 0.000 description 1
- AFQIYTIJXGTIEY-UHFFFAOYSA-N hydrogen carbonate;triethylazanium Chemical compound OC(O)=O.CCN(CC)CC AFQIYTIJXGTIEY-UHFFFAOYSA-N 0.000 description 1
- 125000002887 hydroxy group Chemical group [H]O* 0.000 description 1
- 230000002055 immunohistochemical effect Effects 0.000 description 1
- 230000001976 improved effect Effects 0.000 description 1
- 238000000338 in vitro Methods 0.000 description 1
- 238000011065 in-situ storage Methods 0.000 description 1
- 230000002779 inactivation Effects 0.000 description 1
- 238000011534 incubation Methods 0.000 description 1
- 239000003999 initiator Substances 0.000 description 1
- 208000014674 injury Diseases 0.000 description 1
- 238000010212 intracellular staining Methods 0.000 description 1
- 239000007927 intramuscular injection Substances 0.000 description 1
- 238000010255 intramuscular injection Methods 0.000 description 1
- 238000007912 intraperitoneal administration Methods 0.000 description 1
- 239000007928 intraperitoneal injection Substances 0.000 description 1
- 238000002955 isolation Methods 0.000 description 1
- 230000002045 lasting effect Effects 0.000 description 1
- 239000003446 ligand Substances 0.000 description 1
- 125000005647 linker group Chemical group 0.000 description 1
- 150000002632 lipids Chemical class 0.000 description 1
- MHCFAGZWMAWTNR-UHFFFAOYSA-M lithium perchlorate Chemical compound [Li+].[O-]Cl(=O)(=O)=O MHCFAGZWMAWTNR-UHFFFAOYSA-M 0.000 description 1
- 229910001486 lithium perchlorate Inorganic materials 0.000 description 1
- 210000002540 macrophage Anatomy 0.000 description 1
- 238000012423 maintenance Methods 0.000 description 1
- 239000000463 material Substances 0.000 description 1
- 150000002739 metals Chemical class 0.000 description 1
- 229930182817 methionine Natural products 0.000 description 1
- 125000002496 methyl group Chemical group [H]C([H])([H])* 0.000 description 1
- 230000025608 mitochondrion localization Effects 0.000 description 1
- 230000002297 mitogenic effect Effects 0.000 description 1
- 230000000394 mitotic effect Effects 0.000 description 1
- 239000000203 mixture Substances 0.000 description 1
- 230000009456 molecular mechanism Effects 0.000 description 1
- 239000003068 molecular probe Substances 0.000 description 1
- 238000012544 monitoring process Methods 0.000 description 1
- 210000005087 mononuclear cell Anatomy 0.000 description 1
- 230000005914 myocardial expression Effects 0.000 description 1
- 230000032929 negative regulation of cell cycle Effects 0.000 description 1
- 229930027945 nicotinamide-adenine dinucleotide Natural products 0.000 description 1
- 102000037979 non-receptor tyrosine kinases Human genes 0.000 description 1
- 108091008046 non-receptor tyrosine kinases Proteins 0.000 description 1
- 231100000252 nontoxic Toxicity 0.000 description 1
- 230000003000 nontoxic effect Effects 0.000 description 1
- 239000002777 nucleoside Substances 0.000 description 1
- 125000003835 nucleoside group Chemical group 0.000 description 1
- 230000030648 nucleus localization Effects 0.000 description 1
- 235000015097 nutrients Nutrition 0.000 description 1
- 229940046166 oligodeoxynucleotide Drugs 0.000 description 1
- 230000003287 optical effect Effects 0.000 description 1
- 125000001181 organosilyl group Chemical group [SiH3]* 0.000 description 1
- 238000012261 overproduction Methods 0.000 description 1
- 150000002926 oxygen Chemical class 0.000 description 1
- 125000004430 oxygen atom Chemical group O* 0.000 description 1
- 102000041788 p53 family Human genes 0.000 description 1
- 108091075611 p53 family Proteins 0.000 description 1
- 230000008506 pathogenesis Effects 0.000 description 1
- 230000037361 pathway Effects 0.000 description 1
- 239000000863 peptide conjugate Substances 0.000 description 1
- 230000010412 perfusion Effects 0.000 description 1
- 210000003516 pericardium Anatomy 0.000 description 1
- 150000008298 phosphoramidates Chemical class 0.000 description 1
- 150000003014 phosphoric acid esters Chemical class 0.000 description 1
- DHRLEVQXOMLTIM-UHFFFAOYSA-N phosphoric acid;trioxomolybdenum Chemical compound O=[Mo](=O)=O.O=[Mo](=O)=O.O=[Mo](=O)=O.O=[Mo](=O)=O.O=[Mo](=O)=O.O=[Mo](=O)=O.O=[Mo](=O)=O.O=[Mo](=O)=O.O=[Mo](=O)=O.O=[Mo](=O)=O.O=[Mo](=O)=O.O=[Mo](=O)=O.OP(O)(O)=O DHRLEVQXOMLTIM-UHFFFAOYSA-N 0.000 description 1
- 229910052698 phosphorus Inorganic materials 0.000 description 1
- 239000011574 phosphorus Substances 0.000 description 1
- 230000026731 phosphorylation Effects 0.000 description 1
- 238000006366 phosphorylation reaction Methods 0.000 description 1
- 102000020233 phosphotransferase Human genes 0.000 description 1
- QWYZFXLSWMXLDM-UHFFFAOYSA-M pinacyanol iodide Chemical compound [I-].C1=CC2=CC=CC=C2N(CC)C1=CC=CC1=CC=C(C=CC=C2)C2=[N+]1CC QWYZFXLSWMXLDM-UHFFFAOYSA-M 0.000 description 1
- 229920002647 polyamide Polymers 0.000 description 1
- 229920000570 polyether Polymers 0.000 description 1
- 229920000656 polylysine Polymers 0.000 description 1
- 102000040430 polynucleotide Human genes 0.000 description 1
- 108091033319 polynucleotide Proteins 0.000 description 1
- 239000002157 polynucleotide Substances 0.000 description 1
- 239000011148 porous material Substances 0.000 description 1
- 230000003389 potentiating effect Effects 0.000 description 1
- 239000002243 precursor Substances 0.000 description 1
- 230000037452 priming Effects 0.000 description 1
- 230000000861 pro-apoptotic effect Effects 0.000 description 1
- 108090000765 processed proteins & peptides Proteins 0.000 description 1
- 229960000286 proflavine Drugs 0.000 description 1
- 230000000750 progressive effect Effects 0.000 description 1
- 230000002062 proliferating effect Effects 0.000 description 1
- 230000000069 prophylactic effect Effects 0.000 description 1
- 238000001243 protein synthesis Methods 0.000 description 1
- ZCCUUQDIBDJBTK-UHFFFAOYSA-N psoralen Chemical class C1=C2OC(=O)C=CC2=CC2=C1OC=C2 ZCCUUQDIBDJBTK-UHFFFAOYSA-N 0.000 description 1
- 238000000746 purification Methods 0.000 description 1
- 150000003230 pyrimidines Chemical class 0.000 description 1
- 238000004445 quantitative analysis Methods 0.000 description 1
- 230000002285 radioactive effect Effects 0.000 description 1
- 238000002601 radiography Methods 0.000 description 1
- 238000011552 rat model Methods 0.000 description 1
- 230000009257 reactivity Effects 0.000 description 1
- 230000008707 rearrangement Effects 0.000 description 1
- 238000006479 redox reaction Methods 0.000 description 1
- 230000008439 repair process Effects 0.000 description 1
- 230000003362 replicative effect Effects 0.000 description 1
- 230000008943 replicative senescence Effects 0.000 description 1
- 238000011160 research Methods 0.000 description 1
- 230000027756 respiratory electron transport chain Effects 0.000 description 1
- 230000008458 response to injury Effects 0.000 description 1
- 230000004043 responsiveness Effects 0.000 description 1
- 238000004007 reversed phase HPLC Methods 0.000 description 1
- 229920006395 saturated elastomer Polymers 0.000 description 1
- 230000037387 scars Effects 0.000 description 1
- 230000019491 signal transduction Effects 0.000 description 1
- 230000005476 size effect Effects 0.000 description 1
- 210000002460 smooth muscle Anatomy 0.000 description 1
- 210000000329 smooth muscle myocyte Anatomy 0.000 description 1
- 159000000000 sodium salts Chemical class 0.000 description 1
- 230000009870 specific binding Effects 0.000 description 1
- 238000012453 sprague-dawley rat model Methods 0.000 description 1
- 239000008223 sterile water Substances 0.000 description 1
- 238000007920 subcutaneous administration Methods 0.000 description 1
- 239000007929 subcutaneous injection Substances 0.000 description 1
- 239000005720 sucrose Substances 0.000 description 1
- 238000005987 sulfurization reaction Methods 0.000 description 1
- 230000004083 survival effect Effects 0.000 description 1
- 230000002459 sustained effect Effects 0.000 description 1
- 230000002195 synergetic effect Effects 0.000 description 1
- 230000008685 targeting Effects 0.000 description 1
- 238000012360 testing method Methods 0.000 description 1
- ZRKFYGHZFMAOKI-QMGMOQQFSA-N tgfbeta Chemical compound C([C@H](NC(=O)[C@H](C(C)C)NC(=O)CNC(=O)[C@H](CCC(O)=O)NC(=O)[C@H](CCCNC(N)=N)NC(=O)[C@H](CC(N)=O)NC(=O)[C@H](CC(C)C)NC(=O)[C@H]([C@@H](C)O)NC(=O)[C@H](CCC(O)=O)NC(=O)[C@H]([C@@H](C)O)NC(=O)[C@H](CC(C)C)NC(=O)CNC(=O)[C@H](C)NC(=O)[C@H](CO)NC(=O)[C@H](CCC(N)=O)NC(=O)[C@@H](NC(=O)[C@H](C)NC(=O)[C@H](C)NC(=O)[C@@H](NC(=O)[C@H](CC(C)C)NC(=O)[C@@H](N)CCSC)C(C)C)[C@@H](C)CC)C(=O)N[C@@H]([C@@H](C)O)C(=O)N[C@@H](C(C)C)C(=O)N[C@@H](CC=1C=CC=CC=1)C(=O)N[C@@H](C)C(=O)N1[C@@H](CCC1)C(=O)N[C@@H]([C@@H](C)O)C(=O)N[C@@H](CC(N)=O)C(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](C)C(=O)N[C@@H](CC=1C=CC=CC=1)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](C)C(=O)N[C@@H](CC(C)C)C(=O)N1[C@@H](CCC1)C(=O)N1[C@@H](CCC1)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CO)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CC(C)C)C(O)=O)C1=CC=C(O)C=C1 ZRKFYGHZFMAOKI-QMGMOQQFSA-N 0.000 description 1
- 125000003396 thiol group Chemical group [H]S* 0.000 description 1
- 239000005495 thyroid hormone Substances 0.000 description 1
- 229940036555 thyroid hormone Drugs 0.000 description 1
- 238000010361 transduction Methods 0.000 description 1
- 230000026683 transduction Effects 0.000 description 1
- 238000011269 treatment regimen Methods 0.000 description 1
- LENZDBCJOHFCAS-UHFFFAOYSA-N tris Chemical compound OCC(N)(CO)CO LENZDBCJOHFCAS-UHFFFAOYSA-N 0.000 description 1
- 101150072359 trx gene Proteins 0.000 description 1
- 239000000225 tumor suppressor protein Substances 0.000 description 1
- OUYCCCASQSFEME-UHFFFAOYSA-N tyrosine Natural products OC(=O)C(N)CC1=CC=C(O)C=C1 OUYCCCASQSFEME-UHFFFAOYSA-N 0.000 description 1
- 238000000870 ultraviolet spectroscopy Methods 0.000 description 1
- 230000009452 underexpressoin Effects 0.000 description 1
- 230000003827 upregulation Effects 0.000 description 1
- 229940035893 uracil Drugs 0.000 description 1
- 230000002792 vascular Effects 0.000 description 1
- 229940124676 vascular endothelial growth factor receptor Drugs 0.000 description 1
- 230000000982 vasogenic effect Effects 0.000 description 1
- 230000007998 vessel formation Effects 0.000 description 1
- 230000035899 viability Effects 0.000 description 1
- 108010047303 von Willebrand Factor Proteins 0.000 description 1
- 102100036537 von Willebrand factor Human genes 0.000 description 1
- 238000001262 western blot Methods 0.000 description 1
- 239000008096 xylene Substances 0.000 description 1
Images
Classifications
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K38/00—Medicinal preparations containing peptides
- A61K38/16—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- A61K38/17—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
- A61K38/19—Cytokines; Lymphokines; Interferons
- A61K38/20—Interleukins [IL]
- A61K38/2053—IL-8
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K38/00—Medicinal preparations containing peptides
- A61K38/16—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- A61K38/17—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
- A61K38/19—Cytokines; Lymphokines; Interferons
- A61K38/193—Colony stimulating factors [CSF]
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/075—Ethers or acetals
- A61K31/085—Ethers or acetals having an ether linkage to aromatic ring nuclear carbon
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/33—Heterocyclic compounds
- A61K31/395—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
- A61K31/495—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
- A61K31/505—Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
- A61K31/506—Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim not condensed and containing further heterocyclic rings
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/70—Carbohydrates; Sugars; Derivatives thereof
- A61K31/7088—Compounds having three or more nucleosides or nucleotides
- A61K31/7105—Natural ribonucleic acids, i.e. containing only riboses attached to adenine, guanine, cytosine or uracil and having 3'-5' phosphodiester links
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/70—Carbohydrates; Sugars; Derivatives thereof
- A61K31/7088—Compounds having three or more nucleosides or nucleotides
- A61K31/711—Natural deoxyribonucleic acids, i.e. containing only 2'-deoxyriboses attached to adenine, guanine, cytosine or thymine and having 3'-5' phosphodiester links
-
- 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
- A61K35/34—Muscles; Smooth muscle cells; Heart; Cardiac stem cells; Myoblasts; Myocytes; Cardiomyocytes
-
- 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
- A61K35/44—Vessels; Vascular smooth muscle cells; Endothelial cells; Endothelial progenitor cells
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K38/00—Medicinal preparations containing peptides
- A61K38/16—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- A61K38/17—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
- A61K38/1703—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
- A61K38/1709—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K38/00—Medicinal preparations containing peptides
- A61K38/16—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- A61K38/17—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
- A61K38/18—Growth factors; Growth regulators
- A61K38/1825—Fibroblast growth factor [FGF]
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K38/00—Medicinal preparations containing peptides
- A61K38/16—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- A61K38/17—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
- A61K38/18—Growth factors; Growth regulators
- A61K38/1858—Platelet-derived growth factor [PDGF]
- A61K38/1866—Vascular endothelial growth factor [VEGF]
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K38/00—Medicinal preparations containing peptides
- A61K38/16—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- A61K38/17—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
- A61K38/18—Growth factors; Growth regulators
- A61K38/1891—Angiogenesic factors; Angiogenin
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K38/00—Medicinal preparations containing peptides
- A61K38/16—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- A61K38/17—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
- A61K38/19—Cytokines; Lymphokines; Interferons
- A61K38/195—Chemokines, e.g. RANTES
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K45/00—Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
- A61K45/06—Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P1/00—Drugs for disorders of the alimentary tract or the digestive system
- A61P1/04—Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P1/00—Drugs for disorders of the alimentary tract or the digestive system
- A61P1/16—Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P11/00—Drugs for disorders of the respiratory system
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P13/00—Drugs for disorders of the urinary system
- A61P13/10—Drugs for disorders of the urinary system of the bladder
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P21/00—Drugs for disorders of the muscular or neuromuscular system
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P25/00—Drugs for disorders of the nervous system
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P29/00—Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P31/00—Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
- A61P31/04—Antibacterial agents
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P35/00—Antineoplastic agents
-
- 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
- A61P7/00—Drugs for disorders of the blood or the extracellular fluid
- A61P7/02—Antithrombotic agents; Anticoagulants; Platelet aggregation inhibitors
-
- 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
-
- 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
- A61P9/04—Inotropic agents, i.e. stimulants of cardiac contraction; Drugs for heart failure
-
- 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
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K16/00—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
- C07K16/18—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
- C07K16/28—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
- C07K16/2866—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against receptors for cytokines, lymphokines, interferons
-
- 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
- C12N15/00—Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
- C12N15/09—Recombinant DNA-technology
- C12N15/11—DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
- C12N15/113—Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
-
- 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
- C12N15/00—Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
- C12N15/09—Recombinant DNA-technology
- C12N15/11—DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
- C12N15/113—Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
- C12N15/1137—Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing against enzymes
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N5/00—Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
- C12N5/06—Animal cells or tissues; Human cells or tissues
- C12N5/0602—Vertebrate cells
- C12N5/069—Vascular Endothelial cells
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K2039/505—Medicinal preparations containing antigens or antibodies comprising antibodies
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2317/00—Immunoglobulins specific features
- C07K2317/70—Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
- C07K2317/76—Antagonist effect on antigen, e.g. neutralization or inhibition of binding
-
- G—PHYSICS
- G01—MEASURING; TESTING
- G01N—INVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
- G01N2510/00—Detection of programmed cell death, i.e. apoptosis
Definitions
- Vascular network formation is the end result of a complex process that begins in the pre-natal period with induction of vasculogenesis by hemangioblasts—cells derived from the human ventral aorta which give rise to both endothelial and hematopoietic elements (8-11).
- CXC chemokines containing the ELR motif regulate migration of human bone marrow-derived endothelial progenitor cells to sites of tissue ischemia.
- selective bone marrow homing and engraftment of hematopoietic progenitors depends on CXCR4 binding to SDF-1 expressed constitutively in the bone marrow (28-30)
- interruption of CXCR4/SDF-1 interactions could redirect trafficking of human bone marrow-derived endothelial progenitor cells to sites of tissue ischemia, thereby augmenting therapeutic vasculogenesis.
- CXC chemokines, including IL-8, Gro-alpha, and SDF-1 play a central role in regulating human adult bone marrow-dependent vasculogenesis.
- This invention provides a method of treating a disorder of a subject's heart involving loss of cardiomyocytes which comprises administering to the subject an amount of an agent effective to cause cardiomyocyte proliferation within the subject's heart so as to thereby treat the disorder.
- This invention further provides the instant method wherein the agent is human endothelial progenitor cells.
- This invention further provides the instant method comprising administering an effective amount of a second agent that increases the cardiomyocyte proliferation caused by the human endothelial progenitor cells.
- This invention also provides a method of determining the susceptibility of a cardiomyocyte in a subject to apoptosis comprising:
- FIGS. 1 A- 1 D IL-8/Gro-Alpha CXC Chemokines Regulate Migration Of Human Endothelial progenitor cells (angioblasts) To Myocardial Tissue In Vivo And Subsequent Development Of Vasculogenesis.
- FIGS. 2 A- 2 C Blocking CXCR4/SDF-1 Interactions Redirects Intravenously Injected Human Endothelial progenitor cells From Bone Marrow To Ischemic Myocardium.
- (B) and (C) depict the effects of mAbs against CXCR4, SDF-1 or CD34 on trafficking of human CD34+endothelial progenitor cells (angioblasts) to rat bone marrow and myocardium following LAD ligation.
- Co-administration of anti-CXCR4 or anti-SDF-1 significantly reduced trafficking of intravenously injected human CD34+ cells to rat bone marrow at 48 hours and increased trafficking to ischemic myocardium, whereas anti-CD34 mAb had no effect (results are expressed as mean+sem of bone marrow and cardiac studies performed in three LAD-ligated animals at 48 hours after injection).
- FIGS. 3 A- 3 F Redirected Trafficking Of Human Endothelial progenitor cells (angioblasts) To The Site Of Infarction Induces Vasculogenesis And Protects Cardiomyocytes against Apoptosis.
- the infarct zones of rats receiving either 10 3 or 10 5 endothelial progenitor cells (angioblasts) show myocardial scars composed of paucicellular, dense fibrous tissue stained blue by trichrome (x400).
- the infarct zones of rats injected with 10 5 endothelial progenitor cells plus anti-CXCR4 mAb show significant increase in cellularity of granulation tissue, minimal matrix deposition and fibrosis, and numerous medium-sized capillaries of human origin.
- the infarct zones of rats injected with 2 ⁇ 10 5 endothelial progenitor cells show a similar reduction in fibrous tissue and increase in medium-sized capillaries, and an additional increase in large-sized vessels of human origin.
- (B) and (C) show the relationship between the number of human CD117 bright endothelial progenitor cells injected intravenously (10 3 , 10 5 , 10 5 plus anti-CXCR4 mAb, and 2 ⁇ 10 5 ) and development of rat infarct bed vasculogenesis at two weeks, defined as the mean number of capillaries/high power field (hpf) with medium- or large-sized lumen diameter (respectively, 0.02 mm mean diameter with 3-6 contiguous endothelial lining cells and 0.05 mm mean diameter with >6 contiguous endothelial lining cells). Results are expressed as the mean+sem of at least 15 hpf in three separate experiments.
- (D) shows that co-administration of anti-CXCR4 mAb together with the highest concentration of endothelial progenitor cells (angioblasts), 2 ⁇ 10 5 , resulted in a further 23% increase in growth of large-lumen capillaries. More strikingly, there was a further 2-fold increase in capillary numbers when 2 ⁇ 10 5 endothelial progenitor cells were injected intravenously after direct intracardiac delivery of 1.0 ⁇ g/ml SDF-1 into infarcted hearts (p ⁇ 0.01) (results are expressed as mean+sem of three separate experiments).
- (E) shows that at 2 weeks the numbers of apoptotic myocytes at the peri-infarct rim, defined by concomitant staining with anti-desmin mAb and DNA end-labeling using TUNEL technique, are significantly reduced in rats receiving either 2.0 ⁇ 10 5 endothelial progenitor cells or 10 5 endothelial progenitor cells together with anti-CXCR4 mAb in comparison to rats receiving 10 3 or 10 5 endothelial progenitor cells (p ⁇ 0.01) (results are expressed as mean+sem of three separate experiments).
- (F) shows that co-administration of anti-CXCR4 mAb or intracardiac injection of SDF-1 resulted in further reductions in cardiomyocyte apoptosis of 65% and 76%, respectively, at two weeks (both p ⁇ 0.001) (results are expressed as mean+sem of three separate experiments).
- FIGS. 4 A- 4 H Infarct Bed Vasculogenesis Improves Long-Term Myocardial Function Through Mechanisms Involving Both Cardiomyocyte Protection and Proliferation/Regeneration.
- (A) and (B) show the relationship between the number of human CD117 bright endothelial progenitor cells (angioblasts) injected intravenously (10 3 , 10 5 , 10 5 plus anti-CXCR4 mAb, and 2 ⁇ 10 5 ) and improvement in myocardial function at 15 weeks, defined as mean improvement in left ventricular ejection fraction (LVEF) (A) and mean reduction in left ventricular area at end-systole (LVAs)
- Bar graph shows that the group of animals receiving 2 ⁇ 10 5 human endothelial progenitor cells has a significantly higher index of cell cycling cardiomyocytes at the peri-infarct region than saline controls or sham operated animals (both p ⁇ 0.01). No difference between the groups is seen at sites distal to the infarct.
- (E) shows that the index of cell-cycling cardiomyocytes at the peri-infarct rim was increased by a further 1.9-fold when 2 ⁇ 10 5 human endothelial progenitor cells were intravenously co-administered together with SDF-1 injected directly into the ischemic myocardium alone (p ⁇ 0.01), or an 8-fold cumulative increase in cell-cycling cardiomyocytes at two weeks compared with LAD-ligated controls receiving saline (results are expressed as mean+sem of three separate experiments).
- (F) shows that intravenous co-administration of anti-CXCR4 mAb, or intracardiac co-administration of SDF-1, but not IL-8, results in 2.8 to 4-fold greater LVEF improvement, determined by echocardiography, compared with intravenous injection of 2 ⁇ 10 5 endothelial progenitor cells alone (p ⁇ 0.01) (results are expressed as mean+sem of three separate experiments).
- (G) shows that at 15 weeks the mean proportion of scar/normal left ventricular myocardium in rats receiving either or 10 5 endothelial progenitor cells together with anti-CXCR4 mAb was significantly reduced in comparison to rats receiving either 10 3 or 10 5 endothelial progenitor cells (angioblasts) alone, or saline (p ⁇ 0.01).
- the group receiving 2.0 ⁇ 10 5 endothelial progenitor cells demonstrated still 38% greater reduction in the ratio of scar/muscle tissue (results are expressed as mean+sem of three separate experiments).
- FIG. 1 Sections of rat hearts stained with Masson's trichrome at 15 weeks after LAD ligation and injection of 2.0 ⁇ 10 6 G-CSF mobilized human cells containing 10 3 (left) or 2.0 ⁇ 10 5 (right) CD117 bright endothelial progenitor cells.
- Hearts of rats receiving 10 3 endothelial progenitor cells had greater loss of anterior wall mass, collagen deposition (lighter gray), and septal hypertrophy compared with hearts of rats receiving 2.0 ⁇ 10 5 endothelial progenitor cells.
- FIG. 5 This figure shows mRNA expression of three genes in the ischemic rat hearts at various time points. You will see that at 48 hours and 2 weeks after LAD ligation and ischemia, HBP23 (the rat homologue of human PAG/NKEF-A,B,C, all of which are part of the family of peroxiredoxins (Prx)) is decreased, and vitamin D3 upregulated protein VDUP-1 is increased. The early (48 hour) reduction in PRX and increase in VDUP-1 results in a compensatory increase in thiol reductase thioredoxin (TRX). Note that endothelial progenitor cell therapy reverses this pattern of mRNA expression.
- HBP23 the rat homologue of human PAG/NKEF-A,B,C, all of which are part of the family of peroxiredoxins (Prx)
- Prx peroxiredoxins
- FIG. 6 DNA sequence (SEQ ID NO:1) corresponding to mRNA encoding VDUP-1.
- FIG. 7 This figure shows catalytic DNA 5′-AT-3′ cleavage sites on VDUP-1 DNA (SEQ ID NO:3) corresponding to the coding region of VDUP-1 mRNA. Cleavage site pairs are uppercase.
- FIG. 8 This figure shows catalytic DNA 5′-GC-3′ cleavage sites on VDUP-1 DNA (SEQ ID NO:3) corresponding to the VDUP-1 mRNA. Cleavage site pairs are uppercase.
- FIG. 9 This figure shows catalytic DNA 5′-GT-3′ cleavage sites on VDUP-1 DNA (SEQ ID NO:3) corresponding to the VDUP-1 mRNA. Cleavage site pairs are uppercase.
- FIG. 10 This figure shows catalytic DNA 5′-AC-3′ cleavage sites on VDUP-1 DNA (SEQ ID NO:3) corresponding to the VDUP-1 mRNA. Cleavage site pairs are uppercase.
- FIG. 11 This figure shows sites which can be cleaved by a hammerhead ribozyme in VDUP-1 DNA (SEQ ID NO:3) corresponding to the VDUP-1 mRNA coding region.
- Uppercase “T” represents cleavage site.
- VEGF vascular endothelial growth factor
- VEGF-R vascular endothelial growth factor receptor
- FGF fibroblast growth factor
- IGF Insulin-like growth factor
- SCF stem cell factor
- G-CSF granulocyte colony stimulating factor
- M-CSF macrophage colony stimulating factor
- GM-CSF granulocyte-macrophage colony stimulating factor
- MCP monocyte chemoattractant protein.
- angioblasts is synonymous with the term “endothelial progenitor cells”.
- CXC chemokine refers to the structure of the chemokine. Each “C” represents a cysteine and “X” represents any amino acid. “CXCR4” refers to CXC receptor 4.
- CC chemokine refers to the structure of the chemokine.
- Each “C” represents a cysteine.
- Catalytic shall mean the functioning of an agent as a catalyst, i.e. an agent that increases the rate of a chemical reaction without itself undergoing a permanent structural change.
- Nucleic acid shall include without limitation any nucleic acid, including, without limitation, DNA, RNA, oligonucleotides, or polynucleotides, and analogs or derivatives thereof.
- the nucleotides that form the nucleic acid may be nucleotide analogs or derivatives thereof.
- the nucleic acid may incorporate non nucleotides.
- nucleotides shall include without limitation nucleotides and analogs or derivatives thereof.
- nucleotides may comprise the bases A, C, G, T and U, as well as derivatives thereof. Derivatives of these bases are well known in the art, and are exemplified in PCR Systems, Reagents and Consumables (Perkin Elmer Catalogue 1996-1997, Roche Molecular Systems, Inc., Branchburg, N.J., U.S.A.).
- “Proliferation” and “regeneration” are used synonymously when referring to cardiomyocytes in this application. “Proliferation” in respect to cardiomyocytes, shall mean a fold increase in proportion of cardiomyocytes entering the cell cycle relative to untreated rat heart.
- Trafficking means the blood-borne migration of cells, in particular angioblast/endothelial progenitor cells.
- This invention provides a method of treating a disorder of a subject's heart involving loss of cardiomyocytes which comprises administering to the subject an amount of an agent effective to cause cardiomyocyte proliferation within the subject's heart so as to thereby treat the disorder.
- the agent is human endothelial progenitor cells.
- the endothelial progenitor cells are bone marrow-derived. In another they are derived from cord blood, or embryonic or fetal sources. Effective amounts of endothelial progenitor cells sufficient to cause cardiomyocyte proliferation can be done based on animal data using routine computational methods. In one embodiment the effective amount is about 1.5 ⁇ 10 5 endothelial progenitor cells per kg body mass to about 3 ⁇ 10 5 per kg body mass. In another embodiment the effective amount is about 3 ⁇ 10 5 per kg body mass to about 4.5 ⁇ 10 5 endothelial progenitor cells per kg body mass.
- the effective amount is about 4.5 ⁇ 10 5 per kg body mass to about 5.5 ⁇ 10 5 endothelial progenitor cells per kg body mass. In another embodiment the effective amount is about 5.5 ⁇ 10 5 per kg body mass to about 7 ⁇ 10 5 endothelial progenitor cells per kg body mass. In another embodiment the effective amount is about 7 ⁇ 10 5 per kg body mass to about 1 ⁇ 10 6 endothelial progenitor cells per kg body mass. In another embodiment the effective amount is about 1 ⁇ 10 6 per kg body mass to about 1.5 ⁇ 10 6 endothelial progenitor cells per kg body mass.
- the effective amount of human endothelial progenitor cells is between about 1.5 ⁇ 10 6 and 4.5 ⁇ 10 6 endothelial progenitor cells per kg of the subject's body mass and In a preferred embodiment the effective amount is about 5 ⁇ 10 5 endothelial progenitor cells per kg of the subject's body mass.
- the endothelial progenitor cells are allogeneic with respect to the subject.
- the subject is an adult or an embryo or a fetus.
- the endothelial progenitor cells are derived from cloned autologous embryonic stem cells.
- the agent induces expression of a mRNA encoding a peroxiredoxin.
- the expression of peroxiredoxin mRNA may be increased, for example, by administration of 2(3)-t-butyl-4-hydroxyanisole (BHA) (see 106) which has been shown to increase expression of Peroxiredoxin-1 when administered by diet.
- BHA 2(3)-t-butyl-4-hydroxyanisole
- Peroxiredoxin mRNA expression such as that of thiol peroxidases, may also be induced by heme, cadmium or cobalt (see 108).
- Peroxiredoxins include, but are not limited to, PAG, HBP23, MSP23, NKEF.
- the agent induces expression of a mRNA encoding NF-E2-related factor 2 (Nrf2).
- Cytomplasmic NF-E2-related factor 2 (Nrf2) expression can be indirectly increased by raising free Nrf2 levels. Since Nrf2 is tightly bound to keap1 in the cytoplasm then reducing expression of keap1 mRNA is a suitable target e.g. by keap1 antisense oligonucleotides or catalytic nucleic acids (see 109 for keap1). Also, blocking the interaction between Nrf2 and Keap1 by inhibiting the interaction of the Neh2 domain of Nrf2 and the DGR domain of keap1, e.g.
- the agent induces dissociation of a Nrf2 protein from a Keap-1.
- the agent inhibits association of a Nrf2 protein with a Keap-1.
- the agent inhibits association of a thiol reductase thioredoxin with a VDUP-1 protein.
- the agent inhibits c-Abl tyrosine kinase activation.
- the agent is STI-571.
- the agent is a CXC chemokine.
- the agent is Stromal-Derived Factor-1, Il-8 or Gro-Alpha.
- the amount of CXC chemokine administered is between 0.2 and 5 ⁇ g/ml at a max volume of 10 ml for a 70 kg human subject. In a preferred embodiment the amount is about 1 ⁇ g/ml.
- the agent is an inhibitor of plasminogen activator inhibitor-1.
- the agent is an antibody directed against an epitope of CXCR4.
- the amount of antibody directed against an epitope of CXCR4 is between 25 and 75 ⁇ g/ml, at a max volume of 10 ml for a 70 kg human subject. A simple calculation is performed for subjects of different mass. In a preferred embodiment the amount is about 5 ⁇ g/ml.
- the instant method further comprises administering an effective amount of a second agent that increases the cardiomyocyte proliferation caused by the human endothelial progenitor cells.
- Effective amounts of the second agent are amounts sufficient to enhance or accelerate cardiomyocyte proliferation in the presence of administered endothelial progenitor cells.
- the endothelial progenitor cells express CD117, CD34, AC133 or a high level of intracellular GATA-2 activity.
- the administering comprises injecting directly into the subject's peripheral circulation, heart muscle, left ventricle, right ventricle, coronary artery, cerebro-spinal fluid, neural tissue, ischemic tissue, or post-ischemic tissue.
- the second agent is an antisense oligonucleotide which specifically inhibits translation of Vitamin D3 Up-Regulated Protein-1 (VDUP-1) mRNA.
- VDUP-1 Vitamin D3 Up-Regulated Protein-1
- VDUP-1 protein (SEQ ID NO:2) expression
- Antisense oligonucleotides are small fragments of DNA and derivatives thereof complementary to a defined sequence on a specified mRNA.
- a VDUP-1 antisense oligonucleotide specifically binds to targets on the VDUP-1 mRNA (SEQ ID NO:1) molecule and in doing so inhibits the translation thereof into VDUP-1 protein (SEQ ID NO:2).
- Antisense oligonucleotide molecules synthesized with a phosphorothioate backbone have proven particularly resistant to exonuclease damage compared to standard deoxyribonucleic acids, and so they are used in preference.
- a phosphorothioate antisense oligonucleotide for VDUP-1 mRNA can be synthesized on an Applied Biosystems (Foster City, Calif.) model 380B DNA synthesizer by standard methods. E.g. Sulfurization can be performed using tetraethylthiuram disulfide/acetonitrile.
- oligodeoxynucleotides can be base deblocked in ammonium hydroxide at 60° C. for 8 h and purified by reversed-phase HPLC [0.1M triethylammonium bicarbonate/acetonitrile; PRP-1 support]. Oligomers can be detritylated in 3% acetic acid and precipitated with 2% lithiumperchlorate/acetone, dissolved in sterile water and reprecipitated as the sodium salt from 1 M NaCl/ethanol. Concentrations of the full length species can be determined by UV spectroscopy.
- Any other means for such synthesis known in the art may additionally or alternatively be employed. It is well known to use similar techniques to prepare oligonucleotides such as the phosphorothioates and alkylated derivatives.
- Hybridization of antisense oligonucleotides with VDUP-1 mRNA interferes with one or more of the normal functions of VDUP-1 1 mRNA.
- the functions of mRNA to be interfered with include all vital functions such as, for example, translocation of the RNA to the site of protein translation, translation of protein from the RNA, splicing of the RNA to yield one or more mRNA species, and catalytic activity which may be engaged in by the RNA.
- Preferred modified oligonucleotide backbones include, for example, phosphorothioates, chiral phosphorothioates, phosphorodithioates, phosphotriesters, aminoalkylphosphotriesters, methyl and other alkyl phosphonates including 3′-alkylene phosphonates, 5′-alkylene phosphonates and chiral phosphonates, phosphinates, phosphoramidates including 3′-amino phosphoramidate and aminoalkylphosphoramidates, thionophosphoramidates, thionoalkylphosphonates, thionoalkylphosphotriesters, selenophosphates and borano-phosphates having normal 3′-5′ linkages, 2′-5′ linked analogs of these, and those having inverted polarity wherein one or more internucleotide linkages is a 3′ to 3′, 5′ to 5′ or 2′ to 2′ linkage.
- oligonucleotides having inverted polarity comprise a single 3′ to 3′ linkage at the 3′-most internucleotide linkage i.e. a single inverted nucleoside residue which may be abasic (the nucleobase is missing or has a hydroxyl group in place thereof) are included.
- Various salts, mixed salts and free acid forms are also included.
- messenger RNA includes not only the information to encode a protein using the three letter genetic code, but also associated ribonucleotides which form a region known to such persons as the 5′-untranslated region, the 3′-untranslated region, the 5′ cap region and intron/exon junction ribonucleotides.
- antisense oligonucleotides or the catalytic nucleic acids described below may be formulated in accordance with this invention which are targeted wholly or in part to these associated ribonucleotides as well as to the informational ribonucleotides.
- the antisense oligonucleotides may therefore be specifically hybridizable with a transcription initiation site region, a translation initiation codon region, a 5′ cap region, an intron/exon junction, coding sequences, a translation termination codon region or sequences in the 5′- or 3′-untranslated region.
- the catalytic nucleic acids may specifically cleave a transcription initiation site region, a translation initiation codon region, a 5′ cap region, an intron/exon junction, coding sequences, a translation termination codon region or sequences in the 5′- or 3′-untranslated region.
- the translation initiation codon is typically 5′-AUG (in transcribed mRNA molecules; 5′-ATG in the corresponding DNA molecule).
- 5′-AUG in transcribed mRNA molecules
- 5′-ATG in the corresponding DNA molecule
- a minority of genes have a translation initiation codon having the RNA sequence 5′-GUG, 5′UUG or 5′-CUG, and 5′-AUA, 5′-ACG and 5′-CUG have been shown to function in vivo.
- the term “translation initiation codon” can encompass many codon sequences, even though the initiator amino acid in each instance is typically methionine in eukaryotes.
- translation initiation codon refers to the codon or codons that are used in vivo to initiate translation of an mRNA molecule transcribed from a gene encoding VDUP-1, regardless of the sequence(s) of such codons.
- a translation termination codon of a gene may have one of three sequences, i.e., 5′-UAA, 5′-UAG and 5′-UGA (the corresponding DNA sequences are 5′-TAA, 5′-TAG and 5′-TGA, respectively).
- the term “translation initiation codon region” refers to a portion of such an mRNA or gene that encompasses from about 25 to about 50 contiguous nucleotides in either direction (i.e., 5′ or 3′) from a translation initiation codon. This region is one preferred target region.
- translation termination codon region refers to a portion of such an mRNA or gene that encompasses from about 25 to about 50 contiguous nucleotides in either direction (i.e., 5′ or 3′) from a translation termination codon. This region is also one preferred target region.
- Other preferred target regions include the 5′ untranslated region (5′UTR), known in the art to refer to the portion of an mRNA in the 5′ direction from the translation initiation codon, and thus including nucleotides between the 5′ cap site and the translation initiation codon of an mRNA or corresponding nucleotides on the gene, and the 3′ untranslated region (3′UTR), known in the art to refer to the portion of an mRNA in the 3′ direction from the translation termination codon, and thus including nucleotides between the translation termination codon and 3′ end of an mRNA or corresponding nucleotides on the gene.
- 5′UTR 5′ untranslated region
- 3′UTR 3′ untranslated region
- mRNA splice sites may also be preferred target regions, and are particularly useful in situations where aberrant splicing is implicated in disease, or where an overproduction of a particular mRNA splice product is implicated in disease. Aberrant fusion junctions due to rearrangements or deletions may also be preferred targets.
- antisense oligonucleotides can be chosen which are sufficiently complementary to the target, i.e., hybridize sufficiently well and with sufficient specificity, to give the desired disruption of the function of the molecule.
- “Hybridization”, in the context of this invention means hydrogen bonding, also known as Watson-Crick base pairing, between complementary bases, usually on opposite nucleic acid strands or two regions of a nucleic acid strand. Guanine and cytosine are examples of complementary bases which are known to form three hydrogen bonds between them. Adenine and thymine are examples of complementary bases which form two hydrogen bonds between them.
- Specifically hybridizable and “complementary” are terms which are used to indicate a sufficient degree of complementarity such that stable and specific binding occurs between the DNA or RNA target and the catalytic nucleic acids.
- catalytic nucleic acids are synthesized once cleavage target sites on the VDUP-1 mRNA molecule.
- antisense oligonucleotides or catalytic nucleic acids or analogs thereof it is preferred to administer antisense oligonucleotides or catalytic nucleic acids or analogs thereof to mammals suffering from cardiovascular disease, in either native form or suspended in a carrier medium in amounts and upon treatment schedules which are effective to therapeutically treat the mammals to reduce the detrimental effects of cardiovascular disease.
- One or more different catalytic nucleic acids or antisense oligonucleotides or analogs thereof targeting different sections of the nucleic acid sequence of VDUP-1 mRNA may be administered together in a single dose or in different doses and at different amounts and times depending upon the desired therapy.
- the catalytic nucleic acids or antisense oligonucleotides can be administered to mammals in a manner capable of getting the oligonucleotides initially into the blood stream and subsequently into cells, or alternatively in a manner so as to directly introduce the catalytic nucleic acids or antisense oligonucleotides into the cells or groups of cells, for example cardiomyocytes, by such means by electroporation or by direct injection into the heart.
- Antisense oligonucleotides whose presence in cells can inhibit transcription or protein synthesis can be administered by intravenous injection, intravenous drip, subcutaneous, intraperitoneal or intramuscular injection, orally or rectally.
- Doses of the oligonucleotides or analogs thereof of the present invention in a pharmaceutical dosage unit will be an efficacious, nontoxic quantity administered to a human patient in need of cardiomyocyte regeneration (or inhibition of VDUP-1 expression) from 1-6 or more times daily or every other day. Dosage is dependent on severity and responsiveness of the effects of abnormal cardiovascular disease to be treated, with course of treatment lasting from several days to months or until a cure is effected or a reduction of the effects is achieved. Oral dosage units for human administration generally use lower doses. The actual dosage administered may take into account the size and weight of the patient, whether the nature of the treatment is prophylactic or therapeutic in nature, the age, weight, health and sex of the patient, the route of administration, and other factors.
- the second agent is a pro-angiogenic agent.
- the pro-angiogenic agent is vascular endothelial growth factor, fibroblast growth factor or angiopoietin.
- the second agent induces expression of a pro-angiogenic factor.
- the second agent is Hypoxia Inducible Factor-1.
- the second agent is a catalytic nucleic acid which specifically inhibits translation of Vitamin D3 Up-Regulated Protein-1 mRNA.
- the catalytic nucleic acid comprises deoxyribonucleotides.
- the catalytic nucleic acid comprises ribonucleotides.
- Catalytic nucleic acid molecules can cleave Vitamin D3 Up-Regulated Protein-1 (VDUP-1) mRNA (corresponding DNA shown in SEQ ID NO:1, FIG. 5) at each and any of the consensus sequences therein. Since catalytic ribo- and deoxyribo-nucleic acid consensus sequences are known, and the VDUP-1 Protein mRNA sequence is known, one of ordinary skill could readily construct a catalytic ribo- or deoxyribo nucleic acid molecule directed to any of the VDUP-1 protein mRNA consensus sequences based on the instant specification. In preferred embodiments of this invention the catalytic deoxyribonucleic acids include the 10-23 structure.
- catalytic ribonucleic acids examples include hairpin and hammerhead ribozymes.
- the catalytic ribonucleic acid molecule is formed in a hammerhead (50) or hairpin motif (51,52,53), but may also be formed in the motif of a hepatitis delta virus (54), group I intron (60), RNaseP RNA (in association with an RNA guide sequence) (55,56) or Neurospora VS RNA (57,58,59).
- catalytic nucleic acids can be designed based on the consensus cleavage sites 5′-purine:pyrimidine-3′ in the VDUP-1 mRNA sequence (104) (see FIGS. 6 - 10 ) for cleavage sites on DNA corresponding to the mRNA encoding VDUP-1 (SEQ ID NO:2). Those potential cleavage sites located on an open loop of the mRNA according to RNA folding software e.g. RNADRaw 2.1 are particularly preferred as targets (61).
- the DNA based catalytic nucleic acids can utilize the structure where two sequence-specific arms are attached to a catalytic core based on the VDUP-1 mRNA sequence.
- catalytic DNA structure is detailed in (62) and (63).
- Commercially available mouse brain polyA-RNA (Ambion) can serve as a template in the in vitro cleavage reaction to test the efficiency of the catalytic deoxyribonucleic acids.
- Catalytic RNA is described above, is designed similarly.
- Hammerhead ribozymes can cleave any 5′-NUH-3′ triplets of a mRNA, where U is conserved and N is any nucleotide and H can be C,U,A, but not G.
- the sites which can be cleaved by a hammerhead ribozyme in human VDUP-1 mRNA coding region are shown in FIG. 10.
- Cleaving of VDUP-1 mRNA with catalytic nucleic acids interferes with one or more of the normal functions of VDUP-1 mRNA.
- the functions of mRNA to be interfered with include all vital functions such as, for example, translocation of the RNA to the site of protein translation, translation of protein from the RNA, splicing of the RNA to yield one or more mRNA species, and catalytic activity which may be engaged in by the RNA.
- the nucleotides may comprise other bases such as inosine, deoxyinosine, hypoxanthine may be used.
- isoteric purine 2′deoxy-furanoside analogs, 2′-deoxynebularine or 2′deoxyxanthosine, or other purine or pyrimidine analogs may also be used.
- inosine may be used to reduce hybridization specificity
- diaminopurines may be used to increase hybridization specificity.
- Adenine and guanine may be modified at positions N3, N7, N9, C2, C4, C5, C6, or C8 and still maintain their hydrogen bonding abilities.
- Cytosine, thymine and uracil may be modified at positions N1, C2, C4, C5, or C6 and still maintain their hydrogen bonding abilities.
- Some base analogs have different hydrogen bonding attributes than the naturally occurring bases. For example, 2-amino-2′-dA forms three (3), instead of the usual two (2), hydrogen bonds to thymine (T).
- Examples of base analogs that have been shown to increase duplex stability include, but are not limited to, 5-fluoro-2′-dU, 5-bromo-2′-dU, 5-methyl-2′-dC, 5-propynyl-2′-dC, 5-propynyl-2′-dU, 2-amino-2′-dA, 7-deazaguanosine, 7-deazadenosine, and N2-Imidazoylpropyl-2′-dG.
- Nucleotide analogs may be created by modifying and/or replacing a sugar moiety.
- the sugar moieties of the nucleotides may also be modified by the addition of one or more substituents.
- one or more of the sugar moieties may contain one or more of the following substituents: amino, alkylamino, araalkyl, heteroalkyl, heterocycloalkyl, aminoalkylamino, O, H, an alkyl, polyalkylamino, substituted silyl, F, Cl, Br, CN, CF 3 , OCF 3 , OCN, O-alkyl, S-alkyl, SOMe, SO 2 Me, ONO 2 , NH-alkyl, OCH 2 CH ⁇ CH 2 , OCH 2 CCH, OCCHO, allyl, O-allyl, NO 2 , N 3 , and NH 2 .
- the 2′ position of the sugar may be modified to contain one of the following groups: H, OH, OCN, O-alkyl, F, CN, CF 3 , allyl, O-allyl, OCF 3 , S-alkyl, SOMe, SO 2 Me, ONO 2 , NO 2 , N 3 , NH 2 , NH-alkyl, or OCH ⁇ CH 2 , OCCH, wherein the alkyl may be straight, branched, saturated, or unsaturated.
- the nucleotide may have one or more of its sugars modified and/or replaced so as to be a ribose or hexose (i.e. glucose, galactose) or have one or more anomeric sugars.
- the nucleotide may also have one or more L-sugars.
- the sugar may be modified to contain one or more linkers for attachment to other chemicals such as fluorescent labels.
- the sugar is linked to one or more aminoalkyloxy linkers.
- the sugar contains one or more alkylamino linkers. Aminoalkyloxy and alkylamino linkers may be attached to biotin, cholic acid, fluorescein, or other chemical moieties through their amino group.
- Nucleotide analogs or derivatives may have pendant groups attached.
- Pendant groups serve a variety of purposes which include, but are not limited to, increasing cellular uptake of the oligonucleotide, enhancing degradation of the target nucleic acid, and increasing hybridization affinity.
- Pendant groups can be linked to any portion of the oligonucleotide but are commonly linked to the end(s) of the oligonucleotide chain. Examples of pendant groups include, but are not limited to: acridine derivatives (i.e.
- cross-linkers such as psoralen derivatives, azidophenacyl, proflavin, and azidoproflavin; artificial endonucleases; metal complexes such as EDTA-Fe(II), o-phenanthroline-Cu(I), and porphyrin-Fe(II); alkylating moieties; nucleases such as amino-1-hexanolstaphylococcal nuclease and alkaline phosphatase; terminal transferases; abzymes; cholesteryl moieties; lipophilic carriers; peptide conjugates; long chain alcohols; phosphate esters; amino; mercapto groups; radioactive markers; nonradioactive markers such as dyes; and polylysine or other polyamines.
- cross-linkers such as psoralen derivatives, azidophenacyl, proflavin, and azidoproflavin
- artificial endonucleases such as EDTA-Fe(II), o
- the nucleic acid comprises an oligonucleotide conjugated to a carbohydrate, sulfated carbohydrate, or gylcan.
- Conjugates may be regarded as a way as to introduce a specificity into otherwise unspecific DNA binding molecules by covalently linking them to a selectively hybridizing oligonucleotide.
- the catalytic nucleic acid binding domains i.e. the non-catalytic domains
- antisense oligonucleotide may comprise modified bonds.
- internucleosides bonds of the oligonucleotide may comprise phosphorothioate linkages.
- the nucleic acid may comprise nucleotides having moiety may be modified by replacing one or both of the two bridging oxygen atoms of the linkage with analogues such as —NH, —CH 2 , or —S. Other oxygen analogues known in the art may also be used.
- the phosphorothioate bonds may be stereo regular or stereo random.
- the oligonucleotide moiety may have one or more of its sugars modified or replaced so as to be ribose, glucose, sucrose, or galactose, or any other sugar.
- the phosphorothioate oligonucleotide may have one or more of its sugars substituted or modified in its 2′ position, i.e. 2′allyl or 2′-O-allyl.
- An example of a 2′-O-allyl sugar is a 2′-O-methylribonucleotide.
- the phosphorothioate oligonucleotide may have one or more of its sugars substituted or modified to form an ⁇ -anomeric sugar.
- a catalytic nucleic acid may include non-nucleotide substitution.
- the non-nucleotide substitution includes either abasic nucleotide, polyether, polyamine, polyamide, peptide, carbohydrate, lipid or polyhydrocarbon compounds.
- abasic or “abasic nucleotide” as used herein encompasses sugar moieties lacking a base or having other chemical groups in place of base at the 1′ position.
- Determining the effective amount of the instant nucleic acid molecules can be done based on animal data using routine computational methods.
- the effective amount contains between about 10 ng and about 100 ⁇ g of the instant nucleic acid molecules per kg body mass. In another embodiment, the effective amount contains between about 100 ng and about 10 ⁇ g of the nucleic acid molecules per kg body mass. In a further embodiment, the effective amount contains between about 1 ⁇ g and about 5 ⁇ g, and in a further embodiment about 2 ⁇ g, of the nucleic acid molecules per kg body mass.
- the second agent is cardiomyocyte progenitor cells.
- the second agent is skeletal muscle progenitor cells. Either of theses cell types can be derived from embryonic, fetal or adult subjects.
- the second agent promotes trafficking of the endothelial progenitor cells to the subject's heart.
- the second agent that promotes trafficking is an antibody directed against an epitope of CXCR4.
- the second agent that promotes trafficking is a CC chemokine.
- the CC chemokine is RANTES, EOTAXIN, monocyte chemoattractant protein-1 (MCP-1), MCP-2, MCP-3, or MCP.
- the second agent that promotes trafficking is a CXC chemokine.
- the CXC chemokine is Interleukin-8, Gro-Alpha, or Stromal-Derived Factor-1.
- This invention also provides a method of determining the susceptibility of a cardiomyocyte in a subject to apoptosis comprising:
- This invention also provides a method of determining the susceptibility of a cardiomyocyte in a subject to apoptosis comprising:
- a ratio that is at least two standard deviations above that found in control patients with normal hearts is considered a high ratio.
- Quantitation of expression of protein in cardiomyocytes is performed using routine methods known to those of skill in the art, for example, northern and western blots.
- the subject of any of the above methods is a mammal and in a preferred further embodiment the mammal is a human being.
- the subject has a cardiovascular disease.
- the subject has congestive heart failure, has suffered a myocardial infarct, has suffered myocardial ischemia, has angina, or has a cardiomyopathy.
- LAD coronary artery ligation resulted in trafficking of intravenously injected human endothelial progenitor cells to the site of ischemic myocardium, it was also accompanied by increased distribution of human cells to rat bone marrow.
- FIG. 2 a at 2-14 days after intravenous injection of 2 ⁇ 10 6 human CD34+ cells bone marrow from LAD-ligated rats contained 5-8 fold higher levels of human CD117 bright endothelial progenitor cells compared with bone marrow from normal rats, p ⁇ 0.001.
- co-administration of mAbs against either human CXCR4 or rat SDF-1 significantly inhibited migration of intravenously administered human endothelial progenitor cells to ischemic rat bone marrow compared with anti-CD34 control antibody (both p ⁇ 0.001).
- co-administration of mAbs against either human CXCR4 or rat SDF-1 increased trafficking of CD34+ human endothelial progenitor cells to ischemic rat myocardium by means of 24% and 17%, respectively (both p ⁇ 0.001), FIG. 2 c.
- Induction of neovascularization at two weeks was measured by performing quantitative analysis of medium- and large-sized capillaries, defined, respectively, as having 3-6 or >6 contiguous endothelial lining cells.
- Medium-sized capillaries had mean lumen diameter of 0.020 mm+0.002, while large-sized capillaries had mean lumen diameter of 0.053 mm+0.004 (p ⁇ 0.001).
- large-lumen capillaries overlapped in size with arterioles which could be distinguished by a thin layer containing 2-3 smooth muscle cells of rat origin, as determined by positive staining with desmin and rat MHC class I mAbs. As shown in FIGS.
- both the group receiving 2 ⁇ 10 5 endothelial progenitor cells and the one receiving 10 5 endothelial progenitor cells plus anti-CXCR4 mAb demonstrated 1.7-fold higher numbers of medium-sized capillaries compared with the other two groups (p ⁇ 0.01).
- the group receiving 2 ⁇ 10 5 endothelial progenitor cells additionally demonstrated 3.3-fold higher numbers of large-lumen capillaries compared with the groups receiving 10 3 or 10 5 endothelial progenitor cells (p ⁇ 0.01), and 2-fold higher numbers of large-lumen capillaries compared with the group receiving 10 5 endothelial progenitor cells plus anti-CXCR4 mAb (p ⁇ 0.01).
- FIG. 1 As shown in FIG.
- FIG. 3 e the number of apoptotic cardiomyocytes at the infarct rim was significantly reduced in both rats receiving 10 5 endothelial progenitor cells plus anti-CXCR4 mAb and those receiving 2 ⁇ 10 5 endothelial progenitor cells compared with the groups receiving either 10 3 or 10 5 endothelial progenitor cells alone (both p ⁇ 0.001).
- co-administration of anti-CXCR4 mAb or intracardiac injection of SDF-1 resulted in further reductions in cardiomyocyte apoptosis of 65% and 76%, respectively, FIG. 3 f (both p ⁇ 0.001).
- FIGS. 4 a and b We next examined the effect of increasing the number of human endothelial progenitor cells trafficking to ischemic myocardium on long-term myocardial function, defined as the degree of improvement in left ventricular ejection fraction (LVEF) and reduction in left ventricular end-systolic area (LVAs) at 15 weeks after intravenous injection, FIGS. 4 a and b . No improvement in these parameters was observed in the groups receiving 10 3 or 10 5 endothelial progenitor cells in comparison to rats receiving saline alone.
- LVEF left ventricular ejection fraction
- LVAs left ventricular end-systolic area
- rats receiving 10 5 endothelial progenitor cells plus anti-CXCR4 mAb demonstrated significant improvement in these parameters, 22+2% mean recovery in LVEF and 24+4% mean reduction in LVAs (both p ⁇ 0.001). Even more strikingly, the group receiving 2 ⁇ 10 5 endothelial progenitor cells had a mean recovery in LVEF of 34+4% and a mean reduction in LVAs of 37+6% (both p ⁇ 0.001), or 50% further improvement in both parameters.
- the number of cardiomyocytes progressing through cell cycle at the peri-infarct region of rats receiving 2 ⁇ 10 5 human endothelial progenitor cells was 40-fold higher than that at sites distal to the infarct, where myocyte DNA activity was no different than in sham-operated rats. As shown in FIG.
- animals receiving 2 ⁇ 10 5 human endothelial progenitor cells had a 20-fold higher number of cell-cycling cardiomyocytes at the peri-infarct rim than that found in non-infarcted hearts (1.19+0.2% vs 0.06+0.03%, p ⁇ 0.01) and 3.5-fold higher than in the same region in LAD-ligated controls receiving saline (1.19+0.2% vs 0.344+0.1%, p ⁇ 0.01).
- intracardiac injection of SDF-1 in combination with intravenous injection of 2 ⁇ 10 5 human endothelial progenitor cells resulted in approximately an 8-fold cumulative increase in cell-cycling cardiomyocytes at two weeks compared with LAD-ligated controls receiving saline, and translated into over 4-fold greater LVEF improvement, determined by echocardiography, compared with intravenous injection of 2 ⁇ 10 5 endothelial progenitor cells alone (FIG. 4 f , p ⁇ 0.01).
- Co-administration of anti-CXCR4 mAb augmented LVEF improvement by 2.8-fold (p ⁇ 0.01) while intracardiac injection of IL-8 conferred no additive benefit.
- FIG. 4 h The overall effects of medium- and large-size neovasculature combining to both protect against myocyte apoptosis and induce myocyte proliferation/regeneration are shown dramatically in FIG. 4 h where, in contrast to saline controls, injection with 2 ⁇ 10 5 endothelial progenitor cells resulted in almost complete salvage of the anterior myocardium, normal septal size and minimal collagen deposition.
- E2 catalytic nucleic acid
- Vitamin D3 Up-Regulated Proteinl VDUP1
- H 2 O 2 hydrogen peroxide
- the cell cycle inhibitory activity of p21Cip1/WAF1 is intimately correlated with its nuclear localization and participation in quaternary complexes of cell cycle regulators by binding to G1 cyclin-CDK through its N-terminal domain and to proliferating cell nuclear antigen (PCNA) through its C-terminal domain (68-71).
- PCNA cell nuclear antigen
- the latter interaction blocks the ability of PCNA to activate DNA polymerase, the principal replicative DNA polymerase (72).
- For a growth-arrested cell to subsequently enter an apoptotic pathway requires signals provided by specific apoptotic stimuli in concert with cell-cycle regulators.
- caspase-mediated cleavage of p21 together with upregulation of cyclin A-associated cdk2 activity, have been shown to be critical steps for induction of cellular apoptosis by either deprivation of growth factors (73) or hypoxia of cardiomyocytes (74).
- the apoptosis signal-regulating kinase 1 (ASK1) is a pivotal component in the mechanism of cytokine- and stress-induced apoptosis (75,76). Under basal conditions, resistance to ASK1-mediated apoptosis appears to be the result of complex formation between ASK1, cytoplasmic p21Cip1/WAF1 (77), and the thiol reductase thioredoxin (TRX) (78). Intact cytoplasmic expression of p21Cip1/WAF1 appears to be important for both prevention of apoptosis in response to ASK1 (75) and in maintaining a state of terminal differentiation (77).
- the reduced form of TRX binds to the N-terminal portion of ASK1 and is a physiologic inhibitor of ASK1-mediated cellular apoptosis (78).
- the recently-identified protein VDUP1 has been shown to compete with ASK1 for binding of the reduced form of TRX (78,79), resulting in augmention of ASK1-mediated apoptosis (80). This indicates that ASK1-mediated cellular apoptosis is increased by processes that result in a net dissociation of TRX from ASK1, such as either generation of TRX-VDUP1 complexes or generation of oxidised TRX by changes in cellular redox status accompanying oxidative stress.
- TRX and glutathione constitute the major cellular reducing systems that maintain the thiol-disulfide status of the cytosol (81).
- the redox-active/dithiol active site of TRX is highly conserved across all species, Trp-Cys-Gly-Pro-Cys-Lys.
- the two cysteine residues at the active site, Cys-32 and Cys-35 undergo reversible oxidation-reduction reactions catalyzed by a NADPH-dependent enzyme TRX reductase. These reactions involve electron transfer via disulfide bridges formed with members of a family of antioxidant enzymes known as peroxiredoxins (Prxs), which show peroxidase activity (82,83).
- Prxs peroxiredoxins
- Prxs are distinct from other peroxidases in that they have no cofactors, such as metals or prosthetic groups. Prxs generally have two conserved cysteines at the N- and C-terminal regions (84), and their antioxidant effects are coupled with the physiological electron donor activity of the TRX system (82,85,86). Prxs with 95-97% sequence homology have been identified in rats (Heme-binding protein 23, HBP23) (87), mice (mouse macrophage stress protein 23, MSP23) (88) and humans (proliferation-associated gene product, PAG (89) and human natural killer cell-enhancing factor A (90)).
- Prxs are members of a repertoire of oxidative stress responsive genes whose expression is regulated by NF-E2-related factor 2 (Nrf2) which binds to an anti-oxidant responsive element (ARE) present in the promoter of each (91). These include glutathione-S-transferase, heme oxygenase-1, and TRX. Under basal conditions, Nrf2 is bound to a specific protein, Keap1, in the cytosol (92). However, under conditions of oxidative stress Nrf2 dissociates from Keap1 and translocates to the nucleus where it induces transcriptional activation of the anti-oxidant genes containing ARE motifs.
- Nrf2 NF-E2-related factor 2
- ARE anti-oxidant responsive element
- Nrf2 nuclear translocation of Nrf2 and subsequent ARE activation appear to be dependent on pathways activated by phosphatidylinositol 3-kinase (PI3 kinase) (93).
- PI3 kinase phosphatidylinositol 3-kinase
- hemin is a potent inducer of Nrf2 dissociation from Keap1, resulting in TRX gene transcription through the ARE (94).
- Prxs During periods of rapid changes in cellular redox Prxs presumably serve to maintain the cytosolic levels of reduced TRX by accepting electrons from the oxidized form of TRX. This homeostatic mechanism likely enables maintenance of sufficient levels of reduced TRX to ensure adequate binding to ASK1 and prevention of cellular apoptosis. If the endogenous Prx system is overloaded, as might occur during changes in cellular redox when excess oxidized TRX is generated, cellular apoptosis will occur through the unopposed effects of ASK1. To counteract this, transcriptional activation of Prxs must occur following oxidative stress via nuclear translocation of Nrf2. This can be achieved either by Nrf2 dissociation from Keap1 via hemin- and PI3 kinase-dependent mechanisms (93,94), or by increasing Nrf2 mRNA and protein expression as occurs following increase in oxygen tension (95,96).
- the Prx gene products specifically bind the SH3 domain of c-Abl, a non-receptor tyrosine kinase, inhibiting its activation by various stimuli, including agents that damage DNA (97).
- c-Abl activation through the SH3 domain induces either arrest of the cell cycle in phase G1 or cellular apoptosis (98).
- Cell cycle arrest is dependent on the kinase activity of c-Abl (96) and is mediated by the ability of c-Abl to downregulate the activity of the cyclin-dependent kinase Cdk2 and induce the expression of p21 (99).
- c-Abl The apoptotic effects of c-Abl are dependent on the ability of nuclear c-Abl to phosphorylate p73, a member of the p53 family of tumor-suppressor proteins which can induce apoptosis (100,101). Recently, it has been shown that cytoplasmic, rather than nuclear, forms of c-Abl are activated by H 2 O 2 and that this results in mitochondrial localization of c-Abl, c-Abl dependent cytochrome c release, and cellular apoptosis following oxidative stress (102,103).
- the PAG gene product By associating with c-Abl in vivo, the PAG gene product (and presumably the other Prxs) can inhibit tyrosine phosphorylation induced by c-Abl overexpression and rescue cells from both the cytostatic and pro-apoptotic effects of the activated c-Abl gene product (97).
- Nrf2-dependent oxidative stress responsive genes are downregulated following myocardial ischemia likely reflects direct effects of hemin and oxygen deprivation.
- the end result of Prx downregulation in the ischemic heart would be augmentation in ASK1-dependent cellular apoptosis as well as Abl-dependent apoptosis and cell cycle arrest.
- the observed parallel increase in VDUP1 expression would further augment ASK1-dependent cellular apoptosis.
- the ratio in expression of PAG or other Prx mRNA or protein to VDUP1 mRNA or protein can form the basis of a diagnostic assay to predict the degree of risk for cardiomyocyte apoptosis and cell cycle arrest after ischemia, as well as enable monitoring of the response to specific therapy after myocardial ischemia that protects cardiomyocytes against apototic death and enhances myocardial proliferation/regeneration.
- Nrf2 mRNA or causing dissociation of Nrf2 protein from Keap1, or preferably cause both to occur simultaneously in the setting of myocardial ischemia in order to increase transcription and activity of members of a repertoire of oxidative stress responsive genes whose expression is regulated by binding of Nrf2 to an anti-oxidant responsive element (ARE) in their promoters, including the Prxs, TRX and glutathione-S-transferase would result in both protection of cardiomyocytes against apoptosis as well as induce cardiomyocyte cell cycle progression following oxidative stress.
- ARE anti-oxidant responsive element
- VDUP1 Reducing the expression of VDUP1 following myocardial ischemia would protect the ischemic myocardium against apoptosis by reducing binding of TRX to VDUP1, and consequently increasing TRX-ASK1 interactions.
- Neovascularization of the myocardium by either bone marrow-derived endothelial progenitors or any other process, is an example of one method which causes induction of Prx expression and reduction in VDUP1 expression after myocardial ischemia, and results in both protection against redox-mediated apoptosis and induction of myocardial proliferation/regeneration.
- Single-donor leukopheresis products were obtained from humans treated with recombinant G-CSF 10 mg/kg (Amgen, CA) sc daily for four days. Donors were healthy individuals undergoing standard institutional procedures of bone marrow mobilization, harvesting and isolation for allogeneic stem cell transplants. Mononuclear cells were separated by Ficoll-Hypaque, and highly-purified CD34+ cells (>98% positive) were obtained using magnetic beads coated with anti-CD34 monoclonal antibody (mAb) (Miltenyi Biotech, CA).
- mAb anti-CD34 monoclonal antibody
- CD34 cells were stained with fluorescein-conjugated mAbs against CD34 and CD117 (Becton Dickinson, CA), AC133 (Miltenyi Biotech, CA), CD54 (Immunotech, CA), CD62E (BioSource, MA), VEGFR-2, Tie-2, vWF, eNOS, CXCR1, CXCR2, and CXCR4 (all Santa Cruz Biotech, CA), and analyzed by four-parameter fluorescence using FACScan (Becton Dickinson, CA).
- Cells positively selected for CD34 expression were also stained with phycoerythrin (PE)-conjugated anti-CD117 mAb (Becton Dickinson, CA), and sorted for bright and dim fluorescence using a Facstar Plus (Becton Dickinson) and a PE filter.
- Intracellular staining for GATA-2 was performed by permeabilizing one million cells from each of the brightly and dimly fluorescing cell populations using a Pharmingen Cytofix/CytopermTM kit, incubating for 30 minutes on ice with 10 ⁇ l of fluorochrome-conjugated mAbs against both CD117 and CD34 surface antigens (Becton Dickinson, CA).
- CD34+CD117 brigh t cells were plated in 48-well chemotaxis chambers fitted with membranes (8 mm pores) (Neuro Probe, MD). After incubation for 2 hours at 37° C., chambers were inverted and cells were cultured for 3 hours in medium containing IL-8, SDF-1 alpha/beta, and SCF at 0.2, 1.0 and 5.0 ⁇ g/ml. The membranes were fixed with methanol and stained with LeukostatTM (Fischer Scientific, Ill.). Chemotaxis was calculated by counting migrating cells in 10 high-power fields.
- CD34+ cells obtained from a single donor after G-CSF mobilization were injected into the tail vein 48 hours after LAD ligation either alone or together with 50 ⁇ g/ml monoclonal antibody (mAb) with known functional inhibitory activity against either human CXCR1, human CXCR2, human CXCR4, rat SDF-1 (all R & D Systems, MN), human CD34 (Pharmingen, CA), or rat IL-8 (ImmunoLaboratories, Japan). Controls received either isotype control antibodies at the same concentration or saline after LAD ligation.
- mAb monoclonal antibody
- Quantitation of myocardial infiltration after injection of human cells was performed by assessment of DiI fluorescence in hearts from rats sacrificed 2 days after injection (expressed as number of DiI-positive cells per high power field, minimum 5 fields examined per sample). Quantitation of rat bone marrow infiltration by human cells was performed in 12 rats at baseline, days 2, 7, and 14 by flow cytometric and RT-PCR analysis of the proportion of HLA class I-positive cells relative to the total rat bone marrow population.
- Poly(A)+ mRNA was extracted by standard methods from the hearts of 3 normal and 12 LAD-ligated rats. RT-PCR was used to quantify myocardial expression of rat IL-8 and Gro-alpha mRNA at baseline and at 6, 12, 24 and 48 hours after LAD ligation after normalizing for total rat RNA as measured by GAPDH expression.
- cDNA was amplified in the polymerase chain reaction (PCR) using Taq polymerase (Invitrogen, Carlsbad, Calif., U.S.A.), radiolabeled dideoxy-nucleotide ([a32P]-ddATP: 3,000 Ci/mmol, Amersham, Arlington Heights, Ill.), and primers for rat Cinc (rat homologue of human IL-8/Gro-alpha and GAPDH (Fisher Genosys, CA).
- Taq polymerase Invitrogen, Carlsbad, Calif., U.S.A.
- radiolabeled dideoxy-nucleotide [a32P]-ddATP: 3,000 Ci/mmol, Amersham, Arlington Heights, Ill.
- primers for rat Cinc rat homologue of human IL-8/Gro-alpha and GAPDH (Fisher Genosys, CA).
- Primer pairs (sense/antisense) for rat Cinc and GAPDH were, gaagatagattgcaccgatg (SEQ ID NO:4)/catagcctctcacatttc SEQ ID NO:5), gcgcccgtccgccaatgagctgcgc SEQ ID NO:6)/cttggggacacccttcagcatctttgg SEQ ID NO:7), and ctctacccacggcaagttcaa SEQ ID NO:8)/gggatgaccttgcccacagc SEQ ID NO:9), respectively.
- rat IL-8/Gro-alpha Serum levels of rat IL-8/Gro-alpha were measured at baseline and at 6, 12, 24 and 48 hours after LAD ligation in four rats by a commercial ELISA using polyclonal antibodies against the rat IL-8/Gro homologue Cinc (ImmunoLaboratories, Japan). The amount of protein in each serum sample was calculated according to a standard curve of optical density (OD) values constructed for known levels of rat IL-8/Gro-alpha protein.
- OD optical density
- Anti-Cinc antibodies were also used according to the manufacturer's instructions at 1:200 dilution in immunohistochemical studies to identify the cellular source of Cinc production in rat myocardium after LAD ligation. Positively-staining cells were visualized as brown through the Avidin/Biotin system described below.
- Staining was performed by immunoperoxidase technique using an Avidin/Biotin Blocking Kit, a rat-absorbed biotinylated anti-mouse IgG, and a peroxidase-conjugate (all Vector Laboratories Burlingame, Calif.).
- Capillary density was determined at 2 weeks post infarction from sections labeled with anti-CD31 mAb, and confirmed with anti-factor VIII mAb, and compared to the capillary density of the unimpaired myocardium. Values are expressed as the number of CD31-positive capillaries per HPF (400x).
- Cardiomyocyte DNA synthesis and cell cycling was determined by dual staining of rat myocardial tissue sections obtained from LAD-ligated rats at two weeks after injection of either saline or CD34+ human cells, and from healthy rats as negative controls, for cardiomyocyte-specific troponin I and human- or rat-specific Ki-67. Briefly, paraffin embedded sections were microwaved in a 0.1M EDTA buffer, and stained with either a primary monoclonal antibody against rat Ki-67 at 1:3000 dilution (gift of Giorgio Catoretti, Columbia University) or human Ki-67 at 1:300 dilution (Dako, CA) and incubated overnight at 4 degrees C.
- TUNEL dexynucleotidyl transferase
- tissue sections were then digested with Proteinase K (10 ⁇ g/ml in Tris/HCL) for 30 minutes at 37° C.
- the slides were then washed 3 times in PBS and incubated with 50 ⁇ l of the TUNEL reaction mixture (TdT and fluorescein-labeled dUTP) and incubated in a humid atmosphere for 60 minutes at 37° C.
- TdT was eliminated from the reaction mixture.
- the sections were then incubated for 30 minutes with an antibody specific for fluorescein-conjugated alkaline phosphatase (AP) (Boehringer Mannheim, Mannheim, Germany).
- AP fluorescein-conjugated alkaline phosphatase
- the TUNEL stain was visualized with a substrate system in which nuclei with DNA fragmentation stained blue, (BCIP/NBT substrate system, Dako, Carpinteria, Calif.). The reaction was terminated following three minutes of exposure with PBS. To determine the proportion of blue-staining apoptotic nuclei within myocytes, tissue was counterstained with a monoclonal antibody specific for desmin. Endogenous peroxidase was blocked by using a 3% hydrogen perioxidase solution in PBS for 15 minutes, followed by washing with 20% goat serum solution. An anti-troponin I antibody (Accurate Chemicals, CT) was incubated overnight (1:200) at 40 degrees C.
- BCIP/NBT substrate system Dako, Carpinteria, Calif.
- Tissue sections were examined microscopically at 200 ⁇ magnification. Within each 200 ⁇ field 4 regions were examined, containing at least 250 cells per region and cumulatively approximating 1 mm 2 of tissue, at both the peri-infarct site and distally to this site. Stained cells at the edges of the tissue were not counted. Results were expressed as the mean number of apoptotic myocytes per mm 2 at each site examined.
- Echocardiographic studies were performed using a high frequency liner array transducer (SONOS 5500, Hewlett Packard, Andover, Mass.). 2D images were obtained at mid-papillary and apical levels. End-diastolic (EDV) and end-systolic (ESV) left ventricular volumes were obtained by bi-plane area-length method, and % left ventricular ejection fraction was calculated as [(EDV ⁇ ESV)/EDV] ⁇ 100.
- SONOS 5500 High frequency liner array transducer
- ESV End-diastolic
- ESV end-systolic
- messenger RNA was isolated from each heart, and 1 ⁇ g was used for first-strand cDNA synthesis with random primers.
- the subtractive hybridization was performed with the PCR-select cDNA subtraction kit (CLONTECH), following the manufacturer's recommendations.
- CLONTECH PCR-select cDNA subtraction kit
- the two cDNA libraries were digested with RsaI. Digestion products of the “tester” library were ligated to a specific adapter (T7 promoter), then hybridized with a 30-fold excess of the “driver” library for subtraction. After hybridization, the remaining products were further amplified by PCR.
- the ischemic tissue is the “tester” and the normal tissue is the “driver.”
- the “tester” and the “driver” are switched to determine the genes that are down-regulated in the ischemic sample.
- Nrf2 Transcription factor Nrf2 coordinately regulates a group of oxidative stress-inducible genes in macrophages. J Biological Chem 2000, 275:16023-16029
- Keap1 represses nuclear activation of antioxidant responsive elements by Nrf2 through binding to the amino-terminal Neh2 domain. Genes Dev. Jan. 1, 1999;13(1):76-86.
- Jost, C. A., Marin, M. C. & Kaelin, W. J. p73 is a human p53-related protein that can induce apoptosis. Nature 389, 191-194 (1997).
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Chemical & Material Sciences (AREA)
- Engineering & Computer Science (AREA)
- General Health & Medical Sciences (AREA)
- Medicinal Chemistry (AREA)
- Animal Behavior & Ethology (AREA)
- Veterinary Medicine (AREA)
- Public Health (AREA)
- Pharmacology & Pharmacy (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Organic Chemistry (AREA)
- Zoology (AREA)
- Epidemiology (AREA)
- Biomedical Technology (AREA)
- Genetics & Genomics (AREA)
- Immunology (AREA)
- Biotechnology (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- Gastroenterology & Hepatology (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Molecular Biology (AREA)
- General Chemical & Material Sciences (AREA)
- Chemical Kinetics & Catalysis (AREA)
- Biochemistry (AREA)
- Wood Science & Technology (AREA)
- Vascular Medicine (AREA)
- Cell Biology (AREA)
- General Engineering & Computer Science (AREA)
- Cardiology (AREA)
- Microbiology (AREA)
- Biophysics (AREA)
- Virology (AREA)
- Developmental Biology & Embryology (AREA)
- Plant Pathology (AREA)
- Physics & Mathematics (AREA)
- Heart & Thoracic Surgery (AREA)
- Urology & Nephrology (AREA)
- Neurology (AREA)
- Marine Sciences & Fisheries (AREA)
Priority Applications (20)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
US10/128,738 US20030199464A1 (en) | 2002-04-23 | 2002-04-23 | Regeneration of endogenous myocardial tissue by induction of neovascularization |
EP11157749A EP2366706A1 (fr) | 2002-04-23 | 2003-04-23 | Régénération de tissu du myocarde endogène par induction de néovascularisation |
JP2003587162A JP4897199B2 (ja) | 2002-04-23 | 2003-04-23 | 新血管新生の誘導による内因性の心筋組織の再生 |
EP10164397.1A EP2292631B1 (fr) | 2002-04-23 | 2003-04-23 | Régénération de tissu myocardique endogène par induction de néovascularisation |
AU2003231090A AU2003231090B2 (en) | 2002-04-23 | 2003-04-23 | Regeneration of endogenous myocardial tissue by induction of neovascularization |
CA2482996A CA2482996C (fr) | 2002-04-23 | 2003-04-23 | Proliferation de cardiomyocytes promus sdf-1 pour le traitement de troubles cardiomyocytes induits par la mort |
CNB038147157A CN100379751C (zh) | 2002-04-23 | 2003-04-23 | 通过诱导新血管形成使内生性心肌组织再生 |
US10/512,518 US7887796B2 (en) | 2002-04-23 | 2003-04-23 | Method of inhibiting collagen formation by VDUP1 inhibition |
PCT/US2003/012768 WO2003090512A2 (fr) | 2002-04-23 | 2003-04-23 | Regeneration de tissu myocardique endogene par induction de neovascularisation |
ZA200408777A ZA200408777B (en) | 2002-04-23 | 2003-04-23 | Regeneration of endogenous myocardial tissue by induction of neovascularization |
EP03724217A EP1501852A4 (fr) | 2002-04-23 | 2003-04-23 | Regeneration de tissu myocardique endogene par induction de neovascularisation |
US10/693,480 US8663652B2 (en) | 2002-04-23 | 2003-10-23 | Regeneration of endogenous myocardial tissue |
IL164671A IL164671A (en) | 2002-04-23 | 2004-10-18 | As cxc-type cokin, cells expressing cxc-type cokin or a gene coding for cxc-type cokin for use in improving cardiac activity |
US11/648,769 US8242091B2 (en) | 2002-04-23 | 2006-12-29 | Treatment of tumor with dnazyme directed to peroxiredoxin |
AU2009213032A AU2009213032B2 (en) | 2002-04-23 | 2009-09-10 | Regeneration of endogenous myocardial tissue by induction of neovascularization |
JP2011021983A JP5619644B2 (ja) | 2002-04-23 | 2011-02-03 | 新血管新生の誘導による内因性の心筋組織の再生 |
JP2013249643A JP5911841B2 (ja) | 2002-04-23 | 2013-12-02 | 新血管新生の誘導による内因性の心筋組織の再生 |
US14/196,711 US20150056161A1 (en) | 2002-04-23 | 2014-03-04 | Regeneration of endogenous myocardial tissue |
IL235149A IL235149B (en) | 2002-04-23 | 2014-10-19 | Endogenous tissue regeneration of the heart by induction of neovascularization |
JP2016014321A JP2016155804A (ja) | 2002-04-23 | 2016-01-28 | 新血管新生の誘導による内因性の心筋組織の再生 |
Applications Claiming Priority (1)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
US10/128,738 US20030199464A1 (en) | 2002-04-23 | 2002-04-23 | Regeneration of endogenous myocardial tissue by induction of neovascularization |
Related Child Applications (3)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/US2003/012768 Continuation-In-Part WO2003090512A2 (fr) | 2002-04-23 | 2003-04-23 | Regeneration de tissu myocardique endogene par induction de neovascularisation |
US10/512,518 Continuation-In-Part US7887796B2 (en) | 2002-04-23 | 2003-04-23 | Method of inhibiting collagen formation by VDUP1 inhibition |
US10512518 Continuation-In-Part | 2003-04-23 |
Publications (1)
Publication Number | Publication Date |
---|---|
US20030199464A1 true US20030199464A1 (en) | 2003-10-23 |
Family
ID=29215504
Family Applications (5)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
US10/128,738 Abandoned US20030199464A1 (en) | 2002-04-23 | 2002-04-23 | Regeneration of endogenous myocardial tissue by induction of neovascularization |
US10/512,518 Expired - Fee Related US7887796B2 (en) | 2002-04-23 | 2003-04-23 | Method of inhibiting collagen formation by VDUP1 inhibition |
US10/693,480 Expired - Fee Related US8663652B2 (en) | 2002-04-23 | 2003-10-23 | Regeneration of endogenous myocardial tissue |
US11/648,769 Expired - Fee Related US8242091B2 (en) | 2002-04-23 | 2006-12-29 | Treatment of tumor with dnazyme directed to peroxiredoxin |
US14/196,711 Abandoned US20150056161A1 (en) | 2002-04-23 | 2014-03-04 | Regeneration of endogenous myocardial tissue |
Family Applications After (4)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
US10/512,518 Expired - Fee Related US7887796B2 (en) | 2002-04-23 | 2003-04-23 | Method of inhibiting collagen formation by VDUP1 inhibition |
US10/693,480 Expired - Fee Related US8663652B2 (en) | 2002-04-23 | 2003-10-23 | Regeneration of endogenous myocardial tissue |
US11/648,769 Expired - Fee Related US8242091B2 (en) | 2002-04-23 | 2006-12-29 | Treatment of tumor with dnazyme directed to peroxiredoxin |
US14/196,711 Abandoned US20150056161A1 (en) | 2002-04-23 | 2014-03-04 | Regeneration of endogenous myocardial tissue |
Country Status (9)
Country | Link |
---|---|
US (5) | US20030199464A1 (fr) |
EP (3) | EP2292631B1 (fr) |
JP (4) | JP4897199B2 (fr) |
CN (1) | CN100379751C (fr) |
AU (2) | AU2003231090B2 (fr) |
CA (1) | CA2482996C (fr) |
IL (2) | IL164671A (fr) |
WO (1) | WO2003090512A2 (fr) |
ZA (1) | ZA200408777B (fr) |
Cited By (30)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US20040258670A1 (en) * | 2002-12-05 | 2004-12-23 | Case Western Reserve University | Cell-based therapies for ischemia |
US20050069527A1 (en) * | 2002-12-05 | 2005-03-31 | Case Western Reserve University | Cell-based therapies for ischemia |
US20050158858A1 (en) * | 2000-04-06 | 2005-07-21 | Franco Wayne P. | Growth factor therapy mobilization of stem cells into the periperal blood |
US20050232905A1 (en) * | 2004-03-26 | 2005-10-20 | Yeh Edward T | Use of peripheral blood cells for cardiac regeneration |
US20050233992A1 (en) * | 2002-04-23 | 2005-10-20 | Silviu Itescu | Regeneration of endogeneous myocardial tissue by induction of neovascularization |
US20050265980A1 (en) * | 2004-05-14 | 2005-12-01 | Becton, Dickinson And Company | Cell culture environments for the serum-free expansion of mesenchymal stem cells |
US20060035290A1 (en) * | 2004-08-13 | 2006-02-16 | Medtronic, Inc. | Isolation of endothelial progenitor cell subsets and methods for their use |
US20060111290A1 (en) * | 2000-06-05 | 2006-05-25 | The Trustees Of Columbia University In The City Of New York | Use of SDF-1 or G-CSF to improve myocardial function after ischemic injury |
US20060165667A1 (en) * | 2004-12-03 | 2006-07-27 | Case Western Reserve University | Novel methods, compositions and devices for inducing neovascularization |
US7166280B2 (en) | 2000-04-06 | 2007-01-23 | Franco Wayne P | Combination growth factor therapy and cell therapy for treatment of acute and chronic heart disease |
US20070105217A1 (en) * | 2005-11-07 | 2007-05-10 | Pecora Andrew L | Compositions and methods of vascular injury repair |
US20070135369A1 (en) * | 2005-09-16 | 2007-06-14 | Cooke John P | Methods of modulating angiogenesis and screening compounds for activity in modulating angiogenesis |
US7288521B2 (en) | 2000-04-06 | 2007-10-30 | Franco Wayne P | Growth factor therapy mobilization of stem cells into the peripheral blood |
US20070258943A1 (en) * | 2002-08-22 | 2007-11-08 | Cleveland Clinic Foundation | Genetically engineered cells for therapeutic applications |
US20080241246A1 (en) * | 2006-11-15 | 2008-10-02 | Arteriocyte Inc. | Cell-based therapies for treating liver disease |
EP2094288A2 (fr) * | 2006-10-23 | 2009-09-02 | The Brighamn and Women's Hospital, Inc. | Mutants résistants aux protéases du facteur dérivé des cellules stromales de type 1 utilisés dans la réparation d'une lésion tissulaire |
WO2009155669A1 (fr) * | 2008-06-27 | 2009-12-30 | The Heart Research Institute Ltd | Procédé de traitement de complications vasculaires |
US20100034794A1 (en) * | 2006-10-03 | 2010-02-11 | Van Der Strate Barry W A | Endothelial progenitor cell compositions and neovascularization |
US20100143317A1 (en) * | 2006-10-24 | 2010-06-10 | Andrew Pecora | Infarct area perfusion-improving compositions and methods of vascular injury repair |
US20100166717A1 (en) * | 2002-08-22 | 2010-07-01 | Penn Marc S | Method of treating ischemic disorders |
US20100272679A1 (en) * | 2007-12-14 | 2010-10-28 | Penn Marc S | Compositions and methods of promoting wound healing |
US20100303769A1 (en) * | 2000-04-06 | 2010-12-02 | Franco Wayne P | Combination growth factor therapy and cell therapy for treatment of acute and chronic heart disease |
EP2360242A1 (fr) * | 2004-09-24 | 2011-08-24 | Angioblast Systems, Inc. | Procédé d'amélioration de la prolifération et/ou survie des cellules de précurseurs mesenchymataux (MPC) |
US8343485B2 (en) | 2005-11-07 | 2013-01-01 | Amorcyte, Inc. | Compositions and methods of vascular injury repair |
US8425899B2 (en) | 2005-11-07 | 2013-04-23 | Andrew L. Pecora | Compositions and methods for treating progressive myocardial injury due to a vascular insufficiency |
US8513007B2 (en) | 2009-08-28 | 2013-08-20 | The Cleveland Clinic Foundation | SDF-1 delivery for treating ischemic tissue |
EP2905331A4 (fr) * | 2012-10-05 | 2016-04-06 | Samsung Life Public Welfare Foundation | Composition comprenant un sérum ischémique pour favoriser l'activation d'une cellule souche et procédé favorisant l'activation d'une cellule souche |
US9308277B2 (en) | 2010-02-25 | 2016-04-12 | Mesoblast International Sàrl | Protease-resistant mutants of stromal cell derived factor-1 in the repair of tissue damage |
US9572817B2 (en) | 2008-07-04 | 2017-02-21 | The Heart Research Institute Ltd. | Use of androgens for vascular regeneration and endothelial repair |
US10662234B2 (en) | 2011-06-07 | 2020-05-26 | Mesoblast International Sàrl | Methods for repairing tissue damage using protease-resistant mutants of stromal cell derived factor-1 |
Families Citing this family (47)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US7351421B2 (en) * | 1996-11-05 | 2008-04-01 | Hsing-Wen Sung | Drug-eluting stent having collagen drug carrier chemically treated with genipin |
GB2424273B (en) * | 2002-11-14 | 2007-06-27 | Univ Nottingham | Method for preparing tumour marker protein |
EP1693451B1 (fr) * | 2003-11-21 | 2011-11-09 | Daiichi Sankyo Company, Limited | Procede de culture de cellules myocardiques |
CA2571315C (fr) | 2004-06-21 | 2017-02-14 | The Cleveland Clinic Foundation | Ligands ccr pour domiciliation de cellules souches |
EP1785482B1 (fr) | 2004-08-27 | 2017-10-04 | Daiichi Sankyo Company, Limited | Procede de selection de cellules myocardiques en utilisant des mitochondries intracellulaires comme indice |
EP1856526A4 (fr) * | 2005-01-20 | 2008-11-12 | Univ Rochester | Protéine d'interaction avec la thiorédoxine (txnip) en tant que régulateur de la fonction vasculaire |
TW200734462A (en) | 2006-03-08 | 2007-09-16 | In Motion Invest Ltd | Regulating stem cells |
CN101632020B (zh) * | 2006-09-13 | 2013-11-27 | 昂西免疫有限公司 | 改进的免疫测定方法 |
JP5608635B2 (ja) | 2008-04-07 | 2014-10-15 | カロバイオス ファーマシューティカルズ インコーポレイティッド | 心不全処置のためのgm−csfの中和方法 |
AU2009244790B2 (en) * | 2008-05-09 | 2013-09-12 | Duke University | Treatment for diseases relying on discovery that thioredoxin mediates nitric oxide release in cells |
GB0814302D0 (en) * | 2008-08-05 | 2008-10-01 | Coretherapix Slu | Compounds and methods |
WO2010138180A2 (fr) * | 2009-05-26 | 2010-12-02 | The University Of Vermont And State Agriculture College | Compositions et méthodes utilisables dans le cadre de la réparation du tissu cardiaque |
WO2011071992A1 (fr) * | 2009-12-08 | 2011-06-16 | Health Research, Inc. | Inhibition de l'angiogenèse tumorale par inhibition de la peroxiredoxine 1 (prx1) |
IN2012DN04875A (fr) * | 2009-12-08 | 2015-09-25 | Health Research Inc | |
WO2011081229A1 (fr) * | 2009-12-29 | 2011-07-07 | 이화여자대학교 산학협력단 | Composition pharmaceutique pour l'inhibition de l'angiogenèse |
SG10201913904PA (en) * | 2010-02-25 | 2020-03-30 | Abt Holding Co | Modulation of angiogenesis |
RU2638802C2 (ru) | 2011-05-16 | 2017-12-15 | Джензим Корпорейшн | Применение антагонистов cxcr4 |
AR087364A1 (es) | 2011-07-29 | 2014-03-19 | Pf Medicament | Anticuerpo anti-cxcr4 y su uso para la deteccion y dianostico de canceres |
AR087363A1 (es) | 2011-07-29 | 2014-03-19 | Pf Medicament | Uso del anticuerpo i-3859 para la deteccion y diagnostico de los canceres |
CN102382127A (zh) * | 2011-09-23 | 2012-03-21 | 复旦大学 | 特异性促进心肌细胞发生增殖的心肌小分子化合物及其应用 |
EP2776057B1 (fr) * | 2011-11-07 | 2020-04-15 | Hina W. Chaudhry | Une population de cellules comprenant des cellules dérivées du placenta exprimant cdx2 pour une utilisation dans le traitement d'un tissu cardiaque endommagé ou dégénéré en induisant la régénération cardiaque |
US9585915B2 (en) | 2014-01-23 | 2017-03-07 | Emory University | Polypeptide hydrogels and uses related thereto |
CN106536056B (zh) | 2014-06-13 | 2021-07-16 | 儿童医学中心公司 | 分离线粒体的产品和方法 |
US9375406B2 (en) * | 2014-09-30 | 2016-06-28 | Taigen Biotechnology Co., Ltd. | Substituted pyrimidines for mobilizing cells expressing a type 4 CXC chemokine receptor |
CA2982179A1 (fr) * | 2015-04-09 | 2016-10-13 | Regencor, Inc. | Facteurs paracrine derives de l'epicarde pour la reparation de tissu cardiaque |
US11433136B2 (en) | 2015-12-18 | 2022-09-06 | The General Hospital Corporation | Polyacetal polymers, conjugates, particles and uses thereof |
CN105602888A (zh) * | 2016-01-12 | 2016-05-25 | 温州医科大学附属第一医院 | 一种内皮祖细胞快速扩增体系 |
EP3402490B1 (fr) * | 2016-01-15 | 2022-06-01 | The Children's Medical Center Corporation | Utilisation thérapeutique de mitochondries et d'agents mitochondriaux combinés |
WO2018106738A1 (fr) | 2016-12-05 | 2018-06-14 | Massachusetts Institute Of Technology | Polymères en étoile à bras en brosse, conjugués et particules, et leurs utilisations |
WO2018107686A1 (fr) | 2016-12-15 | 2018-06-21 | 深圳瑞健生命科学研究院有限公司 | Méthode de traitement de l'athérosclérose et de ses complications |
JP7168990B2 (ja) | 2016-12-15 | 2022-11-10 | タレンゲン インターナショナル リミテッド | 肥満症を予防および治療するための方法および薬物 |
US11389515B2 (en) | 2016-12-15 | 2022-07-19 | Talengen International Limited | Method for mitigating heart disease |
WO2018191476A1 (fr) | 2017-04-12 | 2018-10-18 | Magenta Therapeutics, Inc. | Antagonistes de récepteur d'hydrocarbure aryle et utilisations de ceux-ci |
WO2019028094A1 (fr) * | 2017-08-01 | 2019-02-07 | Temple University-Of The Commonwealth System Of Higher Education | Exosomes dérivés de cellules souches d'os cortical pouvant augmenter la fonction cardiaque après une lésion cardiaque |
WO2019089826A1 (fr) | 2017-10-31 | 2019-05-09 | Magenta Therapeutics Inc. | Compositions et procédés de multiplication de cellules souches et progénitrices hématopoïétiques |
CN111683669A (zh) | 2017-10-31 | 2020-09-18 | 美真达治疗公司 | 用于造血干细胞和祖细胞移植疗法的组合物和方法 |
US10058573B1 (en) | 2017-12-06 | 2018-08-28 | Magenta Therapeutics, Inc. | Dosing regimens for the mobilization of hematopoietic stem cells |
US11260079B2 (en) | 2017-12-06 | 2022-03-01 | Magenta Therapeutics, Inc. | Dosing regimens for the mobilization of hematopoietic stem and progenitor cells |
WO2019113375A2 (fr) | 2017-12-06 | 2019-06-13 | Magenta Therapeutics, Inc. | Régimes posologiques pour la mobilisation de cellules souches et progénitrices hématopoïétiques |
CA3087527A1 (fr) | 2018-01-03 | 2019-07-11 | Magenta Therapeutics, Inc. | Compositions et procedes pour l'expansion de cellules souches hematopoietiques et progenitrices et le traitement de troubles metaboliques hereditaires |
WO2019168685A1 (fr) * | 2018-02-27 | 2019-09-06 | Cytori Therapeutics, Inc. | Amélioration de la fonction des cellules endothéliales |
CN109589339A (zh) * | 2018-12-26 | 2019-04-09 | 山西医科大学 | 人子宫内膜内皮祖细胞在改善损伤心肌细胞上的应用 |
WO2021016663A1 (fr) * | 2019-07-30 | 2021-02-04 | Victor Chang Cardiac Research Institute | Cardiomyogenèse induite par klf |
US20220401481A1 (en) | 2019-11-01 | 2022-12-22 | Magenta Therapeutics, Inc. | Dosing regimens for the mobilization of hematopoietic stem and progenitor cells |
IL297690A (en) | 2020-04-27 | 2022-12-01 | Magenta Therapeutics Inc | Methods and compositions for transduction of hematopoietic stem cells and progenitor cells in a living body. |
WO2022197776A1 (fr) | 2021-03-16 | 2022-09-22 | Magenta Therapeutics, Inc. | Schémas posologiques pour la mobilisation des cellules souches hématopoïétiques en vue d'une greffe de cellules souches chez les patients atteints de myélome multiple |
CN117919400B (zh) * | 2024-03-22 | 2024-05-28 | 再少年(北京)生物科技有限公司 | iPS诱导定向分化的成内皮祖细胞和抗体联合治疗心脑血管疾病 |
Citations (6)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US5061620A (en) * | 1990-03-30 | 1991-10-29 | Systemix, Inc. | Human hematopoietic stem cell |
US5552381A (en) * | 1989-07-21 | 1996-09-03 | Washington University | Recombinantly produced human membrane cofactor protein (MCP) pharmaceutical composition, and method of inhibiting complement activity |
US5559703A (en) * | 1993-12-14 | 1996-09-24 | Nissan Motor Co., Ltd. | Fuel cut and ignition timing control system for controlling acceleration slip |
US5880090A (en) * | 1997-09-19 | 1999-03-09 | The Hope Heart Institute | Treatment of vascular graft implants with G-CSF |
US5980887A (en) * | 1996-11-08 | 1999-11-09 | St. Elizabeth's Medical Center Of Boston | Methods for enhancing angiogenesis with endothelial progenitor cells |
US6288103B1 (en) * | 1997-08-07 | 2001-09-11 | Zeneca Limited | Indole derivatives as MCP-1 receptor antagonists |
Family Cites Families (66)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US3687808A (en) | 1969-08-14 | 1972-08-29 | Univ Leland Stanford Junior | Synthetic polynucleotides |
US4469863A (en) | 1980-11-12 | 1984-09-04 | Ts O Paul O P | Nonionic nucleic acid alkyl and aryl phosphonates and processes for manufacture and use thereof |
US5023243A (en) | 1981-10-23 | 1991-06-11 | Molecular Biosystems, Inc. | Oligonucleotide therapeutic agent and method of making same |
US4476301A (en) | 1982-04-29 | 1984-10-09 | Centre National De La Recherche Scientifique | Oligonucleotides, a process for preparing the same and their application as mediators of the action of interferon |
US5550111A (en) | 1984-07-11 | 1996-08-27 | Temple University-Of The Commonwealth System Of Higher Education | Dual action 2',5'-oligoadenylate antiviral derivatives and uses thereof |
US4987071A (en) | 1986-12-03 | 1991-01-22 | University Patents, Inc. | RNA ribozyme polymerases, dephosphorylases, restriction endoribonucleases and methods |
US5264423A (en) | 1987-03-25 | 1993-11-23 | The United States Of America As Represented By The Department Of Health And Human Services | Inhibitors for replication of retroviruses and for the expression of oncogene products |
US5276019A (en) | 1987-03-25 | 1994-01-04 | The United States Of America As Represented By The Department Of Health And Human Services | Inhibitors for replication of retroviruses and for the expression of oncogene products |
US4924624A (en) | 1987-10-22 | 1990-05-15 | Temple University-Of The Commonwealth System Of Higher Education | 2,',5'-phosphorothioate oligoadenylates and plant antiviral uses thereof |
US5188897A (en) | 1987-10-22 | 1993-02-23 | Temple University Of The Commonwealth System Of Higher Education | Encapsulated 2',5'-phosphorothioate oligoadenylates |
EP0406309A4 (en) | 1988-03-25 | 1992-08-19 | The University Of Virginia Alumni Patents Foundation | Oligonucleotide n-alkylphosphoramidates |
US5278302A (en) | 1988-05-26 | 1994-01-11 | University Patents, Inc. | Polynucleotide phosphorodithioates |
CA1340323C (fr) | 1988-09-20 | 1999-01-19 | Arnold E. Hampel | Catalyseur d'arn pour le clivage de sequences specifiques d'arn |
US5194599A (en) | 1988-09-23 | 1993-03-16 | Gilead Sciences, Inc. | Hydrogen phosphonodithioate compositions |
US5008284A (en) * | 1989-02-15 | 1991-04-16 | E. R. Squibb & Sons, Inc. | Method of reducing pre- and post-ischemic myocardial arrhythmias and fibrillation |
US5721218A (en) | 1989-10-23 | 1998-02-24 | Gilead Sciences, Inc. | Oligonucleotides with inverted polarity |
US5399676A (en) | 1989-10-23 | 1995-03-21 | Gilead Sciences | Oligonucleotides with inverted polarity |
JPH05503512A (ja) | 1989-11-29 | 1993-06-10 | ブリガム・アンド・ウイメンズ・ホスピタル | 白血球接着阻害因子としての[ala il―8]↓7↓7 |
US5177198A (en) | 1989-11-30 | 1993-01-05 | University Of N.C. At Chapel Hill | Process for preparing oligoribonucleoside and oligodeoxyribonucleoside boranophosphates |
US5587361A (en) | 1991-10-15 | 1996-12-24 | Isis Pharmaceuticals, Inc. | Oligonucleotides having phosphorothioate linkages of high chiral purity |
US5321131A (en) | 1990-03-08 | 1994-06-14 | Hybridon, Inc. | Site-specific functionalization of oligodeoxynucleotides for non-radioactive labelling |
IE912365A1 (en) * | 1990-07-23 | 1992-01-29 | Zeneca Ltd | Continuous release pharmaceutical compositions |
US5177196A (en) | 1990-08-16 | 1993-01-05 | Microprobe Corporation | Oligo (α-arabinofuranosyl nucleotides) and α-arabinofuranosyl precursors thereof |
US5672697A (en) | 1991-02-08 | 1997-09-30 | Gilead Sciences, Inc. | Nucleoside 5'-methylene phosphonates |
JP2697495B2 (ja) | 1991-06-19 | 1998-01-14 | 富士レビオ株式会社 | アルデヒド誘導体 |
US5571799A (en) | 1991-08-12 | 1996-11-05 | Basco, Ltd. | (2'-5') oligoadenylate analogues useful as inhibitors of host-v5.-graft response |
US5476925A (en) | 1993-02-01 | 1995-12-19 | Northwestern University | Oligodeoxyribonucleotides including 3'-aminonucleoside-phosphoramidate linkages and terminal 3'-amino groups |
GB9304618D0 (en) | 1993-03-06 | 1993-04-21 | Ciba Geigy Ag | Chemical compounds |
US5599703A (en) * | 1993-10-28 | 1997-02-04 | The United States Of America As Represented By The Secretary Of The Navy | In vitro amplification/expansion of CD34+ stem and progenitor cells |
US5625050A (en) | 1994-03-31 | 1997-04-29 | Amgen Inc. | Modified oligonucleotides and intermediates useful in nucleic acid therapeutics |
US5824784A (en) * | 1994-10-12 | 1998-10-20 | Amgen Inc. | N-terminally chemically modified protein compositions and methods |
DE4442665A1 (de) | 1994-11-30 | 1996-06-05 | Gruenenthal Gmbh | Chimäre Proteine mit fibrinolytischen und thrombinhemmenden Eigenschaften |
US6506770B1 (en) * | 1996-06-06 | 2003-01-14 | Anormed, Inc. | Antiviral compounds |
US5980884A (en) * | 1996-02-05 | 1999-11-09 | Amgen, Inc. | Methods for retreatment of patients afflicted with Hepatitis C using consensus interferon |
US6110889A (en) | 1996-06-14 | 2000-08-29 | Board Of Regents, The University Of Texas System | Peptide tumor cell growth inhibitors |
JP3866800B2 (ja) * | 1996-08-29 | 2007-01-10 | 東菱薬品工業株式会社 | アポトーシス関連疾患の予防及び/又は治療薬 |
JP2001507354A (ja) * | 1996-12-20 | 2001-06-05 | クリエイティブ バイオモレキュールズ,インコーポレイテッド | 局所的モルフォゲンまたは形態形成処理された筋形成前駆体細胞による哺乳動物心筋層の処置 |
US6248878B1 (en) | 1996-12-24 | 2001-06-19 | Ribozyme Pharmaceuticals, Inc. | Nucleoside analogs |
US6251666B1 (en) | 1997-03-31 | 2001-06-26 | Ribozyme Pharmaceuticals, Inc. | Nucleic acid catalysts comprising L-nucleotide analogs |
CN1068517C (zh) * | 1997-04-04 | 2001-07-18 | 北京万业经贸发展公司 | 2(3)叔丁基-4-羟基茴香醚在制备防治组织器官损伤性疾病药物中的应用 |
DE69806244T2 (de) | 1997-05-27 | 2002-11-07 | Unichema Chemie B.V., Gouda | Fettsäuren-oligomere |
EP0897980A3 (fr) | 1997-08-20 | 2002-04-17 | Smithkline Beecham Corporation | CXCR4B: Un variant d'épissage humain du récepteur de chemokine CXCR4 |
JP4891477B2 (ja) | 1997-10-02 | 2012-03-07 | マックス−プランク−ゲゼルシャフト ツール フォーデルング デル ヴィッセンシャフテン エー.ヴェー. | 血管新生及び/または既存細動脈網から側枝動脈及び/または他の動脈の発達の調節に関する方法 |
CA2305234A1 (fr) | 1997-10-06 | 1999-04-15 | Acculase Inc. | Techniques d'ablation de tissus et dispositif correspondant |
WO1999037779A1 (fr) | 1998-01-22 | 1999-07-29 | Genentech, Inc. | Conjugues fragments d'anticorps-polymeres et anticorps monoclonaux humanises anti-il-8, et leurs utilisations |
WO1999037751A1 (fr) | 1998-01-23 | 1999-07-29 | Imclone Systems Incorporated | Populations purifiees de cellules souches |
CN100352930C (zh) | 1998-02-27 | 2007-12-05 | 宾夕法尼亚州立大学托管会 | 疫苗、免疫治疗剂及其应用方法 |
CA2322559C (fr) * | 1998-03-09 | 2012-07-17 | St. Elizabeth's Medical Center | Compositions et methodes modulant la vascularisation |
WO1999065507A1 (fr) * | 1998-06-19 | 1999-12-23 | The General Hospital Corporation | Modulation de la fonction plaquettaire |
US20020107195A1 (en) * | 1998-07-21 | 2002-08-08 | Smithkline Beecham Corporation | Method for inducing chemotaxis in endothelial cells by administering stromal cell derived factor-1alpha |
US6124131A (en) * | 1998-08-25 | 2000-09-26 | The Johns Hopkins University School Of Medicine | Mutant hypoxia inducible factor-1 HIF-1 |
DK1171165T3 (da) * | 1999-03-30 | 2007-09-03 | Ran Kornowski | Injektion af autolog knoglemarv i myokardiet |
US20060051334A1 (en) * | 1999-03-30 | 2006-03-09 | Myocardial Therapeutics, Inc. | Injection of bone marrow-derived conditioned medium for angiogenesis |
US20060057722A1 (en) * | 1999-03-30 | 2006-03-16 | Myocardial Therapeutics, Inc. | Conditioned medium of autologous or allogenic progenitor cells for angiogenesis treatment |
EP1207900A2 (fr) * | 1999-08-13 | 2002-05-29 | Chiron Corporation | Dose d'un facteur angiogenique et procede d'administration correspondant destines a ameliorer le debit sanguin du myocarde |
JP5414958B2 (ja) | 2000-06-05 | 2014-02-12 | ザ・トラスティーズ・オブ・コランビア・ユニバーシティー・イン・ザ・シティー・オブ・ニューヨーク | ヒト骨髄由来内皮前駆細胞の同定、及び虚血性傷害後の心筋細胞の機能を改善するためのヒト骨髄由来内皮前駆細胞の使用 |
US7547674B2 (en) * | 2001-06-06 | 2009-06-16 | New York Medical College | Methods and compositions for the repair and/or regeneration of damaged myocardium |
WO2002009650A2 (fr) * | 2000-07-31 | 2002-02-07 | New York Medical College | Compositions et procedes destines a la reparation et/ou la regeneration d'un myocarde endommage |
JP2004506443A (ja) * | 2000-08-22 | 2004-03-04 | ブライハム アンド ウィメンズ ホスピタル インコーポレーテッド | 心血管状態の診断および処置 |
WO2002020848A2 (fr) * | 2000-09-08 | 2002-03-14 | The Regents Of The University Of California | Gene et variation de sequence associes au cancer |
JP3866197B2 (ja) | 2000-09-13 | 2007-01-10 | 中外製薬株式会社 | 虚血性疾患治療剤 |
WO2002036078A2 (fr) | 2000-11-05 | 2002-05-10 | University Of Florida | Ciblage de cellules souches pluripotentielles dans des tissus |
IL146970A0 (en) | 2001-12-06 | 2002-08-14 | Yeda Res & Dev | Migration of haematopoietic stem cells and progenitor cells to the liver |
US20030199464A1 (en) | 2002-04-23 | 2003-10-23 | Silviu Itescu | Regeneration of endogenous myocardial tissue by induction of neovascularization |
US7178755B2 (en) * | 2003-07-30 | 2007-02-20 | Lincoln Global, Inc | Retainer ring for wire package |
US7220407B2 (en) | 2003-10-27 | 2007-05-22 | Amgen Inc. | G-CSF therapy as an adjunct to reperfusion therapy in the treatment of acute myocardial infarction |
-
2002
- 2002-04-23 US US10/128,738 patent/US20030199464A1/en not_active Abandoned
-
2003
- 2003-04-23 EP EP10164397.1A patent/EP2292631B1/fr not_active Expired - Lifetime
- 2003-04-23 EP EP03724217A patent/EP1501852A4/fr not_active Withdrawn
- 2003-04-23 CA CA2482996A patent/CA2482996C/fr not_active Expired - Lifetime
- 2003-04-23 US US10/512,518 patent/US7887796B2/en not_active Expired - Fee Related
- 2003-04-23 WO PCT/US2003/012768 patent/WO2003090512A2/fr active Application Filing
- 2003-04-23 JP JP2003587162A patent/JP4897199B2/ja not_active Expired - Fee Related
- 2003-04-23 EP EP11157749A patent/EP2366706A1/fr not_active Ceased
- 2003-04-23 CN CNB038147157A patent/CN100379751C/zh not_active Expired - Fee Related
- 2003-04-23 ZA ZA200408777A patent/ZA200408777B/en unknown
- 2003-04-23 AU AU2003231090A patent/AU2003231090B2/en not_active Ceased
- 2003-10-23 US US10/693,480 patent/US8663652B2/en not_active Expired - Fee Related
-
2004
- 2004-10-18 IL IL164671A patent/IL164671A/en active IP Right Grant
-
2006
- 2006-12-29 US US11/648,769 patent/US8242091B2/en not_active Expired - Fee Related
-
2009
- 2009-09-10 AU AU2009213032A patent/AU2009213032B2/en not_active Ceased
-
2011
- 2011-02-03 JP JP2011021983A patent/JP5619644B2/ja not_active Expired - Fee Related
-
2013
- 2013-12-02 JP JP2013249643A patent/JP5911841B2/ja not_active Expired - Fee Related
-
2014
- 2014-03-04 US US14/196,711 patent/US20150056161A1/en not_active Abandoned
- 2014-10-19 IL IL235149A patent/IL235149B/en active IP Right Grant
-
2016
- 2016-01-28 JP JP2016014321A patent/JP2016155804A/ja active Pending
Patent Citations (6)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US5552381A (en) * | 1989-07-21 | 1996-09-03 | Washington University | Recombinantly produced human membrane cofactor protein (MCP) pharmaceutical composition, and method of inhibiting complement activity |
US5061620A (en) * | 1990-03-30 | 1991-10-29 | Systemix, Inc. | Human hematopoietic stem cell |
US5559703A (en) * | 1993-12-14 | 1996-09-24 | Nissan Motor Co., Ltd. | Fuel cut and ignition timing control system for controlling acceleration slip |
US5980887A (en) * | 1996-11-08 | 1999-11-09 | St. Elizabeth's Medical Center Of Boston | Methods for enhancing angiogenesis with endothelial progenitor cells |
US6288103B1 (en) * | 1997-08-07 | 2001-09-11 | Zeneca Limited | Indole derivatives as MCP-1 receptor antagonists |
US5880090A (en) * | 1997-09-19 | 1999-03-09 | The Hope Heart Institute | Treatment of vascular graft implants with G-CSF |
Cited By (63)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US7166280B2 (en) | 2000-04-06 | 2007-01-23 | Franco Wayne P | Combination growth factor therapy and cell therapy for treatment of acute and chronic heart disease |
US20100303769A1 (en) * | 2000-04-06 | 2010-12-02 | Franco Wayne P | Combination growth factor therapy and cell therapy for treatment of acute and chronic heart disease |
US20050158858A1 (en) * | 2000-04-06 | 2005-07-21 | Franco Wayne P. | Growth factor therapy mobilization of stem cells into the periperal blood |
US20090155227A1 (en) * | 2000-04-06 | 2009-06-18 | Franco Wayne P | Combination growth factor therapy and cell therapy for treatment of acute and chronic heart disease |
US7291597B2 (en) | 2000-04-06 | 2007-11-06 | Franco Wayne P | Growth factor therapy mobilization of stem cells into the peripheral blood |
US7288521B2 (en) | 2000-04-06 | 2007-10-30 | Franco Wayne P | Growth factor therapy mobilization of stem cells into the peripheral blood |
US20070196343A1 (en) * | 2000-04-06 | 2007-08-23 | Franco Wayne P | Combination growth factor therapy and cell therapy for treatment of acute and chronic heart disease |
US20060111290A1 (en) * | 2000-06-05 | 2006-05-25 | The Trustees Of Columbia University In The City Of New York | Use of SDF-1 or G-CSF to improve myocardial function after ischemic injury |
US7662392B2 (en) | 2000-06-05 | 2010-02-16 | The Trustees Of Columbia University In The City Of New York | Use of SDF-1 or G-CSF to improve myocardial function after ischemic injury |
US8486416B2 (en) | 2000-06-05 | 2013-07-16 | The Trustees Of Columbia University In The City Of New York | Use of SDF-1 to improve ischemic myocardial function |
US8153113B2 (en) | 2000-06-05 | 2012-04-10 | The Trustees Of Columbia University In The City Of New York | Method of increasing trafficking of endothelial progenitor cells to ischemia-damaged tissue |
US9387234B2 (en) | 2000-06-05 | 2016-07-12 | The Trustees Of Columbia University In The City Of New York | Use of SDF-1 to improve ischemic myocardial function |
US20090142296A1 (en) * | 2000-06-05 | 2009-06-04 | The Trustees Of Columbia University In The City Of New York | Method of increasing trafficking of endothelial progenitor cells to ischemia-damaged tissue |
US7887796B2 (en) | 2002-04-23 | 2011-02-15 | The Trustees Of Columbia University In The City Of New York | Method of inhibiting collagen formation by VDUP1 inhibition |
US8663652B2 (en) | 2002-04-23 | 2014-03-04 | The Trustees Of Columbia University In The City Of New York | Regeneration of endogenous myocardial tissue |
US20050233992A1 (en) * | 2002-04-23 | 2005-10-20 | Silviu Itescu | Regeneration of endogeneous myocardial tissue by induction of neovascularization |
US20070172467A1 (en) * | 2002-04-23 | 2007-07-26 | The Trustees Of Columbia University In The City Of New York | Regeneration of endogenous myocardial tissue by induction of neovascularization |
US8242091B2 (en) | 2002-04-23 | 2012-08-14 | The Trustees Of Columbia University In The City Of New York | Treatment of tumor with dnazyme directed to peroxiredoxin |
US20070258943A1 (en) * | 2002-08-22 | 2007-11-08 | Cleveland Clinic Foundation | Genetically engineered cells for therapeutic applications |
US20100166717A1 (en) * | 2002-08-22 | 2010-07-01 | Penn Marc S | Method of treating ischemic disorders |
US9226978B2 (en) | 2002-08-22 | 2016-01-05 | The Cleveland Clinic Foundation | Method of treating ischemic disorders |
US7470538B2 (en) | 2002-12-05 | 2008-12-30 | Case Western Reserve University | Cell-based therapies for ischemia |
US20040258670A1 (en) * | 2002-12-05 | 2004-12-23 | Case Western Reserve University | Cell-based therapies for ischemia |
US20050069527A1 (en) * | 2002-12-05 | 2005-03-31 | Case Western Reserve University | Cell-based therapies for ischemia |
US20050232905A1 (en) * | 2004-03-26 | 2005-10-20 | Yeh Edward T | Use of peripheral blood cells for cardiac regeneration |
US20050265980A1 (en) * | 2004-05-14 | 2005-12-01 | Becton, Dickinson And Company | Cell culture environments for the serum-free expansion of mesenchymal stem cells |
US20060035290A1 (en) * | 2004-08-13 | 2006-02-16 | Medtronic, Inc. | Isolation of endothelial progenitor cell subsets and methods for their use |
EP2360242A1 (fr) * | 2004-09-24 | 2011-08-24 | Angioblast Systems, Inc. | Procédé d'amélioration de la prolifération et/ou survie des cellules de précurseurs mesenchymataux (MPC) |
US20060165667A1 (en) * | 2004-12-03 | 2006-07-27 | Case Western Reserve University | Novel methods, compositions and devices for inducing neovascularization |
US8236772B2 (en) * | 2005-09-16 | 2012-08-07 | The Board Of Trustees Of The Leland Stanford Junior University | Methods of modulating angiogenesis and screening compounds for activity in modulating angiogenesis |
US20070135369A1 (en) * | 2005-09-16 | 2007-06-14 | Cooke John P | Methods of modulating angiogenesis and screening compounds for activity in modulating angiogenesis |
US20070105217A1 (en) * | 2005-11-07 | 2007-05-10 | Pecora Andrew L | Compositions and methods of vascular injury repair |
US7794705B2 (en) | 2005-11-07 | 2010-09-14 | Amorcyte, Inc. | Compositions and methods of vascular injury repair |
US8637005B2 (en) | 2005-11-07 | 2014-01-28 | Amorcyte, Inc. | Compositions and methods of vascular injury repair |
US8425899B2 (en) | 2005-11-07 | 2013-04-23 | Andrew L. Pecora | Compositions and methods for treating progressive myocardial injury due to a vascular insufficiency |
US8088370B2 (en) | 2005-11-07 | 2012-01-03 | Amorcyte, Inc. | Compositions and methods of vascular injury repair |
US20090226402A1 (en) * | 2005-11-07 | 2009-09-10 | Andrew Pecora | Compositions and Methods of Vascular Injury Repair |
US9534202B2 (en) | 2005-11-07 | 2017-01-03 | Amorcyte, Inc. | Compositions and methods for treating progressive myocardial injury due to a vascular insufficiency |
US8343485B2 (en) | 2005-11-07 | 2013-01-01 | Amorcyte, Inc. | Compositions and methods of vascular injury repair |
US20100034794A1 (en) * | 2006-10-03 | 2010-02-11 | Van Der Strate Barry W A | Endothelial progenitor cell compositions and neovascularization |
EP2094288A4 (fr) * | 2006-10-23 | 2010-09-08 | Brighamn And Women S Hospital | Mutants résistants aux protéases du facteur dérivé des cellules stromales de type 1 utilisés dans la réparation d'une lésion tissulaire |
US20100184950A1 (en) * | 2006-10-23 | 2010-07-22 | The Brigham And Women's Hospital, Inc. | Protease resistant mutants of stromal cell derived factor-1 in the repair of tissue damage |
US7999067B2 (en) | 2006-10-23 | 2011-08-16 | The Brigham And Women's Hospital, Inc. | Protease resistant mutants of stromal cell derived factor-1 in the repair of tissue damage |
US10774124B2 (en) | 2006-10-23 | 2020-09-15 | The Brigham And Women's Hospital, Inc. | Protease resistant mutants of stromal cell derived factor-1 in the repair of tissue damage |
US9631005B2 (en) | 2006-10-23 | 2017-04-25 | The Brigham And Women's Hospital, Inc. | Protease resistant mutants of stromal cell derived factor-1 in the repair of tissue damage |
EP2094288A2 (fr) * | 2006-10-23 | 2009-09-02 | The Brighamn and Women's Hospital, Inc. | Mutants résistants aux protéases du facteur dérivé des cellules stromales de type 1 utilisés dans la réparation d'une lésion tissulaire |
EP2676674A1 (fr) * | 2006-10-23 | 2013-12-25 | The Brigham and Women's Hospital, Inc. | Mutants résistants aux protéases du facteur 1 dérivé des cellules stromales dans la réparation d'une lésion tissulaire |
US9034316B2 (en) | 2006-10-24 | 2015-05-19 | Amorcyte, Llc | Infarct area perfusion-improving compositions and methods of vascular injury repair |
US20100143317A1 (en) * | 2006-10-24 | 2010-06-10 | Andrew Pecora | Infarct area perfusion-improving compositions and methods of vascular injury repair |
US20080241246A1 (en) * | 2006-11-15 | 2008-10-02 | Arteriocyte Inc. | Cell-based therapies for treating liver disease |
US8679477B2 (en) | 2007-12-14 | 2014-03-25 | The Cleveland Clinic Foundation | Use of SDF-1 to mitigate scar formation |
US20100272679A1 (en) * | 2007-12-14 | 2010-10-28 | Penn Marc S | Compositions and methods of promoting wound healing |
EP2565275A1 (fr) * | 2008-06-27 | 2013-03-06 | The Heart Research Institute Limited | Procédé de traitement de complications vasculaires en utilisant des modulateurs de TRX et TRXNIP |
WO2009155669A1 (fr) * | 2008-06-27 | 2009-12-30 | The Heart Research Institute Ltd | Procédé de traitement de complications vasculaires |
US9572817B2 (en) | 2008-07-04 | 2017-02-21 | The Heart Research Institute Ltd. | Use of androgens for vascular regeneration and endothelial repair |
US8513213B2 (en) | 2009-08-28 | 2013-08-20 | The Cleveland Clinic Foundation | SDF-1 delivery for treating ischemic tissue |
US8513007B2 (en) | 2009-08-28 | 2013-08-20 | The Cleveland Clinic Foundation | SDF-1 delivery for treating ischemic tissue |
US9844581B2 (en) | 2009-08-28 | 2017-12-19 | The Cleveland Clinic | SDF-1 delivery for treating ischemic tissue |
US8883756B2 (en) | 2009-08-28 | 2014-11-11 | Juventas Therapeutics, Inc. | SDF-1 delivery for treating ischemic tissue |
US9308277B2 (en) | 2010-02-25 | 2016-04-12 | Mesoblast International Sàrl | Protease-resistant mutants of stromal cell derived factor-1 in the repair of tissue damage |
US10456451B2 (en) | 2010-02-25 | 2019-10-29 | Mesoblast International Sàrl | Protease-resistant mutants of stromal cell derived factor-1 in the repair of tissue damage |
US10662234B2 (en) | 2011-06-07 | 2020-05-26 | Mesoblast International Sàrl | Methods for repairing tissue damage using protease-resistant mutants of stromal cell derived factor-1 |
EP2905331A4 (fr) * | 2012-10-05 | 2016-04-06 | Samsung Life Public Welfare Foundation | Composition comprenant un sérum ischémique pour favoriser l'activation d'une cellule souche et procédé favorisant l'activation d'une cellule souche |
Also Published As
Publication number | Publication date |
---|---|
CA2482996A1 (fr) | 2003-11-06 |
CN1662548A (zh) | 2005-08-31 |
US20050233992A1 (en) | 2005-10-20 |
WO2003090512A2 (fr) | 2003-11-06 |
US20070172467A1 (en) | 2007-07-26 |
IL164671A (en) | 2014-11-30 |
EP2292631B1 (fr) | 2017-02-15 |
US20150056161A1 (en) | 2015-02-26 |
US7887796B2 (en) | 2011-02-15 |
CN100379751C (zh) | 2008-04-09 |
EP2366706A1 (fr) | 2011-09-21 |
EP2292631A1 (fr) | 2011-03-09 |
AU2003231090B2 (en) | 2009-10-01 |
WO2003090512A3 (fr) | 2004-11-04 |
AU2009213032A1 (en) | 2009-10-08 |
ZA200408777B (en) | 2007-04-25 |
JP5911841B2 (ja) | 2016-05-11 |
EP1501852A4 (fr) | 2008-10-01 |
IL164671A0 (en) | 2005-12-18 |
AU2009213032B2 (en) | 2013-06-27 |
EP1501852A2 (fr) | 2005-02-02 |
JP2005534290A (ja) | 2005-11-17 |
JP2011137011A (ja) | 2011-07-14 |
IL235149B (en) | 2019-03-31 |
US8663652B2 (en) | 2014-03-04 |
JP2016155804A (ja) | 2016-09-01 |
US20040247564A1 (en) | 2004-12-09 |
US8242091B2 (en) | 2012-08-14 |
JP2014088390A (ja) | 2014-05-15 |
AU2003231090A1 (en) | 2003-11-10 |
JP4897199B2 (ja) | 2012-03-14 |
JP5619644B2 (ja) | 2014-11-05 |
CA2482996C (fr) | 2017-02-07 |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
US8242091B2 (en) | Treatment of tumor with dnazyme directed to peroxiredoxin | |
EP2324839B1 (fr) | Filgrastim pour usage de traitement de l' infarctus du myocard | |
Grote et al. | The angiogenic factor CCN1 promotes adhesion and migration of circulating CD34+ progenitor cells: potential role in angiogenesis and endothelial regeneration | |
US10780128B2 (en) | Mesenchymal stem cell particles |
Legal Events
Date | Code | Title | Description |
---|---|---|---|
AS | Assignment |
Owner name: TRUSTEES OF COLUMBIA UNIVERSITY IN THE CITY OF NEW Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:ITESCU, SILVIU;REEL/FRAME:015537/0863 Effective date: 20040616 |
|
STCB | Information on status: application discontinuation |
Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION |