WO2008016995A2 - Methods of identifying modulators of insulin signalling - Google Patents
Methods of identifying modulators of insulin signalling Download PDFInfo
- Publication number
- WO2008016995A2 WO2008016995A2 PCT/US2007/075001 US2007075001W WO2008016995A2 WO 2008016995 A2 WO2008016995 A2 WO 2008016995A2 US 2007075001 W US2007075001 W US 2007075001W WO 2008016995 A2 WO2008016995 A2 WO 2008016995A2
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- jnk
- cdc42
- sample
- mammal
- insulin sensitivity
- Prior art date
Links
- 238000000034 method Methods 0.000 title claims abstract description 91
- 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 title claims description 178
- 102000004877 Insulin Human genes 0.000 title claims description 92
- 108090001061 Insulin Proteins 0.000 title claims description 92
- 229940125396 insulin Drugs 0.000 title claims description 89
- 230000011664 signaling Effects 0.000 title description 20
- 206010022489 Insulin Resistance Diseases 0.000 claims abstract description 87
- 239000003814 drug Substances 0.000 claims abstract description 8
- 108010055717 JNK Mitogen-Activated Protein Kinases Proteins 0.000 claims description 128
- 150000001875 compounds Chemical class 0.000 claims description 112
- 230000000694 effects Effects 0.000 claims description 112
- 108010051348 cdc42 GTP-Binding Protein Proteins 0.000 claims description 96
- 102000013515 cdc42 GTP-Binding Protein Human genes 0.000 claims description 96
- 230000004913 activation Effects 0.000 claims description 86
- 102000014160 PTEN Phosphohydrolase Human genes 0.000 claims description 69
- 108010011536 PTEN Phosphohydrolase Proteins 0.000 claims description 69
- 238000006366 phosphorylation reaction Methods 0.000 claims description 65
- 230000026731 phosphorylation Effects 0.000 claims description 64
- 238000012360 testing method Methods 0.000 claims description 62
- 102100023274 Dual specificity mitogen-activated protein kinase kinase 4 Human genes 0.000 claims description 58
- 108090000623 proteins and genes Proteins 0.000 claims description 44
- 102100025207 Mitogen-activated protein kinase kinase kinase 11 Human genes 0.000 claims description 41
- 230000007423 decrease Effects 0.000 claims description 35
- 241000124008 Mammalia Species 0.000 claims description 32
- 230000037361 pathway Effects 0.000 claims description 30
- 102000004169 proteins and genes Human genes 0.000 claims description 26
- 230000027455 binding Effects 0.000 claims description 20
- 230000001965 increasing effect Effects 0.000 claims description 20
- 150000003384 small molecules Chemical class 0.000 claims description 13
- 108090000765 processed proteins & peptides Proteins 0.000 claims description 10
- 230000002829 reductive effect Effects 0.000 claims description 6
- 230000019491 signal transduction Effects 0.000 claims description 6
- 239000000816 peptidomimetic Substances 0.000 claims description 4
- 238000010171 animal model Methods 0.000 claims description 2
- 229940124597 therapeutic agent Drugs 0.000 claims description 2
- 101001005602 Homo sapiens Mitogen-activated protein kinase kinase kinase 11 Proteins 0.000 claims 11
- 101001115395 Homo sapiens Dual specificity mitogen-activated protein kinase kinase 4 Proteins 0.000 claims 9
- 102000019145 JUN kinase activity proteins Human genes 0.000 claims 8
- 208000001072 type 2 diabetes mellitus Diseases 0.000 abstract description 27
- 206010012601 diabetes mellitus Diseases 0.000 abstract description 14
- 229940079593 drug Drugs 0.000 abstract description 6
- 241000699670 Mus sp. Species 0.000 description 103
- 210000004027 cell Anatomy 0.000 description 60
- 101000927796 Homo sapiens Rho guanine nucleotide exchange factor 7 Proteins 0.000 description 58
- 101000652725 Drosophila melanogaster Transcription initiation factor TFIID subunit 5 Proteins 0.000 description 57
- 101000752722 Homo sapiens Apoptosis-stimulating of p53 protein 1 Proteins 0.000 description 57
- 101001120056 Homo sapiens Phosphatidylinositol 3-kinase regulatory subunit alpha Proteins 0.000 description 57
- 101001120097 Homo sapiens Phosphatidylinositol 3-kinase regulatory subunit beta Proteins 0.000 description 57
- 101001116549 Homo sapiens Protein CBFA2T2 Proteins 0.000 description 57
- 101001000998 Homo sapiens Protein phosphatase 1 regulatory subunit 12C Proteins 0.000 description 57
- 102100026169 Phosphatidylinositol 3-kinase regulatory subunit alpha Human genes 0.000 description 57
- 108090000430 Phosphatidylinositol 3-kinases Proteins 0.000 description 57
- 102000003993 Phosphatidylinositol 3-kinases Human genes 0.000 description 57
- 210000004185 liver Anatomy 0.000 description 52
- 108091007960 PI3Ks Proteins 0.000 description 50
- 108010068304 MAP Kinase Kinase 4 Proteins 0.000 description 48
- 230000001105 regulatory effect Effects 0.000 description 34
- 230000014509 gene expression Effects 0.000 description 31
- 108010041596 mitogen-activated protein kinase kinase kinase 11 Proteins 0.000 description 30
- 108091008611 Protein Kinase B Proteins 0.000 description 29
- 241000701161 unidentified adenovirus Species 0.000 description 29
- ZSZXYWFCIKKZBT-IVYVYLGESA-N 1,2-dihexadecanoyl-sn-glycero-3-phospho-(1D-myo-inositol-3,4,5-trisphosphate) Chemical compound CCCCCCCCCCCCCCCC(=O)OC[C@@H](OC(=O)CCCCCCCCCCCCCCC)COP(O)(=O)O[C@@H]1[C@H](O)[C@H](OP(O)(O)=O)[C@@H](OP(O)(O)=O)[C@H](OP(O)(O)=O)[C@H]1O ZSZXYWFCIKKZBT-IVYVYLGESA-N 0.000 description 24
- 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 24
- 210000003494 hepatocyte Anatomy 0.000 description 24
- 239000008103 glucose Substances 0.000 description 23
- 230000003247 decreasing effect Effects 0.000 description 22
- 208000008589 Obesity Diseases 0.000 description 18
- 238000012217 deletion Methods 0.000 description 18
- 230000037430 deletion Effects 0.000 description 18
- 210000001519 tissue Anatomy 0.000 description 18
- 241001465754 Metazoa Species 0.000 description 17
- 102000001708 Protein Isoforms Human genes 0.000 description 17
- 108010029485 Protein Isoforms Proteins 0.000 description 17
- 238000001727 in vivo Methods 0.000 description 17
- 235000020824 obesity Nutrition 0.000 description 17
- 230000002440 hepatic effect Effects 0.000 description 16
- 235000018102 proteins Nutrition 0.000 description 16
- 230000009471 action Effects 0.000 description 15
- 235000004400 serine Nutrition 0.000 description 15
- 102100026753 Lymphokine-activated killer T-cell-originated protein kinase Human genes 0.000 description 14
- MTCFGRXMJLQNBG-UHFFFAOYSA-N Serine Natural products OCC(N)C(O)=O MTCFGRXMJLQNBG-UHFFFAOYSA-N 0.000 description 14
- 230000007246 mechanism Effects 0.000 description 14
- 235000009200 high fat diet Nutrition 0.000 description 13
- 150000002632 lipids Chemical class 0.000 description 13
- 102000014400 SH2 domains Human genes 0.000 description 12
- 108050003452 SH2 domains Proteins 0.000 description 12
- 210000004369 blood Anatomy 0.000 description 12
- 239000008280 blood Substances 0.000 description 12
- 239000000047 product Substances 0.000 description 12
- 108091000080 Phosphotransferase Proteins 0.000 description 11
- 102000020233 phosphotransferase Human genes 0.000 description 11
- 210000002966 serum Anatomy 0.000 description 11
- 230000000638 stimulation Effects 0.000 description 11
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 description 10
- 239000012634 fragment Substances 0.000 description 10
- UBXIJOJXUFYNRG-RJKBCLGNSA-N PIP[3'](17:0/20:4(5Z,8Z,11Z,14Z)) Chemical compound CCCCC\C=C/C\C=C/C\C=C/C\C=C/CCCC(=O)O[C@H](COC(=O)CCCCCCCCCCCCCCCC)COP(O)(=O)O[C@H]1C(O)C(O)C(O)[C@@H](OP(O)(O)=O)C1O UBXIJOJXUFYNRG-RJKBCLGNSA-N 0.000 description 9
- 239000006166 lysate Substances 0.000 description 9
- 230000001404 mediated effect Effects 0.000 description 9
- 230000002503 metabolic effect Effects 0.000 description 9
- IJGRMHOSHXDMSA-UHFFFAOYSA-N Atomic nitrogen Chemical compound N#N IJGRMHOSHXDMSA-UHFFFAOYSA-N 0.000 description 8
- 238000003556 assay Methods 0.000 description 8
- 230000001419 dependent effect Effects 0.000 description 8
- 238000003119 immunoblot Methods 0.000 description 8
- 239000012133 immunoprecipitate Substances 0.000 description 8
- -1 phospho Chemical class 0.000 description 8
- 238000012216 screening Methods 0.000 description 8
- 238000001262 western blot Methods 0.000 description 8
- 102000004190 Enzymes Human genes 0.000 description 7
- 108090000790 Enzymes Proteins 0.000 description 7
- 208000035475 disorder Diseases 0.000 description 7
- 229940088598 enzyme Drugs 0.000 description 7
- QTBSBXVTEAMEQO-UHFFFAOYSA-N Acetic acid Chemical compound CC(O)=O QTBSBXVTEAMEQO-UHFFFAOYSA-N 0.000 description 6
- 102100025092 Insulin receptor substrate 2 Human genes 0.000 description 6
- 101710201820 Insulin receptor substrate 2 Proteins 0.000 description 6
- OKKJLVBELUTLKV-UHFFFAOYSA-N Methanol Chemical compound OC OKKJLVBELUTLKV-UHFFFAOYSA-N 0.000 description 6
- 241000699666 Mus <mouse, genus> Species 0.000 description 6
- 150000001413 amino acids Chemical class 0.000 description 6
- 230000003197 catalytic effect Effects 0.000 description 6
- 238000011161 development Methods 0.000 description 6
- 238000002474 experimental method Methods 0.000 description 6
- 210000004602 germ cell Anatomy 0.000 description 6
- DCWXELXMIBXGTH-QMMMGPOBSA-N phosphonotyrosine Chemical compound OC(=O)[C@@H](N)CC1=CC=C(OP(O)(O)=O)C=C1 DCWXELXMIBXGTH-QMMMGPOBSA-N 0.000 description 6
- YFDSDPIBEUFTMI-UHFFFAOYSA-N tribromoethanol Chemical compound OCC(Br)(Br)Br YFDSDPIBEUFTMI-UHFFFAOYSA-N 0.000 description 6
- 102000003746 Insulin Receptor Human genes 0.000 description 5
- 108010001127 Insulin Receptor Proteins 0.000 description 5
- 208000001145 Metabolic Syndrome Diseases 0.000 description 5
- 102000000395 SH3 domains Human genes 0.000 description 5
- 108050008861 SH3 domains Proteins 0.000 description 5
- FAPWRFPIFSIZLT-UHFFFAOYSA-M Sodium chloride Chemical compound [Na+].[Cl-] FAPWRFPIFSIZLT-UHFFFAOYSA-M 0.000 description 5
- 201000000690 abdominal obesity-metabolic syndrome Diseases 0.000 description 5
- 230000003213 activating effect Effects 0.000 description 5
- 235000001014 amino acid Nutrition 0.000 description 5
- 238000004458 analytical method Methods 0.000 description 5
- 230000002596 correlated effect Effects 0.000 description 5
- 230000006870 function Effects 0.000 description 5
- 230000002068 genetic effect Effects 0.000 description 5
- 238000000338 in vitro Methods 0.000 description 5
- PBGKTOXHQIOBKM-FHFVDXKLSA-N insulin (human) Chemical compound C([C@@H](C(=O)N[C@@H](CC(C)C)C(=O)N[C@H]1CSSC[C@H]2C(=O)N[C@H](C(=O)N[C@@H](CO)C(=O)N[C@H](C(=O)N[C@H](C(N[C@@H](CO)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CC=3C=CC(O)=CC=3)C(=O)N[C@@H](CCC(N)=O)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](CC(N)=O)C(=O)N[C@@H](CC=3C=CC(O)=CC=3)C(=O)N[C@@H](CSSC[C@H](NC(=O)[C@H](C(C)C)NC(=O)[C@H](CC(C)C)NC(=O)[C@H](CC=3C=CC(O)=CC=3)NC(=O)[C@H](CC(C)C)NC(=O)[C@H](C)NC(=O)[C@H](CCC(O)=O)NC(=O)[C@H](C(C)C)NC(=O)[C@H](CC(C)C)NC(=O)[C@H](CC=3NC=NC=3)NC(=O)[C@H](CO)NC(=O)CNC1=O)C(=O)NCC(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](CCCNC(N)=N)C(=O)NCC(=O)N[C@@H](CC=1C=CC=CC=1)C(=O)N[C@@H](CC=1C=CC=CC=1)C(=O)N[C@@H](CC=1C=CC(O)=CC=1)C(=O)N[C@@H]([C@@H](C)O)C(=O)N1[C@@H](CCC1)C(=O)N[C@@H](CCCCN)C(=O)N[C@@H]([C@@H](C)O)C(O)=O)C(=O)N[C@@H](CC(N)=O)C(O)=O)=O)CSSC[C@@H](C(N2)=O)NC(=O)[C@H](CCC(N)=O)NC(=O)[C@H](CCC(O)=O)NC(=O)[C@H](C(C)C)NC(=O)[C@@H](NC(=O)CN)[C@@H](C)CC)[C@@H](C)CC)[C@@H](C)O)NC(=O)[C@H](CCC(N)=O)NC(=O)[C@H](CC(N)=O)NC(=O)[C@@H](NC(=O)[C@@H](N)CC=1C=CC=CC=1)C(C)C)C1=CN=CN1 PBGKTOXHQIOBKM-FHFVDXKLSA-N 0.000 description 5
- 230000003993 interaction Effects 0.000 description 5
- 239000000758 substrate Substances 0.000 description 5
- 102100023332 Dual specificity mitogen-activated protein kinase kinase 7 Human genes 0.000 description 4
- 108010025076 Holoenzymes Proteins 0.000 description 4
- 101000624594 Homo sapiens Dual specificity mitogen-activated protein kinase kinase 7 Proteins 0.000 description 4
- 102100037809 Mitogen-activated protein kinase 9 Human genes 0.000 description 4
- 241000283973 Oryctolagus cuniculus Species 0.000 description 4
- 102000003728 Peroxisome Proliferator-Activated Receptors Human genes 0.000 description 4
- 108090000029 Peroxisome Proliferator-Activated Receptors Proteins 0.000 description 4
- 102000004160 Phosphoric Monoester Hydrolases Human genes 0.000 description 4
- 108090000608 Phosphoric Monoester Hydrolases Proteins 0.000 description 4
- 238000000540 analysis of variance Methods 0.000 description 4
- 210000004899 c-terminal region Anatomy 0.000 description 4
- 230000008859 change Effects 0.000 description 4
- 235000005911 diet Nutrition 0.000 description 4
- 230000037213 diet Effects 0.000 description 4
- 230000003828 downregulation Effects 0.000 description 4
- 238000001114 immunoprecipitation Methods 0.000 description 4
- 238000001802 infusion Methods 0.000 description 4
- 238000011813 knockout mouse model Methods 0.000 description 4
- 230000004807 localization Effects 0.000 description 4
- 108020004999 messenger RNA Proteins 0.000 description 4
- 230000009456 molecular mechanism Effects 0.000 description 4
- 210000003205 muscle Anatomy 0.000 description 4
- 230000002018 overexpression Effects 0.000 description 4
- 102000004196 processed proteins & peptides Human genes 0.000 description 4
- 230000009467 reduction Effects 0.000 description 4
- 230000004044 response Effects 0.000 description 4
- 125000003607 serino group Chemical group [H]N([H])[C@]([H])(C(=O)[*])C(O[H])([H])[H] 0.000 description 4
- 238000012546 transfer Methods 0.000 description 4
- 150000003626 triacylglycerols Chemical class 0.000 description 4
- 241000283707 Capra Species 0.000 description 3
- 206010018429 Glucose tolerance impaired Diseases 0.000 description 3
- PEDCQBHIVMGVHV-UHFFFAOYSA-N Glycerine Chemical compound OCC(O)CO PEDCQBHIVMGVHV-UHFFFAOYSA-N 0.000 description 3
- 101000950669 Homo sapiens Mitogen-activated protein kinase 9 Proteins 0.000 description 3
- 101000714920 Homo sapiens Taste receptor type 2 member 13 Proteins 0.000 description 3
- 101000766345 Homo sapiens Tribbles homolog 3 Proteins 0.000 description 3
- DCWXELXMIBXGTH-QMMMGPOBSA-L O(4)-phosphonato-L-tyrosine(2-) Chemical compound [O-]C(=O)[C@@H]([NH3+])CC1=CC=C(OP([O-])([O-])=O)C=C1 DCWXELXMIBXGTH-QMMMGPOBSA-L 0.000 description 3
- 102100026390 Tribbles homolog 3 Human genes 0.000 description 3
- 238000002679 ablation Methods 0.000 description 3
- 239000012190 activator Substances 0.000 description 3
- 230000004075 alteration Effects 0.000 description 3
- 230000008901 benefit Effects 0.000 description 3
- 230000033228 biological regulation Effects 0.000 description 3
- 230000015572 biosynthetic process Effects 0.000 description 3
- 230000037396 body weight Effects 0.000 description 3
- 238000006243 chemical reaction Methods 0.000 description 3
- 238000010367 cloning Methods 0.000 description 3
- 230000000875 corresponding effect Effects 0.000 description 3
- 201000010099 disease Diseases 0.000 description 3
- 230000002708 enhancing effect Effects 0.000 description 3
- 235000021588 free fatty acids Nutrition 0.000 description 3
- 230000009229 glucose formation Effects 0.000 description 3
- 230000014101 glucose homeostasis Effects 0.000 description 3
- 230000004190 glucose uptake Effects 0.000 description 3
- 239000000833 heterodimer Substances 0.000 description 3
- 230000006872 improvement Effects 0.000 description 3
- 239000003112 inhibitor Substances 0.000 description 3
- 239000007924 injection Substances 0.000 description 3
- 238000002347 injection Methods 0.000 description 3
- 239000007788 liquid Substances 0.000 description 3
- 239000000463 material Substances 0.000 description 3
- 229910052757 nitrogen Inorganic materials 0.000 description 3
- 229940103453 novolin Drugs 0.000 description 3
- 102000039446 nucleic acids Human genes 0.000 description 3
- 108020004707 nucleic acids Proteins 0.000 description 3
- 150000007523 nucleic acids Chemical class 0.000 description 3
- 238000013116 obese mouse model Methods 0.000 description 3
- 210000003240 portal vein Anatomy 0.000 description 3
- 230000003389 potentiating effect Effects 0.000 description 3
- 230000009822 protein phosphorylation Effects 0.000 description 3
- 238000010379 pull-down assay Methods 0.000 description 3
- 230000035945 sensitivity Effects 0.000 description 3
- 102000030938 small GTPase Human genes 0.000 description 3
- 108060007624 small GTPase Proteins 0.000 description 3
- 239000011780 sodium chloride Substances 0.000 description 3
- 239000000243 solution Substances 0.000 description 3
- 230000001225 therapeutic effect Effects 0.000 description 3
- 230000007306 turnover Effects 0.000 description 3
- 230000003827 upregulation Effects 0.000 description 3
- 238000011144 upstream manufacturing Methods 0.000 description 3
- 108091032973 (ribonucleotides)n+m Proteins 0.000 description 2
- 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
- 108010013238 70-kDa Ribosomal Protein S6 Kinases Proteins 0.000 description 2
- 102000009027 Albumins Human genes 0.000 description 2
- 108010088751 Albumins Proteins 0.000 description 2
- 102100026008 Breakpoint cluster region protein Human genes 0.000 description 2
- 101100205088 Caenorhabditis elegans iars-1 gene Proteins 0.000 description 2
- HEDRZPFGACZZDS-UHFFFAOYSA-N Chloroform Chemical compound ClC(Cl)Cl HEDRZPFGACZZDS-UHFFFAOYSA-N 0.000 description 2
- 108010019804 Class Ia Phosphatidylinositol 3-Kinase Proteins 0.000 description 2
- 102000020849 Class Ia Phosphatidylinositol 3-Kinase Human genes 0.000 description 2
- 102000027487 Fructose-Bisphosphatase Human genes 0.000 description 2
- 108010017464 Fructose-Bisphosphatase Proteins 0.000 description 2
- 102000001267 GSK3 Human genes 0.000 description 2
- 108060006662 GSK3 Proteins 0.000 description 2
- 102000030595 Glucokinase Human genes 0.000 description 2
- 108010021582 Glucokinase Proteins 0.000 description 2
- 208000002705 Glucose Intolerance Diseases 0.000 description 2
- 102000003638 Glucose-6-Phosphatase Human genes 0.000 description 2
- 108010086800 Glucose-6-Phosphatase Proteins 0.000 description 2
- 108010067218 Guanine Nucleotide Exchange Factors Proteins 0.000 description 2
- 102000016285 Guanine Nucleotide Exchange Factors Human genes 0.000 description 2
- 206010019708 Hepatic steatosis Diseases 0.000 description 2
- 101150030450 IRS1 gene Proteins 0.000 description 2
- 102000043136 MAP kinase family Human genes 0.000 description 2
- 108091054455 MAP kinase family Proteins 0.000 description 2
- 102100037808 Mitogen-activated protein kinase 8 Human genes 0.000 description 2
- 108091034117 Oligonucleotide Proteins 0.000 description 2
- 108010043958 Peptoids Proteins 0.000 description 2
- 102000001253 Protein Kinase Human genes 0.000 description 2
- 108010024221 Proto-Oncogene Proteins c-bcr Proteins 0.000 description 2
- 238000010240 RT-PCR analysis Methods 0.000 description 2
- 241000283984 Rodentia Species 0.000 description 2
- 238000000692 Student's t-test Methods 0.000 description 2
- 102000006467 TATA-Box Binding Protein Human genes 0.000 description 2
- 108010044281 TATA-Box Binding Protein Proteins 0.000 description 2
- 108700019146 Transgenes Proteins 0.000 description 2
- ZKHQWZAMYRWXGA-KNYAHOBESA-N [[(2r,3s,4r,5r)-5-(6-aminopurin-9-yl)-3,4-dihydroxyoxolan-2-yl]methoxy-hydroxyphosphoryl] dihydroxyphosphoryl hydrogen phosphate Chemical compound C1=NC=2C(N)=NC=NC=2N1[C@@H]1O[C@H](COP(O)(=O)OP(O)(=O)O[32P](O)(O)=O)[C@@H](O)[C@H]1O ZKHQWZAMYRWXGA-KNYAHOBESA-N 0.000 description 2
- 238000009825 accumulation Methods 0.000 description 2
- 239000012131 assay buffer Substances 0.000 description 2
- 230000004071 biological effect Effects 0.000 description 2
- 239000000872 buffer Substances 0.000 description 2
- 230000015556 catabolic process Effects 0.000 description 2
- 230000005754 cellular signaling Effects 0.000 description 2
- 239000002299 complementary DNA Substances 0.000 description 2
- 230000007547 defect Effects 0.000 description 2
- 238000001514 detection method Methods 0.000 description 2
- 229950006137 dexfosfoserine Drugs 0.000 description 2
- 230000003292 diminished effect Effects 0.000 description 2
- 229910001873 dinitrogen Inorganic materials 0.000 description 2
- 238000009510 drug design Methods 0.000 description 2
- 239000012636 effector Substances 0.000 description 2
- 238000005516 engineering process Methods 0.000 description 2
- 239000003623 enhancer Substances 0.000 description 2
- 230000001610 euglycemic effect Effects 0.000 description 2
- 239000000284 extract Substances 0.000 description 2
- 230000037406 food intake Effects 0.000 description 2
- 235000012631 food intake Nutrition 0.000 description 2
- 230000001890 gluconeogenic effect Effects 0.000 description 2
- 238000007446 glucose tolerance test Methods 0.000 description 2
- 239000011539 homogenization buffer Substances 0.000 description 2
- 230000005764 inhibitory process Effects 0.000 description 2
- 238000000021 kinase assay Methods 0.000 description 2
- 230000004132 lipogenesis Effects 0.000 description 2
- 229930014626 natural product Natural products 0.000 description 2
- 230000007310 pathophysiology Effects 0.000 description 2
- 230000002093 peripheral effect Effects 0.000 description 2
- YBYRMVIVWMBXKQ-UHFFFAOYSA-N phenylmethanesulfonyl fluoride Chemical compound FS(=O)(=O)CC1=CC=CC=C1 YBYRMVIVWMBXKQ-UHFFFAOYSA-N 0.000 description 2
- 150000003905 phosphatidylinositols Chemical class 0.000 description 2
- 229930029653 phosphoenolpyruvate Natural products 0.000 description 2
- DTBNBXWJWCWCIK-UHFFFAOYSA-N phosphoenolpyruvic acid Chemical compound OC(=O)C(=C)OP(O)(O)=O DTBNBXWJWCWCIK-UHFFFAOYSA-N 0.000 description 2
- 229920001184 polypeptide Polymers 0.000 description 2
- 230000004481 post-translational protein modification Effects 0.000 description 2
- 230000008569 process Effects 0.000 description 2
- BDERNNFJNOPAEC-UHFFFAOYSA-N propan-1-ol Chemical compound CCCO BDERNNFJNOPAEC-UHFFFAOYSA-N 0.000 description 2
- 108060006633 protein kinase Proteins 0.000 description 2
- 230000002797 proteolythic effect Effects 0.000 description 2
- 238000011160 research Methods 0.000 description 2
- 238000010839 reverse transcription Methods 0.000 description 2
- 238000003757 reverse transcription PCR Methods 0.000 description 2
- 238000007619 statistical method Methods 0.000 description 2
- 238000005556 structure-activity relationship Methods 0.000 description 2
- 230000004960 subcellular localization Effects 0.000 description 2
- 238000006467 substitution reaction Methods 0.000 description 2
- 238000003786 synthesis reaction Methods 0.000 description 2
- 235000008521 threonine Nutrition 0.000 description 2
- 229950004616 tribromoethanol Drugs 0.000 description 2
- 210000001631 vena cava inferior Anatomy 0.000 description 2
- GZCGUPFRVQAUEE-NKKIJKBOSA-N (2r,3s,4r,5r)-2,3,4,5,6-pentahydroxy-3-tritiohexanal Chemical compound O=C[C@H](O)[C@](O)([3H])[C@H](O)[C@H](O)CO GZCGUPFRVQAUEE-NKKIJKBOSA-N 0.000 description 1
- KJAXEBRGQOHHOY-VXRVIWLSSA-N (4s)-4-[[(2s)-2-[[(2s)-2-[[(2s)-2-[[(2s)-2-[[(2s,3r)-2-[[(2s)-2-[[(2s)-1-[(2s)-2-[(2-aminoacetyl)amino]-5-(diaminomethylideneamino)pentanoyl]pyrrolidine-2-carbonyl]amino]-5-(diaminomethylideneamino)pentanoyl]amino]-3-hydroxybutanoyl]amino]-3-hydroxypropan Chemical compound N([C@@H](CCCN=C(N)N)C(=O)N[C@@H]([C@H](O)C)C(=O)N[C@@H](CO)C(=O)N[C@@H](CO)C(=O)N[C@@H](CC=1C=CC=CC=1)C(=O)N[C@@H](C)C(=O)N[C@@H](CCC(O)=O)C(=O)NCC(O)=O)C(=O)[C@@H]1CCCN1C(=O)[C@H](CCCN=C(N)N)NC(=O)CN KJAXEBRGQOHHOY-VXRVIWLSSA-N 0.000 description 1
- MYGCFWRBKKQKCG-GBWOLBBFSA-N (z,2r,3s,4r)-hex-5-ene-1,2,3,4,6-pentol Chemical compound OC[C@@H](O)[C@@H](O)[C@H](O)\C=C/O MYGCFWRBKKQKCG-GBWOLBBFSA-N 0.000 description 1
- BHNQPLPANNDEGL-UHFFFAOYSA-N 2-(4-octylphenoxy)ethanol Chemical compound CCCCCCCCC1=CC=C(OCCO)C=C1 BHNQPLPANNDEGL-UHFFFAOYSA-N 0.000 description 1
- FWBHETKCLVMNFS-UHFFFAOYSA-N 4',6-Diamino-2-phenylindol Chemical compound C1=CC(C(=N)N)=CC=C1C1=CC2=CC=C(C(N)=N)C=C2N1 FWBHETKCLVMNFS-UHFFFAOYSA-N 0.000 description 1
- 206010001258 Adenoviral infections Diseases 0.000 description 1
- 230000007730 Akt signaling Effects 0.000 description 1
- 239000012099 Alexa Fluor family Substances 0.000 description 1
- 239000004475 Arginine Substances 0.000 description 1
- 201000004569 Blindness Diseases 0.000 description 1
- 102000002110 C2 domains Human genes 0.000 description 1
- 108050009459 C2 domains Proteins 0.000 description 1
- 101100322915 Caenorhabditis elegans akt-1 gene Proteins 0.000 description 1
- 102000008186 Collagen Human genes 0.000 description 1
- 108010035532 Collagen Proteins 0.000 description 1
- 102000029816 Collagenase Human genes 0.000 description 1
- 108060005980 Collagenase Proteins 0.000 description 1
- 108010069514 Cyclic Peptides Proteins 0.000 description 1
- 102000001189 Cyclic Peptides Human genes 0.000 description 1
- 108090000695 Cytokines Proteins 0.000 description 1
- 102000004127 Cytokines Human genes 0.000 description 1
- 108020004414 DNA Proteins 0.000 description 1
- 101710146518 Dual specificity mitogen-activated protein kinase kinase 4 Proteins 0.000 description 1
- KCXVZYZYPLLWCC-UHFFFAOYSA-N EDTA Chemical compound OC(=O)CN(CC(O)=O)CCN(CC(O)=O)CC(O)=O KCXVZYZYPLLWCC-UHFFFAOYSA-N 0.000 description 1
- 238000002965 ELISA Methods 0.000 description 1
- 108700024394 Exon Proteins 0.000 description 1
- 102000010834 Extracellular Matrix Proteins Human genes 0.000 description 1
- 108010037362 Extracellular Matrix Proteins Proteins 0.000 description 1
- 101150023900 G6PC1 gene Proteins 0.000 description 1
- 101000834253 Gallus gallus Actin, cytoplasmic 1 Proteins 0.000 description 1
- 206010019663 Hepatic failure Diseases 0.000 description 1
- 241000282412 Homo Species 0.000 description 1
- 101000950695 Homo sapiens Mitogen-activated protein kinase 8 Proteins 0.000 description 1
- 101150088952 IGF1 gene Proteins 0.000 description 1
- 206010056997 Impaired fasting glucose Diseases 0.000 description 1
- 108090000723 Insulin-Like Growth Factor I Proteins 0.000 description 1
- 102000004218 Insulin-Like Growth Factor I Human genes 0.000 description 1
- ONIBWKKTOPOVIA-BYPYZUCNSA-N L-Proline Chemical compound OC(=O)[C@@H]1CCCN1 ONIBWKKTOPOVIA-BYPYZUCNSA-N 0.000 description 1
- OUYCCCASQSFEME-QMMMGPOBSA-N L-tyrosine Chemical compound OC(=O)[C@@H](N)CC1=CC=C(O)C=C1 OUYCCCASQSFEME-QMMMGPOBSA-N 0.000 description 1
- 108700000058 Medicago sativa MMK4 Proteins 0.000 description 1
- 108700027648 Mitogen-Activated Protein Kinase 8 Proteins 0.000 description 1
- 108700027653 Mitogen-Activated Protein Kinase 9 Proteins 0.000 description 1
- 101100299504 Mus musculus Pten gene Proteins 0.000 description 1
- 229910020700 Na3VO4 Inorganic materials 0.000 description 1
- BZQFBWGGLXLEPQ-UHFFFAOYSA-N O-phosphoryl-L-serine Natural products OC(=O)C(N)COP(O)(O)=O BZQFBWGGLXLEPQ-UHFFFAOYSA-N 0.000 description 1
- 238000010222 PCR analysis Methods 0.000 description 1
- 108010014971 PDZ-binding kinase Proteins 0.000 description 1
- 108010067902 Peptide Library Proteins 0.000 description 1
- 102100032543 Phosphatidylinositol 3,4,5-trisphosphate 3-phosphatase and dual-specificity protein phosphatase PTEN Human genes 0.000 description 1
- 101710132081 Phosphatidylinositol 3,4,5-trisphosphate 3-phosphatase and dual-specificity protein phosphatase PTEN Proteins 0.000 description 1
- 108700019535 Phosphoprotein Phosphatases Proteins 0.000 description 1
- 102000045595 Phosphoprotein Phosphatases Human genes 0.000 description 1
- 208000001280 Prediabetic State Diseases 0.000 description 1
- 102100033237 Pro-epidermal growth factor Human genes 0.000 description 1
- ONIBWKKTOPOVIA-UHFFFAOYSA-N Proline Natural products OC(=O)C1CCCN1 ONIBWKKTOPOVIA-UHFFFAOYSA-N 0.000 description 1
- 229940124158 Protease/peptidase inhibitor Drugs 0.000 description 1
- 108090000315 Protein Kinase C Proteins 0.000 description 1
- 102000003923 Protein Kinase C Human genes 0.000 description 1
- 208000001647 Renal Insufficiency Diseases 0.000 description 1
- 102100033200 Rho guanine nucleotide exchange factor 7 Human genes 0.000 description 1
- 240000004808 Saccharomyces cerevisiae Species 0.000 description 1
- 101100221606 Saccharomyces cerevisiae (strain ATCC 204508 / S288c) COS7 gene Proteins 0.000 description 1
- 102100025252 StAR-related lipid transfer protein 13 Human genes 0.000 description 1
- 101710173511 Tensin homolog Proteins 0.000 description 1
- AYFVYJQAPQTCCC-UHFFFAOYSA-N Threonine Natural products CC(O)C(N)C(O)=O AYFVYJQAPQTCCC-UHFFFAOYSA-N 0.000 description 1
- 239000004473 Threonine Substances 0.000 description 1
- JLCPHMBAVCMARE-UHFFFAOYSA-N [3-[[3-[[3-[[3-[[3-[[3-[[3-[[3-[[3-[[3-[[3-[[5-(2-amino-6-oxo-1H-purin-9-yl)-3-[[3-[[3-[[3-[[3-[[3-[[5-(2-amino-6-oxo-1H-purin-9-yl)-3-[[5-(2-amino-6-oxo-1H-purin-9-yl)-3-hydroxyoxolan-2-yl]methoxy-hydroxyphosphoryl]oxyoxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(5-methyl-2,4-dioxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxyoxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(5-methyl-2,4-dioxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(5-methyl-2,4-dioxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(5-methyl-2,4-dioxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methyl [5-(6-aminopurin-9-yl)-2-(hydroxymethyl)oxolan-3-yl] hydrogen phosphate Polymers Cc1cn(C2CC(OP(O)(=O)OCC3OC(CC3OP(O)(=O)OCC3OC(CC3O)n3cnc4c3nc(N)[nH]c4=O)n3cnc4c3nc(N)[nH]c4=O)C(COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3CO)n3cnc4c(N)ncnc34)n3ccc(N)nc3=O)n3cnc4c(N)ncnc34)n3ccc(N)nc3=O)n3ccc(N)nc3=O)n3ccc(N)nc3=O)n3cnc4c(N)ncnc34)n3cnc4c(N)ncnc34)n3cc(C)c(=O)[nH]c3=O)n3cc(C)c(=O)[nH]c3=O)n3ccc(N)nc3=O)n3cc(C)c(=O)[nH]c3=O)n3cnc4c3nc(N)[nH]c4=O)n3cnc4c(N)ncnc34)n3cnc4c(N)ncnc34)n3cnc4c(N)ncnc34)n3cnc4c(N)ncnc34)O2)c(=O)[nH]c1=O JLCPHMBAVCMARE-UHFFFAOYSA-N 0.000 description 1
- 239000012574 advanced DMEM Substances 0.000 description 1
- 235000004279 alanine Nutrition 0.000 description 1
- 125000003295 alanine group Chemical group N[C@@H](C)C(=O)* 0.000 description 1
- 230000008848 allosteric regulation Effects 0.000 description 1
- 150000001408 amides Chemical class 0.000 description 1
- 238000002266 amputation Methods 0.000 description 1
- 239000005557 antagonist Substances 0.000 description 1
- 229940045988 antineoplastic drug protein kinase inhibitors Drugs 0.000 description 1
- 230000006907 apoptotic process Effects 0.000 description 1
- ODKSFYDXXFIFQN-UHFFFAOYSA-N arginine Natural products OC(=O)C(N)CCCNC(N)=N ODKSFYDXXFIFQN-UHFFFAOYSA-N 0.000 description 1
- 230000002238 attenuated effect Effects 0.000 description 1
- 238000000376 autoradiography Methods 0.000 description 1
- 230000003115 biocidal effect Effects 0.000 description 1
- 210000004556 brain Anatomy 0.000 description 1
- 210000001593 brown adipocyte Anatomy 0.000 description 1
- 102200062311 c.1096A>G Human genes 0.000 description 1
- AIYUHDOJVYHVIT-UHFFFAOYSA-M caesium chloride Chemical compound [Cl-].[Cs+] AIYUHDOJVYHVIT-UHFFFAOYSA-M 0.000 description 1
- 230000000453 cell autonomous effect Effects 0.000 description 1
- 230000010261 cell growth Effects 0.000 description 1
- 239000013592 cell lysate Substances 0.000 description 1
- 230000001413 cellular effect Effects 0.000 description 1
- 230000030570 cellular localization Effects 0.000 description 1
- 230000019522 cellular metabolic process Effects 0.000 description 1
- 210000003169 central nervous system Anatomy 0.000 description 1
- 229920001436 collagen Polymers 0.000 description 1
- 229960002424 collagenase Drugs 0.000 description 1
- 238000004891 communication Methods 0.000 description 1
- 230000001447 compensatory effect Effects 0.000 description 1
- 239000013078 crystal Substances 0.000 description 1
- 238000005520 cutting process Methods 0.000 description 1
- 230000003436 cytoskeletal effect Effects 0.000 description 1
- 238000000326 densiometry Methods 0.000 description 1
- 230000030609 dephosphorylation Effects 0.000 description 1
- 238000006209 dephosphorylation reaction Methods 0.000 description 1
- 239000008121 dextrose Substances 0.000 description 1
- 230000004069 differentiation Effects 0.000 description 1
- 238000009826 distribution Methods 0.000 description 1
- 230000000667 effect on insulin Effects 0.000 description 1
- 230000002124 endocrine Effects 0.000 description 1
- 201000010063 epididymitis Diseases 0.000 description 1
- 150000002148 esters Chemical class 0.000 description 1
- DEFVIWRASFVYLL-UHFFFAOYSA-N ethylene glycol bis(2-aminoethyl)tetraacetic acid Chemical compound OC(=O)CN(CC(O)=O)CCOCCOCCN(CC(O)=O)CC(O)=O DEFVIWRASFVYLL-UHFFFAOYSA-N 0.000 description 1
- 210000002744 extracellular matrix Anatomy 0.000 description 1
- 230000037433 frameshift Effects 0.000 description 1
- 239000000446 fuel Substances 0.000 description 1
- 238000010230 functional analysis Methods 0.000 description 1
- 108020001507 fusion proteins Proteins 0.000 description 1
- 102000037865 fusion proteins Human genes 0.000 description 1
- 208000004104 gestational diabetes Diseases 0.000 description 1
- 201000007116 gestational trophoblastic neoplasm Diseases 0.000 description 1
- 230000004110 gluconeogenesis Effects 0.000 description 1
- ZDXPYRJPNDTMRX-UHFFFAOYSA-N glutamine Natural products OC(=O)C(N)CCC(N)=O ZDXPYRJPNDTMRX-UHFFFAOYSA-N 0.000 description 1
- 230000012010 growth Effects 0.000 description 1
- 239000003102 growth factor Substances 0.000 description 1
- 230000036541 health Effects 0.000 description 1
- 208000019622 heart disease Diseases 0.000 description 1
- 125000000623 heterocyclic group Chemical group 0.000 description 1
- 230000013632 homeostatic process Effects 0.000 description 1
- 238000009396 hybridization Methods 0.000 description 1
- 239000001257 hydrogen Substances 0.000 description 1
- 229910052739 hydrogen Inorganic materials 0.000 description 1
- 238000010166 immunofluorescence Methods 0.000 description 1
- 238000003125 immunofluorescent labeling Methods 0.000 description 1
- 230000002621 immunoprecipitating effect Effects 0.000 description 1
- 238000000099 in vitro assay Methods 0.000 description 1
- 230000002779 inactivation Effects 0.000 description 1
- 230000028709 inflammatory response Effects 0.000 description 1
- 230000002401 inhibitory effect Effects 0.000 description 1
- 238000003780 insertion Methods 0.000 description 1
- 230000037431 insertion Effects 0.000 description 1
- 238000007912 intraperitoneal administration Methods 0.000 description 1
- 239000007928 intraperitoneal injection Substances 0.000 description 1
- 238000011835 investigation Methods 0.000 description 1
- 238000002955 isolation Methods 0.000 description 1
- 210000004731 jugular vein Anatomy 0.000 description 1
- 201000006370 kidney failure Diseases 0.000 description 1
- 231100000225 lethality Toxicity 0.000 description 1
- 239000003446 ligand Substances 0.000 description 1
- 230000000670 limiting effect Effects 0.000 description 1
- 230000004576 lipid-binding Effects 0.000 description 1
- 208000007903 liver failure Diseases 0.000 description 1
- 231100000835 liver failure Toxicity 0.000 description 1
- 230000003908 liver function Effects 0.000 description 1
- 238000012423 maintenance Methods 0.000 description 1
- 239000003550 marker Substances 0.000 description 1
- 239000011159 matrix material Substances 0.000 description 1
- 230000037353 metabolic pathway Effects 0.000 description 1
- 239000000203 mixture Substances 0.000 description 1
- 230000004048 modification Effects 0.000 description 1
- 238000012986 modification Methods 0.000 description 1
- 238000010172 mouse model Methods 0.000 description 1
- 238000001964 muscle biopsy Methods 0.000 description 1
- 208000010125 myocardial infarction Diseases 0.000 description 1
- NFVJNJQRWPQVOA-UHFFFAOYSA-N n-[2-chloro-5-(trifluoromethyl)phenyl]-2-[3-(4-ethyl-5-ethylsulfanyl-1,2,4-triazol-3-yl)piperidin-1-yl]acetamide Chemical compound CCN1C(SCC)=NN=C1C1CN(CC(=O)NC=2C(=CC=C(C=2)C(F)(F)F)Cl)CCC1 NFVJNJQRWPQVOA-UHFFFAOYSA-N 0.000 description 1
- 230000007935 neutral effect Effects 0.000 description 1
- 238000005457 optimization Methods 0.000 description 1
- 230000000803 paradoxical effect Effects 0.000 description 1
- 230000036961 partial effect Effects 0.000 description 1
- 239000000137 peptide hydrolase inhibitor Substances 0.000 description 1
- 230000010412 perfusion Effects 0.000 description 1
- 230000009984 peri-natal effect Effects 0.000 description 1
- 239000012071 phase Substances 0.000 description 1
- 150000003904 phospholipids Chemical class 0.000 description 1
- BZQFBWGGLXLEPQ-REOHCLBHSA-N phosphoserine Chemical compound OC(=O)[C@@H](N)COP(O)(O)=O BZQFBWGGLXLEPQ-REOHCLBHSA-N 0.000 description 1
- 230000008288 physiological mechanism Effects 0.000 description 1
- 230000007180 physiological regulation Effects 0.000 description 1
- 230000008092 positive effect Effects 0.000 description 1
- 230000009596 postnatal growth Effects 0.000 description 1
- 201000009104 prediabetes syndrome Diseases 0.000 description 1
- 238000004321 preservation Methods 0.000 description 1
- 230000002265 prevention Effects 0.000 description 1
- 230000002035 prolonged effect Effects 0.000 description 1
- 238000002731 protein assay Methods 0.000 description 1
- 239000003909 protein kinase inhibitor Substances 0.000 description 1
- 238000000746 purification Methods 0.000 description 1
- 238000011002 quantification Methods 0.000 description 1
- 239000000700 radioactive tracer Substances 0.000 description 1
- 239000011535 reaction buffer Substances 0.000 description 1
- 238000011084 recovery Methods 0.000 description 1
- 230000003938 response to stress Effects 0.000 description 1
- 230000002441 reversible effect Effects 0.000 description 1
- 108010004650 rho GTPase-activating protein Proteins 0.000 description 1
- 230000022932 ruffle assembly Effects 0.000 description 1
- 230000009919 sequestration Effects 0.000 description 1
- 150000003355 serines Chemical class 0.000 description 1
- 210000002027 skeletal muscle Anatomy 0.000 description 1
- FQENQNTWSFEDLI-UHFFFAOYSA-J sodium diphosphate Chemical compound [Na+].[Na+].[Na+].[Na+].[O-]P([O-])(=O)OP([O-])([O-])=O FQENQNTWSFEDLI-UHFFFAOYSA-J 0.000 description 1
- 238000002415 sodium dodecyl sulfate polyacrylamide gel electrophoresis Methods 0.000 description 1
- 239000001488 sodium phosphate Substances 0.000 description 1
- 229910000162 sodium phosphate Inorganic materials 0.000 description 1
- 239000007790 solid phase Substances 0.000 description 1
- 239000006104 solid solution Substances 0.000 description 1
- 241000894007 species Species 0.000 description 1
- 238000010186 staining Methods 0.000 description 1
- 238000010561 standard procedure Methods 0.000 description 1
- 210000003518 stress fiber Anatomy 0.000 description 1
- 239000000126 substance Substances 0.000 description 1
- 230000001629 suppression Effects 0.000 description 1
- 230000002459 sustained effect Effects 0.000 description 1
- 230000002194 synthesizing effect Effects 0.000 description 1
- 230000009897 systematic effect Effects 0.000 description 1
- 230000008685 targeting Effects 0.000 description 1
- 150000003588 threonines Chemical class 0.000 description 1
- RYFMWSXOAZQYPI-UHFFFAOYSA-K trisodium phosphate Chemical compound [Na+].[Na+].[Na+].[O-]P([O-])([O-])=O RYFMWSXOAZQYPI-UHFFFAOYSA-K 0.000 description 1
- IHIXIJGXTJIKRB-UHFFFAOYSA-N trisodium vanadate Chemical compound [Na+].[Na+].[Na+].[O-][V]([O-])([O-])=O IHIXIJGXTJIKRB-UHFFFAOYSA-N 0.000 description 1
- OUYCCCASQSFEME-UHFFFAOYSA-N tyrosine Natural products OC(=O)C(N)CC1=CC=C(O)C=C1 OUYCCCASQSFEME-UHFFFAOYSA-N 0.000 description 1
- LSGOVYNHVSXFFJ-UHFFFAOYSA-N vanadate(3-) Chemical compound [O-][V]([O-])([O-])=O LSGOVYNHVSXFFJ-UHFFFAOYSA-N 0.000 description 1
- 210000003462 vein Anatomy 0.000 description 1
- 230000035899 viability Effects 0.000 description 1
- 238000005406 washing Methods 0.000 description 1
- 230000003442 weekly effect Effects 0.000 description 1
- 230000004584 weight gain Effects 0.000 description 1
- 235000019786 weight gain Nutrition 0.000 description 1
- QDLHCMPXEPAAMD-UHFFFAOYSA-N wortmannin Natural products COCC1OC(=O)C2=COC(C3=O)=C2C1(C)C1=C3C2CCC(=O)C2(C)CC1OC(C)=O QDLHCMPXEPAAMD-UHFFFAOYSA-N 0.000 description 1
- QDLHCMPXEPAAMD-QAIWCSMKSA-N wortmannin Chemical compound C1([C@]2(C)C3=C(C4=O)OC=C3C(=O)O[C@@H]2COC)=C4[C@@H]2CCC(=O)[C@@]2(C)C[C@H]1OC(C)=O QDLHCMPXEPAAMD-QAIWCSMKSA-N 0.000 description 1
Classifications
-
- G—PHYSICS
- G01—MEASURING; TESTING
- G01N—INVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
- G01N33/00—Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
- G01N33/48—Biological material, e.g. blood, urine; Haemocytometers
- G01N33/50—Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
- G01N33/68—Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
- G01N33/6893—Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids related to diseases not provided for elsewhere
-
- G—PHYSICS
- G01—MEASURING; TESTING
- G01N—INVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
- G01N33/00—Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
- G01N33/48—Biological material, e.g. blood, urine; Haemocytometers
- G01N33/50—Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
- G01N33/5005—Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
- G01N33/5008—Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
- G01N33/502—Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics for testing non-proliferative effects
- G01N33/5041—Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics for testing non-proliferative effects involving analysis of members of signalling pathways
-
- G—PHYSICS
- G01—MEASURING; TESTING
- G01N—INVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
- G01N2500/00—Screening for compounds of potential therapeutic value
Definitions
- This invention relates to methods of identifying drugs for the treatment of insulin resistance and diabetes.
- Type 2 diabetes is the leading cause of kidney failure, blindness, and amputations, and is a major risk factor for heart disease and stroke.
- Hepatic steatosis is the second most common cause of liver failure in the U.S., and the metabolic syndrome is a major risk factor in as many as 60% of individuals suffering heart attack or stroke.
- the metabolic syndrome is a major risk factor in as many as 60% of individuals suffering heart attack or stroke.
- PI 3-kinase phosphatidylinositol 3-kinase
- the present invention is based, at least in part, on novel mechanistic insights into the connection between PI3K/JNK signalling and improved insulin sensitivity.
- Provided herein are a number of screening methods that use the proteins in this pathway as targets.
- the invention provides methods for identifying candidate compounds for improving insulin sensitivity, e.g., in a mammal in need thereof.
- the methods include providing a sample comprising p85 ⁇ and cdc42; contacting the sample with a test compound, and evaluating binding of p85 ⁇ to cdc42 in the sample.
- a test compound that decreases binding of p85 ⁇ to cdc42 is a candidate compound for improving insulin sensitivity in a mammal.
- the invention provides methods for identifying candidate compounds for improving insulin sensitivity, e.g., in a mammal in need thereof.
- the methods include providing a sample comprising cdc42 and MLK3; contacting the sample with a test compound, and evaluating phosphorylation of MLK3 by cdc42 in the sample.
- a test compound that decreases phosphorylation of MLK3 by cdc42 is a candidate compound for improving insulin sensitivity in a mammal.
- the invention provides methods for identifying candidate compounds for improving insulin sensitivity, e.g., in a mammal in need thereof.
- the methods include providing a sample comprising MLK3 and MKK4; contacting the sample with a test compound, and evaluating phosphorylation of MKK4 by MLK3 in the sample.
- a test compound that decreases phosphorylation of MKK4 by MLK3 is a candidate compound for improving insulin sensitivity in a mammal.
- the invention provides methods for identifying candidate compounds for improving insulin sensitivity, e.g., in a mammal in need thereof.
- the methods include providing a sample comprising MKK4 and JNK; contacting the sample with a test compound, and evaluating phosphorylation of JNK by MKK4 in the sample.
- a test compound that decreases phosphorylation of JNK by MKK4 is a candidate compound for improving insulin sensitivity in a mammal.
- the invention provides methods for identifying candidate compounds for improving insulin sensitivity, e.g., in a mammal in need thereof.
- the methods include providing a sample comprising PTEN and JNK; contacting the sample with a test compound, and evaluating phosphorylation of PTEN by JNK in the sample.
- a test compound that decreases phosphorylation of PTEN by JNK is a candidate compound for improving insulin sensitivity in a mammal.
- the invention provides methods for identifying candidate compounds for improving insulin sensitivity, e.g., in a mammal in need thereof.
- the methods include providing a sample comprising one or more target proteins selected from the group consisting of cdc42, MLK3, or MKK4; contacting the sample with a test compound; evaluating binding of the test compounds to the target protein; and selecting the test compound as a candidate compound if it binds to the target protein.
- the methods include providing a cell having a functional insulin signalling pathway comprising p85 ⁇ , cdc42, MLK3, MKK4, and JNK; contacting the cell with the candidate compound; contacting the cell with an amount of insulin sufficient to activate said pathway; evaluating activation of said pathway in the cell in the presence of the test compound; comparing activation of said pathway in the cell in the presence of the test compound to a reference representing activation of said pathway in the cell in the absence of the test compound, and selecting the candidate compound as a candidate therapeutic agent for improving insulin sensitivity in a mammal if activation of said pathway is reduced in the presence of the test compound as compared to activation of said pathway in the absence of the test compound.
- activation of said pathway is determined by one or more of detecting cdc42 activation of MLK3; detecting binding of cdc42 to MLK3; detecting MLK3 kinase activity; detecting phosphorylation of MKK4; or detecting JNK activation.
- the test compound can be, e.g., a small molecule, or a peptide or peptidomimetic.
- small molecules refers to small organic or inorganic molecules of molecular weight below about 3,000 Daltons.
- the sample is or includes a cell, e.g., a cell expressing the recited proteins, either endogenously or exogenously..
- the methods described herein further include administering the candidate compound to a mammal, e.g., a mammal in need of increased insulin sensitivity, and evaluating whether the candidate compound increases insulin sensitivity in the mammal.
- a mammal e.g., a mammal in need of increased insulin sensitivity
- the methods further include selecting the compound if is increases insulin sensitivity and evaluating the compound in a clinical trial.
- FIG. IA is a set of Western blots for P ⁇ kirl gene products with an antibody against the N-terminal SH2 domain (pan-p85) in tissues lysates, as indicated, from control, heterozygous KO and L-Pik3rlKO mice. Tissues were collected from mice after an overnight fast, and proteins were extracted and processed as described in the Methods section. Each lane represents lysates from a different mouse.
- FIGs. IB are line graphs of fasted blood glucose (IB) and fasted serum insulin levels (1C) at 8, 16, and 24 weeks of age in lox/lox and KO mice. Open circles (O) — lox/lox; closed circles (•)— L-Pik3rlKO.
- FIGs. ID and IE are bar graphs of serum triglycerides (ID) and serum non- esterified free fatty acid levels (IE) from lox/lox or KO mice in the fasted state.
- 2A-2E are bar graphs illustrating the results of hyperinsulinemic- euglycemic clamp analyses and gene expression changes in L-Pik3rlKO mice.
- Male mice (n l 1) of the indicated genotype at 10-12 weeks of age were subjected to hyperinsulinemic-euglycemic clamp analysis.
- 2E is a bar graph showing the results of quantitative RT-PCR analysis of mRNA levels in lox/lox and L-Pik3r IKO mice of phosphoenolpyruvate carboxykinase (Pckl), glucose-6-phosphatase (G6Pc), fructose- 1,6-bisphosphatase (Fbpl), peroxisome proliferator-activated receptor (PPAR)- ⁇ coactivator-1 alpha (Ppargcl) and tribbles3 (trib3) and glucokinase (Gckl).
- Pckl phosphoenolpyruvate carboxykinase
- G6Pc glucose-6-phosphatase
- Fbpl fructose- 1,6-bisphosphatase
- PPAR peroxisome proliferator-activated receptor
- trib3 tribbles3
- glucokinase Gckl
- 3A is a bar graph illustrating enhanced Akt activation in L-Pik3rlKO mice.
- FIGs. 3B is a Western blot of pi 10a and p85 ⁇ from pi 10a immunoprecipitates.
- FIG. 3 C is a par of blots of pTyr (upper panel) and insulin receptor (lower panel) in insulin receptor ( ⁇ -subunit) immunoprecipitates.
- FIG. 3D is a blot showing Ser473 phosphorylation of Akt.
- FIG. 4A is a set of six photomicrographs illustrating enhanced PIP3 levels in L-Pik3rlKO mice (bottom three panels) as compared to lox/lox mice (top three panels) due to decreased PTEN activity.
- Immunofluorescent staining with a primary anti-PIP 3 antibody (IgM) and an anti-mouse secondary antibody conjugated to Alexafluor Red and counterstained with DAPI. Following an overnight fast, mice were injected with saline (time 0) or 5U of insulin for 5 or 15 minutes.
- N six mice in each genotype/treatment group.
- FIG. 4B is a bar graph of the results of quantification of the immunofluorescence from PIP3 staining shown in FIG. 4A. Representative slides were chosen from each mouse and the fluorescence intensity was measured and analyzed with VH-H 1A5 Analyzer software (KEYENCE, Osaka, Japan).
- FIGs. 4C and 4D are bar graphs of insulin-stimulated pTyr-associated PI3K activity (4C) and PTEN activity (4D) in lox/lox or KO animals at the indicated timepoints.
- FIG. 4E is a Western blot showing PTEN levels in lox/lox and KO mice at the indicated timepoints.
- FIG. 5 A is a blot showing expression levels of hepatic p85 ⁇ and p50 in lox/lox and KO mice.
- Six-week-old L-Pik3rl KO mice and lox/lox controls were fed a normal chow (NC) or a high fat diet (HFD) for a total of 8 weeks.
- NC normal chow
- HFD high fat diet
- FIG. 5B is a line graph indicating body weight for each week on either diet. Open squares (D)- lox/lox, NC, Open circles (O)- L-Pik3r IKO, NC; Closed squares ( ⁇ )— lox/lox, HFD; closed circles (•)— L-Pik3rlK0, HFD.
- FIG. 5C is a trio of Western blots performed against liver lysates of mice of indicated genotype and diet using the phosphoserine 473 Akt antibody, phospho-JNK antibody, phosphoserine307 IRS-I antibodies.
- the phospho-specific antibody blots were stripped and re-probed with the antibody for total levels of the corresponding proteins, which in each case did not change and are therefore not shown.
- FIG. 5D is a line graph of fasting blood glucose from mice of the indicated genotype and diet. *p ⁇ 0.05, **p ⁇ 0.01
- FIG. 6 is a series of seven immunoblots of pJNK, JNK, pS307IRS-l, IRS-I, pAkt, Akt, and p85 in lox/lox and KO mice.
- FIG. 7 A is a bar graph illustrating cdc42 activity as determined by PAKl pulldown assay from liver lysates after three minutes of insulin stimulation via the portal vein.
- FIG. 7B is a quartet of Western blots against phosphor-MKK4 (pMM4),
- MKK4 phospho-JNK (pJNK) and JNK from liver lysates of mice of indicated genotypes.
- FIG. 7C is a series of five phosphoimmunoblots from primary hepatocytes against pJNK, pMKK4, pAkt, Myc tag, and p85.
- FIG. 8A is an immunoblot of LacZ, p85 ⁇ , p55 ⁇ , and p50 ⁇ . Recombinant adenoviruses were injected via tail-vein into 10-12 week old male mice of the indicated genotype. Mice were injected with adenoviruses encoding control LacZ, or one of the Pik3rl gene products, p85 ⁇ , p55 ⁇ , and p50 ⁇ . An extra band of approximately 5OkD appears in the livers treated with p55 ⁇ adenovirus, and this likely represents a proteolytic breakdown product of p55 ⁇ .
- FIG. 8B is a line graph of PBK activity from the mice injected with the indicated adenoviruses is 8A.
- FIG. 8C is a trio of Western blots performed against liver lysates of mice treated with adenovirus, using phospho-JNK and phosphoserine307 antibodies. The phospho-specific antibody blots were stripped and re-probed with the antibody for total levels of the corresponding proteins (data not shown).
- FIGs. 8D and 8E are bar graphs of fasted blood glucose (8D) and fasted serum o insulin (8E) in mice treated with the indicated adenoviruses.
- FIG. 9A is trio of immunoblots of LacZ, p85 ⁇ , p55 ⁇ , and p50 ⁇ .
- Recombinant5 adenoviruses were injected via tail-vein into 10-12 week old male mice of the indicated genotype. Mice were injected with adenoviruses encoding control LacZ, or one of the Pik3rl gene products, p85 ⁇ , p55 ⁇ , and p50 ⁇ .
- An extra band of approximately 5OkD appears in the livers treated with p55 ⁇ adenovirus, and this likely represents a proteolytic breakdown product of p55 ⁇ .
- FIGs. 9B and 9C are bar graphs of PDK activity (9B) and cdc42 activity (9C) in the mice injected with the indicated adenoviruses.
- FIG. 1OA is an immunoblot of LacZ, p85 ⁇ , ⁇ SH3, ⁇ BH, and ⁇ expression,5 showing that the Activation of cdc42 Requires an Intact N-terminus of p85 ⁇ .
- FIGs. 1OB and 1OC are bar graphs of PI3K activity (10B) and cdc42 activity
- FIG. 11 is a schematic illustration of a hypothetical assignment of functions to the regions of p85 ⁇ .
- This regulatory subunit of PI3K regulates insulin sensitivity through both positive and negative mechanisms.
- p85 ⁇ regulates PIP3 levels through its traditional role as a regulator of PDK activity, but it also independently regulates PIP 3 levels via the activation of JNK via cdc42 and possibly through the activation of a lipid phosphatase or by the alteration of subcellular localization of PDK.
- FIG. 12A is a gar graph illustrating the average weekly food intake of lox/lox (FLOX) or KO mice of either normal chow (NC) or high- fat diet HFD) over a week, expressed in grams. Genotypes of animals are indicated directly on the bars.
- FIGs. 12B and 12C are bar graphs illustrating fasting blood glucose (12B)and fasting serum insulin (12C) in lox/lox or KO mice after an eight-week treatment with either normal chow or high-fat diet.
- FIGs. 13A-B are schematic illustrations of natural variants (13A) or artificial mutants (13B) of p85 ⁇ , with their effect on PDK activity, JNK activity, or insulin sensitivity.
- FIG. 14 is a schematic illustration of the structure of the PTEN (top) and a phosphoimmunoblot of PTEN showing phosphorylation by JNK (bottom).
- the present invention describes methods of identifying novel modulators of the PI3-kinase/p85 ⁇ signalling pathway that eventually modulate JNK and PTEN activity, thereby regulating insulin action and sensitivity.
- PI 3-kinase regulatory subunits as modulators of the stress kinases JNK and p38
- the enzyme phosphatidylinositol 3-kinase is central to the metabolic actions of insulin.
- the enzyme itself is comprised of a regulatory subunit and a catalytic subunit.
- the catalytic subunit is either pi 10a (GenBank Ace. No. NM_006218.2) or pi lO ⁇ (GenBank Ace. No. BCl 14432.1).
- the most common forms of regulatory subunit are p85 ⁇ (GenBank Ace. No. NM_181523.1) and p85 ⁇ (GenBank Ace. No. BC090249.1 or BC070082.1), which are products of separate genes.
- GRBl the gene encoding p85 ⁇
- GRBl also produces several alternatively spliced variants, p55 ⁇ , p50 ⁇ and forms with small additional inserted exons.
- These different regulatory subunits are expressed to different levels in different tissues and also differentially regulated in disease states such as obesity.
- US Pat. App. Pub. No. 2002-0051786-A1 the present inventors demonstrated that heterozygous deletion of p85 ⁇ improves insulin sensitivity and can protect mice with genetic and acquired forms of insulin resistance, including the insulin resistance associated with high fat diet, from developing diabetes. Furthermore, this effect can be mimicked by reducing expression of p85 ⁇ in liver only via tissue specific knockout (see Barbour et al, J. Biol. Chem. 280(45):37489-94 (2005). Epub 2005 Sep 8).
- the work described herein demonstrates novel mechanistic insights into this connection between p85 ⁇ and improved insulin sensitivity.
- One aspect of the improved sensitivity is the result of a decrease in serine phosphorylation of the insulin receptor substrate IRS-I .
- Serine phosphorylation of IRS-I has been previously shown to reduce its tyrosine phosphorylation and reduce its ability to activate the PI 3-kinase pathway, thus reducing insulin's metabolic actions.
- Several protein kinases have been shown to phosphorylate IRS-I on serine residues, but a key kinase in this pathway is JNK.
- p85 ⁇ plays a novel role in JNK activation by binding to and activating the small GTPase cdc42, which leads to the activation of Mixed Lineage Kinase-3 (MLK3), which phosphorylates Mitogen-Activated Protein Kinase Kinase 4 (MKK4), leading to JNK activation.
- MKK3 Mixed Lineage Kinase-3
- MKK4 Mitogen-Activated Protein Kinase Kinase 4
- this pathway is normally increased in obesity-related insulin resistance, and reducing the level of p85 improves insulin sensitivity, as least in part, by reducing activation of cdc42, MMK4 and JNK, leading to reduced serine phosphorylation of IRS-I . Therefore, modulation of this pathway provides a number of novel targets for drugs that improve insulin sensitivity.
- the methods described herein can be used to identify and optimize small molecule inhibitors that reduce the interaction of cdc42 with p85 and/or reduce cdc42- mediated activation of MKK4, thereby reducing JNK activation and serine phosphorylation of IRS-I, and enhancing insulin sensitivity in insulin resistant states.
- the methods include the use of a high throughput in vitro assay system to identify test compounds, e.g., small molecules, that have this property and can therefore serve as drugs for diabetes, metabolic syndrome and related disorders.
- PI 3-kinase regulatory subunits as modulators of the lipid phosphatase PTEN
- PI 3-kinase is central to the metabolic actions of insulin. This occurs via formation of its phospholipid products, in particular PIP3, which activates downstream enzymes like Akt and the atypical PKCs ⁇ and ⁇ . This process is antagonized or reverse by the action of lipid phosphatases, which break down PIP3, the most important of which is the enzyme "phosphatase and tensin homolog," or PTEN.
- PTEN activity is multi-factorial and includes allosteric regulation of PTEN by its lipid products, subcellular targeting, cofactor interactions, as well as post-translational modifications (Gericke, Gene. 2006 Jun 7;374: l-9. Epub 2006 Mar 14).
- PTEN phosphorylation occurs on serine residues (>90%), especially serine residues 370 and 385.
- phosphorylation has been reported on other serines (e.g., serine 229, 360, 362, and/or 380) and threonines (e.g., threonine 223, 319, 321, 366, 382, 383, and/or 401), residues that map to the C2 lipid binding domain (amino acids (a. a.) 190-351) and a region proximal to the PDZ ligand sequence (a.a. 401-403) at the C-terminus.
- serines e.g., serine 229, 360, 362, and/or 380
- threonines e.g., threonine 223, 319, 321, 366, 382, 383, and/or 401
- residues that map to the C2 lipid binding domain amino acids (a. a.) 190-351)
- a region proximal to the PDZ ligand sequence a.a. 401-403
- JNK is the critical PTEN Kinase.
- K/RXXXXLXL conserved JNK-binding motif
- JNK/MAP kinase family two potential phosphorylation sites (Ser 338 and Thr 366 ) for JNK/MAP kinase family exist in the C-terminal region of PTEN, and this region has been shown to be important for stability and/or activity of PTEN. Since JNK activity is decreased o in p85 ⁇ KO mice and cells, a decrease in JNK would also be expected to result in a change in serine phosphorylation and activity of PTEN.
- sequences for the protein and nucleic acid targets useful in the methods described herein are known in the art. The following is a list of exemplary sequences5 that can be used. As one of skill in the art will appreciate, homologs of these sequences from other species can also be used. For example, nucleic acids that hybridize under stringent conditions to a sequence listed herein, or a polypeptide encoded by such a sequence, can equally be used. As used herein, the term “hybridizes under stringent conditions” describes conditions for hybridization and0 washing. Stringent conditions are known to those skilled in the art and can be found in Current Protocols in Molecular Biology, John Wiley & Sons, N.Y. (1989), 6.3.1- 6.3.6.
- Aqueous and nonaqueous methods are described in that reference and either can be used.
- stringency conditions are 0.5 M sodium phosphate, 7% SDS at 65°C, followed by one or more washes at 0.2 X SSC, 1% SDS at 65°C. 5
- a sequence that is at least 80%, e.g., at least 85%, 90%, 95%, 96%, 97%, 98%, 99%, or more identical to a sequence listed herein can be used.
- the comparison of sequences and determination of percent identity between two sequences can be accomplished using a mathematical algorithm. For purposes of the present methods, the percent identity between two amino acid sequences is 0 determined using the Needleman and Wunsch ((1970) J. MoI. Biol.
- mouse PTEN sequences are available at:
- JNKl also known as mitogen-activated protein kinase 8
- JNK2 also known as mitogen activated protein kinase 9
- MLK3 also known as Mitogen- Activated Protein Kinase Kinase Kinase 11 (MAPKKl 1) can be found at:
- MKK4 mitogen-activated protein kinase kinase 4
- PIK3R1 phosphoinositide-3-kinase, regulatory subunit 1 (PIK3R1)
- the invention includes methods for screening of test compounds, to identify compounds that modulate a pathway described herein, e.g., compounds that (i) reduce the interaction of cdc42 with p85, thereby reducing cdc42 activity, or (ii) reduce the activation of MLK3 by cdc42, thereby reducing MLK3 activity, or (iii) reduce the phosphorylation of MKK4 by MLK3, thereby reducing MKK4 activity, thus reducing JNK activation and serine phosphorylation of IRS-I, and enhancing insulin action and sensitivity.
- each of cdc42, MLK3, and MKK4 are novel targets for inhibition, and can be used as targets for a screening method described herein.
- the methods described herein can be used to identify compounds that bind to p85, cdc42, MLK3, or MKK4.
- the effect of a test compound on the p85 signalling pathway can be determined.
- the methods can be used to identify compounds that demonstrate (i) binding to p85 and/or cdc42, and/or decrease p85-mediated activation of cdc42, (ii) binding to cdc42 and/or MLK3, and/or decrease csc42-mediated activation of MLK3, or (iii) binding to MLK3 and/or MKK4, and/or decrease MLK3 -mediated phosphorylation of MKK4.
- the methods will include providing a sample that includes one or more of p85, cdc42, MLK3, and MKK4.
- the sample can include p85 ⁇ and cdc42, e.g., isolated and purified p85 ⁇ and cdc42 proteins, and the methods include identifying a compound that affects binding between p85 and cdc42.
- Screening methods suitable for use in these embodiments are known in the art and include, but are not limited to, yeast or mammalian 2-hybrid systems, tagged protein assays, immunoprecipitation assays, and proteomics assays. These methods can be used to identify natural (i.e., endogenous) regulators of cdc42, for example.
- the sample includes isolated and purified cdc42 and MLK3, with or without MKK4, and the methods include detecting identifying compounds that affect activation of MLK3 by cdc42, and/or phosphorylation of MKK4. In some embodiments, the sample includes isolated and purified MLK3 and
- MKK4 and the methods include detecting identifying compounds that affect phosphorylation of MKK4.
- MKK7 can be used in place of MKK4.
- the methods include identifying isoform-specific inhibitors of JNK, i.e., inhibitors that substantially reduce the activity of JNKl while not substantially affecting the activity of JNK2, or vice-versa.
- the different isoforms have different activities depending on the tissues, therefore, it may be desirable to affect only the isoform that is active in the particular cell type.
- the methods can include adding the compound to cells that express all of these proteins, and exhibit p85 signalling via cdc42 and MKK4 that results in JNK activation and serine phosphorylation of IRS- 1 and/or PTEN.
- the methods can then include contacting the cells with the compound, and evaluating an effect of the compound on JNK activity (e.g., serine phosphorylation of IRS-I or PTEN).
- the methods further include determining whether the compound has an effect on phosphorylation of other substrates by JNK, and selecting a test compound if it selectively inhibits JNK phosphorylation of PTEN.
- such a compounds might be designed such that it binds a JNK-recognition site, or a JNK-phosphorylation site, on PTEN.
- a number of suitable assays are known in the art, see, e.g., Methods in Enzymology. Volume 201 : Protein Phosphorylation. Part B: Analysis of Protein Phosphorylation. Protein Kinase Inhibitors, and Protein (Methods in Enzymology) by John N. Abelson, Melvin I. Simon, Tony Hunter, and Bartholomew M. Sefton (Hardcover - Jan 15, 1991), Protein Phosphorylation. Part A: Protein Kinases: Assays. Purification.
- volume 200 Volume 200, Part A (Methods in Enzymology) by John N. Abelson, Melvin I. Simon, Tony Hunter, and Bartholomew M. Sefton (Hardcover - JuI 28, 1991).
- test compounds can be, e.g., natural products or members of a combinatorial chemistry library.
- the test compounds are initially members of a library, e.g., an inorganic or organic chemical library, peptide library, oligonucleotide library, or mixed-molecule library.
- the methods include screening small molecules, e.g., natural products or members of a combinatorial chemistry library. These methods can also be used, for example, to screen a library of proteins or fragments thereof, e.g., proteins that are expressed in liver or pancreatic cells.
- a given library can comprise a set of structurally related or unrelated test compounds.
- a set of diverse molecules should be used to cover a variety of functions such as charge, aromaticity, hydrogen bonding, flexibility, size, length of side chain, hydrophobicity, and rigidity.
- Combinatorial techniques suitable for creating libraries are known in the art, e.g., methods for synthesizing libraries of small molecules, e.g., as exemplified by Obrecht and Villalgordo, Solid-Supported Combinatorial and Parallel Synthesis of Small-Molecular- Weight Compound Libraries. Pergamon-Elsevier Science Limited (1998). Such methods include the "split and pool” or “parallel” synthesis techniques, solid-phase and solution-phase techniques, and encoding techniques (see, for example, Czarnik, Curr. Opin. Chem. Bio. 1 :60-6 (1997)). In addition, a number of libraries, including small molecule libraries, are commercially available.
- the test compounds are peptide or peptidomimetic molecules, e.g., peptide analogs including peptides comprising non-naturally occurring amino acids or having non-peptide linkages; peptidomimetics (e.g., peptoid oligomers, e.g., peptoid amide or ester analogues, ⁇ -peptides, D-peptides, L-peptides, oligourea or oligocarbamate); small peptides (e.g., pentapeptides, hexapeptides, heptapeptides, octapeptides, nonapeptides, decapeptides, or larger, e.g., 20-mers or more); cyclic peptides; other non-natural or unnatural peptide-like structures; and inorganic molecules (e.g., heterocyclic ring molecules).
- the test compounds are nucleic acids,
- test compounds and libraries thereof can be obtained by systematically altering the structure of a first test compound.
- a small molecule e.g., a first small molecule is selected that is, e.g., structurally similar to a known phosphorylation or protein recognition site.
- a general library of small molecules is screened, e.g., using the methods described herein, to select a fist test small molecule.
- the structure of that small molecule is identified if necessary and correlated to a resulting biological activity, e.g., by a structure-activity relationship study.
- test compounds identified as "hits" e.g., test compounds that demonstrate activity in a method described herein
- a first screen is selected and optimized by being systematically altered, e.g., using rational design, to optimize binding affinity, avidity, specificity, or other parameter.
- Such potentially optimized structures can also be screened using the methods described herein.
- the invention includes screening a first library of test compounds using a method described herein, identifying one or more hits in that library, subjecting those hits to systematic structural alteration to create one or more second generation compounds structurally related to the hit, and screening the second generation compound. Additional rounds of optimization can be used to identify a test compound with a desirable therapeutic profile.
- Test compounds identified as hits can be considered candidate therapeutic compounds, useful in treating disorders described herein.
- the invention also includes compounds identified as "hits" by a method described herein, and methods for their administration and use in the treatment, prevention, or delay of development or progression of a disease described herein.
- Example 1 Phosphoinositide 3-Kinase Regulatory Subunit p85 ⁇ Suppresses Insulin Action via Positive Regulation of PTEN
- Insulin action on the liver is required for the proper maintenance of metabolic homeostasis. Under normal conditions, insulin inhibits gluconeogenesis and activates lipogenesis to promote proper fuel utilization in the fed state. The failure of insulin to regulate hepatic function can lead to unfettered hepatic glucose output and elevated lipogenesis in the liver, which are two key features of the metabolic syndrome (Shimomura, et al., (2000) MoI. Cell, 6(l):77-86). Not surprisingly, an overwhelming amount of epidemiological (Tripathy et al., (2004) Diabetologia, 47(5):782-93) and physiological (Fisher and Kahn, (2003) J. Clin. Invest., 111(4):463-468) evidence links hepatic insulin resistance to the development of type 2 diabetes.
- PI3K phosphoinositide 3-kinase pathway
- Saltiel and Kahn, (2001) Nature, 414(6865):799-806 PI3K is an obligate heterodimer, with an SH2-containing regulatory subunit (p85) and a catalytic subunit (pi 10).
- the regulatory subunit mediates the binding, activation and localization of the PI3K enzyme (Virkamaki, et al., (1999) J. Clin. Invest.
- the negative effects of p85 ⁇ on insulin action may have important consequences in the pathophysiology of insulin resistance and diabetes.
- the increased expression of p85 ⁇ in mouse models of gestational diabetes (Barbour et al., (2004) Endocrinology 145(3): 1144-50) and in obese humans (Bandyopadhyay et al., (2005) Diabetes 54(8):2351-9) is strongly linked with insulin resistance.
- heterozygosity for P ⁇ kirl prevents the onset of diabetes in genetically insulin resistant mice (Mauvais-Jarvis et al, (2002) J. Clin. Invest. 109, 141-149).
- mice with a liver-specific deletion oiPik3rl were created.
- This conditional knockout oiPik3rl (p85 ⁇ , p55 ⁇ , and p50 ⁇ ) in hepatocytes circumvents the perinatal lethality observed in the corresponding germline knockout mice and furthermore allows the investigation of the specific role of the liver in the physiological actions of p85 ⁇ .
- L-Pik3rlKO mice have enhanced hepatic and whole body insulin sensitivity as well as increased Akt activity in liver, despite decreased total hepatic PBK activity.
- p85 ⁇ mediates its negative effect on insulin sensitivity at least in part via the activation of PTEN lipid phosphatase activity.
- mice used in this study were on a 129Sv-C57BL/6-FVB mixed genetic background. Metabolic studies.
- GTT glucose tolerance testing
- blood samples were obtained at 0, 15, 30, 60, and 120 minutes after intraperitoneal injection of 2 g/kg dextrose. Insulin tolerance tests were performed by injecting 1 U/kg insulin (Novolin, Novo Nordisk, Denmark) intraperitoneally, followed by blood collection at 0, 15, 30 and 60 minutes after injection. Blood glucose values were determined using a One Touch II glucose monitor (Lifescan Inc., Milipitas, CA).
- Plasma insulin levels were measured by ELISA using mouse insulin as a standard (Crystal Chem Inc., Chicago, IL). Non-esterified free fatty acid levels were measured from random fed mice using a kit from Wako Diagnostics, while serum triglycerides were measured by Anilytics (Gaithersburg, MD).
- Hyperinsulinemic-euglycemic clamp Mice were anaesthetized with a 1.2% solution of 2,2,2-tribromoethanol in normal saline, followed by the microsurgical insertion of a catheter into the right jugular vein. Approximately 7 days of recovery, mice were fasted for 5 hours and were infused with a constant (2.5 mU/kg/min) dose of insulin and a variable glucose infusion rate to maintain euglycemia and assess whole-body insulin sensitivity.
- mice were anesthetized with Avertin (2,2,2-tribromoethanol in PBS), and injected with 5 U of regular human insulin (Novolin, Novo Nordisk, Denmark) via the inferior vena cava. Five minutes after the insulin bolus, tissues were removed and frozen in liquid nitrogen. Immunoprecipitation and immunoblot analysis of insulin signaling molecules was performed as previously described (Taniguchi et al, (2005) J. Clin. Invest. 115(3):718-27).
- TATA box binding protein TBP
- Antibodies Rabbit polyclonal anti-IRS-1 antibody (IRS-I), anti-IRS-2 antibody (IRS-2), anti-IR antibody (IR) and pan-p85 ⁇ antibody were generated as described previously (Ueki et al., (2002) Proc. Natl. Acad. Sci. U.S.A., 99(l):419-24).
- the anti-TRB3 antibody was a gift from Marc Montminy (Du et al., (2003) Science 300, 1574-7).
- Rabbit polyclonal anti-Akt, anti-phospho Akt (S473) anti-PTEN were purchased from Cell Signaling Technology (Beverly, MA).
- the phosphotyrosine (pTyr) antibody, 4G10 was purchased from Upstate Biotechnology, Inc. (Lake Placid, NY).
- Goat polyclonal anti-Aktl/2 antibody (Akt) was purchased from Santa Cruz Biotechnology, Inc. (Santa Cruz, CA).
- Phosphatidylinositol (Avanti Polar Lipids) was in vitro phosphorylated to form phosphatidylinositol 3 -phosphate (PI-3P) by recombinant PI3K (Upstate) with [ 32 P] ⁇ -ATP.
- the PI-3P was re- extracted with methanol/choloroform and run on a TLC plate with a n-propanaol:2M acetic acid (65:35). The intensity of PI-3P spots were quantitated with NIH Image. PTEN activity was determined relative to IgG immunoprecipiates from lox/lox livers. Relative PTEN activity was compared with lox/lox without insulin stimulation. Statistics. Data are presented as ⁇ s.e.m. Student's t-test was used for statistical analysis between two groups, while statistical significance between multiple treatment groups was determined by analysis of variance (ANOVA) and Tukey's t- test.
- ANOVA analysis of variance
- mice with a liver-specific deletion oiPik3rl were generated via the Cre-loxP system using animals carrying a floxed exon 7, which encodes the N-terminal SH2 domain (Luo, et al., (2005) MoI. Cell. Biol. 25:491-502) and mice carrying the Cre transgene driven by the albumin promoter (Postic and Magnuson, (2000) Genesis 26(2): 149-50).
- the presence of loxP sites in the Pik3rl gene did not affect expression of p85 ⁇ as compared to WT littermate controls (Luo et al., (2005) MoI. Cell. Biol. 25:491-502) nor did the presence of albumin-Cre (Michael et al., (2000) MoI Cell
- L-Pik3rl KO mice were born in a normal Mendelian distribution and exhibited normal postnatal growth (data not shown).
- Western blots of liver extracts of L-Pik3rl KO mice revealed an 80-90% decrease in p85 ⁇ and a complete loss of p50 ⁇ (FIG. IA), consistent with complete ablation in hepatocytes.
- the expression of p85 ⁇ was unaltered in brain, skeletal muscle and fat.
- L-Pik3rl KO mice exhibited significant improvements in serum metabolic chemistries.
- L-Pik3rl KO mice displayed lower fasted blood glucose and fasted serum insulin levels at 8, 16, and 24 weeks of age (FIG. IB and 1C). In addition, L-Pik3rlKO mice exhibited decreased levels of serum triglycerides and circulating free fatty acids (FIGs. ID and IE). At 16 weeks of age, L-Pik3rl KO mice were significantly more glucose tolerant to an intraperitoneal glucose challenge (FIG. IF), but this was observed as early as eight weeks of age and was maintained through 24 weeks of age (data not shown).
- FIG. IF intraperitoneal glucose challenge
- L-Pik3rl KO mice were subjected to a hyperinsulinemic- euglycemic clamp. This revealed increased hepatic insulin sensitivity such that hepatic glucose production was suppressed by 25% more in the L-Pik3rl KO mice compared to controls (FIG. 2A).
- L-Pik3rl KO mice displayed improved peripheral insulin sensitivity, as determined by a two-fold increase in glucose infusion rates (FIG. 2B). This correlated with a 1.5-fold increase in glucose uptake in muscle (FIG. 2C) and a three-fold increase in glucose uptake in fat (FIG. 2D).
- FIG. 2B shows improved peripheral insulin sensitivity
- the physiologic data indicating increased hepatic insulin sensitivity corresponded with decreased expression of several key gluconeogenic genes.
- Quantitative RT-PCR analysis found a decrease in phosphoenolpyruvate carboxykinase (Pckl), glucose-6 phosphatase (G6pc) and fructose- 1,6-bisphosphatase (Fbpl) mRNAs in liver by 30%, 70%, and 60%, respectively (FIG. 2e).
- PI3K function was characterized in L-Pik3rlKO mice.
- a 50% decrease in IRS-I -associated PBK activity was observed, as was a 60% decrease in IRS-2- associated PDK activity, which together resulted in a 50% reduction in total PBK activity, as measured from pTyr-immunoprecipitates (FIG. 3A).
- These decreases in PBK were concordant with a 70% decrease in the expression of the pi 10a catalytic subunit (FIG. 3B), but were not associated with decreased insulin receptor activation in the L-Pik3rl KO mice (FIG. 3C).
- the parallel decrease in expression of pi 10a was expected, since the regulatory subunits are known to stabilize the catalytic subunit (Yu et al., (1998) MoI. Cell. Biol. 18(3): 1379-87).
- PBK activity or decreased turnover, total PBK activity and the activity of the lipid phosphatase, PTEN, which degrades the PIP3 formed by PBK were directly assessed.
- pTyr-associated PBK activity was not changed in the livers of either the lox/lox or L-Pik3rlKO mice, though the insulin-stimulated PI3K activity of the knockout mice remained at only half the level of the controls (FIG. 4C).
- the lipid phosphatase activity of the negative regulator PTEN was consistently decreased by 40% in L-Pik3rlKO at all timepoints (FIG. 4D).
- insulin had no effect on PTEN activation in either the control or knockout mice. This decrease in PTEN activity is not due to decreased PTEN expression (FIG. 4E), but must be due to some other aspect of the actions of the regulatory subunit of PI3K.
- Example 2 The p85 ⁇ Regulatory Subunit of Phosphoinositide 3 -Kinase activates JNK via a cd c42/MKK4 pathway
- Insulin resistance is an underlying feature of type 2 diabetes and the metabolic syndrome (Reaven, (2005) Cell. Metab. 1 :9-14). Physiologic and epidemiologic studies have demonstrated strong links between obesity and the development of insulin resistance (Hu et al., (2001) N. Engl. J. Med. 345:790-7; Sinha et al., (2002) N. Engl. J. Med. 346:802-10). Not surprisingly, the rise of type 2 diabetes in the United States over the last decade has paralleled the rapid rise in obesity (Mokdad et al., (2003) JAMA 289:76-9).
- JNK stress kinase
- c-Jun-N-terminal kinase JNK
- cytokines Baud et al., (1999) Genes Dev. 13: 1297-308
- JNK c-Jun-N-terminal kinase
- JNK phosphoinositide 3-kinase
- PI3K is an obligate heterodimer, with an SH2 -containing regulatory subunit (p85) and a catalytic subunit (pi 10).
- the regulatory subunit mediates the binding, activation and localization of the PBK enzyme (Backer et al., (1992) EMBO J.
- mice carrying germline deletion of the P ⁇ kirl gene that encodes p85 ⁇ and its shorter isoforms p55 ⁇ and p50 ⁇ die perinatally (Fruman et al., (2000) Nat Genet 26:379-82).
- mice with a liver-specific deletion oiPik3rl gene (L-Pik3rlKO) were created.
- mice lacking p85 ⁇ in liver have diminished hepatic activation of JNK and improved whole body insulin sensitivity.
- p85 ⁇ activates JNK via the cdc42-MKK4 pathway and that this interaction requires both an intact N-terminus and functional SH2 domains in the C-terminus of the p85a regulatory subunit.
- p85 ⁇ may regulate insulin sensitivity in both lean and obese mice via crosstalk with the stress kinase pathway.
- mice and high fat diet Animals and high fat diet. All animals were housed on a 12-h light-dark cycle and fed a standard rodent chow (Purina). High fat chow (45% kcal from fat) was purchased from Research Diets. All mice in this study were on a 129Sv- C57BL/6-FVB mixed genetic background, and littermates of the same mixed genetic background were used as controls.
- Metabolic studies were performed as described above in Example 1.
- the p50 ⁇ and p55 ⁇ adenoviruses were constructed as previously described (Ueki et al., (2000) MoI. Cell. Biol. 20:8035-46).
- the constitutively active MKK4 adenovirus was purchased from CellBio Labs (San Diego, CA), and the constitutively active cdc42 adenovirus was a gift from James Bamburg (Kuhn et al., (2000) J. Neurobiol. 44: 126-144).
- the wild type (WT) p85 ⁇ , ⁇ SH3, ⁇ BH, ⁇ iSH2, RARA and ⁇ p85 constructs were made as follows. Cloning ofp85 constructs
- the p85 constructs were cloned from a cDNA of human p85 ⁇ using the following PCR primers:
- the PCR fragments from the WT, and ⁇ SH3 reactions were digested with Xbal/Sall then ligated into pBluescript (pBS).
- the 5' portion of the ⁇ BH reaction was digested with Xbal/BamHI and the 3' fragment was digested with BamHI/Sall.
- the two fragments were ligated together in pBS.
- the ⁇ iSH2 construct was made by cutting the above 1.5 kb PCR fragment with EcoRV and Sail, then fusing it with the Xbal-EcoRV fragment of the WTp85 ⁇ .
- the ⁇ p85 construct was made by ligating the Xbal-EcoRV fragment of WTp85 ⁇ FLAG construct (N-terminus) to the EcoRV- SaII ⁇ iSH2 fragment (C-terminal half).
- adenoviruses were then produced according to the standard AdEasy protocol (He et al, (1998) Proc. Natl. Acad. Sci. U.S.A. 95:2509-14).
- Adenovirus-mediated gene transfer and in vivo insulin stimulation Prior to use on primary hepatocytes or in vivo, all adenoviruses were purified on sequential cesium chloride gradients then dialyzed into PBS containing 10% glycerol. 10-12 week-old male mice were injected via tail vein with an adenoviral dose of 5xl ⁇ 8 pfu/g body weight as described previously (Taniguchi et al., (2005) J. Clin. Invest. 1 15:718- 27).
- mice On the fifth day after injection, following an overnight fast, the mice were anesthetized with Avertin (1.2% 2,2,2-tribromoethanol in PBS), and injected with 5 U of regular human insulin (Novolin, Novo Nordisk, Denmark) via the inferior vena cava. Five minutes after the insulin bolus, tissues were removed and frozen in liquid nitrogen.
- Avertin (1.2% 2,2,2-tribromoethanol in PBS
- regular human insulin Novolin, Novo Nordisk, Denmark
- tissue homogenates prepared in a tissue homogenization buffer that contained 25 mM Tris-HCl (pH 7.4), 10 mM Na 3 VO 4 , 100 mM NaF, 50 mM Na 4 P 2 O 7 , 10 mM EGTA, 10 mM EDTA, 2 mM phenylmethylsulfonyl fluoride, 1% Nonidet-P40 supplemented with the Complete protease inhibitor cocktail (Roche). All protein expression data were quantified by densitometry using NIH Image software. Antibodies.
- Rabbit polyclonal anti-IRS-1 antibody (IRS-I), anti-IRS-2 antibody (IRS-2), anti-IR antibody (IR) and pan-p85 ⁇ antibody were generated as described previously (Ueki et al., (2002) Proc. Natl. Acad. Sci. U.S.A. 99:419-24).
- mice were anesthetized and injected with 5U of insulin via the portal vein. Three minutes after the injection, the right lobe of the liver was quickly dissected and snap frozen directly into liquid nitrogen.
- cdc42 activity in the livers was measured with a PAKl pulldown assay (Upstate).
- the kit was used essentially as directed, but with the addition of 10 mM ortho vanadate to the reaction buffer.
- Hepatocytes were isolated as described previously (Block et al, (1996). J. Cell. Biol. 132: 1133-49). Briefly, the mice were anesthetized with a 1.2% solution of 2,2,2-tribromoethanol and the portal vein was cannulated with a 24.5 G catheter, and the liver perfused for 15 minutes at a rate of 7 mL/min with calcium-free perfusion buffer. The blanched liver is perfused with collagenase solution (200U/mL) for 10 minutes at 7 mL/min to release hepatocytes from the extracellular matrix.
- collagenase solution 200U/mL
- the digested liver was excised and placed in preservation buffer, where the digested cells were gently scraped from the liver sac, washed and purified with Percoll to remove dead cells and enrich the hepatocyte fraction.
- Typical viabilities are between 85-90%, with cell yields of 1.0-1.5 x 10 6 cells/g mouse.
- the isolated hepatocytes are then grown on collagen-coated plates in Advanced DMEM (Gibco) supplemented with glutamine, antibiotic cocktail and 10% FBS.
- mice with a liver-specific deletion oiPik3rl via the Cre-loxP system were generated as described previously (Postic and Magnuson. (2000) Genesis 26: 149-50).
- Mice carrying a floxed exon 7, which encodes the N-terminal SH2 domain common to all three transcripts, p85 ⁇ , p55 ⁇ and p50 ⁇ were crossed with mice carrying the Cre transgene driven by the albumin promoter.
- L-Pik3rlKO mice maintained lower fasting blood glucose and fasting serum insulin levels when fed either a high- fat diet or normal chow (FIGs. 12B and 12C).
- obese lox/lox mice were severely glucose intolerant
- obese L-Pik3rlKO mice exhibited normal to improved glucose tolerance even when compared against control mice on normal chow (FIG. 5C).
- FIG. 5C the loss of p85 ⁇ expression in liver protected against obesity-induced insulin resistance and diabetes.
- L-Pik3rlKO Hepatocytes are Resistant to JNK-induced insulin resistance.
- JNKl was overexpressed in primary hepatocytes isolated from lox/lox or L- Pik3rlKO mice using adenovirus-mediate gene transfer.
- the JNKl isoform was chosen because it is the only one of the three JNK isoforms that has been shown to have a significant role in mediating obesity -related insulin resistance in the liver
- JNKl was overexpressed by six-fold in both p85 ⁇ knockout and control primary hepatocytes (FIG. 6). This forced expression of JNKl led to significant increases in JNK phosphorylation and serine phosphorylation of IRS-I in lox/lox hepatocytes. By contrast, an equal level of JNKl overexpression resulted in only a 2-fold increase in cells derived from L-Pik3rl mice (FIG. 6) (p ⁇ 0.05 knockout vs. control cells).
- the p85 ⁇ regulatory subunit activates JNK via a cdc42/MKK4 pathway
- One candidate effector is the small GTPase cdc42, which is known to activate both SEK1/MKK4 and JNK (Gallo and Johnson, (2002) Nat. Rev. MoI. Cell. Biol. 3:663-72), and has been shown to interact with p85 ⁇ , but it was unknown whether this interaction had any functional consequences in vivo (Zheng et al, (1994) J. Biol. Chem.
- cdc42/MKK4/JNK signaling pathways is an intrinsic property of hepatic insulin signaling
- primary hepatocytes from L-Pik3rlKO livers and lox/lox controls were infected with constitutively active forms of MKK4 and cdc42 and stimulated with either insulin or saline control (FIG. 7C).
- Expression of activated MKK4 or cdc42 enhanced JNK phosphorylation, indicating that these enzymes were upstream of this pathway in hepatocytes.
- Akt activation was differentially affected by the expression of p85 ⁇ .
- control LacZ, p55 ⁇ , or p50 ⁇ maintained the elevated Akt activation observed in L-Pik3rlKO mice
- the expression of p85 ⁇ caused a relative decrease in Akt phosphorylation to a level similar to lox/lox controls.
- re-expression of p85 ⁇ specifically restored several mechanisms of negative regulation to L-Pik3rlKO animals.
- the expression of p85 ⁇ restored insulin- stimulated JNK activation and levels of IRS-I serine phosphorylation back to levels comparable to lox/lox controls (FIG. 8C).
- the ⁇ iSH2 construct When overexpressed in cells or in mouse livers, the ⁇ iSH2 construct has a dominant negative effect (Miyake et al., (2002) J. Clin. Invest. 110: 1483-91).
- the other p85 mutant contains arginine to alanine substitutions in critical residues in both SH2 domains in the C-terminus (RARA); this mutant is able to bind pi 10, but cannot bind to phosphorylated IRS proteins, which is required for the proper activation and localization of the PI3K holoenzyme (Hill et al., (2001) J. Biol. Chem. 276: 16374-8).
- the ⁇ iSH2 mutant caused significant glucose intolerance consistent with diabetes, probably due to the inhibition of the positive effects of PBK in addition to the negative effects of JNK activation. Consistent with the cdc42/JNK data, the RARA p85a mutant had negligible effects on insulin sensitivity.
- the N-terminus of p85 ⁇ is 339 amino acids long and contains an SH3 domain, two proline rich regions, and a domain homologous to a portion of breakpoint cluster region (bcr) gene product (BH domain).
- adenoviral p85 constructs were created which substitute a FLAG tag for either the -80 amino acid SH3 domain ( ⁇ SH3) or the -170 amindacid BH domain ( ⁇ BH), which effectively deleted the domain while providing an epitope tag for easy detection by Western.
- ⁇ SH3 domain -80 amino acid SH3 domain
- ⁇ BH -170 amindacid BH domain
- One construct with a combination deletion of both the SH3 domain and inter-SH2 domain ( ⁇ p85) was also created to serve as a control for PI3K activity (FIG. 10A).
- JNK The activation of JNK, particularly in the liver, has been shown previously to be a major mediator of the insulin resistance that occurs in obesity (Ozcan et al., (2004) Science 306:457-61). Consequently, one of the mechanisms by which p85 ⁇ suppresses insulin action in vivo may occur through the JNK-mediated negative feedback on insulin signaling.
- deletion of either the SH3 domain or BH domain from the N-terminus is sufficient to ablate the ability of p85 to activate cdc42, while fully maintaining PI3K activity (FIG. 1OB and Beeton et al, (1999) MoI. Cell Biol. Res. Commun. 1 : 153-7).
- the functional inactivation of the SH2 domains in p85 also rendered it unable to activate either PI3K or cdc42/JNK, suggesting that the proper localization in the cell or within a complex may be required for JNK activation.
- the data define the minimal requirements for the activation of JNK by p85 as a fully intact N- terminus of p85 and functional SH2 domains.
- p85 has been demonstrated as an essential activator of small GTPases such as cdc42 or Rac (reviewed in Burridge and Wennerberg, (2004) Cell 116: 167-79) that mediate PDGF or EGF-induced cytoskeletal changes, such as membrane ruffling or stress fiber disassembly (Brachmann et al., (2005) MoI. Cell. Biol. 25:2593-606; Hill et al., (2001) J. Biol. Chem. 276: 16374-8).
- small GTPases such as cdc42 or Rac (reviewed in Burridge and Wennerberg, (2004) Cell 116: 167-79) that mediate PDGF or EGF-induced cytoskeletal changes, such as membrane ruffling or stress fiber disassembly (Brachmann et al., (2005) MoI. Cell. Biol. 25:2593-606; Hill et al., (2001) J. Biol. Chem. 276: 16374
- the BH domain is similar in structure to the Rho-GTPase activating protein (GAP) domain of the breakpoint cluster region (bcr) protein (Musacchio et al., (1996) Proc. Natl. Acad. Sci. U.S.A.
- these domains might be responsible for the proper intracellular localization of the cdc42/JNK-activating complex.
- the p85 subunit has been found to form insulin-dependent protein aggregates that do not generate PIP 3 (Luo et al., (2005) J. Cell. Biol. 170(3):455-64). These aggregates were proposed as sequestration complexes, but an alternate interpretation is that these complexes could be active negative regulatory complexes that activate cdc42 and JNK.
- p85 is an essential part of the PDK heterodimer, it also plays a novel role in regulating a cdc42/MKK4/JNK pathway that suppresses insulin action in both lean and obese mice.
- These mechanisms not only provide a level of internal negative feedback on this critical node (Taniguchi et al, (2006) Nat. Rev. MoI. Cell. Biol. 7:85-96) in insulin and growth factor signaling, but also allow crosstalk between the PBK signaling pathway and the stress or inflammatory responses, thus creating an important connection that could have broad impact in the basic understanding of cell growth and metabolism.
- This powerful link between p85 ⁇ and JNK activation might also represent an exciting new therapeutic intervention into type 2 diabetes.
- Example 3 JNK phosphorylates PTEN Since JNK activity is decreased in p85 ⁇ KO mice and cells, it is possible that a decrease in JNK could also result in a change in serine phosphorylation and activity of PTEN.
- PTEN Activity Assay Phosphatidylinositol (Avanti Polar Lipids) was in vitro- phosphorylated to form phosphatidylinositol 3 -phosphate (PI-3-P) by recombinant PI3K (Upstate) with [gamma-32P]ATP.
- the phosphorylated lipid was extracted with 1: 1 methanolxhloroform, dried under nitrogen gas, reconstituted into PTEN assay buffer (10mMTris-HCl/25mMNaCl, pH 7.5), and incubated with PTEN immunoprecipitates from lox/lox or L-Pik3rlKO liver lysates (Miller et al., FEBS Lett. 528:145-153 (2005)).
- the PI-3-P was re-extracted with methanokchloroform and run on a TLC plate with n-propanol:2 M acetic acid (65:35).
- PTEN activity was determined by decreased intensity of the PI-3-P spot relative to IgG immunoprecipitates from lox/lox livers. Relative PTEN activity was determined by normalizing PTEN activity to lox/lox livers without insulin stimulation. SDS-PAGE and autoradiography revealed that JNK was able to phosphorylate PTEN in vitro (FIG. 14). To further assess this phosphorylation of PTEN by JNK, mutants of PTEN were generated with substitution of the two potential JNK phosphorylation sites (PTEN-S338A, and PTEN-T366A).
- mutants as well as wild-type PTEN, were then subcloned into pCMV-Tag2 vector that introduced an N- terminal FLAG tag. 5 ⁇ g of each construct was transfected into COS7 cells and the activities of Akt and p70S6 kinase following IGF-I stimulation was evaluated. In cells expressing these mutants, two downstream targets of the PI 3-kinase pathway (Akt and p70S6 kinase) were maintained at higher levels of activity than observed in wild-type control cells.
- Akt and p70S6 kinase two downstream targets of the PI 3-kinase pathway
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Engineering & Computer Science (AREA)
- Biomedical Technology (AREA)
- Immunology (AREA)
- Molecular Biology (AREA)
- Chemical & Material Sciences (AREA)
- Urology & Nephrology (AREA)
- Hematology (AREA)
- Cell Biology (AREA)
- General Health & Medical Sciences (AREA)
- Biotechnology (AREA)
- Pathology (AREA)
- Food Science & Technology (AREA)
- Medicinal Chemistry (AREA)
- Physics & Mathematics (AREA)
- Analytical Chemistry (AREA)
- Biochemistry (AREA)
- Microbiology (AREA)
- General Physics & Mathematics (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Toxicology (AREA)
- Tropical Medicine & Parasitology (AREA)
- Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
- Investigating Or Analysing Biological Materials (AREA)
- Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
Abstract
This invention relates to methods of identifying drugs for the treatment of insulin resistance and diabetes.
Description
Methods of Identifying Modulators of Insulin Signalling
FEDERALLY SPONSORED RESEARCH OR DEVELOPMENT
This invention was made with Government support under Grant Nos. DK33201, and DK55545 awarded by the National Institutes of Health. The Government has certain rights in the invention.
TECHNICAL FIELD
This invention relates to methods of identifying drugs for the treatment of insulin resistance and diabetes.
BACKGROUND
Twenty-one million people in the U.S. have diabetes mellitus, and -95% of these have the Type 2 form of the disease. In addition, over 25 million people have impaired fasting glucose, i.e., prediabetes, and many more have the metabolic syndrome, hepatic steatosis and/or obesity. Furthermore, these disorders are increasing at epidemic rates in both adults and children, and each is a source of major morbidity and mortality. Type 2 diabetes is the leading cause of kidney failure, blindness, and amputations, and is a major risk factor for heart disease and stroke.
Hepatic steatosis is the second most common cause of liver failure in the U.S., and the metabolic syndrome is a major risk factor in as many as 60% of individuals suffering heart attack or stroke. As a result, over 12% of healthcare dollars and 25% of Medicare dollars are spent on people with type 2 diabetes and related disorders, and these numbers grow as the number of people affected continues to increase.
A central component of nearly all of these disorders is insulin resistance at the cellular level. The key intermediate in insulin signaling to control these metabolic pathways is the enzyme phosphatidylinositol 3-kinase (PI 3-kinase or PI3K). This enzyme pathway also plays key regulatory roles in growth, differentiation and control of apoptosis, and is a regulator of longevity in lower organisms, such as C. elegans.
SUMMARY
The present invention is based, at least in part, on novel mechanistic insights into the connection between PI3K/JNK signalling and improved insulin sensitivity.
Provided herein are a number of screening methods that use the proteins in this pathway as targets.
In one aspect, the invention provides methods for identifying candidate compounds for improving insulin sensitivity, e.g., in a mammal in need thereof. The methods include providing a sample comprising p85α and cdc42; contacting the sample with a test compound, and evaluating binding of p85α to cdc42 in the sample. A test compound that decreases binding of p85α to cdc42 is a candidate compound for improving insulin sensitivity in a mammal.
In another aspect, the invention provides methods for identifying candidate compounds for improving insulin sensitivity, e.g., in a mammal in need thereof. The methods include providing a sample comprising cdc42 and MLK3; contacting the sample with a test compound, and evaluating phosphorylation of MLK3 by cdc42 in the sample. A test compound that decreases phosphorylation of MLK3 by cdc42 is a candidate compound for improving insulin sensitivity in a mammal. In an additional aspect, the invention provides methods for identifying candidate compounds for improving insulin sensitivity, e.g., in a mammal in need thereof. The methods include providing a sample comprising MLK3 and MKK4; contacting the sample with a test compound, and evaluating phosphorylation of MKK4 by MLK3 in the sample. A test compound that decreases phosphorylation of MKK4 by MLK3 is a candidate compound for improving insulin sensitivity in a mammal.
In a further aspect, the invention provides methods for identifying candidate compounds for improving insulin sensitivity, e.g., in a mammal in need thereof. The methods include providing a sample comprising MKK4 and JNK; contacting the sample with a test compound, and evaluating phosphorylation of JNK by MKK4 in the sample. A test compound that decreases phosphorylation of JNK by MKK4 is a candidate compound for improving insulin sensitivity in a mammal.
In yet another aspect, the invention provides methods for identifying candidate compounds for improving insulin sensitivity, e.g., in a mammal in need thereof. The methods include providing a sample comprising PTEN and JNK; contacting the sample with a test compound, and evaluating phosphorylation of PTEN by JNK in the sample. A test compound that decreases phosphorylation of PTEN by JNK is a candidate compound for improving insulin sensitivity in a mammal.
In one aspect, the invention provides methods for identifying candidate compounds for improving insulin sensitivity, e.g., in a mammal in need thereof. The methods include providing a sample comprising one or more target proteins selected from the group consisting of cdc42, MLK3, or MKK4; contacting the sample with a test compound; evaluating binding of the test compounds to the target protein; and selecting the test compound as a candidate compound if it binds to the target protein.
In some embodiments, the methods include providing a cell having a functional insulin signalling pathway comprising p85α, cdc42, MLK3, MKK4, and JNK; contacting the cell with the candidate compound; contacting the cell with an amount of insulin sufficient to activate said pathway; evaluating activation of said pathway in the cell in the presence of the test compound; comparing activation of said pathway in the cell in the presence of the test compound to a reference representing activation of said pathway in the cell in the absence of the test compound, and selecting the candidate compound as a candidate therapeutic agent for improving insulin sensitivity in a mammal if activation of said pathway is reduced in the presence of the test compound as compared to activation of said pathway in the absence of the test compound. In some embodiments, activation of said pathway is determined by one or more of detecting cdc42 activation of MLK3; detecting binding of cdc42 to MLK3; detecting MLK3 kinase activity; detecting phosphorylation of MKK4; or detecting JNK activation.
In some embodiments of the methods described herein, the test compound can be, e.g., a small molecule, or a peptide or peptidomimetic. As used herein, "small molecules" refers to small organic or inorganic molecules of molecular weight below about 3,000 Daltons. In some embodiments of the methods described herein, the sample is or includes a cell, e.g., a cell expressing the recited proteins, either endogenously or exogenously..
In some embodiments, the methods described herein further include administering the candidate compound to a mammal, e.g., a mammal in need of increased insulin sensitivity, and evaluating whether the candidate compound increases insulin sensitivity in the mammal. In some embodiments, the mammal is a non-human experimental animal.
In some embodiments, the methods further include selecting the compound if is increases insulin sensitivity and evaluating the compound in a clinical trial.
Unless otherwise defined, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. Methods and materials are described herein for use in the present invention; other, suitable methods and materials known in the art can also be used. The materials, methods, and examples are illustrative only and not intended to be limiting. All publications, patent applications, patents, sequences, database entries, and other references mentioned herein are incorporated by reference in their entirety, including the priority application, USSN 60/821, 118, filed August 1 , 2006. In case of conflict, the present specification, including definitions, will control.
Other features and advantages of the invention will be apparent from the following detailed description and figures, and from the claims.
DESCRIPTION OF DRAWINGS FIG. IA is a set of Western blots for Pϊkirl gene products with an antibody against the N-terminal SH2 domain (pan-p85) in tissues lysates, as indicated, from control, heterozygous KO and L-Pik3rlKO mice. Tissues were collected from mice after an overnight fast, and proteins were extracted and processed as described in the Methods section. Each lane represents lysates from a different mouse. FIGs. IB are line graphs of fasted blood glucose (IB) and fasted serum insulin levels (1C) at 8, 16, and 24 weeks of age in lox/lox and KO mice. Open circles (O) — lox/lox; closed circles (•)— L-Pik3rlKO.
FIGs. ID and IE are bar graphs of serum triglycerides (ID) and serum non- esterified free fatty acid levels (IE) from lox/lox or KO mice in the fasted state. FIG. IF is a line graph showing the results of Glucose tolerance tests (2 g/kg, intraperitoneally) performed on mice following a 16 hour fast for five days; blood samples were collected and glucose measured at the times indicated. All values are presented as mean±SEM (n=6-20). Open circles (O) — lox/lox; closed circles (•) — L-Pik3rlKO. FIGs. 2A-2E are bar graphs illustrating the results of hyperinsulinemic- euglycemic clamp analyses and gene expression changes in L-Pik3rlKO mice. Male mice (n=l 1) of the indicated genotype at 10-12 weeks of age were subjected to
hyperinsulinemic-euglycemic clamp analysis. A number of parameters were evaluated, including insulin suppression of hepatic glucose production (2A), glucose infusion rates (2B), and in vivo 14C-deoxyglucose uptake in muscle (2C) and epididymal fat tissue (2D). FIG. 2E is a bar graph showing the results of quantitative RT-PCR analysis of mRNA levels in lox/lox and L-Pik3r IKO mice of phosphoenolpyruvate carboxykinase (Pckl), glucose-6-phosphatase (G6Pc), fructose- 1,6-bisphosphatase (Fbpl), peroxisome proliferator-activated receptor (PPAR)-γ coactivator-1 alpha (Ppargcl) and tribbles3 (trib3) and glucokinase (Gckl). N=8 for each genotype for RT-PCR experiments. *p<0.05 vs lox/lox mice. FIG. 3A is a bar graph illustrating enhanced Akt activation in L-Pik3rlKO mice. PI3K activity was assayed in IRS-I, IRS-2 and phospho-Tyrosine (pTyr) immunoprecipitates (bars represent mean ± SEM, n=5, *p<0.05).
FIGs. 3B is a Western blot of pi 10a and p85α from pi 10a immunoprecipitates. FIG. 3 C is a par of blots of pTyr (upper panel) and insulin receptor (lower panel) in insulin receptor (β-subunit) immunoprecipitates.
FIG. 3D is a blot showing Ser473 phosphorylation of Akt. FIG. 3 E is a bar graph illustrating Akt kinase activity as measured from Akt immunoprecipitates using Crosstide as a substrate. Bars represent mean±SEM, n=8, *p<0.05.
FIG. 4A is a set of six photomicrographs illustrating enhanced PIP3 levels in L-Pik3rlKO mice (bottom three panels) as compared to lox/lox mice (top three panels) due to decreased PTEN activity. Immunofluorescent staining with a primary anti-PIP3 antibody (IgM) and an anti-mouse secondary antibody conjugated to Alexafluor Red and counterstained with DAPI. Following an overnight fast, mice were injected with saline (time=0) or 5U of insulin for 5 or 15 minutes. N = six mice in each genotype/treatment group.
FIG. 4B is a bar graph of the results of quantification of the immunofluorescence from PIP3 staining shown in FIG. 4A. Representative slides were chosen from each mouse and the fluorescence intensity was measured and analyzed with VH-H 1A5 Analyzer software (KEYENCE, Osaka, Japan).
FIGs. 4C and 4D are bar graphs of insulin-stimulated pTyr-associated PI3K activity (4C) and PTEN activity (4D) in lox/lox or KO animals at the indicated timepoints.
FIG. 4E is a Western blot showing PTEN levels in lox/lox and KO mice at the indicated timepoints.
FIG. 5 A is a blot showing expression levels of hepatic p85α and p50 in lox/lox and KO mice. Six-week-old L-Pik3rl KO mice and lox/lox controls were fed a normal chow (NC) or a high fat diet (HFD) for a total of 8 weeks.
FIG. 5B is a line graph indicating body weight for each week on either diet. Open squares (D)- lox/lox, NC, Open circles (O)- L-Pik3r IKO, NC; Closed squares (■)— lox/lox, HFD; closed circles (•)— L-Pik3rlK0, HFD.
FIG. 5C is a trio of Western blots performed against liver lysates of mice of indicated genotype and diet using the phosphoserine 473 Akt antibody, phospho-JNK antibody, phosphoserine307 IRS-I antibodies. The phospho-specific antibody blots were stripped and re-probed with the antibody for total levels of the corresponding proteins, which in each case did not change and are therefore not shown. The bars represent ± SEM (n=6-8).
FIG. 5D is a line graph of fasting blood glucose from mice of the indicated genotype and diet. *p<0.05, **p<0.01 FIG. 6 is a series of seven immunoblots of pJNK, JNK, pS307IRS-l, IRS-I, pAkt, Akt, and p85 in lox/lox and KO mice.
FIG. 7 A is a bar graph illustrating cdc42 activity as determined by PAKl pulldown assay from liver lysates after three minutes of insulin stimulation via the portal vein. FIG. 7B is a quartet of Western blots against phosphor-MKK4 (pMM4),
MKK4, phospho-JNK (pJNK) and JNK from liver lysates of mice of indicated genotypes.
FIG. 7C is a series of five phosphoimmunoblots from primary hepatocytes against pJNK, pMKK4, pAkt, Myc tag, and p85. FIG. 8A is an immunoblot of LacZ, p85α, p55α, and p50α. Recombinant adenoviruses were injected via tail-vein into 10-12 week old male mice of the indicated genotype. Mice were injected with adenoviruses encoding control LacZ, or one of the Pik3rl gene products, p85α, p55α, and p50α. An extra band of
approximately 5OkD appears in the livers treated with p55α adenovirus, and this likely represents a proteolytic breakdown product of p55α.
FIG. 8B is a line graph of PBK activity from the mice injected with the indicated adenoviruses is 8A. 5 FIG. 8C is a trio of Western blots performed against liver lysates of mice treated with adenovirus, using phospho-JNK and phosphoserine307 antibodies. The phospho-specific antibody blots were stripped and re-probed with the antibody for total levels of the corresponding proteins (data not shown).
FIGs. 8D and 8E are bar graphs of fasted blood glucose (8D) and fasted serum o insulin (8E) in mice treated with the indicated adenoviruses.
FIG. 8F is a line graph of the results of GTTs in mice whose livers were reconstituted with one of the Pϊkirl gene products. The bars represent ±SEM (n=6- 8).
FIG. 9A is trio of immunoblots of LacZ, p85α, p55α, and p50α. Recombinant5 adenoviruses were injected via tail-vein into 10-12 week old male mice of the indicated genotype. Mice were injected with adenoviruses encoding control LacZ, or one of the Pik3rl gene products, p85α, p55α, and p50α. An extra band of approximately 5OkD appears in the livers treated with p55α adenovirus, and this likely represents a proteolytic breakdown product of p55α. 0 FIGs. 9B and 9C are bar graphs of PDK activity (9B) and cdc42 activity (9C) in the mice injected with the indicated adenoviruses.
FIG. 9D is a line graph of fasted blood glucose levels in mice treated with the indicated adenoviruses. The bars represent ± SEM (n=6-8).
FIG. 1OA is an immunoblot of LacZ, p85α, ΔSH3, ΔBH, and ΔΔ expression,5 showing that the Activation of cdc42 Requires an Intact N-terminus of p85α.
Recombinant adenoviruses were injected via tail-vein into 10-12 week old male mice of the indicated genotype. Mice were injected with adenoviruses encoding control LacZ, or one of the Pik3rl gene products, p85α, p55α, and p50α. Immunoblots were then performed. 0 FIGs. 1OB and 1OC are bar graphs of PI3K activity (10B) and cdc42 activity
(10C) from the mice injected with the indicated adenoviruses. The bars represent ±SEM (n=6-8).
FIG. 11 is a schematic illustration of a hypothetical assignment of functions to the regions of p85α. This regulatory subunit of PI3K regulates insulin sensitivity through both positive and negative mechanisms. p85α regulates PIP3 levels through its traditional role as a regulator of PDK activity, but it also independently regulates PIP3 levels via the activation of JNK via cdc42 and possibly through the activation of a lipid phosphatase or by the alteration of subcellular localization of PDK.
FIG. 12A is a gar graph illustrating the average weekly food intake of lox/lox (FLOX) or KO mice of either normal chow (NC) or high- fat diet HFD) over a week, expressed in grams. Genotypes of animals are indicated directly on the bars. FIGs. 12B and 12C are bar graphs illustrating fasting blood glucose (12B)and fasting serum insulin (12C) in lox/lox or KO mice after an eight-week treatment with either normal chow or high-fat diet.
FIGs. 13A-B are schematic illustrations of natural variants (13A) or artificial mutants (13B) of p85α, with their effect on PDK activity, JNK activity, or insulin sensitivity.
FIG. 14 is a schematic illustration of the structure of the PTEN (top) and a phosphoimmunoblot of PTEN showing phosphorylation by JNK (bottom).
DETAILED DESCRIPTION
The present invention describes methods of identifying novel modulators of the PI3-kinase/p85α signalling pathway that eventually modulate JNK and PTEN activity, thereby regulating insulin action and sensitivity.
PI 3-kinase regulatory subunits as modulators of the stress kinases JNK and p38
The enzyme phosphatidylinositol 3-kinase is central to the metabolic actions of insulin. The enzyme itself is comprised of a regulatory subunit and a catalytic subunit. The catalytic subunit is either pi 10a (GenBank Ace. No. NM_006218.2) or pi lOβ (GenBank Ace. No. BCl 14432.1). The most common forms of regulatory subunit are p85α (GenBank Ace. No. NM_181523.1) and p85β (GenBank Ace. No. BC090249.1 or BC070082.1), which are products of separate genes. In addition, GRBl, the gene encoding p85α, also produces several alternatively spliced variants, p55α, p50α and forms with small additional inserted exons. These different regulatory subunits are expressed to different levels in different tissues and also
differentially regulated in disease states such as obesity. In US Pat. App. Pub. No. 2002-0051786-A1, the present inventors demonstrated that heterozygous deletion of p85α improves insulin sensitivity and can protect mice with genetic and acquired forms of insulin resistance, including the insulin resistance associated with high fat diet, from developing diabetes. Furthermore, this effect can be mimicked by reducing expression of p85α in liver only via tissue specific knockout (see Barbour et al, J. Biol. Chem. 280(45):37489-94 (2005). Epub 2005 Sep 8).
The work described herein demonstrates novel mechanistic insights into this connection between p85α and improved insulin sensitivity. One aspect of the improved sensitivity is the result of a decrease in serine phosphorylation of the insulin receptor substrate IRS-I . Serine phosphorylation of IRS-I has been previously shown to reduce its tyrosine phosphorylation and reduce its ability to activate the PI 3-kinase pathway, thus reducing insulin's metabolic actions. Several protein kinases have been shown to phosphorylate IRS-I on serine residues, but a key kinase in this pathway is JNK. As described herein, p85α plays a novel role in JNK activation by binding to and activating the small GTPase cdc42, which leads to the activation of Mixed Lineage Kinase-3 (MLK3), which phosphorylates Mitogen-Activated Protein Kinase Kinase 4 (MKK4), leading to JNK activation. A potential schematic is as follows:
P85 -> cdc42 -*?^ MLK3 -> MKK4 -> JNK
Furthermore, this pathway is normally increased in obesity-related insulin resistance, and reducing the level of p85 improves insulin sensitivity, as least in part, by reducing activation of cdc42, MMK4 and JNK, leading to reduced serine phosphorylation of IRS-I . Therefore, modulation of this pathway provides a number of novel targets for drugs that improve insulin sensitivity.
Thus, the methods described herein can be used to identify and optimize small molecule inhibitors that reduce the interaction of cdc42 with p85 and/or reduce cdc42- mediated activation of MKK4, thereby reducing JNK activation and serine phosphorylation of IRS-I, and enhancing insulin sensitivity in insulin resistant states. In some embodiments, the methods include the use of a high throughput in vitro assay system to identify test compounds, e.g., small molecules, that have this property and can therefore serve as drugs for diabetes, metabolic syndrome and related disorders.
PI 3-kinase regulatory subunits as modulators of the lipid phosphatase PTEN
As noted above, PI 3-kinase is central to the metabolic actions of insulin. This occurs via formation of its phospholipid products, in particular PIP3, which activates downstream enzymes like Akt and the atypical PKCs λ and ζ. This process is antagonized or reverse by the action of lipid phosphatases, which break down PIP3, the most important of which is the enzyme "phosphatase and tensin homolog," or PTEN.
Regulation of PTEN activity is multi-factorial and includes allosteric regulation of PTEN by its lipid products, subcellular targeting, cofactor interactions, as well as post-translational modifications (Gericke, Gene. 2006 Jun 7;374: l-9. Epub 2006 Mar 14). Although there is phosphorylation of PTEN in a number of cell types, the functional consequences of PTEN phosphorylation have been largely uncharacterized. The vast majority of PTEN phosphorylation occurs on serine residues (>90%), especially serine residues 370 and 385. In addition, phosphorylation has been reported on other serines (e.g., serine 229, 360, 362, and/or 380) and threonines (e.g., threonine 223, 319, 321, 366, 382, 383, and/or 401), residues that map to the C2 lipid binding domain (amino acids (a. a.) 190-351) and a region proximal to the PDZ ligand sequence (a.a. 401-403) at the C-terminus. Some recent evidence suggests that the phosphorylation of PTEN may regulate its stability and play an important role in modulating biological activity (Vazquez et al, MoI. Cell. Biol. 20:5010-5018 (2000); Okahara et al., J. Biol. Chem. 279:45300-45303 (2004)), serving to inactivate the enzyme. Upon dephosphorylation, PTEN becomes active with a commensurate decrease in protein stability (Vazquez, 2000, supra; Vazquez et al., J.Biol. Chem. (2001) 276(52):48627-30. Epub 2001 Nov 13; Okahara, 2004, supra).
In immunoblotting experiments performed with an antibody that recognizes the phosphorylation of PTEN on sites in the distal C2 domain (Ser380/Thr382/383), it was discovered that PTEN undergoes phosphorylation in response to insulin stimulation. More important, this phosphorylation is increased substantially in p85 ~'~ cells as compared to p85 +/+ cells, and that this correlates with a decrease in PTEN activity. This results in more sustained PIP3 levels following insulin stimulation in liver p85α knockout mice and another mechanism for enhanced insulin sensitivity in these animals.
Based in the above observations, a key regulator of PTEN would be the kinase that phosphorylates PTEN in response to p85 regulation. As described herein, there is considerable evidence that JNK is the critical PTEN Kinase. First, there is a conserved JNK-binding motif (K/RXXXXLXL) at residues 313-320 of PTEN based 5 upon similar motifs obtained from established JNK substrates (Dickens et al., Science, 277(5326):693-696 (1997); Yang et al., EMBO J., 17(6):1740-1749 (1998)). Moreover, two potential phosphorylation sites (Ser338 and Thr366) for JNK/MAP kinase family exist in the C-terminal region of PTEN, and this region has been shown to be important for stability and/or activity of PTEN. Since JNK activity is decreased o in p85α KO mice and cells, a decrease in JNK would also be expected to result in a change in serine phosphorylation and activity of PTEN.
Target Sequences
Sequences for the protein and nucleic acid targets useful in the methods described herein are known in the art. The following is a list of exemplary sequences5 that can be used. As one of skill in the art will appreciate, homologs of these sequences from other species can also be used. For example, nucleic acids that hybridize under stringent conditions to a sequence listed herein, or a polypeptide encoded by such a sequence, can equally be used. As used herein, the term "hybridizes under stringent conditions" describes conditions for hybridization and0 washing. Stringent conditions are known to those skilled in the art and can be found in Current Protocols in Molecular Biology, John Wiley & Sons, N.Y. (1989), 6.3.1- 6.3.6. Aqueous and nonaqueous methods are described in that reference and either can be used. As used herein, stringency conditions are 0.5 M sodium phosphate, 7% SDS at 65°C, followed by one or more washes at 0.2 X SSC, 1% SDS at 65°C. 5 Alternatively, a sequence that is at least 80%, e.g., at least 85%, 90%, 95%, 96%, 97%, 98%, 99%, or more identical to a sequence listed herein can be used. The comparison of sequences and determination of percent identity between two sequences can be accomplished using a mathematical algorithm. For purposes of the present methods, the percent identity between two amino acid sequences is 0 determined using the Needleman and Wunsch ((1970) J. MoI. Biol. 48:444-453 ) algorithm which has been incorporated into the GAP program in the GCG software package (available on the world wide web at gcg.com), using the default parameters,
i.,e., a Blossum 62 scoring matrix with a gap penalty of 12, a gap extend penalty of 4, and a frameshift gap penalty of 5.
PTEN - Phosphatase and Tensin Homolog
The mouse PTEN sequences are available at:
See also: HomoloGene:265.
JNKl /2 - c-Jun N -Terminal Kinases 1 and 2
Sequences of JNKl, also known as mitogen-activated protein kinase 8, can be found at:
Sequences of JNK2, also known as mitogen activated protein kinase 9
(MAPK9), can be found at:
MLKi - Mixed Lineage Kinase 3
Sequences of MLK3, also known as Mitogen- Activated Protein Kinase Kinase Kinase 11 (MAPKKl 1) can be found at:
MKK4 - MAP Kinase Kinase 4
Sequences of MKK4, also known as mitogen-activated protein kinase kinase 4 (MAP2K4), can be found at:
p85a-p85 alpha, regulatory subunit
Sequences of p85, also known as PIK3R1 phosphoinositide-3-kinase, regulatory subunit 1 (PIK3R1), can be found at:
The invention includes methods for screening of test compounds, to identify compounds that modulate a pathway described herein, e.g., compounds that (i) reduce the interaction of cdc42 with p85, thereby reducing cdc42 activity, or (ii) reduce the activation of MLK3 by cdc42, thereby reducing MLK3 activity, or (iii) reduce the phosphorylation of MKK4 by MLK3, thereby reducing MKK4 activity, thus reducing JNK activation and serine phosphorylation of IRS-I, and enhancing insulin action and sensitivity. In addition, each of cdc42, MLK3, and MKK4 are novel targets for inhibition, and can be used as targets for a screening method described herein. The methods described herein can be used to identify compounds that bind to p85, cdc42, MLK3, or MKK4. In addition, the effect of a test compound on the p85 signalling pathway can be determined. For example, the methods can be used to identify compounds that demonstrate (i) binding to p85 and/or cdc42, and/or decrease p85-mediated activation of cdc42, (ii) binding to cdc42 and/or MLK3, and/or decrease csc42-mediated activation of MLK3, or (iii) binding to MLK3 and/or MKK4, and/or decrease MLK3 -mediated phosphorylation of MKK4.
Thus, in general, the methods will include providing a sample that includes one or more of p85, cdc42, MLK3, and MKK4.
For example, in some embodiments, the sample can include p85α and cdc42, e.g., isolated and purified p85α and cdc42 proteins, and the methods include identifying a compound that affects binding between p85 and cdc42. Screening methods suitable for use in these embodiments are known in the art and include, but are not limited to, yeast or mammalian 2-hybrid systems, tagged protein assays, immunoprecipitation assays, and proteomics assays. These methods can be used to identify natural (i.e., endogenous) regulators of cdc42, for example.
In some embodiments, the sample includes isolated and purified cdc42 and MLK3, with or without MKK4, and the methods include detecting identifying compounds that affect activation of MLK3 by cdc42, and/or phosphorylation of MKK4. In some embodiments, the sample includes isolated and purified MLK3 and
MKK4, and the methods include detecting identifying compounds that affect phosphorylation of MKK4.
In some embodiments of the methods described herein, MKK7 can be used in place of MKK4.
In some embodiments, the methods include identifying isoform-specific inhibitors of JNK, i.e., inhibitors that substantially reduce the activity of JNKl while not substantially affecting the activity of JNK2, or vice-versa. As described herein, the different isoforms have different activities depending on the tissues, therefore, it may be desirable to affect only the isoform that is active in the particular cell type. In addition, it may be desirable to affect only the ability of JNK to phosphorylate PTEN, e.g., drugs that bind to either JNK1/2 or PTEN and prevent phosphorylation.
In order to evaluate the ability of a compound to affect signalling, it will generally be desirable to test the compound in a cell. For example, the methods can include adding the compound to cells that express all of these proteins, and exhibit p85 signalling via cdc42 and MKK4 that results in JNK activation and serine phosphorylation of IRS- 1 and/or PTEN. The methods can then include contacting the cells with the compound, and evaluating an effect of the compound on JNK activity (e.g., serine phosphorylation of IRS-I or PTEN). In some embodiments, the methods further include determining whether the compound has an effect on phosphorylation of other substrates by JNK, and selecting a test compound if it selectively inhibits JNK phosphorylation of PTEN. For example, such a compounds might be designed such that it binds a JNK-recognition site, or a JNK-phosphorylation site, on PTEN. A number of suitable assays are known in the art, see, e.g., Methods in Enzymology. Volume 201 : Protein Phosphorylation. Part B: Analysis of Protein Phosphorylation. Protein Kinase Inhibitors, and Protein (Methods in Enzymology) by John N. Abelson, Melvin I. Simon, Tony Hunter, and Bartholomew M. Sefton (Hardcover - Jan 15, 1991), Protein Phosphorylation. Part A: Protein Kinases: Assays. Purification.
Antibodies. Functional Analysis. Cloning, and Expression. Volume 200: Volume 200, Part A (Methods in Enzymology) by John N. Abelson, Melvin I. Simon, Tony Hunter, and Bartholomew M. Sefton (Hardcover - JuI 28, 1991).
Test Compounds The test compounds can be, e.g., natural products or members of a combinatorial chemistry library.
In some embodiments, the test compounds are initially members of a library, e.g., an inorganic or organic chemical library, peptide library, oligonucleotide library, or mixed-molecule library. In some embodiments, the methods include screening small molecules, e.g., natural products or members of a combinatorial chemistry library. These methods can also be used, for example, to screen a library of proteins or fragments thereof, e.g., proteins that are expressed in liver or pancreatic cells. A given library can comprise a set of structurally related or unrelated test compounds. Preferably, a set of diverse molecules should be used to cover a variety of functions such as charge, aromaticity, hydrogen bonding, flexibility, size, length of side chain, hydrophobicity, and rigidity. Combinatorial techniques suitable for creating libraries are known in the art, e.g., methods for synthesizing libraries of small molecules, e.g., as exemplified by Obrecht and Villalgordo, Solid-Supported Combinatorial and Parallel Synthesis of Small-Molecular- Weight Compound Libraries. Pergamon-Elsevier Science Limited (1998). Such methods include the "split and pool" or "parallel" synthesis techniques, solid-phase and solution-phase techniques, and encoding techniques (see, for example, Czarnik, Curr. Opin. Chem. Bio. 1 :60-6 (1997)). In addition, a number of libraries, including small molecule libraries, are commercially available.
In some embodiments, the test compounds are peptide or peptidomimetic molecules, e.g., peptide analogs including peptides comprising non-naturally occurring amino acids or having non-peptide linkages; peptidomimetics (e.g., peptoid oligomers, e.g., peptoid amide or ester analogues, β-peptides, D-peptides, L-peptides, oligourea or oligocarbamate); small peptides (e.g., pentapeptides, hexapeptides, heptapeptides, octapeptides, nonapeptides, decapeptides, or larger, e.g., 20-mers or more); cyclic peptides; other non-natural or unnatural peptide-like structures; and inorganic molecules (e.g., heterocyclic ring molecules). In some embodiments, the test compounds are nucleic acids, e.g., DNA or RNA oligonucleotides.
In some embodiments, test compounds and libraries thereof can be obtained by systematically altering the structure of a first test compound. Taking a small molecule as an example, e.g., a first small molecule is selected that is, e.g., structurally similar to a known phosphorylation or protein recognition site. For example, in one embodiment, a general library of small molecules is screened, e.g., using the methods described herein, to select a fist test small molecule. Using
methods known in the art, the structure of that small molecule is identified if necessary and correlated to a resulting biological activity, e.g., by a structure-activity relationship study. As one of skill in the art will appreciate, there are a variety of standard methods for creating such a structure-activity relationship. Thus, in some instances, the work may be largely empirical, and in others, the three-dimensional structure of an endogenous polypeptide or portion thereof can be used as a starting point for the rational design of a small molecule compound or compounds. In some embodiments, test compounds identified as "hits" (e.g., test compounds that demonstrate activity in a method described herein) in a first screen are selected and optimized by being systematically altered, e.g., using rational design, to optimize binding affinity, avidity, specificity, or other parameter. Such potentially optimized structures can also be screened using the methods described herein. Thus, in one embodiment, the invention includes screening a first library of test compounds using a method described herein, identifying one or more hits in that library, subjecting those hits to systematic structural alteration to create one or more second generation compounds structurally related to the hit, and screening the second generation compound. Additional rounds of optimization can be used to identify a test compound with a desirable therapeutic profile.
Test compounds identified as hits can be considered candidate therapeutic compounds, useful in treating disorders described herein. Thus, the invention also includes compounds identified as "hits" by a method described herein, and methods for their administration and use in the treatment, prevention, or delay of development or progression of a disease described herein.
EXAMPLES
The invention is further described in the following examples, which do not limit the scope of the invention described in the claims.
Example 1 : Phosphoinositide 3-Kinase Regulatory Subunit p85α Suppresses Insulin Action via Positive Regulation of PTEN
Insulin action on the liver is required for the proper maintenance of metabolic homeostasis. Under normal conditions, insulin inhibits gluconeogenesis and activates lipogenesis to promote proper fuel utilization in the fed state. The failure of insulin to
regulate hepatic function can lead to unfettered hepatic glucose output and elevated lipogenesis in the liver, which are two key features of the metabolic syndrome (Shimomura, et al., (2000) MoI. Cell, 6(l):77-86). Not surprisingly, an overwhelming amount of epidemiological (Tripathy et al., (2004) Diabetologia, 47(5):782-93) and physiological (Fisher and Kahn, (2003) J. Clin. Invest., 111(4):463-468) evidence links hepatic insulin resistance to the development of type 2 diabetes.
The molecular mechanisms that maintain hepatic insulin sensitivity are likely linked to the class IA phosphoinositide 3-kinase (PI3K) pathway, which is a critical regulator of insulin action (Saltiel and Kahn, (2001) Nature, 414(6865):799-806). PI3K is an obligate heterodimer, with an SH2-containing regulatory subunit (p85) and a catalytic subunit (pi 10). The regulatory subunit mediates the binding, activation and localization of the PI3K enzyme (Virkamaki, et al., (1999) J. Clin. Invest. 103(7):931-43), (Backer et al., (1992) EMBO J., 11(9):3469-79), (Fruman et al., (1996) Genomics, 37(1): 113-21). Despite the crucial role that the regulatory subunits play in mediating insulin- dependent PI3K signaling, insulin sensitivity correlates inversely with regulatory subunit expression. This paradoxical relationship is demonstrated by knockouts of the different regulatory subunits. For instance, the deletion of the less abundant isoforms of Pik3rl (p55α/p50α), (Chen et al., (2004) MoI. Cell. Biol. 24:320-9) or the minor isoform p85beta (Ueki et al., (2002) Proc. Natl. Acad. Sci. U.S.A., 99(l):419-24) mildly improve insulin sensitivity, while the knockout of more abundant p85α markedly improves glucose homeostasis (Terauchi et al., (1999) Nat. Genet. 21(2):230-235). Germline deletion of all three products of the Pik3rl gene (p85α, p55α, and p50α) also enhances insulin sensitivity, though these mice die perinatally (Fruman et al., (2000) Nat. Genet. 26(3):379-82). These data indicate that in addition to its traditional positive function as a component of the PI3K holoenzyme, p85α is also a potent negative regulator of insulin signaling.
The negative effects of p85α on insulin action may have important consequences in the pathophysiology of insulin resistance and diabetes. For example, the increased expression of p85α in mouse models of gestational diabetes (Barbour et al., (2004) Endocrinology 145(3): 1144-50) and in obese humans (Bandyopadhyay et al., (2005) Diabetes 54(8):2351-9) is strongly linked with insulin resistance.
Conversely, heterozygosity for Pϊkirl prevents the onset of diabetes in genetically insulin resistant mice (Mauvais-Jarvis et al, (2002) J. Clin. Invest. 109, 141-149).
The physiologic and molecular mechanisms that underlie this negative regulation by p85α have been difficult to determine. At the physiologic level, germline knockouts that delete p85 isoforms in all tissues throughout development cannot discriminate whether the negative action of p85α occurs as a tissue- autonomous effect or as a consequence of altered endocrine communication between insulin responsive tissues. This situation may also be complicated by potential compensatory adjustments to loss of p85 during development. Similarly, the molecular mechanisms of p85α's negative effects are still understood only at rudimentary level. To clarify the mechanisms by which p85α regulates insulin sensitivity in vivo, at the physiologic and molecular level, mice with a liver-specific deletion oiPik3rl (L-Pik3rlKO) were created. This conditional knockout oiPik3rl (p85α, p55α, and p50α) in hepatocytes circumvents the perinatal lethality observed in the corresponding germline knockout mice and furthermore allows the investigation of the specific role of the liver in the physiological actions of p85α. As described herein, L-Pik3rlKO mice have enhanced hepatic and whole body insulin sensitivity as well as increased Akt activity in liver, despite decreased total hepatic PBK activity. Furthermore, p85α mediates its negative effect on insulin sensitivity at least in part via the activation of PTEN lipid phosphatase activity.
Experimental Procedures
All animals were housed on a 12-hour light-dark cycle and fed a standard rodent chow. All mice used in this study were on a 129Sv-C57BL/6-FVB mixed genetic background. Metabolic studies. For glucose tolerance testing (GTT), blood samples were obtained at 0, 15, 30, 60, and 120 minutes after intraperitoneal injection of 2 g/kg dextrose. Insulin tolerance tests were performed by injecting 1 U/kg insulin (Novolin, Novo Nordisk, Denmark) intraperitoneally, followed by blood collection at 0, 15, 30 and 60 minutes after injection. Blood glucose values were determined using a One Touch II glucose monitor (Lifescan Inc., Milipitas, CA). Plasma insulin levels were measured by ELISA using mouse insulin as a standard (Crystal Chem Inc., Chicago, IL). Non-esterified free fatty acid levels were measured from random fed mice using
a kit from Wako Diagnostics, while serum triglycerides were measured by Anilytics (Gaithersburg, MD).
Hyperinsulinemic-euglycemic clamp. Mice were anaesthetized with a 1.2% solution of 2,2,2-tribromoethanol in normal saline, followed by the microsurgical insertion of a catheter into the right jugular vein. Approximately 7 days of recovery, mice were fasted for 5 hours and were infused with a constant (2.5 mU/kg/min) dose of insulin and a variable glucose infusion rate to maintain euglycemia and assess whole-body insulin sensitivity. An infusion of a [3-3H]-glucose tracer was used to ascertain hepatic glucose production, and a bolus of 2-deoxy -D-[I -14C] glucose was administered during steady state conditions to determine fat and muscle-specific glucose uptake.
In vivo insulin signaling. Following an overnight fast, the mice were anesthetized with Avertin (2,2,2-tribromoethanol in PBS), and injected with 5 U of regular human insulin (Novolin, Novo Nordisk, Denmark) via the inferior vena cava. Five minutes after the insulin bolus, tissues were removed and frozen in liquid nitrogen. Immunoprecipitation and immunoblot analysis of insulin signaling molecules was performed as previously described (Taniguchi et al, (2005) J. Clin. Invest. 115(3):718-27).
Quantitative Reverse Transcription (RT) -PCR analysis. Total RNA was isolated from mouse tissues using an RNeasy kit (QIAGEN, Valencia, CA), and cDNA was prepared using the Advantage RT-PCR kit (BD Biosciences, Palo Alto, CA) with random hexamer primers, according to manufacturer's instructions. PCR reactions were run in triplicate and quantitated in the ABI Prism 7700 Sequence Detection System. Ct values were normalized to TATA box binding protein (TBP) expression, and the results were expressed as a fold change of mRNA compared to control lox/lox mice.
Antibodies. Rabbit polyclonal anti-IRS-1 antibody (IRS-I), anti-IRS-2 antibody (IRS-2), anti-IR antibody (IR) and pan-p85α antibody were generated as described previously (Ueki et al., (2002) Proc. Natl. Acad. Sci. U.S.A., 99(l):419-24). The anti-TRB3 antibody was a gift from Marc Montminy (Du et al., (2003) Science 300, 1574-7). Rabbit polyclonal anti-Akt, anti-phospho Akt (S473) anti-PTEN were purchased from Cell Signaling Technology (Beverly, MA). The phosphotyrosine (pTyr) antibody, 4G10, was purchased from Upstate Biotechnology, Inc. (Lake
Placid, NY). Goat polyclonal anti-Aktl/2 antibody (Akt) was purchased from Santa Cruz Biotechnology, Inc. (Santa Cruz, CA).
In vitro kinase assays. Tissue homogenates from liver, muscle or fat were extracted with tissue homogenization buffer and subjected to immunoprecipitation with IRS-I, IRS-2, or pTyr antibodies followed PI3K assay as described previously (Taniguchi et al, (2005) J. Clin. Invest. 115(3):718-27). For Akt kinase activity, the enzyme was immunoprecipitated from lysates with a goat polyclonal Akt 1/2 antibody, and kinase activity measured using CROSSTIDE™ (Upstate) as a substrate peptide. PTEN activity assay. Phosphatidylinositol (Avanti Polar Lipids) was in vitro phosphorylated to form phosphatidylinositol 3 -phosphate (PI-3P) by recombinant PI3K (Upstate) with [32P]γ-ATP. The phosphorylated lipid was extracted with methanol/chloroform, dried under nitrogen gas, reconstituted into PTEN assay buffer (10 mM Tris HCl, 25 mM NaCl, />H=7.5) then incubated with PTEN immunoprecipitates from lox/lox or L-Pik3rlKO liver lysates. The PI-3P was re- extracted with methanol/choloroform and run on a TLC plate with a n-propanaol:2M acetic acid (65:35). The intensity of PI-3P spots were quantitated with NIH Image. PTEN activity was determined relative to IgG immunoprecipiates from lox/lox livers. Relative PTEN activity was compared with lox/lox without insulin stimulation. Statistics. Data are presented as ± s.e.m. Student's t-test was used for statistical analysis between two groups, while statistical significance between multiple treatment groups was determined by analysis of variance (ANOVA) and Tukey's t- test.
Results L-Pik3rl KO mice are insulin sensitive
Mice with a liver-specific deletion oiPik3rl were generated via the Cre-loxP system using animals carrying a floxed exon 7, which encodes the N-terminal SH2 domain (Luo, et al., (2005) MoI. Cell. Biol. 25:491-502) and mice carrying the Cre transgene driven by the albumin promoter (Postic and Magnuson, (2000) Genesis 26(2): 149-50). The presence of loxP sites in the Pik3rl gene did not affect expression of p85α as compared to WT littermate controls (Luo et al., (2005) MoI. Cell. Biol. 25:491-502) nor did the presence of albumin-Cre (Michael et al., (2000) MoI Cell
6(l):87-97). The L-Pik3rl KO mice were born in a normal Mendelian distribution
and exhibited normal postnatal growth (data not shown). Western blots of liver extracts of L-Pik3rl KO mice revealed an 80-90% decrease in p85α and a complete loss of p50α (FIG. IA), consistent with complete ablation in hepatocytes. The expression of p85α was unaltered in brain, skeletal muscle and fat. Despite the lack of p85α and p50α in liver, L-Pik3rl KO mice exhibited significant improvements in serum metabolic chemistries. Compared to littermate lox/lox controls, L-Pik3rl KO mice displayed lower fasted blood glucose and fasted serum insulin levels at 8, 16, and 24 weeks of age (FIG. IB and 1C). In addition, L-Pik3rlKO mice exhibited decreased levels of serum triglycerides and circulating free fatty acids (FIGs. ID and IE). At 16 weeks of age, L-Pik3rl KO mice were significantly more glucose tolerant to an intraperitoneal glucose challenge (FIG. IF), but this was observed as early as eight weeks of age and was maintained through 24 weeks of age (data not shown).
To further quantify this insulin sensitivity and to determine which tissues were involved in the phenotype, L-Pik3rl KO mice were subjected to a hyperinsulinemic- euglycemic clamp. This revealed increased hepatic insulin sensitivity such that hepatic glucose production was suppressed by 25% more in the L-Pik3rl KO mice compared to controls (FIG. 2A). Somewhat surprisingly, L-Pik3rl KO mice displayed improved peripheral insulin sensitivity, as determined by a two-fold increase in glucose infusion rates (FIG. 2B). This correlated with a 1.5-fold increase in glucose uptake in muscle (FIG. 2C) and a three-fold increase in glucose uptake in fat (FIG. 2D). Thus, improving insulin sensitivity in the liver secondarily improved insulin sensitivity in peripheral tissues.
The physiologic data indicating increased hepatic insulin sensitivity corresponded with decreased expression of several key gluconeogenic genes. Quantitative RT-PCR analysis found a decrease in phosphoenolpyruvate carboxykinase (Pckl), glucose-6 phosphatase (G6pc) and fructose- 1,6-bisphosphatase (Fbpl) mRNAs in liver by 30%, 70%, and 60%, respectively (FIG. 2e). The mRNA levels of the gluconeogenic regulators, peroxisome proliferator-activated receptor (PPAR)-γ coactivator-1 alpha (Ppargcla) and tribbles3 (Trb3) were also decreased by 50% and 60%, respectively, while glucokinase was upregulated by three-fold. These molecular and physiologic data suggest that the loss of Pik3rl in the liver improves hepatic insulin sensitivity.
L-Pik3rl KO mice display significantly improved activation of Akt despite reduced total PI3K activity
To determine a molecular mechanism of the improved insulin sensitivity, PI3K function was characterized in L-Pik3rlKO mice. As expected, a 50% decrease in IRS-I -associated PBK activity was observed, as was a 60% decrease in IRS-2- associated PDK activity, which together resulted in a 50% reduction in total PBK activity, as measured from pTyr-immunoprecipitates (FIG. 3A). These decreases in PBK were concordant with a 70% decrease in the expression of the pi 10a catalytic subunit (FIG. 3B), but were not associated with decreased insulin receptor activation in the L-Pik3rl KO mice (FIG. 3C). The parallel decrease in expression of pi 10a was expected, since the regulatory subunits are known to stabilize the catalytic subunit (Yu et al., (1998) MoI. Cell. Biol. 18(3): 1379-87).
Despite these decreases in IRS-I, IRS-2 and pTyr-associated PBK activity, both Ser473 phosphorylation of Akt and Akt kinase activity were upregulated by 1.5 fold (FIG. 3E). This enhanced Akt activation could be in part related to the decreased TRB3 levels (FIG. 3F and FIG. 2E), however it seems more likely that the decreased TRB3 expression is merely an indirect marker of insulin sensitivity, since TRB3 levels are directly regulated by insulin (Koo et al., (2004) Nat. Med. 10(5):530-4). Akt is activated by the lipid product of PBK, phosphatidylinositol(3,4,5)- trisphosphate (PIP3). To determine if changes in PIP3 levels might help to explain the increased Akt activation, an immunofluorescent histology technique with an anti-PIP3 antibody was used to estimate the accumulation of PIP3 levels in vivo (FIG. 4A and (Kitamura et al., (2004) J. Clin. Invest. 113(2):209-19). After 5 minutes of insulin stimulation, PIP3 levels increased, but did not differ between control and KO animals; however, after 15 minutes of stimulation, PIP3 levels in the L-Pik3rlKO remained high at 74 ± 3.3% of maximum stimulation, while PIP3 levels in the control decreased to 37.7 ± 2.5% of peak levels (FIG. 4B). These data indicate that although less PBK is associated with IRS-I and IRS-2 in the Pik3rlKO livers, more PIP3 accumulates over time. To determine whether this increased accumulation of PIP3 is due to enhanced
PBK activity or decreased turnover, total PBK activity and the activity of the lipid phosphatase, PTEN, which degrades the PIP3 formed by PBK were directly assessed. Over 15 minutes, pTyr-associated PBK activity was not changed in the livers of
either the lox/lox or L-Pik3rlKO mice, though the insulin-stimulated PI3K activity of the knockout mice remained at only half the level of the controls (FIG. 4C). On the other hand, the lipid phosphatase activity of the negative regulator PTEN was consistently decreased by 40% in L-Pik3rlKO at all timepoints (FIG. 4D). Moreover, insulin had no effect on PTEN activation in either the control or knockout mice. This decrease in PTEN activity is not due to decreased PTEN expression (FIG. 4E), but must be due to some other aspect of the actions of the regulatory subunit of PI3K.
Additional References
Cai et al, (2005) Nat. Med. 11,(2): 183-90. Luo et al., (2005) Proc. Natl. Acad. Sci. U.S.A. 102(29): 10238-43.
Example 2: The p85α Regulatory Subunit of Phosphoinositide 3 -Kinase activates JNK via a cd c42/MKK4 pathway
Insulin resistance is an underlying feature of type 2 diabetes and the metabolic syndrome (Reaven, (2005) Cell. Metab. 1 :9-14). Physiologic and epidemiologic studies have demonstrated strong links between obesity and the development of insulin resistance (Hu et al., (2001) N. Engl. J. Med. 345:790-7; Sinha et al., (2002) N. Engl. J. Med. 346:802-10). Not surprisingly, the rise of type 2 diabetes in the United States over the last decade has paralleled the rapid rise in obesity (Mokdad et al., (2003) JAMA 289:76-9). While there are multiple mechanisms involved in obesity linked insulin resistance, one important mediators of the process is the stress kinase, c-Jun-N-terminal kinase (JNK), which is activated by insulin (Miller et al., (1996) Biochemistry 35:8769-75) and cytokines (Baud et al., (1999) Genes Dev. 13: 1297-308) in obesity and other insulin resistant states and phosphorylates IRS-I on serine residues decreasing its ability to mediate insulin signaling (Ozcan et al., (2004) Science 306:457-61, Pirola et al, (2004) Diabetologia 47: 170-84). The importance of this stress kinase is illustrated by that fact that genetic deletion of JNKl can prevent insulin resistance in severely obese mice (Hirosumi et al., (2002) Nature 420:333-6).
While it is clear that the activation of JNK contributes to the pathophysiology of obesity, the molecules that are directly involved with the activating JNK in this disorder not well understood. One interesting and perhaps counterintuitive candidate molecule is the p85α regulatory subunit of phosphoinositide 3-kinase. PI3K is an
obligate heterodimer, with an SH2 -containing regulatory subunit (p85) and a catalytic subunit (pi 10). The regulatory subunit mediates the binding, activation and localization of the PBK enzyme (Backer et al., (1992) EMBO J. 1 1 :3469-79, Fruman et al., (1996) Genomics 37: 113-21, Virkamaki et al., (1999) J. Clin. Invest. 103 :931- 43). While the p85 subunit is an important link in the metabolic actions of insulin as a component of the PI3K holoenzyme (Taniguchi et al., (2006) Cell. Metab. 3 :343-53), it also plays a role as an independent negative regulator. Indeed, full or partial deletions of the p85 subunit significantly can prevent high-fat diet- induced induced insulin resistance (Terauchi et al., (2004) Diabetes 53:2261-70), as well as diabetes in mice with heterozygous deletions of the insulin receptor and IRS-I (Mauvais-Jarvis et al., (2002) J. Clin. Invest. 109: 141-9).
Several studies have linked p85α circumstantially to the activation of JNK. For instance, cells lacking p85α have diminished JNK activation in response to insulin/IGF- 1, and this is reversed with re-expression of p85α. The p85 regulatory subunit has also been identified as part of a complex that is involved in JNK activation (Zhu et al., (1998) J. Biol. Chem. 273:33864-75), and has been shown to bind to the small GTPase cdc42 (Tolias et al., (1995) J. Biol. Chem. 270: 17656-9; Zheng et al., (1994) J. Biol. Chem. 269: 18727-30) which is an upstream activator of JNK (Minden et al., (1995) Cell 81 : 1 147-57). Moreover, muscle biopsies from obese diabetic patients found a strong positive correlation of p85 expression and JNK.
Although these data suggest an intriguing and novel connection between the PI3K and stress kinase pathways, this question has been difficult to address in vivo, because mice carrying germline deletion of the Pϊkirl gene that encodes p85α and its shorter isoforms p55α and p50α die perinatally (Fruman et al., (2000) Nat Genet 26:379-82). To circumvent the problems of germline knockout models of p85, and to investigate the links between p85α and JNK activation in vivo, mice with a liver- specific deletion oiPik3rl gene (L-Pik3rlKO) were created. As described herein, mice lacking p85α in liver have diminished hepatic activation of JNK and improved whole body insulin sensitivity. In addition, p85α activates JNK via the cdc42-MKK4 pathway and that this interaction requires both an intact N-terminus and functional SH2 domains in the C-terminus of the p85a regulatory subunit. Thus, p85α may regulate insulin sensitivity in both lean and obese mice via crosstalk with the stress kinase pathway.
Experimental Procedures
Animals and high fat diet. All animals were housed on a 12-h light-dark cycle and fed a standard rodent chow (Purina). High fat chow (45% kcal from fat) was purchased from Research Diets. All mice in this study were on a 129Sv- C57BL/6-FVB mixed genetic background, and littermates of the same mixed genetic background were used as controls.
Metabolic studies. Metabolic studies were performed as described above in Example 1.
Recombinant Adenoviruses. The p50α and p55α adenoviruses were constructed as previously described (Ueki et al., (2000) MoI. Cell. Biol. 20:8035-46). The constitutively active MKK4 adenovirus was purchased from CellBio Labs (San Diego, CA), and the constitutively active cdc42 adenovirus was a gift from James Bamburg (Kuhn et al., (2000) J. Neurobiol. 44: 126-144). The wild type (WT) p85α, ΔSH3, ΔBH, ΔiSH2, RARA and ΔΔp85 constructs were made as follows. Cloning ofp85 constructs
The p85 constructs were cloned from a cDNA of human p85α using the following PCR primers:
1. WT p85α with C-terminal FLAG tag
5' : AGTGCTGAGGGGTACCAGTACAGA (SEQ ID NO: ) 3' (3 'p85 FLAG tag primer):
GCGTCGACTAACTACTTATCGTCGTCATCCTTGTAATCTCGCCTCTGCTG TGCATATAC (SEQ ID NO: )
2. ΔSH3 (Deleted aa 1-81) (Xbal on 5', Sail on 3', FLAG tagged on N and Cterm):
5' ITAGGCTCTAGACGCCGCCACCATGGATTACAAGGATGACGACGA TAAG AAAATCTCGCCTCCCACAC (SEQ ID NO: ) 3' : 3'p85 FLAG tag primer (see above)
3. ΔBH (Deleted aa 136-308, Xbal on 5', Sail on 3', internal FLAG and C- term FLAG)
1st half: (Xba on 5', BamHI on 3 ')
5':
TAGTAGGCTCTAGACGCCGCCACCATGTACCCATACGATGTTCCAGATTAC GCAAGTGCTGAGGGGTACCAGTACAGA (SEQ ID NO: )
3': CTACTAGGATCCTTTCTTTTCAATGGCTTCCAC (SEQ ID NO: ) 2nd half (BamHI on 5', Sail on 3 ')
5' ITAGTAGGGATCCGATTACAAGGATGACGACGATAAGCCTCCTAA ACCACCAAAACCT (SEQ ID NO: )
3': 3'p85 FLAG tag primer (see above)
4. ΔiSH2 fragment (Deleted pi 10 binding domain from aa 480-510, internal and C-terminal FLAG tag) lst half (BamHI on 3' end)
5': CCTCCTAAACCACCAAAACCTAC (SEQ ID NO: ) 3': CTAATCGATAGAGGTCTGGCACTGTTCTTCAAAT(SEQ ID NO: ) 2nd half (BamHI on 5', Sail on 3')
5 ' : AGT ATCGATGATTAC AAGGATGACGACGAT AAGTTGAAGTCTCG AATCAGTGAAA (SEQ ID NO: )
3': 3'p85 FLAG tag primer (see above)
The PCR fragments from the WT, and ΔSH3 reactions were digested with Xbal/Sall then ligated into pBluescript (pBS). The 5' portion of the ΔBH reaction was digested with Xbal/BamHI and the 3' fragment was digested with BamHI/Sall. The two fragments were ligated together in pBS. The ΔiSH2 construct was made by cutting the above 1.5 kb PCR fragment with EcoRV and Sail, then fusing it with the Xbal-EcoRV fragment of the WTp85α. The ΔΔp85 construct was made by ligating the Xbal-EcoRV fragment of WTp85α FLAG construct (N-terminus) to the EcoRV- SaII ΔiSH2 fragment (C-terminal half).
The above fragments (excised by either Notl/Sall or Xbal/Sall) were ligated into a custom pShuttle vector with a chicken β-actin/CMV enhacer sequence (CAG) and BGH poly A sequence (CAG pShuttle), which was made by simply ligating the CAG enhancer into an empty pShuttle vector. A new multiple cloning site (MCS) was then inserted between the CAG enhancer and the polyA sequence. The MCS was as follows: NotI-NheI-SwaI-EcoRV-Hind3-SbfI-SalI. After subcloning the p85
constructs into this vector, the adenoviruses were then produced according to the standard AdEasy protocol.
To create the recombinant adenoviruses, the above constructs were ligated into an empty pShuttle vector with a CAG promoter and BGH poly A sequence (CAGpShuttle). A new multicloning site was inserted into the vector containing the following restriction sites in sequence: NotI-NheI-SwaI-EcoRV-Hind3-Sbfl-SalI. The adenoviruses were then produced according to the standard AdEasy protocol (He et al, (1998) Proc. Natl. Acad. Sci. U.S.A. 95:2509-14).
Adenovirus-mediated gene transfer and in vivo insulin stimulation. Prior to use on primary hepatocytes or in vivo, all adenoviruses were purified on sequential cesium chloride gradients then dialyzed into PBS containing 10% glycerol. 10-12 week-old male mice were injected via tail vein with an adenoviral dose of 5xlθ8 pfu/g body weight as described previously (Taniguchi et al., (2005) J. Clin. Invest. 1 15:718- 27). On the fifth day after injection, following an overnight fast, the mice were anesthetized with Avertin (1.2% 2,2,2-tribromoethanol in PBS), and injected with 5 U of regular human insulin (Novolin, Novo Nordisk, Denmark) via the inferior vena cava. Five minutes after the insulin bolus, tissues were removed and frozen in liquid nitrogen. Immunoprecipitation and immunoblot analysis of insulin signaling molecules were performed using tissue homogenates prepared in a tissue homogenization buffer that contained 25 mM Tris-HCl (pH 7.4), 10 mM Na3VO4, 100 mM NaF, 50 mM Na4P2O7, 10 mM EGTA, 10 mM EDTA, 2 mM phenylmethylsulfonyl fluoride, 1% Nonidet-P40 supplemented with the Complete protease inhibitor cocktail (Roche). All protein expression data were quantified by densitometry using NIH Image software. Antibodies. Rabbit polyclonal anti-IRS-1 antibody (IRS-I), anti-IRS-2 antibody (IRS-2), anti-IR antibody (IR) and pan-p85α antibody were generated as described previously (Ueki et al., (2002) Proc. Natl. Acad. Sci. U.S.A. 99:419-24). Rabbit polyclonal anti-Akt, anti-phospho Akt (S473) anti-phospho GSK3 (Ser9), anti- GSK3, anti-phospho FoxOl (Ser256), anti FoxOl, anti-phospho MAPK, anti-MAPK, anti-phospho JNK, anti- JNK, anti-phospho MKK4, and anti-MKK4 antibodies were purchased from Cell Signaling Technology (Beverly, MA). Anti-phosphoserine 307 IRS-I antibody, cdc42 antibody and phosphotyrosine (pTyr) antibody, 4G10, was purchased from Upstate Biotechnology, Inc. (Lake Placid, NY). Goat polyclonal anti-
Aktl/2 antibody (Akt) was purchased from Santa Cruz Biotechnology, Inc. (Santa Cruz, CA).
In vivo cdc42 activation assay. Mice were anesthetized and injected with 5U of insulin via the portal vein. Three minutes after the injection, the right lobe of the liver was quickly dissected and snap frozen directly into liquid nitrogen.
Approximately 200-300 mg of the liver sample was used to measure cdc42 activity in the livers with a PAKl pulldown assay (Upstate). The kit was used essentially as directed, but with the addition of 10 mM ortho vanadate to the reaction buffer.
Primary Hepatocyte Isolation. Hepatocytes were isolated as described previously (Block et al, (1996). J. Cell. Biol. 132: 1133-49). Briefly, the mice were anesthetized with a 1.2% solution of 2,2,2-tribromoethanol and the portal vein was cannulated with a 24.5 G catheter, and the liver perfused for 15 minutes at a rate of 7 mL/min with calcium-free perfusion buffer. The blanched liver is perfused with collagenase solution (200U/mL) for 10 minutes at 7 mL/min to release hepatocytes from the extracellular matrix. The digested liver was excised and placed in preservation buffer, where the digested cells were gently scraped from the liver sac, washed and purified with Percoll to remove dead cells and enrich the hepatocyte fraction. Typical viabilities are between 85-90%, with cell yields of 1.0-1.5 x 106 cells/g mouse. The isolated hepatocytes are then grown on collagen-coated plates in Advanced DMEM (Gibco) supplemented with glutamine, antibiotic cocktail and 10% FBS.
Statistics. Data are presented as ±s.e.m. Student's t-test was used for statistical analysis between two groups, while statistical significance between multiple treatment groups was determined by analysis of variance (ANOVA) and Tukey's t- test.
Results
Blunted JNK Activation in L-Pik3rlKO Mice Ameliorates Obesity- Induced Diabetes
To study the crosstalk between p85α and JNK, mice with a liver-specific deletion oiPik3rl via the Cre-loxP system (L-Pik3rlKO) were generated as described previously (Postic and Magnuson. (2000) Genesis 26: 149-50). Mice carrying a floxed exon 7, which encodes the N-terminal SH2 domain common to all three transcripts, p85α, p55α and p50α (Luo et al., (2005) MoI. Cell. Biol. 25:491-502), were crossed
with mice carrying the Cre transgene driven by the albumin promoter. Western blots of liver extracts of L-Pik3rl KO mice revealed an 80-90% decrease in p85α and a complete loss of p50α (FIG. 5A), consistent with complete ablation oiPik3rl in hepatocytes. Obesity is a powerful physiologic activator of JNK, particularly in the liver.
To investigate if p85α played a role in activation of JNK in vivo, six-week old L- Pik3rlKO mice were placed on a high fat diet (45% of calories from fat) for a total of eight weeks. At the end of the treatment, both lox/lox and L-Pik3rlKO mice were equally obese (FIG. 5B) and consumed an equal amount of calories (FIG. 12A). Interestingly, while insulin increased JNK activity by three-fold in lean lox/lox animals, L-Pik3rlKO animals showed a 60% reduction in insulin-simulated JNK activation (FIG. 5C). After high-fat feeding, basal and insulin-stimulated JNK activation increased by two-fold in both lox/lox and L-Pik3rlKO animals, but JNK activation in obese knockout animals reached only 50-60% the level observed in obese control animals. The attenuated JNK activity in both lean and obese L- Pik3rlKO mice directly correlated with decreased levels of serine-307 phosphorylation of IRS-I and increased Akt activity compared to controls.
These reductions in stress kinase activation translated to marked improvements in glucose homeostasis. L-Pik3rlKO mice maintained lower fasting blood glucose and fasting serum insulin levels when fed either a high- fat diet or normal chow (FIGs. 12B and 12C). In addition, while obese lox/lox mice were severely glucose intolerant, obese L-Pik3rlKO mice exhibited normal to improved glucose tolerance even when compared against control mice on normal chow (FIG. 5C). Thus, the loss of p85α expression in liver protected against obesity-induced insulin resistance and diabetes.
L-Pik3rlKO Hepatocytes are Resistant to JNK-induced insulin resistance.
To determine if JNK resistance was a cell autonomous effect oiPik3rl deletion, JNKl was overexpressed in primary hepatocytes isolated from lox/lox or L- Pik3rlKO mice using adenovirus-mediate gene transfer. The JNKl isoform was chosen because it is the only one of the three JNK isoforms that has been shown to have a significant role in mediating obesity -related insulin resistance in the liver
(Hirosumi et al, (2002) Nature 420:333-6). Following adenoviral infection, JNKl
was overexpressed by six-fold in both p85α knockout and control primary hepatocytes (FIG. 6). This forced expression of JNKl led to significant increases in JNK phosphorylation and serine phosphorylation of IRS-I in lox/lox hepatocytes. By contrast, an equal level of JNKl overexpression resulted in only a 2-fold increase in cells derived from L-Pik3rl mice (FIG. 6) (p < 0.05 knockout vs. control cells).
Likewise, overexpression of JNK reduced insulin-stimulated Akt phosphorylation in lox/lox hepatocytes by 85%, while the same level of overexpression in L-Pik3rlKO hepatocytes did not significantly reduce insulin stimulated Akt signaling.
The p85α regulatory subunit activates JNK via a cdc42/MKK4 pathway The defects in JNK activation by insulin in the L-Pik3rlKO hepatocytes in vivo and in vitro indicated that the one of the gene products oiPik3rl regulated some effector of this system. One candidate effector is the small GTPase cdc42, which is known to activate both SEK1/MKK4 and JNK (Gallo and Johnson, (2002) Nat. Rev. MoI. Cell. Biol. 3:663-72), and has been shown to interact with p85α, but it was unknown whether this interaction had any functional consequences in vivo (Zheng et al, (1994) J. Biol. Chem. 269: 18727-30). To assess activation of cdc42, the ability of this protein in its activated form to bind to p21 -associated kinase- 1 (PAKl) was utilized. Using tagged or embolized PAKl protein in a pulldown assay, the level of activated cdc42 was significantly reduced in knockout animals (FIG. 7A). This decrease is cdc42 activation correlated with a decrease in MKK4 phosphorylation/activation (FIG. 7B). Since MKK4 is the direct upstream kinase of JNK, this would result in the observed decrease in JNK activation.
To confirm that this cdc42/MKK4/JNK signaling pathways is an intrinsic property of hepatic insulin signaling, primary hepatocytes from L-Pik3rlKO livers and lox/lox controls were infected with constitutively active forms of MKK4 and cdc42 and stimulated with either insulin or saline control (FIG. 7C). Expression of activated MKK4 or cdc42 enhanced JNK phosphorylation, indicating that these enzymes were upstream of this pathway in hepatocytes. Moreover, consistent with the role of JNK as an antagonist of insulin signaling, expression of active cdc42 or MKK4 led to significant reduction in Akt phosphorylation by insulin, indicating that these molecules could play a role in regulating hepatic insulin sensitivity in vivo.
The Negative Effects on Insulin Signaling are Specific to p85α
Since Pϊkirl encodes three different regulatory subunit isoforms, the possibility emerged that the negative regulation of insulin signaling could be isoform specific. To address this possibility, adenovirus-mediated gene transfer was used to reconstitute the livers of L-Pik3rlKO mice with each of the three Pik3rl gene products: p85α, p55α, or p50α. As was previously observed in other cell types, the hepatic reconstitution of L-Pik3rl KO mice with each of the three Pik3rl isoforms restored pTyr-associated PI3K activity to similar extents (FIG. 8B).
Despite the fact the PI3K activity enabled by each of these isoforms was nearly equal, Akt activation was differentially affected by the expression of p85α. Thus, while the expression of control LacZ, p55α, or p50α maintained the elevated Akt activation observed in L-Pik3rlKO mice, the expression of p85α caused a relative decrease in Akt phosphorylation to a level similar to lox/lox controls. In addition, re-expression of p85α specifically restored several mechanisms of negative regulation to L-Pik3rlKO animals. The expression of p85α restored insulin- stimulated JNK activation and levels of IRS-I serine phosphorylation back to levels comparable to lox/lox controls (FIG. 8C).
These changes at the molecular level correlated with changes at the physiologic level. The reconstitution of L-Pik3rlKO mice with p85α, but not its short isoforms, reversed the improvements in fasting blood glucose and fasting serum insulin levels and restored them to the levels of lox/lox controls (FIG. 8D). Moreover, re-expression of p85α produced a relative glucose intolerance in L- Pik3rlKO mice compared to LacZ-treated KO mice (FIG. 8E), such that the glucose excursion curves of p85α-expressing mice were indistinguishable from lox/lox controls.
Potentiation of JNK Activation by p85 Occurs Independently of PI3K Activity
Since insulin can activate small GTPases like Rac and cdc42 by PB K- dependent mechanisms, experiments were performed to determine whether the decreased cdc42/JNK activation in L-Pik3rl merely reflect decreased PI3K activity, or is due specifically to some aspect p85α expression. To this end, the livers of L- Pik3rlKO animals were reconstituted with either WT p85α, or one of two p85α
mutants that are incapable of activating PB K by adenovirus-mediated gene transfer. One mutant substitutes a FLAG tag for the inter-SH2 (ΔiSH2) domain, which mediates the binding and activation of pi 10 (Dhand et al., (1994) EMBO J. 13:511- 21). When overexpressed in cells or in mouse livers, the ΔiSH2 construct has a dominant negative effect (Miyake et al., (2002) J. Clin. Invest. 110: 1483-91). The other p85 mutant contains arginine to alanine substitutions in critical residues in both SH2 domains in the C-terminus (RARA); this mutant is able to bind pi 10, but cannot bind to phosphorylated IRS proteins, which is required for the proper activation and localization of the PI3K holoenzyme (Hill et al., (2001) J. Biol. Chem. 276: 16374-8). Interestingly, JNK activity and serine phosphorylation of IRS-I was restored by either WTp85α or the ΔiSH2 mutant, but not by the RARA mutant (FIG. 9A). This activation of JNK could not have occurred by a PI3K dependent mechanisms, since the ΔiSH2 mutant drastically inhibited total PI3K in these livers, while the re- expression of WT p85a restored PI3K back to levels of lox/lox animals (FIG. 9B). These experiments suggest that p85α expression may thus be the more critical link to JNK activation than the generation of PIP3, at least in the liver. Interestingly, the RARA mutant, which also cannot activate PI3K, was also unable to activate JNK, which indicates that functional SH2 domains are necessary for p85-JNK crosstalk.
To further determine the extent to which PI3K activity is necessary to activate cdc42, the same p85α mutants were expressed in L-Pik3rlKO primary hepatocytes and insulin-stimulated cdc42 activity was measured (FIG. 9C). As with the JNK activity in whole livers, both the WT and the ΔiSH2 versions of p85a fully restored the cdc42 response, despite their dichotomous effects on PI3K activity. On the other hand, the RARA mutant had neutral effects on both PI3K and cdc42 activity. These data correspond with the in vivo JNK data (FIG. 9A) that indicates that full-length p85α with functional SH2 domains is necessary for the insulin-dependent activation of cdc42/JNK, while PI3K activity is not.
These molecular changes in cdc42/JNK corresponded to physiologic changes, where increased JNK activity led to decreased insulin sensitivity. For instance, the expression of either WTp85α and ΔiSH2, which increased cdc42 and JNK activation, led to worsened glucose tolerance compared to L-Pik3rlKO mice treated with control LacZ adenovirus (FIG. 9D), which normally possess heightened insulin sensitivity
(Taniguchi et al., (2006) Cell. Metab. 3:343-53). The ΔiSH2 mutant caused
significant glucose intolerance consistent with diabetes, probably due to the inhibition of the positive effects of PBK in addition to the negative effects of JNK activation. Consistent with the cdc42/JNK data, the RARA p85a mutant had negligible effects on insulin sensitivity.
An intact N-terminus of p85 is required for the activation of cdc42
That only full-length forms of p85α are able to activate JNK and suppress insulin action suggested that unique structural features of p85α may account for the functional differences. All regulatory subunits of Class IA PDK share a common C- terminus, but diverge greatly in the length and composition of their N-termini (Carpenter and Cantley, (1996) Curr. Opin. Cell. Biol. 8: 153-8). While the N-termini of short isoforms p55α and p50α are only 34 and 6 amino acids long, respectively (Antonetti et al, (1996) MoI. Cell. Biol .16:2195-203), Inukai et al, (1996) J. Biol. Chem. 271 :5317-20), the N-terminus of p85α is 339 amino acids long and contains an SH3 domain, two proline rich regions, and a domain homologous to a portion of breakpoint cluster region (bcr) gene product (BH domain).
To investigate whether the negative effects of p85α are specific to one of the domains in the N-terminus, adenoviral p85 constructs were created which substitute a FLAG tag for either the -80 amino acid SH3 domain (ΔSH3) or the -170 amindacid BH domain (ΔBH), which effectively deleted the domain while providing an epitope tag for easy detection by Western. One construct with a combination deletion of both the SH3 domain and inter-SH2 domain (ΔΔp85) was also created to serve as a control for PI3K activity (FIG. 10A). Primary hepatocytes from L-Pik3rlKO mice were infected with ΔSH3, ΔBH, ΔΔp85 adenoviruses and total insulin-stimulated (pTyr- associated) PI3K and cdc42 activity were measured. The loss of either the SH3 or BH domain from the N-terminus of p85α did not affect the ability of p85 to rescue PI3K activity in L-Pik3rlKO hepatocytes (FIG. 10B). On the other hand, the control ΔΔp85 adenovirus, which lacks the pi 10 binding region in the C-terminus fully inhibited PI3K activation (FIG. 10B). However, despite normal PI3K activity in the cells infected with ΔSH3 and ΔBH, cdc42 activity was significantly ablated (FIG. 10C). This abrogation of cdc42 activity also occurred in the ΔΔp85 adenovirus cells, which indicates that a fully intact N-terminus of p85 is also necessary for the activation of cdc42. These data support the notion that the expression so some aspect
of the N-terminus of p85, and not PDK activity, is required for the insulin-stimulated activation of cdc42/JNK.
Discussion
Selective ablation oiPik3rl in the liver improves insulin sensitivity and protects mice against obesity-induced insulin resistance. These physiologic changes appear to be due, at least in part, to a diminution of JNK activation and a decrease in the serine phosphorylation of IRS- 1 normally observed in obese insulin resistant states. Thus, obese L-Pik3rlKO mice remained insulin sensitive compared to obese controls due to abrogated JNK activity. Moreover, even in the face of forced overexpression of JNKl, L-Pik3rlKO hepatocytes exhibit decreased JNKl phosphorylation/activity and remain fully capable of activating Akt, while control cells exhibit significant defects in Akt activation. The activation of JNK, particularly in the liver, has been shown previously to be a major mediator of the insulin resistance that occurs in obesity (Ozcan et al., (2004) Science 306:457-61). Consequently, one of the mechanisms by which p85α suppresses insulin action in vivo may occur through the JNK-mediated negative feedback on insulin signaling.
These results are similar to those observed in the whole-body knockout of
JNKl (Hirosumi et al., (2002) Nature 420:333-6), except that that latter was against both insulin resistance and obesity of HFD feeding, whereas in L-Pik3rlKO mice improved insulin sensitivity occurs without protectect against HFD-induced obesity. These data suggest that the chief disorders of obesity — weight gain and insulin resistance — have physiologically distinct mechanisms, where the liver plays a critical role in maintaining insulin sensitivity, even in the face of massive obesity. On the other hand, the mechanisms that control body weight and food intake must lie in other tissues, mostly likely in the central nervous system (Howard et al., (2004) Nat. Med. 10:734-8).
The data of this study also directly identify p85α as an important regulator of cdc42 and JNK activity in vivo, as L-Pik3rlKO mice exhibited a three-fold decrease in insulin-stimulated cdc42 activity, which correlates with a -75% decrease in the activity of downstream kinases MKK4 and JNK. Conversely, the expression of constitutively active forms of cdc42 or MKK4 leads to marked upregulation in JNK activity in Pik3rlKO hepatocytes. The possibility that MKK7, which is similar in structure to MKK4 and also activates JNK (Gallo and Johnson, (2002) Nat. Rev. MoI.
Cell. Biol. 3:663-72), may also contribute to the effects of p85 cannot be excluded, since no good antibodies exist to activated forms of MKK7. Thus, in the methods described herein, MKK7 may be substituted for MKK4.
While the exact mechanism by which p85α activates cdc42 and JNK needs further study, the current work has revealed several interesting aspects of this regulation. First, JNK activity is restored by p85α, but not by p55α and p50α, indicating that the activation of cdc42 and JNK is dependent on some particular property of p85α as compared to the shorter regulatory subunits. As described herein, this effect of p85α is not dependent upon PI3K activation, since dominant negative forms of p85 can fully activate cdc42 and JNK, while inhibiting PI3K. On the other hand, deletion of either the SH3 domain or BH domain from the N-terminus is sufficient to ablate the ability of p85 to activate cdc42, while fully maintaining PI3K activity (FIG. 1OB and Beeton et al, (1999) MoI. Cell Biol. Res. Commun. 1 : 153-7). Interestingly, the functional inactivation of the SH2 domains in p85 also rendered it unable to activate either PI3K or cdc42/JNK, suggesting that the proper localization in the cell or within a complex may be required for JNK activation. Thus, the data define the minimal requirements for the activation of JNK by p85 as a fully intact N- terminus of p85 and functional SH2 domains.
The data of the current study are consistent with other reports that suggest that the N-terminus of p85 may have unique properties apart from its role as a component of PI3K holoenzyme. Notably, p85 has been demonstrated as an essential activator of small GTPases such as cdc42 or Rac (reviewed in Burridge and Wennerberg, (2004) Cell 116: 167-79) that mediate PDGF or EGF-induced cytoskeletal changes, such as membrane ruffling or stress fiber disassembly (Brachmann et al., (2005) MoI. Cell. Biol. 25:2593-606; Hill et al., (2001) J. Biol. Chem. 276: 16374-8). Interestingly, consistent with these findings, two other studies have found that p85 can potentiate small GTPases like cdc42 even in the presence of wortmannin (Jimenez et al., (2000) J. Cell. Biol. 151 :249-62, Kang et al., (2002) J. Biol. Chem. 277:912-21). Furthermore, in the study by Hill, et al (Hill et al., (2001) J. Biol. Chem. 276: 16374- 8), it appeared that while wild-type p85 could activate JNK, deletions in either the N- terminus or SH2 domains rendered p85 ineffective in activating JNK.
The mechanism by which the N-terminus of p85 activates cdc42, while still unknown, likely involves specific roles for each of the N-terminal domains (FIG. 11
and Okkenhaug and Vanhaesebroeck. (2001) Sci. STKE 2001:PEl). The BH domain is similar in structure to the Rho-GTPase activating protein (GAP) domain of the breakpoint cluster region (bcr) protein (Musacchio et al., (1996) Proc. Natl. Acad. Sci. U.S.A. 93: 14373-8), and can bind activated cdc42, but has no intrinsic GAP activity because it lacks certain conserved residues in the switch domains (Fidyk and Cerione, (2002) Biochemistry 41: 15644-53). The data also indicate that the SH3 and SH2 domains must also be involved in the activation of since deletion of either of these domains abrogated cdc42 activation (FIG. 11). These domains could function to recruit a cdc42 guanine nucleotide exchange factor (GEF), thereby providing a positive feedback loop for further activation of cdc42 protein, and ultimately, JNK (Bertagnolo et al., (2004) Cell Signal. 16:423-33). In addition, these domains might be responsible for the proper intracellular localization of the cdc42/JNK-activating complex. The p85 subunit has been found to form insulin-dependent protein aggregates that do not generate PIP3 (Luo et al., (2005) J. Cell. Biol. 170(3):455-64). These aggregates were proposed as sequestration complexes, but an alternate interpretation is that these complexes could be active negative regulatory complexes that activate cdc42 and JNK. Interestingly, these complexes do not form with RARA p85 or with the shorter p55α or p50α isoforms, suggesting a similar requirement of an intact N-terminus and functional SH2 domains for full negative regulation by p85 (Luo et al., (2005) J. Cell. Biol. 170(3):455-64).
The apparent paradox of the improved insulin action afforded by p85 deletion may be partially resolved by the finding that p85 may regulate the activity of PTEN in vivo. L-Pik3rlKO mice displayed decreased PIP3 turnover during prolonged insulin stimulation (FIG. 8A). PIP3 levels were also elevated in a previous germline knockout of p85α but it was unclear whether the increase was due to an alteration in PIP3 turnover or increased kinase activity. That previous study attributed the elevated PIP3 levels to the upregulation of the short isoforms, p55α and p50α (Dhand et al., (1994) EMBO J. 13:511-21). This mechanism, however, cannot account for the elevated PIP3 levels observed in L-Pik3rlKO mice, since deletion oiPϊkirl ablates both p55α and p50αin addition to p85α. Exactly how p85α might regulate PTEN is not known, though it is possible that p85α could modulate PTEN indirectly by altering some post-translational modification of PTEN or possibly its subcellular localization.
Taken together, these data demonstrate a unique role for the p85α regulatory subunit in insulin signaling and the physiologic regulation of glucose homeostasis. Although p85 is an essential part of the PDK heterodimer, it also plays a novel role in regulating a cdc42/MKK4/JNK pathway that suppresses insulin action in both lean and obese mice. These mechanisms not only provide a level of internal negative feedback on this critical node (Taniguchi et al, (2006) Nat. Rev. MoI. Cell. Biol. 7:85-96) in insulin and growth factor signaling, but also allow crosstalk between the PBK signaling pathway and the stress or inflammatory responses, thus creating an important connection that could have broad impact in the basic understanding of cell growth and metabolism. This powerful link between p85α and JNK activation might also represent an exciting new therapeutic intervention into type 2 diabetes.
Additional Reference:
Ueki et al., (2003) J. Biol. Chem. 278(48):48453-66
Example 3; JNK phosphorylates PTEN Since JNK activity is decreased in p85α KO mice and cells, it is possible that a decrease in JNK could also result in a change in serine phosphorylation and activity of PTEN.
To begin to assess this possibility, an in vitro kinase assay was performed using 100 ng GST-PTEN fusion protein (Upstate Biotechnology) and activated JNK prepared by immunoprecipitating cell lysate from insulin-stimulated brown adipocytes with JNK antibody or control IgG in the presence of 5 μCi of [γ-32P]-ATP. PTEN Activity Assay: Phosphatidylinositol (Avanti Polar Lipids) was in vitro- phosphorylated to form phosphatidylinositol 3 -phosphate (PI-3-P) by recombinant PI3K (Upstate) with [gamma-32P]ATP. The phosphorylated lipid was extracted with 1: 1 methanolxhloroform, dried under nitrogen gas, reconstituted into PTEN assay buffer (10mMTris-HCl/25mMNaCl, pH 7.5), and incubated with PTEN immunoprecipitates from lox/lox or L-Pik3rlKO liver lysates (Miller et al., FEBS Lett. 528:145-153 (2005)). The PI-3-P was re-extracted with methanokchloroform and run on a TLC plate with n-propanol:2 M acetic acid (65:35). The intensities of PI-3-P spots were quantified with NIH IMAGE (rsb.info.nih.gov/nih-image). PTEN activity was determined by decreased intensity of the PI-3-P spot relative to IgG immunoprecipitates from lox/lox livers. Relative PTEN activity was determined by normalizing PTEN activity to lox/lox livers without insulin stimulation.
SDS-PAGE and autoradiography revealed that JNK was able to phosphorylate PTEN in vitro (FIG. 14). To further assess this phosphorylation of PTEN by JNK, mutants of PTEN were generated with substitution of the two potential JNK phosphorylation sites (PTEN-S338A, and PTEN-T366A). These mutants, as well as wild-type PTEN, were then subcloned into pCMV-Tag2 vector that introduced an N- terminal FLAG tag. 5 μg of each construct was transfected into COS7 cells and the activities of Akt and p70S6 kinase following IGF-I stimulation was evaluated. In cells expressing these mutants, two downstream targets of the PI 3-kinase pathway (Akt and p70S6 kinase) were maintained at higher levels of activity than observed in wild-type control cells.
These data suggest that insulin-dependent JNK activation via p85-mediated signaling may result in the increased phosphorylation of PTEN, thereby enhancing its lipid phosphatase activity and/or increasing its stability. These data also indicate that drugs which target the PTEN kinase and reduce phosphorylation of PTEN should also reduce its activity, thereby potentiating insulin action and improving insulin sensitivity.
Additional Reference:
Taniguchi et al, Proc. Natl. Acad. Sci. U.S.A. 103(32)12093-12097 (2006)
OTHER EMBODIMENTS It is to be understood that while the invention has been described in conjunction with the detailed description thereof, the foregoing description is intended to illustrate and not limit the scope of the invention, which is defined by the scope of the appended claims. Other aspects, advantages, and modifications are within the scope of the following claims.
Claims
1. A method of identifying a candidate compound for improving insulin sensitivity in a mammal in need thereof, the method comprising: providing a sample comprising p85α and cdc42; contacting the sample with a test compound, and evaluating binding of p85α to cdc42 in the sample, wherein a test compound that decreases binding of p85α to cdc42 is a candidate compound for improving insulin sensitivity in a mammal.
2. A method of identifying a candidate compound for improving insulin sensitivity in a mammal in need thereof, the method comprising: providing a sample comprising cdc42 and MLK3; contacting the sample with a test compound, and evaluating phosphorylation of MLK3 by cdc42 in the sample, wherein a test compound that decreases phosphorylation of MLK3 by cdc42 is a candidate compound for improving insulin sensitivity in a mammal.
3. A method of identifying a candidate compound for improving insulin sensitivity in a mammal in need thereof, the method comprising: providing a sample comprising MLK3 and MKK4; contacting the sample with a test compound, and evaluating phosphorylation of MKK4 by MLK3 in the sample, wherein a test compound that decreases phosphorylation of MKK4 by MLK3 is a candidate compound for improving insulin sensitivity in a mammal.
4. A method of identifying a candidate compound for improving insulin sensitivity in a mammal in need thereof, the method comprising: providing a sample comprising MKK4 and JNK; contacting the sample with a test compound, and evaluating phosphorylation of JNK by MKK4 in the sample, wherein a test compound that decreases phosphorylation of JNK by MKK4 is a candidate compound for improving insulin sensitivity in a mammal.
5. A method of identifying a candidate compound for improving insulin sensitivity in a mammal in need thereof, the method comprising: providing a sample comprising PTEN and JNK; contacting the sample with a test compound, and evaluating phosphorylation of PTEN by JNK in the sample, wherein a test compound that decreases phosphorylation of PTEN by JNK is a candidate compound for improving insulin sensitivity in a mammal.
6. A method of identifying a candidate compound for improving insulin sensitivity in a mammal in need thereof, the method comprising: providing a sample comprising one or more target proteins selected from the group consisting of cdc42, MLK3, or MKK4; contacting the sample with a test compound; evaluating binding of the test compounds to the target protein; and selecting the test compound as a candidate compound if it binds to the target protein.
7. The method of claim 6, further comprising: providing a cell having a functional insulin signalling pathway comprising p85α, cdc42, MLK3, MKK4, and JNK; contacting the cell with the candidate compound; contacting the cell with an amount of insulin sufficient to activate said pathway; evaluating activation of said pathway in the cell in the presence of the test compound; comparing activation of said pathway in the cell in the presence of the test compound to a reference representing activation of said pathway in the cell in the absence of the test compound, and selecting the candidate compound as a candidate therapeutic agent for improving insulin sensitivity in a mammal if activation of said pathway is reduced in the presence of the test compound as compared to activation of said pathway in the absence of the test compound.
8. The method of claim 7, wherein activation of said pathway is determined by one or more of detecting cdc42 activation of MLK3; detecting binding of cdc42 to MLK3; detecting MLK3 kinase activity; detecting phosphorylation of MKK4; or detecting JNK activation.
9. The method of any of claims 1-6, wherein the test compound is a small molecule.
10. The method of any of claims 1-6, wherein the test compound is a peptide or peptidomimetic.
11. The method of any of claims 1-6, wherein the sample comprises a cell.
12. The method of any of claims 1-6, further comprising administering the candidate compound to a mammal, and evaluating whether the candidate compound increases insulin sensitivity in the mammal.
13. The method of claim 12, wherein the mammal is in need of increased insulin sensitivity.
14. The method of claim 12, wherein the mammal is a non-human experimental animal.
15. The method of claim 12, further comprising selecting the compound if is increases insulin sensitivity and evaluating the compound in a clinical trial.
Applications Claiming Priority (2)
| Application Number | Priority Date | Filing Date | Title |
|---|---|---|---|
| US82111806P | 2006-08-01 | 2006-08-01 | |
| US60/821,118 | 2006-08-01 |
Publications (2)
| Publication Number | Publication Date |
|---|---|
| WO2008016995A2 true WO2008016995A2 (en) | 2008-02-07 |
| WO2008016995A3 WO2008016995A3 (en) | 2008-11-27 |
Family
ID=38997846
Family Applications (1)
| Application Number | Title | Priority Date | Filing Date |
|---|---|---|---|
| PCT/US2007/075001 WO2008016995A2 (en) | 2006-08-01 | 2007-08-01 | Methods of identifying modulators of insulin signalling |
Country Status (1)
| Country | Link |
|---|---|
| WO (1) | WO2008016995A2 (en) |
Family Cites Families (1)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| WO2002085396A1 (en) * | 2001-04-24 | 2002-10-31 | President And Fellows Of Harvard College | Inhibition of jun kinase |
-
2007
- 2007-08-01 WO PCT/US2007/075001 patent/WO2008016995A2/en active Application Filing
Also Published As
| Publication number | Publication date |
|---|---|
| WO2008016995A3 (en) | 2008-11-27 |
Similar Documents
| Publication | Publication Date | Title |
|---|---|---|
| Li et al. | Atrogin-1/muscle atrophy F-box inhibits calcineurin-dependent cardiac hypertrophy by participating in an SCF ubiquitin ligase complex | |
| Sanders et al. | Defining the mechanism of activation of AMP-activated protein kinase by the small molecule A-769662, a member of the thienopyridone family | |
| Mauvais-Jarvis et al. | Reduced expression of the murine p85α subunit of phosphoinositide 3-kinase improves insulin signaling and ameliorates diabetes | |
| Fayard et al. | Protein kinase B (PKB/Akt), a key mediator of the PI3K signaling pathway | |
| Egawa et al. | Membrane-targeted phosphatidylinositol 3-kinase mimics insulin actions and induces a state of cellular insulin resistance | |
| EP3072555B1 (en) | Mk2/3 inhibitors to treat metabolic disturbances of obesity | |
| Vantler et al. | Systematic evaluation of anti-apoptotic growth factor signaling in vascular smooth muscle cells: only phosphatidylinositol 3′-kinase is important | |
| US10226514B2 (en) | Methods and compositions for reducing blood glucose levels | |
| Straubinger et al. | Amplified pathogenic actions of angiotensin II in cysteine‐rich LIM‐only protein 4–negative mouse hearts | |
| EP1539177A2 (en) | Compositions and methods for treating heart disease | |
| AU2013290266B2 (en) | Method and composition for metabolic regulation | |
| US20140234321A1 (en) | Hunk, a snf1-related kinase essential for mammary tumor metastasis | |
| WO2008016995A2 (en) | Methods of identifying modulators of insulin signalling | |
| EP1658504B1 (en) | Inhibition of s6 kinase activity for the treatment of insulin resistance | |
| WO2004092395A2 (en) | Modulators of telomere stability | |
| Qu et al. | Knockout of cardiac troponin I‐interacting kinase leads to cardiac dysfunction and remodelling | |
| Baumgartener | SHIP2: an emerging target for the treatment of type 2 diabetes mellitus | |
| US20070053910A1 (en) | Modulation of s6 kinase activity for the treatment of obesity | |
| Lebeche et al. | Transgenic models of heart failure: elucidation of the molecular mechanisms of heart disease | |
| Quon et al. | Tyr 612 and Tyr 632 in Human Insulin Receptor Substrate-1 Are Important for Full Activation of Insulin-Stimulated Phosphatidylinositol 3-Kinase Activity and Translocation of GLUT4 in Adipose Cells 1 | |
| EP1556505A1 (en) | Modulation of s6 kinase activity for the treatment of obestiy | |
| Buch | Identifying a Functional Interaction between the Plasma Membrane Calcium ATPase Pump and Calcineurin-A Potential Role in Cardiac Hypertrophy | |
| Taniguchi et al. | The p85α Regulatory Subunit of Phosphoinositide 3-Kinase 3 Potentiates JNK-mediated Insulin Resistance 4 | |
| Colomer et al. | The Roles of CaMKII in the Genesis of Cardiac Hypertrophy | |
| Taniguchi | The dissection of insulin signaling isoforms in vivo: Molecular mechanisms of hepatic insulin action |
Legal Events
| Date | Code | Title | Description |
|---|---|---|---|
| 121 | Ep: the epo has been informed by wipo that ep was designated in this application |
Ref document number: 07840640 Country of ref document: EP Kind code of ref document: A2 |
|
| NENP | Non-entry into the national phase |
Ref country code: DE |
|
| NENP | Non-entry into the national phase |
Ref country code: RU |
|
| 122 | Ep: pct application non-entry in european phase |
Ref document number: 07840640 Country of ref document: EP Kind code of ref document: A2 |