WO2020077030A1 - Aggf1 and aggf1-primed cells for treating diseases and conditions - Google Patents
Aggf1 and aggf1-primed cells for treating diseases and conditions Download PDFInfo
- Publication number
- WO2020077030A1 WO2020077030A1 PCT/US2019/055513 US2019055513W WO2020077030A1 WO 2020077030 A1 WO2020077030 A1 WO 2020077030A1 US 2019055513 W US2019055513 W US 2019055513W WO 2020077030 A1 WO2020077030 A1 WO 2020077030A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- aggf1
- peptide
- seq
- composition
- subject
- Prior art date
Links
- 201000010099 disease Diseases 0.000 title claims abstract description 36
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 title claims abstract description 36
- 101000690725 Homo sapiens Angiogenic factor with G patch and FHA domains 1 Proteins 0.000 claims abstract description 373
- 102100026683 Angiogenic factor with G patch and FHA domains 1 Human genes 0.000 claims abstract description 262
- NOESYZHRGYRDHS-UHFFFAOYSA-N insulin Chemical compound N1C(=O)C(NC(=O)C(CCC(N)=O)NC(=O)C(CCC(O)=O)NC(=O)C(C(C)C)NC(=O)C(NC(=O)CN)C(C)CC)CSSCC(C(NC(CO)C(=O)NC(CC(C)C)C(=O)NC(CC=2C=CC(O)=CC=2)C(=O)NC(CCC(N)=O)C(=O)NC(CC(C)C)C(=O)NC(CCC(O)=O)C(=O)NC(CC(N)=O)C(=O)NC(CC=2C=CC(O)=CC=2)C(=O)NC(CSSCC(NC(=O)C(C(C)C)NC(=O)C(CC(C)C)NC(=O)C(CC=2C=CC(O)=CC=2)NC(=O)C(CC(C)C)NC(=O)C(C)NC(=O)C(CCC(O)=O)NC(=O)C(C(C)C)NC(=O)C(CC(C)C)NC(=O)C(CC=2NC=NC=2)NC(=O)C(CO)NC(=O)CNC2=O)C(=O)NCC(=O)NC(CCC(O)=O)C(=O)NC(CCCNC(N)=N)C(=O)NCC(=O)NC(CC=3C=CC=CC=3)C(=O)NC(CC=3C=CC=CC=3)C(=O)NC(CC=3C=CC(O)=CC=3)C(=O)NC(C(C)O)C(=O)N3C(CCC3)C(=O)NC(CCCCN)C(=O)NC(C)C(O)=O)C(=O)NC(CC(N)=O)C(O)=O)=O)NC(=O)C(C(C)CC)NC(=O)C(CO)NC(=O)C(C(C)O)NC(=O)C1CSSCC2NC(=O)C(CC(C)C)NC(=O)C(NC(=O)C(CCC(N)=O)NC(=O)C(CC(N)=O)NC(=O)C(NC(=O)C(N)CC=1C=CC=CC=1)C(C)C)CC1=CN=CN1 NOESYZHRGYRDHS-UHFFFAOYSA-N 0.000 claims abstract description 106
- 239000000203 mixture Substances 0.000 claims abstract description 64
- 108090001061 Insulin Proteins 0.000 claims abstract description 58
- 238000000034 method Methods 0.000 claims abstract description 57
- 102000004877 Insulin Human genes 0.000 claims abstract description 54
- 229940125396 insulin Drugs 0.000 claims abstract description 53
- 239000013604 expression vector Substances 0.000 claims abstract description 45
- 230000003511 endothelial effect Effects 0.000 claims abstract description 44
- 210000000130 stem cell Anatomy 0.000 claims abstract description 31
- 108090000765 processed proteins & peptides Proteins 0.000 claims abstract description 15
- 230000000302 ischemic effect Effects 0.000 claims abstract description 14
- 230000002159 abnormal effect Effects 0.000 claims abstract description 13
- 230000004153 glucose metabolism Effects 0.000 claims abstract description 12
- 208000019553 vascular disease Diseases 0.000 claims abstract description 12
- 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 claims description 66
- 239000008103 glucose Substances 0.000 claims description 64
- 150000001413 amino acids Chemical class 0.000 claims description 55
- 206010064911 Pulmonary arterial hypertension Diseases 0.000 claims description 35
- 125000003275 alpha amino acid group Chemical group 0.000 claims description 31
- 208000001072 type 2 diabetes mellitus Diseases 0.000 claims description 24
- 201000001421 hyperglycemia Diseases 0.000 claims description 22
- 208000029078 coronary artery disease Diseases 0.000 claims description 19
- 210000004369 blood Anatomy 0.000 claims description 15
- 239000008280 blood Substances 0.000 claims description 15
- 208000030613 peripheral artery disease Diseases 0.000 claims description 12
- 208000010125 myocardial infarction Diseases 0.000 claims description 10
- 238000012360 testing method Methods 0.000 claims description 9
- 102000052716 human AGGF1 Human genes 0.000 claims description 7
- 238000004519 manufacturing process Methods 0.000 claims description 7
- 206010019280 Heart failures Diseases 0.000 claims description 6
- 239000007788 liquid Substances 0.000 claims description 6
- 208000024172 Cardiovascular disease Diseases 0.000 claims description 5
- 208000002815 pulmonary hypertension Diseases 0.000 claims description 5
- 206010007572 Cardiac hypertrophy Diseases 0.000 claims description 4
- 208000006029 Cardiomegaly Diseases 0.000 claims description 4
- 208000007536 Thrombosis Diseases 0.000 claims description 4
- 206010067584 Type 1 diabetes mellitus Diseases 0.000 claims description 4
- 238000012806 monitoring device Methods 0.000 claims description 4
- 208000024891 symptom Diseases 0.000 claims description 4
- 206010060378 Hyperinsulinaemia Diseases 0.000 claims description 3
- 230000003451 hyperinsulinaemic effect Effects 0.000 claims description 3
- 201000008980 hyperinsulinism Diseases 0.000 claims description 3
- 206010072557 Peripheral artery restenosis Diseases 0.000 claims description 2
- 238000002399 angioplasty Methods 0.000 claims description 2
- 108091028043 Nucleic acid sequence Proteins 0.000 claims 2
- 150000007523 nucleic acids Chemical group 0.000 claims 2
- 206010012601 diabetes mellitus Diseases 0.000 abstract description 76
- 239000012634 fragment Substances 0.000 abstract description 4
- 102000004196 processed proteins & peptides Human genes 0.000 abstract description 4
- 241000699670 Mus sp. Species 0.000 description 167
- 238000011282 treatment Methods 0.000 description 62
- 238000012217 deletion Methods 0.000 description 53
- 230000037430 deletion Effects 0.000 description 52
- 230000000694 effects Effects 0.000 description 49
- 101000588302 Homo sapiens Nuclear factor erythroid 2-related factor 2 Proteins 0.000 description 47
- 102100031701 Nuclear factor erythroid 2-related factor 2 Human genes 0.000 description 47
- 235000009200 high fat diet Nutrition 0.000 description 46
- 210000004027 cell Anatomy 0.000 description 40
- 230000001225 therapeutic effect Effects 0.000 description 35
- 230000006870 function Effects 0.000 description 34
- 230000014509 gene expression Effects 0.000 description 28
- 238000013508 migration Methods 0.000 description 28
- 230000001965 increasing effect Effects 0.000 description 26
- 230000005012 migration Effects 0.000 description 26
- 108020004459 Small interfering RNA Proteins 0.000 description 21
- 230000033115 angiogenesis Effects 0.000 description 20
- 210000005087 mononuclear cell Anatomy 0.000 description 20
- 108010021466 Mutant Proteins Proteins 0.000 description 19
- 102000008300 Mutant Proteins Human genes 0.000 description 19
- 238000002560 therapeutic procedure Methods 0.000 description 19
- 238000001262 western blot Methods 0.000 description 19
- 238000003556 assay Methods 0.000 description 18
- 238000005516 engineering process Methods 0.000 description 18
- 239000007924 injection Substances 0.000 description 17
- 238000002347 injection Methods 0.000 description 17
- 230000015572 biosynthetic process Effects 0.000 description 16
- 230000037396 body weight Effects 0.000 description 16
- 210000002889 endothelial cell Anatomy 0.000 description 16
- 210000003141 lower extremity Anatomy 0.000 description 16
- 238000010232 migration assay Methods 0.000 description 16
- 238000009163 protein therapy Methods 0.000 description 16
- 238000012744 immunostaining Methods 0.000 description 14
- 230000035755 proliferation Effects 0.000 description 14
- 108090000623 proteins and genes Proteins 0.000 description 12
- 230000002829 reductive effect Effects 0.000 description 12
- 208000028867 ischemia Diseases 0.000 description 11
- 230000003247 decreasing effect Effects 0.000 description 10
- 239000001963 growth medium Substances 0.000 description 10
- 201000002818 limb ischemia Diseases 0.000 description 10
- 239000013642 negative control Substances 0.000 description 10
- 239000003642 reactive oxygen metabolite Substances 0.000 description 10
- 230000002792 vascular Effects 0.000 description 10
- QDLHCMPXEPAAMD-QAIWCSMKSA-N wortmannin Chemical compound C1([C@]2(C)C3=C(C4=O)OC=C3C(=O)O[C@@H]2COC)=C4[C@@H]2CCC(=O)[C@@]2(C)C[C@H]1OC(C)=O QDLHCMPXEPAAMD-QAIWCSMKSA-N 0.000 description 10
- QDLHCMPXEPAAMD-UHFFFAOYSA-N wortmannin Natural products COCC1OC(=O)C2=COC(C3=O)=C2C1(C)C1=C3C2CCC(=O)C2(C)CC1OC(C)=O QDLHCMPXEPAAMD-UHFFFAOYSA-N 0.000 description 10
- 101000851007 Homo sapiens Vascular endothelial growth factor receptor 2 Proteins 0.000 description 9
- 102100024616 Platelet endothelial cell adhesion molecule Human genes 0.000 description 9
- 102100033177 Vascular endothelial growth factor receptor 2 Human genes 0.000 description 9
- 208000027418 Wounds and injury Diseases 0.000 description 9
- 239000003795 chemical substances by application Substances 0.000 description 9
- 239000003814 drug Substances 0.000 description 9
- 239000000243 solution Substances 0.000 description 9
- 238000002054 transplantation Methods 0.000 description 9
- 102100031650 C-X-C chemokine receptor type 4 Human genes 0.000 description 8
- 101000922348 Homo sapiens C-X-C chemokine receptor type 4 Proteins 0.000 description 8
- 102000005789 Vascular Endothelial Growth Factors Human genes 0.000 description 8
- 108010019530 Vascular Endothelial Growth Factors Proteins 0.000 description 8
- 238000004458 analytical method Methods 0.000 description 8
- 230000002491 angiogenic effect Effects 0.000 description 8
- 230000017531 blood circulation Effects 0.000 description 8
- 210000001185 bone marrow Anatomy 0.000 description 8
- 230000004663 cell proliferation Effects 0.000 description 8
- 230000004942 nuclear accumulation Effects 0.000 description 8
- 238000006366 phosphorylation reaction Methods 0.000 description 8
- 210000001147 pulmonary artery Anatomy 0.000 description 8
- 102100031181 Glyceraldehyde-3-phosphate dehydrogenase Human genes 0.000 description 7
- 206010052428 Wound Diseases 0.000 description 7
- 230000001464 adherent effect Effects 0.000 description 7
- 230000027455 binding Effects 0.000 description 7
- 230000006735 deficit Effects 0.000 description 7
- 229940079593 drug Drugs 0.000 description 7
- 108020004445 glyceraldehyde-3-phosphate dehydrogenase Proteins 0.000 description 7
- 230000001771 impaired effect Effects 0.000 description 7
- 230000001404 mediated effect Effects 0.000 description 7
- 108020004999 messenger RNA Proteins 0.000 description 7
- 238000010172 mouse model Methods 0.000 description 7
- 230000036542 oxidative stress Effects 0.000 description 7
- 230000026731 phosphorylation Effects 0.000 description 7
- 238000012755 real-time RT-PCR analysis Methods 0.000 description 7
- 230000012292 cell migration Effects 0.000 description 6
- 150000001875 compounds Chemical class 0.000 description 6
- 230000004064 dysfunction Effects 0.000 description 6
- 230000010595 endothelial cell migration Effects 0.000 description 6
- 108010048367 enhanced green fluorescent protein Proteins 0.000 description 6
- 238000002474 experimental method Methods 0.000 description 6
- 238000003197 gene knockdown Methods 0.000 description 6
- 230000003345 hyperglycaemic effect Effects 0.000 description 6
- 238000001727 in vivo Methods 0.000 description 6
- 208000015181 infectious disease Diseases 0.000 description 6
- 238000011068 loading method Methods 0.000 description 6
- 230000007246 mechanism Effects 0.000 description 6
- 230000002685 pulmonary effect Effects 0.000 description 6
- 230000006496 vascular abnormality Effects 0.000 description 6
- XLYOFNOQVPJJNP-UHFFFAOYSA-N water Substances O XLYOFNOQVPJJNP-UHFFFAOYSA-N 0.000 description 6
- 206010048554 Endothelial dysfunction Diseases 0.000 description 5
- 241000713666 Lentivirus Species 0.000 description 5
- 206010028851 Necrosis Diseases 0.000 description 5
- 102000038030 PI3Ks Human genes 0.000 description 5
- 108091007960 PI3Ks Proteins 0.000 description 5
- 239000013543 active substance Substances 0.000 description 5
- 230000003078 antioxidant effect Effects 0.000 description 5
- 230000008901 benefit Effects 0.000 description 5
- 210000004899 c-terminal region Anatomy 0.000 description 5
- 230000000747 cardiac effect Effects 0.000 description 5
- 238000004113 cell culture Methods 0.000 description 5
- 235000005911 diet Nutrition 0.000 description 5
- 230000037213 diet Effects 0.000 description 5
- 239000012149 elution buffer Substances 0.000 description 5
- 230000008694 endothelial dysfunction Effects 0.000 description 5
- 239000007928 intraperitoneal injection Substances 0.000 description 5
- 230000017074 necrotic cell death Effects 0.000 description 5
- 230000030648 nucleus localization Effects 0.000 description 5
- -1 polyoxyethylene Polymers 0.000 description 5
- 230000019491 signal transduction Effects 0.000 description 5
- 239000007929 subcutaneous injection Substances 0.000 description 5
- 238000010254 subcutaneous injection Methods 0.000 description 5
- 238000001356 surgical procedure Methods 0.000 description 5
- 108010010803 Gelatin Proteins 0.000 description 4
- 206010021143 Hypoxia Diseases 0.000 description 4
- 206010022489 Insulin Resistance Diseases 0.000 description 4
- 239000004698 Polyethylene Substances 0.000 description 4
- 208000032594 Vascular Remodeling Diseases 0.000 description 4
- 230000008081 blood perfusion Effects 0.000 description 4
- 210000000988 bone and bone Anatomy 0.000 description 4
- 238000003352 cell adhesion assay Methods 0.000 description 4
- 230000001684 chronic effect Effects 0.000 description 4
- 230000004087 circulation Effects 0.000 description 4
- 230000000254 damaging effect Effects 0.000 description 4
- 230000034994 death Effects 0.000 description 4
- 231100000517 death Toxicity 0.000 description 4
- 238000013118 diabetic mouse model Methods 0.000 description 4
- 239000003085 diluting agent Substances 0.000 description 4
- 238000000684 flow cytometry Methods 0.000 description 4
- 239000008273 gelatin Substances 0.000 description 4
- 229920000159 gelatin Polymers 0.000 description 4
- 235000019322 gelatine Nutrition 0.000 description 4
- 235000011852 gelatine desserts Nutrition 0.000 description 4
- 230000010030 glucose lowering effect Effects 0.000 description 4
- 238000002513 implantation Methods 0.000 description 4
- 239000003112 inhibitor Substances 0.000 description 4
- 230000002045 lasting effect Effects 0.000 description 4
- 239000002609 medium Substances 0.000 description 4
- 239000008194 pharmaceutical composition Substances 0.000 description 4
- 239000000546 pharmaceutical excipient Substances 0.000 description 4
- 238000007747 plating Methods 0.000 description 4
- 102000004169 proteins and genes Human genes 0.000 description 4
- 230000008439 repair process Effects 0.000 description 4
- 230000002441 reversible effect Effects 0.000 description 4
- IZTQOLKUZKXIRV-YRVFCXMDSA-N sincalide Chemical compound C([C@@H](C(=O)N[C@@H](CCSC)C(=O)NCC(=O)N[C@@H](CC=1C2=CC=CC=C2NC=1)C(=O)N[C@@H](CCSC)C(=O)N[C@@H](CC(O)=O)C(=O)N[C@@H](CC=1C=CC=CC=1)C(N)=O)NC(=O)[C@@H](N)CC(O)=O)C1=CC=C(OS(O)(=O)=O)C=C1 IZTQOLKUZKXIRV-YRVFCXMDSA-N 0.000 description 4
- ZSJLQEPLLKMAKR-GKHCUFPYSA-N streptozocin Chemical compound O=NN(C)C(=O)N[C@H]1[C@@H](O)O[C@H](CO)[C@@H](O)[C@@H]1O ZSJLQEPLLKMAKR-GKHCUFPYSA-N 0.000 description 4
- 230000002459 sustained effect Effects 0.000 description 4
- 210000001519 tissue Anatomy 0.000 description 4
- 210000004509 vascular smooth muscle cell Anatomy 0.000 description 4
- 230000007730 Akt signaling Effects 0.000 description 3
- WVDDGKGOMKODPV-UHFFFAOYSA-N Benzyl alcohol Chemical compound OCC1=CC=CC=C1 WVDDGKGOMKODPV-UHFFFAOYSA-N 0.000 description 3
- 108091003079 Bovine Serum Albumin Proteins 0.000 description 3
- 238000011746 C57BL/6J (JAX™ mouse strain) Methods 0.000 description 3
- 208000002705 Glucose Intolerance Diseases 0.000 description 3
- 102100031573 Hematopoietic progenitor cell antigen CD34 Human genes 0.000 description 3
- 241000282412 Homo Species 0.000 description 3
- 101000777663 Homo sapiens Hematopoietic progenitor cell antigen CD34 Proteins 0.000 description 3
- 101000738771 Homo sapiens Receptor-type tyrosine-protein phosphatase C Proteins 0.000 description 3
- 206010020880 Hypertrophy Diseases 0.000 description 3
- 208000013016 Hypoglycemia Diseases 0.000 description 3
- WQZGKKKJIJFFOK-ZZWDRFIYSA-N L-glucose Chemical compound OC[C@@H]1OC(O)[C@@H](O)[C@H](O)[C@H]1O WQZGKKKJIJFFOK-ZZWDRFIYSA-N 0.000 description 3
- 229930040373 Paraformaldehyde Natural products 0.000 description 3
- DNIAPMSPPWPWGF-UHFFFAOYSA-N Propylene glycol Chemical compound CC(O)CO DNIAPMSPPWPWGF-UHFFFAOYSA-N 0.000 description 3
- 102100037422 Receptor-type tyrosine-protein phosphatase C Human genes 0.000 description 3
- 108010087230 Sincalide Proteins 0.000 description 3
- ZSJLQEPLLKMAKR-UHFFFAOYSA-N Streptozotocin Natural products O=NN(C)C(=O)NC1C(O)OC(CO)C(O)C1O ZSJLQEPLLKMAKR-UHFFFAOYSA-N 0.000 description 3
- 230000032683 aging Effects 0.000 description 3
- 125000000539 amino acid group Chemical group 0.000 description 3
- 230000000692 anti-sense effect Effects 0.000 description 3
- 239000003963 antioxidant agent Substances 0.000 description 3
- 230000004872 arterial blood pressure Effects 0.000 description 3
- 230000002238 attenuated effect Effects 0.000 description 3
- 239000000872 buffer Substances 0.000 description 3
- 230000015556 catabolic process Effects 0.000 description 3
- 238000010609 cell counting kit-8 assay Methods 0.000 description 3
- 238000002659 cell therapy Methods 0.000 description 3
- 238000012512 characterization method Methods 0.000 description 3
- 239000011248 coating agent Substances 0.000 description 3
- 239000002299 complementary DNA Substances 0.000 description 3
- 230000007423 decrease Effects 0.000 description 3
- 238000006731 degradation reaction Methods 0.000 description 3
- 238000000432 density-gradient centrifugation Methods 0.000 description 3
- 230000001419 dependent effect Effects 0.000 description 3
- 238000011161 development Methods 0.000 description 3
- 230000018109 developmental process Effects 0.000 description 3
- 238000010586 diagram Methods 0.000 description 3
- 238000009826 distribution Methods 0.000 description 3
- 210000003038 endothelium Anatomy 0.000 description 3
- 239000012091 fetal bovine serum Substances 0.000 description 3
- 239000000499 gel Substances 0.000 description 3
- 238000007446 glucose tolerance test Methods 0.000 description 3
- 230000012010 growth Effects 0.000 description 3
- 230000036541 health Effects 0.000 description 3
- 238000007490 hematoxylin and eosin (H&E) staining Methods 0.000 description 3
- 230000002218 hypoglycaemic effect Effects 0.000 description 3
- 230000007954 hypoxia Effects 0.000 description 3
- 230000001976 improved effect Effects 0.000 description 3
- 230000003993 interaction Effects 0.000 description 3
- 238000012423 maintenance Methods 0.000 description 3
- 108010082117 matrigel Proteins 0.000 description 3
- 210000004379 membrane Anatomy 0.000 description 3
- 239000012528 membrane Substances 0.000 description 3
- 230000004048 modification Effects 0.000 description 3
- 238000012986 modification Methods 0.000 description 3
- 230000009456 molecular mechanism Effects 0.000 description 3
- 210000003205 muscle Anatomy 0.000 description 3
- 229920002866 paraformaldehyde Polymers 0.000 description 3
- 230000002093 peripheral effect Effects 0.000 description 3
- 239000013612 plasmid Substances 0.000 description 3
- 201000009104 prediabetes syndrome Diseases 0.000 description 3
- 238000000746 purification Methods 0.000 description 3
- 230000001105 regulatory effect Effects 0.000 description 3
- 150000003839 salts Chemical class 0.000 description 3
- 230000011664 signaling Effects 0.000 description 3
- 210000002027 skeletal muscle Anatomy 0.000 description 3
- 238000002415 sodium dodecyl sulfate polyacrylamide gel electrophoresis Methods 0.000 description 3
- 230000002269 spontaneous effect Effects 0.000 description 3
- 229960001052 streptozocin Drugs 0.000 description 3
- 239000000725 suspension Substances 0.000 description 3
- 210000002303 tibia Anatomy 0.000 description 3
- 210000000689 upper leg Anatomy 0.000 description 3
- 230000006492 vascular dysfunction Effects 0.000 description 3
- 210000003462 vein Anatomy 0.000 description 3
- 230000002861 ventricular Effects 0.000 description 3
- AZQWKYJCGOJGHM-UHFFFAOYSA-N 1,4-benzoquinone Chemical compound O=C1C=CC(=O)C=C1 AZQWKYJCGOJGHM-UHFFFAOYSA-N 0.000 description 2
- VBICKXHEKHSIBG-UHFFFAOYSA-N 1-monostearoylglycerol Chemical compound CCCCCCCCCCCCCCCCCC(=O)OCC(O)CO VBICKXHEKHSIBG-UHFFFAOYSA-N 0.000 description 2
- IZHVBANLECCAGF-UHFFFAOYSA-N 2-hydroxy-3-(octadecanoyloxy)propyl octadecanoate Chemical compound CCCCCCCCCCCCCCCCCC(=O)OCC(O)COC(=O)CCCCCCCCCCCCCCCCC IZHVBANLECCAGF-UHFFFAOYSA-N 0.000 description 2
- FJKROLUGYXJWQN-UHFFFAOYSA-N 4-hydroxybenzoic acid Chemical compound OC(=O)C1=CC=C(O)C=C1 FJKROLUGYXJWQN-UHFFFAOYSA-N 0.000 description 2
- 238000011537 Coomassie blue staining Methods 0.000 description 2
- 208000002249 Diabetes Complications Diseases 0.000 description 2
- 208000032781 Diabetic cardiomyopathy Diseases 0.000 description 2
- 239000006144 Dulbecco’s modified Eagle's medium Substances 0.000 description 2
- WZUVPPKBWHMQCE-UHFFFAOYSA-N Haematoxylin Chemical compound C12=CC(O)=C(O)C=C2CC2(O)C1C1=CC=C(O)C(O)=C1OC2 WZUVPPKBWHMQCE-UHFFFAOYSA-N 0.000 description 2
- 108010057186 Insulin Glargine Proteins 0.000 description 2
- COCFEDIXXNGUNL-RFKWWTKHSA-N Insulin glargine Chemical compound C([C@@H](C(=O)N[C@@H](CC(C)C)C(=O)N[C@H]1CSSC[C@H]2C(=O)N[C@H](C(=O)N[C@@H](CO)C(=O)N[C@H](C(=O)N[C@H](C(N[C@@H](CO)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CC=3C=CC(O)=CC=3)C(=O)N[C@@H](CCC(N)=O)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](CC(N)=O)C(=O)N[C@@H](CC=3C=CC(O)=CC=3)C(=O)N[C@@H](CSSC[C@H](NC(=O)[C@H](C(C)C)NC(=O)[C@H](CC(C)C)NC(=O)[C@H](CC=3C=CC(O)=CC=3)NC(=O)[C@H](CC(C)C)NC(=O)[C@H](C)NC(=O)[C@H](CCC(O)=O)NC(=O)[C@H](C(C)C)NC(=O)[C@H](CC(C)C)NC(=O)[C@H](CC=3NC=NC=3)NC(=O)[C@H](CO)NC(=O)CNC1=O)C(=O)NCC(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](CCCNC(N)=N)C(=O)NCC(=O)N[C@@H](CC=1C=CC=CC=1)C(=O)N[C@@H](CC=1C=CC=CC=1)C(=O)N[C@@H](CC=1C=CC(O)=CC=1)C(=O)N[C@@H]([C@@H](C)O)C(=O)N1[C@@H](CCC1)C(=O)N[C@@H](CCCCN)C(=O)N[C@@H]([C@@H](C)O)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CCCNC(N)=N)C(O)=O)C(=O)NCC(O)=O)=O)CSSC[C@@H](C(N2)=O)NC(=O)[C@H](CCC(N)=O)NC(=O)[C@H](CCC(O)=O)NC(=O)[C@H](C(C)C)NC(=O)[C@@H](NC(=O)CN)[C@@H](C)CC)[C@@H](C)CC)[C@@H](C)O)NC(=O)[C@H](CCC(N)=O)NC(=O)[C@H](CC(N)=O)NC(=O)[C@@H](NC(=O)[C@@H](N)CC=1C=CC=CC=1)C(C)C)C1=CN=CN1 COCFEDIXXNGUNL-RFKWWTKHSA-N 0.000 description 2
- 241001465754 Metazoa Species 0.000 description 2
- 241000699666 Mus <mouse, genus> Species 0.000 description 2
- 206010029113 Neovascularisation Diseases 0.000 description 2
- 239000002033 PVDF binder Substances 0.000 description 2
- 238000011529 RT qPCR Methods 0.000 description 2
- 206010039163 Right ventricular failure Diseases 0.000 description 2
- 208000000924 Right ventricular hypertrophy Diseases 0.000 description 2
- 102000005157 Somatostatin Human genes 0.000 description 2
- 108010056088 Somatostatin Proteins 0.000 description 2
- OUUQCZGPVNCOIJ-UHFFFAOYSA-M Superoxide Chemical compound [O-][O] OUUQCZGPVNCOIJ-UHFFFAOYSA-M 0.000 description 2
- 108091023040 Transcription factor Proteins 0.000 description 2
- 102000040945 Transcription factor Human genes 0.000 description 2
- 241000700605 Viruses Species 0.000 description 2
- 230000004913 activation Effects 0.000 description 2
- 239000002870 angiogenesis inducing agent Substances 0.000 description 2
- 239000003472 antidiabetic agent Substances 0.000 description 2
- 230000006907 apoptotic process Effects 0.000 description 2
- 230000000903 blocking effect Effects 0.000 description 2
- 239000002775 capsule Substances 0.000 description 2
- 239000000969 carrier Substances 0.000 description 2
- 239000003153 chemical reaction reagent Substances 0.000 description 2
- RNFNDJAIBTYOQL-UHFFFAOYSA-N chloral hydrate Chemical compound OC(O)C(Cl)(Cl)Cl RNFNDJAIBTYOQL-UHFFFAOYSA-N 0.000 description 2
- 229960002327 chloral hydrate Drugs 0.000 description 2
- 208000020832 chronic kidney disease Diseases 0.000 description 2
- 239000006071 cream Substances 0.000 description 2
- 230000006378 damage Effects 0.000 description 2
- 238000013461 design Methods 0.000 description 2
- 238000003745 diagnosis Methods 0.000 description 2
- 239000002552 dosage form Substances 0.000 description 2
- 230000007783 downstream signaling Effects 0.000 description 2
- 239000003937 drug carrier Substances 0.000 description 2
- 230000000081 effect on glucose Effects 0.000 description 2
- 201000000523 end stage renal failure Diseases 0.000 description 2
- 210000001105 femoral artery Anatomy 0.000 description 2
- 239000007903 gelatin capsule Substances 0.000 description 2
- 208000019622 heart disease Diseases 0.000 description 2
- 238000001802 infusion Methods 0.000 description 2
- 230000002401 inhibitory effect Effects 0.000 description 2
- 208000014674 injury Diseases 0.000 description 2
- 238000002955 isolation Methods 0.000 description 2
- 210000003734 kidney Anatomy 0.000 description 2
- 231100000518 lethal Toxicity 0.000 description 2
- 230000001665 lethal effect Effects 0.000 description 2
- 239000006210 lotion Substances 0.000 description 2
- 210000005244 lower chamber Anatomy 0.000 description 2
- 210000004072 lung Anatomy 0.000 description 2
- 239000003550 marker Substances 0.000 description 2
- 239000011159 matrix material Substances 0.000 description 2
- 238000005259 measurement Methods 0.000 description 2
- 239000002480 mineral oil Substances 0.000 description 2
- 235000010446 mineral oil Nutrition 0.000 description 2
- 239000001788 mono and diglycerides of fatty acids Substances 0.000 description 2
- 239000007922 nasal spray Substances 0.000 description 2
- 230000030147 nuclear export Effects 0.000 description 2
- 239000002674 ointment Substances 0.000 description 2
- 230000008506 pathogenesis Effects 0.000 description 2
- 230000037361 pathway Effects 0.000 description 2
- 235000019271 petrolatum Nutrition 0.000 description 2
- 229920005862 polyol Polymers 0.000 description 2
- 150000003077 polyols Chemical class 0.000 description 2
- 229920002981 polyvinylidene fluoride Polymers 0.000 description 2
- 239000011148 porous material Substances 0.000 description 2
- 239000000843 powder Substances 0.000 description 2
- 239000003755 preservative agent Substances 0.000 description 2
- 210000002966 serum Anatomy 0.000 description 2
- 239000002356 single layer Substances 0.000 description 2
- 210000003491 skin Anatomy 0.000 description 2
- 239000002904 solvent Substances 0.000 description 2
- NHXLMOGPVYXJNR-ATOGVRKGSA-N somatostatin Chemical compound C([C@H]1C(=O)N[C@H](C(N[C@@H](CO)C(=O)N[C@@H](CSSC[C@@H](C(=O)N[C@@H](CCCCN)C(=O)N[C@@H](CC(N)=O)C(=O)N[C@@H](CC=2C=CC=CC=2)C(=O)N[C@@H](CC=2C=CC=CC=2)C(=O)N[C@@H](CC=2C3=CC=CC=C3NC=2)C(=O)N[C@@H](CCCCN)C(=O)N[C@H](C(=O)N1)[C@@H](C)O)NC(=O)CNC(=O)[C@H](C)N)C(O)=O)=O)[C@H](O)C)C1=CC=CC=C1 NHXLMOGPVYXJNR-ATOGVRKGSA-N 0.000 description 2
- 229960000553 somatostatin Drugs 0.000 description 2
- 238000010186 staining Methods 0.000 description 2
- 239000006228 supernatant Substances 0.000 description 2
- 239000000375 suspending agent Substances 0.000 description 2
- 235000020357 syrup Nutrition 0.000 description 2
- 239000006188 syrup Substances 0.000 description 2
- 230000035488 systolic blood pressure Effects 0.000 description 2
- 229940124597 therapeutic agent Drugs 0.000 description 2
- 230000000699 topical effect Effects 0.000 description 2
- 238000013518 transcription Methods 0.000 description 2
- 230000035897 transcription Effects 0.000 description 2
- 238000001890 transfection Methods 0.000 description 2
- 230000001960 triggered effect Effects 0.000 description 2
- 238000010798 ubiquitination Methods 0.000 description 2
- 230000034512 ubiquitination Effects 0.000 description 2
- 235000015112 vegetable and seed oil Nutrition 0.000 description 2
- 239000008158 vegetable oil Substances 0.000 description 2
- 108091032973 (ribonucleotides)n+m Proteins 0.000 description 1
- LEACJMVNYZDSKR-UHFFFAOYSA-N 2-octyldodecan-1-ol Chemical compound CCCCCCCCCCC(CO)CCCCCCCC LEACJMVNYZDSKR-UHFFFAOYSA-N 0.000 description 1
- 229940090248 4-hydroxybenzoic acid Drugs 0.000 description 1
- 235000007173 Abies balsamea Nutrition 0.000 description 1
- 102000007469 Actins Human genes 0.000 description 1
- 108010085238 Actins Proteins 0.000 description 1
- 102000008076 Angiogenic Proteins Human genes 0.000 description 1
- 108010074415 Angiogenic Proteins Proteins 0.000 description 1
- 206010003210 Arteriosclerosis Diseases 0.000 description 1
- 239000004857 Balsam Substances 0.000 description 1
- 201000006474 Brain Ischemia Diseases 0.000 description 1
- 229940127291 Calcium channel antagonist Drugs 0.000 description 1
- 241000283707 Capra Species 0.000 description 1
- 102100035882 Catalase Human genes 0.000 description 1
- 108010053835 Catalase Proteins 0.000 description 1
- 101800005151 Cholecystokinin-8 Proteins 0.000 description 1
- 102400000888 Cholecystokinin-8 Human genes 0.000 description 1
- 229920002261 Corn starch Polymers 0.000 description 1
- 108060002063 Cyclotide Proteins 0.000 description 1
- 108020004414 DNA Proteins 0.000 description 1
- 241000252212 Danio rerio Species 0.000 description 1
- 101710088194 Dehydrogenase Proteins 0.000 description 1
- 108010008532 Deoxyribonuclease I Proteins 0.000 description 1
- 102100030012 Deoxyribonuclease-1 Human genes 0.000 description 1
- 206010070901 Diabetic dyslipidaemia Diseases 0.000 description 1
- 208000008960 Diabetic foot Diseases 0.000 description 1
- 208000032928 Dyslipidaemia Diseases 0.000 description 1
- 241001198387 Escherichia coli BL21(DE3) Species 0.000 description 1
- 108700024394 Exon Proteins 0.000 description 1
- 241000282326 Felis catus Species 0.000 description 1
- 206010016654 Fibrosis Diseases 0.000 description 1
- 229940121710 HMGCoA reductase inhibitor Drugs 0.000 description 1
- 229920000209 Hexadimethrine bromide Polymers 0.000 description 1
- 101100001157 Homo sapiens AGGF1 gene Proteins 0.000 description 1
- 101000946889 Homo sapiens Monocyte differentiation antigen CD14 Proteins 0.000 description 1
- 101000604901 Homo sapiens Phenylalanine-4-hydroxylase Proteins 0.000 description 1
- 101001023770 Homo sapiens Transcription factor NF-E2 45 kDa subunit Proteins 0.000 description 1
- 208000001953 Hypotension Diseases 0.000 description 1
- 244000018716 Impatiens biflora Species 0.000 description 1
- 206010061218 Inflammation Diseases 0.000 description 1
- 102100034343 Integrase Human genes 0.000 description 1
- 238000012313 Kruskal-Wallis test Methods 0.000 description 1
- GUBGYTABKSRVRQ-QKKXKWKRSA-N Lactose Natural products OC[C@H]1O[C@@H](O[C@H]2[C@H](O)[C@@H](O)C(O)O[C@@H]2CO)[C@H](O)[C@@H](O)[C@H]1O GUBGYTABKSRVRQ-QKKXKWKRSA-N 0.000 description 1
- 108090001090 Lectins Proteins 0.000 description 1
- 102000004856 Lectins Human genes 0.000 description 1
- 208000017170 Lipid metabolism disease Diseases 0.000 description 1
- 239000012097 Lipofectamine 2000 Substances 0.000 description 1
- 108090001030 Lipoproteins Proteins 0.000 description 1
- 102000004895 Lipoproteins Human genes 0.000 description 1
- 108010092217 Long-Acting Insulin Proteins 0.000 description 1
- 102000016261 Long-Acting Insulin Human genes 0.000 description 1
- 229940100066 Long-acting insulin Drugs 0.000 description 1
- 235000019759 Maize starch Nutrition 0.000 description 1
- 241000124008 Mammalia Species 0.000 description 1
- 102100035877 Monocyte differentiation antigen CD14 Human genes 0.000 description 1
- 208000036110 Neuroinflammatory disease Diseases 0.000 description 1
- 208000008589 Obesity Diseases 0.000 description 1
- 108700026244 Open Reading Frames Proteins 0.000 description 1
- 239000012124 Opti-MEM Substances 0.000 description 1
- 241000283973 Oryctolagus cuniculus Species 0.000 description 1
- 101710160107 Outer membrane protein A Proteins 0.000 description 1
- 208000005764 Peripheral Arterial Disease Diseases 0.000 description 1
- 208000030831 Peripheral arterial occlusive disease Diseases 0.000 description 1
- 208000018262 Peripheral vascular disease Diseases 0.000 description 1
- 102000003992 Peroxidases Human genes 0.000 description 1
- 239000004264 Petrolatum Substances 0.000 description 1
- 229920003171 Poly (ethylene oxide) Polymers 0.000 description 1
- 239000002202 Polyethylene glycol Substances 0.000 description 1
- 229920001214 Polysorbate 60 Polymers 0.000 description 1
- 229940079156 Proteasome inhibitor Drugs 0.000 description 1
- 108091008611 Protein Kinase B Proteins 0.000 description 1
- 102000005765 Proto-Oncogene Proteins c-akt Human genes 0.000 description 1
- 102000016971 Proto-Oncogene Proteins c-kit Human genes 0.000 description 1
- 108010014608 Proto-Oncogene Proteins c-kit Proteins 0.000 description 1
- 208000010378 Pulmonary Embolism Diseases 0.000 description 1
- 239000013614 RNA sample Substances 0.000 description 1
- 108010092799 RNA-directed DNA polymerase Proteins 0.000 description 1
- 101001109714 Rhizobium meliloti (strain 1021) NAD(P)H dehydrogenase (quinone) 1 Proteins 0.000 description 1
- FAPWRFPIFSIZLT-UHFFFAOYSA-M Sodium chloride Chemical compound [Na+].[Cl-] FAPWRFPIFSIZLT-UHFFFAOYSA-M 0.000 description 1
- HVUMOYIDDBPOLL-XWVZOOPGSA-N Sorbitan monostearate Chemical compound CCCCCCCCCCCCCCCCCC(=O)OC[C@@H](O)[C@H]1OC[C@H](O)[C@H]1O HVUMOYIDDBPOLL-XWVZOOPGSA-N 0.000 description 1
- 235000021355 Stearic acid Nutrition 0.000 description 1
- 238000000692 Student's t-test Methods 0.000 description 1
- 239000006180 TBST buffer Substances 0.000 description 1
- 102100035412 Transcription factor NF-E2 45 kDa subunit Human genes 0.000 description 1
- 240000003864 Ulex europaeus Species 0.000 description 1
- 235000010730 Ulex europaeus Nutrition 0.000 description 1
- 208000024248 Vascular System injury Diseases 0.000 description 1
- 208000012339 Vascular injury Diseases 0.000 description 1
- 230000005856 abnormality Effects 0.000 description 1
- 238000002835 absorbance Methods 0.000 description 1
- 238000010521 absorption reaction Methods 0.000 description 1
- 230000009471 action Effects 0.000 description 1
- 230000003213 activating effect Effects 0.000 description 1
- 239000002671 adjuvant Substances 0.000 description 1
- 230000002411 adverse Effects 0.000 description 1
- 238000002266 amputation Methods 0.000 description 1
- 229940127003 anti-diabetic drug Drugs 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
- 238000013459 approach Methods 0.000 description 1
- 208000011775 arteriosclerosis disease Diseases 0.000 description 1
- QVGXLLKOCUKJST-UHFFFAOYSA-N atomic oxygen Chemical compound [O] QVGXLLKOCUKJST-UHFFFAOYSA-N 0.000 description 1
- 210000002469 basement membrane Anatomy 0.000 description 1
- 235000019445 benzyl alcohol Nutrition 0.000 description 1
- 239000011230 binding agent Substances 0.000 description 1
- 230000004071 biological effect Effects 0.000 description 1
- 239000002981 blocking agent Substances 0.000 description 1
- 230000036772 blood pressure Effects 0.000 description 1
- 229960001467 bortezomib Drugs 0.000 description 1
- GXJABQQUPOEUTA-RDJZCZTQSA-N bortezomib Chemical compound C([C@@H](C(=O)N[C@@H](CC(C)C)B(O)O)NC(=O)C=1N=CC=NC=1)C1=CC=CC=C1 GXJABQQUPOEUTA-RDJZCZTQSA-N 0.000 description 1
- 239000000480 calcium channel blocker Substances 0.000 description 1
- 230000021164 cell adhesion Effects 0.000 description 1
- 230000010261 cell growth Effects 0.000 description 1
- 230000009087 cell motility Effects 0.000 description 1
- 238000001516 cell proliferation assay Methods 0.000 description 1
- 239000006285 cell suspension Substances 0.000 description 1
- 230000002032 cellular defenses Effects 0.000 description 1
- 230000001413 cellular effect Effects 0.000 description 1
- 230000008614 cellular interaction Effects 0.000 description 1
- 230000007248 cellular mechanism Effects 0.000 description 1
- 230000005754 cellular signaling Effects 0.000 description 1
- 229940081733 cetearyl alcohol Drugs 0.000 description 1
- 238000006243 chemical reaction Methods 0.000 description 1
- 239000003593 chromogenic compound Substances 0.000 description 1
- 238000000576 coating method Methods 0.000 description 1
- 239000003086 colorant Substances 0.000 description 1
- 238000002648 combination therapy Methods 0.000 description 1
- 230000000052 comparative effect Effects 0.000 description 1
- 238000004624 confocal microscopy Methods 0.000 description 1
- 230000001268 conjugating effect Effects 0.000 description 1
- 230000008602 contraction Effects 0.000 description 1
- 230000001120 cytoprotective effect Effects 0.000 description 1
- 230000002354 daily effect Effects 0.000 description 1
- 230000007547 defect Effects 0.000 description 1
- 230000007812 deficiency Effects 0.000 description 1
- 230000001934 delay Effects 0.000 description 1
- 230000003111 delayed effect Effects 0.000 description 1
- 230000003831 deregulation Effects 0.000 description 1
- 239000007933 dermal patch Substances 0.000 description 1
- 201000009101 diabetic angiopathy Diseases 0.000 description 1
- 230000003205 diastolic effect Effects 0.000 description 1
- 230000004069 differentiation Effects 0.000 description 1
- 238000010790 dilution Methods 0.000 description 1
- 239000012895 dilution Substances 0.000 description 1
- 230000003828 downregulation Effects 0.000 description 1
- 239000000890 drug combination Substances 0.000 description 1
- 238000010410 dusting Methods 0.000 description 1
- 238000013399 early diagnosis Methods 0.000 description 1
- 230000013020 embryo development Effects 0.000 description 1
- 239000003995 emulsifying agent Substances 0.000 description 1
- 239000008387 emulsifying waxe Substances 0.000 description 1
- 239000000839 emulsion Substances 0.000 description 1
- 208000028208 end stage renal disease Diseases 0.000 description 1
- 230000012202 endocytosis Effects 0.000 description 1
- 230000002708 enhancing effect Effects 0.000 description 1
- 239000006167 equilibration buffer Substances 0.000 description 1
- 150000002148 esters Chemical class 0.000 description 1
- BEFDCLMNVWHSGT-UHFFFAOYSA-N ethenylcyclopentane Chemical compound C=CC1CCCC1 BEFDCLMNVWHSGT-UHFFFAOYSA-N 0.000 description 1
- 230000003203 everyday effect Effects 0.000 description 1
- 230000029142 excretion Effects 0.000 description 1
- 238000013401 experimental design Methods 0.000 description 1
- 239000000284 extract Substances 0.000 description 1
- 239000003925 fat Substances 0.000 description 1
- 230000004761 fibrosis Effects 0.000 description 1
- 239000000796 flavoring agent Substances 0.000 description 1
- 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 1
- MHMNJMPURVTYEJ-UHFFFAOYSA-N fluorescein-5-isothiocyanate Chemical compound O1C(=O)C2=CC(N=C=S)=CC=C2C21C1=CC=C(O)C=C1OC1=CC(O)=CC=C21 MHMNJMPURVTYEJ-UHFFFAOYSA-N 0.000 description 1
- 238000000799 fluorescence microscopy Methods 0.000 description 1
- 239000006260 foam Substances 0.000 description 1
- 235000013305 food Nutrition 0.000 description 1
- 235000013355 food flavoring agent Nutrition 0.000 description 1
- 230000037406 food intake Effects 0.000 description 1
- 235000012631 food intake Nutrition 0.000 description 1
- 235000003599 food sweetener Nutrition 0.000 description 1
- 239000003205 fragrance Substances 0.000 description 1
- 230000005714 functional activity Effects 0.000 description 1
- 230000002496 gastric effect Effects 0.000 description 1
- 210000001035 gastrointestinal tract Anatomy 0.000 description 1
- 238000001415 gene therapy Methods 0.000 description 1
- 230000002068 genetic effect Effects 0.000 description 1
- 230000002641 glycemic effect Effects 0.000 description 1
- 229940074045 glyceryl distearate Drugs 0.000 description 1
- 229940075507 glyceryl monostearate Drugs 0.000 description 1
- 239000008187 granular material Substances 0.000 description 1
- 150000003278 haem Chemical class 0.000 description 1
- 239000000017 hydrogel Substances 0.000 description 1
- 239000002471 hydroxymethylglutaryl coenzyme A reductase inhibitor Substances 0.000 description 1
- 229940126904 hypoglycaemic agent Drugs 0.000 description 1
- 230000036543 hypotension Effects 0.000 description 1
- 230000001146 hypoxic effect Effects 0.000 description 1
- 238000010166 immunofluorescence Methods 0.000 description 1
- 238000003125 immunofluorescent labeling Methods 0.000 description 1
- 238000002991 immunohistochemical analysis Methods 0.000 description 1
- 238000000338 in vitro Methods 0.000 description 1
- 230000001939 inductive effect Effects 0.000 description 1
- 230000004054 inflammatory process Effects 0.000 description 1
- 239000004026 insulin derivative Substances 0.000 description 1
- 229960002869 insulin glargine Drugs 0.000 description 1
- 230000003914 insulin secretion Effects 0.000 description 1
- 238000000185 intracerebroventricular administration Methods 0.000 description 1
- 238000007917 intracranial administration Methods 0.000 description 1
- 238000007918 intramuscular administration Methods 0.000 description 1
- 238000007912 intraperitoneal administration Methods 0.000 description 1
- 238000001990 intravenous administration Methods 0.000 description 1
- 230000002147 killing effect Effects 0.000 description 1
- 238000011813 knockout mouse model Methods 0.000 description 1
- 239000008101 lactose Substances 0.000 description 1
- 229940060975 lantus Drugs 0.000 description 1
- 239000002523 lectin Substances 0.000 description 1
- 210000002414 leg Anatomy 0.000 description 1
- 238000012417 linear regression Methods 0.000 description 1
- 150000002632 lipids Chemical class 0.000 description 1
- 229940057995 liquid paraffin Drugs 0.000 description 1
- 244000144972 livestock Species 0.000 description 1
- 230000004807 localization Effects 0.000 description 1
- 230000033001 locomotion Effects 0.000 description 1
- 230000005923 long-lasting effect Effects 0.000 description 1
- 230000007774 longterm Effects 0.000 description 1
- 239000000314 lubricant Substances 0.000 description 1
- 239000006166 lysate Substances 0.000 description 1
- 239000012139 lysis buffer Substances 0.000 description 1
- 238000013227 male C57BL/6J mice Methods 0.000 description 1
- 239000000463 material Substances 0.000 description 1
- 208000030159 metabolic disease Diseases 0.000 description 1
- 230000002503 metabolic effect Effects 0.000 description 1
- 239000012120 mounting media Substances 0.000 description 1
- 210000002200 mouth mucosa Anatomy 0.000 description 1
- 208000031225 myocardial ischemia Diseases 0.000 description 1
- 229940097496 nasal spray Drugs 0.000 description 1
- 230000003959 neuroinflammation Effects 0.000 description 1
- 230000007935 neutral effect Effects 0.000 description 1
- 230000009871 nonspecific binding Effects 0.000 description 1
- 230000005937 nuclear translocation Effects 0.000 description 1
- 235000020824 obesity Nutrition 0.000 description 1
- GLDOVTGHNKAZLK-UHFFFAOYSA-N octadecan-1-ol Chemical compound CCCCCCCCCCCCCCCCCCO GLDOVTGHNKAZLK-UHFFFAOYSA-N 0.000 description 1
- QIQXTHQIDYTFRH-UHFFFAOYSA-N octadecanoic acid Chemical compound CCCCCCCCCCCCCCCCCC(O)=O QIQXTHQIDYTFRH-UHFFFAOYSA-N 0.000 description 1
- OQCDKBAXFALNLD-UHFFFAOYSA-N octadecanoic acid Natural products CCCCCCCC(C)CCCCCCCCC(O)=O OQCDKBAXFALNLD-UHFFFAOYSA-N 0.000 description 1
- 230000009437 off-target effect Effects 0.000 description 1
- 239000003921 oil Substances 0.000 description 1
- 239000012053 oil suspension Substances 0.000 description 1
- 235000019198 oils Nutrition 0.000 description 1
- 238000001543 one-way ANOVA Methods 0.000 description 1
- 210000000056 organ Anatomy 0.000 description 1
- 230000002018 overexpression Effects 0.000 description 1
- 229910052760 oxygen Inorganic materials 0.000 description 1
- 239000001301 oxygen Substances 0.000 description 1
- 238000004806 packaging method and process Methods 0.000 description 1
- 239000012188 paraffin wax Substances 0.000 description 1
- 238000007911 parenteral administration Methods 0.000 description 1
- 230000036961 partial effect Effects 0.000 description 1
- 230000001575 pathological effect Effects 0.000 description 1
- 230000007170 pathology Effects 0.000 description 1
- 230000007310 pathophysiology Effects 0.000 description 1
- 210000003899 penis Anatomy 0.000 description 1
- 239000000137 peptide hydrolase inhibitor Substances 0.000 description 1
- 108040007629 peroxidase activity proteins Proteins 0.000 description 1
- 229940066842 petrolatum Drugs 0.000 description 1
- 239000008024 pharmaceutical diluent Substances 0.000 description 1
- 239000008363 phosphate buffer Substances 0.000 description 1
- 230000004962 physiological condition Effects 0.000 description 1
- 229920001223 polyethylene glycol Polymers 0.000 description 1
- 239000001818 polyoxyethylene sorbitan monostearate Substances 0.000 description 1
- 235000010989 polyoxyethylene sorbitan monostearate Nutrition 0.000 description 1
- 229920001451 polypropylene glycol Polymers 0.000 description 1
- 229940113124 polysorbate 60 Drugs 0.000 description 1
- 230000029279 positive regulation of transcription, DNA-dependent Effects 0.000 description 1
- 230000003389 potentiating effect Effects 0.000 description 1
- 230000002028 premature Effects 0.000 description 1
- 238000002360 preparation method Methods 0.000 description 1
- 239000000651 prodrug Substances 0.000 description 1
- 229940002612 prodrug Drugs 0.000 description 1
- 208000037821 progressive disease Diseases 0.000 description 1
- 230000002062 proliferating effect Effects 0.000 description 1
- 235000013772 propylene glycol Nutrition 0.000 description 1
- 239000003207 proteasome inhibitor Substances 0.000 description 1
- 230000001681 protective effect Effects 0.000 description 1
- 238000000159 protein binding assay Methods 0.000 description 1
- 230000036593 pulmonary vascular resistance Effects 0.000 description 1
- 238000011002 quantification Methods 0.000 description 1
- 238000003753 real-time PCR Methods 0.000 description 1
- 230000008929 regeneration Effects 0.000 description 1
- 238000011069 regeneration method Methods 0.000 description 1
- 238000007634 remodeling Methods 0.000 description 1
- 230000036454 renin-angiotensin system Effects 0.000 description 1
- 238000010839 reverse transcription Methods 0.000 description 1
- 238000012552 review Methods 0.000 description 1
- 210000005241 right ventricle Anatomy 0.000 description 1
- 239000000523 sample Substances 0.000 description 1
- 238000000926 separation method Methods 0.000 description 1
- 230000003584 silencer Effects 0.000 description 1
- 235000020183 skimmed milk Nutrition 0.000 description 1
- WXMKPNITSTVMEF-UHFFFAOYSA-M sodium benzoate Chemical compound [Na+].[O-]C(=O)C1=CC=CC=C1 WXMKPNITSTVMEF-UHFFFAOYSA-M 0.000 description 1
- 235000010234 sodium benzoate Nutrition 0.000 description 1
- 239000004299 sodium benzoate Substances 0.000 description 1
- 239000011780 sodium chloride Substances 0.000 description 1
- 239000007787 solid Substances 0.000 description 1
- 235000010199 sorbic acid Nutrition 0.000 description 1
- 239000004334 sorbic acid Substances 0.000 description 1
- 229940075582 sorbic acid Drugs 0.000 description 1
- 239000001587 sorbitan monostearate Substances 0.000 description 1
- 235000011076 sorbitan monostearate Nutrition 0.000 description 1
- 229940035048 sorbitan monostearate Drugs 0.000 description 1
- 239000003381 stabilizer Substances 0.000 description 1
- 238000007619 statistical method Methods 0.000 description 1
- 239000008117 stearic acid Substances 0.000 description 1
- 210000002784 stomach Anatomy 0.000 description 1
- 230000006354 stress signaling Effects 0.000 description 1
- 238000007920 subcutaneous administration Methods 0.000 description 1
- 239000000126 substance Substances 0.000 description 1
- 239000000758 substrate Substances 0.000 description 1
- 235000000346 sugar Nutrition 0.000 description 1
- 150000008163 sugars Chemical class 0.000 description 1
- 230000004083 survival effect Effects 0.000 description 1
- 238000013268 sustained release Methods 0.000 description 1
- 239000012730 sustained-release form Substances 0.000 description 1
- 239000003765 sweetening agent Substances 0.000 description 1
- 230000002194 synthesizing effect Effects 0.000 description 1
- 230000009897 systematic effect Effects 0.000 description 1
- 230000009885 systemic effect Effects 0.000 description 1
- 230000002123 temporal effect Effects 0.000 description 1
- MHXBHWLGRWOABW-UHFFFAOYSA-N tetradecyl octadecanoate Chemical compound CCCCCCCCCCCCCCCCCC(=O)OCCCCCCCCCCCCCC MHXBHWLGRWOABW-UHFFFAOYSA-N 0.000 description 1
- 230000008719 thickening Effects 0.000 description 1
- 239000003053 toxin Substances 0.000 description 1
- 231100000765 toxin Toxicity 0.000 description 1
- 108700012359 toxins Proteins 0.000 description 1
- 239000012096 transfection reagent Substances 0.000 description 1
- 230000005945 translocation Effects 0.000 description 1
- 238000011277 treatment modality Methods 0.000 description 1
- 238000002604 ultrasonography Methods 0.000 description 1
- 210000003606 umbilical vein Anatomy 0.000 description 1
- 241000701161 unidentified adenovirus Species 0.000 description 1
- 238000011144 upstream manufacturing Methods 0.000 description 1
- 210000003556 vascular endothelial cell Anatomy 0.000 description 1
- 239000013598 vector Substances 0.000 description 1
- 230000003612 virological effect Effects 0.000 description 1
- 238000005406 washing Methods 0.000 description 1
- 239000001993 wax Substances 0.000 description 1
- 239000000080 wetting agent Substances 0.000 description 1
- 239000003871 white petrolatum Substances 0.000 description 1
Classifications
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K14/00—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- C07K14/435—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
- C07K14/475—Growth factors; Growth regulators
- C07K14/515—Angiogenesic factors; Angiogenin
-
- 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
- A61K38/18—Growth factors; Growth regulators
- A61K38/1891—Angiogenesic factors; Angiogenin
-
- 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
Definitions
- compositions, systems, kits, and methods for treating a subject with a disease or condition characterized by abnormal glucose metabolism (e.g., diabetes) or ischemic vascular disease (e.g., PAD or CAD) by administering a composition comprising AGGF1 peptides, or an expression vector encoding said peptides, or AGGF1 -primed endothelial progenitor cells, to a subject.
- a composition comprising AGGF1 peptides, or an expression vector encoding said peptides, or AGGF1 -primed endothelial progenitor cells, to a subject.
- the AGGF1 peptide is full- length AGGF1, or a biologically active fragment or mutant thereof.
- the AGGF1 peptides or expression vectors are administered around the same time insulin is administered to the subject.
- Diabetes mellitus is a common and heterogeneous disease with high morbidity and mortality worldwide. An estimated 422 million adults are affected with diabetes based on the WHO’s most recent 2016 data.
- the prevalence of diabetes, and in particular, of type 2 diabetes (T2DM) increases rapidly each year due to lifestyle habits and increased rates of aging, inactivity, and obesity, and poses an important challenge to human health.
- T2DM type 2 diabetes
- the treatment of diabetes remains suboptimal in a significant proportion of patients.
- >50% of T2DM patients have poor glycemic control and 18% develop severe complications.
- Population HbAlc concentrations failed to decline as much as expected due to poor medication compliance and other causes. To address this grand challenge, much is needed to understand the pathophysiology of diabetes and to develop new innovative therapies. In particular, long-lasting treatment can dramatically reduce poor medication compliance.
- compositions, systems, kits, and methods for treating a subject with a disease or condition characterized by abnormal glucose metabolism (e.g., diabetes) or ischemic vascular disease (e.g., PAD or CAD) by administering a composition comprising AGGF1 peptides, or an expression vector encoding said peptides, or AGGF1 -primed endothelial progenitor cells, to a subject.
- a composition comprising AGGF1 peptides, or an expression vector encoding said peptides, or AGGF1 -primed endothelial progenitor cells, to a subject.
- the AGGF1 peptide is full- length AGGF1, or a biologically active fragment or mutant thereof.
- the AGGF1 peptides or expression vectors are administered around the same time insulin is administered to the subject.
- provided herein are methods of treating a subject with a disease or condition characterized by abnormal glucose metabolism comprising:
- the methods further comprise administering insulin to the subject.
- the insulin is administered at about the same time as the composition.
- kits for treating a subject comprising: administering a composition to a subject, wherein the subject has at least one of the following: a disease or condition characterized by abnormal glucose metabolism, ischemic vascular disease, pulmonary hypertension, cardiac hypertrophy, and heart failure: wherein said composition comprises: i) an AGGF1 peptide, and/or ii) an expression vector encoding said AGGF1 peptide.
- a cardiovascular disease or condition characterized comprising: administering a composition comprising: i) an AGGF1 peptide that is no longer than 600 amino acids (e.g., 100-600 or 100-300 or 100-150 amino acids in length), or ii) an expression vector encoding the AGGF1 peptide, to the subject with the disease or condition.
- the disease or condition is selected from the group consisting of: coronary artery disease, myocardial infarction, cardiac hypertrophy, heart failure, peripheral artery disease, restenosis and in-stent thrombosis after angioplasty and stenting, and pulmonary arterial hypertension.
- pulmonary arterial hypertension comprising: administering a composition comprising: i) an AGGF1 peptide, or ii) an expression vector encoding the AGGF1 peptide, to the subject with the PAH.
- the subject in any of the methods herein is a human and: A) the composition comprises said AGGF1 peptide, and wherein the dosage is selected from the group consisting of: 0.1-15 mg/kg (e.g., 0.1 mg/kg ... 4.0 mg/kg ... 8.0 mg/k ... 12.0 mg/kg ... and 15 mg/kg), 0.5-10 mg/kg, and 1-7 mg/kg; or B) said composition comprises said expression vector encoding said AGGF1 peptide, and wherein said dosage is about 1.0 x 10 11 - 5.0 x 10 13 (e.g., 1.0 x 10 11 ... 1.0 x 10 12 ... or 5.0 x 10 13 ).
- kits comprising: a) a blood glucose monitoring device or article of manufacture; and b) a composition comprising: i) an AGGF1 peptide, or ii) an expression vector encoding the AGGF1 peptide.
- the glucose monitoring device or article of manufacture is selected from the group consisting of: a blood glucose test strip, a reader for reading the glucose test strip, FreeStyle Libre, GlucoTrack, Eversense, GlucoWise, NovioSense, GlucoSense, and a smart contact lens.
- kits comprising: a) a composition comprising: i) an AGGF1 peptide, or ii) an expression vector encoding the AGGF1 peptide; and b) an implantable pump containing the composition, wherein the implantable pump is configured to deliver an amount of the composition into the bloodstream of a subject.
- compositions comprising: a) an AGGF1 peptide, or an expression vector encoding the AGGF1 peptide; and b) insulin peptide, or an expression vector encoding the insulin peptide.
- kits for treating a subject comprising: administering a composition to a subject, wherein the subject has a disease or condition characterized by abnormal glucose metabolism and/or ischemic vascular disease, and wherein the composition comprises an AGGFl-primed endothelial progenitor cell.
- the disease or condition is selected from the group consisting of peripheral artery disease (PAD), coronary artery disease (CAD), coronary heart disease (CHD), and myocardial infarction (MI).
- the AGGFl-primed endothelial progenitor cell comprises an endothelial progenitor cell (EPC) from the subject, wherein the EPC has been exposed to AGGF1 peptides ex vivo.
- the AGGFl- primed endothelial progenitor cell comprises an endothelial progenitor cell (EPC) from a relative of the subject, wherein the EPC has been exposed to AGGF1 peptides ex vivo.
- the AGGFl-primed endothelial progenitor cell comprises an endothelial progenitor cell (EPC) from a non-relative of the subject, wherein the EPC has been exposed to AGGF1 peptides ex vivo.
- EPC endothelial progenitor cell
- compositions comprising: an AGGFl- primed endothelial progenitor cell.
- the composition further comprises cell growth media.
- the AGGFl-primed endothelial progenitor cell comprises an endothelial progenitor cell (EPC) from a subject with a disease or condition characterized by abnormal glucose metabolism and/or ischemic vascular disease, wherein the EPC has been exposed to AGGF1 peptides ex vivo.
- the compositions further comprise a physiological tolerable liquid.
- kits and systems comprising: a) an endothelial progenitor cell (EPC), and b) a composition comprising: i) an AGGF1 peptide, and/or ii) an expression vector encoding the AGGF1 peptide.
- EPC endothelial progenitor cell
- the AGGF1 peptide comprises full length human AGGF1 shown in SEQ ID NO: 1.
- the AGGF1 peptide has at least 97% sequence identity with SEQ ID NO: 1 (e.g., 97 ... 98 .. 99 .. 99.5% identify).
- the AGGF1 peptide is selected from any of SEQ ID Nos:2-l6 or i) a peptide comprising the amino acid sequence from 574 to 614 of SEQ ID NO:l, ii) a peptide comprising the amino acid sequence from 574 to 624 of SEQ ID NO:l, or iii) a peptide comprising the amino acid sequence from 604 to 714 of SEQ ID NO:l.
- the AGGF1 peptide has at least 97% sequence identity (e.g., 97 ... 98 .. 99 ..
- the subject is a human.
- the AGGF1 peptide consists of, consists essentially of, or comprises QRDDAPASVH (SEQ ID NO: 18).
- an expression vector encoding SEQ ID NO: 18 is employed.
- the sequence encoding SEQ ID NO: 18 is CAAAGAGAUGAUG
- CUCCUGCAUCUGUUCAU (SEQ ID NO:25).
- the expression vectors herein comprise SEQ ID NO:25.
- AGGF1 regulates glucose metabolism and insulin resistance.
- A Body weight of AGGFl +/_ KO mice and littermate WT mice fed a standard chow diet or a high fat diet (HFD).
- B Fasting glucose levels in AGGFl +/_ KO mice and littermate WT mice fed HFD.
- C Fasting insulin levels in AGGFl +/ KO mice and littermate WT mice fed HFD.
- D Glucose tolerance tests in AGGFl +/_ KO mice and littermate WT mice fed HFD.
- E Insulin tolerance tests in AGGFl +/ KO mice and littermate WT mice fed HFD. *P ⁇ 0.01,
- Fig. 3 Therapeutic effects of chronic AGGF1 protein therapy on lowering fasting glucose levels.
- 24-week-old male HFD-induced diabetic mice were treated by intraperitoneal injection of 0.25 mg/kg AGGF1 protein or PBS control every 2 days for 36 days (chronic treatment).
- A Fasting serum glucose levels.
- AGGF1 protein therapy is insulin-dependent.
- A Therapeutic effects of insulin alone (1.00 ug/kg body weight) and in combination with AGGF1 (0.25 mg/kg body weight) in 24-week-old male HFD-induced diabetic mice.
- B No therapeutic effect for AGGF1 protein injection on fasting glucose levels in 16-week-old male diabetic mice induced by streptozotocin (STZ) for 4 weeks.
- Figure 5a shows the amino acid sequence of full-length human AGGF1 (714 amino acid residues) (SEQ ID NO:l).
- Figure 5b shows the amino acid sequence of AGGF1-N1 (696 amino acids; 19 amino acid deletion from the N-terminus of AGGF1) (SEQ ID NO:2).
- Figure 5c shows the amino acid sequence of AGGF1-N2 (629 amino acids; 86 amino acid deletion from the N-terminus of AGGF1) (SEQ ID NOG).
- Figure 6a shows the amino acid sequence of AGGF1-N3 (550 amino acids; 165 amino acid deletion from the N-terminus of AGGF1) (SEQ ID NO:4).
- Figure 6b shows the amino acid sequence of AGGF1-N4 (496 amino acids; 219 amino acid deletion from the N- terminus of AGGF1) (SEQ ID NOG).
- Figure 6c shows the amino acid sequence of AGGF1- N5 (431 amino acids; 284 amino acid deletion from the N-terminus of AGGF1) (SEQ ID NO:6).
- Figure 6d shows the amino acid sequence of AGGF1-N6 (366 amino acids; 349 amino acid deletion from the N-terminus of AGGF1) (SEQ ID NO:7).
- Figure 7a shows the amino acid sequence of AGGF1-N7 (302 amino acids; 413 amino acid deletion from the N-terminus of AGGF1) (SEQ ID NOG).
- Figure 7b shows the amino acid sequence of AGGF1-N8 (244 amino acids; 472 amino acid deletion from the N- terminus of AGGF1) (SEQ ID NO:9).
- Figure 7c shows the amino acid sequence of AGGF1- N9 (193 amino acids; 522 amino acid deletion from the N-terminus of AGGF1) (SEQ ID NO: 10).
- Figure 7d show the amino acid sequence of AGGF1-N10 (141 amino acids; 574 amino acid deletion from the N-terminus of AGGF1) (SEQ ID NO: 11).
- Figure 7e shows the amino acid sequence of AGGF1-N101 (131 amino acids; 584 amino acid deletion from the N-terminus of AGGF1) (SEQ ID NO: 12).
- Figure 7f shows the amino acid sequence of AGGF1-N102 (121 amino acids; 594 amino acid deletion from the N-terminus of AGGF1) (SEQ ID NO: 13).
- Figure 8a shows the amino acid sequence of AGGF1-N103 (110 amino acids; 604 amino acid deletion from the N-terminus of AGGF1) (SEQ ID NO: 14).
- Figure 8b shows the amino acid sequence of AGGF1-C1 (664 amino acids; 50 amino acid deletion from the C- terminus of AGGF1) (SEQ ID NO: 15).
- Figure 8c shows the amino acid sequence of AGGF1-C2 (614 amino acids; 100 amino acid deletion from the C-terminus of AGGF1) (SEQ ID NO: 16).
- Figure 9 shows the full length human AGGF1 cDNA (SEQ ID NO: 17), with arrows showing truncation mutants.
- FIG 10 shows a diagram showing the full length AGGF1 (WT) and its serial N- terminal and C-terminal deletion mutant proteins.
- Figure 11 shows results of purification of mutant AGGF1 proteins with serial N- and C-terminal deletions.
- Figure 12 shows results of AGGF1 -endothelial cell adhesion (binding) assays for serial N- and C-terminal AGGF1 deletion mutants.
- Figure 13 shows results of Endothelial cell migration assays for serial N- and C- terminal AGGF1 deletion mutants.
- Figure 14 shows results of angiogenesis assays for serial N- and C-terminal AGGF1 deletion mutants (capillary endothelial tube formation assays).
- FIG. 15 Micro-deletion analysis identified the short mutant form of AGGF1 with full functions of the full-length AGGF1 protein.
- A A diagram showing the full length AGGF1 (WT) and serial N-terminal deletion mutant proteins.
- B Endothelial cell adhesion assay with AGGF1 and its N-terminal deletion mutants. Coating buffer (CB) with and without BSA was used as a negative control. *P ⁇ 0.05.
- C Endothelial cell migration assay with AGGF1 and its N-terminal deletion mutants. BSA was used as a negative control. *P ⁇ 0.05.
- D Endothelial tube formation assay AGGF1 and its N-terminal deletion mutants. BSA was used as a negative control (left panel).
- FIG. 16 Aggfl +/ KO mice spontaneously developed PAH.
- Male Agg[] 0,‘o/+ KO mice (16-20 weeks) showed much higher mPAP (mean pulmonary arterial pressure) (A, B), total pulmonary resistance (TPR) (C), and right ventricular hypertrophy compared to WT mice (D).
- mPAP mean pulmonary arterial pressure
- TPR total pulmonary resistance
- D right ventricular hypertrophy compared to WT mice
- FIG. 17 Aggfl +/ KO causes vascular remodeling.
- A Representative H&E of pulmonary arteries.
- B Mean medial wall thickness.
- C Resistance pulmonary arterial muscularization.
- lung sections (5 pm) were co-immunostained with an a-smooth muscle actin antibody (a-SMA, VSMC marker) and a CD31 antibody (EC maker). Vessels were classified as fully (100%), partially, or non-muscularized (0%) vessels based on the percentage of CD31 signal surrounded by a-SMA signal.
- Aggfl is required for essential functions of EPCs in capillary tube formation, proliferation, transendothelial migration, and migration as in a diabetic mouse model ( db/db mice).
- EPCs were isolated from bone marrow of WT, heterozygous Aggfl +/ KO mice, db/db mice, and Aggfl +/ db/db mice and characterized.
- Aggfl is required for essential functions of EPCs in capillary tube formation, proliferation, transendothelial migration, and migration as in a diabetic mouse model ( db/db mice).
- EPCs were isolated from bone marrow of WT, heterozygous Aggfl +/ KO mice, db/db mice, and Aggfl +/ db/db mice and characterized.
- haploinsufficiency inhibits angiogensis mediated by EPCs.
- B Images from (A) were analyzed, quantified and plotted.
- C Aggfl haploinsufficiency inhibits EPC proliferation.
- FIG. 20 AGGF1 dramatically improves essential functions of EPCs impaired by HG.
- A Western blot analysis showing increased AGGF1 expression in EPCs.
- B AGGF1 treatment reversed the impairment of angiogenic function of EPCs by HG.
- C Images from (B) were analyzed, quantified and plotted.
- D AGGF1 treatment reversed the reduced cell proliferation of EPCs by HG.
- E AGGF1 treatment reversed the HG-impaired
- FIG. 21 AGGF1 protein treatment robustly potentiates the therapeutic effects of EPCs on peripheral vascular complications in a hindlimb ischemia model in db/db mice.
- a hindlimb ischemia model was created in db/db mice.
- C Therapeutic effects of AGGF1 -pretreated EPCs on ambulatory impairment compared with EPCs without AGGF1 pretreatment.
- D Effects of AGGF1 -pretreated EPCs on the density of CD3l-positive capillary vessels compared with EPCs without AGGF1 pretreatment.
- FIG. 22 AGGF1 regulates the nuclear accumulation of Nrf2.
- A Western blot analysis for the effect of AGGF1, HG and HG+AGGF1 on the expression levels of nuclear Nrf2 (n-Nrf2) and its downstream signaling molecules, including HQ-l, NQO-l, and CAT. GAPDH was used as loading control.
- FIG. 23 AGGF1 activates AKT-Fyn-Nrf2 signaling in EPCs.
- A-B Western blot analysis showing the effect of siRNA for A KT (siAKT) (A) and wortmannin (B) on AGGF1- activated phosphorylation of AKT and nuclear accumulation of Fyn (n-Fyn).
- FIG. 24 Knockdown of AKT expression, wortmannin treatment, and knockdown of Nrf2 expression attenuate the protective effects of AGGF1 on EPCs.
- A Western blot analysis for AGGF1 in EPCs.
- B-E Effects of AKT siRNA on AGGF1 -mediated rescue of HG-impaired cell proliferation (B), tube formation (C), transendothelial migration (D), and cell migration (E) by EPCs.
- G-J Effects of wortmannin on AGGF1 -mediated rescue of HG-impaired cell proliferation (G), tube formation (H), transendothelial migration (I), and cell migration (J) by EPCs.
- FIG. 25 Isolation and characterization of bone marrow derived EPCs from mice.
- Bone marrow mononuclear cells (MNCs) were washed out from the femurs and tibias of mice and isolated by density gradient centrifugation with histopaque-l083.
- MNCs were plated on gelatin-coated culture dishes and maintained in endothelial growth factor- supplemented media (EGM-2 bullet kit). Ten days after maintenance in endothelial-specific media, non-adherent MNCs were removed and the remaining adherent cells were subjected to immunostaining to verify their EPC identity.
- EMM-2 bullet kit Endothelial growth factor- supplemented media
- FIG. 26 Aggfl haploinsufficiency significantly reduces the number of bone marrow mononuclear cells (MNCs).
- MNCs were washed out from the femurs and tibias of WT and Aggfl +/ KO mice and purified by density gradient centrifugation with histopaque-l083.
- MNCs were plated into gelatin-coated culture dishes and maintained in endothelial growth factor-supplemented media (EGM-2 bullet kit for ten days). Non-adherent MNCs were removed and the remaining adherent cells were subjected to immunostaining to verify their EPC identity.
- FIG. 27 The expression level of the AGGF1 protein is increased in EPCs isolated from db/db mice or mice treated with HFD.
- A Western blot analysis showing increased AGGF1 expression in isolated EPCs from db/db mice.
- FIG. 28 Increased glucose levels in mice fed with a high fat diet (HFD).
- HFD high fat diet
- C57BL/6J mice were fed with a high fat diet alone to induce T2DM.
- Mice fed with a chow diet were sued as controls.
- blood glucose levels were measured using a glucometer, and then a hind-limb ischemia model was established with the mice.
- FIG. 30 Aggfl is required for essential functions of EPCs in capillary tube formation, proliferation, transendothelial migration, and migration as in a HFD-induced diabetic mouse model.
- EPCs were isolated from bone marrow of wild-type (WT) and heterozygous Aggfl +/ knockout (KO) mice fed with and without a high glucose diet (HFD), and characterized.
- WT wild-type
- KO heterozygous Aggfl +/ knockout mice fed with and without a high glucose diet
- HFD high glucose diet
- Aggfl haploinsufficiency inhibits angiogensis mediated by EPCs. Angiogenic function of EPCs was assessed by a matrigel-based capillary tube formation.
- B Images from (A) were analyzed, quantified and plotted.
- C Aggfl haploinsufficiency inhibits EPC proliferation.
- EPCs Proliferation of EPCs was examined using the CCK-8 kit.
- D Aggfl haploinsufficiency inhibits transendothelial migration of EPCs in transwell assays.
- HUVECs about lxlO 4 cells per well
- EPCs were harvested, resuspended in basal culture medium (EBM-2, 0.5% BSA), and added to the upper chamber of the transwell plate (0.2 ml). Images were captured 24 hours after EPCs plating with an inverted Nikon Eclipse Ti microscope.
- E Images from (D) were quantified and plotted.
- F Aggfl haploinsufficiency inhibits EPC migration in wound scratch migration assays.
- EPCs monolayers in 6-well cell culture plates were wounded with a scratch by a 200 pl pipette tip and examined 24 hours after wounding. Images were captured with an inverted Nikon Eclipse Ti microscope.
- FIG 32 AGGF1 protein treatment robustly potentiates the therapeutic effects of EPCs on angiogenesis and homing in a hindlimb ischemia model in db/db mice.
- FIG. 33 AGGF1 protein treatment robustly potentiates the therapeutic effects of EPCs on peripheral vascular complications in a hindlimb ischemia model.
- a hindlimb ischemia model was created in mice. Two days after the surgery, the mice were injected with EPCs, AGGFl-pretrearted EPCs ( ⁇ lxl0 6 ), and control PBS via the tail vein. The therapeutic effects were evaluated using a Vevo 2100 High- Resolution Micro-Ultrasound System before the ischemic surgery and 7, 14 and 28 days after ischemia.
- A Transplantation of AGGF1- pretreated EPCs dramatically improved blood perfusion compared with EPCs without AGGF1 pretreatment.
- Scale bar l00 pm.
- DCFH-DA Dichlorodihydrofluorescein diacetate
- FIG. 36 AGGF1 increases the level of nuclear Nrf2 in EPCs.
- FIG. 37 AGGF1 does not affect the expression levels of Nrf2 and KEAP-l in EPCs.
- EPCs were cultured in media with or without high glucose (HG, 30 mM) as well as a combination of high glucose and AGGF1 protein.
- A Total RNA was extracted with Trizol reagent and used for real-time RT-PCR analysis for the level of Nrf2 mRNA.
- B Western blot analysis with an anti-Nrf2 antibody.
- FIG 38 Western blot analysis for nuclear accumulation of nNrf2 in EPCs treated with Aggfl siRNA and Fyn siRNA under the normal condition (Control) and under a high glucose condition (HG). GAPDH was used as loading control.
- FIG 39 A schematic diagram showing the hypothetical molecular signaling pathway by which AGGF1 protein therapy potentiates the therapeutic effects of EPCs on diabetes-induced vascular complications. It is noted that the present invention is not limited to this particular mechanism, and an understanding of the mechanism is not necessary to practice the inventin.
- AGGF1 activates AKT signaling, which inhibits Fyn-mediated export and degradation of nuclear Nrf2. Nrf2 binds to the promoter and regulatory region and activate the transcription of CAT, H(2-l, and NQO-1, which function as antioxidants to block generation of ROS, reducing oxidative stress and increasing function of EPCs. Overall, AGGF1 functions as an antioxidant regulator to protect the function of EPCs, and becomes a potential therapeutic target for improving the ischemia-reparative capacity of EPCs transplantation in DM.
- Figure 40 shows the duration of the glucose-lowering effect of injection of a single dose of AGGF1 protein is more than 10 times longer than insulin in db/db diabetic mice.
- A Experimental design of the 48-hour AGGF1 treatment assay in Example 4. The time points for AGGF1 injection, glucose measurement, fasting period, and eating period are shown.
- the term“or” is an inclusive“or” operator and is equivalent to the term“and/or” unless the context clearly dictates otherwise.
- the meaning of“a”,“an”, and“the” include plural references.
- the meaning of“in” includes“in” and“on.”
- the terms“subject” and“patient” refer to any animal, such as a mammal like a dog, cat, bird, livestock, and preferably a human (e.g., a human with a disease such as diabetes, cardiovascular disease, or related conditions).
- the term“administration” refers to the act of giving a drug, prodrug, or other agent, or therapeutic treatment to a subject.
- exemplary routes of administration to the human body can be through the mouth (oral), skin (transdermal, topical), nose (nasal), lungs (inhalant), oral mucosa (buccal), inhaled, an implanted pump activation, by injection (e.g., intravenously, subcutaneously, intratumorally, intraocular, intraperitoneally, etc.), and the like.
- compositions, systems, kits, and methods for treating a subject with a disease or condition characterized by abnormal glucose metabolism (e.g., diabetes) or ischemic vascular disease (e.g., PAD or CAD) by administering a composition comprising AGGF1 peptides, or an expression vector encoding said peptides, or AGGF1 -primed endothelial progenitor cells, to a subject.
- a composition comprising AGGF1 peptides, or an expression vector encoding said peptides, or AGGF1 -primed endothelial progenitor cells, to a subject.
- the AGGF1 peptide is full- length AGGF1, or a biologically active fragment or mutant thereof.
- the AGGF1 peptides or expression vectors are administered around the same time insulin is administered to the subject.
- AGGF1 protein and its derivatives can effectively lower glucose levels.
- therapies with AGGF1 and AGGF1 derivatives have a longer therapeutic effect, and do not cause hypoglycemia.
- work conducted during development of embodiments herein demonstrated AGGF1 treatment has a much longer therapeutic effect with a peak effect at 18-24 hours and lasting for >42 hours.
- AGGF1 peptides or expression vector encoding AGGF1
- insulin are administered at about the same time (e.g., within 1-25 minutes of each other).
- Combined treatment with both insulin and AGGF1 can quickly decrease the fasting glucose level within 30 minutes, but also significantly extend the effect of insulin. Therefore, the therapy with combined insulin and AGGF1 can have a sustained lowering effect on glucose levels for a much longer time than insulin alone.
- patients typically require insulin injections 3 times per day. Even the long-acting insulin such as insulin glargine (Lantus, Sanofi) requires injection once a day.
- the combined AGGF1 and insulin treatment reduces the frequency of injection to once per >2 days.
- the various embodiments described herein can enable more patients with type 1 and type 2 diabetes mellitus to reach better glucose targets by reducing the frequency of injections of drugs and enhancing medication compliance, and maintain a better quality of life.
- AGGF1 is an angiogenic protein that has been proposed previously for use in treating cardiovascular diseases. In work conducted during the development of embodiments herein, it was shown that AGGF1 has activities similar to insulin. In AGGF1 KO mice, fasting glucose levels were significantly elevated when mice were fed a high fat diet and were insulin resistant. In diabetic mice, AGGF1 administration significantly lowered fasting glucose levels. In streptozotocin mice, the effects of AGGF1 seemed to depend on and potentiate insulins activities.
- AGGF1 therapy e.g., protein or expression vector encoding protein
- benefits of AGGF1 therapy include, but are not limited to, AGGF1 peptides regulate glucose levels more precisely than insulin, no evidence of hypoglycemia, longer therapeutic window, peak glucose lowering at 18-24 hours, lasting for >42 hrs.
- AGGF1 peptide and expression vectors encoding AGGF1 can be used to better control glucose levels over extended periods of time compared with insulin alone.
- the AGGF1 gene encodes a 714 amino acid angiogenic factor essential for embryonic development.
- KO heterozygous AGGF1 knockout mice develop diabetes with an increased fasting glucose level, impaired glucose tolerance, and insulin resistance, and that AGGF1 protein therapy dramatically reduces the fasting glucose level in high fat diet (HFD)-induced diabetic mice.
- HFD high fat diet
- AGGF1 treatment has a much longer therapeutic effect with a peak effect at 18-24 hours and lasting for >42 hours. It was found that AGGF1 protein therapy of one injection per two days maintained the low glucose level constantly.
- long term expression can allow a subject to receive a single administration without further treatment of for a longer period of time (e.g., 2 months).
- diabetic patients are treated with AGGF1 therapy who also have coronary artery disease, at risk for heart attack, or heart failure, and receive a benefit for both conditions.
- compositions according to the present disclosure comprises or consists of a pharmaceutically acceptable carrier, diluent, or excipient (including combinations thereof).
- a pharmaceutically acceptable carrier e.g., a pharmaceutically acceptable carrier, diluent, or excipient (including combinations thereof).
- Acceptable carriers or diluents for therapeutic use are well known in the pharmaceutical art and are described, for example, in Remington’s Pharmaceutical Sciences, Mack Publishing Co. (A. R. Gennaro edit. 1985).
- the choice of pharmaceutical carrier, excipient, or diluent is selected with regard to the intended route of administration and standard pharmaceutical practice.
- pharmaceutical comprise as, or in addition to, the carrier, excipient, or diluent any suitable binder(s), lubricant(s), suspending agent(s), coating agent(s), and/or solubilizing agent(s).
- This pharmaceutical composition will desirably be provided in a sterile form. It may be provided in unit dosage form and will generally be provided in a sealed container. A plurality of unit dosage forms may be provided.
- composition inside of pumps that are surgical embedded within a patient such that the AGGF1 peptide or expression vector can be injected (e.g., in a manner similar to insulin) to the subject’s blood stream when needed (see, e.g., Shah et ak, Int J Pharm Investig. 2016 Jan-Mar; 6(1): 1-9, herein incorporated by reference).
- compositions within the scope of the present technology may include one or more of the following: preserving agents, solubilizing agents, stabilizing agents, wetting agents, emulsifiers, sweeteners, colorants, flavoring agents, odorants, and/or salts.
- Compounds of the present technology may themselves be provided in the form of a pharmaceutically acceptable salt.
- embodiments may comprise buffers, coating agents, antioxidants, suspending agents, adjuvants, excipients, and/or diluents. Examples of preservatives include sodium benzoate, sorbic acid, and esters of p-hydroxybenzoic acid.
- compositions of the present technology may also contain other therapeutically active agents in addition to compounds of the present technology (e.g., insulin). Where two or more therapeutic agents are used they may be administered separately (e.g., at different times and/or via different routes) and therefore do not always need to be present in a single composition. Thus, combination therapy is within the scope of the present technology.
- compositions herein contain an expression vector encoding the AGGF1 peptide.
- the expression vectors is a plasmid, an adenovirus-associated viruses, or other viruses.
- the expression vectors are delivered by nano-technologies and/or targeted delivery systems.
- the routes for administration include, but are not limited to, one or more of: oral (e.g. as a tablet, capsule, or as an ingestable solution), implantable pump, topical, mucosal (e.g. as a nasal spray or aerosol for inhalation), nasal, parenteral (e.g. by an injectable form), gastrointestinal, intraspinal, intraperitoneal, intramuscular, intravenous, intrauterine, intraocular, intradermal, intracranial, intratracheal, intravaginal,
- AGGF1 peptides and expression vectors described herein is administered parenterally, then examples of such administration include one or more of: intravenously, intra-arterially, intraperitoneally, intrathecally, intraventricularly, intraurethrally,
- compositions adapted for oral administration are provided as capsules or tablets; as powders or granules; as solutions, food product, syrups or suspensions (in aqueous or non-aqueous liquids); as edible foams or whips; or as emulsions.
- Tablets or hard gelatin capsules may comprise lactose, maize starch or derivatives thereof, stearic acid or salts thereof.
- Soft gelatin capsules may comprise vegetable oils, waxes, fats, semi-solid, or liquid polyols etc.
- Solutions and syrups may comprise water, polyols and sugars.
- oils e.g., vegetable oils
- An active agent intended for oral administration may be coated with or admixed with a material that delays disintegration and/or absorption of the active agent in the gastrointestinal tract (e.g., glyceryl monostearate or glyceryl distearate may be used).
- a material that delays disintegration and/or absorption of the active agent in the gastrointestinal tract e.g., glyceryl monostearate or glyceryl distearate may be used.
- the AGGF1 peptides described herein may be applied topically in the form of a gel, hydrogel, lotion, solution, cream, ointment or dusting powder.
- the agent of the present technology may also be dermally or transdermally administered, for example, by the use of a skin patch.
- the agent of the present technology can be formulated as a suitable ointment containing the active compound suspended or dissolved in, for example, a mixture with one or more of the following: mineral oil, liquid petrolatum, white petrolatum, propylene glycol, polyoxyethylene
- polyoxypropylene compound emulsifying wax and water.
- it can be formulated as a suitable lotion or cream, suspended or dissolved in, for example, a mixture of one or more of the following: mineral oil, sorbitan monostearate, a polyethylene glycol, liquid paraffin, polysorbate 60, cetyl esters wax, cetearyl alcohol, 2-octyldodecanol, benzyl alcohol and water.
- AGGF1 peptides are administered parenterally, then examples of such administration include one or more of: intravenously, intra-arterially, intraperitoneally, intrathecally, intraventricularly, intraurethrally, intrasternally, intracranially, intramuscularly or subcutaneously administering the agent; and/or by using infusion techniques.
- the dosage is between 0.1 and 5 mg per kilogram of the subject (e.g., 0.1 ... 0.8 ... 1.5 ... 2.5 ... 4.0 ... 5.0 mg per kg).
- the agent and/or the pharmaceutical composition of the present technology may be administered in accordance with a regimen of from 1 to 10 times per day, such as once or twice per day, or only once every other day or every third day.
- the daily dosage level of the agent may be in single or divided doses.
- AGGF1 may be regarded as the“the 2 nd insulin” for treatment of diabetes. Compared to typical insulin treatment, which has a fast-acting effect in 30 minutes but lasts about 3 hours, AGGF1 treatment, in this Example, has a longer therapeutic effect with a peak effect at 18-24 hours and lasting for >42 hours. AAV-AGGF1 gene therapy can make its therapeutic effect last even for months.
- GTT glucose tolerance test
- AGGF1 protein (0.25 mg/kg body weight) was injected into HFD-induced hyperglycemic mice. Surprisingly, injection of AGGF1 protein dramatically reduced the fasting glucose level in HFD hyperglycemic mice compared with control PBS buffer (Fig. 2). The maximal glucose lowering effect of AGGF1 was achieved at 18-24 hours after injection, and the sustained effect can be observed 42 hours after the injection (Fig. 2). Chronic AGGF1 treatment of HFD-induced hyperglycemic mice every 2 days for 36 days maintained the significantly low fasting glucose level for the entire treatment period (Fig. 3).
- HFD-induced hyperglycemic mice were administered with insulin alone (1 ug/kg body weight) or combination of insulin (1 ug/kg body weight) and AGGF1 protein (0.25 mg/kg body weight) by intraperitoneal injection.
- Insulin treatment quickly decreased the fasting glucose level within 30 minutes and the effect of insulin was sustained for only 150 minutes.
- AGGF1 protein treatment significantly extended the effect of insulin (Fig. 4A).
- AGGF1 protein therapy is dependent on insulin.
- STZ streptozotocin
- Fig. 4B the glucose-lowering effect of AGGF1 was severely blunted when insulin secretion was inhibited by somatostatin
- Fig. 4C the glucose-lowering effect of AGGF1 protein therapy lowers blood glucose levels in an insulin-dependent manner.
- the interaction domain between AGGF1 and endothelial cells is localized between amino acids 574 and 624.
- endothelial cell adhesion assays were performed with human umbilical vascular endothelial cells (HUVECs) for full-length AGGF1 (WT) and serial N-terminal deletion mutant proteins. Significantly stronger binding (i.e. high fluorescence) was observed with WT-AGGF1 and deletion mutant proteins Nl to N10 than deletion mutant proteins Nl 1 and N12 ( Figure 12, left panel). These results suggest that the potential interaction domain of AGGF1 is between amino acid 574 and 624.
- Endothelial cell adhesion assays were also performed with WT-AGGF1 and serial C- terminal deletion mutant proteins of AGGF1. Significantly higher binding (i.e. fluorescence) was observed with WT-AGGF1 and deletion mutants Cl and C2 than deletion mutants C3 to C13 ( Figure 12, right panel). These results suggest that the potential interaction domain of AGGF1 with endothelial cells is between amino acid 564 and 614 ( Figure 10). Together, these results narrow the AGGFl-endothelail cell interaction domain to a 40-amino acid region (amino acids 574-614) at the C-terminus of AGGF1.
- AGGF1 responsible for endothelial cell migration is localized between amino acids 574 and 624.
- the cellular mechanism of angiogenesis involves endothelial cell motility.
- Endothelial cell migration assays were performed with WT-AGGF1 and serial N- and C- terminal deletion mutant AGGF1 proteins. Significantly more HUVECs migrated into the ‘wounded area’ after 24 hours in presence of WT-AGGF1 and deletion mutant proteins Nl to N10 than deletion mutant proteins Nll and N12 ( Figure 13). Significantly more HUVECs migration was also observed with deletion mutant proteins Cl and C2 than deletion mutant proteins C3 to C13 ( Figure 13). These endothelial cell migration assays demonstrate that the 40 amino acid AGGF1 binding domain (amino acids 574-614) is also responsible for inducing endothelial cell migration.
- AGGF1 responsible for angiogenesis is localized between amino acids 574 and 624.
- Angiogenesis assays were performed using the capillary endothelial tube formation assays.
- a significantly higher number of‘enclosed circles’ i.e. angiogenesis
- were formed by HUVECs after 16 hours in presence of WT-AGGF1 and deletion mutant proteins Nl to N10 than deletion mutant proteins Nll and N12 Figure 14
- a significantly higher number of‘enclosed circles’ were formed by HUVECs in presence of WT-AGGF1 and deletion mutant proteins Cl and C2 than deletion mutant proteins C3 to C13 ( Figure 14).
- These endothelial tube formation assays demonstrate that the 40 amino acid AGGF1 binding domain (amino acids 574-614) is responsible for angiogenesis.
- a short mutant AGGF1 protein with amino acids 604-714 has full functions of full- length AGGF1 in endothelial cell proliferation, migration, and angiogenesis
- AGGFl604-7i4 has full AGGF1 functions, we can utilize it for treatment of, for example, cardiac and vascular diseases, and the other diseases listed herein. Because AGGFl604-7i4has only 110 amino acid residues, it can be synthesized in vitro and used for clinical treatment of cardiac and vascular diseases. AGGFl 6 04-7i4 can be further optimized by many different modifications to increase its stability and functional activities in treatment of diseases.
- AGGF U04-714 can be further modified by removing more amino acids to identify the shortest form of AGGF1, but with full activities, which can be used for treatment of diseases (e.g., using the assays and procedures described in this Example).
- AGGFl604-7i4 can be further modified by adding some amino acids to increase its stability and activities.
- AGGFl604-7i4 can be further modified by adding some amino acids (e.g. mimotope) or by conjugating carriers for its targeted delivery to specific organs, tissues and cells for treatment of specific diseases.
- AGGFI604-714 can be further modified by synthesizing as a circular form (cyclotide) to increase its stability for disease treatment.
- Pulmonary arterial hypertension is characterized by an increased mean pulmonary arterial pressure (mPAP) of >25 mmHg, which is caused by functional and structural remodeling of pulmonary arteries, and in turn increases the burden on the right ventricle and eventually leads to right ventricular failure and premature death 5 ’ 6 .
- the pathogenesis of PAH is complex and involves pulmonary endothelial dysfunction, proliferation of vascular smooth muscle cells (VSMCs), inflammation, fibrosis, as well as abnormal thickening and contraction of pulmonary arteries, thrombus formation, and genetic abnormalities, leading to incremental increases of pulmonary vascular resistance, increased right ventricular loading, and eventual right ventricular failure and death. 7 11 Accurate and early diagnosis of PAH is seriously lacking because PAH symptoms appear late.
- Aggfl +/ KO mice develop spontaneous PAH.
- AGGF1 protein therapy can block PAH.
- RVSP right ventricular systolic pressure
- Figure 18 AGGF1 protein therapy also reduced RV hypertrophy (RV/LV+S) compared with IgG control ( Figure 18) although additional mice are needed to achieve statistical significance.
- AGGF1 protein therapy for PAH has several advantages.
- other VSMC proliferation blocking agents proteasome inhibitors bortezomib BTZ and carfizomib CFZ
- VSMC proliferation blocking agents have off-target effects (causing cardiac apoptosis in RV and LV).
- existing therapy for PAH such as calcium channel blockers causes hypotension, but AGGF1 treatment does not affect blood pressure under a normal physiological condition.
- Galie et ak Eur Heart J. 2016;37:67-119. 6. Galie et ak; 2015 ESC/ERS Guidelines for the Diagnosis and Treatment of Pulmonary Hypertension. Rev Esp Cardiol (Engl Ed). 20l6;69:l77.
- EPCs endothelial progenitor cells
- Aggfl +/ mice showed impairment of proliferation, migration, angiogenesis, and transendothelial migration as in hyperglycemic mice fed with a high fat diet (HFD) or db/db mice.
- HFD high fat diet
- db/db mice mice fed with a high fat diet
- the number of EPCs from Aggfl +/ mice was significantly reduced.
- AGGF1 protein can fully reverse damaging effects of hyperglycemia on EPCs.
- transplantation of AGGF1 -primed EPCs successfully restores blood flow, and blocks tissue necrosis and ambulatory impairment in HFD-induced hyperglycemic mice or db/db mice with diabetic hind-limb ischemia.
- AGGF1 activates AKT and reduces nuclear localization of Fyn, which increases the nuclear level of Nrf2 and expression of anti-oxidative genes, and inhibits ROS generation.
- DM Diabetes mellitus
- cardiovascular disease cardiovascular disease and diabetic peripheral arterial disease (PAD)
- PAD diabetic peripheral arterial disease
- EPCs endothelial progenitor cells
- EPCs are progenitors of endothelial cells (ECs) and have the potential to proliferate, migrate, home into the disrupted endothelium and differentiate into ECs to maintain endothelium integrity, restore endothelial dysfunction, promote neovascularization and repair damaged vessels (11; 13-15) .
- Cell-based therapy based on implantation of EPCs has emerged as a potential therapy for myocardial ischemia, brain ischemia and pulmonary embolism (16; 17).
- the efficacy of a therapy using EPC implantation for diabetic vascular complications is uncertain (18).
- autologous EPCs transplantation may not work efficiently because of the impaired function of such EPCs in DM by hyperglycemia (9; 11; 18).
- Wild-type (WT) C57BL/6J mice and db/db mice (Jackson Lab) were used in the study.
- Aggfl+/- KO mice with exons 2-11 deleted were described previously (20). Because homozygous Aggfl-/- KO mice die before E8.5, heterozygous Aggfl+/- KO mice were studied.
- a high fat diet (HFD) -induced mouse model for type 2 diabetes mellitus (T2DM) was previously described (25-27).
- EPCs were isolated from the bone marrow of WT, HFD-induced diabetic mice, and db/db mice, and cultured as described (11; 31). Mice were anesthetized with chloral hydrate (3%, m/v) via intraperitoneal injection. Bone marrow mononuclear cells (MNCs) were washed out from the femurs and tibias of mice with phosphate buffer saline (PBS), and then purified by density gradient centrifugation (2000 r/min) with histopaque-l083 (Sigma- Aldrich, St. Louis, MO) for 30 min at room temperature with a horizontal rotor centrifuge (Anke, LCJ-2B, Shanghai, China).
- PBS phosphate buffer saline
- the volume ratio of single cell suspension and histopaque-l083 separation liquid was 2:1.
- the cells were then washed twice using PBS, resuspended gently, plated in 0.1% (m/v) gelatin-coated cell culture dishes, and maintained in endothelial growth factor-supplemented media (EGM-2 bullet kit; Lonza, Switzerland) with 10% fetal bovine serum (FBS).
- EGM-2 bullet kit Lonza, Switzerland
- FBS fetal bovine serum
- Cells were cultured at 37°C with 5% C02 in a humidified water jacket incubator (Thermo Fisher Scientific, MA, USA).
- the EGM-2 medium was replaced every 3 days after first plating, and cellular morphology was monitored every day.
- MNCs Characterization of MNCs was performed as described (11; 31). Seven days after maintenance in endothelial-specific media and the removal of non-adherent MNCs, the remaining cells were subjected to immunostaining analysis. Cells were plated on gelatin- coated slide (ibidi) for one day, and then incubated with 10 pg/mL of acetylated Dil lipoprotein from human plasma (Dil-Ac-LDL, Thermo Fisher Scientific, MA, USA) at 37°C for 4 hours.
- the cells were washed 3 times with PBS, and incubated with 10 pg/mL of fluorescein isothiocyanate-labeled ulex europaeus lectin-l (Sigma-Aldrich, St. Louis, MO, USA) for 1 hour at 37°C.
- the cells were rinsed 3 times with PBS again, and visualized under a confocal microscopy.
- MNCs were also characterized by flow cytometry analysis.
- Cells were incubated with 5% BSA (Sigma-Aldrich, St. Louis, MO) for 15 minutes for to block non-specific binding, and then stained with anti-mouse CD34-phycoerythrin (PE), CD31-PE, CD45-PE or CD144- PE (BD Biosciences, San Jose, CA, USA) at room temperature for 1 hour, respectively.
- BSA Sigma-Aldrich, St. Louis, MO
- PE anti-mouse CD34-phycoerythrin
- CD31-PE CD45-PE
- CD144- PE BD Biosciences, San Jose, CA, USA
- the same fluorescein-labeled isotype IgG served as negative control.
- Cells were analyzed using a Beckman CytoFLEX, and data were analyzed using FlowJo software version 10 (TreeStar, Ashland, OR, USA). Immunostaining for CD31
- muscle samples were fixed overnight in paraformaldehyde (4%), embedded in paraffin and sectioned. Sections were deparaffinized, dehydrated and rehydrated after being sectioned. After microwave antigen retrieval, endogenous peroxidase blocking and normal goat serum blocking in equilibration buffer, sections were subject to
- AGGF1 protein was purified as described previously (21; 33). EPCs isolated from mice were treated with purified AGGF1 protein (0.5 pg/ml) or negative control elution buffer for purification at 37°C for l2h. Two days after the diabetic hind-limb ischemia surgery, AGGFl-primed EPCs (-1x106) were injected into mice via the tail vein. Blood flow in both legs was measured in mice anesthetized with chloral hydrate (3%, m/v) using a Vevo 2100 High- Resolution Microultrasound System (Visualsonics Inc) immediately before the ischemic surgery and at time-points of 7, 14 and 28 days after ischemia.
- Visualsonics Inc Vevo 2100 High- Resolution Microultrasound System
- Vs peak systolic velocity
- Vd minimal end diastolic flow velocity
- Va temporal average velocity per cardiac cycle
- Human embryonic kidney 293 cells (HEK293) were transfected with a lentiviral vector with EGFP, the packaging plasmid psPAX2 and the envelope plasmid pMD2.G using Lipofectamine 2000 (Invitrogen, Carlsbad, CA, USA). Viral supernatants were produced from the transfected HEK293 as described (11). HEK293 cells were maintained at 37°C in the high-glucose Dulbecco’s Modified Eagle’s medium (DMEM) supplemented with 10% (V/V) FBS (Gibico Life Technologies, New York, USA) in a humidified water jacket incubator with 5% C02. The supernatant was harvested 72 hours after transfection, filtered through 0.45-pm polyvinylidene fluoride filters, and stored at -80°C until use.
- DMEM Modified Eagle’s medium
- EPCs were infected with the GFP-lentiviruses overnight at a multiplicity of infection of 50 with 2.5 pg/mL polybrene supplemented, and the culture medium was replaced with the fresh growth medium 24 hours after infection. After transfection for 72 hours, the infection efficiency was determined by GFP expression under a Nikon EclipseTi micorscopy (Nikon, Japan).
- siRNAs for Nrf2 and AKT negative control siRNAs were chemically synthesized by Ribobio.
- the control siRNA is the silencer scrambled negative control siRNA (siNC) having no significant homology to any known gene sequences from the mouse genome.
- siRNAs are as follows: Nrf2 sense: 5’-CGAGAAGUGUUUGACUUUATT-3’ (SEQ ID NO:l9), Nrf2 antisense: 5’-UAAAGUCAAACA-CUUCUCGTT-3’ (SEQ ID NO:20); AKT sense: 5'-UGCCCUUCUACAACCAGGATT-3' (SEQ ID NO:2l), AKT antisense: 5’-UCCUGGUUGUAGAAGGGCATT-3’ (SEQ ID NO:22); siNC sense: 5’- UUCUC-CGAACGUGUCACGUTT-3’ (SEQ ID NO:23), and siNC antisense: 5’- ACGU G AC AC- GUUCGG AG AATT- 3’ (SEQ ID NO:24). EPCs were transfected with siRNA in the Opti-MEM medium overnight using transfection reagent (Santa Cruz).
- EPCs were seeded into 96-well cell culture plates, and treated with AGGF1 (0.5 pg/mL) or a solution with high D-glucose (30 mM, Sigma, St. Louis, MO, control, L-glucose). Then, 10 pl/well of CCK-8 solution was added, and the plates were incubated for 1-4 hours in dark. Cell proliferation was determined by the measurement of absorbance at 450 nm using a Molecular Devices VERSA max microplate reader as described (39; 40).
- TEM transendothelial migration
- EPCs Human umbilical vein endothelial cells (HUVECs) (about lxlO 4 cells per well) were cultured in the upper chamber of a 24-transwell insert (8.0 pm pores; BD Falcon, San Jose, CA, USA) to confluency. Monolayer confluency was confirmed under an inverted fluorescence microscopy before each experiment.
- EPCs were treated with AGGF1 vs. elution buffer, a high glucose solution vs. control, and siRNAs, and cultured for 24 hours. The EPCs were harvested and resuspended in the basal culture medium (EBM-2, 0.5% BSA).
- EPC suspension ( ⁇ 0.2 mL) was added to the upper chamber of Transwell, and 0.5 ml of EBM-2 medium was added to the lower chamber.
- Cells were cultured for 24 hours at 37°C, and EPCs that migrated from the upper chamber to the lower chamber of Transwell were visualized. Images were captured at 40x magnification under an inverted Nikon Eclipse Ti microscope (Nikon, Japan). The number of cells was counted and analyzed.
- EPCs in cell culture dishes were trypsinized and resuspended in growth media and allowed to grow to a confluent monolayer in 6-well plates. EPCs were starved for 6 hours by replacing the regular growth media with serum-free growth media. EPC monolayers were then wounded with a 200 m ⁇ pipette tip gently through the cell sheet to remove some cells and make a wound. The growth medium was aspirated out and wells washed twice with PBS to remove cell debris. Media supplemented with AGGF1 (0.5 pg/ml) were added to the wells in a total volume of 1 ml of serum-free growth media. Cells were cultured for 24 hours by incubating the plate at 37°C and 5% C02. Images were captured at 40X magnification under an inverted Nikon Eclipse Ti microscope (Nikon, Japan). The movement of cells into the scraped area was measured.
- Protein extracts were prepared from cultured EPCs with Western-IP lysis buffer (Beyotime, Beijing, China) supplemented with a proteinase inhibitor cocktail (Roche, Basel, Switzerland), separated by 12% SDS-PAGE, and transferred to polyvinylidene-fluoride membranes.
- the membranes were blocked with skim milk and incubated with appropriate primary antibodies overnight at 4°C with gentle shaking. The membranes were then incubated with appropriate secondary antibodies for 2 hours at room temperature after standard TBST washing procedures.
- Nrf2 The primary antibodies against Nrf2 (1:1000 dilution), heme oxygenase-l (HQ-l, 1:2000), NAD(P)H dehydrogenase quinone 1 (NQO-l, 1:1000), catalase (1:1000), and GAPDH (1:5000) were purchased from ProteinTech (Wuhan, China).
- Ser473; 1:1000 were purchased from Cell Signaling Technology (Danvers, MA, USA). Images from Western blot analysis were developed using a ChemiDoc XRS (Bio-Rad Laboratories, Richmond, CA, USA) with the SuperSignal West Pico Chemiluminescent Substrate (Pierce Chemical Co., Rockford, IL, USA), and further analyzed with a Gel-Pro analyzer. GAPDH was used as the loading control.
- RNA samples were isolated from EPCs using Trizol reagent (TaKaRa Biotech, Japan), and reversed transcribed into cDNA with M-MLV reverse transcriptase (Promega, Madison, wi) according to the manufacturer’s instruction.
- DNase I Thermo Fisher
- Quantitative real-time RT-PCR analysis was performed in triplicate with a 10 pl reaction system using the FastStart Universal SYBR Green Master (Roche, Basel, Switzerland) and a 7900 HT Fast Real-Time PCR System (ABI, US).
- the PCR profile was 94°C for 5 min, and 40 cycles of 94°C for 10 s and 60°C for 15 s. Fold differences between samples were assessed with a comparative cycle time (Ct) method.
- Ct comparative cycle time
- GAPDH served as an internal standard. The data were analyzed using 2-AACt relative expression quantity as reported (41).
- MNCs bone marrow-derived mononuclear cells
- WT wild type
- Aggfl +/- KO mice The isolated MNCs appeared to be centrally rounded EPCs with a peripheral spindle shape four days after culture (Fig. 25 A). The cells appeared to be spindle-shaped adherent EPCs and some showed cobblestone-like morphology at day 8 (Fig. 25A).
- the isolated EPCs were confirmed by a Dil-Ac-LDL endocytosis assay (red fluorescence) and a UEA-l binding assay (green fluorescence).
- the isolated EPCs were able to endocytose Ac-LDL and bind UEA-l (Fig. 25B), suggesting that they are endothelial lineage cells with characteristics of EPCs as previously described (32;
- Fig. 19D-E neovascularization in a transwell assay
- Fig. 19F-G reduced migration in a scratch- wound assay
- WT EPCs isolated form wild type mice
- Aggfl +/- showed similar effects as db/db mice (Fig. 19A-G).
- Aggfl +/- db/db double KO mice showed exacerbated effects compared with Aggfl +/- KO mice (Fig. 19A-
- AGGF1 is an angiogenic factor that can be secreted outside of cells (21), therefore, we treated EPCs with the purified human AGGF1 protein in culture to determine the effects of AGGF1 on functions of EPCs (Fig. 20A). All the defects by high glucose on EPC functions were blocked by AGGF1 treatment (Fig. 20B-H).
- Fig. 20A All the defects by high glucose on EPC functions were blocked by AGGF1 treatment (Fig. 20B-H).
- Fig. 20B-H We analyzed the expression levels of VEGF, VEGFR2 and CXCR4 mRNA in EPCs treated with or without AGGF1 in combination with or without high glucose.
- AGGF1 did not affect the expression levels of VEGF, VEGFR2 and CXCR4 in EPCs (Fig. 31).
- AGGF1 Boosts EPC-Mediated Angiogenesis and Blood Perfusion in T2MD
- AGGF1 In a T2DM mouse model (db/db mice), AGGF1 was found to greatly boost the therapeutic effects of cell therapy with EPC implantation on vascular complications under ischemia in vivo. As AGGF1 can reverse the damaging effects of T2DM on EPCs, we hypothesized that AGGFl-primed EPCs can serve as a successful treatment tool for vascular complications in T2MD in vivo.
- AGGFl-primed EPCs were significantly more effective than elution-buff er- pretreated EPCs in increasing blood flow in diabetic mice with ischemia (Fig. 21A).
- the rate of tissue necrosis was significantly decreased by AGGFl-primed EPCs compared with elution-buffer-pretreated control EPCs (Fig. 21B).
- Similar significant inhibitory effect on ambulatory impairment was detected for AGGFl-primed EPCs (Fig. 21C).
- Immunostaining showed that the density of CD3l-postiive vessels was significantly higher for AGGFl- primed EPCs than for elution-buffer-pretreated control EPCs at day 28 after transplantation (Fig. 21D and Fig. 32A).
- the therapeutic effect of AGGFl-primed EPCs may be related to the possibility that AGGF1 increases the homing of transplanted EPCs to the ischemic areas.
- AGGF1 increases the homing of transplanted EPCs to the ischemic areas.
- AGGF1 can activate PI3K and AKT signaling in endothelial cells and in zebrafish (19; 20).
- PI3K and AKT were shown to regulate phosphorylation and nuclear localization of Fyn (19; 20).
- Fyn was shown to phosphorylate Nrf2 at Y568, resulting in nuclear export and degradation of Nrf2 (19; 20). Therefore, we hypothesized that AGGF1 regulates ROS generation and EPC functions via an AGGF1- AKT-Fyn-Nrf2 signaling pathway.
- EPCs showed a significantly reduced level of phosphorylation of AKT, however, the effect was reversed by AGGF1 treatment (Fig. 23A).
- Nrf2 is a transcription factor important to cellular defense against oxidative stress. Nrf2 needs to be translocated into the nucleus to execute its function, and its nuclear localization is regulated by AKT-Fyn signaling. AGGF1 activates AKT, which leads to de-phosphorylation of Fyn, resulting in reduced translocation of Fyn into the nucleus and decreased nuclear Fyn (n-Fyn).
- n-Fyn leads to decreased phosphorylation of Nrf2 and inhibits the nuclear export, ubiquitination and degradation of Nrf2, increasing the expression levels of nuclear Nrf2 and downstream cytoprotective genes such as NQO-l and CAT (Fig. 22).
- Oxidative stress was considered to be a critical factor accounting for EPC dysfunction in T2DM (44; 45).
- Our results suggest that AGGF1 protects EPCs partly by inhibiting hyperglycemia-induced ROS generation through a novel anti-oxidative stress signaling pathway (Fig. 39).
- Endothelial dysfunction is a major problem in T2DM, and lack of endothelial regeneration and impaired angiogenesis by EPCs are responsible for the vascular abnormalities in DM (10; 11).
- DM vascular abnormalities
- the data in this Example establish AGGF1 as an important molecule to repair diabetic EPC dysfunction induced by hyperglycemia or dyslipidemia in db/db mice and HFD-induced hyperglycemia mice (Fig. 21, Fig.32 and Fig. 33).
- An AGGFl-based EPC therapy for diabetic ischemia may have additional benefits for DM patients who are often affected with CAD/MI, heart failure, peripheral vascular disease, and other conditions. Furthermore, DM can also cause many other diseases including diabetic cardiomyopathy, end-stage renal disease (ESRD) and vascular aging, which are all associated with abnormal angiogenesis and vascular dysfunction. Based on our data, it is likely that AGGF1 -primed EPC therapy may serve as a new strategy not only for treating DM-associated vascular abnormalities, and those with vascular abnormalities without DM, but also for many other diabetes mellitus complications such as diabetic cardiomyopathy, ESRD and vascular aging.
- ESRD end-stage renal disease
- AGGF1 protein therapy reduces plasma glucose levels in db/db diabetic mice.
- AGGF1 protein therapy is effective in lowering fasting glucose levels in HFD- induced diabetic mice.
- the db/db mice are a well-established murine model for diabetes.
- the overall study design is shown in Figure 40A.
- Figure 40B compared to IgG control, AGGF1 significantly lowers the plasma glucose levels for more than 24 hours. The effect was lost at the time point of 30 hours.
- AGGF1 The therapeutic effects of AGGF1 is more than 10 times longer than insulin treatment.
- U.K. prospective diabetes study 16 Overview of 6 years' therapy of type II diabetes: a progressive disease. U.K. Prospective Diabetes Study Group. Diabetes.
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Chemical & Material Sciences (AREA)
- Medicinal Chemistry (AREA)
- Vascular Medicine (AREA)
- Engineering & Computer Science (AREA)
- General Health & Medical Sciences (AREA)
- Zoology (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Organic Chemistry (AREA)
- Pharmacology & Pharmacy (AREA)
- Veterinary Medicine (AREA)
- Public Health (AREA)
- Animal Behavior & Ethology (AREA)
- Immunology (AREA)
- Epidemiology (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- Cell Biology (AREA)
- Gastroenterology & Hepatology (AREA)
- Biomedical Technology (AREA)
- Developmental Biology & Embryology (AREA)
- Virology (AREA)
- Urology & Nephrology (AREA)
- Cardiology (AREA)
- Heart & Thoracic Surgery (AREA)
- Chemical Kinetics & Catalysis (AREA)
- General Chemical & Material Sciences (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Biotechnology (AREA)
- Toxicology (AREA)
- Biochemistry (AREA)
- Biophysics (AREA)
- Genetics & Genomics (AREA)
- Molecular Biology (AREA)
- Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
- Peptides Or Proteins (AREA)
Abstract
Provided herein are compositions, systems, kits, and methods for treating a subject with a disease or condition characterized by abnormal glucose metabolism (e.g., diabetes) or ischemic vascular disease (e.g., PAD or CAD) by administering a composition comprising AGGF1 peptides, or an expression vector encoding said peptides, or AGGF1-primed endothelial progenitor cells, to a subject. In certain embodiments, the AGGF1 peptide is full-length AGGF1, or a biologically active fragment or mutant thereof. In some embodiments, the AGGF1 peptides or expression vectors are administered around the same time insulin is administered to the subject.
Description
AGGF1 AND AGGF1-PRIMED CELLS FOR TREATING DISEASES AND
CONDITIONS
The present application claims priority to U.S. Provisional applications serial number 62/744,488 filed October 11, 2018, and serial number 62/842,283, filed May 2, 2019, both of which are herein incorporated by reference.
GOVERNMENT FUNDING
This invention was made with government support under grant number HL077107 awarded by National Institutes of Health. The government has certain rights in the invention.
FIELD
Provided herein are compositions, systems, kits, and methods for treating a subject with a disease or condition characterized by abnormal glucose metabolism (e.g., diabetes) or ischemic vascular disease (e.g., PAD or CAD) by administering a composition comprising AGGF1 peptides, or an expression vector encoding said peptides, or AGGF1 -primed endothelial progenitor cells, to a subject. In certain embodiments, the AGGF1 peptide is full- length AGGF1, or a biologically active fragment or mutant thereof. In some embodiments, the AGGF1 peptides or expression vectors are administered around the same time insulin is administered to the subject.
BACKGROUND
Diabetes mellitus is a common and heterogeneous disease with high morbidity and mortality worldwide. An estimated 422 million adults are affected with diabetes based on the WHO’s most recent 2016 data. The prevalence of diabetes, and in particular, of type 2 diabetes (T2DM), increases rapidly each year due to lifestyle habits and increased rates of aging, inactivity, and obesity, and poses an important challenge to human health. However, the treatment of diabetes remains suboptimal in a significant proportion of patients. Despite intensive use of current antidiabetic drugs, >50% of T2DM patients have poor glycemic control and 18% develop severe complications. Population HbAlc concentrations failed to decline as much as expected due to poor medication compliance and other causes. To address this grand challenge, much is needed to understand the pathophysiology of diabetes
and to develop new innovative therapies. In particular, long-lasting treatment can dramatically reduce poor medication compliance.
SUMMARY
Provided herein are compositions, systems, kits, and methods for treating a subject with a disease or condition characterized by abnormal glucose metabolism (e.g., diabetes) or ischemic vascular disease (e.g., PAD or CAD) by administering a composition comprising AGGF1 peptides, or an expression vector encoding said peptides, or AGGF1 -primed endothelial progenitor cells, to a subject. In certain embodiments, the AGGF1 peptide is full- length AGGF1, or a biologically active fragment or mutant thereof. In some embodiments, the AGGF1 peptides or expression vectors are administered around the same time insulin is administered to the subject.
In some embodiments, provided herein are methods of treating a subject with a disease or condition characterized by abnormal glucose metabolism comprising:
administering a composition comprising: i) an AGGF1 peptide, or ii) an expression vector encoding the AGGF1 peptide, to the subject with the disease or condition. In particular embodiments, the disease or condition is Type I or Type II diabetes. In certain embodiments, the disease or condition is selected from the group consisting of: hyperglycemia, inadequate glucose clearance, and hyperinsulinemia. In particular embodiments, the methods further comprise administering insulin to the subject. In other embodiments, the insulin is administered at about the same time as the composition.
In certain embodiments, provided herein are methods of treating a subject comprising: administering a composition to a subject, wherein the subject has at least one of the following: a disease or condition characterized by abnormal glucose metabolism, ischemic vascular disease, pulmonary hypertension, cardiac hypertrophy, and heart failure: wherein said composition comprises: i) an AGGF1 peptide, and/or ii) an expression vector encoding said AGGF1 peptide.
In some embodiments, provided herein are methods of treating a subject with a cardiovascular disease or condition characterized comprising: administering a composition comprising: i) an AGGF1 peptide that is no longer than 600 amino acids (e.g., 100-600 or 100-300 or 100-150 amino acids in length), or ii) an expression vector encoding the AGGF1 peptide, to the subject with the disease or condition. In certain embodiments, the disease or condition is selected from the group consisting of: coronary artery disease, myocardial
infarction, cardiac hypertrophy, heart failure, peripheral artery disease, restenosis and in-stent thrombosis after angioplasty and stenting, and pulmonary arterial hypertension.
In further embodiments, provided herein are methods of treating a subject with pulmonary arterial hypertension (PAH) comprising: administering a composition comprising: i) an AGGF1 peptide, or ii) an expression vector encoding the AGGF1 peptide, to the subject with the PAH.
In some embodiments, the subject in any of the methods herein is a human and: A) the composition comprises said AGGF1 peptide, and wherein the dosage is selected from the group consisting of: 0.1-15 mg/kg (e.g., 0.1 mg/kg ... 4.0 mg/kg ... 8.0 mg/k ... 12.0 mg/kg ... and 15 mg/kg), 0.5-10 mg/kg, and 1-7 mg/kg; or B) said composition comprises said expression vector encoding said AGGF1 peptide, and wherein said dosage is about 1.0 x 1011 - 5.0 x 1013 (e.g., 1.0 x 1011 ... 1.0 x 1012 ... or 5.0 x 1013).
In particular embodiments, provided herein are systems and kits comprising: a) a blood glucose monitoring device or article of manufacture; and b) a composition comprising: i) an AGGF1 peptide, or ii) an expression vector encoding the AGGF1 peptide. In some embodiments, the glucose monitoring device or article of manufacture is selected from the group consisting of: a blood glucose test strip, a reader for reading the glucose test strip, FreeStyle Libre, GlucoTrack, Eversense, GlucoWise, NovioSense, GlucoSense, and a smart contact lens.
In further embodiments, provided herein are systems and kits comprising: a) a composition comprising: i) an AGGF1 peptide, or ii) an expression vector encoding the AGGF1 peptide; and b) an implantable pump containing the composition, wherein the implantable pump is configured to deliver an amount of the composition into the bloodstream of a subject.
In certain embodiments, provided herein are compositions comprising: a) an AGGF1 peptide, or an expression vector encoding the AGGF1 peptide; and b) insulin peptide, or an expression vector encoding the insulin peptide.
In some embodiments, provided herein are methods of treating a subject comprising: administering a composition to a subject, wherein the subject has a disease or condition characterized by abnormal glucose metabolism and/or ischemic vascular disease, and wherein the composition comprises an AGGFl-primed endothelial progenitor cell. In certain
embodiments, the disease or condition is selected from the group consisting of peripheral artery disease (PAD), coronary artery disease (CAD), coronary heart disease (CHD), and myocardial infarction (MI). In other embodiments, the AGGFl-primed endothelial progenitor cell comprises an endothelial progenitor cell (EPC) from the subject, wherein the EPC has been exposed to AGGF1 peptides ex vivo. In further embodiments, the AGGFl- primed endothelial progenitor cell comprises an endothelial progenitor cell (EPC) from a relative of the subject, wherein the EPC has been exposed to AGGF1 peptides ex vivo. In additional embodiments, the AGGFl-primed endothelial progenitor cell comprises an endothelial progenitor cell (EPC) from a non-relative of the subject, wherein the EPC has been exposed to AGGF1 peptides ex vivo.
In some embodiments, provided herein are compositions comprising: an AGGFl- primed endothelial progenitor cell. In further embodiments, the composition further comprises cell growth media. In other embodiments, the AGGFl-primed endothelial progenitor cell comprises an endothelial progenitor cell (EPC) from a subject with a disease or condition characterized by abnormal glucose metabolism and/or ischemic vascular disease, wherein the EPC has been exposed to AGGF1 peptides ex vivo. In additional embodiments, the compositions further comprise a physiological tolerable liquid.
In other embodiments, provided herein are kits and systems comprising: a) an endothelial progenitor cell (EPC), and b) a composition comprising: i) an AGGF1 peptide, and/or ii) an expression vector encoding the AGGF1 peptide.
In certain embodiments, the AGGF1 peptide comprises full length human AGGF1 shown in SEQ ID NO: 1. In other embodiments, the AGGF1 peptide has at least 97% sequence identity with SEQ ID NO: 1 (e.g., 97 ... 98 .. 99 .. 99.5% identify). In other embodiments, the AGGF1 peptide is selected from any of SEQ ID Nos:2-l6 or i) a peptide comprising the amino acid sequence from 574 to 614 of SEQ ID NO:l, ii) a peptide comprising the amino acid sequence from 574 to 624 of SEQ ID NO:l, or iii) a peptide comprising the amino acid sequence from 604 to 714 of SEQ ID NO:l. In further embodiments, the AGGF1 peptide has at least 97% sequence identity (e.g., 97 ... 98 .. 99 ..
99.5% identify) with any one of SEQ ID Nos:2-l6. In certain embodiments, the
administering reduces or eliminates at least one symptom of the disease or condition. In some embodiments, the subject is a human. In certain embodiments, the AGGF1 peptide consists of, consists essentially of, or comprises QRDDAPASVH (SEQ ID NO: 18). In certain embodiments, an expression vector encoding SEQ ID NO: 18 is employed. In some
embodiments, the sequence encoding SEQ ID NO: 18 is CAAAGAGAUGAUG
CUCCUGCAUCUGUUCAU (SEQ ID NO:25). In certain embodiments, the expression vectors herein comprise SEQ ID NO:25.
DESCRIPTION OF THE FIGURES
The patent or application file contains at least one drawing executed in color. Copies of this patent or patent application publication with color drawings will be provided by the Office upon request and payment of the necessary fee.
Fig. 1. AGGF1 regulates glucose metabolism and insulin resistance. (A) Body weight of AGGFl+/_ KO mice and littermate WT mice fed a standard chow diet or a high fat diet (HFD). (B) Fasting glucose levels in AGGFl+/_ KO mice and littermate WT mice fed HFD. (C) Fasting insulin levels in AGGFl+/ KO mice and littermate WT mice fed HFD. (D) Glucose tolerance tests in AGGFl+/_ KO mice and littermate WT mice fed HFD. (E) Insulin tolerance tests in AGGFl+/ KO mice and littermate WT mice fed HFD. *P<0.01,
**P<0.005, n=8.
Fig. 2. AGGF1 protein therapy for diabetes. Intraperitoneal injection of AGGF1 protein dramatically lowers fasting glucose levels in HFD-induced diabetic mice (24-week- old male mice fed HFD for 16 weeks). *P<0.01, **P<0.005, n=8.
Fig. 3. Therapeutic effects of chronic AGGF1 protein therapy on lowering fasting glucose levels. 24-week-old male HFD-induced diabetic mice were treated by intraperitoneal injection of 0.25 mg/kg AGGF1 protein or PBS control every 2 days for 36 days (chronic treatment). (A) Fasting serum glucose levels. (B) Body weight. *P<0.01, **P<0.005, n=8.
Fig. 4. AGGF1 protein therapy is insulin-dependent. (A) Therapeutic effects of insulin alone (1.00 ug/kg body weight) and in combination with AGGF1 (0.25 mg/kg body weight) in 24-week-old male HFD-induced diabetic mice. (B) No therapeutic effect for AGGF1 protein injection on fasting glucose levels in 16-week-old male diabetic mice induced by streptozotocin (STZ) for 4 weeks. (C) Therapeutic effect of AGGF1 protein injection is abolished in 24- week-old male HFD-induced diabetic mice treated with somatostatin. *P<0.01, **P<0.005, NS, not significant, n=8.
Figure 5a shows the amino acid sequence of full-length human AGGF1 (714 amino acid residues) (SEQ ID NO:l). Figure 5b shows the amino acid sequence of AGGF1-N1
(696 amino acids; 19 amino acid deletion from the N-terminus of AGGF1) (SEQ ID NO:2). Figure 5c shows the amino acid sequence of AGGF1-N2 (629 amino acids; 86 amino acid deletion from the N-terminus of AGGF1) (SEQ ID NOG).
Figure 6a shows the amino acid sequence of AGGF1-N3 (550 amino acids; 165 amino acid deletion from the N-terminus of AGGF1) (SEQ ID NO:4). Figure 6b shows the amino acid sequence of AGGF1-N4 (496 amino acids; 219 amino acid deletion from the N- terminus of AGGF1) (SEQ ID NOG). Figure 6c shows the amino acid sequence of AGGF1- N5 (431 amino acids; 284 amino acid deletion from the N-terminus of AGGF1) (SEQ ID NO:6). Figure 6d shows the amino acid sequence of AGGF1-N6 (366 amino acids; 349 amino acid deletion from the N-terminus of AGGF1) (SEQ ID NO:7).
Figure 7a shows the amino acid sequence of AGGF1-N7 (302 amino acids; 413 amino acid deletion from the N-terminus of AGGF1) (SEQ ID NOG). Figure 7b shows the amino acid sequence of AGGF1-N8 (244 amino acids; 472 amino acid deletion from the N- terminus of AGGF1) (SEQ ID NO:9). Figure 7c shows the amino acid sequence of AGGF1- N9 (193 amino acids; 522 amino acid deletion from the N-terminus of AGGF1) (SEQ ID NO: 10). Figure 7d show the amino acid sequence of AGGF1-N10 (141 amino acids; 574 amino acid deletion from the N-terminus of AGGF1) (SEQ ID NO: 11). Figure 7e shows the amino acid sequence of AGGF1-N101 (131 amino acids; 584 amino acid deletion from the N-terminus of AGGF1) (SEQ ID NO: 12). Figure 7f shows the amino acid sequence of AGGF1-N102 (121 amino acids; 594 amino acid deletion from the N-terminus of AGGF1) (SEQ ID NO: 13).
Figure 8a shows the amino acid sequence of AGGF1-N103 (110 amino acids; 604 amino acid deletion from the N-terminus of AGGF1) (SEQ ID NO: 14). Figure 8b shows the amino acid sequence of AGGF1-C1 (664 amino acids; 50 amino acid deletion from the C- terminus of AGGF1) (SEQ ID NO: 15). Figure 8c shows the amino acid sequence of AGGF1-C2 (614 amino acids; 100 amino acid deletion from the C-terminus of AGGF1) (SEQ ID NO: 16).
Figure 9 shows the full length human AGGF1 cDNA (SEQ ID NO: 17), with arrows showing truncation mutants.
Figure 10 shows a diagram showing the full length AGGF1 (WT) and its serial N- terminal and C-terminal deletion mutant proteins.
Figure 11 shows results of purification of mutant AGGF1 proteins with serial N- and C-terminal deletions.
Figure 12 shows results of AGGF1 -endothelial cell adhesion (binding) assays for serial N- and C-terminal AGGF1 deletion mutants.
Figure 13 shows results of Endothelial cell migration assays for serial N- and C- terminal AGGF1 deletion mutants.
Figure 14 shows results of angiogenesis assays for serial N- and C-terminal AGGF1 deletion mutants (capillary endothelial tube formation assays).
Figure 15. Micro-deletion analysis identified the short mutant form of AGGF1 with full functions of the full-length AGGF1 protein. (A) A diagram showing the full length AGGF1 (WT) and serial N-terminal deletion mutant proteins. (B) Endothelial cell adhesion assay with AGGF1 and its N-terminal deletion mutants. Coating buffer (CB) with and without BSA was used as a negative control. *P<0.05. (C) Endothelial cell migration assay with AGGF1 and its N-terminal deletion mutants. BSA was used as a negative control. *P<0.05. (D) Endothelial tube formation assay AGGF1 and its N-terminal deletion mutants. BSA was used as a negative control (left panel). *P<0.05. (E) Western blot analysis of pAKTSer473 in HUVEC lysates. The cells were stimulated with AGGF1 and its N-terminal deletion mutants respectively. BSA was used as a negative control (left panel). *P<0.05.
Figure 16. Aggfl+/ KO mice spontaneously developed PAH. Male Agg[]0,‘o/+ KO mice (16-20 weeks) showed much higher mPAP (mean pulmonary arterial pressure) (A, B), total pulmonary resistance (TPR) (C), and right ventricular hypertrophy compared to WT mice (D). We assessed right ventricular hypertrophy by measuring the ratio of RV weight over the weight of the LV and septum, i.e. the RV/(LV + S) weight. **R<0.01; n=6.
Figure 17. Aggfl+/ KO causes vascular remodeling. (A) Representative H&E of pulmonary arteries. (B) Mean medial wall thickness. (C) Resistance pulmonary arterial muscularization. For muscularization studies, lung sections (5 pm) were co-immunostained with an a-smooth muscle actin antibody (a-SMA, VSMC marker) and a CD31 antibody (EC maker). Vessels were classified as fully (100%), partially, or non-muscularized (0%) vessels based on the percentage of CD31 signal surrounded by a-SMA signal. (D) PASMC proliferation (Ki67-positive dividing cells). **R<0.01; n=6.
Figure 18. AGGF1 attenuates the development of PAH in a hypoxic mouse model. Three-month-old male C567BL/6 mice were induced to develop PAH by hypoxia (10% oxygen for three weeks). Recombinant AGGF1 protein or control IgG (0.25 mg/KG body weight) was delivered by tail vein injection one day prior to hypoxia and continued twice a week for 3 weeks. RVSP, right-ventricular systolic pressure; RV/LV+S, RV hypertrophy ratio.
Figure 19. Aggfl is required for essential functions of EPCs in capillary tube formation, proliferation, transendothelial migration, and migration as in a diabetic mouse model ( db/db mice). EPCs were isolated from bone marrow of WT, heterozygous Aggfl +/ KO mice, db/db mice, and Aggfl +/ db/db mice and characterized. (A) Aggfl
haploinsufficiency inhibits angiogensis mediated by EPCs. (B) Images from (A) were analyzed, quantified and plotted. (C) Aggfl haploinsufficiency inhibits EPC proliferation.
(D) Aggfl haploinsufficiency inhibits transendothelial migration of EPCs in transwell assays.
(E) Images from (D) were quantified and plotted. (F) Aggfl haploinsufficiency inhibits EPC migration in wound scratch migration assays. (G) Representative images from EPC migration assays. Data are shown as mean ± SD. *P<0.05, **R<0.01, (n=6 mice per group).
Figure 20. AGGF1 dramatically improves essential functions of EPCs impaired by HG. (A) Western blot analysis showing increased AGGF1 expression in EPCs. (B) AGGF1 treatment reversed the impairment of angiogenic function of EPCs by HG. (C) Images from (B) were analyzed, quantified and plotted. (D) AGGF1 treatment reversed the reduced cell proliferation of EPCs by HG. (E) AGGF1 treatment reversed the HG-impaired
transendothelial migration of EPCs. (F) Images from (E) were quantified and plotted. (G) AGGF1 treatment reversed the HG-impaired migration of EPCs in scratch wound migration assays. (H) Representative images from EPC migration assays. Data are shown as mean ± SD. *P<0.05, **R<0.01, (n=6 mice per group).
Figure 21. AGGF1 protein treatment robustly potentiates the therapeutic effects of EPCs on peripheral vascular complications in a hindlimb ischemia model in db/db mice. A hindlimb ischemia model was created in db/db mice. (A) Transplantation of AGGF1- pretreated EPCs dramatically improved blood perfusion compared with EPCs without AGGF1 pretreatment in db/db mice. (B) Therapeutic effects of AGGF1 -pretreated EPCs on necrosis compared with EPCs without AGGF1 pretreatment. (C) Therapeutic effects of AGGF1 -pretreated EPCs on ambulatory impairment compared with EPCs without AGGF1
pretreatment. (D) Effects of AGGF1 -pretreated EPCs on the density of CD3l-positive capillary vessels compared with EPCs without AGGF1 pretreatment.
Figure 22. AGGF1 regulates the nuclear accumulation of Nrf2. (A) Western blot analysis for the effect of AGGF1, HG and HG+AGGF1 on the expression levels of nuclear Nrf2 (n-Nrf2) and its downstream signaling molecules, including HQ-l, NQO-l, and CAT. GAPDH was used as loading control. (B) AGGF1 protein treatment induces expression of Nrf2 downstream anti-oxidative genes in EPCs. Data are shown as mean ± SD. *P<0.05, **P<0.0l, (n=3 per group).
Figure 23. AGGF1 activates AKT-Fyn-Nrf2 signaling in EPCs. (A-B) Western blot analysis showing the effect of siRNA for A KT (siAKT) (A) and wortmannin (B) on AGGF1- activated phosphorylation of AKT and nuclear accumulation of Fyn (n-Fyn). (C-E) Western blot analysis showing the effect of siAKT (C), wortmannin (D) and siRNA for Nrf2 (siNrf2) (E) on AGGF1 -induced increases of n-Nrf2 and its downstream signaling molecules. Data are shown as mean ± SD. *P<0.05, **P<0.0l, NS, not significant (n=3 per group).
Figure 24. Knockdown of AKT expression, wortmannin treatment, and knockdown of Nrf2 expression attenuate the protective effects of AGGF1 on EPCs. (A) Western blot analysis for AGGF1 in EPCs. (B-E) Effects of AKT siRNA on AGGF1 -mediated rescue of HG-impaired cell proliferation (B), tube formation (C), transendothelial migration (D), and cell migration (E) by EPCs. (F) Western blot analysis for AGGF1 in EPCs. (G-J) Effects of wortmannin on AGGF1 -mediated rescue of HG-impaired cell proliferation (G), tube formation (H), transendothelial migration (I), and cell migration (J) by EPCs. (K) Western blot analysis for AGGF1 in EPCs. (L-O) Effects of Nrf2 siRNA on AGGF1 -mediated rescue of HG-impaired cell proliferation (L), tube formation (M), transendothelial migration (N), and cell migration (O) by EPCs. Data are shown as mean ± SD. *P<0.05, **R<0.01, (n=6 mice per group).
Figure 25. Isolation and characterization of bone marrow derived EPCs from mice. Bone marrow mononuclear cells (MNCs) were washed out from the femurs and tibias of mice and isolated by density gradient centrifugation with histopaque-l083. MNCs were plated on gelatin-coated culture dishes and maintained in endothelial growth factor- supplemented media (EGM-2 bullet kit). Ten days after maintenance in endothelial-specific media, non-adherent MNCs were removed and the remaining adherent cells were subjected to immunostaining to verify their EPC identity. (A) Morphology of MNCs on day 4 and day 8 after plating on gelatin-coated dishes. Scale bar=50 pm. (B) Representative images from assays for uptake of Dil-acLDL (red fluorescence) and FITC-UEA-l (green fluorescence).
The isolated EPCs were Dil-acLDL and FITC-UEA-l positive. Blue, staining of the nuclei by Hoechst. Scale bar=50 pm (upper) , Scale bar=l00 pm (lower) . Three independent experiments were performed. (C) MNCs were cultured in the bullet kit medium in gelatin- coated cell culture dishes. Ten days after plating, adherent cells were further analyzed using flow cytometry with the cell surface antigen markers including antibodies for CD31 , CD34, CD45 and CD 144. Isotype IgG was used as a control antibody. Six mice per group
(n=6/group) were studied each time.
Figure 26. Aggfl haploinsufficiency significantly reduces the number of bone marrow mononuclear cells (MNCs). MNCs were washed out from the femurs and tibias of WT and Aggfl+/ KO mice and purified by density gradient centrifugation with histopaque-l083. MNCs were plated into gelatin-coated culture dishes and maintained in endothelial growth factor-supplemented media (EGM-2 bullet kit for ten days). Non-adherent MNCs were removed and the remaining adherent cells were subjected to immunostaining to verify their EPC identity. (A) Representative images from assays for uptake of Dil-Ac-LDL (red fluorescence) and FITC-UEA-l (green fluorescence) are shown in (A). The isolated EPCs were Dil-ALDL- and FITC-UEA-l positive. Blue, staining of the nuclei by Hoechst. (B)
The number of the Dil-acLDL/FITC-UEA-l (lectin) positive cells was plotted and compared between WT and Aggfl +/ KO mice. Scale bar=l00 pm. Three independent experiments were performed. Six mice per group (n=6/group) were studied each time. Data are shown as mean ± SD. *P<0.05.
Figure 27. The expression level of the AGGF1 protein is increased in EPCs isolated from db/db mice or mice treated with HFD. (A) Western blot analysis showing increased AGGF1 expression in isolated EPCs from db/db mice. (B) Western blot analysis showing increased AGGF1 expression in isolated EPCs from HFD-induced T2DM mice. GAPDH was used as loading control. Data are shown as mean ± SD. **R<0.01, (n=6).
Figure 28. Increased glucose levels in mice fed with a high fat diet (HFD). C57BL/6J mice were fed with a high fat diet alone to induce T2DM. Mice fed with a chow diet were sued as controls. After 16 weeks, blood glucose levels were measured using a glucometer, and then a hind-limb ischemia model was established with the mice. Three independent experiments (A, B, and C) were performed. Data are shown as mean ± S.D. **R<0.01 (n=6).
Figure 29. Analysis of the expression levels of CXCR4 (Fig. 29a), VEGF (Fig. 29b) and VEGFR2 (Fig. 29c) mRNA in EPCs from different groups of mice. Bone marrow-
derived EPCs were isolated and used for real-time RT-PCR analysis. Data are shown as mean ± SD. NS not significant, (n=3).
Figure 30. Aggfl is required for essential functions of EPCs in capillary tube formation, proliferation, transendothelial migration, and migration as in a HFD-induced diabetic mouse model. EPCs were isolated from bone marrow of wild-type (WT) and heterozygous Aggfl +/ knockout (KO) mice fed with and without a high glucose diet (HFD), and characterized. (A) Aggfl haploinsufficiency inhibits angiogensis mediated by EPCs. Angiogenic function of EPCs was assessed by a matrigel-based capillary tube formation. (B) Images from (A) were analyzed, quantified and plotted. (C) Aggfl haploinsufficiency inhibits EPC proliferation. Proliferation of EPCs was examined using the CCK-8 kit. (D) Aggfl haploinsufficiency inhibits transendothelial migration of EPCs in transwell assays. HUVECs (about lxlO4 cells per well) were cultured in the upper chamber of a 24-transwell insert (8.0-pm pores). EPCs were harvested, resuspended in basal culture medium (EBM-2, 0.5% BSA), and added to the upper chamber of the transwell plate (0.2 ml). Images were captured 24 hours after EPCs plating with an inverted Nikon Eclipse Ti microscope. (E) Images from (D) were quantified and plotted. (F) Aggfl haploinsufficiency inhibits EPC migration in wound scratch migration assays. EPCs monolayers in 6-well cell culture plates were wounded with a scratch by a 200 pl pipette tip and examined 24 hours after wounding. Images were captured with an inverted Nikon Eclipse Ti microscope. (G) Representative images from EPC migration assays. Data are shown as mean ± SD. *P<0.05, **P<0.0l, ***R<0.001 (n=6 mice per group).
Figure 31. Analysis of the expression levels of CXCR4 (Fig. 3la), VEGF (Fig. 3lb) and VEGFR2 (Fig. 3lc) mRNA in EPCs from mice treated with the AGGF1 protein and high glucose. EPCs were isolated, and used for real-time RT-PCR analysis. Data are shown as mean ± SD. NS not significant, (n=3).
Figure 32. AGGF1 protein treatment robustly potentiates the therapeutic effects of EPCs on angiogenesis and homing in a hindlimb ischemia model in db/db mice. (A) Representative images for H&E staining and immunostaining for CD31. Scale bar=l00 pm. (B) Representative images for immunostaining with an anti-GFP antibody. EPCs were infected with EGFP lentivirus, and transplanted into db/db mice with diabetic hind-limb ischemia. Scale bar=200 pm. (n=l2 mice per group).
Figure 33. AGGF1 protein treatment robustly potentiates the therapeutic effects of EPCs on peripheral vascular complications in a hindlimb ischemia model. A hindlimb ischemia model was created in mice. Two days after the surgery, the mice were injected with
EPCs, AGGFl-pretrearted EPCs (~lxl06), and control PBS via the tail vein. The therapeutic effects were evaluated using a Vevo 2100 High- Resolution Micro-Ultrasound System before the ischemic surgery and 7, 14 and 28 days after ischemia. (A) Transplantation of AGGF1- pretreated EPCs dramatically improved blood perfusion compared with EPCs without AGGF1 pretreatment. (B) Therapeutic effects of AGGF1 -pretreated EPCs on necrosis compared with EPCs without AGGF1 pretreatment. (C) Therapeutic effects of AGGF1- pretreated EPCs on ambulatory impairment compared with EPCs without AGGF1 pretreatment. (D) Effects of AGGF1 -pretreated EPCs on the density of CD3l-postive capillary vessels compared with EPCs without AGGF1 pretreatment. Capillary density was measured in HFD-fed HLI mice. The gastrocnemius muscle from HLI mice were fixed in 4% paraformaldehyde for 24 hours and used for immunostaining analysis with an anti-CD31 antibody. (E) Representative images for H&E staining and immunostaining for CD31. Scale bar=l00 pm. (F) Representative images for immunostaining with an anti-GFP antibody. EPCs were infected with EGFP lentivirus, and transplanted into mice with diabetic hind-limb ischemia. Four weeks after transplantation, the gastrocnemius muscle were fixed in 4% paraformaldehyde for 24 hours and then subjected to immunostaining analysis with an anti- EGFP antibody to determine whether EPCs were homed into the ischemic areas. Scale bar=l00 pm. Data are shown as mean ± SD. *P<0.05, **R<0.01 (n=l2 mice per group).
Figure 34. EPCs were infected lentiviruses with GFP expression as an easily identifiable marker. The infection efficiency and the level of GFP expression were evaluated 72 h after infection. Scale bar=20 pm. Three independent experiments were performed and representative images were shown. Images were captured at the magnification of 40x.
Figure 35. AGGF1 inhibits HG-induced superoxide production by EPCs.
Dichlorodihydrofluorescein diacetate (DCFH-DA, sigma) was used to detect superoxide in EPCs with flow cytometry (Beckman CytoFLEX). Fluorescence intensity was recorded, analyzed and plotted. Data are shown as mean ± SD. *P<0.05, **P<0.0l, (n=6 per group).
Figure 36. AGGF1 increases the level of nuclear Nrf2 in EPCs. EPCs were treated with a high glucose solution (HG) in the presence or absence of purified AGGF1 protein for 12 hours, and used for immunofluorescent staining with an anti-Nrf2 antibody to analyze nuclear translocation of Nrf2. Scale bar=200 pm.
Figure 37. AGGF1 does not affect the expression levels of Nrf2 and KEAP-l in EPCs. EPCs were cultured in media with or without high glucose (HG, 30 mM) as well as a combination of high glucose and AGGF1 protein. (A) Total RNA was extracted with Trizol reagent and used for real-time RT-PCR analysis for the level of Nrf2 mRNA. (B) Western
blot analysis with an anti-Nrf2 antibody. (C) Western blot analysis using an anti-KEAP-l antibody. GAPDH was used as loading control. Three independent experiments were performed. Data are shown as mean ± S.D, (n=6).
Figure 38. Western blot analysis for nuclear accumulation of nNrf2 in EPCs treated with Aggfl siRNA and Fyn siRNA under the normal condition (Control) and under a high glucose condition (HG). GAPDH was used as loading control.
Figure 39. A schematic diagram showing the hypothetical molecular signaling pathway by which AGGF1 protein therapy potentiates the therapeutic effects of EPCs on diabetes-induced vascular complications. It is noted that the present invention is not limited to this particular mechanism, and an understanding of the mechanism is not necessary to practice the inventin. AGGF1 activates AKT signaling, which inhibits Fyn-mediated export and degradation of nuclear Nrf2. Nrf2 binds to the promoter and regulatory region and activate the transcription of CAT, H(2-l, and NQO-1, which function as antioxidants to block generation of ROS, reducing oxidative stress and increasing function of EPCs. Overall, AGGF1 functions as an antioxidant regulator to protect the function of EPCs, and becomes a potential therapeutic target for improving the ischemia-reparative capacity of EPCs transplantation in DM.
Figure 40 shows the duration of the glucose-lowering effect of injection of a single dose of AGGF1 protein is more than 10 times longer than insulin in db/db diabetic mice.
(A) Experimental design of the 48-hour AGGF1 treatment assay in Example 4. The time points for AGGF1 injection, glucose measurement, fasting period, and eating period are shown. (B) Therapeutic effects of the AGGF1 protein on blood glucose levels in db/db mice compared to IgG control after subcutaneous injection of IgG or AGGF1 (3 nmole/g body weight) over a time period of 48 hours. *P<0.05, **P<0.0l, n=4. (C) Therapeutic effects of the AGGF1 protein on blood glucose levels in db/db mice compared to IgG control after subcutaneous injection of insulin (0.75 IU/kg body weight). *P<0.05, **R<0.01, n=4.
DEFINITIONS
To facilitate an understanding of the present technology, a number of terms and phrases are defined below. Additional definitions are set forth throughout the detailed description.
Throughout the specification and claims, the following terms take the meanings explicitly associated herein, unless the context clearly dictates otherwise. The phrase“in
some embodiments” as used herein does not necessarily refer to the same embodiment, though it may. Furthermore, the phrase“in another embodiment” as used herein does not necessarily refer to a different embodiment, although it may. Thus, as described below, various embodiments of the technology may be readily combined, without departing from the scope or spirit of the technology.
In addition, as used herein, the term“or” is an inclusive“or” operator and is equivalent to the term“and/or” unless the context clearly dictates otherwise. In addition, throughout the specification, the meaning of“a”,“an”, and“the” include plural references. The meaning of“in” includes“in” and“on.”
As used herein, the terms“subject” and“patient” refer to any animal, such as a mammal like a dog, cat, bird, livestock, and preferably a human (e.g., a human with a disease such as diabetes, cardiovascular disease, or related conditions).
As used herein, the term“administration” refers to the act of giving a drug, prodrug, or other agent, or therapeutic treatment to a subject. Exemplary routes of administration to the human body can be through the mouth (oral), skin (transdermal, topical), nose (nasal), lungs (inhalant), oral mucosa (buccal), inhaled, an implanted pump activation, by injection (e.g., intravenously, subcutaneously, intratumorally, intraocular, intraperitoneally, etc.), and the like.
DETAILED DESCRIPTION
Provided herein are compositions, systems, kits, and methods for treating a subject with a disease or condition characterized by abnormal glucose metabolism (e.g., diabetes) or ischemic vascular disease (e.g., PAD or CAD) by administering a composition comprising AGGF1 peptides, or an expression vector encoding said peptides, or AGGF1 -primed endothelial progenitor cells, to a subject. In certain embodiments, the AGGF1 peptide is full- length AGGF1, or a biologically active fragment or mutant thereof. In some embodiments, the AGGF1 peptides or expression vectors are administered around the same time insulin is administered to the subject.
Similar to insulin, recombinant AGGF1 protein and its derivatives (e.g. short AGGF1 peptides or mutants with biological activity) can effectively lower glucose levels. However, different from insulin, therapies with AGGF1 and AGGF1 derivatives have a longer therapeutic effect, and do not cause hypoglycemia. Compared to typical insulin treatment,
which has a fast-acting effect in 30 minutes but lasts about 3 hours, work conducted during development of embodiments herein demonstrated AGGF1 treatment has a much longer therapeutic effect with a peak effect at 18-24 hours and lasting for >42 hours.
In certain embodiments, AGGF1 peptides (or expression vector encoding AGGF1) and insulin are administered at about the same time (e.g., within 1-25 minutes of each other). Combined treatment with both insulin and AGGF1 can quickly decrease the fasting glucose level within 30 minutes, but also significantly extend the effect of insulin. Therefore, the therapy with combined insulin and AGGF1 can have a sustained lowering effect on glucose levels for a much longer time than insulin alone. For insulin treatment, patients typically require insulin injections 3 times per day. Even the long-acting insulin such as insulin glargine (Lantus, Sanofi) requires injection once a day. In certain embodiments, the combined AGGF1 and insulin treatment reduces the frequency of injection to once per >2 days. The various embodiments described herein can enable more patients with type 1 and type 2 diabetes mellitus to reach better glucose targets by reducing the frequency of injections of drugs and enhancing medication compliance, and maintain a better quality of life.
AGGF1 is an angiogenic protein that has been proposed previously for use in treating cardiovascular diseases. In work conducted during the development of embodiments herein, it was shown that AGGF1 has activities similar to insulin. In AGGF1 KO mice, fasting glucose levels were significantly elevated when mice were fed a high fat diet and were insulin resistant. In diabetic mice, AGGF1 administration significantly lowered fasting glucose levels. In streptozotocin mice, the effects of AGGF1 seemed to depend on and potentiate insulins activities.
Benefits of AGGF1 therapy (e.g., protein or expression vector encoding protein) include, but are not limited to, AGGF1 peptides regulate glucose levels more precisely than insulin, no evidence of hypoglycemia, longer therapeutic window, peak glucose lowering at 18-24 hours, lasting for >42 hrs. In combination with insulin, AGGF1 peptide and expression vectors encoding AGGF1 , can be used to better control glucose levels over extended periods of time compared with insulin alone.
The AGGF1 gene encodes a 714 amino acid angiogenic factor essential for embryonic development. As described in the Example below, it was found that heterozygous AGGF1 knockout (KO) mice develop diabetes with an increased fasting glucose level, impaired glucose tolerance, and insulin resistance, and that AGGF1 protein therapy dramatically
reduces the fasting glucose level in high fat diet (HFD)-induced diabetic mice. Compared to typical insulin treatment, which has a fast-acting effect in 30 minutes but lasts about 3 hours, AGGF1 treatment has a much longer therapeutic effect with a peak effect at 18-24 hours and lasting for >42 hours. It was found that AGGF1 protein therapy of one injection per two days maintained the low glucose level constantly. In certain embodiments, where expression vectors are administered to a subject, long term expression can allow a subject to receive a single administration without further treatment of for a longer period of time (e.g., 2 months). In certain embodiments, diabetic patients are treated with AGGF1 therapy who also have coronary artery disease, at risk for heart attack, or heart failure, and receive a benefit for both conditions.
In certain embodiments, the compositions according to the present disclosure (e.g., with an AGGF1 peptide or expression vector) comprises or consists of a pharmaceutically acceptable carrier, diluent, or excipient (including combinations thereof). Acceptable carriers or diluents for therapeutic use are well known in the pharmaceutical art and are described, for example, in Remington’s Pharmaceutical Sciences, Mack Publishing Co. (A. R. Gennaro edit. 1985). The choice of pharmaceutical carrier, excipient, or diluent is selected with regard to the intended route of administration and standard pharmaceutical practice. The
pharmaceutical comprise as, or in addition to, the carrier, excipient, or diluent any suitable binder(s), lubricant(s), suspending agent(s), coating agent(s), and/or solubilizing agent(s).
This pharmaceutical composition will desirably be provided in a sterile form. It may be provided in unit dosage form and will generally be provided in a sealed container. A plurality of unit dosage forms may be provided.
In certain embodiments, the composition inside of pumps that are surgical embedded within a patient, such that the AGGF1 peptide or expression vector can be injected (e.g., in a manner similar to insulin) to the subject’s blood stream when needed (see, e.g., Shah et ak, Int J Pharm Investig. 2016 Jan-Mar; 6(1): 1-9, herein incorporated by reference).
Pharmaceutical compositions within the scope of the present technology may include one or more of the following: preserving agents, solubilizing agents, stabilizing agents, wetting agents, emulsifiers, sweeteners, colorants, flavoring agents, odorants, and/or salts. Compounds of the present technology may themselves be provided in the form of a pharmaceutically acceptable salt. In addition, embodiments may comprise buffers, coating
agents, antioxidants, suspending agents, adjuvants, excipients, and/or diluents. Examples of preservatives include sodium benzoate, sorbic acid, and esters of p-hydroxybenzoic acid.
They may also contain other therapeutically active agents in addition to compounds of the present technology (e.g., insulin). Where two or more therapeutic agents are used they may be administered separately (e.g., at different times and/or via different routes) and therefore do not always need to be present in a single composition. Thus, combination therapy is within the scope of the present technology.
In certain embodiments, the compositions herein contain an expression vector encoding the AGGF1 peptide. In certain embodiments, the expression vectors is a plasmid, an adenovirus-associated viruses, or other viruses. In certain embodiments, the expression vectors are delivered by nano-technologies and/or targeted delivery systems.
The routes for administration (delivery) include, but are not limited to, one or more of: oral (e.g. as a tablet, capsule, or as an ingestable solution), implantable pump, topical, mucosal (e.g. as a nasal spray or aerosol for inhalation), nasal, parenteral (e.g. by an injectable form), gastrointestinal, intraspinal, intraperitoneal, intramuscular, intravenous, intrauterine, intraocular, intradermal, intracranial, intratracheal, intravaginal,
intracerebroventricular, intracerebral, subcutaneous, ophthalmic (including intravitreal or intracameral), transdermal, rectal, buccal, via the penis, vaginal, epidural, sublingual. It is to be understood that not all of the agent need be administered by the same route. Likewise, if the composition comprises more than one active component, then those components may be administered by different routes.
If the AGGF1 peptides and expression vectors described herein is administered parenterally, then examples of such administration include one or more of: intravenously, intra-arterially, intraperitoneally, intrathecally, intraventricularly, intraurethrally,
intrastemally, intracranially, intramuscularly, or subcutaneously administering the agent; and/or by using infusion techniques.
In some embodiments, pharmaceutical compositions adapted for oral administration are provided as capsules or tablets; as powders or granules; as solutions, food product, syrups or suspensions (in aqueous or non-aqueous liquids); as edible foams or whips; or as emulsions. Tablets or hard gelatin capsules may comprise lactose, maize starch or derivatives thereof, stearic acid or salts thereof. Soft gelatin capsules may comprise vegetable oils, waxes, fats, semi-solid, or liquid polyols etc. Solutions and syrups may comprise water,
polyols and sugars. For the preparation of suspensions, oils (e.g., vegetable oils) may be used to provide oil-in-water or water-in-oil suspensions. An active agent intended for oral administration may be coated with or admixed with a material that delays disintegration and/or absorption of the active agent in the gastrointestinal tract (e.g., glyceryl monostearate or glyceryl distearate may be used). Thus, the sustained release of an active agent may be achieved over many hours and, if necessary, the active agent can be protected from being degraded within the stomach.
In certain embodiments, the AGGF1 peptides described herein may be applied topically in the form of a gel, hydrogel, lotion, solution, cream, ointment or dusting powder. The agent of the present technology may also be dermally or transdermally administered, for example, by the use of a skin patch. For application topically to the skin, the agent of the present technology can be formulated as a suitable ointment containing the active compound suspended or dissolved in, for example, a mixture with one or more of the following: mineral oil, liquid petrolatum, white petrolatum, propylene glycol, polyoxyethylene
polyoxypropylene compound, emulsifying wax and water. Alternatively, it can be formulated as a suitable lotion or cream, suspended or dissolved in, for example, a mixture of one or more of the following: mineral oil, sorbitan monostearate, a polyethylene glycol, liquid paraffin, polysorbate 60, cetyl esters wax, cetearyl alcohol, 2-octyldodecanol, benzyl alcohol and water. If the AGGF1 peptides are administered parenterally, then examples of such administration include one or more of: intravenously, intra-arterially, intraperitoneally, intrathecally, intraventricularly, intraurethrally, intrasternally, intracranially, intramuscularly or subcutaneously administering the agent; and/or by using infusion techniques.
Typically, a physician will determine the actual dosage of the AGGF1 peptides or expression vector which will be most suitable for an individual subject. The specific dose level and frequency of dosage for any particular patient may be varied and will depend upon a variety of factors including the activity of the specific compound employed; the metabolic stability and length of action of that compound; the age, body weight, general health, sex, diet, mode and time of administration; rate of excretion; drug combination; the severity of the particular condition; and the individual undergoing therapy. In some embodiments, the dosage is between 0.1 and 5 mg per kilogram of the subject (e.g., 0.1 ... 0.8 ... 1.5 ... 2.5 ... 4.0 ... 5.0 mg per kg). The agent and/or the pharmaceutical composition of the present technology may be administered in accordance with a regimen of from 1 to 10 times per day, such as once or twice per day, or only once every other day or every third day. For oral and
parenteral administration to human patients, the daily dosage level of the agent may be in single or divided doses.
EXAMPLES
EXAMPLE 1
AGGF1 may be regarded as the“the 2nd insulin” for treatment of diabetes. Compared to typical insulin treatment, which has a fast-acting effect in 30 minutes but lasts about 3 hours, AGGF1 treatment, in this Example, has a longer therapeutic effect with a peak effect at 18-24 hours and lasting for >42 hours. AAV-AGGF1 gene therapy can make its therapeutic effect last even for months.
Increased fasting glucose levels in heterozygous AGGF1+/ KO mice.
In order to assess the effect of AGGF1 on diabetes, we characterized AGGF1 knockout mice developed by our laboratory10 13. Because homozygous AGGFl_/ KO mice are embryonically lethal13, we studied heterozygous AGGFl+/ KO mice. We found no significant differences in body weight and food intake between AGGFl /_ KO mice and their littermate WT control mice fed a standard chow diet for up to 60 weeks (P>0.05, n=8/group) (Fig. 1 A) or a high fat diet (HFD) (P>0.05, n=8/group) (Fig. 1 A). Interestingly, when we measured the fasting glucose levels, we found that AGGFl+/_ KO mice showed a significantly higher level of fasting blood glucose than littermate WT mice fed HFD (P<0.05, n=8/group) (Fig. 1B). The fasting insulin level was also higher in AGGFl /_ KO mice than in littermate WT mice fed HFD (P<0.05, n=8/group) (Fig. 1C).
Impaired glucose tolerance in heterozygous AGGF1+/ KO mice.
We performed the glucose tolerance test (GTT). As shown in Fig. 1D, the GTT revealed significantly impaired glucose tolerance in AGGFl_/ KO mice compared with littermate WT control mice (P<0.05, n=8/group).
Insulin resistance in heterozygous AGGF1+/ KO mice.
We performed insulin tolerance tests (ITT). As shown in Fig. 1E, AGGFl_/ KO mice showed characteristics of insulin resistance compared with littermate WT mice (P<0.05, n= 8/group).
AGGF1 protein therapy dramatically lowers fasting glucose levels in diabetic mice.
Because AGGFl /_ KO mice developed diabetes, we hypothesized that intraperitoneal injection of AGGF1 protein may inhibit diabetes. To test the hypothesis, AGGF1 protein (0.25 mg/kg body weight) was injected into HFD-induced hyperglycemic mice. Surprisingly, injection of AGGF1 protein dramatically reduced the fasting glucose level in HFD hyperglycemic mice compared with control PBS buffer (Fig. 2). The maximal glucose lowering effect of AGGF1 was achieved at 18-24 hours after injection, and the sustained effect can be observed 42 hours after the injection (Fig. 2). Chronic AGGF1 treatment of HFD-induced hyperglycemic mice every 2 days for 36 days maintained the significantly low fasting glucose level for the entire treatment period (Fig. 3).
Effect of combined treatment with insulin and AGGF1.
HFD-induced hyperglycemic mice were administered with insulin alone (1 ug/kg body weight) or combination of insulin (1 ug/kg body weight) and AGGF1 protein (0.25 mg/kg body weight) by intraperitoneal injection. Insulin treatment quickly decreased the fasting glucose level within 30 minutes and the effect of insulin was sustained for only 150 minutes. AGGF1 protein treatment significantly extended the effect of insulin (Fig. 4A). These data suggest that the therapy with combined insulin and AGGF1 can have a sustained lowering effect on glucose levels for a much longer time than insulin alone.
Interestingly, we found that AGGF1 protein therapy is dependent on insulin. In a diabetic mouse model induced by streptozotocin (STZ, killing b-cells), AGGF1 protein therapy failed to lower blood glucose levels (Fig. 4B). Moreover, the glucose-lowering effect of AGGF1 was severely blunted when insulin secretion was inhibited by somatostatin (Fig. 4C). These data suggest that AGGF1 protein therapy lowers blood glucose levels in an insulin-dependent manner.
EXAMPLE 2
This Examples describes the identification of truncated AGGF1 peptides that function in a manner similar to the full length AGGF1 protein.
Identification of short, functional AGGF1 domain required for endothelial cell proliferation, migration, and angiogenesis between amino acids 574 and 624
In order to localize the functional domain of AGGF1, we created serial deletions of AGGF1 by systematic truncation of AGGF1 by 50 amino acids each time from the N- terminus or C-terminus (Figure 10). Successful creation of AGGF1 deletions are
demonstrated by coomassie-blue staining of N-terminal and C-terminal serial deletion mutant proteins on SDS-PAGE gels shown in Figure 11.
The interaction domain between AGGF1 and endothelial cells is localized between amino acids 574 and 624.
To identify the AGGF1 domain responsible for binding to endothelial cells, endothelial cell adhesion assays were performed with human umbilical vascular endothelial cells (HUVECs) for full-length AGGF1 (WT) and serial N-terminal deletion mutant proteins. Significantly stronger binding (i.e. high fluorescence) was observed with WT-AGGF1 and deletion mutant proteins Nl to N10 than deletion mutant proteins Nl 1 and N12 (Figure 12, left panel). These results suggest that the potential interaction domain of AGGF1 is between amino acid 574 and 624.
Endothelial cell adhesion assays were also performed with WT-AGGF1 and serial C- terminal deletion mutant proteins of AGGF1. Significantly higher binding (i.e. fluorescence) was observed with WT-AGGF1 and deletion mutants Cl and C2 than deletion mutants C3 to C13 (Figure 12, right panel). These results suggest that the potential interaction domain of AGGF1 with endothelial cells is between amino acid 564 and 614 (Figure 10). Together, these results narrow the AGGFl-endothelail cell interaction domain to a 40-amino acid region (amino acids 574-614) at the C-terminus of AGGF1.
The functional domain of AGGF1 responsible for endothelial cell migration is localized between amino acids 574 and 624.
The cellular mechanism of angiogenesis involves endothelial cell motility.
Endothelial cell migration assays were performed with WT-AGGF1 and serial N- and C- terminal deletion mutant AGGF1 proteins. Significantly more HUVECs migrated into the ‘wounded area’ after 24 hours in presence of WT-AGGF1 and deletion mutant proteins Nl to N10 than deletion mutant proteins Nll and N12 (Figure 13). Significantly more HUVECs migration was also observed with deletion mutant proteins Cl and C2 than deletion mutant
proteins C3 to C13 (Figure 13). These endothelial cell migration assays demonstrate that the 40 amino acid AGGF1 binding domain (amino acids 574-614) is also responsible for inducing endothelial cell migration.
The functional domain of AGGF1 responsible for angiogenesis is localized between amino acids 574 and 624.
Angiogenesis assays were performed using the capillary endothelial tube formation assays. A significantly higher number of‘enclosed circles’ (i.e. angiogenesis) were formed by HUVECs after 16 hours in presence of WT-AGGF1 and deletion mutant proteins Nl to N10 than deletion mutant proteins Nll and N12 (Figure 14). Similarly, a significantly higher number of‘enclosed circles’ were formed by HUVECs in presence of WT-AGGF1 and deletion mutant proteins Cl and C2 than deletion mutant proteins C3 to C13 (Figure 14). These endothelial tube formation assays demonstrate that the 40 amino acid AGGF1 binding domain (amino acids 574-614) is responsible for angiogenesis.
A short mutant AGGF1 protein with amino acids 604-714 has full functions of full- length AGGF1 in endothelial cell proliferation, migration, and angiogenesis
To further narrow down the amino acid residues involved in AGGF1 functions, we created four more serial N-terminal deletions, each truncating AGGF1 by 10 amino acids between amino acids 574 and 624, referred to as N101, N102, N103, and N104 (Figure 15A)). Endothelial cell adhesion assays showed that WT-AGGF1 and deletion mutant proteins N101 to N103 can bind to endothelial cells, but not deletion N104 (Figure 15B). Similar findings were found for HUVEC migration (Figure 15C).
For endothelial tube formation (angiogenesis) assays (Figure 15D), WT-AGGF1 and deletion mutant proteins N101 to N 103 showed a significantly increased angiogenesis than deletion N104 and control BSA. N102 appeared to present with a stronger angiogenic activity than N103 (Figure 15D).
We also analyzed the WT and deletion mutant AGGF1 for their function in activation of the AKT signaling pathway involved in angiogenesis and insulin signaling. Compared to control BSA, a significantly higher phosphorylation level of AKT (Ser473) was shown in
presence of WT-AGGF1. Similarly, a high phosphorylation level of AKT (Ser473) was shown with deletion mutant proteins N101 to N103, but a significantly less phosphorylation level of AKT (Ser473) was shown with deletion mutant protein N104 (Figure 15E).
In summary, by analyzing serial AGGF1 deletions, we have identified a short mutant form of AGGF1 spanning amino acids 604-714 (N103), which retains the full function of the full-length AGGF1 protein. This mutant AGGF1 is referred to as AGGFI604-714. Because AGGFl604-7i4has full AGGF1 functions, we can utilize it for treatment of, for example, cardiac and vascular diseases, and the other diseases listed herein. Because AGGFl604-7i4has only 110 amino acid residues, it can be synthesized in vitro and used for clinical treatment of cardiac and vascular diseases. AGGFl604-7i4 can be further optimized by many different modifications to increase its stability and functional activities in treatment of diseases. For example, AGGF U04-714 can be further modified by removing more amino acids to identify the shortest form of AGGF1, but with full activities, which can be used for treatment of diseases (e.g., using the assays and procedures described in this Example). AGGFl604-7i4 can be further modified by adding some amino acids to increase its stability and activities.
AGGFl604-7i4 can be further modified by adding some amino acids (e.g. mimotope) or by conjugating carriers for its targeted delivery to specific organs, tissues and cells for treatment of specific diseases. AGGFI604-714 can be further modified by synthesizing as a circular form (cyclotide) to increase its stability for disease treatment.
Utilization of recombinant AGGF1 and AGGF1 derivatives to treat PAH
Pulmonary arterial hypertension (PAH) is characterized by an increased mean pulmonary arterial pressure (mPAP) of >25 mmHg, which is caused by functional and structural remodeling of pulmonary arteries, and in turn increases the burden on the right ventricle and eventually leads to right ventricular failure and premature death5’ 6. The pathogenesis of PAH is complex and involves pulmonary endothelial dysfunction, proliferation of vascular smooth muscle cells (VSMCs), inflammation, fibrosis, as well as abnormal thickening and contraction of pulmonary arteries, thrombus formation, and genetic abnormalities, leading to incremental increases of pulmonary vascular resistance, increased right ventricular loading, and eventual right ventricular failure and death.7 11 Accurate and early diagnosis of PAH is seriously lacking because PAH symptoms appear late. Hence, the best and early treatment for patients is delayed, resulting in a low survival rate.5, 6 PAH
remains a fatal disease with 31-45% of patients dying within 3 years even with the best treatments.12 18 Moreover, current treatments cannot reverse vascular remodeling.8’ 9
AGGF1 deficiency causes PAH:
We recorded the pulmonary artery pressure (PAP) from Aggfl+/_ (Aggfl /_ is embryonically lethal) and wild type (WT) mice (Aggfl+/+ littermates) at the age of 16-20 weeks (male mice, n=6/group) (Figure 16A). It was striking that mPAP in WT mice (16.4+/- 1.20 mmHg) falls within the normal range of 9-18 mmHg in humans, whereas mPAP in Aggfl+/ KO mice was significantly increased to 27.3+/-1.90 mmHg (P=0.007, n=6/group), which is higher than the threshold for the diagnosis of PAH in humans (Figure 16B). We conclude that Aggfl+/ KO mice develop spontaneous PAH. Compared with wild type mice, Aggfl+/ KO mice showed significantly higher total pulmonary resistance (TPR) (1.24+/-0.17 mmHg/mL/min for KO mice vs. 0.72+/-0.10 for WT, P=0.001 , n=6/group) (Figure 16C). Aggfl+/ KO mice showed right ventricular (RV) hypertrophy because their RV/(LV+S) weight is significantly higher than that from WT mice (32.8+/-1.8 for KO vs. 26.1+/-1.30 for WT, P=0.003, n=6/group) (Figure 16D). All of these findings are important features of PAH. We therefore conclude that Aggfl+/_ KO mice develop spontaneous PAH.
We analyzed vascular remodeling associated with PAH in Aggfl+/ KO mice. The pulmonary arteries were isolated from Aggfl+/_ KO and WT mice (16-20 weeks, male mice, n=6/group), and stained with H&E. H&E staining showed that the pulmonary artery from Aggfl+/_ KO mice had a significantly increased medial wall thickness compared to WT mice (KO 42.6%+/-2.l vs. WT 3l.7%+/-l.4, P=0.002, n=6/group) (Figure 17A-B). The pulmonary arteries of Aggfl+/ KO mice had a significantly increased full muscularization (see Figure 17 description for method of quantitation: KO 21.6+/- 4.4 vs. WT 12.4+/- 2.5 %, P=0.002, n=6/group) and partial muscularization (KO 34.3+/- 7.9 vs. WT 20.4+/- 5.7 %, P=0.002, n=6/group) compared to that of WT mice (Figure 17C).
We analyzed cell proliferation using immunostaining with an anti-Ki67 antibody, which identifies Ki67-positive proliferating or dividing cells. The density of positive Ki67 cells was much higher in pulmonary arteries from Aggfl+/- KO mice than that from WT mice (6.87+/-1.15 % for KO vs. 3.21+/-0.62 for WT, P=0.008, n=6/group) (Figure 17D).
All of these data indicate that Aggfl+/_ KO mice develop spontaneous PAH and vascular remodeling that fully recapitulate human PAH pathology.
AGGF1 can block PAH:
Even with currently available therapies, the 3-year death rate for PAH patients is as high as 3l%-45%.12-18 Therefore, there is an urgent medical need to develop new or improved therapeutic agents to manage PAH patients. We found that AGGF1 protein therapy can block PAH. Systemic injection of recombinant AGGF1 protein significantly reduced right ventricular systolic pressure (RVSP) in a hypoxia-induced mouse model for PAH compared with control IgG (P=0.019) (Figure 18). AGGF1 protein therapy also reduced RV hypertrophy (RV/LV+S) compared with IgG control (Figure 18) although additional mice are needed to achieve statistical significance.
We have created a new mutant form of AGGF1, AGGFI604-714, which has the full activities of the full-length AGGF1, and can be utilized for treatment of PAH. The AGGF1 protein therapy for PAH has several advantages. First, because our study indicates that Aggf 1 KO and deregulation are directly related to the pathogenesis of PAH in mice and humans, AGGF1 treatment can be a specific therapy for PAH. On the other hand, other VSMC proliferation blocking agents (proteasome inhibitors bortezomib BTZ and carfizomib CFZ) have off-target effects (causing cardiac apoptosis in RV and LV).19 Existing therapy for PAH such as calcium channel blockers causes hypotension, but AGGF1 treatment does not affect blood pressure under a normal physiological condition.
References cited for this Example:
1. Lu et al„ PLoS Biol. 20l6;l4:el002529.
2. Zhang et a , Hum Mol Genet. 2016;25:5094-5110.
3. Yao et ak, Nat Commun. 20l7;8:l33.
4. Yao et ak, J Am Heart Assoc. 20l7;6:e005889.
5. Galie et ak, Eur Heart J. 2016;37:67-119.
6. Galie et ak; 2015 ESC/ERS Guidelines for the Diagnosis and Treatment of Pulmonary Hypertension. Rev Esp Cardiol (Engl Ed). 20l6;69:l77.
7. Bazan IS and Fares WH., Ther. Clin Risk Manag. 2015;11:1221-1233.
8. Malenfant et ak, Expert Rev Respir Med. 2013;7:43-55. 9. Malenfant et ak, Pulm Circ. 2013;3:278-293.
10. Nogueira-Ferreira et ak, Biochim Biophys Acta. 2014;1843:885-893.
11. Tuder et ak, J Am Coll Cardiol. 20l3;62:D4-l2.
12. Fox et ak, Curr Opin Anaesthesiol. 2008;21:467-472.
13. George et ak, Pulmonary hypertension surveillance: United States, 2001 to 2010. Chest. 2014;146:476-495.
14. Humbert et ak, Circulation. 2010;122:156-163.
15. McLaughlin et ak, J Am Coll Cardiol. 2015;65:1976-1997.
16. Prins et ak, Cardiol Clin. 2016;34:363-374.
17. Rich et ak, Ann Intern Med. 1986;105:499-502. 18. Thenappan et ak, Eur Respir J. 2010;35:1079-1087.
19. Wang et ak, Cardiovasc Res. 2016;110:188-199.
EXAMPLE 3
In Vivo Treatment with AGGFl-Primed EPCs Hyperglycemia-triggered vascular abnormalities are the most serious complications of diabetes mellitus (DM). The major cause of vascular dysfunction in DM is endothelial injury and dysfunction associated with the reduced number and dysfunction of endothelial progenitor cells (EPCs). However, the major regulators and underlying molecular mechanism are poorly characterized. In this Example, it is shown that EPCs from
heterozygous knockout (KO) Aggfl+/ mice showed impairment of proliferation, migration, angiogenesis, and transendothelial migration as in hyperglycemic mice fed with a high fat diet (HFD) or db/db mice. The number of EPCs from Aggfl +/ mice was significantly
reduced. Ex vivo, AGGF1 protein can fully reverse damaging effects of hyperglycemia on EPCs. In vivo transplantation of AGGF1 -primed EPCs successfully restores blood flow, and blocks tissue necrosis and ambulatory impairment in HFD-induced hyperglycemic mice or db/db mice with diabetic hind-limb ischemia. While not limited to any particular mechanism, it is believed that AGGF1 activates AKT and reduces nuclear localization of Fyn, which increases the nuclear level of Nrf2 and expression of anti-oxidative genes, and inhibits ROS generation. These results suggest that Aggfl is required for essential function of EPCs, AGGF1 reverses the damaging effects of hyperglycemia on EPCs, and AGGFl-priming of EPCs is a useful treatment modality for vascular complications in DM.
Diabetes mellitus (DM) is a chronic metabolic disorder, however, hyperglycemia- triggered vascular abnormalities, including cardiovascular disease and diabetic peripheral arterial disease (PAD), are the most serious complications, contributing to numerous deaths, diabetic foot wounds and lower-extremity amputation (1-8). The major causes of vascular dysfunction in DM are endothelial injury and dysfunctions associated with the reduced number and dysfunction of endothelial progenitor cells (EPCs) (9-12). EPCs are progenitors of endothelial cells (ECs) and have the potential to proliferate, migrate, home into the disrupted endothelium and differentiate into ECs to maintain endothelium integrity, restore endothelial dysfunction, promote neovascularization and repair damaged vessels (11; 13-15) . Cell-based therapy based on implantation of EPCs has emerged as a potential therapy for myocardial ischemia, brain ischemia and pulmonary embolism (16; 17). However, the efficacy of a therapy using EPC implantation for diabetic vascular complications is uncertain (18). In particular, autologous EPCs transplantation may not work efficiently because of the impaired function of such EPCs in DM by hyperglycemia (9; 11; 18).
DESIGN AND METHODS
Animals
Wild-type (WT) C57BL/6J mice and db/db mice (Jackson Lab) were used in the study. Aggfl+/- KO mice with exons 2-11 deleted were described previously (20). Because homozygous Aggfl-/- KO mice die before E8.5, heterozygous Aggfl+/- KO mice were studied. A high fat diet (HFD) -induced mouse model for type 2 diabetes mellitus (T2DM) was previously described (25-27). Male C57BL/6J mice fed with HFD or db/db mice were used to create a hind-limb ischemia model associated with T2DM by ligation of the femoral artery with 6/0 Ethilon sutures as described previously by us and others (28-30).
Isolation and Culture of Bone Marrow-Derived EPCs
EPCs were isolated from the bone marrow of WT, HFD-induced diabetic mice, and db/db mice, and cultured as described (11; 31). Mice were anesthetized with chloral hydrate (3%, m/v) via intraperitoneal injection. Bone marrow mononuclear cells (MNCs) were washed out from the femurs and tibias of mice with phosphate buffer saline (PBS), and then purified by density gradient centrifugation (2000 r/min) with histopaque-l083 (Sigma- Aldrich, St. Louis, MO) for 30 min at room temperature with a horizontal rotor centrifuge (Anke, LCJ-2B, Shanghai, China). The volume ratio of single cell suspension and histopaque-l083 separation liquid was 2:1. The cells were then washed twice using PBS, resuspended gently, plated in 0.1% (m/v) gelatin-coated cell culture dishes, and maintained in endothelial growth factor-supplemented media (EGM-2 bullet kit; Lonza, Switzerland) with 10% fetal bovine serum (FBS). Cells were cultured at 37°C with 5% C02 in a humidified water jacket incubator (Thermo Fisher Scientific, MA, USA). The EGM-2 medium was replaced every 3 days after first plating, and cellular morphology was monitored every day.
Characterization of Bone Marrow-Derived MNCs
Characterization of MNCs was performed as described (11; 31). Seven days after maintenance in endothelial-specific media and the removal of non-adherent MNCs, the remaining cells were subjected to immunostaining analysis. Cells were plated on gelatin- coated slide (ibidi) for one day, and then incubated with 10 pg/mL of acetylated Dil lipoprotein from human plasma (Dil-Ac-LDL, Thermo Fisher Scientific, MA, USA) at 37°C for 4 hours. The cells were washed 3 times with PBS, and incubated with 10 pg/mL of fluorescein isothiocyanate-labeled ulex europaeus lectin-l (Sigma-Aldrich, St. Louis, MO, USA) for 1 hour at 37°C. The cells were rinsed 3 times with PBS again, and visualized under a confocal microscopy.
MNCs were also characterized by flow cytometry analysis. Cells were incubated with 5% BSA (Sigma-Aldrich, St. Louis, MO) for 15 minutes for to block non-specific binding, and then stained with anti-mouse CD34-phycoerythrin (PE), CD31-PE, CD45-PE or CD144- PE (BD Biosciences, San Jose, CA, USA) at room temperature for 1 hour, respectively. The same fluorescein-labeled isotype IgG served as negative control. Cells were analyzed using a Beckman CytoFLEX, and data were analyzed using FlowJo software version 10 (TreeStar, Ashland, OR, USA).
Immunostaining for CD31
In brief, muscle samples were fixed overnight in paraformaldehyde (4%), embedded in paraffin and sectioned. Sections were deparaffinized, dehydrated and rehydrated after being sectioned. After microwave antigen retrieval, endogenous peroxidase blocking and normal goat serum blocking in equilibration buffer, sections were subject to
immunohistochemical analysis using a rabbit polyclonal antibody against CD31 (1:200, Proteintech). DAB (3, 30-diaminobenzidine) was used as a chromogenic substrate, and the sections were counterstained with hematoxylin. All ready sections were sealed with clear nail polish or neutral balsam (mounting medium). Images were photographed with an inverted Nikon Eclipse Ti microscope (Nikon USA, Melville, NY, USA) and further analyzed using the Image-Pro Plus version 6.0 software (Media Cybernetics Inc., Bethesda, MD, USA).
Cell Therapy with Implantation of EPCs Primed with and without AGGF1
Recombinant AGGF1 protein was purified as described previously (21; 33). EPCs isolated from mice were treated with purified AGGF1 protein (0.5 pg/ml) or negative control elution buffer for purification at 37°C for l2h. Two days after the diabetic hind-limb ischemia surgery, AGGFl-primed EPCs (-1x106) were injected into mice via the tail vein. Blood flow in both legs was measured in mice anesthetized with chloral hydrate (3%, m/v) using a Vevo 2100 High- Resolution Microultrasound System (Visualsonics Inc) immediately before the ischemic surgery and at time-points of 7, 14 and 28 days after ischemia. We measured the peak systolic velocity (Vs), the minimal end diastolic flow velocity (Vd), and the temporal average velocity per cardiac cycle (Va) in the femoral artery. The blood flow ratio was computed using the formula of (Vs-Vd)/Va using four continuous cardiac cycles (22; 34).
Production of Lentiviruses and Infection
Human embryonic kidney 293 cells (HEK293) were transfected with a lentiviral vector with EGFP, the packaging plasmid psPAX2 and the envelope plasmid pMD2.G using Lipofectamine 2000 (Invitrogen, Carlsbad, CA, USA). Viral supernatants were produced from the transfected HEK293 as described (11). HEK293 cells were maintained at 37°C in the high-glucose Dulbecco’s Modified Eagle’s medium (DMEM) supplemented with 10% (V/V) FBS (Gibico Life Technologies, New York, USA) in a humidified water jacket
incubator with 5% C02. The supernatant was harvested 72 hours after transfection, filtered through 0.45-pm polyvinylidene fluoride filters, and stored at -80°C until use.
EPCs were infected with the GFP-lentiviruses overnight at a multiplicity of infection of 50 with 2.5 pg/mL polybrene supplemented, and the culture medium was replaced with the fresh growth medium 24 hours after infection. After transfection for 72 hours, the infection efficiency was determined by GFP expression under a Nikon EclipseTi micorscopy (Nikon, Japan).
AGGF1 protein
The open reading frame of AGGF1 cDNA was cloned into pET-28b (Novagen) and then transformed into Escherichia coli BL21 (DE3) Star. 6xHis-AGGFl protein was overexpressed, purified and quality of purification was examined by SDS-PAGE with coomassie blue staining and Western blot analysis (35). siRNAs and inhibitors of PI3K
siRNAs for Nrf2 and AKT negative control siRNAs were chemically synthesized by Ribobio. The control siRNA is the silencer scrambled negative control siRNA (siNC) having no significant homology to any known gene sequences from the mouse genome. The sequences of siRNAs are as follows: Nrf2 sense: 5’-CGAGAAGUGUUUGACUUUATT-3’ (SEQ ID NO:l9), Nrf2 antisense: 5’-UAAAGUCAAACA-CUUCUCGTT-3’ (SEQ ID NO:20); AKT sense: 5'-UGCCCUUCUACAACCAGGATT-3' (SEQ ID NO:2l), AKT antisense: 5’-UCCUGGUUGUAGAAGGGCATT-3’ (SEQ ID NO:22); siNC sense: 5’- UUCUC-CGAACGUGUCACGUTT-3’ (SEQ ID NO:23), and siNC antisense: 5’- ACGU G AC AC- GUUCGG AG AATT- 3’ (SEQ ID NO:24). EPCs were transfected with siRNA in the Opti-MEM medium overnight using transfection reagent (Santa Cruz
Biotechnology, Dallas, TX) as described (36-38). The knockdown efficiency was determined by real time RT-PCR analysis described below. Wortmannin, inhibitors of PI3K was purchased from Selleck Chemicals (Houston, TX).
Tube Formation Assays
Fourteen days after maintenance in endothelial- specific media, adherent cells were subjected to tube formation assays. The matrigel basement membrane matrix (BD
Biosciences, San Jose, CA, USA) was thawed and pre-coated into 48-well plate wells with pre-chilled tips (100 pl/well). The plates were centrifuged at room temperature at 1500 rpm
for 30 minutes to make a flat surface, and incubated at 37 °C for another hour to make the matrix gel solidified. Purified AGGF1 protein (0.5 pg/mL) or D-glucose (30 mM, Sigma- Aldrich, St. Louis, MO, USA, control, L-glucose) was added into wells, and incubated at 37°C for 12 hours. Images were visualized and captured at 40 x magnification under an inverted Nikon Eclipse Ti microscope (Nikon, Japan). For quantification, the number of enclosed circles formed by the endothelial tube-like structures was counted in each field.
Cell Proliferation Assays
Cells were plated into 96-well plates for 48 hours and analyzed for proliferation using a CCK-8 kit according to the manufacturer’s instruction (Dojindo Laboratories, Japan).
EPCs were seeded into 96-well cell culture plates, and treated with AGGF1 (0.5 pg/mL) or a solution with high D-glucose (30 mM, Sigma, St. Louis, MO, control, L-glucose). Then, 10 pl/well of CCK-8 solution was added, and the plates were incubated for 1-4 hours in dark. Cell proliferation was determined by the measurement of absorbance at 450 nm using a Molecular Devices VERSA max microplate reader as described (39; 40).
Transendothelial Migration Assays
We evaluated transendothelial migration (TEM) capabilities of EPCs with a transwell assay as described (11). Human umbilical vein endothelial cells (HUVECs) (about lxlO4 cells per well) were cultured in the upper chamber of a 24-transwell insert (8.0 pm pores; BD Falcon, San Jose, CA, USA) to confluency. Monolayer confluency was confirmed under an inverted fluorescence microscopy before each experiment. EPCs were treated with AGGF1 vs. elution buffer, a high glucose solution vs. control, and siRNAs, and cultured for 24 hours. The EPCs were harvested and resuspended in the basal culture medium (EBM-2, 0.5% BSA). EPC suspension (~0.2 mL) was added to the upper chamber of Transwell, and 0.5 ml of EBM-2 medium was added to the lower chamber. Cells were cultured for 24 hours at 37°C, and EPCs that migrated from the upper chamber to the lower chamber of Transwell were visualized. Images were captured at 40x magnification under an inverted Nikon Eclipse Ti microscope (Nikon, Japan). The number of cells was counted and analyzed.
Scratch- Wound Cell Migration Assays
EPCs in cell culture dishes were trypsinized and resuspended in growth media and allowed to grow to a confluent monolayer in 6-well plates. EPCs were starved for 6 hours by replacing the regular growth media with serum-free growth media. EPC monolayers were
then wounded with a 200 mΐ pipette tip gently through the cell sheet to remove some cells and make a wound. The growth medium was aspirated out and wells washed twice with PBS to remove cell debris. Media supplemented with AGGF1 (0.5 pg/ml) were added to the wells in a total volume of 1 ml of serum-free growth media. Cells were cultured for 24 hours by incubating the plate at 37°C and 5% C02. Images were captured at 40X magnification under an inverted Nikon Eclipse Ti microscope (Nikon, Japan). The movement of cells into the scraped area was measured.
Western Blot Analysis
Protein extracts were prepared from cultured EPCs with Western-IP lysis buffer (Beyotime, Beijing, China) supplemented with a proteinase inhibitor cocktail (Roche, Basel, Switzerland), separated by 12% SDS-PAGE, and transferred to polyvinylidene-fluoride membranes. The membranes were blocked with skim milk and incubated with appropriate primary antibodies overnight at 4°C with gentle shaking. The membranes were then incubated with appropriate secondary antibodies for 2 hours at room temperature after standard TBST washing procedures. The primary antibodies against Nrf2 (1:1000 dilution), heme oxygenase-l (HQ-l, 1:2000), NAD(P)H dehydrogenase quinone 1 (NQO-l, 1:1000), catalase (1:1000), and GAPDH (1:5000) were purchased from ProteinTech (Wuhan, China). Antibodies for total protein kinase B or AKT (tAkt) and phosphorylated AKT (p-Akt,
Ser473; 1:1000) were purchased from Cell Signaling Technology (Danvers, MA, USA). Images from Western blot analysis were developed using a ChemiDoc XRS (Bio-Rad Laboratories, Richmond, CA, USA) with the SuperSignal West Pico Chemiluminescent Substrate (Pierce Chemical Co., Rockford, IL, USA), and further analyzed with a Gel-Pro analyzer. GAPDH was used as the loading control.
Quantitative Real-Time RT-PCR Analysis
Total RNA samples were isolated from EPCs using Trizol reagent (TaKaRa Biotech, Japan), and reversed transcribed into cDNA with M-MLV reverse transcriptase (Promega, Madison, wi) according to the manufacturer’s instruction. DNase I (Thermo Fisher
Scientific, MA, USA) was used to remove contaminating genomic DNA before reverse transcription. Quantitative real-time RT-PCR analysis was performed in triplicate with a 10 pl reaction system using the FastStart Universal SYBR Green Master (Roche, Basel, Switzerland) and a 7900 HT Fast Real-Time PCR System (ABI, US). The PCR profile was 94°C for 5 min, and 40 cycles of 94°C for 10 s and 60°C for 15 s. Fold differences between
samples were assessed with a comparative cycle time (Ct) method. GAPDH served as an internal standard. The data were analyzed using 2-AACt relative expression quantity as reported (41).
Statistical Analysis
Data are presented as mean +/- standard deviation (SD). The comparison of means of two groups was made by a Student’ s t test or a nonparametric Wilcoxon rank test when the sample size was small and/or the distribution was not normal. To compare the means of more than 2 groups, one-way ANOVA or the generalized linear regression approach was employed for data with normal distribution and the Kruskal-Wallis test for non- normal distribution data or small samples. P<0.05 was considered to be statistically significant.
RESULTS
Aggfl Haploinsufficiency (Heterozygous Aggfl+/- KO) Significantly Reduces the Number of Bone Marrow-Derived EPCs
We isolated and characterized bone marrow-derived mononuclear cells (MNCs) from wild type (WT) and heterozygous Aggfl +/- KO mice. The isolated MNCs appeared to be centrally rounded EPCs with a peripheral spindle shape four days after culture (Fig. 25 A). The cells appeared to be spindle-shaped adherent EPCs and some showed cobblestone-like morphology at day 8 (Fig. 25A). The isolated EPCs were confirmed by a Dil-Ac-LDL endocytosis assay (red fluorescence) and a UEA-l binding assay (green fluorescence). The isolated EPCs were able to endocytose Ac-LDL and bind UEA-l (Fig. 25B), suggesting that they are endothelial lineage cells with characteristics of EPCs as previously described (32;
42; 43). Previous studies indicated that early EPCs (<14 days) were positive for CD34, CD31, CD14, CD45, Scal-l, c-Kit, and VEGFR2, but mostly negative for CD144 (11). Our flow cytometry analysis showed that the isolated EPCs were positive for CD31, CD34, and CD45, and negative for CD144 at day 10 of culture (Fig. 25C). We compared the number of Dil-Ac-LDL/FITC-UEA- 1 positive cells from heterozygous Aggfl +/- KO mice with the cells from WT mice. The number of Dil-Ac-LDL and FITC-UEA- 1 positive cells isolated from heterozygous Aggfl +/- KO mice was moderately but significantly reduced compared with WT mice (P<0.05; n=6/group) (Fig. 26). These data suggest that Aggfl may be involved in differentiation of EPCs.
Aggfl Is Required for Angiogenic and Other Functions of EPCs
Western blot analysis showed that AGGF1 expression was significantly increased in EPCs isolated from db/db mice and HFD-induced T2DM mice (Fig. 27). Interestingly, we also found that Aggfl is required for angiogenic and other functions of EPCs. We studied db/db mice and a mouse model for type 2 DM (T2DM) by treating C57BL/6J mice with HFD. HFD treatment successfully induced a significant increase of blood glucose levels in mice (Fig. 28). The EPCs isolated from db/db mice showed a significantly decreased angiogenic function in a matrigel-based capillary tube formation assay (Fig. 19A-B), decreased proliferation in a CCK8 assay (Fig. 19C), decreased transendothelial migration required for homing of EPCs to sites of endothelial disruption for repair and
neovascularization in a transwell assay (Fig. 19D-E), and reduced migration in a scratch- wound assay (Fig. 19F-G) compared with EPCs isolated form wild type mice (WT).
Interestingly, Aggfl +/- showed similar effects as db/db mice (Fig. 19A-G). Aggfl +/- db/db double KO mice showed exacerbated effects compared with Aggfl +/- KO mice (Fig. 19A-
G). We analyzed blood glucose levels between WT mice and Aggfl +/- KO mice at baseline, but no significant difference was detected (WT: 6.54+0.78 mM vs. Aggfl +/- KO: 7.14+0.92 mM). We analyzed the expression levels of VEGF, VEGFR2 and CXCR4 mRNA in EPCs isolated from different groups of mice. Our real-time RT-PCR analysis showed that Aggfl haploinsufficiency did not affect the expression of VEGF, VEGFR2 and CXCR4 in EPCs (Fig. 29), suggesting that the effect of Aggfl haploinsufficiency is independent of VEGF, VEGFR2 and CXCR4. In addition, similar results were obtained in the HFD-induced mouse model for T2DM (Fig. 30). These data suggest that Aggfl is required for important functions of EPCs.
AGGF1 Protein Treatment Reverses Hyperglycemia-Impaired Angiogenic and Other Functions of EPCs
We found that the AGGF1 protein treatment reversed the impaired angiogenesis, proliferation, migration and transendothelial migration of EPCs induced by high glucose (HG). When cultured in HG media (30 mM D-glucose mimicking the pathological environment of diabetic dyslipidemia or hyperglycemia; 30 mM L-glucose as negative control), mouse EPCs showed significantly decreased angiogenic function (Fig. 20B, C), proliferation (Fig. 20D), transendothelial migration (Fig. 20E, F), and migration (Fig. 20G,
H) compared with the EPCs cultured in control regular media. AGGF1 is an angiogenic factor that can be secreted outside of cells (21), therefore, we treated EPCs with the purified human AGGF1 protein in culture to determine the effects of AGGF1 on functions of EPCs
(Fig. 20A). All the defects by high glucose on EPC functions were blocked by AGGF1 treatment (Fig. 20B-H). We analyzed the expression levels of VEGF, VEGFR2 and CXCR4 mRNA in EPCs treated with or without AGGF1 in combination with or without high glucose. AGGF1 did not affect the expression levels of VEGF, VEGFR2 and CXCR4 in EPCs (Fig. 31). These data suggest that AGGF1 can counter the damaging effects of hyperglycemia on EPCs, however, its effects appear independent of CXCR4, VEGF or VEGFR2.
AGGF1 Boosts EPC-Mediated Angiogenesis and Blood Perfusion in T2MD
In a T2DM mouse model (db/db mice), AGGF1 was found to greatly boost the therapeutic effects of cell therapy with EPC implantation on vascular complications under ischemia in vivo. As AGGF1 can reverse the damaging effects of T2DM on EPCs, we hypothesized that AGGFl-primed EPCs can serve as a successful treatment tool for vascular complications in T2MD in vivo. We pre-treated EPCs with recombinant AGGF1 and transplanted the AGGFl-primed EPCs into db/db mice after a hind- limb ischemia surgery. Notably, AGGFl-primed EPCs were significantly more effective than elution-buff er- pretreated EPCs in increasing blood flow in diabetic mice with ischemia (Fig. 21A). The rate of tissue necrosis was significantly decreased by AGGFl-primed EPCs compared with elution-buffer-pretreated control EPCs (Fig. 21B). Similar significant inhibitory effect on ambulatory impairment was detected for AGGFl-primed EPCs (Fig. 21C). Immunostaining showed that the density of CD3l-postiive vessels was significantly higher for AGGFl- primed EPCs than for elution-buffer-pretreated control EPCs at day 28 after transplantation (Fig. 21D and Fig. 32A). Similar therapeutic efficacy for AGGFl-primed EPCs was obtained in HFD-induced hyperglycemia mice (Fig. 31A-E). Together, these data demonstrate that AGGFl-primed EPCs improve blood perfusion and angiogenesis in T2MD mice with hind- limb ischemia, indicating that AGGF1 can boost the therapeutic effects of cell therapy with EPCs.
The therapeutic effect of AGGFl-primed EPCs may be related to the possibility that AGGF1 increases the homing of transplanted EPCs to the ischemic areas. To test the hypothesis, we infected EPCs with EGFP lentivirus (Fig. 34) and performed EPC
transplantation into db/db mice. Four weeks after transplantation, the gastrocnemius muscle of DM mice with hind-limb ischemia was characterized by immunostaining with an anti-GFP antibody. As shown in Fig. 32B, EPCs labeled by overexpression of EGFP and primed by AGGF1 induced significantly more homing or mobilization of GFP-labeled EPCs in the
vascular injury site in the muscle than elution-buffer-pretreated control EPCs. Similar results were obtained in HFD-induced hyperglycemia mice (Fig. 33F).
AGGF1 Attenuates Hyperglycemia-Induced Oxidative Stress in EPCs
To identify the molecular mechanism by which AGGF1 boosts the therapeutic effects of EPCs, we analyzed the effect of AGGF1 on oxidative stress induced by hyperglycemia in EPCs. Oxidative stress is a key factor accounting for endothelial dysfunction in diabetic EPCs (44-47). Therefore, we analyzed the effect of AGGF1 on high glucose-induced oxidative stress in EPCs. The level of reactive oxygen species (ROS) in EPCs after treatment with a high glucose solution was significantly increased, however, the effect was attenuated by treatment with AGGF1 (Fig. 35). However, AGGF1 treatment did not fully reverse the high glucose-induced ROS increase (P<0.05 between the control group and HG+AGGF1 group) (Fig. 35). The data suggest that AGGF1 can attenuate the production of ROS in EPCs, which may be a factor for its effects on boosting the therapeutic effects of EPCs on endothelial dysfunction in DM.
AGGF1 Activates Nuclear Localization of Nrf2 via the AKT/ Fyn/Nrf2 Signaling Pathway in EPCs
To identify the molecular mechanism by which AGGF1 inhibits ROS generation, we characterized the effect of AGGF1 on anti-oxidative transcription factor Nrf2 (NF-E2 p45- related factor 2) (43-46). We found that AGGF1 promoted nuclear accumulation of Nrf2, thereby activating the Nrf2 pathway in EPCs. When cultured in high glucose media, the nuclear accumulation of Nrf2 was significantly reduced, however, the effect was abolished by treatment with AGGF1 (Fig. 22). Nrf2 activates the transcription from many downstream genes, including HQ1, NQO-l, and CAT (11; 49). Western blot analysis showed that AGGF1 increased the expression levels of HQ1, NQO-l, and CAT (Fig. 22A). Real-time RT-PCR analysis showed that AGGF1 increased the expression levels of HQ1, NQO-l, and CAT mRNA (Fig. 22B). Furthermore, immunostaining with an anti-Nrf2 antibody confirmed the finding of increased nuclear immunofluorescence/localization for Nrf2 by AGGF1 (Fig. 36). Interestingly, real-time RT-PCR analysis showed that AGGF1 did not affect the expression level of Nrf2 mRNA (Fig. 37A). Western blot analysis also showed that AGGF1 did not affect the expression level of Nrf2 and REAP- 1 (binding to Nrf2 to facilitate its ubiquitination) (50) (Fig. 37B-C). These data demonstrate that AGGF1 increases the nuclear
accumulation of Nrf2, but not the expression levels of Nrf2 and KEAP-l, and results in transcriptional activation of Nrf2 downstream target genes NQO-l and CAT.
It was previously shown that AGGF1 can activate PI3K and AKT signaling in endothelial cells and in zebrafish (19; 20). PI3K and AKT were shown to regulate phosphorylation and nuclear localization of Fyn (19; 20). Fyn was shown to phosphorylate Nrf2 at Y568, resulting in nuclear export and degradation of Nrf2 (19; 20). Therefore, we hypothesized that AGGF1 regulates ROS generation and EPC functions via an AGGF1- AKT-Fyn-Nrf2 signaling pathway. When cultured in high glucose media, EPCs showed a significantly reduced level of phosphorylation of AKT, however, the effect was reversed by AGGF1 treatment (Fig. 23A). On the other hand, the effect of AGGF1 was blocked by siRNA for AKT or wortmannin, a specific inhibitor of PI3K upstream of AKT (Fig. 23). EPCs treated with high glucose showed a significantly increased level of nuclear Fyn (n- Fyn), however, the effect was reversed by AGGF1 treatment (Fig. 23A-B). The effect of AGGF1 was blocked by siRNA for AKT or wortmannin (Fig. 23A-B). Similarly, siRNA for AKT, wortmannin or siRNA for Nrf2 blocked the rescue effects of AGGF1 on the HG- induced decrease of Nrf2 nuclear accumulation and down-regulation of Nrf2 downstream genes NQO-l, and CAT (Fig. 23C-E). Furthermore, the nuclear accumulation of nNrf2 was decreased by siRNA for AGGF1 under either a normal or HG condition, however, Fyn knock down with siRNA blocked the effect of AGGF1 (Fig. 38). These data suggest that AGGF1 promotes Nrf2 nuclear localization and activates Nrf2 downstream target genes by regulating the AKT-Fyn signaling pathway.
To further demonstrate that AGGF1 regulates the function of EPCs by Nrf2, we studied the effects of wortmannin and siRNAs for AKT and Nrf2 on EPCs treated with AGGF1 (Fig. 24A, F, K). Treatment of EPCs with AGGF1 reversed the impaired angiogenesis (Fig. 24C, H, M), proliferation (Fig. 24B, G, L), transendothelial migration (Fig. 24D, I, N), and migration by hyperglycemia (Fig. 24E, J, O). However, these effects of AGGF1 were attenuated by wortmannin, knockdown of AKT, and knockdown of Nrf2 (Fig. 24). These data further indicate that AGGF1 regulates the functions of EPCs by affecting Nrf2.
DISCUSSION
While the results presented above, and discussed below, discuss possible mechanisms, the present invention is not limited to any particular mechanism and an understanding of the mechanism is not necessary to practice the invention. We show that AGGF1 activates the
AKT-Fyn-Nrf2 signaling pathway in EPCs (Fig. 39). Nrf2 is a transcription factor important to cellular defense against oxidative stress. Nrf2 needs to be translocated into the nucleus to execute its function, and its nuclear localization is regulated by AKT-Fyn signaling. AGGF1 activates AKT, which leads to de-phosphorylation of Fyn, resulting in reduced translocation of Fyn into the nucleus and decreased nuclear Fyn (n-Fyn). Decreased n-Fyn leads to decreased phosphorylation of Nrf2 and inhibits the nuclear export, ubiquitination and degradation of Nrf2, increasing the expression levels of nuclear Nrf2 and downstream cytoprotective genes such as NQO-l and CAT (Fig. 22). This blocks hyperglycemia- induced ROS generation in EPCs and potentiates the functions of EPCs. Oxidative stress was considered to be a critical factor accounting for EPC dysfunction in T2DM (44; 45). Our results suggest that AGGF1 protects EPCs partly by inhibiting hyperglycemia-induced ROS generation through a novel anti-oxidative stress signaling pathway (Fig. 39).
Endothelial dysfunction is a major problem in T2DM, and lack of endothelial regeneration and impaired angiogenesis by EPCs are responsible for the vascular abnormalities in DM (10; 11). Hence, there is an unmet need for therapeutic interventions to accelerate the repair of dysfunctional endothelium and restore blood flow by EPCs in treatment of DM patients. The data in this Example establish AGGF1 as an important molecule to repair diabetic EPC dysfunction induced by hyperglycemia or dyslipidemia in db/db mice and HFD-induced hyperglycemia mice (Fig. 21, Fig.32 and Fig. 33). Robust therapeutic effects were observed for AGGF1 -primed EPCs in boosting angiogenesis, restoring blood flow, reducing tissue necrosis and ambulatory impairment in vivo in a diabetic hind-limb ischemia model in db/db mice and HFD-induced hyperglycemia mice (Fig. 21, Fig. 32 and Fig. 33). Although some hypoglycemic agents (e.g. metaformin), lipid lowering drugs (e.g. statins) and renin- angiotensin system inhibitors were shown to increase circulating EPC levels, however, the increases were moderate as compared to the levels in healthy study subjects (53; 54). Moreover, EPC functions in DM patients are severely impaired, therefore, the therapeutic effects may be limited even with the increased level of autologous EPCs by these drugs. We showed that AGGF1 fully reversed hyperglycemia- impaired functions of EPCs (Fig. 20), therefore, one potential advantage of the AGGFl-based EPC therapy is that dysfunctional EPCs from DM patients may be repaired and/or functions enhanced, which may achieve a maximum efficacy for the treatment for DM-associated vascular abnormalities.
An AGGFl-based EPC therapy for diabetic ischemia may have additional benefits for DM patients who are often affected with CAD/MI, heart failure, peripheral vascular disease,
and other conditions. Furthermore, DM can also cause many other diseases including diabetic cardiomyopathy, end-stage renal disease (ESRD) and vascular aging, which are all associated with abnormal angiogenesis and vascular dysfunction. Based on our data, it is likely that AGGF1 -primed EPC therapy may serve as a new strategy not only for treating DM-associated vascular abnormalities, and those with vascular abnormalities without DM, but also for many other diabetes mellitus complications such as diabetic cardiomyopathy, ESRD and vascular aging. This Example showed that in db/db mice or HFD-induced diabetic mice with dysfunctional EPCs, AGGFl-primed EPCs attenuated vascular complications by increasing blood flow and CD3l-positive capillary density. Because AGGFl-primed EPCs showed increased capillary formation capability, they may be able to increase blood flow and capillary density in lean and non-diabetic mice with functional EPCs, too.
References for Example 3:
1. Menon et al., Front Endocrinol (Lausanne) 8:263, 2017
2. Cao et al., Life Sci 181:9-16, 2017
3. Muniyappa and Sowers, Diabetes 2014 Apr; 63(4): 1194-1197.
4. Kitabchi et al., Diabetes Care 32:1335-1343, 2009
5. Smith et al., Circulation l05:el65-el69, 2002
6. Hill et al., PLoS One l2:el86554, 2017
8. Tong et al., Clin J Am Soc Nephrol, 2017
9. Yu et al., Stem Cells Int 2016:1803989, 2016
10. Wils et al., Pharmacol Ther 170:98-115, 2017
11. Dai et al., Circ Res l20:e7-e23, 2017
12. Georgescu et al., Eur J Pharmacol 669:1-6, 2011
13. Asahara et a., Stem Cells 29:1650-1655, 2011
14. Silvestre et al., Physiological Reviews 93:1743-1802, 2013
15. Dimmeler et al., Arteriosclerosis, Thrombosis, and Vascular Biology 30:1088-1093, 2010
16. Meamar et al., J Res Med Sci 21:47, 2016
17. Wan et al., Exp Biol Med (Maywood) 235:989-998, 2010
18. Jarajapu et al., Circ Res 106:854-869, 2010
19. Chen et al., Hum Mol Genet 22:963-976, 2013.
20. Zhang et al., Hum Mol Genet 25:5094-5110, 2016
21. Tian et al., Nature 427:640-645, 2004.
22. Lu et al., PLoS One 7:e46998, 2012
23. Li et al., Blood 123:501-508, 2014
24. Cao et al., Anat Rec (Hoboken) 294:580-588, 2011
25. Surwit et al., Diabetes 37:1163-1167, 1988
26. Srinivasan et al., Pharmacol Res 52:313-320, 2005
27. Winzell et al., Diabetes 53 Suppl 3:S2l5-S2l9, 2004
28. Helisch et al., Arterioscler Thromb Vase Biol 26:520-526, 2006
29. Limbourg et al., Nat Protoc 4:1737-1746, 2009
30. Lu et al., PLoS One 7:e46998, 2012
31. Asahara et al., Science 275:964-967, 1997
32. Kalka et al., Proceedings of the National Academy of Sciences 97:3422-3427, 2000
33. Yao et al., Nat Commun 8:133, 2017
34. Xu et al., Cell Physiol Biochem 48:2114-2122, 2018
35. Lu et al., PLoS Biol l4:el002529, 2016
36. Li et al., Stem Cell Res Ther 8:237, 2017
37. Hayakawa et al., Proc Natl Acad Sci U S A 109:7505-7510, 2012
38. Liu et al., Diabetes 63:1763-1778, 2014
39. Ye et al., Sci Rep 6:34034, 2016
40. Xu et al., Am J Transl Res 8:1708-1718, 2016
41. Wang et al., Nat Genet 43:345-349, 2011
42. Urbich et al., Circulation 108:2511-2516, 2003
43. Fadini et al., Circ Res 110:624-637, 2012
44. Sorrentino et al., Circulation 116:163-173, 2007
45. Dembach, Blood 104:3591-3597, 2004
46. Annuk et al., Kidney Int Suppl:S50-S53, 2003
47. Cai et al., Circ Res 87:840-844, 2000
48. Zhang et al., Oxid Med Cell Longev 2016:4350965, 2016
49. Kuang et al., Int J Biol Sci 9:656-665, 2013
50. Limonciel et al., Toxins (Basel) 6:371-379, 2014
51. Niture et al., Free Radic Biol Med 66:36-44, 2014
52. Tonelli et al., Antioxid Redox Signal 29:1727-1745, 2018
53. Churdchomjan et al., BMC Endocr Disord 10:5, 2010
54. Reinhard et al., Diabetologia 53:2129-2133, 2010
55. Yao et al., J Am Heart Assoc 6, 2017
56. Liu et al., Apoptosis 19:1254-1268, 2014
57. Zhu et al., J Neuroinflammation 15: 178, 2018
58. Ackermann et al., Int J Mol Med 33:833-839, 2014
59. Zemani et al., Arterioscler Thromb Vase Biol 28:644-650, 2008
60. Kang et al., Lancet 363:751-756, 2004
61. Kang et al., Lancet 363:751-756, 2004
EXAMPLE 4
In Vivo Treatment with AGGF1 Peptides
1. AGGF1 protein therapy reduces plasma glucose levels in db/db diabetic mice.
AGGF1 protein therapy is effective in lowering fasting glucose levels in HFD- induced diabetic mice. To further conform this, we examined the efficacy of AGGF1 protein therapy in another independent diabetic mouse model. The db/db mice are a well-established murine model for diabetes. We administered recombinant His-tagged AGGF1 protein (3 nmole or 0.25 ug/g body weight) vs. IgG negative control (3 nmole/g body weight) to 4 db/db mice via subcutaneous injection over a time period of 48 hours. The overall study design is shown in Figure 40A. As shown in Figure 40B, compared to IgG control, AGGF1 significantly lowers the plasma glucose levels for more than 24 hours. The effect was lost at the time point of 30 hours. These data confirm that subcutaneous injection of recombinant AGGF1 protein is an effective therapy for lowering plasma glucose levels in diabetic mice.
2. The therapeutic effects of AGGF1 is more than 10 times longer than insulin treatment.
We performed a similar treatment study for insulin in db/db mice. As shown in Figure 40C, subcutaneous injection of insulin (0.75 IU/kg body weight) also lowers plasma glucose levels in db/db diabetic mice. The maximum effect of insulin was found to be achieved at 1.0- 1.5 hours after injection, however, the effect was lost 2.5 hours after injection. Comparing Figure 40B and 40C, it is clear that the therapeutic effect of AGGF1 is more than 10 times longer than insulin in in db/db diabetic mice. It is also important to note that AGGF1 therapy did not cause hypoglycemia, one of the major adverse effect of insulin treatment.
REFERENCES:
1. Mathieu et al., Nat Rev Endocrinol. 2017;13:385-399.
2. Pandyarajan et al., Curr Diab Rep. 2012;12:697-704.
3. Nathan, JAMA. 2015;314:1052-62.
4. Levitt et al., J Pediatr. 2018;196:208-216 e2.
5. Nathan, N Engl J Med. 1993;328:1676-85.
6. U.K. prospective diabetes study 16. Overview of 6 years' therapy of type II diabetes: a progressive disease. U.K. Prospective Diabetes Study Group. Diabetes.
1995;44:1249-58.
7. Klein, Diabetes Care. 1995;18:258-68.
8. Bailey, Trends Pharmacol Sci. 2000;21:259-65.
9. Gloyn and Drucker, Lancet Diabetes Endocrinol. 2018.
10. Lu et al., PLoS Biol. 20l6;l4:el002529.
11. Yao et al., Nat Commun. 20l7;8:l33.
12. Yao et al., J Am Heart Assoc. 20l7;6:e005889.
13. Zhang et al., Hum Mol Genet. 2016;25:5094-5110.
All publications and patents mentioned in the above specification are herein incorporated by reference in their entirety for all purposes. Various modifications and variations of the described compositions, methods, and uses of the technology will be apparent to those skilled in the art without departing from the scope and spirit of the technology as described. Although the technology has been described in connection with specific exemplary embodiments, it should be understood that the technology as claimed should not be unduly limited to such specific embodiments. Indeed, various modifications of the described modes for carrying out the technology that are obvious to those skilled in pharmacology, biochemistry, medical science, or related fields are intended to be within the scope of the following claims.
Claims
1. A method of treating a subject comprising: administering a composition to a subject, wherein said subject has at least one of the following: a disease or condition characterized by abnormal glucose metabolism, ischemic vascular disease, pulmonary hypertension, cardiac hypertrophy, and heart failure:
wherein said composition comprises:
i) an AGGF1 peptide, and/or
ii) an expression vector encoding said AGGF1 peptide.
2. The method of Claim 1, wherein said AGGF1 peptide consists of, consists essentially of, or comprises QRDDAPASVH (SEQ ID NO: 18).
3. The method of Claim 1, wherein said expression vector comprises the nucleic acid sequence in SEQ ID NO:25.
4. The method of Claim 1, wherein said disease or condition is selected from the group consisting of: pulmonary hypertension, peripheral artery disease (PAD), coronary artery disease (CAD), coronary heart disease (CHD), myocardial infarction (MI), Type I diabetes, Type II diabetes, hyperglycemia, inadequate glucose clearance, and hyperinsulinemia.
5. The method of Claim 1, wherein said method further comprises administering insulin to said subject.
6. The method of Claim 1, wherein said AGGF1 peptide comprises full length human AGGF1 shown in SEQ ID NO:l or wherein said AGGF1 peptide has at least 97% sequence identity with SEQ ID NO:l.
7. The method of Claim 1, wherein said AGGF1 peptide is selected from any of SEQ ID Nos:2-l6 or i) a peptide comprising the amino acid sequence from 574 to 614 of SEQ ID NO: l, ii) a peptide comprising the amino acid sequence from 574 to 624 of SEQ ID NO:l, or iii) a peptide comprising the amino acid sequence from 604 to 714 of SEQ ID NO:l.
8. The method of Claim 1, wherein said AGGF1 peptide has at least 97% sequence identity with any one of SEQ ID Nos:2-l6.
9. The method of Claim 1, wherein said administering reduces or eliminates at least one symptom of said disease or condition.
10. The method of Claim 1, wherein said composition comprises said AGGF1 peptide.
11. The method of Claim 1, wherein said composition comprises said an expression vector encoding said AGGF1 peptide.
12. The method of Claim 1, wherein said subject is a human.
13. The method of Claim 12, wherein said composition comprises said AGGF1 peptide, and wherein the dosage is selected from the group consisting of: 0.1-15 mg/kg, 0.5-10 mg/kg, and 1-7 mg/kg.
14. The method of Claim 12, wherein said composition comprises said an expression vector encoding said AGGF1 peptide, and wherein said dosage is about 1.0 x 1011 - 5.0 x 1013.
15. A system or kit comprising:
a) a blood glucose monitoring device or article of manufacture; and
b) a composition comprising: i) an AGGF1 peptide, or ii) an expression vector encoding said AGGF1 peptide.
16. The system or kit of Claim 15, wherein said composition comprises said AGGF1 peptide.
17. The system of kit of Claim 1, wherein said glucose monitoring device or article of manufacture is selected from the group consisting of: a blood glucose test strip, a reader for reading said glucose test strip, FreeStyle Libre, GlucoTrack, Eversense, GlucoWise, NovioSense, GlucoSense, and a smart contact lens.
18. A system or kit comprising:
a) a composition comprising: i) an AGGF1 peptide, or ii) an expression vector encoding said AGGF1 peptide; and
b) an implantable pump containing said composition, wherein said implantable pump is configured to deliver an amount of said composition into the bloodstream of a subject.
19. The system or kit of Claim 18, wherein said composition comprises said AGGF1 peptide.
20. A composition comprising:
a) an AGGF1 peptide, or an expression vector encoding said AGGF1 peptide; and
b) insulin peptide, or an expression vector encoding said insulin peptide.
21. The composition of Claim 21, wherein said AGGF1 peptide comprises full length human AGGF1 shown in SEQ ID NO:l, or wherein said AGGF1 peptide has at least 97% sequence identity with SEQ ID NO: l.
22. The composition of Claim 21, wherein said AGGF1 peptide consists of, consists essentially of, or comprises QRDDAPASVH (SEQ ID NO: 18).
23. The composition of Claim 21, wherein said AGGF1 peptide is selected from any of SEQ ID Nos:2-l6.
24. The composition of Claim 21, wherein said AGGF1 peptide has at least 97% sequence identity with any one of SEQ ID Nos:2-l6.
25. A method of treating a subject with a cardiovascular disease or condition comprising: administering a composition comprising: i) an AGGF1 peptide that is no longer than 600 amino acids, ii) an expression vector encoding said AGGF1 peptide, to said subject with said disease or condition, or iv) the AGGF1 peptide consists of, consists essentially of, or comprises QRDDAPASVH (SEQ ID NO: 18).
26. The method of Claim 25, wherein said disease or condition is selected from the group consisting of: coronary artery disease, myocardial infarction, cardiac hypertrophy, heart failure, peripheral artery disease, restenosis and in-stent thrombosis after angioplasty and stenting, and pulmonary arterial hypertension.
27. The method of Claim 25, wherein, wherein said AGGF1 peptide is selected from any of SEQ ID Nos:2-l6.
28. The method of Claim 25, wherein said subject is a human and: A) said composition comprises said AGGF1 peptide, and wherein the dosage is selected from the group consisting of: 0.1-15 mg/kg, 0.5-10 mg/kg, and 1-7 mg/kg; or B) said composition comprises said expression vector encoding said AGGF1 peptide, and wherein said dosage is about 1.0 x 1011 - 5.0 x 1013.
29. The method of Claim 25, wherein said AGGF1 peptide is no longer than 300, or no longer than 150, amino acids in length and said peptide comprises, consists essentially of, or consists of: i) the amino acid sequence from 574 to 614 of SEQ ID NO:l, ii) the amino acid sequence from 574 to 624 of SEQ ID NO:l, iii) the amino acid sequence from 604 to 714 of SEQ ID NO:l, or iv) the AGGF1 peptide consists of, consists essentially of, or comprises QRDDAPASVH (SEQ ID NO: 18).
30. The method of Claim 25, wherein said AGGF1 peptide has at least 97% sequence identity with any one of SEQ ID Nos:2-l6.
31. A method of treating a subject with pulmonary arterial hypertension (PAH) comprising: administering a composition comprising: i) an AGGF1 peptide, or ii) an expression vector encoding said AGGF1 peptide, to said subject with said PAH.
32. The method of Claim 31, wherein said AGGF1 peptide is no longer than 300, or no longer than 150, amino acids in length and said peptide comprises, consists essentially of, or consists of: i) the amino acid sequence from 574 to 614 of SEQ ID NO:l, ii) the amino acid sequence from 574 to 624 of SEQ ID NO:l, iii) the amino acid sequence from 604 to 714 of
SEQ ID NO:l; or iv) the AGGF1 peptide consists of, consists essentially of, or comprises QRDDAPASVH (SEQ ID NO: 18).
33. The method of Claim 31, wherein said AGGF1 peptide has at least 97% sequence identity with any one of SEQ ID Nos:2-l6.
34. A method of treating a subject comprising: administering a composition to a subject, wherein said subject has a disease or condition characterized by abnormal glucose metabolism and/or ischemic vascular disease, and
wherein said composition comprises an AGGF1 -primed endothelial progenitor cell.
35. The method of Claim 34, wherein said disease or condition is selected from the group consisting of peripheral artery disease (PAD), coronary artery disease (CAD), coronary heart disease (CHD), and myocardial infarction (MI).
36. The method of Claim 34, wherein said disease or condition is selected from the group consisting of: Type I diabetes, Type II diabetes, hyperglycemia, inadequate glucose clearance, and hyperinsulinemia.
37. The method of Claim 34, wherein said AGGF1 peptide comprises full length human AGGF1 shown in SEQ ID NO:l.
38. The method of Claim 34, wherein said AGGF1 peptide has at least 97% sequence identity with SEQ ID NO:l.
39. The method of Claim 34, wherein said AGGF1 peptide is selected from any of SEQ ID Nos:2-l6 or i) a peptide comprising the amino acid sequence from 574 to 614 of SEQ ID NO: l, ii) a peptide comprising the amino acid sequence from 574 to 624 of SEQ ID NO:l, iii) a peptide comprising the amino acid sequence from 604 to 714 of SEQ ID NO:l, or iv) the AGGF1 peptide consists of, consists essentially of, or comprises QRDDAPASVH (SEQ ID NO: 18).
40. The method of Claim 34, wherein said AGGF1 peptide has at least 97% sequence identity with any one of SEQ ID Nos:2-l6.
41. The method of Claim 34, wherein said administering reduces or eliminates at least one symptom of said disease or condition.
42. The method of Claim 34, wherein said composition comprises a said AGGF1 peptide.
43. The method of Claim 34, wherein said composition comprises said an expression vector encoding said AGGF1 peptide.
44. The method of Claim 34, wherein said subject is a human.
45. The method of Claim 34, wherein said AGGFl-primed endothelial progenitor cell comprises an endothelial progenitor cell (EPC) from said subject, wherein said EPC has been exposed to AGGF1 peptides ex vivo.
46. The method of Claim 34, wherein said AGGFl-primed endothelial progenitor cell comprises an endothelial progenitor cell (EPC) from a relative of said subject, wherein said EPC has been exposed to AGGF1 peptides ex vivo.
47. The method of Claim 34, wherein said AGGFl-primed endothelial progenitor cell comprises an endothelial progenitor cell (EPC) from a non-relative of said subject, wherein said EPC has been exposed to AGGF1 peptides ex vivo.
48. A composition comprising: an AGGFl-primed endothelial progenitor cell.
49. The composition of Claim 48, wherein said AGGFl-primed endothelial progenitor cell comprises an endothelial progenitor cell (EPC) from a subject with a disease or condition characterized by abnormal glucose metabolism and/or ischemic vascular disease, wherein said EPC has been exposed to AGGF1 peptides ex vivo.
50. The composition of Claim 48, further comprising a physiological tolerable liquid.
51. A kit or system comprising:
a) an endothelial progenitor cell (EPC), and
b) a composition comprising:
i) an AGGF1 peptide, and/or
ii) an expression vector encoding said AGGF1 peptide.
52. The kit or system of Claim 51, wherein said AGGF1 peptide consists of, consists essentially of, or comprises QRDDAPASVH (SEQ ID NO: 18).
53. The kit or system of Claim 51, wherein said expression vector comprises the nucleic acid sequence in SEQ ID NO:25.
Applications Claiming Priority (4)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
US201862744488P | 2018-10-11 | 2018-10-11 | |
US62/744,488 | 2018-10-11 | ||
US201962842283P | 2019-05-02 | 2019-05-02 | |
US62/842,283 | 2019-05-02 |
Publications (1)
Publication Number | Publication Date |
---|---|
WO2020077030A1 true WO2020077030A1 (en) | 2020-04-16 |
Family
ID=70164705
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/US2019/055513 WO2020077030A1 (en) | 2018-10-11 | 2019-10-10 | Aggf1 and aggf1-primed cells for treating diseases and conditions |
Country Status (1)
Country | Link |
---|---|
WO (1) | WO2020077030A1 (en) |
Cited By (2)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
CN115957300A (en) * | 2022-12-22 | 2023-04-14 | 华中科技大学 | Application of recombinant protein in preparation of drug for inhibiting aneurysm |
CN116059319A (en) * | 2022-07-13 | 2023-05-05 | 华中科技大学 | Application of recombinant protein in preparation of muscle atrophy inhibiting drugs |
Citations (7)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US20060074404A1 (en) * | 2002-04-22 | 2006-04-06 | Medtronic, Inc. | Implantable drug delivery system responsive to intra-cardiac pressure |
US20070059288A1 (en) * | 2005-03-31 | 2007-03-15 | Dinsmore Jonathan H | Treatment for heart disease |
WO2007049096A1 (en) * | 2005-10-25 | 2007-05-03 | Inserm (Institut National De La Sante Et De La Recherche Medicale) | A method of expansion of endothelial progenitor cells |
US20120122227A1 (en) * | 2010-11-15 | 2012-05-17 | Roche Diagnostics Operations, Inc. | Guided structured testing kit |
US20130058940A1 (en) * | 2003-08-20 | 2013-03-07 | The Cleveland Clinic Foundation | Novel Gene And Protein Associated With Angiogenesis And Endothelial Cell Specific Apoptosis |
US20170087217A1 (en) * | 2005-03-31 | 2017-03-30 | Mannkind Corporation | Superior control of blood glucose in diabetes treatment |
US20170312317A1 (en) * | 1998-03-27 | 2017-11-02 | Northern Therapeutics Inc. | Cell-based therapy for the pulmonary system |
-
2019
- 2019-10-10 WO PCT/US2019/055513 patent/WO2020077030A1/en active Application Filing
Patent Citations (7)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US20170312317A1 (en) * | 1998-03-27 | 2017-11-02 | Northern Therapeutics Inc. | Cell-based therapy for the pulmonary system |
US20060074404A1 (en) * | 2002-04-22 | 2006-04-06 | Medtronic, Inc. | Implantable drug delivery system responsive to intra-cardiac pressure |
US20130058940A1 (en) * | 2003-08-20 | 2013-03-07 | The Cleveland Clinic Foundation | Novel Gene And Protein Associated With Angiogenesis And Endothelial Cell Specific Apoptosis |
US20070059288A1 (en) * | 2005-03-31 | 2007-03-15 | Dinsmore Jonathan H | Treatment for heart disease |
US20170087217A1 (en) * | 2005-03-31 | 2017-03-30 | Mannkind Corporation | Superior control of blood glucose in diabetes treatment |
WO2007049096A1 (en) * | 2005-10-25 | 2007-05-03 | Inserm (Institut National De La Sante Et De La Recherche Medicale) | A method of expansion of endothelial progenitor cells |
US20120122227A1 (en) * | 2010-11-15 | 2012-05-17 | Roche Diagnostics Operations, Inc. | Guided structured testing kit |
Non-Patent Citations (3)
Title |
---|
BARKER ET AL.: "Is the E133K allele of VG5Q associated with Klippel-Trenaunay and other overgrowth syndromes?", J MED GENET, vol. 43, 27 January 2006 (2006-01-27), pages 613 - 614, XP055701474 * |
LIU ET AL.: "AGGF1 protects from myocardial ischemia/reperfusion injury by regulating myocardial apoptosis and angiogenesis", APOPTOSIS, vol. 19, 4 June 2014 (2014-06-04), pages 1254 - 1268, XP055701477 * |
YAO ET AL.: "Angiogenic Factor AGGF1-Primed Endothelial Progenitor Cells Repair Vascular Defect in Diabetic Mice", DIABETES, vol. 68, 15 May 2019 (2019-05-15), pages 1635 - 1648, XP055701468 * |
Cited By (4)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
CN116059319A (en) * | 2022-07-13 | 2023-05-05 | 华中科技大学 | Application of recombinant protein in preparation of muscle atrophy inhibiting drugs |
CN115957300A (en) * | 2022-12-22 | 2023-04-14 | 华中科技大学 | Application of recombinant protein in preparation of drug for inhibiting aneurysm |
CN115957300B (en) * | 2022-12-22 | 2024-04-05 | 华中科技大学 | Application of a recombinant protein in preparing aneurysm inhibition drug |
WO2024130872A1 (en) * | 2022-12-22 | 2024-06-27 | 华中科技大学 | Use of recombinant protein in preparing drug for inhibiting aneurysm |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
Short et al. | IL‐10 promotes endothelial progenitor cell infiltration and wound healing via STAT3 | |
Dai et al. | Elevating CXCR7 improves angiogenic function of EPCs via Akt/GSK-3β/Fyn-mediated Nrf2 activation in diabetic limb ischemia | |
Song et al. | CREG protects from myocardial ischemia/reperfusion injury by regulating myocardial autophagy and apoptosis | |
Song et al. | Down-regulation of microRNA-320 suppresses cardiomyocyte apoptosis and protects against myocardial ischemia and reperfusion injury by targeting IGF-1 | |
KR102251773B1 (en) | Pharmaceutical Composition for Treating Cardiac Fibrosis | |
Zhang et al. | CXCR4-dependent macrophage-to-fibroblast signaling contributes to cardiac diastolic dysfunction in heart failure with preserved ejection fraction | |
Mofid et al. | Cardiac overexpression of S100A6 attenuates cardiomyocyte apoptosis and reduces infarct size after myocardial ischemia‐reperfusion | |
Liu et al. | Crosstalk between the activated Slit2–Robo1 pathway and TGF‐β1 signalling promotes cardiac fibrosis | |
EP3530275A1 (en) | Treatment of heart failure and related conditions | |
CN105194651B (en) | CREG albumen is used for the medical usage of Ischemic myocardium reperfusion injury | |
KR20050059180A (en) | Compositions for the inhibition of protein kinase c alpha for treatment of diabetes mellitus and cardiovascular diseases | |
Li et al. | Inhibition of Dectin-1 in mice ameliorates cardiac remodeling by suppressing NF-κB/NLRP3 signaling after myocardial infarction | |
EP2565275B1 (en) | Method of treatment of vascular complications using modulators of TRX and TRXNIP | |
JP6978774B2 (en) | Pharmaceutical composition for the prevention or treatment of heart failure | |
Li et al. | Renal Fibrosis Is Alleviated through Targeted Inhibition of IL-11–Induced Renal Tubular Epithelial-to-Mesenchymal Transition | |
WO2020077030A1 (en) | Aggf1 and aggf1-primed cells for treating diseases and conditions | |
Li et al. | Postinfarction gene therapy with adenoviral vector expressing decorin mitigates cardiac remodeling and dysfunction | |
Wang et al. | Cardioprotection of ischemic preconditioning in rats involves upregulating adiponectin | |
US20120058086A1 (en) | Compositions, kits, and methods for promoting ischemic and diabetic wound healing | |
US20240342244A1 (en) | Isthmin Protein Therapeutics for the Treatment of Non-Alcoholic Fatty Liver Disease | |
Tian et al. | Systemic AAV10. COMP-Ang1 rescues renal glomeruli and pancreatic islets in type 2 diabetic mice | |
WO2025106913A1 (en) | Peptides for promoting neovascularization and wound healing and methods using the same | |
Xu et al. | Inhibition of Bif-1 confers cardio-protection in myocardial infarction | |
CN106692974A (en) | Application of estrogen receptor inhibitor in preparing medicine for treating ischemic diseases | |
TW202327566A (en) | Use of composition containing fucoxanthin and oligo-fucoidan for preparing pharmaceutical product treating or improving chronic kidney disease, composition containing fucoxanthin, oligo- fucoidan and L-carnitine, and pharmaceutical product containing the same effectively treating or slowing down the chronic kidney disease |
Legal Events
Date | Code | Title | Description |
---|---|---|---|
121 | Ep: the epo has been informed by wipo that ep was designated in this application |
Ref document number: 19870585 Country of ref document: EP Kind code of ref document: A1 |
|
NENP | Non-entry into the national phase |
Ref country code: DE |
|
122 | Ep: pct application non-entry in european phase |
Ref document number: 19870585 Country of ref document: EP Kind code of ref document: A1 |