CN115515969A - Application of FGF4 and Fgfr1 binding ligand in diabetes - Google Patents
Application of FGF4 and Fgfr1 binding ligand in diabetes Download PDFInfo
- Publication number
- CN115515969A CN115515969A CN202080099938.7A CN202080099938A CN115515969A CN 115515969 A CN115515969 A CN 115515969A CN 202080099938 A CN202080099938 A CN 202080099938A CN 115515969 A CN115515969 A CN 115515969A
- Authority
- CN
- China
- Prior art keywords
- fgf4
- fgfr1
- mice
- glucose
- once
- Prior art date
- Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
- Pending
Links
- 101150016624 fgfr1 gene Proteins 0.000 title claims abstract description 110
- 239000003446 ligand Substances 0.000 title claims abstract description 38
- 102100028072 Fibroblast growth factor 4 Human genes 0.000 title claims description 181
- 101001060274 Homo sapiens Fibroblast growth factor 4 Proteins 0.000 title claims description 179
- 206010012601 diabetes mellitus Diseases 0.000 title claims description 36
- 238000011282 treatment Methods 0.000 claims abstract description 23
- 238000000034 method Methods 0.000 claims abstract description 18
- 208000030159 metabolic disease Diseases 0.000 claims abstract description 14
- 239000003814 drug Substances 0.000 claims abstract description 9
- 238000004519 manufacturing process Methods 0.000 claims abstract description 8
- 239000008194 pharmaceutical composition Substances 0.000 claims description 25
- 208000001072 type 2 diabetes mellitus Diseases 0.000 claims description 24
- 201000001421 hyperglycemia Diseases 0.000 claims description 22
- 239000002502 liposome Substances 0.000 claims description 17
- 206010022489 Insulin Resistance Diseases 0.000 claims description 11
- 208000016097 disease of metabolism Diseases 0.000 claims description 8
- 208000008589 Obesity Diseases 0.000 claims description 7
- 235000020824 obesity Nutrition 0.000 claims description 7
- 230000001684 chronic effect Effects 0.000 claims description 6
- 208000002705 Glucose Intolerance Diseases 0.000 claims description 5
- 206010018429 Glucose tolerance impaired Diseases 0.000 claims description 5
- 206010060378 Hyperinsulinaemia Diseases 0.000 claims description 5
- 230000003451 hyperinsulinaemic effect Effects 0.000 claims description 5
- 201000008980 hyperinsulinism Diseases 0.000 claims description 5
- 239000008177 pharmaceutical agent Substances 0.000 claims 2
- 101100446513 Mus musculus Fgf4 gene Proteins 0.000 abstract description 26
- 108090000381 Fibroblast growth factor 4 Proteins 0.000 abstract description 7
- 102000003969 Fibroblast growth factor 4 Human genes 0.000 abstract 3
- 241000699670 Mus sp. Species 0.000 description 141
- WQZGKKKJIJFFOK-GASJEMHNSA-N Glucose Natural products OC[C@H]1OC(O)[C@H](O)[C@@H](O)[C@@H]1O WQZGKKKJIJFFOK-GASJEMHNSA-N 0.000 description 122
- 239000008103 glucose Substances 0.000 description 122
- 210000002569 neuron Anatomy 0.000 description 88
- 210000004369 blood Anatomy 0.000 description 67
- 239000008280 blood Substances 0.000 description 67
- 230000037396 body weight Effects 0.000 description 40
- 102000018233 Fibroblast Growth Factor Human genes 0.000 description 30
- 108050007372 Fibroblast Growth Factor Proteins 0.000 description 30
- 210000004027 cell Anatomy 0.000 description 30
- 229940126864 fibroblast growth factor Drugs 0.000 description 30
- 238000002347 injection Methods 0.000 description 27
- 239000007924 injection Substances 0.000 description 27
- 230000008859 change Effects 0.000 description 26
- 230000002267 hypothalamic effect Effects 0.000 description 25
- 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 description 24
- 230000000694 effects Effects 0.000 description 23
- 210000004556 brain Anatomy 0.000 description 21
- 210000003016 hypothalamus Anatomy 0.000 description 21
- 238000010304 firing Methods 0.000 description 18
- 238000012360 testing method Methods 0.000 description 18
- 230000002459 sustained effect Effects 0.000 description 17
- 235000009200 high fat diet Nutrition 0.000 description 13
- 230000010412 perfusion Effects 0.000 description 13
- 230000004044 response Effects 0.000 description 13
- 102000004877 Insulin Human genes 0.000 description 12
- 108090001061 Insulin Proteins 0.000 description 12
- 101150046735 LEPR gene Proteins 0.000 description 12
- 101150063827 LEPROT gene Proteins 0.000 description 12
- 230000010030 glucose lowering effect Effects 0.000 description 12
- 229940125396 insulin Drugs 0.000 description 12
- 102000003973 Fibroblast growth factor 21 Human genes 0.000 description 11
- 108090000376 Fibroblast growth factor 21 Proteins 0.000 description 11
- 230000002058 anti-hyperglycaemic effect Effects 0.000 description 11
- 230000001965 increasing effect Effects 0.000 description 11
- 108090000623 proteins and genes Proteins 0.000 description 11
- 241001465754 Metazoa Species 0.000 description 10
- 210000003169 central nervous system Anatomy 0.000 description 10
- 238000002474 experimental method Methods 0.000 description 10
- 239000000243 solution Substances 0.000 description 10
- 230000001154 acute effect Effects 0.000 description 9
- 102000005962 receptors Human genes 0.000 description 9
- 108020003175 receptors Proteins 0.000 description 9
- 101150021185 FGF gene Proteins 0.000 description 8
- 230000033228 biological regulation Effects 0.000 description 8
- 230000002218 hypoglycaemic effect Effects 0.000 description 8
- 238000007912 intraperitoneal administration Methods 0.000 description 8
- 239000000203 mixture Substances 0.000 description 8
- 229920001184 polypeptide Polymers 0.000 description 8
- 102000004196 processed proteins & peptides Human genes 0.000 description 8
- 108090000765 processed proteins & peptides Proteins 0.000 description 8
- FAPWRFPIFSIZLT-UHFFFAOYSA-M sodium chloride Inorganic materials [Na+].[Cl-] FAPWRFPIFSIZLT-UHFFFAOYSA-M 0.000 description 8
- 230000001052 transient effect Effects 0.000 description 8
- 101100227089 Danio rerio fgfr1a gene Proteins 0.000 description 7
- 102000003971 Fibroblast Growth Factor 1 Human genes 0.000 description 7
- 108090000386 Fibroblast Growth Factor 1 Proteins 0.000 description 7
- 102100031734 Fibroblast growth factor 19 Human genes 0.000 description 7
- 230000003178 anti-diabetic effect Effects 0.000 description 7
- 230000037406 food intake Effects 0.000 description 7
- 235000012631 food intake Nutrition 0.000 description 7
- 230000006870 function Effects 0.000 description 7
- 210000004185 liver Anatomy 0.000 description 7
- 230000002503 metabolic effect Effects 0.000 description 7
- 235000018102 proteins Nutrition 0.000 description 7
- 102000004169 proteins and genes Human genes 0.000 description 7
- 230000009467 reduction Effects 0.000 description 7
- 101000846394 Homo sapiens Fibroblast growth factor 19 Proteins 0.000 description 6
- 238000004458 analytical method Methods 0.000 description 6
- HVYWMOMLDIMFJA-DPAQBDIFSA-N cholesterol Chemical compound C1C=C2C[C@@H](O)CC[C@]2(C)[C@@H]2[C@@H]1[C@@H]1CC[C@H]([C@H](C)CCCC(C)C)[C@@]1(C)CC2 HVYWMOMLDIMFJA-DPAQBDIFSA-N 0.000 description 6
- 229940029303 fibroblast growth factor-1 Drugs 0.000 description 6
- 235000013305 food Nutrition 0.000 description 6
- 230000005764 inhibitory process Effects 0.000 description 6
- 230000004060 metabolic process Effects 0.000 description 6
- 108091008794 FGF receptors Proteins 0.000 description 5
- HTTJABKRGRZYRN-UHFFFAOYSA-N Heparin Chemical compound OC1C(NC(=O)C)C(O)OC(COS(O)(=O)=O)C1OC1C(OS(O)(=O)=O)C(O)C(OC2C(C(OS(O)(=O)=O)C(OC3C(C(O)C(O)C(O3)C(O)=O)OS(O)(=O)=O)C(CO)O2)NS(O)(=O)=O)C(C(O)=O)O1 HTTJABKRGRZYRN-UHFFFAOYSA-N 0.000 description 5
- 241000699666 Mus <mouse, genus> Species 0.000 description 5
- 230000002124 endocrine Effects 0.000 description 5
- 239000008273 gelatin Substances 0.000 description 5
- 229920000159 gelatin Polymers 0.000 description 5
- 229960002897 heparin Drugs 0.000 description 5
- 229920000669 heparin Polymers 0.000 description 5
- 210000003140 lateral ventricle Anatomy 0.000 description 5
- 230000001404 mediated effect Effects 0.000 description 5
- 230000003076 paracrine Effects 0.000 description 5
- 230000002093 peripheral effect Effects 0.000 description 5
- 239000011780 sodium chloride Substances 0.000 description 5
- 241000702423 Adeno-associated virus - 2 Species 0.000 description 4
- 241000702421 Dependoparvovirus Species 0.000 description 4
- 229940125830 FGFR1 inhibitor Drugs 0.000 description 4
- 101150057182 GFAP gene Proteins 0.000 description 4
- 108010010803 Gelatin Proteins 0.000 description 4
- DHMQDGOQFOQNFH-UHFFFAOYSA-N Glycine Chemical compound NCC(O)=O DHMQDGOQFOQNFH-UHFFFAOYSA-N 0.000 description 4
- 229930006000 Sucrose Natural products 0.000 description 4
- CZMRCDWAGMRECN-UGDNZRGBSA-N Sucrose Chemical compound O[C@H]1[C@H](O)[C@@H](CO)O[C@@]1(CO)O[C@@H]1[C@H](O)[C@@H](O)[C@H](O)[C@@H](CO)O1 CZMRCDWAGMRECN-UGDNZRGBSA-N 0.000 description 4
- 230000004913 activation Effects 0.000 description 4
- 210000000577 adipose tissue Anatomy 0.000 description 4
- 210000001130 astrocyte Anatomy 0.000 description 4
- 235000019322 gelatine Nutrition 0.000 description 4
- 235000011852 gelatine desserts Nutrition 0.000 description 4
- 230000002440 hepatic effect Effects 0.000 description 4
- 239000003112 inhibitor Substances 0.000 description 4
- 238000000185 intracerebroventricular administration Methods 0.000 description 4
- 108020004999 messenger RNA Proteins 0.000 description 4
- 230000037081 physical activity Effects 0.000 description 4
- 230000002829 reductive effect Effects 0.000 description 4
- 230000011664 signaling Effects 0.000 description 4
- 210000002027 skeletal muscle Anatomy 0.000 description 4
- 239000005720 sucrose Substances 0.000 description 4
- 239000000375 suspending agent Substances 0.000 description 4
- 210000001519 tissue Anatomy 0.000 description 4
- 108091032973 (ribonucleotides)n+m Proteins 0.000 description 3
- FWBHETKCLVMNFS-UHFFFAOYSA-N 4',6-Diamino-2-phenylindol Chemical compound C1=CC(C(=N)N)=CC=C1C1=CC2=CC=C(C(N)=N)C=C2N1 FWBHETKCLVMNFS-UHFFFAOYSA-N 0.000 description 3
- 239000000275 Adrenocorticotropic Hormone Substances 0.000 description 3
- ZEOWTGPWHLSLOG-UHFFFAOYSA-N Cc1ccc(cc1-c1ccc2c(n[nH]c2c1)-c1cnn(c1)C1CC1)C(=O)Nc1cccc(c1)C(F)(F)F Chemical compound Cc1ccc(cc1-c1ccc2c(n[nH]c2c1)-c1cnn(c1)C1CC1)C(=O)Nc1cccc(c1)C(F)(F)F ZEOWTGPWHLSLOG-UHFFFAOYSA-N 0.000 description 3
- 102400000739 Corticotropin Human genes 0.000 description 3
- 101800000414 Corticotropin Proteins 0.000 description 3
- FBPFZTCFMRRESA-FSIIMWSLSA-N D-Glucitol Natural products OC[C@H](O)[C@H](O)[C@@H](O)[C@H](O)CO FBPFZTCFMRRESA-FSIIMWSLSA-N 0.000 description 3
- FBPFZTCFMRRESA-KVTDHHQDSA-N D-Mannitol Chemical compound OC[C@@H](O)[C@@H](O)[C@H](O)[C@H](O)CO FBPFZTCFMRRESA-KVTDHHQDSA-N 0.000 description 3
- 102000044168 Fibroblast Growth Factor Receptor Human genes 0.000 description 3
- 102100023593 Fibroblast growth factor receptor 1 Human genes 0.000 description 3
- 101710182386 Fibroblast growth factor receptor 1 Proteins 0.000 description 3
- 101710193519 Glial fibrillary acidic protein Proteins 0.000 description 3
- PEDCQBHIVMGVHV-UHFFFAOYSA-N Glycerine Chemical compound OCC(O)CO PEDCQBHIVMGVHV-UHFFFAOYSA-N 0.000 description 3
- 229920002527 Glycogen Polymers 0.000 description 3
- 229930195725 Mannitol Natural products 0.000 description 3
- DNIAPMSPPWPWGF-UHFFFAOYSA-N Propylene glycol Chemical compound CC(O)CO DNIAPMSPPWPWGF-UHFFFAOYSA-N 0.000 description 3
- 241000700159 Rattus Species 0.000 description 3
- 102000007056 Recombinant Fusion Proteins Human genes 0.000 description 3
- 108010008281 Recombinant Fusion Proteins Proteins 0.000 description 3
- 206010067584 Type 1 diabetes mellitus Diseases 0.000 description 3
- 241000700605 Viruses Species 0.000 description 3
- 230000003213 activating effect Effects 0.000 description 3
- 239000003472 antidiabetic agent Substances 0.000 description 3
- WQZGKKKJIJFFOK-VFUOTHLCSA-N beta-D-glucose Chemical compound OC[C@H]1O[C@@H](O)[C@H](O)[C@@H](O)[C@@H]1O WQZGKKKJIJFFOK-VFUOTHLCSA-N 0.000 description 3
- 230000004071 biological effect Effects 0.000 description 3
- 229940107161 cholesterol Drugs 0.000 description 3
- KRKNYBCHXYNGOX-UHFFFAOYSA-N citric acid Chemical compound OC(=O)CC(O)(C(O)=O)CC(O)=O KRKNYBCHXYNGOX-UHFFFAOYSA-N 0.000 description 3
- 229960000258 corticotropin Drugs 0.000 description 3
- IDLFZVILOHSSID-OVLDLUHVSA-N corticotropin Chemical compound C([C@@H](C(=O)N[C@@H](CO)C(=O)N[C@@H](CCSC)C(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](CC=1NC=NC=1)C(=O)N[C@@H](CC=1C=CC=CC=1)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CC=1C2=CC=CC=C2NC=1)C(=O)NCC(=O)N[C@@H](CCCCN)C(=O)N1[C@@H](CCC1)C(=O)N[C@@H](C(C)C)C(=O)NCC(=O)N[C@@H](CCCCN)C(=O)N[C@@H](CCCCN)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N1[C@@H](CCC1)C(=O)N[C@@H](C(C)C)C(=O)N[C@@H](CCCCN)C(=O)N[C@@H](C(C)C)C(=O)N[C@@H](CC=1C=CC(O)=CC=1)C(=O)N1[C@@H](CCC1)C(=O)N[C@@H](CC(N)=O)C(=O)NCC(=O)N[C@@H](C)C(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](CC(O)=O)C(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](CO)C(=O)N[C@@H](C)C(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](C)C(=O)N[C@@H](CC=1C=CC=CC=1)C(=O)N1[C@@H](CCC1)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](CC=1C=CC=CC=1)C(O)=O)NC(=O)[C@@H](N)CO)C1=CC=C(O)C=C1 IDLFZVILOHSSID-OVLDLUHVSA-N 0.000 description 3
- 230000001419 dependent effect Effects 0.000 description 3
- 235000005911 diet Nutrition 0.000 description 3
- 230000037213 diet Effects 0.000 description 3
- 238000009472 formulation Methods 0.000 description 3
- 239000012634 fragment Substances 0.000 description 3
- 210000005046 glial fibrillary acidic protein Anatomy 0.000 description 3
- 230000009229 glucose formation Effects 0.000 description 3
- 229940096919 glycogen Drugs 0.000 description 3
- 229940088597 hormone Drugs 0.000 description 3
- 239000005556 hormone Substances 0.000 description 3
- 230000002401 inhibitory effect Effects 0.000 description 3
- 239000000594 mannitol Substances 0.000 description 3
- 235000010355 mannitol Nutrition 0.000 description 3
- 239000000463 material Substances 0.000 description 3
- 230000001537 neural effect Effects 0.000 description 3
- 210000004498 neuroglial cell Anatomy 0.000 description 3
- 239000000843 powder Substances 0.000 description 3
- 238000004445 quantitative analysis Methods 0.000 description 3
- 238000003753 real-time PCR Methods 0.000 description 3
- 230000001105 regulatory effect Effects 0.000 description 3
- 238000011160 research Methods 0.000 description 3
- 230000000241 respiratory effect Effects 0.000 description 3
- 230000035945 sensitivity Effects 0.000 description 3
- 210000003625 skull Anatomy 0.000 description 3
- 239000000600 sorbitol Substances 0.000 description 3
- 230000035924 thermogenesis Effects 0.000 description 3
- 239000003981 vehicle Substances 0.000 description 3
- XLYOFNOQVPJJNP-UHFFFAOYSA-N water Substances O XLYOFNOQVPJJNP-UHFFFAOYSA-N 0.000 description 3
- QKNYBSVHEMOAJP-UHFFFAOYSA-N 2-amino-2-(hydroxymethyl)propane-1,3-diol;hydron;chloride Chemical compound Cl.OCC(N)(CO)CO QKNYBSVHEMOAJP-UHFFFAOYSA-N 0.000 description 2
- WRMNZCZEMHIOCP-UHFFFAOYSA-N 2-phenylethanol Chemical compound OCCC1=CC=CC=C1 WRMNZCZEMHIOCP-UHFFFAOYSA-N 0.000 description 2
- 239000013607 AAV vector Substances 0.000 description 2
- CIWBSHSKHKDKBQ-JLAZNSOCSA-N Ascorbic acid Chemical compound OC[C@H](O)[C@H]1OC(=O)C(O)=C1O CIWBSHSKHKDKBQ-JLAZNSOCSA-N 0.000 description 2
- 241000283707 Capra Species 0.000 description 2
- OMFXVFTZEKFJBZ-UHFFFAOYSA-N Corticosterone Natural products O=C1CCC2(C)C3C(O)CC(C)(C(CC4)C(=O)CO)C4C3CCC2=C1 OMFXVFTZEKFJBZ-UHFFFAOYSA-N 0.000 description 2
- FBPFZTCFMRRESA-JGWLITMVSA-N D-glucitol Chemical compound OC[C@H](O)[C@@H](O)[C@H](O)[C@H](O)CO FBPFZTCFMRRESA-JGWLITMVSA-N 0.000 description 2
- KCXVZYZYPLLWCC-UHFFFAOYSA-N EDTA Chemical compound OC(=O)CN(CC(O)=O)CCN(CC(O)=O)CC(O)=O KCXVZYZYPLLWCC-UHFFFAOYSA-N 0.000 description 2
- 102100028071 Fibroblast growth factor 7 Human genes 0.000 description 2
- WSFSSNUMVMOOMR-UHFFFAOYSA-N Formaldehyde Chemical compound O=C WSFSSNUMVMOOMR-UHFFFAOYSA-N 0.000 description 2
- 102100039289 Glial fibrillary acidic protein Human genes 0.000 description 2
- 239000004471 Glycine Substances 0.000 description 2
- 101001060261 Homo sapiens Fibroblast growth factor 7 Proteins 0.000 description 2
- MHAJPDPJQMAIIY-UHFFFAOYSA-N Hydrogen peroxide Chemical compound OO MHAJPDPJQMAIIY-UHFFFAOYSA-N 0.000 description 2
- 208000013016 Hypoglycemia Diseases 0.000 description 2
- DGAQECJNVWCQMB-PUAWFVPOSA-M Ilexoside XXIX Chemical compound C[C@@H]1CC[C@@]2(CC[C@@]3(C(=CC[C@H]4[C@]3(CC[C@@H]5[C@@]4(CC[C@@H](C5(C)C)OS(=O)(=O)[O-])C)C)[C@@H]2[C@]1(C)O)C)C(=O)O[C@H]6[C@@H]([C@H]([C@@H]([C@H](O6)CO)O)O)O.[Na+] DGAQECJNVWCQMB-PUAWFVPOSA-M 0.000 description 2
- PIWKPBJCKXDKJR-UHFFFAOYSA-N Isoflurane Chemical compound FC(F)OC(Cl)C(F)(F)F PIWKPBJCKXDKJR-UHFFFAOYSA-N 0.000 description 2
- 102000016267 Leptin Human genes 0.000 description 2
- 108010092277 Leptin Proteins 0.000 description 2
- MWUXSHHQAYIFBG-UHFFFAOYSA-N Nitric oxide Chemical compound O=[N] MWUXSHHQAYIFBG-UHFFFAOYSA-N 0.000 description 2
- WCUXLLCKKVVCTQ-UHFFFAOYSA-M Potassium chloride Chemical compound [Cl-].[K+] WCUXLLCKKVVCTQ-UHFFFAOYSA-M 0.000 description 2
- 241000283984 Rodentia Species 0.000 description 2
- 229940124639 Selective inhibitor Drugs 0.000 description 2
- DKGAVHZHDRPRBM-UHFFFAOYSA-N Tert-Butanol Chemical compound CC(C)(C)O DKGAVHZHDRPRBM-UHFFFAOYSA-N 0.000 description 2
- 239000013504 Triton X-100 Substances 0.000 description 2
- 229920004890 Triton X-100 Polymers 0.000 description 2
- 210000000593 adipose tissue white Anatomy 0.000 description 2
- 239000002671 adjuvant Substances 0.000 description 2
- 238000010171 animal model Methods 0.000 description 2
- 208000022531 anorexia Diseases 0.000 description 2
- 239000003963 antioxidant agent Substances 0.000 description 2
- 235000006708 antioxidants Nutrition 0.000 description 2
- 210000003295 arcuate nucleus Anatomy 0.000 description 2
- 238000003556 assay Methods 0.000 description 2
- QVGXLLKOCUKJST-UHFFFAOYSA-N atomic oxygen Chemical compound [O] QVGXLLKOCUKJST-UHFFFAOYSA-N 0.000 description 2
- WPYMKLBDIGXBTP-UHFFFAOYSA-N benzoic acid Chemical compound OC(=O)C1=CC=CC=C1 WPYMKLBDIGXBTP-UHFFFAOYSA-N 0.000 description 2
- 230000015572 biosynthetic process Effects 0.000 description 2
- 230000000903 blocking effect Effects 0.000 description 2
- 239000000872 buffer Substances 0.000 description 2
- RYYVLZVUVIJVGH-UHFFFAOYSA-N caffeine Chemical compound CN1C(=O)N(C)C(=O)C2=C1N=CN2C RYYVLZVUVIJVGH-UHFFFAOYSA-N 0.000 description 2
- 239000003795 chemical substances by application Substances 0.000 description 2
- 235000012000 cholesterol Nutrition 0.000 description 2
- OMFXVFTZEKFJBZ-HJTSIMOOSA-N corticosterone Chemical compound O=C1CC[C@]2(C)[C@H]3[C@@H](O)C[C@](C)([C@H](CC4)C(=O)CO)[C@@H]4[C@@H]3CCC2=C1 OMFXVFTZEKFJBZ-HJTSIMOOSA-N 0.000 description 2
- 206010061428 decreased appetite Diseases 0.000 description 2
- 238000011161 development Methods 0.000 description 2
- 230000018109 developmental process Effects 0.000 description 2
- 239000003085 diluting agent Substances 0.000 description 2
- 229940079593 drug Drugs 0.000 description 2
- 238000005516 engineering process Methods 0.000 description 2
- 230000005284 excitation Effects 0.000 description 2
- 102000052178 fibroblast growth factor receptor activity proteins Human genes 0.000 description 2
- 238000007446 glucose tolerance test Methods 0.000 description 2
- 230000004190 glucose uptake Effects 0.000 description 2
- 102000057231 human FGF4 Human genes 0.000 description 2
- 230000003345 hyperglycaemic effect Effects 0.000 description 2
- 230000004179 hypothalamic–pituitary–adrenal axis Effects 0.000 description 2
- 238000007901 in situ hybridization Methods 0.000 description 2
- 238000011065 in-situ storage Methods 0.000 description 2
- 230000001939 inductive effect Effects 0.000 description 2
- 238000010253 intravenous injection Methods 0.000 description 2
- 229960002725 isoflurane Drugs 0.000 description 2
- NRYBAZVQPHGZNS-ZSOCWYAHSA-N leptin Chemical compound O=C([C@H](CO)NC(=O)[C@H](CC(C)C)NC(=O)[C@H](CC(O)=O)NC(=O)[C@H](CC(C)C)NC(=O)[C@H](CCC(N)=O)NC(=O)[C@H](CC=1C2=CC=CC=C2NC=1)NC(=O)[C@H](CC(C)C)NC(=O)[C@@H](NC(=O)[C@H](CC(O)=O)NC(=O)[C@H](CCC(N)=O)NC(=O)[C@H](CC(C)C)NC(=O)[C@H](CO)NC(=O)CNC(=O)[C@H](CCC(N)=O)NC(=O)[C@@H](N)CC(C)C)CCSC)N1CCC[C@H]1C(=O)NCC(=O)N[C@@H](CS)C(O)=O NRYBAZVQPHGZNS-ZSOCWYAHSA-N 0.000 description 2
- 229940039781 leptin Drugs 0.000 description 2
- 108010019813 leptin receptors Proteins 0.000 description 2
- 102000005861 leptin receptors Human genes 0.000 description 2
- 230000000670 limiting effect Effects 0.000 description 2
- 230000007774 longterm Effects 0.000 description 2
- 230000007246 mechanism Effects 0.000 description 2
- 210000001616 monocyte Anatomy 0.000 description 2
- 238000010172 mouse model Methods 0.000 description 2
- 239000002105 nanoparticle Substances 0.000 description 2
- 210000005036 nerve Anatomy 0.000 description 2
- 239000001301 oxygen Substances 0.000 description 2
- 229910052760 oxygen Inorganic materials 0.000 description 2
- 235000020030 perry Nutrition 0.000 description 2
- 230000002085 persistent effect Effects 0.000 description 2
- 229920001223 polyethylene glycol Polymers 0.000 description 2
- 239000001267 polyvinylpyrrolidone Substances 0.000 description 2
- 229920000036 polyvinylpyrrolidone Polymers 0.000 description 2
- 235000013855 polyvinylpyrrolidone Nutrition 0.000 description 2
- 230000003389 potentiating effect Effects 0.000 description 2
- 238000002360 preparation method Methods 0.000 description 2
- 230000002035 prolonged effect Effects 0.000 description 2
- 239000002994 raw material Substances 0.000 description 2
- 238000003757 reverse transcription PCR Methods 0.000 description 2
- YGSDEFSMJLZEOE-UHFFFAOYSA-N salicylic acid Chemical compound OC(=O)C1=CC=CC=C1O YGSDEFSMJLZEOE-UHFFFAOYSA-N 0.000 description 2
- 239000000523 sample Substances 0.000 description 2
- 239000011734 sodium Substances 0.000 description 2
- GEHJYWRUCIMESM-UHFFFAOYSA-L sodium sulfite Chemical compound [Na+].[Na+].[O-]S([O-])=O GEHJYWRUCIMESM-UHFFFAOYSA-L 0.000 description 2
- 239000002904 solvent Substances 0.000 description 2
- 210000004272 stretch cell Anatomy 0.000 description 2
- 238000001356 surgical procedure Methods 0.000 description 2
- 230000002889 sympathetic effect Effects 0.000 description 2
- 230000001225 therapeutic effect Effects 0.000 description 2
- 210000000211 third ventricle Anatomy 0.000 description 2
- GETQZCLCWQTVFV-UHFFFAOYSA-N trimethylamine Chemical compound CN(C)C GETQZCLCWQTVFV-UHFFFAOYSA-N 0.000 description 2
- 210000003462 vein Anatomy 0.000 description 2
- QYSXJUFSXHHAJI-YRZJJWOYSA-N vitamin D3 Chemical compound C1(/[C@@H]2CC[C@@H]([C@]2(CCC1)C)[C@H](C)CCCC(C)C)=C\C=C1\C[C@@H](O)CCC1=C QYSXJUFSXHHAJI-YRZJJWOYSA-N 0.000 description 2
- JNYAEWCLZODPBN-JGWLITMVSA-N (2r,3r,4s)-2-[(1r)-1,2-dihydroxyethyl]oxolane-3,4-diol Chemical class OC[C@@H](O)[C@H]1OC[C@H](O)[C@H]1O JNYAEWCLZODPBN-JGWLITMVSA-N 0.000 description 1
- UCTWMZQNUQWSLP-VIFPVBQESA-N (R)-adrenaline Chemical compound CNC[C@H](O)C1=CC=C(O)C(O)=C1 UCTWMZQNUQWSLP-VIFPVBQESA-N 0.000 description 1
- 229930182837 (R)-adrenaline Natural products 0.000 description 1
- PZNPLUBHRSSFHT-RRHRGVEJSA-N 1-hexadecanoyl-2-octadecanoyl-sn-glycero-3-phosphocholine Chemical compound CCCCCCCCCCCCCCCCCC(=O)O[C@@H](COP([O-])(=O)OCC[N+](C)(C)C)COC(=O)CCCCCCCCCCCCCCC PZNPLUBHRSSFHT-RRHRGVEJSA-N 0.000 description 1
- IIZPXYDJLKNOIY-JXPKJXOSSA-N 1-palmitoyl-2-arachidonoyl-sn-glycero-3-phosphocholine Chemical compound CCCCCCCCCCCCCCCC(=O)OC[C@H](COP([O-])(=O)OCC[N+](C)(C)C)OC(=O)CCC\C=C/C\C=C/C\C=C/C\C=C/CCCCC IIZPXYDJLKNOIY-JXPKJXOSSA-N 0.000 description 1
- JKMHFZQWWAIEOD-UHFFFAOYSA-N 2-[4-(2-hydroxyethyl)piperazin-1-yl]ethanesulfonic acid Chemical compound OCC[NH+]1CCN(CCS([O-])(=O)=O)CC1 JKMHFZQWWAIEOD-UHFFFAOYSA-N 0.000 description 1
- AOYNUTHNTBLRMT-SLPGGIOYSA-N 2-deoxy-2-fluoro-aldehydo-D-glucose Chemical compound OC[C@@H](O)[C@@H](O)[C@H](O)[C@@H](F)C=O AOYNUTHNTBLRMT-SLPGGIOYSA-N 0.000 description 1
- UFMBOFGKHIXOTA-UHFFFAOYSA-N 2-methylterephthalic acid Chemical compound CC1=CC(C(O)=O)=CC=C1C(O)=O UFMBOFGKHIXOTA-UHFFFAOYSA-N 0.000 description 1
- NAIGRTFQKPRBJX-UHFFFAOYSA-N 2-propylterephthalic acid Chemical compound CCCC1=CC(C(O)=O)=CC=C1C(O)=O NAIGRTFQKPRBJX-UHFFFAOYSA-N 0.000 description 1
- 102000007469 Actins Human genes 0.000 description 1
- 108010085238 Actins Proteins 0.000 description 1
- 102000000263 Anoctamin-4 Human genes 0.000 description 1
- 108050008802 Anoctamin-4 Proteins 0.000 description 1
- 239000004475 Arginine Substances 0.000 description 1
- 235000005781 Avena Nutrition 0.000 description 1
- 244000075850 Avena orientalis Species 0.000 description 1
- 239000005711 Benzoic acid Substances 0.000 description 1
- 101710104526 Beta-klotho Proteins 0.000 description 1
- 102100020683 Beta-klotho Human genes 0.000 description 1
- BVKZGUZCCUSVTD-UHFFFAOYSA-M Bicarbonate Chemical class OC([O-])=O BVKZGUZCCUSVTD-UHFFFAOYSA-M 0.000 description 1
- 241000237519 Bivalvia Species 0.000 description 1
- 241000283690 Bos taurus Species 0.000 description 1
- 241001631457 Cannula Species 0.000 description 1
- 241000700199 Cavia porcellus Species 0.000 description 1
- 206010008401 Changes in physical activity Diseases 0.000 description 1
- GHXZTYHSJHQHIJ-UHFFFAOYSA-N Chlorhexidine Chemical compound C=1C=C(Cl)C=CC=1NC(N)=NC(N)=NCCCCCCN=C(N)N=C(N)NC1=CC=C(Cl)C=C1 GHXZTYHSJHQHIJ-UHFFFAOYSA-N 0.000 description 1
- 102000011045 Chloride Channels Human genes 0.000 description 1
- 108010062745 Chloride Channels Proteins 0.000 description 1
- 229920000858 Cyclodextrin Polymers 0.000 description 1
- 108090000695 Cytokines Proteins 0.000 description 1
- 102000004127 Cytokines Human genes 0.000 description 1
- WQZGKKKJIJFFOK-QTVWNMPRSA-N D-mannopyranose Chemical compound OC[C@H]1OC(O)[C@@H](O)[C@@H](O)[C@@H]1O WQZGKKKJIJFFOK-QTVWNMPRSA-N 0.000 description 1
- 239000004375 Dextrin Substances 0.000 description 1
- 229920001353 Dextrin Polymers 0.000 description 1
- 208000032781 Diabetic cardiomyopathy Diseases 0.000 description 1
- 101150086096 Eif2ak3 gene Proteins 0.000 description 1
- 241000283073 Equus caballus Species 0.000 description 1
- 241000588724 Escherichia coli Species 0.000 description 1
- 108010007005 Estrogen Receptor alpha Proteins 0.000 description 1
- 102100038595 Estrogen receptor Human genes 0.000 description 1
- CTKXFMQHOOWWEB-UHFFFAOYSA-N Ethylene oxide/propylene oxide copolymer Chemical compound CCCOC(C)COCCO CTKXFMQHOOWWEB-UHFFFAOYSA-N 0.000 description 1
- 108010037362 Extracellular Matrix Proteins Proteins 0.000 description 1
- 102000010834 Extracellular Matrix Proteins Human genes 0.000 description 1
- 239000001116 FEMA 4028 Substances 0.000 description 1
- 101150081880 FGF1 gene Proteins 0.000 description 1
- 101710153349 Fibroblast growth factor 19 Proteins 0.000 description 1
- 102000003974 Fibroblast growth factor 2 Human genes 0.000 description 1
- 108090000379 Fibroblast growth factor 2 Proteins 0.000 description 1
- 102100031361 Fibroblast growth factor 20 Human genes 0.000 description 1
- 102100037680 Fibroblast growth factor 8 Human genes 0.000 description 1
- 102100037665 Fibroblast growth factor 9 Human genes 0.000 description 1
- 108090000367 Fibroblast growth factor 9 Proteins 0.000 description 1
- 101150052535 GYS2 gene Proteins 0.000 description 1
- 241000287828 Gallus gallus Species 0.000 description 1
- 102000053171 Glial Fibrillary Acidic Human genes 0.000 description 1
- 102400000321 Glucagon Human genes 0.000 description 1
- 108060003199 Glucagon Proteins 0.000 description 1
- 239000007995 HEPES buffer Substances 0.000 description 1
- 238000013218 HFD mouse model Methods 0.000 description 1
- 102000002812 Heat-Shock Proteins Human genes 0.000 description 1
- 108010004889 Heat-Shock Proteins Proteins 0.000 description 1
- 229920002971 Heparan sulfate Polymers 0.000 description 1
- 241000282412 Homo Species 0.000 description 1
- 101100120051 Homo sapiens FGF1 gene Proteins 0.000 description 1
- 101000846532 Homo sapiens Fibroblast growth factor 20 Proteins 0.000 description 1
- 101001027382 Homo sapiens Fibroblast growth factor 8 Proteins 0.000 description 1
- 101001105486 Homo sapiens Proteasome subunit alpha type-7 Proteins 0.000 description 1
- 101000575685 Homo sapiens Synembryn-B Proteins 0.000 description 1
- 108060003951 Immunoglobulin Proteins 0.000 description 1
- 229940122355 Insulin sensitizer Drugs 0.000 description 1
- LPHGQDQBBGAPDZ-UHFFFAOYSA-N Isocaffeine Natural products CN1C(=O)N(C)C(=O)C2=C1N(C)C=N2 LPHGQDQBBGAPDZ-UHFFFAOYSA-N 0.000 description 1
- ODKSFYDXXFIFQN-BYPYZUCNSA-P L-argininium(2+) Chemical compound NC(=[NH2+])NCCC[C@H]([NH3+])C(O)=O ODKSFYDXXFIFQN-BYPYZUCNSA-P 0.000 description 1
- CKLJMWTZIZZHCS-REOHCLBHSA-N L-aspartic acid Chemical compound OC(=O)[C@@H](N)CC(O)=O CKLJMWTZIZZHCS-REOHCLBHSA-N 0.000 description 1
- ZDXPYRJPNDTMRX-VKHMYHEASA-N L-glutamine Chemical compound OC(=O)[C@@H](N)CCC(N)=O ZDXPYRJPNDTMRX-VKHMYHEASA-N 0.000 description 1
- KDXKERNSBIXSRK-YFKPBYRVSA-N L-lysine Chemical compound NCCCC[C@H](N)C(O)=O KDXKERNSBIXSRK-YFKPBYRVSA-N 0.000 description 1
- KDXKERNSBIXSRK-UHFFFAOYSA-N Lysine Natural products NCCCCC(N)C(O)=O KDXKERNSBIXSRK-UHFFFAOYSA-N 0.000 description 1
- 239000004472 Lysine Substances 0.000 description 1
- 241000124008 Mammalia Species 0.000 description 1
- 108010008364 Melanocortins Proteins 0.000 description 1
- 101001129925 Mus musculus Leptin receptor Proteins 0.000 description 1
- 101100370100 Mus musculus Tor3a gene Proteins 0.000 description 1
- KCWZGJVSDFYRIX-YFKPBYRVSA-N N(gamma)-nitro-L-arginine methyl ester Chemical compound COC(=O)[C@@H](N)CCCN=C(N)N[N+]([O-])=O KCWZGJVSDFYRIX-YFKPBYRVSA-N 0.000 description 1
- 230000004988 N-glycosylation Effects 0.000 description 1
- 102100022397 Nitric oxide synthase, brain Human genes 0.000 description 1
- 101710111444 Nitric oxide synthase, brain Proteins 0.000 description 1
- 101150039326 PCK1 gene Proteins 0.000 description 1
- 238000009004 PCR Kit Methods 0.000 description 1
- 229910019142 PO4 Inorganic materials 0.000 description 1
- 241000009328 Perro Species 0.000 description 1
- 101150025052 Pklr gene Proteins 0.000 description 1
- 239000002202 Polyethylene glycol Substances 0.000 description 1
- 229920001213 Polysorbate 20 Polymers 0.000 description 1
- HLCFGWHYROZGBI-JJKGCWMISA-M Potassium gluconate Chemical compound [K+].OC[C@@H](O)[C@@H](O)[C@H](O)[C@@H](O)C([O-])=O HLCFGWHYROZGBI-JJKGCWMISA-M 0.000 description 1
- 102100021201 Proteasome subunit alpha type-7 Human genes 0.000 description 1
- 206010037660 Pyrexia Diseases 0.000 description 1
- 238000011529 RT qPCR Methods 0.000 description 1
- 102000007562 Serum Albumin Human genes 0.000 description 1
- 108010071390 Serum Albumin Proteins 0.000 description 1
- DWAQJAXMDSEUJJ-UHFFFAOYSA-M Sodium bisulfite Chemical compound [Na+].OS([O-])=O DWAQJAXMDSEUJJ-UHFFFAOYSA-M 0.000 description 1
- 239000012505 Superdex™ Substances 0.000 description 1
- 241000282898 Sus scrofa Species 0.000 description 1
- 102100026014 Synembryn-B Human genes 0.000 description 1
- 102000008219 Uncoupling Protein 2 Human genes 0.000 description 1
- 108010021111 Uncoupling Protein 2 Proteins 0.000 description 1
- 230000002159 abnormal effect Effects 0.000 description 1
- 230000009471 action Effects 0.000 description 1
- 239000004480 active ingredient Substances 0.000 description 1
- 230000003044 adaptive effect Effects 0.000 description 1
- 239000000853 adhesive Substances 0.000 description 1
- 230000001070 adhesive effect Effects 0.000 description 1
- 210000001943 adrenal medulla Anatomy 0.000 description 1
- 229910052783 alkali metal Inorganic materials 0.000 description 1
- 150000001340 alkali metals Chemical class 0.000 description 1
- 230000004075 alteration Effects 0.000 description 1
- 235000001014 amino acid Nutrition 0.000 description 1
- 150000001413 amino acids Chemical class 0.000 description 1
- 230000003321 amplification Effects 0.000 description 1
- 230000019552 anatomical structure morphogenesis Effects 0.000 description 1
- 210000003484 anatomy Anatomy 0.000 description 1
- 230000001772 anti-angiogenic effect Effects 0.000 description 1
- 239000004599 antimicrobial Substances 0.000 description 1
- ODKSFYDXXFIFQN-UHFFFAOYSA-N arginine Natural products OC(=O)C(N)CCCNC(N)=N ODKSFYDXXFIFQN-UHFFFAOYSA-N 0.000 description 1
- 238000013528 artificial neural network Methods 0.000 description 1
- 235000010323 ascorbic acid Nutrition 0.000 description 1
- 239000011668 ascorbic acid Substances 0.000 description 1
- 229960005070 ascorbic acid Drugs 0.000 description 1
- 235000003704 aspartic acid Nutrition 0.000 description 1
- 230000003305 autocrine Effects 0.000 description 1
- 210000000227 basophil cell of anterior lobe of hypophysis Anatomy 0.000 description 1
- 230000009286 beneficial effect Effects 0.000 description 1
- 235000010233 benzoic acid Nutrition 0.000 description 1
- 229960004365 benzoic acid Drugs 0.000 description 1
- OQFSQFPPLPISGP-UHFFFAOYSA-N beta-carboxyaspartic acid Natural products OC(=O)C(N)C(C(O)=O)C(O)=O OQFSQFPPLPISGP-UHFFFAOYSA-N 0.000 description 1
- WHGYBXFWUBPSRW-FOUAGVGXSA-N beta-cyclodextrin Chemical compound OC[C@H]([C@H]([C@@H]([C@H]1O)O)O[C@H]2O[C@@H]([C@@H](O[C@H]3O[C@H](CO)[C@H]([C@@H]([C@H]3O)O)O[C@H]3O[C@H](CO)[C@H]([C@@H]([C@H]3O)O)O[C@H]3O[C@H](CO)[C@H]([C@@H]([C@H]3O)O)O[C@H]3O[C@H](CO)[C@H]([C@@H]([C@H]3O)O)O3)[C@H](O)[C@H]2O)CO)O[C@@H]1O[C@H]1[C@H](O)[C@@H](O)[C@@H]3O[C@@H]1CO WHGYBXFWUBPSRW-FOUAGVGXSA-N 0.000 description 1
- 235000011175 beta-cyclodextrine Nutrition 0.000 description 1
- 229960004853 betadex Drugs 0.000 description 1
- 239000003124 biologic agent Substances 0.000 description 1
- 230000008827 biological function Effects 0.000 description 1
- 230000008512 biological response Effects 0.000 description 1
- IISBACLAFKSPIT-UHFFFAOYSA-N bisphenol A Chemical group C=1C=C(O)C=CC=1C(C)(C)C1=CC=C(O)C=C1 IISBACLAFKSPIT-UHFFFAOYSA-N 0.000 description 1
- 150000001642 boronic acid derivatives Chemical class 0.000 description 1
- 239000005388 borosilicate glass Substances 0.000 description 1
- 229960001948 caffeine Drugs 0.000 description 1
- VJEONQKOZGKCAK-UHFFFAOYSA-N caffeine Natural products CN1C(=O)N(C)C(=O)C2=C1C=CN2C VJEONQKOZGKCAK-UHFFFAOYSA-N 0.000 description 1
- 150000001720 carbohydrates Chemical class 0.000 description 1
- 235000014633 carbohydrates Nutrition 0.000 description 1
- 230000000747 cardiac effect Effects 0.000 description 1
- 239000000969 carrier Substances 0.000 description 1
- 230000003915 cell function Effects 0.000 description 1
- 230000004663 cell proliferation Effects 0.000 description 1
- 230000007248 cellular mechanism Effects 0.000 description 1
- 230000019522 cellular metabolic process Effects 0.000 description 1
- 239000004568 cement Substances 0.000 description 1
- 238000005119 centrifugation Methods 0.000 description 1
- 210000001175 cerebrospinal fluid Anatomy 0.000 description 1
- 238000012512 characterization method Methods 0.000 description 1
- 239000002738 chelating agent Substances 0.000 description 1
- 238000006243 chemical reaction Methods 0.000 description 1
- 239000003153 chemical reaction reagent Substances 0.000 description 1
- 229960003260 chlorhexidine Drugs 0.000 description 1
- 235000020639 clam Nutrition 0.000 description 1
- 238000003776 cleavage reaction Methods 0.000 description 1
- 238000010367 cloning Methods 0.000 description 1
- 239000003086 colorant Substances 0.000 description 1
- 239000002299 complementary DNA Substances 0.000 description 1
- 239000008139 complexing agent Substances 0.000 description 1
- 238000002591 computed tomography Methods 0.000 description 1
- 239000000356 contaminant Substances 0.000 description 1
- 230000003247 decreasing effect Effects 0.000 description 1
- 230000007812 deficiency Effects 0.000 description 1
- 230000002950 deficient Effects 0.000 description 1
- 238000012217 deletion Methods 0.000 description 1
- 230000037430 deletion Effects 0.000 description 1
- 210000003520 dendritic spine Anatomy 0.000 description 1
- 239000003479 dental cement Substances 0.000 description 1
- 238000013461 design Methods 0.000 description 1
- 238000001514 detection method Methods 0.000 description 1
- 235000019425 dextrin Nutrition 0.000 description 1
- UQLDLKMNUJERMK-UHFFFAOYSA-L di(octadecanoyloxy)lead Chemical compound [Pb+2].CCCCCCCCCCCCCCCCCC([O-])=O.CCCCCCCCCCCCCCCCCC([O-])=O UQLDLKMNUJERMK-UHFFFAOYSA-L 0.000 description 1
- 238000013118 diabetic mouse model Methods 0.000 description 1
- 238000010586 diagram Methods 0.000 description 1
- 238000013229 diet-induced obese mouse Methods 0.000 description 1
- 235000020931 dietary conditions Nutrition 0.000 description 1
- 238000009792 diffusion process Methods 0.000 description 1
- 239000000539 dimer Substances 0.000 description 1
- 238000006471 dimerization reaction Methods 0.000 description 1
- 230000000447 dimerizing effect Effects 0.000 description 1
- 230000003467 diminishing effect Effects 0.000 description 1
- 150000002016 disaccharides Chemical class 0.000 description 1
- 201000010099 disease Diseases 0.000 description 1
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 description 1
- 238000004090 dissolution Methods 0.000 description 1
- 238000009826 distribution Methods 0.000 description 1
- 239000012154 double-distilled water Substances 0.000 description 1
- 239000003937 drug carrier Substances 0.000 description 1
- 238000012377 drug delivery Methods 0.000 description 1
- 238000002296 dynamic light scattering Methods 0.000 description 1
- 230000000081 effect on glucose Effects 0.000 description 1
- 230000013020 embryo development Effects 0.000 description 1
- 239000003995 emulsifying agent Substances 0.000 description 1
- 229960005139 epinephrine Drugs 0.000 description 1
- BEFDCLMNVWHSGT-UHFFFAOYSA-N ethenylcyclopentane Chemical compound C=CC1CCCC1 BEFDCLMNVWHSGT-UHFFFAOYSA-N 0.000 description 1
- 238000013401 experimental design Methods 0.000 description 1
- 210000002744 extracellular matrix Anatomy 0.000 description 1
- 239000000945 filler Substances 0.000 description 1
- 230000004992 fission Effects 0.000 description 1
- 239000000796 flavoring agent Substances 0.000 description 1
- 235000019634 flavors Nutrition 0.000 description 1
- 239000012530 fluid Substances 0.000 description 1
- 235000021588 free fatty acids Nutrition 0.000 description 1
- 230000002496 gastric effect Effects 0.000 description 1
- 101150036914 gck gene Proteins 0.000 description 1
- 239000000499 gel Substances 0.000 description 1
- 230000030279 gene silencing Effects 0.000 description 1
- 238000002695 general anesthesia Methods 0.000 description 1
- 239000011521 glass Substances 0.000 description 1
- 229960004666 glucagon Drugs 0.000 description 1
- MASNOZXLGMXCHN-ZLPAWPGGSA-N glucagon Chemical compound C([C@@H](C(=O)N[C@H](C(=O)N[C@@H](CCC(N)=O)C(=O)N[C@@H](CC=1C2=CC=CC=C2NC=1)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CCSC)C(=O)N[C@@H](CC(N)=O)C(=O)N[C@@H]([C@@H](C)O)C(O)=O)C(C)C)NC(=O)[C@H](CC(O)=O)NC(=O)[C@H](CCC(N)=O)NC(=O)[C@H](C)NC(=O)[C@H](CCCNC(N)=N)NC(=O)[C@H](CCCNC(N)=N)NC(=O)[C@H](CO)NC(=O)[C@H](CC(O)=O)NC(=O)[C@H](CC(C)C)NC(=O)[C@H](CC=1C=CC(O)=CC=1)NC(=O)[C@H](CCCCN)NC(=O)[C@H](CO)NC(=O)[C@H](CC=1C=CC(O)=CC=1)NC(=O)[C@H](CC(O)=O)NC(=O)[C@H](CO)NC(=O)[C@@H](NC(=O)[C@H](CC=1C=CC=CC=1)NC(=O)[C@@H](NC(=O)CNC(=O)[C@H](CCC(N)=O)NC(=O)[C@H](CO)NC(=O)[C@@H](N)CC=1NC=NC=1)[C@@H](C)O)[C@@H](C)O)C1=CC=CC=C1 MASNOZXLGMXCHN-ZLPAWPGGSA-N 0.000 description 1
- 230000001890 gluconeogenic effect Effects 0.000 description 1
- 229960001031 glucose Drugs 0.000 description 1
- 230000014101 glucose homeostasis Effects 0.000 description 1
- 125000002791 glucosyl group Chemical group C1([C@H](O)[C@@H](O)[C@H](O)[C@H](O1)CO)* 0.000 description 1
- ZDXPYRJPNDTMRX-UHFFFAOYSA-N glutamine Natural products OC(=O)C(N)CCC(N)=O ZDXPYRJPNDTMRX-UHFFFAOYSA-N 0.000 description 1
- 230000034659 glycolysis Effects 0.000 description 1
- 230000002414 glycolytic effect Effects 0.000 description 1
- 238000009499 grossing Methods 0.000 description 1
- 239000003102 growth factor Substances 0.000 description 1
- 229940093915 gynecological organic acid Drugs 0.000 description 1
- 230000003284 homeostatic effect Effects 0.000 description 1
- 229960002163 hydrogen peroxide Drugs 0.000 description 1
- 229920001477 hydrophilic polymer Polymers 0.000 description 1
- 238000003384 imaging method Methods 0.000 description 1
- 102000018358 immunoglobulin Human genes 0.000 description 1
- 238000003364 immunohistochemistry Methods 0.000 description 1
- 238000002513 implantation Methods 0.000 description 1
- 238000000338 in vitro Methods 0.000 description 1
- 238000001727 in vivo Methods 0.000 description 1
- 238000011534 incubation Methods 0.000 description 1
- 230000006698 induction Effects 0.000 description 1
- 230000036512 infertility Effects 0.000 description 1
- 238000001802 infusion Methods 0.000 description 1
- 230000003914 insulin secretion Effects 0.000 description 1
- 230000003993 interaction Effects 0.000 description 1
- 239000007928 intraperitoneal injection Substances 0.000 description 1
- 238000007914 intraventricular administration Methods 0.000 description 1
- 238000011835 investigation Methods 0.000 description 1
- 229930027917 kanamycin Natural products 0.000 description 1
- SBUJHOSQTJFQJX-NOAMYHISSA-N kanamycin Chemical compound O[C@@H]1[C@@H](O)[C@H](O)[C@@H](CN)O[C@@H]1O[C@H]1[C@H](O)[C@@H](O[C@@H]2[C@@H]([C@@H](N)[C@H](O)[C@@H](CO)O2)O)[C@H](N)C[C@@H]1N SBUJHOSQTJFQJX-NOAMYHISSA-N 0.000 description 1
- 229960000318 kanamycin Drugs 0.000 description 1
- 229930182823 kanamycin A Natural products 0.000 description 1
- 238000011813 knockout mouse model Methods 0.000 description 1
- 230000002045 lasting effect Effects 0.000 description 1
- 239000000787 lecithin Substances 0.000 description 1
- 235000010445 lecithin Nutrition 0.000 description 1
- 229940067606 lecithin Drugs 0.000 description 1
- 230000005923 long-lasting effect Effects 0.000 description 1
- 239000006166 lysate Substances 0.000 description 1
- 230000010311 mammalian development Effects 0.000 description 1
- 238000005259 measurement Methods 0.000 description 1
- 239000002865 melanocortin Substances 0.000 description 1
- 230000007102 metabolic function Effects 0.000 description 1
- 230000037323 metabolic rate Effects 0.000 description 1
- DJGAAPFSPWAYTJ-UHFFFAOYSA-M metamizole sodium Chemical compound [Na+].O=C1C(N(CS([O-])(=O)=O)C)=C(C)N(C)N1C1=CC=CC=C1 DJGAAPFSPWAYTJ-UHFFFAOYSA-M 0.000 description 1
- QUMVIIAJMDRLJX-JEDNCBNOSA-N methyl (2s)-2-amino-5-[carbamimidoyl(nitro)amino]pentanoate;hydrochloride Chemical compound Cl.COC(=O)[C@@H](N)CCCN(C(N)=N)[N+]([O-])=O QUMVIIAJMDRLJX-JEDNCBNOSA-N 0.000 description 1
- 238000004377 microelectronic Methods 0.000 description 1
- 230000027939 micturition Effects 0.000 description 1
- 230000003278 mimic effect Effects 0.000 description 1
- 230000002438 mitochondrial effect Effects 0.000 description 1
- 230000000394 mitotic effect Effects 0.000 description 1
- 239000011259 mixed solution Substances 0.000 description 1
- 230000004048 modification Effects 0.000 description 1
- 238000012986 modification Methods 0.000 description 1
- 239000002808 molecular sieve Substances 0.000 description 1
- 238000012544 monitoring process Methods 0.000 description 1
- 150000002772 monosaccharides Chemical class 0.000 description 1
- 210000003205 muscle Anatomy 0.000 description 1
- 230000035772 mutation Effects 0.000 description 1
- 230000000869 mutational effect Effects 0.000 description 1
- 210000004165 myocardium Anatomy 0.000 description 1
- 239000013642 negative control Substances 0.000 description 1
- 210000000653 nervous system Anatomy 0.000 description 1
- 230000008904 neural response Effects 0.000 description 1
- 230000007658 neurological function Effects 0.000 description 1
- 238000003199 nucleic acid amplification method Methods 0.000 description 1
- 238000001543 one-way ANOVA Methods 0.000 description 1
- 230000003287 optical effect Effects 0.000 description 1
- 210000000056 organ Anatomy 0.000 description 1
- 230000005305 organ development Effects 0.000 description 1
- 150000007524 organic acids Chemical class 0.000 description 1
- 235000005985 organic acids Nutrition 0.000 description 1
- 210000000496 pancreas Anatomy 0.000 description 1
- FJKROLUGYXJWQN-UHFFFAOYSA-N papa-hydroxy-benzoic acid Natural products OC(=O)C1=CC=C(O)C=C1 FJKROLUGYXJWQN-UHFFFAOYSA-N 0.000 description 1
- 230000036961 partial effect Effects 0.000 description 1
- 239000002245 particle Substances 0.000 description 1
- 244000052769 pathogen Species 0.000 description 1
- 230000007170 pathology Effects 0.000 description 1
- 230000007310 pathophysiology Effects 0.000 description 1
- 230000037361 pathway Effects 0.000 description 1
- 229960001412 pentobarbital Drugs 0.000 description 1
- WEXRUCMBJFQVBZ-UHFFFAOYSA-N pentobarbital Chemical compound CCCC(C)C1(CC)C(=O)NC(=O)NC1=O WEXRUCMBJFQVBZ-UHFFFAOYSA-N 0.000 description 1
- 230000000737 periodic effect Effects 0.000 description 1
- 230000009955 peripheral mechanism Effects 0.000 description 1
- 230000035699 permeability Effects 0.000 description 1
- 239000000546 pharmaceutical excipient Substances 0.000 description 1
- 230000000144 pharmacologic effect Effects 0.000 description 1
- 230000009038 pharmacological inhibition Effects 0.000 description 1
- 229940067107 phenylethyl alcohol Drugs 0.000 description 1
- -1 phenylmethanoxy chloride Chemical compound 0.000 description 1
- 235000021317 phosphate Nutrition 0.000 description 1
- 239000008363 phosphate buffer Substances 0.000 description 1
- 150000003013 phosphoric acid derivatives Chemical class 0.000 description 1
- IYDGMDWEHDFVQI-UHFFFAOYSA-N phosphoric acid;trioxotungsten Chemical compound O=[W](=O)=O.O=[W](=O)=O.O=[W](=O)=O.O=[W](=O)=O.O=[W](=O)=O.O=[W](=O)=O.O=[W](=O)=O.O=[W](=O)=O.O=[W](=O)=O.O=[W](=O)=O.O=[W](=O)=O.O=[W](=O)=O.OP(O)(O)=O IYDGMDWEHDFVQI-UHFFFAOYSA-N 0.000 description 1
- 230000035479 physiological effects, processes and functions Effects 0.000 description 1
- 239000002504 physiological saline solution Substances 0.000 description 1
- 229920001993 poloxamer 188 Polymers 0.000 description 1
- 229940044519 poloxamer 188 Drugs 0.000 description 1
- 239000000256 polyoxyethylene sorbitan monolaurate Substances 0.000 description 1
- 235000010486 polyoxyethylene sorbitan monolaurate Nutrition 0.000 description 1
- 239000000244 polyoxyethylene sorbitan monooleate Substances 0.000 description 1
- 235000010482 polyoxyethylene sorbitan monooleate Nutrition 0.000 description 1
- 229920000136 polysorbate Polymers 0.000 description 1
- 229940068977 polysorbate 20 Drugs 0.000 description 1
- 229940068968 polysorbate 80 Drugs 0.000 description 1
- 229920000053 polysorbate 80 Polymers 0.000 description 1
- 229940068965 polysorbates Drugs 0.000 description 1
- 239000001103 potassium chloride Substances 0.000 description 1
- 235000011164 potassium chloride Nutrition 0.000 description 1
- 239000004224 potassium gluconate Substances 0.000 description 1
- 229960003189 potassium gluconate Drugs 0.000 description 1
- 235000013926 potassium gluconate Nutrition 0.000 description 1
- 239000002243 precursor Substances 0.000 description 1
- 238000004321 preservation Methods 0.000 description 1
- 239000003755 preservative agent Substances 0.000 description 1
- 230000002265 prevention Effects 0.000 description 1
- 230000008569 process Effects 0.000 description 1
- 230000000069 prophylactic effect Effects 0.000 description 1
- 239000002510 pyrogen Substances 0.000 description 1
- 238000011002 quantification Methods 0.000 description 1
- 108091008598 receptor tyrosine kinases Proteins 0.000 description 1
- 102000027426 receptor tyrosine kinases Human genes 0.000 description 1
- 108091006091 regulatory enzymes Proteins 0.000 description 1
- 238000007634 remodeling Methods 0.000 description 1
- 238000012552 review Methods 0.000 description 1
- 238000011808 rodent model Methods 0.000 description 1
- 229960004889 salicylic acid Drugs 0.000 description 1
- 230000007017 scission Effects 0.000 description 1
- 238000001338 self-assembly Methods 0.000 description 1
- 210000002966 serum Anatomy 0.000 description 1
- 230000019491 signal transduction Effects 0.000 description 1
- 230000012488 skeletal system development Effects 0.000 description 1
- 229910052708 sodium Inorganic materials 0.000 description 1
- 235000015424 sodium Nutrition 0.000 description 1
- URGAHOPLAPQHLN-UHFFFAOYSA-N sodium aluminosilicate Chemical compound [Na+].[Al+3].[O-][Si]([O-])=O.[O-][Si]([O-])=O URGAHOPLAPQHLN-UHFFFAOYSA-N 0.000 description 1
- 238000002415 sodium dodecyl sulfate polyacrylamide gel electrophoresis Methods 0.000 description 1
- 235000010267 sodium hydrogen sulphite Nutrition 0.000 description 1
- 235000010265 sodium sulphite Nutrition 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
- 238000001179 sorption measurement Methods 0.000 description 1
- 239000008347 soybean phospholipid Substances 0.000 description 1
- 238000004611 spectroscopical analysis Methods 0.000 description 1
- 230000002269 spontaneous effect Effects 0.000 description 1
- 239000003381 stabilizer Substances 0.000 description 1
- 238000010186 staining Methods 0.000 description 1
- 238000007619 statistical method Methods 0.000 description 1
- 210000000130 stem cell Anatomy 0.000 description 1
- 230000000638 stimulation Effects 0.000 description 1
- 239000002438 stress hormone Substances 0.000 description 1
- 150000005846 sugar alcohols Chemical class 0.000 description 1
- 239000000725 suspension Substances 0.000 description 1
- 238000003786 synthesis reaction Methods 0.000 description 1
- 230000009885 systemic effect Effects 0.000 description 1
- 238000002560 therapeutic procedure Methods 0.000 description 1
- RTKIYNMVFMVABJ-UHFFFAOYSA-L thimerosal Chemical compound [Na+].CC[Hg]SC1=CC=CC=C1C([O-])=O RTKIYNMVFMVABJ-UHFFFAOYSA-L 0.000 description 1
- 229940033663 thimerosal Drugs 0.000 description 1
- 230000035922 thirst Effects 0.000 description 1
- 230000030968 tissue homeostasis Effects 0.000 description 1
- 230000017423 tissue regeneration Effects 0.000 description 1
- 238000012546 transfer Methods 0.000 description 1
- 230000001131 transforming effect Effects 0.000 description 1
- 230000014616 translation Effects 0.000 description 1
- 238000004627 transmission electron microscopy Methods 0.000 description 1
- ODLHGICHYURWBS-LKONHMLTSA-N trappsol cyclo Chemical compound CC(O)COC[C@H]([C@H]([C@@H]([C@H]1O)O)O[C@H]2O[C@@H]([C@@H](O[C@H]3O[C@H](COCC(C)O)[C@H]([C@@H]([C@H]3O)O)O[C@H]3O[C@H](COCC(C)O)[C@H]([C@@H]([C@H]3O)O)O[C@H]3O[C@H](COCC(C)O)[C@H]([C@@H]([C@H]3O)O)O[C@H]3O[C@H](COCC(C)O)[C@H]([C@@H]([C@H]3O)O)O3)[C@H](O)[C@H]2O)COCC(O)C)O[C@@H]1O[C@H]1[C@H](O)[C@@H](O)[C@@H]3O[C@@H]1COCC(C)O ODLHGICHYURWBS-LKONHMLTSA-N 0.000 description 1
- UFTFJSFQGQCHQW-UHFFFAOYSA-N triformin Chemical compound O=COCC(OC=O)COC=O UFTFJSFQGQCHQW-UHFFFAOYSA-N 0.000 description 1
- GPRLSGONYQIRFK-MNYXATJNSA-N triton Chemical compound [3H+] GPRLSGONYQIRFK-MNYXATJNSA-N 0.000 description 1
- 238000007492 two-way ANOVA Methods 0.000 description 1
- MDYZKJNTKZIUSK-UHFFFAOYSA-N tyloxapol Chemical compound O=C.C1CO1.CC(C)(C)CC(C)(C)C1=CC=C(O)C=C1 MDYZKJNTKZIUSK-UHFFFAOYSA-N 0.000 description 1
- 229920001664 tyloxapol Polymers 0.000 description 1
- 229960004224 tyloxapol Drugs 0.000 description 1
- 238000002604 ultrasonography Methods 0.000 description 1
- 241000701447 unidentified baculovirus Species 0.000 description 1
- 238000012762 unpaired Student’s t-test Methods 0.000 description 1
- 238000010865 video microscopy Methods 0.000 description 1
- 235000019786 weight gain Nutrition 0.000 description 1
Images
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
- C07K14/50—Fibroblast growth factor [FGF]
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P3/00—Drugs for disorders of the metabolism
- A61P3/04—Anorexiants; Antiobesity agents
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P3/00—Drugs for disorders of the metabolism
- A61P3/08—Drugs for disorders of the metabolism for glucose homeostasis
- A61P3/10—Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P5/00—Drugs for disorders of the endocrine system
- A61P5/48—Drugs for disorders of the endocrine system of the pancreatic hormones
- A61P5/50—Drugs for disorders of the endocrine system of the pancreatic hormones for increasing or potentiating the activity of insulin
Landscapes
- Health & Medical Sciences (AREA)
- Diabetes (AREA)
- Chemical & Material Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Organic Chemistry (AREA)
- General Health & Medical Sciences (AREA)
- Medicinal Chemistry (AREA)
- Pharmacology & Pharmacy (AREA)
- Public Health (AREA)
- Veterinary Medicine (AREA)
- Animal Behavior & Ethology (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- General Chemical & Material Sciences (AREA)
- Chemical Kinetics & Catalysis (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Engineering & Computer Science (AREA)
- Hematology (AREA)
- Endocrinology (AREA)
- Obesity (AREA)
- Biochemistry (AREA)
- Molecular Biology (AREA)
- Toxicology (AREA)
- Zoology (AREA)
- Biophysics (AREA)
- Genetics & Genomics (AREA)
- Emergency Medicine (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- Gastroenterology & Hepatology (AREA)
- Child & Adolescent Psychology (AREA)
- Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
Abstract
Fibroblast growth factor 4 (FGF 4) and Fgfr1 binding ligands are provided for use in the treatment of metabolic disorders, methods of treatment using fibroblast growth factor 4 (FGF 4) and Fgfr1 binding ligands, and use of fibroblast growth factor 4 (FGF 4) and Fgfr1 binding ligands in the manufacture of a medicament.
Description
Technical Field
The present invention relates to FGF4 and Fgfr1 binding ligands for use in the treatment of metabolic diseases, methods of treatment using FGF4 and Fgfr1 binding ligands, and the use of FGF4 and Fgfr1 binding ligands in the manufacture of a medicament.
Background
The Fibroblast Growth Factor (FGF) family contains 22 structurally related polypeptides that act in an autocrine, paracrine or endocrine manner, mediating many key events in mammalian development, tissue homeostasis/repair, and body metabolism (Beenken and Mohammadi,2009 itoh and Ornitz, 2011. Over the past decades, there has been substantial evidence that endocrine members FGF19 and FGF21, as well as the paracrine member FGF1, have great potential for the treatment of obesity and type 2 diabetes (T2D) (degrolamo et al, 2016 fu et al, 2004 huang et al, 2017 jonker et al, 2012 lan et al, 2017. Notably, a single dose of FGF1 administered to the Central Nervous System (CNS) induces sustained diabetes remission in T2D rodents, in contrast to the transient effects seen following central administration of endocrine FGFs (Morton et al, 2013 perry et al, 2015 sarrouf et al, 2010 scarlet et al, 2016. However, the mechanism by which FGF1 induces this sustained action remains unclear. FGF1 administration was associated with transient reduction of food intake, stimulation of hepatic glucose uptake, and preservation of islet β cells (Scarlett et al, 2019, tennant et al, 2019. Studies also suggest that the assembly of monocytes, glial cells and neuronal networks (PNN) at the arcuate nucleus and adjacent intermediate bulge (ARC-ME) sites may play an important role in the sustained hypoglycemic effect of FGF1 (Alonge et al, 2020 bentsen et al, 2020. For example, central administration of FGF1 rapidly induces activation of MAPK-ERK signaling in monocytes and results in increased coverage of dendritic spines by processes from perisynaptic astrocytes in the ARC neurons of ob/ob mice (Bentsen et al, 2020, brown et al, 2019. Although not essential for initial anorexia and glucose lowering, melanocortin signaling and PNN within the ARC are partially essential for sustained action (Alonge et al, 2020. However, the key receptors and cellular mechanisms that mediate this sustained action remain unclear. Also, it is not known whether this surprising antidiabetic activity is unique to FGF1 in the FGF family. These gaps in knowledge have prevented the development of more effective methods for FGF-based treatment of T2D.
FGF mediates multiple biological functions by binding, dimerizing and thereby activating seven subtypes of homologous FGF receptors (FGFRs), each FGFR subtype having a unique spatio-temporal expression profile (Fon Tacer et al, 2010 goetz and Mohammadi,2013 kurosu et al, 2007. It has recently been found that different FGFs can modulate the intensity of FGFR dimerization to perform corresponding cellular functions in metabolic regulation or cell proliferation (Huang et al, 2017). Thus, it is speculated that other FGFs may exist that have anti-diabetic potential similar to or more pronounced than FGF1.
Fgfr1 is abundantly expressed in white adipose tissue and in the Central Nervous System (CNS) (Fon Tacer et al, 2010). Previous studies have shown that Fgfr1 is a key receptor target in white adipose tissue that mediates the regulation of FGF metabolism (Adams et al, 2012 ye et al, 2016. Fgfr1 in the CNS is also thought to be a receptor target mediating the regulation of FGF19 and FGF21 metabolism (Lan et al, 2017). However, it remains unclear whether Fgfr1 mediates the sustained hypoglycemic effect of FGF1 in the CNS. Glucose Sensing Neurons (GSNs) include glucose-stimulated neurons (GEs) and glucose-inhibited neurons (GIs). GSNs have been found to sense changes in extracellular glucose levels and provide feedback to regulate blood glucose (Anand et al, 1964, marty et al, 2007. Therefore, based on the important roles of Fgfr1 and GSN in metabolic regulation, it is hypothesized that FGF may exert its sustained anti-hyperglycemic effect by acting on GSN and by activating Fgfr1 on the relevant cells.
In this study, several representative members of the FGF paracrine family were screened to test their antihyperglycemic effect in the mouse db/db model of type 2 diabetes, and it was found that central administration of FGF4 sustains relief of hyperglycemia. Notably, FGF4 has significantly better anti-diabetic efficacy than FGF1 in diet-induced obese mice. Mechanistic studies suggest that FGF4 needs to mediate its lowering of glucose levels in T2D rodent models through Fgfr1 and GSN in the hypothalamus. Acute perfusion of FGF4 inhibits GI neurons, while chronic administration of FGF4 re-sensitizes GI and GE neurons and resets the ratio of GI to GE neurons. Notably, non-invasive nasal administration of FGF4 was able to induce sustained diabetes remission. Thus, the data indicate that FGF4 is a new biological agent with high transforming potential and can achieve long-lasting remission of diabetes.
Disclosure of Invention
The present invention relates to Fgfr1 ligands (e.g. FGF 4) for use in the treatment of metabolic disorders, methods of treatment using Fgfr1 ligands, and the use of Fgfr1 ligands in the manufacture of a medicament.
In a first aspect, the invention provides a method for treating a metabolic disease, the method comprising the step of administering to a patient in need thereof a therapeutically effective amount of a ligand of Fgfr1.
In a second aspect, the invention provides a ligand of Fgfr1 for use in the treatment of a metabolic disease.
In a third aspect, the invention provides the use of a ligand of Fgfr1 in the manufacture of a medicament for the treatment of a metabolic disorder.
Preferably, the Fgfr1 ligand is FGF4.
Preferably, the metabolic disorder is selected from the group consisting of obesity, diabetes, insulin resistance, hyperinsulinemia, glucose intolerance and hyperglycemia. In an embodiment, the diabetes is type 1 diabetes or type 2 diabetes, particularly type 2 diabetes.
The Fgfr1 ligand is isolated. The isolated Fgfr1 ligand can be comprised in a pharmaceutical composition. In an embodiment, FGF4 is the only active ingredient in the pharmaceutical composition.
Preferably, the Fgfr1 ligand or the pharmaceutical composition comprising the Fgfr1 ligand is administered intracerebrally or intranasally. Thus, the pharmaceutical composition is a pharmaceutical composition suitable for intracerebral administration or intranasal administration, and the medicament of the third aspect is a medicament suitable for intracerebral administration or intranasal administration.
Preferably, the intracerebral administration is chronic intracerebral administration.
The pharmaceutical composition may further comprise pharmaceutically acceptable formulating agents such as carriers, adjuvants, solubilizers, stabilizers and/or antioxidants. In an embodiment, the pharmaceutical composition further comprises liposomes suitable for intranasal administration.
In embodiments, the frequency of intracerebral administration is less than once every 2 weeks, preferably less than once every 5 weeks, for example once every 6 to 16 weeks.
In another embodiment, the frequency of intranasal administration is less than once every 2 days, preferably less than once every 5 days, for example, once every 6 to 9 days.
Drawings
Figure 1 shows that a single injection of FGF4 into the CNS reduces hyperglycemia in diabetic mice.
(A) Representative FGFs are from each subfamily.
(B-C) changes in blood glucose (B) and body weight (C) following a single icv administration of paracrine FGF (3.0 μ g each) to db/db mice.
(D-E) changes in blood glucose (left) and area under the curve (AUC) (right) in db/db mice subjected to ipGTT 7 days (D) or 50 days (E) after icv FGF1 or FGF4 (3.0 μ g), as indicated.
(F) By intravenous injection 18 Representative micro-PET/CT images (left) and quantitative analysis (right) were performed 30 minutes after F-FDG in db/db mice that received icv FGF1 or FGF4 (3.0. Mu.g) 3 days before.
(G) Expression of mRNA encoding glucose regulatory enzymes in the liver of db/db mice 7 days after icv FGF4 (3.0. Mu.g).
(H) 7 days after icv FGF4 (3.0. Mu.g), glycogen in the liver and skeletal muscle of db/db mice was stained with periodic acid-Schiff (PAS). Scale bar, 100 μm.
(I) Change in blood glucose after a single icv injection of FGF4 (3.0. Mu.g) in DIO mice.
(J) Blood glucose (left) and AUC (right) changes in DIO mice subjected to ipGTT 7 days post icv FGF4 (3.0 μ g).
(K) Change in body weight after a single dose of icv FGF4 (3.0. Mu.g) in DIO mice.
(L-M) changes in physical Activity (L) and thermogenesis (M) in DIO mice 7 days after icv FGF4 (3.0. Mu.g).
Data are shown as mean ± s.e.m,. P ≦ 0.05 for the corresponding control group; * P is less than or equal to 0.01; * P is less than or equal to 0.001; compared with FGF1 group, # p is less than or equal to 0.05; # p is not more than 0.001,ns, not significant.
Figure 2 shows that FGF4 requires Fgfr1 in hypothalamic neurons to exert anti-hyperglycemic effects.
(A-C) Fgfr1 is expressed by the hypothalamus in which mRNA hybridizes in situ. Scale bar, 1mm (A) and 500 μm (B, C).
(D) For injection of AAV8-CMV-creGFP into Fgfr1 flox/flox A hypothalamic Fgfr1 knockout protocol was generated in mice.
(E) Hypothalamic expression of AAV 8-CMV-creGFP. Scale bar, 200 μm.
(F)Fgfr1 CMV-Cre The blood glucose of the mice was significantly increased. Virus was injected at t = 0.
(G) In Fgfr1 CMV-Cre Blocking the antihyperglycemic effect of FGF4 in mice.
(H) Designed by injecting AAV8-hSyn-Cre into Fgfr1 flox/flox Fgfr1 was knocked out in hypothalamic neurons in mice.
(I-J) Fgfr1 under different dietary conditions (Normal food or HFD) hSyn-Cre Changes in blood glucose (I) and body weight (J) in mice and GFP control.
(K-N)Fgfr1 hSyn-Cre Changes in blood glucose (K, L) and body weight (M, N) after icv FGF4 (3.0 μ g) in mice (K, M) and GFP control (L, N) mice.
(O-P) changes in blood glucose (O) and body weight (P) after FGF4 (1.5. Mu.g) was injected into VMH.
(Q) change in blood glucose (left) and AUC (right) in DIO mice subjected to ipGTT 24 hours after FGF4 (1.5 μ g) was injected into VMH.
VMH, ventral hypothalamus; DMH, dorsal hypothalamus; LH, lateral hypothalamus; ARC, hypothalamic arcuate nucleus; 3V, third ventricle. Data are shown as mean ± s.e.m,. P ≦ 0.05 for the corresponding control group; * P is less than or equal to 0.01; * P.ltoreq.0.001,ns, not significant.
Figure 3 shows that FGF4 restores the function of VMH GSN in diabetic mice.
(A-B) representative firing rates associated with VMH GE neurons (A) and GI neurons (B) from wt, db/db and db + FGF4 mice respond to changes in low glucose (0.1 mM). db + FGF4 means db/db mice injected with FGF4 (3.0. Mu.g, 7 to 10 days post-icv).
(C) Relative percentages of GSN cell types in wt, db/db and db + FGF4 mice. INS, glucose insensitive.
(D-F) GE neurons respond to mean firing rate change (D), peak firing rate change (E) and peak time (F) of 0.1mM glucose.
(G-I) mean firing rate change (G), peak firing rate change (H), and peak time (I) of GI neurons in response to 0.1mM glucose.
(J) The experiment was designed to test the acute effect of FGF4 on neurons in db/db mice.
(K) Representative traces show inhibition of GI neurons by FGF4 perfusion.
(L) mean firing rate changes associated with GI neurons and their percentage of perfusion by FGF4.
(M) the assay was designed to test the effect of blocking Fgfr1 by PD 166866.
(N) representative trace shows that D166866 inhibits FGF 4-inhibited GI neurons.
(O) mean firing rate change and its percentage in GI neurons treated with FGF4 plus PD 166866.
(P) Central administration of GI inhibitors to lower blood glucose in db/db mice.
(Q) the experiment was designed to test the acute effect of FGF4 on neurons in DIO mice.
(R) representative traces show that FGF4 perfusion inhibits GI neurons.
(S) mean firing rate changes of GI neurons and their percentage perfused by FGF4.
(T) the experiment was designed to test the effect of PD166866 on GI neurons stimulated or inhibited by FGF4.
(U) the Fgfr1 inhibitor partially abrogates the effect of FGF4 on GI neurons.
(V) mean firing rate change and percentage thereof in FGF 4-inhibited GI neurons treated with PD 166866.
Data are shown as mean ± s.e.m,. P ≦ 0.05 for the corresponding group; * P is less than or equal to 0.01; * P is less than or equal to 0.001.
Fig. 4 shows that nasal delivery of FGF4 to the brain relieves hyperglycemia.
(A) Representative pictures of liposome nanoparticles loaded with gelatin solution (control) and FGF 4-gelatin solution (FGF 4). Scale bar, 100nm.
(B) Distribution of liposome nanoparticle diameters.
(C-D) changes in blood glucose (C) and body weight (D) of db/db mice following intranasal delivery of FGF4 (30 μ g).
(E-F) changes in blood glucose (left) and AUC (right) in db/db mice subjected to ipGTT (E) or ipITT (F) 24 hours after intranasal FGF4 delivery.
(G) Blood glucose in db/db mice after repeated intranasal delivery of FGF4.
Data are shown as mean ± s.e.m,. P.ltoreq.0.05 compared to the corresponding control group; * P is less than or equal to 0.01; * P is less than or equal to 0.001.
FIG. S1 shows the phenotypes in wt and db/db mice after FGF injection, which are related to FIG. 1.
(A) There was no significant change in blood glucose following a single icv administration of endocrine FGF21 (3.0 μ g) in db/db mice.
(B-C) blood glucose (B) and body weight (C) in response to increasing FGF4 dose.
(D) icv FGF4 (3.0 μ g) indicates the change in blood glucose (left) and AUC (right) in db/db mice subjected to ipGTT 7 days after dose.
(E) db/db mice had no decrease in blood glucose after ip injection of FGF4 (3.0. Mu.g).
(F) Transient decrease in blood glucose after ip injection of FGF4 (30. Mu.g).
(G) Change in blood glucose after icv FGF4 (3.0 μ g) in wt mice.
(H) Blood glucose (left) and area under the curve (AUC) (right) changes in wt mice subjected to ipGTT 7 days post icv FGF4 (3.0 μ g).
(I) Change in body weight after a single icv administration of FGF4 (3.0. Mu.g) to wt mice.
(J-O) changes in plasma hormone (J), food intake (K), body fat content (L), physical activity level (M), respiratory exchange ratio (RER, N) and thermogenesis (O) in db/db mice on the indicated days following injection of icv FGF4 (3.0 μ g). The 12-light-dim light cycle is indicated.
(P) blood glucose (left) and AUC (right) changes in db/db mice subjected to ipITT 10 days after icv FGF4 (3.0 μ g).
(Q) representative micro-PET/CT images and intravenous injection 18 Quantitative analysis 15 min (left) and 60 min (right) after F-FDG was performed in db/db mice that had received icv FGF1 or FGF4 (3.0. Mu.g) 3 days ago.
Data are shown as mean ± s.e.m,. P.ltoreq.0.05 compared to the corresponding control group; * P is less than or equal to 0.01; * P is less than or equal to 0.001; compared with FGF4 (0.3. Mu.g) group, # # # p ≦ 0.001,ns, not significant.
Figure S2 shows the phenotype in DIO mice after FGF injection, which correlates with figure 1.
(A-B) changes in blood glucose (A) and body weight (B) following a single injection of FGF1 (3.0. Mu.g) in DIO mice.
(C) Blood glucose (left) and AUC (right) changes were made 7 days after icv FGF4 (3.0 μ g) in ipott DIO mice.
(D-F) Change in body fat content (D), food intake (E) and RER (F) after a single icv FGF4 (3.0 μ g) in DIO mice. Days post injection and 12 hours light-dim light cycle are indicated.
(G) Transient reduction of blood glucose after ip injection of 30 μ g FGF4 in DIO mice.
Data are shown as mean ± s.e.m,. P ≦ 0.05 for the corresponding control group; * P is less than or equal to 0.001.
Figure S3 shows that hypothalamic Fgfr1 is required for FGF 4-mediated glucose lowering, in relation to figure 2.
(A) The hypothalamus expresses Fgfr1. Scale bar, 1mm (left); 100 μm (right).
(B)Fgfr1 + The proportion of cells, expressed as the number of Fgfr1+ cells divided by all cells from wt mice (Dapi +).
(C) Relative expression of Fgfr1 in different hypothalamic regions was normalized to VMH (n =8 slides from 5 mice).
(D) For injection of AAV8-CMV-CreGFP into Fgfr1 flox/flox Protocol for generating hypothalamic Fgfr1 knockdown in mice.
(E) AAV8-CMV-creGFP is expressed in the hypothalamus of different brain regions, brain region B.
(F)GFP + Cells and all cells from the indicated hypothalamic nuclei (Dapi) + ) The ratio of (a) to (b).
(G) Reduction of Fgfr1 mRNA after CMV-Cre expression.
(H) Weight change in CMV-GFP and CMV-Cre mice.
(I) Blood glucose (left) and AUC (right) of CMV-Cre mice subjected to ipGTT 6 weeks after virus injection.
(J) Change in body weight of CMV-cre mice after icv FGF4 (3.0. Mu.g).
(K-L) change in blood glucose (K) and body weight (L) in CMV-GFP mice after icv FGF4 (3.0. Mu.g).
Data are shown as mean ± s.e.m,. P ≦ 0.05 for the corresponding control group; * P is less than or equal to 0.01; * P is less than or equal to 0.001.
FIG. S4 shows a graph at Lepr + And lack of Fgfr1 in astrocytes but none block the hypoglycemic effect of FGF4.
(A-B) Lepr changes in body weight (A) and blood glucose (B) in CKO mice.
(C-D) Lepr that experienced either ipGTT (C) or ipITT (D) changes in blood glucose (left) and AUC (right) in CKO mice (14 weeks of age).
(E) Lepr weight change in CKO mice under HFD.
(F) Lepr CKO mice blood glucose at HFD at week 12.
(G-H) Lepr changes in blood glucose (G) and body weight (H) in CKO mice after icv FGF4 (3.0. Mu.g).
(I-J) Gfap: weight change in CKO mice under diet food (I) or HFD (J).
(K) Gfap CKO mice blood glucose at HFD at week 12.
(L-M) Gfap changes in blood glucose (L) and body weight (M) of CKO mice after icv FGF4 (3.0. Mu.g).
Data are shown as mean ± s.e.m,. P.ltoreq.0.05 compared to the corresponding control group; * P is less than or equal to 0.01; * P is less than or equal to 0.001; ns, not significant.
Figure S5 shows the phenotype associated with figure 2 following Fgfr1 delay in hypothalamic neurons.
(A) By injecting AAV8-hSyn-CreGFP into Fgfr1 flox/flox Fgfr1 knock-outs were designed in mice in hypothalamic neurons.
(B-C) changes in body weight (B) and blood glucose (C) of syn-GFP and syn-cre mice.
(D) AAV8-hSyn-CreGFP is expressed in the hypothalamus of different brain regions, brain region B.
(E) Expression of Fgfr1 in Syn-GFP and Syn-Cre mice.
(F) Reduction of Fgfr1 expression levels following Syn-Cre expression.
Data are shown as mean ± s.e.m,. P ≦ 0.01 for the corresponding control group; * P is less than or equal to 0.001.
FIG. S6 shows the effect of FGF4 on VMH GE neurons from db/db mice, in relation to FIG. 3.
(A) The experiments were designed to test the acute effect of FGF4 on neurons in db/db mice.
(B-C) shows representative traces of either excitation (B) or inhibition (C) of GE neurons perfused by FGF4.
(D) The mean firing rate change associated with VMH GE neurons and their percentage of perfusion by FGF4.
(E) Schematic diagram showing the experimental design to test the effect of the Fgfr1 inhibitor PD 166866.
(F-G) mean firing rate changes in FGF 4-stimulated (F) or FGF 4-inhibited (G) GE neurons treated with PD 166866.
ACSF, artificial cerebrospinal fluid. Data are shown as mean ± s.e.m.
Figure S7 shows the effect of FGF4 on VMH GE neurons from DIO mice, in relation to figure 3.
(A) The experiment was designed to test the response of GSN to high (5.0 mM) glucose and low (0.1 mM) glucose.
(B-D) mean firing rate changes associated with GE (B) neurons, GI (C) neurons, and INS (D) neurons by treatment with 5.0mM glucose and 0.1mM glucose.
(E) The experiment was designed to test the acute effect of FGF4 on brain sections.
(F) Representative traces show the firing, inhibition, and insensitivity of GE neurons perfused by FGF4.
(G) Mean firing rate changes associated with GE neurons and their percentage of perfusion by FGF4.
(H) The experiment was designed to test the effect of the Fgfr1 inhibitor PD166866 on GE neurons.
(I) PD166866 partially blocks the FGF 4-stimulated and inhibited GE neurons.
(J) Change in mean firing rate and percentage of FGF 4-stimulated GE neurons treated with PD 166866.
Data are shown as mean ± s.e.m.
Detailed Description
As used herein, the terms "a" or "an" mean one or more, unless expressly specified otherwise.
The term "isolated" molecule refers to a molecule that is substantially free of any other contaminating molecules or other contaminants that are found in its natural environment and which would interfere with its use in production or its therapeutic, diagnostic, prophylactic or research use. Thus, typically an isolated polypeptide of the invention is a purified polypeptide and free of pathogens and pyrogens, having a purity of at least 80%, preferably at least 90%, more preferably at least 95%, in particular having a pharmaceutical purity, i.e. at least 98%. Preferably, the isolated polypeptide of the invention may be substantially free of other polypeptides, in particular polypeptides from animals.
The term "FGF4", an abbreviation for fibroblast growth factor 4, means full length FGF4 or an active fragment thereof, e.g. a fragment from Ala67 to Leu 206. Preferably, FGF4 is human FGF4. Human FGF4 is a 176 AA long protein derived by cleavage of the N-terminal 30 AA of the precursor protein. FGF-4 contains a single N-linked glycosylation site. The unglycosylated FGF-4 is a polypeptide truncated by two NH 2-terminal residues with better heparin affinity and biological activity than the wild-type protein.
Recombinant FGF4 protein is commercially available (e.g., from Sigma Aldrich, where recombinant FGF4 protein is produced in baculovirus; or from Invitrogen, where recombinant FGF4 protein is produced in E.coli).
The term "pharmaceutically acceptable carrier" as used herein refers to one or more formulation materials suitable to effect or enhance FGF4 delivery.
The term "therapeutically effective amount" refers to an amount of FGF4 to support observable levels of one or more biological activities, such as the ability to lower blood glucose, insulin, triglyceride, or cholesterol levels; reducing body weight; or improve glucose tolerance, energy expenditure or insulin sensitivity. In the present invention, the "patient" is preferably a human, but may also be an animal, preferably a mammal such as a dog, pig, cow, horse, rat, mouse, guinea pig, and the like.
For human subjects, "obesity" can be defined as a body weight that is more than 20% higher than the ideal body weight for a given population (r.h. williams. Endocrinology textbook 1974, pages 904-906).
"diabetes" is characterized by excessive urination and persistent thirst and includes both types. Type II diabetes is characterized by excessive glucose production despite the availability of insulin, and the body's glucose levels remain too high due to insufficient glucose clearance. In this embodiment, the diabetes may be type 1 diabetes, and the effect of FGF4 on the treatment of type 2 diabetes is believed to be much stronger than the effect of type 1 diabetes. The present inventors have found that FGF4 is more beneficial for the treatment of type 2 diabetes.
The term "insulin resistance" refers to a state where a normal amount of insulin produces a subnormal biological response.
The term "hyperinsulinemia" is defined as higher than normal levels of insulin in the blood.
The term "glucose intolerance" as used herein is defined as an abnormal sensitivity to glucose.
The term "hyperglycemia" is defined as excess sugar (glucose) in the blood.
The pharmaceutical composition may comprise one or more pharmaceutically acceptable formulations for modifying, maintaining or preserving, for example, pH, permeability, viscosity, clarity, color, isotope, odor, sterility, stability, dissolution or release rate, adsorption or permeation of the composition. Suitable agents include, but are not limited to, amino acids (such as glycine, glutamine, aspartic acid, arginine, or lysine), antimicrobials, antioxidants (such as ascorbic acid, sodium sulfite, or sodium bisulfite), buffers (such as borates, bicarbonates, tris-HCl, citric acid, phosphates, or other organic acids), co-raw materials (such as mannitol or glycine), chelating agents (such as ethylenediaminetetraacetic acid (EDTA)), complexing agents (such as caffeine, polyvinylpyrrolidone, beta-cyclodextrin, or hydroxypropyl beta-cyclodextrin), fillers, monosaccharides, disaccharides, and other carbohydrates (such as glucose, mannose, or dextrin), proteins (such as serum albumin, gelatin, or immunoglobulin), colorants, flavors and diluents, emulsifiers, hydrophilic polymers (such as polyvinylpyrrolidone), low molecular weight polypeptides, salt-forming counter-ions (such as sodium), preservatives (such as phenylmethanoxy chloride, benzoic acid, salicylic acid, thimerosal, phenylethyl alcohol, methyl terephthalic acid, propyl terephthalic acid, chlorhexidine, sorbic acid, or hydrogen peroxide), solvents (such as glycerol, propylene glycol, or polyethylene glycol), sugar alcohol (such as mannitol or sorbitol), suspending agents (such as suspending agents, suspending agents or suspending agents; PEG, sorbitan esters, polysorbates such as polysorbate 20 or polysorbate 80, triton, trimethylamine, lecithin, cholesterol or tyloxapol), stability sensitizers (such as sucrose or sorbitol), sensitizers (such as alkali metal halides-preferably sodium or potassium chloride or mannitol sorbitol), delivery vehicles, diluents, adjuvants and/or pharmaceutical adjuvants (pharmaceutical science of remgton, a.r. gennaro, editors, mikawa).
The primary vehicle or carrier in the pharmaceutical composition may be aqueous or non-aqueous. For example, a suitable vehicle or carrier for injection may be water or a physiological saline solution.
The pharmaceutical compositions of the present invention may be administered by any route known to those skilled in the art, such as oral administration, rectal administration, sublingual administration, intrapulmonary administration, intraperitoneal administration, transdermal administration, iontophoretic administration, vaginal administration, intracerebral administration, and intranasal administration. However, intraperitoneal administration of large doses of FGF4 only resulted in a temporary reduction of blood glucose levels in db/db mice. Preferably, the pharmaceutical composition of the invention is administered by intracerebral administration or intranasally, according to the mechanism discovered by the inventors.
The pharmaceutical compositions for intranasal administration of the present invention may be in the form of liposomes. In embodiments, FGF4 is loaded into a 100nm liposome.
A single intracerebral or intranasal administration of FGF4 may result in a sustained chronic hypoglycemic effect. The present inventors found that a single intracerebral administration of FGF4 might even produce a hypoglycemic effect lasting more than 4 months. At least the frequency of intracerebral administration may be less than once every 2 weeks, preferably less than once every 5 weeks, for example, once every 6 to 16 weeks. In addition, the frequency of intranasal administration may be less than once every 2 days, preferably less than once every 5 days, for example, once every 6 to 9 days.
The dosage administered will vary depending on the formulation, the desired time course, and the patient to be treated, and the physician can determine the feasible dosage in treatment based on the actual circumstances (e.g., the condition, weight, etc. of the patient). For an average adult, the dosage of the pharmaceutical composition of the present invention may be 1ng to 10mg of FGF4 or an variant thereof per kg of body weight of the adult. For the injection route, the dose is preferably 10ng to 1mg per kg body weight, more preferably 100ng to 100ug per kg body weight, still more preferably 1ug to 10ug such as 2ug, 5ug, 7ug or 8ug per kg body weight.
Publications cited in this application are provided herein for purposes of illustration and are incorporated herein by reference as if fully set forth herein.
For a better understanding of the present invention, it will now be described in more detail with reference to specific examples. It should be noted that these examples are merely illustrative of the present invention and should not be construed as limiting the scope of the present invention. Various modifications and alterations of this invention will become apparent to those skilled in the art from this description.
Examples of the invention
The following examples are illustrative of embodiments of the present invention and various uses thereof. The following examples are for illustrative purposes only and should not be construed as limiting the scope of the invention in any way. The materials used in the examples are commercially available.
EXAMPLE 1 materials and methods
Experimental model and subject details
Mouse model
Adult Male db/db (B6. BKS (D) -Lepr) db /J,8-12 weeks old) was purchased from seikagaku limited company (nanjing, china). C57BL/6J (wt) mice were from Chighui laboratory animal feeding Co., ltd. Fgfr1 flox/flox Mice (accession number 007671) were obtained from Jackson laboratories. By Fgfr1 flox/flox Mice and Lepr Cre/+ Mice (# 008320, jackson laboratory) and Gfap Cre/+ Mice (gifts from J.W.Zhou) were crossed to generate Fgfr1 conditional knockout mice (Lepr: CKO, lepr stands for leptin receptor; GFAP: CKO, GFAP stands for glial fibrillary acidic protein). Mice were housed under constant temperature and humidity in a 12 hour light/12 hour dark cycle (light, 8PM-8 AM). Mice were randomly assigned to experimental groups and water and either standard laboratory food or a high fat diet (HFD; D12492, high fat diet, usa) were ad libitum (ad libitum) obtained unless otherwise stated. All animal procedures were performed strictly following animal studies: in vivo experimental guidelines are reported. Care and use of animals is provided by Shanghai university of science and technology, shanghai model animals Co., ltd, zhang JiangLaboratory national protein science facilities animal facilities and government regulations review and approval.
Details of the method
Expression constructs and recombinant protein production
Human FGF1 (Met 1-Asp 155), FGF4 (Ala 67-Leu 206), FGF7 (Cys 32-Thr 194), FGF8a (Gln 23-Arg 204), FGF20 (Met 1-Thr 211) and FGF21 (His 29-Ser 209) were expressed and purified according to published protocols (benken et al, 2012, bellosta et al, 2001. Briefly, a cDNA fragment encoding FGF4 (Ala 67-Leu 206) was PCR amplified and subcloned into pET-15b at the NcoI site and the Xhol site, respectively. Coli BL-21 (DE 3) cells transformed with the FGF4 construct were cultured in LB medium (2% glucose and 30kg/mL kanamycin) at 37 ℃ and 200rpm in an incubation shake flask. Cells were induced at an optical density of 0.8-1.0 (measured as 600) with 1.0mM isopropyl-L-thio-D-galactopyranoside (IPTG) for 4 hours at 37 ℃. Then, the cells were collected and lysed in 25mM Na/K phosphate buffer (pH 7.5) containing 300mM NaCl using an Emulsiflex-C3 (Avena protein, ontario, canada) high volume homogenizer. The lysates were clarified by centrifugation at 20,000rpm for 30 min at 4 ℃ and the soluble recombinant proteins were purified by a heparin affinity column (Hitrap heparin HP, GE Healthcare, piscataway, NJ, column volume 5 ml) eluting with a linear gradient of NaCl (0M-2.0M) in HEPES (25mm, ph 7.5). Purity of the protein of interest will be determined by 12% SDS-PAGE analysis; further, the fractions were combined, concentrated and further purified using gel molecular sieves (Superdex TM-75GE healthcare, piscataway, NJ) in a mobile phase buffer (1.0M NaCl,25mM Tris-HCl, pH 8.0). The purity of the recombinant protein was estimated to be > 98%. Other FGFs were prepared in a similar manner.
Brain stereotaxic injection
FGF (. About.3.0. Mu.g, unless otherwise specified) or AAV virus (. About.200 nl) was delivered by a drawn glass pipette and pen-type oil pressure syringe with custom designed robot (MO-10, tritech research, USA). Injection coordinates were calculated from Paxinos & Franklin mouse brain coordinates (3 rd edition) by injection under isoflurane general anesthesia using a small animal stereotaxic instrument (David Kopf instrument, # PF-3983 rwd life sciences, #68030; qiano family nanjing biotechnology limited, # SH 01A). The feedback heater is used to keep the mouse warm during surgery. Mice were recovered in warm racks before being transferred to containment cages. The coordinates are as follows: 1) For intraventricular (i.c.v.) administration, lateral Ventricle (LV): bregma is-0.40 mm behind; 1.10mm on the side, 2.50mm below the skull surface; 2) For hypothalamic delivery, ventral hypothalamic nuclei (VMH): bregma is-1.65 mm; 0.40mm lateral, 5.70mm below the skull surface, anatomy is shown directly on the relevant figures. For pharmacological studies, cannulas of mice (RWD life sciences) were placed in the LV.
Physiological measurement
Glucose levels were determined by a hand-held glucometer (agco-dukele golgi glucometer, roche, switzerland). Unless otherwise stated, blood samples were collected from the tail vein of mice fed ad libitum. In the food intake assay, mice were implanted with cannulae in the lateral ventricle (RWD life sciences, 62002, 62102) and the cannulae were fixed to the skull with dental cement (C & B metabolic rapid adhesive cement system). All intubated mice were individually housed for 7-10 days post-surgery and recovered with food ad libitum and water. FGF4 (3.0 μ g) or saline was delivered to the mice via cannula and the food was weighed daily. Body composition was measured using quantitative magnetic resonance spectroscopy (minisec body composition analyzer LF50, bruker, karlsruhe, german). Heat production, physical activity and RER (respiratory exchange rate) were monitored with a comprehensive laboratory animal monitoring system (CLAMS, columbus instruments). For plasma analysis, samples were collected from mice fed ad libitum. Plasma insulin, corticosterone, ACTH, epinephrine, and non-esterified free fatty acid (NEFA) concentrations were determined by the Elisa kit according to the manufacturer's instructions (# E0448m, # E0540Ge, # E0836m, # E0858Ge from EIAab, wuhan, china, # a042-2-1 from the institute of bioengineering, tokyo, china).
Intraperitoneal glucose tolerance test (ipGTT) and intraperitoneal insulin tolerance test (ipITT)
ipGTTs were performed in mice overnight after intraperitoneal injection of 1.0g/kg body weight of glucose (20% glucose) followed by fasting for 16 hours. ipITT was performed in fasted 6-hour mice by injection of 0.75 units/kg body weight (for wt mice) or 1.0 units/kg (for db/db and HFD mice) insulin (# 40112es25, yeasenl2016, china). Blood samples were collected from the tail vein at the indicated times.
Microelectronic emission computed tomography (micro PET-CT) imaging and analysis
MicroPET-CT analysis was performed in db/db mice 3 days after icv injection of FGF (. About.3.0. Mu.g). Receiving 18 Overnight fasted mice of F-FDG (259.1 + -17.76 μ Ci, i.v.) were transiently anesthetized with 1.5% isoflurane in oxygen at 2L/min and scanned in Inveon preclinical multimodal PET-CT (Siemens, munich, germany) for 1 hour. The image is reconstructed using a three-dimensional ordered subset expectation maximization (3 DOSEM) algorithm and then using a Maximum A Posteriori (MAP). Under the guidance of CT images, a 3D region of interest (ROI) is rendered on the brain, liver and muscles. The radioactivity of the corresponding tissues was measured using the Inveon research workplace (Siemens, munich, germany).
AAV vectors
AAV vectors carrying specific genes (titre > 10) 12 ) Purchased from Taitol Bioscience (Shanghai, china), including AAV2/8-CMV _ bGI-Cre-EGFP-pA, AAV2/8-CMV _ bGI-EGFP-WPRE-pA, AAV2/8-hSyn-Cre-EGFP-WPRE-pA, AAV2/8-hSyn-EGFP-WPRE-pA.
Reverse transcription and quantitative real-time PCR
Total RNA was extracted from hypothalamus or liver using Trizol reagent (Invitrogen, usa). Reverse transcription PCR was performed using PrimeScript RT kit (RR 047A, takara, japan), and ViiA was performed using SYBR PCR kit (RR 820A, takara, japan) TM 7 real-time PCR System (applied biosystems, USA), real-time PCR was determined according to the manufacturer's instructions. Actin was used as an internal control for real-time PCR amplification.
Immunohistochemistry and RNA in situ hybridization
Mice were anesthetized and perfused with PBS followed by 10% formalin. Brains were excised, fixed overnight and cut into-50 μm sections. After collection and three washes with PBST (0.3% Triton X-100 in PBS), cryostat sections were incubated withBlocking solution (PBS containing 2.5% goat serum and 0.3% Triton X-100) was incubated together, then treated with primary antibody (chicken anti-GFP, ab13970, abcam,1 1000) overnight at 4 ℃, and then the samples were incubated with secondary antibody (goat anti-rabbit Alexa 488 secondary antibody, invitrogen, a 11039) for 2 hours at room temperature. Images were captured on a Zeiss LSM800 confocal microscope or Olympus VS120 virtual microscope slide scanning system. According to the manufacturer's instructions, use2.5HD kit (# 322350, advanced cell diagnostics) for Fgfr1 RNA in situ detection. The Fgfr1 probe was designed by advanced cytodiagnostics and the DapB probe was used as a negative control.
Electrophysiological recording slice
Briefly, hypothalamic sections containing VMH regions were prepared from adult males wt, db/db and db/db with icv FGF4. Oxygen content in ice-cold condition (95% 2 And 5% of CO 2 ) Before cardiac perfusion, mice were anesthetized with sodium pentobarbital (100 mg/kg i.p.), a high sucrose solution consisting of (in mM): 213 sucrose, 26NaHCO 3 、1.25NaH 2 PO 4 、2.0Na 2 HPO 4 2.5KCl and 2MgSO 4 (osmolality adjusted to 300mOsm/kg, pH 7.4). Brains were removed quickly and placed in ice-cold oxygenated high sucrose solution. The coronal hypothalamic sections (250 μm) were cut with a vibrating microtome (VT 1200S, leica microsystems, germany). Sections were incubated at 31 ℃ in oxygenated artificial cerebrospinal fl fluid (ACSF in mM:124NaCl,5KCl, 1.24KH) prior to transfer to the recording chamber 2 PO 4 ,1.3MgSO 4 ,26NaHCO 3 ,2.4CaCl 2 and 2.5glucose, pH 7.2-7.4,300-310 mOsm/kg)>For 1 hour. ACSF was perfused at 1.2 ml/min. The hypothalamic sections were observed by infrared differential interference contrast (ID-DIC) video microscopy using an upright microscope (AxioDetector, zeiss, germany) and an infrared camera (IR-1000, DAGE-MTI, USA).
The recording pipette was pulled from the borosilicate glass capillary tube using a P-97 pipette puller (Sutter instruments, usa). To use electricitySpontaneous discharges were recorded extracellularly in sections by flow-clamping, using solutions (in mM: 130 potassium gluconate, 1 CaCl) 2 ,10EGTA,10HEPES,2.0Mg-ATP,2.0Na 3 -GTP) (pH 7.2-7.4,295 mosm/kg) fill pipettes. The voltage signals were recorded using EPC-10 amplifier and PatchMaster10 acquisition software (HEKA, germany). These signals were digitized at 10kHz and filtered with a 2kHz low pass filter. To determine the glucose sensitivity of each recorded VMH neuron, 2.5mM glucose was replaced with 0.1mM glucose in ACSF, as shown in the figure. The spontaneous firing activity of the neurons was continuously recorded and no holding current was applied to the neurons during the extracellular glucose change. After identification, neurons were recorded for 20 min during perfusion with ACSF containing 2.5mM glucose. Control ACSF was then replaced by experimental ACSF with or without 10 μ M FGFR1 selective inhibitor (PD 166866) containing 9 μ g/ml FGF4 for about 5 minutes according to the experiment. The experimental ACSF was then replaced with the control ACSF for 20 minutes to wash the drug away.
For GE/GI neurons, the peak in firing rate change was defined as the minimum/maximum firing rate minus 100% (baseline, t) in 0.1mM glucose perfusion 0 ). The response curves of the GE/GI neurons to 0.1mM glucose were fitted by 2nd order smoothing. The time to peak of GE/GI neurons is defined as t 0 (0.1 mM glucose perfusion) to the time duration that the FR reaches the minimum/maximum. The criteria for excitation or inhibition were set to give a rate change of-30% (Toda et al, 2016).
Preparation, characterization and treatment of FGF 4-loaded liposomes
FGF4 was dissolved in a 20% poloxamer 188 covalently bound heparin copolymer solution and further diluted with a 2.0% gelatin solution to a final concentration of 1.68mg/ml. The mixture was then sonicated (110w, 15 ℃,15 s) and lyophilized to obtain a powder containing FGF4. Thereafter, the powder was dispersed in a tert-butanol solution containing Hydrogenated Soybean Phospholipids (HSPC) and cholesterol. The mixed solution was sonicated (90w, 25 ℃,20 s) and lyophilized again to obtain a powder containing FGF 4-liposomes. Finally, the FGF 4-liposomes were reconstituted in double distilled water to form a liposome suspension. In these procedures, a gelatin solution without FGF4 was used as a blank.
The morphology of the blank and FGF4 loaded liposomes was observed by Transmission Electron Microscopy (TEM) (1230, jeol Jem, tokyo, japan) using negative staining with 1% phosphotungstic acid. Zeta potential of blanks and FGF 4-liposomes Zeta potential by dynamic light Scattering Using zeta potential/particle Sizer Nicomp as described previously TM 380 Nils (pss. Nicomp, san babara, ca, usa) (Zhao et al, 2016).
Then, db/db mice were intranasally administered equal volumes of blank or FGF 4-liposomes (30 μ g FGF 4) to achieve nasal to brain delivery, respectively. Blood glucose was measured at the indicated times and ipGTTs/ipITT experiments were performed as described above.
References to methods only
Benken, a., eliseenkova, a.v., ibrahimi, o.a., olsen, s.k., and Mohammadi, m. (2012). The plasticity of the N-terminus of fibroblast growth factor 1 (FGF 1) interacting with FGF receptors forms the basis of FGF1 versatility. J Biol Chem 287,3067-3078.
Bellosta, p., iwahori, a., plotnikov, a.n., eliseenkova, a.v., basilico, c., and Mohammadi, m. (2001) identification of heparin binding sites in receptors and fibroblast growth factor 4 based on structural mutation. Mol Cell Biol 21,5946-5957.
Kaliina, j., byron, s.a., makarenkova, h.p., olsen, s.k., eliseenkova, a.v., larochelle, w.j., dhanabal, m., blais, s.ornitz, d.m., day, l.a., et al (2009). Homodimerization produces receptor binding of the fibroblast growth factor 9 subfamily and heparan sulfate-dependent diffusion in the extracellular matrix. Mol Cell biol29,4663-4678.
Kharitonnkov, a., shiyanova, t.l., koester, a., ford, a.m., micarovic, r., galbreth, e.j., sandusky, g.e., hammond, l.j., moyers, j.s., owens, r.a., et al. (2005). A novel metabolic regulator FGF-21.J Clin Invest 115,1627-1635.
Olsen, s.k., li, j.y., bromleigh, c., eliseenkova, a.v., ibreahimi, o.a., lao, z., zhang, f., lindardt, r.j., joyner, a.l., and Mohammadi, m. (2006). Alternative splicing modulates the structural basis of FGF8 tissue activity in the brain. Genes Dev 20,185-198.
Structure-based mutational analysis in Sher, i., yeh, b.k., mohammadi, m., adir, n., and Ron, d. (2003) FGF7 identified new residues associated with FGFR2IIb specific interactions. FEBS Lett 552,150-154.
Toda, c., kim, j.d., improvizzeri, d., cuzzocura, s., liu, z.w., and Diano, s. (2016.) regulation of UCP2 mitochondrial fission and glucose-responsive control of the ventral medial nucleus. Cell 164,872-883.
Zhao, y.z., zhang, m., tian, x.q., zheng, l., and Lu, c.t. (2016) early intervention in diabetic cardiomyopathy was performed using basic fibroblast growth factor nanoliposomes in combination with ultrasound technology. Int J Nanomedicine 11,675-686.
Quantitative and statistical analysis
Statistical information
Mice were randomly assigned to each group, and the age and body weight of the mice were matched. Animals were not excluded from the study unless otherwise indicated. Two sets of comparisons were made using unpaired Student's t-test. RM one-way ANOVA was used for multiple group comparisons, two-way ANOVA followed by Sidak's or Tukey's multiple comparisons for groups mixed by time factor design. Data were analyzed using Prism 8.0 and expressed as mean ± s.e.m.
Example 2 results
Single injection of FGF4 into CNS for reducing hyperglycemia
First, four human recombinant FGFs (rFGF 4, rFGF7, rFGF8a, and rFGF 20) were purified as representative members of their paracrine subfamilies (fig. 1A). These rFGF's were then administered into the CNS of db/db mice and tested for their anti-hyperglycemic activity. Human recombinant FGF1 and FGF21 (rFGF 1 and rFGF 21) were used as controls. Notably, single dose administration of rFGF4 (FGF 4,3.0 μ g) into the lateral ventricles of db/db mice significantly reduced blood glucose for > 7 weeks without affecting body weight, similar to the effect of FGF1 (fig. 1B and fig. 1C). In contrast, rFGF7, rFGF8a, rFGF20 and rFGF21 lack this antihyperglycemic activity (fig. 1B, fig. 1C and fig. S1A). Interestingly, intracerebroventricular (icv) administration of FGF4 (hereinafter icv FGF 4) had a more pronounced effect on glucose tolerance than FGF1 (measured using the intraperitoneal glucose tolerance test, ipGTT) 7 days after injection (fig. 1D). This difference was no longer evident 50 days after injection (fig. 1E). As expected, higher doses of FGF4 were more effective in lowering blood glucose and improving glucose tolerance, as well as not affecting body weight (fig. S1B-S1D). As little as 0.3 μ g of FGF4 was sufficient to lower blood glucose for 4 days (fig. S1B). Like FGF1 (Scarlett et al, 2016), intraperitoneal (ip) administration of FGF4 (3.0 μ g) did not affect blood glucose in db/db mice (FIG. S1E), while the 30 μ g dose induced a transient decrease (FIG. S1F). Notably, central administration of FGF1 or FGF4 to wild-type (wt) mice did not affect blood glucose, body weight, or glucose tolerance (fig. S1G through S1L and (Scarlett et al, 2016)), suggesting that the risk of hypoglycemia induced by these two FGFs is negligible.
Next, the peripheral response after central FGF administration was recorded. Plasma hormones from db/db mice were measured 7 days after icv FGF4 and no significant change in insulin or corticosterone concentration was found, while adrenocorticotropic hormone (ACTH) levels were significantly increased (fig. S1J). These results indicate that the glucose lowering effect of FGF4 is unlikely due to an increase in insulin or a decrease in stress hormones associated with the hypothalamic-pituitary-adrenal (HPA) axis. Furthermore, there was a transient decrease in food intake after icv FGF4 (fig. S1K), similar to the effect after icv FGF1 (Scarlett et al, 2016). Meanwhile, body fat/lean content, physical activity and Respiratory Exchange Rate (RER) were not changed 7 days after icv FGF4 (fig. S1 to S1N). However, heat production increased (fig. S10). In addition, insulin resistance was significantly improved (as measured by the intraperitoneal insulin resistance test (IPITT)) 10 days after icv FGF4 (fig. S1P). To identify the target organs responsible for handling glucose, micro PET/CT studies were performed on db/db mice 3 days after icv FGF4 or FGF1. Deep enrichment of radiolabeled fluorodeoxyglucose (18F-FDG) was observed in the liver, skeletal muscle and brain of FGF4 and FGF1 treated mice (fig. 1F and S1Q). Consistent with the results, hepatic glycolytic genes Pklr, gck and Gys2 were up-regulated, while gluconeogenic gene Pck1 was down-regulated (fig. 1G). Liver and skeletal muscle glycogen also increased significantly (fig. 1H). Taken together, these data indicate that transient anorexia, elevated caloric production, and increased glucose uptake and use are important peripheral responses for centrally administered FGF to lower blood glucose.
To test whether these FGFs were able to induce sustained diabetes remission in other diabetes models, a hyperglycemic mouse model of diet-induced obesity (DIO) was developed by feeding a High Fat Diet (HFD). Significantly, central administration of FGF4 significantly reduced blood glucose, and the effect was maintained for > 2 weeks (fig. 1I). In contrast, FGF1 did not alleviate hyperglycemia in DIO mice nor affected their body weight (fig. S2A and S2B), consistent with previous reports (Scarlett et al, 2016). As expected, FGF4 can significantly improve glucose tolerance and insulin sensitivity in DIO mice (fig. 1J and S2C). In addition, glucose lowering in DIO mice was independent of body weight, fat/lean tissue content, food intake, physical activity and RER (fig. 1K, 1L and S2D-S2F). Like db/db mice, thermogenesis was increased (FIG. 1M), and ip administration of 30 μ G FGF4 only transiently reduced blood glucose in DIO mice (FIG. S2G). Thus, unlike FGF1, central injection of FGF4 induces sustained remission in diabetic DIO mice.
Fgfr1 in hypothalamic neurons is essential for FGF4 to exert anti-hyperglycemic function
To determine whether Fgfr1 in the CNS is a target for FGF, the expression pattern of Fgfr1 was first characterized by mRNA in situ hybridization. Fgfr1 + Relative to Dapi + Quantification of cells revealed that Fgfr1 is expressed in most cells throughout the hypothalamus, including VMH, dorsolateral hypothalamus (DMH), and ARC (fig. 2A-2C and S3A-S3C). The highest levels of expression were observed in VMH (fig. 2C and S3C). Then, the mixture is passed to Fgfr1 flox/flox Mice were injected in the hypothalamus with adeno-associated virus expressing Cre under it by a constitutive CMV promoter (AAV 8-CMV-CreGFP) to non-selectively knock out Fgfr1 in the hypothalamus (fig. 2D). The CMV promoter drives the expression of creGFP in-33% of the cells within VMH, in-24% of the cells in ARC, and in-53% of the cells in DMH (FIGS. S3E and S3F, resulting in an overall reduction of-40% of Fgfr1 expression in the hypothalamus (FIG. S3G). Note that mice lacking Fgfr1 in the hypothalamus (Fgfr 1) CMV-Cre ) Hyperglycemia and glucose tolerance were shown 6 weeks after virus injectionAnd (fig. 2F and S3I). Interestingly, these mice began to gain weight 3 weeks after injection (fig. S3H), which means that Fgfr1 also has an important regulatory effect on body weight. To directly test the function of Fgfr1 in FGF 4-mediated antihyperglycemic, FGF4 was centrally injected into hyperglycemic Fgfr1 CMV-Cre In mice. Icv FGF4 was found not to alleviate hyperglycemia, demonstrating that the ability of FGF4 to reduce glucose was blocked (fig. 2G) and body weight was not affected (fig. S3J). Therefore, fgfr1 is a key receptor in the hypothalamus to which FGF4 is centrally administered to lower blood glucose.
To determine which hypothalamic cell type mediates the effect of FGF4 on blood glucose, the Lepr-Cre line was used to knock out Fgfr1 in cells expressing the leptin receptor (Lepr) because Lepr is known to regulate metabolism (Xu et al, 2018 zhang et al, 1994. Surprisingly, specific Fgfr1 conditional knockdown in these cells did not affect blood glucose or body weight in standard food and HFD fed mice (FIGS. S4A-S4F). This knockout also did not affect the ability of icv FGF4 to reduce blood glucose in HFD fed mice (fig. S4G and S4H). Then, fgfr1 in astrocytes was knocked out using the Gfap-Cre line, and it was found that it did not block the glucose lowering effect of FGF4 (FIGS. S4I to S4M). AAV8-hSyn-CreGFP was then injected into Fgfr1 flox/flox In the hypothalamus of mice (FIG. S5A), fgfr1 (Fgfr 1) was selectively deleted in hypothalamic neurons hSyn -Cre ). Interestingly, fgfr1 hSyn-Cre Mice showed a significant increase in body weight, but blood glucose was unaffected (fig. S5B and S5C), indicating that the effects of Fgfr1 loss on body weight gain and hyperglycemia were separable. The mice were then replaced with HFD and Fgfr1 was found hSyn-Cre Body weight and blood glucose in mice were dramatically increased compared to GFP control (fig. 2H to fig. 2J). More importantly, icv FGF4 does not reduce Fgfr1 hSyn-Cre Hyperglycemia in mice (FIG. 2K), in sharp contrast to GFP control (FIG. 2L). Post icv FGF4 or GFP control body weights were not affected in Fgfr1 hson-cre mice (fig. 2M and fig. 2N). Taken together, these results suggest that the glucose lowering effect of FGF4 requires Fgfr1 in hypothalamic neurons.
Fgfr1 showed the highest level of expression in VMH (FIG. 2C), which led to the hypothesis that VMH was the primary target of FGF4. Therefore, FGF4 was directly injected into VMH of DIO mice to test its effect. Indeed, half the icv dose (1.5 μ g) was sufficient to alleviate hyperglycemia and improve glucose tolerance without affecting body weight (fig. 2O to fig. 2Q), suggesting that VMH may be the primary target region for FGF4 to exert its glucose lowering effect.
FGF4 restoration of VMH GSN function in diabetic mice
It is well known that GSN in VMH regulates blood glucose and may be important for the role of FGF4. To test the function of GSNs, their self-release rates (FR) were recorded using slice preparations in wt mice, db/db mice and FGF4 treated db/db mice (7-10 days post icv FGF 4). As expected, the conversion from normal glucose (2.5 mM) to low glucose (0.1 mM) inhibited GE neurons and stimulated GI neurons (fig. 3A and 3B). Then, the number of GE, GI and glucose Insensitive (INS) neurons were analyzed and it was found that GE neurons were present in excess of db/db, whereas GI neurons were present in deficiency of db/db mice compared to wt mice (fig. 3C). The relative numbers of GI neurons were restored to wt levels by the focused administration of FGF4 in db/db mice (FIG. 3C). The peak response and time to peak of the GSN in response to low glucose was further analyzed. For the GE neurons, the peak response to low glucose appeared to be smaller in db/db mice compared to the wt control, although the difference was not significant. The peak response increased after treatment of db/db with FGF4 (FIG. 3D and FIG. 3E). The time to peak was significantly prolonged in db/db mice compared to wt, and was fully restored by FGF4 treatment (fig. 3F). In GI neurons, the peak responses were indistinguishable between wt, db/db and FGF 4-treated db/db mice (FIGS. 3G and 3H). The time to peak was prolonged in db/db mice compared to wt, and was also restored by FGF4 treatment (fig. 3I). Taken together, these results indicate that glucose sensitivity and the relative amounts of GSNs are altered in diabetic conditions and are restored by FGF4 treatment.
In the inspired of these findings, an attempt was made to determine whether GSN is sensitive to FGF4 if FGF4 is directly applied (fig. 3J). Notably, most of the VMH GI neurons recorded (7/9, 78%) were inhibited by FGF4 perfusion, while 11% were either stimulated or insensitive to FGF4 (fig. 3K and fig. 3L). In contrast, FGF4 infusion stimulated or inhibited an equivalent number of GE neurons (fig. S6A-S6D). To test whether this acute effect is dependent on Fgfr1, PD166866, a selective Fgfr1 inhibitor, was added (Panek et al, 1998), which prevented Fgfr1 during slice recording (FIG. 3M). Remarkably, the inhibitor removed almost all FGF 4-induced neural responses (13/14, 93%); fig. 3N and fig. 3O), which indicates that FGF4 requires intact Fgfr1 for inhibition of gastrointestinal neurons. In contrast, PD166866 had essentially no effect on FGF 4-stimulated GE neurons and partially blocked FGF 4-inhibited GE neurons (fig. S6E to S6G).
In view of the ability of FGF4 to inhibit GI neurons, it was tested whether central administration of GI inhibitors could mimic the effect of FGF4 in alleviating hyperglycemia. As expected, a single icv injection of N' -nitro-L-arginine methyl ester hydrochloride (L-name), which inhibits GI neurons by inhibiting nNOS (Canabal et al, 2007), significantly ameliorates hyperglycemia for 2 days (fig. 3P). Recently, anoctamin 4 was identified as a chloride channel in the glucose-sensing pathway involving VMH GI neurons (He et al, 2020). Its inhibitor, caCciNH-A01, was found to also lower blood glucose in db/db mice for 2 days (FIG. 3P).
To explore whether FGF4 has a similar acute effect on VMH GSNs in DIO mice, the above experiments in db/db mice were therefore repeated in DIO mice. It was first demonstrated that GE neurons could be easily identified by switching to high glucose (5 mM) or low glucose, whereas GI neurons were more sensitive to low glucose and showed a small response to high glucose (fig. S7A to S7D). Therefore, for convenience, low glucose is used to distinguish between GSN subtypes. As expected, most of the recorded VMHGI neurons (6/11, 55%) were inhibited by FGF4 perfusion, while 2/11 was stimulated and 3/11 was insensitive (FIG. 3Q to FIG. 3S). To test whether Fgfr1 is required for this acute effect, fgfr1 was blocked using PD166866 (fig. 3T), resulting in 60% (3/5) of FGF 4-inhibited GI neuronal silencing (fig. 3U and fig. 3V). Thus, as in db/db mice, most GI neurons are acutely inhibited by FGF4, this acute effect being mediated primarily by Fgfr1. In contrast, a similar number of VMH GE neurons were found to be (6/18) stimulated, (7/18) inhibited, or (5/18) insensitive to FGF4 perfusion (FIGS. S7E-S7G). In addition, PD166866 blocked 60% (3/5) FGF 4-stimulated GE neurons and 50% (1/2) FGF 4-inhibited GE neurons (fig. S7H to S7J). Taken together, these results indicate that FGF4 inhibits most GI neurons in DIO mice, and that most inhibition requires Fgfr1, whereas FGF4 has an equal chance to acutely excite or inhibit GE neurons, and Fgfr1 mediates only half of this effect.
Nasal to brain delivery of FGF4 to alleviate hyperglycemia
Finally, to explore the therapeutic potential of FGF in the treatment of diabetic patients. A nasal to brain delivery system that has been used for brain-targeted drug delivery in humans has been tried by reference (Chapman et al, 2013). FGF4 was loaded into 100-nm liposomes (FIGS. 4A and 4B) and they were administered to db/db mice by the nasal drip route. Notably, nasal to brain delivery significantly reduced blood glucose in db/db mice for nearly a week without affecting body weight (fig. 4C and 4D). Thus, nasally delivered FGF4 improved glucose tolerance and insulin resistance (fig. 4E and 4F). To determine whether repeated central administration of FGF4 would cause resistance, FGF4 liposomes were administered intranasally to db/db mice every 2 weeks-3 weeks, and FGF4 was found to be able to reduce blood glucose repeatedly and efficiently without diminishing antihyperglycemic effects (fig. 4G). Therefore, intranasal administration is an effective way to deliver FGF4 for long-term anti-diabetic effects.
Discussion of the related Art
In this study, representative FGF ability to reduce blood glucose was evaluated systematically. Strong evidence is given that central or intranasal administration of FGF4 can effectively and chronically alleviate hyperglycemia in diabetic db/db mice without inducing hypoglycemia (fig. 1 and 4). Notably, FGF4 had more potent antidiabetic activity than FGF1 in diabetic DIO mice (figure 1). FGF4 is a well-established essential factor in various stages of embryonic development, including during implantation, morphogenesis and organogenesis (Beenken and Mohammadi,2009 itoh and Ornitz, 2008). This study extends the role of FGF4 in treating T2D in a sustained manner.
Attempts to identify receptors and cell classes that underlie the glucose lowering effect of FGFType, and Fgfr1 in the hypothalamus was found to be a key factor (fig. 2G). Fgfr1 is widely expressed in different hypothalamic cell types (including neurons and glia). Neuronal Fgfr1 was shown to be critical for FGF 4-mediated diabetes remission as its deletion substantially prevented the glucose lowering effect of FGF4 in DIO mice (fig. 2K). In contrast, fgfr1 is in lepr + Expression in cells and astrocytes was not necessary for FGF4 mediated glucose lowering (fig. S4). Fgfr1 was also required for sustained action of FGF1, as FGF1 was unable to lower blood glucose in DIO mice (FIG. S2A) and (Scarlett et al, 2016). However, based on the strong binding affinity between FGF1 and Fgfr1 (Suh et al, 2014), it is reasonable to assume that FGF1 will also function via Fgfr1.
GSNs are important players in glucose homeostasis. The lowering of blood glucose stimulates GI neurons, which leads to sympathetic nerve activation of the liver, adrenal medulla and pancreas, thereby increasing glucose production (Shimazu and Minokoshi, 2017). In contrast, an increase in blood glucose stimulates GE neurons, then activates sympathetic nerves innervating BAT, heart and skeletal muscles to increase glucose use (Shimazu and Minokoshi, 2017). However, the role of GSNs in diabetes pathology and prevention has not been fully explored. Here, FGF4 was shown to directly inhibit GI neurons by Fgfr1 in VMH, and pharmacological inhibition of GI neurons was sufficient to alleviate hyperglycemia (fig. 3). Consistent with known GI neurological function, FGF4 administration decreased hepatic glucose production and increased hepatic glycolysis and glycogen formation (fig. 1F-1H). Thus, the data collectively suggest that FGF4 exerts its antihyperglycemic function by inhibiting GI neurons. For GE neurons, FGF4 was observed to have a mixed effect on GE neurons, and only partial effects were dependent on Fgfr1. However, a subset of FGF 4-stimulated GE neurons may still mediate this glucose lowering effect, since activation of GE leads to glucose lowering. Whether GE neurons are involved in the antihyperglycemic effects of FGF4 remains to be investigated further.
In addition to GSN, fgfr1 is expressed in other cell types of the hypothalamus, including tanycytes. Heat shock protein 25 and pERK activation were reported in villous cells following central FGF1 administration (Brown et al, 2019, scarlett et al, 2016). In addition, tanycytes is also sensitive to glucose changes (Benford et al, 2017, fraying et al, 2011). Whether FGF requires Fgfr1 signaling in stem cells to affect blood glucose is also an important topic for future studies.
With respect to the peripheral response associated with central administration of FGF, insulin and glucagon levels did not change immediately after central administration of FGF1 or FGF4 (fig. S1J and (Scarlett et al, 2019)), suggesting that these hormones may not be the major drivers for the induction of diabetes remission. In contrast, transient decrease in food intake (fig. S1K and (Tennant et al, 2019)), increase in glucose intake (fig. 1F to 1H), and increase in energy expenditure (fig. 1M and S10) may be important contributing factors.
In conclusion, FGF4 was found to be effective and long-term in relieving hyperglycemia in diabetic db/db and DIO mice. FGF4 accomplishes this function through Fgfr1 and GSNs in the hypothalamus. FGF4 acutely inhibits GI neurons to achieve hypoglycemic effects, and re-sensitizes GSNs and restores their relative numbers over a long period of time. The identification of FGF4 as a potent and persistent antidiabetic factor may provide a new avenue for the treatment of T2D and related metabolic diseases.
Reference documents:
adams, a.c., yang, c., coskun, t., cheng, c.c., gimeno, r.e., luo, y, and kharitonnkov, a. (2012). Breadth of FGF21 metabolism is controlled by FGFR1 in adipose tissue. Mol Metab 2,31-37.
Alonge, k.m., mirzadeh, z., scarlett, j.m., logsdon, a.f., brown, j.m., cabrales, e., chan, c.k., kaiyala, k.j., bentsen, m.a., banks, w.a., et al (2020). In rats, fibroblast growth factor 1-induced diabetes is continuously alleviated requiring self-assembly of the hypothalamic peripheral neural network.
Anand, b.k., khina, g.s., sharma, k.n., dua, s., and Singh, b. (1964). The effect of glucose. Am J Physiol 207,1146-1154.
Beenken, a., and Mohammadi, m. (2009) FGF family: biology, pathophysiology, and therapy. Nat Rev Drug Discov 8,235-253.
Benford, h., bolborrea, m., pollatzek, e., lossow, k., hermans-Borgmeyer, i., liu, b., meyerhof, w., kasparov, s., and Dale, n. (2017). Glia 65,773-789.
Bentsen, m.a., rausch, d.m., mirzadeh, z., muta, k.k., scarlett, j.m., brown, j.m., herranz-Perez, v., baquero, a.f., thompson, j.e., alonge, k.m., et al (2020) transcriptomics analysis linked different hypothalamic cell types to the sustained remission of fibroblast growth factor 1-induced diabetes. Nat Commun 11,4458.
Brown, j.m., scarlett, j.m., matsen, m.e., nguyen, h.t., seer, a.l., jorgensen, r., morton, g.j., and Schwartz, m.w. (2019.) hypothalamic arcus-median eminence is a target for sustained remission of fibroblast growth factor 1-induced diabetes. Diabetes 68,1054-1061.
Canabal, d.d., song, z., potian, j.g., beuve, a., mcArdle, j.j., and Routh, v.h. (2007). Glucose, insulin and leptin signaling pathways modulate nitric oxide synthesis in hypothalamic ventral glucose-inhibited neurons. Am J Physiol Regul Integr Comp Physiol 292, R1418-1428.
Chapman, c.d., frey, w.h.,2nd, craft, s., danielyan, l., hallschmid, m., schioth, h.b., and Benedict, c. (2013). Pharm Res 30,2475-2484.
Degirolamo, C., sabba, C., and Moschetta, A. (2016.) therapeutic effects of endocrine fibroblast growth factors FGF19, FGF21, and FGF 23. Nat Rev Drug Discov 15,51-69.
Fon Tacer, k., bookout, a.l., ding, x., kurosu, h., john, g.b., wang, l., goetz, r., mohammadi, m., kuro-o, m., magelsdorf, d.j., et al. (2010) research resources: comprehensive expression profiles of fibroblast growth factor system in adult mice. Mol Endocrinol 24,2050-2064.
Frayling, c., britton, r., and Dale, n. (2011). J Physiol 589,2275-2286.
Fu, l., john, l.m., adams, s.h., yu, x.x., tomlinson, e., renz, m., williams, p.m., soriano, r., corpuz, r., moffat, b., et al (2004) fibroblast growth factor 19 increases metabolic rate, reverses diet and leptin-deficient diabetes. Endocrinology 145,2594-2603.
Goetz, r., and Mohammadi, m. (2013). Nat Rev Mol Cell Bio 14,166-180.
He, y, xu, p, wang, c, xia, y, yu, m, yang, y, yu, k, cai, x, qu, n, saito, k, et al, (2020) estrogen receptor α expression in ventral VMH regulates glucose balance. Nat Commun 11,2165.
Huang, z., tan, y., gu, j., liu, y., song, l., niu, j., zhao, l., srinivasan, l., lin, q., deng, j., et al. (2017). The mitotic and metabolic functions of FGF1 are decoupled by modulating FGF1-FGF receptor dimer stability. Cell Rep 20,1717-1728.
History of functional evolution of the mouse family of Fgf genes (2008). Dev Dyn 237,18-27.
Itoh, n., and Ornitz, d.m. (2011). From molecular evolution to roles in development, metabolism, and disease. J Biochem 149,121-130.
Jonker, j.w., suh, j.m., atkins, a.r., ahmadian, m., li, p., whiyte, j.he, m., jukuilon, h.s., yin, y.q., phillips, c.t., et al (2012) PPARgamma-FGF1 axis is necessary in adaptive fat remodeling and metabolic balance. Nature 485,391-394.
Kharitonnkov, a., shiyanova, t.l., koester, a., ford, a.m., micarovic, r., galbreth, e.j., sandusky, g.e., hammond, l.j., moyers, j.s., owens, r.a., et al.27 (2005). FGF-21 as a novel metabolic modulator. Journal of Clinical Investigation 115,1627-1635.
Tissue-specific expression of Kurosu, h., choi, m., ogawa, y., dickson, a.s., goetz, r., eliseenkova, a.v., mohammadi, m., rosenblatt, k.p., kliewer, s.a., and Kuro-o, m. (2007), betaKlotho and Fibroblast Growth Factor (FGF) receptor subtypes determines the metabolic activity of FGF19 and FGF 21.J Biol Chem 282,26687-26695.
Lan, t, morgan, d.a., rahmouni, k, sonoda, j, fu, x.r., burgess, s.c., holland, w.l., kliewer, s.a., and magelsdorf, d.j. (2017). FGF19, FGF21, and FGFR1/betaKloth activating antibodies act on the nervous system to regulate body weight and blood glucose. Cell Metabolism 26,709- +.
Marty, n., dallaporta, m., and Thorens, b. (2007). Physiology (Bethesda) 22,241-251.
Morton, g.j., matsen, m.e., bracy, d.p., meek, t.h., nguyen, h.t., stefanovski, d., bergman, r.n., wasserman, d.h., and Schwartz, m.w. (2013) FGF19 action in the brain: inducing insulin independent blood sugar reduction. J Clin Invest 123,4799-4808.
Oomura, y., ono, t., ooyama, h., and Wayner, m.j. (1969). Nature 222,282-284.
Ornitz, d.m. (2005). FGF signaling in endochondral skeletal development. Cytokine Growth Factor Rev16,205-213.
In vitro biological properties and anti-angiogenic effects of Panek, r.l., lu, g.h., dahring, t.k., butley, b.l., connolly, c., hamby, j.m., and Brown, k.j. (1998). PD 166866: selective inhibitors of FGF-1 receptor tyrosine kinase. J Pharmacol Exp Ther 286,569-577.
Perry, r.j., lee, s, ma, l, zhang, d, schlesssinger, j, and Shulman, g.i. (2015). FGF1 and FGF19 cure diabetes by inhibiting the hypothalamic-pituitary-adrenal axis. Nat Commun 6,6980.
The role of fibroblast growth factor 21 in the brain, sarrouf, d.a., thaler, j.p., morton, g.j., german, j., fischer, j.d., agimoto, k., and Schwartz, m.w. (2010): increase the energy expenditure and insulin sensitivity in obese rats. Diabetes 59,1817-1824.
Scarlett, j.m., muta, k., brown, j.m., rojas, j.m., matsen, m.e., achaya, n.k., seer, a., ingvorsen, c., jorgensen, r., hoeg-Jensen, t., et al (2019) peripheral mechanisms in brain mediate FGF1 sustained antidiabetic effects. Diabetes 68,654-664.
Central injection of fibroblast growth factor 1 induces sustained relief of diabetic hyperglycemia in rodents. Nat Med 22,800-806.
Shimazu, t., and Minokoshi, y. (2017). Systemic blood glucose regulation by glucose-sensing neurons in the hypothalamic ventral medial nucleus (VMH). J Endocr Soc 1,449-459.
Steinbusch, l., labouebe, g., and Thorens, b. (2015). Homeostatic and in feature regulation brain glucose sensing. Trends Endocrinol Metab 26,455-466.
The endocrine of Suh, j.m., jonker, j.w., ahmadian, m., goetz, r., lackey, d., osborn, o., huang, z., liu, w., yoshihara, e., van Dijk, t.h., et al (2014) FGF1 produces a new modality and effective insulin sensitizer. Nature 513,436-439.
Central and peripheral administration of Tennant, k.g., lindsley, s.r., kirigiti, m.a., true, c., and kievt, p. (2019) fibroblast growth factor 1 improved insulin secretion in diabetic mouse models. Diabetes.
Xu, j., bartolome, c.l., low, c.s., yi, x., chien, c. -h., wang, p., and Kong, d. (2018). Nature.
Ye,M.,Lu,W.,Wang,X.,Wang,C.,Abbruzzese,J.L.,Liang,G.,Li,X.,and Luo,Y.
(2016) FGF21-FGFR1 coordinates phospholipid homeostasis, lipid droplet function and endoplasmic reticulum stress in obesity. Endocrinology157,4754-4769.
Zhang, y., proenca, r., maffei, m., barone, m., leopold, l., and Friedman, j.m. (1994). Positional cloning of the mouse adiposity gene and its human homolog. Nature 372,425-432.
Claims (18)
1. A method for treating a metabolic disease comprising the step of administering a therapeutically effective dose of a Fgfr1 ligand or a pharmaceutical composition comprising the Fgfr1 ligand to a patient in need thereof via intracerebral or intranasally, wherein the metabolic disease comprises obesity, diabetes (particularly type 2 diabetes), insulin resistance, hyperinsulinemia, glucose intolerance or hyperglycemia.
2. The method of claim 1, wherein the ligand is FGF4.
3. The method of claim 2, wherein the intracerebral administration is chronic intracerebral administration.
4. The method according to claim 3, wherein the frequency of intracerebral administration is less than once every 2 weeks, preferably less than once every 5 weeks, such as once every 6 to 16 weeks.
5. The method according to claim 2, wherein the frequency of intranasal administration is less than once every 2 days, preferably less than once every 5 days, such as once every 6 to 9 days.
6. The method of claim 1, wherein the pharmaceutical composition further comprises a pharmaceutically acceptable pharmaceutical agent, such as a liposome.
7. An Fgfr1 ligand or a pharmaceutical composition comprising the Fgfr1 ligand for use in the treatment of a metabolic disease by intracerebral or intranasal administration, wherein the metabolic disease is obesity, diabetes (particularly type 2 diabetes), insulin resistance, hyperinsulinemia, glucose intolerance or hyperglycemia.
8. The Fgfr1 ligand or the pharmaceutical composition comprising the Fgfr1 ligand of claim 7 wherein the ligand is FGF4.
9. The Fgfr1 ligand or the pharmaceutical composition comprising the Fgfr1 ligand of claim 8, wherein the intracerebral administration is chronic intracerebral administration.
10. The Fgfr1 ligand or the pharmaceutical composition comprising the Fgfr1 ligand of claim 9, wherein the frequency of intracerebral administration is less than once every 2 weeks, preferably less than once every 5 weeks, such as once every 6 to 9 weeks.
11. The Fgfr1 ligand or the pharmaceutical composition comprising the Fgfr1 ligand of claim 8, wherein the frequency of intranasal administration is less than once every 2 days, preferably less than once every 5 days, such as once every 6 to 9 days.
12. The Fgfr1 ligand or the pharmaceutical composition comprising the Fgfr1 ligand of claim 7 wherein the pharmaceutical composition further comprises a pharmaceutically acceptable pharmaceutical agent such as a liposome.
13. Use of an Fgfr1 ligand or a pharmaceutical composition comprising the Fgfr1 ligand in the manufacture of a medicament for the treatment of a metabolic disease by intracerebral or intranasal administration, wherein the metabolic disease is obesity, diabetes (particularly type 2 diabetes), insulin resistance, hyperinsulinemia, glucose intolerance or hyperglycemia.
14. The use according to claim 13, wherein the ligand is FGF4.
15. The use of claim 14, wherein the intracerebral administration is chronic intracerebral administration.
16. Use according to claim 15, wherein the frequency of intracerebral administration is less than once every 2 weeks, preferably less than once every 5 weeks, such as once every 6 to 9 weeks.
17. Use according to claim 14, wherein the frequency of intranasal administration is less than once every 2 days, preferably less than once every 5 days, such as once every 6 to 9 days.
18. The use according to claim 13, wherein the pharmaceutical composition further comprises a pharmaceutically acceptable pharmaceutical formulation, such as a liposome.
Applications Claiming Priority (1)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
PCT/CN2020/126449 WO2022094789A1 (en) | 2020-11-04 | 2020-11-04 | The application of fgf4 and fgfr1 binding ligands in diabetes |
Publications (1)
Publication Number | Publication Date |
---|---|
CN115515969A true CN115515969A (en) | 2022-12-23 |
Family
ID=81458487
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
CN202080099938.7A Pending CN115515969A (en) | 2020-11-04 | 2020-11-04 | Application of FGF4 and Fgfr1 binding ligand in diabetes |
Country Status (2)
Country | Link |
---|---|
CN (1) | CN115515969A (en) |
WO (1) | WO2022094789A1 (en) |
Citations (4)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO2007110079A2 (en) * | 2006-03-29 | 2007-10-04 | Enkam Pharmaceuticals A/S | Targeted delivery of fgfr ligands into the brain |
CN105601748A (en) * | 2011-07-01 | 2016-05-25 | 恩格姆生物制药公司 | Compositions, application and method for disease metabolism and treatment of disease |
US20180228869A1 (en) * | 2015-10-30 | 2018-08-16 | Salk Institute For Biological Studies | Treatment of steroid-induced hyperglycemia with fibroblast growth factor (fgf) 1 analogs |
CN111944035A (en) * | 2019-05-14 | 2020-11-17 | 温州医科大学 | FGF4 and application thereof |
-
2020
- 2020-11-04 CN CN202080099938.7A patent/CN115515969A/en active Pending
- 2020-11-04 WO PCT/CN2020/126449 patent/WO2022094789A1/en active Application Filing
Patent Citations (4)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO2007110079A2 (en) * | 2006-03-29 | 2007-10-04 | Enkam Pharmaceuticals A/S | Targeted delivery of fgfr ligands into the brain |
CN105601748A (en) * | 2011-07-01 | 2016-05-25 | 恩格姆生物制药公司 | Compositions, application and method for disease metabolism and treatment of disease |
US20180228869A1 (en) * | 2015-10-30 | 2018-08-16 | Salk Institute For Biological Studies | Treatment of steroid-induced hyperglycemia with fibroblast growth factor (fgf) 1 analogs |
CN111944035A (en) * | 2019-05-14 | 2020-11-17 | 温州医科大学 | FGF4 and application thereof |
Non-Patent Citations (5)
Title |
---|
GASSER E等: "FGF1 - a new weapon to control type 2 diabetes mellitus", NAT REV ENDOCRINOL, vol. 13, no. 10, 30 June 2017 (2017-06-30), pages 599 - 609, XP055929006, DOI: 10.1038/nrendo.2017.78 * |
JAZWA A等: "Combined vascular endothelial growth factor-A and fibroblast growth factor 4 gene transfer improves wound healing in diabetic mice", GENET VACCINES THER, vol. 8, no. 6, 30 August 2010 (2010-08-30), pages 1 - 16 * |
SUN H等: "Sustained remission of type 2 diabetes in rodents by centrally administered fibroblast growth factor 4", CELL METAB, vol. 35, no. 6, 10 May 2023 (2023-05-10), pages 1022 - 1037, XP087330460, DOI: 10.1016/j.cmet.2023.04.018 * |
XIE, Y等: "FGF/FGFR signaling in health and disease", SIG TRANSDUCT TARGET THER 5, vol. 181, 2 September 2020 (2020-09-02), pages 1 - 38 * |
刘姗姗等: "成纤维细胞生长因子及其在代谢性疾病中的作用与临床应用研究进展", 药学进展, vol. 43, no. 1, 25 January 2019 (2019-01-25), pages 19 - 28 * |
Also Published As
Publication number | Publication date |
---|---|
WO2022094789A1 (en) | 2022-05-12 |
WO2022094789A9 (en) | 2023-04-13 |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
Lindahl et al. | Unconventional neurotrophic factors CDNF and MANF: structure, physiological functions and therapeutic potential | |
Lindholm et al. | Cerebral dopamine neurotrophic factor protects and repairs dopamine neurons by novel mechanism | |
Sun et al. | Modulation of the astrocyte-neuron lactate shuttle system contributes to neuroprotective action of fibroblast growth factor 21 | |
Zhang et al. | Regulation of neuropeptide Y in body microenvironments and its potential application in therapies: A review | |
Xiong et al. | Erythropoietin improves histological and functional outcomes after traumatic brain injury in mice in the absence of the neural erythropoietin receptor | |
Sheikh et al. | Polylysine-modified polyethylenimine (PEI-PLL) mediated VEGF gene delivery protects dopaminergic neurons in cell culture and in rat models of Parkinson's Disease (PD) | |
JP6403062B2 (en) | Tissue repair active composition and use thereof | |
Miao et al. | Overexpression of adiponectin improves neurobehavioral outcomes after focal cerebral ischemia in aged mice | |
US20200071372A1 (en) | C-terminal CDNF and MANF fragments, pharmaceutical compositions comprising same and uses thereof | |
Huang et al. | Glucagon-like peptide-1 cleavage product GLP-1 (9–36) reduces neuroinflammation from stroke via the activation of insulin-like growth factor 1 receptor in astrocytes | |
US20090038022A1 (en) | IGF-1 Novel peptides | |
JP6474549B2 (en) | Evaluation index of stem cell culture product and its use | |
PT792160E (en) | NEUROTROPHIC FACTOR DERIVED FROM GLIAL CELLS USED AS A NEUROPROTECTOR AGENT | |
EP2098244A1 (en) | Pharmaceutical composition for the treatment of myocardial infarction | |
Benissan-Messan et al. | Multi-cellular functions of MG53 in muscle calcium signaling and regeneration | |
JP4581115B2 (en) | Treatment or prevention agent for pruritic skin diseases | |
CN115279392A (en) | Pharmaceutical composition for treating TRPV1 activity mediated diseases | |
Magnan et al. | Pleiotropic effects of prokineticin 2 in the control of energy metabolism | |
CN115515969A (en) | Application of FGF4 and Fgfr1 binding ligand in diabetes | |
KR101906578B1 (en) | Pharmaceutical composition for preventing or treating neurodegenerative disease comprising cyclodextrin and VEGF overexpressing stem cells as an active agent | |
JPWO2007139120A1 (en) | Amyloid β clearance promoter | |
WO2022212268A1 (en) | Methods and compositions for treating lysosomal storage disorders | |
CN111944035B (en) | FGF4 and application thereof | |
JP2024530355A (en) | Small extracellular vesicles expressing dominant negative AMPKα1 mutants for use in the treatment of obesity | |
KR20130122927A (en) | Composition for preventing or treating erectile dysfunction comprising dkk2 protein or gene therefor and use thereof |
Legal Events
Date | Code | Title | Description |
---|---|---|---|
PB01 | Publication | ||
PB01 | Publication | ||
SE01 | Entry into force of request for substantive examination | ||
SE01 | Entry into force of request for substantive examination |