KR100755872B1 - Pharmaceutical composition for preventing and treating inflammation containing midazolam - Google Patents
Pharmaceutical composition for preventing and treating inflammation containing midazolam Download PDFInfo
- Publication number
- KR100755872B1 KR100755872B1 KR1020060096894A KR20060096894A KR100755872B1 KR 100755872 B1 KR100755872 B1 KR 100755872B1 KR 1020060096894 A KR1020060096894 A KR 1020060096894A KR 20060096894 A KR20060096894 A KR 20060096894A KR 100755872 B1 KR100755872 B1 KR 100755872B1
- Authority
- KR
- South Korea
- Prior art keywords
- midazolam
- inflammation
- inhibition
- pharmaceutical composition
- lps
- Prior art date
Links
- 229960003793 midazolam Drugs 0.000 title claims abstract description 70
- DDLIGBOFAVUZHB-UHFFFAOYSA-N midazolam Chemical compound C12=CC(Cl)=CC=C2N2C(C)=NC=C2CN=C1C1=CC=CC=C1F DDLIGBOFAVUZHB-UHFFFAOYSA-N 0.000 title claims abstract description 70
- 230000004054 inflammatory process Effects 0.000 title claims abstract description 35
- 206010061218 Inflammation Diseases 0.000 title claims abstract description 34
- 239000008194 pharmaceutical composition Substances 0.000 title claims abstract description 18
- 230000000694 effects Effects 0.000 claims abstract description 36
- 230000005764 inhibitory process Effects 0.000 claims abstract description 25
- 238000004519 manufacturing process Methods 0.000 claims abstract description 23
- 230000014509 gene expression Effects 0.000 claims abstract description 14
- 230000003110 anti-inflammatory effect Effects 0.000 claims abstract description 11
- 230000005012 migration Effects 0.000 claims abstract description 11
- 238000013508 migration Methods 0.000 claims abstract description 11
- 210000003855 cell nucleus Anatomy 0.000 claims abstract description 4
- 239000003085 diluting agent Substances 0.000 claims abstract description 4
- 239000003937 drug carrier Substances 0.000 claims abstract description 4
- 239000000546 pharmaceutical excipient Substances 0.000 claims abstract description 4
- 102100038280 Prostaglandin G/H synthase 2 Human genes 0.000 claims description 23
- 102000011779 Nitric Oxide Synthase Type II Human genes 0.000 claims description 22
- 108010076864 Nitric Oxide Synthase Type II Proteins 0.000 claims description 22
- 108050003267 Prostaglandin G/H synthase 2 Proteins 0.000 claims description 21
- 102000002574 p38 Mitogen-Activated Protein Kinases Human genes 0.000 claims description 14
- 108010068338 p38 Mitogen-Activated Protein Kinases Proteins 0.000 claims description 14
- 238000000034 method Methods 0.000 claims description 10
- 108090000623 proteins and genes Proteins 0.000 claims description 6
- 230000002265 prevention Effects 0.000 claims description 5
- 230000027405 negative regulation of phosphorylation Effects 0.000 claims description 3
- 108010037462 Cyclooxygenase 2 Proteins 0.000 claims description 2
- 230000002401 inhibitory effect Effects 0.000 abstract description 12
- 230000026731 phosphorylation Effects 0.000 abstract description 6
- 238000006366 phosphorylation reaction Methods 0.000 abstract description 6
- 239000000203 mixture Substances 0.000 abstract description 4
- 108010057466 NF-kappa B Proteins 0.000 abstract 2
- 102000003945 NF-kappa B Human genes 0.000 abstract 2
- 101150071146 COX2 gene Proteins 0.000 abstract 1
- 101150109636 Inos gene Proteins 0.000 abstract 1
- 102100029438 Nitric oxide synthase, inducible Human genes 0.000 abstract 1
- 210000004027 cell Anatomy 0.000 description 38
- 210000002540 macrophage Anatomy 0.000 description 23
- 150000003254 radicals Chemical class 0.000 description 17
- 230000004913 activation Effects 0.000 description 12
- 238000002474 experimental method Methods 0.000 description 12
- 102000004127 Cytokines Human genes 0.000 description 10
- 108090000695 Cytokines Proteins 0.000 description 10
- MWUXSHHQAYIFBG-UHFFFAOYSA-N Nitric oxide Chemical compound O=[N] MWUXSHHQAYIFBG-UHFFFAOYSA-N 0.000 description 10
- 102100035100 Transcription factor p65 Human genes 0.000 description 9
- 230000000770 proinflammatory effect Effects 0.000 description 9
- 102100039337 NF-kappa-B inhibitor alpha Human genes 0.000 description 8
- 101710083073 NF-kappa-B inhibitor alpha Proteins 0.000 description 8
- FAPWRFPIFSIZLT-UHFFFAOYSA-M Sodium chloride Chemical compound [Na+].[Cl-] FAPWRFPIFSIZLT-UHFFFAOYSA-M 0.000 description 8
- 230000015556 catabolic process Effects 0.000 description 8
- 238000006731 degradation reaction Methods 0.000 description 8
- 230000002757 inflammatory effect Effects 0.000 description 8
- 239000003642 reactive oxygen metabolite Substances 0.000 description 8
- 238000001262 western blot Methods 0.000 description 8
- 102000043136 MAP kinase family Human genes 0.000 description 7
- 108091054455 MAP kinase family Proteins 0.000 description 7
- KNJDBYZZKAZQNG-UHFFFAOYSA-N lucigenin Chemical compound [O-][N+]([O-])=O.[O-][N+]([O-])=O.C12=CC=CC=C2[N+](C)=C(C=CC=C2)C2=C1C1=C(C=CC=C2)C2=[N+](C)C2=CC=CC=C12 KNJDBYZZKAZQNG-UHFFFAOYSA-N 0.000 description 7
- 108060001084 Luciferase Proteins 0.000 description 6
- 239000005089 Luciferase Substances 0.000 description 6
- 229940079593 drug Drugs 0.000 description 6
- 239000003814 drug Substances 0.000 description 6
- MZOFCQQQCNRIBI-VMXHOPILSA-N (3s)-4-[[(2s)-1-[[(2s)-1-[[(1s)-1-carboxy-2-hydroxyethyl]amino]-4-methyl-1-oxopentan-2-yl]amino]-5-(diaminomethylideneamino)-1-oxopentan-2-yl]amino]-3-[[2-[[(2s)-2,6-diaminohexanoyl]amino]acetyl]amino]-4-oxobutanoic acid Chemical compound OC[C@@H](C(O)=O)NC(=O)[C@H](CC(C)C)NC(=O)[C@H](CCCN=C(N)N)NC(=O)[C@H](CC(O)=O)NC(=O)CNC(=O)[C@@H](N)CCCCN MZOFCQQQCNRIBI-VMXHOPILSA-N 0.000 description 5
- ACFIXJIJDZMPPO-NNYOXOHSSA-N NADPH Chemical compound C1=CCC(C(=O)N)=CN1[C@H]1[C@H](O)[C@H](O)[C@@H](COP(O)(=O)OP(O)(=O)OC[C@@H]2[C@H]([C@@H](OP(O)(O)=O)[C@@H](O2)N2C3=NC=NC(N)=C3N=C2)O)O1 ACFIXJIJDZMPPO-NNYOXOHSSA-N 0.000 description 5
- 206010040047 Sepsis Diseases 0.000 description 5
- 108060008682 Tumor Necrosis Factor Proteins 0.000 description 5
- 102000000852 Tumor Necrosis Factor-alpha Human genes 0.000 description 5
- 208000027866 inflammatory disease Diseases 0.000 description 5
- 229930027945 nicotinamide-adenine dinucleotide Natural products 0.000 description 5
- SVUOLADPCWQTTE-UHFFFAOYSA-N 1h-1,2-benzodiazepine Chemical compound N1N=CC=CC2=CC=CC=C12 SVUOLADPCWQTTE-UHFFFAOYSA-N 0.000 description 4
- 108010085238 Actins Proteins 0.000 description 4
- 102000007469 Actins Human genes 0.000 description 4
- 102000007665 Extracellular Signal-Regulated MAP Kinases Human genes 0.000 description 4
- 238000003556 assay Methods 0.000 description 4
- 229940049706 benzodiazepine Drugs 0.000 description 4
- 210000004940 nucleus Anatomy 0.000 description 4
- 229940094443 oxytocics prostaglandins Drugs 0.000 description 4
- 150000003180 prostaglandins Chemical class 0.000 description 4
- 241000894006 Bacteria Species 0.000 description 3
- 102000004190 Enzymes Human genes 0.000 description 3
- 108090000790 Enzymes Proteins 0.000 description 3
- 108010007457 Extracellular Signal-Regulated MAP Kinases Proteins 0.000 description 3
- 102000008299 Nitric Oxide Synthase Human genes 0.000 description 3
- 108010021487 Nitric Oxide Synthase Proteins 0.000 description 3
- 231100000504 carcinogenesis Toxicity 0.000 description 3
- 238000001378 electrochemiluminescence detection Methods 0.000 description 3
- 239000002158 endotoxin Substances 0.000 description 3
- 238000003125 immunofluorescent labeling Methods 0.000 description 3
- 208000015181 infectious disease Diseases 0.000 description 3
- 230000028709 inflammatory response Effects 0.000 description 3
- 230000007246 mechanism Effects 0.000 description 3
- 230000001404 mediated effect Effects 0.000 description 3
- 239000012528 membrane Substances 0.000 description 3
- 229940021182 non-steroidal anti-inflammatory drug Drugs 0.000 description 3
- XJMOSONTPMZWPB-UHFFFAOYSA-M propidium iodide Chemical compound [I-].[I-].C12=CC(N)=CC=C2C2=CC=C(N)C=C2[N+](CCC[N+](C)(CC)CC)=C1C1=CC=CC=C1 XJMOSONTPMZWPB-UHFFFAOYSA-M 0.000 description 3
- 230000000451 tissue damage Effects 0.000 description 3
- 231100000827 tissue damage Toxicity 0.000 description 3
- CURLTUGMZLYLDI-UHFFFAOYSA-N Carbon dioxide Chemical compound O=C=O CURLTUGMZLYLDI-UHFFFAOYSA-N 0.000 description 2
- 208000005623 Carcinogenesis Diseases 0.000 description 2
- DHCLVCXQIBBOPH-UHFFFAOYSA-N Glycerol 2-phosphate Chemical compound OCC(CO)OP(O)(O)=O DHCLVCXQIBBOPH-UHFFFAOYSA-N 0.000 description 2
- 239000007995 HEPES buffer Substances 0.000 description 2
- 108010001336 Horseradish Peroxidase Proteins 0.000 description 2
- 206010028980 Neoplasm Diseases 0.000 description 2
- 102100022397 Nitric oxide synthase, brain Human genes 0.000 description 2
- 101710111444 Nitric oxide synthase, brain Proteins 0.000 description 2
- 102100028452 Nitric oxide synthase, endothelial Human genes 0.000 description 2
- 101710090055 Nitric oxide synthase, endothelial Proteins 0.000 description 2
- 102000004005 Prostaglandin-endoperoxide synthases Human genes 0.000 description 2
- 108090000459 Prostaglandin-endoperoxide synthases Proteins 0.000 description 2
- 108010071390 Serum Albumin Proteins 0.000 description 2
- 102000007562 Serum Albumin Human genes 0.000 description 2
- 239000004480 active ingredient Substances 0.000 description 2
- 230000003444 anaesthetic effect Effects 0.000 description 2
- 238000004458 analytical method Methods 0.000 description 2
- 239000007864 aqueous solution Substances 0.000 description 2
- 230000033228 biological regulation Effects 0.000 description 2
- 230000000903 blocking effect Effects 0.000 description 2
- 210000001772 blood platelet Anatomy 0.000 description 2
- 230000036952 cancer formation Effects 0.000 description 2
- 230000008859 change Effects 0.000 description 2
- 230000001419 dependent effect Effects 0.000 description 2
- 238000011161 development Methods 0.000 description 2
- 230000006870 function Effects 0.000 description 2
- 230000002496 gastric effect Effects 0.000 description 2
- 210000003734 kidney Anatomy 0.000 description 2
- 210000000265 leukocyte Anatomy 0.000 description 2
- 238000003670 luciferase enzyme activity assay Methods 0.000 description 2
- 239000000463 material Substances 0.000 description 2
- 239000003226 mitogen Substances 0.000 description 2
- 230000037361 pathway Effects 0.000 description 2
- 230000002093 peripheral effect Effects 0.000 description 2
- 230000028327 secretion Effects 0.000 description 2
- 230000019491 signal transduction Effects 0.000 description 2
- 239000011780 sodium chloride Substances 0.000 description 2
- UCSJYZPVAKXKNQ-HZYVHMACSA-N streptomycin Chemical compound CN[C@H]1[C@H](O)[C@@H](O)[C@H](CO)O[C@H]1O[C@@H]1[C@](C=O)(O)[C@H](C)O[C@H]1O[C@@H]1[C@@H](NC(N)=N)[C@H](O)[C@@H](NC(N)=N)[C@H](O)[C@H]1O UCSJYZPVAKXKNQ-HZYVHMACSA-N 0.000 description 2
- 239000000126 substance Substances 0.000 description 2
- 208000024891 symptom Diseases 0.000 description 2
- 238000013518 transcription Methods 0.000 description 2
- 230000035897 transcription Effects 0.000 description 2
- NHBKXEKEPDILRR-UHFFFAOYSA-N 2,3-bis(butanoylsulfanyl)propyl butanoate Chemical compound CCCC(=O)OCC(SC(=O)CCC)CSC(=O)CCC NHBKXEKEPDILRR-UHFFFAOYSA-N 0.000 description 1
- 108091003079 Bovine Serum Albumin Proteins 0.000 description 1
- 230000006820 DNA synthesis Effects 0.000 description 1
- 239000006144 Dulbecco’s modified Eagle's medium Substances 0.000 description 1
- KCXVZYZYPLLWCC-UHFFFAOYSA-N EDTA Chemical compound OC(=O)CN(CC(O)=O)CCN(CC(O)=O)CC(O)=O KCXVZYZYPLLWCC-UHFFFAOYSA-N 0.000 description 1
- 241000588724 Escherichia coli Species 0.000 description 1
- 208000018522 Gastrointestinal disease Diseases 0.000 description 1
- 208000012671 Gastrointestinal haemorrhages Diseases 0.000 description 1
- 101000611183 Homo sapiens Tumor necrosis factor Proteins 0.000 description 1
- 108010055717 JNK Mitogen-Activated Protein Kinases Proteins 0.000 description 1
- BFVQTKQTUCQRPI-YYEZTRBPSA-N LPS with O-antigen Chemical compound O([C@@H]1[C@@H](O[C@H]2[C@@H]([C@@H](O[C@H]3[C@@H]([C@@H](O[C@@H]4[C@@H]([C@@H](O)[C@@H](O)[C@@H](CO[C@@H]5[C@@H]([C@@H](O)[C@@H](O)[C@@H](CO)O5)O)O4)O)[C@@H](O)[C@@H](CO)O3)NC(C)=O)[C@@H](O)[C@@H](CO[C@H]3[C@@H]([C@@H](O)[C@H](O)[C@@H](CO)O3)NC(C)=O)O2)NC(C)=O)[C@H](O)[C@@H](CO)OC1O[C@@H]1[C@@H](O)[C@@H](O)[C@@H](CO)O[C@@H]1O[C@H]1[C@H](O)[C@@H](CO[C@@H]2[C@@H]([C@@H](O)[C@@H](O)[C@@H](CO)O2)O)OC([C@@H]1O)O[C@H]1[C@H](O)[C@@H]([C@@H](O)COC2[C@H]([C@@H](O)[C@H](OP(O)(O)=O)[C@@H]([C@@H](O)CO)O2)O)OC([C@H]1O)O[C@H]1[C@H](OP(O)(=O)OP(O)(=O)OCCN)[C@@H]([C@@H](O)CO)OC([C@H]1O)O[C@H]1[C@H](O[C@]2(O[C@@H]([C@@H](O)[C@H](O[C@]3(O[C@@H]([C@@H](O)[C@H](OP(O)(=O)OCCN)C3)[C@@H](O)CO)C(O)=O)C2)[C@@H](O)CO)C(O)=O)C[C@](O[C@@H]1[C@@H](O)CO)(OC[C@H]1O[C@@H](OC[C@@H]2[C@H]([C@H](OC(=O)C[C@H](O)CCCCCCCCCCC)[C@@H](NC(=O)C[C@H](O)CCCCCCCCCCC)[C@@H](OP(O)(O)=O)O2)O)[C@H](NC(=O)C[C@@H](CCCCCCCCCCC)OC(=O)CCCCCCCCCCC)[C@H]([C@@H]1OP(O)(O)=O)OC(=O)C[C@@H](CCCCCCCCCCC)OC(=O)CCCCCCCCCCCCC)C(O)=O)[C@@H]1O[C@H](CO)[C@@H](O)[C@H](O)[C@H]1NC(C)=O BFVQTKQTUCQRPI-YYEZTRBPSA-N 0.000 description 1
- 239000012097 Lipofectamine 2000 Substances 0.000 description 1
- 208000034486 Multi-organ failure Diseases 0.000 description 1
- 241000699666 Mus <mouse, genus> Species 0.000 description 1
- 241000699670 Mus sp. Species 0.000 description 1
- 241000283973 Oryctolagus cuniculus Species 0.000 description 1
- 229930182555 Penicillin Natural products 0.000 description 1
- JGSARLDLIJGVTE-MBNYWOFBSA-N Penicillin G Chemical compound N([C@H]1[C@H]2SC([C@@H](N2C1=O)C(O)=O)(C)C)C(=O)CC1=CC=CC=C1 JGSARLDLIJGVTE-MBNYWOFBSA-N 0.000 description 1
- 108091000080 Phosphotransferase Proteins 0.000 description 1
- 102100038277 Prostaglandin G/H synthase 1 Human genes 0.000 description 1
- 108050003243 Prostaglandin G/H synthase 1 Proteins 0.000 description 1
- 229940124158 Protease/peptidase inhibitor Drugs 0.000 description 1
- 206010037660 Pyrexia Diseases 0.000 description 1
- 108010052090 Renilla Luciferases Proteins 0.000 description 1
- 108091006619 SLC11A1 Proteins 0.000 description 1
- 238000000692 Student's t-test Methods 0.000 description 1
- 102000002689 Toll-like receptor Human genes 0.000 description 1
- 108020000411 Toll-like receptor Proteins 0.000 description 1
- 108091023040 Transcription factor Proteins 0.000 description 1
- 102000040945 Transcription factor Human genes 0.000 description 1
- 229920004890 Triton X-100 Polymers 0.000 description 1
- 239000013504 Triton X-100 Substances 0.000 description 1
- 241000700605 Viruses Species 0.000 description 1
- 210000000683 abdominal cavity Anatomy 0.000 description 1
- 230000001154 acute effect Effects 0.000 description 1
- 230000006907 apoptotic process Effects 0.000 description 1
- QVGXLLKOCUKJST-UHFFFAOYSA-N atomic oxygen Chemical compound [O] QVGXLLKOCUKJST-UHFFFAOYSA-N 0.000 description 1
- 230000001580 bacterial effect Effects 0.000 description 1
- 230000000975 bioactive effect Effects 0.000 description 1
- 230000000740 bleeding effect Effects 0.000 description 1
- 210000004204 blood vessel Anatomy 0.000 description 1
- 201000011510 cancer Diseases 0.000 description 1
- 239000002775 capsule Substances 0.000 description 1
- 229910002092 carbon dioxide Inorganic materials 0.000 description 1
- 239000001569 carbon dioxide Substances 0.000 description 1
- 230000003915 cell function Effects 0.000 description 1
- 230000005754 cellular signaling Effects 0.000 description 1
- 238000006243 chemical reaction Methods 0.000 description 1
- 239000003795 chemical substances by application Substances 0.000 description 1
- 208000037976 chronic inflammation Diseases 0.000 description 1
- 208000037893 chronic inflammatory disorder Diseases 0.000 description 1
- 230000005796 circulatory shock Effects 0.000 description 1
- 230000016396 cytokine production Effects 0.000 description 1
- 230000003413 degradative effect Effects 0.000 description 1
- 238000000326 densiometry Methods 0.000 description 1
- 238000010790 dilution Methods 0.000 description 1
- 239000012895 dilution Substances 0.000 description 1
- XPPKVPWEQAFLFU-UHFFFAOYSA-J diphosphate(4-) Chemical compound [O-]P([O-])(=O)OP([O-])([O-])=O XPPKVPWEQAFLFU-UHFFFAOYSA-J 0.000 description 1
- 235000011180 diphosphates Nutrition 0.000 description 1
- 239000002552 dosage form Substances 0.000 description 1
- 230000009977 dual effect Effects 0.000 description 1
- 210000002889 endothelial cell Anatomy 0.000 description 1
- DEFVIWRASFVYLL-UHFFFAOYSA-N ethylene glycol bis(2-aminoethyl)tetraacetic acid Chemical compound OC(=O)CN(CC(O)=O)CCOCCOCCN(CC(O)=O)CC(O)=O DEFVIWRASFVYLL-UHFFFAOYSA-N 0.000 description 1
- 239000012091 fetal bovine serum Substances 0.000 description 1
- MHMNJMPURVTYEJ-UHFFFAOYSA-N fluorescein-5-isothiocyanate Chemical compound O1C(=O)C2=CC(N=C=S)=CC=C2C21C1=CC=C(O)C=C1OC1=CC(O)=CC=C21 MHMNJMPURVTYEJ-UHFFFAOYSA-N 0.000 description 1
- 208000030304 gastrointestinal bleeding Diseases 0.000 description 1
- 239000011521 glass Substances 0.000 description 1
- 230000005283 ground state Effects 0.000 description 1
- 235000008216 herbs Nutrition 0.000 description 1
- 230000013632 homeostatic process Effects 0.000 description 1
- 102000057041 human TNF Human genes 0.000 description 1
- 230000008105 immune reaction Effects 0.000 description 1
- 238000003365 immunocytochemistry Methods 0.000 description 1
- 238000010166 immunofluorescence Methods 0.000 description 1
- 238000010820 immunofluorescence microscopy Methods 0.000 description 1
- 230000008595 infiltration Effects 0.000 description 1
- 238000001764 infiltration Methods 0.000 description 1
- 230000006749 inflammatory damage Effects 0.000 description 1
- 239000007924 injection Substances 0.000 description 1
- 238000002347 injection Methods 0.000 description 1
- 208000014674 injury Diseases 0.000 description 1
- 239000000543 intermediate Substances 0.000 description 1
- 230000003834 intracellular effect Effects 0.000 description 1
- 238000001990 intravenous administration Methods 0.000 description 1
- 230000002147 killing effect Effects 0.000 description 1
- 150000002632 lipids Chemical class 0.000 description 1
- 229920006008 lipopolysaccharide Polymers 0.000 description 1
- 210000004698 lymphocyte Anatomy 0.000 description 1
- 239000006166 lysate Substances 0.000 description 1
- 239000012139 lysis buffer Substances 0.000 description 1
- 210000004962 mammalian cell Anatomy 0.000 description 1
- 238000005259 measurement Methods 0.000 description 1
- 210000001616 monocyte Anatomy 0.000 description 1
- 210000000214 mouth Anatomy 0.000 description 1
- 210000004877 mucosa Anatomy 0.000 description 1
- 208000029744 multiple organ dysfunction syndrome Diseases 0.000 description 1
- 230000035772 mutation Effects 0.000 description 1
- 231100000957 no side effect Toxicity 0.000 description 1
- 230000017111 nuclear migration Effects 0.000 description 1
- 230000004768 organ dysfunction Effects 0.000 description 1
- 230000002018 overexpression Effects 0.000 description 1
- 238000012261 overproduction Methods 0.000 description 1
- 230000036542 oxidative stress Effects 0.000 description 1
- 229910052760 oxygen Inorganic materials 0.000 description 1
- 239000001301 oxygen Substances 0.000 description 1
- 229940049954 penicillin Drugs 0.000 description 1
- 239000000137 peptide hydrolase inhibitor Substances 0.000 description 1
- 230000035699 permeability Effects 0.000 description 1
- 239000008177 pharmaceutical agent Substances 0.000 description 1
- 239000000825 pharmaceutical preparation Substances 0.000 description 1
- 230000000144 pharmacologic effect Effects 0.000 description 1
- 102000020233 phosphotransferase Human genes 0.000 description 1
- 230000036470 plasma concentration Effects 0.000 description 1
- 239000013612 plasmid Substances 0.000 description 1
- 229920002981 polyvinylidene fluoride Polymers 0.000 description 1
- 230000035755 proliferation Effects 0.000 description 1
- 102000004169 proteins and genes Human genes 0.000 description 1
- 230000011514 reflex Effects 0.000 description 1
- 230000001105 regulatory effect Effects 0.000 description 1
- 230000001624 sedative effect Effects 0.000 description 1
- 125000003607 serino group Chemical group [H]N([H])[C@]([H])(C(=O)[*])C(O[H])([H])[H] 0.000 description 1
- 235000020183 skimmed milk Nutrition 0.000 description 1
- 238000002415 sodium dodecyl sulfate polyacrylamide gel electrophoresis Methods 0.000 description 1
- 239000000243 solution Substances 0.000 description 1
- 239000002904 solvent Substances 0.000 description 1
- 238000007619 statistical method Methods 0.000 description 1
- 229960005322 streptomycin Drugs 0.000 description 1
- 238000007920 subcutaneous administration Methods 0.000 description 1
- 239000006228 supernatant Substances 0.000 description 1
- 230000008961 swelling Effects 0.000 description 1
- 230000005062 synaptic transmission Effects 0.000 description 1
- 239000006188 syrup Substances 0.000 description 1
- 235000020357 syrup Nutrition 0.000 description 1
- 230000009897 systematic effect Effects 0.000 description 1
- 239000003826 tablet Substances 0.000 description 1
- 230000001225 therapeutic effect Effects 0.000 description 1
- 230000002103 transcriptional effect Effects 0.000 description 1
- 238000001890 transfection Methods 0.000 description 1
- 230000001052 transient effect Effects 0.000 description 1
- 230000008733 trauma Effects 0.000 description 1
- GPRLSGONYQIRFK-MNYXATJNSA-N triton Chemical compound [3H+] GPRLSGONYQIRFK-MNYXATJNSA-N 0.000 description 1
- 230000034512 ubiquitination Effects 0.000 description 1
- 238000010798 ubiquitination Methods 0.000 description 1
- XLYOFNOQVPJJNP-UHFFFAOYSA-N water Substances O XLYOFNOQVPJJNP-UHFFFAOYSA-N 0.000 description 1
Images
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
- A61K31/33—Heterocyclic compounds
- A61K31/395—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
- A61K31/55—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole
- A61K31/551—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole having two nitrogen atoms, e.g. dilazep
- A61K31/5513—1,4-Benzodiazepines, e.g. diazepam or clozapine
- A61K31/5517—1,4-Benzodiazepines, e.g. diazepam or clozapine condensed with five-membered rings having nitrogen as a ring hetero atom, e.g. imidazobenzodiazepines, triazolam
Landscapes
- Health & Medical Sciences (AREA)
- Chemical & Material Sciences (AREA)
- Medicinal Chemistry (AREA)
- Pharmacology & Pharmacy (AREA)
- Epidemiology (AREA)
- Life Sciences & Earth Sciences (AREA)
- Animal Behavior & Ethology (AREA)
- General Health & Medical Sciences (AREA)
- Public Health (AREA)
- Veterinary Medicine (AREA)
- Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
Abstract
본 발명은 유효량의 미다졸람(midazolam) 및 약학적으로 허용 가능한 담체, 희석제 또는 부형제를 함유하며 염증유발의 억제를 통한 염증억제 효과를 갖는 염증 예방 및 치료용 약제학적 조성물에 관한 것이다.The present invention relates to a pharmaceutical composition for preventing and treating inflammation containing an effective amount of midazolam and a pharmaceutically acceptable carrier, diluent or excipient and having an anti-inflammatory effect through inhibition of inflammation.
본 발명에 의한 조성물은 iNOS 또는 COX-2 유전자의 발현억제, NF-kB의 활성억제, NF-kB p65의 세포핵으로의 이동억제, MAPK p38의 인산화 억제 또는 활성산소의 생성억제 등을 통해 염증억제 효과를 나타낸다.The composition according to the present invention inhibits inflammation by inhibiting iNOS or COX-2 gene expression, inhibiting NF-kB activity, inhibiting NF-kB p65 migration into the cell nucleus, inhibiting phosphorylation of MAPK p38 or inhibiting the production of free radicals. Effect.
Description
도 1은 미다졸람이 대식세포의 전염증 매개물질 생성을 억제함을 보여주는 블럿 사진 및 그래프.1 is a blot photograph and graph showing that midazolam inhibits proinflammatory mediator production of macrophages.
도 2는 미다졸람이 LPS-유도성 NF-kB 활성을 억제함을 보여주는 블럿 사진, 그래프 및 면역형광 결과 사진.2 is a blot picture, graph and immunofluorescence results showing that midazolam inhibits LPS-induced NF-kB activity.
도 3은 미다졸람이 LPS 유도성 p38MAPK 활성을 저해함을 보여주는 블럿 사진.3 is a blot photograph showing that midazolam inhibits LPS-induced p38MAPK activity.
도 4는 미다졸람이 LPS에 의해 유도되는 활성산소의 생성을 억제함을 보여주는 그래프. 4 is a graph showing that midazolam inhibits the production of free radicals induced by LPS.
본 발명은 미다졸람을 유효성분으로 하는 염증성 질환을 예방 및 억제, 치료하는 약제학적 조성물에 관한 것이다.The present invention relates to a pharmaceutical composition for preventing, inhibiting and treating inflammatory diseases comprising midazolam as an active ingredient.
염증 질환은 감염, 외상, 면역학적 반응을 포함한 인체 내의 반응이다. 발열, 홍조, 부종, 통증 등의 급성 염증 증후가 있으며, 염증과정의 후반에는 염증부위로의 백혈구의 이동 및 사이토카인(cytokine), 분해효소(degradative enzyme), 생활성 지질 중간체(bioactive lipid intermediate), 일과성 반응성 산소종(transient reactive oxygen species), 림프구(sensitized lmphocyte)의 생성 등 세포내 변화가 일어난다. 만성적인 염증 질환에서는 백혈구의 침윤에 의해 세포활성 및 세포사멸이 일어난다. 염증반응의 화학매개물 중 prostaglandins(PG)과 nitric oxide(NO)는 발암 및 염증의 진행과정에 중요한 매개물질로 알려져 있다.Inflammatory diseases are reactions in the human body, including infections, trauma, and immunological reactions. Acute inflammatory symptoms such as fever, redness, swelling, and pain are present. Later in the inflammatory process, white blood cells move to the inflammatory site and cytokines, degradative enzymes, and bioactive lipid intermediates. Intracellular changes occur, such as the generation of transient reactive oxygen species and lymphocytes (sensitized lmphocytes). In chronic inflammatory diseases, cell activity and apoptosis occur due to infiltration of white blood cells. Prostaglandins (PG) and nitric oxide (NO) are known as important mediators of carcinogenesis and inflammation.
염증이나 통증을 조절하는 약물 중 가장 흔히 사용되는 약물인 비스테로이드계 염증질환제(nonsteroidal antiinflammatory drugs) (NSAIDs)는 체내에 PG를 생성하는 COX(cyclooxygenase)를 저해함으로써 약리작용을 나타나는데 이 효소는 COX-1과 COX-2의 두 가지 이성체로 존재한다는 것이 밝혀졌다. 이중 COX-1은 정상상태에서 위장관 점막과 혈소판, 신장에서 세포 보호 작용과 조절 작용에 관여하는 PG류 생성에 관여하는 효소인데 반해, COX-2는 정상세포에서는 그 농도가 매우 낮으나 염증관련세포에서 여러 자극(cytokine, endotoxin, mitogen)에 의해 유도 발현되어 통증이나 염증에 관여하는 PG류 생성에 관여한다. 또한 nitric oxide synthase(NOS)는 혈관의 긴장도(vascular tone), 신경전달(neurotransmission), 세균 및 암세포의 사멸과 항상성 조절에 관여한다. 또한 높은 농도의 NO는 순환기 쇼크, 염증, 발암 등의 생리반응에서 나타난다. 신경성 NOS(Neuronal NOS)와 내피 성 NOS(endothelial NOS)는 정상적인 상태에서 존재하며, 반면 iNOS(NOS type2)는 LPS, 사이토카인(cytokine) 등에 의해 유도 발현된다.Nonsteroidal antiinflammatory drugs (NSAIDs), the most commonly used drugs that control inflammation or pain, are pharmacological by inhibiting cyclooxygenase (COX), which produces PG in the body. It was found to exist as two isomers, -1 and COX-2. COX-1 is an enzyme that is involved in the production of PGs, which are involved in the protection and regulation of gastrointestinal mucosa, platelets, and kidneys in the normal state, whereas COX-2 is very low in normal cells but in inflammation-related cells. It is induced by various stimuli (cytokine, endotoxin, mitogen) and is involved in the production of PGs that are involved in pain or inflammation. Nitric oxide synthase (NOS) is also involved in the regulation of blood vessel tone, neurotransmission, killing and homeostasis of bacteria and cancer cells. High levels of NO also occur in physiological reactions such as circulatory shock, inflammation and carcinogenesis. Neuronal NOS (Neuronal NOS) and endothelial NOS (endothelial NOS) are present in the normal state, while iNOS (NOS type2) is induced and expressed by LPS, cytokine (cytokine).
패혈증(Sepsis)은 다기관부전증(multi organ failure)의 가장 큰 원인의 하나로, 아직도 높은 사망률을 보이고 있다. 그람-음성 박테리아의 바깥 막의 구성요소인 LPS(lipopplysaccharide)는 패혈증을 유발하는 가장 중요한 요인으로 알려져 왔다. 활성화된 대식세포(macrophage)는 염증 반응에 관여하며 다양한 사이토카인(cytokine)을 생산하여 순환계에 분비하여 패혈증에서 발생하는 생리특성에 영향을 미친다. 그람-음성 박테리아의 주된 외막(outer membrane)인 LPS는 단핵구/대식세포와 혈관내피세포(endothelial cell)과 같은 다양한 포유동물 세포를 활성화시키고 패혈증에서 체계적인 변이를 일으킨다. LPS는 NF-kB와 MAPK의 활성화를 유도하는 signaling pathway를 시작하게 하는 것으로 알려져 왔다. NF-kB의 활성화는 IkB의 serine 잔기 32와 36번이 인산화와 인산화에 이은 ubiquitination, IkB의 분해, 그에 수반된 NF-kB의 분비 및 이동을 일컫는다.Sepsis is one of the leading causes of multi organ failure and still has a high mortality rate. Lipopplysaccharide (LPS), a component of the outer membrane of Gram-negative bacteria, has been known to be the most important factor causing sepsis. Activated macrophage (macrophage) is involved in the inflammatory response and produces a variety of cytokines (cytokine) secretion into the circulatory system affects the physiological characteristics that occur in sepsis. LPS, the main outer membrane of Gram-negative bacteria, activates various mammalian cells such as monocytes / macrophages and endothelial cells and causes systematic mutations in sepsis. LPS has been known to initiate signaling pathways that induce the activation of NF-kB and MAPK. NF-kB activation refers to the phosphorylation and phosphorylation of serine residues 32 and 36 of IkB followed by ubiquitination, degradation of IkB, and subsequent secretion and migration of NF-kB.
사이토카인(cytokine)이나 nitric oxide(NO)와 같은 전염증 매개물질는 다양한 염증질환에서 중추적인 역할을 한다. 패혈증에서, NO의 과잉 발현은 조직의 손상에 가장 중요한 영향을 미치는 요인의 하나로 여겨지고 있다. mitogen, cytokine이나 박테리아의 LPS와 같이 염증을 야기하는 자극에 의해 대식세포는 COX-2나 iNOS를 발현한다. 축적된 데이터에 의하면 이러한 COX-2와 iNOS는 많은 염증 과정에 포함되어 있고 다양한 암을 유발하는 것으로 알려져 있어, COS-2와 iNOS가 염증과 암유발에서 중요한 역할을 담당하는 것을 알 수 있다. 그러므로 이러한 염증 인자들을 조율할 수 있다면 LPS 유도성 염증반응을 호전시킬 수 있을 것이라 여겨진다.Proinflammatory mediators such as cytokines and nitric oxides (NO) play a central role in various inflammatory diseases. In sepsis, overexpression of NO is considered to be one of the most important factors affecting tissue damage. Inflammatory stimuli, such as mitogen, cytokine or bacterial LPS, cause macrophages to express COX-2 or iNOS. Accumulated data suggest that COX-2 and iNOS are involved in many inflammatory processes and cause various cancers, indicating that COS-2 and iNOS play an important role in inflammation and cancer induction. Therefore, if these inflammatory factors can be tuned, it is thought that the LPS-induced inflammatory response can be improved.
대식세포는 조직손상에 대한 염증반응에 주요한 구성요소 중 하나이다. 활성 산소종(ROS)의 급격한 증가는 LPS와 전염증 cytokine(예를 들면 TNF-α나 IL-1β)과 같은 염증 자극에 의해 유발되며 DNA 합성, 전사 요소 활성화, 유전자 발현 및 증식과 같은 다수의 세포 기능의 변화를 야기한다. MAPK pathway와 MAPK에 의해 매개된 전염증 매개물질의 생성은 적어도 부분적으로는 NF-kB/ROS sensitive 메카니즘에 의존적인 산화환원반응(redox)으로 알려져 왔다. Macrophages are one of the major components of the inflammatory response to tissue damage. Sudden increases in reactive oxygen species (ROS) are caused by inflammatory stimuli such as LPS and pro-inflammatory cytokine (e.g. TNF-α or IL-1β), and are associated with numerous factors such as DNA synthesis, transcription factor activation, gene expression and proliferation. Causes changes in cell function. The production of MAPK pathways and pro-inflammatory mediators mediated by MAPK has been known as redox, which depends at least in part on NF-kB / ROS sensitive mechanisms.
염증이나 통증을 조절하는 약물 중 NSAIDs는 인체 내에서 염증 반응에 관여하는 PG류 이외에 위점막 보호, 혈소판기능과 관련된 PG의 생성도 억제하여, 위장관 장해나 출혈 등의 부작용도 함께 나타나게 된다. NSAIDs, among the drugs that control inflammation and pain, inhibit the production of PG related to gastric mucosal protection and platelet function in addition to PGs involved in inflammatory reactions in the human body, and also have side effects such as gastrointestinal disorders and bleeding.
따라서 부작용이 없고 염증관련 유전자 및/또는 관련물질의 활성억제 등을 선택적으로 할 수 있는 새로운 염증예방/치료제의 개발이 요구되고 있다.Therefore, there is a need for the development of new inflammation prevention / treatment agents that have no side effects and can selectively inhibit the activity of inflammation-related genes and / or related substances.
본 발명자들은 종래 마취제로 널리 사용되며 부작용이 보고된 바 없는 벤조다이아제핀(benzodiazepine) 계열의 진정효과를 가지는 유도체인 미다졸람(midazolam)이 염증예방 및 치료효과를 가진다는 사실을 최초로 발견하여 깊이 연구한 결과 본 발명을 완성하게 되었다.The present inventors have found for the first time the fact that midazolam, a derivative that has a sedative effect of the benzodiazepine family, widely used as an anesthetic and has not been reported as a side effect for the first time, deeply studies the fact that it has an inflammatory and therapeutic effect. As a result, the present invention has been completed.
본 발명의 목적은 배양된 대식세포에서 미다졸람의 항염증 작용을 확인하는 것이다. An object of the present invention is to identify the anti-inflammatory action of midazolam in cultured macrophages.
본 발명의 또 다른 목적은, 종래 마취제로 사용되는 미다졸람의 새로운 기능을 밝혀내고 이에 기초하여 미다졸람을 유효성분으로 하는 염증성 질환을 예방 및 억제, 치료하는 약제학적 조성물을 제공하는 것이다.Still another object of the present invention is to discover a new function of midazolam, which is used as a conventional anesthetic, and to provide a pharmaceutical composition for preventing, inhibiting and treating an inflammatory disease comprising midazolam as an active ingredient.
전술한 목적을 달성하기 위한 본 발명은, 유효량의 미다졸람(midazolam) 및 약학적으로 허용 가능한 담체, 희석제 또는 부형제를 함유하며 염증유발의 억제를 통한 염증억제 효과를 갖는 염증 예방 및 치료용 약제학적 조성물에 관한 것이다.In order to achieve the above object, the present invention provides a pharmaceutical agent for preventing and treating inflammation containing an effective amount of midazolam and a pharmaceutically acceptable carrier, diluent or excipient and having an anti-inflammatory effect through the inhibition of inflammation. It relates to a composition.
본 발명에서 상기 염증유발의 억제는 ① iNOS(inducible nitric oxide synthase) 또는 COX-2(cyclooxygenase-2) 유전자의 발현억제 ② NF-kB(Nuclear factor-kB)의 활성억제 ③ NF-kB p65(Nuclear factor-kB p65)의 세포핵으로의 이동억제 ④ MAPK p38(mitogen-activated protein kinase p38)의 인산화 억제 또는 ⑤ 활성산소의 생성억제를 통해 이루어지는 것을 특징으로 한다.In the present invention, the inhibition of the inflammation-induced ① inhibition of iNOS (inducible nitric oxide synthase) or COX-2 (cyclooxygenase-2)
본 발명에 의한 약제학적 조성물에서 미다졸람의 유효함량은 0.001㎎/㎖~0.01mg/㎖인 것이 바람직하다.In the pharmaceutical composition according to the present invention, the effective content of midazolam is preferably 0.001 mg / ml to 0.01 mg / ml.
본 발명에 의한 약제학적 조성물에 사용되는 미다졸람의 작용효과를 확인하기 위해 다음과 같은 실험이 수행되었다.The following experiments were carried out to confirm the effect of the midazolam used in the pharmaceutical composition according to the present invention.
대식세포 셀라인 중 하나인 RAW264.7 cell을 이용하여, western blot analysis를 통해 미다졸람이 Mitogen-activated protein kinase의 작용과 전염증 매개물질(proinflammatory mediator)에 미치는 영향을 측정하였다. Nuclear factor-kB(NF-kB)의 활성화와 NF-kB의 p65 서브유닛의 이동을 luciferase assay와 면역세포화학(immunocytochemistry)을 이용하여 측정하였다. 활성산소(superoxide)의 생성정도는 lucigenin chemiluminescence를 이용하여 측정하였다. RAW264.7 cells, one of the macrophage cell lines, were used to measure the effects of midazolam on mitogen-activated protein kinase and proinflammatory mediators by western blot analysis. Activation of nuclear factor-kB (NF-kB) and migration of p65 subunits of NF-kB were measured using luciferase assay and immunocytochemistry. The production of free radicals was measured using lucigenin chemiluminescence.
여러 실험 결과, 미다졸람은 LPS(lipopolysaccharide)에 의해 유도되는, COX-2와 iNOS의 과잉조절을 농도 의존적(약 3~30uM)으로 억제함을 확인하였다. 또한 LPS에 의해 유도된 IkB-α의 분해와 NF-kB 전사 활성 역시 미다졸람에 의해 억제되었다. NF-kB의 p65 서브유닛의 세포핵으로의 이동도 미다졸람에 의해 저해되었다. 더불어 미다졸람은 p38 MAPK의 인산화와 LPS에 의해 유도되는 활성산소의 생성을 억제함도 확인되었다.Several experiments confirmed that midazolam inhibits overregulation of COX-2 and iNOS induced by LPS (lipopolysaccharide) in a concentration-dependent manner (about 3-30 μM). The degradation of IkB-α induced by LPS and NF-kB transcriptional activity were also inhibited by midazolam. The migration of NF-kB to the cell nucleus of the p65 subunit was also inhibited by midazolam. Midazolam also inhibited the phosphorylation of p38 MAPK and the production of free radicals induced by LPS.
본 발명에 따른 약제학적 조성물은 유효량의 미다졸람을 포함하여 염증유발 생체인자의 유전자 발현을 억제하는 등의 작용을 하므로 소염 또는 염증억제 효과를 갖는다. 이때 본 발명에 따른 약제학적 조성물은 미다졸람 및 약제학적으로 허용되는 담체, 희석제 또는 부형제와 함께 약제학적으로 허용되는 약학적 제제, 예 를 들면 주사제, 액제, 시럽제, 정제, 캡슐제, 약침액 등으로 제제화하여 염증질환 치료 및 예방용 약으로 이용될 수 있다.The pharmaceutical composition according to the present invention has an anti-inflammatory or anti-inflammatory effect because it contains an effective amount of midazolam to inhibit gene expression of inflammatory-induced biofactors. At this time, the pharmaceutical composition according to the present invention is a pharmaceutically acceptable pharmaceutical preparation, such as injections, solutions, syrups, tablets, capsules, medicinal herbs, etc. together with midazolam and pharmaceutically acceptable carriers, diluents or excipients. It can be formulated as a drug for the treatment and prevention of inflammatory diseases.
또한 본 발명에 따른 약제학적 조성물은 임상적으로 투여시 정맥, 척추강, 경구, 복강, 피하 투여 등의 방법으로 통상적인 투여방법에 따라 투여할 수 있다.In addition, the pharmaceutical composition according to the present invention may be administered according to a conventional administration method by the method of intravenous, spinal cavity, oral, abdominal cavity, subcutaneous administration, etc. when clinically administered.
또한 본 발명에 따른 약제학적 조성물의 임상적 투여용량은 환자의 연령, 증상, 투여제형 또는 약물의 종류에 따라 다양하게 조절할 수 있다. 특정한 상태에서 바람직한 투여량을 결정하는 것은 공지된 기술에 해당한다.In addition, the clinical dosage of the pharmaceutical composition according to the present invention can be variously adjusted according to the age, symptoms, dosage form or type of drug of the patient. Determining the desired dosage in a particular condition corresponds to known techniques.
이하 실시예를 통하여 본 발명을 보다 상세히 설명한다. 실시예는 본 발명을 설명하기 위해 예시적으로 열거한 것일 뿐 이에 의해 본 발명의 기술적 사상의 범위나 내용이 변경되거나 축소되는 것은 아니다. 실시예에서는 미다졸람의 기작과 효과에 대해서만 검토·실험하였으나 실시예에 의해 항염증 작용이 있음이 확인된 미다졸람을 절절한 농도로 조성물화하는 것과, 만들어진 조성물을 다양한 방식으로 투여할 수 있음은 당업자에게 있어 용이한 일일 것이다.Hereinafter, the present invention will be described in more detail with reference to the following examples. The embodiments are only listed by way of example to illustrate the present invention, thereby not changing or reducing the scope or content of the technical spirit of the present invention. In the Examples, only the mechanism and effect of midazolam were examined and tested, but the composition of the midazolam, which was confirmed to have anti-inflammatory action by the Example, at an appropriate concentration, and the resulting composition can be administered in various ways. It would be easy for me.
실시예에 사용된 세포주 및 물질과 실험방법 및 분석방법은 다음과 같았다.The cell lines and materials used in the examples and the experimental and analytical methods were as follows.
(1) 사용된 세포주 및 물질(1) Cell lines and materials used
본 실시예에서는 쥐의 대식 세포주 RAW264.7이 사용되었다. RAW264.7 세포 주는 Abelson Ieukemia 바이러스가 형질도입된 BALB/c mice(Nramp 1D169 / D169)로부터 분리한 불사화(immortalized) 대식 세포주이다. RAW264.7과 human embryonic kidney 293(HEK293) 세포주는 American Type Culture Collection(Rockvile, MD)에서 분주받았다. 이들 세포주는 5%의 이산화탄소와 95%의 공기가 혼합된 37℃ 수분 공급형 인큐베이터 환경에서 유지·배양되었다. 배양액으로는 10% Fetal Bovine Serum, penicillin 100U/ml, streptomycin 100㎍/ml을 보충한 Dulbecco's Modified Eagle's Medium(Gibco, Grand Island, NY)을 사용하였다. In this example, a rat macrophage line RAW264.7 was used. RAW264.7 cell line is an immortalized macrophage cell line isolated from BALB / c mice transfected with Abelson Ieukemia virus (Nramp 1 D169 / D169 ). RAW264.7 and human embryonic kidney 293 (HEK293) cell lines were divided from the American Type Culture Collection (Rockvile, MD). These cell lines were maintained and cultured in a 37 ° C. water incubator environment with 5% carbon dioxide and 95% air. Dulbecco's Modified Eagle's Medium (Gibco, Grand Island, NY) supplemented with 10% Fetal Bovine Serum, penicillin 100U / ml, and streptomycin 100µg / ml were used.
예비실험에서, 0.9% NaCl을 이용하여 미다졸람을 희석하였을 때, 미다졸람의 용해도에 아무런 영향이 없었다. 게다가 0.9% NaCl에 의한 세포의 처리는 LPS 유도성 COX-2의 발현에 아무런 영향력을 보이지 않았다(데이터 미도시). 이러한 이유로 미다졸람(Roche)은 0.9% NaCl 수용액에 녹여서 사용하였으며, 용매 대조군으로는 0.9% NaCl 수용액을 선택하였다.In preliminary experiments, dilution of midazolam with 0.9% NaCl had no effect on the solubility of midazolam. In addition, treatment of cells with 0.9% NaCl showed no effect on the expression of LPS-inducible COX-2 (data not shown). For this reason, midazolam (Roche) was used by dissolving in 0.9% NaCl aqueous solution, and 0.9% NaCl aqueous solution was selected as a solvent control.
대식세포의 활성화를 유도하기 위해 박테리아의 내독소(endotoxin) LPS (300ng/ml)를 사용하였다. 기타 E. coli serotype O26:B6의 LPS, 인간 TNF-α, β-actin, NADPH의 환원형(Sigma), Horseradish peroxidase-labeled anti-rabbit과 anti-mouse antibody(Amersham, UK), 웨스턴 블럿에 사용된 항체 anti-iNOS, IkB-α(Santa Cruz), anti-COX-2(BD Biosciences), anti-phospho-p38 및 anti-extracellular signal-regulated kinase(Cell Signaling) 등을 사용하였다. Bacterial endotoxin LPS (300 ng / ml) was used to induce macrophage activation. Used for LPS of other E. coli serotype O26: B6, human TNF-α, β-actin, reduced form of NADPH (Sigma), Horseradish peroxidase-labeled anti-rabbit and anti-mouse antibody (Amersham, UK), Western blot Antibodies such as anti-iNOS, IkB-α (Santa Cruz), anti-COX-2 (BD Biosciences), anti-phospho-p38 and anti-extracellular signal-regulated kinase (Cell Signaling) were used.
(2) 실험방법 및 분석방법(2) Experimental method and analysis method
① Western blot analysis① Western blot analysis
웨스턴 블럿 분석을 위하여, RAW264.7 cell은 20mM Tris-Cl, pH 7.5 100mM NaCl, 2mM EDTA, 2mM EGTA, 1mM Na3VO3, 1mM β-glycerophosphate, 4mM Na pyrophosphate, 5mM NaF, 1% Triton X-100 및 protease inhibitor cocktail을 포함하는 100㎕의 lysis 완충용액에서 harvest되었다. lysate는 12,000rpm에서 20분간 원심 분리하여 상층액을 분리하였다. 40㎍의 단백질(9% iNOS, 10% COX-2, 12.5% IkB-α, phospho-p38 및 phosphoextracellular signal-regulatedkinase)을 SDS-PAGE를 이용하여 분리하고 polyvinylidene difluoride membrane에서 전기영동하였다. 5% skim milk로 상온에서 2시간 동안 블록킹한 후 블럿은 특이적인 primary antibody와 함께 1:1000의 비율로 4℃에서 overnight incubation 하고, horseradish peroxidase-conjugated secondary antibody로 검출하였다. 블럿을 위해 ECL kit(Pierce)를 이용하였다.For Western blot analysis, RAW264.7 cells were treated with 20 mM Tris-Cl, pH 7.5 100 mM NaCl, 2 mM EDTA, 2 mM EGTA, 1 mM Na 3 VO 3 , 1 mM β-glycerophosphate, 4 mM Na pyrophosphate, 5 mM NaF, 1% Triton X- 100 μl of lysis buffer containing protease inhibitor cocktail was harvested. Lysate was centrifuged at 12,000 rpm for 20 minutes to separate the supernatant. 40 μg of proteins (9% iNOS, 10% COX-2, 12.5% IkB-α, phospho-p38 and phosphoextracellular signal-regulatedkinase) were isolated using SDS-PAGE and electrophoresed on polyvinylidene difluoride membrane. After blocking for 2 hours at room temperature with 5% skim milk, the blot was incubated overnight at 4 ° C at a ratio of 1: 1000 with specific primary antibodies, and detected with horseradish peroxidase-conjugated secondary antibody. ECL kit (Pierce) was used for the blot.
② Transfection과 Reporter assay② Transfection and Reporter assay
NF-kB의 활성화에 대한 미다졸람의 영향을 관찰하기 위해, NF-kB가 감염된 HEK293 세포를 사용하여 reporter assay를 실행하였다. LPS의 toll-like receptor는 HEK293 cell에서는 발현되지 않고, TNF-α가 대식세포에서 LPS에 의해 생성되므로 LPS대신 NF-kB 활성화를 자극하기 위하여 TNF-α를 사용하였다. 감염을 위해서 6 well plate에서 5X104 cells을 Lipofectamine 2000(Invitrogen)을 사용하여 1㎍ NF-kB luciferase 혹은 Renilla luciferase로 감염시켰다. cotransfection 실험을 위해서 동일 몰량의 각 plasmid를 1㎍/well로 감염시켰다. 감염 1일 후 세포들을 미다졸람이 처리된 환경과 그렇지 않은 환경 하에서 TNF-α (15ng/ml)처리로 자극하여 luciferase 생성이 측정가능해지는 8시간 동안 배양하였다. 세포들을 수확하고 제조자(Promega)의 매뉴얼에 따라 dual luciferase assay를 하였다. Luciferase activity는 Luminoskan Ascent(Thermo Labsystems)를 이용 측정하였다.To observe the effect of midazolam on the activation of NF-kB, a reporter assay was performed using HEK293 cells infected with NF-kB. Toll-like receptors of LPS were not expressed in HEK293 cells, and since TNF-α is produced by LPS in macrophages, TNF-α was used to stimulate NF-kB activation instead of LPS. For infection, 5 × 10 4 cells were infected with 1 μg NF-kB luciferase or Renilla luciferase using Lipofectamine 2000 (Invitrogen) in 6 well plates. For cotransfection experiments, the same molar amount of each plasmid was infected at 1 µg / well. After 1 day of infection, cells were cultured for 8 hours in a midazolam-treated and non-moldazol-treated environment with TNF-α (15ng / ml) treatment to allow luciferase production to be measured. Cells were harvested and subjected to dual luciferase assay according to the manufacturer's manual. Luciferase activity was measured using Luminoskan Ascent (Thermo Labsystems).
③ Immunofluorescent staining③ Immunofluorescent staining
LPS에 의해 유도되는 NF-kB p65의 이동에 미치는 미다졸람의 영향을 RAW264.7 세포의 면역형광 염색 (immunofluorescent staining)에 의해 측정하였다. NF-kB의 이동은 LPS 처리 후 30분 이내에 일어나기 때문에, RAW264.7 세포를 미다졸람의 존재 또는 부재 하에 30분간 LPS(300 ng/ml)로 처리하였다. 면역형광 염색을 위하여, RAW265.7 세포를 glass coverslip 위에 성장시켰다. 미다졸람 또는 LPS에 노출시킨 후, 1% glutaldehyde로 세포를 고정하고 0.25% triton X-100으로 세포투과능을 향상시킨 후 2% bovin serum albumin으로 1시간 동안 blocking하였다. Coverslip을 1% bovin serum albumin에서 rabbit anti-p65(1:100) primary antibody를 사용하여 4℃에서 overnight하여 배양하였다. 세포를 세척하고, fluorescein isothiocyanate-labeled secondary antibody에서 1시간 동안 배양하였 다. 세포를 다시 세척하고, 5㎍/㎖ propidium iodide로 핵을 볼 수 있도록 10분간 염색한 후 세척하였다. Zeiss confocal microscope (Oberkochen)으로 coverslip의 형광을 관측하였다.The effect of midazolam on the migration of NF-kB p65 induced by LPS was measured by immunofluorescent staining of RAW264.7 cells. Since NF-kB migration occurs within 30 minutes after LPS treatment, RAW264.7 cells were treated with LPS (300 ng / ml) for 30 minutes in the presence or absence of midazolam. For immunofluorescence staining, RAW265.7 cells were grown on glass coverslip. After exposure to midazolam or LPS, cells were fixed with 1% glutaldehyde, cell permeability was enhanced with 0.25% triton X-100, and blocked with 2% bovin serum albumin for 1 hour. Coverslip was incubated overnight at 4 ° C using rabbit anti-p65 (1: 100) primary antibody in 1% bovin serum albumin. The cells were washed and incubated for 1 hour in fluorescein isothiocyanate-labeled secondary antibody. The cells were washed again and stained for 10 minutes to see the nuclei with 5 μg / ml propidium iodide and washed. Fluorescence of the coverslip was observed with a Zeiss confocal microscope (Oberkochen).
④ 활성산소 측정④ Free radical measurement
LPS에 의해 생성되는 활성산소(O2-)에 미다졸람이 어떤 영향을 미치는가를 알아보기 위해 Raw264.7 세포를 18시간 동안 미다졸람이 있는 환경과 없는 환경에서 LPS에 노출시키고 Lucigenin-enhanced chemiluminescence assay를 시행하였다. Lucigenin-enhanced chemiluminescene assay는 이전에 보고된 것처럼(17~19) 활성산소 생산 정도를 분석하기 위해 수행되었다. Lucigenin(bis-N-methylacridinium nitrate)는 활성산소의 존재하에 특히 발광한다. 간단히, RAW264.7(1X105세포)를 Krebs-HEPES buffer(100mM NaCl, 4.7mM KCl, 1.97mM CaCl2, 1.2mM MgSO4, 1.03mM K2HPO4, 25mM NaHCO3, 20mM Na-HEPES, pH 7.4)를 함유하는 scintillation vial에 옮겼으며, 이때 최종농도 5μM이 되도록 lucigenin을 첨가하였다. chemiluminescence는 100μM의 NADPH를 첨가한 후 2분간 기록하였으며, 발광량에서 blank의 발광량을 감하여 활성산소가 생성되었는지를 측정하는데 이용하였다. 수치는 1X105 세포 당 상대적인 빛의 양으로 나타내었다.To determine the effect of midazolam on the free radicals produced by LPS (O 2- ), Raw264.7 cells were exposed to LPS for 18 hours in and without midazolam and Lucigenin-enhanced chemiluminescence assay. Was implemented. Lucigenin-enhanced chemiluminescene assays were performed to analyze the extent of free radical production, as reported previously (17–19). Lucigenin (bis-N-methylacridinium nitrate) specifically emits light in the presence of free radicals. Briefly, RAW264.7 (1 × 10 5 cells) was added to Krebs-HEPES buffer (100 mM NaCl, 4.7 mM KCl, 1.97 mM CaCl 2 , 1.2 mM MgSO 4 , 1.03 mM K 2 HPO 4 , 25 mM NaHCO 3 , 20 mM Na-HEPES, pH 7.4) was transferred to a scintillation vial containing lucigenin to a final concentration of 5 μM. Chemiluminescence was recorded for 2 minutes after the addition of 100μM NADPH. The chemiluminescence was used to determine whether free radicals were generated by subtracting the amount of light emitted from the blank. The figures are 1x10 5 It is expressed as the relative amount of light per cell.
⑤ 통계 분석⑤ Statistical analysis
결과는 평균±표준편차로 나타내었다. Student t test를 이용하여 대조군과 drug 처리군 간의 통계적 유의성을 검증하였다. P value<0.05 것을 유의하다고 판정하였다.The results are expressed as mean ± standard deviation. Student t test was used to verify statistical significance between the control and drug treatment groups. It was determined that P value <0.05 was significant.
실시예 1 : LPS에 의해 유도되는 COX-2 및 iNOS의 발현 억제효과 실험Example 1 Experiment Inhibitory Effect of COX-2 and iNOS Induced by LPS
대식세포에서 전염증 매개물질의 발현에 미다졸람이 어떤 영향을 미치는지 고찰하였다. The effect of midazolam on the expression of pro-inflammatory mediators in macrophages was investigated.
대식세포에 미다졸람(3~30μM)을 전처리하고, 300ng/㎖의 LPS에 18시간 동안 노출시켰다. iNOS와 COX-2의 발현 정도는 Western blot analysis를 통하여 측정하였다(도 1). 도에서 (A) 미다졸람은 RAW264.7 세포에서 LPS에 의해 유도되는 iNOS와 COX-2 단백질의 발현을 보여주는 웨스턴 블럿 사진(이때 대조구로 β-Actin을 사용)이며, (B)는 densitometry 데이터로서 세 개의 각각의 실험에 대해서 평균±SEM을 나타낸다. * 및 #는 p<0.05인 유의한 데이터를 의미한다.Macrophages were pretreated with midazolam (3-30 μM) and exposed to 300 ng / ml LPS for 18 hours. Expression levels of iNOS and COX-2 were measured by Western blot analysis (FIG. 1). In the figure, (A) midazolam is a Western blot photograph showing the expression of iNOS and COX-2 proteins induced by LPS in RAW264.7 cells (in which β-Actin is used as a control), and (B) is densitometry data. Mean ± SEM is shown for each of the three experiments. * And # mean significant data with p <0.05.
도 1에 나타난 것과 같이 LPS는 iNOS와 COX-2의 발현을 유도하였으며, LPS의 자극을 받지 않은 세포에서는 iNOS와 COX-2가 감지되지 않았다. 그러나 미다졸람을 전처리한 경우 LPS의 자극을 받은 세포라도 농도 의존적(약 3~30μM)으로 COX-2와 iNOS의 발현이 억제되었다. 특히 고농도(30μM)의 미다졸람은 RAW264.7 세포에서 LPS에 의해 유도되는 iNOS와 COX-2발현을 완벽하게 억제함을 알 수 있다.As shown in FIG. 1, LPS induced expression of iNOS and COX-2, and iNOS and COX-2 were not detected in cells not stimulated by LPS. However, pretreatment with midazolam inhibited the expression of COX-2 and iNOS even in LPS-stimulated cells in a concentration-dependent manner (about 3 to 30 μM). In particular, high concentrations (30 μM) of midazolam completely inhibited iNOS and COX-2 expression induced by LPS in RAW264.7 cells.
정리하자면, 3~30μM 범위에서 미다졸람은 전염증 매개물질로 알려진 iNOS와 COX-2의 발현을 억제하였다. 임상에서 쓰이는 benzodiazepine계열의 혈장농도는 0.1~50μM 이다. 본 실시예에 의하면 임상에서 투여된 미다졸람이 대식세포에서 전염증 매개물질의 억제를 통하여 항염증작용을 나타낼 수 있음을 시사한다. 말초형태의 benzodiazepine 결합부위가 쥐의 대식세포에 존재한다. 따라서 본 실시예는 미다졸람이 말초형태의 benzodiazepine 결합부위를 통하여 대식세포에서 항염증 효과를 나타내는 것을 보여준다. In summary, midazolam inhibited the expression of iNOS and COX-2, known as pro-inflammatory mediators, in the range of 3-30 μM. The plasma concentration of the benzodiazepine family used in clinical practice is 0.1 ~ 50μM. This example suggests that midazolam administered clinically can exhibit anti-inflammatory activity through inhibition of pro-inflammatory mediators in macrophages. Peripheral benzodiazepine binding sites are present in mouse macrophages. Therefore, this example shows that midazolam has an anti-inflammatory effect in macrophages through peripheral benzodiazepine binding sites.
실시예 2 : NF-kB 활성 억제효과 실험Example 2 NF-kB activity inhibitory effect experiment
(1) 미다졸람이 IkB의 분해를 직접적으로 저해하는지 여부를 확인하기 위하여 대식세포 RAW264.7를 LPS에 30분간 노출시킨 후 IkB-α 단백질의 양의 변화를 측정하였다(도 2의 A). 도 2의 A는 RAW264.7세포에서 IkB-α의 분해에 대한 미다졸람의 효과를 보여주는 웨스턴 블럿 사진(이때 대조구로 β-Actin을 사용)이다. (1) In order to confirm whether midazolam directly inhibits the degradation of IkB, the macrophage RAW264.7 was exposed to LPS for 30 minutes, and then the change in the amount of IkB-α protein was measured (A of FIG. 2). FIG. 2A is a Western blot photograph showing the effect of midazolam on the degradation of IkB-α in RAW264.7 cells, wherein β-Actin was used as a control.
도 2의 A에서 볼 수 있듯이, LPS는 IkB-α의 분해를 야기하였다. 그러나 미다졸람을 전처리한 경우 IkB-α의 분해를 유의적으로 억제하였다. 따라서 미다졸람이 LPS에 의해 유도되는 IkB-α의 분해를 차단하여 NF-kB 활성을 저해할 수 있음을 유추할 수 있다.As can be seen in A of FIG. 2, LPS caused degradation of IkB-α. However, pretreatment with midazolam significantly inhibited the degradation of IkB-α. Therefore, it can be inferred that midazolam can inhibit NF-kB activity by blocking the degradation of IkB-α induced by LPS.
(2) NF-kB는 COX-2와 iNOS의 프로모터 유전자의 cis-elements에 결합하여 이들의 전사를 조절하는 것으로 잘 알려져 있다. 이를 확인하기 위하여 NF-kB luciferase 활성에 대한 미다졸람의 영향을 확인하였다(도 2의 B). 도 2의 B에서 luciferase의 활성을 fold change %로 나타내었다. 효소활성값은 4번의 분리된 실험에 대한 평균±표준편차로 나타내었다. (2) NF-kB is well known to bind to cis- elements of promoter genes of COX-2 and iNOS and regulate their transcription. In order to confirm this, the effect of midazolam on NF-kB luciferase activity was confirmed (B of FIG. 2). In Figure 2 B, luciferase activity is expressed as fold change%. Enzyme values were expressed as mean ± standard deviation for 4 separate experiments.
도 2의 B에서 볼 수 있듯이 미다졸람은 TNF-α에 의해 유도된 luciferase 활성의 증가를 완벽하게 억제하였다. 본 실시예에 의하면 미다졸람은 LPS-유도성 IkB 분해를 저해하고(하거나) promoter elements에 binding 하는 능력을 억제하는 것에 의해 NF-kB 활성을 저해하는 것으로 판단된다. As shown in B of FIG. 2, midazolam completely inhibited the increase of luciferase activity induced by TNF-α. According to this example, midazolam is believed to inhibit NF-kB activity by inhibiting LPS-induced IkB degradation and / or inhibiting its ability to bind promoter elements.
(3) LPS에 노출된 후 NF-kB에 의해 매개되는 전사를 위해서는 NF-kB p50/p65 heterodimer가 핵으로 이동되어야 한다. NF-kB 이동에 대한 미다졸람의 효과를 관찰하기 위하여, LPS로 자극된 RAW264.7 대식세포에서 p65 핵 이동에 대한 미다졸람의 효과를 알아보았다. (3) NF-kB p50 / p65 heterodimers must be transferred to the nucleus for NF-kB mediated transcription after exposure to LPS. To observe the effect of midazolam on NF-kB migration, we examined the effect of midazolam on p65 nuclear migration in LPS-stimulated RAW264.7 macrophages.
먼저 RAW264.7 세포를 미다졸람의 존재(30μM) 또는 부재하에 LPS (300ng/㎖)로 30분간 처리하였다. 처리된 세포를 고정하고 NF-kB p65 항체(green)와 propidium iodide(PI)로 핵을 염색한 후 면역형광 현미경으로 관측하였다(도 2의 C). 도 2의 C에 의하면 LPS에 의한 RAW 세포의 처리는 확연하게 NF-kB p65 subunit의 핵으로의 이동을 유도하며, 미다졸람은 이러한 이동을 차단함을 알 수 있다.RAW264.7 cells were first treated with LPS (300 ng / ml) for 30 minutes in the presence or absence of midazolam (30 μM). The treated cells were fixed and stained with nuclei with NF-kB p65 antibody (green) and propidium iodide (PI) and observed by immunofluorescence microscopy (FIG. 2C). According to FIG. 2C, the treatment of RAW cells by LPS obviously induces migration of the NF-kB p65 subunit to the nucleus, and midazolam blocks this migration.
실시예 3 : p38 MAPK의 인산화 저해효과 실험Example 3 Inhibition of Phosphorylation of p38 MAPK
미다졸람이 LPS에 의해 유도되는 MAPK(mitogen-activated protein kinase)의 활성을 저해하는지 여부를 확인하는 실험을 수행하였다.Experiments were conducted to determine whether midazolam inhibits the activity of MAPK (mitogen-activated protein kinase) induced by LPS.
이를 위해 미다졸람을 처리한 대식세포에서 p38 MAPK의 인산화 정도를 관찰하였다(도 3). 도 3은 RAW264.7세포에서 MAPK 및 ERK의 활성화에 대한 미다졸람의 효과를 보여주는 웨스턴 블럿 사진(이때 대조구로 β-Actin을 사용)이다. To this end, the degree of phosphorylation of p38 MAPK was observed in macrophages treated with midazolam (FIG. 3). FIG. 3 is a Western blot photograph showing the effect of midazolam on the activation of MAPK and ERK in RAW264.7 cells using β-Actin as a control.
LPS 처리 30분 경과시에 p38 MAPK의 인산화가 유도되었다가 50분 내에 기저 수준으로 회복되었다. 미다졸람의 처리는 LPS에 의해 유도된 p38 활성을 완벽히 억제하였다(도 3의 A). 반면 미다졸람은 LPS에 의해 유도된 extracellular signal-regulated kinase(ERK)의 활성화는 억제하지 않았다(도 3의 B).Phosphorylation of p38 MAPK was induced 30 minutes after LPS treatment and then returned to baseline within 50 minutes. Treatment of midazolam completely inhibited p38 activity induced by LPS (FIG. 3A). Midazolam, on the other hand, did not inhibit the activation of extracellular signal-regulated kinase (ERK) induced by LPS (FIG. 3B).
p38MAPK pathway는 단핵구-대식세포 계통(lineage)의 세포에서 cytokine 생성에 포함되어 있다고 알려져 있다. 본 실시예에서 미다졸람은 LPS에 의해 유도된 p38 활성화를 저해하였다. c-Jun N-terminal kinase, extracellular signal-regulated kinase, p38과 같은 MAPK는 LPS에 의해 매개되는 COX-2와 iNOS의 발현에 매우 중요한 역할을 한다고 알려져 있다. 특히 p38MAPK의 저해는 iNOS와 COX-2의 프로모터 활성의 저해를 통해 전염증 cytokine의 생성을 방지하는 것으로 알려져 있다. 따라서 미다졸람에 의한 p38의 저해는 iNOS와 COX-2의 발현을 감소시켜 궁극적으로 항염증 효과를 나타냄을 알 수 있다.The p38MAPK pathway is known to be involved in cytokine production in cells of the monocyte-macrophage lineage. In this example, midazolam inhibited p38 activation induced by LPS. MAPKs such as c-Jun N-terminal kinase, extracellular signal-regulated kinase, and p38 are known to play an important role in the expression of COX-2 and iNOS mediated by LPS. In particular, inhibition of p38MAPK is known to prevent the production of pro-inflammatory cytokine through inhibition of promoter activity of iNOS and COX-2. Thus, inhibition of p38 by midazolam reduced the expression of iNOS and COX-2 and ultimately showed anti-inflammatory effect.
실시예 4 :활성산소종(Reactive Oxygen species) 생성 억제효과 실험Example 4 Experiment to Inhibit the Production of Reactive Oxygen Species
미다졸람이 활성산소(superoxide)의 생성을 저해하는지 연구하기 위하여, NADPH에 의한 활성산소의 생성을 lucigenin-enhanced chemiluminescence 방법에 따 라 측정하였다(도 4). In order to study whether midazolam inhibits the production of free radicals, the production of free radicals by NADPH was measured according to the lucigenin-enhanced chemiluminescence method (FIG. 4).
LPS는 RAW 264.7 세포에서 NADPH에 의해 활성산소의 생성을 야기하였다. 미다졸람 처리시 기본적인 NADPH에 의한 활성산소의 생성은 억제되지 않았지만 LPS에 의해 유도되는 활성산소의 생성은 현저하게 억제되었다.LPS caused the production of free radicals by NADPH in RAW 264.7 cells. In the midazolam treatment, the production of free radicals by basic NADPH was not inhibited, but the production of free radicals induced by LPS was significantly suppressed.
실시예 3 및 4의 결과에 의하면 대식세포에서 p38 MAPK 활성화와 LPS에 의해 유도되는 활성 산소의 생성는 미다졸람에 의해 억제·차단됨을 알 수 있다. 따라서 p38 MAPK의 저해는 미다졸람에 의해 유도된 활성산소 생성의 저해에 기인하는 것이라 유추할 수 있다. The results of Examples 3 and 4 show that p38 MAPK activation and production of free radicals induced by LPS in macrophages are inhibited and blocked by midazolam. Therefore, it can be inferred that inhibition of p38 MAPK is due to inhibition of free radical production induced by midazolam.
미다졸람의 항염증 작용을 설명하는 가능한 메카니즘은 LPS에 의해 유도된 활성산소의 저해를 포함한다. 실시예에 의하면 기저상태의 ROS 생성은 미다졸람에 의해 유의하게 저해되지는 않았으나, LPS에 의해 증가된 활성산소는 미다졸람의 전처리에 의해 크게 저해되는데, 이는 미다졸람이 대식세포에서 LPS에 의해 유도된 신호전달(signaling pathway)을 특이적으로 저해할 수 있음을 시사한다. 대식세포의 활성화에 의한 ROS의 과량 생성은 염증과 조직손상의 발병에 중요한 역할을 한다. 따라서 미다졸람에 의한 ROS 생성의 억제는 산화 스트레스에 의한 기관의 장애(dysfunction)에 유효할 것이다.Possible mechanisms that explain the anti-inflammatory action of midazolam include inhibition of free radicals induced by LPS. According to the examples, the ROS production in the ground state was not significantly inhibited by midazolam, but the active oxygen increased by LPS was greatly inhibited by the pretreatment of midazolam, which was induced by LPS in macrophages. Suggests specific inhibition of signaling pathways. Overproduction of ROS by activation of macrophages plays an important role in the development of inflammation and tissue damage. Thus, inhibition of ROS production by midazolam would be effective for organ dysfunction due to oxidative stress.
본 발명에 의하여 염증관련 유전자 및/또는 관련물질의 활성억제 등을 선택적으로 할 수 있는 미다졸람을 함유하며, 부작용이 없는 새로운 염증 예방 및 치료 용 약제학적 조성물이 가능하게 된다.According to the present invention, a new pharmaceutical composition for preventing and treating inflammation containing midazolam, which can selectively inhibit the activity of inflammation-related genes and / or related substances, can be selected.
본 발명에 의하여 염증환자를 위한 염증 예방/치료를 위한 약제의 선택폭을 넓히는 효과를 얻을 수 있다.According to the present invention, it is possible to obtain an effect of widening the choice of drugs for preventing / treating inflammation for inflammatory patients.
Claims (7)
Priority Applications (1)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
KR1020060096894A KR100755872B1 (en) | 2006-10-02 | 2006-10-02 | Pharmaceutical composition for preventing and treating inflammation containing midazolam |
Applications Claiming Priority (1)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
KR1020060096894A KR100755872B1 (en) | 2006-10-02 | 2006-10-02 | Pharmaceutical composition for preventing and treating inflammation containing midazolam |
Publications (1)
Publication Number | Publication Date |
---|---|
KR100755872B1 true KR100755872B1 (en) | 2007-09-05 |
Family
ID=38736631
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
KR1020060096894A KR100755872B1 (en) | 2006-10-02 | 2006-10-02 | Pharmaceutical composition for preventing and treating inflammation containing midazolam |
Country Status (1)
Country | Link |
---|---|
KR (1) | KR100755872B1 (en) |
Citations (3)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
JP2000186041A (en) | 1998-12-21 | 2000-07-04 | Yamanouchi Pharmaceut Co Ltd | Pain therapeutic agent |
EP1323422A1 (en) | 2001-12-24 | 2003-07-02 | Special Products Limited | Pharmaceutical composition comprising midazolam for buccal administration |
KR20040095352A (en) * | 2002-04-08 | 2004-11-12 | 길포드 파마슈티컬스 인코포레이티드 | Pharmaceutical compositons containing water-soluble prodrugs of propofol and methods of administering same |
-
2006
- 2006-10-02 KR KR1020060096894A patent/KR100755872B1/en active IP Right Grant
Patent Citations (4)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
JP2000186041A (en) | 1998-12-21 | 2000-07-04 | Yamanouchi Pharmaceut Co Ltd | Pain therapeutic agent |
EP1323422A1 (en) | 2001-12-24 | 2003-07-02 | Special Products Limited | Pharmaceutical composition comprising midazolam for buccal administration |
US6936605B2 (en) | 2001-12-24 | 2005-08-30 | Special Products Limited | Pharmaceutical composition |
KR20040095352A (en) * | 2002-04-08 | 2004-11-12 | 길포드 파마슈티컬스 인코포레이티드 | Pharmaceutical compositons containing water-soluble prodrugs of propofol and methods of administering same |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
Zhao et al. | Baicalin alleviates bleomycin‑induced pulmonary fibrosis and fibroblast proliferation in rats via the PI3K/AKT signaling pathway | |
Li et al. | Prophylactic treatment with MSC-derived exosomes attenuates traumatic acute lung injury in rats | |
Zeng et al. | 4-PBA inhibits LPS-induced inflammation through regulating ER stress and autophagy in acute lung injury models | |
Deng et al. | Metformin protects against intestinal barrier dysfunction via AMPKα1‐dependent inhibition of JNK signalling activation | |
Shen et al. | Anti-proliferation and anti-inflammation effects of corilagin in rheumatoid arthritis by downregulating NF-κB and MAPK signaling pathways | |
Fu et al. | Dexmedetomidine attenuates lipopolysaccharide-induced acute lung injury by inhibiting oxidative stress, mitochondrial dysfunction and apoptosis in rats | |
Liu et al. | Leonurine (SCM-198) attenuates myocardial fibrotic response via inhibition of NADPH oxidase 4 | |
Zhao et al. | Effect and mechanism of evodiamine against ethanol-induced gastric ulcer in mice by suppressing Rho/NF-кB pathway | |
Qiu et al. | Asiatic acid alleviates ischemic myocardial injury in mice by modulating mitophagy-and glycophagy-based energy metabolism | |
Li et al. | Ulinastatin inhibits the inflammation of LPS-induced acute lung injury in mice via regulation of AMPK/NF-κB pathway | |
Wang et al. | Alleviation of acute lung injury in rats with sepsis by resveratrol via the phosphatidylinositol 3-kinase/nuclear factor-erythroid 2 related factor 2/heme oxygenase-1 (PI3K/Nrf2/HO-1) pathway | |
Tu et al. | Geniposide attenuates hyperglycemia‐induced oxidative stress and inflammation by activating the Nrf2 signaling pathway in experimental diabetic retinopathy | |
Luo et al. | Rutaecarpine derivative R3 attenuates atherosclerosis via inhibiting NLRP3 inflammasome‐related inflammation and modulating cholesterol transport | |
Wu et al. | Ribosylation-derived advanced glycation end products induce tau hyperphosphorylation through brain-derived neurotrophic factor reduction | |
Zhang et al. | Total flavone of Abelmoschus manihot ameliorates Crohn's disease by regulating the NF-κB and MAPK signaling pathways | |
Yang et al. | Cordycepin inhibits LPS-induced inflammatory responses by modulating NOD-Like Receptor Protein 3 inflammasome activation | |
Fu et al. | Protective effect of Cordyceps sinensis extract on lipopolysaccharide-induced acute lung injury in mice | |
Guo et al. | ACT001 suppressing M1 polarization against inflammation via NF-κB and STAT1 signaling pathways alleviates acute lung injury in mice | |
Shan et al. | Inhibition of epidermal growth factor receptor attenuates LPS-induced inflammation and acute lung injury in rats | |
Yu et al. | Guhong Injection Alleviates Cerebral Ischemia–Reperfusion Injury via the PKC/HIF-1α Pathway in Rats | |
Zhao et al. | Syringin exerts anti‐inflammatory and antioxidant effects by regulating SIRT1 signaling in rat and cell models of acute myocardial infarction | |
Guo et al. | Autophagy inhibition protects from alveolar barrier dysfunction in LPS-induced ALI mice by targeting alveolar epithelial cells | |
Jin et al. | Oxymatrine attenuates lipopolysaccharide-induced acute lung injury by activating the epithelial sodium channel and suppressing the JNK signaling pathway | |
Yang et al. | Polygonatum kingianum saponins delay cellular senescence through SASP down-regulation and prolong the healthy lifespan of Caenorhabditis elegans by activating sir-2.1/autophagy | |
Sun et al. | Daming capsule protects against myocardial infarction by promoting mitophagy via the SIRT1/AMPK signaling pathway |
Legal Events
Date | Code | Title | Description |
---|---|---|---|
A201 | Request for examination | ||
PA0109 | Patent application |
Patent event code: PA01091R01D Comment text: Patent Application Patent event date: 20061002 |
|
PA0201 | Request for examination | ||
E701 | Decision to grant or registration of patent right | ||
PE0701 | Decision of registration |
Patent event code: PE07011S01D Comment text: Decision to Grant Registration Patent event date: 20070828 |
|
GRNT | Written decision to grant | ||
PR0701 | Registration of establishment |
Comment text: Registration of Establishment Patent event date: 20070830 Patent event code: PR07011E01D |
|
PR1002 | Payment of registration fee |
Payment date: 20070831 End annual number: 3 Start annual number: 1 |
|
PG1601 | Publication of registration | ||
PR1001 | Payment of annual fee |
Payment date: 20100728 Start annual number: 4 End annual number: 4 |
|
FPAY | Annual fee payment |
Payment date: 20110722 Year of fee payment: 5 |
|
PR1001 | Payment of annual fee |
Payment date: 20110722 Start annual number: 5 End annual number: 5 |
|
FPAY | Annual fee payment |
Payment date: 20111129 Year of fee payment: 7 |
|
PR1001 | Payment of annual fee |
Payment date: 20111129 Start annual number: 6 End annual number: 7 |
|
FPAY | Annual fee payment |
Payment date: 20150807 Year of fee payment: 9 |
|
PR1001 | Payment of annual fee |
Payment date: 20150807 Start annual number: 9 End annual number: 9 |
|
FPAY | Annual fee payment |
Payment date: 20160725 Year of fee payment: 10 |
|
PR1001 | Payment of annual fee |
Payment date: 20160725 Start annual number: 10 End annual number: 10 |
|
FPAY | Annual fee payment |
Payment date: 20170728 Year of fee payment: 11 |
|
PR1001 | Payment of annual fee |
Payment date: 20170728 Start annual number: 11 End annual number: 11 |
|
FPAY | Annual fee payment |
Payment date: 20180723 Year of fee payment: 12 |
|
PR1001 | Payment of annual fee |
Payment date: 20180723 Start annual number: 12 End annual number: 12 |
|
FPAY | Annual fee payment |
Payment date: 20190730 Year of fee payment: 13 |
|
PR1001 | Payment of annual fee |
Payment date: 20190730 Start annual number: 13 End annual number: 13 |
|
PR1001 | Payment of annual fee |
Payment date: 20200803 Start annual number: 14 End annual number: 14 |
|
PR1001 | Payment of annual fee |
Payment date: 20220728 Start annual number: 16 End annual number: 16 |
|
PR1001 | Payment of annual fee |
Payment date: 20230727 Start annual number: 17 End annual number: 17 |