WO2011072132A1 - Procédés de traitement d'états inflammatoires - Google Patents
Procédés de traitement d'états inflammatoires Download PDFInfo
- Publication number
- WO2011072132A1 WO2011072132A1 PCT/US2010/059699 US2010059699W WO2011072132A1 WO 2011072132 A1 WO2011072132 A1 WO 2011072132A1 US 2010059699 W US2010059699 W US 2010059699W WO 2011072132 A1 WO2011072132 A1 WO 2011072132A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- gpr120
- arrestin2
- dsrna
- compound
- cells
- Prior art date
Links
- 238000000034 method Methods 0.000 title claims abstract description 130
- 230000004968 inflammatory condition Effects 0.000 title claims description 24
- 102100040134 Free fatty acid receptor 4 Human genes 0.000 claims abstract description 270
- 101000890672 Homo sapiens Free fatty acid receptor 4 Proteins 0.000 claims abstract description 270
- 150000001875 compounds Chemical class 0.000 claims abstract description 87
- 230000001419 dependent effect Effects 0.000 claims abstract description 45
- 230000001404 mediated effect Effects 0.000 claims abstract description 39
- 230000019491 signal transduction Effects 0.000 claims abstract description 34
- 108010032967 beta-Arrestin 2 Proteins 0.000 claims abstract description 33
- 206010061218 Inflammation Diseases 0.000 claims abstract description 29
- 230000004054 inflammatory process Effects 0.000 claims abstract description 26
- 208000008589 Obesity Diseases 0.000 claims abstract description 25
- 235000020824 obesity Nutrition 0.000 claims abstract description 25
- 230000004044 response Effects 0.000 claims abstract description 23
- 206010012601 diabetes mellitus Diseases 0.000 claims abstract description 12
- 108091032973 (ribonucleotides)n+m Proteins 0.000 claims description 185
- 102000040650 (ribonucleotides)n+m Human genes 0.000 claims description 155
- 210000004027 cell Anatomy 0.000 claims description 129
- 235000014113 dietary fatty acids Nutrition 0.000 claims description 61
- 229930195729 fatty acid Natural products 0.000 claims description 61
- 239000000194 fatty acid Substances 0.000 claims description 61
- 150000004665 fatty acids Chemical class 0.000 claims description 60
- 230000014509 gene expression Effects 0.000 claims description 60
- 230000000694 effects Effects 0.000 claims description 59
- 108090000623 proteins and genes Proteins 0.000 claims description 50
- 230000000295 complement effect Effects 0.000 claims description 45
- 241001465754 Metazoa Species 0.000 claims description 42
- 239000003795 chemical substances by application Substances 0.000 claims description 38
- 230000000692 anti-sense effect Effects 0.000 claims description 35
- 108020004999 messenger RNA Proteins 0.000 claims description 32
- 239000002158 endotoxin Substances 0.000 claims description 31
- 229920006008 lipopolysaccharide Polymers 0.000 claims description 30
- 102100029648 Beta-arrestin-2 Human genes 0.000 claims description 28
- 235000020660 omega-3 fatty acid Nutrition 0.000 claims description 27
- 230000006461 physiological response Effects 0.000 claims description 27
- 108060008682 Tumor Necrosis Factor Proteins 0.000 claims description 26
- 102100040247 Tumor necrosis factor Human genes 0.000 claims description 26
- 239000003446 ligand Substances 0.000 claims description 25
- 230000028327 secretion Effects 0.000 claims description 21
- 230000027455 binding Effects 0.000 claims description 20
- 102000004127 Cytokines Human genes 0.000 claims description 18
- 108090000695 Cytokines Proteins 0.000 claims description 18
- 230000028709 inflammatory response Effects 0.000 claims description 18
- 230000002401 inhibitory effect Effects 0.000 claims description 18
- 238000012216 screening Methods 0.000 claims description 18
- 102000004169 proteins and genes Human genes 0.000 claims description 16
- 108091081021 Sense strand Proteins 0.000 claims description 14
- 230000003213 activating effect Effects 0.000 claims description 11
- 210000001616 monocyte Anatomy 0.000 claims description 7
- 229940012843 omega-3 fatty acid Drugs 0.000 claims description 5
- CYQFCXCEBYINGO-IAGOWNOFSA-N delta1-THC Chemical compound C1=C(C)CC[C@H]2C(C)(C)OC3=CC(CCCCC)=CC(O)=C3[C@@H]21 CYQFCXCEBYINGO-IAGOWNOFSA-N 0.000 claims description 3
- 102000007379 beta-Arrestin 2 Human genes 0.000 abstract 4
- NOESYZHRGYRDHS-UHFFFAOYSA-N insulin Chemical compound N1C(=O)C(NC(=O)C(CCC(N)=O)NC(=O)C(CCC(O)=O)NC(=O)C(C(C)C)NC(=O)C(NC(=O)CN)C(C)CC)CSSCC(C(NC(CO)C(=O)NC(CC(C)C)C(=O)NC(CC=2C=CC(O)=CC=2)C(=O)NC(CCC(N)=O)C(=O)NC(CC(C)C)C(=O)NC(CCC(O)=O)C(=O)NC(CC(N)=O)C(=O)NC(CC=2C=CC(O)=CC=2)C(=O)NC(CSSCC(NC(=O)C(C(C)C)NC(=O)C(CC(C)C)NC(=O)C(CC=2C=CC(O)=CC=2)NC(=O)C(CC(C)C)NC(=O)C(C)NC(=O)C(CCC(O)=O)NC(=O)C(C(C)C)NC(=O)C(CC(C)C)NC(=O)C(CC=2NC=NC=2)NC(=O)C(CO)NC(=O)CNC2=O)C(=O)NCC(=O)NC(CCC(O)=O)C(=O)NC(CCCNC(N)=N)C(=O)NCC(=O)NC(CC=3C=CC=CC=3)C(=O)NC(CC=3C=CC=CC=3)C(=O)NC(CC=3C=CC(O)=CC=3)C(=O)NC(C(C)O)C(=O)N3C(CCC3)C(=O)NC(CCCCN)C(=O)NC(C)C(O)=O)C(=O)NC(CC(N)=O)C(O)=O)=O)NC(=O)C(C(C)CC)NC(=O)C(CO)NC(=O)C(C(C)O)NC(=O)C1CSSCC2NC(=O)C(CC(C)C)NC(=O)C(NC(=O)C(CCC(N)=O)NC(=O)C(CC(N)=O)NC(=O)C(NC(=O)C(N)CC=1C=CC=CC=1)C(C)C)CC1=CN=CN1 NOESYZHRGYRDHS-UHFFFAOYSA-N 0.000 description 98
- 241000699670 Mus sp. Species 0.000 description 71
- 125000003729 nucleotide group Chemical group 0.000 description 71
- 239000002773 nucleotide Substances 0.000 description 70
- 235000009200 high fat diet Nutrition 0.000 description 61
- 102000004877 Insulin Human genes 0.000 description 49
- 108090001061 Insulin Proteins 0.000 description 49
- 229940125396 insulin Drugs 0.000 description 49
- 210000001789 adipocyte Anatomy 0.000 description 40
- 230000003110 anti-inflammatory effect Effects 0.000 description 37
- 238000011282 treatment Methods 0.000 description 36
- 108020004459 Small interfering RNA Proteins 0.000 description 35
- 210000002540 macrophage Anatomy 0.000 description 31
- 102000003688 G-Protein-Coupled Receptors Human genes 0.000 description 29
- 108090000045 G-Protein-Coupled Receptors Proteins 0.000 description 29
- 235000005911 diet Nutrition 0.000 description 29
- 230000037213 diet Effects 0.000 description 28
- 238000002474 experimental method Methods 0.000 description 28
- 206010022489 Insulin Resistance Diseases 0.000 description 27
- 210000000577 adipose tissue Anatomy 0.000 description 27
- 108010080367 beta-Arrestins Proteins 0.000 description 27
- 230000004913 activation Effects 0.000 description 23
- 102000000072 beta-Arrestins Human genes 0.000 description 22
- 230000000638 stimulation Effects 0.000 description 22
- 230000007423 decrease Effects 0.000 description 21
- 230000003247 decreasing effect Effects 0.000 description 21
- 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 20
- 108010029223 MAP kinase kinase kinase 7 Proteins 0.000 description 19
- 102100026888 Mitogen-activated protein kinase kinase kinase 7 Human genes 0.000 description 19
- 238000012228 RNA interference-mediated gene silencing Methods 0.000 description 19
- 230000009368 gene silencing by RNA Effects 0.000 description 19
- 239000008103 glucose Substances 0.000 description 18
- 208000001072 type 2 diabetes mellitus Diseases 0.000 description 18
- 238000001727 in vivo Methods 0.000 description 17
- 230000037361 pathway Effects 0.000 description 17
- 238000003556 assay Methods 0.000 description 16
- 102000005962 receptors Human genes 0.000 description 16
- 108020003175 receptors Proteins 0.000 description 16
- 230000011664 signaling Effects 0.000 description 16
- FAPWRFPIFSIZLT-UHFFFAOYSA-M Sodium chloride Chemical compound [Na+].[Cl-] FAPWRFPIFSIZLT-UHFFFAOYSA-M 0.000 description 15
- 238000010322 bone marrow transplantation Methods 0.000 description 15
- -1 for example Proteins 0.000 description 15
- 230000006870 function Effects 0.000 description 15
- 150000002632 lipids Chemical class 0.000 description 15
- 230000026731 phosphorylation Effects 0.000 description 15
- 238000006366 phosphorylation reaction Methods 0.000 description 15
- 210000001519 tissue Anatomy 0.000 description 15
- 102000058061 Glucose Transporter Type 4 Human genes 0.000 description 14
- 108091006300 SLC2A4 Proteins 0.000 description 14
- OPTASPLRGRRNAP-UHFFFAOYSA-N cytosine Chemical compound NC=1C=CNC(=O)N=1 OPTASPLRGRRNAP-UHFFFAOYSA-N 0.000 description 14
- 230000002757 inflammatory effect Effects 0.000 description 14
- 230000005764 inhibitory process Effects 0.000 description 14
- 230000000770 proinflammatory effect Effects 0.000 description 14
- 239000013598 vector Substances 0.000 description 14
- 210000004369 blood Anatomy 0.000 description 13
- 239000008280 blood Substances 0.000 description 13
- UYTPUPDQBNUYGX-UHFFFAOYSA-N guanine Chemical compound O=C1NC(N)=NC2=C1N=CN2 UYTPUPDQBNUYGX-UHFFFAOYSA-N 0.000 description 13
- 230000002440 hepatic effect Effects 0.000 description 13
- 108091034117 Oligonucleotide Proteins 0.000 description 12
- 230000001965 increasing effect Effects 0.000 description 12
- 230000001235 sensitizing effect Effects 0.000 description 12
- 102100021228 TGF-beta-activated kinase 1 and MAP3K7-binding protein 1 Human genes 0.000 description 11
- ISAKRJDGNUQOIC-UHFFFAOYSA-N Uracil Chemical compound O=C1C=CNC(=O)N1 ISAKRJDGNUQOIC-UHFFFAOYSA-N 0.000 description 11
- 210000004185 liver Anatomy 0.000 description 11
- 239000011780 sodium chloride Substances 0.000 description 11
- 238000001890 transfection Methods 0.000 description 11
- 108091023037 Aptamer Proteins 0.000 description 10
- 101000674731 Homo sapiens TGF-beta-activated kinase 1 and MAP3K7-binding protein 1 Proteins 0.000 description 10
- 239000000556 agonist Substances 0.000 description 10
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 description 10
- 230000003993 interaction Effects 0.000 description 10
- 230000007246 mechanism Effects 0.000 description 10
- 230000004048 modification Effects 0.000 description 10
- 238000012986 modification Methods 0.000 description 10
- 150000007523 nucleic acids Chemical class 0.000 description 10
- 238000003753 real-time PCR Methods 0.000 description 10
- 230000005945 translocation Effects 0.000 description 10
- 239000013603 viral vector Substances 0.000 description 10
- 238000001262 western blot Methods 0.000 description 10
- 101000831567 Homo sapiens Toll-like receptor 2 Proteins 0.000 description 9
- 241000699666 Mus <mouse, genus> Species 0.000 description 9
- 102100024333 Toll-like receptor 2 Human genes 0.000 description 9
- 238000004458 analytical method Methods 0.000 description 9
- 230000004190 glucose uptake Effects 0.000 description 9
- 102000039446 nucleic acids Human genes 0.000 description 9
- 108020004707 nucleic acids Proteins 0.000 description 9
- 230000004936 stimulating effect Effects 0.000 description 9
- PEDCQBHIVMGVHV-UHFFFAOYSA-N Glycerine Chemical compound OCC(O)CO PEDCQBHIVMGVHV-UHFFFAOYSA-N 0.000 description 8
- 101001047515 Homo sapiens Lethal(2) giant larvae protein homolog 1 Proteins 0.000 description 8
- 102100022956 Lethal(2) giant larvae protein homolog 1 Human genes 0.000 description 8
- 239000002253 acid Substances 0.000 description 8
- 125000000217 alkyl group Chemical group 0.000 description 8
- 230000004071 biological effect Effects 0.000 description 8
- 239000003636 conditioned culture medium Substances 0.000 description 8
- 201000010099 disease Diseases 0.000 description 8
- 230000006377 glucose transport Effects 0.000 description 8
- 239000005090 green fluorescent protein Substances 0.000 description 8
- 125000001570 methylene group Chemical group [H]C([H])([*:1])[*:2] 0.000 description 8
- 210000003205 muscle Anatomy 0.000 description 8
- 238000002360 preparation method Methods 0.000 description 8
- 150000003839 salts Chemical class 0.000 description 8
- 230000009469 supplementation Effects 0.000 description 8
- RWQNBRDOKXIBIV-UHFFFAOYSA-N thymine Chemical compound CC1=CNC(=O)NC1=O RWQNBRDOKXIBIV-UHFFFAOYSA-N 0.000 description 8
- VRYALKFFQXWPIH-PBXRRBTRSA-N (3r,4s,5r)-3,4,5,6-tetrahydroxyhexanal Chemical compound OC[C@@H](O)[C@@H](O)[C@H](O)CC=O VRYALKFFQXWPIH-PBXRRBTRSA-N 0.000 description 7
- 229930024421 Adenine Natural products 0.000 description 7
- 102000004889 Interleukin-6 Human genes 0.000 description 7
- 108090001005 Interleukin-6 Proteins 0.000 description 7
- 229960000643 adenine Drugs 0.000 description 7
- 210000004982 adipose tissue macrophage Anatomy 0.000 description 7
- PMMURAAUARKVCB-UHFFFAOYSA-N alpha-D-ara-dHexp Natural products OCC1OC(O)CC(O)C1O PMMURAAUARKVCB-UHFFFAOYSA-N 0.000 description 7
- 229940104302 cytosine Drugs 0.000 description 7
- 238000012217 deletion Methods 0.000 description 7
- 230000037430 deletion Effects 0.000 description 7
- 239000003814 drug Substances 0.000 description 7
- 235000019197 fats Nutrition 0.000 description 7
- 238000007446 glucose tolerance test Methods 0.000 description 7
- 238000000338 in vitro Methods 0.000 description 7
- 230000003834 intracellular effect Effects 0.000 description 7
- 102000040430 polynucleotide Human genes 0.000 description 7
- 108091033319 polynucleotide Proteins 0.000 description 7
- 239000002157 polynucleotide Substances 0.000 description 7
- 238000011160 research Methods 0.000 description 7
- 239000000243 solution Substances 0.000 description 7
- 238000009482 thermal adhesion granulation Methods 0.000 description 7
- 229940035893 uracil Drugs 0.000 description 7
- 102100033409 40S ribosomal protein S3 Human genes 0.000 description 6
- IJGRMHOSHXDMSA-UHFFFAOYSA-N Atomic nitrogen Chemical compound N#N IJGRMHOSHXDMSA-UHFFFAOYSA-N 0.000 description 6
- 101710155857 C-C motif chemokine 2 Proteins 0.000 description 6
- 102000000018 Chemokine CCL2 Human genes 0.000 description 6
- 238000002965 ELISA Methods 0.000 description 6
- OKKJLVBELUTLKV-UHFFFAOYSA-N Methanol Chemical compound OC OKKJLVBELUTLKV-UHFFFAOYSA-N 0.000 description 6
- 108091008611 Protein Kinase B Proteins 0.000 description 6
- 108091027967 Small hairpin RNA Proteins 0.000 description 6
- 102000002689 Toll-like receptor Human genes 0.000 description 6
- 108020000411 Toll-like receptor Proteins 0.000 description 6
- 150000007513 acids Chemical class 0.000 description 6
- 210000001185 bone marrow Anatomy 0.000 description 6
- 239000000872 buffer Substances 0.000 description 6
- 230000001413 cellular effect Effects 0.000 description 6
- 238000000749 co-immunoprecipitation Methods 0.000 description 6
- IPCSVZSSVZVIGE-UHFFFAOYSA-N hexadecanoic acid Chemical compound CCCCCCCCCCCCCCCC(O)=O IPCSVZSSVZVIGE-UHFFFAOYSA-N 0.000 description 6
- 230000002503 metabolic effect Effects 0.000 description 6
- 239000000203 mixture Substances 0.000 description 6
- 239000002953 phosphate buffered saline Substances 0.000 description 6
- 239000000047 product Substances 0.000 description 6
- 108010033804 ribosomal protein S3 Proteins 0.000 description 6
- 210000002966 serum Anatomy 0.000 description 6
- 230000001225 therapeutic effect Effects 0.000 description 6
- 241000701161 unidentified adenovirus Species 0.000 description 6
- XLYOFNOQVPJJNP-UHFFFAOYSA-N water Substances O XLYOFNOQVPJJNP-UHFFFAOYSA-N 0.000 description 6
- 239000013607 AAV vector Substances 0.000 description 5
- GFFGJBXGBJISGV-UHFFFAOYSA-N Adenine Chemical compound NC1=NC=NC2=C1N=CN2 GFFGJBXGBJISGV-UHFFFAOYSA-N 0.000 description 5
- 102100026148 Free fatty acid receptor 1 Human genes 0.000 description 5
- 101710198884 GATA-type zinc finger protein 1 Proteins 0.000 description 5
- DTHNMHAUYICORS-KTKZVXAJSA-N Glucagon-like peptide 1 Chemical compound C([C@@H](C(=O)N[C@@H]([C@@H](C)CC)C(=O)N[C@@H](C)C(=O)N[C@@H](CC=1C2=CC=CC=C2NC=1)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](C(C)C)C(=O)N[C@@H](CCCCN)C(=O)NCC(=O)N[C@@H](CCCNC(N)=N)C(N)=O)NC(=O)[C@H](CCC(O)=O)NC(=O)[C@H](CCCCN)NC(=O)[C@H](C)NC(=O)[C@H](C)NC(=O)[C@H](CCC(N)=O)NC(=O)CNC(=O)[C@H](CCC(O)=O)NC(=O)[C@H](CC(C)C)NC(=O)[C@H](CC=1C=CC(O)=CC=1)NC(=O)[C@H](CO)NC(=O)[C@H](CO)NC(=O)[C@@H](NC(=O)[C@H](CC(O)=O)NC(=O)[C@H](CO)NC(=O)[C@@H](NC(=O)[C@H](CC=1C=CC=CC=1)NC(=O)[C@@H](NC(=O)CNC(=O)[C@H](CCC(O)=O)NC(=O)[C@H](C)NC(=O)[C@@H](N)CC=1N=CNC=1)[C@@H](C)O)[C@@H](C)O)C(C)C)C1=CC=CC=C1 DTHNMHAUYICORS-KTKZVXAJSA-N 0.000 description 5
- 102400000322 Glucagon-like peptide 1 Human genes 0.000 description 5
- 101000912510 Homo sapiens Free fatty acid receptor 1 Proteins 0.000 description 5
- 102100025354 Macrophage mannose receptor 1 Human genes 0.000 description 5
- 108091093037 Peptide nucleic acid Proteins 0.000 description 5
- YASAKCUCGLMORW-UHFFFAOYSA-N Rosiglitazone Chemical compound C=1C=CC=NC=1N(C)CCOC(C=C1)=CC=C1CC1SC(=O)NC1=O YASAKCUCGLMORW-UHFFFAOYSA-N 0.000 description 5
- 239000003153 chemical reaction reagent Substances 0.000 description 5
- 230000035605 chemotaxis Effects 0.000 description 5
- 230000001684 chronic effect Effects 0.000 description 5
- 239000000562 conjugate Substances 0.000 description 5
- 235000021323 fish oil Nutrition 0.000 description 5
- 230000000910 hyperinsulinemic effect Effects 0.000 description 5
- 238000001802 infusion Methods 0.000 description 5
- 125000002496 methyl group Chemical group [H]C([H])([H])* 0.000 description 5
- 210000000107 myocyte Anatomy 0.000 description 5
- 239000002777 nucleoside Substances 0.000 description 5
- 229910052698 phosphorus Inorganic materials 0.000 description 5
- 230000003389 potentiating effect Effects 0.000 description 5
- 230000000153 supplemental effect Effects 0.000 description 5
- 230000001629 suppression Effects 0.000 description 5
- 238000012360 testing method Methods 0.000 description 5
- 102100029649 Beta-arrestin-1 Human genes 0.000 description 4
- 108020004414 DNA Proteins 0.000 description 4
- 102100023274 Dual specificity mitogen-activated protein kinase kinase 4 Human genes 0.000 description 4
- 102100025682 Dystroglycan 1 Human genes 0.000 description 4
- 108091006027 G proteins Proteins 0.000 description 4
- 102000030782 GTP binding Human genes 0.000 description 4
- 108091000058 GTP-Binding Proteins 0.000 description 4
- 101001115395 Homo sapiens Dual specificity mitogen-activated protein kinase kinase 4 Proteins 0.000 description 4
- 101000831496 Homo sapiens Toll-like receptor 3 Proteins 0.000 description 4
- 101000669447 Homo sapiens Toll-like receptor 4 Proteins 0.000 description 4
- 102100024324 Toll-like receptor 3 Human genes 0.000 description 4
- 102100039360 Toll-like receptor 4 Human genes 0.000 description 4
- 230000009471 action Effects 0.000 description 4
- 150000001408 amides Chemical group 0.000 description 4
- 239000003708 ampul Substances 0.000 description 4
- 108010032969 beta-Arrestin 1 Proteins 0.000 description 4
- 230000015556 catabolic process Effects 0.000 description 4
- 210000000170 cell membrane Anatomy 0.000 description 4
- HVYWMOMLDIMFJA-DPAQBDIFSA-N cholesterol Chemical group C1C=C2C[C@@H](O)CC[C@]2(C)[C@@H]2[C@@H]1[C@@H]1CC[C@H]([C@H](C)CCCC(C)C)[C@@]1(C)CC2 HVYWMOMLDIMFJA-DPAQBDIFSA-N 0.000 description 4
- 230000021615 conjugation Effects 0.000 description 4
- 238000006731 degradation reaction Methods 0.000 description 4
- 238000010586 diagram Methods 0.000 description 4
- 208000037765 diseases and disorders Diseases 0.000 description 4
- 239000003937 drug carrier Substances 0.000 description 4
- 238000004520 electroporation Methods 0.000 description 4
- 230000001610 euglycemic effect Effects 0.000 description 4
- 238000001943 fluorescence-activated cell sorting Methods 0.000 description 4
- 238000009396 hybridization Methods 0.000 description 4
- 210000001865 kupffer cell Anatomy 0.000 description 4
- 238000005259 measurement Methods 0.000 description 4
- 238000010606 normalization Methods 0.000 description 4
- 125000003835 nucleoside group Chemical group 0.000 description 4
- 125000004437 phosphorous atom Chemical group 0.000 description 4
- 239000004417 polycarbonate Substances 0.000 description 4
- 108090000765 processed proteins & peptides Proteins 0.000 description 4
- 239000000700 radioactive tracer Substances 0.000 description 4
- 230000002829 reductive effect Effects 0.000 description 4
- 238000003757 reverse transcription PCR Methods 0.000 description 4
- 125000002652 ribonucleotide group Chemical group 0.000 description 4
- 210000002027 skeletal muscle Anatomy 0.000 description 4
- 239000000126 substance Substances 0.000 description 4
- 229940113082 thymine Drugs 0.000 description 4
- 238000013518 transcription Methods 0.000 description 4
- 230000035897 transcription Effects 0.000 description 4
- 238000012546 transfer Methods 0.000 description 4
- 150000003626 triacylglycerols Chemical class 0.000 description 4
- 239000003981 vehicle Substances 0.000 description 4
- 101710129000 Arginase-1 Proteins 0.000 description 3
- 102100021723 Arginase-1 Human genes 0.000 description 3
- WVDDGKGOMKODPV-UHFFFAOYSA-N Benzyl alcohol Chemical compound OCC1=CC=CC=C1 WVDDGKGOMKODPV-UHFFFAOYSA-N 0.000 description 3
- 108090000565 Capsid Proteins Proteins 0.000 description 3
- 108090000994 Catalytic RNA Proteins 0.000 description 3
- 102000053642 Catalytic RNA Human genes 0.000 description 3
- 102100023321 Ceruloplasmin Human genes 0.000 description 3
- 241000701022 Cytomegalovirus Species 0.000 description 3
- LFQSCWFLJHTTHZ-UHFFFAOYSA-N Ethanol Chemical compound CCO LFQSCWFLJHTTHZ-UHFFFAOYSA-N 0.000 description 3
- 101710154606 Hemagglutinin Proteins 0.000 description 3
- 102000003814 Interleukin-10 Human genes 0.000 description 3
- 108090000174 Interleukin-10 Proteins 0.000 description 3
- 102000016267 Leptin Human genes 0.000 description 3
- 108010092277 Leptin Proteins 0.000 description 3
- 101100102907 Mus musculus Wdtc1 gene Proteins 0.000 description 3
- 102100029438 Nitric oxide synthase, inducible Human genes 0.000 description 3
- 101710089543 Nitric oxide synthase, inducible Proteins 0.000 description 3
- 101710093908 Outer capsid protein VP4 Proteins 0.000 description 3
- 101710135467 Outer capsid protein sigma-1 Proteins 0.000 description 3
- 239000004793 Polystyrene Substances 0.000 description 3
- DNIAPMSPPWPWGF-UHFFFAOYSA-N Propylene glycol Chemical compound CC(O)CO DNIAPMSPPWPWGF-UHFFFAOYSA-N 0.000 description 3
- 101710176177 Protein A56 Proteins 0.000 description 3
- 108091028664 Ribonucleotide Proteins 0.000 description 3
- HEMHJVSKTPXQMS-UHFFFAOYSA-M Sodium hydroxide Chemical compound [OH-].[Na+] HEMHJVSKTPXQMS-UHFFFAOYSA-M 0.000 description 3
- RYYWUUFWQRZTIU-UHFFFAOYSA-N Thiophosphoric acid Chemical class OP(O)(S)=O RYYWUUFWQRZTIU-UHFFFAOYSA-N 0.000 description 3
- ZMANZCXQSJIPKH-UHFFFAOYSA-N Triethylamine Chemical compound CCN(CC)CC ZMANZCXQSJIPKH-UHFFFAOYSA-N 0.000 description 3
- 230000001594 aberrant effect Effects 0.000 description 3
- 230000001154 acute effect Effects 0.000 description 3
- 239000000427 antigen Substances 0.000 description 3
- 108091007433 antigens Proteins 0.000 description 3
- 102000036639 antigens Human genes 0.000 description 3
- 238000013459 approach Methods 0.000 description 3
- WQZGKKKJIJFFOK-VFUOTHLCSA-N beta-D-glucose Chemical compound OC[C@H]1O[C@@H](O)[C@H](O)[C@@H](O)[C@@H]1O WQZGKKKJIJFFOK-VFUOTHLCSA-N 0.000 description 3
- 230000037396 body weight Effects 0.000 description 3
- 210000000234 capsid Anatomy 0.000 description 3
- 239000000969 carrier Substances 0.000 description 3
- 230000004700 cellular uptake Effects 0.000 description 3
- 238000003776 cleavage reaction Methods 0.000 description 3
- 238000004624 confocal microscopy Methods 0.000 description 3
- 239000008121 dextrose Substances 0.000 description 3
- 230000004069 differentiation Effects 0.000 description 3
- 229940079593 drug Drugs 0.000 description 3
- 201000010063 epididymitis Diseases 0.000 description 3
- 239000011521 glass Substances 0.000 description 3
- 235000011187 glycerol Nutrition 0.000 description 3
- 239000000185 hemagglutinin Substances 0.000 description 3
- 210000003958 hematopoietic stem cell Anatomy 0.000 description 3
- 238000001114 immunoprecipitation Methods 0.000 description 3
- 230000006698 induction Effects 0.000 description 3
- 239000003112 inhibitor Substances 0.000 description 3
- NRYBAZVQPHGZNS-ZSOCWYAHSA-N leptin Chemical compound O=C([C@H](CO)NC(=O)[C@H](CC(C)C)NC(=O)[C@H](CC(O)=O)NC(=O)[C@H](CC(C)C)NC(=O)[C@H](CCC(N)=O)NC(=O)[C@H](CC=1C2=CC=CC=C2NC=1)NC(=O)[C@H](CC(C)C)NC(=O)[C@@H](NC(=O)[C@H](CC(O)=O)NC(=O)[C@H](CCC(N)=O)NC(=O)[C@H](CC(C)C)NC(=O)[C@H](CO)NC(=O)CNC(=O)[C@H](CCC(N)=O)NC(=O)[C@@H](N)CC(C)C)CCSC)N1CCC[C@H]1C(=O)NCC(=O)N[C@@H](CS)C(O)=O NRYBAZVQPHGZNS-ZSOCWYAHSA-N 0.000 description 3
- 229940039781 leptin Drugs 0.000 description 3
- 230000007774 longterm Effects 0.000 description 3
- 239000006166 lysate Substances 0.000 description 3
- 239000000463 material Substances 0.000 description 3
- 239000002609 medium Substances 0.000 description 3
- 239000012528 membrane Substances 0.000 description 3
- 229910052751 metal Inorganic materials 0.000 description 3
- 239000002184 metal Substances 0.000 description 3
- 108091070501 miRNA Proteins 0.000 description 3
- VLKZOEOYAKHREP-UHFFFAOYSA-N n-Hexane Chemical compound CCCCCC VLKZOEOYAKHREP-UHFFFAOYSA-N 0.000 description 3
- 229910052757 nitrogen Inorganic materials 0.000 description 3
- 208000008338 non-alcoholic fatty liver disease Diseases 0.000 description 3
- 239000008194 pharmaceutical composition Substances 0.000 description 3
- 150000003904 phospholipids Chemical class 0.000 description 3
- 229920001223 polyethylene glycol Polymers 0.000 description 3
- 230000000069 prophylactic effect Effects 0.000 description 3
- 238000000746 purification Methods 0.000 description 3
- 230000001105 regulatory effect Effects 0.000 description 3
- 239000002336 ribonucleotide Substances 0.000 description 3
- 108091092562 ribozyme Proteins 0.000 description 3
- 239000000523 sample Substances 0.000 description 3
- 230000007017 scission Effects 0.000 description 3
- 150000003384 small molecules Chemical class 0.000 description 3
- 238000010186 staining Methods 0.000 description 3
- QAOWNCQODCNURD-UHFFFAOYSA-N sulfuric acid group Chemical class S(O)(O)(=O)=O QAOWNCQODCNURD-UHFFFAOYSA-N 0.000 description 3
- 238000011144 upstream manufacturing Methods 0.000 description 3
- 210000003462 vein Anatomy 0.000 description 3
- BHQCQFFYRZLCQQ-UHFFFAOYSA-N (3alpha,5alpha,7alpha,12alpha)-3,7,12-trihydroxy-cholan-24-oic acid Natural products OC1CC2CC(O)CCC2(C)C2C1C1CCC(C(CCC(O)=O)C)C1(C)C(O)C2 BHQCQFFYRZLCQQ-UHFFFAOYSA-N 0.000 description 2
- QGVQZRDQPDLHHV-DPAQBDIFSA-N (3s,8s,9s,10r,13r,14s,17r)-10,13-dimethyl-17-[(2r)-6-methylheptan-2-yl]-2,3,4,7,8,9,11,12,14,15,16,17-dodecahydro-1h-cyclopenta[a]phenanthrene-3-thiol Chemical compound C1C=C2C[C@@H](S)CC[C@]2(C)[C@@H]2[C@@H]1[C@@H]1CC[C@H]([C@H](C)CCCC(C)C)[C@@]1(C)CC2 QGVQZRDQPDLHHV-DPAQBDIFSA-N 0.000 description 2
- VBICKXHEKHSIBG-UHFFFAOYSA-N 1-monostearoylglycerol Chemical compound CCCCCCCCCCCCCCCCCC(=O)OCC(O)CO VBICKXHEKHSIBG-UHFFFAOYSA-N 0.000 description 2
- FZWGECJQACGGTI-UHFFFAOYSA-N 2-amino-7-methyl-1,7-dihydro-6H-purin-6-one Chemical compound NC1=NC(O)=C2N(C)C=NC2=N1 FZWGECJQACGGTI-UHFFFAOYSA-N 0.000 description 2
- OVONXEQGWXGFJD-UHFFFAOYSA-N 4-sulfanylidene-1h-pyrimidin-2-one Chemical compound SC=1C=CNC(=O)N=1 OVONXEQGWXGFJD-UHFFFAOYSA-N 0.000 description 2
- RYVNIFSIEDRLSJ-UHFFFAOYSA-N 5-(hydroxymethyl)cytosine Chemical compound NC=1NC(=O)N=CC=1CO RYVNIFSIEDRLSJ-UHFFFAOYSA-N 0.000 description 2
- LRSASMSXMSNRBT-UHFFFAOYSA-N 5-methylcytosine Chemical compound CC1=CNC(=O)N=C1N LRSASMSXMSNRBT-UHFFFAOYSA-N 0.000 description 2
- UJBCLAXPPIDQEE-UHFFFAOYSA-N 5-prop-1-ynyl-1h-pyrimidine-2,4-dione Chemical compound CC#CC1=CNC(=O)NC1=O UJBCLAXPPIDQEE-UHFFFAOYSA-N 0.000 description 2
- HCGHYQLFMPXSDU-UHFFFAOYSA-N 7-methyladenine Chemical compound C1=NC(N)=C2N(C)C=NC2=N1 HCGHYQLFMPXSDU-UHFFFAOYSA-N 0.000 description 2
- KDCGOANMDULRCW-UHFFFAOYSA-N 7H-purine Chemical compound N1=CNC2=NC=NC2=C1 KDCGOANMDULRCW-UHFFFAOYSA-N 0.000 description 2
- LRFVTYWOQMYALW-UHFFFAOYSA-N 9H-xanthine Chemical compound O=C1NC(=O)NC2=C1NC=N2 LRFVTYWOQMYALW-UHFFFAOYSA-N 0.000 description 2
- CIWBSHSKHKDKBQ-JLAZNSOCSA-N Ascorbic acid Chemical compound OC[C@H](O)[C@H]1OC(=O)C(O)=C1O CIWBSHSKHKDKBQ-JLAZNSOCSA-N 0.000 description 2
- 241000273930 Brevoortia tyrannus Species 0.000 description 2
- OYPRJOBELJOOCE-UHFFFAOYSA-N Calcium Chemical compound [Ca] OYPRJOBELJOOCE-UHFFFAOYSA-N 0.000 description 2
- 102000009410 Chemokine receptor Human genes 0.000 description 2
- 108050000299 Chemokine receptor Proteins 0.000 description 2
- 102000019034 Chemokines Human genes 0.000 description 2
- 108010012236 Chemokines Proteins 0.000 description 2
- HEDRZPFGACZZDS-UHFFFAOYSA-N Chloroform Chemical compound ClC(Cl)Cl HEDRZPFGACZZDS-UHFFFAOYSA-N 0.000 description 2
- 239000004380 Cholic acid Substances 0.000 description 2
- 102000029816 Collagenase Human genes 0.000 description 2
- 108060005980 Collagenase Proteins 0.000 description 2
- 239000006144 Dulbecco’s modified Eagle's medium Substances 0.000 description 2
- KCXVZYZYPLLWCC-UHFFFAOYSA-N EDTA Chemical compound OC(=O)CN(CC(O)=O)CCN(CC(O)=O)CC(O)=O KCXVZYZYPLLWCC-UHFFFAOYSA-N 0.000 description 2
- LYCAIKOWRPUZTN-UHFFFAOYSA-N Ethylene glycol Chemical compound OCCO LYCAIKOWRPUZTN-UHFFFAOYSA-N 0.000 description 2
- 238000012413 Fluorescence activated cell sorting analysis Methods 0.000 description 2
- WSFSSNUMVMOOMR-UHFFFAOYSA-N Formaldehyde Chemical compound O=C WSFSSNUMVMOOMR-UHFFFAOYSA-N 0.000 description 2
- 208000002705 Glucose Intolerance Diseases 0.000 description 2
- 206010018429 Glucose tolerance impaired Diseases 0.000 description 2
- 102100031181 Glyceraldehyde-3-phosphate dehydrogenase Human genes 0.000 description 2
- 238000013218 HFD mouse model Methods 0.000 description 2
- 206010019708 Hepatic steatosis Diseases 0.000 description 2
- DGAQECJNVWCQMB-PUAWFVPOSA-M Ilexoside XXIX Chemical compound C[C@@H]1CC[C@@]2(CC[C@@]3(C(=CC[C@H]4[C@]3(CC[C@@H]5[C@@]4(CC[C@@H](C5(C)C)OS(=O)(=O)[O-])C)C)[C@@H]2[C@]1(C)O)C)C(=O)O[C@H]6[C@@H]([C@H]([C@@H]([C@H](O6)CO)O)O)O.[Na+] DGAQECJNVWCQMB-PUAWFVPOSA-M 0.000 description 2
- 229930010555 Inosine Natural products 0.000 description 2
- UGQMRVRMYYASKQ-KQYNXXCUSA-N Inosine Chemical compound O[C@@H]1[C@H](O)[C@@H](CO)O[C@H]1N1C2=NC=NC(O)=C2N=C1 UGQMRVRMYYASKQ-KQYNXXCUSA-N 0.000 description 2
- 102100025087 Insulin receptor substrate 1 Human genes 0.000 description 2
- 101710201824 Insulin receptor substrate 1 Proteins 0.000 description 2
- 102100034349 Integrase Human genes 0.000 description 2
- 101710203526 Integrase Proteins 0.000 description 2
- 241000713666 Lentivirus Species 0.000 description 2
- 239000005089 Luciferase Substances 0.000 description 2
- TWRXJAOTZQYOKJ-UHFFFAOYSA-L Magnesium chloride Chemical compound [Mg+2].[Cl-].[Cl-] TWRXJAOTZQYOKJ-UHFFFAOYSA-L 0.000 description 2
- MBBZMMPHUWSWHV-BDVNFPICSA-N N-methylglucamine Chemical compound CNC[C@H](O)[C@@H](O)[C@H](O)[C@H](O)CO MBBZMMPHUWSWHV-BDVNFPICSA-N 0.000 description 2
- REYJJPSVUYRZGE-UHFFFAOYSA-N Octadecylamine Chemical compound CCCCCCCCCCCCCCCCCCN REYJJPSVUYRZGE-UHFFFAOYSA-N 0.000 description 2
- 235000021314 Palmitic acid Nutrition 0.000 description 2
- 208000037273 Pathologic Processes Diseases 0.000 description 2
- 239000004698 Polyethylene Substances 0.000 description 2
- 239000002202 Polyethylene glycol Substances 0.000 description 2
- ZLMJMSJWJFRBEC-UHFFFAOYSA-N Potassium Chemical compound [K] ZLMJMSJWJFRBEC-UHFFFAOYSA-N 0.000 description 2
- 206010070834 Sensitisation Diseases 0.000 description 2
- 108091027568 Single-stranded nucleotide Proteins 0.000 description 2
- 108091081024 Start codon Proteins 0.000 description 2
- 241000906446 Theraps Species 0.000 description 2
- 241000711975 Vesicular stomatitis virus Species 0.000 description 2
- RLXCFCYWFYXTON-JTTSDREOSA-N [(3S,8S,9S,10R,13S,14S,17R)-3-hydroxy-10,13-dimethyl-17-[(2R)-6-methylheptan-2-yl]-2,3,4,7,8,9,11,12,14,15,16,17-dodecahydro-1H-cyclopenta[a]phenanthren-16-yl] N-hexylcarbamate Chemical group C1C=C2C[C@@H](O)CC[C@]2(C)[C@@H]2[C@@H]1[C@@H]1CC(OC(=O)NCCCCCC)[C@H]([C@H](C)CCCC(C)C)[C@@]1(C)CC2 RLXCFCYWFYXTON-JTTSDREOSA-N 0.000 description 2
- 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 2
- XVIYCJDWYLJQBG-UHFFFAOYSA-N acetic acid;adamantane Chemical compound CC(O)=O.C1C(C2)CC3CC1CC2C3 XVIYCJDWYLJQBG-UHFFFAOYSA-N 0.000 description 2
- 125000001931 aliphatic group Chemical group 0.000 description 2
- 125000003342 alkenyl group Chemical group 0.000 description 2
- 125000000304 alkynyl group Chemical group 0.000 description 2
- 229910052782 aluminium Inorganic materials 0.000 description 2
- XAGFODPZIPBFFR-UHFFFAOYSA-N aluminium Chemical compound [Al] XAGFODPZIPBFFR-UHFFFAOYSA-N 0.000 description 2
- 125000003277 amino group Chemical group 0.000 description 2
- 238000010171 animal model Methods 0.000 description 2
- 239000011324 bead Substances 0.000 description 2
- SRSXLGNVWSONIS-UHFFFAOYSA-M benzenesulfonate Chemical compound [O-]S(=O)(=O)C1=CC=CC=C1 SRSXLGNVWSONIS-UHFFFAOYSA-M 0.000 description 2
- 230000000903 blocking effect Effects 0.000 description 2
- 210000004979 bone marrow derived macrophage Anatomy 0.000 description 2
- 210000004899 c-terminal region Anatomy 0.000 description 2
- 239000011575 calcium Substances 0.000 description 2
- 229910052791 calcium Inorganic materials 0.000 description 2
- 238000004113 cell culture Methods 0.000 description 2
- 238000006243 chemical reaction Methods 0.000 description 2
- 235000012000 cholesterol Nutrition 0.000 description 2
- 150000001840 cholesterol esters Chemical class 0.000 description 2
- 235000019416 cholic acid Nutrition 0.000 description 2
- BHQCQFFYRZLCQQ-OELDTZBJSA-N cholic acid Chemical compound C([C@H]1C[C@H]2O)[C@H](O)CC[C@]1(C)[C@@H]1[C@@H]2[C@@H]2CC[C@H]([C@@H](CCC(O)=O)C)[C@@]2(C)[C@@H](O)C1 BHQCQFFYRZLCQQ-OELDTZBJSA-N 0.000 description 2
- 229960002471 cholic acid Drugs 0.000 description 2
- 229960002424 collagenase Drugs 0.000 description 2
- 239000002299 complementary DNA Substances 0.000 description 2
- 125000000753 cycloalkyl group Chemical group 0.000 description 2
- 230000006735 deficit Effects 0.000 description 2
- KXGVEGMKQFWNSR-UHFFFAOYSA-N deoxycholic acid Natural products C1CC2CC(O)CCC2(C)C2C1C1CCC(C(CCC(O)=O)C)C1(C)C(O)C2 KXGVEGMKQFWNSR-UHFFFAOYSA-N 0.000 description 2
- 238000013461 design Methods 0.000 description 2
- 238000011161 development Methods 0.000 description 2
- 230000018109 developmental process Effects 0.000 description 2
- 239000010432 diamond Substances 0.000 description 2
- LOKCTEFSRHRXRJ-UHFFFAOYSA-I dipotassium trisodium dihydrogen phosphate hydrogen phosphate dichloride Chemical compound P(=O)(O)(O)[O-].[K+].P(=O)(O)([O-])[O-].[Na+].[Na+].[Cl-].[K+].[Cl-].[Na+] LOKCTEFSRHRXRJ-UHFFFAOYSA-I 0.000 description 2
- 229940042399 direct acting antivirals protease inhibitors Drugs 0.000 description 2
- 208000035475 disorder Diseases 0.000 description 2
- 238000009826 distribution Methods 0.000 description 2
- 238000011067 equilibration Methods 0.000 description 2
- 238000011156 evaluation Methods 0.000 description 2
- 239000000284 extract Substances 0.000 description 2
- 235000019387 fatty acid methyl ester Nutrition 0.000 description 2
- 239000012091 fetal bovine serum Substances 0.000 description 2
- 235000013305 food Nutrition 0.000 description 2
- 238000009472 formulation Methods 0.000 description 2
- 235000021588 free fatty acids Nutrition 0.000 description 2
- 238000003304 gavage Methods 0.000 description 2
- 239000000499 gel Substances 0.000 description 2
- 230000009229 glucose formation Effects 0.000 description 2
- 230000014101 glucose homeostasis Effects 0.000 description 2
- 108020004445 glyceraldehyde-3-phosphate dehydrogenase Proteins 0.000 description 2
- 125000003827 glycol group Chemical group 0.000 description 2
- 125000001475 halogen functional group Chemical group 0.000 description 2
- 210000003494 hepatocyte Anatomy 0.000 description 2
- 125000005842 heteroatom Chemical group 0.000 description 2
- 238000004128 high performance liquid chromatography Methods 0.000 description 2
- FDGQSTZJBFJUBT-UHFFFAOYSA-N hypoxanthine Chemical compound O=C1NC=NC2=C1NC=N2 FDGQSTZJBFJUBT-UHFFFAOYSA-N 0.000 description 2
- 238000012744 immunostaining Methods 0.000 description 2
- 230000001976 improved effect Effects 0.000 description 2
- 238000011534 incubation Methods 0.000 description 2
- 230000001939 inductive effect Effects 0.000 description 2
- 238000002347 injection Methods 0.000 description 2
- 239000007924 injection Substances 0.000 description 2
- 150000007529 inorganic bases Chemical class 0.000 description 2
- 229960003786 inosine Drugs 0.000 description 2
- 239000007788 liquid Substances 0.000 description 2
- 238000004519 manufacturing process Methods 0.000 description 2
- 239000003550 marker Substances 0.000 description 2
- 230000005012 migration Effects 0.000 description 2
- 238000013508 migration Methods 0.000 description 2
- 230000001617 migratory effect Effects 0.000 description 2
- 230000009456 molecular mechanism Effects 0.000 description 2
- 125000004573 morpholin-4-yl group Chemical group N1(CCOCC1)* 0.000 description 2
- WQEPLUUGTLDZJY-UHFFFAOYSA-N n-Pentadecanoic acid Natural products CCCCCCCCCCCCCCC(O)=O WQEPLUUGTLDZJY-UHFFFAOYSA-N 0.000 description 2
- 230000007935 neutral effect Effects 0.000 description 2
- 231100000252 nontoxic Toxicity 0.000 description 2
- 230000003000 nontoxic effect Effects 0.000 description 2
- 150000003833 nucleoside derivatives Chemical class 0.000 description 2
- 238000013116 obese mouse model Methods 0.000 description 2
- 230000009437 off-target effect Effects 0.000 description 2
- 239000003921 oil Substances 0.000 description 2
- 150000007530 organic bases Chemical class 0.000 description 2
- 238000004806 packaging method and process Methods 0.000 description 2
- 125000000913 palmityl group Chemical group [H]C([*])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])[H] 0.000 description 2
- 230000009054 pathological process Effects 0.000 description 2
- ONTNXMBMXUNDBF-UHFFFAOYSA-N pentatriacontane-17,18,19-triol Chemical compound CCCCCCCCCCCCCCCCC(O)C(O)C(O)CCCCCCCCCCCCCCCC ONTNXMBMXUNDBF-UHFFFAOYSA-N 0.000 description 2
- 239000000137 peptide hydrolase inhibitor Substances 0.000 description 2
- 239000000546 pharmaceutical excipient Substances 0.000 description 2
- 239000004033 plastic Substances 0.000 description 2
- 229920003023 plastic Polymers 0.000 description 2
- 229920000768 polyamine Polymers 0.000 description 2
- 229920000515 polycarbonate Polymers 0.000 description 2
- 239000011591 potassium Substances 0.000 description 2
- 229910052700 potassium Inorganic materials 0.000 description 2
- 210000000229 preadipocyte Anatomy 0.000 description 2
- MFDFERRIHVXMIY-UHFFFAOYSA-N procaine Chemical compound CCN(CC)CCOC(=O)C1=CC=C(N)C=C1 MFDFERRIHVXMIY-UHFFFAOYSA-N 0.000 description 2
- 229960004919 procaine Drugs 0.000 description 2
- 230000008569 process Effects 0.000 description 2
- 230000009467 reduction Effects 0.000 description 2
- 229960004586 rosiglitazone Drugs 0.000 description 2
- 238000007423 screening assay Methods 0.000 description 2
- 230000008313 sensitization Effects 0.000 description 2
- 239000004055 small Interfering RNA Substances 0.000 description 2
- 239000011734 sodium Substances 0.000 description 2
- 229910052708 sodium Inorganic materials 0.000 description 2
- 239000008223 sterile water Substances 0.000 description 2
- 238000006467 substitution reaction Methods 0.000 description 2
- 239000000758 substrate Substances 0.000 description 2
- 208000024891 symptom Diseases 0.000 description 2
- 230000008685 targeting Effects 0.000 description 2
- 150000003568 thioethers Chemical class 0.000 description 2
- RYYWUUFWQRZTIU-UHFFFAOYSA-K thiophosphate Chemical compound [O-]P([O-])([O-])=S RYYWUUFWQRZTIU-UHFFFAOYSA-K 0.000 description 2
- ZMANZCXQSJIPKH-UHFFFAOYSA-O triethylammonium ion Chemical compound CC[NH+](CC)CC ZMANZCXQSJIPKH-UHFFFAOYSA-O 0.000 description 2
- DCXXMTOCNZCJGO-UHFFFAOYSA-N tristearoylglycerol Chemical compound CCCCCCCCCCCCCCCCCC(=O)OCC(OC(=O)CCCCCCCCCCCCCCCCC)COC(=O)CCCCCCCCCCCCCCCCC DCXXMTOCNZCJGO-UHFFFAOYSA-N 0.000 description 2
- 125000002948 undecyl group Chemical group [H]C([*])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])[H] 0.000 description 2
- 230000002792 vascular Effects 0.000 description 2
- 210000005167 vascular cell Anatomy 0.000 description 2
- DTOSIQBPPRVQHS-UHFFFAOYSA-N α-Linolenic acid Chemical compound CCC=CCC=CCC=CCCCCCCCC(O)=O DTOSIQBPPRVQHS-UHFFFAOYSA-N 0.000 description 2
- INNTZVXVIZIYBF-PXSLIBMESA-N (2S)-6-amino-2-[[(2S)-6-amino-2-[[(2S)-6-amino-2-[[(2S)-6-amino-2-[[(2S)-2-[[(2R)-3-[2,3-di(hexadecanoyloxy)propylsulfanyl]-2-(hexadecanoylamino)propanoyl]amino]-3-hydroxypropanoyl]amino]hexanoyl]amino]hexanoyl]amino]hexanoyl]amino]hexanoic acid trihydrochloride Chemical compound Cl.Cl.Cl.CCCCCCCCCCCCCCCC(=O)N[C@@H](CSCC(COC(=O)CCCCCCCCCCCCCCC)OC(=O)CCCCCCCCCCCCCCC)C(=O)N[C@@H](CO)C(=O)N[C@@H](CCCCN)C(=O)N[C@@H](CCCCN)C(=O)N[C@@H](CCCCN)C(=O)N[C@@H](CCCCN)C(O)=O INNTZVXVIZIYBF-PXSLIBMESA-N 0.000 description 1
- JNTMAZFVYNDPLB-PEDHHIEDSA-N (2S,3S)-2-[[[(2S)-1-[(2S,3S)-2-amino-3-methyl-1-oxopentyl]-2-pyrrolidinyl]-oxomethyl]amino]-3-methylpentanoic acid Chemical compound CC[C@H](C)[C@H](N)C(=O)N1CCC[C@H]1C(=O)N[C@@H]([C@@H](C)CC)C(O)=O JNTMAZFVYNDPLB-PEDHHIEDSA-N 0.000 description 1
- YIMATHOGWXZHFX-WCTZXXKLSA-N (2r,3r,4r,5r)-5-(hydroxymethyl)-3-(2-methoxyethoxy)oxolane-2,4-diol Chemical compound COCCO[C@H]1[C@H](O)O[C@H](CO)[C@H]1O YIMATHOGWXZHFX-WCTZXXKLSA-N 0.000 description 1
- FYADHXFMURLYQI-UHFFFAOYSA-N 1,2,4-triazine Chemical class C1=CN=NC=N1 FYADHXFMURLYQI-UHFFFAOYSA-N 0.000 description 1
- TZCPCKNHXULUIY-RGULYWFUSA-N 1,2-distearoyl-sn-glycero-3-phosphoserine Chemical compound CCCCCCCCCCCCCCCCCC(=O)OC[C@H](COP(O)(=O)OC[C@H](N)C(O)=O)OC(=O)CCCCCCCCCCCCCCCCC TZCPCKNHXULUIY-RGULYWFUSA-N 0.000 description 1
- ZOBPZXTWZATXDG-UHFFFAOYSA-N 1,3-thiazolidine-2,4-dione Chemical compound O=C1CSC(=O)N1 ZOBPZXTWZATXDG-UHFFFAOYSA-N 0.000 description 1
- RYCNUMLMNKHWPZ-SNVBAGLBSA-N 1-acetyl-sn-glycero-3-phosphocholine Chemical compound CC(=O)OC[C@@H](O)COP([O-])(=O)OCC[N+](C)(C)C RYCNUMLMNKHWPZ-SNVBAGLBSA-N 0.000 description 1
- SWTYBBUBEPPYCX-YTQNUIGOSA-N 17(S)-HDoHE Chemical compound CC\C=C/C[C@H](O)\C=C\C=C/C\C=C/C\C=C/C\C=C/CCC(O)=O SWTYBBUBEPPYCX-YTQNUIGOSA-N 0.000 description 1
- UHUHBFMZVCOEOV-UHFFFAOYSA-N 1h-imidazo[4,5-c]pyridin-4-amine Chemical compound NC1=NC=CC2=C1N=CN2 UHUHBFMZVCOEOV-UHFFFAOYSA-N 0.000 description 1
- IHPYMWDTONKSCO-UHFFFAOYSA-N 2,2'-piperazine-1,4-diylbisethanesulfonic acid Chemical compound OS(=O)(=O)CCN1CCN(CCS(O)(=O)=O)CC1 IHPYMWDTONKSCO-UHFFFAOYSA-N 0.000 description 1
- QSHACTSJHMKXTE-UHFFFAOYSA-N 2-(2-aminopropyl)-7h-purin-6-amine Chemical compound CC(N)CC1=NC(N)=C2NC=NC2=N1 QSHACTSJHMKXTE-UHFFFAOYSA-N 0.000 description 1
- PIINGYXNCHTJTF-UHFFFAOYSA-N 2-(2-azaniumylethylamino)acetate Chemical group NCCNCC(O)=O PIINGYXNCHTJTF-UHFFFAOYSA-N 0.000 description 1
- MIJDSYMOBYNHOT-UHFFFAOYSA-N 2-(ethylamino)ethanol Chemical compound CCNCCO MIJDSYMOBYNHOT-UHFFFAOYSA-N 0.000 description 1
- JKMHFZQWWAIEOD-UHFFFAOYSA-N 2-[4-(2-hydroxyethyl)piperazin-1-yl]ethanesulfonic acid Chemical compound OCC[NH+]1CCN(CCS([O-])(=O)=O)CC1 JKMHFZQWWAIEOD-UHFFFAOYSA-N 0.000 description 1
- 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
- ZLAQATDNGLKIEV-UHFFFAOYSA-N 5-methyl-2-sulfanylidene-1h-pyrimidin-4-one Chemical compound CC1=CNC(=S)NC1=O ZLAQATDNGLKIEV-UHFFFAOYSA-N 0.000 description 1
- GOZMBJCYMQQACI-UHFFFAOYSA-N 6,7-dimethyl-3-[[methyl-[2-[methyl-[[1-[3-(trifluoromethyl)phenyl]indol-3-yl]methyl]amino]ethyl]amino]methyl]chromen-4-one;dihydrochloride Chemical compound Cl.Cl.C=1OC2=CC(C)=C(C)C=C2C(=O)C=1CN(C)CCN(C)CC(C1=CC=CC=C11)=CN1C1=CC=CC(C(F)(F)F)=C1 GOZMBJCYMQQACI-UHFFFAOYSA-N 0.000 description 1
- KXBCLNRMQPRVTP-UHFFFAOYSA-N 6-amino-1,5-dihydroimidazo[4,5-c]pyridin-4-one Chemical compound O=C1NC(N)=CC2=C1N=CN2 KXBCLNRMQPRVTP-UHFFFAOYSA-N 0.000 description 1
- DCPSTSVLRXOYGS-UHFFFAOYSA-N 6-amino-1h-pyrimidine-2-thione Chemical compound NC1=CC=NC(S)=N1 DCPSTSVLRXOYGS-UHFFFAOYSA-N 0.000 description 1
- QNNARSZPGNJZIX-UHFFFAOYSA-N 6-amino-5-prop-1-ynyl-1h-pyrimidin-2-one Chemical compound CC#CC1=CNC(=O)N=C1N QNNARSZPGNJZIX-UHFFFAOYSA-N 0.000 description 1
- LOSIULRWFAEMFL-UHFFFAOYSA-N 7-deazaguanine Chemical compound O=C1NC(N)=NC2=C1CC=N2 LOSIULRWFAEMFL-UHFFFAOYSA-N 0.000 description 1
- HRYKDUPGBWLLHO-UHFFFAOYSA-N 8-azaadenine Chemical compound NC1=NC=NC2=NNN=C12 HRYKDUPGBWLLHO-UHFFFAOYSA-N 0.000 description 1
- LPXQRXLUHJKZIE-UHFFFAOYSA-N 8-azaguanine Chemical compound NC1=NC(O)=C2NN=NC2=N1 LPXQRXLUHJKZIE-UHFFFAOYSA-N 0.000 description 1
- 229960005508 8-azaguanine Drugs 0.000 description 1
- MSSXOMSJDRHRMC-UHFFFAOYSA-N 9H-purine-2,6-diamine Chemical compound NC1=NC(N)=C2NC=NC2=N1 MSSXOMSJDRHRMC-UHFFFAOYSA-N 0.000 description 1
- 101710190443 Acetyl-CoA carboxylase 1 Proteins 0.000 description 1
- GUBGYTABKSRVRQ-XLOQQCSPSA-N Alpha-Lactose Chemical compound O[C@@H]1[C@@H](O)[C@@H](O)[C@@H](CO)O[C@H]1O[C@@H]1[C@@H](CO)O[C@H](O)[C@H](O)[C@H]1O GUBGYTABKSRVRQ-XLOQQCSPSA-N 0.000 description 1
- QGZKDVFQNNGYKY-UHFFFAOYSA-O Ammonium Chemical compound [NH4+] QGZKDVFQNNGYKY-UHFFFAOYSA-O 0.000 description 1
- 108020000948 Antisense Oligonucleotides Proteins 0.000 description 1
- 210000002237 B-cell of pancreatic islet Anatomy 0.000 description 1
- 102100021334 Bcl-2-related protein A1 Human genes 0.000 description 1
- 108091003079 Bovine Serum Albumin Proteins 0.000 description 1
- COVZYZSDYWQREU-UHFFFAOYSA-N Busulfan Chemical compound CS(=O)(=O)OCCCCOS(C)(=O)=O COVZYZSDYWQREU-UHFFFAOYSA-N 0.000 description 1
- 239000004322 Butylated hydroxytoluene Substances 0.000 description 1
- NLZUEZXRPGMBCV-UHFFFAOYSA-N Butylhydroxytoluene Chemical compound CC1=CC(C(C)(C)C)=C(O)C(C(C)(C)C)=C1 NLZUEZXRPGMBCV-UHFFFAOYSA-N 0.000 description 1
- 102100040840 C-type lectin domain family 7 member A Human genes 0.000 description 1
- 238000011740 C57BL/6 mouse Methods 0.000 description 1
- 101100205088 Caenorhabditis elegans iars-1 gene Proteins 0.000 description 1
- 101100504320 Caenorhabditis elegans mcp-1 gene Proteins 0.000 description 1
- UXVMQQNJUSDDNG-UHFFFAOYSA-L Calcium chloride Chemical compound [Cl-].[Cl-].[Ca+2] UXVMQQNJUSDDNG-UHFFFAOYSA-L 0.000 description 1
- 108010004103 Chylomicrons Proteins 0.000 description 1
- 108091026890 Coding region Proteins 0.000 description 1
- 102000008186 Collagen Human genes 0.000 description 1
- 108010035532 Collagen Proteins 0.000 description 1
- 108020004394 Complementary RNA Proteins 0.000 description 1
- SBJKKFFYIZUCET-JLAZNSOCSA-N Dehydro-L-ascorbic acid Chemical compound OC[C@H](O)[C@H]1OC(=O)C(=O)C1=O SBJKKFFYIZUCET-JLAZNSOCSA-N 0.000 description 1
- 241000702421 Dependoparvovirus Species 0.000 description 1
- 108010067722 Dipeptidyl Peptidase 4 Proteins 0.000 description 1
- 102100025012 Dipeptidyl peptidase 4 Human genes 0.000 description 1
- 201000011001 Ebola Hemorrhagic Fever Diseases 0.000 description 1
- 102100038132 Endogenous retrovirus group K member 6 Pro protein Human genes 0.000 description 1
- 102100031780 Endonuclease Human genes 0.000 description 1
- 108010042407 Endonucleases Proteins 0.000 description 1
- 244000148064 Enicostema verticillatum Species 0.000 description 1
- 101710091045 Envelope protein Proteins 0.000 description 1
- 230000010665 Enzyme Interactions Effects 0.000 description 1
- 102000004190 Enzymes Human genes 0.000 description 1
- 108090000790 Enzymes Proteins 0.000 description 1
- 108700039887 Essential Genes Proteins 0.000 description 1
- PIICEJLVQHRZGT-UHFFFAOYSA-N Ethylenediamine Chemical compound NCCN PIICEJLVQHRZGT-UHFFFAOYSA-N 0.000 description 1
- 102100040133 Free fatty acid receptor 2 Human genes 0.000 description 1
- 102100040136 Free fatty acid receptor 3 Human genes 0.000 description 1
- 102100033864 G-protein coupled receptor 84 Human genes 0.000 description 1
- 108010010803 Gelatin Proteins 0.000 description 1
- 102100036264 Glucose-6-phosphatase catalytic subunit 1 Human genes 0.000 description 1
- 101710099339 Glucose-6-phosphatase catalytic subunit 1 Proteins 0.000 description 1
- ZWZWYGMENQVNFU-UHFFFAOYSA-N Glycerophosphorylserin Natural products OC(=O)C(N)COP(O)(=O)OCC(O)CO ZWZWYGMENQVNFU-UHFFFAOYSA-N 0.000 description 1
- 241000282412 Homo Species 0.000 description 1
- 101000749325 Homo sapiens C-type lectin domain family 7 member A Proteins 0.000 description 1
- 101000890668 Homo sapiens Free fatty acid receptor 2 Proteins 0.000 description 1
- 101000890662 Homo sapiens Free fatty acid receptor 3 Proteins 0.000 description 1
- 101001069589 Homo sapiens G-protein coupled receptor 84 Proteins 0.000 description 1
- 101000603962 Homo sapiens Oxysterols receptor LXR-alpha Proteins 0.000 description 1
- 101000635799 Homo sapiens Run domain Beclin-1-interacting and cysteine-rich domain-containing protein Proteins 0.000 description 1
- 206010060378 Hyperinsulinaemia Diseases 0.000 description 1
- UGQMRVRMYYASKQ-UHFFFAOYSA-N Hypoxanthine nucleoside Natural products OC1C(O)C(CO)OC1N1C(NC=NC2=O)=C2N=C1 UGQMRVRMYYASKQ-UHFFFAOYSA-N 0.000 description 1
- 101150030450 IRS1 gene Proteins 0.000 description 1
- 102000008394 Immunoglobulin Fragments Human genes 0.000 description 1
- 108010021625 Immunoglobulin Fragments Proteins 0.000 description 1
- 102100037850 Interferon gamma Human genes 0.000 description 1
- 108010074328 Interferon-gamma Proteins 0.000 description 1
- HNDVDQJCIGZPNO-YFKPBYRVSA-N L-histidine Chemical compound OC(=O)[C@@H](N)CC1=CN=CN1 HNDVDQJCIGZPNO-YFKPBYRVSA-N 0.000 description 1
- GUBGYTABKSRVRQ-QKKXKWKRSA-N Lactose Natural products OC[C@H]1O[C@@H](O[C@H]2[C@H](O)[C@@H](O)C(O)O[C@@H]2CO)[C@H](O)[C@@H](O)[C@H]1O GUBGYTABKSRVRQ-QKKXKWKRSA-N 0.000 description 1
- 108091026898 Leader sequence (mRNA) Proteins 0.000 description 1
- 108010013563 Lipoprotein Lipase Proteins 0.000 description 1
- 102100022119 Lipoprotein lipase Human genes 0.000 description 1
- WHXSMMKQMYFTQS-UHFFFAOYSA-N Lithium Chemical compound [Li] WHXSMMKQMYFTQS-UHFFFAOYSA-N 0.000 description 1
- 108060001084 Luciferase Proteins 0.000 description 1
- 229920005479 Lucite® Polymers 0.000 description 1
- KDXKERNSBIXSRK-UHFFFAOYSA-N Lysine Natural products NCCCCC(N)C(O)=O KDXKERNSBIXSRK-UHFFFAOYSA-N 0.000 description 1
- 239000004472 Lysine Substances 0.000 description 1
- 210000004322 M2 macrophage Anatomy 0.000 description 1
- 102000019149 MAP kinase activity proteins Human genes 0.000 description 1
- 108040008097 MAP kinase activity proteins Proteins 0.000 description 1
- 108091054455 MAP kinase family Proteins 0.000 description 1
- 102000043136 MAP kinase family Human genes 0.000 description 1
- FYYHWMGAXLPEAU-UHFFFAOYSA-N Magnesium Chemical compound [Mg] FYYHWMGAXLPEAU-UHFFFAOYSA-N 0.000 description 1
- 241000124008 Mammalia Species 0.000 description 1
- 108010031099 Mannose Receptor Proteins 0.000 description 1
- 102000018697 Membrane Proteins Human genes 0.000 description 1
- 108010052285 Membrane Proteins Proteins 0.000 description 1
- 241000714177 Murine leukemia virus Species 0.000 description 1
- 101100227062 Mus musculus Ffar4 gene Proteins 0.000 description 1
- 235000021360 Myristic acid Nutrition 0.000 description 1
- TUNFSRHWOTWDNC-UHFFFAOYSA-N Myristic acid Natural products CCCCCCCCCCCCCC(O)=O TUNFSRHWOTWDNC-UHFFFAOYSA-N 0.000 description 1
- GXCLVBGFBYZDAG-UHFFFAOYSA-N N-[2-(1H-indol-3-yl)ethyl]-N-methylprop-2-en-1-amine Chemical compound CN(CCC1=CNC2=C1C=CC=C2)CC=C GXCLVBGFBYZDAG-UHFFFAOYSA-N 0.000 description 1
- 101710163270 Nuclease Proteins 0.000 description 1
- 239000005642 Oleic acid Substances 0.000 description 1
- 241000283973 Oryctolagus cuniculus Species 0.000 description 1
- 240000007594 Oryza sativa Species 0.000 description 1
- 235000007164 Oryza sativa Nutrition 0.000 description 1
- 102000016979 Other receptors Human genes 0.000 description 1
- 102100038476 Oxysterols receptor LXR-alpha Human genes 0.000 description 1
- 239000012828 PI3K inhibitor Substances 0.000 description 1
- 239000007990 PIPES buffer Substances 0.000 description 1
- 229910019142 PO4 Inorganic materials 0.000 description 1
- 229930040373 Paraformaldehyde Natural products 0.000 description 1
- 235000019483 Peanut oil Nutrition 0.000 description 1
- 108010079855 Peptide Aptamers Proteins 0.000 description 1
- 102000003993 Phosphatidylinositol 3-kinases Human genes 0.000 description 1
- 108090000430 Phosphatidylinositol 3-kinases Proteins 0.000 description 1
- 108090000472 Phosphoenolpyruvate carboxykinase (ATP) Proteins 0.000 description 1
- 102100034792 Phosphoenolpyruvate carboxykinase [GTP], mitochondrial Human genes 0.000 description 1
- 102000004160 Phosphoric Monoester Hydrolases Human genes 0.000 description 1
- 108090000608 Phosphoric Monoester Hydrolases Proteins 0.000 description 1
- ABLZXFCXXLZCGV-UHFFFAOYSA-N Phosphorous acid Chemical class OP(O)=O ABLZXFCXXLZCGV-UHFFFAOYSA-N 0.000 description 1
- OAICVXFJPJFONN-UHFFFAOYSA-N Phosphorus Chemical compound [P] OAICVXFJPJFONN-UHFFFAOYSA-N 0.000 description 1
- 108091000080 Phosphotransferase Proteins 0.000 description 1
- 102000009516 Protein Serine-Threonine Kinases Human genes 0.000 description 1
- 108010009341 Protein Serine-Threonine Kinases Proteins 0.000 description 1
- 101710188315 Protein X Proteins 0.000 description 1
- CZPWVGJYEJSRLH-UHFFFAOYSA-N Pyrimidine Chemical compound C1=CN=CN=C1 CZPWVGJYEJSRLH-UHFFFAOYSA-N 0.000 description 1
- 238000001190 Q-PCR Methods 0.000 description 1
- 238000002123 RNA extraction Methods 0.000 description 1
- 206010037742 Rabies Diseases 0.000 description 1
- 241000700159 Rattus Species 0.000 description 1
- 102100030852 Run domain Beclin-1-interacting and cysteine-rich domain-containing protein Human genes 0.000 description 1
- VYPSYNLAJGMNEJ-UHFFFAOYSA-N Silicium dioxide Chemical compound O=[Si]=O VYPSYNLAJGMNEJ-UHFFFAOYSA-N 0.000 description 1
- DWAQJAXMDSEUJJ-UHFFFAOYSA-M Sodium bisulfite Chemical compound [Na+].OS([O-])=O DWAQJAXMDSEUJJ-UHFFFAOYSA-M 0.000 description 1
- 241000251131 Sphyrna Species 0.000 description 1
- 229920002472 Starch Polymers 0.000 description 1
- 229930006000 Sucrose Natural products 0.000 description 1
- CZMRCDWAGMRECN-UGDNZRGBSA-N Sucrose Chemical compound O[C@H]1[C@H](O)[C@@H](CO)O[C@@]1(CO)O[C@@H]1[C@H](O)[C@@H](O)[C@H](O)[C@@H](CO)O1 CZMRCDWAGMRECN-UGDNZRGBSA-N 0.000 description 1
- UCKMPCXJQFINFW-UHFFFAOYSA-N Sulphide Chemical compound [S-2] UCKMPCXJQFINFW-UHFFFAOYSA-N 0.000 description 1
- 102000019197 Superoxide Dismutase Human genes 0.000 description 1
- 108010012715 Superoxide dismutase Proteins 0.000 description 1
- 101710178958 TGF-beta-activated kinase 1 and MAP3K7-binding protein 1 Proteins 0.000 description 1
- 108700012920 TNF Proteins 0.000 description 1
- FEWJPZIEWOKRBE-UHFFFAOYSA-N Tartaric Acid Chemical class [H+].[H+].[O-]C(=O)C(O)C(O)C([O-])=O FEWJPZIEWOKRBE-UHFFFAOYSA-N 0.000 description 1
- 229940123464 Thiazolidinedione Drugs 0.000 description 1
- 108091036066 Three prime untranslated region Proteins 0.000 description 1
- 229920004890 Triton X-100 Polymers 0.000 description 1
- 239000013504 Triton X-100 Substances 0.000 description 1
- GBOGMAARMMDZGR-UHFFFAOYSA-N UNPD149280 Natural products N1C(=O)C23OC(=O)C=CC(O)CCCC(C)CC=CC3C(O)C(=C)C(C)C2C1CC1=CC=CC=C1 GBOGMAARMMDZGR-UHFFFAOYSA-N 0.000 description 1
- 241000700605 Viruses Species 0.000 description 1
- HCHKCACWOHOZIP-UHFFFAOYSA-N Zinc Chemical compound [Zn] HCHKCACWOHOZIP-UHFFFAOYSA-N 0.000 description 1
- 238000010521 absorption reaction Methods 0.000 description 1
- QPMSXSBEVQLBIL-CZRHPSIPSA-N ac1mix0p Chemical compound C1=CC=C2N(C[C@H](C)CN(C)C)C3=CC(OC)=CC=C3SC2=C1.O([C@H]1[C@]2(OC)C=CC34C[C@@H]2[C@](C)(O)CCC)C2=C5[C@]41CCN(C)[C@@H]3CC5=CC=C2O QPMSXSBEVQLBIL-CZRHPSIPSA-N 0.000 description 1
- 238000009825 accumulation Methods 0.000 description 1
- 235000011054 acetic acid Nutrition 0.000 description 1
- 150000001242 acetic acid derivatives Chemical class 0.000 description 1
- 230000002378 acidificating effect Effects 0.000 description 1
- 239000008186 active pharmaceutical agent Substances 0.000 description 1
- 239000000654 additive Substances 0.000 description 1
- 230000000996 additive effect Effects 0.000 description 1
- 239000002671 adjuvant Substances 0.000 description 1
- 230000008484 agonism Effects 0.000 description 1
- 150000001336 alkenes Chemical class 0.000 description 1
- 125000005083 alkoxyalkoxy group Chemical group 0.000 description 1
- 125000002877 alkyl aryl group Chemical group 0.000 description 1
- 125000005600 alkyl phosphonate group Chemical group 0.000 description 1
- 239000000956 alloy Substances 0.000 description 1
- 229910045601 alloy Inorganic materials 0.000 description 1
- 230000001668 ameliorated effect Effects 0.000 description 1
- 150000001413 amino acids Chemical group 0.000 description 1
- 125000005122 aminoalkylamino group Chemical group 0.000 description 1
- 239000005557 antagonist Substances 0.000 description 1
- 239000003242 anti bacterial agent Substances 0.000 description 1
- 230000003178 anti-diabetic effect Effects 0.000 description 1
- 239000000611 antibody drug conjugate Substances 0.000 description 1
- 229940049595 antibody-drug conjugate Drugs 0.000 description 1
- 239000003472 antidiabetic agent Substances 0.000 description 1
- 239000003963 antioxidant agent Substances 0.000 description 1
- 235000006708 antioxidants Nutrition 0.000 description 1
- 239000000074 antisense oligonucleotide Substances 0.000 description 1
- 238000012230 antisense oligonucleotides Methods 0.000 description 1
- 230000006907 apoptotic process Effects 0.000 description 1
- 229940114079 arachidonic acid Drugs 0.000 description 1
- 125000003710 aryl alkyl group Chemical group 0.000 description 1
- 235000010323 ascorbic acid Nutrition 0.000 description 1
- 239000011668 ascorbic acid Substances 0.000 description 1
- 229960005070 ascorbic acid Drugs 0.000 description 1
- 230000002238 attenuated effect Effects 0.000 description 1
- 230000037429 base substitution Effects 0.000 description 1
- 230000009286 beneficial effect Effects 0.000 description 1
- 230000008901 benefit Effects 0.000 description 1
- WPYMKLBDIGXBTP-UHFFFAOYSA-N benzoic acid group Chemical group C(C1=CC=CC=C1)(=O)O WPYMKLBDIGXBTP-UHFFFAOYSA-N 0.000 description 1
- 235000019445 benzyl alcohol Nutrition 0.000 description 1
- 229960004217 benzyl alcohol Drugs 0.000 description 1
- 230000015572 biosynthetic process Effects 0.000 description 1
- 235000010354 butylated hydroxytoluene Nutrition 0.000 description 1
- 229940095259 butylated hydroxytoluene Drugs 0.000 description 1
- 239000001110 calcium chloride Substances 0.000 description 1
- 229910001628 calcium chloride Inorganic materials 0.000 description 1
- 238000003965 capillary gas chromatography Methods 0.000 description 1
- 125000003178 carboxy group Chemical group [H]OC(*)=O 0.000 description 1
- 238000012754 cardiac puncture Methods 0.000 description 1
- 230000001364 causal effect Effects 0.000 description 1
- 239000013592 cell lysate Substances 0.000 description 1
- 230000005754 cellular signaling Effects 0.000 description 1
- 239000000919 ceramic Substances 0.000 description 1
- 230000008859 change Effects 0.000 description 1
- 239000002738 chelating agent Substances 0.000 description 1
- 239000002975 chemoattractant Substances 0.000 description 1
- 230000003399 chemotactic effect Effects 0.000 description 1
- VDANGULDQQJODZ-UHFFFAOYSA-N chloroprocaine Chemical compound CCN(CC)CCOC(=O)C1=CC=C(N)C=C1Cl VDANGULDQQJODZ-UHFFFAOYSA-N 0.000 description 1
- 229960002023 chloroprocaine Drugs 0.000 description 1
- OEYIOHPDSNJKLS-UHFFFAOYSA-N choline Chemical compound C[N+](C)(C)CCO OEYIOHPDSNJKLS-UHFFFAOYSA-N 0.000 description 1
- 229960001231 choline Drugs 0.000 description 1
- 150000001860 citric acid derivatives Chemical class 0.000 description 1
- 230000008045 co-localization Effects 0.000 description 1
- 229920001436 collagen Polymers 0.000 description 1
- 239000003184 complementary RNA Substances 0.000 description 1
- 239000000470 constituent Substances 0.000 description 1
- 238000012937 correction Methods 0.000 description 1
- 230000008878 coupling Effects 0.000 description 1
- 238000010168 coupling process Methods 0.000 description 1
- 238000005859 coupling reaction Methods 0.000 description 1
- 238000012258 culturing Methods 0.000 description 1
- 238000013211 curve analysis Methods 0.000 description 1
- WZHCOOQXZCIUNC-UHFFFAOYSA-N cyclandelate Chemical compound C1C(C)(C)CC(C)CC1OC(=O)C(O)C1=CC=CC=C1 WZHCOOQXZCIUNC-UHFFFAOYSA-N 0.000 description 1
- 125000001995 cyclobutyl group Chemical group [H]C1([H])C([H])([H])C([H])(*)C1([H])[H] 0.000 description 1
- GBOGMAARMMDZGR-JREHFAHYSA-N cytochalasin B Natural products C[C@H]1CCC[C@@H](O)C=CC(=O)O[C@@]23[C@H](C=CC1)[C@H](O)C(=C)[C@@H](C)[C@@H]2[C@H](Cc4ccccc4)NC3=O GBOGMAARMMDZGR-JREHFAHYSA-N 0.000 description 1
- GBOGMAARMMDZGR-TYHYBEHESA-N cytochalasin B Chemical compound C([C@H]1[C@@H]2[C@@H](C([C@@H](O)[C@@H]3/C=C/C[C@H](C)CCC[C@@H](O)/C=C/C(=O)O[C@@]23C(=O)N1)=C)C)C1=CC=CC=C1 GBOGMAARMMDZGR-TYHYBEHESA-N 0.000 description 1
- 230000009089 cytolysis Effects 0.000 description 1
- 238000012303 cytoplasmic staining Methods 0.000 description 1
- 210000000172 cytosol Anatomy 0.000 description 1
- 230000001086 cytosolic effect Effects 0.000 description 1
- 238000007405 data analysis Methods 0.000 description 1
- 230000002950 deficient Effects 0.000 description 1
- 230000003111 delayed effect Effects 0.000 description 1
- 210000004443 dendritic cell Anatomy 0.000 description 1
- 239000005547 deoxyribonucleotide Substances 0.000 description 1
- 125000002637 deoxyribonucleotide group Chemical group 0.000 description 1
- 238000000586 desensitisation Methods 0.000 description 1
- 238000001514 detection method Methods 0.000 description 1
- 235000021045 dietary change Nutrition 0.000 description 1
- 230000000378 dietary effect Effects 0.000 description 1
- ZBCBWPMODOFKDW-UHFFFAOYSA-N diethanolamine Chemical compound OCCNCCO ZBCBWPMODOFKDW-UHFFFAOYSA-N 0.000 description 1
- 229940043237 diethanolamine Drugs 0.000 description 1
- 230000001079 digestive effect Effects 0.000 description 1
- 208000010643 digestive system disease Diseases 0.000 description 1
- 239000003085 diluting agent Substances 0.000 description 1
- 108010054812 diprotin A Proteins 0.000 description 1
- ZGSPNIOCEDOHGS-UHFFFAOYSA-L disodium [3-[2,3-di(octadeca-9,12-dienoyloxy)propoxy-oxidophosphoryl]oxy-2-hydroxypropyl] 2,3-di(octadeca-9,12-dienoyloxy)propyl phosphate Chemical compound [Na+].[Na+].CCCCCC=CCC=CCCCCCCCC(=O)OCC(OC(=O)CCCCCCCC=CCC=CCCCCC)COP([O-])(=O)OCC(O)COP([O-])(=O)OCC(OC(=O)CCCCCCCC=CCC=CCCCCC)COC(=O)CCCCCCCC=CCC=CCCCCC ZGSPNIOCEDOHGS-UHFFFAOYSA-L 0.000 description 1
- 229910000397 disodium phosphate Inorganic materials 0.000 description 1
- BFMYDTVEBKDAKJ-UHFFFAOYSA-L disodium;(2',7'-dibromo-3',6'-dioxido-3-oxospiro[2-benzofuran-1,9'-xanthene]-4'-yl)mercury;hydrate Chemical compound O.[Na+].[Na+].O1C(=O)C2=CC=CC=C2C21C1=CC(Br)=C([O-])C([Hg])=C1OC1=C2C=C(Br)C([O-])=C1 BFMYDTVEBKDAKJ-UHFFFAOYSA-L 0.000 description 1
- NAGJZTKCGNOGPW-UHFFFAOYSA-N dithiophosphoric acid Chemical class OP(O)(S)=S NAGJZTKCGNOGPW-UHFFFAOYSA-N 0.000 description 1
- 239000002552 dosage form Substances 0.000 description 1
- 230000003828 downregulation Effects 0.000 description 1
- 230000007783 downstream signaling Effects 0.000 description 1
- 230000000081 effect on glucose Effects 0.000 description 1
- 229920001971 elastomer Polymers 0.000 description 1
- 239000003995 emulsifying agent Substances 0.000 description 1
- 230000012202 endocytosis Effects 0.000 description 1
- 239000006274 endogenous ligand Substances 0.000 description 1
- 210000002889 endothelial cell Anatomy 0.000 description 1
- 238000012407 engineering method Methods 0.000 description 1
- 230000002708 enhancing effect Effects 0.000 description 1
- 229940088598 enzyme Drugs 0.000 description 1
- 229940012017 ethylenediamine Drugs 0.000 description 1
- 239000013604 expression vector Substances 0.000 description 1
- 210000004905 finger nail Anatomy 0.000 description 1
- 235000013312 flour Nutrition 0.000 description 1
- 238000000684 flow cytometry Methods 0.000 description 1
- 239000011888 foil Substances 0.000 description 1
- 238000013467 fragmentation Methods 0.000 description 1
- 238000006062 fragmentation reaction Methods 0.000 description 1
- 230000004927 fusion Effects 0.000 description 1
- 239000007789 gas Substances 0.000 description 1
- 238000004817 gas chromatography Methods 0.000 description 1
- 230000002496 gastric effect Effects 0.000 description 1
- 238000001502 gel electrophoresis Methods 0.000 description 1
- 239000008273 gelatin Substances 0.000 description 1
- 229920000159 gelatin Polymers 0.000 description 1
- 235000019322 gelatine Nutrition 0.000 description 1
- 235000011852 gelatine desserts Nutrition 0.000 description 1
- 230000030279 gene silencing Effects 0.000 description 1
- 238000007429 general method Methods 0.000 description 1
- YQEMORVAKMFKLG-UHFFFAOYSA-N glycerine monostearate Natural products CCCCCCCCCCCCCCCCCC(=O)OC(CO)CO YQEMORVAKMFKLG-UHFFFAOYSA-N 0.000 description 1
- SVUQHVRAGMNPLW-UHFFFAOYSA-N glycerol monostearate Natural products CCCCCCCCCCCCCCCCC(=O)OCC(O)CO SVUQHVRAGMNPLW-UHFFFAOYSA-N 0.000 description 1
- 244000144993 groups of animals Species 0.000 description 1
- 239000001963 growth medium Substances 0.000 description 1
- 229940093915 gynecological organic acid Drugs 0.000 description 1
- 238000003306 harvesting Methods 0.000 description 1
- 230000036541 health Effects 0.000 description 1
- 238000007490 hematoxylin and eosin (H&E) staining Methods 0.000 description 1
- 210000003547 hepatic macrophage Anatomy 0.000 description 1
- 125000000623 heterocyclic group Chemical group 0.000 description 1
- 125000000592 heterocycloalkyl group Chemical group 0.000 description 1
- 238000010842 high-capacity cDNA reverse transcription kit Methods 0.000 description 1
- HNDVDQJCIGZPNO-UHFFFAOYSA-N histidine Natural products OC(=O)C(N)CC1=CN=CN1 HNDVDQJCIGZPNO-UHFFFAOYSA-N 0.000 description 1
- 230000002962 histologic effect Effects 0.000 description 1
- 235000011167 hydrochloric acid Nutrition 0.000 description 1
- 150000004679 hydroxides Chemical class 0.000 description 1
- WGCNASOHLSPBMP-UHFFFAOYSA-N hydroxyacetaldehyde Natural products OCC=O WGCNASOHLSPBMP-UHFFFAOYSA-N 0.000 description 1
- 230000003451 hyperinsulinaemic effect Effects 0.000 description 1
- 201000008980 hyperinsulinism Diseases 0.000 description 1
- 238000003365 immunocytochemistry Methods 0.000 description 1
- 238000010166 immunofluorescence Methods 0.000 description 1
- 230000008595 infiltration Effects 0.000 description 1
- 238000001764 infiltration Methods 0.000 description 1
- 239000004615 ingredient Substances 0.000 description 1
- 230000000977 initiatory effect Effects 0.000 description 1
- 230000003914 insulin secretion Effects 0.000 description 1
- 230000010354 integration Effects 0.000 description 1
- 239000000138 intercalating agent Substances 0.000 description 1
- 229940100601 interleukin-6 Drugs 0.000 description 1
- 239000007925 intracardiac injection Substances 0.000 description 1
- 238000010212 intracellular staining Methods 0.000 description 1
- 230000010189 intracellular transport Effects 0.000 description 1
- 238000007912 intraperitoneal administration Methods 0.000 description 1
- 125000001449 isopropyl group Chemical group [H]C([H])([H])C([H])(*)C([H])([H])[H] 0.000 description 1
- JJWLVOIRVHMVIS-UHFFFAOYSA-N isopropylamine Chemical compound CC(C)N JJWLVOIRVHMVIS-UHFFFAOYSA-N 0.000 description 1
- 208000017169 kidney disease Diseases 0.000 description 1
- 239000008101 lactose Substances 0.000 description 1
- 125000005647 linker group Chemical group 0.000 description 1
- 229960004488 linolenic acid Drugs 0.000 description 1
- 230000037356 lipid metabolism Effects 0.000 description 1
- 229910052744 lithium Inorganic materials 0.000 description 1
- 238000003468 luciferase reporter gene assay Methods 0.000 description 1
- 210000004072 lung Anatomy 0.000 description 1
- 230000022275 macrophage chemotaxis Effects 0.000 description 1
- 239000011777 magnesium Substances 0.000 description 1
- 229910052749 magnesium Inorganic materials 0.000 description 1
- 229910001629 magnesium chloride Inorganic materials 0.000 description 1
- 238000012423 maintenance Methods 0.000 description 1
- 210000004962 mammalian cell Anatomy 0.000 description 1
- 229960003194 meglumine Drugs 0.000 description 1
- 230000004060 metabolic process Effects 0.000 description 1
- 150000002739 metals Chemical class 0.000 description 1
- 125000000956 methoxy group Chemical group [H]C([H])([H])O* 0.000 description 1
- 235000010270 methyl p-hydroxybenzoate Nutrition 0.000 description 1
- 239000002480 mineral oil Substances 0.000 description 1
- 235000010446 mineral oil Nutrition 0.000 description 1
- 150000007522 mineralic acids Chemical class 0.000 description 1
- 238000012544 monitoring process Methods 0.000 description 1
- 239000000178 monomer Substances 0.000 description 1
- 229910000402 monopotassium phosphate Inorganic materials 0.000 description 1
- 210000000663 muscle cell Anatomy 0.000 description 1
- 210000003098 myoblast Anatomy 0.000 description 1
- 230000031990 negative regulation of inflammatory response Effects 0.000 description 1
- 238000007899 nucleic acid hybridization Methods 0.000 description 1
- 235000019198 oils Nutrition 0.000 description 1
- ZQPPMHVWECSIRJ-KTKRTIGZSA-N oleic acid group Chemical group C(CCCCCCC\C=C/CCCCCCCC)(=O)O ZQPPMHVWECSIRJ-KTKRTIGZSA-N 0.000 description 1
- 239000006014 omega-3 oil Substances 0.000 description 1
- 238000001543 one-way ANOVA Methods 0.000 description 1
- 150000007524 organic acids Chemical class 0.000 description 1
- 235000005985 organic acids Nutrition 0.000 description 1
- 229920000620 organic polymer Polymers 0.000 description 1
- 125000001181 organosilyl group Chemical group [SiH3]* 0.000 description 1
- 235000006408 oxalic acid Nutrition 0.000 description 1
- 229910052760 oxygen Inorganic materials 0.000 description 1
- 239000006179 pH buffering agent Substances 0.000 description 1
- SECPZKHBENQXJG-FPLPWBNLSA-N palmitoleic acid Chemical compound CCCCCC\C=C/CCCCCCCC(O)=O SECPZKHBENQXJG-FPLPWBNLSA-N 0.000 description 1
- 229920002866 paraformaldehyde Polymers 0.000 description 1
- 239000000312 peanut oil Substances 0.000 description 1
- 239000000816 peptidomimetic Substances 0.000 description 1
- 230000002093 peripheral effect Effects 0.000 description 1
- 239000003208 petroleum Substances 0.000 description 1
- 230000003285 pharmacodynamic effect Effects 0.000 description 1
- 235000021317 phosphate Nutrition 0.000 description 1
- WTJKGGKOPKCXLL-RRHRGVEJSA-N phosphatidylcholine Chemical compound CCCCCCCCCCCCCCCC(=O)OC[C@H](COP([O-])(=O)OCC[N+](C)(C)C)OC(=O)CCCCCCCC=CCCCCCCCC WTJKGGKOPKCXLL-RRHRGVEJSA-N 0.000 description 1
- 150000004713 phosphodiesters Chemical group 0.000 description 1
- 229940043441 phosphoinositide 3-kinase inhibitor Drugs 0.000 description 1
- 150000008298 phosphoramidates Chemical class 0.000 description 1
- 235000011007 phosphoric acid Nutrition 0.000 description 1
- 150000003013 phosphoric acid derivatives Chemical class 0.000 description 1
- 239000011574 phosphorus Substances 0.000 description 1
- 102000020233 phosphotransferase Human genes 0.000 description 1
- 230000035479 physiological effects, processes and functions Effects 0.000 description 1
- 230000036470 plasma concentration Effects 0.000 description 1
- 239000013612 plasmid Substances 0.000 description 1
- 230000010287 polarization Effects 0.000 description 1
- 229920002401 polyacrylamide Polymers 0.000 description 1
- 229940115272 polyinosinic:polycytidylic acid Drugs 0.000 description 1
- 229920000642 polymer Polymers 0.000 description 1
- 239000004926 polymethyl methacrylate Substances 0.000 description 1
- 229920002223 polystyrene Polymers 0.000 description 1
- 229920002981 polyvinylidene fluoride Polymers 0.000 description 1
- 238000001556 precipitation Methods 0.000 description 1
- 102000004196 processed proteins & peptides Human genes 0.000 description 1
- 238000012545 processing Methods 0.000 description 1
- 230000007114 proinflammatory cascade Effects 0.000 description 1
- 230000008741 proinflammatory signaling process Effects 0.000 description 1
- 230000001737 promoting effect Effects 0.000 description 1
- QQONPFPTGQHPMA-UHFFFAOYSA-N propylene Natural products CC=C QQONPFPTGQHPMA-UHFFFAOYSA-N 0.000 description 1
- 125000004805 propylene group Chemical group [H]C([H])([H])C([H])([*:1])C([H])([H])[*:2] 0.000 description 1
- 230000004853 protein function Effects 0.000 description 1
- 238000000164 protein isolation Methods 0.000 description 1
- 230000002685 pulmonary effect Effects 0.000 description 1
- 150000003212 purines Chemical class 0.000 description 1
- 238000011002 quantification Methods 0.000 description 1
- 230000033300 receptor internalization Effects 0.000 description 1
- 230000007115 recruitment Effects 0.000 description 1
- 125000006853 reporter group Chemical group 0.000 description 1
- 239000000837 restrainer Substances 0.000 description 1
- 230000000717 retained effect Effects 0.000 description 1
- 230000002441 reversible effect Effects 0.000 description 1
- PYWVYCXTNDRMGF-UHFFFAOYSA-N rhodamine B Chemical compound [Cl-].C=12C=CC(=[N+](CC)CC)C=C2OC2=CC(N(CC)CC)=CC=C2C=1C1=CC=CC=C1C(O)=O PYWVYCXTNDRMGF-UHFFFAOYSA-N 0.000 description 1
- 235000009566 rice Nutrition 0.000 description 1
- 229920002477 rna polymer Polymers 0.000 description 1
- 239000005060 rubber Substances 0.000 description 1
- 239000012723 sample buffer Substances 0.000 description 1
- 229920006395 saturated elastomer Polymers 0.000 description 1
- 238000010187 selection method Methods 0.000 description 1
- 239000008159 sesame oil Substances 0.000 description 1
- 235000011803 sesame oil Nutrition 0.000 description 1
- 239000000741 silica gel Substances 0.000 description 1
- 229910002027 silica gel Inorganic materials 0.000 description 1
- 235000020183 skimmed milk Nutrition 0.000 description 1
- 238000002415 sodium dodecyl sulfate polyacrylamide gel electrophoresis Methods 0.000 description 1
- 235000010267 sodium hydrogen sulphite Nutrition 0.000 description 1
- RYYKJJJTJZKILX-UHFFFAOYSA-M sodium octadecanoate Chemical compound [Na+].CCCCCCCCCCCCCCCCCC([O-])=O RYYKJJJTJZKILX-UHFFFAOYSA-M 0.000 description 1
- 239000007787 solid Substances 0.000 description 1
- 239000002904 solvent Substances 0.000 description 1
- 239000003549 soybean oil Substances 0.000 description 1
- 235000012424 soybean oil Nutrition 0.000 description 1
- 238000010561 standard procedure Methods 0.000 description 1
- 239000008107 starch Substances 0.000 description 1
- 235000019698 starch Nutrition 0.000 description 1
- 230000007863 steatosis Effects 0.000 description 1
- 231100000240 steatosis hepatitis Toxicity 0.000 description 1
- 238000003860 storage Methods 0.000 description 1
- 125000001424 substituent group Chemical group 0.000 description 1
- 239000005720 sucrose Substances 0.000 description 1
- IIACRCGMVDHOTQ-UHFFFAOYSA-N sulfamic acid Chemical group NS(O)(=O)=O IIACRCGMVDHOTQ-UHFFFAOYSA-N 0.000 description 1
- 150000003456 sulfonamides Chemical group 0.000 description 1
- BDHFUVZGWQCTTF-UHFFFAOYSA-M sulfonate Chemical compound [O-]S(=O)=O BDHFUVZGWQCTTF-UHFFFAOYSA-M 0.000 description 1
- 150000003457 sulfones Chemical group 0.000 description 1
- 150000003462 sulfoxides Chemical class 0.000 description 1
- 229910052717 sulfur Inorganic materials 0.000 description 1
- 239000006228 supernatant Substances 0.000 description 1
- 239000013589 supplement Substances 0.000 description 1
- 238000001356 surgical procedure Methods 0.000 description 1
- 238000003786 synthesis reaction Methods 0.000 description 1
- 230000002194 synthesizing effect Effects 0.000 description 1
- 230000009885 systemic effect Effects 0.000 description 1
- 239000000454 talc Substances 0.000 description 1
- 229910052623 talc Inorganic materials 0.000 description 1
- 235000002906 tartaric acid Nutrition 0.000 description 1
- 238000002560 therapeutic procedure Methods 0.000 description 1
- 238000013519 translation Methods 0.000 description 1
- 230000032258 transport Effects 0.000 description 1
- 125000000876 trifluoromethoxy group Chemical group FC(F)(F)O* 0.000 description 1
- 125000002023 trifluoromethyl group Chemical group FC(F)(F)* 0.000 description 1
- 230000010415 tropism Effects 0.000 description 1
- 241001529453 unidentified herpesvirus Species 0.000 description 1
- 241001430294 unidentified retrovirus Species 0.000 description 1
- 238000010200 validation analysis Methods 0.000 description 1
- 235000013311 vegetables Nutrition 0.000 description 1
- 230000003612 virological effect Effects 0.000 description 1
- 238000005406 washing Methods 0.000 description 1
- 238000009736 wetting Methods 0.000 description 1
- 239000000080 wetting agent Substances 0.000 description 1
- 229940075420 xanthine Drugs 0.000 description 1
- 229910052725 zinc Inorganic materials 0.000 description 1
- 239000011701 zinc Substances 0.000 description 1
Classifications
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/185—Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
- A61K31/19—Carboxylic acids, e.g. valproic acid
- A61K31/20—Carboxylic acids, e.g. valproic acid having a carboxyl group bound to a chain of seven or more carbon atoms, e.g. stearic, palmitic, arachidic acids
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P29/00—Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/185—Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
- A61K31/19—Carboxylic acids, e.g. valproic acid
- A61K31/20—Carboxylic acids, e.g. valproic acid having a carboxyl group bound to a chain of seven or more carbon atoms, e.g. stearic, palmitic, arachidic acids
- A61K31/202—Carboxylic acids, e.g. valproic acid having a carboxyl group bound to a chain of seven or more carbon atoms, e.g. stearic, palmitic, arachidic acids having three or more double bonds, e.g. linolenic
Definitions
- Sequence Listing which is a part of the present disclosure, includes a computer readable file "RUC_1 12WO_ST25.txt" generated by U.S. Patent & Trademark Office Patentin version 3.5 software comprising nucleotide and/or amino acid sequences of the present invention.
- the subject matter of the Sequence Listing is incorporated herein by reference in its entirety.
- the present teachings relate to methods of treating inflammatory conditions, including diabetes mellitus, by modulating P-arrestin2 and/or GPR120 mediated responses in a subject, as well as methods of screening compounds predicted to bind ⁇ - arrestin2 and/or GPR120.
- GPCRs G protein-coupled receptors
- the present teachings include methods of treating inflammation in a subject.
- the method comprises administering a therapeutically effective amount of a compound that binds GPR120, the compound selectively activating a P-arrestin2-dependent signaling pathway of GPR120.
- the compound that selectively activates a P-arrestin2-dependent signaling pathway is an co-3 fatty acid., including DHA and EPA.
- the compound can selectively activate a P-arrestin2-dependent signaling pathway and not activate a ⁇ -arrestinl -dependent signaling pathway.
- the inflammatory condition can be diabetes, an inflammatory condition is associated with obesity, and obesity.
- the subject is an animal, particularly a human.
- a method is also provided of treating an inflammatory condition associated with P-arrestin2 function.
- the method comprises administering a therapeutically effective amount of a -arrestin2 modulating agent to a subject in need thereof.
- the inflammatory condition can be diabetes, an inflammatory condition is associated with obesity, and obesity.
- the compound that selectively activates a P-arrestin2-dependent signaling pathway is an co-3 fatty acid., including DHA and EPA. The compound can selectively activate a P-arrestin2-dependent signaling pathway and not activate a ⁇ -arrestinl -dependent signaling pathway.
- the subject is an animal, particularly a human.
- a method is also provided of treating an inflammatory condition associated with P-arrestin2 function and GPR120 function.
- the method comprises administering a therapeutically effective amount of a P-arrestin2 modulating agent and a GPR120 modulating agent to a subject in need thereof.
- the inflammatory condition can be diabetes, an inflammatory condition is associated with obesity, and obesity.
- the compound that selectively activates a ⁇ - arrestin2-dependent signaling pathway is an co-3 fatty acid., including DHA and EPA. The compound can selectively activate a P-arrestin2-dependent signaling pathway and not activate a ⁇ -arrestinl -dependent signaling pathway.
- the subject is an animal, particularly a human.
- a method is provided of treating a p-arrestin2 mediated response.
- the method comprises administering to a subject in need thereof a therapeutically effective amount of a compound predicted to bind GPR120, the compound selectively activating a P-arrestin2-dependent signaling pathway of said
- the P-arrestin2 mediated response includes inflammation, including a lipopolysaccharide-induced inflammatory response and a TNFa-induced inflammatory response.
- a method is also provided of inhibiting cytokine secretion.
- the method comprising administering to a cell an effective amount of a compound predicted to bind GPR120, the compound selectively activating a beta-arrestin2- dependent signaling pathway of GPR120.
- the P-arrestin2 mediated response includes inflammation, including a lipopolysaccharide-induced inflammatory response.
- the cytokine secretion inhibited is a lipopolysaccharide- induced cytokine secretion.
- a method is also provided of screening a compound predicted to bind GPR120 for P-arrestin2 biased activity.
- the method comprises (a) administering to a subject an amount of a compound predicted to bind GPR120 sufficient to generate a physiological response, (b) evaluating at a first time said physiological response, (c) administering to said subject an effective amount of a dsRNA capable of binding a ⁇ - arrestin2 RNA, (d) allowing a period of time to pass sufficient for said dsRNA to substantially bind said P-arrestin2 RNA, and (e) evaluating at a second time said physiological response, wherein if said physiological response is reversed at said second time as compared to said first time, said compound predicted to bind GPR120 is a ⁇ - arrestin2 biased compound.
- the physiological response relates to a lipopolysaccharide-induced inflammatory response. In various aspects, the physiological response relates to a TNF-a-induced inflammatory response. In various aspects, the physiological response relates to a lipopolysaccharide-induced cytokine secretion.
- kits can comprise an effective amount of at least one dsRNA molecule capable of binding a P-arrestin2 RNA, and written indicia providing a user of said kit with instructions for using said kit in conjunction with the methods above.
- the kit can further comprise cells suitable for use in evaluating a compound predicted to bind GPR120, and provided in an amount sufficient for use in evaluating said GPR120 ligand.
- the cells can be mouse monocyte cells.
- the mouse monocyte cells are from cell line Raw264.7.
- a dsRNA is also provided for inhibiting functional p-arrestin2 protein expression in a cell.
- the dsRNA comprising a sense strand and an antisense strand, wherein said antisense strand comprises a region of complementarity having a sequence substantially complementary to a P-arrestin2 target sequence, wherein said sense strand is substantially complementary to said antisense strand, and wherein said dsRNA, upon contact with a cell expressing functional P-arrestin2 protein, inhibits functional -arrestin2 protein expression.
- the P-arrestin2 target sequence compries a sequence having SEQ ID NO. 2, or sequences complementary thereto.
- a method is provided of inhibiting expression of a P-arrestin2 gene in a cell.
- the method comprises introducing into the cell a dsRNA, wherein the dsRNA comprises two sequences that are compementary to each other and wherein a sense strand comprises a first sequence and an antisense strand comprises a second sequence comprising a region of complimentarity that is substantially
- the dsRNA comprises a sequence selected having SEQ ID NO. 2, or sequences complementary thereto.
- FIG. 1 Expression level of GPR120 and GPR120-mediated antiinflammatory response in RAW 264.7 cells.
- A The mRNA expression pattern of various lipid sensing GPCRs is shown in adipose tissue
- B CD1 lc+ bone marrow-derived dendritic cells (BMDCs), bone marrow-derived macrophages (BMDMs), IPMacs, 3T3-L1 preadipocytes, differentiated 3T3-L1 adipocytes, RAW 264.7 cells and L6 myocytes.
- BMDCs bone marrow-derived dendritic cells
- BMDMs bone marrow-derived macrophages
- IPMacs IPMacs
- 3T3-L1 preadipocytes differentiated 3T3-L1 adipocytes
- RAW 264.7 cells and L6 myocytes.
- Ribosomal protein S3 was used as internal control.
- C Expression of GPR120 in SVF, adipocytes and hepatic Kupffer cells from chow (NC)- or HFD-fed mice was examined by q-PCR. Data are expressed as the mean ⁇ SEM of at least three independent experiments in triplicate. *, p ⁇ 0.05 versus NC.
- D RAW 264.7 cells, transfected with scrambled (Scr) or GPR120 #2 siRNA (GPR120 KD), were treated with 100 ⁇ of GW9508 for 1 hr prior to LPS (100 ng/ml) treatment for 10 min and then subjected to western blotting. Left panel is a representative image from three independent experiments, and the scanned bar graph (right panel) shows fold induction over basal conditions.
- FIG. 1 Omega-3 FA stimulates GPR120 and mediates anti-inflammatory effects.
- A GPR120-mediated SRE-luc activity after treatment with various FAs (circles GPR120-GW9508; diamonds GPR120-DHA; triangles GPR120-EPA; squares GPR120- Palmitoleate; Xs GPR120-Oleic acid; empty diamonds GPR120-Arachidonic acid; empty circles GPR120-y-linoleic acid; asterics GPR120-Palmitic acid; empty squares GPR120- Myristic acid). Results are fold activities over basal. Each data point represents mean ⁇ SEM of three independent experiments performed in triplicate.
- Black lines indicate SRE-luc activities without GPR120 transfection or with non-stimulating FAs.
- DHA inhibits LPS- induced inflammatory signaling (B), cytokine secretion (C) and inflammatory gene mRNA expression level (D) in RAW 264.7 cells, but not in GPR120 knockdown cells.
- B LPS- induced inflammatory signaling
- C cytokine secretion
- D inflammatory gene mRNA expression level
- E and F GPR120 stimulation inhibits LPS-induced inflammatory response in WT primary macrophage.
- Data are expressed as the mean ⁇ SEM of three independent experiments. *, p ⁇ 0.05 versus LPS treatment in scrambled siR A transfected cells or WT IPMacs. See also Fig. 9.
- FIG. 1 GPR120 internalization with P-arrestin2 mediates antiinflammatory effects.
- RAW 264.7 cells were transfected with siRNA as indicated and stimulated with or without 100 ⁇ of DHA 1 hr prior to LPS (100 ng/ml) treatment for 10 min and then subjected to western blotting.
- B TNF-a secretion was measured in RAW 264.7 cell cultured media with or without RNA interference as indicated.
- C C
- HEK 293 cells were co-transfected with HA-GPR120 and ⁇ - arrestin2 » GFP to analyze GPR120 internalization after DHA stimulation for the indicated times. GPR120 (left column) and P-arrestin2 (middle column) were localized by confocal microscopy.
- E Co-immunoprecipitation between GPR120 and P-arrestin2 with DHA stimulation for 30 min in RAW 264.7 cells and,
- F HEK 293 cells (HA-GPR120 and ⁇ - arrestin2 » GFP), respectively. Lysate indicates 1/10 input in each experiment.
- FIG. 4 GPR120 activation enhances GLUT4 translocation and glucose uptake.
- A 3T3-L1 adipocytes were transfected with a dually tagged HA-GLUT4-GFP construct. Total GLUT4 expression was determined by GFP fluorescence, and GLUT4 translocation to the cell surface after 100 ng/ml insulin or 100 ⁇ DHA stimulation for 30 min was determined by indirect immunofluorescence of the HA-conjugated with Alexa 594 in fixed cells. Translocation following insulin stimulation was expressed as a percentage of the maximum response. Bar graph represents the mean ⁇ SEM data from four independent experiments. *, p ⁇ 0.05 vs. vehicle treatment.
- Glucose uptake was measured in WT and GPR120 KO mouse primary adipose tissue and in (C-F) 3T3-L1 adipocytes ⁇ siRNA with the indicated treatment. Data are expressed as mean ⁇ SEM of three independent experiments in triplicate. *, p ⁇ 0.05 vs. basal activity. The indicated siRNA knockdown efficiency was validated by western blotting. See also Fig. 10.
- FIG. 5 In vivo metabolic studies in GPR120 KO mice.
- B Insulin concentration were measured at the indicated time points and
- C area-under-curve analysis of the insulin data shows a significant difference between WT and GPR120 KO mice on NC.
- D
- FIG. Omega-3 FA enriched diet decreases inflammatory macrophage infiltration in adipose tissue.
- FIG. 7 Ml and M2 inflammatory gene expression levels in adipose tissue from WT vs. GPR120 KO mice.
- FIG 8 related to Figure 1. Validation of GPR120 siRNA knockdown efficiency.
- A Relative gene expression level of GPR120 and GPR40 in IPMac, RAW 264.7 cells, and 3T3-L1 adipocytes by q-PCR.
- B SiRNA transfection did not lead to an increase of IFN- ⁇ expression.
- FIG. 9 related to Figures 2 and 3.
- DHA-stimulated GPR120 inhibits TNF-a, TLR2 and TLR3-induced inflammation via a P-arrestin2 dependent mechanism.
- A ERK1/2 phosphorylation stimulated by GPR120 activation in RAW 264.7 cells transfected with scrambled or GPR120 siRNA. Cells were treated with 100 ⁇ of DHA, a-linolenic acid or palmitic acid for 5 min. Left panel is a representative image from three independent experiments, and the scanned bar graph (right panel) shows fold induction over the vehicle treatment. Data are expressed as the mean ⁇ SEM of three independent experiments. *, p ⁇ 0.05 versus scrambled siRNA transfected cells.
- FIG. 10 shows Akt phosphorylation in a GPR120-dependent manner.
- A Differentiated 3T3-L1 adipocytes were transfected with scrambled or GPR120 siRNA and then 48 hr after siRNA transfection, cells were treated with 100 ⁇ of DHA 30 min prior to insulin (3 ng/ml) treatment for 7 min followed by western blotting with phosphorylated Akt (S473) antibody.
- Left panel is a representative image from three independent experiments, and the scanned bar graph (right panel) shows fold induction over basal after normalization for total Akt. Data are expressed as the mean ⁇ SEM of three independent experiments. *, p ⁇ 0.05 versus basal.
- FIG. 1 related to Figure 5.
- WT or GPR120 KO mouse bone marrow (BM) was injected into irradiated WT C57B1/6 animals after 8 weeks for BM reconstitution, GPR120 was >90% reduced in circulating monocytes. Animals were then kept on either chow or switched to 60% HFD for 15 weeks ( ⁇ supplemention with ⁇ -3 FAs for 5 weeks).
- BMT GPR120 KO animals on chow are insulin resistant compared to WT controls.
- BMT GPR120 KO animals on HFD supplemented with co-3 FAs (+ ⁇ 3) are insulin resistant compared to WT controls. This effect is shown by decreased GIR, decreased IS-GDR, and a greater suppression of HGP in WT animals compared to KOs.
- C GPR120 expression levels in various tissues. RPS3 was the internal control for normalization. GPR120 is not expressed in soleus, or EDL muscle.
- D Glucose uptake is not altered by DHA treatment (100 ⁇ ) in L6 myocytes, compared to vehicle treated cells.
- FIG. 13 related to Figures 5 and 7.
- Omega-3 FAs decrease hepatic steatosis and metabolic gene expression, and increase M2 anti-inflammatory gene expression in liver.
- A Hepatic TAG was measured by lipidomic analysis in WT and GPR120 KO mice. Hepatic TAG was decreased in HFD+co3-fed WT, but not in GPR120 KOs. Data represent mean ⁇ SEM.*, p ⁇ 0.05 and ⁇ - ⁇ per group.
- B H&E staining of frozen sections of livers obtained from chow (NC), HFD and HFD+co3-fed WT and GPR120 KO mice.
- FIG. 14 related to Figure 7.
- G,” “C,” “A,” “T,” and “U” each generally stand for a nucleotide that contains guanine, cytosine, adenine, thymine, and uracil as a base, respectively. It will be understood, however, that the term “ribonucleotide” or “nucleotide” can also refer to a modified ribonucleotide, as further define below, or a surrogate replacement moiety.
- guanine, cytosine, adenine, thymine, and uracil may be replaced by other moieties without substantially altering the base pairing properties of an oligonucleotide comprising a nucleotide bearing such replacement moiety.
- a nucleotide comprising inosine as its base may base pair with nucleotides containing adenine, cytosine, or uracil.
- nucleotides containing uracil, guanine, or adenine may be replaced in the nucleotide sequence of the invention by a nucleotide containing, for example, inosine. Sequences comprising such replacement moieties are embodiments of the invention.
- Antisense Strand refers to the strand of a dsRNA which includes a region that is substantially complementary to a target sequence.
- region of complementarity refers to the region on the antisense strand that is substantially complementary to a sequence, for example a target sequence, as defined herein. Where the region of complementarity is not fully
- the mismatches are most tolerated in the terminal regions and, if present, are generally in a terminal region or regions, e.g., within 6, 5, 4, 3, or 2 nucleotides of the 5' and/or 3' terminus. Most preferably, the mismatches are located within 6, 5, 4, 3, or 2 nucleotides of the 5' terminus of the antisense strand and/or the 3' terminus of the sense strand.
- the term “complementary,” when used to describe a first nucleotide sequence in relation to a second nucleotide sequence, refers to the ability of an oligonucleotide or polynucleotide comprising the first nucleotide sequence to hybridize and form a duplex structure under certain conditions with an oligonucleotide or polynucleotide comprising the second nucleotide sequence, as will be understood by the skilled person.
- Such conditions can, for example, be stringent conditions, where stringent conditions may include: 400 mM NaCl, 40 mM PIPES pH 6.4, 1 mM EDTA, 50°C or 70°C for 12-16 hours followed by washing.
- stringent conditions may include: 400 mM NaCl, 40 mM PIPES pH 6.4, 1 mM EDTA, 50°C or 70°C for 12-16 hours followed by washing.
- Other conditions such as physiologically relevant conditions as may be encountered inside an organism, can apply. The skilled person will be able to determine the set of conditions most appropriate for a test of complementarity of two sequences in accordance with the ultimate application of the hybridized nucleotides.
- sequences can be referred to as “fully complementary” with respect to each other herein.
- first sequence is referred to as “substantially complementary” with respect to a second sequence herein
- the two sequences can be fully complementary, or they may form one or more, but generally not more than 4, 3, or 2 mismatched base pairs upon hybridization, while retaining the ability to hybridize under the conditions most relevant to their ultimate application.
- a dsRNA comprising one oligonucleotide 21 nucleotides in length and another oligonucleotide 23 nucleotides in length, wherein the longer
- oligonucleotide comprises a sequence of 21 nucleotides that is fully complementary to the shorter oligonucleotide, may yet be referred to as "fully complementary” for the purposes of the invention.
- Complementary sequences may also include, or be formed entirely from, non- Watson-Crick base pairs and/or base pairs formed from natural and modified nucleotides, in as far as the above requirements with respect to their ability to hybridize are fulfilled.
- Double-Stranded RNA or dsRNA refers to a complex of ribonucleic acid molecules, having a duplex structure comprising two anti-parallel and substantially complementary, as defined above, nucleic acid strands.
- the two strands forming the duplex structure may be different portions of one larger RNA molecule, or they may be separate RNA molecules.
- the connecting RNA chain is referred to as a "hairpin loop” and the entire structure is referred to as a "short hairpin RNA" or "shRNA.”
- the connecting structure is referred to as the "linker.”
- the RNA strands may have the same or a different number of nucleotides.
- a dsRNA may comprise one or more nucleotide overhangs.
- nucleotide overhang refers to the unpaired nucleotide or nucleotides that protrude from the duplex structure of a dsRNA when a 3 '-end of one strand of the dsRNA extends beyond the 5 '-end of the other strand, or vice versa.
- Bount or “blunt end” means that there are no unpaired nucleotides at that end of the dsRNA, i.e. no nucleotide overhang.
- a “blunt ended" dsRNA is a dsRNA that has no nucleotide overhang at either end of the molecule.
- compositions are pharmaceutically acceptable.
- pharmaceutically acceptable means approved by a regulatory agency of the Federal or a state government or listed in the U.S. Pharmacopeia or other generally recognized pharmacopeia for use in animals, and more particularly in humans.
- “pharmaceutically acceptable carrier” refers to a diluent, adjuvant, excipient, or vehicle with which a compound is administered.
- Such carriers can be sterile liquids, such as water and oils, including those of petroleum, animal, vegetable, or synthetic origin, such as peanut oil, soybean oil, mineral oil, sesame oil and the like, polyethylene glycols, glycerine, propylene glycol, or other synthetic solvents. Water is a preferred carrier when a compound is administered intravenously.
- Saline solutions and aqueous dextrose and glycerol solutions can also be employed as liquid carriers, particularly for injectable solutions.
- Suitable excipients include starch, glucose, lactose, sucrose, gelatin, malt, rice, flour, chalk, silica gel, sodium stearate, glycerol monostearate, talc, sodium chloride, dried skim milk, glycerol, propylene, glycol, water, ethanol, and the like.
- a compound, if desired, can also combine minor amount of wetting or emulsifying agents, or pH buffering agents such as acetates, citrates, or phosphates.
- Antibacterial agents such as a benzyl alcohol or methyl parabens; antioxidants such as ascorbic acid or sodium bisulfite; chelating agents such as ethylenediaminetetraacetic acid; and agents for the adjustment of tonicity such as sodium chloride or dextrose may also be a carrier.
- “pharmaceutically acceptable salt” includes those salts of a pharmaceutically acceptable compound formed with free amino groups such as those derived from hydrochloric, phosphoric, acetic, oxalic, and tartaric acids, and those formed with free carboxyl groups such as those derived from sodium, potassium, ammonium, calcium, ferric hydroxides, isopropylamine, triethylamine, 2-ethylamino ethanol, histidine, and procaine. If the compound is basic, salts may be prepared from pharmaceutically acceptable non-toxic acids including inorganic and organic acids. Such acids include acetic, benzene-sulfonic
- besylate benzoic, camphorsulfonic, citric, ethenesulfonic, fumaric, gluconic, glutamic, hydrobromic, hydrochloric, isethionic, lactic, maleic, malic, mandelic, methanesulfonic, mucic, nitric, pamoic, pantothenic, phosphoric, succinic, sulfuric, tartaric, p-toluenesulfonic, and the like. Particularly preferred are besylate, hydrobromic, hydrochloric, phosphoric, and sulfuric acids. If the compound is acidic, salts may be prepared from pharmaceutically acceptable organic and inorganic bases.
- Suitable organic bases include, but are not limited to, lysine, ⁇ , ⁇ '-dibenzylethylenediamine, chloroprocaine, choline, diethanolamine, ethylene diamine, meglumine (N-methyl-glucamine) and procaine.
- Suitable inorganic bases include, but are not limited to, alkaline and earth-alkaline metals such as aluminum, calcium, lithium, magnesium, potassium, sodium, and zinc. Methods for synthesizing such salts are known to those of skill in the art.
- Sense Strand refers to the strand of a dsR A that includes a region that is substantially complementary to a region of the antisense strand.
- siRNA and inhibit the expression of insofar as they refer to a ⁇ -arrestin gene, e.g. a P-arrestin2 gene, refer to at least the partial suppression of the expression of a ⁇ -arrestin gene as manifested by a reduction of the amount of mRNA transcribed from a ⁇ -arrestin gene which may be isolated from a first cell or group of cells in which a ⁇ -arrestin gene is transcribed and which has or have been treated such that the expression of a ⁇ -arrestin gene is inhibited, as compared to a second cell or group of cells substantially identical to the first cell or group of cells but which has or have not been so treated (control cells).
- the degree of inhibition is usually expressed in terms of
- the degree of inhibition may be given in terms of a reduction of a parameter that is functionally linked to ⁇ -arrestin gene transcription, e.g. the amount of protein encoded by a ⁇ -arrestin gene which is secreted by a cell, or found in solution after lysis of such cells, or the number of cells displaying a certain phenotype, e.g. apoptosis or cell surface CFTR.
- ⁇ -arrestin silencing may be determined in any cell expressing the target, either constitutively or by genomic engineering, and by any appropriate assay.
- the methods provided in the Examples below shall serve as such reference.
- Strand Comprising a Sequence refers to an oligonucleotide comprising a chain of nucleotides that is described by the sequence referred to using the standard nucleotide nomenclature.
- a polynucleotide that is "substantially complementary" to at least part of a messenger RNA (mRNA) refers to a polynucleotide that is substantially complementary to a contiguous portion of the mRNA of interest.
- mRNA messenger RNA
- a polynucleotide is complementary to at least part of a ⁇ -arrestin mRNA if the sequence is substantially complementary to a non-interrupted portion of an mRNA encoding ⁇ -arrestin.
- Target Sequence refers to a contiguous portion of the nucleotide sequence of an mRNA molecule formed during the transcription of a ⁇ -arrestin gene, including mRNA that is a product of RNA processing of a primary transcription product.
- the target sequence of any given RNAi agent of the invention means an mRNA-sequence of X nucleotides that is targeted by the RNAi agent by virtue of the complementarity of the antisense strand of the RNAi agent to such sequence and to which the antisense strand may hybridize when brought into contact with the mRNA, wherein X is the number of nucleotides in the antisense strand plus the number of nucleotides in a single stranded overhang of the sense strand, if any.
- therapeutically effective amount refers to those amounts that, when administered to a particular subject in view of the nature and severity of that subject's disease or condition, will have a desired therapeutic effect, e.g. an amount that will cure, prevent, inhibit, or at least partially arrest or partially prevent a target disease or condition.
- Transformed Cell As used herein, the term "transformed cell” is a cell into which a vector has been introduced from which a dsRNA molecule may be expressed.
- Treatment refers to relief from or alleviation of pathological processes mediated by ⁇ -arrestin expression.
- the terms “treat”, “treatment”, and the like mean to relieve or alleviate at least one symptom associated with such condition, or to slow or reverse the progression of such condition.
- the present invention provides methods of treating a P-arrestin2 mediated and/or GPR120 mediated response in a subject.
- the ⁇ -arrestin mediated and/or GPCR mediated response is inflammation.
- the method can comprise administering to a subject in whom inflammation is to be treated a therapeutically effective amount of a compound predicted to bind a ⁇ - ⁇ 8 ⁇ 2 molecule and/or a G protein-coupled receptor, where the compound selectively activates a ⁇ -arrestin2-dependent signaling pathway of the GPCR.
- a GenBank Gene ID No. 109689 and ⁇ -arrestinl has a GenBank Gene ID No. 216869.
- the invention further provides that GPR120, a GPCR contemplated in the invention and having GenBank Accession No. NP 859529, is expressed in proinflammatory macrophages and is induced in macrophages and Kupffer cells in obesity. DHA and EPA, major omega-3 FAs (co-3 FAs) in fish oil, and GPR120 function as an co-3 FA
- co-3 FAs exert robust and broad anti-inflammatory effects through GPR120 in macrophages.
- an co-3 FA supplemented diet exerts both antiinflammatory and anti-diabetic/insulin sensitizing effects in obese mice, but is without effect in GPR120 KO mice.
- the invention provides methods for treating conditions associated with inflammation and insulin sensitization including, but not limited to, diabetes and obesity.
- Diseases and disorders that are characterized by P-arrestin2 and/or GPR120 biological activity may be treated with therapeutics that agonize GPR120 activity.
- Agonists may be administered in a therapeutic or prophylactic manner.
- Therapeutics that may be used include omega-3 fatty acids, analogs thereof and derivatives thereof, and other modulators that alter the interaction between GPR120 and its binding partners.
- methods for screening for other agonists are provided herein, including in the Examples section.
- the agonist can selectively activate a P-arrestin2 -dependent signaling pathway and not activate a ⁇ - arrestinl -dependent signaling pathway.
- the agonist can selectively activate a -arrestin2-dependent signaling pathway and not activate a G protein-dependent signaling pathway.
- the agonist can selectively activate a G protein-dependent signaling pathway and not activate a P-arrestin2-dependent signaling pathway.
- Diseases and disorders that are characterized by decreased GPR120 levels or biological activity may be treated with therapeutics that increase (i.e., are agonists to) activity.
- Therapeutics that upregulate activity may be administered therapeutically or prophylactically.
- the invention provides a method for preventing, in a subject, a disease or condition associated with an aberrant GPR120 activity by administering an agent that modulates GPR120 activity.
- aberrant activity includes inflammatory conditions, including diabetes, inflammatory condition is associated with obesity, and obesity.
- Subjects at risk for a disease that is caused or contributed to by aberrant GPR120 expression or activity can be identified by, for example, an assay provided in the Examples section herein.
- Administration of a prophylactic agent can occur prior to the manifestation of symptoms characteristic of the GPR120 aberrancy, such that a disease or disorder is prevented or, alternatively, delayed in its progression.
- a GPR120 agonist can be used to treat the subject.
- the appropriate agent can be determined based on screening assays.
- the subject can be an animal, and in particular a human.
- Another aspect of the invention pertains to methods of modulating ⁇ - arrestin2 and/or GPR120 biological activity for therapeutic purposes.
- the modulatory methods of the invention involve contacting a cell with an agent that modulates one or more of the activities of P-arrestin2 and/or GPR120 biological activity associated with the cell.
- An agent that modulates p-arrestin2 and/or GPCR biological activity can be a naturally occurring cognate ligand of GPR120 such as an omega-3 fatty acid, a peptide, a GPR120 and/or P-arrestin2 peptidomimetic, dsRNA, small molecule, and the like.
- Modulatory methods can be performed in vitro (e.g., by culturing the cell with the agent) or in vivo (e.g., by administering the agent to a subject).
- the invention provides methods of treating an individual afflicted with a disease or disorder characterized by expression or activity of a ⁇ -arrestin and/or GPCR.
- the method involves
- an agent e.g., an agent identified by a screening assay
- agents that modulates e.g., up-regulates
- GPR120 biological activity e.g., administering an agent (e.g., an agent identified by a screening assay), or combination of agents that modulates (e.g., up-regulates) -arrestin2 and/or GPR120 biological activity.
- the inflammation being treated by the method is a lipopolysaccharide ("LPS") induced inflammatory response.
- the inflammation being treated by the method is a TNF-a-induced inflammatory response.
- the P-arrestin2 mediated response is cytokine secretion.
- cytokines include, but are not limited to TNF-a and interleukin-6.
- the method can comprise administering to a subject in whom cytokine secretion is sought to be inhibited a therapeutically effective amount of a compound predicted to bind GPR120, where the compound selectively activates a P-arrestin2- dependent signaling pathway of GPR120.
- the cytokine secretion being inhibited is a lipopolysaccharide-induced cytokine secretion.
- a method for screening compounds predicted to bind GPR120 in such a manner as to produce -arrestin2 biased activity includes administering to a subject an amount of a compound predicted to bind a GPCR sufficient to generate a physiological response, evaluating that response, administering to the subject an effect amount of an siRNA capable of binding a P-arrestin2 RNA, allowing the binding to occur, and then evaluating the response once again. If the physiological response is reversed at the time of the second evaluation, then the compound being evaluated is a P-arrestin2 biased compound.
- the method for screening described above is provided, wherein the physiological response evaluated is a
- the method for screening described above is provided, wherein the physiological response is a lipopolysaccharide-induced cytokine secretion.
- agents that modulate functional P-arrestin2 and/or functional GPR120 are contemplated by certain aspects of the present invention. Such agents are useful in treating conditions associated with P-arrestin2 function and/or GPR120 function. Such agents are also useful for screening for P-arrestin2 biased ligands or other compounds for GPCRs. These agents are useful in both in vivo and in vitro screening methods.
- Agents that decrease levels of functional P-arrestin2 can target functional P-arrestin2 or nucleotides encoding therefor, resulting in a descrease in P-arrestin2 activity.
- the various classes of agents for use herein as agents that decrease levels of functional ⁇ - arrestin2 generally include, but are not limited to, RNA interference molecules, antibodies, small inorganic molecules, antisense oligonucleotides, and aptamers.
- Agents that agonize functional GPCR, and specifically GPR120 can include the ⁇ -3 FAs identified herein.
- Naturally occurring ⁇ -3 FAs are well known in the art.
- modified co-3 FAs can be made by methods known in the art. See, e.g., Bailey 's Industrial Oil and Fat Products, Sixth Edition, Six Volume Set. Edited by
- co-3 FAs having a biological activity useful in performing the methods of the invention can be indentified by employing the methods described herein.
- RNA interference can be used to decrease the levels of functional P-arrestin2.
- RNAi methods can utilize double-stranded RNAs, for example, small interfering RNAs (siRNA), short hairpin RNAs (shRNA), and micro RNAs (miRNA).
- siRNA small interfering RNAs
- shRNA short hairpin RNAs
- miRNA micro RNAs
- dsRNA molecule design programs using a variety of algorithms are known to the art (see e.g., Cenix algorithm, Ambion; BLOCK-iTTM RNAi Designer, Invitrogen; dsRNA Whitehead Institute Design Tools, Bioinofrmatics & Research
- Traits influential in defining optimal dsRNA sequences include G/C content at the termini of the dsRNAs, T m of specific internal domains of the dsRNA, dsRNA length, position of the target sequence within the CDS (coding region), and nucleotide content of the 3' overhangs.
- dsRNA molecules specific for functional P-arrestin2, and/or other related molecules with similar functions can effect the RNAi-mediated degradation of the target (P-arrestin2) mRNA.
- a therapeutically effective amount of dsRNA specific for p-arrestin2 can be adminstered to patient in need thereof to treat a condition mediated by -arrestin2.
- an effective amount of dsRNA molecule comprises an intercellular concentration at or near the site of misfolding from about 1 nanomolar (nM) to about 100 nM, preferably from about 2 nM to about 50 nM, more preferably from about 2.5 nM to about 10 nM. It is contemplated that greater or lesser amounts of dsRNA can be administered.
- the dsRNA can be administered to the subject by any means suitable for delivering the RNAi molecules to the cells of interest.
- dsRNA molecules can be administered by gene gun, electroporation, or by other suitable parenteral or enteral administration routes, such as intravitreous injection.
- RNAi molecules can also be administered locally (lung tissue) or systemically (circulatory system) via pulmonary delivery.
- RNAi molecules can be used in conjunction with a variety of delivery and targeting systems, as described in further detail below.
- dsRNA can be encapsulated into targeted polymeric delivery systems designed to promote payload internalization.
- the dsRNA can generally be targeted to any stretch of less than 30 contiguous nucleotides, generally about 19-25 contiguous nucleotides, in the functional ⁇ - arrestin2 mRNA target sequences.
- Exemplary siRNAs targeting ⁇ -arrestin 1 and 2 are provided in Table 1 and Table 2, below. Searches of the human genome database (BLAST) can be carried out to ensure that selected dsRNA sequence will not target other gene transcripts. Techniques for selecting target sequences for dsRNA are known in the art (see e.g., Reynolds et al. (2004) Nature Biotechnology 22(3), 326 - 330).
- the sense strand of the present dsRNA can comprise a nucleotide sequence identical to any contiguous stretch of about 19 to about 25 nucleotides in the target mRNA of functional P-arrestin2.
- a target sequence on the target mRNA can be selected from a given cDNA sequence corresponding to the target mRNA, preferably beginning 50 to 100 nt downstream (i.e., in the 3' direction) from the start codon.
- the target sequence can, however, be located in the 5' or 3' untranslated regions, or in the region nearby the start codon.
- the dsRNA of the invention can comprise an RNA strand (the antisense strand) having a region which is less than 30 nucleotides in length, generally 19-25 nucleotides in length, and is substantially complementary to at least part of an mRNA transcript of a P-arrestin2 gene.
- RNA strand the antisense strand
- very low dosages of these dsRNA can specifically and efficiently mediate RNAi, resulting in significant inhibition of expression of a -arrestin2 gene.
- the methods and compositions of the invention comprising these dsRNAs are useful for screening compounds for selective activating a P-arrestin2 pathway when binding a GPCR.
- compositions containing dsRNA to inhibit the expression of a P-arrestin2 gene.
- the pharmaceutical compositions of the invention comprise a dsRNA having an antisense strand comprising a region of complementarity which is less than 30 nucleotides in length, generally 19-25 nucleotides in length, and is substantially complementary to at least part of an RNA transcript of a P-arrestin2 gene, together with a pharmaceutically acceptable carrier.
- These pharmaceutical compositions are useful in administering the dsRNA to an animal for use in animal models for screening compounds that can selectively activate a ⁇ - arrestin2 pathway when binding a GPCR.
- compositions comprising the dsRNA of the invention together with a pharmaceutically acceptable carrier, as well as methods of using the compositions to inhibit expression of a ⁇ - arrestin2 gene.
- dsRNA molecules for inhibiting the expression of a P-arrestin2 gene in a cell or mammal, wherein the dsRNA comprises an antisense strand comprising a region of complementarity that is complementary to at least a part of an mRNA formed in the expression of a P-arrestin2 gene, and wherein the region of complementarity is less than 30 nucleotides in length, generally 19-25 nucleotides in length.
- the dsRNA has at least 5, at least 10, at least 15, at least 18, or at least 20 contiguous nucleotides per strand in common with at least one strand, but preferably both strands, of one of the dsRNAs shown in Tables 1 and 2.
- Alternative dsRNAs that target elsewhere in the target sequence of one of the dsRNAs provided in Tables 1 and 2 can readily be determined using the target sequence and the flanking ⁇ - arrestin2 sequence.
- the dsRNA comprises two RNA strands that are complementary to hybridize to form a duplex structure.
- One strand of the dsRNA (the antisense strand) comprises a region of complementarity that is substantially complementary, and generally fully complementary, to a target sequence, derived from the sequence of an mRNA formed during the expression of a P-arrestin2 gene
- the other strand (the sense strand) comprises a region which is complementary to the antisense strand, such that the two strands hybridize and form a duplex structure when combined under suitable conditions.
- the duplex structure is between 15 and 30, more generally between 18 and 25, yet more generally between 19 and 24, and most generally between 19 and 21 base pairs in length.
- the region of complementarity to the target sequence is between 15 and 30, more generally between 18 and 25, yet more generally between 19 and 24, and most generally between 19 and 21 nucleotides in length.
- the dsRNA of the invention may further comprise one or more single-stranded nucleotide overhang(s). For example,
- deoxyribonucleotide sequence "tt” or ribonucleotide sequence "UU” can be connected to the 3 '-end of both sense and antisense strands to form overhangs.
- the dsRNA can be synthesized by standard methods known in the art as further discussed below, e.g., by use of an automated DNA synthesizer, such as are commercially available from, for example, Biosearch, Applied Biosystems, Inc.
- dsRNAs comprising a duplex structure of between 20 and 23, but specifically 21 , base pairs have been hailed as particularly effective in inducing RNA interference (Elbashir et al., EMBO 2001 , 20:6877- 6888). However, others have found that shorter or longer dsRNAs can be effective as well.
- the dsRNA of the invention can contain one or more mismatches to the target sequence. If the antisense strand of the dsRNA contains mismatches to a target sequence, it is preferable that the area of mismatch not be located in the center of the region of complementarity. If the antisense strand of the dsRNA contains mismatches to the target sequence, it is preferable that the mismatch be restricted to 5 nucleotides from either end, for example 5, 4, 3, 2, or 1 nucleotide from either the 5' or 3' end of the region of complementarity, and preferably from the 5'-end.
- the dsRNA generally does not contain any mismatch within the central 13 nucleotides.
- the methods described within the invention can be used to determine whether a dsRNA containing a mismatch to a target sequence is effective in inhibiting the expression of a P-arrestin2 gene. Consideration of the efficacy of dsRNAs with mismatches in inhibiting expression of a P-arrestin2 gene is important, especially if the particular region of complementarity in a p-arrestin2 gene is known to have polymorphic sequence variation within the population.
- At least one end of the dsRNA has a single-stranded nucleotide overhang of 1 to 4, generally 1 or 2 nucleotides.
- dsRNAs having at least one nucleotide overhang have unexpectedly superior inhibitory properties than their blunt-ended counterparts.
- the present inventors have discovered that the presence of only one nucleotide overhang strengthens the interference activity of the dsRNA, without affecting its overall stability.
- dsRNA having only one overhang has proven particularly stable and effective in vivo, as well as in a variety of cells, cell culture mediums, blood, and serum.
- the single-stranded overhang is located at the 3'-terminal end of the antisense strand or, alternatively, at the 3'-terminal end of the sense strand.
- the dsRNA may also have a blunt end, generally located at the 5 '-end of the antisense strand.
- Such dsRNAs have improved stability and inhibitory activity, thus allowing administration at low dosages, i.e., less than 5 mg/kg body weight of the recipient per day.
- the antisense strand of the dsRNA has a nucleotide overhang at the 3'-end, and the 5'-end is blunt.
- one or more of the nucleotides in the overhang is replaced with a nucleoside thiophosphate.
- the dsRNA is chemically modified to enhance stability.
- the nucleic acids of the invention may be synthesized and/or modified by methods well established in the art, such as those described in "Current protocols in nucleic acid chemistry", Beaucage, S.L. et al. (Edrs.), John Wiley & Sons, Inc., New York, NY, USA, which is hereby incorporated herein by reference.
- Specific examples of preferred dsRNA compounds useful in this invention include dsRNAs containing modified backbones or no natural internucleoside linkages.
- dsRNAs having modified backbones include those that retain a phosphorus atom in the backbone and those that do not have a phosphorus atom in the backbone.
- modified dsRNAs that do not have a phosphorus atom in their internucleoside backbone can also be considered to be oligonucleosides.
- Preferred modified dsRNA backbones include, for example,
- phosphorothioates chiral phosphorothioates, phosphorodithioates, phosphotriesters, aminoalkylphosphotriesters, methyl and other alkyl phosphonates including 3'-alkylene phosphonates and chiral phosphonates, phosphinates, phosphoramidates including 3'-amino phosphoramidate and aminoalkylphosphoramidates, thionophosphoramidates,
- thionoalkylphosphonates having normal 3'-5' linkages, 2 -5' linked analogs of these, and those having inverted polarity wherein the adjacent pairs of nucleoside units are linked 3'-5' to 5 -3' or 2'-5' to 5'-2'.
- Representative U.S. patents that teach the preparation of the above phosphorus-containing linkages include, but are not limited to, U.S. Pat. Nos. 3,687,808; 4,469,863; 4,476,301 ; 5,023,243; 5,177,195; 5,188,897; 5,264,423; 5,276,019; 5,278,302; 5,286,717; 5,321,131 ; 5,399,676; 5,405,939; 5,453,496; 5,455,233; 5,466,677; 5,476,925; 5,519,126; 5,536,821 ; 5,541 ,316; 5,550,1 1 1 ; 5,563,253; 5,571 ,799; 5,587,361 ; and
- Preferred modified dsR A backbones that do not include a phosphorus atom therein have backbones that are formed by short chain alkyl or cycloalkyl
- internucleoside linkages mixed heteroatoms and alkyl or cycloalkyl internucleoside linkages, or one or more short chain heteroatomic or heterocyclic internucleoside linkages.
- These include those having morpholino linkages (formed in part from the sugar portion of a nucleoside); siloxane backbones; sulfide, sulfoxide and sulfone backbones; formacetyl and thioformacetyl backbones; methylene formacetyl and thioformacetyl backbones; alkene containing backbones; sulfamate backbones; methyleneimino and methylenehydrazino backbones; sulfonate and sulfonamide backbones; amide backbones; and others having mixed N, O, S and CH 2 component parts.
- both the sugar and the internucleoside linkage, i.e., the backbone, of the nucleotide units are replaced with novel groups.
- the base units are maintained for hybridization with an appropriate nucleic acid target compound.
- a dsRNA mimetic that has been shown to have excellent hybridization properties, is referred to as a peptide nucleic acid (PNA).
- PNA peptide nucleic acid
- the sugar backbone of a dsRNA is replaced with an amide containing backbone, in particular an aminoethylglycine backbone.
- the nucleobases are retained and are bound directly or indirectly to aza nitrogen atoms of the amide portion of the backbone.
- phosphorothioate backbones and oligonucleosides with heteroatom backbones and in particular -CH 2- NH.CH 2- , -CH 2- N(CH 3 )-0-CH 2- [known as a methylene (methylimino) or MMI backbone], -CH 2- 0-N(CH 3 ).CH 2- , -CH 2 .N(CH 3 )-N(CH 3 )-CH 2- and -N(CH 3 ).CH 2 .CH 2- [wherein the native phosphodiester backbone is represented as -O.P.O.CH 2 .] of the above- referenced U.S. Pat. No.
- Modified dsRNAs may also contain one or more substituted sugar moieties.
- Preferred dsRNAs comprise one of the following at the 2' position: OH; F; 0-, S-, or N-alkyl; 0-, S-, or N-alkenyl; 0-, S- or N-alkynyl; or O-alkyl-O-alkyl, wherein the alkyl, alkenyl and alkynyl may be substituted or unsubstituted C ⁇ to Ci 0 alkyl or C 2 to Ci 0 alkenyl and alkynyl.
- n and m are from 1 to about 10.
- dsRNAs comprise one of the following at the 2' position: C
- a preferred modification includes 2'-methoxyethoxy (2'-0.CH 2 CH 2 0CH 3 , also known as 2'-0-(2-methoxyethyl) or 2'-MOE) (Martin et al., Helv. Chim. Acta, 1995, 78, 486-504) i.e., an alkoxy-alkoxy group.
- a further preferred modification includes 2'-dimethylaminooxyethoxy, i.e., a 0(CH 2 ) 2 ON(CH 3 ) 2 group, also known as 2'-DMAOE, as described in examples hereinbelow, and 2'- dimethylaminoethoxyethoxy (also known in the art as 2'-0-dimethylaminoethoxyethyl or 2'- DMAEOE), i.e., 2'-O.CH 2- O.CH 2- N(CH 2 ) 2 , also described in examples hereinbelow.
- 2'-dimethylaminooxyethoxy i.e., a 0(CH 2 ) 2 ON(CH 3 ) 2 group
- 2'-DMAOE 2'- dimethylaminoethoxyethoxy
- 2'-O.CH 2- O.CH 2- N(CH 2 ) 2 also described in examples hereinbelow.
- dsRNAs may also include nucleobase (often referred to in the art simply as “base”) modifications or substitutions.
- nucleobases include the purine bases adenine (A) and guanine (G), and the pyrimidine bases thymine (T), cytosine (C) and uracil (U).
- Modified nucleobases include other synthetic and natural nucleobases such as 5-methylcytosine (5-me-C), 5-hydroxymethyl cytosine, xanthine, hypoxanthine, 2-aminoadenine, 6-methyl and other alkyl derivatives of adenine and guanine, 2-propyl and other alkyl derivatives of adenine and guanine, 2-thiouracil, 2- thiothymine and 2-thiocytosine, 5-halouracil and cytosine, 5-propynyl uracil and cytosine,
- nucleobases such as 5-methylcytosine (5-me-C), 5-hydroxymethyl cytosine, xanthine, hypoxanthine, 2-aminoadenine, 6-methyl and other alkyl derivatives of adenine and guanine, 2-propyl and other alkyl derivatives of adenine and guanine, 2-thiouracil, 2- thiothymine and 2-thi
- nucleobases include those disclosed in U.S. Pat. No. 3,687,808, those disclosed in The Concise Encyclopedia Of Polymer Science And Engineering, pages 858-859, Kroschwitz, J. L, ed. John Wiley & Sons, 1990, these disclosed by Englisch et al., Angewandte Chemie, International Edition, 1991, 30, 613, and those disclosed by Sanghvi, Y S., Chapter 15, DsRNA Research and Applications, pages 289-302, Crooke, S. T. and Lebleu, B., Ed., CRC Press, 1993.
- nucleobases are particularly useful for increasing the binding affinity of the oligomeric compounds of the invention.
- These include 5-substituted pyrimidines, 6- azapyrimidines and N-2, N-6 and 0-6 substituted purines, including 2-aminopropyladenine, 5-propynyluracil and 5-propynylcytosine. 5-methylcytosine substitutions have been shown to increase nucleic acid duplex stability by 0.6-1.2. degree. C. (Sanghvi, Y. S., Crooke, S. T. and Lebleu, B., Eds., DsRNA Research and Applications, CRC Press, Boca Raton, 1993, pp.
- dsRNAs of the invention involves chemically linking to the dsRNA one or more moieties or conjugates which enhance the activity, cellular distribution or cellular uptake of the dsRNA.
- moieties include but are not limited to lipid moieties such as a cholesterol moiety (Letsinger et al., Proc. Natl. Acid. Sci. USA, 199, 86, 6553-6556), cholic acid (Manoharan et al., Biorg. Med. Chem. Let., 1994 4 1053-1060), a thioether, e.g., beryl-S-tritylthiol (Manoharan et al., Ann. N.Y. Acad.
- Acids Res., 1990, 18, 3777-3783 a polyamine or a polyethylene glycol chain (Manoharan et al., Nucleosides & Nucleotides, 1995, 14, 969- 973), or adamantane acetic acid (Manoharan et al., Tetrahedron Lett., 1995, 36, 3651 -3654), a palmityl moiety (Mishra et al., Biochim. Biophys. Acta, 1995, 1264, 229-237), or an octadecylamine or hexylamino-carbonyloxycholesterol moiety (Crooke et al., J. Pharmacol. Exp. Ther., 1996, 277, 923-937).
- the present invention also includes dsRNA compounds which are chimeric compounds.
- dsRNA compounds are dsRNA compounds, particularly dsRNAs, which contain two or more chemically distinct regions, each made up of at least one monomer unit, i.e., a nucleotide in the case of a dsRNA compound.
- dsRNAs typically contain at least one region wherein the dsRNA is modified so as to confer upon the dsRNA increased resistance to nuclease degradation, increased cellular uptake, and/or increased binding affinity for the target nucleic acid.
- An additional region of the dsRNA may serve as a substrate for enzymes capable of cleaving RNA:DNA or RNA:RNA hybrids.
- RNase H is a cellular endonuclease which cleaves the RNA strand of an RNA:DNA duplex. Activation of RNase H, therefore, results in cleavage of the RNA target, thereby greatly enhancing the efficiency of dsRNA inhibition of gene expression. Consequently, comparable results can often be obtained with shorter dsRNAs when chimeric dsRNAs are used, compared to phosphorothioate deoxydsRNAs hybridizing to the same target region. Cleavage of the RNA target can be routinely detected by gel electrophoresis and, if necessary, associated nucleic acid hybridization techniques known in the art.
- the dsRNA may be modified by a non-ligand group.
- a number of non-ligand molecules have been conjugated to dsRNAs in order to enhance the activity, cellular distribution or cellular uptake of the dsRNA, and procedures for performing such conjugations are available in the scientific literature.
- Such non-ligand moieties have included lipid moieties, such as cholesterol (Letsinger et al., Proc. Natl. Acad. Sci. USA, 1989, 86:6553), cholic acid (Manoharan et al., Bioorg. Med. Chem.
- a thioether e.g., hexyl-S-tritylthiol
- a thiocholesterol Olet al., Nucl.
- Acids Res., 1990, 18:3777 a polyamine or a polyethylene glycol chain (Manoharan et al., Nucleosides & Nucleotides, 1995, 14:969), or adamantane acetic acid (Manoharan et al., Tetrahedron Lett., 1995, 36:3651), a palmityl moiety (Mishra et al., Biochim. Biophys. Acta, 1995, 1264:229), or an octadecylamine or hexylamino-carbonyl-oxycholesterol moiety (Crooke et al., J. Pharmacol. Exp. Ther., 1996, 277:923).
- Typical conjugation protocols involve the synthesis of dsRNAs bearing an aminolinker at one or more positions of the sequence. The amino group is then reacted with the molecule being conjugated using appropriate coupling or activating reagents. The conjugation reaction may be performed either with the dsRNA still bound to the solid support or following cleavage of the dsRNA in solution phase. Purification of the dsRNA conjugate by HPLC typically affords the pure conjugate.
- a dsRNA for inhibiting functional ⁇ -arrestin protein expression in a cell, the dsRNA having a sense strand and an antisense strand.
- the antisense strand includes a region of
- the sense strand is substantially complementary to said antisense strand.
- the dsRNA upon contact with a cell expressing functional ⁇ -arrestin protein, inhibits functional ⁇ -arrestin protein expression.
- a ⁇ -arrestin target sequence includes a sequence selected from the group consisting of SEQ ID NO. 1 and SEQ ID NO. 2 or sequences complementary thereto.
- Another aspect of the present invention provides a method for inhibiting expression of a ⁇ -arrestin gene in a cell.
- the method includes introducing into the cell a dsRNA having two sequences that are compementary to each other.
- a sense strand includes a first sequence and an antisense strand includes a second sequence having a region of complimentarity that is substantially complementary to at least a part of an mRNA encoding ⁇ -arrestin.
- a method for inhibiting ⁇ -arrestin expression is provided, where the dsRNA used includes a sequence selected from the group consisting of SEQ ID NO. 1 and SEQ ID NO. 2, or sequences complementary thereto.
- the dsRNA of the invention can also be expressed from recombinant viral vectors intracellularly in vivo.
- the recombinant viral vectors of the invention comprise sequences encoding the dsRNA of the invention and any suitable promoter for expressing the dsRNA sequences. Suitable promoters include, for example, the U6 or HI RNA pol III promoter sequences and the cytomegalovirus promoter. Selection of other suitable promoters is within the skill in the art.
- the recombinant viral vectors of the invention can also comprise inducible or regulatable promoters for expression of the dsRNA in a particular tissue or in a particular intracellular environment. The use of recombinant viral vectors to deliver dsRNA of the invention to cells in vivo is discussed in more detail below.
- dsRNA of the invention can be expressed from a recombinant viral vector either as two separate, complementary RNA molecules, or as a single RNA molecule with two complementary regions.
- Any viral vector capable of accepting the coding sequences for the dsRNA molecule(s) to be expressed can be used, for example vectors derived from adenovirus (AV); adeno-associated virus (AAV); retroviruses (e.g, lentiviruses (LV), Rhabdoviruses, murine leukemia virus); herpes virus, and the like.
- AV adenovirus
- AAV adeno-associated virus
- retroviruses e.g, lentiviruses (LV), Rhabdoviruses, murine leukemia virus
- herpes virus and the like.
- the tropism of viral vectors can be modified by pseudotyping the vectors with envelope proteins or other surface antigens from other viruses, or by substituting different viral capsid proteins, as appropriate.
- lentiviral vectors of the invention can be pseudotyped with surface proteins from vesicular stomatitis virus (VSV), rabies, Ebola, Mokola, and the like.
- AAV vectors of the invention can be made to target different cells by engineering the vectors to express different capsid protein serotypes.
- an AAV vector expressing a serotype 2 capsid on a serotype 2 genome is called AAV 2/2.
- This serotype 2 capsid gene in the AAV 2/2 vector can be replaced by a serotype 5 capsid gene to produce an AAV 2/5 vector.
- AAV vectors which express different capsid protein serotypes are within the skill in the art; see, e.g., Rabinowitz J E et al. (2002), J Virol 76:791 -801 , the entire disclosure of which is herein incorporated by reference.
- Preferred viral vectors are those derived from AV and AAV.
- the dsRNA of the invention is expressed as two separate, complementary single-stranded RNA molecules from a recombinant AAV vector comprising, for example, either the U6 or HI RNA promoters, or the cytomegalovirus (CMV) promoter.
- CMV cytomegalovirus
- a suitable AV vector for expressing the dsRNA of the invention a method for constructing the recombinant AV vector, and a method for delivering the vector into target cells, are described in Xia H et al. (2002), Nat. Biotech. 20: 1006-1010.
- Suitable AAV vectors for expressing the dsRNA of the invention, methods for constructing the recombinant AV vector, and methods for delivering the vectors into target cells are described in Samulski R et al. (1987), J. Virol. 61 : 3096-3101 ; Fisher K J et al. (1996), J. Virol, 70: 520-532; Samulski R et al. (1989), J. Virol. 63 : 3822-3826; U.S. Pat. No. 5,252,479; U.S. Pat. No. 5, 139,941 ; International Patent Application No. WO
- Antibodies can be used to decrease levels of functional -arrestin2.
- Antibodies within the scope of the invention include, for example, polyclonal antibodies, monoclonal antibodies, antibody fragments, and antibody-based fusion molecules.
- Antibodies can be altered or selected so as to achieve efficient antibody internalization. As such, the antibodies can more effectively interact with target intracellular molecules, such as functional p ⁇ arrestin2. Further, antibody-drug conjugates can increase the efficiency of antibody internalization. Efficient antibody internalization can be desirable for delivering functional P-arrestin2 specific antibodies to the intracellular environment for screening ligands or compounds predicted to bind GPCRs for selective activation of P-arrestin2 pathways. Conjugation of antibodies to a variety of agents that can facilitate cellular internalization of antibodies is known in the art (see generally Wu et al. (2005) Nat
- aptamers that specifically recognize and bind to functional ⁇ - arrestin2 nucleotides or proteins can be used to decrease the level of functional P-arrestin2.
- Aptamers are nucleic acids or peptide molecules selected from a large random sequence pool to bind to specific target molecule. The small size of aptamers makes them easier to synthesize and chemically modify and enables them to access epitopes that otherwise might be blocked or hidden. And aptamers are generally nontoxic and weak antigens because of their close resemblance to endogenous molecules. Generation, selection, and delivery of aptamers is within the skill of the art (see e.g., Lee et al. (2006) Curr Opin Chem Biol.
- aptamers can finely discriminate between molecular variants.
- Aptamers can also be used to temporally and spatially regulate protein function (e.g., functional P-arrestin2 function) in cells and organisms.
- protein function e.g., functional P-arrestin2 function
- the ligand-regulated peptide (LiRP) system provides a general method where the binding activity of intracellular peptides is controlled by a peptide aptamer in turn regulated by a cell-permeable small molecule (see e.g., Binkowski (2005) Chem & Biol.
- aptamers can provide an effective means to decrease functional -arrestin2 levels by, for example, directly binding the functional P-arrestin2 mRNA and/or functional P-arrestin2 expressed protein.
- Antisense nucleic acid molecules within the invention are those that specifically hybridize (e.g., bind) under cellular conditions to cellular mRNA and/or genomic DNA encoding, for example functional p-arrestin2 protein, in a manner that inhibits expression of that protein, e.g., by inhibiting transcription and/or translation.
- Antisense molecules effective for decreasing functional p-arrestin2 levels, can be designed, produced, and administered by methods commonly known to the art (see e.g., Chan et al. (2006) Clinical and Experimental Pharmacology and Physiology 33(5-6), 533-540).
- Ribozyme molecules designed to catalytically cleave target mRNA transcripts can also be used to decrease levels of functional P-arrestin2.
- Ribozyme molecules specific for functional P-arrestin2 can be designed, produced, and administered by methods commonly known to the art (see e.g., Fanning and Symonds (2006) Handbook Experimental Pharmacology 173, 289-303G, reviewing therapeutic use of hammerhead ribozymes and small hairpin RNA).
- Triplex-forming oligonucleotides can also be used to decrease levels of related molecules with similar activity (see generally, Rogers et al. (2005) Current Medicinal Chemistry 5(4), 319-326).
- dsRNAs or other compounds for inhibiting functional -arrestin2 are used in animal models, these dsRNAs or other compounds must be effectively administered to the animals in question. Any suitable methods of administration may be used, and it is understood that many such methods of administration will be readily apparent to those of skill in the art upon reading this disclosure. [0120] Screening
- another aspect of the invention is directed to a system for screening candidate agents predicted to bind GPCRs for selective activation of functional ⁇ - arrestin2 expression.
- Assays can be performed on living mammalian cells, which more closely approximate the effects of a particular serum level of drug in the body. Studies using extracts offer the possibility of a more rigorous determination of direct agent/enzyme interactions. Exemplary screening methods are detailed in the examples, below.
- screening is accomplished by administering to a subject an amount of a compound predicted to bind a GPCR sufficient to generate a physiological response, evaluating that response, administering to the subject an effect amount of a dsRNA (such as, for example, a siRNA) capable of binding a P-arrestin2 RNA, allowing the binding to occur, and then evaluating the response once again. If the physiological response is reversed at the time of the second evaluation, then the compound being evaluated is a P-arrestin2 biased compound.
- the physiological response can be any response mediated by -arrestin2.
- a reversal may include an increase of an observed physiological response, while with respect to other responses a reversal may include a decrease of an observed physiological response.
- the physiological reponse may, for example, relate to a LPS-induced or TNF-a-induced inflammatory response, or may relate to a LPS-induced cytokine secretion.
- the phrase "relate to" with respect to a physiological response can refer to an increase in the measured physiological response, a decrease in the measured physiological response, or any other measurable variation in the physiological response.
- kits can include a compound of the present invention, optionally one or more ingredients for preparing a pharmaceutically acceptable formulation of the compound, and instructions for use (e.g., administration).
- a kit can include a compound of the present invention, optionally one or more ingredients for preparing a pharmaceutically acceptable formulation of the compound, and instructions for use (e.g., administration).
- different components of a compound formulation can be packaged in separate containers and admixed immediately before use.
- Such packaging of the components separately can, if desired, be presented in a pack or dispenser device which may contain one or more unit dosage forms containing the compound.
- the pack may, for example, comprise metal or plastic foil such as a blister pack.
- Such packaging of the components separately can also, in certain instances, permit long-term storage without losing activity of the components.
- the different components can be packaged separately and not mixed prior to use.
- the different components can be packaged in one combination for
- Kits may include reagents in separate containers such as, for example, sterile water or saline to be added to a lyophilized active component packaged separately.
- sealed glass ampules may contain lyophilized superoxide dismutase mimetics and in a separate ampule, sterile water, sterile saline or sterile each of which has been packaged under a neutral non-reacting gas, such as nitrogen.
- Ampules may consist of any suitable material, such as glass, organic polymers, such as polycarbonate, polystyrene, ceramic, metal or any other material typically employed to hold reagents.
- suitable containers include bottles that may be fabricated from similar substances as ampules, and envelopes that may consist of foil-lined interiors, such as aluminum or an alloy.
- Other containers include test tubes, vials, flasks, bottles, syringes, and the like.
- Containers may have a sterile access port, such as a bottle having a stopper that can be pierced by a hypodermic injection needle.
- Other containers may have two compartments that are separated by a readily removable membrane that upon removal permits the components to mix.
- Removable membranes may be glass, plastic, rubber, and the like.
- kits can be supplied with instructional materials. Instructions may be printed on paper or other substrate, and/or may be supplied as an electronic-readable medium, such as a floppy disc, mini-CD-ROM, CD-ROM, DVD-ROM, videotape, audio tape, and the like. Detailed instructions may not be physically associated with the kit; instead, a user may be directed to an Internet web site specified by the manufacturer or distributor of the kit, or supplied as electronic mail.
- kits in another aspect of the present invention, includes an effective amount of at least one siR A molecule capable of binding a ⁇ -arrestin RNA, and written indicia providing a user of the kit with instructions for using the kit in accordance with one of the methods described herein.
- the kit further includes cells suitable for use in evaluating a compound predicted to bind a GPCR in an amount sufficient to evaluate the compound.
- the cells provided in the kit are mouse monocyte cells.
- the mouse monocyte cells provided are from the cell line Raw 264.7.
- Example 1 GPR120, Inflammation, Diabetes and Obesity
- Fatty acids can function as endogenous ligands modulating inflammatory responses, but not all FAs work in the same way.
- saturated FAs SFAs
- unsaturated FAs are weakly pro-inflammatory or neutral
- G)3-FAs can be anti-inflammatory (Lee et al., 2003; Calder, 2005; Solinas et al., 2007).
- GPR40, 41 , 43, 84, and 120 GPR40, 41 , 43, 84, and 120. Based on its tissue expression pattern, GPR120 emerged as a receptor of particular interest. As seen in Fig.
- GPR120 is the only lipid sensing GPCR which is highly expressed in adipose tissue, pro-inflammatory CD1 lc+ macrophages (BMDCs), mature adipocytes, and monocytic RAW 264.7 cells (Fig. 1A and I B).
- BMDCs pro-inflammatory CD1 lc+ macrophages
- Fig. 1A and I B monocytic RAW 264.7 cells
- GPR120 is induced in the stromal vascular fraction (SVF) of adipose tissue (which contains the macrophages), as well as in hepatic Kupffer cells, during high fat diet (HFD) feeding in mice (Fig. 1 C).
- GPR120 is also expressed in enteroendocrine L cells with negligible expression in muscle (Fig. 1 1 C), hepatocytes or other cell types (Hirasawa et al., 2005, Gotoh et al., 2007).
- GPR120 signals via a Gaq/1 1 -coupled pathway and can respond to long chain FAs (Hirasawa et al., 2005).
- Glaxo published GW9508 as a GPR40 selective agonist.
- this compound was not specific and also stimulated GPR120 (Briscoe et al., 2006). Since macrophages and adipocytes do not express GPR40 (this was confirmed by repeated q-PCR and RT-PCR measures, Fig. 8A), GW9508 is a functional GPR120 specific compound in these cell types.
- GW9508 treatment broadly and markedly repressed the ability of the TLR4 ligand LPS to stimulate inflammatory responses in RAW 264.7 cells (Fig. ID and E).
- GW9508 inhibited LPS stimulated phosphorylation of ⁇ and JN , prevented ⁇ degradation, and inhibited TNF-a and IL-6 secretion. All of these effects of GW9508 were completely abrogated by siRNA mediated knockdown of GPR120 (Fig. ID, E and Fig. 8F).
- GW9508 ligand we found that GW9508, the ⁇ -3 FAs (DHA and EPA) and palmitoleate (CI 6: ln7), all activated the SRE-luc reporter with an EC 50 of 1-10 ⁇ (Fig. 2 A), while SFAs were without effect. GW9508 and DHA were used at 100 ⁇ in all subsequent studies to achieve maximal action.
- the co-3 FAs (DHA and oc-linolenic acid), and SFA (palmitic acid (C16:0)) activated ERK phosphorylation in RAW 264.7 cells, but only DHA- and a-linolenic acid-mediated ERK phosphorylation were abolished by GPR120
- RNA interference was not affected by ⁇ -arrestin 1 , 2 or Gaq/1 1 knockdown.
- ⁇ -arrestin2 knockdown DHA-mediated anti-inflammatory signaling was extinguished, while ⁇ -arrestinl and Gaq/1 1 knockdown were without effect (Fig. 3 A).
- Fig. 3 A and Fig. 9 show that GPR120 stimulation inhibits both TLR4- and TNF-a mediated inflammatory responses. Since the TNF-a and TLR signaling cascades converge downstream of GPR120 activation, these results indicate that the site of GPR120- induced inhibition is either at, or upstream, of JNK/ ⁇ . LPS activates inflammation through the TLR4 pathway by engaging the serine kinase IRAK, leading to phosphorylation of transforming growth factor- ⁇ activated kinase 1 (TAK1) which is upstream of
- TNF-a and TLR2/3 also leads to stimulation of TAK1 , resulting in activation of ⁇ and JNK (Takaesu et al., 2003).
- ⁇ can translocate to a number of GPCRs where it mediates receptor internalization and signaling (Barak et al., 1997).
- ⁇ can translocate to a number of GPCRs where it mediates receptor internalization and signaling (Barak et al., 1997).
- Fig. 3D In the basal state, GPR120 was localized to the plasma membrane as assessed by immunostaining (red fluorescence, Fig. 3D), while ⁇ - arrestin2 exhibited a diffuse, largely cytoplasmic staining pattern (green, Fig. 3D).
- TAK1 binding protein 1 TAK1 binding protein 1
- Fig. 3H shows that LPS stimulation of RAW 264.7 cells causes TAB1/TAK1 association.
- DHA treatment leads to the association of ⁇ - arrestin2 with TAB 1 (Fig. 3G) and largely blocks TAK1/TAB 1 association (Fig. 3H).
- Fig. 9D To further examine the interaction site of p-arrestin2 and GPR120 or TAB1 , we pursued co- immunoprecipitation with a series of -arrestin2 truncation/deletion mutants.
- GPR120 activation enhances glucose uptake in 3T3-L1 adipocytes.
- GPR120 KO mice and WT littermates were evaluated on normal chow diet (NC). Body weights were similar in both groups, and as summarized in Fig. 5, glucose tolerance tests (GTT) showed a mild degree of impairment in GPR120 KO animals compared to WTs (Fig. 5A). More impressively, insulin secretion was more than 2-fold greater in the KO animals, and the combination of hyperinsulinemia and mild glucose intolerance indicates the presence of insulin resistance (Fig. 5B and C). This was confirmed by performing
- hyperinsulinemic/euglycemic clamp studies in the chow fed WT and KO mice (Fig. 5D). These studies revealed a 31% decrease in the glucose infusion rate (GIR) required to maintain euglycemia in the KO mice. Since 70-80% of total body insulin stimulated glucose disposal is accounted for by skeletal muscle glucose uptake (Baron et al, 1988), the decreased insulin stimulated (IS)-glucose disposal rate (GDR) provides direct evidence for skeletal muscle insulin resistance in the KO mice. Likewise, the GPR120 KO mice exhibited a marked decrease in the ability of insulin to suppress hepatic glucose production (HGP), demonstrating the presence of hepatic insulin resistance.
- HGP hepatic glucose production
- the decreased GIR was -50% related to muscle and -50% due to liver insulin resistance, respectively. Since the chow diet contains exogenous ⁇ -3 FAs, we conclude that blunted ⁇ -3 FA signaling in the KO mice, accounts for the decreased insulin sensitivity.
- WT and GPR120 KO mice were placed on 60% HFD for 15 weeks. At this point, separate groups of 15 mice each, were treated for five additional weeks with 60% HFD or an isocaloric HFD diet containing 27% fish oil supplementation enriched in ⁇ -3 FAs. This diet provided 50 and 100 mg of DHA and EPA, respectively, per mouse, per day. Fig.
- 5E shows that administration of the co-3 FA diet led to improved insulin sensitivity with increased glucose infusion rates, enhanced muscle insulin sensitivity (increased IS-GDR), greater hepatic insulin sensitivity (increased HGP suppression), and decreased hepatic steatosis (Fig. 13A and B).
- the ⁇ -3 FA diet was completely without effect in the GPR120 KO mice.
- a separate group of WT mice were treated with the insulin sensitizing thiazolidinedione Rosiglitazone, and the effects of co-3 FAs were equal to or greater (HGP suppression) than the effects of this clinically used insulin sensitizing drug.
- ⁇ -3 FA treatment had a beneficial effect on hepatic lipid metabolism, causing decreased liver triglycerides, DAGs, along with reduced SFA and co-6 FA content in the various lipid classes (Fig. 13A-C and Table 2).
- the co-3 FA supplementation was entirely without effect, or much less effective, at reducing hepatic lipid levels in the GPR120 KOs.
- BMT bone marrow transplantation
- HFD led to a large but comparable increase in CD1 lb+ and CD1 lc+ ATM content in WT and GPR120 KO mice (Fig. 6B, middle panel).
- Treatment with the co-3 FA-enriched HFD caused a striking decrease in CD1 lb+ and CD1 l c+ ATMs in WT mice, but was without effect in the GPR120 KO group (Fig. 6B, right panel).
- the FACS analysis was fully consistent with the histological results.
- CD1 lc+ ATM content was also greater in the GPR120 KOs on the chow diet relative to WT consistent with the insulin resistance in the KO animals.
- Omega-3 FAs decrease Ml pro-inflammatory gene and increase M2 anti-inflammatory gene expression in adipose tissue
- Ml inflammatory genes such as IL-6, TNF- , MCP-1 , IL- ⁇ ⁇ , iNOS, and CD1 lc was increased by HFD compared to chow diet in both genotypes, and was reduced in the co-3 FA treated WT mice, but not in the GPR120 KO mice. Even on chow diet, expression of several inflammatory genes was higher in GPR120 KOs compared to WT, consistent with the insulin resistance observed in the chow- fed KO mice.
- siRNAs were obtained from Dharmacon. Sequences for GPR120, ⁇ - arrestinl/2, GLUT4 and Gaq/1 1 siRNA duplexes are available upon request. RAW 264.7 cells and 3T3-L1 adipocytes at day 8 from differentiation protocol were collected and electroporated with siRNAs as described previously (GENE PULSER, Bio-Rad) (Yoshizaki et al., 2009). Further experiments were performed after 48 hr of electroporation.
- GMI Four-Color Real-Time System
- Arginase 1 ATGGAAGAGACCTTCAGCTAC 27
- Clec7a C-type lectin domain family 7, member a
- MMR macrophage mannose receptor
- HEK 293 and 3T3-L1 cells were maintained as described (Liao et al., 2007). Adipocytes were studied at 10-14 days post-differentiation. RAW 264.7 cells were maintained in 10% low endotoxin fetal bovine serum (Hyclone)/DMEM (1 g/1 of glucose). IPMacs were obtained from WT and GPR120 KO mice and cultured as described previously (Nguyen et al., 2007, Patsouris et al., 2009). L6 myoblast cells were maintained in 10% FBS/MEM until cells become confluent, and then changed medium to 2% serum contained MEM to differentiate into myocyte.
- HE 293 cells were plated in 60 mm culture dish at day before transfection.
- HA-tagged GPR120 or/and FLAG-tagged P-arrestin2 mutant were transfected using with Effectene reagent (Qiagen, Valencia CA) following as manufacturer's instructions.
- Effectene reagent Qiagen, Valencia CA
- cells were stimulated with 100 ⁇ DHA for 30 min (for interaction between GPR120 and P-arrestin2) or 1 hr (For endogenous TAB1 and P-arrestin2) before harvested cells subjected to immunoprecipitation and western blotting.
- HEK 293 cells were co-transfected with SRE-luc and either mouse GPR120 or pcDNA3 control plasmid (without GPR120) in 24-well plate. Forty-eight hr after transfection, cells were treated with the indicated concentration of various fatty acids for 6 hr. Cells were harvested, and luciferase activities in the cell extracts were measured as previously described (Oh et al., 2005, Fan et al., 2009).
- HA-GLUT4-eGFP expression vector were electroporated into day 8 post differentiated 3T3-L1 adipocytes for monitoring of GLUT4 translocation as previously described (Yoshizaki et al., 2007).
- 3T3-L1 adipocytes were pretreated for 30 min with the indicated drug; and stimulated with 3 ng/ml of insulin for 30 min at 37°C.
- L6 myocytes were pretreated for 30 min with 100 ⁇ DHA and then stimulated with 100 ng/ml of insulin for 30 min. Glucose uptake was measured as previously described
- Epididymal fat pads were collected from chow fed WT and GPR120 KO mice, respectively and then mince them by razor blade into smaller pieces (1 -2 cubic mm size). The minced tissues were suspended in Hepes-phosphate salt buffer (Hepes, 10 mM, KCl 4 mM, NaCl 125 mM, KH 2 P0 4 0.8 mM, Na 2 HP0 4 1.3 mM, MgCl 2 ImM, CaCl 2 ImM) and briefly centrifuged at 400 x g. Supernatant was removed from the bottom with a 20 gauge needle syringe.
- Hepes-phosphate salt buffer Hepes, 10 mM, KCl 4 mM, NaCl 125 mM, KH 2 P0 4 0.8 mM, Na 2 HP0 4 1.3 mM, MgCl 2 ImM, CaCl 2 ImM
- the floating fat explants were washed once and add the Hepes- phosphate salt buffer as 25% vol/vol (for example, 0.5 ml explants + 1.5 ml buffer) to plate desired aliquot into 12-well plate.
- the fat tissue explants were gently rotated at 37°C for 30 minutes and then pretreated with or without 100 ⁇ DHA for 30 minutes followed by 100 ng/ml of insulin treatment for 30 min to subject 2-DOG uptake assay.
- [3H]-2-DOG (1 ⁇ / ⁇ ) was added for 10 min and then terminate the incubation by adding of 10 ⁇ Cytochalasin B.
- collagenase (Type I; 5 mg/ml) solution was added to each well and incubated plate for another 30 min. Collagen digested sample were washed once and resuspended in IN NaOH. Transfer the rest to a vial and count for radioactivity.
- Proteins from tissues or cell lysates were extracted with radioimmune precipitation buffer in the presence of phosphatase inhibitors and protease inhibitors (Roche Applied Science). Twenty ⁇ of proteins/lane were separated on a 10% polyacrylamide, precast SDS gel (Bio-Rad) followed by transfer on polyvinylidene difluoride membrane (Immobilon, Millipore), and western blotting was performed as described (Nguyen et al., 2005) with indicated antibodies.
- lysates were incubated with 1 ⁇ g of anti-p-arrestin2, TAK1 antibody (Cell signaling), or GFP antibody (Santa Cruz Biotechnology) overnight at 4°C and immune complexes precipitated with Protein A/G- conjugated beads (Invitrogen). Beads were washed with PBS and resuspended in sample buffer. Lysates and immune complexes were separated by SDS-PAGE, subjected to western blotting.
- TNF-a and IL-6 were directly quantified from RAW 264.7 cells with or without GPR120 knockdown, or IPMacs culture medium. Cells were treated with 100 ⁇ GW9508 or DHA for 1 hr prior to treat 100 ng/ml LPS for 6 hr and then collect medium from the cells and subjected to ELISA according to the manufacturer's instructions
- BIOSOURCE For GLP-1 measurement, mouse blood was collected lhr after indicated diet feeding. Adequate inhibitors (protease inhibitors and Diprotin A as DPP IV inhibitor) were used while blood was collecting and then subjected to ELISA to the manufacturer's instructions (ALPCO, Salem, NH).
- mice were euthanized and slowly perfused by intracardiac injection with 10 ml of 1% paraformaldehyde diluted in PBS. Finger nail sized fat pad samples were excised and blocked for 1 hr in 5% BSA in PBS with gentle rocking at RT. For detection of intracellular antigens, blocking and subsequent incubations were done in 5% BSA in PBS with 0.3% Triton X-100. Primary antibodies were diluted in blocking buffer to 0.5-1 ⁇ g/ml and added to fat samples for overnight at 4°C.
- fluorochrome-conjugated secondary antibodies were added for 1 hr at RT. Fat pads were imaged on an inverted confocal microscope (Olympus Fluoview 1000). Anti-mouse antibodies used were against F4/80 and MGL1 (Abeam); Caveolinl (BD Biosciences).
- In vitro chemotaxis assay was performed as previously described (Patsouris et al., 2009). Briefly, mature 3T3-L1 adipocytes, more than 99% of cells showing large lipids droplets (12 days after differentiation protocol initiation), were used for preparation of conditioned media. Treatment with the 100 ⁇ DHA for 6 hr was performed in serum-free DMEM cultured IPMacs. For the migration per se, 100,000 IPMacs from WT or GPR120 KO mice were used per condition. The IPMacs were placed in the upper chamber of an 8 ⁇ polycarbonate filter (24-transwell format; Corning, Lowell, MA), whereas adipocyte conditioned medium or MCP-1 treatment was placed in the lower chamber. After 3 hr of migration, cells were fixed in formalin and stained with 4', 6-diamidino-2-phenylindole and observed.
- FACS Fluorescence-activated Cell Sorting
- Epididymal fat pads were weighed, rinsed in phosphate-buffered saline, and then minced in FACS buffer (phosphate-buffered saline plus 1% BSA).
- Adipocytes and stromal vascular cells were prepared from collagenase digested adipose tissue as described previously (Nguyen et al., 2007). FACS analysis of stromal vascular cells for macrophage content and subtypes were performed as previously described (Nguyen et al., 2007).
- GTT Glucose tolerance tests
- Glucose infusion rate was adjusted until steady-state blood glucose (120 mg/dl, ⁇ 5 mg/dl) was achieved.
- the mouse were exsanguinated by cardiac puncture (>1 ml, whole blood collected), and tissues were harvested, mass recorded, and preserved as required for future analysis.
- clamp studies were performed on WT animals on 60% HFD for 15 weeks.
- BMT bone marrow transplantation
- DHA and EPA concentrations in plasma TG and liver total TAG were measured by Lipomics, Inc. (West Sacramento, CA).
- the lipids were extracted from plasma and tissues in the presence of authentic internal standards using chloroform mixed with methanol (2: 1 vol/vol) (Folch et al., 1957) and individual lipid classes were separated by HPLC (Cao et al., 2008).
- Lipid class fractions were transesterified in 1% sulfuric acid (in methanol) in a sealed vial with nitrogen at 100°C for 45 min.
- Fatty acid methyl esters were extracted from the mixture with hexane containing 0.05% butylated hydroxytoluene, and readied for gas chromatography under nitrogen.
- fatty acid methyl esters were separated and quantified by capillary gas chromatography equipped with a 30 m DB-88MS capillary column and a flame-ionization detector.
- GPR120 functions as an co-3 FA receptor/sensor in pro-inflammatory macrophages and mature adipocytes.
- DHA and EPA the major natural co-3 FA constituents of fish oil
- GPR120-mediated anti-inflammation involves inhibition of TAKl through a P-arrestin2/TAB 1 dependent effect. Since chronic tissue inflammation is an important mechanism causing insulin resistance (Xu et al., 2003, Shoelson et al., 2007, Schenk et al., 2008), the anti-inflammatory actions of ⁇ -3 FAs exert potent insulin sensitizing effects.
- GPR120 The in vivo anti-inflammatory and insulin sensitizing effects of co-3 FAs are dependent on expression of GPR120, as demonstrated in studies of obese GPR120 KO animals and WT littermates.
- GPR120 is highly expressed in pro-inflammatory macrophages and functions as an co-3 FA receptor, mediating the anti-inflammatory effects of this class of FAs to inhibit both the TLR2/3/4 and the TNF-a response pathways and cause systemic insulin sensitization.
- GPR120 is a Gaq/1 1-coupled receptor, and since it is expressed in enteroendocrine L cells, past interest in this receptor has focused on its potential ability to stimulate L cell GLP-1 secretion.
- GPR120 is highly expressed in pro-inflammatory, Ml -like macrophages and mature adipocytes, with negligible expression in muscle, pancreatic ⁇ -cells, and hepatocytes (Gotoh et al., 2007).
- GPR120 expression is highly induced in ATMs as well as resident liver macrophages (Kupffer cells).
- TAKl activation stimulates both the ⁇ /NFKB and JNK/ API pathways, and the TLR and TNF-a signaling pathways converge at this step.
- Our data show that stimulation of GPR120 specifically inhibits TAKl phosphorylation and activation providing a common mechanism for the inhibition of both TLR and TNF-a signaling.
- Beta-arrestins can serve as important adaptor and scaffold molecules mediating the functions of a number of different GPCRs, as well as other receptor subtypes (Miller and Lefkowitz, 2001).
- the C-terminal region of GPR120 contains several putative P-arrestin2 binding motifs ((S/T)X4-5(S/T); Cen et al., 2001), but whether ⁇ -arrestins play any role in GPR120 function was unknown.
- P-arrestin2 binding motifs (S/T)X4-5(S/T); Cen et al., 2001)
- ⁇ -arrestins play any role in GPR120 function was unknown.
- activation of GPR120 by DHA stimulation leads to association of the receptor with P-arrestin2, but not ⁇ -arrestinl , and that the anti-inflammatory effects of GPR120 are completely P-arrestin2 dependent.
- Functional immunocytochemical studies showed that DHA stimulation leads to recruitment of P-arrestin2 to the plasma membrane where it co-localizes with GPR120.
- TAB 1 is the activating protein for TAK1 and our results show that following DHA-stimulated internalization of the GPR120/p-arrestin2 complex, ⁇ - arrestin2 can now associate with TAB 1 , as measured in co-immunoprecipitation experiments; only full-length p-arrestin2 was capable of interacting with GPR120 and TAB1. This apparently blocks the association of TAB 1 with TAK1 , inhibiting TAK1 activation and downstream signaling to the ⁇ /NFKB and JNK/AP1 system.
- GPR120 is not expressed in muscle, and DHA did not stimulated glucose uptake in L6 myocytes (Fig. 1 1C and 1 ID).
- acute administration of DHA had no stimulatory effects on IS-GDR (Fig. 1 IE). This reports the conclusion that the in vivo stimulatory effects of DHA on GDR are related to anti-inflammation, and that the glucose transport stimulatory effects in adipocytes contribute little to the overall phenotype.
- DHA treatment inhibited the ability of primary WT macrophages to migrate towards adipocyte CM. This could be due to DHA-induced decreased chemokine secretion or down regulation of chemokine receptors, or both.
- DHA by signaling through GPR120, can mediate heterologous desensitization of other GPCR chemokine receptors.
- M2 markers such as IL-10, arginase 1 , MGL1 , Ym-1 , Clec7a, and MMR.
- M2 markers such as IL-10, arginase 1 , MGL1 , Ym-1 , Clec7a, and MMR.
- co-3 FAs can redirect ATMs from an Ml to an M2 polarization state.
- these mechanisms account for the decreased inflammatory state.
- the in vivo anti-inflammatory actions of ⁇ -3 FAs are consistent with the insulin sensitizing effects of these agents and are fully dependent on the presence of GPR120, indicating a causal relationship.
- CE Cholesterol ester
- DAG diacylgiycerol
- FFA free fatty acid
- LYPC lysophosphatidylcholine
- PC phosphatidylcholine
- PE phosphatitylethanolamine
- Dietary DHA is rapidly esterified into chylomicrons during the process of gastrointestinal absorption, and is also packaged into VLDL triglycerides by the liver. DHA can also be esterified into phospholipids and cholesterol esters associated with circulating lipopoproteins and only a small proportion ( ⁇ 5%) of total plasma DHA is found in the FFA pool.
- co-3 FAs are cleaved from circulating triglycerides where they can act as ligands or be taken up by peripheral tissues (Polozova and Salem Jr., 2007).
- GPR120 functions as an co-3 FA receptor/sensor and mediates robust and broad anti-inflammatory effects, particularly in macrophages.
- GPR120 couples to P-arrestin2 which is followed by receptor endocytosis and inhibition of TAB 1 -mediated activation of TAK1 , providing a mechanism for inhibition of both the TLR and TNF-a pro-inflammatory signaling pathways.
- TAB 1 receptor endocytosis
- TAB 1 TAB 1 -mediated activation of TAK1
- GW9508 was purchased from Tocris bioscience (Ellisville, MO) and DHA was from Cayman chemical (Ann Arbor, MI). All other chemicals were purchased from Sigma unless mentioned otherwise.
- mice were switched to an isocaloric HFD-containing 27% menhaden fish oil replacement (wt/wt; menhaden fish oil: 16% EPA (C20:5n3), 9%, DHA (C22:6n3), Research Diet) (Jucker et al., 1999, Neschen et al., 2007) and fed for 5 weeks. Mice received fresh diet every 3rd day, and food
- beta-arrestin2 as a G protein-coupled receptor-stimulated regulator of NF- kappaB pathways. Mol Cell 14, 303-317.
- Phenotypic switching of adipose tissue macrophages with obesity is generated by spatiotemporal differences in macrophage subtypes. Diabetes 57, 3239-3246.
- TAK1 is critical for IkappaB kinase-mediated activation of the NF-kappaB pathway. J Mol Biol 326, 105-1 15.
- Bone marrow-specific Cap gene deletion protects against high-fat diet-induced insulin resistance. Nat Med 13, 455-462.
- Myosin 5a is an insulin-stimulated Akt2 (protein kinase Bbeta) substrate modulating GLUT4 vesicle translocation. Mol Cell Biol 27, 5172-5183.
- SIRT1 exerts anti -inflammatory effects and improves insulin sensitivity in adipocytes. Mol Cell Biol 29, 1363-1374.
Landscapes
- Health & Medical Sciences (AREA)
- Veterinary Medicine (AREA)
- Chemical & Material Sciences (AREA)
- Medicinal Chemistry (AREA)
- Pharmacology & Pharmacy (AREA)
- Life Sciences & Earth Sciences (AREA)
- Animal Behavior & Ethology (AREA)
- General Health & Medical Sciences (AREA)
- Public Health (AREA)
- Epidemiology (AREA)
- Pain & Pain Management (AREA)
- Rheumatology (AREA)
- Chemical Kinetics & Catalysis (AREA)
- General Chemical & Material Sciences (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Organic Chemistry (AREA)
- Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
- Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
Abstract
La présente invention concerne des procédés de traitement d'une réponse à médiation par la ß-arrestine2 et/ou GPR 120 chez un sujet. La réponse à médiation par la ß-arrestine2 et/ou GPR 120 peut être une inflammation, notamment le diabète, une inflammation associée à l'obésité et l'obésité. Les procédés peuvent comprendre l'administration à un sujet d'une quantité thérapeutiquement efficace d'un composé prévu pour se lier à une molécule de ß-arrestine2 et/ou GPR 120, le composé activant de manière sélective une voie de signalisation dépendante de la ß-arrestine2 de GPR 120.
Priority Applications (1)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
US13/493,241 US8987332B2 (en) | 2009-12-09 | 2012-06-11 | Methods of treating inflammatory conditions |
Applications Claiming Priority (6)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
US28508609P | 2009-12-09 | 2009-12-09 | |
US61/285,086 | 2009-12-09 | ||
US37760110P | 2010-08-27 | 2010-08-27 | |
US61/377,601 | 2010-08-27 | ||
US37962610P | 2010-09-02 | 2010-09-02 | |
US61/379,626 | 2010-09-02 |
Related Child Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
US13/493,241 Continuation US8987332B2 (en) | 2009-12-09 | 2012-06-11 | Methods of treating inflammatory conditions |
Publications (1)
Publication Number | Publication Date |
---|---|
WO2011072132A1 true WO2011072132A1 (fr) | 2011-06-16 |
Family
ID=44145916
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/US2010/059699 WO2011072132A1 (fr) | 2009-12-09 | 2010-12-09 | Procédés de traitement d'états inflammatoires |
Country Status (2)
Country | Link |
---|---|
US (1) | US8987332B2 (fr) |
WO (1) | WO2011072132A1 (fr) |
Cited By (6)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO2014011926A1 (fr) | 2012-07-11 | 2014-01-16 | Elcelyx Therapeutics, Inc. | Compositions comportant des statines, des biguanides et d'autres agents pour réduire un risque cardiométabolique |
WO2015125085A1 (fr) | 2014-02-19 | 2015-08-27 | Piramal Enterprises Limited | Composés pouvant être utilisés à titre d'agonistes de gpr120 |
WO2016125182A1 (fr) | 2015-02-05 | 2016-08-11 | Piramal Enterprises Limited | Composés contenant une séquence de liaison carbone-carbone en tant qu'agonistes gpr120 |
WO2017158355A1 (fr) * | 2016-03-15 | 2017-09-21 | Queen Mary University Of London | Méthode de traitement de l'obésité |
US10214521B2 (en) | 2014-09-11 | 2019-02-26 | Piramal Enterprises Limited | Fused heterocyclic compounds as GPR120 agonists |
US10273230B2 (en) | 2014-07-25 | 2019-04-30 | Piramal Enterprises Limited | Substituted phenyl alkanoic acid compounds as GPR120 agonists and uses thereof |
Families Citing this family (1)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US10682397B2 (en) * | 2015-12-04 | 2020-06-16 | Massachusetts Institute Of Technology | Methods of treating fragile X syndrome and related disorders |
Citations (4)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US20060026702A1 (en) * | 2002-03-19 | 2006-02-02 | Duke University | Phosphoinositide 3-kinase mediated inhibition of GPCRs |
WO2007134613A1 (fr) * | 2006-05-24 | 2007-11-29 | Rheoscience A/S | Modulation de l'activité des gpr120 dans les adipocytes/tissu graisseux |
WO2008074839A2 (fr) * | 2006-12-19 | 2008-06-26 | Ablynx N.V. | Séquences d'acides aminés dirigées contre les gpcr et polypeptides les contenant pour le traitement de maladies et de troubles liés au gpcr |
US20090274730A1 (en) * | 2007-10-25 | 2009-11-05 | Revalesio Corporation | Compositions and methods for treating inflammation |
-
2010
- 2010-12-09 WO PCT/US2010/059699 patent/WO2011072132A1/fr active Application Filing
-
2012
- 2012-06-11 US US13/493,241 patent/US8987332B2/en active Active
Patent Citations (4)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US20060026702A1 (en) * | 2002-03-19 | 2006-02-02 | Duke University | Phosphoinositide 3-kinase mediated inhibition of GPCRs |
WO2007134613A1 (fr) * | 2006-05-24 | 2007-11-29 | Rheoscience A/S | Modulation de l'activité des gpr120 dans les adipocytes/tissu graisseux |
WO2008074839A2 (fr) * | 2006-12-19 | 2008-06-26 | Ablynx N.V. | Séquences d'acides aminés dirigées contre les gpcr et polypeptides les contenant pour le traitement de maladies et de troubles liés au gpcr |
US20090274730A1 (en) * | 2007-10-25 | 2009-11-05 | Revalesio Corporation | Compositions and methods for treating inflammation |
Non-Patent Citations (5)
Cited By (9)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO2014011926A1 (fr) | 2012-07-11 | 2014-01-16 | Elcelyx Therapeutics, Inc. | Compositions comportant des statines, des biguanides et d'autres agents pour réduire un risque cardiométabolique |
WO2015125085A1 (fr) | 2014-02-19 | 2015-08-27 | Piramal Enterprises Limited | Composés pouvant être utilisés à titre d'agonistes de gpr120 |
US10227360B2 (en) | 2014-02-19 | 2019-03-12 | Piramal Enterprises Limited | Compounds for use as GPR120 agonists |
US10273230B2 (en) | 2014-07-25 | 2019-04-30 | Piramal Enterprises Limited | Substituted phenyl alkanoic acid compounds as GPR120 agonists and uses thereof |
US10214521B2 (en) | 2014-09-11 | 2019-02-26 | Piramal Enterprises Limited | Fused heterocyclic compounds as GPR120 agonists |
WO2016125182A1 (fr) | 2015-02-05 | 2016-08-11 | Piramal Enterprises Limited | Composés contenant une séquence de liaison carbone-carbone en tant qu'agonistes gpr120 |
WO2017158355A1 (fr) * | 2016-03-15 | 2017-09-21 | Queen Mary University Of London | Méthode de traitement de l'obésité |
US10925849B2 (en) | 2016-03-15 | 2021-02-23 | Queen Mary University Of London | Method of treatment of obesity |
US12023313B2 (en) | 2016-03-15 | 2024-07-02 | Queen Mary University Of London | Method of treatment of obesity |
Also Published As
Publication number | Publication date |
---|---|
US20120295975A1 (en) | 2012-11-22 |
US8987332B2 (en) | 2015-03-24 |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
US8987332B2 (en) | Methods of treating inflammatory conditions | |
Talukdar et al. | GPR120 is an omega-3 fatty acid receptor mediating potent anti-inflammatory and insulin-sensitizing effects | |
KR20210010529A (ko) | 안지오텐시노젠(AGT) iRNA 조성물 및 이들의 사용 방법 | |
US8906875B2 (en) | Methods of treating vascular inflammatory disorders | |
US20130150431A1 (en) | Mir-33 inhibitors and uses thereof | |
Kawasaki et al. | Blockade of B-cell-activating factor signaling enhances hepatic steatosis induced by a high-fat diet and improves insulin sensitivity | |
US11319368B2 (en) | Treatment of hepatotoxicity with IL-11 antibody | |
US20210177940A2 (en) | Treatment and prevention of metabolic diseases | |
WO2014122660A1 (fr) | Inhibiteurs de cd14 en tant que traitement efficace pour l'infection par vhc | |
Wang et al. | Pentraxin-3 is a TSH-inducible protein in human fibrocytes and orbital fibroblasts | |
Jin et al. | Babam2 negatively regulates osteoclastogenesis by interacting with Hey1 to inhibit Nfatc1 transcription | |
WO2014209947A2 (fr) | Méthodes et compositions permettant d'inhiber le trpv4 | |
WO2006068133A1 (fr) | Procede de suppression de l’expression de nfat2 | |
US20200182860A1 (en) | Methods for identifying therapeutic agents which interact with stk24 | |
US9732346B2 (en) | SiRNA targeting PRK2, which is hepatitis C virus therapeutic agent | |
US8815799B2 (en) | Inhibiting striated muscle activator of RHO (stars) to improve glycemic control | |
US20110015123A1 (en) | Modulation of GLUT4 Gene Promoter activity BY AHNAK | |
KR101668954B1 (ko) | HCV IRES를 표적으로 하는 C형 간염 바이러스 치료제 siRNA | |
EA047713B1 (ru) | Лечение гепатотоксичности | |
JP2008245581A (ja) | カルパイン遺伝子をノックダウンする核酸 | |
KR20050012082A (ko) | 노화세포의 생물학적 기능 회복 방법 |
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: 10836687 Country of ref document: EP Kind code of ref document: A1 |
|
NENP | Non-entry into the national phase |
Ref country code: DE |
|
122 | Ep: pct application non-entry in european phase |
Ref document number: 10836687 Country of ref document: EP Kind code of ref document: A1 |