NL2034780B1 - Combination therapy glucocorticoids and ginsenoside - Google Patents
Combination therapy glucocorticoids and ginsenoside Download PDFInfo
- Publication number
- NL2034780B1 NL2034780B1 NL2034780A NL2034780A NL2034780B1 NL 2034780 B1 NL2034780 B1 NL 2034780B1 NL 2034780 A NL2034780 A NL 2034780A NL 2034780 A NL2034780 A NL 2034780A NL 2034780 B1 NL2034780 B1 NL 2034780B1
- Authority
- NL
- Netherlands
- Prior art keywords
- ginsenoside
- treatment
- micelle
- beclomethasone
- combination
- Prior art date
Links
- 229930182494 ginsenoside Natural products 0.000 title claims abstract description 350
- 229940089161 ginsenoside Drugs 0.000 title claims abstract description 279
- 239000003862 glucocorticoid Substances 0.000 title claims abstract description 237
- 229940037128 systemic glucocorticoids Drugs 0.000 title description 5
- 238000002648 combination therapy Methods 0.000 title description 2
- 238000011282 treatment Methods 0.000 claims abstract description 210
- 239000000693 micelle Substances 0.000 claims abstract description 107
- 239000000203 mixture Substances 0.000 claims abstract description 86
- 238000000034 method Methods 0.000 claims abstract description 66
- 238000009472 formulation Methods 0.000 claims abstract description 45
- NBMKJKDGKREAPL-DVTGEIKXSA-N beclomethasone Chemical compound C1CC2=CC(=O)C=C[C@]2(C)[C@]2(Cl)[C@@H]1[C@@H]1C[C@H](C)[C@@](C(=O)CO)(O)[C@@]1(C)C[C@@H]2O NBMKJKDGKREAPL-DVTGEIKXSA-N 0.000 claims description 185
- 229940092705 beclomethasone Drugs 0.000 claims description 183
- 230000000694 effects Effects 0.000 claims description 141
- JYGXADMDTFJGBT-VWUMJDOOSA-N hydrocortisone Chemical compound O=C1CC[C@]2(C)[C@H]3[C@@H](O)C[C@](C)([C@@](CC4)(O)C(=O)CO)[C@@H]4[C@@H]3CCC2=C1 JYGXADMDTFJGBT-VWUMJDOOSA-N 0.000 claims description 84
- 229960003957 dexamethasone Drugs 0.000 claims description 59
- UREBDLICKHMUKA-CXSFZGCWSA-N dexamethasone Chemical compound C1CC2=CC(=O)C=C[C@]2(C)[C@]2(F)[C@@H]1[C@@H]1C[C@@H](C)[C@@](C(=O)CO)(O)[C@@]1(C)C[C@@H]2O UREBDLICKHMUKA-CXSFZGCWSA-N 0.000 claims description 59
- XLYOFNOQVPJJNP-UHFFFAOYSA-N water Substances O XLYOFNOQVPJJNP-UHFFFAOYSA-N 0.000 claims description 55
- LFQSCWFLJHTTHZ-UHFFFAOYSA-N Ethanol Chemical compound CCO LFQSCWFLJHTTHZ-UHFFFAOYSA-N 0.000 claims description 43
- 229960000890 hydrocortisone Drugs 0.000 claims description 42
- 229920005989 resin Polymers 0.000 claims description 39
- 239000011347 resin Substances 0.000 claims description 39
- 230000002829 reductive effect Effects 0.000 claims description 35
- 239000008194 pharmaceutical composition Substances 0.000 claims description 32
- BBEUDPAEKGPXDG-UHFFFAOYSA-N protopanaxatriol Natural products CC(CCC=C(C)C)C1CCC2(C)C1C(O)CC3C4(C)CCC(O)C(C)(C)C4C(O)CC23C BBEUDPAEKGPXDG-UHFFFAOYSA-N 0.000 claims description 30
- 239000003125 aqueous solvent Substances 0.000 claims description 27
- 239000000284 extract Substances 0.000 claims description 26
- 241000196324 Embryophyta Species 0.000 claims description 23
- 210000001519 tissue Anatomy 0.000 claims description 22
- 230000003247 decreasing effect Effects 0.000 claims description 21
- 208000027866 inflammatory disease Diseases 0.000 claims description 21
- 206010061218 Inflammation Diseases 0.000 claims description 15
- 230000004054 inflammatory process Effects 0.000 claims description 15
- 239000003937 drug carrier Substances 0.000 claims description 14
- 235000002791 Panax Nutrition 0.000 claims description 13
- 241000208343 Panax Species 0.000 claims description 13
- 240000004371 Panax ginseng Species 0.000 claims description 13
- OKKJLVBELUTLKV-UHFFFAOYSA-N Methanol Chemical compound OC OKKJLVBELUTLKV-UHFFFAOYSA-N 0.000 claims description 12
- 208000024780 Urticaria Diseases 0.000 claims description 12
- 201000006417 multiple sclerosis Diseases 0.000 claims description 12
- 238000004042 decolorization Methods 0.000 claims description 11
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 claims description 11
- 230000012010 growth Effects 0.000 claims description 11
- 208000010668 atopic eczema Diseases 0.000 claims description 10
- 208000035475 disorder Diseases 0.000 claims description 10
- 230000029663 wound healing Effects 0.000 claims description 10
- 208000001132 Osteoporosis Diseases 0.000 claims description 9
- 208000006673 asthma Diseases 0.000 claims description 9
- 239000000017 hydrogel Substances 0.000 claims description 9
- 208000035285 Allergic Seasonal Rhinitis Diseases 0.000 claims description 8
- 208000006545 Chronic Obstructive Pulmonary Disease Diseases 0.000 claims description 8
- 208000011231 Crohn disease Diseases 0.000 claims description 8
- 206010012438 Dermatitis atopic Diseases 0.000 claims description 8
- 206010020772 Hypertension Diseases 0.000 claims description 8
- 208000022559 Inflammatory bowel disease Diseases 0.000 claims description 8
- 208000007048 Polymyalgia Rheumatica Diseases 0.000 claims description 8
- 206010039085 Rhinitis allergic Diseases 0.000 claims description 8
- 201000010105 allergic rhinitis Diseases 0.000 claims description 8
- 201000008937 atopic dermatitis Diseases 0.000 claims description 8
- 230000007850 degeneration Effects 0.000 claims description 8
- 206010012601 diabetes mellitus Diseases 0.000 claims description 8
- 210000001503 joint Anatomy 0.000 claims description 8
- 206010025135 lupus erythematosus Diseases 0.000 claims description 8
- 210000003205 muscle Anatomy 0.000 claims description 8
- 210000002435 tendon Anatomy 0.000 claims description 8
- 230000004584 weight gain Effects 0.000 claims description 8
- 235000019786 weight gain Nutrition 0.000 claims description 8
- 235000008434 ginseng Nutrition 0.000 claims description 7
- 239000002904 solvent Substances 0.000 claims description 7
- KFZMGEQAYNKOFK-UHFFFAOYSA-N Isopropanol Chemical compound CC(C)O KFZMGEQAYNKOFK-UHFFFAOYSA-N 0.000 claims description 6
- 239000000499 gel Substances 0.000 claims description 6
- 235000002789 Panax ginseng Nutrition 0.000 claims description 5
- 230000005526 G1 to G0 transition Effects 0.000 claims description 4
- 244000062730 Melissa officinalis Species 0.000 claims description 3
- 235000010654 Melissa officinalis Nutrition 0.000 claims description 3
- 239000000443 aerosol Substances 0.000 claims description 3
- 239000002775 capsule Substances 0.000 claims description 3
- 238000004440 column chromatography Methods 0.000 claims description 3
- 239000006071 cream Substances 0.000 claims description 3
- 239000006260 foam Substances 0.000 claims description 3
- 239000000865 liniment Substances 0.000 claims description 3
- 239000006210 lotion Substances 0.000 claims description 3
- 239000007937 lozenge Substances 0.000 claims description 3
- 239000002674 ointment Substances 0.000 claims description 3
- BDERNNFJNOPAEC-UHFFFAOYSA-N propan-1-ol Chemical compound CCCO BDERNNFJNOPAEC-UHFFFAOYSA-N 0.000 claims description 3
- 239000007921 spray Substances 0.000 claims description 3
- 239000006188 syrup Substances 0.000 claims description 3
- 235000020357 syrup Nutrition 0.000 claims description 3
- 239000003826 tablet Substances 0.000 claims description 3
- 229940100613 topical solution Drugs 0.000 claims description 3
- 229940061102 topical suspension Drugs 0.000 claims description 3
- 230000001476 alcoholic effect Effects 0.000 claims description 2
- 206010003246 arthritis Diseases 0.000 claims 2
- 206010009887 colitis Diseases 0.000 claims 2
- SHCBCKBYTHZQGZ-DLHMIPLTSA-N protopanaxatriol Chemical compound C1C[C@H](O)C(C)(C)[C@@H]2[C@@H](O)C[C@@]3(C)[C@]4(C)CC[C@H]([C@](C)(O)CCC=C(C)C)[C@H]4[C@H](O)C[C@@H]3[C@]21C SHCBCKBYTHZQGZ-DLHMIPLTSA-N 0.000 claims 2
- 230000002411 adverse Effects 0.000 claims 1
- 210000000952 spleen Anatomy 0.000 claims 1
- 230000002265 prevention Effects 0.000 abstract description 14
- YURJSTAIMNSZAE-HHNZYBFYSA-N ginsenoside Rg1 Chemical compound O([C@@](C)(CCC=C(C)C)[C@@H]1[C@@H]2[C@@]([C@@]3(C[C@@H]([C@H]4C(C)(C)[C@@H](O)CC[C@]4(C)[C@H]3C[C@H]2O)O[C@H]2[C@@H]([C@@H](O)[C@H](O)[C@@H](CO)O2)O)C)(C)CC1)[C@@H]1O[C@H](CO)[C@@H](O)[C@H](O)[C@H]1O YURJSTAIMNSZAE-HHNZYBFYSA-N 0.000 description 100
- 241000252212 Danio rerio Species 0.000 description 84
- 102000003676 Glucocorticoid Receptors Human genes 0.000 description 80
- 108090000079 Glucocorticoid Receptors Proteins 0.000 description 80
- 210000004027 cell Anatomy 0.000 description 67
- 230000014509 gene expression Effects 0.000 description 60
- 238000002474 experimental method Methods 0.000 description 58
- 150000001875 compounds Chemical class 0.000 description 56
- 108090000623 proteins and genes Proteins 0.000 description 47
- 239000003981 vehicle Substances 0.000 description 46
- 230000003110 anti-inflammatory effect Effects 0.000 description 45
- 238000011866 long-term treatment Methods 0.000 description 41
- 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 33
- 238000011278 co-treatment Methods 0.000 description 32
- 239000008103 glucose Substances 0.000 description 32
- 108020004999 messenger RNA Proteins 0.000 description 30
- 210000000440 neutrophil Anatomy 0.000 description 30
- 238000003753 real-time PCR Methods 0.000 description 30
- SHCBCKBYTHZQGZ-CJPZEJHVSA-N protopanaxatriol Chemical group C1C[C@H](O)C(C)(C)[C@@H]2[C@@H](O)C[C@@]3(C)[C@]4(C)CC[C@H]([C@@](C)(O)CCC=C(C)C)[C@H]4[C@H](O)C[C@@H]3[C@]21C SHCBCKBYTHZQGZ-CJPZEJHVSA-N 0.000 description 29
- IAZDPXIOMUYVGZ-UHFFFAOYSA-N Dimethylsulphoxide Chemical compound CS(C)=O IAZDPXIOMUYVGZ-UHFFFAOYSA-N 0.000 description 27
- 108010048367 enhanced green fluorescent protein Proteins 0.000 description 27
- 235000001727 glucose Nutrition 0.000 description 24
- 239000000126 substance Substances 0.000 description 23
- YPHMISFOHDHNIV-FSZOTQKASA-N cycloheximide Chemical compound C1[C@@H](C)C[C@H](C)C(=O)[C@@H]1[C@H](O)CC1CC(=O)NC(=O)C1 YPHMISFOHDHNIV-FSZOTQKASA-N 0.000 description 22
- 235000013601 eggs Nutrition 0.000 description 22
- 230000023603 positive regulation of transcription initiation, DNA-dependent Effects 0.000 description 22
- 230000007774 longterm Effects 0.000 description 21
- 230000005012 migration Effects 0.000 description 21
- 238000013508 migration Methods 0.000 description 21
- 230000001965 increasing effect Effects 0.000 description 19
- 102000004169 proteins and genes Human genes 0.000 description 18
- 239000000243 solution Substances 0.000 description 17
- RJURFGZVJUQBHK-UHFFFAOYSA-N actinomycin D Natural products CC1OC(=O)C(C(C)C)N(C)C(=O)CN(C)C(=O)C2CCCN2C(=O)C(C(C)C)NC(=O)C1NC(=O)C1=C(N)C(=O)C(C)=C2OC(C(C)=CC=C3C(=O)NC4C(=O)NC(C(N5CCCC5C(=O)N(C)CC(=O)N(C)C(C(C)C)C(=O)OC4C)=O)C(C)C)=C3N=C21 RJURFGZVJUQBHK-UHFFFAOYSA-N 0.000 description 16
- 238000011284 combination treatment Methods 0.000 description 16
- 239000000523 sample Substances 0.000 description 16
- 210000002257 embryonic structure Anatomy 0.000 description 15
- 230000001418 larval effect Effects 0.000 description 15
- 230000002441 reversible effect Effects 0.000 description 15
- 230000035945 sensitivity Effects 0.000 description 15
- 108060008682 Tumor Necrosis Factor Proteins 0.000 description 14
- 102100040247 Tumor necrosis factor Human genes 0.000 description 14
- 210000002540 macrophage Anatomy 0.000 description 13
- 239000006180 TBST buffer Substances 0.000 description 12
- 238000003556 assay Methods 0.000 description 12
- 235000020710 ginseng extract Nutrition 0.000 description 11
- 238000004128 high performance liquid chromatography Methods 0.000 description 11
- 230000008929 regeneration Effects 0.000 description 11
- 238000011069 regeneration method Methods 0.000 description 11
- 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 10
- 101150073411 gba-2 gene Proteins 0.000 description 10
- PWAOOJDMFUQOKB-WCZZMFLVSA-N ginsenoside Re Chemical compound O[C@@H]1[C@H](O)[C@@H](O)[C@H](C)O[C@H]1O[C@H]1[C@H](O[C@@H]2[C@H]3C(C)(C)[C@@H](O)CC[C@]3(C)[C@@H]3[C@@]([C@@]4(CC[C@@H]([C@H]4[C@H](O)C3)[C@](C)(CCC=C(C)C)O[C@H]3[C@@H]([C@@H](O)[C@H](O)[C@@H](CO)O3)O)C)(C)C2)O[C@H](CO)[C@@H](O)[C@@H]1O PWAOOJDMFUQOKB-WCZZMFLVSA-N 0.000 description 10
- 125000002791 glucosyl group Chemical group C1([C@H](O)[C@@H](O)[C@H](O)[C@H](O1)CO)* 0.000 description 10
- 238000005259 measurement Methods 0.000 description 10
- 239000002953 phosphate buffered saline Substances 0.000 description 10
- 108091032973 (ribonucleotides)n+m Proteins 0.000 description 9
- 230000017423 tissue regeneration Effects 0.000 description 9
- 238000001262 western blot Methods 0.000 description 9
- 108010092160 Dactinomycin Proteins 0.000 description 8
- 238000002965 ELISA Methods 0.000 description 8
- 101001033249 Homo sapiens Interleukin-1 beta Proteins 0.000 description 8
- RJURFGZVJUQBHK-IIXSONLDSA-N actinomycin D Chemical compound C[C@H]1OC(=O)[C@H](C(C)C)N(C)C(=O)CN(C)C(=O)[C@@H]2CCCN2C(=O)[C@@H](C(C)C)NC(=O)[C@H]1NC(=O)C1=C(N)C(=O)C(C)=C2OC(C(C)=CC=C3C(=O)N[C@@H]4C(=O)N[C@@H](C(N5CCC[C@H]5C(=O)N(C)CC(=O)N(C)[C@@H](C(C)C)C(=O)O[C@@H]4C)=O)C(C)C)=C3N=C21 RJURFGZVJUQBHK-IIXSONLDSA-N 0.000 description 8
- 238000002266 amputation Methods 0.000 description 8
- 239000012736 aqueous medium Substances 0.000 description 8
- 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 8
- 230000027455 binding Effects 0.000 description 8
- 230000015572 biosynthetic process Effects 0.000 description 8
- 229960000640 dactinomycin Drugs 0.000 description 8
- 230000007423 decrease Effects 0.000 description 8
- 230000006870 function Effects 0.000 description 8
- 230000005764 inhibitory process Effects 0.000 description 8
- 230000009467 reduction Effects 0.000 description 8
- 238000013518 transcription Methods 0.000 description 8
- 230000035897 transcription Effects 0.000 description 8
- 101000961071 Homo sapiens NF-kappa-B inhibitor alpha Proteins 0.000 description 7
- 102100039065 Interleukin-1 beta Human genes 0.000 description 7
- 102100039337 NF-kappa-B inhibitor alpha Human genes 0.000 description 7
- 229930040373 Paraformaldehyde Natural products 0.000 description 7
- 208000021017 Weight Gain Diseases 0.000 description 7
- 238000007398 colorimetric assay Methods 0.000 description 7
- 230000002860 competitive effect Effects 0.000 description 7
- 229920002866 paraformaldehyde Polymers 0.000 description 7
- 230000000270 postfertilization Effects 0.000 description 7
- 238000001542 size-exclusion chromatography Methods 0.000 description 7
- 108020004463 18S ribosomal RNA Proteins 0.000 description 6
- 108091003079 Bovine Serum Albumin Proteins 0.000 description 6
- 206010009900 Colitis ulcerative Diseases 0.000 description 6
- 239000006144 Dulbecco’s modified Eagle's medium Substances 0.000 description 6
- XEKOWRVHYACXOJ-UHFFFAOYSA-N Ethyl acetate Chemical compound CCOC(C)=O XEKOWRVHYACXOJ-UHFFFAOYSA-N 0.000 description 6
- 208000007465 Giant cell arteritis Diseases 0.000 description 6
- 101000596277 Homo sapiens TSC22 domain family protein 3 Proteins 0.000 description 6
- 208000010428 Muscle Weakness Diseases 0.000 description 6
- 206010028372 Muscular weakness Diseases 0.000 description 6
- 102100035260 TSC22 domain family protein 3 Human genes 0.000 description 6
- 201000006704 Ulcerative Colitis Diseases 0.000 description 6
- 231100000673 dose–response relationship Toxicity 0.000 description 6
- 230000003828 downregulation Effects 0.000 description 6
- AOGZLQUEBLOQCI-UHFFFAOYSA-N ginsenoside-Re Natural products CC1OC(OCC2OC(OC3CC4(C)C(CC(O)C5C(CCC45C)C(C)(CCC=C(C)C)OC6OC(CO)C(O)C(O)C6O)C7(C)CCC(O)C(C)(C)C37)C(O)C(O)C2O)C(O)C(O)C1O AOGZLQUEBLOQCI-UHFFFAOYSA-N 0.000 description 6
- 239000005090 green fluorescent protein Substances 0.000 description 6
- 230000002401 inhibitory effect Effects 0.000 description 6
- 230000005937 nuclear translocation Effects 0.000 description 6
- 101150067708 pckG gene Proteins 0.000 description 6
- 229960004618 prednisone Drugs 0.000 description 6
- XOFYZVNMUHMLCC-ZPOLXVRWSA-N prednisone Chemical compound O=C1C=C[C@]2(C)[C@H]3C(=O)C[C@](C)([C@@](CC4)(O)C(=O)CO)[C@@H]4[C@@H]3CCC2=C1 XOFYZVNMUHMLCC-ZPOLXVRWSA-N 0.000 description 6
- 235000000346 sugar Nutrition 0.000 description 6
- 206010043207 temporal arteritis Diseases 0.000 description 6
- 238000012360 testing method Methods 0.000 description 6
- 230000005945 translocation Effects 0.000 description 6
- 238000007492 two-way ANOVA Methods 0.000 description 6
- 101000859679 Homo sapiens Non-lysosomal glucosylceramidase Proteins 0.000 description 5
- 102000004890 Interleukin-8 Human genes 0.000 description 5
- 108090001007 Interleukin-8 Proteins 0.000 description 5
- 240000005373 Panax quinquefolius Species 0.000 description 5
- 238000010162 Tukey test Methods 0.000 description 5
- 239000008365 aqueous carrier Substances 0.000 description 5
- 238000006243 chemical reaction Methods 0.000 description 5
- 230000001419 dependent effect Effects 0.000 description 5
- 238000010790 dilution Methods 0.000 description 5
- 239000012895 dilution Substances 0.000 description 5
- 239000003814 drug Substances 0.000 description 5
- 238000010828 elution Methods 0.000 description 5
- 238000002073 fluorescence micrograph Methods 0.000 description 5
- 239000011521 glass Substances 0.000 description 5
- 210000005260 human cell Anatomy 0.000 description 5
- 239000003112 inhibitor Substances 0.000 description 5
- 238000001543 one-way ANOVA Methods 0.000 description 5
- 229960005205 prednisolone Drugs 0.000 description 5
- 239000000651 prodrug Substances 0.000 description 5
- 229940002612 prodrug Drugs 0.000 description 5
- 230000000770 proinflammatory effect Effects 0.000 description 5
- 230000004083 survival effect Effects 0.000 description 5
- 230000035903 transrepression Effects 0.000 description 5
- PYXFVCFISTUSOO-HKUCOEKDSA-N (20S)-protopanaxadiol Chemical compound C1C[C@H](O)C(C)(C)[C@@H]2CC[C@@]3(C)[C@]4(C)CC[C@H]([C@@](C)(O)CCC=C(C)C)[C@H]4[C@H](O)C[C@@H]3[C@]21C PYXFVCFISTUSOO-HKUCOEKDSA-N 0.000 description 4
- HJCMDXDYPOUFDY-WHFBIAKZSA-N Ala-Gln Chemical compound C[C@H](N)C(=O)N[C@H](C(O)=O)CCC(N)=O HJCMDXDYPOUFDY-WHFBIAKZSA-N 0.000 description 4
- 108091058560 IL8 Proteins 0.000 description 4
- 101100242976 Neurospora crassa (strain ATCC 24698 / 74-OR23-1A / CBS 708.71 / DSM 1257 / FGSC 987) acu-6 gene Proteins 0.000 description 4
- 102100027814 Non-lysosomal glucosylceramidase Human genes 0.000 description 4
- 241000168720 Panax japonicus Species 0.000 description 4
- 241000180649 Panax notoginseng Species 0.000 description 4
- 235000003140 Panax quinquefolius Nutrition 0.000 description 4
- 230000009471 action Effects 0.000 description 4
- 230000004913 activation Effects 0.000 description 4
- 231100000215 acute (single dose) toxicity testing Toxicity 0.000 description 4
- 238000011047 acute toxicity test Methods 0.000 description 4
- 239000000654 additive Substances 0.000 description 4
- 230000000996 additive effect Effects 0.000 description 4
- 238000004458 analytical method Methods 0.000 description 4
- 230000003042 antagnostic effect Effects 0.000 description 4
- 239000000872 buffer Substances 0.000 description 4
- 239000003153 chemical reaction reagent Substances 0.000 description 4
- 239000003795 chemical substances by application Substances 0.000 description 4
- 239000002299 complementary DNA Substances 0.000 description 4
- 238000001514 detection method Methods 0.000 description 4
- IINNWAYUJNWZRM-UHFFFAOYSA-L erythrosin B Chemical compound [Na+].[Na+].[O-]C(=O)C1=CC=CC=C1C1=C2C=C(I)C(=O)C(I)=C2OC2=C(I)C([O-])=C(I)C=C21 IINNWAYUJNWZRM-UHFFFAOYSA-L 0.000 description 4
- 229940011411 erythrosine Drugs 0.000 description 4
- 235000012732 erythrosine Nutrition 0.000 description 4
- 239000004174 erythrosine Substances 0.000 description 4
- 238000001704 evaporation Methods 0.000 description 4
- 239000012894 fetal calf serum Substances 0.000 description 4
- SYWHXTATXSMDSB-GSLJADNHSA-N fludrocortisone acetate Chemical compound C1CC2=CC(=O)CC[C@]2(C)[C@]2(F)[C@@H]1[C@@H]1CC[C@@](C(=O)COC(=O)C)(O)[C@@]1(C)C[C@@H]2O SYWHXTATXSMDSB-GSLJADNHSA-N 0.000 description 4
- 229960003336 fluorocortisol acetate Drugs 0.000 description 4
- 230000013595 glycosylation Effects 0.000 description 4
- 238000006206 glycosylation reaction Methods 0.000 description 4
- 230000012447 hatching Effects 0.000 description 4
- 230000002209 hydrophobic effect Effects 0.000 description 4
- 238000003384 imaging method Methods 0.000 description 4
- 238000003365 immunocytochemistry Methods 0.000 description 4
- 230000006698 induction Effects 0.000 description 4
- 239000007924 injection Substances 0.000 description 4
- 238000002347 injection Methods 0.000 description 4
- 210000001161 mammalian embryo Anatomy 0.000 description 4
- 230000001404 mediated effect Effects 0.000 description 4
- 239000002609 medium Substances 0.000 description 4
- -1 ocotillol saponin Chemical class 0.000 description 4
- 239000013612 plasmid Substances 0.000 description 4
- 239000013641 positive control Substances 0.000 description 4
- OIGNJSKKLXVSLS-VWUMJDOOSA-N prednisolone Chemical compound O=C1C=C[C@]2(C)[C@H]3[C@@H](O)C[C@](C)([C@@](CC4)(O)C(=O)CO)[C@@H]4[C@@H]3CCC2=C1 OIGNJSKKLXVSLS-VWUMJDOOSA-N 0.000 description 4
- 238000002360 preparation method Methods 0.000 description 4
- 102000005962 receptors Human genes 0.000 description 4
- 108020003175 receptors Proteins 0.000 description 4
- 238000007619 statistical method Methods 0.000 description 4
- 150000003431 steroids Chemical group 0.000 description 4
- 239000006228 supernatant Substances 0.000 description 4
- 230000002195 synergetic effect Effects 0.000 description 4
- 230000009885 systemic effect Effects 0.000 description 4
- 230000001225 therapeutic effect Effects 0.000 description 4
- 230000009261 transgenic effect Effects 0.000 description 4
- LWIHDJKSTIGBAC-UHFFFAOYSA-K tripotassium phosphate Chemical compound [K+].[K+].[K+].[O-]P([O-])([O-])=O LWIHDJKSTIGBAC-UHFFFAOYSA-K 0.000 description 4
- WEVYAHXRMPXWCK-UHFFFAOYSA-N Acetonitrile Chemical compound CC#N WEVYAHXRMPXWCK-UHFFFAOYSA-N 0.000 description 3
- TWCMVXMQHSVIOJ-UHFFFAOYSA-N Aglycone of yadanzioside D Natural products COC(=O)C12OCC34C(CC5C(=CC(O)C(O)C5(C)C3C(O)C1O)C)OC(=O)C(OC(=O)C)C24 TWCMVXMQHSVIOJ-UHFFFAOYSA-N 0.000 description 3
- PLMKQQMDOMTZGG-UHFFFAOYSA-N Astrantiagenin E-methylester Natural products CC12CCC(O)C(C)(CO)C1CCC1(C)C2CC=C2C3CC(C)(C)CCC3(C(=O)OC)CCC21C PLMKQQMDOMTZGG-UHFFFAOYSA-N 0.000 description 3
- 229920002307 Dextran Polymers 0.000 description 3
- 101000878253 Homo sapiens Peptidyl-prolyl cis-trans isomerase FKBP5 Proteins 0.000 description 3
- 101000864800 Homo sapiens Serine/threonine-protein kinase Sgk1 Proteins 0.000 description 3
- 235000005035 Panax pseudoginseng ssp. pseudoginseng Nutrition 0.000 description 3
- BELBBZDIHDAJOR-UHFFFAOYSA-N Phenolsulfonephthalein Chemical compound C1=CC(O)=CC=C1C1(C=2C=CC(O)=CC=2)C2=CC=CC=C2S(=O)(=O)O1 BELBBZDIHDAJOR-UHFFFAOYSA-N 0.000 description 3
- 229920001213 Polysorbate 20 Polymers 0.000 description 3
- 102100030070 Serine/threonine-protein kinase Sgk1 Human genes 0.000 description 3
- 102000040945 Transcription factor Human genes 0.000 description 3
- 108091023040 Transcription factor Proteins 0.000 description 3
- 239000005557 antagonist Substances 0.000 description 3
- 239000007864 aqueous solution Substances 0.000 description 3
- 229960002537 betamethasone Drugs 0.000 description 3
- UREBDLICKHMUKA-DVTGEIKXSA-N betamethasone Chemical compound C1CC2=CC(=O)C=C[C@]2(C)[C@]2(F)[C@@H]1[C@@H]1C[C@H](C)[C@@](C(=O)CO)(O)[C@@]1(C)C[C@@H]2O UREBDLICKHMUKA-DVTGEIKXSA-N 0.000 description 3
- 238000004113 cell culture Methods 0.000 description 3
- 230000001684 chronic effect Effects 0.000 description 3
- 208000037976 chronic inflammation Diseases 0.000 description 3
- OORMXZNMRWBSTK-LGFJJATJSA-N dammarane Chemical group C1CCC(C)(C)[C@@H]2CC[C@@]3(C)[C@]4(C)CC[C@H]([C@H](C)CCCC(C)C)[C@H]4CC[C@@H]3[C@]21C OORMXZNMRWBSTK-LGFJJATJSA-N 0.000 description 3
- 238000001378 electrochemiluminescence detection Methods 0.000 description 3
- 230000008020 evaporation Effects 0.000 description 3
- 210000001035 gastrointestinal tract Anatomy 0.000 description 3
- PFOARMALXZGCHY-UHFFFAOYSA-N homoegonol Natural products C1=C(OC)C(OC)=CC=C1C1=CC2=CC(CCCO)=CC(OC)=C2O1 PFOARMALXZGCHY-UHFFFAOYSA-N 0.000 description 3
- 238000000338 in vitro Methods 0.000 description 3
- 210000000265 leukocyte Anatomy 0.000 description 3
- 239000003446 ligand Substances 0.000 description 3
- 230000007246 mechanism Effects 0.000 description 3
- CXKWCBBOMKCUKX-UHFFFAOYSA-M methylene blue Chemical compound [Cl-].C1=CC(N(C)C)=CC2=[S+]C3=CC(N(C)C)=CC=C3N=C21 CXKWCBBOMKCUKX-UHFFFAOYSA-M 0.000 description 3
- 229960000907 methylthioninium chloride Drugs 0.000 description 3
- 238000002156 mixing Methods 0.000 description 3
- 230000009456 molecular mechanism Effects 0.000 description 3
- 230000002018 overexpression Effects 0.000 description 3
- 229960003531 phenolsulfonphthalein Drugs 0.000 description 3
- 239000000256 polyoxyethylene sorbitan monolaurate Substances 0.000 description 3
- 235000010486 polyoxyethylene sorbitan monolaurate Nutrition 0.000 description 3
- 238000000746 purification Methods 0.000 description 3
- 238000011002 quantification Methods 0.000 description 3
- 235000002639 sodium chloride Nutrition 0.000 description 3
- 239000000758 substrate Substances 0.000 description 3
- 230000001629 suppression Effects 0.000 description 3
- 238000002560 therapeutic procedure Methods 0.000 description 3
- RWXIFXNRCLMQCD-JBVRGBGGSA-N (20S)-ginsenoside Rg3 Chemical compound O([C@@H]1[C@@H](O)[C@H](O)[C@@H](CO)O[C@H]1O[C@H]1CC[C@]2(C)[C@H]3C[C@@H](O)[C@H]4[C@@]([C@@]3(CC[C@H]2C1(C)C)C)(C)CC[C@@H]4[C@@](C)(O)CCC=C(C)C)[C@@H]1O[C@H](CO)[C@@H](O)[C@H](O)[C@H]1O RWXIFXNRCLMQCD-JBVRGBGGSA-N 0.000 description 2
- RAQNTCRNSXYLAH-RFCGZQMISA-N (20S)-ginsenoside Rh1 Chemical compound O([C@@H]1[C@H]2C(C)(C)[C@@H](O)CC[C@]2(C)[C@H]2C[C@@H](O)[C@H]3[C@@]([C@@]2(C1)C)(C)CC[C@@H]3[C@@](C)(O)CCC=C(C)C)[C@@H]1O[C@H](CO)[C@@H](O)[C@H](O)[C@H]1O RAQNTCRNSXYLAH-RFCGZQMISA-N 0.000 description 2
- ZUXNULGHCOXCFL-UHFFFAOYSA-N 2-(4-tert-butyl-2,6-dimethylphenyl)acetonitrile Chemical compound CC1=CC(C(C)(C)C)=CC(C)=C1CC#N ZUXNULGHCOXCFL-UHFFFAOYSA-N 0.000 description 2
- GHWSMQHJFMAATF-DSHMRAQASA-N 20(S)-protopanaxadiol Natural products CC(=CCC[C@@](O)(CO)[C@H]1CC[C@]2(C)[C@@H]1CC[C@H]3[C@@]2(C)CC[C@H]4C(C)(C)[C@@H](O)CC[C@]34CO)C GHWSMQHJFMAATF-DSHMRAQASA-N 0.000 description 2
- 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 2
- 229920000936 Agarose Polymers 0.000 description 2
- PQSUYGKTWSAVDQ-ZVIOFETBSA-N Aldosterone Chemical compound C([C@@]1([C@@H](C(=O)CO)CC[C@H]1[C@@H]1CC2)C=O)[C@H](O)[C@@H]1[C@]1(C)C2=CC(=O)CC1 PQSUYGKTWSAVDQ-ZVIOFETBSA-N 0.000 description 2
- PQSUYGKTWSAVDQ-UHFFFAOYSA-N Aldosterone Natural products C1CC2C3CCC(C(=O)CO)C3(C=O)CC(O)C2C2(C)C1=CC(=O)CC2 PQSUYGKTWSAVDQ-UHFFFAOYSA-N 0.000 description 2
- 101100282794 Caenorhabditis elegans gba-2 gene Proteins 0.000 description 2
- CURLTUGMZLYLDI-UHFFFAOYSA-N Carbon dioxide Chemical compound O=C=O CURLTUGMZLYLDI-UHFFFAOYSA-N 0.000 description 2
- 206010008342 Cervix carcinoma Diseases 0.000 description 2
- 108010072220 Cyclophilin A Proteins 0.000 description 2
- 108020004414 DNA Proteins 0.000 description 2
- 201000004624 Dermatitis Diseases 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
- TWLLPUMZVVGILS-UHFFFAOYSA-N Ethyl 2-aminobenzoate Chemical compound CCOC(=O)C1=CC=CC=C1N TWLLPUMZVVGILS-UHFFFAOYSA-N 0.000 description 2
- BGHNZAWRRWLKPO-UHFFFAOYSA-N Ginsenoside F1 Natural products CC(=C)CCCC(C)(OC1OC(CO)C(O)C(O)C1O)C2CCC3(C)C2C(O)CC4C5(C)CCC(O)C(C)(C)C5C(O)CC34C BGHNZAWRRWLKPO-UHFFFAOYSA-N 0.000 description 2
- 101000926939 Homo sapiens Glucocorticoid receptor Proteins 0.000 description 2
- 101001071608 Homo sapiens Glutathione reductase, mitochondrial Proteins 0.000 description 2
- 101000990902 Homo sapiens Matrix metalloproteinase-9 Proteins 0.000 description 2
- 206010021033 Hypomenorrhoea Diseases 0.000 description 2
- 241000124008 Mammalia Species 0.000 description 2
- 102100030412 Matrix metalloproteinase-9 Human genes 0.000 description 2
- 241001465754 Metazoa Species 0.000 description 2
- FQISKWAFAHGMGT-SGJOWKDISA-M Methylprednisolone sodium succinate Chemical compound [Na+].C([C@@]12C)=CC(=O)C=C1[C@@H](C)C[C@@H]1[C@@H]2[C@@H](O)C[C@]2(C)[C@@](O)(C(=O)COC(=O)CCC([O-])=O)CC[C@H]21 FQISKWAFAHGMGT-SGJOWKDISA-M 0.000 description 2
- 241000283973 Oryctolagus cuniculus Species 0.000 description 2
- 102100034539 Peptidyl-prolyl cis-trans isomerase A Human genes 0.000 description 2
- 102100037026 Peptidyl-prolyl cis-trans isomerase FKBP5 Human genes 0.000 description 2
- 108010020062 Peptidylprolyl Isomerase Proteins 0.000 description 2
- 102000009658 Peptidylprolyl Isomerase Human genes 0.000 description 2
- NBIIXXVUZAFLBC-UHFFFAOYSA-N Phosphoric acid Chemical compound OP(O)(O)=O NBIIXXVUZAFLBC-UHFFFAOYSA-N 0.000 description 2
- 239000013614 RNA sample Substances 0.000 description 2
- 229920005654 Sephadex Polymers 0.000 description 2
- 239000012507 Sephadex™ Substances 0.000 description 2
- 102000004142 Trypsin Human genes 0.000 description 2
- 108090000631 Trypsin Proteins 0.000 description 2
- 208000006105 Uterine Cervical Neoplasms Diseases 0.000 description 2
- 229960002478 aldosterone Drugs 0.000 description 2
- 229940124599 anti-inflammatory drug Drugs 0.000 description 2
- 230000005784 autoimmunity Effects 0.000 description 2
- PYXFVCFISTUSOO-UHFFFAOYSA-N betulafolienetriol Natural products C1CC(O)C(C)(C)C2CCC3(C)C4(C)CCC(C(C)(O)CCC=C(C)C)C4C(O)CC3C21C PYXFVCFISTUSOO-UHFFFAOYSA-N 0.000 description 2
- 239000000090 biomarker Substances 0.000 description 2
- 238000009835 boiling Methods 0.000 description 2
- 229940098773 bovine serum albumin Drugs 0.000 description 2
- 238000010804 cDNA synthesis Methods 0.000 description 2
- 238000010805 cDNA synthesis kit Methods 0.000 description 2
- 229960004424 carbon dioxide Drugs 0.000 description 2
- 235000011089 carbon dioxide Nutrition 0.000 description 2
- 201000010881 cervical cancer Diseases 0.000 description 2
- 230000008859 change Effects 0.000 description 2
- 230000006020 chronic inflammation Effects 0.000 description 2
- 230000002060 circadian Effects 0.000 description 2
- 230000009260 cross reactivity Effects 0.000 description 2
- 238000002425 crystallisation Methods 0.000 description 2
- 230000008025 crystallization Effects 0.000 description 2
- 229960001145 deflazacort Drugs 0.000 description 2
- FBHSPRKOSMHSIF-GRMWVWQJSA-N deflazacort Chemical compound C1CC2=CC(=O)C=C[C@]2(C)[C@@H]2[C@@H]1[C@@H]1C[C@H]3OC(C)=N[C@@]3(C(=O)COC(=O)C)[C@@]1(C)C[C@@H]2O FBHSPRKOSMHSIF-GRMWVWQJSA-N 0.000 description 2
- 230000022811 deglycosylation Effects 0.000 description 2
- 238000004925 denaturation Methods 0.000 description 2
- 230000036425 denaturation Effects 0.000 description 2
- 238000011161 development Methods 0.000 description 2
- 230000018109 developmental process Effects 0.000 description 2
- 229940079593 drug Drugs 0.000 description 2
- 238000001035 drying Methods 0.000 description 2
- 235000013399 edible fruits Nutrition 0.000 description 2
- 210000000750 endocrine system Anatomy 0.000 description 2
- 230000007717 exclusion Effects 0.000 description 2
- 238000010195 expression analysis Methods 0.000 description 2
- 230000004720 fertilization Effects 0.000 description 2
- 238000002875 fluorescence polarization Methods 0.000 description 2
- XNGXWSFSJIQMNC-FIYORUNESA-N ginsenoside F1 Chemical compound O([C@@](C)(CCC=C(C)C)[C@@H]1[C@@H]2[C@@]([C@@]3(C[C@H](O)[C@H]4C(C)(C)[C@@H](O)CC[C@]4(C)[C@H]3C[C@H]2O)C)(C)CC1)[C@@H]1O[C@H](CO)[C@@H](O)[C@H](O)[C@H]1O XNGXWSFSJIQMNC-FIYORUNESA-N 0.000 description 2
- YURJSTAIMNSZAE-UHFFFAOYSA-N ginsenoside a2 Polymers C1CC(C2(CC(C3C(C)(C)C(O)CCC3(C)C2CC2O)OC3C(C(O)C(O)C(CO)O3)O)C)(C)C2C1C(C)(CCC=C(C)C)OC1OC(CO)C(O)C(O)C1O YURJSTAIMNSZAE-UHFFFAOYSA-N 0.000 description 2
- 230000028709 inflammatory response Effects 0.000 description 2
- 150000002632 lipids Chemical class 0.000 description 2
- 239000007788 liquid Substances 0.000 description 2
- 238000012423 maintenance Methods 0.000 description 2
- 238000004519 manufacturing process Methods 0.000 description 2
- 239000000463 material Substances 0.000 description 2
- 230000002503 metabolic effect Effects 0.000 description 2
- 229960004584 methylprednisolone Drugs 0.000 description 2
- 239000013642 negative control Substances 0.000 description 2
- 239000002798 polar solvent Substances 0.000 description 2
- 229910000160 potassium phosphate Inorganic materials 0.000 description 2
- 235000011009 potassium phosphates Nutrition 0.000 description 2
- 239000001397 quillaja saponaria molina bark Substances 0.000 description 2
- 239000011541 reaction mixture Substances 0.000 description 2
- 230000001172 regenerating effect Effects 0.000 description 2
- 230000008439 repair process Effects 0.000 description 2
- 229930182490 saponin Natural products 0.000 description 2
- 235000017709 saponins Nutrition 0.000 description 2
- 229910052594 sapphire Inorganic materials 0.000 description 2
- 239000010980 sapphire Substances 0.000 description 2
- 210000002966 serum Anatomy 0.000 description 2
- 239000003270 steroid hormone Substances 0.000 description 2
- 229960005294 triamcinolone Drugs 0.000 description 2
- GFNANZIMVAIWHM-OBYCQNJPSA-N triamcinolone Chemical compound O=C1C=C[C@]2(C)[C@@]3(F)[C@@H](O)C[C@](C)([C@@]([C@H](O)C4)(O)C(=O)CO)[C@@H]4[C@@H]3CCC2=C1 GFNANZIMVAIWHM-OBYCQNJPSA-N 0.000 description 2
- 229940072040 tricaine Drugs 0.000 description 2
- FQZJYWMRQDKBQN-UHFFFAOYSA-N tricaine methanesulfonate Chemical compound CS([O-])(=O)=O.CCOC(=O)C1=CC=CC([NH3+])=C1 FQZJYWMRQDKBQN-UHFFFAOYSA-N 0.000 description 2
- 239000012588 trypsin Substances 0.000 description 2
- 230000003827 upregulation Effects 0.000 description 2
- 238000012800 visualization Methods 0.000 description 2
- 238000005406 washing Methods 0.000 description 2
- YOSRLTNUOCHBEA-UHFFFAOYSA-N (3beta)-28-(beta-D-glucopyranosyloxy)-28-oxoolean-12-en-3-yl beta-D-glucopyranosiduronic acid Natural products C12CC(C)(C)CCC2(C(=O)OC2C(C(O)C(O)C(CO)O2)O)CCC(C2(CCC3C4(C)C)C)(C)C1=CCC2C3(C)CCC4OC1OC(C(O)=O)C(O)C(O)C1O YOSRLTNUOCHBEA-UHFFFAOYSA-N 0.000 description 1
- YRIZYWQGELRKNT-UHFFFAOYSA-N 1,3,5-trichloro-1,3,5-triazinane-2,4,6-trione Chemical compound ClN1C(=O)N(Cl)C(=O)N(Cl)C1=O YRIZYWQGELRKNT-UHFFFAOYSA-N 0.000 description 1
- FUFLCEKSBBHCMO-UHFFFAOYSA-N 11-dehydrocorticosterone Natural products O=C1CCC2(C)C3C(=O)CC(C)(C(CC4)C(=O)CO)C4C3CCC2=C1 FUFLCEKSBBHCMO-UHFFFAOYSA-N 0.000 description 1
- VCNKUCWWHVTTBY-UHFFFAOYSA-N 18alpha-Oleanane Natural products C1CCC(C)(C)C2CCC3(C)C4(C)CCC5(C)CCC(C)(C)CC5C4CCC3C21C VCNKUCWWHVTTBY-UHFFFAOYSA-N 0.000 description 1
- SDYWXFYBZPNOFX-UHFFFAOYSA-N 3,4-dichloroaniline Chemical compound NC1=CC=C(Cl)C(Cl)=C1 SDYWXFYBZPNOFX-UHFFFAOYSA-N 0.000 description 1
- JFUAWXPBHXKZGA-IBGZPJMESA-N 4-fluoro-2-[(4r)-5,5,5-trifluoro-4-hydroxy-2-methyl-4-(1h-pyrrolo[2,3-c]pyridin-2-ylmethyl)pentan-2-yl]phenol Chemical compound C([C@@](O)(CC=1NC2=CN=CC=C2C=1)C(F)(F)F)C(C)(C)C1=CC(F)=CC=C1O JFUAWXPBHXKZGA-IBGZPJMESA-N 0.000 description 1
- 241000251468 Actinopterygii Species 0.000 description 1
- 208000024893 Acute lymphoblastic leukemia Diseases 0.000 description 1
- 208000014697 Acute lymphocytic leukaemia Diseases 0.000 description 1
- 208000017194 Affective disease Diseases 0.000 description 1
- 239000012103 Alexa Fluor 488 Substances 0.000 description 1
- 208000023275 Autoimmune disease Diseases 0.000 description 1
- 241000167854 Bourreria succulenta Species 0.000 description 1
- 238000009010 Bradford assay Methods 0.000 description 1
- 101100191768 Caenorhabditis elegans pbs-4 gene Proteins 0.000 description 1
- 241000283707 Capra Species 0.000 description 1
- 229930192360 Chikusetsusaponin Natural products 0.000 description 1
- 239000000055 Corticotropin-Releasing Hormone Substances 0.000 description 1
- MFYSYFVPBJMHGN-ZPOLXVRWSA-N Cortisone Chemical compound O=C1CC[C@]2(C)[C@H]3C(=O)C[C@](C)([C@@](CC4)(O)C(=O)CO)[C@@H]4[C@@H]3CCC2=C1 MFYSYFVPBJMHGN-ZPOLXVRWSA-N 0.000 description 1
- MFYSYFVPBJMHGN-UHFFFAOYSA-N Cortisone Natural products O=C1CCC2(C)C3C(=O)CC(C)(C(CC4)(O)C(=O)CO)C4C3CCC2=C1 MFYSYFVPBJMHGN-UHFFFAOYSA-N 0.000 description 1
- 208000028399 Critical Illness Diseases 0.000 description 1
- 102000004127 Cytokines Human genes 0.000 description 1
- 108090000695 Cytokines Proteins 0.000 description 1
- 238000000116 DAPI staining Methods 0.000 description 1
- 208000035859 Drug effect increased Diseases 0.000 description 1
- 238000008157 ELISA kit Methods 0.000 description 1
- 101100001669 Emericella variicolor andD gene Proteins 0.000 description 1
- 102000004190 Enzymes Human genes 0.000 description 1
- 108090000790 Enzymes Proteins 0.000 description 1
- 101150066002 GFP gene Proteins 0.000 description 1
- 241001123946 Gaga Species 0.000 description 1
- 208000015872 Gaucher disease Diseases 0.000 description 1
- XIRZPICFRDZXPF-UHFFFAOYSA-N Ginsenoside Rg3 Natural products CC(C)=CCCC(C)(O)C1CCC(C2(CC(O)C3C4(C)C)C)(C)C1C(O)CC2C3(C)CCC4OC1OC(CO)C(O)C(O)C1OC1OC(CO)C(O)C(O)C1O XIRZPICFRDZXPF-UHFFFAOYSA-N 0.000 description 1
- RAQNTCRNSXYLAH-UHFFFAOYSA-N Ginsenoside Rh1 Natural products CC(C)=CCCC(C)(O)C1CCC(C2(C3)C)(C)C1C(O)CC2C1(C)CCC(O)C(C)(C)C1C3OC1OC(CO)C(O)C(O)C1O RAQNTCRNSXYLAH-UHFFFAOYSA-N 0.000 description 1
- 208000010412 Glaucoma Diseases 0.000 description 1
- 108700011498 Glucocorticoid Receptor Deficiency Proteins 0.000 description 1
- 101001055222 Homo sapiens Interleukin-8 Proteins 0.000 description 1
- 101001128634 Homo sapiens NADH dehydrogenase [ubiquinone] 1 beta subcomplex subunit 2, mitochondrial Proteins 0.000 description 1
- 101000990915 Homo sapiens Stromelysin-1 Proteins 0.000 description 1
- 102100026236 Interleukin-8 Human genes 0.000 description 1
- 235000005769 Japanese ginseng Nutrition 0.000 description 1
- 239000007836 KH2PO4 Substances 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
- 239000012741 Laemmli sample buffer Substances 0.000 description 1
- FSNCEEGOMTYXKY-JTQLQIEISA-N Lycoperodine 1 Natural products N1C2=CC=CC=C2C2=C1CN[C@H](C(=O)O)C2 FSNCEEGOMTYXKY-JTQLQIEISA-N 0.000 description 1
- 229910015667 MoO4 Inorganic materials 0.000 description 1
- 208000019022 Mood disease Diseases 0.000 description 1
- 241001508003 Mycobacterium abscessus Species 0.000 description 1
- 102000003896 Myeloperoxidases Human genes 0.000 description 1
- 108090000235 Myeloperoxidases Proteins 0.000 description 1
- OKIZCWYLBDKLSU-UHFFFAOYSA-M N,N,N-Trimethylmethanaminium chloride Chemical compound [Cl-].C[N+](C)(C)C OKIZCWYLBDKLSU-UHFFFAOYSA-M 0.000 description 1
- 102100032194 NADH dehydrogenase [ubiquinone] 1 beta subcomplex subunit 2, mitochondrial Human genes 0.000 description 1
- 108010057466 NF-kappa B Proteins 0.000 description 1
- 102000003945 NF-kappa B Human genes 0.000 description 1
- 239000000020 Nitrocellulose Substances 0.000 description 1
- 108091028043 Nucleic acid sequence Proteins 0.000 description 1
- RQBNSDSKUAGBOI-GGWQMWHBSA-N Ocotillol Natural products O1[C@@H](C(C)(O)C)CC[C@@]1(C)[C@@H]1[C@@H](CC[C@H]2[C@]3(CC[C@H]4C(C)(C)[C@@H](O)CC[C@@]42C)C)[C@@]3(C)CC1 RQBNSDSKUAGBOI-GGWQMWHBSA-N 0.000 description 1
- RQBNSDSKUAGBOI-VVGBCXFDSA-N Ocotillol II Natural products O1[C@H](C(C)(O)C)CC[C@@]1(C)[C@@H]1[C@@H](CC[C@H]2[C@]3(CC[C@H]4C(C)(C)[C@@H](O)CC[C@@]42C)C)[C@@]3(C)CC1 RQBNSDSKUAGBOI-VVGBCXFDSA-N 0.000 description 1
- 206010030043 Ocular hypertension Diseases 0.000 description 1
- 229910019142 PO4 Inorganic materials 0.000 description 1
- 235000003174 Panax japonicus Nutrition 0.000 description 1
- 241000190702 Panax japonicus var. bipinnatifidus Species 0.000 description 1
- 235000003143 Panax notoginseng Nutrition 0.000 description 1
- 241000168721 Panax stipuleanatus Species 0.000 description 1
- 244000133018 Panax trifolius Species 0.000 description 1
- 241001527087 Panax vietnamensis Species 0.000 description 1
- 241000180664 Panax wangianus Species 0.000 description 1
- 241000168719 Panax zingiberensis Species 0.000 description 1
- 241000452638 Parasaissetia nigra Species 0.000 description 1
- 206010057249 Phagocytosis Diseases 0.000 description 1
- 244000010922 Plantago major Species 0.000 description 1
- 241000146405 Primula sikkimensis Species 0.000 description 1
- 229940123573 Protein synthesis inhibitor Drugs 0.000 description 1
- 244000173166 Pyrus ussuriensis Species 0.000 description 1
- 108091027981 Response element Proteins 0.000 description 1
- 101100372017 Saccharomyces cerevisiae (strain ATCC 204508 / S288c) UPS3 gene Proteins 0.000 description 1
- 239000013504 Triton X-100 Substances 0.000 description 1
- 229920004890 Triton X-100 Polymers 0.000 description 1
- 239000012963 UV stabilizer Substances 0.000 description 1
- 206010052428 Wound Diseases 0.000 description 1
- 208000027418 Wounds and injury Diseases 0.000 description 1
- 206010000269 abscess Diseases 0.000 description 1
- 210000004100 adrenal gland Anatomy 0.000 description 1
- 230000002776 aggregation Effects 0.000 description 1
- 238000004220 aggregation Methods 0.000 description 1
- 239000000556 agonist Substances 0.000 description 1
- 239000003905 agrochemical Substances 0.000 description 1
- 238000013459 approach Methods 0.000 description 1
- 239000012223 aqueous fraction Substances 0.000 description 1
- 238000003149 assay kit Methods 0.000 description 1
- 230000009286 beneficial effect Effects 0.000 description 1
- 230000008901 benefit Effects 0.000 description 1
- 230000000903 blocking effect Effects 0.000 description 1
- 210000004369 blood Anatomy 0.000 description 1
- 239000008280 blood Substances 0.000 description 1
- 238000000339 bright-field microscopy Methods 0.000 description 1
- 239000007975 buffered saline Substances 0.000 description 1
- 238000004364 calculation method Methods 0.000 description 1
- 150000001720 carbohydrates Chemical class 0.000 description 1
- 235000014633 carbohydrates Nutrition 0.000 description 1
- 230000015556 catabolic process Effects 0.000 description 1
- 230000011712 cell development Effects 0.000 description 1
- 230000003915 cell function Effects 0.000 description 1
- 210000000170 cell membrane Anatomy 0.000 description 1
- 230000005754 cellular signaling Effects 0.000 description 1
- 239000003610 charcoal Substances 0.000 description 1
- 229920001429 chelating resin Polymers 0.000 description 1
- 235000019693 cherries Nutrition 0.000 description 1
- 210000000038 chest Anatomy 0.000 description 1
- 208000037893 chronic inflammatory disorder Diseases 0.000 description 1
- 238000003776 cleavage reaction Methods 0.000 description 1
- 238000000942 confocal micrograph Methods 0.000 description 1
- 238000004624 confocal microscopy Methods 0.000 description 1
- 239000003246 corticosteroid Substances 0.000 description 1
- 229960001334 corticosteroids Drugs 0.000 description 1
- IDLFZVILOHSSID-OVLDLUHVSA-N corticotropin Chemical compound C([C@@H](C(=O)N[C@@H](CO)C(=O)N[C@@H](CCSC)C(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](CC=1NC=NC=1)C(=O)N[C@@H](CC=1C=CC=CC=1)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CC=1C2=CC=CC=C2NC=1)C(=O)NCC(=O)N[C@@H](CCCCN)C(=O)N1[C@@H](CCC1)C(=O)N[C@@H](C(C)C)C(=O)NCC(=O)N[C@@H](CCCCN)C(=O)N[C@@H](CCCCN)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N1[C@@H](CCC1)C(=O)N[C@@H](C(C)C)C(=O)N[C@@H](CCCCN)C(=O)N[C@@H](C(C)C)C(=O)N[C@@H](CC=1C=CC(O)=CC=1)C(=O)N1[C@@H](CCC1)C(=O)N[C@@H](CC(N)=O)C(=O)NCC(=O)N[C@@H](C)C(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](CC(O)=O)C(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](CO)C(=O)N[C@@H](C)C(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](C)C(=O)N[C@@H](CC=1C=CC=CC=1)C(=O)N1[C@@H](CCC1)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](CC=1C=CC=CC=1)C(O)=O)NC(=O)[C@@H](N)CO)C1=CC=C(O)C=C1 IDLFZVILOHSSID-OVLDLUHVSA-N 0.000 description 1
- 229960000258 corticotropin Drugs 0.000 description 1
- 150000001886 cortisols Chemical class 0.000 description 1
- 229960004544 cortisone Drugs 0.000 description 1
- 239000013078 crystal Substances 0.000 description 1
- 239000000625 cyclamic acid and its Na and Ca salt Substances 0.000 description 1
- 230000034994 death Effects 0.000 description 1
- 230000007812 deficiency Effects 0.000 description 1
- 230000002950 deficient Effects 0.000 description 1
- 238000006731 degradation reaction Methods 0.000 description 1
- 229910003460 diamond Inorganic materials 0.000 description 1
- 239000010432 diamond Substances 0.000 description 1
- 239000000539 dimer Substances 0.000 description 1
- 238000006471 dimerization reaction Methods 0.000 description 1
- 201000010099 disease Diseases 0.000 description 1
- 239000002270 dispersing agent Substances 0.000 description 1
- 230000004064 dysfunction Effects 0.000 description 1
- 239000012636 effector Substances 0.000 description 1
- 238000001962 electrophoresis Methods 0.000 description 1
- 239000003974 emollient agent Substances 0.000 description 1
- 239000003995 emulsifying agent Substances 0.000 description 1
- 238000005516 engineering process Methods 0.000 description 1
- 239000002031 ethanolic fraction Substances 0.000 description 1
- 238000013401 experimental design Methods 0.000 description 1
- 238000000605 extraction Methods 0.000 description 1
- 238000001914 filtration Methods 0.000 description 1
- GNBHRKFJIUUOQI-UHFFFAOYSA-N fluorescein Chemical compound O1C(=O)C2=CC=CC=C2C21C1=CC=C(O)C=C1OC1=CC(O)=CC=C21 GNBHRKFJIUUOQI-UHFFFAOYSA-N 0.000 description 1
- 238000000799 fluorescence microscopy Methods 0.000 description 1
- 239000013022 formulation composition Substances 0.000 description 1
- CBEHEBUBNAGGKC-UHFFFAOYSA-N ginsenoside Rg1 Natural products CC(=CCCC(C)(OC1OC(CO)C(O)C(O)C1O)C2CCC3(C)C2C(O)CC4C5(C)CCC(O)C(C)(C)C5CC(OC6OC(CO)C(O)C(O)C6O)C34C)C CBEHEBUBNAGGKC-UHFFFAOYSA-N 0.000 description 1
- 229940124750 glucocorticoid receptor agonist Drugs 0.000 description 1
- 208000026352 glucocorticoid resistance Diseases 0.000 description 1
- 150000002304 glucoses Chemical class 0.000 description 1
- 230000005484 gravity Effects 0.000 description 1
- 229940088597 hormone Drugs 0.000 description 1
- 239000005556 hormone Substances 0.000 description 1
- 239000003906 humectant Substances 0.000 description 1
- SIOMFBXUIJKTMF-UHFFFAOYSA-N hypoglauterpenic acid Natural products C1CC(O)C(C)(C)C2=CCC3(C)C4(C)CCC5(C(O)=O)CCC(C)(C)CC5C4=CCC3C21C SIOMFBXUIJKTMF-UHFFFAOYSA-N 0.000 description 1
- 230000004179 hypothalamic–pituitary–adrenal axis Effects 0.000 description 1
- 230000001506 immunosuppresive effect Effects 0.000 description 1
- 229940124589 immunosuppressive drug Drugs 0.000 description 1
- 238000001727 in vivo Methods 0.000 description 1
- 230000001939 inductive effect Effects 0.000 description 1
- 201000006747 infectious mononucleosis Diseases 0.000 description 1
- 102000027411 intracellular receptors Human genes 0.000 description 1
- 108091008582 intracellular receptors Proteins 0.000 description 1
- 238000002372 labelling Methods 0.000 description 1
- 239000008101 lactose Substances 0.000 description 1
- 230000009571 larval growth Effects 0.000 description 1
- 238000000670 ligand binding assay Methods 0.000 description 1
- 230000000670 limiting effect Effects 0.000 description 1
- 238000004811 liquid chromatography Methods 0.000 description 1
- 210000004698 lymphocyte Anatomy 0.000 description 1
- 239000012139 lysis buffer Substances 0.000 description 1
- 239000012528 membrane Substances 0.000 description 1
- 238000000520 microinjection Methods 0.000 description 1
- 210000001616 monocyte Anatomy 0.000 description 1
- 229910000402 monopotassium phosphate Inorganic materials 0.000 description 1
- 235000019796 monopotassium phosphate Nutrition 0.000 description 1
- 230000035772 mutation Effects 0.000 description 1
- 239000002105 nanoparticle Substances 0.000 description 1
- 230000031990 negative regulation of inflammatory response Effects 0.000 description 1
- 230000003448 neutrophilic effect Effects 0.000 description 1
- 229920001220 nitrocellulos Polymers 0.000 description 1
- 231100000957 no side effect Toxicity 0.000 description 1
- 230000036963 noncompetitive effect Effects 0.000 description 1
- 238000010606 normalization Methods 0.000 description 1
- BPAWXSVOAOLSRP-UHFFFAOYSA-N oleanane Natural products CCCCCCCCCCCCCCCC(=O)OC1CCC2(C)C(CCC3(C)C2CC=C4C5CC(C)(C)CCC5(C)C(O)CC34C)C1(C)C BPAWXSVOAOLSRP-UHFFFAOYSA-N 0.000 description 1
- 210000000056 organ Anatomy 0.000 description 1
- 239000003960 organic solvent Substances 0.000 description 1
- 230000008520 organization Effects 0.000 description 1
- 239000003002 pH adjusting agent Substances 0.000 description 1
- 230000036961 partial effect Effects 0.000 description 1
- 230000037361 pathway Effects 0.000 description 1
- 239000008188 pellet Substances 0.000 description 1
- 230000008782 phagocytosis Effects 0.000 description 1
- NBIIXXVUZAFLBC-UHFFFAOYSA-K phosphate Chemical compound [O-]P([O-])([O-])=O NBIIXXVUZAFLBC-UHFFFAOYSA-K 0.000 description 1
- 239000010452 phosphate Substances 0.000 description 1
- 239000000590 phytopharmaceutical Substances 0.000 description 1
- 239000004014 plasticizer Substances 0.000 description 1
- 230000010287 polarization Effects 0.000 description 1
- 229920000136 polysorbate Polymers 0.000 description 1
- GNSKLFRGEWLPPA-UHFFFAOYSA-M potassium dihydrogen phosphate Chemical compound [K+].OP(O)([O-])=O GNSKLFRGEWLPPA-UHFFFAOYSA-M 0.000 description 1
- 239000003755 preservative agent Substances 0.000 description 1
- 230000002335 preservative effect Effects 0.000 description 1
- 239000000047 product Substances 0.000 description 1
- 230000001737 promoting effect Effects 0.000 description 1
- 239000000007 protein synthesis inhibitor Substances 0.000 description 1
- 101150025733 pub2 gene Proteins 0.000 description 1
- 230000005180 public health Effects 0.000 description 1
- 238000001525 receptor binding assay Methods 0.000 description 1
- 230000030541 receptor transactivation Effects 0.000 description 1
- 238000009877 rendering Methods 0.000 description 1
- 238000011160 research Methods 0.000 description 1
- 206010039073 rheumatoid arthritis Diseases 0.000 description 1
- 210000003296 saliva Anatomy 0.000 description 1
- 230000007017 scission Effects 0.000 description 1
- 238000013207 serial dilution Methods 0.000 description 1
- 150000003384 small molecules Chemical class 0.000 description 1
- 238000002415 sodium dodecyl sulfate polyacrylamide gel electrophoresis Methods 0.000 description 1
- 239000007787 solid Substances 0.000 description 1
- 238000000527 sonication Methods 0.000 description 1
- 241000894007 species Species 0.000 description 1
- 239000012798 spherical particle Substances 0.000 description 1
- 239000003381 stabilizer Substances 0.000 description 1
- 238000010186 staining Methods 0.000 description 1
- 229930002534 steroid glycoside Natural products 0.000 description 1
- 239000011550 stock solution Substances 0.000 description 1
- 239000012536 storage buffer Substances 0.000 description 1
- 238000003786 synthesis reaction Methods 0.000 description 1
- 239000012085 test solution Substances 0.000 description 1
- 230000000699 topical effect Effects 0.000 description 1
- 231100000419 toxicity Toxicity 0.000 description 1
- 230000001988 toxicity Effects 0.000 description 1
- 230000002103 transcriptional effect Effects 0.000 description 1
- 238000001890 transfection Methods 0.000 description 1
- 239000012096 transfection reagent Substances 0.000 description 1
- 230000014616 translation Effects 0.000 description 1
- 238000013519 translation Methods 0.000 description 1
- 239000003656 tris buffered saline Substances 0.000 description 1
- 229930182493 triterpene saponin Natural products 0.000 description 1
- 210000003462 vein Anatomy 0.000 description 1
- 108700023427 zebrafish GBA2 Proteins 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
- A61K31/56—Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids
- A61K31/57—Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids substituted in position 17 beta by a chain of two carbon atoms, e.g. pregnane or progesterone
- A61K31/573—Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids substituted in position 17 beta by a chain of two carbon atoms, e.g. pregnane or progesterone substituted in position 21, e.g. cortisone, dexamethasone, prednisone or aldosterone
-
- 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
- A61K31/70—Carbohydrates; Sugars; Derivatives thereof
- A61K31/7042—Compounds having saccharide radicals and heterocyclic rings
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K9/00—Medicinal preparations characterised by special physical form
- A61K9/10—Dispersions; Emulsions
- A61K9/107—Emulsions ; Emulsion preconcentrates; Micelles
-
- 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]
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Chemical & Material Sciences (AREA)
- General Health & Medical Sciences (AREA)
- Veterinary Medicine (AREA)
- Pharmacology & Pharmacy (AREA)
- Medicinal Chemistry (AREA)
- Animal Behavior & Ethology (AREA)
- Public Health (AREA)
- Epidemiology (AREA)
- Dispersion Chemistry (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)
- Molecular Biology (AREA)
- Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
Abstract
The invention relates to a ginsenoside formulation comprising a PPT- type ginsenoside at le ast partly incorporated in a micelle, preferably at least partly incorporated in an outer layer of the micelle, said micelle having a molecular weight of at least 10.000 Da and a ginsenoside for use in a method of prevention or treatment of a side-effect of glucocorticoid (GC) treatment, said method comprising administering a ginsenoside to a subject that is suffering from, or at risk of suffering from, one or more side-effect(s) of GC treatment.
Description
P134912NL00
Title: Combination therapy glucocorticoids and ginsenoside
The invention relates to a ginsenoside for use in the treatment of a side- effect of glucocorticoid (GC) treatment, a combination of a GC and a ginsenoside for use in the treatment of an inflammatory disease, a ginsenoside formulation and a pharmaceutical composition comprising a ginsenoside and a GC. The invention further relates to methods for preparing such a ginsenoside formulation and pharmaceutical composition and a product obtainable by a method for preparing a ginsenoside formulation.
GCs are widely used drugs for treating overactive inflammatory responses and chronic inflammatory diseases [1-8]. GCs such as dexamethasone, prednisolone, and beclomethasone are synthetic analogs of the steroid hormone cortisol, the primary GC hormone in our body which is secreted by the adrenal glands after stress. GCs can diffuse across the cell membrane and bind and activate an intracellular receptor, the glucocorticoid receptor (GR)[1-3]. Upon activation, GR dimers translocate to the nucleus where they function as transcription factors to regulate the transcription of a plethora of target genes.
They modulate gene transcription through several mechanisms. First, they bind to
DNA sequences known as glucocorticoid response elements (GREs) to alter the transcription of responsive genes [1-3]. This mode of action is referred to as transactivation as in most cases it leads to increased gene transcription. Second,
GR can induce changes in gene transcription by interacting with other transcription factors such as NF-kB and AP-1, and thereby modulate their activity.
Since this type of activity is suppressive in most cases, this mechanism is generally called transrepression [4-6]. (GCs are highly effective immunosuppressive and anti-inflammatory drugs. They inhibit the production of pro-inflammatory cytokines and other effector molecules, alter monocyte recirculation, and induce lymphocyte death [5,6].
Classically, the anti-inflammatory function of GCs is based on the transrepression activity of GR, but in recent years it has become clear that transactivation of anti- inflammatory genes plays a role as well. The main issue that limits the clinical use of GCs, especially in patients requiring long-term treatment, is the severity of their side effects, which include diabetes, decreased cortisol levels, reduced growth velocity, wound healing disorders, tissue degeneration, osteoporosis, hypertension, weight gain and muscle weakness [5-11]. Most of the side effects of GCs are caused by the transactivation activity of GR [7-9], although GR’s transrepression activity has been shown to contribute to decreased cortisol levels and osteoporosis.
Another problem accompanying chronic GC treatment is resistance to this therapy. Over time, the treatment may become less effective due to decreased
GR activity and higher doses will often be required for longer periods to elicit anti- inflammatory activity, potentially worsening the side effects [12]. Although the exact mechanisms behind acquired GC resistance are yet to be fully understood, ligand-mediated downregulation (homologous downregulation) of GR is considered the hallmark of acquired GC resistance [12,13].
Therefore, there is an urgent need for developing more selective anti- inflammatory GC drugs, which maintain the anti-inflammatory activity, and do not cause severe side effects and the resistance to GC therapy.
The inventors realized that one or more of the above-mentioned drawbacks may be overcome by administering a ginsenoside to a subject that is suffering from or at risk of suffering from side-effects of GC treatment.
Accordingly, the invention relates to a ginsenoside for use in a method of prevention or treatment of a side-effect of GC treatment, said method comprising administering a ginsenoside to a subject that is suffering from, or at risk of suffering from, one or more side-effect(s) of GC treatment.
The inventors further realized that the side-effects of GC treatment may be effectively prevented or treated, whilst maintaining an anti-inflammatory effect, when a GC and a ginsenoside are administered in a specific molar ratio.
Accordingly, the invention further relates to a pharmaceutical composition comprising a GC, a ginsenoside and a pharmaceutically acceptable carrier, wherein the molar ratio between said GC and said ginsenoside is between about 1:1 and about 1: 2000.
The inventors even further realized that solubility of a ginsenoside and optionally said GC may be significantly improved when incorporating said ginsenoside at least partly in a micelle. Improved solubility is particularly advantageous in case a relatively high concentration of a ginsenoside is desirable,
for example to enable administration of a relatively high concentration of a ginsenoside to a subject in need thereof, or to facilitate the manufacturing of a pharmaceutical composition comprising a ginsenoside.
Accordingly, the invention further relates to a ginsenoside formulation comprising a PPT-type ginsenoside at least partly incorporated in a micelle, preferably at least partly incorporated in an outer layer of the micelle, said micelle having a molecular weight of at least 10.000 Da.
Figure 1. Rgl induces additive anti-inflammatory effects to beclomethasone in the zebrafish tail fin wounding assay. (A) The chemical structures of the synthetic GC beclomethasone and the ginsenoside Rgl, glucose groups were indicated in Blue. (B) Schematic overview of the experimental design of the tail wounding assay. At 72 hpf, zebrafish larvae of the
Tg(mpx:GFEP:*%mpeg1:mCherry»sf09) line were treated with (combinations of) compounds for 6 h (2 h pre-wounding and 4 h post-wounding). At 74 hpf, tail fin wounding was performed at the indicated site (red line). At 78 hpf, the larvae were imaged by fluorescence microscopy, and the area for quantification of the fluorescently labeled neutrophils and macrophages (i.e, the area posterior to the tail vein) is indicated (red box). (C) The effects of vehicle (Veh) and different doses of beclomethasone (Bec), Rgl, and 10 nM beclomethasone in combination with different doses of Rg1 on the number of neutrophils that have migrated to the wounded site. The data shown (means + SEM) are pooled data from three independent experiments, each performed with 15 larvae per group. (D) Numbers of neutrophils and macrophages that have migrated towards the wounded site, after treatment with vehicle, 10 pM beclomethasone, 50 nM Rgl, or the combination of 10 pM beclomethasone with 50 pM Rgl. The data shown (means +
SEM) are pooled from three independent experiments, each performed with 20 larvae per group. (E) Representative fluorescence microscopy images of wounding- induced migration of (GFP-labeled) neutrophils after treatment with vehicle, beclomethasone, dexamethasone, Rg1, or Rgl in combination with beclomethasone.
Scale bar: 100 pm. (F) The effect of beclomethasone, Rg1, and beclomethasone with
Rg1 on the expression of illb and il6. Expression analysis by qPCR was performed using total RNA from non-wounded larvae and wounded larvae, treated with either vehicle, 10 pM beclomethasone, 50 nM Rgl, or 10 nM beclomethasone with 50 pM
Rgl. The relative expression levels in (F) were normalized to those of ppial in zebrafish. Data shown are means + SEM of three independent experiments (each performed in triplicate, with technical duplicates). Statistical significance in C, D and F is indicated: P < 0.05 (*), 0.01 (**), and 0.001 (***) compared to the wounded vehicle group; P < 0.05 #), 0.01 @#), and 0.001 4) for the combination treatment compared to beclomethasone.
Figure S1. (A) Representative fluorescence microscopy images of wounding-induced migration of (GFP-labeled) neutrophils and (mCherry-labeled) macrophages after treatment with vehicle, beclomethasone (10 pM), Rg1 (50 pM), or Rg1 (50 pM) in combination with beclomethasone (10 pM). Scale bar: 100 pm.
The white box indicates the counting area. (B) Representative fluorescence microscopy images of wounding-induced migration of neutrophils after treatment with dexamethasone (10 pM), or Rg1 (50 nM) in combination with dexamethasone (10 pM). Scale bar: 100 nm. (C) Numbers of neutrophils that have migrated towards the wounded site after treatment with vehicle, 10 pM dexamethasone, or the combination of 10 pM dexamethasone and 50 pM Rgl. The data shown (means + SEM) are pooled from three independent experiments, each performed with 15 larvae per group. Statistical significance in (C) is indicated: P < 0.001 (***) compared to the Vehicle group; P < 0.001 G4#%) for the combination treatment compared to dexamethasone.
Figure 2. Rgl binds with GR receptors and trigger an additive anti-inflammatory effects to beclomethasone in the Hela cell line. (A) In vitro determination of relative GR binding affinities using the PolarScreen
Glucocorticoid Receptor (GR) Competitor Assay. Fluorescence polarization levels are plotted, reflecting the binding of a fluorescent ligand that is competed off the receptor by increasing concentrations of the compounds beclomethasone (Bee), dexamethasone (Dex), and Rgl. IC50 levels for each compound are indicated. Data shown are means + SEM of two individual experiments (performed in duplicate). (B) Nuclear translocation levels of GR in HeLa cells upon treatment with beclomethasone and Rgl, determined using immunocytochemistry and confocal microscopy, and the relative translocation levels (means = SEM of three individual experiments (performed in triplicate). (C) Representative confocal microscopy images of nuclear translocation levels of GR in HeLa cells upon treatment with 5 beclomethasone and Rgl, showing GR (green) and DAPI staining (blue). (D) The effect of beclomethasone, Rgl, and beclomethasone with Rg1 on the expression of
IL1B and IL8. Expression analysis was performed by qPCR using total RNA from
HeLa cells treated without, or with TNF-a, and co-treated with vehicle, 0.01 pM beclomethasone, 20 pM Rg1, or 0.01 pM beclomethasone with 20 pM Rgl. The relative expression levels were normalized to those of 18S rRNA in HeLa cells and are shown on a log2 scale. Data shown are means + SEM of two individual experiments (performed in duplicate) in A and of three independent experiments (each performed in triplicate, with technical duplicates) in B and D. Statistical significance in A, andD is indicated: P < 0.05 (*), 0.01 (**), and 0.001 (**¥) compared to the wounded vehicle group; P < 0.05 #), 0.01 @#), and 0.001 G4#%) for the combination treatment compared to beclomethasone.
Figure S2. Rgl reduces both beclomethasone- and dexamethasone-induced side effects. (A) Representative images of the tail fins of zebrafish larvae at 5 dpf, wounded at 2 dpf and treated with Rg1 (50 uM) alone or in combination with beclomethasone (10 uM) or dexamethasone (10 uM). Scale bar: 100 nm. (B) Length of the regenerated tail fin at 5 dpf after tail fin amputation at 2 dpf, and treatment with vehicle (Veh), 10 uM dexamethasone (Dex), or 10 uM dexamethasone with 50 uM Rg1 from 0 to 5 dpf. (C) Relative EGFP intensities of
Tg(9xGCRE-HSV.UI23:EGFP)#2 reporter zebrafish larvae after exposure to the indicated treatments for 24 h at 2 dpf. The data shown in B and C (means + SEM) are pooled from three independent experiments, each performed with 15 larvae per group. Statistical significance in (C) is indicated: P < 0.001 (***) compared to the vehicle group; P < 0.001 G4#) for the combination treatment compared to dexamethasone.
Figure 3. Rgl co-treatment strongly reduces beclomethasone- induced side effects. (A) Length of the regenerated tail fin at 5 dpf after tail fin wounding at 2 dpf, and treatment with vehicle (Veh), 10 uM beclomethasone (Bec), 50 uM Rg1, or 10 uM beclomethasone with 50 uM Rg1 from 0 to 5 dpf. The data shown (means + SEM) are pooled from three independent experiments, each performed with 15 larvae per group. (B) Length of larvae at 5 dpf, after 5-day treatment with vehicle, beclomethasone, Rgl, or beclomethasone with Rgl. The data shown (means + SEM) are pooled from three independent experiments, each performed with 15 larvae per group. (C, D) Whole body glucose levels, determined using a colorimetric assay on samples from 5 dpf larvae treated from 0 to 4 dpf with vehicle, 10 uM beclomethasone, 50 uM Rg1, or 10 uM beclomethasone with 50 uM Rg1 from 0 to 5 dpf (in C), and with different beclomethasone doses (0.1, 1, 5, 10 and 15 nM) alone and in combination with 50 pM Rg1 (in D). Data shown are means + SEM of three independent experiments (each performed in triplicate, with technical duplicates). (E, F) Whole body cortisol levels, determined using ELISA on samples from 5 dpf larvae treated from 0 to 4 dpf with vehicle, 10 uM beclomethasone, 50 uM Rgl, or 10 uM beclomethasone with 50 uM Rg1 (in E), and with different beclomethasone doses (0.1, 1, 5, 10 and 15 nM) alone and in combination with 50 pM Rg1 (in F). ((3) Relative EGFP intensities of Tg(9xGCRE-
HSV.UI23:EGFP)22 reporter zebrafish larvae (3 dpf) after exposure to the indicated treatments for 24 h (starting at 2 dpf). The data shown (means + SEM) are pooled from three independent experiments, each performed with 15 larvae per group. (H) Representative fluorescence microscopy images of Tg(9xGCRE-
HSV. UI23: EGFP) #20 reporter zebrafish larvae (3 dpf) after exposure to the indicated treatments for 24 h (starting at 2 dpf). Scale bar: 100 nm. (I) The fkbp5 and pck1 mRNA levels, studied by qPCR using total RNA from 3 dpf larvae without wounding or with wounding after treatment with vehicle, beclomethasone,
Rg1, or beclomethasone with Rgl. The treatment lasted 6 h (2 h pre- and 4 h post- wounding). The relative expression values were normalized to those of ppial and shown on a log2 scale. The data shown are means + SEM of three independent experiments, each performed in triplicate with technical duplicates. In graphs
A B,C,E,G, and I, statistical significance is indicated: P < 0.05 (¥), 0.01 (*%), and 0.001 (***) compared to the vehicle group, and wounded vehicle group in (I); P <
0.05 @), 0.01 GA), and 0.001 GAH) for the combination treatment compared to beclomethasone. In graphs D, F statistical significance indicated: P < 0.05 (¥), 0.01 (**), and 0.001 (***) compared to the Rgl group.
Figure 4. GC sensitivity is not reduced after long-term beclomethasone and Rgl co-treatment. The mRNA levels of fkbp5 (A), pck1 (B), nfkbiaa (C), and gr (D) were studied by qPCR using total RNA from 5 dpf larvae without wounding, after short- or long-term treatment with vehicle, beclomethasone (10 uM), Rg1(50 uM), or beclomethasone(10 uM) with Rg1(50 uM).
The short-term treatment lasted 6 h. In the long-term treatment, larvae were treated for 5 days (from 0 to 5 dpf). The relative expression values were normalized to those of ppial and shown on a log2 scale. The data shown are means + SEM of three independent experiments, each performed in triplicate with technical duplicates. Statistical significance is indicated: P < 0.001 (***) compared to the vehicle group; P < 0.001 #48) for the combination treatment compared to treatment with beclomethasone alone; P < 0.05 (+), 0.01 (++), and 0.001 (+++) for the long-term treatment compared to the corresponding short-term treatment.
Figure S4. Rgl restores the GC sensitivity in HeLa cells that is reduced by long-term high-dose GC treatment. (A, B) The effects of Rgl, beclomethasone as an individual treatment or in combination on the relative IL1B mRNA level (determined by qPCR) at different time points after TNF-a treatment during short-term treatment (in A) or long-term treatment (in B). The, control group reflects a vehicle-treated group in the absence of TNF-a. (C,D) The effects of the combination treatment of Rg1 with dexamethasone on the expression of IL1B (in C) and NFKBIA (in D) after short- or long-term treatment. In A-D, the short- term treatment consisted of 6 h of compound co-treatment with TNF-a, and in the long-term treatment, cells were treated with the compounds for 30 h, consisting of 24 h plus 6 h of compound co-treatment with TNF-a. The relative expression values were normalized to those of 18S rRNA and shown on a log2 scale. The data shown are means = SEM of three independent experiments, performed in triplicate with technical duplicates. Statistical significance is indicated: P < 0.05 (*), 0.01 (**), and 0.001 (***) compared to the corresponding vehicle group; P < 0.05 @#), 0.01 ##), and
0.001 G5#) for the combination treatment compared to beclomethasone or dexamethasone alone; P < 0.05 (+), 0.01 (++), and 0.001 (+++) for the long-term treatment compared to the corresponding short-term treatment.
Figure 5. Rgl restores the GC sensitivity in HeLa cells that is reduced by long-term high-dose GC treatment. (A-G) Relative mRNA levels in
HeLa cells for IL1B (A), MMP9 (B), IL8 (C), FKBP5 MD), NFKBIA (E), GILZ (F),
SGK1 (G), determined by qPCR. In A, Hela cells were treated (short- or long-term) with increasing doses of beclomethasone (0.01, 0.1 and 1 pM), Rg1 (20, 100 and 500 pM) or co-treatment of Rg1 (20, 100 and 500 pM) with beclomethasone (0.01 pM).
In B-G, HeLa cells were treated with vehicle, beclomethasone (1 pM), Rg1 (20 pM), or beclomethasone with Rg1, in the absence and presence of TNF-a. The short-term treatment consisted of 6 h of compound co-treatment with TNF-q, and in the long- term treatment, cells were treated with the compounds for 30 h, consisting of 24 h plus 6 h of compound co-treatment with TNF-a. The relative expression values were normalized to those of 18S rRNA and shown on a log2 scale. The data shown in (A-G) are means + SEM of three independent experiments, each performed in triplicate with technical duplicates. Statistical significance is indicated: P < 0.05 (*), 0.01 (*%), and 0.001 (***) compared to the corresponding vehicle group; P < 0.05 #), 0.01 G#), and 0.001 G4#) for the combination treatment compared to beclomethasone; P < 0.05 (+), 0.01 (++), and 0.001 (+++) for the long-term treatment compared to the corresponding short-term treatment.
Figure S5. Representative gels of western blots showing that Rgl inhibits the effect of beclomethasone-induced downregulation of GR in
HeLa cells. (A) Western blots for GR (and U-actin) were obtained using (duplicate) protein samples from Hela cells after short-term treatment (6 h) with the indicated compounds. (B) Western blots for GR (and B-actin) were obtained using (duplicate) protein samples from HeLa cells after long-term treatment (24 h) with the indicated compounds. (C) Western blots for GR (and B-actin) were obtained using protein samples from HeLa cells after long-term treatment (24 h) with the indicated compounds, in the absence and presence of cycloheximide (5 pg/ml).
Imaged was used to determine the integrated intensity of the GR and B-actin protein bands.
Figure 6. Rgl inhibits the beclomethasone-induced downregulation of GR in HeLa cells. (A) The effects of beclomethasone (1 pM) and/or Rgl (20 pM) treatment on GR mRNA levels, determined by qPCR, after short- and long-term treatment, in the absence and presence of actinomycin-D (1 ng/ml). The relative expression values were normalized to those of 18S rRNA. Data shown are means + SEM of three independent experiments (each performed in triplicate with technical duplicates) and are plotted on a log2 scale. (B) The effects of beclomethasone and/or Rgl treatment on GR protein levels, determined using western blotting, after short- and long-term treatment (6 and 24 h respectively). (C) The effects of beclomethasone and/or Rgl treatment on GR protein levels in the absence and presence of cycloheximide (5 pg/ml), after long-term treatment. Data shown have been normalized to the level of B-actin and are means + SEM of four (in
A) independent experiments, each performed in duplicate, or six (in B) independent experiments, each performed as a single measurement. Statistical significance is indicated: P < 0.05 (*), 0.01 (*%), and 0.001 (***) compared to the corresponding vehicle group: P < 0.05 #), 0.01 FH), and 0.001 644) for the combination treatment compared to beclomethasone alone; P < 0.05 (+), 0.01 (++), and 0.001 (+++) for the long-term treatment compared to the corresponding short-term treatment.
Figure 7. The anti-inflammatory action of monoglycosylated ginsenosides requires Gr function, but not glucosylceramidase beta 2 (Gba2). A. Structures of the compounds used for treatment: Beclomethasone (Bec),
Protopanaxatriol (PPT), F1, Rh1, and Rgl. Glucose (Glc) groups conjugated to the steroid backbone are indicated in blue. B. Schematic overview of the experimental approach. Zebrafish larvae from the Tg(mpx:GFP!!!4#mpeg1:mCherry-FumsF%3) line were subjected to tail wounding at 74 hpf. Chemical compound treatments started at 2 h before wounding and were continued for 4 h after wounding. At this time point, the number of neutrophils that had migrated to the wounded area (indicated by the red box) was determined. C. The number of migrated neutrophils and macrophages upon compound treatment at 4 hours after wounding. D. The number of migrated neutrophils upon compound treatment in the absence and presence of the Gba2-inhibitor MZ31 at 4 hours after wounding. E. Relative mRNA levels for il1b, il6, il8, mmp9, and mmp13, determined by qPCR, before and after wounding and upon compound treatment. Data shown are means + SEM and represent the average of data pooled from three individual experiments (n=60) in C and D, or the average of three individual experiments (performed in triplicate) in E. Statistical significance was determined by one- or two-way ANOVA and Tukey’s post hoc test.
Significant difference from the wounded Veh groups (in CE), or from the corresponding Veh group (Veh/Veh or Veh/MZ31; in D) is indicated by *** (P<0.001). The difference from the Veh group treated with the same compound (hatched versus non-Hatehed bars) is indicated by ### (P<0.001). Significant difference from the PPT group (in E) is indicated by $$$ (P<0.001) and the difference from the F1 group (in E) is indicated by &&& (P<0.001).
Figure S7. Fish Embryo Acute Toxicity Test (FET) for studied ginsenosides and beclomethasone (A, B). Embryos were exposed to different concentrations of those compounds to assess their toxicity. A negative control (nC), solvent control (sC), and positive control (pC) group were included in the experiment. A. Hatching rate (%) at 48 and 72 hpf. B. Survival rate (%) at 96 and 120 hpf. C. The number of migrated neutrophils upon compound treatment in wild type (gr**) and Gr deficient (gr) individuals upon treatment with Bee, PPT, F1,
Rhl, or Rgl. Data shown are means + SEM and represent the average of data pooled from two individual experiments (n=100) in A and B, and from three individual experiments (n=60) in C. Statistical significance was determined by one- or two-way ANOVA and Tukey's post hoc test. A significant difference from the
Veh (NC) group (same time point) in (A,B), or from the corresponding Veh group (gr** or gr”; C) is indicated by *** (P<0.001) and the difference from the gr** group treated with the same compound (hatched versus non-hatched bars; C) is indicated by ### (P<0.001).
Figure 8. Ginsenosides show a strong reduction in side effects compared to beclomethasone. A. Whole-body glucose levels, determined by colorimetric assay, in 5 dpf zebrafish larvae treated (from 2 hpf until 5 dpf) with beclomethasone or ginsenosides PPT, F1, Rh1 or Rgl from 2 hpf. B. Whole-body glucose levels in 5 dpf larvae, upon wounding at 2 dpf, and compound treatment with or without MZ31 from 2 hpf until 5 dpf. C. Whole-body cortisol levels, determined by ELISA, in 5 dpf zebrafish larvae, treated with beclomethasone or ginsenosides PPD, F2, or Rb1 (from 2 hpf until 4 dpf). D. Whole-body cortisol levels in 5 dpf larvae, upon wounding at 2 dpf, and compound treatment with or without
MZ31 (from 2 hpf until 4dpf). E. Body length of zebrafish larvae at 5 dpf upon compound treatment from 2 hpf until 5 dpf. F. Length of the regenerated tail fin of 5 dpf larvae, wounded at 2 dpf, upon compound treatment with or without MZ31 (from 2 hpf until 5 dpf). G. Representative fluorescence microscopy images of 3 dpf embryos from experimental groups presented in H. Scale bar: 200 pm. H. Relative
GFP fluorescence levels after 24 h compound treatment in 3 dpf embryos from the
Tg(9x GCRE-HSV.UI23: EGFP) line, which is a reporter line for the transactivation activity of Gr. I. Relative mRNA levels for fhbp 5, determined by qPCR, before and after wounding and upon compound treatment. Data shown in A, B, C, D and I are means + SEM and averages of three individual experiments (performed in triplicate). Data shown in E, F,and H are means + SEM and represent the average of data pooled from three individual experiments (n=60). Statistical significance was determined by one- or two-way ANOVA and Tukey’s post hoe test. Significant difference from the Veh group (in A, C, E, H) or the corresponding Veh group (Veh/Veh or Veh/MZ31; in B, D, F) is indicated by * (P<0.05), ** (P<0.01), *** (P<0.001), the difference from the group treated with the same compound (hatched versus non-hatched bars; in B, D, F) is indicated by # (P<0.05), ## (P<0.01), ## (P<0.001)
Figure S8A. Representative brightfield microscopy images of tail fins of 5 dpf larvae from experimental groups presented in Fig.2F.
Arrowheads indicate the site of amputation and any tissue observed to right of this point is regenerated. Scale bar: 100 pm. B. Relative mRNA levels for pck1 and nfkbiaa determined by qPCR, before and after wounding and upon compound treatment. Data shown are means + SEM and averages of three individual experiments (performed in triplicate). Statistical significance was determined by two-way ANOVA and Tukey’s post hoc test. Significant difference from the Veh group is indicated by *** (P<0.001).
Figure 9. Anti-inflammatory effects and putative side effects of ginsenoside Re on zebrafish after wound induction by tail amputation.
A: . effect of Re and GC (prednisone) on the number on neutrophils at the amputation site; B: effect of Re and GC (prednisone) on the number of macrophages at the amputation site; C: effect of Re and GC (prednisone) on whole body glucose level; D: effect of Re and GC (prednisone) on whole body cortisol and E: effect of Re and GC (prednisone) on regenerative tissue. Veh: vehicle.
Figure 10 A. Appearance of solutions of ginsenoside in water after micelle formation. From left to right: 10 mg/ml pure Re solved in ethanol prior to the micelle formatting procedure; 10 mg/mL pure Re solved in H20 after the procedure of micelle formation; 150 mg/mL Re/Rgl enriched ginseng extract in ethanol obtained by purification and decolorization on a D101 and D941 resin respectively; 150 mg/mL Re/Rgl enriched ginseng extract after micelle formation.
B: microscopic view (400x) of crystals of Re formed by solving the Re film in H20 during the procedure of micelle formation
Figure 11: Size exclusion chromatography to determine the molecular weight and composition of the micelles. 1 mL of an aqueous solution containing Re/Rgl enriched ginseng extract and dexamethasone was loaded on a 17 mL Sephadex G50 size exclusion chromatography column after micelle formation and eluted with H20 at a rate of 0.4 mL/min. Fractions of 2 mL were collected. The column was calibrated using blue dextran (MW 2.000.000 Da) and erythrosine (MW 880 Da) as molecular weight markers (see arrows for peak elution of the markers). The presence of and concentration of ginsenosides Re, Rgl and dexamethasone was determined and calculated by means of HPLC.
Figure 12: HPLC chromatograms of fractions relevant for or obtained during size exclusion chromatography. A: HPLC profile and peak location of pure dexamethasone at 203 nm (upper panel) and 241 nm (lower panel);
B: 203 nm HPLC chromatogram of fraction 15 eluted at 30 mL showing the presence of Rgl and Re as the major peaks; C: 241 nm HPLC chromatogram of fraction 15 eluted at 30 mL showing the presence of dexamethasone
The invention relates to a ginsenoside formulation comprising a PPT-type ginsenoside at least partly incorporated in a micelle, preferably at least partly incorporated in an outer layer of the micelle, said micelle having a molecular weight of at least 10.000 Da, preferably a molecular weight of at most 100.000 Da.
In a preferred aspect, said ginsenoside is a PPT-type ginsenoside, preferably selected from Rgl, Re and a combination thereof.
The invention further relates to a method for preparing a ginsenoside formulation according to the invention, said method comprising (a) providing a ginsenoside extract from a plant or plant part of the genus Panax, preferably Panax ginseng; (b) contacting said ginsenoside extract with an aqueous solvent mixture, preferably wherein said aqueous solvent mixture comprises a mixture of water and a non- aqueous solvent, preferably an alcoholic solvent, more preferably selected from methanol, ethanol, propanol and isopropanol, to obtain a ginsenoside mixture; (c) at least partly removing said aqueous solvent mixture from said ginsenoside mixture; and optionally (d) reconstituting the ginsenoside mixture obtained in step c) in water to obtain a ginsenoside formulation.
In preferred methods, the method comprises subjecting said ginsenoside extract to a step of column chromatography using a non-polar resin as a stationary phase, preferably a D101 macroporous resin and/or subjecting said ginsenoside extract to a step of decolorization, preferably using a D941 macroporous resin.
The invention further relates to a ginsenoside formulation obtainable by a method according to the invention, preferably comprising a PPT-type ginsenoside at least partly incorporated in a micelle, preferably at least partly incorporated in an outer layer of the micelle, said micelle having a molecular weight of at least 10.000 Da and/or a molecular weight of at most 100.000 Da.
In a preferred ginsenoside formulation obtainable by a method according to the invention, said ginsenoside is a PPT-type ginsenoside, preferably selected from Rgl, Re and a combination thereof.
Further, the invention relates to a ginsenoside for use in a method of prevention or treatment of a side-effect of GC treatment, said method comprising administering a ginsenoside to a subject that is suffering from, or at risk of suffering from, one or more side-effect(s) of GC treatment.
Preferably, said side-effect is selected from GC-induced diabetes, GC- induced decreased cortisol levels, GC-induced reduced growth velocity, GC-induced wound healing disorders, GC-induced tissue degeneration, GC-induced osteoporosis, GC-induced hypertension, GC-induced weight gain and GC-induced muscle weakness.
The invention further relates to ginsenoside formulations according to the invention for use according to the invention.
The invention further relates to a combination of a GC, preferably selected from the group of beclomethasone and dexamethasone, and a ginsenoside for use in a method of prevention or treatment of an inflammatory disease, wherein said method comprises administering a GC and a ginsenoside in a molar ratio of between about 1:1 and about 1: 2000 to a subject in need thereof, wherein said molar ratio is between about 1:4 and about 1:1000, more preferably between about 1:8 and about 1:20.
In preferred embodiments, the invention relates to the combination for use according to the invention, wherein said GC is administered in a therapeutically ineffective amount, preferably at a dose of at most 0.07 mg/kg/day, preferably at most 0.007 mg/kg/day and/or wherein said ginsenoside is administered in a therapeutic ineffective amount, preferably at a dose of at most 5.5 mg/kg/day, more preferably at most 0.05 mg/kg/day.
In preferred embodiments, the invention relates to the combination for use according to the invention, wherein said inflammatory disease is selected from asthma, allergic rhinitis, hay fever, urticaria, atopic eczema, chronic obstructive pulmonary disease, inflammation of the joints, muscles and tendons, lupus, inflammatory bowel disease, such as Crohn's disease and ulcerative colitis, giant cell arteritis and polymyalgia rheumatica, and multiple sclerosis.
Preferably, the invention relates to combinations for use according to any one of claims 8 to 13, wherein said ginsenoside is at least partly incorporated into a micelle, preferably at least partly incorporated in an outer layer of a micelle, said micelle having a molecular weight of at least 10.000 Da and/or a molecular weight of at most 100.000 Da. Preferably, said ginsenoside is a PPT-type ginsenoside, preferably selected from Rg1, Re and a combination thereof.
The invention further relates to a pharmaceutical composition comprising a GC, a ginsenoside, preferably wherein the ginsenoside is a protopanaxatriol (PPT)-type ginsenoside, more preferably selected from Rgl and Re and a combination thereof and a pharmaceutically acceptable carrier, wherein the molar ratio between said GC and said ginsenoside is between about 1:1 and about 1: 2000, preferably between about 1:4 and about 1:1000, more preferably between 1:8 and 1:20.
Preferably, in the pharmaceutical composition according to the invention, said ginsenoside is at least partly incorporated into a micelle, preferably at least partly incorporated in an outer layer of a micelle, said micelle having a molecular weight of at least 10.000 Da and/or at most 100.000 Da.
In an aspect, the invention relates to a pharmaceutical composition according to the invention, wherein said pharmaceutical composition is formulated as acream, a lotion, a balm, a hydrogel, an ointment, a foam, a gel, a spray, a tablet, a capsule, a lozenge, a topical solution, a topical suspension, an aerosol and a syrup. Most preferably, the pharmaceutical composition is formulated as a hydrogel.
The invention further relates to the pharmaceutical composition for use in a method of prevention or treatment of an inflammatory disease, preferably wherein said inflammatory disease is selected from asthma, allergic rhinitis, hay fever, urticaria, atopic eczema, chronic obstructive pulmonary disease, inflammation of the joints, muscles and tendons, lupus, inflammatory bowel disease, such as Crohn’s disease and ulcerative colitis, giant cell arteritis and polymyalgia rheumatica, and multiple sclerosis.
The term “or” as used herein is defined as “and/or” unless specified otherwise.
The term “a” or “ an” as used herein is defined as “at least one” unless specified otherwise.
The term “substantial(ly)” or “essential(ly)” is generally used herein to indicate that it has the general character or function of that which is specified.
When referring to a quantifiable feature, these terms are in particular used to indicate that it is for at least 75 %, more in particular at least 90 %, even more in particular at least 95 % of the indicated feature.
In the context of the present application, the term “about” means generally a deviation of 15% or less from the given value, in particular a deviation of 10% or less, more in particular a deviation of 5% or less.
When referring to a noun in the singular, the plural is meant to be included, unless it follows from the context that it should refer to the singular only. “Ginsenosides” are a class of steroid glycosides and triterpene saponins found in the plant genus Panax. Ginsenosides are commonly referred to by the name “Rx”, wherein the x describes the chromatographic polarity in alphabetical order, although there are also ginsenosides described using a different name, such as ginsenoside F1. Thus, Ra is one of the least polar ginsenoside described thus far and Rb is more polar than Ra. (Ginsenosides are characterized by a dammarane scaffold — also known under the systemic name (1R,3aR,3bR,5a5,9a5,9bR,11aR)-1-[(2R)-6-
Methylheptan-2-yl]-3a,3b,6,6,9a-pentamethylhexadecahydro-1H- cyclopentala|lphenanthrene — typically with a sugar moiety attached thereto, although ginsenosides lacking a sugar moiety have also been described, such as ginsenoside “aglycon-PPT”. Ginsenosides are usually divided into two major subclasses, i.e. 20(S)-protopanaxadiol (PPD) ginsenosides (formula I) and 20(S)- protopanaxatriol (PPT) ginsenosides (formula IT). PPD and PPT-type ginsenosides are distinguished from one another in the position of the sugar moiety to the dammarane scaffold. In PTT-type ginsenosides, the sugar moiety is attached to the 6-position of the scaffold, whereas in PPD-type ginsenosides the sugar moiety is attached to the 3-position of the scaffold.
RO § - OH 7
NN N
N
Me Ve
OR,
RO 4
Formula (1) Formula (II)
Examples of protopanaxadiol-type ginsenosides include Rb1, Rb2, Rb3,
Re, Rd, Rg3, Rh2 and Rsl. Examples of protopanaxatriol-type ginsenosides include
Re, Rf, Rg1, Rg2 and Rhl. Further, some rare ginsenosides are described, such as the ocotillol saponin F11 (24-R-pseudoginsenoside) and the pentaeyclic oleanane saponin Ro (3,28-0-bisdesmoside). “Glucocorticoids” (GC) refer herein to a class of steroid hormones that have an ability to bind to the glucocorticoid receptor. GCs are commonly prescribed in the treatment of (chronic) inflammatory diseases, including eczema, asthma and rheumatoid arthritis. Cortisol, also referred to as hydrocortisone, is an endogenous
GC. Synthetic analogues of cortisol are also described, which typically have one or more changed properties compared to cortisol. Examples of synthetic GCs include dexamethasone, betamethasone, prednisolone, methylprednisolone triamcinolone, deflazacort, fludrocortisone acetate, fludrocortisone acetate, aldosterone, beclomethasone or a prodrug thereof.
With the term prodrug as used herein is meant an analogue of a GC that is converted to a biologically active GC in vivo. Examples of prodrugs of GCs include cortisone and prednisone.
The term “micelle” refers to a substantially spherical particle, comprising an outer layer comprising one or more amphiphilic molecules and an inner cavity.
The outer layer typically comprises a hydrophilic outer surface and a hydrophobic inner centre. Typically, in an aqueous solution, the hydrophilic part of the micelle is in contact with an aqueous medium, whereas the hydrophobic part of the micelle is shielded from the aqueous medium. Said inner cavity is usually formed by the outer layer, and may encompass one or more small molecules, such as a therapeutic molecule.
In the context of the present application, the hydrophilic part of the micelle is usually formed by the hydrophilic part of the ginsenosides, typically the sugar moiety, whereas the hydrophobic part of the micelle is usually formed by the hydrophobic part of the ginsenoside, typically the dammarane moiety of the ginsenoside.
With the term “therapeutically effective amount” as used herein, is meant an amount, when administered during a period of time necessary, that is effective to achieve an anti-inflammatory effect in a subject in need thereof. With the term “therapeutically ineffective amount” is meant any amount that is lower than a therapeutic effective amount, and which essentially does not exhibit a significant anti-inflammatory effect in a subject in need thereof, when administered for a sufficient period of time and in absence of another anti- inflammatory compound.
Ginsenoside for use in the treatment of a side-effect of GC treatment
Although GCs are effective anti-inflammatory drugs, their long-term use is associated with a number of drawbacks.
For example, long-term use of GC’s is associated with decreased glucocorticoid receptor (GR) activity and thus increased GC resistance.
Consequently, in order to maintain effectiveness of GCs, the dosage is typically increased.
However, in particular long-term use of GCs is associated with the occurrence of severe side-effects, including diabetes, decreased cortisol levels, reduced growth velocity, wound healing disorders, tissue degeneration, osteoporosis, hypertension, weight gain and muscle weakness.
The occurrence and severity of these side-effects is further usually enhanced at increased dosage, which is often required to overcome the decreased glucocorticoid receptor activity and maintain efficacy.
The inventors surprisingly realized that a ginsenoside can effectively prevent or alleviate the side-effects of GC treatment and can furthermore prevent
GC resistance. This would significantly increase the therapeutic options for the treatment of anti-inflammatory diseases.
Without wishing to be bound by any theory, the inventors believe that a ginsenoside, in particular Rgl, acts as a competitive antagonist of the glucocorticoid receptor (see e.g. Examples and Figure 3D, F). In more detail, the inventors envisage that selective binding of a ginsenoside to the glucocorticoid receptor induces transrepression but not transactivation, the latter being linked to the occurrence of side-effects.
Accordingly, in an aspect the invention relates to a ginsenoside for use in a method of prevention or treatment of a side-effect of GC treatment, said method comprising administering a ginsenoside to a subject that is suffering from, or at risk of suffering from, one or more side-effect(s) of GC treatment.
Ginsenoside
The ginsenoside for use according to the invention may in principle be any ginsenoside that exerts an anti-inflammatory effect and that is capable of mediating the side-effects of GC treatment.
Said ginsenoside is preferably a 20(S)-protopanaxadiol (PPD)-type ginsenoside or a 20(S)-protopaxanatriol (PPT)-type ginsenoside, more preferably a
PPT-type ginsenoside.
Preferably, said ginsenoside is selected from Rg1, Re, Rf, Rg2, F1, aglycon-PPT and Rh1 or a combination thereof, more preferably said at least one ginsenoside is selected from Rgl and Re or a combination thereof.
Rgl is a PPT-type ginsenoside with molecular formula C4zH72014 and a molecular weight of 801 g/mol. It is also known under the chemical names (3B,6a,128)-3,12-Dihydroxydammar-24-ene-6,20-diyl bis-8-D-glucopyranoside,
Ginsenoside A2, Ginsenoside gl, Panaxoside A, Panaxoside Rg1, Sanchinoside C1 and Sanchinoside Rgl. The molecular structure of Rgl is depicted in Figure 1A.
Further, Re is another PPT-type ginsenoside and has molecular formula
CasHszO1s and a molecular weight of 947.15 g/mol. It is also known under the chemical names (38,64, 128)-20-(B-D-Glucopyranosyloxy)-3,12-dihydroxydammar- 24-en-6-yl 2-0-(6-deoxy-a-L-mannopyranosyl)-8-D-glucopyranoside,
Chikusetsusaponin IVe, Ginsenoside B2, NSC 308877, Panaxoside Re and
Sanchinoside Re.
Preferably, said ginsenoside comprises Rg1, Re, or a combination of Rgl and Re, more preferably, if said ginsenoside comprises a combination of Re and
Rgl, said Rgl and Re are present in a wt. ratio of between about 1:2 and about 2:1, more preferably between about 1:1.5 and about 1.5:1.
Said ginsenoside may be obtained from a natural source, or may be synthetically prepared.
For example, said ginsenoside may be obtained from the plant genus
Panax. Preferably, said ginsenoside may be obtained from one or more of the species Panax bipinnatifidus, Panax, elegantior, P. ginseng, P. japonicus, P. major,
P. notoginseng, P. ometensis, P. pseudoginseng, P. quinquefolius, P.sikkimensis, P. sinensis, P. stipuleanatus, P. trifolius, P.vietnamensis, P. wangianus and P. zingiberensis.
Most preferably, said ginsenoside may be obtained from P. quinquefolius (also referred to as American ginseng), P. japonicus (also referred to as Japanese ginseng), P. ginseng (also referred to as Korean ginseng) or P. notoginseng (also referred to as South China ginseng).
Preferably, said ginsenoside may be obtained from one or more of the roots, the stems, the fruits, the rhizomes, the flowers or the leaves of a plant of the genus Panax, more preferably from one or more of the roots, stems, fruits, rhizomes, flowers or leaves of P. ginseng, P. japonicus, P. notoginseng or P. quinquefolius.
In a plant of the genus Panax, the content of ginsenosides is typically in the range of about 1 to about 10 wt.%, based on the dry weight of the plant, such as between about 2 and about 8 wt.%, between about 3 and about 6 wt.%, in particular about 4 wt.%, based on the dry weight of the plant.
Said ginsenoside may be isolated from a plant of the genus Panax, using any suitable method known in the art. For example, said ginsenoside may be obtained by drying a part, such as a root of a plant of the genus Panax, followed by extraction of the dried plant or plant part with a suitable solvent. Methods for preparing a ginseng extract is described in Chinese pharmacopeia edition 2015 (English language), volume I, page 524-526.
Optionally, said plant part, such as a root of a plant of Panax is steamed at 100 °C for at least two hours prior to drying. An extract obtained using this method is typically referred to as “red ginseng”.
Ginseng extracts are also commercially available, for example G115 from
P. ginseng (Pharmaton SA, Switzerland) and NAGE from P. quinquefolius (Canadian Phytopharmaceuticals Corporation, Canada).
Preferably, the content of ginsenosides in a ginseng extract is at least 1 wt.%, more preferably at least 2 wt.%, at least 3 wt.%, at least 4 wt.%. Typically, the content of ginsenosides In a ginseng extract is at most 99 wt.%, more preferably at most 98 wt.%, even more preferably at most 95 wt.%
A preferred range of ginsenosides in a ginseng extract is between about 4 and about 50 wt.%, more preferably between about 8 wt.% and about 20 wt.%, based on the dry weight of the ginseng extract.
In an embodiment, said ginsenoside is provided as a ginseng extract.
Said ginseng extract optionally contains one or more components other than ginsenosides, such as ginseng proteins and ginseng carbohydrates.
Alternatively, said ginsenoside is provided in isolated form, i.e. essentially separated from components naturally present in a plant from the genus
Panax. Isolated ginsenosides are commercially available, for example Ginsenoside-
Rb1 (CAS 41753-43-9), Ginsenoside-Rh1 (CAS 63223-86-9), Ginsenoside Re (CAS 52286-59-6) and Ginsenoside Rgl (CAS 22427-39-0) are commercially available from Sigma Aldrich.
An isolated ginsenoside may also be optionally provided in synthetic form if a chemical synthetic pathway is known. For example, the chemical synthesis of ginsenoside Rg3 is described by Anufriev et al, Carbohydr Res. 1997; 304(2):179- 182.
Preferably, the purity of said ginsenoside is at least 90 wt.%. More preferably, the purity of said ginsenoside is at least 92 wt.%, at least 94 wt.%, at least 96 wt.%, at least 98 wt.%, at least 99 wt.%, such as 100 wt.%.
Micelle
The inventors further recognized that the solubility of ginsenosides in water was a limiting factor in the medical use of ginsenosides in the treatment or prevention of side effects caused by GC treatment or in the preparation of a pharmaceutical composition according to the invention. Further, ginsenosides notoriously suffer from degradation in the gastro-intestinal tract rendering them ineffective and hampering their systemic use.
The inventors therefore developed a novel formulation of ginsenosides in which one or more of the abovementioned drawbacks have been overcome.
Accordingly, said ginsenoside for use according to the invention is preferably at least partly incorporated into a micelle, more preferably in an outer layer thereof.
The inventors realized that when ginsenosides were formulated as a micelle, solubility in water could be markedly increased and stability in the intestinal tract was improved compared to a regular ginsenoside formulation, wherein the ginsenoside was not incorporated in (an outer layer of) a micelle.
Such increased water solubility is beneficial as it allows to obtain a sufficiently high concentration of a ginsenoside in an aqueous medium, typically required to counteract one or more side-effects of GC treatment.
Further, improved stability in the intestinal tract advantageously improves efficacy of systemic use of said ginsenoside.
A micelle having at least one ginsenoside incorporated therein, preferably at least partly incorporated in an outer layer of the micelle, may in principle be prepared using any suitable method known in the art, depending on the type of ginsenoside to be incorporated into the micelle. For example, for a micelle having one or more PPD-type ginsenosides incorporated therein, said micelle may be prepared by slowly evaporating a solution of a PPD-type ginsenoside in an organic or aqueous solvent (e.g. ethanol or a mixture of ethanol and water) under reduced pressure, followed by slowly resolving the obtained film of ginsenosides in water to obtain a micelle having one or more PPD-type ginsenosides incorporated therein.
However, the inventors surprisingly realized that when this method was applied using the PPT-type ginsenoside Re, crystallization of Re was observed, but no micelle formation occurred. This is demonstrated in Example 4.
The inventors surprisingly realized that a micelle having at least one
PPT-type ginsenoside incorporated therein could however be prepared using a
(crude) ginsenoside extract comprising at least one PPT-type ginsenoside instead of an isolated PPT-type ginsenoside as substrate.
Accordingly, the invention further relates to a ginsenoside formulation comprising a PPT-type ginsenoside at least partly incorporated therein, preferably at least partly incorporated in an outer layer of the micelle, said micelle having a molecular weight of at least 10.000 Da.
Said ginsenoside is as defined herein above in the section “ginsenoside” providing it is a PPT-type ginsenoside.
The micelles have a molecular weight of at least 10.000 Da, preferably at least 20.000 Da, at least 40.000 Da, at least 50.000 Da, at least 60.000 Da, at least 70.000 Da, at least 80.000 Da, at last 90.000 Da, most preferably at least 100.000
Da.
The micelles typically have a molecular weight of at most 100.000.000
Da, preferably at most 10.000.000 Da, most preferably at most 1.000.000 Da.
The micelles preferably have a molecular weight in the range of between about 10.000 and about 100.000.000 Da, more preferably in the range of between about 50.000 and about 10.000.000 Da, even more preferably in the range of between about 80.000 and about 1.000.000 Da.
Said molecular weight may be determined using any suitable method known in the art, for example using size-exclusion chromatography or High-
Performance Liquid Chromatography, in particular using the method as described in Example 4.
Typically, the micelles comprise between about 10 and about 100.000 molecules of one or more ginsenosides, more preferably between about 20 and about 10.000 molecules of one or more ginsenosides, even more preferably between about 50 and about 10.000 molecules of one or more ginsenosides, in particular between about 100 and about 1000 molecules of one or more ginsenosides.
The solubility in an aqueous medium, preferably water, of a ginsenoside formulation according to the invention, or a ginsenoside which is at least partly incorporated in (an outer layer of) a micelle, is preferably at least 0.03 mmol/L, more preferably at least 0.05 mmol/L, at least 0.1 mmol/L, at least 1 mmol/L, at least 5 mmol/L, at least 10 mmol/L, at least 50 mmol/L, at least 0.1 mol/L, at least
0.15 mol/L, at least 0.2 mol/L, at least 0.25 mol/L, most preferably at least 0.3 mol/L.
Typically, the solubility in an aqueous medium, preferably water, of a ginsenoside formulation according to the invention, or a ginsenoside which is at least partly incorporated in (an outer layer of) a micelle is in the range of between about 0.03 mmol/L and about 10 mol/L, preferably between about 0.05 mmol/L and about 5 mol/L, more preferably between about 0.1 mmol/L and about 2 mol/L, between about 1 mmol/L and about 1 mol/L, even more preferably between about 5 mmol/L and about 0.5 mol/L, in particular between about 10 mmol/L and about 0.3 mol/L.
The solubility in an aqueous medium, preferably water, of a ginsenoside formulation according to the invention, or a ginsenoside which is at least partly incorporated in (an outer layer of) a micelle is preferably at least 30 mg/L, more preferably at least 50 mg/L, at least 100 mg/L, at least 1 g/L, at least 5 g/L, at least 10 g/L, at least 50 g/L, at least 100 g/L, at least 150 g/L, at least 200 g/L, at least 250 g/L, most preferably at least 300 g/L.
The solubility in an aqueous medium, preferably water, of a ginsenoside formulation according to the invention, or a ginsenoside which is at least partly incorporated in (an outer layer of) a micelle, is preferably in the range of between about 30 mg/L and about 1000 g/L, more preferably between about 50 mg/L and about 5000 g/L, more preferably between about 100 mg/L and about 2000 g/L, even more preferably between about 1 g/L and about 1000 g/L, between about 5 g/L and about 500 g/L, in particular between about 10 g/L and about 300 g/L.
Particularly good results have been obtained with a ginsenoside formulation according to the invention or a ginsenoside which is at least partly incorporated in (an outer layer of) a micelle, comprising Rgl and/or Re which are at least partly incorporated into said micelle, preferably wherein Rg1 and Re are present in a wt. ratio of between about 1:2 and about 2:1, more preferably between about 1:1 and about 1:1.5. Preferably, said micelle has a molecular weight of between about 50.000 and about 100.000 Da.
The invention further relates to a method for preparing a ginsenoside formulation according to the invention or a ginsenoside which is at least partly incorporated in (an outer layer of) a micelle, said method comprising:
(a) providing a ginsenoside extract from a plant or plant part of the genus
Panax, preferably Panax ginseng; (b) contacting said ginsenoside extract with an aqueous solvent mixture to obtain a ginsenoside mixture; (c) at least partly removing said aqueous solvent mixture from said ginsenoside mixture; and optionally (d) reconstituting the ginsenoside mixture obtained in step ¢) in water to obtain a ginsenoside formulation according to the invention.
Said ginsenoside extract may be obtained using any suitable method known in the art, for example by boiling a dried plant or plant part from the genus
Panax in water, followed by filtering the water phase to obtain a crude ginsenoside extract.
The amount of water to be used herein is dependent on the amount of dried plant or dried plant parts to be extracted. It is within the ability of the skilled person to select appropriate amounts of water, based on his common general knowledge and the information provided herein.
Preferably, said ginsenoside extract is subjected to one or more purification steps prior to or during contacting the ginsenoside extract with said aqueous solvent mixture in step b. Preferably, said ginsenoside extract is subjected to a step of column chromatography using a non-polar resin as stationary phase.
Said non-polar resin is preferably a D101-type resin, a D201-tpe resin, a
D113-type resin, a D285-type resin, a D296-type resin, a D941-type resin, a D945- type resin, a DM 130-type resin, a DM131-type resin, a Dt-type resin, a HPD100- type resin, a HPD300-type resin, an NKA-type resin, an LK37-type resin, an
LK1300S-type resin, an LK20-type resin, a 388-type resin, Amberlite IRA900, an
AB-8-type resin, Amberlite XAD16, an SA-2-type resin or an LX-TS4-type resin.
Said ginsenoside is preferably eluted from said non-polar resin is with a suitable aqueous solvent mixture to obtain a purified ginsenoside extract. Said aqueous solvent mixture preferably is a mixture of water and a non-aqueous solvent. Preferably said non-aqueous solvent has a boiling point lower than water.
Preferably, said aqueous solvent mixture is a mixture of alcohol in water, such as between about 40% and about 80% alcohol in water. More preferably, said aqueous solvent is a mixture of methanol, ethanol, propanol or isopropanol in water, most preferably a mixture of between about 40% and about 80% of ethanol in water.
Said non-polar resin is preferably first washed with a polar solvent, preferably water, prior to elution of the ginsenoside from the non-polar resin using the aqueous solvent mixture as defined herein above.
Said ginsenoside extract is preferably subjected to a step of decolorization. Preferably said purified ginsenoside extract is contacted with a decolorization agent, preferably a D941 macroporous resin. Preferably said ginsenoside extract is subjected to a step of decolorization after having been subjected to contacting with a non-polar resin. Preferably, said step of decolorization is carried out in step b), e.g. said ginsenoside extract is contacted with a an aqueous solvent mixture and said aqueous solvent mixture comprising said ginsenoside extract is contacted with a decolorization agent.
The amount of solvents, non-polar resin and decolorization agent to be used herein is dependent on the amount of ginsenoside extract to be subjected to the method. It is within the ability of the skilled person to select appropriate amounts of non-aqueous solvent, polar solvent, non-polar resin and decolorization agent, based on his common general knowledge and the information provided herein.
Said aqueous solvent mixture is preferably removed from said ginsenoside mixture in step c. This can be achieved using any suitable method known in the art, preferably by evaporation under reduced pressure. Optionally, said ginsenoside mixture may be heated, typically to a temperature between 30 °C and 90 °C, such as between 40 °C and about 80 °C to aid evaporation of (part of) the aqueous solvent mixture.
Said ginsenoside mixture obtained in step ©) is preferably dissolved in water to obtain a ginsenoside formulation according to the invention.
Alternatively, said non-aqueous solvent present in said aqueous solvent mixture is at least partly evaporated to obtain a water fraction or an aqueous solvent mixture enriched in water, comprising a ginsenoside formulation according to the invention.
Said ginsenoside in said ginsenoside formulation according to the invention is at least partly incorporated into a micelle, preferably at least partly incorporated in an outer layer of a micelle. This can be determined using any suitable analytic method known in the art, for example size exclusion chromatography, optionally in combination with HPLC, e.g. as described in
Example 4.
Alternatively or additionally, said micelle further comprises a GC, preferably selected from dexamethasone and beclomethasone, at least partly incorporated therein. In this embodiment, said ginsenoside is preferably incorporated in an outer layer of a micelle and said GC is preferably present in an inner cavity of the micelle. Said micelle may be prepared using the method as described herein above, provided said GC is contacted with said ginsenoside extract in step b).
The invention further preferably relates to a ginsenoside formulation obtainable by method according to the invention.
Said ginsenoside formulation preferably comprises a PPT-type ginsenoside at least partly incorporated in a micelle, preferably at least partly incorporated in an outer layer of the micelle, said micelle having a molecular weight of at least 10.000 Da and/or a molecular weight of at most 100.000 Da.
Further, said ginsenoside is preferably a PPT-type ginsenoside, preferably selected from Rgl, Re and a combination thereof.
Glucocorticoid
Said GC in a combination for use according to the invention may be any natural or synthetic GC, or a pro-drug thereof.
Preferably, said GC is selected from dexamethasone, betamethasone, prednisolone, methylprednisolone triamcinolone, deflazacort, fludrocortisone acetate, fludrocortisone acetate, aldosterone and beclomethasone. More preferably said GC is beclomethasone or dexamethasone.
Advantageously, a combination for use according to the invention allows to apply GCs in a treatment which are normally associated with very severe side- effects and therefore typically not administered to a subject. Such GCs may however be applied in a combination for use according to the invention, due to the presence of the ginsenoside to mediate the side-effects. This advantageously expands the options for treatment of an inflammatory disease in a subject in need thereof.
Medical use
Said side-effect of GC treatment may be any side effect that is induced by (chronic) use of one or more GC's). Examples of side-effects that may occur include diabetes, decreased cortisol levels, reduced growth velocity, wound healing disorders, tissue degeneration, osteoporosis, hypertension, weight gain and muscle weakness.
Preferably, said side-effect of GC treatment is selected from decreased cortisol levels, reduced growth velocity and wound healing disorders.
Accordingly, in another aspect the invention relates to a ginsenoside for use in a method of prevention or treatment of (GC-induced) diabetes, (GC-induced) decreased cortisol levels, (GC-induced) reduced growth velocity, (GC-induced) wound healing disorders, GC-induced tissue degeneration, (GC-induced) osteoporosis, (GC-induced) hypertension, (GC-induced) weight gain and (GC- induced) muscle weakness, said method comprising administering a ginsenoside to a subject that is suffering from, or at risk of suffering from, one or more side- effect(s) of GC treatment. Preferably, the invention relates to a ginsenoside for use in a method of prevention or treatment of (GC-induced) decreased cortisol levels, (GC-induced) reduced growth velocity and (GC-induced) wound healing disorders.
Said subject may be any subject that suffers from GC-induced side- effects, preferably a mammal, more preferably a human.
In particular, said subject is a subject that is subjected to or has been subjected to GC treatment. Preferably, said subject is subjected to a daily dosage of
GC.
Typically, said subject is subjected to long-term treatment with one or more GC(s), such as during a period of at least 6 weeks, preferably at least 8 weeks, more preferably at least 12 weeks, more preferably at least 16 weeks, at least 20 weeks, at least 25 weeks, at least 30 weeks, at least 35 weeks, at least 40 weeks, at least 45 weeks, at least 50 weeks, more preferably at least one year, at least 1.5 year, at least 2 years, at least 2.5 years, at least 3 years, at least 3.5 years, at least 5 years, most preferably for at least 10 years.
Typically, said subject has been subjected to treatment with one or more
GC(s) during a period of between about 6 weeks and about 10 years, between about 8 weeks and about 5 years, between about 12 weeks and about 3 years, between about 16 weeks and about 1 year.
Preferably, said subject is under treatment with one or more GC(s) simultaneously with said treatment of a side-effect of GC treatment. Alternatively, said subject has been subjected to GC treatment prior to said treatment of said side-effect. Preferably said treatment with one or more GC(s) is less than 5 years ago, more preferably less than 4 years, less than 3 years, less than 2 years, less than 1 year, less than 9 months, less than 6 months, less than 4 months, less than 2 months, most preferably less than 1 month prior to said treatment of said side- effect of GC treatment.
Typically, said treatment with one or more GC(s) is between about 1 week and about 5 years prior to said treatment of said side-effects of GC treatment, preferably between about 2 weeks and about 4 years, between about 4 weeks and about 3 years, between about 2 months and about 2 years, such as between about 4 months and about 1 year.
A typical dosage for GC treatment varies between about 0.5 mg to about 50 mg per day, such as between about 4 mg and about 20 mg of GC per day.
Usually, the dose of GC is about 0.007 to about 0.8 mg/kg/day, such as between 0.05 and about 0.3 mg/kg/day.
As the skilled person will appreciate, said dosage of said ginsenoside depends on the circumstances of the case, such as the patient, the side-effect to be treated and the dosage of GC the subject is being subjected to or was subjected to.
A typical dose of ginsenoside is between about 4 mg and about 400 mg/day, such as between about 30 mg/day and about 70 mg/day. Usually, the dose of GC is about 0.05 to about 5.5 mg/kg/day, such as between about 0.4 and about 1.0 mg/kg/day.
Said dosage of ginsenoside may be provided to a subject in a single dose, or may be provided in multiple doses per day, such as between 2 to 4 doses per day, provided the total amount per day is the same.
Preferably, in particular in case a subject is subjected to GC treatment, said ginsenoside is administered to said subject in a molar amount that is at least equal to the amount of GC. More preferably, said ginsenoside is administered in a molar excess, with regards to GC, preferably a molar excess of at least 4, more preferably at least 8, even more preferably at least 10, at least 20, at least 50, at least 100, at least 1000, at least 2000.
Preferably, said ginsenoside is administered to said subject in a molar ratio, with regards to GC of between about 1 (GC) to 2000 (ginsenoside), preferably between about 4 to about 1000, about 6 to about 100, about 8 to about 20.
Said ginsenoside may be administered in any suitable way. For example, said ginsenoside may be administered systemically, such as by oral administration, sublingual administration, buccal administration or rectal administration.
Alternatively or additionally, said combination may be administered locally, such as transdermal, or via inhalation or via transnasal administration. Said combination may also be administered by means of injection, e.g. subcutaneously, intravenously or intramuscularly. Preferably, said ginsenoside is administered orally or transdermally.
Said ginsenoside is preferably administered in a formulation with a suitable carrier, preferably an aqueous carrier, more preferably a hydrogel. In a specific embodiment, said ginsenoside is at least partly incorporated into a micelle, preferably in an outer layer of said micelle, said micelle having a molecular weight of at least 10.000 Da and is administered in a formulation comprising an aqueous carrier. Such a ginsenoside was particularly effective in treatment or prevention of
GC treatment.
The invention further relates to a method of treatment of a side-effect of
GC treatment, preferably (GC-induced) diabetes, (GC-induced) decreased cortisol levels, (GC-induced) reduced growth velocity, (GC-induced) wound healing disorders, (GC-induced) tissue degeneration, (GC-induced) osteoporosis, (GC- induced) hypertension, (GC-induced) weight gain and (GC-inducedymuscle weakness, comprising administering a ginsenoside to a subject that is suffering from, or at risk of suffering from, one or more side-effect(s) of GC treatment.
The invention further relates to a use of ginsenoside for the preparation of a medicament for the treatment of a side-effect of GC treatment, preferably (GC- induced) diabetes, (GC-induced) decreased cortisol levels, (GC-induced) reduced growth velocity, (GC-induced) wound healing disorders, (GC-induced) tissue degeneration, (GC-induced) osteoporosis, (GC-induced) hypertension, (GC-induced) weight gain and (GC-induced) muscle weakness.
Combination of ginsenoside and GC for use according to the invention
The inventors further realized that co-treatment of a ginsenoside and a
GC has several advantages over the individual use of a GC or a ginsenoside as anti-inflammatory medicaments.
For example, a ginsenoside advantageously reduces resistance against said GC treatment. Without wishing to be bound by any theory, it is believed that a ginsenoside advantageously stabilizes the receptor mRNA and the receptor protein and accordingly prevents down regulation of the GR, thereby reducing sensitivity to GC treatment (Example 1).
In addition, the inventors found that a ginsenoside and a GC advantageously exhibit a synergistic anti-inflammatory effect. This is advantageous, as lower dosages of GC and ginsenosides are required, whilst still achieving an effective anti-inflammatory response (Figure lc). Using a lower dosage of GC may also advantageously decrease the risk of obtaining one or more side-effects typically associated with GC treatment.
Accordingly, the invention further pertains to a combination of a GC and a ginsenoside for use in a method of prevention or treatment of an anti- inflammatory disease. The inventors further realized that the side-effects of GC treatment may be effectively prevented or treated when a GC and a ginsenoside are administered in a specific molar ratio, whilst maintaining an anti- inflammatory effect.
Without wishing to be bound by any theory, the inventors believe that said ginsenoside acts as a competitive antagonist on the glucocorticoid receptor by inhibiting dimerization of the glucocorticoid receptor (see Example 1).
Accordingly, the invention relates to a combination of a GC and a ginsenoside for use in a method of prevention or treatment of an anti-inflammatory disease, wherein said method comprises administering a GC and a ginsenoside in a molar ratio of between about 1:1 and about 1: 2000, preferably between about 1:4 and about 1:1000, more preferably between 1:8 and 1:20 to a subject in need thereof.
Said ginsenoside and said GC is as described herein above.
Preferably, said molar ratio between said GC and said ginsenoside is between about 1:2 and about 1:1000, more preferably between 1:4 and 1:500, more preferably between about 1:6 and about 1:250, even more preferably between about 1:7 and about 1:100, in particular between about 1:8 and about 1:20. Without wishing to be bound by any theory it is believed that within these molar ranges, optimal competitive antagonistic effect of said ginsenoside is obtained. Further, at these molar ranges, solubility of said ginsenoside at a therapeutically effective amount is feasible, especially when said ginsenoside is administered in a micelle formulation, as described herein above.
Said GC and said ginsenoside may be administered together, or separately. Accordingly, in an embodiment, said GC and ginsenoside are administered as a pharmaceutical composition according to the invention.
Alternatively, said GC and said ginsenoside may be administered separately. Said GC and ginsenoside are preferably administered within a time window of 1 hour or less, preferably within 45 minutes or less, such as within 30 minutes or less, in particular within 15 minutes or less. Most preferably, said GC and ginsenoside are administered essentially at the same time.
Alternatively, said ginsenoside may be administered slightly before or after the GC, for example approximately 60 minutes before or after administering the GC, preferably approximately 45 minutes before or after administering the GC, more preferably approximately 30 minutes before or after administering the GC, such as 15 minutes before or after administering the GC.
As mentioned herein, said GC and ginsenoside advantageously act synergistically together to achieve an anti-inflammatory effect. This synergistic effect allows administration of GC and ginsenoside at a lower dosage than typically required to induce an anti-inflammatory effect, if the GC and ginsenoside are administered in separate treatments.
Accordingly, the invention preferably relates to a combination of a GC and a ginsenoside for use according to the invention, wherein said GC and/or said ginsenoside are administered in a therapeutically ineffective amount. In other words, said GC and/or said ginsenoside are preferably administered in an amount that is lower than the amount of GC or ginsenoside that would be administered in a mono-treatment of GC or a ginsenoside to a subject in need thereof for the treatment of the same anti-inflammatory disease.
As the skilled person will appreciate, the dose of GC or ginsenoside that would normally be prescribed to a subject depends on the patient, the illness and the circumstances. For example, for a subject with higher weight, typically a higher dose is required to achieve an effect. Further, the severity of the illness may require a higher dosing to be effective. The skilled person is capable of selecting a suitable dosage, based on the information provided herein, common general knowledge and a reasonable amount of trial and error.
A typical dosage for GC treatment varies between about 0.5 mg to about 50 mg per day, such as between about 4 mg and about 20 mg of GC per day.
Usually, the dose of GC is about 0.007 to about 0.7 mg/kg/day, such as between 0.05 and about 0.3 mg/kg/day.
Said dosage may be provided to a subject in a single dose, or may be provided in multiple doses per day, such as between 2 to 4 doses per day, provided the total amount per day is the same.
Accordingly, the invention preferably relates to a combination of GC and ginsenoside for use according to the invention, wherein said GC is administered at a dose of at most about 50 mg/day, preferably at most 25 mg/day, at most 10 mg/day, more preferably at most 5 mg/day, even more preferably at most 0.5 mg/day.
A dose for ginsenosides usually varies between about 4 mg and about 400 mg/day, such as between about 30 mg/day and about 70 mg/day. Usually, the dose of GC is about 0.05 to about 5.5 mg/kg/day, such as between 0.4 and about 1.0 mg/kg/day.
Said dosage may be provided to a subject in a single dose, or may be provided in multiple doses per day, such as between 2 to 4 doses per day, provided the total amount per day is the same.
Accordingly, the invention preferably relates to a combination of GC and ginsenoside for use according to the invention, wherein said ginsenoside is administered at a dose of at most about 400 mg/day, preferably at most 200 mg/day, at most 120 mg/day, more preferably at most 70 mg/day, at most 20 mg/day, even more preferably at most 4 mg/day.
Said combination may be administered in any suitable way. For example, said combination may be administered systemically, such as by oral administration, sublingual administration, buccal administration or rectal administration. Alternatively or additionally, said combination may be administered locally, such as transdermal, or via inhalation or via transnasal administration. Said combination may also be administered by means of injection, e.g. subcutaneously, intravenously or intramuscularly.
Optionally, said GC may be administered via a different mode of administration compared to said ginsenoside. Preferably, said GC and said ginsenoside are administered via the same mode of administration, preferably transdermal (topically), via inhalation or via oral administration.
Said combination is preferably administered in a formulation with a suitable carrier, preferably an aqueous carrier. In a specific embodiment, said ginsenoside present in said formulation is at least partly incorporated into a micelle, preferably in an outer layer of a micelle, said micelle having a molecular weight of at least 10.000 Da and is preferably administered in a formulation comprising an aqueous medium, such as a hydrogel. Such a combination was particularly effective in achieving an anti-inflammatory effect, whilst preventing the occurrence of side-effects.
Said subject may be any subject suffering from or predisposed of suffering from an inflammatory disease, preferably chronic inflammation. Preferably, said subject is a mammal, more preferably a human.
In principle said inflammatory disease may be any inflammatory disease in which activation of the glucocorticoid receptor may reduce inflammation.
Examples include, but are not limited to, asthma, allergic rhinitis, hay fever, urticaria (hives), atopic eczema, chronic obstructive pulmonary disease (COPD), inflammation of the joints, muscles and tendons, lupus, inflammatory bowel disease, such as Crohn's disease and ulcerative colitis, giant cell arteritis and polymyalgia rheumatica, and multiple sclerosis (MS).
Accordingly, the invention further relates to a combination of GC and a ginsenoside for use in the prevention or treatment of asthma, allergic rhinitis, hay fever, urticaria (hives), atopic eczema, chronic obstructive pulmonary disease (COPD), inflammation of the joints, muscles and tendons, lupus, inflammatory bowel disease, such as Crohn's disease and ulcerative colitis, giant cell arteritis and polymyalgia rheumatica or multiple sclerosis (MS).
The invention further relates to a method of treatment of an inflammatory disease, preferably selected from asthma, allergic rhinitis, hay fever, urticaria (hives), atopic eczema, chronic obstructive pulmonary disease (COPD), inflammation of the joints, muscles and tendons, lupus, inflammatory bowel disease, such as Crohn's disease and ulcerative colitis, giant cell arteritis and polymyalgia rheumatica or multiple sclerosis (MS), comprising administering a combination of GC and a ginsenoside to a subject in need thereof.
The invention further relates to the use of a combination of a GC and a ginsenoside for the preparation of a medicament for the treatment of an inflammatory disease, preferably selected from asthma, allergic rhinitis, hay fever, urticaria (hives), atopic eczema, chronic obstructive pulmonary disease (COPD), inflammation of the joints, muscles and tendons, lupus, inflammatory bowel disease, such as Crohn’s disease and ulcerative colitis, giant cell arteritis and polymyalgia rheumatica or multiple sclerosis (MS), comprising administering a combination of GC and a ginsenoside to a subject in need thereof.
In a specific embodiment, the invention preferably relates to a combination of a GC, preferably selected from dexamethasone and beclomethasone, and a ginsenoside, preferably a PTT-type ginsenoside, preferably selected from
Rg1, Re and a combination thereof for use in a method of prevention or treatment of an inflammatory disease.
Pharmaceutical composition
The invention further relates to a pharmaceutical composition comprising
GC, a ginsenoside and a pharmaceutically acceptable carrier, wherein the molar ratio between said GC and said ginsenoside is between about 1:1 and about 1: 2000.
Said ginsenoside, GC and said molar ratio are as described herein above.
Pharmaceutical carrier
The pharmaceutical composition according to the invention comprises a pharmaceutically acceptable carrier. Said carrier may in principle be any pharmaceutically acceptable carrier known in the art, e.g. a liquid carrier, preferably an aqueous carrier, or a solid carrier such as lactose.
Preferably, said pharmaceutically acceptable carrier is a water-based carrier, more preferably a hydrogel. With such a carrier, a pharmaceutical composition may be formulated as a formulation for topical application, such as a cream or the like. Such a pharmaceutical composition is in particular suitable for dermatological applications, such as eczema.
Alternatively, said pharmaceutical carrier may also be a lipid carrier, or a combination of a liquid carrier and an aqueous carrier.
Preferably, the pharmaceutical composition according to the invention is formulated as a cream, a lotion, a balm, a hydrogel, an ointment, a foam, a gel, a spray, a tablet, a capsule, a lozenge, a topical solution, a topical suspension, an aerosol and a syrup, most preferably a hydrogel.
Said pharmaceutical composition according to the invention may further comprise one or more additives, such as a humectant, a stabilizer, a dispersant, a uv-stabilizer, a plasticizer, an emulsifier, an emollient, a preservative or a pH adjusting agent.
In a specific embodiment, the invention relates to a pharmaceutical composition comprising a GC, preferably selected from dexamethasone and betamethasone, a gmsenoside, preferably a PTT-type ginsenoside, preferably selected from Rgl, Re and a combination thereof and a pharmaceutically acceptable carrier, wherein the molar ratio between said GC and said ginsenoside is between about 1:1 and about 1: 2000, preferably between about 1:8 and about 1:20. Preferably, herein, said ginsenoside is at least substantially incorporated in a micelle, preferably in an outer layer of a micelle, said micelle having a molecular weight of at least 10.000 Da. Such pharmaceutical composition was found particularly effective in obtaining an anti-inflammatory effect whilst reducing the occurrence of side-effects, in particular side-effects associated with GC-treatment.
The invention further relates to a method for preparing a pharmaceutical composition according to the invention. Said method comprises mixing a ginsenoside, a GC and a pharmaceutical carrier to obtain a pharmaceutical composition according to the invention,
Preferably, said method comprises mixing a ginsenoside formulation according to the invention with a GC, preferably dexamethasone and a pharmaceutical carrier, preferably a hydrogel to obtain a pharmaceutical composition according to the invention.
Alternatively or additionally, said method comprises mixing a ginsenoside which is at least partly incorporated in an outer layer of a micelle, said micelle having a molecular weight of at least 10.000 Da with a GC and a pharmaceutical carrier. Optionally, said (GC may be at least partly incorporated into (an inner cavity of) the micelle and said micelle comprising said GC is mixed with said pharmaceutical carrier to obtain a pharmaceutical composition according to the invention.
For the purpose of clarity and a concise description, features are described herein as part of the same or separate embodiments, however, it will be appreciated that the scope of the invention may include embodiments having combinations of all or some of the features described.
The invention is demonstrated by the following examples.
Example 1: Co-treatment of Rgl and GC
Material and methods
Zebrafish lines and maintenance
Zebrafish (Danio rerio) were maintained and handled according to the guidelines from the Zebrafish Model Organism Database (http://zfin.org) and in compliance with the directives of the local animal welfare committee of Leiden
University. Zebrafish were exposed to a 14 h light and 10 h dark cycle to maintain circadian rhythmicity. Fertilization was performed by natural spawning at the beginning of the light period. Eggs were collected and raised at 28°C in egg water
(60 pg/mL Instant Ocean sea salts and 0.0025% methylene blue). The following zebrafish lines were used in this study: the double transgenic line
Te(mpx:GFPit1i/ mpeg 1:m Cherryuwmstool) in which neutrophils and macrophages are fluorescently labeled (green and red, respectively) [14,15], and the Tg(9xGCRE-
HSV.UI23:EGFP)e2 reporter line, in which the gene encoding enhanced green fluorescent protein (EGFP) is driven by a GRE-containing promoter [16].
Chemicals
The chemical compounds beclomethasone, dexamethasone, Rgl, TNF-q, actinomycin-D, and cycloheximide were purchased from Sigma-Aldrich (St. Louis,
MO, USA).
Tail fin wounding assay in zebrafish larvae
For the tail fin wounding experiments, 20 larvae at 2 or 3 days post- fertilization (dpf) were utilized for each experimental group. For wounding, larvae were anesthetized in egg water containing 0.02 % buffered aminobenzoic acid ethyl ester (tricaine; Sigma-Aldrich). Larvae were placed on Petri dishes coated with 2% agarose under a Leica M165C stereomicroscope (Leica Microsystems, Wetzlar,
Germany), and the tails were partly amputated using a 1-mm sapphire blade (World Precision Instruments, Sarasota, FL, USA). For quantification of leukocyte migration, larvae were fixed overnight in 4% paraformaldehyde (PFA) at 4°C. The next day, fixed larvae were washed with Phosphate Buffered Saline (PBS) containing 0.1% Tween 20, and stored at 4°C until imaging.
In the tail fin wounding experiments, the larvae were treated with vehicle (DMSO 0.01%) diluted in egg water, beclomethasone (or dexamethasone), and/or Rgl at indicated concentrations for 6 h (2 h pre-wounding and 4 h post- wounding).
Imaging of the Te(mpx:GEP2:4/mpeg I:mCherrywsF%1) line larvae was performed utilizing a Leica MZ16FA fluorescence stereomicroscope supported by
LAS 3.7 software (Leica Microsystems). The macrophages were detected based on their red (m Cherry) fluorescence, and neutrophils based on their green (EGFP) fluorescence. To quantify the number of macrophages and neutrophils recruited to the wounded area, the cells in a defined area of the tail were determined by blinded manual counting.
Determination of the Gr transactivation activity in zebrafish larvae
To investigate the Gr transactivation activity in zebrafish larvae, the
Te(9xGCRE-HSV.UI23: EGFP)«Zi reporter line was used, which expresses enhanced green fluorescent protein (EGFP) under the control of a promoter containing an array of nine GREs [16]. To study the effect of treatment with vehicle, 10 uM beclomethasone (or dexamethasone), 50 uM Rg1, and 10 uM beclomethasone (or dexamethasone) with 50 uM Rg1, 15 embryos per group (2 dpf) were treated with the indicated compounds for 24 h and fixed for visualization to measure the whole-body fluorescence intensity utilizing a Leica MZ16FA fluorescence stereomicroscope, supported by LAS 3.7 software (Leica
Microsystems). The integrated intensity of the EGFP signal in the larvae was determined using Imaged software.
Measurement of larval body length and regeneration of the tail fin
To determine the length of regenerated tail fin tissue after wounding or the larval body length, chemical treatment (beclomethasone/dexamethasone (10 uM), or Rg1 (50 nM) or beclomethasone/dexamethasone (10 uM) with Rg1 (50 pM) of the embryos (15 per treatment group) was started at 2 hours post-fertilization (hpf) and continued until 5 dpf. During this period, solutions were refreshed daily.
For the regeneration experiments, tail fins were wounded at 2 dpf. For determination of the larval length and the regeneration of the tail fins, larvae were fixed at 5 dpf overnight in 4% PFA at 4°C and imaged the next day by using a
Leica MZ16FA fluorescence stereomicroscope supported by LAS 3.7 software (Leica
Microsystems), and the length of newly grown tail tissue or the entire larva was measured using Imaged software. The newly grown tissue can visually be distinguished from the old tissue, enabling precise measurement of the size of the regenerated tissue.
Whole-body glucose measurement of zebrafish larvae
Zebrafish embryos at 2 hpf received a chemical treatment (vehicle, 10 pM beclomethasone, 50 pM Rgl, or 10 uM beclomethasone with 50 pM Rgl), with a daily refreshment of the solutions, until 5 dpf. For dose response curve (0.1, 1,5, 10, or 15 pM of beclomethasone were introduced alone or in combination with 50 pM of
Rgl. At 5 dpf, the larvae (15 per sample) were collected in an Eppendorf tube, washed in egg water (3 times for 10 min), and placed in egg water for 1 h.
Subsequently, 100 uL of ice-cold glucose buffer was added to each sample, and the larvae were homogenized using a BulletBlender® for 3 min at 8,000 rpm. The homogenates were then centrifuged at 4°C for 8 min at 11,000 rpm and the supernatant was stored at -20°C. Whole-body glucose concentrations were determined using a Glucose Colorimetric Assay kit (Cayman Chemical, Ann Arbor,
MI, USA), according to the manufacturer’s instructions. In each experiment, three biological replicates were used for each treatment group, and the colorimetric assay was performed using technical duplicates.
Whole-body cortisol measurement of zebrafish larvae
Zebrafish embryos at 2 hpf received a chemical treatment (vehicle, 10 pM beclomethasone, 50 pM Rgl, or 10 uM beclomethasone with 50 pM Rgl), with a daily refreshment of the solutions, until 96 hpf. For dose response curve (0.1, 1, 5, 10, or 15 pM of beclomethasone were introduced alone or in combination with 50 pM of Rgl. Then, the treatments were stopped and larvae were incubated in egg water until 5 dpf to avoid cross-reactivity with the compounds of the cortisol antibody used in the ELISA. Then, the treatments were stopped and replaced with egg water. At 5 dpf, the larvae were collected in an Eppendorf tube (30 larvae per sample) and 100 ul of ice-cold egg water was added. After the excess water had been taken out, the samples were glaciated in ethanol (EtOH)/dry-ice bath.
Subsequently, the larvae were homogenized using a BulletBlender® for 3 min at 8,000 rpm. Ethyl acetate was added to the homogenate. The homogenates were then centrifuged at 4°C for 8 min at 11,000 rpm and the supernatant was collected and vaporized. A volume of 150 nl of 0.2% Bovine serum albumin (Sigma-Aldrich) dissolved in PBS was added to the samples and frozen. Whole-body cortisol levels of the zebrafish larvae were determined by ELISA (Demeditec Diagnostics GmbH,
Kiel-Wellsee, Germany), following the manufacturer's instructions. In each experiment, three biological replicates were used for each treatment group, and the
ELISA was performed using technical duplicates.
Competitive Glucocorticoid Receptor binding assay
To determine the relative binding affinities of the different compounds for the human GR in vitro, the PolarScreen™ Glucocorticoid Receptor Competitor
Assay Kit (ThermoFisher Scientific, Waltham, MA, USA) was used following the manufacturer's instructions. Briefly, test compounds were first dissolved in DMSO to a 10 mM stock concentration and further diluted in GR buffer (100 mM potassium phosphate (pH 7.4), 200 mM Na:MoO4, 1 mM EDTA, and 20% DMSO).
Serial dilutions of the compounds were transferred to a Corning® black 384-well plate and Fluormone GS red™ was added, followed by GR Full Length (partially purified receptor in storage buffer). The plate was incubated for 4 h at RT in the dark and fluorescence polarization was measured with a CLARIOStar Microplate
Reader (BMG Labtech, Ortenberg, Germany). Obtained values were normalized to the assay maximum (no ligand) and minimum control (10 pM dexamethasone), and relative percentages of polarization were determined. Dose-response curves were fitted enabling the calculations of IC50 values for each compound.
Cell cultures
HeLa (human cervical cancer) cells were cultured in Dulbecco's Modified
Eagle's (DMEM) High Glucose (HG) medium without phenol red supplemented with 10% Fetal Calf Serum (FCS) and 10% Glutamax (Sigma-Aldrich). The cells were maintained at 37°C and 5% CO:. At 24 h before adding the treatment, the medium was replaced by DMEM HG with 10% charcoal-inactivated serum and 10%
Glutamax (Sigma-Aldrich). The cells were exposed to short- and long-term compound treatments. In the short-term treatment, cells were treated with TNF-a (10 ng/ml, Sigma-Aldrich) and either vehicle (0.01% DMSO), beclomethasone (0.01, 0.1, or 1 uM) or dexamethasone (1 uM), Rg1 (20, 100, or 500 uM), or beclomethasone (0.01 or 1uM, or dexamethasone (1 uM)) in combination with Rgl (20, 100, 500 uM) for 6 h. In the long-term treatments, the cells were treated for 24 h with one of the following treatments: vehicle (0.01% DMSO), beclomethasone
(0.01, 0.1, or 1uM) or dexamethasone (1uM), Rg1 (20, 100, or 500 uM), or beclomethasone (0.01 or 1uM, or dexamethasone (1uM)) in combination with Rgl (20, 100 or 200 uM), with or without actinomycin-D (1 ng/ml) or cycloheximide (5 pg/ml). Subsequently, TNF-a (10 ng/ml) was added along with the compound treatments for 6 h (this last step was not added in the actinomycin-D and cycloheximide experiments).
Immunocytochemistry
To determine the nuclear translocation of GR, immunocytochemistry on
GR in HeLa cells was performed. The cells were seeded in Nunc™ Lab-Tek™ II
Chamber Slide™ System (ThermoFisher Scientific), and cultured in DMEM HG (without phenol red) supplemented with 10% charcoal inactivated serum and 100 mM Glutamax (Sigma-Aldrich) for 48 h. Subsequently, the cells were treated with increasing doses of beclomethasone or Rgl for 6 h, and fixed using 4% PFA at 4°C overnight. After washing cells 3 times with PBS for 5 min, PBS with 0.1% Tween (PBST) for 15 min, and PBST with 3% BSA for 30 min, a primary GR antibody (Glucocorticoid Receptor (D6H2L) XP® Rabbit mAb (Cell Signaling Technology,
Danvers, MA, USA) was added, diluted in PBST in with 3% BSA (1:1000), and incubated overnight at 4°C. The next day, samples were washed in PBS for 5 min 20 four times. Then, an Alexa Fluor 488% goat anti-rabbit antibody (Sigma-Aldrich) diluted in PBST with 3% BSA (1:500) was added and incubated at room temperature for 2 h. Then, samples were washed in PBS 4 times for 5 min. The cells were mounted using ProLong™ Diamond Antifade Mountant with DAPI (ThermoFisher Scientific) and imaged on a Leica TCS SP8 confocal microscope using a 40x (1.25 NA) objective, (Leica Microsystems). Experiments were performed 3 times in triplicate for each treatment group. In each experiment, 40 randomly selected cells from each treatment group were analyzed. The relative nuclear translocation of the GR was quantified by determining the integrated fluorescence intensity in the nucleus and the whole cell using Fiji Imaged v1.53c, and these values were corrected for the mean background fluorescence. The percentage of nuclear translocation was assessed by determining the corrected density in the nucleus relative to the corrected density in the whole cell (in %).
Quantitative PCR (qPCR) analysis
To determine levels of gene expression in zebrafish larvae, 3 dpf larvae were used in both wounded and non-wounded conditions, and after short- and long- term treatments. Groups of 15 larvae were collected in TRIzol reagent (ThermoFisher) and total RNA was isolated using miRNeasy mini kit (Qiagen,
Hilden, Germany). For experiments in Hela cells, cells were seeded in 6 well plates and after treatment, the cells were removed from the wells using TRIzol reagent and mRNA was isolated using miRNeasy mini kit (Qiagen) according to the manufacturer's instructions. The RNA samples were DNAse-treated utilizing the
DNA-free™ DNA Removal Kit (ThermoFisher Scientific). The cDNA synthesis was performed using the iScript cDNA synthesis kit (Bio-Rad Laboratories, Hercules,
CA, USA) using 1 pg of RNA per sample. For qPCR, 10 uM of forward primer and 10 uM of reverse primer, 12.5 ul of IQ SYBR Green Supermix (Bio-Rad
Laboratories), and 2 ul of cDNA were added to the qPCR reaction mixture. Each mixture had a total volume of 25 ul, which was split as duplicates with a volume of 12.5 ul. The qPCR reactions were performed on a MyiQ-single-color real-time PCR detection system (Bio-Rad Laboratories), with initial denaturation for 3 min, 95°C, and 40 cycles of 15 s at 95.5°C, 15 s at 60°C, and 30 s at 72°C. Cycle threshold values (Ct values, i.e. the cycle numbers at which a threshold value of the fluorescence intensity was reached) were determined for each sample. The gene expression level for each sample was normalized using the expression of ppial (peptidylprolyl isomerase Ab (cyclophilin A)) for zebrafish samples, and 18S rRNA for human cells. The fold change per sample (compared to the respective control group) was calculated using the OOCt method. In each experiment, three biological replicates were used for each treatment group, and reactions were performed in duplicates. The sequences of the qPCR primers for experiments on zebrafish and
Hel.a cells are presented in Table S1.
Table 51: qPCR primers for zebrafish and humanprimers for zebrafish and human qPCR primers for gPCR primers for zebrafish human
NAME Sequence 5-3’ NAME Sequence 5-3’
CATCCACAACCTTCCC ppail forward GAACAC hS§18 forward GATGGGCGGGGGAAAAT
ACACTGAAACACGGA CTTGTACTGGCGTGGAT ppail reverse GGCAAAG hS18_ reverse TCTGC
TGTGIGTTTGGGAATC GCGAAGGAGAAGACCAC
1116 forward TCCA hAEKBPS forward GACAT
CTGATAAACCAACCGG TAGGCTTCCCTGCCTCT il1b_reverse GACA hFKBP5_ reverse CCAAA
TGTGTTATTGTTTTCC CATTGCETGGATGAAGT
18 Jorward TGGCATTTC hPCKI forward TTGACG
GCGACAGCGTGGATC GGGTTGGTCTTCACTGA il8_reverse TACAG hPCKI1_reverse AGTCC
CTGCCAGCATAAGA hGILZ forward CGTGGTGGCCATAGACA:
Jkbp5_ forward TTCGTGAGC ACA
GACCCTGCTTATTC hGILZ_reverse CCTCTCTCACAGCATACA fkbp3_reverse TGATCGGAAA TCAGATG
AACTGGCAACGGTTCT ANFKBIA jorwa CTCCGAGACTTTCGAGG gr. forward ATCAGCTCA rd AAATAC
TTCTGGTGAAAGAGCA hNFKBIA revers GCCATTGAAGTTGGTAG gr_reverse GCGG e CCTTCA
CAGGGCGATCTGGCG hILIB forward CCACAGACCTTCCAGGA pekl forward TCTCT GAATG
CTGCTGTCGATGAACT hILIB revese GTGCAGTTCAGTGATCG pckl1_reverse CCCG TACAGG nfkbiaa forwar CTTGGGCTAAAGT hCAXCL8 forward GAGAGTGATTGAGAGTG d AGTCACCG GACCAC
GATGGCAAGGTGCAG hCXCLS8 reverse CACAACCCTCTGCACCC nfkbiaa_reverse ATACGTG AGTTT hGR forward GAAAAGCUATCGTCAAA
AGGG
TGGAAGCAGTAGGTAAG hGR_reverse GAGA hMMP3 forward GCCACTACTGTGCCTTT
GAGTC hMMP9_reverse CCCTCAGAGAATCGCCA
GTACT
Western blotting
Western blots were conducted to determine GR protein levels in HeLa cells. The cells were seeded as described previously in 6-well plates. The cells were exposed to short- and long-term compound treatments. In the short-term treatment, cells were treated with vehicle (0.01% DMSO), beclomethasone (1 uM),
Rgl (20 uM), or beclomethasone in combination with Rg1 (1 and 20 uM, respectively) for 6 h. In the long-term treatments, the cells were treated for 30 h with one of the following treatments: vehicle, beclomethasone (1 uM), Rg1(20 uM), or beclomethasone in combination with Rg1(20 and 1 uM, respectively), with or without cycloheximide (5 pg/ml). Cells were harvested using trypsin (0.25% (v/v) trypsin in PBS, no EDTA), washed twice with PBS, and centrifuged. After centrifuging for 3 min at 3,000 rpm, cell pellets were stored at -80°C until use.
Cells were homogenized in potassium phosphate (KPi lysis buffer; 25 mM K:HPO+-
KH2PO4 (pH 6.5) + 0.1% (v/v) Triton X-100 (Merck, Darmstadt, Germany)) by sonication (20% amplitude, 3 s on, 3 s off for 4 cycles), using a Vibra-Cell™ VCX 130 sonicator (Sonics, Newtown, CT, USA), while on ice. The total protein concentration of the homogenates was determined using the Quickstart Bradford protein assay (Bio-Rad Laboratories) and measured using an EMax® plus microplate reader (Molecular Devices, Sunnyvale, CA, USA). Hel.a cell homogenates (30 ng protein in 10 pl) were added to 30 pl Laemmli sample buffer, vortexed, and, boiled for 5 min at 98°C. Of each sample, 15 nl was loaded in duplicate on a 12% gradient precast SDS-PAGE gel (Bio-Rad Laboratories), and run for approximately 2 h at 90 V (no duplicates were used in the eycloheximide experiment). The experiments were performed 4 times (6 times for the cycloheximide experiment). After running the gel, proteins were transferred to a nitrocellulose membrane (Thermo Fisher Scientific) by using Bio-Rad Power Pae
Basic Mini Electrophoresis System at 100 V for 1 h. The blot was washed in a blocking buffer (Tris-buffered saline containing 0.1% Tween-20 (TBST) with 3%
BSA) 3 times for 15 min at room temperature. A primary GR antibody (Glucocorticoid Receptor Recombinant Rabbit Monoclonal Antibody (2D8,
ThermoFisher Scientific)) at a 1:500 dilution in TBST with 3% BSA was added to the blots and incubated overnight at 4°C. The next day, the blot was washed with
TBST 3 times for 5 min. Then, an HRP-conjugated anti-rabbit secondary antibody (Sigma-Aldrich) at 1:2000 dilution in TBST with 3% BSA was added and incubated at room temperature for 2 h. The blot was washed 3 times for 5 min in TBST with 3% BSA and twice for 5 min with TBST. The blot was incubated with 10 ml of
Pierce™ enhanced chemiluminescence (ECL) western blotting substrate (ThermoFisher Scientific) for 1-2 min. Chemiluminescence was detected using a
ChemiDoc MP imager (Bio-Rad Laboratories) at an exposure time of 1 min. The blot was washed 3 times for 5 min in TBST, then washed for 1 h in TBST with 3%
BSA. A B-actin antibody (MA1-91399, ThermoFisher Scientific) was added at a 1:2000 dilution in TBST with 3% BSA to the blot and incubated overnight at 4°C.
The next day, the blot was washed 3 times for 5 min in TBST with 3% BSA. Then, an HRP-conjugated anti-mouse secondary antibody (Sigma-Aldrich) at a 1:2000 dilution in TBST with 3% BSA was added and incubated at room temperature for 2 h. Subsequently, the blot was washed 3 times for 5 min in TBST. The blot was incubated with 10 ml of ECL substrate for 1-2 min, and chemiluminescence was visualized. The intensities of the GR and B-actin bands were quantified using
Imaged software, and the GR level was normalized to the level of B-actin.
Statistical analysis
Statistical analysis of the experiments was performed using GraphPad
Prism software by performing one- or two-way ANOVA with Tukey's post hoc test.
The statistics of qPCR data were done on log2-transformed data. Significance was accepted at P<0.05 and different significance levels are indicated in the graphs: *P<0.05; **P<0.01; and ***P<0.001 for treatments compared to the corresponding vehicle group. #P<0.05; #P<0.01; and ##P<0.001 for combination treatment compared to beclomethasone or dexamethasone alone. +P<0.05; ++P<0.01; and +++P<0.001 for long-term treatment compared to the corresponding short-term treatment.
Results
Rg1l has a synergistic anti-inflammatory effect when administered as GC co-treatment
To study the co-treatment of Rg1 and beclomethasone, we first determined the anti-inflammatory effects of beclomethasone, Rgl, and the combination of these compounds on the migration of neutrophils in the zebrafish tail wounding assay, at 4 h after wounding. The chemical structures of the compounds are shown in Fig. 1A, and a schematic overview of the experiment is presented in Fig. 1B. Beclomethasone was administered at concentrations of 1, 5, 10, 20, and 40 uM, and Rg1 at 10, 20, 40, 80, and 160 uM. The results showed that treatment with 10 uM beclomethasone or lower did not affect the migration of neutrophils towards the wounded site, but that the 20 uM and 40 uM concentrations did significantly reduce the neutrophil migration (Fig. 1C). For Rgl, administration of 40 uM and lower did not alter the neutrophil migration, whereas
80 uM and 160 uM did (Fig.1C). For the co-treatment, 10 pM beclomethasone was combined with different concentrations of Rg1 (10, 20, 40, and 80 uM). All groups treated with beclomethasone in combination with Rg1 showed decreased neutrophil migration towards the damaged site, compared to the vehicle-treated control groups, and increasing doses of Rg1 resulted in larger inhibition (Fig. 1C).
Apparently, co-treatment with Rgl has a synergistic effect on the anti- inflammatory effect of beclomethasone.
Based on these observations, we further examined the anti-inflammatory effect of the combination of 10 uM beclomethasone with 50 pM Rgl. This treatment significantly reduced neutrophil migration, but left the migration of macrophages unaffected (Fig. 1D,E,S1A), as previously shown in this assay upon treatment with various GCs [17][18]. Moreover, we investigated the effects of Rgl in combination with dexamethasone, another commonly used synthetic GC. As observed for beclomethasone, the data showed that 10 pM dexamethasone did not affect neutrophil migration, but co-treatment with 50 nM Rg1 caused a significant inhibition of the neutrophil migration to the wounded site (Fig.S1B,C). This result indicated that the additive anti-inflammatory effect of Rg1 is not only observed upon co-treatment with beclomethasone but can be observed with other GR agonists as well.
To explain the observed effects and translate the results obtained in our zebrafish model to the human situation, we first studied the binding of beclomethasone and Rg1 to the human GR in vitro using a competitive ligand binding assay. The results showed that beclomethasone had a high relative affinity binding to GR, as shown by the IC50 value of 4.9 nM (Fig.2A). Rgl had a dramatically (-4,000 times) lower relative binding affinity than the beclomethasone, reflected by an IC50 of 22 uM (Fig.2A). Using immunocytochemistry on Hela cell cultures, the translocation of the GR to the nucleus was determined after 6 h of administration of different concentrations of beclomethasone (0.01, 0. 1, and 1 pM), and Rg1 (5, 10, and 20 pM). The concentration of 0.01 pM beclomethasone significantly increased the translocation level compared to vehicle, and maximal nuclear translocation was observed after treatment with 0.1 and 1 pM beclomethasone (Fig.2B,C). Rgl only increased the translocation level compared to the vehicle treatment at 20 pM, still inducing only partial translocation (Fig.2B,C).
To study the anti-inflammatory effects of Rgl in combination with beclomethasone in human cells, we determined the anti-inflammatory effects of beclomethasone, Rgl, and the combination of these compounds on the pro- inflammatory genes IL1B, and IL8 after short-term treatment (6 h). For this purpose, beclomethasone was administered at a concentration of 0.01 pM and Rgl at 20 pM, which are both concentrations that did not trigger maximal translocation of GR. HeLa cells were treated with TNF-a, which induced the expression of IL 1B, and LS. This induction was slightly suppressed by 0.01 uM beclomethasone and 20 uM Rg1 as individual treatments (Fig.2D). Interestingly, Rg1 (20 pM) in combination with beclomethasone (0.01 uM) strongly suppressed the induction of the IL1B, and IL8 after (6 h) treatment significantly (Fig.2D).
To confirm the anti-inflammatory effects of Rgl in combination with GCs, we determined the mRNA levels of two inflammation-related genes, il1b and il6 by qPCR at 4 h after wounding (Fig.1F). The cytokine-encoding genes il/1b and 7/6 have previously been shown to be transrepressed in this assay upon treatment with beclomethasone or Rg1 [17]. The results showed that the cytokine-encoding genes tl 1b and 7/6 were upregulated after wounding, and that both 10 nM beclomethasone and 50 pM Rg1 as individual treatments slightly decreased the expression of these genes (Fig. 1F). Interestingly, the combination treatment further reduced the expression of il 1b and 716 significantly (Fig. 1F).
Ginsenoside Rgl antagonizes GC-induced side effects in zebrafish
In zebrafish larvae, different biomarkers can be studied as readouts to model side effects of GCs: tissue regeneration [17], larval length, glucose and cortisol levels [19], and the expression of several Gr target genes. In the present study, we have used these biomarkers to examine whether co-treatment with Rgl could reduce the side effects induced by GCs.
First, a tail fin regeneration assay was performed, in which tail fins are amputated (similarly to the wounding assay) at 2 dpf, and the length of the regenerated fin tissue is determined at 5 dpf. The zebrafish were incubated with compound treatments from 0 to 5 dpf. Our data showed that the vehicle-treated larvae fully regenerated their amputated tail, that beclomethasone (10 uM) inhibited the regeneration, and that Rgl (50 uM) did not affect the regenerative process, as previously shown [17]. Interestingly, the Rg1 (50 uM)/beclomethasone (10 uM) combination treatment resulted in inhibition of the regeneration, but this inhibition was strongly reduced compared to the beclomethasone treatment (Fig.3A,S2A). Thus, our data showed that Rgl decreased the beclomethasone- induced inhibition of tissue regeneration. To confirm this result using another GC, we studied the effects of Rgl in combination with dexamethasone (10 uM) in the same assay (Fig.S2A,B). The results showed that Rg1 also reduced the dexamethasone-induced inhibition of the regenerative process, which suggests that
Rg1 generally antagonizes the inhibition of regeneration by GCs.
Second, we studied the effect of the Rgl/beclomethasone combination on the growth of zebrafish. For this purpose, we measured the larval length at 5 dpf after exposing the zebrafish to compound treatments for the entire five-day period (from 0 to 5 dpf). Our data demonstrated that the larval length was significantly decreased when beclomethasone was administered, compared to the effect of vehicle treatment. However, administration of Rgl did not affect the larval length, and neither did the co-treatment (Fig.3B), suggesting that Rgl antagonized the effect of beclomethasone on larval growth.
Third, we studied the effects on whole body glucose and cortisol levels at 5 dpf, which are indicators for GC-induced disruption of the metabolic and endocrine systems, respectively. Larvae were treated over a five-day period (0-5 dpf) for the glucose measurements (assessed using a colorimetric assay) and for four days (0-4 dpf) to determine the effects on cortisol levels (by ELISA). Our results demonstrated that the larvae treated with beclomethasone displayed elevated whole body glucose and reduced whole body cortisol levels compared to the levels of the vehicle-treated larvae (Fig.3C,E). In contrast, Rgl did not affect the whole-body glucose and cortisol concentrations. In the larvae treated with beclomethasone in combination with Rg1, the whole body glucose level was not affected, while the whole body cortisol concentration was only slightly decreased compared to the vehicle-treated larvae (Fig.3C,E). These data indicate that Rgl antagonizes the effects of beclomethasone on the metabolic and endocrine systems.
To determine whether the antagonistic effects of Rgl on beclomethasone on the glucose and cortisol levels are competitive or non-competitive, dose-response curves were generated for beclomethasone (0.1, 1, 5, 10, and 15 pM) in the absence and presence of Rg1 (50 uM). Our data showed a dose-dependent effect of beclomethasone on the whole-body glucose and cortisol levels. Interestingly, there were significant differences between the groups treated with beclomethasone and the groups treated with the same concentration of beclomethasone in combination with 50 uM Rg1, and the curves of the co-treatment showed a shift to the right compared to the beclomethasone dose-response curve, suggesting that Rgl acts as a competitive antagonist to beclomethasone (Fig.3D,F).
It is generally believed that many of the side effects of GC treatment are a result of the transactivation activity of GR. To study if Rgl inhibits the transactivation activity of the beclomethasone-activated Gr in zebrafish, the
Te(9xGCRE-HSV. UI23: EGFP)ia2 reporter zebrafish line was used. In this line, the
EGFP gene expression is driven by a GRE-containing promoter, so the transactivation activity of the Gr can be determined by measuring the EGFP intensity in the larvae.
Larvae at 2 dpf were treated for 24 h, after which they were fixed and visualized under a stereomicroscope to determine the EGFP signal intensity in their bodies, and the relative fluorescence intensity was determined by normalization of the EGFP signal to that of the vehicle group. Our results demonstrated that beclomethasone, as well as dexamethasone significantly increased the relative EGFP intensity (Fig. 3G,H,S2C). Importantly, Rg1 treatment did not affect the EGFP signal, and in the co-treatment groups, Rg1 abolished the beclomethasone- (and dexamethasone-)induced increase in the EGFP signal (Fig.3G,H,S2C). These data indicate that Rg1 antagonizes the GC-induced transactivation activity of Gr in zebrafish.
To further investigate the inhibitory effects of Rg1 on the transactivation activity of the beclomethasone-activated Gr, the expression levels of two endogenous Gr target genes, pckl1, and fhbp 5, were determined by qPCR at 8 dpf after 6 h term treatment in wounded zebrafish larvae. Our data showed that, while beclomethasone upregulated the expression of peck 1 and {kbp5, Rg1 did not affect the expression of these genes (Fig.3D). Interestingly, the beclomethasone/Rg1 co-
treatment did not affect the expression of either of the genes (Fig.31). These data confirm the antagonistic effect of Rgl on the transactivation activity of Gr in zebrafish.
GC sensitivity is not reduced after long-term beclomethasone and
Rgl co-treatment
We studied if treatment with beclomethasone for a longer time can cause
GC resistance in the zebrafish model. To do this, we studied the effects of beclomethasone on the endogenous Gr target genes {kbp5, pck 1, and nfkbiaa at 5dpf after short-term (6 h) and long-term (5 days, from 0 to 5 dpf) treatment. Our data showed that beclomethasone (10 uM) upregulated the expression of fkbp5 (Fig. 4A), pck 1 (Fig.4B), and nfkbiaa (Fig.4C) after short-term treatment, but this upregulation of pek I and nfkbiaa was significantly reduced after long-term treatment (Fig.4A,B), while the upregulation of fkbp5 did not decrease (Fig.4A).
Rgl (50 uM) did not affect the expression of the genes after either the short- or long-term treatment. Importantly, Rgl (50 uM) in combination with beclomethasone (10 uM) upregulated the expression of those genes similarly after short- and long-term treatment, indicating that the larvae do not lose GC sensitivity during the long-term combination treatment (Fig.4A-C).
To investigate the effects of beclomethasone and Rg1 treatment on the expression levels of gr in the zebrafish larvae, a qPCR analysis was performed after short- and long-term treatment at 5 dpf (Fig.4D). The results of this experiment demonstrated that beclomethasone suppressed the gr mRNA level only after long- term treatment. Rgl did not affect the gr mRNA level, but did reduce the inhibitory effect of beclomethasone on gr expression after long-term co-treatment. In fact, the co-treatment groups did not show an inhibitory effect on the gr mRNA concentration (Fig.4D). These data indicate that Rgl can reduce the beclomethasone-induced suppression of the gr expression, which may explain why the GC sensitivity is not reduced after the Rgl/beclomethasone co-treatment.
To study the effects of Rg1 on the beclomethasone-induced reduction in
GR sensitivity in human cells, we first determined the effects of beclomethasone,
Rgl, and the combination of these compounds on the pro-inflammatory gene ILIB after short- (6 h) and long-term (30 h) treatment in HeLa cells. For this purpose,
beclomethasone was administered at concentrations of 0.01, 0.1, and 1 uM and Rgl at 20, 100, and 500 uM. Cells were co-treated with TNF-a for the final 6 h. This
TNF-a treatment increased the expression of IL 1B, and this increase was strongly suppressed by 0.1 and 1 pM beclomethasone after short-term treatment. However, after long-term treatment a significant reduction in the suppressive effects of these high doses of beclomethasone was observed (Fig.5A). All three doses of Rgl caused a decrease in IL 1B expression after short-term treatment, which was not significantly different after long-term treatment. Similarly, Rg1 in combination with beclomethasone inhibited the induction of the IL 1B expression after both short- and long-term treatment, indicating that no loss of GC sensitivity occurs during the long-term combination treatment. Similar data were obtained for Rgl (20 uM) in combination with dexamethasone (1 uM) on the expression of ILIB (Fig.S4C).
To investigate the effect of Rg1 on the reduced GC sensitivity after long- term treatment in more detail, the effect of two different concentrations of beclomethasone (0.01 and 1 pM) in combination with Rg1 (20 nM) was studied on the expression of IL1B at several time points during short- and long-term treatment in combination with TNF-a treatment. The results demonstrated that the high dose of beclomethasone (1 nM) exhibited a strong suppressive effect on the
ILIB expression after short-term treatment, whereas the low concentration (0.01 pM) hardly showed an effect. The inhibitory effect of the high dose of beclomethasone was relatively stable over the 6 h of treatment, and Rgl had a minor additive effect when it was co-administered (Fig.S4A). However, after long- term treatment, the high dose of beclomethasone (1 nM) did not show a significant suppressive effect on the expression of IL 1B, whereas the low dose (0.01 pM) did (Fig.S4B). Interestingly, Rg1 co-treatment with the high dose strongly suppressed the IL1B. These data indicated that the GC sensitivity of HeLa cells which is reduced after a long-term and high-dose treatment with beclomethasone can be prevented by Rg1 co-treatment.
Subsequently, we investigated the effects of Rgl (20 nM), beclomethasone (1 uM), and Rg1 in combination with beclomethasone on the expression of the pro- inflammatory genes MMP3 and ILS after short- and long-term treatments. The results showed that the suppressive effects of beclomethasone were significantly reduced after long-term treatment. In contrast, Rgl slightly suppressed the expression of MMP9, and IL8 after short-term treatment, and more strongly after long-term treatment (Fig.5B,C). Interestingly, Rg1 in combination with beclomethasone strongly decreased the expression of both genes after both short- and long-term treatment (Fig. 5B,C). These data confirm that Rgl co-treatment prevents the reduced GC sensitivity that is observed after long-term GC treatment.
In addition to these effects on the transrepression activity of GR in HeLa cells, we studied the effects of beclomethasone (1 uM) and Rg1 (50 uM) on the transactivation activity of GR. For this purpose, we measured the expression of the
GR target genes FKBP5, NFKBIA, GILZ and SGK1 after short- and long-term treatment. Beclomethasone strongly increased the expression of these genes after short-term treatment (Fig.5D-G). The beclomethasone-induced expression level of
FKBP5 was increased over time (i.e., higher after long-term treatment compared to short-term treatment), while the beclomethasone-induced expression levels of
NFKBIA, GILZ, and SGK1 were reduced over time. Rg1 alone did not affect the expression of these genes after either short- or long-term treatment. After short- term co-treatment, Rgl had an antagonistic effect on the transactivation activity, reducing the beclomethasone-induced expression of the studied genes, while after long-term co-treatment Rg1 abolished the beclomethasone-induced increase in
FKBPS5 expression, slightly increased the transactivation activities of NFKBIA and
GILZ, and did not significantly alter the expression of SGA (Fig.5D-G). Similar data were observed for the expression of NFKBIA for the dexamethasone(1uM)/Rg1(20uM) combination (Fig.S4D). These data show that the reduced sensitivity after long-term GC treatment is not common to all genes that are transactivated by GR, but for genes that display this reduced sensitivity, it may be prevented by co-treatment with Rgl.
Rgl inhibits the beclomethasone-induced homologous downregulation of GR
To explain the observed changes in GC sensitivity and confirm the observed effect on the zebrafish gr expression in human cells, the GR mRNA and protein levels were assessed in HeLa cells by qPCR and western blot analysis after short- (6 h) and long-term (24 h) treatment with beclomethasone (1 pM) and Rgl
(20 nM). Our data showed that beclomethasone inhibited the expression of GR at both the mRNA and protein level, and that this inhibitory effect increased over time, whereas Rgl did not affect the GR mRNA or GR protein level (Fig.6A,B,S5A,B). Interestingly, the Rgl/beclomethasone combination treatment did not affect the GR mRNA concentration after either the short- or long-term treatment compared to the vehicle treatment (Fig.6A), induced only a slight reduction of the GR protein level in the short-term treatment, and did not affect the protein level in the long-term treatment (Fig.6B,S5A,B). These data indicated that beclomethasone decreases the GR mRNA and GR protein levels, and that Rgl does not affect these levels, but that co-treatment with Rg1 can inhibit the beclomethasone-induced reduction in GR expression.
To discriminate between effects on transcription and translation versus effects on mRNA and protein stability, we studied the effect of long-term beclomethasone/Rgl co-treatment in the presence of the transcription inhibitor actinomycin-D (1 ng/ml) or the protein synthesis inhibitor cycloheximide (5 pg/ml).
This way, the effects of different treatments on mRNA and protein stability could be determined. Our results showed that, as expected, actinomycin-D and cycloheximide significantly reduced the GR mRNA and protein concentration in
Hela cells treated with vehicle, beclomethasone, Rg1, or Rgl in combination with beclomethasone, although the effect on the mRNA level in the beclomethasone- treated group did not reach significance (Fig. 6A,C,S5C). When we compared the mRNA levels of the actinomycin-D-treated groups, we found that beclomethasone decreased the mRNA levels when transcription is blocked, whereas Rgl and beclomethasone/Rg1 did not alter the GR mRNA levels (Fig. 6A). Apparently, beclomethasone, and not Rgl, decreases the stability of GR mRNA. Similar data were observed for the protein levels in the presence of cycloheximide (Fig. 6C,S5C), indicating that protein stability is affected in a similar way as the mRNA levels by beclomethasone, and not Rgl. Taken together, these data demonstrate that the effect of beclomethasone on the GR expression level is largely due to a reduced mRNA and protein stability and that this reduction in stability is prevented by co- treatment with Rgl.
Example 2: Anti-inflammatory effect of PPT, F1, Rgl and Rh1 and their effects on glucose and cortisol levels, the larval length, and tissue regeneration upon wounding
Materials and Methods
Zebrafish Lines and Maintenance
Zebrafish (Danio rerio) were maintained and handled according to the guidelines from the Zebrafish Model Organism Database (http://zfin.org) and in compliance with the directives of the local animal welfare committee of Leiden
University. Zebrafish were exposed to a 14 h light and 10 h dark cycle to maintain circadian rhythmicity. Fertilization was performed by natural spawning at the beginning of the light period. Eggs were collected and raised at 28°C in egg water (60 pg/ml Instant Ocean sea salts and 0.0025% methylene blue). The following zebrafish lines were used in this study: AB/TL wild type, the transgenic lines Te(mpx:GEFP414/ mpeg 1:mCherryersF00)[ 14, 15] and Te(9xGCRE-
HSV.UI23:EGFPia®){16], and the mutant lines grs857{20].
Chemicals
The chemieal compounds beclomethasone, prednisolone, dexamethasone, glucuronide-dexamethasone (GDex), PPT, Rhl, F1, Rgl, actinomycin-D, and cycloheximide were purchased from Sigma-Aldrich (St. Louis, MO, USA). Glucose-- prednisolone (GPdn) was synthesized in our laboratory, as described previously
[21]. MZ31 was kindly provided by Prof. Dr. Hans Aerts (Leiden University, The
Netherlands).
Fish Embryo Acute Toxicity Test (FET)
Fish Embryo Acute Toxicity Test (FET) was performed to determine the tolerated doses of the studied compounds. Because of the relatively high costs of the studied ginsenosides, the FET guidelines of the Organization for Economie
Cooperation and Development (OECD,
No.236 hitpdidx.dolorg/10.1787/4 3788284203708 en) [22]) were adapted according to a previously published protocol that has succesfully been used for testing nanoparticles[23], [17,21]. A range of three test concentrations (100, 120, and 150 pM) of Rg1, Rhl, and F1 and (25, 50, and 75nM) of PPT was used. A range of four test concentrations of Beclomethasone was used (5, 10, 20, and 25 pM).
Stock solutions were made in DMSO, and final dilutions in egg water (60 pg/mL
Instant Ocean Sea salts and 0.0025% methylene blue), such that the final DMSO concentration were 0.01%. The following controls were used: a negative control (nC, egg water), a solvent control (sC, 0.01% DMSO in egg water), and positive control (pC), 4 mg/L 3,4-dichloroaniline (Sigma-Aldrich, St. Louis, MO, USA) in egg water.
Embryos were collected around at 1.5 hpf and distributed over standard twenty-four well plates (10 embryos per well), with each well containing 2 mL of test solution. The transgenic line Tg (mpx: GFPi114/ mpeg I:mCherry-FumsF00D) was used for proper comparison with other experiments in this study. Five 24-well plates were prepared, each with 3 wells containing the Rg1, F1, or Rh1 solutions (100, 120, 150nM), 3 wells containing the PPT (25, 50, and 75uM), two for nC and $C, and one for pC (on each plate, 7 wells remained empty). All solutions were refreshed daily.
The plates were kept at a temperature between 26 and 27 °C and exposed to a 12 h light and 12 h dark cycle. At 96, and 120 hpf, the average survival rate (in %) for all experimental groups was determined. Additionally, hatching was recorded at 48 and 72 hpf. Survival rates for the studied compound and pC-treated groups from the five individual plates were averaged. At 96 hpf, the hatching rates (280% for nC and sC) and the survival rates (290% for nC and sC, <70% for pC) were within the criteria for test validity.
Tail fin wounding assay in zebrafish larvae
For the tail fin wounding experiments, 2- or 3- days post-fertilization (dpf) larvae were anesthetized in egg water containing 0.02% buffered aminobenzoic acid ethyl ester (tricaine; Sigma-Aldrich). Larvae were placed in
Petri dishes coated with 2% agarose under a Leica M165C stereomicroscope (Leica
Microsystems, Wetzlar, Germany), and the tails were partly amputated using a 1- mm sapphire blade (World Precision Instruments, Sarasota, FL, USA).
Quantification of leukocyte migration
In experiments in which the leukocyte migration was used as a readout, larvae were subjected to treatment with chemicals as indicated, starting at 2 h before the amputation (pretreatment), and treatment was continued for 4 h after the tail fin amputation (20 larvae per group, unless indicated otherwise). In some experiments, a 24 h treatment with MZ31 (10 pM) was started at 2 dpf, and these treatments were continued during the pretreatment and treatment with the other chemicals. At 4 h after amputation, the larvae were fixed in 4% paraformaldehyde (PFA) overnight at 4°C. The next day, fixed larvae were washed with Phosphate
Buffered Saline (PBS) containing 0.1% Tween 20 and stored at 4°C until imaging.
Imaging of larvae from the Tg (mpx: GFP / mpeg I:mCherrywsF001) line was performed utilizing a Leica MZ16FA fluorescence stereomicroscope supported by
LAS 3.7 software (Leica Microsystems). The macrophages were detected based on red (mCherry) fluorescence and neutrophils on their green (EGFP) fluorescence. To quantify the number of macrophages and neutrophils recruited to the wounded area, the cells in a defined area of the tail were determined by blinded manual counting. When larvae from the 275357 mutant lines were used, neutrophil labeling was performed with the TSA fluorescein detection kit (PerkinElmer) for specific staining of Myeloperoxidase (Mpx)-positive cells, following the manufacturer’s instructions.
Measurement of larval body length and regeneration of the tail fin
In experiments in which the length of regenerated tail fin tissue after wounding, or the larval body length, was used as a readout, chemical treatments were started at 2 hours post-fertilization (hpf) and continued until 5 dpf, as indicated (15 larvae per treatment group). During this period, solutions were refreshed daily. For the regeneration experiments, tail fins were amputated at 2 dpf. At 5 dpf, larvae were fixed overnight in 4% PFA at 4°C. For determination of the larval length and the regeneration of the tail fins, larvae were imaged using a
LeicaMZ16FA fluorescence stereomicroscope supported by LAS 3.7 software. The length of the entire larva or the newly grown tissue was measured using Image.
software. The newly grown tissue can be visually distinguished from the old tissue, enabling precise measurement of its length.
Whole-body glucose measurement of zebrafish larvae
Zebrafish embryos at 2 hpf received a chemical treatment with or without
MZ31, with a daily refreshment of the solutions, until 5 dpf. Tail fin wounding were performed at 2 days post-fertilization (dpf). At 5 dpf, the larvae (15 per sample) were placed in egg water for 1 h and collected in an Eppendorf tube, washed in egg water (3 times for 10 min). Subsequently, 100 pL of ice-cold glucose buffer was added to each sample, and the larvae were homogenized using a
BulletBlender® for 3 min at 8,000 rpm. The homogenates were then centrifuged at 4°C for 8 min at 11,000 rpm and the supernatant was stored at -20°C. Whole-body glucose concentrations were determined using a Glucose Colorimetric Assay kit (Cayman Chemical, Ann Arbor, MI, USA), according to the manufacturer’s instructions. In each experiment, three biological replicates were used for each treatment group, and the colorimetric assay was performed using technical duplicates.
Whole-body cortisol measurement of zebrafish larvae
Zebrafish embryos at 2 hpf received a chemical treatment with or without
MZ31, with a daily refreshment of the solutions, until 96 hpf. Tail fin wounding were performed at 2 days post-fertilization (dpf). At 96hpf, the treatments were stopped, and larvae were incubated in egg water until 5 dpf to avoid cross- reactivity between the compounds and the cortisol antibody used in the ELISA. At 5 dpf the larvae were collected in an Eppendorf tube (30 larvae per sample), and 100 ul of ice-cold egg water was added. After the excess water had been taken out, the samples were glaciated in ethanol (EtOH)/dry-ice bath. Subsequently, the larvae were homogenized using a BulletBlender® for 3 min at 8,000 rpm. Ethyl acetate was added to the homogenate. The homogenates were then centrifuged at 4°C for 8 min at 11,000 rpm, and the supernatant was collected and vaporized. A volume of 150 nl of 0.2% bovine serum albumin (Sigma-Aldrich) dissolved in PBS was added to the samples and frozen. Whole-body cortisol levels of the zebrafish larvae were determined using a Cortisol free in Saliva ELISA kit (Demeditec
Diagnostics GmbH, Kiel-Wellsee, Germany), following the manufacturer's instructions. In each experiment, three biological replicates were used for each treatment group, and the ELISA was performed using technical duplicates.
Determination of the Gr transactivation activity in zebrafish larvae
To investigate the Gr transactivation activity in zebrafish larvae, the Tg(9xGCRE-HSV.UI23: EGF Pie?) reporter line was used, which expresses enhanced green fluorescent protein (EGFP) under the control of a promoter containing an array of nine GREs [16]. To study the effect of chemical treatment, 2 dpf zebrafish embryos (15 per group) were treated with indicated compounds for 24 h and fixed for visualization to measure the whole-body fluorescence intensity utilizing a Leica MZ16FA fluorescence stereomicroscope, supported by LAS 3.7 software (Leica Microsystems). The integrated intensity of the EGFP signal in the larvae was determined using Imaged software.
Overexpression of gba2 in zebrafish
For overexpression of gba2 in zebrafish, a plasmid containing the cDNA encoding the zebrafish gba2 gene fused to a CMV promoter (pDEST-zeo-zGBA2) [21,23,24] was used. One-cell stage zebrafish embryos were injected with this plasmid, diluted in nuclease-free water (1 nl/egg with a final concentration of 80 pg/egg), using the Automated Microinjection System Version 3 AMS-03 (Life
Science Methods BV, Leiden, The Netherlands). After the injection, chemical treatment was performed as indicated.
Quantitative PCR (qPCR) analysis
To determine mRNA levels of specifie genes in zebrafish, 3 dpf larvae were used (wounded and non-wounded) and after 6 h of chemical treatments (2 h pre-wounding, 4 h post-wounding), as indicated. Per sample, groups of 15 larvae were collected in TRIzol reagent (Thermo Fisher Scientific, Waltham, MA, USA), and total RNA was isolated using the miRNeasy mini kit (Qiagen, Hilden,
Germany). For similar experiments in Hela cells, cells were seeded in 6 well plates, and after 6 h of chemical treatment, the cells were removed from the wells using TRIzol reagent, and total RNA was isolated using the miRNeasy mini kit according to the manufacturer's instructions. The RNA samples were DNAse- treated utilizing the DNA-free™ DNA Removal Kit (Thermo Fisher Scientific). The cDNA synthesis was performed using the iScript cDNA synthesis kit (Bio-Rad
Laboratories, Hercules, CA, USA) using 1 ng of RNA per sample. For the PCR reactions, 10 uM of forward and 10 uM of reverse primer, 12.5 ul of iQ SYBR Green
Supermix (Bio-Rad), and 2 pl of cDNA were added to the qPCR reaction mixture.
Each mixture had a total volume of 25 uL, which was split as duplicates with a volume of 12.5 uL. The reactions were performed on a MyiQ-single-color real-time
PCR detection system (Bio-Rad Laboratories) with initial denaturation for 3 min at 95°C and 40 cycles of 15 s at 95.5°C, 15 s at 60°C, and 30 s at 72°C. Cycle threshold values (Ct values, i.e. the cycle numbers at which a threshold value of the fluorescence intensity was reached) were determined for each sample. The gene expression level for each sample was normalized using the expression of ppial (peptidylprolyl isomerase Ab (cyclophilin A)) for zebrafish samples, 18S rRNA for human cells. The fold change per sample (compared to the respective control group) was calculated using the AACt method. In each experiment, three biological replicates were used for each treatment group, and reactions were performed in duplicates. The sequences of the qPCR primers for experiments on zebrafish, and HeLa cells are presented in Table 1 and 2, respectively.
Table 1. Zebrafish qPCR primers
Name Sequence 57-3 ppail forward CATCCACAACCTTCCCGAACAC ppail_reverse ACACTGAAACACGGAGGCAAAG ith forward TGTOTGTITIGGGAATCTECA il1h_reverse CTGATAAACCAACCGGGACA ilo forward COCTAAGGCAACTGGAAGAC i16_reverse CCAGACCACTGGGAAACACT iS: forward TGTGTITATTGTITICCTGGCATTIC iI8 reverse GCGACAGCGTGGATCTACAG mp9 forward CATTAAAGATGCCECTGATGTATECE mmp9_reverse AGTGGTGGTCCGTGGTTGAG wonpl3a forward ATGGTGCAAGGUTATCCCAAGAGT mmpl3a_reverse GUCTGTITGTTGGAGCCAAACTCAA
JkbpS: forward TETGCEAGCACAAGATTEGTGAGE fkbp5_reverse GACCCTGCTTATTCTGATCGGAAA pekl forward CAGGGEGATETGGEGTETET pekl_reverse CTGCTGTCGATGAACTCCCG nfkliaa forward CTTGGGCTAAAGTAGTCACCE nfkbiaa reverse GATGGCAAGGTGCAGATACGTG
Table2. Human qPCR primers
Name Sequence 5°-3°
SIR forward GHIGOGUGEGGE EAE $18 reverse CEPGT ACTH GNA TION
FKBPS forward GOEAAGEAGAAGACCATGACAT
FKBP5_ reverse TAGGCTTCCCTGCOTCTCOAAA
PCE forward CATTGOCTA IAT GAAGTITGACG
PCKL reverse GGGTTGGTCTTCACTGAAGTCC
GILZ forward CETGGTGGECATAGAC AAC
GILZ reverse COTCTCTCACAGCATACATCAGATG
NEKBIA forward CTCCGAGACTIFCGAGGAAATAC
NFKBIA reverse GOCATTGAAGTTGGTAGCCTTCA
ILIB forward CEHCAGACCTTOCAGGAGAATG
ILIB revese GTGCAGTTCAGTIGATCGTACAGG
CXCLS forward GACAGIGATPGAGAGTGGACEAE
CXCL8 reverse CACAACCCTCTGCACCCAGTIT
MMPI3. forward CCTTGATGECATTACCAGTE TEC
MMP13_reverse AAACAGCTCCGCATCAACCTGC
GR-forward
GR- reverse TGGAAGCAGTAGGTAAGGAGA
Cell culture and transfection
HeLa (human cervical cancer) cells, purchased from (ATCC), were cultured in Dulbecco's Modified Eagle's (DMEM) High Glucose (HG) medium without Phenol Red, supplemented with 10% Fetal Calf Serum (FCS) and 10%
Glutamax (Sigma-Aldrich). The cells were maintained at 37°C and 5% CO». At 24 hours before treatment, cells were seeded in 6 well plates and allowed to adhere in the presence or absence of MZ31(1nM). MZ31 was added overnight. After adherence and reaching 80% confluence, cells were treated with indicated chemicals for 6 h, in the presence or the absence of TNF-a (10 ng/ml) (Sigma-
Aldrich) and/or MZ31(1pM).
For overexpression of GBAZ in Hel.a cells, a plasmid containing the cDNA encoding the human GBA2 gene fused to a CMV promoter (pDEST-zeo- hGBA2)[21,23,24] was transfected into ~70% confluent HeLa cells. This plasmid was mixed with FuGENE HD transfection reagent (Promega, Madison, WI, USA) and 500 nl of serum-free DMEM. The mixture was incubated for 20 min at room temperature and added to the HeLa cells cultured in supplemented DMEM. After 2 days, the medium was changed, and chemical treatments were performed, as indicated, for 6 h.
Statistical analysis
Statistical analysis of the experiments was performed using GraphPad
Prism software by performing one- or two-way ANOVA with Tukey's post hoc test.
The anti-inflammatory action of monoglycosylated ginsenosides depends on Glucocorticoid receptor function, and not on deglycosylation by Glucosylceramidase beta 2
To study whether monoglycosylated ginsenosides, such as F1 and Rh1, have similar effects and dependency on Gr and/or GbaZ2, we studied their anti- inflammatory action in zebrafish larvae. To investigate the structure-function relationship of these compounds, we used the ginsenosides Protopanaxatriol (PPT),
F1, Rh1 and Rg1, and the glucocorticoid drug beclomethasone (Bec) as a positive control (compounds structures are shown in Fig. 7A). PPT is not glycosylated, and
F1 and Rh1 consist of the aglycone PPT with one glucose group at the C-20 or the
C-6 position of the PPT backbone structure, respectively. Rg1 consists of the aglycone PPT with one glucose group attached at the C-6 and one glucose group at the C-20 position. First, to determine the appropriate doses of the studied ginsenosides in this study, a Fish Embryo Acute Toxicity Test (FET) was performed in which zebrafish embryos were exposed to a concentration range of these compounds between 0 and 120 hpf, and the effects on hatching and survival were determined (Fig.S7A, B). For PPT, no effects were observed until 50 pM, whereas considerable effects were shown at 75 pM. For F1, Rh1, and Rgl, no effects were observed until 100 pM, and minimal effects were observed at 120 pM.
Based on these results, we selected doses of 50 pM for PPT, and 100 nM for F1,
Rh1, and Rgl.
Using these doses, we determined the anti-inflammatory effect of these compounds in zebrafish larvae. We induced a local inflammation in zebrafish larvae at 3 days post fertilization (dpf) by wounding the tail fin (a schematic drawing of the experiment is shown in Fig.7B). Our results showed that treatment of the larvae with the ginsenosides PPT, F1, Rhl, and Rgl, like Bec treatment, inhibited the migration of neutrophils without affecting the macrophage migration (Fig.7C). For all compounds, the inhibition of neutrophil migration was abolished in zebrafish larvae from a mutant line with a deficiency in Gr function (Fig.S7C), indicating that the anti-inflammatory effects of studied ginsenosides are mediated by Gr.
Further, we investigated whether the anti-inflammatory effects of monoglycosylated ginsenosides also required the action of Gba2 using the specific
GBA2 inhibitor MZ31. Treatment with MZ31 did not affect the reduction in neutrophil migration by the monoglycosylated ginsenosides F1 and Rh1, or the aglycone ginsenoside PPT. In contrast, this inhibitor abolished the effect of the polyglycosylated ginsenoside Rg1 (Fig.7D). Taken together, these data indicate that the anti-inflammatory effects of monoglycosylated ginsenosides F1 and Rh1 are independent of Gba2, suggesting that addition of a single glucose group at either the C-6 or C-20 position of PPT does not interfere with its activation of Gr, whereas the presence of glucose groups at both positions requires deglycosylation by Gba2 before it can activate Gr.
To further study the anti-inflammatory effects of the ginsenosides, we studied their effects on the expression of genes encoding pro-inflammatory proteins. Our results demonstrated that there is selectivity in the suppression of inflammation-induced gene expression by ginsenosides. All studied ginsenosides effectively suppressed the expression levels of il1b, il6 and mmp9, similarly to Bec (Fig.7E). However, ginsenosides do not suppress the expression of 1/8 and mmp 13 as effectively as Bec. Only a minor effect of PPT and F1 on the expression of these two genes was found, and no effect of Rh1 and Rgl was observed (Fig.7E). The characteristic that distinguishes Rh1 and Rg1 from PPT and F1 is the glucose group at the C-6 position, so based on these data we suggest that the glucose at the
C-6 position in Rh1 and Rgl is not cleaved off by GbaZ2, whereas this enzyme effectively removes the glucose group from C-20 in F1. As a result, PPT and F1 have a similar effect, as well as Rh1 and Rg1 on the 1/8 and mmp 13 expression.
Glycosylation of ginsenosides causes a significant reduction in side effects
To evaluate the side effects of the monoglycosylated ginsenosides, zebrafish embryos were treated with Bec, PPT, F1, Rh1, and Rgl, and effects were monitored on the glucose and cortisol levels, the larval length, and tissue regeneration upon wounding. Treatment with Bec, used as a positive control, induced an increase in the glucose level (Fig.8A), a decrease in cortisol level (Fig.8C), a decrease in larval length (Fig.8E), and inhibited the tissue regeneration. (Fig.8F, S8A). The aglyconic ginsenoside PPT did not affect the glucose level (Fig.8A) but did decrease the cortisol concentration (Fig.8C), showed a minor effect on larval length (Fig.8E), and considerably inhibited the regeneration of the tail fin after wounding (Fig.8F, S8A). Interestingly, the glycosylated ginsenosides F1, Rh1, and Rgl did not show any side effects, either on the glucose level (Fig.8A), the cortisol concentration (Fig.8C), the larval length (Fig.8E), or the tissue regeneration (Fig.8F, S8A). Thus, we conclude that glycosylation of PPT abolishes all its side effects.
Interestingly, upon wounding, F1 did not affect the glucose level (Fig.8B) but did show a minor effect on the cortisol level and on tissue regeneration (Fig.8D, F, S8A). These latter effects were Gba2-dependent, which was demonstrated using the Gba2 inhibitor MZ31 (Fig.8D, F), suggesting that upon wounding F1 is deglycosylated due to increased activity of Gba2. In contrast,
Rh1 and Rgl did, upon wounding, still not show any effect on the glucose or cortisol level or on tissue regeneration (Fig.8B, D, F, SBA). These results indicated that
Gba2 could cleave off the glucose groups at C20 in ginsenoside F1 (thereby converting it to PPT), but not at C6 in Rh1 and Rgl, leaving the latter two ginsenosides glycosylated at this position. As a result, glycosylation at the C-6 position (as in Rh1) abolished the side effects of PPT also after wounding, but glycosylation at the C-20 position (as in F1) failed to abolish the side effects of PPT, probably because the glucoses are cleaved off due to the increased local Gba2 activity upon wounding.
To study whether ginsenosides PPT, F1, and Rh1 can trigger GR transactivation activities, we used a reporter fish line in which the GFP gene is fused to a GRE-containing promoter Tg (9xGCRE-HSV.U123:EGFP)s20. Treatment of larvae from this line with beclomethasone resulted in a significant increase in the GFP signal throughout the body of the larvae, whereas none of the ginsenosides PPT, F1, Rhl, and Rgl altered the fluorescence intensity (Fig.8G, H).
Moreover, those ginsenosides did not increase the expression of fkbp5, pck1, and nfkbiaa, which are well-known target genes of the transactivation activity of the Gr (Fig.81, S8B). These data indicate that it is a general characteristic of ginsenosides that they do not induce the transactivation activity of the Gr.
Example 3 Effect of ginsenoside Re in zebrafish
In addition to Rgl, the effect of addition of ginsenoside Re in zebrafish was tested on anti-inflammatory effects and putative side effects as described in
Example 1. It was shown that Re inhibited the migration of neutrophils to the wounded tail area and did not inhibit the migration of macrophages (Figure 9A,B).
In contrast to glucocorticosteroids no side effects could be observed after addition of
Re with respect to whole body glucose and whole body cortisol (Figure 9C,D). In addition Re did not affect regenerative tissue (Figure 9E).
Example 4: Preparation of micelles from ginseng leaves
Dried leaves of the plant Panax ginseng (8 gram) were boiled in water (350 mL), filtrated and loaded on a 20 mL Bed Volume (BV) column filled with macroporous resin (type D101). After extensively washing with water (15 BV's) the ginsenosides were eluted with 60% EtOH. The ginsenoside solution was decolorized by passing through a 20 mL BV column filled with type D941 macroporous resin.
Subsequently, the solvents were slowly evaporated, after at 80 °C under reduced pressure (375 hPa) using a glass vials and a evaporator (P12-Multivapor, Buchi).
After evaporation of both the ethanol and the water, a transparent film was formed on the wall of the glass vial. The film was solved in HzO resulting in a clear solution (see figure 12A) containing about 15 mg/mL ginsenosides.
In parallel 100 mg pure Re was solved in 10 mL of ethanol. This resulted in a clear solution (see figure 12A). The ethanol was slowly removed at 80 °C under low pressure (375 hPa) using a glass vials and a evaporator (P12-Multivapor ).
Again a clear film was formed on the wall of the glass tubes. The film was solved in 10 mL of H20 resulting a cloudy solution (see Figure 12A). Microscopic determination revealed crystallization of Re (Figure 12B) and no micelles were obtained.
Micelle formation of ginsenoside extract obtained from dried leaves as described above was repeated in the presence of 0.6 mg of dexamethasone.
Dexamethasone was first dissolved in ethanol in a concentration of 10 mg/mL and 0.6 mg of dexamethasone was added to the ethanolic ginsenoside fraction obtained after purification and decolorization by D101 and D941 resins respectively, using the method described herein above. The content of Re and Rg1 in this ethanolic ginsenoside fraction was 4 mg/10 mL as determined by HPLC (see below). The molecular ratio between dexamethasone and Re/Rel in this solution was around 5:70. The ethanol fraction was slowly removed at 80 °C under reduced pressure (375 hPa) using a glass vials and a evaporator to obtain a film. The film was dissolved in H20 resulting in a clear aqueous solution. The content and the presence of micelles were assessed by size exclusion chromatography as described below.
The size and content of the micelles were determined by means of size exclusion chromatography. 1 mL of aqueous ginsenoside/dexamethasone solution was loaded on a Sephadex (550 size exclusion column (size 25 x 1 cm) and eluted under gravity with water. Blue dextran (MW = 2.000.000 Da) and erythrosine (880
Da) were used as molecular weight markers. Fractions of 2.0 mL were collected.
The fractions were dried and dissolved in 70% methanol and the ginsenoside content was analyzed by means of HPLC. HPLC conditions were: - HPLC equipment: HPLC/DAD Aligent 1200 series, a quaternary pump connected to a DAD and autosampler. - Stationary phase: Kinetex 2.6 um, C18 100 A, 100 x 4.6 mm — Phenomenex,
USA
- Mobile phase: o A: 0.03% o-phosphoric acid (H2PO.) in Ultrapure H20 o B: 0.03% o-phosphoric acid (HsPO4) in Acetonitrile (LiChrosolv,
Millipore-Merck, Darmstadt, Germany; quality far UV) - Gradient: 12.00 22.0 18.00 30.0 31.00 36.0 33.00 60.0 34.50 60.0 34.51 17.0 36.50 17.0 - Conditions o Flow: 0.8 ml/min o Injection volume: 20 ul. o Column temperature: 35 °C o Detection wavelength: 203 and 241 nm
Figure 13 shows a size exclusion chromatogram showing the molecular weight and content of the micelles.
The highest concentration of micelles with a high content of ginsenosides
Re and Rgl was eluted after 30 mL (Figure 13, Figure 14B). Based on the elution profile of the molecular markers blue dextran (MW 2.000.000 Da; peak elution at 17 mL and erythrosine (MW 880 Da; peak elution at 55 mL) it was estimated that the size of the micelles containing mainly the ginsenosides Re and Rgl was about 80.000 Da
These results indicate that the micelles contain around 50 to 100 molecules of mainly Re, Rg1 or both. Interestingly dexamethasone was also found to be co-eluted with the Re and Rg1 containing micelles. In addition dexamethasone, having a molecular weight of 392.5 Da in a single molecular form, was expected to be eluted after erythrosine e.g. in fractions after 55 mL, but dexamethasone could not be detected in these fractions. This indicates that dexamethasone was encapsulated in a micelle formed by Re, Rg1 or both. Further, it is shown that with this method a high concentration of PPT-type ginsenosides, optionally in combination with dexamethasone, in water, could be obtained.
References 1. Homo-Delarche, F.; Fitzpatrick, F.; Christeff, N.; Nunez, E.A.;
Bach, J.F.; Dardenne, M. Sex steroids, glucocorticoids, stress and autoimmunity. /.
Steroid Biochem. Mol. Biol. 1991, 40, 619-637, doi:10.1016/0960-0760(91)90285-d. 2. Hall, B.M.; Hodgkinson, S.J.; Quin, J. Corticosteroids in autoimmune diseases. Aust. Prescr. 1999, 22 9-11, doi:DOI: 10.18773/austprescr.1999.008. 3. Trombetta, A.C.; Meroni, M.; Cutolo, M. Steroids and
Autoimmunity. Front. Horm. Res. 2017, 48, 121-132. 4. Ashwell, J.D.; Lu, FE.W.M.; Vacchio, M.S. Glucocorticoids in T
Cell Development and Function. Annu. Rev. Immunol. 2000, 18, 309-345. 5. Ratman, D.; Vanden Berghe, W.; Dejager, L.; Libert, C.;
Tavernier, J.; Beck, LM.; De Bosscher, K. How glucocorticoid receptors modulate the activity of other transcription factors: a scope beyond tethering. Mol. Cell.
Endocrinol. 2013, 380, 41-54, doi:10.1016/j.mce.2012.12.014.
6. Reichardt, H.M.; Tuckermann, J.P.; Gottlicher, M.; Vujic, M.;
Weih, F.; Angel, P.; Herrlich, P.; Schutz, G. Repression of inflammatory responses in the absence of DNA binding by the glucocorticoid receptor. EMBO J. 2001, 20, 7168-7173, doi: 10.1093/emboj/20.24.7168. 7. Patel, G.C.; Millar, J.C; Clark, A.F. Glucocorticoid Receptor
Transactivation Is Required for Glucocorticoid-Induced Ocular Hypertension and
Glaucoma. Invest. Ophthalmol. Vis. Sci. 2019, 60, 1967-1978, doi:10.1167/iovs.18- 26383. 8. Tanigawa, K.; Nagase, H.; Ohmori, K.; Tanaka, K.; Miyake, H.;
Kiniwa, M.; Ikizawa, K. Species-specific differences in the glucocorticoid receptor transactivation function upon binding with betamethasone-esters. Int.
Immunopharmacol, 2002, 2, 941-950, doi:10.1016/51567-5769(02)00036-x. 9. Newton, R. Molecular mechanisms of glucocorticoid action: what is important? Thorax 2000, 55, 603-613. 10. Gordijn, M.S.; Gemke, R.J.; van Dalen, E.C.; Rotteveel, J.;
Kaspers, G.J. Hypothalamic-pituitary-adrenal (HPA) axis suppression after treatment with glucocorticoid therapy for childhood acute lymphoblastic leukaemia. Cochrane database Syst. Rev. 2012, CD008727, do0i:10.1002/14651858.CD008727.pub2. 11. Straub, R.H.; Cutolo, M. Glucocorticoids and chronic inflammation. Rheumatology (Oxford). 2016, 55, ii6-iil4, doi:10.1093/rheumatology/kew 348. 12. Schaaf, M.J.M.: Cidlowski, J.A. Molecular mechanisms of glucocorticoid action and resistance. J. Steroid Biochem, Mol. Biol. 2002, 83, 37-48, doi:10.1016/80960-0760(02)00263-7. 13. Wilkinson, L.; Verhoog, N.J.D.; Louw, A. Disease- and treatment-associated acquired glucocorticoid resistance. Endocr. Connect. 2018, 7,
R328-R349, doi:10.1530/EC-18-0421. 14. Renshaw, S.A.; Loynes, C.A; Trushell, D.M.I.; Elworthy, S.;
Ingham, P.W.; Whyte, M.K.B. A transgenic zebrafish model of neutrophilic inflammation. Blood 2006, 108, 3976-3978, doi:10.1182/blood-2006-05-024075. 15 Bernut, A.; Herrmann, J.-L.; Kissa, K.; Dubremetz, J.-F.;
Gaillard, J.-L.; Lutfalla, G.; Kremer, L. Mycobacterium abscessus cording prevents phagocytosis and promotes abscess formation. Proc. Natl. Acad. Sci. U.S. A. 2014, 111, E943-E952, doi:10.1073/pnas. 1821390111. 16 Benato, F.; Colletti, E.; Skobo, T.; Moro, E.; Colombo, L.;
Argenton, F.; Dalla Valle, L. A living biosensor model to dynamically trace glucocorticoid transcriptional activity during development and adult life in zebrafish. Mol. Cell. Endocrinol. 2014, 392, 60-72, doi:10.1016/j.mce.2014.04.015. 17. He, M.; Halima, M.; Xie, Y.; Schaaf, M.J.M.; Meijer, A.H;
Wang, M. Ginsenoside Rg1 Acts as a Selective Glucocorticoid Receptor Agonist with Anti-Inflammatory Action without Affecting Tissue Regeneration in
Zebrafish Larvae. Cells 2020, 9, doi:10.3390/cells9051107. 18. Chatzopoulou, A.; Heijmans, J.P.M.; Burgerhout, E.; Oskam,
N.; Spaink, H.P.; Meijer, A.H; Schaaf, M.J.M. Glucocorticoid-induced attenuation of the inflammatory response in zebrafish. Endocrinology 2016, 157, 2772-2784, doi: 10.1210/en.2015-2050. 19. Wepler, M.; Preuss, J.M.; Merz, T.; McCook, O.; Radermacher,
P.; Tuckermann, J.P.; Vettorazzi, S. Impact of downstream effects of glucocorticoid receptor dysfunction on organ function in critical illness-associated systemic inflammation. Intensive Care Med. Exp. 2020, 8, 37, doi:10.1186/s40635-020-00325-
Z. 20. Ziv, L.; Muto, A.; Schoonheim, P.J.; Meijsing, S.H.; Strasser,
D.; Ingraham, H.A.; Schaaf, M.J.M.; Yamamoto, K.R.; Baier, H. An affective disorder in zebrafish with mutation of the glucocorticoid receptor. Mol. Psychiatry 2013, 18, 681-691, doi: 10.1038/mp.2012.64. 21. Halima, M.; Ayed, K. Al; Kroos, S.; Kampstra, A.S.B.;
Lelieveld, L.T.; Narayanan, A. Glycosylated glucocorticoid prodrugs show strongly reduced side effects due to specific Glucosylceramidase Beta 2 — mediated activation in inflamed tissue Glucocorticoid receptor-dependent anti-inflammatory effects of glycosylated ginsenosides require deg. 22. van Pomeren, M.; Peijnenburg, W.J.G.M.; Brun, N.R.; Vijver,
M.G. A Novel Experimental and Modelling Strategy for Nanoparticle Toxicity
Testing Enabling the Use of Small Quantities. Int. JJ. Environ. Res. Public Health 2017, 14, doi:10.3390/ijerph14111348.
23. Lelieveld, L.T.; Mirzaian, M.; Kuo, C.-L.; Artola, M.; Ferraz,
M.J.; Peter, R.E.A.; Akiyama, H.; Greimel, P.; van den Berg, R.J.B.H.N.;
Overkleeft, H.S; et al. Role of beta-glucosidase 2 in aberrant glycosphingolipid metabolism: model of glucocerebrosidase deficiency in zebrafish. J. Lipid Res. 2019, 60, 1851-1867, doi:10.1194/jlr.RA119000154. 24. Lelieveld, L.T. Zebrafish as research model to study Gaucher disease: Insights into molecular mechanisms; 2020. 25. Park H., Lee, M-K and Park, Q-H. Pattern of molecular aggregation of ginsenosides in aquous solution. J. Korean Agricultural Chemical society 1986, 29(2) 198-206 26. ZFIN The Zebrafish Information Network [Internet]. [cited 2020 Dec 23]. Available from: https://zfin.org/ 27. Halima M, He M. Glycosylated ginsenosides exert their anti- inflammatory activities without promoting side effects by Gba2 mediated cleavage of glucose groups at the inflammation environment. 2020.
Claims (31)
Priority Applications (2)
| Application Number | Priority Date | Filing Date | Title |
|---|---|---|---|
| NL2034780A NL2034780B1 (en) | 2023-05-08 | 2023-05-08 | Combination therapy glucocorticoids and ginsenoside |
| PCT/NL2024/050234 WO2024232756A1 (en) | 2023-05-08 | 2024-05-08 | Combination therapy glucocorticoids and ginsenoside |
Applications Claiming Priority (1)
| Application Number | Priority Date | Filing Date | Title |
|---|---|---|---|
| NL2034780A NL2034780B1 (en) | 2023-05-08 | 2023-05-08 | Combination therapy glucocorticoids and ginsenoside |
Publications (1)
| Publication Number | Publication Date |
|---|---|
| NL2034780B1 true NL2034780B1 (en) | 2024-11-25 |
Family
ID=86852135
Family Applications (1)
| Application Number | Title | Priority Date | Filing Date |
|---|---|---|---|
| NL2034780A NL2034780B1 (en) | 2023-05-08 | 2023-05-08 | Combination therapy glucocorticoids and ginsenoside |
Country Status (2)
| Country | Link |
|---|---|
| NL (1) | NL2034780B1 (en) |
| WO (1) | WO2024232756A1 (en) |
Citations (6)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| CN1985837A (en) * | 2006-12-11 | 2007-06-27 | 广东医学院 | High efficiency and low toxicity externally applied medicine preparation of ginsenoside and triamcinolone acetonide |
| CN101856377A (en) * | 2010-06-19 | 2010-10-13 | 中国人民解放军济南军区第四0一医院 | Compound preparation for treating nephrotic syndromes |
| CN101874820A (en) * | 2010-06-19 | 2010-11-03 | 中国人民解放军济南军区第四○一医院 | Compound preparation for treating systemic lupus erythematosus |
| EP2815746A1 (en) * | 2013-04-28 | 2014-12-24 | Fujian South Pharmaceutical Co., Ltd | Saponin nano-micelle and preparation method, use and pharmaceutical composition thereof |
| CN107648244A (en) * | 2016-07-26 | 2018-02-02 | 中国人民解放军第二军医大学 | Application of the ginsenoside Rg1 in glucocorticoid sensitizer is prepared |
| CN115089593A (en) * | 2022-05-20 | 2022-09-23 | 广东南国药业有限公司 | Dexamethasone preparation and preparation method thereof |
Family Cites Families (3)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| US20180050005A1 (en) * | 2016-08-16 | 2018-02-22 | Janssen Pharmaceutica Nv | Concentrated Solution of 17-Hydroxydocosahexaenoic Acid |
| CN107320448A (en) * | 2017-08-29 | 2017-11-07 | 天津中医药大学 | A kind of spherical micelle for wrapping up ginseng sapoglycoside Rg 3 and preparation method and application |
| WO2021221472A1 (en) * | 2020-04-28 | 2021-11-04 | 주식회사 모든바이오 | Micelle particles comprising amphipathic ginsenoside, composition comprising same, and preparation method thereof |
-
2023
- 2023-05-08 NL NL2034780A patent/NL2034780B1/en active
-
2024
- 2024-05-08 WO PCT/NL2024/050234 patent/WO2024232756A1/en active Pending
Patent Citations (6)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| CN1985837A (en) * | 2006-12-11 | 2007-06-27 | 广东医学院 | High efficiency and low toxicity externally applied medicine preparation of ginsenoside and triamcinolone acetonide |
| CN101856377A (en) * | 2010-06-19 | 2010-10-13 | 中国人民解放军济南军区第四0一医院 | Compound preparation for treating nephrotic syndromes |
| CN101874820A (en) * | 2010-06-19 | 2010-11-03 | 中国人民解放军济南军区第四○一医院 | Compound preparation for treating systemic lupus erythematosus |
| EP2815746A1 (en) * | 2013-04-28 | 2014-12-24 | Fujian South Pharmaceutical Co., Ltd | Saponin nano-micelle and preparation method, use and pharmaceutical composition thereof |
| CN107648244A (en) * | 2016-07-26 | 2018-02-02 | 中国人民解放军第二军医大学 | Application of the ginsenoside Rg1 in glucocorticoid sensitizer is prepared |
| CN115089593A (en) * | 2022-05-20 | 2022-09-23 | 广东南国药业有限公司 | Dexamethasone preparation and preparation method thereof |
Non-Patent Citations (39)
| Title |
|---|
| "Chinese pharmacopeia", vol. I, 2015, pages: 524 - 526 |
| ANUFRIEV ET AL., CARBOHYDR RES, vol. 304, no. 2, 1997, pages 179 - 182 |
| ASHWELL, J.D.LU, F.W.M.VACCHIO, M.S.: "Glucocorticoids in T Cell Development and Function.", ANNU. REV. IMMUNOL., vol. 18, 2000, pages 309 - 345, XP055147070, DOI: 10.1146/annurev.immunol.18.1.309 |
| BENATO, F.COLLETTI, E.SKOBO, T.MORO, E.COLOMBO, L.ARGENTON, F.DALLA VALLE, L.: "A living biosensor model to dynamically trace glucocorticoid transcriptional activity during development and adult life in zebrafish.", MOL. CELL. ENDOCRINOL., vol. 392, 2014, pages 60 - 72, XP028870559, DOI: 10.1016/j.mce.2014.04.015 |
| BERNUT, A.; HERRMANN, J.-L.KISSA, K.DUBREMETZ, J.-F.GAILLARD, J.-L.LUTFALLA, G.KREMER, L.: "Mycobacterium abscessus cording prevents phagocytosis and promotes abscess formation", PROC. NATL. ACAD. SCI. U. S. A., 2014, pages 111 |
| CHATZOPOULOU, A.HEIJMANS, J.P.M.; BURGERHOUT, E.OSKAM, N.SPAINK, H.P.MEIJER, A.H.: "Schaaf, M.J.M. Glucocorticoid-induced attenuation of the inflammatory response in zebrafish.", ENDOCRINOLOGY, vol. 157, 2016, pages 2772 - 2784 |
| DATABASE MEDLINE [online] US NATIONAL LIBRARY OF MEDICINE (NLM), BETHESDA, MD, US; August 2010 (2010-08-01), YOU YAN-LI ET AL: "[Efficacy of ginsenosides combined with prednisone in patients with systemic lupus erythematosus: a prospective, randomized, double-blind, placebo-controlled trial].", XP002810420, Database accession no. NLM20727331 * |
| FANG Q ET AL: "Micelle-mediated extraction and preconcentration of ginsenosides from Chinese herbal medicine", JOURNAL OF CHROMATOGRAPHY A, ELSEVIER, AMSTERDAM, NL, vol. 904, no. 1, 22 December 2000 (2000-12-22), pages 47 - 55, XP004221275, ISSN: 0021-9673, DOI: 10.1016/S0021-9673(00)00911-0 * |
| FENG YINGLU ET AL: "Ginsenoside Rh1 Improves the Effect of Dexamethasone on Autoantibodies Production and Lymphoproliferation in MRL/lpr Mice", EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE, vol. 2015, 1 January 2015 (2015-01-01), US, pages 1 - 8, XP093097467, ISSN: 1741-427X, Retrieved from the Internet <URL:http://downloads.hindawi.com/journals/ecam/2015/727650.pdf> DOI: 10.1155/2015/727650 * |
| GORDIJN, M.S.GEMKE, R.J.VAN DALEN, E.C.ROTTEVEEL, J.KASPERS, G.J.: "Hypothalamic-pituitary-adrenal (HPA) axis suppression after treatment with glucocorticoid therapy for childhood acute lymphoblastic leukaemia.", COCHRANE DATABASE SYST. REV., 2012 |
| HALIMA MHE M., GLYCOSYLATED GINSENOSIDES EXERT THEIR ANTI-INFLAMMATORY ACTIVITIES WITHOUT PROMOTING SIDE EFFECTS BY GBA2 MEDIATED CLEAVAGE OF GLUCOSE GROUPS AT THE INFLAMMATION ENVIRONMENT, 2020 |
| HALIMA, M.AYED, K. ALKROOS, S.KAMPSTRA, A.S.B.LELIEVELD, L.T.NARAYANAN, A.: "Glycosylated glucocorticoid prodrugs show strongly reduced side effects due to specific Glucosylceramidase Beta 2 - mediated activation in inflamed tissue", GLUCOCORTICOID RECEPTOR-DEPENDENT ANTI-INFLAMMATORY EFFECTS OF GLYCOSYLATED GINSENOSIDES REQUIRE DEG |
| HALL, B.M.HODGKINSON, S.J.QUIN, J.: "Corticosteroids in autoimmune diseases.", AUST. PRESCR., vol. 22, 1999, pages 9 - 11 |
| HE, M.HALIMA, M.XIE, Y.SCHAAF, M.J.M.MEIJER, A.H.: "Wang, M. Ginsenoside Rg1 Acts as a Selective Glucocorticoid Receptor Agonist with Anti-Inflammatory Action without Affecting Tissue Regeneration in Zebrafish Larvae.", CELLS, vol. 9, 2020 |
| HOMO-DELARCHE, F.; FITZPATRICK, F.; CHRISTEFF, N.; NUNEZ, E.A.;BACH, J.F.; DARDENNE, M. SEX, J. STEROID BIOCHEM. MOL. BIOL., vol. 40, 1991, pages 619 - 637 |
| HU CATHERINE ET AL: "Comparative analysis of ginsenosides in human glucocorticoid receptor binding, transactivation, and transrepression", EUROPEAN JOURNAL OF PHARMACOLOGY, vol. 815, 2017, pages 501 - 511, XP085245791, ISSN: 0014-2999, DOI: 10.1016/J.EJPHAR.2017.10.019 * |
| JUN LI ET AL: "Ginsenoside Rh1 potentiates dexamethasone's anti-inflammatory effects for chronic inflammatory disease by reversing dexamethasone-induced resistance", ARTHRITIS RESEARCH AND THERAPY, BIOMED CENTRAL, LONDON, GB, vol. 16, no. 3, 1 May 2014 (2014-05-01), pages R106, XP021188883, ISSN: 1478-6354, DOI: 10.1186/AR4556 * |
| LELIEVELD, L.T., ZEBRAFISH AS RESEARCH MODEL TO STUDY GAUCHER DISEASE: INSIGHTS INTO MOLECULAR MECHANISMS, 2020 |
| LELIEVELD, L.T.MIRZAIAN, M.KUO, C.-L.ARTOLA, M.FERRAZ, M.J.PETER, R.E.A.AKIYAMA, H.GREIMEL, P.BERG, R.J.B.H.N.OVERKLEEFT, H.S. ET : "Role of beta-glucosidase 2 in aberrant glycosphingolipid metabolism: model of glucocerebrosidase deficiency in zebrafish.", J. LIPID RES., vol. 60, 2019, pages 1851 - 1867 |
| LI WEI-ZU ET AL: "Protective Effect of Extract of Astragalus on Learning and Memory Impairments and Neurons Apoptosis Induced by Glucocorticoids in 12-Month Male Mice", THE ANATOMICAL RECORD, vol. 294, no. 6, 28 April 2011 (2011-04-28), Hoboken, NJ : Wiley-Liss, pages 1003 - 1014, XP093097225, ISSN: 1932-8486, DOI: 10.1002/ar.21386 * |
| LIFE SCIENCE METHODS BV |
| NEWTON, R.: "Molecular mechanisms of glucocorticoid action: what is important?", THORAX, vol. 55, 2000, pages 603 - 613 |
| PARK H.LEE, M-KPARK, Q-H.: "Pattern of molecular aggregation of ginsenosides in aquous solution.", J. KOREAN AGRICULTURAL CHEMICAL SOCIETY, vol. 29, no. 2, 1986, pages 198 - 206, XP053020568 |
| PATEL, G.C.MILLAR, J.C.CLARK, A.F.: "Glucocorticoid Receptor Transactivation Is Required for Glucocorticoid-Induced Ocular Hypertension and Glaucoma.", INVEST. OPHTHALMOL. VIS. SCI., vol. 60, 2019, pages 1967 - 1978 |
| RATMAN, D.VANDEN BERGHE, W.DEJAGER, L.LIBERT, C.TAVERNIER, J.BECK, I.M.DE BOSSCHER, K.: "How glucocorticoid receptors modulate the activity of other transcription factors: a scope beyond tethering.", MOL. CELL. ENDOCRINOL., vol. 380, 2013, pages 41 - 54, XP028728171, DOI: 10.1016/j.mce.2012.12.014 |
| REICHARDT, H.M.TUCKERMANN, J.P.GOTTLICHER, M.VUJIC, M.WEIH, F.ANGEL, P.HERRLICH, P.: "Schutz, G. Repression of inflammatory responses in the absence of DNA binding by the glucocorticoid receptor", EMBO J., vol. 20, 2001, pages 7168 - 7173 |
| RENSHAW, S.A.LOYNES, C.A.TRUSHELL, D.M.I.ELWORTHY, S.INGHAM, P.W.WHYTE, M.K.B.: "A transgenic zebrafish model of neutrophilic inflammation.", BLOOD, vol. 108, 2006, pages 3976 - 3978, XP002447426, DOI: 10.1182/blood-2006-05-024075 |
| SCHAAF, M.J.M.CIDLOWSKI, J.A.: "Molecular mechanisms of glucocorticoid action and resistance.", J. STEROID BIOCHEM. MOL. BIOL., vol. 83, 2002, pages 37 - 48 |
| STRAUB, R.H.CUTOLO, M.: "Glucocorticoids and chronic inflammation.", RHEUMATOLOGY (OXFORD), vol. 55, 2016, pages 116 - 1114 |
| TANIGAWA, K.NAGASE, H.OHMORI, K.TANAKA, K.MIYAKE, H.KINIWA, M.IKIZAWA, K.: "Species-specific differences in the glucocorticoid receptor transactivation function upon binding with betamethasone-esters.", INT. IMMUNOPHARMACOL., vol. 2, 2002, pages 941 - 950 |
| TROMBETTA, A.C.MERONI, M.CUTOLO, M.: "Steroids and Autoimmunity", FRONT. HORM. RES., vol. 48, 2017, pages 121 - 132 |
| VAN POMEREN, M.PEIJNENBURG, W.J.G.M.BRUN, N.R.VIJVER, M.G.: "Novel Experimental and Modelling Strategy for Nanoparticle Toxicity Testing Enabling the Use of Small Quantities.", INT. J. ENVIRON. RES. PUBLIC HEALTH, vol. 14, 2017 |
| WEPLER, M.PREUSS, J.M.MERZ, T.MCCOOK, O.RADERMACHER, P.TUCKERMANN, J.P.VETTORAZZI, S.: "Impact of downstream effects of glucocorticoid receptor dysfunction on organ function in critical illness-associated systemic inflammation.", INTENSIVE CARE MED. EXP., vol. 8, 2020, pages 37 |
| WILKINSON, L.VERHOOG, N.J.D.LOUW, A.: "Disease- and treatment-associated acquired glucocorticoid resistance.", ENDOCR. CONNECT., vol. 7, 2018 |
| XIONG J ET AL: "Self-micelle formation and the incorporation of lipid in the formulation affect the intestinal absorption of Panax notoginseng", INTERNATIONAL JOURNAL OF PHARMACEUTICS, ELSEVIER, NL, vol. 360, no. 1-2, 6 August 2008 (2008-08-06), pages 191 - 196, XP022853142, ISSN: 0378-5173, [retrieved on 20080416], DOI: 10.1016/J.IJPHARM.2008.04.016 * |
| ZFIN THE ZEBRAFISH INFORMATION NETWORK, 23 December 2020 (2020-12-23), Retrieved from the Internet <URL:https://zfin.org> |
| ZHANG YAODONG ET AL: "Ginsenoside Rg1 protects against neuronal degeneration induced by chronic dexamethasone treatment by inhibiting NLRP-1 inflammasomes in mice", INTERNATIONAL JOURNAL OF MOLECULAR MEDICINE, vol. 40, no. 4, 9 August 2017 (2017-08-09), GR, pages 1134 - 1142, XP093097233, ISSN: 1107-3756, DOI: 10.3892/ijmm.2017.3092 * |
| ZHONG XI YI JIE HE XUE BAO = JOURNAL OF CHINESE INTEGRATIVE MEDICINE AUG 2010, vol. 8, no. 8, August 2010 (2010-08-01), pages 762 - 766, ISSN: 1672-1977 * |
| ZIV, L.MUTO, A.SCHOONHEIM, P.J.MEIJSING, S.H.STRASSER, D.INGRAHAM, H.A.SCHAAF, M.J.M.YAMAMOTO, K.R.BAIER, H.: "An affective disorder in zebrafish with mutation of the glucocorticoid receptor.", MOL. PSYCHIATRY, vol. 18, 2013, pages 681 - 691, XP037790032, DOI: 10.1038/mp.2012.64 |
Also Published As
| Publication number | Publication date |
|---|---|
| WO2024232756A1 (en) | 2024-11-14 |
Similar Documents
| Publication | Publication Date | Title |
|---|---|---|
| Wang et al. | The flavonoid-rich fraction from rhizomes of Smilax glabra Roxb. ameliorates renal oxidative stress and inflammation in uric acid nephropathy rats through promoting uric acid excretion | |
| Hou et al. | Salvianolic acid A protects against diabetic nephropathy through ameliorating glomerular endothelial dysfunction via inhibiting AGE-RAGE signaling | |
| Van Kampen et al. | Neuroprotective actions of the ginseng extract G115 in two rodent models of Parkinson's disease | |
| Li et al. | Metformin and resveratrol inhibit Drp1-mediated mitochondrial fission and prevent ER stress-associated NLRP3 inflammasome activation in the adipose tissue of diabetic mice | |
| US10328046B2 (en) | Topical compositions comprising polyunsaturated fatty acids | |
| Shin et al. | Chrysin, a natural flavone, improves murine inflammatory bowel diseases | |
| DK2441454T3 (en) | Use of isothiocyanatforbindelser for the treatment of prostatitis | |
| Marinelli et al. | Garcinoic acid prevents β-amyloid (Aβ) deposition in the mouse brain | |
| Wang et al. | Comparative study on saponin fractions from Panax notoginseng inhibiting inflammation-induced endothelial adhesion molecule expression and monocyte adhesion | |
| Bao et al. | Eurycomanol alleviates hyperuricemia by promoting uric acid excretion and reducing purine synthesis | |
| Ma et al. | Caloric restriction can improve learning and memory in C57/BL mice probably via regulation of the AMPK signaling pathway | |
| Zhang et al. | Antihyperuricemia and antigouty arthritis effects of Persicaria capitata herba in mice | |
| TW200843735A (en) | Chromones as therapeutic agents | |
| Elmas et al. | The efficacy of Aesculus hippocastanum seeds on diabetic nephropathy in a streptozotocin-induced diabetic rat model | |
| Xiong et al. | Tibetan medicine Kuan-Jin-Teng exerts anti-arthritic effects on collagen-induced arthritis rats via inhibition the production of pro-inflammatory cytokines and down-regulation of MAPK signaling pathway | |
| Zeng et al. | A TCM formula VYAC ameliorates DNCB-induced atopic dermatitis via blocking mast cell degranulation and suppressing NF-κB pathway | |
| Sun et al. | Icariin reduces LPS-induced acute lung injury in mice undergoing bilateral adrenalectomy by regulating GRα | |
| Zhong et al. | Oxyberberrubine, a novel liver microsomes-mediated secondary metabolite of berberine, alleviates hyperuricemic nephropathy in mice | |
| Armanini et al. | Pseudohyperaldosteronism: pathogenetic mechanisms | |
| Ling et al. | Ginsenosides may reverse the dexamethasone-induced down-regulation of glucocorticoid receptor | |
| Jiang et al. | Saponins from Nigella glandulifera seeds attenuate collagen-induced rheumatoid arthritis in rats via the OPG/RANKL/NF-κB and Ang/Tie-2 pathways | |
| Alshagga et al. | Khat (Catha edulis) upregulates lipolytic genes in white adipose tissue of male obese mice (C57BL/6J) | |
| Chen et al. | Strobilanthes sarcorrhiza root phenolic extract prevent diabetic nephropathy in mice by regulating NF-κB/IL-1β signaling and glycerophospholipid metabolism | |
| Lv et al. | Study on the anti-hyperuricemic bioactivity and chemical components of Sterculiae lychnophorae Semen | |
| Zhang et al. | Farnesoid X receptor activation is required for the anti-inflammatory and anti-oxidative stress effects of Alisol B 23-acetate in carbon tetrachloride-induced liver fibrosis in mice |