TWI810644B - Therapeutic herbal composition - Google Patents
Therapeutic herbal composition Download PDFInfo
- Publication number
- TWI810644B TWI810644B TW110132930A TW110132930A TWI810644B TW I810644 B TWI810644 B TW I810644B TW 110132930 A TW110132930 A TW 110132930A TW 110132930 A TW110132930 A TW 110132930A TW I810644 B TWI810644 B TW I810644B
- Authority
- TW
- Taiwan
- Prior art keywords
- lutein
- herbal composition
- therapeutic
- therapeutic herbal
- range
- Prior art date
Links
- 239000000203 mixture Substances 0.000 title claims abstract description 303
- 230000001225 therapeutic effect Effects 0.000 title claims abstract description 247
- KBPHJBAIARWVSC-XQIHNALSSA-N trans-lutein Natural products CC(=C/C=C/C=C(C)/C=C/C=C(C)/C=C/C1=C(C)CC(O)CC1(C)C)C=CC=C(/C)C=CC2C(=CC(O)CC2(C)C)C KBPHJBAIARWVSC-XQIHNALSSA-N 0.000 claims abstract description 206
- KBPHJBAIARWVSC-RGZFRNHPSA-N lutein Chemical compound C([C@H](O)CC=1C)C(C)(C)C=1\C=C\C(\C)=C\C=C\C(\C)=C\C=C\C=C(/C)\C=C\C=C(/C)\C=C\[C@H]1C(C)=C[C@H](O)CC1(C)C KBPHJBAIARWVSC-RGZFRNHPSA-N 0.000 claims abstract description 162
- 229960005375 lutein Drugs 0.000 claims abstract description 159
- FJHBOVDFOQMZRV-XQIHNALSSA-N xanthophyll Natural products CC(=C/C=C/C=C(C)/C=C/C=C(C)/C=C/C1=C(C)CC(O)CC1(C)C)C=CC=C(/C)C=CC2C=C(C)C(O)CC2(C)C FJHBOVDFOQMZRV-XQIHNALSSA-N 0.000 claims abstract description 159
- 239000001656 lutein Substances 0.000 claims abstract description 158
- 235000012680 lutein Nutrition 0.000 claims abstract description 158
- ORAKUVXRZWMARG-WZLJTJAWSA-N lutein Natural products CC(=C/C=C/C=C(C)/C=C/C=C(C)/C=C/C1=C(C)CCCC1(C)C)C=CC=C(/C)C=CC2C(=CC(O)CC2(C)C)C ORAKUVXRZWMARG-WZLJTJAWSA-N 0.000 claims abstract description 158
- 238000000034 method Methods 0.000 claims abstract description 40
- 150000002632 lipids Chemical class 0.000 claims abstract description 33
- 239000003814 drug Substances 0.000 claims abstract description 32
- 238000004519 manufacturing process Methods 0.000 claims abstract description 32
- 201000010099 disease Diseases 0.000 claims abstract description 26
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 claims abstract description 26
- 235000013305 food Nutrition 0.000 claims abstract description 26
- 230000036542 oxidative stress Effects 0.000 claims abstract description 23
- 239000000546 pharmaceutical excipient Substances 0.000 claims abstract description 23
- 239000000047 product Substances 0.000 claims abstract description 22
- 230000002757 inflammatory effect Effects 0.000 claims abstract description 21
- 210000004369 blood Anatomy 0.000 claims abstract description 19
- 239000008280 blood Substances 0.000 claims abstract description 19
- 208000002780 macular degeneration Diseases 0.000 claims abstract description 15
- 206010064930 age-related macular degeneration Diseases 0.000 claims abstract description 14
- 235000013361 beverage Nutrition 0.000 claims abstract description 14
- 235000015872 dietary supplement Nutrition 0.000 claims abstract description 12
- 206010012689 Diabetic retinopathy Diseases 0.000 claims abstract description 11
- 235000013334 alcoholic beverage Nutrition 0.000 claims abstract description 8
- 230000001105 regulatory effect Effects 0.000 claims abstract description 7
- LFQSCWFLJHTTHZ-UHFFFAOYSA-N Ethanol Chemical compound CCO LFQSCWFLJHTTHZ-UHFFFAOYSA-N 0.000 claims description 249
- 210000004027 cell Anatomy 0.000 claims description 109
- 239000012141 concentrate Substances 0.000 claims description 55
- 229930153442 Curcuminoid Natural products 0.000 claims description 53
- VFLDPWHFBUODDF-FCXRPNKRSA-N curcumin Chemical compound C1=C(O)C(OC)=CC(\C=C\C(=O)CC(=O)\C=C\C=2C=C(OC)C(O)=CC=2)=C1 VFLDPWHFBUODDF-FCXRPNKRSA-N 0.000 claims description 51
- JKQXZKUSFCKOGQ-JLGXGRJMSA-N (3R,3'R)-beta,beta-carotene-3,3'-diol Chemical compound C([C@H](O)CC=1C)C(C)(C)C=1/C=C/C(/C)=C/C=C/C(/C)=C/C=C/C=C(C)C=CC=C(C)C=CC1=C(C)C[C@@H](O)CC1(C)C JKQXZKUSFCKOGQ-JLGXGRJMSA-N 0.000 claims description 47
- JKQXZKUSFCKOGQ-LQFQNGICSA-N Z-zeaxanthin Natural products C([C@H](O)CC=1C)C(C)(C)C=1C=CC(C)=CC=CC(C)=CC=CC=C(C)C=CC=C(C)C=CC1=C(C)C[C@@H](O)CC1(C)C JKQXZKUSFCKOGQ-LQFQNGICSA-N 0.000 claims description 47
- QOPRSMDTRDMBNK-RNUUUQFGSA-N Zeaxanthin Natural products CC(=C/C=C/C=C(C)/C=C/C=C(C)/C=C/C1=C(C)CCC(O)C1(C)C)C=CC=C(/C)C=CC2=C(C)CC(O)CC2(C)C QOPRSMDTRDMBNK-RNUUUQFGSA-N 0.000 claims description 47
- JKQXZKUSFCKOGQ-LOFNIBRQSA-N all-trans-Zeaxanthin Natural products CC(=C/C=C/C=C(C)/C=C/C=C(C)/C=C/C1=C(C)CC(O)CC1(C)C)C=CC=C(/C)C=CC2=C(C)CC(O)CC2(C)C JKQXZKUSFCKOGQ-LOFNIBRQSA-N 0.000 claims description 47
- 235000010930 zeaxanthin Nutrition 0.000 claims description 47
- 229940043269 zeaxanthin Drugs 0.000 claims description 47
- 239000001775 zeaxanthin Substances 0.000 claims description 47
- 239000002245 particle Substances 0.000 claims description 37
- 239000000843 powder Substances 0.000 claims description 36
- 239000000839 emulsion Substances 0.000 claims description 35
- 235000020777 polyunsaturated fatty acids Nutrition 0.000 claims description 29
- 239000003642 reactive oxygen metabolite Substances 0.000 claims description 29
- 239000013078 crystal Substances 0.000 claims description 27
- 239000000725 suspension Substances 0.000 claims description 27
- 235000012754 curcumin Nutrition 0.000 claims description 26
- 239000004148 curcumin Substances 0.000 claims description 26
- 229940109262 curcumin Drugs 0.000 claims description 26
- VFLDPWHFBUODDF-UHFFFAOYSA-N diferuloylmethane Natural products C1=C(O)C(OC)=CC(C=CC(=O)CC(=O)C=CC=2C=C(OC)C(O)=CC=2)=C1 VFLDPWHFBUODDF-UHFFFAOYSA-N 0.000 claims description 26
- RWSXRVCMGQZWBV-WDSKDSINSA-N glutathione Chemical compound OC(=O)[C@@H](N)CCC(=O)N[C@@H](CS)C(=O)NCC(O)=O RWSXRVCMGQZWBV-WDSKDSINSA-N 0.000 claims description 26
- 229940079593 drug Drugs 0.000 claims description 25
- 210000002966 serum Anatomy 0.000 claims description 22
- 230000001965 increasing effect Effects 0.000 claims description 21
- 108060008682 Tumor Necrosis Factor Proteins 0.000 claims description 20
- 102000000852 Tumor Necrosis Factor-alpha Human genes 0.000 claims description 20
- 235000003373 curcuma longa Nutrition 0.000 claims description 20
- 244000163122 Curcuma domestica Species 0.000 claims description 19
- 102000004190 Enzymes Human genes 0.000 claims description 19
- 108090000790 Enzymes Proteins 0.000 claims description 19
- WSMYVTOQOOLQHP-UHFFFAOYSA-N Malondialdehyde Chemical compound O=CCC=O WSMYVTOQOOLQHP-UHFFFAOYSA-N 0.000 claims description 18
- HVYWMOMLDIMFJA-DPAQBDIFSA-N cholesterol Chemical compound C1C=C2C[C@@H](O)CC[C@]2(C)[C@@H]2[C@@H]1[C@@H]1CC[C@H]([C@H](C)CCCC(C)C)[C@@]1(C)CC2 HVYWMOMLDIMFJA-DPAQBDIFSA-N 0.000 claims description 18
- 229940118019 malondialdehyde Drugs 0.000 claims description 18
- 102000019197 Superoxide Dismutase Human genes 0.000 claims description 17
- 108010012715 Superoxide dismutase Proteins 0.000 claims description 17
- 239000003963 antioxidant agent Substances 0.000 claims description 16
- 239000003995 emulsifying agent Substances 0.000 claims description 16
- 208000015181 infectious disease Diseases 0.000 claims description 16
- MZOFCQQQCNRIBI-VMXHOPILSA-N (3s)-4-[[(2s)-1-[[(2s)-1-[[(1s)-1-carboxy-2-hydroxyethyl]amino]-4-methyl-1-oxopentan-2-yl]amino]-5-(diaminomethylideneamino)-1-oxopentan-2-yl]amino]-3-[[2-[[(2s)-2,6-diaminohexanoyl]amino]acetyl]amino]-4-oxobutanoic acid Chemical compound OC[C@@H](C(O)=O)NC(=O)[C@H](CC(C)C)NC(=O)[C@H](CCCN=C(N)N)NC(=O)[C@H](CC(O)=O)NC(=O)CNC(=O)[C@@H](N)CCCCN MZOFCQQQCNRIBI-VMXHOPILSA-N 0.000 claims description 15
- 241000196324 Embryophyta Species 0.000 claims description 15
- 239000005913 Maltodextrin Substances 0.000 claims description 15
- 229920002774 Maltodextrin Polymers 0.000 claims description 15
- 229940035034 maltodextrin Drugs 0.000 claims description 15
- XLYOFNOQVPJJNP-UHFFFAOYSA-N water Chemical compound O XLYOFNOQVPJJNP-UHFFFAOYSA-N 0.000 claims description 15
- 235000003392 Curcuma domestica Nutrition 0.000 claims description 14
- 108090001005 Interleukin-6 Proteins 0.000 claims description 14
- 230000006378 damage Effects 0.000 claims description 14
- 235000013976 turmeric Nutrition 0.000 claims description 14
- 108010002352 Interleukin-1 Proteins 0.000 claims description 13
- 235000006708 antioxidants Nutrition 0.000 claims description 13
- 230000006907 apoptotic process Effects 0.000 claims description 13
- 235000008242 dietary patterns Nutrition 0.000 claims description 13
- 229960003180 glutathione Drugs 0.000 claims description 13
- 239000007788 liquid Substances 0.000 claims description 13
- 102000004127 Cytokines Human genes 0.000 claims description 12
- 108090000695 Cytokines Proteins 0.000 claims description 12
- 241000700605 Viruses Species 0.000 claims description 11
- 230000003078 antioxidant effect Effects 0.000 claims description 11
- 238000013467 fragmentation Methods 0.000 claims description 11
- 238000006062 fragmentation reaction Methods 0.000 claims description 11
- 239000004615 ingredient Substances 0.000 claims description 11
- UFTFJSFQGQCHQW-UHFFFAOYSA-N triformin Chemical compound O=COCC(OC=O)COC=O UFTFJSFQGQCHQW-UHFFFAOYSA-N 0.000 claims description 11
- 230000002401 inhibitory effect Effects 0.000 claims description 10
- 208000008338 non-alcoholic fatty liver disease Diseases 0.000 claims description 10
- 230000001717 pathogenic effect Effects 0.000 claims description 10
- 230000000770 proinflammatory effect Effects 0.000 claims description 10
- 238000012545 processing Methods 0.000 claims description 9
- 239000000084 colloidal system Substances 0.000 claims description 8
- 239000008213 purified water Substances 0.000 claims description 8
- 230000003405 preventing effect Effects 0.000 claims description 7
- 108010063907 Glutathione Reductase Proteins 0.000 claims description 6
- 102100036442 Glutathione reductase, mitochondrial Human genes 0.000 claims description 6
- 230000009286 beneficial effect Effects 0.000 claims description 6
- 230000001276 controlling effect Effects 0.000 claims description 6
- 230000000378 dietary effect Effects 0.000 claims description 6
- 230000037406 food intake Effects 0.000 claims description 6
- 235000013336 milk Nutrition 0.000 claims description 6
- 239000008267 milk Substances 0.000 claims description 6
- 210000004080 milk Anatomy 0.000 claims description 6
- 244000052769 pathogen Species 0.000 claims description 6
- 241000894006 Bacteria Species 0.000 claims description 5
- 240000000785 Tagetes erecta Species 0.000 claims description 5
- 230000002195 synergetic effect Effects 0.000 claims description 5
- 241000711573 Coronaviridae Species 0.000 claims description 4
- 230000001476 alcoholic effect Effects 0.000 claims description 4
- 238000002483 medication Methods 0.000 claims description 4
- RVBUGGBMJDPOST-UHFFFAOYSA-N 2-thiobarbituric acid Chemical compound O=C1CC(=O)NC(=S)N1 RVBUGGBMJDPOST-UHFFFAOYSA-N 0.000 claims description 3
- 239000003795 chemical substances by application Substances 0.000 claims description 3
- 238000001035 drying Methods 0.000 claims description 3
- 235000015203 fruit juice Nutrition 0.000 claims description 3
- 235000011868 grain product Nutrition 0.000 claims description 3
- 208000006454 hepatitis Diseases 0.000 claims description 3
- 238000001802 infusion Methods 0.000 claims description 3
- 208000018191 liver inflammation Diseases 0.000 claims description 3
- 235000021067 refined food Nutrition 0.000 claims description 3
- 239000013589 supplement Substances 0.000 claims description 3
- 235000005881 Calendula officinalis Nutrition 0.000 claims description 2
- 235000012311 Tagetes erecta Nutrition 0.000 claims description 2
- 208000026594 alcoholic fatty liver disease Diseases 0.000 claims description 2
- 230000027829 mitochondrial depolarization Effects 0.000 claims description 2
- 235000016709 nutrition Nutrition 0.000 claims description 2
- 208000025721 COVID-19 Diseases 0.000 claims 1
- 244000269722 Thea sinensis Species 0.000 claims 1
- 230000002265 prevention Effects 0.000 claims 1
- 230000004054 inflammatory process Effects 0.000 abstract description 31
- 206010061218 Inflammation Diseases 0.000 abstract description 29
- 230000003110 anti-inflammatory effect Effects 0.000 abstract description 15
- 235000021466 carotenoid Nutrition 0.000 abstract description 11
- 150000001747 carotenoids Chemical class 0.000 abstract description 11
- 239000002417 nutraceutical Substances 0.000 abstract description 5
- 235000021436 nutraceutical agent Nutrition 0.000 abstract description 5
- 230000008569 process Effects 0.000 abstract description 5
- 235000017807 phytochemicals Nutrition 0.000 abstract description 3
- 229930000223 plant secondary metabolite Natural products 0.000 abstract description 3
- 230000000694 effects Effects 0.000 description 73
- SEBFKMXJBCUCAI-UHFFFAOYSA-N NSC 227190 Natural products C1=C(O)C(OC)=CC(C2C(OC3=CC=C(C=C3O2)C2C(C(=O)C3=C(O)C=C(O)C=C3O2)O)CO)=C1 SEBFKMXJBCUCAI-UHFFFAOYSA-N 0.000 description 54
- SEBFKMXJBCUCAI-HKTJVKLFSA-N silibinin Chemical compound C1=C(O)C(OC)=CC([C@@H]2[C@H](OC3=CC=C(C=C3O2)[C@@H]2[C@H](C(=O)C3=C(O)C=C(O)C=C3O2)O)CO)=C1 SEBFKMXJBCUCAI-HKTJVKLFSA-N 0.000 description 54
- 235000017700 silymarin Nutrition 0.000 description 54
- 229960004245 silymarin Drugs 0.000 description 54
- RZVAJINKPMORJF-UHFFFAOYSA-N Acetaminophen Chemical compound CC(=O)NC1=CC=C(O)C=C1 RZVAJINKPMORJF-UHFFFAOYSA-N 0.000 description 52
- 210000004185 liver Anatomy 0.000 description 41
- 235000008504 concentrate Nutrition 0.000 description 40
- 241000700159 Rattus Species 0.000 description 30
- 231100000433 cytotoxic Toxicity 0.000 description 21
- 230000001472 cytotoxic effect Effects 0.000 description 21
- JVJFIQYAHPMBBX-UHFFFAOYSA-N 4-hydroxynonenal Chemical compound CCCCCC(O)C=CC=O JVJFIQYAHPMBBX-UHFFFAOYSA-N 0.000 description 18
- 241001465754 Metazoa Species 0.000 description 18
- MWUXSHHQAYIFBG-UHFFFAOYSA-N Nitric oxide Chemical compound O=[N] MWUXSHHQAYIFBG-UHFFFAOYSA-N 0.000 description 16
- 238000004458 analytical method Methods 0.000 description 15
- 229930027945 nicotinamide-adenine dinucleotide Natural products 0.000 description 15
- BOPGDPNILDQYTO-NNYOXOHSSA-N nicotinamide-adenine dinucleotide Chemical compound C1=CCC(C(=O)N)=CN1[C@H]1[C@H](O)[C@H](O)[C@@H](COP(O)(=O)OP(O)(=O)OC[C@@H]2[C@H]([C@@H](O)[C@@H](O2)N2C3=NC=NC(N)=C3N=C2)O)O1 BOPGDPNILDQYTO-NNYOXOHSSA-N 0.000 description 15
- 239000000524 Thiobarbituric Acid Reactive Substance Substances 0.000 description 14
- 150000001875 compounds Chemical class 0.000 description 14
- 102000007698 Alcohol dehydrogenase Human genes 0.000 description 13
- 108010021809 Alcohol dehydrogenase Proteins 0.000 description 13
- 102000004889 Interleukin-6 Human genes 0.000 description 13
- 229940100601 interleukin-6 Drugs 0.000 description 13
- 210000001519 tissue Anatomy 0.000 description 13
- 102100036475 Alanine aminotransferase 1 Human genes 0.000 description 12
- 108010082126 Alanine transaminase Proteins 0.000 description 12
- 108010003415 Aspartate Aminotransferases Proteins 0.000 description 12
- 102000004625 Aspartate Aminotransferases Human genes 0.000 description 12
- 108010010234 HDL Lipoproteins Proteins 0.000 description 12
- 102000015779 HDL Lipoproteins Human genes 0.000 description 12
- 102000000589 Interleukin-1 Human genes 0.000 description 12
- 230000037396 body weight Effects 0.000 description 12
- 230000002829 reductive effect Effects 0.000 description 11
- 238000009837 dry grinding Methods 0.000 description 10
- 239000007921 spray Substances 0.000 description 10
- 108010007622 LDL Lipoproteins Proteins 0.000 description 9
- 102000007330 LDL Lipoproteins Human genes 0.000 description 9
- 206010067125 Liver injury Diseases 0.000 description 9
- 230000002440 hepatic effect Effects 0.000 description 9
- 231100000304 hepatotoxicity Toxicity 0.000 description 9
- 230000003834 intracellular effect Effects 0.000 description 9
- 230000003859 lipid peroxidation Effects 0.000 description 9
- 230000037361 pathway Effects 0.000 description 9
- 235000012000 cholesterol Nutrition 0.000 description 8
- 210000003494 hepatocyte Anatomy 0.000 description 8
- 230000001404 mediated effect Effects 0.000 description 8
- 210000001700 mitochondrial membrane Anatomy 0.000 description 8
- 239000000126 substance Substances 0.000 description 8
- 108010023302 HDL Cholesterol Proteins 0.000 description 7
- 206010019851 Hepatotoxicity Diseases 0.000 description 7
- 108010028554 LDL Cholesterol Proteins 0.000 description 7
- 230000002159 abnormal effect Effects 0.000 description 7
- 230000015572 biosynthetic process Effects 0.000 description 7
- 206010012601 diabetes mellitus Diseases 0.000 description 7
- 235000005911 diet Nutrition 0.000 description 7
- 231100000753 hepatic injury Toxicity 0.000 description 7
- 230000007686 hepatotoxicity Effects 0.000 description 7
- 238000000338 in vitro Methods 0.000 description 7
- 201000007270 liver cancer Diseases 0.000 description 7
- 208000014018 liver neoplasm Diseases 0.000 description 7
- 210000005228 liver tissue Anatomy 0.000 description 7
- 229960005489 paracetamol Drugs 0.000 description 7
- 238000005502 peroxidation Methods 0.000 description 7
- 230000009467 reduction Effects 0.000 description 7
- 229920000881 Modified starch Polymers 0.000 description 6
- 239000004368 Modified starch Substances 0.000 description 6
- 229940121363 anti-inflammatory agent Drugs 0.000 description 6
- 239000002260 anti-inflammatory agent Substances 0.000 description 6
- 235000014113 dietary fatty acids Nutrition 0.000 description 6
- 229930195729 fatty acid Natural products 0.000 description 6
- 239000000194 fatty acid Substances 0.000 description 6
- 208000010706 fatty liver disease Diseases 0.000 description 6
- 230000036541 health Effects 0.000 description 6
- 230000004483 macular pigment optical density Effects 0.000 description 6
- 239000003550 marker Substances 0.000 description 6
- 239000000463 material Substances 0.000 description 6
- 230000004060 metabolic process Effects 0.000 description 6
- 238000002156 mixing Methods 0.000 description 6
- 235000019426 modified starch Nutrition 0.000 description 6
- 229950006238 nadide Drugs 0.000 description 6
- 230000008506 pathogenesis Effects 0.000 description 6
- 238000011160 research Methods 0.000 description 6
- 210000001525 retina Anatomy 0.000 description 6
- 231100000240 steatosis hepatitis Toxicity 0.000 description 6
- 102000004357 Transferases Human genes 0.000 description 5
- 108090000992 Transferases Proteins 0.000 description 5
- 238000010521 absorption reaction Methods 0.000 description 5
- 230000003247 decreasing effect Effects 0.000 description 5
- 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 5
- ZMMJGEGLRURXTF-UHFFFAOYSA-N ethidium bromide Chemical compound [Br-].C12=CC(N)=CC=C2C2=CC=C(N)C=C2[N+](CC)=C1C1=CC=CC=C1 ZMMJGEGLRURXTF-UHFFFAOYSA-N 0.000 description 5
- 150000004665 fatty acids Chemical class 0.000 description 5
- 239000012467 final product Substances 0.000 description 5
- 238000001727 in vivo Methods 0.000 description 5
- 230000028709 inflammatory response Effects 0.000 description 5
- 235000012054 meals Nutrition 0.000 description 5
- 238000005259 measurement Methods 0.000 description 5
- 210000003470 mitochondria Anatomy 0.000 description 5
- 230000004660 morphological change Effects 0.000 description 5
- 230000004792 oxidative damage Effects 0.000 description 5
- 239000002953 phosphate buffered saline Substances 0.000 description 5
- 239000000523 sample Substances 0.000 description 5
- 150000003626 triacylglycerols Chemical class 0.000 description 5
- 206010019708 Hepatic steatosis Diseases 0.000 description 4
- 102000004895 Lipoproteins Human genes 0.000 description 4
- 108090001030 Lipoproteins Proteins 0.000 description 4
- 238000009833 condensation Methods 0.000 description 4
- 230000005494 condensation Effects 0.000 description 4
- -1 curcumin phospholipid Chemical class 0.000 description 4
- 230000003013 cytotoxicity Effects 0.000 description 4
- 231100000135 cytotoxicity Toxicity 0.000 description 4
- 230000037213 diet Effects 0.000 description 4
- 238000002474 experimental method Methods 0.000 description 4
- 239000007850 fluorescent dye Substances 0.000 description 4
- 210000001035 gastrointestinal tract Anatomy 0.000 description 4
- 239000012678 infectious agent Substances 0.000 description 4
- 230000000670 limiting effect Effects 0.000 description 4
- 210000005229 liver cell Anatomy 0.000 description 4
- 208000019423 liver disease Diseases 0.000 description 4
- 210000000056 organ Anatomy 0.000 description 4
- 210000003462 vein Anatomy 0.000 description 4
- 208000022309 Alcoholic Liver disease Diseases 0.000 description 3
- 240000001432 Calendula officinalis Species 0.000 description 3
- 208000024172 Cardiovascular disease Diseases 0.000 description 3
- 208000017667 Chronic Disease Diseases 0.000 description 3
- IAZDPXIOMUYVGZ-UHFFFAOYSA-N Dimethylsulphoxide Chemical compound CS(C)=O IAZDPXIOMUYVGZ-UHFFFAOYSA-N 0.000 description 3
- 208000004930 Fatty Liver Diseases 0.000 description 3
- 241000282412 Homo Species 0.000 description 3
- 102000015696 Interleukins Human genes 0.000 description 3
- 108010063738 Interleukins Proteins 0.000 description 3
- 206010028851 Necrosis Diseases 0.000 description 3
- 206010028980 Neoplasm Diseases 0.000 description 3
- 208000002193 Pain Diseases 0.000 description 3
- FAPWRFPIFSIZLT-UHFFFAOYSA-M Sodium chloride Chemical compound [Na+].[Cl-] FAPWRFPIFSIZLT-UHFFFAOYSA-M 0.000 description 3
- 208000027418 Wounds and injury Diseases 0.000 description 3
- 240000008042 Zea mays Species 0.000 description 3
- 235000005824 Zea mays ssp. parviglumis Nutrition 0.000 description 3
- 235000002017 Zea mays subsp mays Nutrition 0.000 description 3
- 230000004913 activation Effects 0.000 description 3
- 230000002411 adverse Effects 0.000 description 3
- 238000000540 analysis of variance Methods 0.000 description 3
- 208000029618 autoimmune pulmonary alveolar proteinosis Diseases 0.000 description 3
- 238000012742 biochemical analysis Methods 0.000 description 3
- 201000011510 cancer Diseases 0.000 description 3
- 210000000170 cell membrane Anatomy 0.000 description 3
- 230000001684 chronic effect Effects 0.000 description 3
- 208000037976 chronic inflammation Diseases 0.000 description 3
- 238000011278 co-treatment Methods 0.000 description 3
- 235000005822 corn Nutrition 0.000 description 3
- 230000007850 degeneration Effects 0.000 description 3
- 238000002565 electrocardiography Methods 0.000 description 3
- 238000009472 formulation Methods 0.000 description 3
- 235000012055 fruits and vegetables Nutrition 0.000 description 3
- 208000027866 inflammatory disease Diseases 0.000 description 3
- 210000004072 lung Anatomy 0.000 description 3
- 230000028161 membrane depolarization Effects 0.000 description 3
- 230000017074 necrotic cell death Effects 0.000 description 3
- 238000007254 oxidation reaction Methods 0.000 description 3
- 239000000049 pigment Substances 0.000 description 3
- 235000018102 proteins Nutrition 0.000 description 3
- 102000004169 proteins and genes Human genes 0.000 description 3
- 108090000623 proteins and genes Proteins 0.000 description 3
- QZAYGJVTTNCVMB-UHFFFAOYSA-N serotonin Chemical compound C1=C(O)C=C2C(CCN)=CNC2=C1 QZAYGJVTTNCVMB-UHFFFAOYSA-N 0.000 description 3
- 238000010186 staining Methods 0.000 description 3
- 238000007619 statistical method Methods 0.000 description 3
- 230000007863 steatosis Effects 0.000 description 3
- 208000001072 type 2 diabetes mellitus Diseases 0.000 description 3
- NCYCYZXNIZJOKI-UHFFFAOYSA-N vitamin A aldehyde Natural products O=CC=C(C)C=CC=C(C)C=CC1=C(C)CCCC1(C)C NCYCYZXNIZJOKI-UHFFFAOYSA-N 0.000 description 3
- UEPVWRDHSPMIAZ-IZTHOABVSA-N (1e,4z,6e)-5-hydroxy-7-(4-hydroxy-3-methoxyphenyl)-1-(4-hydroxyphenyl)hepta-1,4,6-trien-3-one Chemical compound C1=C(O)C(OC)=CC(\C=C\C(\O)=C\C(=O)\C=C\C=2C=CC(O)=CC=2)=C1 UEPVWRDHSPMIAZ-IZTHOABVSA-N 0.000 description 2
- 208000007848 Alcoholism Diseases 0.000 description 2
- 201000001320 Atherosclerosis Diseases 0.000 description 2
- 201000004569 Blindness Diseases 0.000 description 2
- 240000007124 Brassica oleracea Species 0.000 description 2
- 235000003899 Brassica oleracea var acephala Nutrition 0.000 description 2
- 235000012905 Brassica oleracea var viridis Nutrition 0.000 description 2
- HEDRZPFGACZZDS-UHFFFAOYSA-N Chloroform Chemical compound ClC(Cl)Cl HEDRZPFGACZZDS-UHFFFAOYSA-N 0.000 description 2
- HJTVQHVGMGKONQ-LUZURFALSA-N Curcumin II Natural products C1=C(O)C(OC)=CC(\C=C\C(=O)CC(=O)\C=C\C=2C=CC(O)=CC=2)=C1 HJTVQHVGMGKONQ-LUZURFALSA-N 0.000 description 2
- 239000012591 Dulbecco’s Phosphate Buffered Saline Substances 0.000 description 2
- 208000032928 Dyslipidaemia Diseases 0.000 description 2
- WSFSSNUMVMOOMR-UHFFFAOYSA-N Formaldehyde Chemical compound O=C WSFSSNUMVMOOMR-UHFFFAOYSA-N 0.000 description 2
- DHMQDGOQFOQNFH-UHFFFAOYSA-N Glycine Chemical compound NCC(O)=O DHMQDGOQFOQNFH-UHFFFAOYSA-N 0.000 description 2
- WZUVPPKBWHMQCE-UHFFFAOYSA-N Haematoxylin Chemical compound C12=CC(O)=C(O)C=C2CC2(O)C1C1=CC=C(O)C(O)=C1OC2 WZUVPPKBWHMQCE-UHFFFAOYSA-N 0.000 description 2
- NTYJJOPFIAHURM-UHFFFAOYSA-N Histamine Chemical compound NCCC1=CN=CN1 NTYJJOPFIAHURM-UHFFFAOYSA-N 0.000 description 2
- 206010020772 Hypertension Diseases 0.000 description 2
- 102000010781 Interleukin-6 Receptors Human genes 0.000 description 2
- 108010038501 Interleukin-6 Receptors Proteins 0.000 description 2
- 102000015617 Janus Kinases Human genes 0.000 description 2
- 108010024121 Janus Kinases Proteins 0.000 description 2
- 208000017170 Lipid metabolism disease Diseases 0.000 description 2
- 102000043136 MAP kinase family Human genes 0.000 description 2
- 108091054455 MAP kinase family Proteins 0.000 description 2
- 206010025421 Macule Diseases 0.000 description 2
- 102000002274 Matrix Metalloproteinases Human genes 0.000 description 2
- 108010000684 Matrix Metalloproteinases Proteins 0.000 description 2
- BAWFJGJZGIEFAR-NNYOXOHSSA-N NAD zwitterion Chemical compound NC(=O)C1=CC=C[N+]([C@H]2[C@@H]([C@H](O)[C@@H](COP([O-])(=O)OP(O)(=O)OC[C@@H]3[C@H]([C@@H](O)[C@@H](O3)N3C4=NC=NC(N)=C4N=C3)O)O2)O)=C1 BAWFJGJZGIEFAR-NNYOXOHSSA-N 0.000 description 2
- 102000003945 NF-kappa B Human genes 0.000 description 2
- 108010057466 NF-kappa B Proteins 0.000 description 2
- 102000008299 Nitric Oxide Synthase Human genes 0.000 description 2
- 108010021487 Nitric Oxide Synthase Proteins 0.000 description 2
- 102000011779 Nitric Oxide Synthase Type II Human genes 0.000 description 2
- 108010076864 Nitric Oxide Synthase Type II Proteins 0.000 description 2
- 208000017442 Retinal disease Diseases 0.000 description 2
- 206010038923 Retinopathy Diseases 0.000 description 2
- 108060008683 Tumor Necrosis Factor Receptor Proteins 0.000 description 2
- 108010062497 VLDL Lipoproteins Proteins 0.000 description 2
- 241000234299 Zingiberaceae Species 0.000 description 2
- 238000009825 accumulation Methods 0.000 description 2
- DPKHZNPWBDQZCN-UHFFFAOYSA-N acridine orange free base Chemical compound C1=CC(N(C)C)=CC2=NC3=CC(N(C)C)=CC=C3C=C21 DPKHZNPWBDQZCN-UHFFFAOYSA-N 0.000 description 2
- 208000038016 acute inflammation Diseases 0.000 description 2
- 230000006022 acute inflammation Effects 0.000 description 2
- 201000007930 alcohol dependence Diseases 0.000 description 2
- 238000010171 animal model Methods 0.000 description 2
- 206010003246 arthritis Diseases 0.000 description 2
- 238000003556 assay Methods 0.000 description 2
- 208000006673 asthma Diseases 0.000 description 2
- 230000002238 attenuated effect Effects 0.000 description 2
- 230000008901 benefit Effects 0.000 description 2
- DZBUGLKDJFMEHC-UHFFFAOYSA-N benzoquinolinylidene Natural products C1=CC=CC2=CC3=CC=CC=C3N=C21 DZBUGLKDJFMEHC-UHFFFAOYSA-N 0.000 description 2
- JYTVKRNTTALBBZ-UHFFFAOYSA-N bis demethoxycurcumin Natural products C1=CC(O)=CC=C1C=CC(=O)CC(=O)C=CC1=CC=CC(O)=C1 JYTVKRNTTALBBZ-UHFFFAOYSA-N 0.000 description 2
- PREBVFJICNPEKM-YDWXAUTNSA-N bisdemethoxycurcumin Chemical compound C1=CC(O)=CC=C1\C=C\C(=O)CC(=O)\C=C\C1=CC=C(O)C=C1 PREBVFJICNPEKM-YDWXAUTNSA-N 0.000 description 2
- 239000002775 capsule Substances 0.000 description 2
- 235000014633 carbohydrates Nutrition 0.000 description 2
- 150000001720 carbohydrates Chemical class 0.000 description 2
- 230000003833 cell viability Effects 0.000 description 2
- 230000006020 chronic inflammation Effects 0.000 description 2
- 230000004087 circulation Effects 0.000 description 2
- 208000029078 coronary artery disease Diseases 0.000 description 2
- 230000003412 degenerative effect Effects 0.000 description 2
- NMRUIRRIQNAQEB-UHFFFAOYSA-N demethoxycurcumin Natural products OC(=CC(C=CC1=CC(=C(C=C1)O)OC)=O)C=CC1=CC=C(C=C1)O NMRUIRRIQNAQEB-UHFFFAOYSA-N 0.000 description 2
- 238000011161 development Methods 0.000 description 2
- 230000018109 developmental process Effects 0.000 description 2
- YXAKCQIIROBKOP-UHFFFAOYSA-N di-p-hydroxycinnamoylmethane Natural products C=1C=C(O)C=CC=1C=CC(=O)C=C(O)C=CC1=CC=C(O)C=C1 YXAKCQIIROBKOP-UHFFFAOYSA-N 0.000 description 2
- 239000002552 dosage form Substances 0.000 description 2
- 230000002222 downregulating effect Effects 0.000 description 2
- 230000002526 effect on cardiovascular system Effects 0.000 description 2
- 150000002066 eicosanoids Chemical class 0.000 description 2
- 230000008030 elimination Effects 0.000 description 2
- 238000003379 elimination reaction Methods 0.000 description 2
- 239000002158 endotoxin Substances 0.000 description 2
- 230000002255 enzymatic effect Effects 0.000 description 2
- 230000004149 ethanol metabolism Effects 0.000 description 2
- 229960005542 ethidium bromide Drugs 0.000 description 2
- 238000011156 evaluation Methods 0.000 description 2
- 208000030533 eye disease Diseases 0.000 description 2
- 235000020937 fasting conditions Nutrition 0.000 description 2
- 235000021149 fatty food Nutrition 0.000 description 2
- 229930003935 flavonoid Natural products 0.000 description 2
- 235000017173 flavonoids Nutrition 0.000 description 2
- 150000002215 flavonoids Chemical class 0.000 description 2
- 238000000799 fluorescence microscopy Methods 0.000 description 2
- 235000021384 green leafy vegetables Nutrition 0.000 description 2
- 230000012010 growth Effects 0.000 description 2
- 231100000234 hepatic damage Toxicity 0.000 description 2
- 230000002443 hepatoprotective effect Effects 0.000 description 2
- 238000007489 histopathology method Methods 0.000 description 2
- 230000013632 homeostatic process Effects 0.000 description 2
- 230000001976 improved effect Effects 0.000 description 2
- 238000011534 incubation Methods 0.000 description 2
- 230000006698 induction Effects 0.000 description 2
- 210000004969 inflammatory cell Anatomy 0.000 description 2
- 239000003112 inhibitor Substances 0.000 description 2
- 208000014674 injury Diseases 0.000 description 2
- 229940047122 interleukins Drugs 0.000 description 2
- 210000003734 kidney Anatomy 0.000 description 2
- 229920006008 lipopolysaccharide Polymers 0.000 description 2
- 230000008818 liver damage Effects 0.000 description 2
- 230000003908 liver function Effects 0.000 description 2
- 230000007056 liver toxicity Effects 0.000 description 2
- 239000007937 lozenge Substances 0.000 description 2
- 210000004379 membrane Anatomy 0.000 description 2
- 239000012528 membrane Substances 0.000 description 2
- 239000002207 metabolite Substances 0.000 description 2
- 230000004048 modification Effects 0.000 description 2
- 238000012986 modification Methods 0.000 description 2
- 239000002105 nanoparticle Substances 0.000 description 2
- 229930014626 natural product Natural products 0.000 description 2
- 229940101270 nicotinamide adenine dinucleotide (nad) Drugs 0.000 description 2
- 230000003647 oxidation Effects 0.000 description 2
- 230000004783 oxidative metabolism Effects 0.000 description 2
- UEPVWRDHSPMIAZ-UHFFFAOYSA-N p-hydroxycinnamoyl feruloylmethane Natural products C1=C(O)C(OC)=CC(C=CC(O)=CC(=O)C=CC=2C=CC(O)=CC=2)=C1 UEPVWRDHSPMIAZ-UHFFFAOYSA-N 0.000 description 2
- 230000036407 pain Effects 0.000 description 2
- 238000010951 particle size reduction Methods 0.000 description 2
- 239000006072 paste Substances 0.000 description 2
- 231100000915 pathological change Toxicity 0.000 description 2
- 230000036285 pathological change Effects 0.000 description 2
- 150000003904 phospholipids Chemical class 0.000 description 2
- 150000008442 polyphenolic compounds Chemical class 0.000 description 2
- 238000002360 preparation method Methods 0.000 description 2
- 230000001681 protective effect Effects 0.000 description 2
- 235000021251 pulses Nutrition 0.000 description 2
- 150000003254 radicals Chemical class 0.000 description 2
- 239000007845 reactive nitrogen species Substances 0.000 description 2
- 230000006950 reactive oxygen species formation Effects 0.000 description 2
- 108020003175 receptors Proteins 0.000 description 2
- 102000005962 receptors Human genes 0.000 description 2
- 230000002207 retinal effect Effects 0.000 description 2
- 238000012216 screening Methods 0.000 description 2
- 230000028327 secretion Effects 0.000 description 2
- 239000011780 sodium chloride Substances 0.000 description 2
- 239000000243 solution Substances 0.000 description 2
- 230000008961 swelling Effects 0.000 description 2
- 239000006188 syrup Substances 0.000 description 2
- 235000020357 syrup Nutrition 0.000 description 2
- 235000013616 tea Nutrition 0.000 description 2
- 231100000331 toxic Toxicity 0.000 description 2
- 230000002588 toxic effect Effects 0.000 description 2
- 102000003298 tumor necrosis factor receptor Human genes 0.000 description 2
- 235000019786 weight gain Nutrition 0.000 description 2
- LOGFVTREOLYCPF-KXNHARMFSA-N (2s,3r)-2-[[(2r)-1-[(2s)-2,6-diaminohexanoyl]pyrrolidine-2-carbonyl]amino]-3-hydroxybutanoic acid Chemical compound C[C@@H](O)[C@@H](C(O)=O)NC(=O)[C@H]1CCCN1C(=O)[C@@H](N)CCCCN LOGFVTREOLYCPF-KXNHARMFSA-N 0.000 description 1
- DMASLKHVQRHNES-UPOGUZCLSA-N (3R)-beta,beta-caroten-3-ol Chemical compound C([C@H](O)CC=1C)C(C)(C)C=1/C=C/C(/C)=C/C=C/C(/C)=C/C=C/C=C(C)C=CC=C(C)C=CC1=C(C)CCCC1(C)C DMASLKHVQRHNES-UPOGUZCLSA-N 0.000 description 1
- CABVTRNMFUVUDM-VRHQGPGLSA-N (3S)-3-hydroxy-3-methylglutaryl-CoA Chemical compound O[C@@H]1[C@H](OP(O)(O)=O)[C@@H](COP(O)(=O)OP(O)(=O)OCC(C)(C)[C@@H](O)C(=O)NCCC(=O)NCCSC(=O)C[C@@](O)(CC(O)=O)C)O[C@H]1N1C2=NC=NC(N)=C2N=C1 CABVTRNMFUVUDM-VRHQGPGLSA-N 0.000 description 1
- VFNKZQNIXUFLBC-UHFFFAOYSA-N 2',7'-dichlorofluorescein Chemical compound O1C(=O)C2=CC=CC=C2C21C1=CC(Cl)=C(O)C=C1OC1=C2C=C(Cl)C(O)=C1 VFNKZQNIXUFLBC-UHFFFAOYSA-N 0.000 description 1
- FWBHETKCLVMNFS-UHFFFAOYSA-N 4',6-Diamino-2-phenylindol Chemical compound C1=CC(C(=N)N)=CC=C1C1=CC2=CC=C(C(N)=N)C=C2N1 FWBHETKCLVMNFS-UHFFFAOYSA-N 0.000 description 1
- 244000215068 Acacia senegal Species 0.000 description 1
- 102000018616 Apolipoproteins B Human genes 0.000 description 1
- 108010027006 Apolipoproteins B Proteins 0.000 description 1
- 206010003694 Atrophy Diseases 0.000 description 1
- 208000023275 Autoimmune disease Diseases 0.000 description 1
- 101800004538 Bradykinin Proteins 0.000 description 1
- 102400000967 Bradykinin Human genes 0.000 description 1
- 235000011299 Brassica oleracea var botrytis Nutrition 0.000 description 1
- 235000017647 Brassica oleracea var italica Nutrition 0.000 description 1
- 240000003259 Brassica oleracea var. botrytis Species 0.000 description 1
- 235000002566 Capsicum Nutrition 0.000 description 1
- 208000002177 Cataract Diseases 0.000 description 1
- 241000675108 Citrus tangerina Species 0.000 description 1
- 208000011231 Crohn disease Diseases 0.000 description 1
- 239000004212 Cryptoxanthin Substances 0.000 description 1
- 238000000116 DAPI staining Methods 0.000 description 1
- 208000019505 Deglutition disease Diseases 0.000 description 1
- 101710088194 Dehydrogenase Proteins 0.000 description 1
- 206010070901 Diabetic dyslipidaemia Diseases 0.000 description 1
- 235000011511 Diospyros Nutrition 0.000 description 1
- 241000723267 Diospyros Species 0.000 description 1
- 206010013654 Drug abuse Diseases 0.000 description 1
- 239000006144 Dulbecco’s modified Eagle's medium Substances 0.000 description 1
- KCXVZYZYPLLWCC-UHFFFAOYSA-N EDTA Chemical compound OC(=O)CN(CC(O)=O)CCN(CC(O)=O)CC(O)=O KCXVZYZYPLLWCC-UHFFFAOYSA-N 0.000 description 1
- 238000008157 ELISA kit Methods 0.000 description 1
- 206010048554 Endothelial dysfunction Diseases 0.000 description 1
- 208000018522 Gastrointestinal disease Diseases 0.000 description 1
- 208000012671 Gastrointestinal haemorrhages Diseases 0.000 description 1
- 108010024636 Glutathione Proteins 0.000 description 1
- 239000004471 Glycine Substances 0.000 description 1
- 229920000084 Gum arabic Polymers 0.000 description 1
- QXZGBUJJYSLZLT-UHFFFAOYSA-N H-Arg-Pro-Pro-Gly-Phe-Ser-Pro-Phe-Arg-OH Natural products NC(N)=NCCCC(N)C(=O)N1CCCC1C(=O)N1C(C(=O)NCC(=O)NC(CC=2C=CC=CC=2)C(=O)NC(CO)C(=O)N2C(CCC2)C(=O)NC(CC=2C=CC=CC=2)C(=O)NC(CCCN=C(N)N)C(O)=O)CCC1 QXZGBUJJYSLZLT-UHFFFAOYSA-N 0.000 description 1
- 206010018985 Haemorrhage intracranial Diseases 0.000 description 1
- HTTJABKRGRZYRN-UHFFFAOYSA-N Heparin Chemical compound OC1C(NC(=O)C)C(O)OC(COS(O)(=O)=O)C1OC1C(OS(O)(=O)=O)C(O)C(OC2C(C(OS(O)(=O)=O)C(OC3C(C(O)C(O)C(O3)C(O)=O)OS(O)(=O)=O)C(CO)O2)NS(O)(=O)=O)C(C(O)=O)O1 HTTJABKRGRZYRN-UHFFFAOYSA-N 0.000 description 1
- 206010019799 Hepatitis viral Diseases 0.000 description 1
- 208000031226 Hyperlipidaemia Diseases 0.000 description 1
- 208000001953 Hypotension Diseases 0.000 description 1
- 206010021143 Hypoxia Diseases 0.000 description 1
- 206010022489 Insulin Resistance Diseases 0.000 description 1
- 102000008070 Interferon-gamma Human genes 0.000 description 1
- 108010074328 Interferon-gamma Proteins 0.000 description 1
- 102000003777 Interleukin-1 beta Human genes 0.000 description 1
- 108090000193 Interleukin-1 beta Proteins 0.000 description 1
- 102000019223 Interleukin-1 receptor Human genes 0.000 description 1
- 108050006617 Interleukin-1 receptor Proteins 0.000 description 1
- 208000008574 Intracranial Hemorrhages Diseases 0.000 description 1
- ZDXPYRJPNDTMRX-VKHMYHEASA-N L-glutamine Chemical compound OC(=O)[C@@H](N)CCC(N)=O ZDXPYRJPNDTMRX-VKHMYHEASA-N 0.000 description 1
- UPYKUZBSLRQECL-UKMVMLAPSA-N Lycopene Natural products CC(=C/C=C/C=C(C)/C=C/C=C(C)/C=C/C1C(=C)CCCC1(C)C)C=CC=C(/C)C=CC2C(=C)CCCC2(C)C UPYKUZBSLRQECL-UKMVMLAPSA-N 0.000 description 1
- 208000001344 Macular Edema Diseases 0.000 description 1
- 206010025415 Macular oedema Diseases 0.000 description 1
- 108020005196 Mitochondrial DNA Proteins 0.000 description 1
- 208000023178 Musculoskeletal disease Diseases 0.000 description 1
- XJLXINKUBYWONI-NNYOXOHSSA-O NADP(+) Chemical compound NC(=O)C1=CC=C[N+]([C@H]2[C@@H]([C@H](O)[C@@H](COP(O)(=O)OP(O)(=O)OC[C@@H]3[C@H]([C@@H](OP(O)(O)=O)[C@@H](O3)N3C4=NC=NC(N)=C4N=C3)O)O2)O)=C1 XJLXINKUBYWONI-NNYOXOHSSA-O 0.000 description 1
- 208000012902 Nervous system disease Diseases 0.000 description 1
- 208000025966 Neurological disease Diseases 0.000 description 1
- 206010067482 No adverse event Diseases 0.000 description 1
- 206010030113 Oedema Diseases 0.000 description 1
- 241000758706 Piperaceae Species 0.000 description 1
- 235000010582 Pisum sativum Nutrition 0.000 description 1
- 240000004713 Pisum sativum Species 0.000 description 1
- 241000288906 Primates Species 0.000 description 1
- 206010061481 Renal injury Diseases 0.000 description 1
- 206010063837 Reperfusion injury Diseases 0.000 description 1
- 241000283984 Rodentia Species 0.000 description 1
- 235000009337 Spinacia oleracea Nutrition 0.000 description 1
- 244000300264 Spinacia oleracea Species 0.000 description 1
- 208000010040 Sprains and Strains Diseases 0.000 description 1
- 208000006011 Stroke Diseases 0.000 description 1
- 229930006000 Sucrose Natural products 0.000 description 1
- 102000003929 Transaminases Human genes 0.000 description 1
- 108090000340 Transaminases Proteins 0.000 description 1
- 238000010162 Tukey test Methods 0.000 description 1
- 206010067584 Type 1 diabetes mellitus Diseases 0.000 description 1
- 206010052428 Wound Diseases 0.000 description 1
- 238000002835 absorbance Methods 0.000 description 1
- 239000000205 acacia gum Substances 0.000 description 1
- 235000010489 acacia gum Nutrition 0.000 description 1
- 230000009471 action Effects 0.000 description 1
- 239000012190 activator Substances 0.000 description 1
- 239000004480 active ingredient Substances 0.000 description 1
- 230000001154 acute effect Effects 0.000 description 1
- 210000000577 adipose tissue Anatomy 0.000 description 1
- 239000002671 adjuvant Substances 0.000 description 1
- 230000032683 aging Effects 0.000 description 1
- 108010081577 aldehyde dehydrogenase (NAD(P)+) Proteins 0.000 description 1
- 150000001299 aldehydes Chemical class 0.000 description 1
- OENHQHLEOONYIE-UKMVMLAPSA-N all-trans beta-carotene Natural products CC=1CCCC(C)(C)C=1/C=C/C(/C)=C/C=C/C(/C)=C/C=C/C=C(C)C=CC=C(C)C=CC1=C(C)CCCC1(C)C OENHQHLEOONYIE-UKMVMLAPSA-N 0.000 description 1
- 229930002945 all-trans-retinaldehyde Natural products 0.000 description 1
- 230000000172 allergic effect Effects 0.000 description 1
- 150000001412 amines Chemical class 0.000 description 1
- 230000003527 anti-angiogenesis Effects 0.000 description 1
- 229940124599 anti-inflammatory drug Drugs 0.000 description 1
- 230000000259 anti-tumor effect Effects 0.000 description 1
- 238000013459 approach Methods 0.000 description 1
- QVGXLLKOCUKJST-UHFFFAOYSA-N atomic oxygen Chemical compound [O] QVGXLLKOCUKJST-UHFFFAOYSA-N 0.000 description 1
- 208000010668 atopic eczema Diseases 0.000 description 1
- 230000037444 atrophy Effects 0.000 description 1
- 239000002012 ayurvedic medicine Substances 0.000 description 1
- 235000013734 beta-carotene Nutrition 0.000 description 1
- 239000011648 beta-carotene Substances 0.000 description 1
- TUPZEYHYWIEDIH-WAIFQNFQSA-N beta-carotene Natural products CC(=C/C=C/C=C(C)/C=C/C=C(C)/C=C/C1=C(C)CCCC1(C)C)C=CC=C(/C)C=CC2=CCCCC2(C)C TUPZEYHYWIEDIH-WAIFQNFQSA-N 0.000 description 1
- 235000002360 beta-cryptoxanthin Nutrition 0.000 description 1
- DMASLKHVQRHNES-ITUXNECMSA-N beta-cryptoxanthin Natural products CC(=C/C=C/C=C(C)/C=C/C=C(C)/C=C/C1=C(C)CC(O)CC1(C)C)C=CC=C(/C)C=CC2=C(C)CCCC2(C)C DMASLKHVQRHNES-ITUXNECMSA-N 0.000 description 1
- 229960002747 betacarotene Drugs 0.000 description 1
- 230000000975 bioactive effect Effects 0.000 description 1
- 230000004071 biological effect Effects 0.000 description 1
- 230000008033 biological extinction Effects 0.000 description 1
- 230000033228 biological regulation Effects 0.000 description 1
- 230000001851 biosynthetic effect Effects 0.000 description 1
- 230000017531 blood circulation Effects 0.000 description 1
- 210000004155 blood-retinal barrier Anatomy 0.000 description 1
- 230000004378 blood-retinal barrier Effects 0.000 description 1
- QXZGBUJJYSLZLT-FDISYFBBSA-N bradykinin Chemical compound NC(=N)NCCC[C@H](N)C(=O)N1CCC[C@H]1C(=O)N1[C@H](C(=O)NCC(=O)N[C@@H](CC=2C=CC=CC=2)C(=O)N[C@@H](CO)C(=O)N2[C@@H](CCC2)C(=O)N[C@@H](CC=2C=CC=CC=2)C(=O)N[C@@H](CCCNC(N)=N)C(O)=O)CCC1 QXZGBUJJYSLZLT-FDISYFBBSA-N 0.000 description 1
- 210000004556 brain Anatomy 0.000 description 1
- 150000001746 carotenes Chemical class 0.000 description 1
- 235000005473 carotenes Nutrition 0.000 description 1
- 238000004113 cell culture Methods 0.000 description 1
- 230000030833 cell death Effects 0.000 description 1
- 230000003915 cell function Effects 0.000 description 1
- 230000001413 cellular effect Effects 0.000 description 1
- 230000010001 cellular homeostasis Effects 0.000 description 1
- 230000004700 cellular uptake Effects 0.000 description 1
- 210000003169 central nervous system Anatomy 0.000 description 1
- 239000003153 chemical reaction reagent Substances 0.000 description 1
- 231100000481 chemical toxicant Toxicity 0.000 description 1
- 238000011976 chest X-ray Methods 0.000 description 1
- 208000037893 chronic inflammatory disorder Diseases 0.000 description 1
- 208000019425 cirrhosis of liver Diseases 0.000 description 1
- 238000010411 cooking Methods 0.000 description 1
- 230000002596 correlated effect Effects 0.000 description 1
- 230000000875 corresponding effect Effects 0.000 description 1
- 235000019244 cryptoxanthin Nutrition 0.000 description 1
- 239000001215 curcuma longa l. root Substances 0.000 description 1
- 235000021438 curry Nutrition 0.000 description 1
- UQHKFADEQIVWID-UHFFFAOYSA-N cytokinin Natural products C1=NC=2C(NCC=C(CO)C)=NC=NC=2N1C1CC(O)C(CO)O1 UQHKFADEQIVWID-UHFFFAOYSA-N 0.000 description 1
- 239000004062 cytokinin Substances 0.000 description 1
- 230000001086 cytosolic effect Effects 0.000 description 1
- 230000007123 defense Effects 0.000 description 1
- 230000018044 dehydration Effects 0.000 description 1
- 238000006297 dehydration reaction Methods 0.000 description 1
- 230000002939 deleterious effect Effects 0.000 description 1
- 238000013461 design Methods 0.000 description 1
- 238000001784 detoxification Methods 0.000 description 1
- 230000035487 diastolic blood pressure Effects 0.000 description 1
- XEYBRNLFEZDVAW-ARSRFYASSA-N dinoprostone Chemical compound CCCCC[C@H](O)\C=C\[C@H]1[C@H](O)CC(=O)[C@@H]1C\C=C/CCCC(O)=O XEYBRNLFEZDVAW-ARSRFYASSA-N 0.000 description 1
- 230000002554 disease preventive effect Effects 0.000 description 1
- 230000003828 downregulation Effects 0.000 description 1
- 239000000975 dye Substances 0.000 description 1
- 235000013345 egg yolk Nutrition 0.000 description 1
- 210000002969 egg yolk Anatomy 0.000 description 1
- 230000002124 endocrine Effects 0.000 description 1
- 230000008694 endothelial dysfunction Effects 0.000 description 1
- YQGOJNYOYNNSMM-UHFFFAOYSA-N eosin Chemical compound [Na+].OC(=O)C1=CC=CC=C1C1=C2C=C(Br)C(=O)C(Br)=C2OC2=C(Br)C(O)=C(Br)C=C21 YQGOJNYOYNNSMM-UHFFFAOYSA-N 0.000 description 1
- 206010015037 epilepsy Diseases 0.000 description 1
- 230000007717 exclusion Effects 0.000 description 1
- 238000013401 experimental design Methods 0.000 description 1
- 239000000284 extract Substances 0.000 description 1
- 238000001125 extrusion Methods 0.000 description 1
- 210000000416 exudates and transudate Anatomy 0.000 description 1
- 239000000835 fiber Substances 0.000 description 1
- 239000000834 fixative Substances 0.000 description 1
- 238000012757 fluorescence staining Methods 0.000 description 1
- 235000012631 food intake Nutrition 0.000 description 1
- 230000007760 free radical scavenging Effects 0.000 description 1
- 230000002496 gastric effect Effects 0.000 description 1
- 210000005095 gastrointestinal system Anatomy 0.000 description 1
- 230000004110 gluconeogenesis Effects 0.000 description 1
- 230000023611 glucuronidation Effects 0.000 description 1
- ZDXPYRJPNDTMRX-UHFFFAOYSA-N glutamine Natural products OC(=O)C(N)CCC(N)=O ZDXPYRJPNDTMRX-UHFFFAOYSA-N 0.000 description 1
- 230000035876 healing Effects 0.000 description 1
- 210000002216 heart Anatomy 0.000 description 1
- 229960002897 heparin Drugs 0.000 description 1
- 229920000669 heparin Polymers 0.000 description 1
- 210000003547 hepatic macrophage Anatomy 0.000 description 1
- 206010073071 hepatocellular carcinoma Diseases 0.000 description 1
- 241000411851 herbal medicine Species 0.000 description 1
- 229960001340 histamine Drugs 0.000 description 1
- 229940088597 hormone Drugs 0.000 description 1
- 239000005556 hormone Substances 0.000 description 1
- 230000000871 hypocholesterolemic effect Effects 0.000 description 1
- 230000036543 hypotension Effects 0.000 description 1
- 230000007954 hypoxia Effects 0.000 description 1
- 230000028993 immune response Effects 0.000 description 1
- 230000000899 immune system response Effects 0.000 description 1
- 229960001438 immunostimulant agent Drugs 0.000 description 1
- 230000006872 improvement Effects 0.000 description 1
- 230000008595 infiltration Effects 0.000 description 1
- 238000001764 infiltration Methods 0.000 description 1
- 239000003978 infusion fluid Substances 0.000 description 1
- 230000000977 initiatory effect Effects 0.000 description 1
- 238000002347 injection Methods 0.000 description 1
- 239000007924 injection Substances 0.000 description 1
- 229910052500 inorganic mineral Inorganic materials 0.000 description 1
- 230000003993 interaction Effects 0.000 description 1
- 229960003130 interferon gamma Drugs 0.000 description 1
- 210000004347 intestinal mucosa Anatomy 0.000 description 1
- 230000008810 intracellular oxidative stress Effects 0.000 description 1
- 230000031146 intracellular signal transduction Effects 0.000 description 1
- 208000012947 ischemia reperfusion injury Diseases 0.000 description 1
- 208000037806 kidney injury Diseases 0.000 description 1
- 238000009533 lab test Methods 0.000 description 1
- 150000002617 leukotrienes Chemical class 0.000 description 1
- 239000002502 liposome Substances 0.000 description 1
- 238000004895 liquid chromatography mass spectrometry Methods 0.000 description 1
- 230000007774 longterm Effects 0.000 description 1
- 210000004698 lymphocyte Anatomy 0.000 description 1
- 210000002540 macrophage Anatomy 0.000 description 1
- 201000010230 macular retinal edema Diseases 0.000 description 1
- 239000011159 matrix material Substances 0.000 description 1
- 230000007246 mechanism Effects 0.000 description 1
- 230000007721 medicinal effect Effects 0.000 description 1
- 239000002609 medium Substances 0.000 description 1
- 230000004066 metabolic change Effects 0.000 description 1
- 229920000609 methyl cellulose Polymers 0.000 description 1
- 239000001923 methylcellulose Substances 0.000 description 1
- 235000010981 methylcellulose Nutrition 0.000 description 1
- 239000000693 micelle Substances 0.000 description 1
- 239000011707 mineral Substances 0.000 description 1
- 210000001616 monocyte Anatomy 0.000 description 1
- 230000000877 morphologic effect Effects 0.000 description 1
- 230000001338 necrotic effect Effects 0.000 description 1
- 230000006036 negative regulation of mitochondrial membrane potential Effects 0.000 description 1
- 210000005036 nerve Anatomy 0.000 description 1
- 230000004770 neurodegeneration Effects 0.000 description 1
- 208000015122 neurodegenerative disease Diseases 0.000 description 1
- 210000000440 neutrophil Anatomy 0.000 description 1
- 231100000957 no side effect Toxicity 0.000 description 1
- 239000000820 nonprescription drug Substances 0.000 description 1
- 231100000028 nontoxic concentration Toxicity 0.000 description 1
- 230000001473 noxious effect Effects 0.000 description 1
- 239000002773 nucleotide Substances 0.000 description 1
- 125000003729 nucleotide group Chemical group 0.000 description 1
- 239000008601 oleoresin Substances 0.000 description 1
- 210000004789 organ system Anatomy 0.000 description 1
- 238000012261 overproduction Methods 0.000 description 1
- 239000007800 oxidant agent Substances 0.000 description 1
- 239000001301 oxygen Substances 0.000 description 1
- 229910052760 oxygen Inorganic materials 0.000 description 1
- 229940094443 oxytocics prostaglandins Drugs 0.000 description 1
- 210000000496 pancreas Anatomy 0.000 description 1
- 239000012188 paraffin wax Substances 0.000 description 1
- 210000004738 parenchymal cell Anatomy 0.000 description 1
- 230000000737 periodic effect Effects 0.000 description 1
- 208000030613 peripheral artery disease Diseases 0.000 description 1
- 230000035699 permeability Effects 0.000 description 1
- 239000012466 permeate Substances 0.000 description 1
- 238000009520 phase I clinical trial Methods 0.000 description 1
- 238000001782 photodegradation Methods 0.000 description 1
- 230000004962 physiological condition Effects 0.000 description 1
- 235000019100 piperine Nutrition 0.000 description 1
- MXXWOMGUGJBKIW-YPCIICBESA-N piperine Chemical compound C=1C=C2OCOC2=CC=1/C=C/C=C/C(=O)N1CCCCC1 MXXWOMGUGJBKIW-YPCIICBESA-N 0.000 description 1
- 229940075559 piperine Drugs 0.000 description 1
- WVWHRXVVAYXKDE-UHFFFAOYSA-N piperine Natural products O=C(C=CC=Cc1ccc2OCOc2c1)C3CCCCN3 WVWHRXVVAYXKDE-UHFFFAOYSA-N 0.000 description 1
- 235000013824 polyphenols Nutrition 0.000 description 1
- 230000008092 positive effect Effects 0.000 description 1
- 238000010149 post-hoc-test Methods 0.000 description 1
- 230000003389 potentiating effect Effects 0.000 description 1
- 239000000955 prescription drug Substances 0.000 description 1
- 230000003449 preventive effect Effects 0.000 description 1
- 102000004196 processed proteins & peptides Human genes 0.000 description 1
- 108090000765 processed proteins & peptides Proteins 0.000 description 1
- 230000000750 progressive effect Effects 0.000 description 1
- 230000002035 prolonged effect Effects 0.000 description 1
- 230000001737 promoting effect Effects 0.000 description 1
- 150000003180 prostaglandins Chemical class 0.000 description 1
- 238000001243 protein synthesis Methods 0.000 description 1
- 230000017854 proteolysis Effects 0.000 description 1
- 208000020016 psychiatric disease Diseases 0.000 description 1
- 238000000746 purification Methods 0.000 description 1
- 230000007115 recruitment Effects 0.000 description 1
- 230000008929 regeneration Effects 0.000 description 1
- 238000011069 regeneration method Methods 0.000 description 1
- 210000004994 reproductive system Anatomy 0.000 description 1
- 230000029058 respiratory gaseous exchange Effects 0.000 description 1
- 230000000284 resting effect Effects 0.000 description 1
- 210000003660 reticulum Anatomy 0.000 description 1
- TUFFYSFVSYUHPA-UHFFFAOYSA-M rhodamine 123 Chemical compound [Cl-].COC(=O)C1=CC=CC=C1C1=C(C=CC(N)=C2)C2=[O+]C2=C1C=CC(N)=C2 TUFFYSFVSYUHPA-UHFFFAOYSA-M 0.000 description 1
- 230000035945 sensitivity Effects 0.000 description 1
- 229940076279 serotonin Drugs 0.000 description 1
- 230000011664 signaling Effects 0.000 description 1
- 210000003491 skin Anatomy 0.000 description 1
- 210000000813 small intestine Anatomy 0.000 description 1
- 230000000391 smoking effect Effects 0.000 description 1
- 235000011888 snacks Nutrition 0.000 description 1
- 238000002798 spectrophotometry method Methods 0.000 description 1
- 235000013599 spices Nutrition 0.000 description 1
- 238000000528 statistical test Methods 0.000 description 1
- 230000000638 stimulation Effects 0.000 description 1
- 239000011550 stock solution Substances 0.000 description 1
- 229960005322 streptomycin Drugs 0.000 description 1
- 208000011117 substance-related disease Diseases 0.000 description 1
- 239000005720 sucrose Substances 0.000 description 1
- 239000006228 supernatant Substances 0.000 description 1
- 238000001356 surgical procedure Methods 0.000 description 1
- 238000003786 synthesis reaction Methods 0.000 description 1
- 230000009885 systemic effect Effects 0.000 description 1
- 230000035488 systolic blood pressure Effects 0.000 description 1
- 238000012360 testing method Methods 0.000 description 1
- 150000003595 thromboxanes Chemical class 0.000 description 1
- 230000000451 tissue damage Effects 0.000 description 1
- 231100000827 tissue damage Toxicity 0.000 description 1
- 230000019432 tissue death Effects 0.000 description 1
- 239000003440 toxic substance Substances 0.000 description 1
- 230000001988 toxicity Effects 0.000 description 1
- 231100000419 toxicity Toxicity 0.000 description 1
- 238000013518 transcription Methods 0.000 description 1
- 230000035897 transcription Effects 0.000 description 1
- 230000014616 translation Effects 0.000 description 1
- 230000032258 transport Effects 0.000 description 1
- 230000004102 tricarboxylic acid cycle Effects 0.000 description 1
- 230000002477 vacuolizing effect Effects 0.000 description 1
- 230000002227 vasoactive effect Effects 0.000 description 1
- 230000035899 viability Effects 0.000 description 1
- 201000001862 viral hepatitis Diseases 0.000 description 1
- 230000000007 visual effect Effects 0.000 description 1
- 239000011782 vitamin Substances 0.000 description 1
- 229930003231 vitamin Natural products 0.000 description 1
- 235000013343 vitamin Nutrition 0.000 description 1
- 229940088594 vitamin Drugs 0.000 description 1
- 150000003722 vitamin derivatives Chemical class 0.000 description 1
- 238000005406 washing Methods 0.000 description 1
- 238000005303 weighing Methods 0.000 description 1
- 235000008210 xanthophylls Nutrition 0.000 description 1
- OENHQHLEOONYIE-JLTXGRSLSA-N β-Carotene Chemical compound CC=1CCCC(C)(C)C=1\C=C\C(\C)=C\C=C\C(\C)=C\C=C\C=C(/C)\C=C\C=C(/C)\C=C\C1=C(C)CCCC1(C)C OENHQHLEOONYIE-JLTXGRSLSA-N 0.000 description 1
Landscapes
- Medicines Containing Material From Animals Or Micro-Organisms (AREA)
- Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
Abstract
Description
本發明係關於一種治療性草藥組合物,其包含植物性化合物(即類薑黃素及類胡蘿蔔素(諸如葉黃素))之新穎組合。更特別地,本發明係關於一種包含類薑黃素及葉黃素之治療性草藥組合物。本發明之治療性草藥組合物有利於人類之整體健康。該治療性草藥組合物有效調節血脂概況及調節發炎效應。本發明亦提供一種用於製造此組合物之方法及此組合物於食物、飲料(酒精飲料)中及作為營養品、膳食補充劑及藥物之用途。 The present invention relates to a therapeutic herbal composition comprising a novel combination of botanical compounds, namely curcuminoids and carotenoids such as lutein. More particularly, the present invention relates to a therapeutic herbal composition comprising curcuminoids and lutein. The therapeutic herbal compositions of the present invention are beneficial to the general health of humans. The therapeutic herbal composition effectively regulates blood lipid profile and modulates inflammatory effects. The invention also provides a method for the manufacture of the composition and the use of the composition in food, beverages (alcoholic beverages) and as nutraceuticals, dietary supplements and medicines.
發炎係免疫系統保護生物體免受感染及傷害之反應並藉由去除受損組織組分發揮作用,使得身體開始癒合。僅持續幾天之發炎反應稱為急性發炎,而持續時間較長之反應稱為慢性發炎。感染物/病原體(諸如病毒及細菌)係一些最常見之發炎刺激物。其他可刺激發炎之因素包括物理藥劑、化學物質、不適當之免疫反應及組織死亡。在許多器官系統(包括心臟、胰、肝、腎、肺、腦、腸道及生殖系統)中觀測到急性及慢性發炎介導之組織損傷。發炎係許多慢性疾病(包括癌症、動脈粥樣硬化、胰島素抗性、2型糖尿病及心血管疾病)之常見發病機制。 Inflammation is the immune system's response to protect an organism from infection and injury and works by removing damaged tissue components so the body can begin healing. Inflammation that lasts only a few days is called acute inflammation, and one that lasts longer is called chronic inflammation. Infectious agents/pathogens such as viruses and bacteria are some of the most common inflammatory stimuli. Other factors that can stimulate inflammation include physical agents, chemicals, inappropriate immune responses, and tissue death. Acute and chronic inflammation-mediated tissue damage has been observed in many organ systems, including heart, pancreas, liver, kidney, lung, brain, gut, and reproductive system. Inflammation is a common pathogenesis of many chronic diseases, including cancer, atherosclerosis, insulin resistance, type 2 diabetes, and cardiovascular disease.
儘管發炎反應過程取決於初始刺激之精確性質及其於體內 之位置,但其等均共用一個共同機制,該機制可總結如下:1)細胞表面模式受體識別有害刺激物;2)活化發炎途徑;3)釋放發炎標記;及4)募集發炎細胞。發炎途徑影響許多慢性疾病之發病機制,且涉及常見發炎介質及調節途徑。自循環系統、發炎細胞及受損組織釋放各種化學介質以調節發炎反應。釋放之化學介質包括(1)血管活性胺,諸如組織胺及血清素,(2)肽(例如,緩激肽),及(3)類花生酸(例如,血栓素、白三烯及前列腺素)。前列腺素E2(PGE2)(一種調節分子之類花生酸家族之成員)於發炎部位大量產生,並參與導致發炎典型徵象之所有過程:發紅;腫脹及疼痛。發紅及水腫由流入發炎組織內之血流增加導致。 Although the course of the inflammatory response depends on the precise nature of the initial stimulus and its However, they all share a common mechanism, which can be summarized as follows: 1) recognition of noxious stimuli by cell surface pattern receptors; 2) activation of inflammatory pathways; 3) release of inflammatory markers; and 4) recruitment of inflammatory cells. Inflammatory pathways influence the pathogenesis of many chronic diseases and involve common inflammatory mediators and regulatory pathways. Various chemical mediators are released from the circulatory system, inflammatory cells and damaged tissues to regulate the inflammatory response. Chemical mediators released include (1) vasoactive amines such as histamine and serotonin, (2) peptides (e.g., bradykinin), and (3) eicosanoids (e.g., thromboxanes, leukotrienes, and prostaglandins ). Prostaglandin E2 (PGE2), a member of the eicosanoid family of regulatory molecules, is produced in large quantities at inflamed sites and is involved in all processes leading to the typical signs of inflammation: redness; swelling and pain. Redness and edema result from increased blood flow into inflamed tissue.
各種研究已表明,發炎細胞介素關於各種發炎疾病發揮重要作用。細胞介素(諸如介白素-1β(IL-1β)、介白素-6(IL-6)及腫瘤壞死因子-α(TNF-α))通過與TLR、IL-1受體(IL-1R)、IL-6受體(IL-6R)及TNF受體(TNFR)相互作用介導發炎。受體活化觸發重要之細胞內訊息傳遞路徑,包括促分裂原活化蛋白激酶(MAPK)、核因子κ-B(NF-κB)及Janus激酶(JAK)-信號轉導與轉錄活化蛋白(STAT)途徑。該腫瘤壞死因子-α(TNF-α)係由多種細胞類型(包括巨噬球、嗜中性球及淋巴球)產生之細胞介素。 Various studies have shown that inflammatory cytokines play an important role in various inflammatory diseases. Interleukins (such as interleukin-1β (IL-1β), interleukin-6 (IL-6) and tumor necrosis factor-α (TNF-α)) through the interaction with TLR, IL-1 receptor (IL- 1R), IL-6 receptor (IL-6R), and TNF receptor (TNFR) interact to mediate inflammation. Receptor activation triggers important intracellular signaling pathways including mitogen-activated protein kinase (MAPK), nuclear factor kappa-B (NF-κB), and Janus kinase (JAK)-signal transducer and activator of transcription (STAT) way. The tumor necrosis factor-α (TNF-α) is a cytokine produced by various cell types including macrophages, neutrophils and lymphocytes.
一氧化氮(NO)係在發炎之發病機制中發揮關鍵作用之訊息傳遞分子。一氧化氮在正常生理條件下產生抗發炎效應。另一方面,一氧化氮由於異常狀況中之過度產生而誘導發炎。一氧化氮已與急性及慢性發炎疾患相關,包括各種類型之關節炎、胃腸道疾病、中樞神經系統之發炎病症及某些形式之哮喘,且視為促發炎標記。NO之活體內產生由一氧化氮合成酶(NOS)酵素家族(包括誘導型一氧化氮合成酶(I-NOS))介導,NOS由許多不同之免疫刺激物(包括脂多醣(LPS)、干擾素γ及介白素-1 (IL-1))活化。因此,認為NO抑制劑適用於治療及/或控制發炎疾患。天然產物提供大量NO抑制劑。水果及蔬菜之疾病預防能力已歸因於此等膳食來源中存在之抗氧化劑/多酚。 Nitric oxide (NO) is a signaling molecule that plays a key role in the pathogenesis of inflammation. Nitric oxide produces anti-inflammatory effects under normal physiological conditions. Nitric oxide, on the other hand, induces inflammation due to its overproduction in abnormal conditions. Nitric oxide has been associated with acute and chronic inflammatory disorders, including various types of arthritis, gastrointestinal disorders, inflammatory disorders of the central nervous system, and certain forms of asthma, and is considered a pro-inflammatory marker. In vivo production of NO is mediated by the nitric oxide synthase (NOS) enzyme family, including inducible nitric oxide synthase (I-NOS), which is mediated by many different immune stimulants, including lipopolysaccharide (LPS), Interferon gamma and interleukin-1 (IL-1)) activation. Therefore, NO inhibitors are considered useful in the treatment and/or control of inflammatory disorders. Natural products provide a large number of NO inhibitors. The disease preventive capabilities of fruits and vegetables have been attributed to the antioxidants/polyphenols present in these dietary sources.
肝發炎保護此器官免受感染及損傷,但過度發炎可導致肝細胞大量損失、缺血再灌注損傷、代謝改變並最終導致永久性肝損傷。發炎可破壞肝實質細胞,增加慢性肝病(諸如非酒精性脂肪肝疾病(NAFLD)或病毒性肝炎)之風險。肝係生命器官,在各種內源性及外源性有害物質之代謝及解毒中發揮重要作用。肝中發生藥物之結構改變,導致生物活性或無活性代謝物且此等中之一些有毒。因此,肝係來自各種化學物質及藥物之損傷之脆弱目標。肝病之主要原因係毒性化學物質、過量飲酒、感染及自體免疫性疾病。 Inflammation of the liver protects this organ from infection and injury, but excessive inflammation can lead to massive loss of hepatocytes, ischemia-reperfusion injury, metabolic changes and eventually permanent liver damage. Inflammation can destroy liver parenchymal cells, increasing the risk of chronic liver diseases such as non-alcoholic fatty liver disease (NAFLD) or viral hepatitis. The liver is a vital organ and plays an important role in the metabolism and detoxification of various endogenous and exogenous harmful substances. Structural changes of the drug occur in the liver leading to bioactive or inactive metabolites and some of these are toxic. Thus, the liver is a vulnerable target for damage from various chemicals and drugs. The main causes of liver disease are toxic chemicals, excessive alcohol consumption, infections and autoimmune diseases.
此外,已報導氧化代謝過程中產生的各種發炎刺激物(諸如過量活性氧物質(ROS)/反應性氮物質(RNS))及一些天然或人工化學物質啟動導致促發炎細胞介素之合成及分泌之發炎過程。氧化壓力係與多種疾病(諸如心血管疾病、癌症、糖尿病、高血壓、衰老及動脈粥樣硬化)之發病機制有關。氧化壓力係指ROS及氧化劑之產生與抗氧化劑之對抗活性之間的失衡。已認為氧化壓力係肝損傷之啟動及進展之主要因素。已知各種風險因素(諸如酒精及藥物)誘導氧化壓力,其進一步可導致慢性疾病(諸如脂肪肝疾病)。脂肪肝疾病包含酒精性肝病(ALD)及非酒精性脂肪肝疾病(NAFLD)。越來越多的證據表明,由活性氧物質(ROS)之產生引起之氧化壓力在ALD及NAFLD之進展中起關鍵作用。ROS之過量產生在細胞功能中產生有害影響,最終導致脂肪肝疾病。在粒線體中,ROS產生增加可引起mtDNA耗竭、攻擊生物分子(即,蛋白質、碳水化合物及脂質)並損害 粒線體膜。值得注意地,粒線體具有大量濃度的含有多不飽和脂肪酸(PUFA)之磷脂。該等PUFA更容易受到氧化損傷,因為其等化學結構中存在雙鍵,其導致脂質過氧化。PUFA過氧化增強ApoB之內質網後分泌前蛋白水解,藉此減少極低密度脂蛋白(VLDL)分泌;此可進一步促進肝中甘油三酯(TG)蓄積。此外,通過PUFA過氧化形成之醛損害細胞穩態,因為此等分子影響核苷酸及蛋白質合成,減少肝麩胱甘肽含量並增加促發炎細胞介素TNF-α之產生。此等影響導致肝細胞死亡及壞死、發炎及肝纖維化。 In addition, various inflammatory stimuli produced during oxidative metabolism (such as excess reactive oxygen species (ROS)/reactive nitrogen species (RNS)) and some natural or artificial chemicals have been reported to initiate the synthesis and secretion of pro-inflammatory cytokines the inflammatory process. Oxidative stress has been implicated in the pathogenesis of various diseases such as cardiovascular disease, cancer, diabetes, hypertension, aging and atherosclerosis. Oxidative stress refers to the imbalance between the production of ROS and oxidants and the counteracting activity of antioxidants. Oxidative stress has been considered a major factor in the initiation and progression of liver injury. Various risk factors such as alcohol and drugs are known to induce oxidative stress which in turn can lead to chronic diseases such as fatty liver disease. Fatty liver disease includes alcoholic liver disease (ALD) and non-alcoholic fatty liver disease (NAFLD). Accumulating evidence indicates that oxidative stress caused by generation of reactive oxygen species (ROS) plays a key role in the progression of ALD and NAFLD. Excessive production of ROS has deleterious effects in cellular functions, ultimately leading to fatty liver disease. In mitochondria, increased ROS production can cause mtDNA depletion, attack biomolecules (ie, proteins, carbohydrates, and lipids) and damage mitochondrial membrane. Notably, mitochondria have substantial concentrations of phospholipids containing polyunsaturated fatty acids (PUFA). These PUFAs are more susceptible to oxidative damage due to the presence of double bonds in their chemical structures, which lead to lipid peroxidation. PUFA peroxidation enhances post-endoplasmic reticulum post-presecretory proteolysis of ApoB, thereby reducing very low-density lipoprotein (VLDL) secretion; this can further promote triglyceride (TG) accumulation in the liver. Furthermore, aldehydes formed by PUFA peroxidation impair cellular homeostasis, as these molecules affect nucleotide and protein synthesis, reduce hepatic glutathione content and increase production of the pro-inflammatory interleukin TNF-α. These effects lead to liver cell death and necrosis, inflammation and liver fibrosis.
肝損傷通過數個相互關聯之途徑發生。酒精代謝主要發生在肝中。乙醇代謝之主要途徑係脫氫酶系統。酒精脫氫酶及乙醛脫氫酶使菸鹼醯胺腺嘌呤二核苷酸(NAD)還原為NADH(NAD之還原形式)。NAD/NADH比率之改變通過抑制糖質新生及脂肪酸氧化促進脂肪肝。慢性酒精曝露亦活化肝巨噬球,其然後產生腫瘤壞死因子-α(TNF-α)。TNF-α誘導粒線體增加活性氧物質(ROS)之產生。此氧化壓力促進肝細胞壞死及細胞凋亡。 Liver injury occurs through several interrelated pathways. Alcohol metabolism occurs primarily in the liver. The main pathway of ethanol metabolism is the dehydrogenase system. Alcohol dehydrogenase and acetaldehyde dehydrogenase reduce nicotinamide adenine dinucleotide (NAD) to NADH (the reduced form of NAD). Changes in the NAD/NADH ratio promote fatty liver by inhibiting gluconeogenesis and fatty acid oxidation. Chronic alcohol exposure also activates hepatic macrophages, which then produce tumor necrosis factor-α (TNF-α). TNF-α induces mitochondria to increase the production of reactive oxygen species (ROS). This oxidative stress promotes hepatocyte necrosis and apoptosis.
因此,氧化壓力產物亦可用作發炎反應之標記。 Therefore, oxidative stress products can also be used as markers of inflammatory responses.
糖尿病性視網膜病變(DR)係糖尿病併發症,其影響眼睛,導致視力殘疾及失明。常伴有脂質滲出。血脂異常導致硬性滲出物及臨床顯著黃斑水腫(CSME)之發展。此等進一步干擾視力。升高之脂質濃度係與內皮功能障礙相關聯,該內皮功能障礙似乎在糖尿病性視網膜病變之發病機制中發揮重要作用,特別與血液-視網膜屏障之破壞相關。最近,糖尿病控制及併發症試驗(DCCT)樣本中血脂概況之分析在DR之發展與1型糖尿病中之血脂異常之間建立緊密關聯,且數項使用降脂藥物之臨床試驗 表明血脂異常與2型糖尿病中之DR進展之間存在關聯(Action to Control Cardiovascular Risk in Diabetes Follow-On,2016,Frank,2014,Matthews,2011,Wright and Dodson,2011)。 Diabetic retinopathy (DR) is a complication of diabetes that affects the eyes, causing visual disability and blindness. Often accompanied by lipid exudation. Dyslipidemia leads to hard exudates and the development of clinically significant macular edema (CSME). These further interfere with vision. Elevated lipid concentrations are associated with endothelial dysfunction, which appears to play an important role in the pathogenesis of diabetic retinopathy, particularly associated with disruption of the blood-retinal barrier. Recently, analysis of lipid profiles in Diabetes Control and Complications Trial (DCCT) samples established a strong association between the development of DR and dyslipidemia in type 1 diabetes, and several clinical trials using lipid-lowering drugs showed an association between dyslipidemia and the progression of DR in type 2 diabetes (Action to Control Cardiovascular Risk in Diabetes Follow-On, 2016, Frank, 2014, Matthews, 2011, Wright and Dodson, 2011).
在靈長類動物視網膜黃斑中及周圍發現葉黃素類胡蘿蔔素葉黃素及玉米黃質,其中將其等稱為黃斑色素(MP)。膳食葉黃素及玉米黃質在腸道中與脂肪一起吸收,且最終傳輸至肝。將肝內之葉黃素及玉米黃質併入脂蛋白分子內,該等分子作為類胡蘿蔔素-脂蛋白複合物之部分在整個血流中運輸。MP及血清脂蛋白兩者均已與神經退化性疾病(諸如年齡相關性黃斑變性(AMD))之風險相關。AMD係慢性、進行性、退化性眼病,其影響負責最高視力之中央視網膜。其已成為發達國家及發展中國家老年人法定失明之主要原因。高密度脂蛋白(HDL)(與AMD風險降低相關)及低密度脂蛋白(LDL)(與風險增加相關)兩者上均攜載葉黃素及玉米黃質。因此,據報導血清葉黃素、視網膜葉黃素及脂蛋白濃度有顯著相關性,及脂蛋白濃度之變化可影響視網膜葉黃素之濃度。亦已顯示與外視網膜中低等級發炎及缺氧相關之氧化壓力在AMD之發病機制中係重要的。此與低黃斑色素光學密度(MPOD)可為AMD之風險因素的觀點一致,因為黃斑類胡蘿蔔素顯示作為抗氧化劑之有效性質。 The lutein carotenoids lutein and zeaxanthin are found in and around the macula of the primate retina, where they are known as the macular pigment (MP). Dietary lutein and zeaxanthin are absorbed with fat in the gut and are eventually transported to the liver. Intrahepatic lutein and zeaxanthin are incorporated into lipoprotein molecules, which are transported throughout the bloodstream as part of carotenoid-lipoprotein complexes. Both MP and serum lipoproteins have been associated with the risk of neurodegenerative diseases such as age-related macular degeneration (AMD). AMD is a chronic, progressive, degenerative eye disease that affects the central retina responsible for highest vision. It has become the leading cause of legal blindness among older adults in both developed and developing countries. Both high-density lipoprotein (HDL) (associated with reduced risk of AMD) and low-density lipoprotein (LDL) (associated with increased risk) carry lutein and zeaxanthin. Therefore, it has been reported that serum lutein, retinal lutein, and lipoprotein concentrations are significantly correlated, and changes in lipoprotein concentrations can affect retinal lutein concentrations. Oxidative stress associated with low-grade inflammation and hypoxia in the outer retina has also been shown to be important in the pathogenesis of AMD. This is consistent with the idea that low macular pigment optical density (MPOD) may be a risk factor for AMD, since macular carotenoids exhibit potent properties as antioxidants.
因此,藉由使用抗發炎劑抑制及/或調節上文描述之發炎介質中之一或多者限制發炎過程對控制此過程並限制由其引起之疾病而言係重要的。然而,廣泛使用之合成抗發炎劑/藥物似乎長期使用將產生副作用。因此,仍需來自天然產物之更新穎且更好的抗發炎劑。 Therefore, limiting the inflammatory process by using anti-inflammatory agents to inhibit and/or modulate one or more of the inflammatory mediators described above is important to control this process and limit the diseases caused by it. However, widely used synthetic anti-inflammatory agents/drugs appear to have side effects with long-term use. Therefore, there remains a need for newer and better anti-inflammatory agents from natural products.
此外,如上文描述,維持身體之脂質概況對預防肝損傷(肝係生命器官)同樣重要,並藉此有助於控制由於諸如發炎、氧化壓力等因 素引起之疾病,及諸如糖尿病性視網膜病變及年齡相關性黃斑變性之疾病。 In addition, as described above, maintaining the lipid profile of the body is also important to prevent liver damage (the liver is a vital organ) and thereby help control Diseases caused by hormones, and diseases such as diabetic retinopathy and age-related macular degeneration.
植物性化合物係植物中存在的用於食物及藥品之生物活性化合物。植物性化合物(諸如類薑黃素及類胡蘿蔔素(諸如葉黃素及玉米黃質))因其促進健康之性質而聞名。 Phytochemicals are biologically active compounds found in plants that are used in food and medicine. Phytochemicals such as curcuminoids and carotenoids such as lutein and zeaxanthin are known for their health-promoting properties.
薑黃(Turmeric(Curcuma longa))係流行之印度香料,其係屬於薑科(ginger family(Zingiberaceae))的多年生根莖草本植物,其原產於南亞熱帶地區。薑黃主要因其在印度菜餚中作為咖喱及其他民族美食中之常見成分而聞名。薑黃在阿育吠陀醫學中亦使用數個世紀,阿育吠陀醫學將草藥之藥用性質與食物相結合。在阿育吠陀醫學系統中,已向薑黃分配許多治療性活性以用於範圍廣泛之疾病及病症中,包括彼等皮膚、肺、腸胃系統、酸痛、疼痛、傷口、扭傷及肝病。類薑黃素係薑黃根莖中之多酚化合物且係薑黃呈黃色之原因。類薑黃素佔乾薑黃根粉末之2至4%。薑黃素係薑黃之主要類薑黃素,及另外兩種類薑黃素係脫甲氧基薑黃素及雙脫甲氧基薑黃素。薑黃素之生物效應範圍從抗氧化、抗發炎至抑制血管生成且亦顯示具有特定抗腫瘤活性。已發現薑黃具有抗氧化性質,以及其具有減少促發炎細胞介素形成之能力。 Turmeric ( Curcuma longa ) is a popular Indian spice, which is a perennial rhizome herbaceous plant belonging to the ginger family ( Zingiberaceae ), which is native to the south subtropical region. Turmeric is primarily known for its use in Indian cuisine as a common ingredient in curries and other ethnic cuisines. Turmeric has also been used for centuries in Ayurvedic medicine, which combines the medicinal properties of the herb with food. In the Ayurvedic system of medicine, turmeric has been assigned many therapeutic activities for use in a wide range of diseases and conditions, including those of the skin, lungs, gastrointestinal system, aches, pains, wounds, sprains and liver diseases. Curcuminoids are polyphenolic compounds in turmeric rhizomes and are responsible for the yellow color of turmeric. Curcuminoids make up 2 to 4% of dry turmeric root powder. Curcumin is the main curcuminoid of turmeric, and the other two curcuminoids are demethoxycurcumin and bisdemethoxycurcumin. The biological effects of curcumin range from anti-oxidative, anti-inflammatory to anti-angiogenesis and have also been shown to have specific anti-tumor activity. Turmeric has been found to have antioxidant properties, as well as its ability to reduce the formation of pro-inflammatory cytokines.
薑黃素可以薑黃、薑黃之濃縮物、類薑黃素粉、基本上純(95%)類薑黃素或薑黃素的形式單獨投與。極大限制薑黃素之有效性及實用性之問題在於其低生體可用率,此歸因於水不溶性、生理pH下之不穩定性、光降解、細胞吸收緩慢及快速代謝成無活性代謝物。薑黃素之水溶性估計為11ng/mL(https://pubchem.ncbi.nlm.nih.gov/compound/curcumin)。 Curcumin may be administered alone in the form of turmeric, turmeric concentrate, curcuminoid powder, substantially pure (95%) curcuminoid, or curcuminoids. A problem that greatly limits the effectiveness and utility of curcumin is its low bioavailability due to water insolubility, instability at physiological pH, photodegradation, slow cellular uptake and rapid metabolism to inactive metabolites. The water solubility of curcumin is estimated to be 11 ng/mL (https://pubchem.ncbi.nlm.nih.gov/compound/curcumin).
動物中之藥物動力學研究已證實40至85%經口劑量之薑黃 素通過胃腸道時無變化,及大多數吸收之類黃酮在腸黏膜及肝中代謝。人類及嚙齒類動物中之許多藥物動力學研究已顯示薑黃素具有非常低之生體可用率,從而限制其益處。此低生體可用率很可能由於其在腸道及肝中之低吸收及快速代謝及快速消除。由於薑黃素的低吸收率,因此薑黃素通常與其他植物化合物調配用於增加吸收並增強抗發炎效應。 Pharmacokinetic studies in animals have demonstrated that 40 to 85% of oral doses of turmeric Flavonoids pass through the gastrointestinal tract unchanged, and most absorbed flavonoids are metabolized in the intestinal mucosa and liver. Numerous pharmacokinetic studies in humans and rodents have shown that curcumin has very low bioavailability, limiting its benefits. This low bioavailability is likely due to its low absorption and rapid metabolism and rapid elimination in the gut and liver. Due to curcumin's low absorption rate, curcumin is often formulated with other plant compounds to increase absorption and enhance anti-inflammatory effects.
I期臨床試驗已顯示,薑黃素即使在高劑量(12g/天)下在人體中亦為安全的,但顯示差生體可用率。限制薑黃素之生體可用率之潛在因素包括差吸收、快速代謝及快速全身消除。由於烹飪或溶解於油中,食物中攝取之薑黃素之生體可用率可增加。為改善生體可用率,已開始採取許多方法。此等方法涉及首先使用干擾葡萄糖醛酸化之佐劑(諸如胡椒鹼);其次,使用脂質體薑黃素;第三,薑黃素奈米顆粒;第四,使用薑黃素磷脂複合物;且第五,使用薑黃素之結構類似物(例如,EF-24)。據報導後者吸收快速,及血漿半衰期達成峰值。儘管生體可用率較低,但類薑黃素針對各種人類疾病(包括癌症、心血管疾病、糖尿病、關節炎、神經疾病及克羅恩病(Crohn’s disease))之治療性效用已記錄在案。奈米顆粒、脂質體、膠束及磷脂複合物係其他具有前景之新穎調配物,其等似乎提供更長之循環、更好的滲透性及對代謝過程之抗性。因此當前需要增強薑黃素之生體可用率,以自薑黃素用於治療人類疾病之治療性性質獲得更大益處。 Phase I clinical trials have shown that curcumin is safe in humans even at high doses (12g/day), but shows poor bioavailability. Potential factors that limit the bioavailability of curcumin include poor absorption, rapid metabolism, and rapid systemic elimination. The bioavailability of curcumin ingested in food can be increased due to cooking or dissolving in oil. A number of approaches have been initiated to improve bioavailability. These methods involve firstly using an adjuvant (such as piperine) that interferes with glucuronidation; secondly, using liposomal curcumin; thirdly, using curcumin nanoparticles; fourthly, using curcumin phospholipid complexes; and fifthly, using Structural analogs of curcumin (eg, EF-24) are used. The latter is reported to be rapidly absorbed, with a peak plasma half-life. Despite their low bioavailability, curcuminoids have documented therapeutic utility against various human diseases including cancer, cardiovascular disease, diabetes, arthritis, neurological disorders and Crohn's disease. Nanoparticles, liposomes, micelles, and phospholipid complexes are other promising novel formulations that appear to offer longer circulation, better permeability, and resistance to metabolic processes. There is therefore an ongoing need to enhance the bioavailability of curcumin to gain greater benefit from the therapeutic properties of curcumin for the treatment of human diseases.
葉黃素屬於一大類植物色素,通常可見其異構體玉米黃質及痕量之其他胡蘿蔔素(諸如β-胡蘿蔔素)及隱黃質,統稱為類胡蘿蔔素。其在人類組織中之存在完全由於攝入植物來源;其非由動物組織合成。葉黃素存在於範圍廣泛之水果及蔬菜中且賦予其所在植物(諸如玉米)黃色。 葉黃素在綠葉蔬菜(諸如菠菜、羽衣甘藍及芥藍)中之濃度特別高。由於動物食用植物產物,因此葉黃素亦存在於一些動物產物(諸如蛋黃)中。葉黃素及玉米黃質一起存在於許多食物來源中。深綠葉蔬菜係葉黃素及玉米黃質之主要來源,但其等亦以較少量存在於其他顏色之水果及蔬菜(諸如綠菜花、橙辣椒、玉米、豌豆、柿子及及紅橘)中。葉黃素通常與玉米黃質一起自萬壽菊油樹脂分離。在人類血液及組織中發現之20至30種類胡蘿蔔素中,僅葉黃素及玉米黃質存在於晶狀體及視網膜中。葉黃素及玉米黃質集中在視網膜之黃斑或中央區域,且稱為黃斑色素。 Lutein belongs to a large class of plant pigments, usually its isomer zeaxanthin and trace amounts of other carotene (such as β-carotene) and cryptoxanthin, collectively referred to as carotenoids. Its presence in human tissues is solely due to ingestion of plant sources; it is not synthesized by animal tissues. Lutein is found in a wide range of fruits and vegetables and imparts its yellow color to plants such as corn. Lutein is found in particularly high concentrations in green leafy vegetables such as spinach, kale and kale. Lutein is also present in some animal products, such as egg yolks, due to the consumption of plant products by animals. Lutein and zeaxanthin are found together in many food sources. Dark green leafy vegetables are the main source of lutein and zeaxanthin, but they are also present in smaller amounts in other colored fruits and vegetables (such as broccoli, orange peppers, corn, peas, persimmons, and tangerines) . Lutein is usually isolated from marigold oleoresin together with zeaxanthin. Of the 20 to 30 carotenoids found in human blood and tissues, only lutein and zeaxanthin are found in the lens and retina. Lutein and zeaxanthin are concentrated in the macula, or central area of the retina, and are called macular pigments.
各種研究表明,葉黃素可降低老年人發展兩種最常見眼病(即,白內障及黃斑變性)之風險。由於強烈之光曝露及視網膜中之高氧化代謝率,因此眼中之氧化壓力高。葉黃素之抗氧化性質可降低氧化損傷促進此等疾病之程度或可藉由限制氧滲透膜之程度最小化由於氧化壓力導致之損傷。此外,已在個體(individual)之間發現最高血清或膳食葉黃素濃度與較低冠心病或中風發病率之間存在關聯。在兩項流行病學研究中,如由頸動脈內膜厚度量測,具有葉黃素加玉米黃質之最高血清濃度之個體患冠心病之風險顯著降低。葉黃素及玉米黃質在血漿中主要由高密度脂蛋白(HDL)運輸。 Various studies have shown that lutein can reduce the risk of developing two of the most common eye diseases (ie, cataracts and macular degeneration) in older adults. Oxidative stress in the eye is high due to intense light exposure and a high rate of oxidative metabolism in the retina. The antioxidant properties of lutein may reduce the extent to which oxidative damage contributes to these diseases or may minimize damage due to oxidative stress by limiting the extent to which oxygen permeates membranes. Furthermore, an association between the highest serum or dietary lutein concentrations and a lower incidence of coronary heart disease or stroke has been found between individuals. In two epidemiological studies, individuals with the highest serum concentrations of lutein plus zeaxanthin had a significantly lower risk of developing coronary heart disease, as measured by carotid intima thickness. Lutein and zeaxanthin are mainly transported in plasma by high-density lipoprotein (HDL).
因此,如上文討論,薑黃素及葉黃素經證明為抗氧化劑且有助於由於氧化壓力引起之疾病。因此,自薑黃素及類胡蘿蔔素(諸如葉黃素及玉米黃質)製備包含類薑黃素之組合物將係有利的,該組合物將在調節脂質概況中具有潛在有利效應,藉由抑制及/或調節一或多種關鍵發炎介質之產生而具有抗發炎效應。 Thus, as discussed above, curcumin and lutein have been shown to be antioxidants and contribute to diseases due to oxidative stress. Therefore, it would be advantageous to prepare compositions comprising curcuminoids from curcumin and carotenoids, such as lutein and zeaxanthin, which would have potentially beneficial effects in modulating lipid profiles by inhibiting and /or regulate the production of one or more key inflammatory mediators to have an anti-inflammatory effect.
因此,本發明之目的係提供一種有效調節血脂概況且具有 抗發炎效應之治療性組合物。另外,該治療性草藥組合物有效控制由於諸如發炎(包括由於病原感染引起之發炎)、氧化壓力等因素引起之疾病,及諸如糖尿病性視網膜病變及年齡相關性黃斑變性之疾病。 Therefore, the purpose of the present invention is to provide an effective regulation of blood lipid profile and has Therapeutic composition for anti-inflammatory effect. In addition, the therapeutic herbal composition is effective in controlling diseases due to factors such as inflammation (including inflammation due to pathogenic infection), oxidative stress, and diseases such as diabetic retinopathy and age-related macular degeneration.
本發明之目的亦係提供一種治療性草藥組合物,其包含類薑黃素及類胡蘿蔔素(諸如葉黃素及玉米黃質),該組合物在非常低之劑量下顯示其成分之協同效應且同時具有高生體可用率。 It is also an object of the present invention to provide a therapeutic herbal composition comprising curcuminoids and carotenoids such as lutein and zeaxanthin which shows a synergistic effect of its ingredients at very low doses and At the same time, it has high bioavailability.
本發明係關於一種治療性草藥組合物,其包含植物性化合物(即類薑黃素及類胡蘿蔔素(諸如葉黃素))之新穎組合。更特別地,該治療性草藥組合物包含類薑黃素及葉黃素。該治療性草藥組合物有效促進健康,更特別地,有效調節血脂概況且具有抗發炎效應。該抗發炎效應係藉由抑制及/或調節一或多種關鍵發炎介質之產生而產生。另外,該治療性草藥組合物有效保護肝免受任何氧化損傷,包括由過氧化多不飽和脂肪酸引起之損傷,藉此有助於控制由於諸如發炎、氧化壓力等因素引起之疾病,及諸如糖尿病性視網膜病變及年齡相關性黃斑變性之疾病。本發明亦提供一種用於製造此組合物之新穎方法及此組合物於食物、飲料(酒精飲料)中及作為營養品、膳食補充劑及藥物之用途。 The present invention relates to a therapeutic herbal composition comprising a novel combination of botanical compounds, namely curcuminoids and carotenoids such as lutein. More particularly, the therapeutic herbal composition comprises curcuminoids and lutein. The therapeutic herbal composition is effective in promoting health, more particularly in regulating blood lipid profile and has anti-inflammatory effect. The anti-inflammatory effect is produced by inhibiting and/or modulating the production of one or more key inflammatory mediators. In addition, the therapeutic herbal composition effectively protects the liver from any oxidative damage, including damage caused by peroxidized polyunsaturated fatty acids, thereby helping to control diseases due to factors such as inflammation, oxidative stress, and diseases such as diabetes Retinopathy and age-related macular degeneration. The invention also provides a novel process for the manufacture of the composition and the use of the composition in food, beverages (alcoholic beverages) and as nutraceuticals, dietary supplements and pharmaceuticals.
根據本發明之一實施例,提供一種包含植物性化合物類薑黃素及葉黃素之治療性草藥組合物。 According to one embodiment of the present invention, there is provided a therapeutic herbal composition comprising the botanical compounds curcuminoids and lutein.
根據本發明之一實施例,類薑黃素可以類薑黃素粉或類薑黃素濃縮物之形式使用及葉黃素可以葉黃素晶體或葉黃素濃縮物之形式使用。此外,如本文使用之術語「葉黃素」包括葉黃素及玉米黃質之混合物,因為玉米黃質通常連同葉黃素一起存在/分離。 According to one embodiment of the present invention, curcuminoids may be used in the form of curcuminoid powder or curcuminoid concentrate and lutein may be used in the form of lutein crystals or lutein concentrate. Furthermore, the term "lutein" as used herein includes mixtures of lutein and zeaxanthin, since zeaxanthin is usually present/isolated together with lutein.
根據本發明之一實施例,該治療性草藥組合物包含在24至49%之範圍內之類薑黃素粉/濃縮物,在1至14.5%之範圍內之葉黃素晶體/濃縮物及在50至64%之範圍內之賦形劑。此外,該類薑黃素粉/濃縮物可具有80至95%之類薑黃素含量及葉黃素晶體/濃縮物可具有70至80%之葉黃素含量及4.5至9%之玉米黃質含量。 According to an embodiment of the present invention, the therapeutic herbal composition comprises curcumin powder/concentrate in the range of 24 to 49%, lutein crystals/concentrate in the range of 1 to 14.5% and Excipients in the range of 50 to 64%. Furthermore, the curcuminoid powder/concentrate may have a curcuminoid content of 80 to 95% and the lutein crystals/concentrate may have a lutein content of 70 to 80% and a zeaxanthin content of 4.5 to 9% .
根據本發明之一實施例,類薑黃素及葉黃素係以其等純化形式存在於治療性草藥組合物中。 According to one embodiment of the present invention, curcuminoids and lutein are present in the therapeutic herbal composition in their purified form.
根據本發明之一實施例,類薑黃素粉/濃縮物可自植物薑黃獲得/萃取及純化及葉黃素晶體/濃縮物可自萬壽菊植物金盞花(Tagetes erecta)或任何其他合適之植物來源獲得/萃取及純化。 According to one embodiment of the present invention, the curcuminoid powder/concentrate can be obtained/extracted and purified from the plant Curcuma longa and the lutein crystals/concentrate can be obtained from the marigold plant Calendula ( Tagetes erecta ) or any other suitable plant source Obtained/extracted and purified.
根據本發明之另一實施例,該治療性草藥組合物有效下調發炎途徑。該組合物亦減少活性氧物質(ROS)之形成並平衡NADH/NAD(菸鹼醯胺腺嘌呤二核苷酸(NAD)與NADH(NAD之還原形式))比率。此外,本發明之治療性草藥組合物亦有效控制脂肪肝形成。 According to another embodiment of the present invention, the therapeutic herbal composition is effective in down-regulating inflammatory pathways. The composition also reduces the formation of reactive oxygen species (ROS) and balances the NADH/NAD (nicotinamide adenine dinucleotide (NAD) to NADH (reduced form of NAD)) ratio. In addition, the therapeutic herbal composition of the present invention is also effective in controlling fatty liver formation.
根據本發明之另一實施例,該治療性草藥組合物有效調節血脂概況且具有抗發炎效應。體內異常脂質概況及發炎之一些致病因素可為不健康飲食模式、攝入大量酒精、攝入含有大量過氧化多不飽和脂肪酸(PUFA)含量之食物或由於服用某些藥物。體內發炎亦可由病原感染引起,該病原感染由感染物/病原體諸如病毒(包括病毒諸如冠狀病毒病(COVID-19)及其他引起感染之病毒)及細菌引起。該抗發炎效應係藉由抑制及/或調節一或多種關鍵發炎介質之產生而產生。此外,本發明之治療性草藥組合物亦有效保護肝免受由不健康飲食模式引起之非酒精性脂肪肝疾病。 According to another embodiment of the present invention, the therapeutic herbal composition is effective in regulating blood lipid profile and has anti-inflammatory effect. Some contributing factors to abnormal lipid profiles and inflammation in the body can be unhealthy eating patterns, high alcohol intake, intake of foods with high peroxidized polyunsaturated fatty acid (PUFA) content, or due to the use of certain medications. Inflammation in the body can also be caused by pathogenic infections caused by infectious agents/pathogens such as viruses (including viruses such as coronavirus disease (COVID-19) and other infection-causing viruses) and bacteria. The anti-inflammatory effect is produced by inhibiting and/or modulating the production of one or more key inflammatory mediators. In addition, the therapeutic herbal composition of the present invention is also effective in protecting the liver from non-alcoholic fatty liver disease caused by unhealthy dietary patterns.
根據本發明之另一實施例,該治療性草藥組合物有效抑制受感染細胞中氧化應激損傷。 According to another embodiment of the present invention, the therapeutic herbal composition is effective in inhibiting oxidative stress damage in infected cells.
根據本發明之另一實施例,該治療性草藥組合物有助於保護肝免受肝過氧化。通過發明人在活體外及活體內進行之效用研究已建立相同結論。 According to another embodiment of the present invention, the therapeutic herbal composition helps to protect the liver from hepatic peroxidation. The same conclusion has been established through the effect studies conducted by the inventors in vitro and in vivo.
根據本發明之又其他實施例,該治療性草藥組合物有效調節血脂概況。此外,該治療性草藥組合物有效維持健康之膽固醇濃度及健康之低密度脂蛋白與高密度脂蛋白(LDL/HDL)比率。該組合物亦有效降低升高之血清甘油三酯。由於該治療性草藥組合物有效調節血脂概況,因此其亦有效控制諸如糖尿病性視網膜病變及年齡相關性黃斑變性之疾病,因為該治療性草藥組合物導致黃斑色素光學密度(MPOD)快速增加。 According to yet other embodiments of the present invention, the therapeutic herbal composition is effective in modulating blood lipid profile. In addition, the therapeutic herbal composition is effective in maintaining healthy cholesterol concentrations and healthy low-density lipoprotein to high-density lipoprotein (LDL/HDL) ratios. The composition is also effective in lowering elevated serum triglycerides. Since the therapeutic herbal composition is effective in modulating the blood lipid profile, it is also effective in controlling diseases such as diabetic retinopathy and age-related macular degeneration because the therapeutic herbal composition results in a rapid increase in macular pigment optical density (MPOD).
根據本發明之另一實施例,該治療性草藥組合物有效增加受感染細胞中非酵素抗氧化劑麩胱甘肽還原酶(GSH)及超氧化物歧化酶(SOD)之濃度。 According to another embodiment of the present invention, the therapeutic herbal composition is effective to increase the concentration of non-enzymatic antioxidants glutathione reductase (GSH) and superoxide dismutase (SOD) in infected cells.
根據本發明之另一實施例,如活體外研究顯示,該治療性草藥組合物有效減少受感染細胞中硫巴比妥酸反應性物質(TBARS)丙二醛(MDA)之產生,其中發現該治療性草藥組合物有效抵抗肝過氧化。 According to another embodiment of the present invention, the therapeutic herbal composition is effective in reducing the production of thiobarbituric acid reactive substances (TBARS) malondialdehyde (MDA) in infected cells as shown in in vitro studies, wherein the Therapeutic herbal composition is effective against hepatic peroxidation.
在本發明之一實施例中,如活體外研究,本發明之治療性草藥組合物有效防止受感染細胞中細胞核凋亡及細胞核碎裂。 In one embodiment of the present invention, the therapeutic herbal composition of the present invention is effective in preventing nuclear apoptosis and nuclear fragmentation in infected cells, as in in vitro studies.
在本發明之一實施例中,本發明之治療性草藥組合物有效降低促發炎細胞介素(TNF-α、IL-6及IL-1)濃度,此有利於減少可由不健康飲食模式、攝入大量酒精、攝入含有大量過氧化多不飽和脂肪酸(PUFA)含量之食物或由於服用某些藥物、由於病原感染,及由於酒精性 及非酒精性脂肪肝疾病引起之肝發炎反應。 In one embodiment of the present invention, the therapeutic herbal composition of the present invention effectively reduces the concentration of pro-inflammatory cytokines (TNF-α, IL-6 and IL-1), which is beneficial to reduce the A large amount of alcohol, intake of food containing a large amount of peroxidized polyunsaturated fatty acids (PUFA) or due to taking certain drugs, due to pathogenic infection, and due to alcoholic And liver inflammation caused by non-alcoholic fatty liver disease.
在本發明之一實施例中,本發明之治療性草藥組合物顯示降脂活性,因此其亦有效抵抗可由於飲酒、藥物及不健康飲食模式引起之肝毒性。 In one embodiment of the present invention, the therapeutic herbal composition of the present invention exhibits lipid-lowering activity, thus it is also effective against liver toxicity that may be caused by alcohol consumption, drugs and unhealthy dietary patterns.
根據本發明之另一實施例,該治療性草藥組合物可作為補充劑定期或在攝入高脂肪食物、藥物及/或酒精之前、期間或之後服用/攝取。 According to another embodiment of the present invention, the therapeutic herbal composition may be taken/ingested as a supplement on a regular basis or before, during or after intake of fatty foods, drugs and/or alcohol.
根據本發明之另一實施例,提供一種用於製造包含植物性化合物類薑黃素及葉黃素之治療性草藥組合物之方法,其中該方法包括:a)使用類薑黃素含量在80至95%之範圍內之類薑黃素粉/濃縮物及葉黃素含量在70至80%之範圍內及玉米黃質含量4.5至9%之葉黃素晶體/濃縮物並以預定量混合該類薑黃素粉/濃縮物及葉黃素晶體/濃縮物以形成摻合物;未處理乾燥摻合物之粒度可落於D50-20μm至50μm及D90-100μm至200μm之範圍內;b)將賦形劑諸如合適之乳化劑及麥芽糊精添加至該摻合物;c)將純化水添加至該摻合物以製造懸浮液;d)使該懸浮液通過液體膠體磨機以形成均勻懸浮液;e)使該懸浮液經受進一步處理諸如通過均質機或高剪切顆粒濕磨機以產生微粉化乳液;f)以25rpm之速度將該微粉化乳液進一步攪動8至12小時以使質量溫度升至25℃至40℃,較佳25℃至30℃之溫度;g)使該微粉化乳液經受進一步處理諸如濃縮及/或乾燥以獲得該治療性草藥組合物。 According to another embodiment of the present invention, there is provided a method for the manufacture of a therapeutic herbal composition comprising the botanical compounds curcuminoids and lutein, wherein the method comprises: a) using a curcuminoid content of 80 to 95% Curcuminoid powder/concentrate in the range and lutein crystals/concentrate with lutein content in the range of 70 to 80% and zeaxanthin content in the range of 4.5 to 9% and mix the curcuminoid powder in a predetermined amount /concentrate and lutein crystals/concentrate to form a blend; the particle size of the untreated dry blend may fall within the range of D 50 -20 μm to 50 μm and D 90 -100 μm to 200 μm; b) excipient Add agents such as suitable emulsifiers and maltodextrin to the blend; c) add purified water to the blend to make a suspension; d) pass the suspension through a liquid colloid mill to form a homogeneous suspension e) subjecting the suspension to further processing such as through a homogenizer or a high shear particle wet mill to produce a micronized emulsion; f) further agitating the micronized emulsion at a speed of 25 rpm for 8 to 12 hours to raise the temperature of the mass to a temperature of 25°C to 40°C, preferably 25°C to 30°C; g) subjecting the micronized emulsion to further processing such as concentration and/or drying to obtain the therapeutic herbal composition.
該方法進一步包括使經乾燥之治療性草藥組合物通過合適之顆粒篩以獲得均勻成品。此經處理治療性草藥組合物之粒度可落於D50-0.36μm至5μm之範圍內及D90在0.60μm至10μm之範圍內。此粒度減小有利於該治療性草藥組合物具有高生體可用率。 The method further comprises passing the dried therapeutic herbal composition through a suitable particle sieve to obtain a homogeneous finished product. The particle size of the treated therapeutic herbal composition may fall within the range of D 50 -0.36 μm to 5 μm and D 90 in the range of 0.60 μm to 10 μm. This particle size reduction facilitates high bioavailability of the therapeutic herbal composition.
根據本發明之一實施例,類薑黃素粉/濃縮物及葉黃素晶體/濃縮物之預定量可為使得該類薑黃素粉/濃縮物在24至49%之範圍內,葉黃素晶體/濃縮物在1至14.5%之範圍內及賦形劑在50至64%之範圍內。 According to one embodiment of the present invention, the predetermined amount of curcuminoid powder/concentrate and lutein crystals/concentrate can be such that the curcuminoid powder/concentrate is in the range of 24 to 49%, lutein crystals /Concentrate in the range of 1 to 14.5% and excipient in the range of 50 to 64%.
根據本發明之又其他實施例,最終經處理治療性草藥組合物包含在22.5%至46.5%之範圍內之類薑黃素、在0.90%至11.25%之範圍內之葉黃素及在0.06%至1.12%之範圍內之玉米黃質。顯而易見,最終組合物中玉米黃質之量構成該最終組合物中葉黃素總含量之大約6至9%。 According to still other embodiments of the present invention, the final processed therapeutic herbal composition comprises curcuminoids in the range of 22.5% to 46.5%, lutein in the range of 0.90% to 11.25%, and lutein in the range of 0.06% to 1.12% zeaxanthin. It is apparent that the amount of zeaxanthin in the final composition constitutes approximately 6 to 9% of the total content of lutein in the final composition.
根據本發明之一實施例,使用之賦形劑係麥芽糊精及乳化劑。 According to one embodiment of the present invention, the excipients used are maltodextrin and emulsifier.
根據本發明之另一實施例,該治療性草藥組合物可添加至食物及/或飲料產品或調配成膳食補充劑。 According to another embodiment of the present invention, the therapeutic herbal composition can be added to food and/or beverage products or formulated as a dietary supplement.
根據本發明之另一實施例,該治療性草藥組合物可用作膳食/營養補充劑或用作各種食物及飲料產品中之治療性/健康成分。其中該治療性草藥組合物可用作健康成分之食物及飲料之一些實例包括(但不限於)茶、輸液、果汁、飲料、牛奶及牛奶製品、穀類產品、酒精飲料及加工食物等。 According to another embodiment of the present invention, the therapeutic herbal composition can be used as a dietary/nutritional supplement or as a therapeutic/health ingredient in various food and beverage products. Some examples of foods and beverages in which the therapeutic herbal composition can be used as a health ingredient include, but are not limited to, teas, infusions, fruit juices, beverages, milk and milk products, cereal products, alcoholic beverages, processed foods, and the like.
根據本發明之另一實施例,該治療性草藥組合物亦可調配成選自由以下組成之群之合適劑型:粉末、糊劑、錠劑、糖漿及/或膠囊等。 According to another embodiment of the present invention, the therapeutic herbal composition can also be formulated into a suitable dosage form selected from the group consisting of powder, paste, lozenge, syrup and/or capsule, etc.
根據本發明之另一實施例,植物性化合物類薑黃素及葉黃素在該治療性草藥組合物中顯示協同活性。此外,當相較於較高劑量之未調配葉黃素及未調配類薑黃素之生體可用率時,該治療性草藥組合物中存在之類薑黃素及葉黃素係高度生體可用。此外,該等類薑黃素及葉黃素係以遠低於建議每日需求量之量存在。 According to another embodiment of the present invention, the botanical compounds curcuminoids and lutein exhibit synergistic activity in the therapeutic herbal composition. Furthermore, the curcuminoids and luteinoids present in the therapeutic herbal composition are highly bioavailable when compared to the bioavailability of higher doses of unformulated lutein and unformulated curcuminoids. Furthermore, these curcuminoids and lutein are present in amounts well below the recommended daily requirement.
在完整說明書中更清楚描述本發明之上文及其他特徵及態樣。 The above and other features and aspects of the invention are more clearly described in the full specification.
本發明可根據下圖描述,其中:圖1(a)闡述不同濃度之未處理類薑黃素(U1)在人類肝癌(HepG2)細胞中之細胞毒性效應。 The present invention can be described according to the following figure, wherein: Figure 1(a) illustrates the cytotoxic effect of different concentrations of untreated curcuminoid (U1) in human liver cancer (HepG2) cells.
圖1(b)闡述不同濃度之未處理葉黃素(U2)在人類肝癌(HepG2)細胞中之細胞毒性效應。 Figure 1(b) illustrates the cytotoxic effect of different concentrations of untreated lutein (U2) in human liver cancer (HepG2) cells.
圖1(c)闡述不同濃度之未處理治療性草藥組合物(U3)在人類肝癌(HepG2)細胞中之細胞毒性效應。 Figure 1(c) illustrates the cytotoxic effect of different concentrations of untreated therapeutic herbal composition (U3) in human liver cancer (HepG2) cells.
圖1(d)闡述不同濃度之經處理類薑黃素(P1)在人類肝癌(HepG2)細胞中之細胞毒性效應。 Figure 1(d) illustrates the cytotoxic effect of different concentrations of processed curcuminoids (P1) in human liver cancer (HepG2) cells.
圖1(e)闡述不同濃度之經處理葉黃素(P2)在HepG2細胞中之細胞毒性效應。 Figure 1(e) illustrates the cytotoxic effect of different concentrations of processed lutein (P2) in HepG2 cells.
圖1(f)闡述不同濃度之經處理治療性草藥組合物(P3)在HepG2細胞中之細胞毒性效應。 Figure 1(f) illustrates the cytotoxic effect of different concentrations of the treated therapeutic herbal composition (P3) in HepG2 cells.
圖2(a)闡述不同濃度之未處理類薑黃素(U1)在乙醇誘導之HepG2細胞中之細胞毒性效應。 Figure 2(a) illustrates the cytotoxic effect of different concentrations of untreated curcuminoid (U1) in ethanol-induced HepG2 cells.
圖2(b)闡述不同濃度之未處理葉黃素(U2)在乙醇誘導之HepG2細胞中之細胞毒性效應。 Figure 2(b) illustrates the cytotoxic effect of different concentrations of untreated lutein (U2) in ethanol-induced HepG2 cells.
圖2(c)闡述不同濃度之未處理治療性草藥組合物(U3)在乙醇誘導之HepG2細胞中之細胞毒性效應。 Figure 2(c) illustrates the cytotoxic effect of different concentrations of untreated therapeutic herbal composition (U3) in ethanol-induced HepG2 cells.
圖2(d)闡述不同濃度之經處理類薑黃素(P1)在乙醇誘導之HepG2細胞中之細胞毒性效應。 Figure 2(d) illustrates the cytotoxic effect of different concentrations of processed curcuminoids (P1) in ethanol-induced HepG2 cells.
圖2(e)闡述不同濃度之經處理葉黃素(P2)在乙醇誘導之HepG2細胞中之細胞毒性效應。 Figure 2(e) illustrates the cytotoxic effect of different concentrations of processed lutein (P2) in ethanol-induced HepG2 cells.
圖2(f)闡述不同濃度之經處理治療性草藥組合物(P3)在乙醇誘導之HepG2細胞中之細胞毒性效應。 Figure 2(f) illustrates the cytotoxic effect of different concentrations of the treated therapeutic herbal composition (P3) in ethanol-induced HepG2 cells.
圖3(a)闡述未處理及經處理治療性草藥組合物對乙醇誘導之人類肝癌(HepG2)細胞中硫巴比妥酸反應性物質(TBARS)活性之治療效應。 Figure 3(a) illustrates the therapeutic effect of untreated and treated therapeutic herbal compositions on the activity of thiobarbituric acid-responsive substances (TBARS) in ethanol-induced human liver cancer (HepG2) cells.
圖3(b)闡述未處理及經處理治療性草藥組合物對乙醇誘導之人類肝癌(HepG2)細胞中超氧化物歧化酶(SOD)活性之治療效應。 Figure 3(b) illustrates the therapeutic effect of untreated and treated therapeutic herbal compositions on superoxide dismutase (SOD) activity in ethanol-induced human liver cancer (HepG2) cells.
圖3(c)闡述未處理及經處理治療性草藥組合物對乙醇誘導之HepG2細胞中麩胱甘肽還原酶(GSH)濃度之治療效應。 Figure 3(c) illustrates the therapeutic effect of untreated and treated therapeutic herbal compositions on the concentration of glutathione reductase (GSH) in ethanol-induced HepG2 cells.
圖4(a)係顯示經處理治療性草藥組合物在經對乙醯胺基酚(APAP)處理之HepG2細胞中之細胞毒性效應的條形圖。 Figure 4(a) is a bar graph showing the cytotoxic effect of treated therapeutic herbal compositions in acetaminophen (APAP)-treated HepG2 cells.
圖4(b)係顯示水飛薊素在經APAP處理之HepG2細胞中之細胞毒性效應的條形圖。 Figure 4(b) is a bar graph showing the cytotoxic effect of silymarin in APAP-treated HepG2 cells.
圖5(a)闡述經處理治療性草藥組合物相比於水飛薊素對經APAP處理之HepG2細胞中TBARS活性之治療效應之比較。 Figure 5(a) illustrates a comparison of the therapeutic effects of treated therapeutic herbal compositions compared to silymarin on TBARS activity in APAP-treated HepG2 cells.
圖5(b)闡述經處理治療性草藥組合物相比於水飛薊素對經APAP處理之HepG2細胞中SOD活性之治療效應之比較。 Figure 5(b) illustrates a comparison of the therapeutic effects of treated therapeutic herbal compositions compared to silymarin on SOD activity in APAP-treated HepG2 cells.
圖5(c)闡述經處理治療性草藥組合物相比於水飛薊素對經APAP處理之HepG2細胞中GSH濃度之治療效應之比較。 Figure 5(c) illustrates a comparison of the therapeutic effects of treated therapeutic herbal compositions compared to silymarin on GSH concentration in APAP-treated HepG2 cells.
圖6闡述未處理及經處理治療性草藥組合物相比於水飛薊素對HepG2細胞中乙醇誘導之細胞內活性氧物質(ROS)產生之影響之比較。 Figure 6 illustrates a comparison of the effect of untreated and treated therapeutic herbal compositions compared to silymarin on ethanol-induced intracellular reactive oxygen species (ROS) production in HepG2 cells.
圖7闡述未處理及經處理治療性草藥組合物相比於水飛薊素對HepG2細胞中乙醇介導之粒線體膜電位(MMP)降低之影響之比較。 Figure 7 illustrates a comparison of the effect of untreated and treated therapeutic herbal compositions compared to silymarin on ethanol-mediated reduction of mitochondrial membrane potential (MMP) in HepG2 cells.
圖8闡述未處理及經處理治療性草藥組合物相比於水飛薊素對HepG2細胞中乙醇誘導之細胞核凋亡之影響之比較。 Figure 8 illustrates a comparison of the effect of untreated and treated therapeutic herbal compositions compared to silymarin on ethanol-induced nuclear apoptosis in HepG2 cells.
圖9闡述未處理及經處理治療性草藥組合物相比於水飛薊素對HepG2細胞中乙醇誘導之細胞核碎裂之影響之比較。 Figure 9 illustrates a comparison of the effect of untreated and treated therapeutic herbal compositions compared to silymarin on ethanol-induced nuclear fragmentation in HepG2 cells.
圖10闡述經處理治療性草藥組合物相比於水飛薊素對HepG2細胞中APAP誘導之細胞內ROS產生之影響之比較。 Figure 10 illustrates a comparison of the effect of treated therapeutic herbal compositions compared to silymarin on APAP-induced intracellular ROS production in HepG2 cells.
圖11闡述經處理治療性草藥組合物相比於水飛薊素對HepG2細胞中APAP介導之MMP減少之影響之比較。 Figure 11 illustrates a comparison of the effect of treated therapeutic herbal compositions compared to silymarin on APAP-mediated MMP reduction in HepG2 cells.
圖12闡述經處理治療性草藥組合物相比於水飛薊素對HepG2細胞中APAP誘導之細胞核凋亡之影響之比較。 Figure 12 illustrates a comparison of the effect of treated therapeutic herbal compositions compared to silymarin on APAP-induced nuclear apoptosis in HepG2 cells.
圖13闡述經處理治療性草藥組合物相比於水飛薊素對HepG2細胞中APAP誘導之細胞核碎裂之影響之比較。 Figure 13 illustrates a comparison of the effect of treated therapeutic herbal compositions compared to silymarin on APAP-induced nuclear fragmentation in HepG2 cells.
圖14闡述在研究之第28天,經處理治療性草藥組合物相比於水飛薊素對大鼠肝中乙醇誘導之組織病理學變化之效應。 Figure 14 illustrates the effect of treated therapeutic herbal composition compared to silymarin on alcohol-induced histopathological changes in rat liver at day 28 of the study.
圖15顯示類薑黃素之平均血漿濃度-時間曲線。 Figure 15 shows mean plasma concentration-time profiles of curcuminoids.
圖16顯示葉黃素之平均血漿濃度-時間曲線。 Figure 16 shows the mean plasma concentration-time profile of lutein.
下文討論本發明之一些代表性實施例。本發明在其更廣泛之態樣中不限於特定細節及代表性方法。說明性實例結合本文提供之實施例及方法一起描述於此章節中。根據本說明書閱讀之隨附申請專利範圍中特別指出並清楚主張根據其各種態樣之本發明。 Some representative embodiments of the invention are discussed below. The invention in its broader aspects is not limited to the specific details and representative methods. Illustrative examples are described in this section in conjunction with the embodiments and methods provided herein. The invention in its various aspects is particularly pointed out and distinctly claimed in the appended claims to be read in light of this specification.
應注意,如本說明書及隨附申請專利範圍中使用,除非內文另有明確規定,否則單數形式「一」、「一個」及「該」包括複數個參考物。因此,例如,包含「一種化合物」之組合物之提及包括兩種或更多種化合物之混合物。亦應注意,除非內文另有明確規定,否則術語「或」一般以其包括「及/或」之意義採用。 It should be noted that, as used in this specification and the appended claims, the singular forms "a", "an" and "the" include plural references unless the context clearly dictates otherwise. Thus, for example, reference to a composition comprising "a compound" includes a mixture of two or more compounds. It should also be noted that the term "or" is generally employed in its sense including "and/or" unless the context clearly requires otherwise.
除非另有規定,否則以「%」表示之各種量意謂總溶液或組合物之重量百分比。 All amounts expressed in "%" mean percent by weight of the total solution or composition unless otherwise specified.
如本文使用之術語「葉黃素」包括葉黃素及玉米黃質之混合物,因為玉米黃質通常連同葉黃素一起存在/分離。 The term "lutein" as used herein includes mixtures of lutein and zeaxanthin, since zeaxanthin is usually present/isolated together with lutein.
所有引用之參考文獻均係以全文引用之方式併入本文中。任何參考文獻之引用並非關於對其作為本發明先前技術之可用性之任何確定之承認。 All references cited are hereby incorporated by reference in their entirety. Citation of any reference is not an admission with any certainty as to its applicability as prior art to the present invention.
除非另有明確定義,否則本文使用之技術及科學術語具有熟習本發明所屬領域之技術者通常瞭解之含義。本文對熟習此項技術者已知的各種方法論及材料作出參考。 Unless clearly defined otherwise, technical and scientific terms used herein have the meaning commonly understood by those skilled in the art to which this invention belongs. Reference is made herein to various methodologies and materials known to those skilled in the art.
熟習此項技術者已知的任何合適之材料及/或方法可用以進 行本發明。然而,本文描述較佳材料及方法。除非另有提及,否則以下說明書及實例中作出參考之材料、試劑及類似物可自商業來源獲得。 Any suitable materials and/or methods known to those skilled in the art may be used to further carry out the invention. However, preferred materials and methods are described herein. Materials, reagents and the like to which reference is made in the following specification and examples are available from commercial sources unless otherwise mentioned.
本發明在其產品及方法態樣中係經如下詳細描述。 The present invention is described in detail as follows in its product and method aspects.
本發明係關於一種治療性組合物,其包含植物性化合物(即類薑黃素及類胡蘿蔔素(諸如葉黃素))之新穎組合。更特別地,該治療性草藥組合物包含類薑黃素及葉黃素。該治療性草藥組合物有效調節血脂概況及調節發炎效應。發炎效應或抗發炎效應之此調節係藉由抑制及/或調節一或多種關鍵發炎介質之產生而產生。另外,該治療性草藥組合物有效保護肝免受任何氧化損傷,包括由過氧化多不飽和脂肪酸引起之損傷,藉此有助於控制由於諸如發炎、氧化壓力等因素引起之疾病,及諸如糖尿病性視網膜病變及年齡相關性黃斑變性之疾病。本發明亦提供一種用於製造此組合物之新穎方法及此組合物於食物、飲料(包括酒精飲料)及營養品中及/或作為膳食補充劑之用途。 The present invention relates to a therapeutic composition comprising a novel combination of botanical compounds, namely curcuminoids and carotenoids such as lutein. More particularly, the therapeutic herbal composition comprises curcuminoids and lutein. The therapeutic herbal composition effectively regulates blood lipid profile and modulates inflammatory effects. Such modulation of inflammatory or anti-inflammatory effects occurs by inhibiting and/or modulating the production of one or more key inflammatory mediators. In addition, the therapeutic herbal composition effectively protects the liver from any oxidative damage, including damage caused by peroxidized polyunsaturated fatty acids, thereby helping to control diseases due to factors such as inflammation, oxidative stress, and diseases such as diabetes Retinopathy and age-related macular degeneration. The invention also provides a novel process for the manufacture of the composition and the use of the composition in food, beverages (including alcoholic beverages) and nutritional products and/or as a dietary supplement.
根據本發明之一實施例,該治療性草藥組合物包含類薑黃素及葉黃素。 According to one embodiment of the present invention, the therapeutic herbal composition comprises curcuminoids and lutein.
根據本發明之一實施例,類薑黃素可以類薑黃素粉或類薑黃素濃縮物之形式使用及葉黃素可以葉黃素晶體或葉黃素濃縮物之形式使用。 According to one embodiment of the present invention, curcuminoids may be used in the form of curcuminoid powder or curcuminoid concentrate and lutein may be used in the form of lutein crystals or lutein concentrate.
根據本發明之一實施例,該治療性草藥組合物包含在24至49%之範圍內之類薑黃素粉/濃縮物、在1至14.5%之範圍內之葉黃素晶體/濃縮物及在50至64%之範圍內之賦形劑。此外,該等類薑黃素可具有80至95%之類薑黃素含量及葉黃素晶體/濃縮物可具有70至80%之葉黃素含量及4.5至9%之玉米黃質含量。 According to an embodiment of the present invention, the therapeutic herbal composition comprises curcumin powder/concentrate in the range of 24 to 49%, lutein crystals/concentrate in the range of 1 to 14.5%, and Excipients in the range of 50 to 64%. Furthermore, the curcuminoids may have a curcuminoid content of 80 to 95% and the lutein crystals/concentrate may have a lutein content of 70 to 80% and a zeaxanthin content of 4.5 to 9%.
根據本發明之一實施例,類薑黃素及葉黃素係以其等經純化形式存在於該治療性草藥組合物中。 According to one embodiment of the present invention, curcuminoids and lutein are present in the therapeutic herbal composition in their purified form.
根據本發明之一實施例,類薑黃素粉/濃縮物可自植物薑黃獲得/萃取及純化及葉黃素晶體/濃縮物可自萬壽菊植物金盞花或任何其他合適之植物來源獲得/萃取及純化。 According to one embodiment of the present invention, the curcuminoid powder/concentrate can be obtained/extracted and purified from the plant Curcuma longa and the lutein crystals/concentrate can be obtained/extracted from the marigold plant Calendula officinalis or any other suitable plant source and purification.
根據本發明之另一實施例,該治療性草藥組合物有效下調發炎途徑及亦有效調節血脂概況。體內異常脂質概況及發炎之一些致病因素可能為不健康飲食模式、攝入大量酒精、服用某些藥物或服用飲食中含有大量過氧化多不飽和脂肪酸(PUFA)之食物。體內發炎亦可由病原感染引起,該病原感染由感染物/病原體諸如病毒(包括病毒諸如冠狀病毒病(COVID-19)及其他引起感染之病毒)及細菌引起。該抗發炎效應係藉由抑制及/或調節一或多種關鍵發炎介質之產生而產生。此外,本發明之治療性草藥組合物亦有效保護肝免受由不健康飲食模式引起之非酒精性脂肪肝疾病。 According to another embodiment of the present invention, the therapeutic herbal composition is effective in down-regulating inflammatory pathways and is also effective in modulating blood lipid profile. Some contributing factors to abnormal lipid profiles and inflammation in the body may be unhealthy eating patterns, high alcohol consumption, certain medications, or foods high in peroxidized polyunsaturated fatty acids (PUFAs) in the diet. Inflammation in the body can also be caused by pathogenic infections caused by infectious agents/pathogens such as viruses (including viruses such as coronavirus disease (COVID-19) and other infection-causing viruses) and bacteria. The anti-inflammatory effect is produced by inhibiting and/or modulating the production of one or more key inflammatory mediators. In addition, the therapeutic herbal composition of the present invention is also effective in protecting the liver from non-alcoholic fatty liver disease caused by unhealthy dietary patterns.
根據本發明之另一實施例,該治療性草藥組合物對細胞中氧化應激損傷具有抑制作用。發明人已研究,該治療性草藥組合物有效抵抗肝細胞中之氧化應激損傷及肝過氧化,藉此指示該治療性草藥組合物抑制受感染細胞中氧化應激損傷,該損傷可由各種因素諸如酒精攝入、藥物攝入或不健康飲食模式(包括在飲食中攝入大量過氧化多不飽和脂肪酸PUFA等)引起。 According to another embodiment of the present invention, the therapeutic herbal composition has an inhibitory effect on oxidative stress damage in cells. The inventors have studied that the therapeutic herbal composition is effective against oxidative stress damage and hepatic peroxidation in liver cells, thereby indicating that the therapeutic herbal composition inhibits oxidative stress damage in infected cells, which can be caused by various factors Such as alcohol intake, drug intake, or unhealthy dietary patterns (including high intake of peroxidized polyunsaturated fatty acids PUFA in the diet, etc.).
根據本發明之另一實施例,如活體外研究中顯示,該治療性草藥組合物有效減少受感染細胞中硫巴比妥酸反應性物質(TBARS)丙二醛(MDA)之產生,其中發現該治療性草藥組合物有效抵抗肝過氧化。 According to another embodiment of the present invention, the therapeutic herbal composition is effective in reducing the production of thiobarbituric acid reactive substances (TBARS) malondialdehyde (MDA) in infected cells as shown in in vitro studies, wherein it was found that The therapeutic herbal composition is effective against hepatic peroxidation.
根據本發明之另一實施例,該治療性草藥組合物有效增加受感染細胞中非酵素抗氧化劑麩胱甘肽還原酶(GSH)及超氧化物歧化酶(SOD)之濃度。該等非酵素抗氧化劑之減少可由各種因素諸如酒精攝入、藥物攝入或不健康飲食模式(包括在飲食中攝入大量過氧化多不飽和脂肪酸PUFA等)引起。 According to another embodiment of the present invention, the therapeutic herbal composition is effective to increase the concentration of non-enzymatic antioxidants glutathione reductase (GSH) and superoxide dismutase (SOD) in infected cells. The reduction of these non-enzyme antioxidants can be caused by various factors such as alcohol intake, drug intake or unhealthy dietary pattern (including intake of large amounts of peroxidized polyunsaturated fatty acids PUFA in the diet, etc.).
根據本發明之另一實施例,該治療性草藥組合物顯示抗發炎活性,因為該治療性草藥組合物有效減弱ROS之形成、平衡NADH/NAD比率及下調發炎途徑。本發明之治療性草藥組合物亦有效控制脂肪肝形成。 According to another embodiment of the present invention, the therapeutic herbal composition exhibits anti-inflammatory activity because the therapeutic herbal composition effectively attenuates ROS formation, balances NADH/NAD ratio and downregulates inflammatory pathways. The therapeutic herbal composition of the present invention is also effective in controlling fatty liver formation.
根據本發明之又其他實施例,本發明之治療性草藥組合物有效防止受感染細胞中活性氧物質(ROS)產生,藉此後續減弱受感染細胞中發炎之進一步退化途徑。ROS產生增加可由各種因素諸如酒精攝入、藥物攝入或不健康飲食模式(包括在飲食中攝入大量過氧化多不飽和脂肪酸PUFA等)引起。該治療性草藥組合物亦有效防止受感染細胞中細胞粒線體膜去極化。 According to yet other embodiments of the present invention, the therapeutic herbal composition of the present invention is effective in preventing reactive oxygen species (ROS) production in infected cells, thereby subsequently attenuating further degenerative pathways of inflammation in infected cells. Increased ROS production can be caused by various factors such as alcohol intake, drug intake, or unhealthy dietary patterns (including high intake of peroxidized polyunsaturated fatty acids PUFA in the diet, etc.). The therapeutic herbal composition is also effective in preventing depolarization of mitochondrial membranes in infected cells.
在本發明之一實施例中,如由活體外研究建立,本發明之治療性草藥組合物藉由防止受感染細胞中細胞核凋亡及細胞核碎裂而有效抵抗由諸如酒精、藥物及不健康飲食模式之因素引起之肝毒性。 In one embodiment of the present invention, as established from in vitro studies, the therapeutic herbal composition of the present invention is effective against conditions such as alcohol, drugs and unhealthy dietary patterns by preventing nuclear apoptosis and nuclear fragmentation in infected cells Hepatotoxicity caused by other factors.
在本發明之又其他實施例中,如由臨床前研究建立,本發明之治療性草藥組合物有效抑制血清酵素,即丙胺酸胺基轉移酶(ALT)、天冬胺酸胺基轉移酶(AST)、酒精脫氫酶(ADH)及γ-麩胺醯轉移酶(GGT)之濃度增加。 In yet other embodiments of the present invention, as established from preclinical studies, the therapeutic herbal composition of the present invention effectively inhibits serum enzymes, namely alanine aminotransferase (ALT), aspartate aminotransferase ( AST), alcohol dehydrogenase (ADH) and γ-glutamine transferase (GGT) concentrations increased.
根據本發明之另一實施例,該治療性草藥組合物有助於保 護肝免受肝過氧化。通過發明人進行之活體外及活體內效用研究已建立相同結論。 According to another embodiment of the present invention, the therapeutic herbal composition helps to maintain Protects the liver from liver peroxidation. The same conclusion has been established through the in vitro and in vivo effect studies conducted by the inventors.
在本發明之一實施例中,本發明之治療性草藥組合物有效降低促發炎細胞介素(TNF-α、IL-6及IL-1)濃度,此有利於減少可由不健康飲食模式、攝入大量酒精、攝入含有大量過氧化多不飽和脂肪酸(PUFA)含量之食物或由於服用某些藥物、由於病原感染,及由於酒精性及非酒精性脂肪肝疾病引起之肝發炎反應。 In one embodiment of the present invention, the therapeutic herbal composition of the present invention effectively reduces the concentration of pro-inflammatory cytokines (TNF-α, IL-6 and IL-1), which is beneficial to reduce the Hepatic inflammation caused by a large amount of alcohol, intake of food containing a large amount of peroxidized polyunsaturated fatty acid (PUFA) or certain drugs, pathogenic infection, and alcoholic and non-alcoholic fatty liver disease.
根據本發明之又其他實施例,該治療性草藥組合物有效正常化並維持脂質概況,藉此調節脂質概況。此外,該治療性草藥組合物有效維持健康之膽固醇濃度及健康之低密度脂蛋白與高密度脂蛋白(LDL/HDL)比率。該組合物亦有效降低升高之血清甘油三酯。此外,由於該治療性草藥組合物有效降低升高之血清甘油三酯並正常化脂質概況,藉此其亦有效控制諸如糖尿病性視網膜病變及年齡相關性黃斑變性疾病,因為該治療性草藥組合物導致黃斑色素光學密度(MPOD)快速增加。 According to still other embodiments of the present invention, the therapeutic herbal composition is effective to normalize and maintain lipid profile, thereby modulating lipid profile. In addition, the therapeutic herbal composition is effective in maintaining healthy cholesterol concentrations and healthy low-density lipoprotein to high-density lipoprotein (LDL/HDL) ratios. The composition is also effective in lowering elevated serum triglycerides. In addition, since the therapeutic herbal composition is effective in reducing elevated serum triglycerides and normalizing lipid profile, thereby it is also effective in controlling diseases such as diabetic retinopathy and age-related macular degeneration, because the therapeutic herbal composition Causes a rapid increase in macular pigment optical density (MPOD).
根據本發明之又其他實施例,由於該治療性草藥組合物顯示降脂活性,因此其亦有效抵抗可由於飲酒、藥物及不健康飲食模式引起之肝毒性。 According to yet other embodiments of the present invention, since the therapeutic herbal composition exhibits lipid-lowering activity, it is also effective against liver toxicity that may be caused by alcohol consumption, drugs, and unhealthy dietary patterns.
根據本發明之另一實施例,類薑黃素及葉黃素獲自天然來源,已知其等無副作用。 According to another embodiment of the present invention, curcuminoids and lutein are obtained from natural sources, which are known to have no side effects.
根據本發明之另一實施例,植物性化合物類薑黃素及葉黃素在治療性草藥組合物中顯示協同活性。此外,當相較於較高劑量之未調配葉黃素及未調配類薑黃素之生體可用率時,存在於該治療性草藥組合物中之類薑黃素及葉黃素係高度生體可用。此外,該等類薑黃素及葉黃素係 以遠低於建議每日需求量之量存在。 According to another embodiment of the present invention, the botanical compounds curcuminoids and lutein exhibit synergistic activity in a therapeutic herbal composition. Furthermore, the curcuminoids and luteinoids present in the therapeutic herbal composition are highly bioavailable when compared to the bioavailability of higher doses of unformulated lutein and unformulated curcuminoids . In addition, these curcuminoids and lutein Present in amounts well below the recommended daily requirement.
根據本發明之另一實施例,該治療性草藥組合物可作為補充劑定期或在攝入高脂肪食物、藥物及/或酒精之前、期間或之後服用/攝取。 According to another embodiment of the present invention, the therapeutic herbal composition may be taken/ingested as a supplement on a regular basis or before, during or after intake of fatty foods, drugs and/or alcohol.
本發明亦提供一種用於製造包含類薑黃素及葉黃素之治療性草藥組合物之方法,其中該方法包括下列步驟:a)使用類薑黃素含量在80至95%之範圍內之類薑黃素粉/濃縮物及葉黃素含量在70至80%之範圍內及玉米黃質含量4.5至9%之葉黃素晶體/濃縮物並以預定量混合該類薑黃素粉/濃縮物及葉黃素晶體/濃縮物以形成摻合物;未處理乾燥摻合物之粒度可落於D50-20μm至50μm及D90-100μm至200μm之範圍內;b)將賦形劑諸如合適之乳化劑及麥芽糊精添加至該摻合物;c)將純化水添加至該摻合物以製造懸浮液;d)使該懸浮液通過液體膠體磨機以形成均勻懸浮液;e)使該懸浮液經受進一步處理諸如通過均質機或高剪切顆粒濕磨機以產生微粉化乳液;f)以25rpm之速度將該微粉化乳液進一步攪動8至12小時以使質量溫度升至25℃至40℃,較佳25℃至30℃之溫度;g)使該微粉化乳液經受進一步處理諸如濃縮及/或乾燥以獲得該治療性草藥組合物。 The present invention also provides a method for the manufacture of a therapeutic herbal composition comprising curcuminoids and lutein, wherein the method comprises the following steps: a) using curcuminoid powder having a curcuminoid content in the range of 80 to 95% Lutein crystals/concentrate with a lutein content in the range of 70 to 80% and a zeaxanthin content of 4.5 to 9% and mix the curcumin powder/concentrate and lutein in a predetermined amount Crystals/concentrate to form a blend; the particle size of the untreated dry blend may fall within the range of D 50 -20 μm to 50 μm and D 90 -100 μm to 200 μm; b) excipients such as suitable emulsifiers and maltodextrin is added to the blend; c) purified water is added to the blend to make a suspension; d) the suspension is passed through a liquid colloid mill to form a homogeneous suspension; e) the suspension is Subjecting to further processing such as through a homogenizer or high shear particle wet mill to produce a micronized emulsion; f) further agitating the micronized emulsion at a speed of 25 rpm for 8 to 12 hours to raise the mass temperature to 25°C to 40°C, Preferably a temperature of 25°C to 30°C; g) subjecting the micronized emulsion to further processing such as concentration and/or drying to obtain the therapeutic herbal composition.
該方法進一步包括使經乾燥之治療性草藥組合物通過合適之顆粒篩以獲得均勻成品。此經處理治療性草藥組合物之粒度可落於D50-0.36μm至5μm之範圍內及D90在0.60μm至10μm之範圍內。此粒度減小 有利於該治療性草藥組合物具有高生體可用率。 The method further comprises passing the dried therapeutic herbal composition through a suitable particle sieve to obtain a homogeneous finished product. The particle size of the treated therapeutic herbal composition may fall within the range of D 50 -0.36 μm to 5 μm and D 90 in the range of 0.60 μm to 10 μm. This particle size reduction facilitates high bioavailability of the therapeutic herbal composition.
此外,類薑黃素粉/濃縮物及葉黃素晶體/濃縮物之預定量可為使得該類薑黃素粉/濃縮物在24至49%之範圍內,葉黃素晶體/濃縮物在1至14.5%之範圍內及賦形劑在50至64%之範圍內。 In addition, the predetermined amounts of curcuminoid powder/concentrate and lutein crystals/concentrate can be such that the curcuminoid powder/concentrate is in the range of 24 to 49%, and the lutein crystals/concentrate is in the range of 1 to 49%. In the range of 14.5% and excipient in the range of 50 to 64%.
使用之賦形劑係麥芽糊精及乳化劑。此外,可使用之乳化劑之一些實例係(但不限於)改質澱粉、哥地膠、阿拉伯膠(Gum Arabic)、亞拉伯膠(Gum Acacia)、甲基纖維素及蔗糖脂肪酸酯等。 The excipients used are maltodextrin and emulsifier. In addition, some examples of emulsifiers that can be used are (but not limited to) modified starch, Gum Gum, Gum Arabic, Gum Acacia, methylcellulose, and sucrose fatty acid esters, etc. .
根據本發明之又其他實施例,最終經處理治療性草藥組合物包含在22.5%至46.5%之範圍內之類薑黃素、在0.90%至11.25%之範圍內之葉黃素及在0.06%至1.12%之範圍內之玉米黃質。顯而易見,最終組合物中玉米黃質之量構成該最終組合物中葉黃素總含量之大約6至9%。 According to still other embodiments of the present invention, the final processed therapeutic herbal composition comprises curcuminoids in the range of 22.5% to 46.5%, lutein in the range of 0.90% to 11.25%, and lutein in the range of 0.06% to 1.12% zeaxanthin. It is apparent that the amount of zeaxanthin in the final composition constitutes approximately 6 to 9% of the total content of lutein in the final composition.
根據本發明之另一實施例,可將該治療性草藥組合物添加至食物及/或飲料產品、調配成膳食補充劑、營養品及/或藥物。 According to another embodiment of the present invention, the therapeutic herbal composition can be added to food and/or beverage products, formulated as dietary supplements, nutraceuticals and/or medicines.
根據本發明之另一實施例,該治療性草藥組合物可用作營養品、藥物、膳食/營養補充劑或用作各種食物及飲料產品中之治療性/健康成分。其中該治療性草藥組合物可用作健康成分之食物及飲料之一些實例包括(但不限於)茶、輸液、果汁、飲料、牛奶及牛奶製品、穀類產品、酒精飲料及加工食物等。 According to another embodiment of the present invention, the therapeutic herbal composition can be used as a nutraceutical, medicine, dietary/nutritional supplement or as a therapeutic/health ingredient in various food and beverage products. Some examples of foods and beverages in which the therapeutic herbal composition can be used as a health ingredient include, but are not limited to, teas, infusions, fruit juices, beverages, milk and milk products, cereal products, alcoholic beverages, processed foods, and the like.
根據本發明之另一實施例,該治療性草藥組合物亦可調配成選自由以下組成之群之合適劑型:粉末、糊劑、錠劑、糖漿、輸液及或膠囊等。 According to another embodiment of the present invention, the therapeutic herbal composition can also be formulated into a suitable dosage form selected from the group consisting of powder, paste, lozenge, syrup, infusion solution and/or capsule and the like.
在本發明之一實施例中,亦可將該治療性草藥組合物添加至任何酒精飲料以抵消人們飲用酒精時的不良影響。 In one embodiment of the present invention, the therapeutic herbal composition can also be added to any alcoholic beverage to counteract the adverse effects of alcohol consumption by people.
下列實例旨在進一步闡述本發明之某些較佳實施例且本質上非限制性的。熟習此項技術者將知曉或可確定,僅使用例行性實驗即可獲得本文描述之特定物質及程序之許多等效物。 The following examples are intended to further illustrate certain preferred embodiments of the invention and are non-limiting in nature. Those skilled in the art will know, or can ascertain, using no more than routine experimentation, many equivalents to the specific materials and procedures described herein.
實例1 Example 1
在此實例中,描述本發明之代表性治療性草藥組合物,其包含按重量計之不同量之類薑黃素、葉黃素及賦形劑。表1顯示具有不同量之類薑黃素、葉黃素及賦形劑之治療性草藥組合物中所使用之組分及其量。此外,該等類薑黃素可以類薑黃素粉或類薑黃素濃縮物之形式使用且葉黃素可獲自葉黃素晶體或葉黃素濃縮物。 In this example, a representative therapeutic herbal composition of the invention is described comprising various amounts by weight of curcuminoids, lutein and excipients. Table 1 shows the components and their amounts used in the therapeutic herbal compositions with different amounts of curcuminoids, lutein and excipients. Furthermore, the curcuminoids can be used in the form of curcuminoid powder or curcuminoid concentrate and lutein can be obtained from lutein crystals or lutein concentrate.
實例2 Example 2
藉由如下文描述之方法製造如實例1中詳細闡述之治療性草藥組合物:以給定量將具有類薑黃素含量80至95%之類薑黃素粉/濃縮物及具有葉黃素含量70至80%及玉米黃質含量4.5至9%之葉黃素晶體與賦形劑(諸如改質澱粉乳化劑及麥芽糊精)混合在一起以製備摻合物(100g)。因此獲得之乾燥摻合物之粒度係D50-20μm及D90-100μm。然後用 333.4ml RO純化水重構此摻合物以製造懸浮液並進一步攪動直至該懸浮液中不存在團塊。然後在液體膠體磨機或液體濕磨機中預乳化該懸浮液以形成均勻乳液。使因此獲得之預乳液通過高壓均質機/高剪切顆粒濕磨機以產生亞微米尺寸乳液液滴。然後以達25rpm之速度將該微粉化乳液進一步攪動8至12小時以使質量溫度升至25℃至40℃,較佳25℃至30℃。此微粉化乳液以不超過110℃之腔室溫度進一步噴霧乾燥。然後收集噴霧乾燥粗粉並經受使用微粉碎機乾磨或空氣輔助高剪切乾磨。收集磨碎顆粒並使其等通過合適之顆粒篩且摻合以獲得最終產物。該最終粉末之粒度係在D50-0.360μm至5μm及D90-0.60μm至10μm之範圍內。 The therapeutic herbal composition as detailed in Example 1 was manufactured by the method as described below: a curcuminoid powder/concentrate having a curcuminoid content of 80 to 95% and a lutein content of 70 to Lutein crystals at 80% and with a zeaxanthin content of 4.5 to 9% were mixed together with excipients such as modified starch emulsifier and maltodextrin to prepare a blend (100 g). The particle sizes of the dry blends thus obtained are D 50 -20 μm and D 90 -100 μm. This blend was then reconstituted with 333.4 ml RO purified water to make a suspension and further agitated until no lumps were present in the suspension. The suspension is then pre-emulsified in a liquid colloid mill or liquid wet mill to form a homogeneous emulsion. The pre-emulsion thus obtained is passed through a high pressure homogenizer/high shear particle wet mill to produce sub-micron sized emulsion droplets. The micronized emulsion is then further agitated at a speed of up to 25 rpm for 8 to 12 hours to raise the mass temperature to 25°C to 40°C, preferably 25°C to 30°C. This micronized emulsion was further spray dried at a chamber temperature not exceeding 110°C. The spray dried meal is then collected and subjected to dry milling using a micronizer or air assisted high shear dry milling. The ground particles are collected and passed through a suitable particle sieve and blended to obtain the final product. The particle size of the final powder is in the range of D 50 −0.360 μm to 5 μm and D 90 −0.60 μm to 10 μm.
藉由上文方法製造之實例1之100gm治療性草藥組合物含有如下活性成分:類薑黃素含量22.5%至46.5%;葉黃素含量0.90%至11.25%及玉米黃質含量0.06%至1.12%。顯而易見,最終組合物中玉米黃質之量構成該最終組合物中葉黃素總含量之大約6至9%。 100 gm of the therapeutic herbal composition of Example 1 manufactured by the above method contains the following active ingredients: curcuminoid content 22.5% to 46.5%; lutein content 0.90% to 11.25% and zeaxanthin content 0.06% to 1.12% . It is apparent that the amount of zeaxanthin in the final composition constitutes approximately 6 to 9% of the total content of lutein in the final composition.
實例3 Example 3
藉由如下文描述之方法製造如實例1中詳細闡述之治療性草藥組合物:將28.13g具有類薑黃素含量80%之類薑黃素粉及14.10g具有葉黃素含量80%及玉米黃質含量5.72%之葉黃素晶體與52.22g改質澱粉乳化劑及10g麥芽糊精混合在一起以製備摻合物(100g)。因此獲得之乾燥摻合物之粒度係D50-40μm及D90-155μm。然後用333.4ml RO純化水重構此乾燥摻合物以製造懸浮液並進一步攪動直至該懸浮液中不存在團塊。 然後在液體膠體磨機或液體濕磨機中預乳化該懸浮液以形成均勻乳液。使因此獲得之預乳液通過高壓均質機/高剪切顆粒濕磨機以產生亞微米尺寸乳液液滴。以達25rpm之速度將該微粉化乳液進一步攪動8至12小時以使質量溫度升至25℃至40℃,較佳25℃至30℃。此微粉化乳液以不超過110℃之腔室溫度進一步噴霧乾燥。然後收集該噴霧乾燥粗粉並經受使用微粉碎機乾磨或空氣輔助高剪切乾磨。收集磨碎顆粒並使其等通過合適之顆粒篩且摻合以獲得最終產物。該最終粉末之粒度係D50-2.76μm及D90-5.44μm。 A therapeutic herbal composition as detailed in Example 1 was manufactured by the method as described below: 28.13 g of curcuminoid powder having a curcuminoid content of 80% and 14.10 g of lutein content of 80% and zeaxanthin Lutein crystals with a content of 5.72% were mixed together with 52.22 g of modified starch emulsifier and 10 g of maltodextrin to prepare a blend (100 g). The particle sizes of the dry blends thus obtained are D 50 -40 μm and D 90 -155 μm. This dry blend was then reconstituted with 333.4 ml RO purified water to make a suspension and further agitated until no lumps were present in the suspension. The suspension is then pre-emulsified in a liquid colloid mill or liquid wet mill to form a homogeneous emulsion. The pre-emulsion thus obtained is passed through a high pressure homogenizer/high shear particle wet mill to produce sub-micron sized emulsion droplets. The micronized emulsion is further agitated at a speed of up to 25 rpm for 8 to 12 hours to raise the mass temperature to 25°C to 40°C, preferably 25°C to 30°C. This micronized emulsion was further spray dried at a chamber temperature not exceeding 110°C. The spray dried meal is then collected and subjected to dry milling using a micronizer or air assisted high shear dry milling. The ground particles are collected and passed through a suitable particle sieve and blended to obtain the final product. The particle size of the final powder is D 50 -2.76 μm and D 90 -5.44 μm.
實例3中製造之100gm治療性草藥組合物含有22.5%類薑黃素、11.25%葉黃素及0.81%玉米黃質。 100 gm of the therapeutic herbal composition manufactured in Example 3 contained 22.5% curcuminoids, 11.25% lutein and 0.81% zeaxanthin.
實例4 Example 4
藉由如下文描述之方法製造如實例1中詳細闡述之治療性草藥組合物:將31.57g具有類薑黃素含量95%之類薑黃素濃縮物及3.85g具有葉黃素含量78%及玉米黃質含量5.10%之葉黃素濃縮物與54.58g改質澱粉乳化劑及10g麥芽糊精混合在一起以製備摻合物(100g)。因此獲得之乾燥摻合物之粒度係D50-50μm及D90-200μm。然後用333.4ml RO純化水重構此乾燥摻合物以製造懸浮液並進一步攪動直至該懸浮液中不存在團塊。然後在液體膠體磨機或液體濕磨機中預乳化該懸浮液以形成均勻乳液。使因此獲得之預乳液通過高壓均質機/高剪切顆粒濕磨機以產生亞微米尺寸乳液液滴。以達25rpm之速度將該微粉化乳液進一步攪動8至12小時以使質量溫度升至25℃至40℃,較佳25℃至30℃。此微粉化乳液以不 超過110℃之腔室溫度進一步噴霧乾燥。然後收集該噴霧乾燥粗粉並經受使用微粉碎機乾磨或空氣輔助高剪切乾磨。收集磨碎顆粒並使其等通過合適之顆粒篩且摻合以獲得最終產物。該最終粉末之粒度係D50-0.6μm及D90-6.8μm。 A therapeutic herbal composition as detailed in Example 1 was manufactured by the method as described below: 31.57 g of curcuminoid concentrate having a curcuminoid content of 95% and 3.85 g of lutein content of 78% and zeaxanthin Lutein concentrate with a content of 5.10% was mixed with 54.58 g of modified starch emulsifier and 10 g of maltodextrin to prepare a blend (100 g). The particle sizes of the dry blends thus obtained are D 50 -50 μm and D 90 -200 μm. This dry blend was then reconstituted with 333.4 ml RO purified water to make a suspension and further agitated until no lumps were present in the suspension. The suspension is then pre-emulsified in a liquid colloid mill or liquid wet mill to form a homogeneous emulsion. The pre-emulsion thus obtained is passed through a high pressure homogenizer/high shear particle wet mill to produce sub-micron sized emulsion droplets. The micronized emulsion is further agitated at a speed of up to 25 rpm for 8 to 12 hours to raise the mass temperature to 25°C to 40°C, preferably 25°C to 30°C. This micronized emulsion was further spray dried at a chamber temperature not exceeding 110°C. The spray dried meal is then collected and subjected to dry milling using a micronizer or air assisted high shear dry milling. The ground particles are collected and passed through a suitable particle sieve and blended to obtain the final product. The particle size of the final powder is D 50 -0.6 μm and D 90 -6.8 μm.
實例4中製造之100gm治療性草藥組合物含有30%類薑黃素、3%葉黃素及0.20%玉米黃質。 100 gm of the therapeutic herbal composition made in Example 4 contained 30% curcuminoids, 3% lutein and 0.20% zeaxanthin.
實例5 Example 5
藉由如下文描述之方法製造如實例1中詳細闡述之治療性草藥組合物:將48.95g具有類薑黃素含量95%之類薑黃素粉及2.65g具有葉黃素含量70%及玉米黃質含量7.0%之葉黃素晶體與38.40g改質澱粉乳化劑及10g麥芽糊精混合在一起以製備摻合物(100g)。因此獲得之乾燥摻合物之粒度係D50-50μm及D90-200μm。然後用333.4ml RO純化水重構此乾燥摻合物以製造懸浮液並進一步攪動直至該懸浮液中不存在團塊。然後在液體膠體磨機或液體濕磨機中預乳化該懸浮液以形成均勻乳液。使因此獲得之預乳液通過高壓均質機/高剪切顆粒濕磨機以產生亞微米尺寸乳液液滴。以達25rpm之速度將該微粉化乳液進一步攪動8至12小時以使質量溫度升至25℃至40℃,較佳25℃至30℃。此微粉化乳液以不超過110℃之腔室溫度進一步噴霧乾燥。然後收集該噴霧乾燥粗粉並經受使用微粉碎機乾磨或空氣輔助高剪切乾磨。收集磨碎顆粒並使其等通過合適之顆粒篩且摻合以獲得最終產物。該最終粉末之粒度係D50-0.36μm及D90-2.2μm。 A therapeutic herbal composition as detailed in Example 1 was manufactured by the method as described below: 48.95 g of curcuminoid powder having a curcuminoid content of 95% and 2.65 g of lutein content of 70% and zeaxanthin Lutein crystals with a content of 7.0% were mixed together with 38.40 g of modified starch emulsifier and 10 g of maltodextrin to prepare a blend (100 g). The particle sizes of the dry blends thus obtained are D 50 -50 μm and D 90 -200 μm. This dry blend was then reconstituted with 333.4 ml RO purified water to make a suspension and further agitated until no lumps were present in the suspension. The suspension is then pre-emulsified in a liquid colloid mill or liquid wet mill to form a homogeneous emulsion. The pre-emulsion thus obtained is passed through a high pressure homogenizer/high shear particle wet mill to produce sub-micron sized emulsion droplets. The micronized emulsion is further agitated at a speed of up to 25 rpm for 8 to 12 hours to raise the mass temperature to 25°C to 40°C, preferably 25°C to 30°C. This micronized emulsion was further spray dried at a chamber temperature not exceeding 110°C. The spray dried meal is then collected and subjected to dry milling using a micronizer or air assisted high shear dry milling. The ground particles are collected and passed through a suitable particle sieve and blended to obtain the final product. The particle size of the final powder is D 50 -0.36 μm and D 90 -2.2 μm.
實例5中製造之100gm治療性草藥組合物含有46.5%類薑黃素、1.86%葉黃素及0.18%玉米黃質。 100 gm of the therapeutic herbal composition manufactured in Example 5 contained 46.5% curcuminoids, 1.86% lutein and 0.18% zeaxanthin.
實例6 Example 6
藉由如下文描述之方法製造如實例1中詳細闡述之治療性草藥組合物:將48.95g具有類薑黃素含量95%之類薑黃素濃縮物及1.20g具有葉黃素含量75%及玉米黃質含量5.65%之葉黃素濃縮物與54.58g改質澱粉乳化劑及10g麥芽糊精混合在一起以製備摻合物(100g)。因此獲得之乾燥摻合物之粒度係D50-50μm及D90-200μm。然後用333.4ml RO純化水重構此乾燥摻合物以製造懸浮液並進一步攪動直至該懸浮液中不存在團塊。然後在液體膠體磨機或液體濕磨機中預乳化該懸浮液以形成均勻乳液。使因此獲得之預乳液通過高壓均質機/高剪切顆粒濕磨機以產生亞微米尺寸乳液液滴。以達25rpm之速度將該微粉化乳液進一步攪動8至12小時以使質量溫度升至25℃至40℃,較佳25℃至30℃。此微粉化乳液以不超過110℃之腔室溫度進一步噴霧乾燥。然後收集該噴霧乾燥粗粉並經受使用微粉碎機乾磨或空氣輔助高剪切乾磨。收集磨碎顆粒並使其等通過合適之顆粒篩且摻合以獲得最終產物。該最終粉末之粒度係D50-4.8μm及D90-9.9μm。 The therapeutic herbal composition as detailed in Example 1 was manufactured by the method as described below: 48.95 g of curcuminoid concentrate having a curcuminoid content of 95% and 1.20 g of lutein content of 75% and zeaxanthin Lutein concentrate with a content of 5.65% was mixed with 54.58 g of modified starch emulsifier and 10 g of maltodextrin to prepare a blend (100 g). The particle sizes of the dry blends thus obtained are D 50 -50 μm and D 90 -200 μm. This dry blend was then reconstituted with 333.4 ml RO purified water to make a suspension and further agitated until no lumps were present in the suspension. The suspension is then pre-emulsified in a liquid colloid mill or liquid wet mill to form a homogeneous emulsion. The pre-emulsion thus obtained is passed through a high pressure homogenizer/high shear particle wet mill to produce sub-micron sized emulsion droplets. The micronized emulsion is further agitated at a speed of up to 25 rpm for 8 to 12 hours to raise the mass temperature to 25°C to 40°C, preferably 25°C to 30°C. This micronized emulsion was further spray dried at a chamber temperature not exceeding 110°C. The spray dried meal is then collected and subjected to dry milling using a micronizer or air assisted high shear dry milling. The ground particles are collected and passed through a suitable particle sieve and blended to obtain the final product. The particle size of the final powder is D 50 -4.8 μm and D 90 -9.9 μm.
實例6中製造之100gm治療性草藥組合物含有46.50%類薑黃素、0.90%葉黃素及0.068%玉米黃質。 100 gm of the therapeutic herbal composition manufactured in Example 6 contained 46.50% curcuminoids, 0.90% lutein and 0.068% zeaxanthin.
實例7 Example 7
評估治療性草藥組合物對肝細胞上乙醇誘導之及藥物誘導之氧化壓力之治療效應之活體外研究。 In vitro studies evaluating the therapeutic effects of therapeutic herbal compositions on ethanol-induced and drug-induced oxidative stress on hepatocytes.
目的: Purpose:
研究之主要目的係研究本發明之治療性草藥組合物相比於水飛薊素對人類肝癌(HepG2)細胞系中乙醇及對乙醯胺基酚(APAP)誘導之氧化應激損傷之影響。由於水飛薊素廣泛用以治療肝病,因此其用以比較本發明之新穎治療性草藥組合物之效用。此外,由於乙醇係一般抗發炎劑且肝係不考慮該抗發炎劑之發炎常見靶部位,因此研究該草藥組合物對肝細胞之影響且該抗發炎劑以乙醇的形式使用。 The main objective of the study was to investigate the effect of the therapeutic herbal composition of the present invention compared to silymarin on ethanol and acetaminophen (APAP) induced oxidative stress damage in human liver cancer (HepG2) cell lines. Since silymarin is widely used in the treatment of liver diseases, it was used to compare the efficacy of the novel therapeutic herbal composition of the present invention. In addition, since ethanol is a general anti-inflammatory agent and the liver is not considered a common target site of inflammation for this anti-inflammatory agent, the effect of the herbal composition on liver cells was studied and the anti-inflammatory agent was used in the form of ethanol.
細胞系及培養基: Cell lines and media:
在人類肝癌(HepG2)細胞中進行此工作。該等HepG2細胞獲自National Centre for Cell Science,Pune,India。在37℃下在5% CO2氣氛中將細胞維持在具有10% FBS、1%麩醯胺酸及100U青黴素-鏈黴素之DMEM培養基中。將儲備液維持在T-75cm2組織培養瓶中。 This work was performed in human hepatoma (HepG2) cells. The HepG2 cells were obtained from National Center for Cell Science, Pune, India. Cells were maintained in DMEM medium with 10% FBS, 1% glutamine and 100 U penicillin-streptomycin at 37°C in a 5% CO2 atmosphere. Stock solutions were maintained in T- 75cm tissue culture flasks.
實驗 experiment
細胞毒性研究 Cytotoxicity studies
藉由MTT分析評估未處理類薑黃素(U1)、未處理葉黃素(U2)、未處理治療性草藥組合物(U3)及經處理類薑黃素(P1)、經處理葉黃素(P2)及經處理治療性草藥組合物(P3)、水飛薊素之無毒濃度及其針對乙醇及對乙醯胺基酚(APAP)誘導之細胞毒性之預防效應。 Evaluation of unprocessed curcuminoids (U1), unprocessed lutein (U2), unprocessed therapeutic herbal composition (U3) and processed curcuminoids (P1), processed lutein (P2) by MTT analysis ) and processed therapeutic herbal composition (P3), non-toxic concentrations of silymarin and its preventive effect against ethanol and acetaminophen (APAP)-induced cytotoxicity.
樣品之製備: Sample preparation:
1.藉由使用經純化之類薑黃素粉並以預定量將其與乾燥成分(即乳化劑及麥芽糊精)混合製備未處理類薑黃素(U1)樣品。 1. Unprocessed curcuminoid (U1) samples were prepared by using purified curcuminoid powder and mixing it with dry ingredients (ie emulsifier and maltodextrin) in predetermined amounts.
2.藉由使用經純化之葉黃素濃縮物並以預定量將其與乾燥成分(即乳化劑及麥芽糊精)混合製備未處理葉黃素(U2)樣品。 2. Prepare unprocessed lutein (U2) samples by using purified lutein concentrate and mixing it with dry ingredients (ie emulsifier and maltodextrin) in predetermined amounts.
3.藉由混合預定量的類薑黃素粉、具有6至9%玉米黃質之葉黃素並與預定量的賦形劑(諸如乳化劑及麥芽糊精)進一步乾燥摻合製備未處理治療性草藥組合物(U3)樣品。 3. Prepare untreated by mixing predetermined amount of curcuminoid powder, lutein with 6 to 9% zeaxanthin and further dry blending with predetermined amount of excipients such as emulsifier and maltodextrin Sample of therapeutic herbal composition (U3).
3.藉由使用經純化之類薑黃素並將其與賦形劑(即乳化劑及麥芽糊精)混合製備經處理類薑黃素(P1)樣品。然後使此乾燥摻合物經受與如針對上文實例2至4之任一者中之治療性草藥組合物描述之處理類似之處理。 3. Prepare processed curcuminoid (P1 ) samples by using purified curcuminoid and mixing it with excipients (ie emulsifier and maltodextrin). This dry blend was then subjected to a treatment similar to that described for the therapeutic herbal composition in any of Examples 2-4 above.
4.藉由使用經純化之葉黃素並將其與賦形劑(即乳化劑及麥芽糊精)混合製備經處理葉黃素(P2)樣品。然後使此乾燥摻合物經受與與如針對上文實例2至4之任一者中之治療性草藥組合物描述之處理類似之處理。 4. Prepare processed lutein (P2) samples by using purified lutein and mixing it with excipients (ie emulsifier and maltodextrin). This dry blend was then subjected to a treatment similar to that described for the therapeutic herbal composition in any of Examples 2-4 above.
5.如上文實例2至6之任一者中描述製備經處理治療性草藥組合物樣品(P3)。 5. Prepare the treated therapeutic herbal composition sample (P3) as described in any one of Examples 2 to 6 above.
收集HepG2細胞並以密度(1x105個細胞/孔)接種於96孔盤中。以確定24小時U1、U2、U3、P1、P2、P3(1.95、3.90、7.81、15.62、31.25、62.50、125、250、500及1000μg/ml)之毒性並進行MTT分析。為評估U1、U2、U3、P1、P2、P3針對乙醇及乙醇與APAP之治療性效用,用不同濃度之U1、U2、U3、P1、P2、P3(3.90、7.81、15.62、31.25、62.50及125μg/ml)及水飛薊素(3.12至200μg)將細胞預處理1h, 然後曝露於乙醇(100mM)及對乙醯胺基酚(APAP)(20mM)。然後,添加100μl MTT溶液(5mg/ml於PBS中)並將該培養再延長4h。然後,添加100μl DMSO並使用多模平板閱讀器在570nm下量測吸光度。 HepG2 cells were harvested and seeded in 96-well dishes at a density ( 1x105 cells/well). To determine the toxicity of U1, U2, U3, P1, P2, P3 (1.95, 3.90, 7.81, 15.62, 31.25, 62.50, 125, 250, 500 and 1000 μg/ml) at 24 hours and perform MTT analysis. In order to evaluate the therapeutic effect of U1, U2, U3, P1, P2, P3 against ethanol and ethanol and APAP, different concentrations of U1, U2, U3, P1, P2, P3 (3.90, 7.81, 15.62, 31.25, 62.50 and 125 μg/ml) and silymarin (3.12 to 200 μg) for 1 h, and then exposed to ethanol (100 mM) and acetaminophen (APAP) (20 mM). Then, 100 μl of MTT solution (5 mg/ml in PBS) was added and the incubation was prolonged for another 4 h. Then, 100 μl of DMSO was added and the absorbance was measured at 570 nm using a multimode plate reader.
脂質過氧化及抗氧化劑酵素活性分析 Analysis of Lipid Peroxidation and Antioxidant Enzyme Activity
基於硫巴比妥酸反應性物質(TBARS)產生測定丙二醛(MDA)(脂質過氧化之指標)之濃度,根據套組方法(Himedia及Sigma)量測超氧化物歧化酶(SOD)活性及麩胱甘肽還原酶(GSH)。藉由標準分光光度法量測樣品。 Measurement of malondialdehyde (MDA) (indicator of lipid peroxidation) concentration based on thiobarbituric acid reactive substances (TBARS) production, measurement of superoxide dismutase (SOD) activity according to a set of methods (Himedia and Sigma) and glutathione reductase (GSH). Samples were measured by standard spectrophotometry.
用乙醇(100mM)U1、U2、U3、P1、P2及P3及水飛薊素將細胞培養24h。量測療性草藥組合物中之總GSH含量、SOD活性及脂質過氧化。 Cells were cultured with ethanol (100 mM) U1, U2, U3, P1, P2 and P3 and silymarin for 24 h. The total GSH content, SOD activity and lipid peroxidation in the therapeutic herbal composition were measured.
用APAP(20mM)及P3、水飛薊素將細胞培養24h。量測保肝組合物中之總GSH含量、SOD活性及脂質過氧化。 Cells were cultured with APAP (20mM), P3 and silymarin for 24h. The total GSH content, SOD activity and lipid peroxidation in the hepatoprotective composition were measured.
細胞內活性氧物質(ROS)之量測 Measurement of intracellular reactive oxygen species (ROS)
使用可滲入細胞之細胞內基質中之非螢光探針2,7-二乙醯二氯螢光素(DCFH-DH)量測ROS,其中由ROS將該探針氧化為螢光二氯螢光素(DCF)。簡而言之,在正常磷酸鹽緩衝鹽水(pH 7.4)中,將分離細胞之等分試樣(8×106個細胞/ml)定容至2ml之最終體積。取出1ml細胞等分試樣,對該等分試樣添加1μl DCFH-DA(1mg/mL)並在37℃下在黑暗條件下培養30min。在具有數位相機(Nikon 4500 coolpix,Japan)之螢光顯微鏡(Nikon,Eclipse TS100,Japan)上拍攝影像。 ROS is measured using the non-fluorescent probe 2,7-diacetyldichlorofluorescein (DCFH-DH), which can penetrate into the intracellular matrix of cells, where the probe is oxidized to the fluorescent dichlorofluorescein by ROS prime (DCF). Briefly, aliquots of isolated cells (8 x 106 cells/ml) were brought up to a final volume of 2 ml in normal phosphate buffered saline (pH 7.4). A 1 ml cell aliquot was removed, 1 μl of DCFH-DA (1 mg/mL) was added to the aliquot and incubated at 37° C. for 30 min in the dark. Images were taken on a fluorescent microscope (Nikon, Eclipse TS100, Japan) with a digital camera (Nikon 4500 coolpix, Japan).
用62.50μg U1、U2、U3、P1、P2、P3及50μg水飛薊素 將HepG2細胞處理1小時,然後用乙醇100mM處理24小時。使用螢光探針DCF-DA量測細胞內ROS蓄積。 With 62.50μg U1, U2, U3, P1, P2, P3 and 50μg silymarin HepG2 cells were treated for 1 hour and then treated with ethanol 100 mM for 24 hours. Intracellular ROS accumulation was measured using the fluorescent probe DCF-DA.
粒線體膜電位(MMP)之測定 Measurement of Mitochondrial Membrane Potential (MMP)
減小之MMP係早期細胞凋亡之徵象。在此研究中,由螢光染料羅丹明123(Rh 123)偵測MMP之差異。在6孔盤(1×106)中培養HepG2細胞並在指示治療結束時收集該等細胞。在用測試化合物及乙醇或APAP培養24小時後,用Rh 123(5mmol/ml)將細胞培養15分鐘。然後用PBS沖洗細胞;在螢光顯微鏡下使用藍色濾光鏡(450至490nm)觀測螢光。 Decreased MMPs are a sign of early apoptosis. In this study, differences in MMPs were detected by the fluorescent dye rhodamine 123 (Rh 123). HepG2 cells were cultured in 6-well dishes (1×10 6 ) and harvested at the indicated end of treatment. After 24 hours of incubation with test compounds and ethanol or APAP, cells were incubated with Rh 123 (5 mmol/ml) for 15 minutes. Cells were then washed with PBS; fluorescence was observed under a fluorescent microscope using a blue filter (450 to 490 nm).
HepG2細胞之雙螢光及DAPI染色 Double fluorescence and DAPI staining of HepG2 cells
HepG2細胞在35mm細胞培養皿中生長,用各候選化合物P3(62.50μg)、水飛薊素(50μg)及乙醇100mM或APAP(20mM)處理24h,並用杜爾貝科磷酸鹽緩衝鹽水(DPBS)清洗。使用吖啶橙/溴化乙錠(AO/EB)螢光染色偵測與乙醇或APAP誘導之肝毒性相關之形態變化。以1:1混合溴化乙錠(100mg/mL)及吖啶橙(100mg/mL)並將DAPI 100mg mL添加至細胞,接著進行螢光顯微術以評估形態特性。 HepG2 cells were grown in 35 mm cell culture dishes, treated with each candidate compound P3 (62.50 μg), silymarin (50 μg), ethanol 100 mM or APAP (20 mM) for 24 h, and washed with Dulbecco's phosphate-buffered saline (DPBS). Morphological changes associated with ethanol- or APAP-induced hepatotoxicity were detected using acridine orange/ethidium bromide (AO/EB) fluorescent staining. Ethidium bromide (100 mg/mL) and acridine orange (100 mg/mL) were mixed 1:1 and DAPI 100 mg mL was added to the cells, followed by fluorescence microscopy to assess morphological properties.
結果: result:
A.未處理及經處理成分及治療性草藥組合物在HepG2細胞中之細胞毒性效應 A. Cytotoxic Effects of Untreated and Processed Components and Therapeutic Herbal Compositions in HepG2 Cells
圖1(a、b、c)顯示不同濃度之未處理成分及治療性草藥組合物在HepG2細胞中之細胞毒性效應。圖1(d、e及f)顯示不同濃度之經處 理成分及經處理治療性草藥組合物在HepG2細胞中之細胞毒性效應。 Figure 1 (a, b, c) shows the cytotoxic effects of different concentrations of untreated components and therapeutic herbal compositions in HepG2 cells. Figure 1(d, e and f) shows the different concentrations of Cytotoxic effects of physiological components and treated therapeutic herbal compositions in HepG2 cells.
圖2(a、b、c)顯示不同濃度之未處理成分及治療性草藥組合物在乙醇誘導之HepG2細胞中之細胞毒性效應。而圖2(d、e、f)顯示不同濃度之經處理成分及治療性草藥組合物在乙醇誘導之HepG2細胞中之細胞毒性效應。 Figure 2 (a, b, c) shows the cytotoxic effects of different concentrations of untreated ingredients and therapeutic herbal compositions in ethanol-induced HepG2 cells. And Fig. 2 (d, e, f) shows the cytotoxic effects of different concentrations of the treated ingredients and therapeutic herbal composition in ethanol-induced HepG2 cells.
MTT分析顯示,在HepG2細胞中,細胞存活率在低濃度1.95至62.50μg/ml下未改變。另外,相較於乙醇,3.9至62.50μg/ml U1、U2、U3、P1、P2、P3導致細胞存活率顯著增加。當曝露於62.50μg/ml時,觀測到細胞之最大存活率(圖2(a、b、c、d、e及f))。 MTT analysis showed that in HepG2 cells, cell viability was unchanged at low concentrations ranging from 1.95 to 62.50 μg/ml. In addition, 3.9 to 62.50 μg/ml U1, U2, U3, P1, P2, P3 resulted in a significant increase in cell viability compared to ethanol. The maximum viability of the cells was observed when exposed to 62.50 μg/ml (Figure 2 (a, b, c, d, e and f)).
因此,選擇62.50μg/ml濃度之U1、U2、U3、P1、P2、P3作為所有其他研究之最佳保護濃度。 Therefore, U1, U2, U3, P1, P2, P3 at a concentration of 62.50 μg/ml were selected as the optimal protective concentration for all other studies.
B.未處理及經處理成分及治療性草藥組合物對乙醇誘導之HepG2細胞中抗氧化劑酵素活性之治療效應 B. Therapeutic Effects of Untreated and Treated Components and Therapeutic Herbal Compositions on Antioxidant Enzyme Activity in Ethanol-Induced HepG2 Cells
進行硫巴比妥酸反應性物質(TBARS)分析以研究U1、U2、U3、P1、P2、P3及水飛薊素改變HepG2細胞中乙醇誘導之脂質過氧化之能力(圖3a)。結果顯示乙醇刺激組相較於對照細胞之更高TBARS形成。但該TBARS形成分別由經處理細胞減少。 Thiobarbituric acid reactive substances (TBARS) assay was performed to investigate the ability of U1, U2, U3, P1, P2, P3 and silymarin to alter ethanol-induced lipid peroxidation in HepG2 cells (Fig. 3a). The results showed higher TBARS formation in ethanol-stimulated group compared to control cells. But the TBARS formation was reduced by treated cells, respectively.
U1、U2、U3、P1、P2、P3及水飛薊素對乙醇誘導之SOD活性及GSH濃度之影響顯示於圖3(b)及3(c)中。相較於對照細胞,乙醇刺激顯著降低HepG2細胞中非酵素抗氧化劑GSH濃度及SOD活性。有趣地,相較於乙醇刺激組,經U1、U2、U3、P1、P2、P3及水飛薊素處理之細胞中SOD及GSH增加。 The effects of U1, U2, U3, P1, P2, P3 and silymarin on ethanol-induced SOD activity and GSH concentration are shown in Figures 3(b) and 3(c). Compared with control cells, ethanol stimulation significantly decreased the concentration of non-enzyme antioxidant GSH and the activity of SOD in HepG2 cells. Interestingly, SOD and GSH increased in cells treated with U1, U2, U3, P1, P2, P3 and silymarin compared to the ethanol-stimulated group.
此外,經處理治療性草藥組合物(P3)之效應堪比水飛薊素及對照組之效應。此外,由P3顯示之效應優於由未處理組合物及個別組分類薑黃素(U1、P1)及葉黃素(U2、P2)顯示之效應。 Furthermore, the effect of the treated therapeutic herbal composition (P3) was comparable to that of silymarin and the control group. Furthermore, the effect shown by P3 was superior to that shown by the untreated composition and individual components of curcumin (U1, P1) and lutein (U2, P2).
因此,可自此等結果推斷,本發明之治療性草藥組合物之經處理形式(P3)顯示個別組分,即類薑黃素及葉黃素(具有6至9%玉米黃質)之協同活性。 Therefore, it can be concluded from these results that the processed form (P3) of the therapeutic herbal composition of the present invention shows a synergistic activity of the individual components, namely curcuminoids and lutein (with 6 to 9% zeaxanthin) .
C.經處理治療性草藥組合物相比於水飛薊素在經對乙醯胺基酚(APAP)處理之HepG2細胞中之細胞毒性效應 C. Cytotoxic effects of treated therapeutic herbal compositions compared to silymarin in HepG2 cells treated with acetaminophen (APAP)
圖4(a)及4(b)顯示經處理治療性草藥組合物(P3)相比於水飛薊素在經APAP處理之HepG2細胞中之細胞毒性效應之比較。 Figures 4(a) and 4(b) show a comparison of the cytotoxic effect of the treated therapeutic herbal composition (P3) compared to silymarin in APAP-treated HepG2 cells.
在此研究中,APAP曝露在HepG2細胞中引起顯著之細胞毒性。然而,經處理治療性草藥組合物(P3)(62.50μg)預處理在HepG2細胞中顯著防止APAP誘導之細胞毒性;結果堪比水飛薊素(50μg)之結果。 In this study, APAP exposure caused significant cytotoxicity in HepG2 cells. However, pretreatment with the treated therapeutic herbal composition (P3) (62.50 μg) significantly prevented APAP-induced cytotoxicity in HepG2 cells; the results were comparable to those of silymarin (50 μg).
D.經處理治療性草藥組合物相比於水飛薊素對對乙醯胺基酚(APAP)誘導之HepG2細胞上之抗氧化劑酵素活性之治療效應 D. Therapeutic Effects of Treated Therapeutic Herbal Compositions Compared to Silymarin on Antioxidant Enzyme Activity on Acetaminophen (APAP)-Induced HepG2 Cells
可自圖5(a)推斷,當相較於對照治療時,APAP誘導之HepG2細胞中TBARS之濃度增加。此外,在APAP誘導前使用62.50μg經處理治療性草藥組合物(P3)之處理顯著阻止APAP介導之TBARS濃度。此等結果堪比由水飛薊素(50μg)顯示之效應。 It can be inferred from Figure 5(a) that the concentration of TBARS was increased in APAP-induced HepG2 cells when compared to control treatment. Furthermore, treatment with 62.50 μg of the treated therapeutic herbal composition (P3) before APAP induction significantly prevented APAP-mediated TBARS concentrations. These results are comparable to the effects shown by silymarin (50 μg).
抗氧化劑充當針對自由基之主要防禦。由於產生過量ROS,因此APAP誘導之HepG2細胞顯著降低細胞抗氧化狀態。相反,如 圖5(b)及(c)中可見,在誘導APAP之前,使用62.50μg經處理治療性草藥組合物(P3)及50μg給定之水飛薊素濃縮物之處理顯著阻止HepG2細胞中APAP誘導之抗氧化狀態(SOD及GSH濃度)損失。 Antioxidants serve as the primary defense against free radicals. APAP-induced HepG2 cells significantly decreased cellular antioxidant status due to excess ROS production. Instead, as in As can be seen in Figure 5(b) and (c), treatment with 62.50 μg of the treated therapeutic herbal composition (P3) and 50 μg of the given silymarin concentrate prior to induction of APAP significantly prevented APAP-induced antioxidant status in HepG2 cells (SOD and GSH concentration) loss.
E.未處理及經處理治療性草藥組合物相比於水飛薊素對乙醇誘導之細胞內ROS產生之影響 E. Effect of Untreated and Treated Therapeutic Herbal Compositions Compared to Silymarin on Ethanol-Induced Intracellular ROS Production
可自圖6推斷,相較於未處理對照組,由乙醇單獨刺激之HepG2細胞中ROS濃度增加。相反,U1、U2、U3、P1、P2、P3(62.50μg/ml)及水飛薊素(50μg/ml)顯著阻止HepG2細胞中乙醇誘導之ROS產生。藉由用62.50μg/mL經處理治療性草藥組合物(P3)處理的ROS產生的減少堪比50μg/mL水飛薊素處理之結果。 It can be inferred from Figure 6 that the concentration of ROS was increased in HepG2 cells stimulated by ethanol alone compared to the untreated control group. In contrast, U1, U2, U3, P1, P2, P3 (62.50 μg/ml) and silymarin (50 μg/ml) significantly prevented ethanol-induced ROS production in HepG2 cells. The reduction in ROS production by treatment with 62.50 μg/mL of the treated therapeutic herbal composition (P3) was comparable to that of 50 μg/mL silymarin treatment.
F.未處理及經處理治療性草藥組合物相比於水飛薊素對乙醇介導之MMP減少之效應 F. Effect of Untreated and Treated Therapeutic Herbal Compositions Compared to Silymarin on Ethanol-Mediated MMP Reduction
圖7描述相較於對照,曝露於乙醇100mM 24小時之細胞中粒線體膜電位顯著降低。相較於對照,藉由破壞外膜誘導之粒線體膜電位之去極化導致該粒線體中染料之損失及細胞內螢光之降低。相反,用62.50μg/mL的U1、U2、U3、P1、P2及P3及50μg/mL水飛薊素預處理24小時減弱乙醇誘導之粒線體膜去極化,其由螢光強度之增加顯示。 Figure 7 depicts a significant decrease in mitochondrial membrane potential in cells exposed to ethanol 100 mM for 24 hours compared to controls. Depolarization of the mitochondrial membrane potential induced by disruption of the outer membrane resulted in a loss of the dye in the mitochondria and a decrease in intracellular fluorescence compared to controls. In contrast, pretreatment with 62.50 μg/mL of U1, U2, U3, P1, P2, and P3 and 50 μg/mL silymarin for 24 hours attenuated ethanol-induced mitochondrial membrane depolarization, as shown by an increase in fluorescence intensity.
結果指示,經處理治療性草藥組合物(P3)可顯著增加細胞內螢光,藉此指示對防止細胞粒線體膜電位之去極化具有積極效應,且此等結果堪比已知保肝劑水飛薊素之效應。 The results indicate that the treated therapeutic herbal composition (P3) can significantly increase intracellular fluorescence, thereby indicating a positive effect on preventing the depolarization of the mitochondrial membrane potential of cells, and these results are comparable to those known for hepatoprotective The effect of silymarin.
G.未處理及經處理治療性草藥組合物相比於水飛薊素對乙醇誘導之細胞核凋亡之影響 G. Effect of Untreated and Treated Therapeutic Herbal Compositions Compared to Silymarin on Ethanol-Induced Nuclear Apoptosis
圖8顯示在用AO/EtBr染色之後,對照及乙醇誘導之HepG2細胞中之螢光顯微術形態變化。該圖正繪示顯微照相,其正顯示經處理治療性草藥組合物(P3)62.50μg及水飛薊素50μg對HepG2細胞中乙醇100mM誘導之細胞凋亡形態變化之影響。可見經處理治療性草藥組合物(P3)抑制如HepG2細胞中藉由AO/EtBr染色進行染色之細胞凋亡,其堪比由水飛薊素顯示之效應。 Figure 8 shows the fluorescence microscopy morphological changes in control and ethanol-induced HepG2 cells after staining with AO/EtBr. The figure is showing a photomicrograph showing the effect of treated therapeutic herbal composition (P3) 62.50 μg and silymarin 50 μg on the morphological changes of apoptosis induced by ethanol 100 mM in HepG2 cells. It can be seen that the treated therapeutic herbal composition (P3) inhibits apoptosis as stained by AO/EtBr staining in HepG2 cells, comparable to the effect shown by silymarin.
H.未處理及經處理治療性草藥組合物相比於水飛薊素對乙醇誘導之細胞核碎裂之效應 H. Effect of Untreated and Treated Therapeutic Herbal Compositions Compared to Silymarin on Ethanol-Induced Nuclear Fragmentation
如圖9中顯示,經100mM乙醇處理的HepG2細胞引起細胞核濃縮及碎裂。可見在乙醇曝露之前經處理治療性草藥組合物(P3)及水飛薊素處理兩者由於其等強自由基清除性質而均減少細胞核濃縮及碎裂。 As shown in Figure 9, treatment of HepG2 cells with 100 mM ethanol caused condensation and fragmentation of nuclei. It can be seen that both the treated therapeutic herbal composition (P3) and silymarin treatment before ethanol exposure reduced nuclear condensation and fragmentation due to their equally strong free radical scavenging properties.
I.經處理治療性草藥組合物相比於水飛薊素對APAP誘導之細胞內ROS產生之影響 I. Effect of Treated Therapeutic Herbal Compositions Compared to Silymarin on APAP-Induced Intracellular ROS Production
經APAP 20mM處理的HepG2細胞引起DCF螢光增加。用經處理治療性草藥組合物(P3)62.50μg/ml預處理細胞降低DCF螢光,且其指示相較於APAP單獨處理組,P3顯著降低APAP誘導之自由基釋放(圖10)。 HepG2 cells treated with APAP 20 mM caused an increase in DCF fluorescence. Pretreatment of cells with treated therapeutic herbal composition (P3) 62.50 μg/ml decreased DCF fluorescence and it indicated that P3 significantly reduced APAP-induced free radical release compared to APAP alone treatment group (Figure 10).
J.經處理治療性草藥組合物相比於水飛薊素對對乙醯胺基酚(APAP)介導之MMP減少之效應 J. Effect of Treated Therapeutic Herbal Compositions Compared to Silymarin on Acetaminophen (APAP)-Mediated MMP Reduction
如圖11中顯示,在用Rh-123處理後,藉由測定綠色螢光比率量測粒線體膜電位(△Ψm)。將經APAP處理之細胞極化並顯示顯著△Ψm及相較於對照減少之綠色螢光。相較於經APAP單獨處理之細胞,用經處理治療性草藥組合物(P3)預處理細胞增加綠色螢光,此指示粒線體膜之去極化狀態。此外,經處理治療性草藥組合物(P3)及水飛薊素分別顯示相似效應。 As shown in Figure 11, after treatment with Rh-123, the mitochondrial membrane potential ( ΔΨm ) was measured by measuring the ratio of green fluorescence. APAP-treated cells were polarized and showed significant ΔΨm and reduced green fluorescence compared to controls. Pretreatment of cells with the treated therapeutic herbal composition (P3) increased green fluorescence, indicative of the depolarized state of the mitochondrial membrane, compared to cells treated with APAP alone. Furthermore, the treated therapeutic herbal composition (P3) and silymarin each showed similar effects.
K.經處理治療性草藥組合物相比於水飛薊素對APAP誘導之細胞核凋亡之效應 K. Effects of Treated Therapeutic Herbal Compositions Compared to Silymarin on APAP-Induced Nuclear Apoptosis
圖12顯示顯微照相,其顯示經處理治療性草藥組合物(P3)62.50μg及水飛薊素50μg對HepG2細胞中APAP 20mM誘導之細胞凋亡形態變化之效應。可自圖12推斷,經處理治療性草藥組合物(P3)抑制HepG2細胞中如藉由AO/EtBr染色進行染色之細胞凋亡,且該等結果堪比由水飛薊素顯示之效應。 Figure 12 shows photomicrographs showing the effect of treated therapeutic herbal composition (P3) 62.50 μg and silymarin 50 μg on APAP 20 mM induced apoptosis morphological changes in HepG2 cells. It can be concluded from Figure 12 that the treated therapeutic herbal composition (P3) inhibited apoptosis in HepG2 cells as stained by AO/EtBr staining and these results were comparable to the effect shown by silymarin.
L.經處理治療性草藥組合物相比於水飛薊素對APAP誘導之細胞核碎裂之效應 L. Effect of Treated Therapeutic Herbal Compositions Compared to Silymarin on APAP-Induced Nuclear Fragmentation
如圖13中可見,在細胞中觀測到經20mM APAP處理的HepG2細胞引起細胞核濃縮及碎裂及形態變化。相較於經APAP單獨處理之細胞,在APAP曝露之前,用經處理治療性草藥組合物(P3)及水飛薊素預處理抑制細胞核濃縮及碎裂。 As can be seen in Figure 13, HepG2 cells treated with 20 mM APAP caused nuclear condensation and fragmentation and morphological changes were observed in the cells. Pretreatment with the treated therapeutic herbal composition (P3) and silymarin prior to APAP exposure inhibited nuclear condensation and fragmentation compared to cells treated with APAP alone.
實例8:活體內研究 Example 8: In Vivo Studies
治療性草藥組合物抵抗韋斯大鼠中酒精誘導之肝損傷之效應 Effects of a therapeutic herbal composition against alcohol-induced liver injury in Weiss rats
研究目的: Research purposes:
1.監測治療性草藥組合物(P3)對實驗大鼠之初始及最終體重中酒精誘導之肝損傷變化之效應。 1. To monitor the effect of the therapeutic herbal composition (P3) on the changes of alcohol-induced liver injury in the initial and final body weight of experimental rats.
2.藉由評估脂質概況濃度甘油三酯(TG)、總膽固醇(TCH)、低密度脂蛋白膽固醇(LDL-C)及高密度脂蛋白膽固醇(HDL-C),評估不同劑量的治療性草藥組合物(P3)對酒精誘導之肝毒性之效應。 2. Evaluation of different doses of therapeutic herbal medicines by assessing lipid profile concentrations of triglycerides (TG), total cholesterol (TCH), low-density lipoprotein cholesterol (LDL-C) and high-density lipoprotein cholesterol (HDL-C) Effect of composition (P3) on alcohol-induced hepatotoxicity.
3.確定肝標記酵素之活性:天冬胺酸胺基轉移酶(AST)、丙胺酸胺基轉移酶(ALT)及γ-麩胺醯轉移酶(GGT)。 3. Determine the activity of liver marker enzymes: aspartate aminotransferase (AST), alanine aminotransferase (ALT) and γ-glutamine transferase (GGT).
4.藉由評估脂質過氧化標記丙二醛(MDA)及4-羥基壬烯醛(4-HNE),評估不同劑量的治療性草藥組合物(P3)對酒精誘導之肝毒性之效應。 4. To assess the effect of different doses of the therapeutic herbal composition (P3) on alcohol-induced hepatotoxicity by assessing the lipid peroxidation markers malondialdehyde (MDA) and 4-hydroxynonenal (4-HNE).
5.在大鼠中在酒精誘導之肝毒性期間評估發炎(腫瘤壞死因子α(TNF-α)及介白素6(IL-6)及介白素1(IL-1))標記。 5. Assessment of markers of inflammation (tumor necrosis factor alpha (TNF-α) and interleukin 6 (IL-6) and interleukin 1 (IL-1 )) during alcohol-induced hepatotoxicity in rats.
材料及方法: Materials and methods:
動物- animal-
雄性韋斯大鼠,稱重130至150g,購買自Biogen Laboratory Animal Facility,India。動物在標準生態箱條件下在25±2℃下以12h光:暗循環適應環境。動物隨意餵食標準大鼠飼料及水。實驗前禁食18至24h。實驗室動物之護理及使用根據印度CPCSEA理事會指令(MCAS/IAEC/2019/6/9/2/2019)關於動物實驗之良好實驗室規範進行。 Male Wess rats, weighing 130 to 150 g, were purchased from Biogen Laboratory Animal Facility, India. Animals were acclimatized with a 12h light:dark cycle at 25±2°C under standard ecobox conditions. Animals were fed standard rat chow and water ad libitum. Fasting for 18 to 24 hours before the experiment. The care and use of laboratory animals was carried out in accordance with the Good Laboratory Practices for Animal Experiments in accordance with the Indian CPCSEA Council Directive (MCAS/IAEC/2019/6/9/2/2019).
實驗設計及治療時間表: Experimental design and treatment schedule:
將實驗雄性韋斯大鼠分成六組,各組含有六隻動物,分析28天總實驗時期及乙醇、水飛薊素及經處理治療性草藥組合物P3之投與劑量如下:
對照及乙醇組經由灌胃針歷時28天分別接受0.9%鹽水。為檢查治療性性質,在乙醇投與前30min,以200mg/Kg體重的劑量對大鼠投與水飛薊素及治療性草藥組合物(P3),歷時28天。 The control group and the ethanol group received 0.9% saline via intragastric injection for 28 days, respectively. To examine the therapeutic properties, silymarin and a therapeutic herbal composition (P3) were administered to rats at a dose of 200 mg/Kg body weight 30 min before ethanol administration for 28 days.
動物飼料之組成: Composition of animal feed:
蛋白質-17.7%、脂肪-4.2%、碳水化合物-50.5%、纖維-3.4%、礦物質-6.7%及維生素-1.7%。 Protein - 17.7%, Fat - 4.2%, Carbohydrate - 50.5%, Fiber - 3.4%, Mineral - 6.7% and Vitamin - 1.7%.
在研究之第28天,使用氯仿麻醉所有動物,通過靜脈從對照及實驗大鼠收集血液樣本。以3000rpm將血液樣本離心30min,將血清樣本保存在-20℃之冷凍器中直至分析。亦立即取出經處死動物之肝,在冰冷生理鹽水中適當沖洗,吸乾並稱重,以記錄器官重量。然後將肝組織之一部分立即浸入適當之固定劑內以供進一步之組織病理學研究。保存肝之剩餘部分以供生化分析。 On day 28 of the study, all animals were anesthetized with chloroform, and blood samples were collected intravenously from control and experimental rats. Blood samples were centrifuged at 3000 rpm for 30 min, and serum samples were stored in a -20°C freezer until analysis. The livers of sacrificed animals were also immediately removed, rinsed appropriately in ice-cold saline, blotted dry and weighed to record organ weights. A portion of the liver tissue was then immediately immersed in an appropriate fixative for further histopathological studies. The remainder of the liver was saved for biochemical analysis.
肝重量與體重之量測 Measurement of liver weight and body weight
記錄治療時期結束時所有動物之體重,連同其等處死當天之肝重量,以測定各組中動物之肝重量與最終體重。 The body weight of all animals at the end of the treatment period was recorded, together with their liver weight on the day of sacrifice, to determine liver weight and final body weight of animals in each group.
組織勻漿之製備 Preparation of tissue homogenate
使用組織均質機,在含有1mM EDTA之冰冷磷酸鹽緩衝鹽水(PBS)(pH 7.4)中製備10% w/v肝組織勻漿。然後在4℃下以10,000rpm將該勻漿離心30分鐘。因此獲得之上清液進一步用於組織氧化壓力標記之後續生化分析。 A 10% w/v liver tissue homogenate was prepared in ice-cold phosphate buffered saline (PBS) (pH 7.4) containing 1 mM EDTA using a tissue homogenizer. The homogenate was then centrifuged at 10,000 rpm for 30 minutes at 4°C. The supernatant thus obtained was further used for subsequent biochemical analysis of tissue oxidative stress markers.
生化分析 biochemical analysis
藉由Lowry等人(1951)之方法評估蛋白質含量。藉由使用市售診斷套組(Micro clinical lab,Tamil Nadu)進行天冬胺酸胺基轉移酶(AST)、丙胺酸胺基轉移酶(ALT)、γ-麩胺醯轉移酶(GGT)、甘油三酯(TG)、總膽固醇(TCH)、低密度脂蛋白膽固醇(LDL-C)、高密度脂蛋白膽固醇(HDL-C)、丙二醛(MDA)及4-羥基壬烯醛(4-HNE)分析。 Protein content was estimated by the method of Lowry et al. (1951). Aspartate aminotransferase (AST), alanine aminotransferase (ALT), γ-glutamine transferase (GGT), Triglycerides (TG), total cholesterol (TCH), low-density lipoprotein cholesterol (LDL-C), high-density lipoprotein cholesterol (HDL-C), malondialdehyde (MDA) and 4-hydroxynonenal (4 -HNE) analysis.
肝ADH活性 Liver ADH activity
藉由Bonnichsen及Brink(1955)之方法測定酒精脫氫酶(ADH)活性。簡而言之,在1ml最終體積中的50mM甘胺酸(pH 9.6)、0.8mM NAD、3mM乙醇及50μl胞質部分中量測ADH活性。在340nm下量測酵素活性並使用6.22×106M-1 cm-1之莫耳消光係數將該活性計算為nmol NADH形成/min/mg蛋白質。 Alcohol dehydrogenase (ADH) activity was determined by the method of Bonnichsen and Brink (1955). Briefly, ADH activity was measured in 50 mM glycine (pH 9.6), 0.8 mM NAD, 3 mM ethanol and 50 μl of the cytosolic fraction in a final volume of 1 ml. Enzyme activity was measured at 340 nm and calculated as nmol NADH formation/min/mg protein using a molar extinction coefficient of 6.22 x 106 M-1 cm-1.
細胞介素評估 Cytokines Assessment
亦使用市售ELISA套組進行促發炎細胞介素,即腫瘤壞死因子α(TNF-α)及介白素6(IL-6)及介白素1(IL-1)之評估,並以皮克每毫升 (pg/mL)(Biovision,Milpitas,CA,USA)表示。 Pro-inflammatory interleukins, namely tumor necrosis factor α (TNF-α) and interleukin 6 (IL-6) and interleukin 1 (IL-1), were also assessed using commercially available ELISA kits, and skin grams per milliliter (pg/mL) (Biovision, Milpitas, CA, USA).
組織病理學研究 Histopathological studies
將來自對照及經處理動物之大鼠肝組織之一部分直接固定於10%福爾馬林緩衝液中並在用鹽水定期清洗及脫水後包埋於石蠟中。在所有過程後,然後自組織塊使用切片機製備3至5μm厚之切片。然後該等組織切片用蘇木精及曙紅(H&E)染色。在顯微鏡下觀測該等組織切片並使用附接至其之數位相機捕獲對應影像。 A portion of rat liver tissue from control and treated animals was directly fixed in 10% buffered formalin and embedded in paraffin after periodic washing with saline and dehydration. After all procedures, 3 to 5 μm thick sections were then prepared from the tissue blocks using a microtome. The tissue sections were then stained with hematoxylin and eosin (H&E). The tissue sections are observed under a microscope and corresponding images are captured using a digital camera attached to them.
統計學分析 Statistical analysis
各實驗重複至少三次。數據表示為平均值±S.E。在確定處理組之間之方差同質性後,使用方差分析(ANOVA)之單因素事後檢驗(圖基HSD檢驗)分析該等處理組之間不同參數之平均值的顯著性。使用SPSS軟體20.0版進行統計學測試。p<0.05之值視為統計學顯著。 Each experiment was repeated at least three times. Data are expressed as mean ± S.E. After determining the homogeneity of variance between treatment groups, the significance of the means of the different parameters between these treatment groups was analyzed using a one-way post-hoc test (Tukey HSD test) of analysis of variance (ANOVA). Statistical tests were performed using SPSS software version 20.0. Values of p<0.05 were considered statistically significant.
結果: result:
A.經處理治療性草藥組合物對一般觀測體重之效應 A. Effects of Treated Therapeutic Herbal Compositions on General Observed Body Weight
表2顯示對照及實驗大鼠中水及食物之攝入量。在整個研究期間,對照組與實驗大鼠之間食物及水消耗量無差異。 Table 2 shows the water and food intake in control and experimental rats. There was no difference in food and water consumption between control and experimental rats throughout the study period.
表3及3a顯示乙醇及本發明之經處理治療性草藥組合物(P3)分別對大鼠之對照組及實驗組中體重、體重增加及生長速率及肝重量之影響。相比於未處理對照(組1)大鼠,乙醇誘導之大鼠中體重降低,而肝重量顯著增加(P<0.05),而當相較於組2大鼠時,在用經處理治療性草 藥組合物(P3)處理之大鼠(組4至7)中其似乎接近正常。當相較於組1大鼠時,在組2大鼠中,體重、體重增加及生長速率顯著降低(p<0.05)。用具有不同量的類薑黃素及葉黃素(此外具有6至9%玉米黃質)之本發明之治療性草藥組合物(P3)處理亦顯示針對藉由乙醇處理大鼠誘導之體重損失之保護,且堪比由水飛薊素顯示之效應。 Tables 3 and 3a show the effects of ethanol and the treated therapeutic herbal composition of the present invention (P3) on body weight, body weight gain and growth rate and liver weight in control and experimental groups of rats, respectively. Body weight decreased and liver weight increased significantly ( P < 0.05) in ethanol-induced rats compared to untreated control (group 1) rats, while liver weight increased significantly (P < 0.05) in ethanol-induced rats when compared to group 2 rats It appeared to be close to normal in the herbal composition (P3) treated rats (groups 4 to 7). Body weight, body weight gain and growth rate were significantly reduced ( p <0.05) in group 2 rats when compared to group 1 rats. Treatment with the therapeutic herbal composition of the invention (P3) with varying amounts of curcuminoids and lutein (in addition with 6 to 9% zeaxanthin) also showed efficacy against body weight loss induced by ethanol treatment of rats. Protective and comparable to the effect shown by silymarin.
*值係以各組之平均SD給定。 *Values are given as mean SD of each group.
*值係以各組之平均SD給定。 *Values are given as mean SD of each group.
*值係以各組之平均SD給定。 *Values are given as mean SD of each group.
B.本發明之經處理治療性草藥組合物(P3)對肝功能標記酵素變化之效應 B. The effect of the processed therapeutic herbal composition (P3) of the present invention on changes in liver function marker enzymes
表4指示對照及實驗大鼠中肝標記酵素活性之變化。乙醇之投與分別顯著增加(p<0.05)AST、ALT及GGT濃度。最終,相較於經乙醇處理之大鼠,具有4mg至52mg量之類薑黃素及1.05mg至2mg量之葉黃素之本發明之經處理治療性草藥組合物(P3)以200mg/Kg之劑量共處理(組4至7)顯著(p<0.05)降低AST、ALT及GGT之升高濃度。此外,由具有4mg至52mg量之類薑黃素及1.05mg至2mg量之葉黃素之本發明之經處理治療性草藥組合物(P3)顯示之結果(組4至7)堪比水飛薊素之結果。 Table 4 indicates the changes in liver marker enzyme activity in control and experimental rats. The administration of ethanol significantly increased ( p <0.05) the concentrations of AST, ALT and GGT, respectively. Finally, the treated therapeutic herbal composition of the present invention (P3) with an amount of 4 mg to 52 mg of curcuminoids and 1.05 mg to 2 mg of lutein at a dose of 200 mg/Kg compared to ethanol-treated rats. Treatment (Groups 4 to 7) significantly ( p <0.05) reduced elevated concentrations of AST, ALT and GGT. Furthermore, the results (groups 4 to 7) shown by the treated therapeutic herbal composition of the invention (P3) with curcuminoids in amounts of 4 mg to 52 mg and lutein in amounts of 1.05 mg to 2 mg were comparable to those of silymarin.
表4:治療性草藥組合物(P3)對各組(n=6)中對照及實驗大鼠之乙醇誘
導之肝功能標記酵素變化之效應。
*值係以各組之平均SD給定。 *Values are given as mean SD of each group.
乙醇消耗增強肝細胞中NADH/NADP之比率,此引起粒線體中脂肪酸之β-氧化之破壞,從而導致脂肪變性。酒精亦增加脂質自小腸運輸至肝,此導致脂肪酸自脂肪組織之移動增強,脂肪酸由肝吸收(1)。此引起肝細胞之細胞膜受損,從而導致血流中轉胺酶(丙胺酸胺基轉移酶(ALT)及AST(天冬胺酸胺基轉移酶))濃度減弱。γ-麩胺醯轉移酶(GGT)在維持細胞內氧化壓力穩態中發揮關鍵作用,其保護細胞免受氧化損傷。γ-麩胺醯轉移酶存在於細胞膜中且當細胞膜受損時在循環中釋放。 Ethanol consumption enhances the NADH/NADP ratio in hepatocytes, which causes disruption of β-oxidation of fatty acids in mitochondria, leading to steatosis. Alcohol also increases lipid transport from the small intestine to the liver, which results in increased mobilization of fatty acids from adipose tissue and fatty acid absorption by the liver (1). This causes damage to the cell membrane of the liver cells, which leads to a decrease in the concentration of transaminases (alanine aminotransferase (ALT) and AST (aspartate aminotransferase)) in the bloodstream. Gamma-glutamine transferase (GGT) plays a key role in maintaining intracellular oxidative stress homeostasis, which protects cells from oxidative damage. Gamma-glutamine transferase is present in the cell membrane and is released in circulation when the cell membrane is damaged.
本發明發現顯示,相較於對照組,乙醇投與觸發血清AST、ALT及GGT酵素活性顯著增加。此等增加由於酒精中毒引起之肝發炎。投與具有不同量之其組分,即類薑黃素及葉黃素(此外具有6至9%玉米黃質)之本發明之經處理治療性草藥組合物(P3)有效緩解由酒精攝入引起之血清酵素濃度增加,且導致後續恢復至正常,其堪比對照組動物。 The findings of the present invention show that ethanol administration triggers a significant increase in serum AST, ALT and GGT enzyme activities compared to the control group. These increases are due to liver inflammation caused by alcoholism. Administration of the treated therapeutic herbal composition of the present invention (P3) with varying amounts of its components, namely curcuminoids and lutein (in addition with 6 to 9% zeaxanthin) was effective in alleviating the Increased serum enzyme concentrations in rats resulted in a subsequent return to normal, which was comparable to that of control animals.
C.本發明之經處理治療性草藥組合物(P3)對乙醇誘導之脂質概況變化之效應。 C. Effect of the treated therapeutic herbal composition of the invention (P3) on ethanol-induced changes in lipid profile.
表5顯示藉由評估對照及實驗大鼠中脂質概況,即TG、TCH、LDL-C濃度及HDL-C濃度,比較具有不同量之其組分,即類薑黃素及葉黃素(此外具有6至9%玉米黃質)之經處理治療性草藥組合物(P3)對乙醇誘導之肝毒性之效應。 Table 5 shows that by assessing the lipid profiles in control and experimental rats, namely TG, TCH, LDL-C concentration and HDL-C concentration, comparing their components with different amounts, namely curcuminoids and lutein (in addition to Effect of a treated therapeutic herbal composition (P3) from 6 to 9% zeaxanthin) on ethanol-induced hepatotoxicity.
可自表5推斷,用乙醇處理亦破壞身體之脂質概況穩態,導致TG、TCH及LDL-C濃度分別增加,連同HDL-C濃度同時降低(p<0.05)。用本發明之經處理治療性草藥組合物(P3)共處理將TG、TCH及LDL-C、HDL-C濃度之上文變化顯著逆轉至接近正常值(p<0.05)。在乙醇代謝期間,產生大量還原型菸鹼醯胺-腺嘌呤二核苷酸(NADH),因此,抑制克氏循環(Krebs cycle)及脂肪酸氧化,此有利於肝脂肪變性及血清高脂血症(2)。 As can be inferred from Table 5, treatment with ethanol also disrupted the body's lipid profile homeostasis, resulting in an increase in TG, TCH and LDL-C concentrations, respectively, along with a concomitant decrease in HDL-C concentration (p<0.05). Co-treatment with the treated therapeutic herbal composition of the present invention (P3) significantly reversed the above changes in TG, TCH and LDL-C, HDL-C concentrations to near normal values (p<0.05). During ethanol metabolism, a large amount of reduced nicotinamide-adenine dinucleotide (NADH) is produced, thereby inhibiting the Krebs cycle and fatty acid oxidation, which is beneficial to hepatic steatosis and serum hyperlipidemia (2).
另外,乙醇基團亦顯示TCH、TG、LDL濃度增加連同HDL濃度同時降低。然而,用經處理治療性草藥組合物(P3)處理減弱脂質概況濃度之上文變化。因此,本發明之治療性草藥組合物(P3)有效維持該脂質概況。此外,該組合物有效維持健康之膽固醇濃度及健康之LDL/HDL比率並降低升高之血清甘油三酯濃度。 In addition, the ethanol group also showed an increase in TCH, TG, and LDL concentrations along with a simultaneous decrease in HDL concentrations. However, treatment with the treated therapeutic herbal composition (P3) attenuated the above changes in lipid profile concentrations. Therefore, the therapeutic herbal composition (P3) of the present invention is effective in maintaining this lipid profile. Furthermore, the composition is effective in maintaining healthy cholesterol concentrations and healthy LDL/HDL ratios and reducing elevated serum triglyceride concentrations.
用經處理治療性草藥組合物(P3)處理之動物之低膽固醇血症活性可歸因於通過升高之血清HDL濃度增加膽固醇之肝廓清率,或歸因於膽固醇生物合成酵素HMG-CoA還原酶(3)之下調。 The hypocholesterolemic activity of animals treated with the treated therapeutic herbal composition (P3) can be attributed to increased hepatic clearance of cholesterol by elevated serum HDL concentrations, or to reduction of the cholesterol biosynthetic enzyme HMG-CoA Enzyme (3) downregulation.
*值係以各組之平均SD給定。 *Values are given as mean SD of each group.
D.本發明之經處理治療性草藥組合物(P3)對乙醇誘導之肝ADH、MDA及4-HNE變化之效應 D. The effect of the processed therapeutic herbal composition (P3) of the present invention on the changes of liver ADH, MDA and 4-HNE induced by alcohol
表6顯示具有不同量之其組分,即類薑黃素及葉黃素(此外具有6至9%玉米黃質)之本發明之經處理治療性草藥組合物(P3)對乙醇誘導之肝ADH、MDA及4-HNE濃度變化之效應之比較。相較於對照組,乙醇處理組(組2)顯示ADH、MDA及4-HNE濃度顯著增加。乙醇對照組中ADH、MDA及4-HNE濃度增加指示存在肝細胞之脂質概況,其係由於乙醇之毒性效應。 Table 6 shows the effect of the treated therapeutic herbal composition of the invention (P3) with different amounts of its components, namely curcuminoids and lutein (in addition with 6 to 9% zeaxanthin) on ethanol-induced hepatic ADH , Comparison of the effects of MDA and 4-HNE concentration changes. Compared with the control group, the ethanol-treated group (group 2) showed a significant increase in ADH, MDA and 4-HNE concentrations. Increased concentrations of ADH, MDA and 4-HNE in the ethanol control group indicated the presence of a lipid profile of hepatocytes due to the toxic effects of ethanol.
相較於經乙醇處理之大鼠,用具有4mg至52mg量之類薑黃素及1.05mg至2mg量之葉黃素之本發明之經處理治療性草藥組合物(P3)以200mg/Kg之劑量共處理(組4至7)顯著(p<0.05)降低升高之ADH、MDA及4-HNE濃度。 Co-treatment with the treated therapeutic herbal composition of the invention (P3) at a dose of 200 mg/Kg with curcuminoids in amounts of 4 mg to 52 mg and lutein in amounts of 1.05 mg to 2 mg compared to ethanol-treated rats (Groups 4 to 7) significantly ( p <0.05) reduced elevated ADH, MDA and 4-HNE concentrations.
*值係以各組之平均SD給定。 *Values are given as mean SD of each group.
E.本發明之經處理治療性草藥組合物(P3)對乙醇誘導之細胞介素濃度變化之效應 E. Effect of the Treated Therapeutic Herbal Composition (P3) of the Invention on Alcohol-Induced Changes in Cytokinin Concentrations
表7指示本發明之經處理治療性組合物(P3)對乙醇誘導之細胞介素濃度變化之效應。可見相較於對照組1,投與乙醇導致血清TNF-α、IL-6及IL-1濃度顯著增加(p<0.001)。表7顯示在經乙醇處理之組2中,血清TNF-α、IL-6及IL-1濃度分別增加。如在組4至7之結果中可見,用經處理治療性草藥組合物(P3)共處理引起血清TNF-α、IL-6及IL-1濃度之顯著降低。 Table 7 indicates the effect of the treated therapeutic composition of the invention (P3) on ethanol-induced changes in cytokine concentrations. It can be seen that compared with the control group 1, the administration of ethanol led to a significant increase in serum TNF-α, IL-6 and IL-1 concentrations (p<0.001). Table 7 shows that in ethanol-treated group 2, serum TNF-α, IL-6 and IL-1 concentrations were increased, respectively. As seen in the results for Groups 4 to 7, co-treatment with the treated therapeutic herbal composition (P3) caused a significant reduction in serum TNF-α, IL-6 and IL-1 concentrations.
表7:本發明之治療性草藥組合物(P3)對乙醇誘導之細胞介素濃度變
化之效應
*值係以各組之平均SD給定。 *Values are given as mean SD of each group.
F.肝組織之組織病理學分析 F. Histopathological analysis of liver tissue
圖14中顯示之組織學特徵指示對照大鼠中正常肝小葉結構及肝之細胞結構。健康對照肝中無病理學變化。在乙醇誘導之大鼠(即,組2)中,在連續攝入2.4gm/kg bw乙醇4週後,藉由組織學分析評估肝損傷程度。例如,小葉中心肝細胞中觀測到肝竇擴張、擠壓及索萎縮,可能表明肝細胞再生;中央靜脈周圍觀測到單核細胞及壞死細胞之輕微浸潤。在小葉中心區域中觀測到空泡變化及可能之脂肪變性。簡而言之,小葉中心區域中肝細胞腫脹、水樣變性及空泡化變得明顯,此反映由酒精性肝損傷引起之早期生化及病理學變化。 The histological features shown in Figure 14 are indicative of normal hepatic lobular architecture and cellularity of the liver in control rats. There were no pathological changes in healthy control livers. In ethanol-induced rats (ie, Group 2), the extent of liver injury was assessed by histological analysis after 4 weeks of continuous ingestion of 2.4 gm/kg bw ethanol. For example, sinusoidal dilatation, extrusion, and cord atrophy were observed in centrilobular hepatocytes, possibly indicative of hepatocyte regeneration; a slight infiltration of monocytes and necrotic cells was observed around the central vein. Cavitary changes and possible steatosis were observed in the centrilobular region. Briefly, hepatocyte swelling, watery degeneration, and vacuolation became evident in the centrilobular region, reflecting early biochemical and pathological changes resulting from alcoholic liver injury.
乙醇處理組2大鼠在中央靜脈周圍顯示發炎反應伴有空泡變性及壞死。在組3、4及5中,肝組織在中央靜脈周圍顯示較少之空泡變性及較少之發炎反應。組6及7肝組織顯示幾乎正常之結構。因此,本發明 之經處理治療性草藥組合物(P3)顯著改善乙醇誘導之組織病理學變化,尤其在具有4mg至52mg量之類薑黃素及1.05mg至2mg量之葉黃素之本發明之組合物(P3)中(組4至7),其相較於經乙醇處理之組,顯著減輕肝中脂肪變性且顯示明顯之改善。 Rats in ethanol-treated group 2 showed inflammation around the central vein with vacuolar degeneration and necrosis. In groups 3, 4 and 5, liver tissue showed less vacuolar degeneration and less inflammatory response around the central vein. Groups 6 and 7 liver tissues showed almost normal structure. Therefore, the present invention The treated therapeutic herbal composition (P3) significantly improved ethanol-induced histopathological changes, especially in the compositions of the present invention (P3) with curcuminoids in amounts from 4 mg to 52 mg and lutein in amounts from 1.05 mg to 2 mg (Groups 4 to 7), which significantly reduced steatosis in the liver and showed significant improvement compared to the ethanol-treated group.
研究之結論 conclusion of the study
總之,活體內研究證實本發明之經處理治療性草藥組合物(P3)有效正常化並維持脂質概況。此外,該組合物有效維持健康之膽固醇濃度及健康之LDL/HDL比率並有效降低升高之血清甘油三酯濃度。此外,本發明之治療性草藥組合物有效正常化並維持血清酵素(即AST、ALT、ADH及GGT)濃度。評估脂質過氧化標記MDA及4-HNE之研究亦顯示,該治療性草藥組合物(P3)有效降低可由各種因素(諸如不健康飲食模式、PUFA之高消耗量及飲酒)引起之脂質過氧化。此外,發現本發明之治療性草藥組合物(P3)有效降低血清促發炎細胞介素(TNF-α、IL-6及IL-1)濃度,其等係發炎反應之標記。據信在體內患有各種發炎之病患中及亦在患有非酒精性及酒精性脂肪肝疾病之病患中,此等血清促發炎細胞介素(TNF-α、IL-6及IL-1)顯著升高。此外,該治療性草藥組合物藉由保護就其抗氧化狀態而言之肝狀態、降低脂質概況、肝酵素標記濃度及抗發炎活性及抵抗慢性酒精曝露,而有利改善乙醇誘導之肝毒性。 In conclusion, in vivo studies demonstrate that the treated therapeutic herbal composition (P3) of the present invention effectively normalizes and maintains the lipid profile. Furthermore, the composition is effective in maintaining healthy cholesterol concentrations and healthy LDL/HDL ratios and is effective in reducing elevated serum triglyceride concentrations. Furthermore, the therapeutic herbal composition of the present invention is effective in normalizing and maintaining serum enzymes (ie AST, ALT, ADH and GGT) concentrations. Studies evaluating lipid peroxidation markers MDA and 4-HNE also showed that the therapeutic herbal composition (P3) effectively reduces lipid peroxidation that can be caused by various factors such as unhealthy dietary pattern, high consumption of PUFA and alcohol consumption. Furthermore, the therapeutic herbal composition (P3) of the present invention was found to be effective in reducing serum concentrations of pro-inflammatory cytokines (TNF-α, IL-6 and IL-1), which are markers of inflammatory response. It is believed that these serum pro-inflammatory cytokines (TNF-α, IL-6 and IL- 1) Significantly increased. Furthermore, the therapeutic herbal composition favorably ameliorate ethanol-induced hepatotoxicity by protecting the liver status in terms of its antioxidant status, reducing lipid profile, liver enzyme marker concentration and anti-inflammatory activity and against chronic alcohol exposure.
自結果亦推斷,本發明之治療性草藥組合物具有降脂及抗發炎活性。體內異常脂質概況及發炎之致病因素中之一些可為不健康飲食模式、攝入大量酒精、攝入含有大量過氧化多不飽和脂肪酸(PUFA)含量之食物或由於服用某些藥物。體內發炎亦可由於病原感染引起,該病原感 染由感染物/病原體諸如病毒(包括病毒諸如冠狀病毒病(COVID-19)及其他引起感染之病毒)及細菌引起。 It is also concluded from the results that the therapeutic herbal composition of the present invention has lipid-lowering and anti-inflammatory activities. Some of the causative factors for abnormal lipid profile and inflammation in the body can be unhealthy eating patterns, high alcohol intake, intake of foods with high peroxidized polyunsaturated fatty acid (PUFA) content, or due to taking certain medications. Inflammation in the body can also be caused by pathogenic infection, the pathogenic infection Infections are caused by infectious agents/pathogens such as viruses (including viruses such as coronavirus disease (COVID-19) and other viruses that cause infection) and bacteria.
肝組織之組織病理學分析指示本發明之經處理治療性草藥組合物(P3)顯著改善由乙醇誘導之變化,尤其在具有4mg至52mg量之類薑黃素及1.05mg至2mg量之葉黃素之本發明之組合物(P3)中。 Histopathological analysis of liver tissue indicated that the treated therapeutic herbal composition of the present invention (P3) significantly improved the changes induced by ethanol, especially in the presence of curcuminoids in amounts of 4 mg to 52 mg and lutein in amounts of 1.05 mg to 2 mg. In the composition (P3) of the invention.
實例9:臨床研究 Example 9: Clinical Research
治療性草藥組合物健康人類成年男性受試者(subject)在禁食條件下之隨機、開放標籤單中心、一個時期、單劑量、三臂生體可用率研究 Randomized, open-label, single-center, one-period, single-dose, three-arm bioavailability study of a therapeutic herbal composition in healthy human adult male subjects under fasting conditions
研究目的: Research purposes:
主要(藥物動力學)目的: Primary (pharmacokinetic) purpose:
確定本發明治療性草藥組合物中之類薑黃素及葉黃素之生體可用率相比於未調配類薑黃素及未調配葉黃素之生體可用率。 The bioavailability of curcuminoids and lutein in the therapeutic herbal composition of the present invention was determined compared to the bioavailability of unformulated curcuminoids and unformulated lutein.
次要(安全性)目的: Secondary (security) purpose:
次要目的為監測單劑量之本發明治療性草藥組合物在禁食條件下之9個健康人類成年男性受試者中之安全性及耐受性。 The secondary objective was to monitor the safety and tolerability of a single dose of the therapeutic herbal composition of the present invention in 9 healthy human adult male subjects under fasting conditions.
納入標準 Inclusion criteria
(a)受試者必須為健康人類男性受試者。 (a) Subjects must be healthy human male subjects.
(b)年齡必須介於18至65歲(包括)之間,體重至少50kg。 (b) Must be between 18 and 65 years old (inclusive) and weigh at least 50kg.
(c)準備提供書面知情同意書並應願意在整個研究過程期間可用且應 遵循方案中提及之指南之受試者。 (c) be prepared to provide written informed consent and should be willing to be available throughout the course of the study and should Subjects following the guidelines mentioned in the protocol.
(d)在研究前篩選期間,無證據表明患有潛在疾病之受試者。如由病史及身體檢查、ECG、胸部X光片(PA視圖)及在研究開始前14天內進行之實驗室測試確定該等受試者必須係健康的。 (d) Subjects with no evidence of underlying disease during pre-study screening. Subjects must be healthy as determined by medical history and physical examination, ECG, chest X-ray (PA view), and laboratory tests performed within 14 days prior to the start of the study.
(e)篩選實驗室值係於正常限值內或醫師/主要研究者認為無臨床顯著性之受試者。 (e) Subjects whose screening laboratory values are within normal limits or considered clinically significant by the physician/principal investigator.
排除標準: Exclusion criteria:
1.對薑黃或其萃取物或類薑黃素或葉黃素或任何食物或其他藥物過敏之受試者。 1. Subjects who are allergic to turmeric or its extracts or curcuminoids or lutein or any food or other drugs.
2.患有靜息性低血壓(BP<90/60)或高血壓(BP>140/90)且脈搏率低於50/min及超過100/min之受試者。 2. Subjects with resting hypotension (BP<90/60) or hypertension (BP>140/90) and pulse rate below 50/min and over 100/min.
3.患有或既往病史或存在顯著心血管、肺、肝、腎、血液、胃腸、內分泌、免疫、皮膚、神經、肌肉骨骼或精神疾病或於入住前最後4週內曾住院或接受手術之受試者。 3. Suffering or past medical history or significant cardiovascular, lung, liver, kidney, blood, gastrointestinal, endocrine, immune, skin, nerve, musculoskeletal or mental diseases, or hospitalization or surgery within the last 4 weeks before admission subject.
4.具有MI、中風、外周動脈疾病、GI出血、肝損傷、哮喘、腎損傷、癲癇及顱內出血病史之受試者。 4. Subjects with a history of MI, stroke, peripheral artery disease, GI hemorrhage, liver injury, asthma, kidney injury, epilepsy and intracranial hemorrhage.
5.於研究前之最後14天內已服用非處方或處方藥(包括任何酵素修飾藥物)之受試者。 5. Subjects who have taken over-the-counter or prescription drugs (including any enzyme-modifying drugs) within the last 14 days before the study.
6.有酗酒、藥物濫用或吸煙史之受試者。 6. Subjects with a history of alcoholism, drug abuse or smoking.
7.對肝素高敏之受試者。 7. Subjects with high sensitivity to heparin.
8.在過去三個月內參與任何其他臨床研究之受試者。 8. Subjects who participated in any other clinical research in the past three months.
9.具有臨床上顯著異常實驗室值/異常ECG/異常胸部X光片(PA視圖) 之受試者。 9. With clinically significant abnormal laboratory values/abnormal ECG/abnormal chest radiograph (PA view) of subjects.
10.有獻血困難之受試者。 10. Subjects who have difficulties in donating blood.
11.有吞嚥困難史之受試者。 11. Subjects with a history of dysphagia.
12.具有不適用於反復靜脈穿刺之靜脈之受試者。 12. Subjects with veins not suitable for repeated venipuncture.
結果評量 outcome assessment
類薑黃素及葉黃素於各別調配物中之生體可用率 Bioavailability of curcuminoids and lutein in respective formulations
研究程序 research program
A.設計-隨機、試點、雙盲、單中心、一個時期、單劑量、三臂生體可用率研究 A. Design - Randomized, Pilot, Double-Blind, Single-Center, One-Period, Single-Dose, Three-Arm Bioavailability Study
研究處理分配 Research Processing Assignment
組I:3個受試者,包含120mg類薑黃素及20mg葉黃素之治療性草藥組合物 Group I: 3 subjects, a therapeutic herbal composition comprising 120 mg of curcuminoids and 20 mg of lutein
組II:3個受試者,包含30mg類薑黃素及5mg葉黃素之治療性草藥組合物 Group II: 3 subjects, a therapeutic herbal composition comprising 30 mg of curcuminoids and 5 mg of lutein
組III:3個受試者,包含500mg類薑黃素及40mg未調配葉黃素之參考未調配類薑黃素 Group III: 3 subjects, reference unformulated curcuminoids containing 500 mg curcuminoids and 40 mg unformulated lutein
B.受試者之數量-9個健康男性志願者 B. Number of subjects - 9 healthy male volunteers
C.隨機化(分配至處理順序)-藉由適當之統計學軟體(SAS 9.13版或更高版本)進行隨機化以在所有臂中達成相等數量之受試者。操作團隊無法 獲得隨機化時間表以保持其等對處理分配不知情。 C. Randomization (Assignment to Treatment Order) - Randomization is performed by appropriate statistical software (SAS version 9.13 or higher) to achieve an equal number of subjects in all arms. The operations team cannot A randomization schedule was obtained to keep their peers blinded to treatment assignment.
D.整體研究計劃 D. Overall research plan
在獲得倫理委員會批准後,要求受試者訪問站點。在獲得健康受試者之書面知情同意書後,詢問受試者有關其等病史且研究人員進行身體檢查。自各受試者採取血液樣本用於分析血液學及生化分析。進行X射線及ECG。在滿足所有IC/EC標準後,將受試者納入該研究中。 Subjects were asked to visit the site after ethics committee approval was obtained. After obtaining written informed consent from healthy subjects, subjects were asked about their medical history and researchers performed a physical examination. Blood samples were taken from each subject for analysis of hematology and biochemical analyses. X-rays and ECGs are performed. Subjects were enrolled in the study after meeting all IC/EC criteria.
受試者在給藥前12小時進入設施。向該等受試者提供不含薑黃之晚餐。進行身體檢查及生命體徵。 Subjects entered the facility 12 hours prior to dosing. The subjects were provided with a turmeric-free dinner. Perform physical examination and vital signs.
在給藥當天,進行身體檢查及生命體徵並記錄。收集5ml給藥前樣本並離心以供藥物動力學分析。對所有受試者進行給藥。根據隨機化時間表,將囊袋中1gm研究產品(IP)與240ml水混合,並要求該等受試者飲用具有IP之水。給藥後,採血時間點為:0.50、1、2、3、4、6及24小時,用於藥物動力學分析。於此等時間點記錄生命體徵:給藥後00.15、02.00、04.00、06.00、09.00及12.00小時。於此等時間點進行身體檢查:給藥後1、2、6、12小時。在給藥後(即,分別午餐、零食及晚餐)04.00、08.00及13.00小時,向該等受試者提供不含薑黃之食物。記錄不良事件(若存在)。 On the day of administration, physical examination and vital signs were performed and recorded. A 5 ml predose sample was collected and centrifuged for pharmacokinetic analysis. All subjects were dosed. According to the randomization schedule, 1 gm of the investigational product (IP) in the sachet was mixed with 240 ml of water and the subjects were asked to drink the water with the IP. After administration, the time points of blood collection were: 0.50, 1, 2, 3, 4, 6 and 24 hours for pharmacokinetic analysis. Vital signs were recorded at these time points: 00.15, 02.00, 04.00, 06.00, 09.00 and 12.00 hours after dosing. Physical examinations were performed at these time points: 1, 2, 6, 12 hours after dosing. At 04.00, 08.00 and 13.00 hours after dosing (ie lunch, snack and dinner respectively), the subjects were provided with food without turmeric. Adverse events (if any) were recorded.
在檢查時(給藥後24小時)進行身體檢查及生命體徵。給藥後24小時收集血液樣本用於藥物動力學分析。記錄不良事件(若存在)。收集血液樣本用於安全性分析。在完成所有給藥後程序後,該等受試者自該設施出院。 Physical examination and vital signs were performed at the time of examination (24 hours after dosing). Blood samples were collected 24 hours after dosing for pharmacokinetic analysis. Adverse events (if any) were recorded. Blood samples were collected for safety analysis. The subjects were discharged from the facility after completion of all post-dose procedures.
E.藥物動力學分析 E. Pharmacokinetic Analysis
使用符合目的之LC-MS方法分析血漿樣本,定量樣本中總類薑黃素(薑黃素、脫甲氧基薑黃素及雙脫甲氧基薑黃素)以及葉黃素(此外具有6至9%玉米黃質)。使用標準分析工具計算藥物動力學參數。比較來自治療性草藥組合物之組分之相對生體可用率並報導關於未調配類薑黃素及未調配葉黃素來自相同分析之AUC(0-24h)。 Plasma samples were analyzed using a fit-for-purpose LC-MS method to quantify total curcuminoids (curcumin, demethoxycurcumin and bisdemethoxycurcumin) and lutein (in addition to 6 to 9% corn Xanthophyll). Pharmacokinetic parameters are calculated using standard analytical tools. The relative bioavailability of the components from the therapeutic herbal composition was compared and the AUC (0-24h) from the same assay was reported for unformulated curcuminoids and unformulated lutein.
F.統計學分析 F. Statistical Analysis
進行統計學分析並使用InStat軟體(GraphPad Prism,USA)計算AUC。採用雙尾ANOVA分析組間及組內之總血漿類薑黃素含量,而小於0.05之「p」值計算為該研究之統計學顯著性。 Statistical analysis was performed and AUC was calculated using InStat software (GraphPad Prism, USA). Two-tailed ANOVA was used to analyze total plasma curcuminoid levels between and within groups, and a "p" value of less than 0.05 was calculated as statistical significance for the study.
G.結果及討論 G. Results and Discussion
篩選所有9個無病史之受試者。納入該研究內之組I之受試者之平均年齡為28.4,組B為25.9及組C為23.0且平均值為25.76。組I中志願者之平均高度為162.00,組II為167.00,組III為163.00且平均值為164.0。在基線訪問時,組I之平均重量為65.0,組II為75.80及組III為63.0且平均值為69.6及平均BMI為25.60kg/m2。 All 9 subjects with no medical history were screened. The mean age of the subjects in Group I included in the study was 28.4, Group B was 25.9 and Group C was 23.0 with a mean of 25.76. The average height of the volunteers in Group I was 162.00, Group II was 167.00, Group III was 163.00 and the mean was 164.0. At the baseline visit, the mean weight was 65.0 for Group I, 75.80 for Group II and 63.0 for Group III with a mean of 69.6 and a mean BMI of 25.60 kg/ m2 .
安全性:在研究期間,在給藥前及在給藥後,於所有時間點量測生命體徵(諸如溫度、收縮壓及舒張壓、脈搏率及呼吸率)並記錄。此清楚證實,所有受試者均無臨床意義之價值且因此所有研究產品(本發明之治療性草藥組合物)對投與而言均完全安全。未觀測到不良事件。 Safety: During the study, vital signs (such as temperature, systolic and diastolic blood pressure, pulse rate and respiration rate) were measured and recorded at all time points before and after dosing. This clearly demonstrated that all subjects had no clinically significant value and therefore all investigational products (therapeutic herbal compositions of the present invention) were completely safe for administration. No adverse events were observed.
因此,可得出結論,1g囊袋調配物中之本發明治療性草藥組合物對人類口服而言完全安全。 Therefore, it can be concluded that the therapeutic herbal composition of the present invention in 1 g sachet formulation is completely safe for human oral administration.
生體可用率: Bioavailability:
A.類薑黃素 A. Curcuminoids
如表8中可見,在所有組中,發現口服產品後達成峰值血漿類薑黃素濃度(Tmax)之中值時間為2小時。 As can be seen in Table 8, in all groups, the median time to reach peak plasma curcuminoid concentration (Tmax) after oral administration of the product was found to be 2 hours.
在組I中,發現曝露(Cmax及AUClast)分別為20.70及61.93,而在組II中,發現曝露(Cmax及AUClast)分別為2.51及45.48,而在組III中,發現曝露(Cmax及AUClast)分別為8.98及58.83。類薑黃素之平均血漿濃度-時間曲線呈現於圖15中。 In group I, exposures (C max and AUC last ) were found to be 20.70 and 61.93, respectively, while in group II, exposures (C max and AUC last ) were found to be 2.51 and 45.48, respectively, while in group III, exposures ( C max and AUC last ) were 8.98 and 58.83, respectively. The mean plasma concentration-time profiles of curcuminoids are presented in FIG. 15 .
因此,可推斷當相較於較高劑量之未調配類薑黃素之生體可用率時,在攝取時,經處理治療性草藥組合物中存在之類薑黃素為高度生體可用。 Therefore, it can be concluded that the curcuminoids present in the treated therapeutic herbal composition are highly bioavailable upon ingestion when compared to the bioavailability of unformulated curcuminoids at higher doses.
B.葉黃素 B. Lutein
如表9中可見,在所有組中,發現口服產品後達成峰值血漿葉黃素濃度(Tmax)之中值時間為6小時。 As can be seen in Table 9, in all groups the median time to reach peak plasma lutein concentration (Tmax) after oral administration of the product was found to be 6 hours.
在組I中,發現曝露(Cmax及AUClast)分別為55.88及424.87,而在組II中,發現曝露(Cmax及AUClast)分別為35.18及287.70,而在組III中,發現曝露(Cmax及AUClast)分別為52.27及365.07。葉黃素之平均血漿濃度-時間曲線呈現於圖16中。 In group I, exposures (C max and AUC last ) were found to be 55.88 and 424.87, respectively, while in group II, exposures (C max and AUC last ) were found to be 35.18 and 287.70, respectively, while in group III, exposures ( C max and AUC last ) were 52.27 and 365.07, respectively. The mean plasma concentration-time profiles of lutein are presented in FIG. 16 .
因此,可推斷當相較於較高劑量之未調配葉黃素之生體可用率時,在攝取時,經處理治療性草藥組合物中存在之葉黃素為高度生體可用。 Therefore, it can be concluded that the lutein present in the treated therapeutic herbal composition is highly bioavailable upon ingestion when compared to the bioavailability of unformulated lutein at higher doses.
儘管已闡述並描述本發明之治療性草藥組合物之特定實施例,但熟習此項技術者將顯而易見,可作出各種其他變化及修飾而不背離本發明之精神及範圍。因此,意欲在隨附申請專利範圍中涵蓋於本發明之範疇內之此等變化及修飾。 While particular embodiments of the therapeutic herbal compositions of the present invention have been illustrated and described, it would be obvious to those skilled in the art that various other changes and modifications can be made without departing from the spirit and scope of the invention. Accordingly, it is intended to cover in the appended claims such changes and modifications as are within the scope of this invention.
參考文獻: references:
(1) Shukla I、Azmi L、Gupta SS、Upreti DK、Rao CV. (2018) Melioration of anti-hepatotoxic effect by Lichen rangiferinus against alcohol induced liver damage in rats. J Ayurveda Integr Med. 2018 Jan 29. pii: S0975-9476(17) 30047-5。 (1) Shukla I, Azmi L, Gupta SS, Upreti DK, Rao CV. (2018) Melioration of anti-hepatotoxic effect by Lichen raniferinus against alcohol induced liver damage in rats. J Ayurveda Integr Med. 2018 Jan 29. pii: S0975 -9476(17) 30047-5.
(2) Rukkumani, R.、Balasubashini, M.S.、Vishwanathan, P.及Menon, V.P. (2002) Comparative effects of curcumin and photo-irradiated curcumin on alcohol and polyunsaturated fatty acid induced hyperlipidemia. Pharmocol. Res. 46, 257-264。 (2) Rukkumani, R., Balasubashini, M.S., Vishwanathan, P. and Menon, V.P. (2002) Comparative effects of curcumin and photo-irradiated curcumin on alcohol and polyunsaturated fatty acid induced hyperlipidemia. Pharmacol. Res. 46, 257- 264 .
(3) Patil, R.H.、Prakash, K.及Maheshwari, V.L. (2011) Hypolipidemic effect of Terminalia arjuna (L.) in experimentally induced hypercholesteremic rats. Acta Biol. Szeged. 55(2), 289-293。 (3) Patil, R.H., Prakash, K. and Maheshwari, V.L. (2011) Hypolipidemic effect of Terminalia arjuna (L.) in experimentally induced hypercholesteremic rats. Acta Biol. Szeged. 55(2), 289-293.
Claims (20)
Priority Applications (1)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
TW110132930A TWI810644B (en) | 2021-09-03 | 2021-09-03 | Therapeutic herbal composition |
Applications Claiming Priority (1)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
TW110132930A TWI810644B (en) | 2021-09-03 | 2021-09-03 | Therapeutic herbal composition |
Publications (2)
Publication Number | Publication Date |
---|---|
TW202310824A TW202310824A (en) | 2023-03-16 |
TWI810644B true TWI810644B (en) | 2023-08-01 |
Family
ID=86690875
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
TW110132930A TWI810644B (en) | 2021-09-03 | 2021-09-03 | Therapeutic herbal composition |
Country Status (1)
Country | Link |
---|---|
TW (1) | TWI810644B (en) |
Citations (3)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
CN101516364A (en) * | 2006-07-14 | 2009-08-26 | 帝斯曼知识产权资产管理有限公司 | Compositions and use thereof for the treatment, co-treatment or prevention of inflammatory disorders |
TW201513851A (en) * | 2013-03-27 | 2015-04-16 | Jx Nippon Oil & Energy Corp | Antiinflammatory drugs |
TW201818919A (en) * | 2016-10-25 | 2018-06-01 | 美商阿爾塞斯商業集團國際有限公司 | Compositions of LITHOSPERMUM ERYTHRORHIZON (GROMWELL ROOT) and methods of making and using the compositions |
-
2021
- 2021-09-03 TW TW110132930A patent/TWI810644B/en active
Patent Citations (3)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
CN101516364A (en) * | 2006-07-14 | 2009-08-26 | 帝斯曼知识产权资产管理有限公司 | Compositions and use thereof for the treatment, co-treatment or prevention of inflammatory disorders |
TW201513851A (en) * | 2013-03-27 | 2015-04-16 | Jx Nippon Oil & Energy Corp | Antiinflammatory drugs |
TW201818919A (en) * | 2016-10-25 | 2018-06-01 | 美商阿爾塞斯商業集團國際有限公司 | Compositions of LITHOSPERMUM ERYTHRORHIZON (GROMWELL ROOT) and methods of making and using the compositions |
Non-Patent Citations (1)
Title |
---|
期刊 陳育忠, 薑黃素(curcumin)的臨床應用, 臺灣醫界雜誌, 第63:6期, 中華民國醫師公會全國聯合會, 2020年6月 14-18 * |
Also Published As
Publication number | Publication date |
---|---|
TW202310824A (en) | 2023-03-16 |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
Abd El‐Hack et al. | Curcumin, the active substance of turmeric: its effects on health and ways to improve its bioavailability | |
Kunnumakkara et al. | Is curcumin bioavailability a problem in humans: Lessons from clinical trials | |
Arcusa et al. | Potential role of ginger (Zingiber officinale Roscoe) in the prevention of neurodegenerative diseases | |
Bhat et al. | Benefits of curcumin in brain disorders | |
Imran et al. | Therapeutic application of carvacrol: A comprehensive review | |
Takahashi et al. | Evaluation of an oral carrier system in rats: bioavailability and antioxidant properties of liposome-encapsulated curcumin | |
Gardener et al. | Diet and inflammation in Alzheimer’s disease and related chronic diseases: a review | |
Naeem et al. | Advancement and future directions towards herbal treatment for various diseases | |
JP3790767B2 (en) | Fat metabolism improving composition | |
JP2008542300A (en) | Compositions and methods for preventing and treating conditions associated with inflammation | |
Talebi et al. | Association of Crocus sativus with cognitive dysfunctions and Alzheimer’s disease: a systematic review | |
JP2024059743A (en) | Synergistic liver protective composition | |
Surma et al. | Curcumin-the nutraceutical with pleiotropic effects? Which cardiometabolic subjects might benefit the most? | |
Busari et al. | In vivo Evaluation of Antidiabetic Properties of Seed Oil of Moringa oleifera Lam. | |
Izadi et al. | Longevity and anti-aging effects of curcumin supplementation | |
US20180235904A1 (en) | Cannabidiol compositions including mixtures and uses thereof | |
Pradeep et al. | Synergy among dietary spices in exerting antidiabetic influences | |
TWI810644B (en) | Therapeutic herbal composition | |
Rashwan et al. | Health properties of bioactive food compounds-loaded micro and nano-encapsulation systems: a review | |
Chand | Standardized turmeric and curcumin | |
Tarantul et al. | Turmeric (lat. Curcuma) | |
Divyashree et al. | Curcumin and its derivatives as nutraceuticals: an update | |
WO2021002334A1 (en) | COMPOSITION FOR INHIBITING TNF-α OR IL-6 PRODUCTION | |
Goswami et al. | Review of curcumin and its different formulations: pharmacokinetics, pharmacodynamics and pharmacokinetic-pharmacodynamic interactions | |
Hapuarachchi et al. | Anti-inflammatory potential of aqueous extract and ethyl acetate fractions of Munronia pinnata (Wall) Theob. and the isolated compound, Senecrassidiol |