CN117858630A - Compositions and methods for combating processes associated with inflammation and aging and supporting cellular energy and/or metabolism - Google Patents
Compositions and methods for combating processes associated with inflammation and aging and supporting cellular energy and/or metabolism Download PDFInfo
- Publication number
- CN117858630A CN117858630A CN202280057212.6A CN202280057212A CN117858630A CN 117858630 A CN117858630 A CN 117858630A CN 202280057212 A CN202280057212 A CN 202280057212A CN 117858630 A CN117858630 A CN 117858630A
- Authority
- CN
- China
- Prior art keywords
- extract
- plant material
- composition
- powder
- inflammatory
- Prior art date
- Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
- Pending
Links
- 239000000203 mixture Substances 0.000 title claims abstract description 286
- 238000000034 method Methods 0.000 title claims abstract description 78
- 230000004054 inflammatory process Effects 0.000 title claims abstract description 43
- 206010061218 Inflammation Diseases 0.000 title claims abstract description 42
- 230000032683 aging Effects 0.000 title claims description 17
- 230000008093 supporting effect Effects 0.000 title claims description 6
- 230000001413 cellular effect Effects 0.000 title description 8
- 230000004060 metabolic process Effects 0.000 title description 3
- 230000008569 process Effects 0.000 title description 3
- 230000000770 proinflammatory effect Effects 0.000 claims abstract description 76
- 150000008442 polyphenolic compounds Chemical class 0.000 claims abstract description 62
- 235000013824 polyphenols Nutrition 0.000 claims abstract description 62
- 230000014509 gene expression Effects 0.000 claims abstract description 51
- 108090000623 proteins and genes Proteins 0.000 claims abstract description 42
- 230000008437 mitochondrial biogenesis Effects 0.000 claims abstract description 29
- 230000009758 senescence Effects 0.000 claims abstract description 23
- 235000001412 Mediterranean diet Nutrition 0.000 claims abstract description 16
- 230000002195 synergetic effect Effects 0.000 claims abstract description 12
- 239000000463 material Substances 0.000 claims description 169
- 241000196324 Embryophyta Species 0.000 claims description 123
- 239000000843 powder Substances 0.000 claims description 117
- 210000004027 cell Anatomy 0.000 claims description 108
- 102000004127 Cytokines Human genes 0.000 claims description 78
- 108090000695 Cytokines Proteins 0.000 claims description 78
- 108090001005 Interleukin-6 Proteins 0.000 claims description 58
- 102000004889 Interleukin-6 Human genes 0.000 claims description 58
- 229940100601 interleukin-6 Drugs 0.000 claims description 58
- 239000000284 extract Substances 0.000 claims description 47
- 108060008682 Tumor Necrosis Factor Proteins 0.000 claims description 44
- 102000000852 Tumor Necrosis Factor-alpha Human genes 0.000 claims description 44
- 244000172533 Viola sororia Species 0.000 claims description 41
- 230000001965 increasing effect Effects 0.000 claims description 38
- 102000014777 Adipokines Human genes 0.000 claims description 28
- 108010078606 Adipokines Proteins 0.000 claims description 28
- 239000000478 adipokine Substances 0.000 claims description 28
- 239000003642 reactive oxygen metabolite Substances 0.000 claims description 27
- 230000009467 reduction Effects 0.000 claims description 26
- 208000024891 symptom Diseases 0.000 claims description 26
- WQZGKKKJIJFFOK-GASJEMHNSA-N Glucose Natural products OC[C@H]1OC(O)[C@H](O)[C@@H](O)[C@@H]1O WQZGKKKJIJFFOK-GASJEMHNSA-N 0.000 claims description 23
- 239000008103 glucose Substances 0.000 claims description 23
- 230000004190 glucose uptake Effects 0.000 claims description 22
- 241001464837 Viridiplantae Species 0.000 claims description 21
- 239000003086 colorant Substances 0.000 claims description 21
- 230000011664 signaling Effects 0.000 claims description 21
- 240000007817 Olea europaea Species 0.000 claims description 20
- 235000019216 blueberry extract Nutrition 0.000 claims description 20
- 229940055416 blueberry extract Drugs 0.000 claims description 20
- 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 claims description 20
- 229940038487 grape extract Drugs 0.000 claims description 20
- 229940065115 grapefruit extract Drugs 0.000 claims description 20
- 229940109529 pomegranate extract Drugs 0.000 claims description 20
- 235000021537 Beetroot Nutrition 0.000 claims description 19
- 235000012905 Brassica oleracea var viridis Nutrition 0.000 claims description 19
- 244000000626 Daucus carota Species 0.000 claims description 19
- 235000002767 Daucus carota Nutrition 0.000 claims description 19
- 235000007688 Lycopersicon esculentum Nutrition 0.000 claims description 19
- 235000001537 Ribes X gardonianum Nutrition 0.000 claims description 19
- 235000001535 Ribes X utile Nutrition 0.000 claims description 19
- 235000016919 Ribes petraeum Nutrition 0.000 claims description 19
- 244000281247 Ribes rubrum Species 0.000 claims description 19
- 235000002355 Ribes spicatum Nutrition 0.000 claims description 19
- 241000208829 Sambucus Species 0.000 claims description 19
- 235000018735 Sambucus canadensis Nutrition 0.000 claims description 19
- 241000533293 Sesbania emerus Species 0.000 claims description 19
- 240000003768 Solanum lycopersicum Species 0.000 claims description 19
- 229940069765 bean extract Drugs 0.000 claims description 19
- 235000007123 blue elder Nutrition 0.000 claims description 19
- 235000007124 elderberry Nutrition 0.000 claims description 19
- 235000008995 european elder Nutrition 0.000 claims description 19
- 229940002508 ginger extract Drugs 0.000 claims description 19
- 235000020708 ginger extract Nutrition 0.000 claims description 19
- 229940072113 onion extract Drugs 0.000 claims description 19
- 229940092258 rosemary extract Drugs 0.000 claims description 19
- 235000020748 rosemary extract Nutrition 0.000 claims description 19
- 239000001233 rosmarinus officinalis l. extract Substances 0.000 claims description 19
- 208000027866 inflammatory disease Diseases 0.000 claims description 17
- 230000036542 oxidative stress Effects 0.000 claims description 16
- 230000037149 energy metabolism Effects 0.000 claims description 15
- 229940088594 vitamin Drugs 0.000 claims description 14
- 229930003231 vitamin Natural products 0.000 claims description 14
- 235000013343 vitamin Nutrition 0.000 claims description 14
- 239000011782 vitamin Substances 0.000 claims description 14
- 230000036541 health Effects 0.000 claims description 12
- 208000030159 metabolic disease Diseases 0.000 claims description 12
- 235000016709 nutrition Nutrition 0.000 claims description 12
- 244000064816 Brassica oleracea var. acephala Species 0.000 claims description 10
- 208000024172 Cardiovascular disease Diseases 0.000 claims description 10
- 208000012902 Nervous system disease Diseases 0.000 claims description 10
- 208000025966 Neurological disease Diseases 0.000 claims description 10
- 230000003834 intracellular effect Effects 0.000 claims description 10
- 210000000663 muscle cell Anatomy 0.000 claims description 10
- 230000004792 oxidative damage Effects 0.000 claims description 10
- 240000007124 Brassica oleracea Species 0.000 claims description 9
- 235000003899 Brassica oleracea var acephala Nutrition 0.000 claims description 9
- 210000005260 human cell Anatomy 0.000 claims description 9
- 229910052500 inorganic mineral Inorganic materials 0.000 claims description 9
- 210000000265 leukocyte Anatomy 0.000 claims description 9
- 235000010755 mineral Nutrition 0.000 claims description 9
- 239000011707 mineral Substances 0.000 claims description 9
- 229930027945 nicotinamide-adenine dinucleotide Natural products 0.000 claims description 9
- 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 claims description 8
- 230000036039 immunity Effects 0.000 claims description 8
- 229950006238 nadide Drugs 0.000 claims description 8
- 235000013406 prebiotics Nutrition 0.000 claims description 8
- 229940126634 CD38 inhibitor Drugs 0.000 claims description 7
- VJQALSOBHVEJQM-UHFFFAOYSA-N COCCOC1CCC(CC1)Nc1cc(=O)n(C)c2ccc(cc12)-c1cncs1 Chemical compound COCCOC1CCC(CC1)Nc1cc(=O)n(C)c2ccc(cc12)-c1cncs1 VJQALSOBHVEJQM-UHFFFAOYSA-N 0.000 claims description 7
- JLEBZPBDRKPWTD-TURQNECASA-O N-ribosylnicotinamide Chemical compound NC(=O)C1=CC=C[N+]([C@H]2[C@@H]([C@H](O)[C@@H](CO)O2)O)=C1 JLEBZPBDRKPWTD-TURQNECASA-O 0.000 claims description 7
- DAYLJWODMCOQEW-TURQNECASA-O NMN(+) Chemical compound NC(=O)C1=CC=C[N+]([C@H]2[C@@H]([C@H](O)[C@@H](COP(O)(O)=O)O2)O)=C1 DAYLJWODMCOQEW-TURQNECASA-O 0.000 claims description 7
- PVNIIMVLHYAWGP-UHFFFAOYSA-N Niacin Chemical compound OC(=O)C1=CC=CN=C1 PVNIIMVLHYAWGP-UHFFFAOYSA-N 0.000 claims description 7
- 235000020956 nicotinamide riboside Nutrition 0.000 claims description 7
- 239000011618 nicotinamide riboside Substances 0.000 claims description 7
- 235000001968 nicotinic acid Nutrition 0.000 claims description 7
- 229960003512 nicotinic acid Drugs 0.000 claims description 7
- 239000011664 nicotinic acid Substances 0.000 claims description 7
- 239000006041 probiotic Substances 0.000 claims description 7
- 235000018291 probiotics Nutrition 0.000 claims description 7
- 235000013619 trace mineral Nutrition 0.000 claims description 7
- 239000011573 trace mineral Substances 0.000 claims description 7
- 102100024458 Cyclin-dependent kinase inhibitor 2A Human genes 0.000 claims description 5
- 108010071563 Proto-Oncogene Proteins c-fos Proteins 0.000 claims description 5
- 102000007568 Proto-Oncogene Proteins c-fos Human genes 0.000 claims description 5
- 235000013361 beverage Nutrition 0.000 claims description 4
- 239000002775 capsule Substances 0.000 claims description 4
- 235000009508 confectionery Nutrition 0.000 claims description 4
- 239000011885 synergistic combination Substances 0.000 claims description 4
- 208000001145 Metabolic Syndrome Diseases 0.000 claims description 3
- 201000000690 abdominal obesity-metabolic syndrome Diseases 0.000 claims description 3
- 229940088597 hormone Drugs 0.000 claims description 3
- 239000005556 hormone Substances 0.000 claims description 3
- ZOBPZXTWZATXDG-UHFFFAOYSA-N 1,3-thiazolidine-2,4-dione Chemical compound O=C1CSC(=O)N1 ZOBPZXTWZATXDG-UHFFFAOYSA-N 0.000 claims description 2
- 229940123464 Thiazolidinedione Drugs 0.000 claims description 2
- 102100026019 Interleukin-6 Human genes 0.000 claims 1
- 230000008901 benefit Effects 0.000 abstract description 14
- 230000007423 decrease Effects 0.000 abstract description 13
- 230000001684 chronic effect Effects 0.000 abstract description 12
- 230000019491 signal transduction Effects 0.000 abstract description 8
- 230000004983 pleiotropic effect Effects 0.000 abstract description 3
- 230000006567 cellular energy metabolism Effects 0.000 abstract 1
- 230000002688 persistence Effects 0.000 abstract 1
- 230000000694 effects Effects 0.000 description 77
- 210000001616 monocyte Anatomy 0.000 description 34
- 230000005764 inhibitory process Effects 0.000 description 29
- 239000002158 endotoxin Substances 0.000 description 28
- 229920006008 lipopolysaccharide Polymers 0.000 description 28
- 208000008589 Obesity Diseases 0.000 description 26
- 235000020824 obesity Nutrition 0.000 description 26
- 108010092277 Leptin Proteins 0.000 description 24
- 102000016267 Leptin Human genes 0.000 description 24
- 229940039781 leptin Drugs 0.000 description 24
- NRYBAZVQPHGZNS-ZSOCWYAHSA-N leptin Chemical compound O=C([C@H](CO)NC(=O)[C@H](CC(C)C)NC(=O)[C@H](CC(O)=O)NC(=O)[C@H](CC(C)C)NC(=O)[C@H](CCC(N)=O)NC(=O)[C@H](CC=1C2=CC=CC=C2NC=1)NC(=O)[C@H](CC(C)C)NC(=O)[C@@H](NC(=O)[C@H](CC(O)=O)NC(=O)[C@H](CCC(N)=O)NC(=O)[C@H](CC(C)C)NC(=O)[C@H](CO)NC(=O)CNC(=O)[C@H](CCC(N)=O)NC(=O)[C@@H](N)CC(C)C)CCSC)N1CCC[C@H]1C(=O)NCC(=O)N[C@@H](CS)C(O)=O NRYBAZVQPHGZNS-ZSOCWYAHSA-N 0.000 description 24
- 238000002474 experimental method Methods 0.000 description 23
- GOZMBJCYMQQACI-UHFFFAOYSA-N 6,7-dimethyl-3-[[methyl-[2-[methyl-[[1-[3-(trifluoromethyl)phenyl]indol-3-yl]methyl]amino]ethyl]amino]methyl]chromen-4-one;dihydrochloride Chemical compound Cl.Cl.C=1OC2=CC(C)=C(C)C=C2C(=O)C=1CN(C)CCN(C)CC(C1=CC=CC=C11)=CN1C1=CC=CC(C(F)(F)F)=C1 GOZMBJCYMQQACI-UHFFFAOYSA-N 0.000 description 20
- 206010022489 Insulin Resistance Diseases 0.000 description 20
- 238000012360 testing method Methods 0.000 description 20
- 101000605172 Aspergillus niger (strain CBS 513.88 / FGSC A1513) Probable endopolygalacturonase E Proteins 0.000 description 19
- 101000605171 Aspergillus niger Endopolygalacturonase E Proteins 0.000 description 19
- 101000941281 Bos taurus Gastric triacylglycerol lipase Proteins 0.000 description 19
- 239000004615 ingredient Substances 0.000 description 19
- 241000699670 Mus sp. Species 0.000 description 18
- YASAKCUCGLMORW-UHFFFAOYSA-N Rosiglitazone Chemical compound C=1C=CC=NC=1N(C)CCOC(C=C1)=CC=C1CC1SC(=O)NC1=O YASAKCUCGLMORW-UHFFFAOYSA-N 0.000 description 18
- 235000019197 fats Nutrition 0.000 description 18
- NOESYZHRGYRDHS-UHFFFAOYSA-N insulin Chemical compound N1C(=O)C(NC(=O)C(CCC(N)=O)NC(=O)C(CCC(O)=O)NC(=O)C(C(C)C)NC(=O)C(NC(=O)CN)C(C)CC)CSSCC(C(NC(CO)C(=O)NC(CC(C)C)C(=O)NC(CC=2C=CC(O)=CC=2)C(=O)NC(CCC(N)=O)C(=O)NC(CC(C)C)C(=O)NC(CCC(O)=O)C(=O)NC(CC(N)=O)C(=O)NC(CC=2C=CC(O)=CC=2)C(=O)NC(CSSCC(NC(=O)C(C(C)C)NC(=O)C(CC(C)C)NC(=O)C(CC=2C=CC(O)=CC=2)NC(=O)C(CC(C)C)NC(=O)C(C)NC(=O)C(CCC(O)=O)NC(=O)C(C(C)C)NC(=O)C(CC(C)C)NC(=O)C(CC=2NC=NC=2)NC(=O)C(CO)NC(=O)CNC2=O)C(=O)NCC(=O)NC(CCC(O)=O)C(=O)NC(CCCNC(N)=N)C(=O)NCC(=O)NC(CC=3C=CC=CC=3)C(=O)NC(CC=3C=CC=CC=3)C(=O)NC(CC=3C=CC(O)=CC=3)C(=O)NC(C(C)O)C(=O)N3C(CCC3)C(=O)NC(CCCCN)C(=O)NC(C)C(O)=O)C(=O)NC(CC(N)=O)C(O)=O)=O)NC(=O)C(C(C)CC)NC(=O)C(CO)NC(=O)C(C(C)O)NC(=O)C1CSSCC2NC(=O)C(CC(C)C)NC(=O)C(NC(=O)C(CCC(N)=O)NC(=O)C(CC(N)=O)NC(=O)C(NC(=O)C(N)CC=1C=CC=CC=1)C(C)C)CC1=CN=CN1 NOESYZHRGYRDHS-UHFFFAOYSA-N 0.000 description 18
- 230000002829 reductive effect Effects 0.000 description 18
- 208000001072 type 2 diabetes mellitus Diseases 0.000 description 18
- 102000003973 Fibroblast growth factor 21 Human genes 0.000 description 17
- 108090000376 Fibroblast growth factor 21 Proteins 0.000 description 17
- 108010047909 Resistin Proteins 0.000 description 17
- 210000002540 macrophage Anatomy 0.000 description 17
- 239000006144 Dulbecco’s modified Eagle's medium Substances 0.000 description 16
- 102000007156 Resistin Human genes 0.000 description 16
- 230000003110 anti-inflammatory effect Effects 0.000 description 16
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 description 16
- 108010022752 Acetylcholinesterase Proteins 0.000 description 15
- 102000012440 Acetylcholinesterase Human genes 0.000 description 15
- 229940022698 acetylcholinesterase Drugs 0.000 description 15
- 230000002757 inflammatory effect Effects 0.000 description 15
- 210000001789 adipocyte Anatomy 0.000 description 14
- 235000018102 proteins Nutrition 0.000 description 14
- 102000004169 proteins and genes Human genes 0.000 description 14
- 108090000100 Hepatocyte Growth Factor Proteins 0.000 description 13
- 108090000723 Insulin-Like Growth Factor I Proteins 0.000 description 13
- 108010002350 Interleukin-2 Proteins 0.000 description 13
- 102000000588 Interleukin-2 Human genes 0.000 description 13
- 210000001744 T-lymphocyte Anatomy 0.000 description 13
- 238000010521 absorption reaction Methods 0.000 description 13
- 230000028327 secretion Effects 0.000 description 13
- 108091003079 Bovine Serum Albumin Proteins 0.000 description 12
- 102000003745 Hepatocyte Growth Factor Human genes 0.000 description 12
- 241000699666 Mus <mouse, genus> Species 0.000 description 12
- 239000012091 fetal bovine serum Substances 0.000 description 12
- 239000002609 medium Substances 0.000 description 12
- 239000000047 product Substances 0.000 description 12
- UCSJYZPVAKXKNQ-HZYVHMACSA-N streptomycin Chemical compound CN[C@H]1[C@H](O)[C@@H](O)[C@H](CO)O[C@H]1O[C@@H]1[C@](C=O)(O)[C@H](C)O[C@H]1O[C@@H]1[C@@H](NC(N)=N)[C@H](O)[C@@H](NC(N)=N)[C@H](O)[C@H]1O UCSJYZPVAKXKNQ-HZYVHMACSA-N 0.000 description 12
- 210000000577 adipose tissue Anatomy 0.000 description 11
- 230000015572 biosynthetic process Effects 0.000 description 11
- 102000014429 Insulin-like growth factor Human genes 0.000 description 10
- 102000004140 Oncostatin M Human genes 0.000 description 10
- 108090000630 Oncostatin M Proteins 0.000 description 10
- 208000037976 chronic inflammation Diseases 0.000 description 10
- 208000035475 disorder Diseases 0.000 description 10
- 238000004519 manufacturing process Methods 0.000 description 10
- 102000004877 Insulin Human genes 0.000 description 9
- 108090001061 Insulin Proteins 0.000 description 9
- 102000004372 Insulin-like growth factor binding protein 2 Human genes 0.000 description 9
- 108090000964 Insulin-like growth factor binding protein 2 Proteins 0.000 description 9
- 102000003815 Interleukin-11 Human genes 0.000 description 9
- 108090000177 Interleukin-11 Proteins 0.000 description 9
- 230000004913 activation Effects 0.000 description 9
- 230000006020 chronic inflammation Effects 0.000 description 9
- 150000001875 compounds Chemical class 0.000 description 9
- 235000005911 diet Nutrition 0.000 description 9
- 230000004069 differentiation Effects 0.000 description 9
- 230000006870 function Effects 0.000 description 9
- 229940125396 insulin Drugs 0.000 description 9
- 229940074383 interleukin-11 Drugs 0.000 description 9
- 230000037361 pathway Effects 0.000 description 9
- 229960004586 rosiglitazone Drugs 0.000 description 9
- 238000003786 synthesis reaction Methods 0.000 description 9
- -1 various cytokines Chemical class 0.000 description 9
- 102000004375 Insulin-like growth factor-binding protein 1 Human genes 0.000 description 8
- 108090000957 Insulin-like growth factor-binding protein 1 Proteins 0.000 description 8
- 230000037213 diet Effects 0.000 description 8
- 230000002401 inhibitory effect Effects 0.000 description 8
- 230000001404 mediated effect Effects 0.000 description 8
- 230000004044 response Effects 0.000 description 8
- 102100038246 Retinol-binding protein 4 Human genes 0.000 description 7
- 210000003205 muscle Anatomy 0.000 description 7
- 238000003753 real-time PCR Methods 0.000 description 7
- 102000005962 receptors Human genes 0.000 description 7
- 108020003175 receptors Proteins 0.000 description 7
- 108091032973 (ribonucleotides)n+m Proteins 0.000 description 6
- 102100025668 Angiopoietin-related protein 3 Human genes 0.000 description 6
- 102100021943 C-C motif chemokine 2 Human genes 0.000 description 6
- 108090001007 Interleukin-8 Proteins 0.000 description 6
- 102000004890 Interleukin-8 Human genes 0.000 description 6
- 229930182555 Penicillin Natural products 0.000 description 6
- JGSARLDLIJGVTE-MBNYWOFBSA-N Penicillin G Chemical compound N([C@H]1[C@H]2SC([C@@H](N2C1=O)C(O)=O)(C)C)C(=O)CC1=CC=CC=C1 JGSARLDLIJGVTE-MBNYWOFBSA-N 0.000 description 6
- 239000003963 antioxidant agent Substances 0.000 description 6
- 230000009286 beneficial effect Effects 0.000 description 6
- 238000006243 chemical reaction Methods 0.000 description 6
- 238000011161 development Methods 0.000 description 6
- 230000018109 developmental process Effects 0.000 description 6
- 229960003957 dexamethasone Drugs 0.000 description 6
- UREBDLICKHMUKA-CXSFZGCWSA-N dexamethasone Chemical compound C1CC2=CC(=O)C=C[C@]2(C)[C@]2(F)[C@@H]1[C@@H]1C[C@@H](C)[C@@](C(=O)CO)(O)[C@@]1(C)C[C@@H]2O UREBDLICKHMUKA-CXSFZGCWSA-N 0.000 description 6
- 235000015872 dietary supplement Nutrition 0.000 description 6
- 201000010099 disease Diseases 0.000 description 6
- 210000004185 liver Anatomy 0.000 description 6
- 239000013642 negative control Substances 0.000 description 6
- 229940049954 penicillin Drugs 0.000 description 6
- 230000035790 physiological processes and functions Effects 0.000 description 6
- 239000013641 positive control Substances 0.000 description 6
- 230000001105 regulatory effect Effects 0.000 description 6
- 230000002441 reversible effect Effects 0.000 description 6
- 229960005322 streptomycin Drugs 0.000 description 6
- 101710155857 C-C motif chemokine 2 Proteins 0.000 description 5
- 102000004190 Enzymes Human genes 0.000 description 5
- 108090000790 Enzymes Proteins 0.000 description 5
- 101000693085 Homo sapiens Angiopoietin-related protein 3 Proteins 0.000 description 5
- 102000013264 Interleukin-23 Human genes 0.000 description 5
- 108010065637 Interleukin-23 Proteins 0.000 description 5
- 235000006708 antioxidants Nutrition 0.000 description 5
- 238000003556 assay Methods 0.000 description 5
- 239000012298 atmosphere Substances 0.000 description 5
- 230000004071 biological effect Effects 0.000 description 5
- 239000000090 biomarker Substances 0.000 description 5
- 229960002986 dinoprostone Drugs 0.000 description 5
- 229940088598 enzyme Drugs 0.000 description 5
- 235000009200 high fat diet Nutrition 0.000 description 5
- 230000028993 immune response Effects 0.000 description 5
- 210000004698 lymphocyte Anatomy 0.000 description 5
- 229940094443 oxytocics prostaglandins Drugs 0.000 description 5
- 230000034190 positive regulation of NF-kappaB transcription factor activity Effects 0.000 description 5
- XEYBRNLFEZDVAW-UHFFFAOYSA-N prostaglandin E2 Natural products CCCCCC(O)C=CC1C(O)CC(=O)C1CC=CCCCC(O)=O XEYBRNLFEZDVAW-UHFFFAOYSA-N 0.000 description 5
- 150000003180 prostaglandins Chemical class 0.000 description 5
- 208000024827 Alzheimer disease Diseases 0.000 description 4
- 208000023275 Autoimmune disease Diseases 0.000 description 4
- 238000002965 ELISA Methods 0.000 description 4
- 241000282412 Homo Species 0.000 description 4
- 101000665882 Homo sapiens Retinol-binding protein 4 Proteins 0.000 description 4
- 102000000589 Interleukin-1 Human genes 0.000 description 4
- 108010002352 Interleukin-1 Proteins 0.000 description 4
- 102100038280 Prostaglandin G/H synthase 2 Human genes 0.000 description 4
- 208000038016 acute inflammation Diseases 0.000 description 4
- 230000006022 acute inflammation Effects 0.000 description 4
- 230000003078 antioxidant effect Effects 0.000 description 4
- 230000003115 biocidal effect Effects 0.000 description 4
- 238000004113 cell culture Methods 0.000 description 4
- 230000019522 cellular metabolic process Effects 0.000 description 4
- 239000000544 cholinesterase inhibitor Substances 0.000 description 4
- ADEBPBSSDYVVLD-UHFFFAOYSA-N donepezil Chemical compound O=C1C=2C=C(OC)C(OC)=CC=2CC1CC(CC1)CCN1CC1=CC=CC=C1 ADEBPBSSDYVVLD-UHFFFAOYSA-N 0.000 description 4
- 235000013305 food Nutrition 0.000 description 4
- 238000009472 formulation Methods 0.000 description 4
- 102000006602 glyceraldehyde-3-phosphate dehydrogenase Human genes 0.000 description 4
- 108020004445 glyceraldehyde-3-phosphate dehydrogenase Proteins 0.000 description 4
- 230000007166 healthy aging Effects 0.000 description 4
- 230000001976 improved effect Effects 0.000 description 4
- 239000003112 inhibitor Substances 0.000 description 4
- 239000002417 nutraceutical Substances 0.000 description 4
- 235000021436 nutraceutical agent Nutrition 0.000 description 4
- 230000002018 overexpression Effects 0.000 description 4
- 230000003389 potentiating effect Effects 0.000 description 4
- 230000008741 proinflammatory signaling process Effects 0.000 description 4
- 230000001737 promoting effect Effects 0.000 description 4
- 150000003254 radicals Chemical class 0.000 description 4
- 239000002994 raw material Substances 0.000 description 4
- 239000003488 releasing hormone Substances 0.000 description 4
- 239000000126 substance Substances 0.000 description 4
- 239000006228 supernatant Substances 0.000 description 4
- 239000013589 supplement Substances 0.000 description 4
- 210000001519 tissue Anatomy 0.000 description 4
- 238000011830 transgenic mouse model Methods 0.000 description 4
- 230000035899 viability Effects 0.000 description 4
- XLYOFNOQVPJJNP-UHFFFAOYSA-N water Substances O XLYOFNOQVPJJNP-UHFFFAOYSA-N 0.000 description 4
- QTBSBXVTEAMEQO-UHFFFAOYSA-N Acetic acid Chemical compound CC(O)=O QTBSBXVTEAMEQO-UHFFFAOYSA-N 0.000 description 3
- 102000011690 Adiponectin Human genes 0.000 description 3
- 108010076365 Adiponectin Proteins 0.000 description 3
- 201000001320 Atherosclerosis Diseases 0.000 description 3
- 108010074051 C-Reactive Protein Proteins 0.000 description 3
- 102100032752 C-reactive protein Human genes 0.000 description 3
- 102000019034 Chemokines Human genes 0.000 description 3
- 108010012236 Chemokines Proteins 0.000 description 3
- PHEDXBVPIONUQT-UHFFFAOYSA-N Cocarcinogen A1 Natural products CCCCCCCCCCCCCC(=O)OC1C(C)C2(O)C3C=C(C)C(=O)C3(O)CC(CO)=CC2C2C1(OC(C)=O)C2(C)C PHEDXBVPIONUQT-UHFFFAOYSA-N 0.000 description 3
- 102000018233 Fibroblast Growth Factor Human genes 0.000 description 3
- 108050007372 Fibroblast Growth Factor Proteins 0.000 description 3
- 208000022559 Inflammatory bowel disease Diseases 0.000 description 3
- 102000004218 Insulin-Like Growth Factor I Human genes 0.000 description 3
- 108010064593 Intercellular Adhesion Molecule-1 Proteins 0.000 description 3
- 102000015271 Intercellular Adhesion Molecule-1 Human genes 0.000 description 3
- ZDXPYRJPNDTMRX-VKHMYHEASA-N L-glutamine Chemical compound OC(=O)[C@@H](N)CCC(N)=O ZDXPYRJPNDTMRX-VKHMYHEASA-N 0.000 description 3
- 229930182816 L-glutamine Natural products 0.000 description 3
- 102000004058 Leukemia inhibitory factor Human genes 0.000 description 3
- 108090000581 Leukemia inhibitory factor Proteins 0.000 description 3
- 241001465754 Metazoa Species 0.000 description 3
- 241000699660 Mus musculus Species 0.000 description 3
- 102100023050 Nuclear factor NF-kappa-B p105 subunit Human genes 0.000 description 3
- 108050003267 Prostaglandin G/H synthase 2 Proteins 0.000 description 3
- 101710137011 Retinol-binding protein 4 Proteins 0.000 description 3
- 108091023040 Transcription factor Proteins 0.000 description 3
- 102000040945 Transcription factor Human genes 0.000 description 3
- 238000004458 analytical method Methods 0.000 description 3
- 239000003472 antidiabetic agent Substances 0.000 description 3
- 238000012230 antisense oligonucleotides Methods 0.000 description 3
- 230000033228 biological regulation Effects 0.000 description 3
- 210000004369 blood Anatomy 0.000 description 3
- 239000008280 blood Substances 0.000 description 3
- 230000004663 cell proliferation Effects 0.000 description 3
- 230000003833 cell viability Effects 0.000 description 3
- 230000000295 complement effect Effects 0.000 description 3
- 231100000673 dose–response relationship Toxicity 0.000 description 3
- 239000003814 drug Substances 0.000 description 3
- 210000002950 fibroblast Anatomy 0.000 description 3
- 229940126864 fibroblast growth factor Drugs 0.000 description 3
- 210000002443 helper t lymphocyte Anatomy 0.000 description 3
- 230000001939 inductive effect Effects 0.000 description 3
- 230000003960 inflammatory cascade Effects 0.000 description 3
- 230000004968 inflammatory condition Effects 0.000 description 3
- 102000028416 insulin-like growth factor binding Human genes 0.000 description 3
- 108091022911 insulin-like growth factor binding Proteins 0.000 description 3
- 239000003446 ligand Substances 0.000 description 3
- 150000002632 lipids Chemical class 0.000 description 3
- 108020004999 messenger RNA Proteins 0.000 description 3
- 230000003818 metabolic dysfunction Effects 0.000 description 3
- 230000002093 peripheral effect Effects 0.000 description 3
- PHEDXBVPIONUQT-RGYGYFBISA-N phorbol 13-acetate 12-myristate Chemical compound C([C@]1(O)C(=O)C(C)=C[C@H]1[C@@]1(O)[C@H](C)[C@H]2OC(=O)CCCCCCCCCCCCC)C(CO)=C[C@H]1[C@H]1[C@]2(OC(C)=O)C1(C)C PHEDXBVPIONUQT-RGYGYFBISA-N 0.000 description 3
- 235000020777 polyunsaturated fatty acids Nutrition 0.000 description 3
- 206010039073 rheumatoid arthritis Diseases 0.000 description 3
- 239000000523 sample Substances 0.000 description 3
- 239000000243 solution Substances 0.000 description 3
- 230000000638 stimulation Effects 0.000 description 3
- 238000013518 transcription Methods 0.000 description 3
- 230000035897 transcription Effects 0.000 description 3
- 231100000582 ATP assay Toxicity 0.000 description 2
- CIWBSHSKHKDKBQ-JLAZNSOCSA-N Ascorbic acid Chemical compound OC[C@H](O)[C@H]1OC(=O)C(O)=C1O CIWBSHSKHKDKBQ-JLAZNSOCSA-N 0.000 description 2
- FERIUCNNQQJTOY-UHFFFAOYSA-N Butyric acid Chemical compound CCCC(O)=O FERIUCNNQQJTOY-UHFFFAOYSA-N 0.000 description 2
- 102000001902 CC Chemokines Human genes 0.000 description 2
- 108010040471 CC Chemokines Proteins 0.000 description 2
- 101150071146 COX2 gene Proteins 0.000 description 2
- 101100114534 Caenorhabditis elegans ctc-2 gene Proteins 0.000 description 2
- 108091008794 FGF receptors Proteins 0.000 description 2
- 102000044168 Fibroblast Growth Factor Receptor Human genes 0.000 description 2
- RFSUNEUAIZKAJO-ARQDHWQXSA-N Fructose Chemical class OC[C@H]1O[C@](O)(CO)[C@@H](O)[C@@H]1O RFSUNEUAIZKAJO-ARQDHWQXSA-N 0.000 description 2
- 208000007882 Gastritis Diseases 0.000 description 2
- MHAJPDPJQMAIIY-UHFFFAOYSA-N Hydrogen peroxide Chemical compound OO MHAJPDPJQMAIIY-UHFFFAOYSA-N 0.000 description 2
- 102100034343 Integrase Human genes 0.000 description 2
- 108010019437 Janus Kinase 2 Proteins 0.000 description 2
- 108010051335 Lipocalin-2 Proteins 0.000 description 2
- 102000013519 Lipocalin-2 Human genes 0.000 description 2
- 102000004895 Lipoproteins Human genes 0.000 description 2
- 108060001084 Luciferase Proteins 0.000 description 2
- 239000005089 Luciferase Substances 0.000 description 2
- 102000009571 Macrophage Inflammatory Proteins Human genes 0.000 description 2
- 108010009474 Macrophage Inflammatory Proteins Proteins 0.000 description 2
- 241000124008 Mammalia Species 0.000 description 2
- MWUXSHHQAYIFBG-UHFFFAOYSA-N Nitric oxide Chemical compound O=[N] MWUXSHHQAYIFBG-UHFFFAOYSA-N 0.000 description 2
- 102100029438 Nitric oxide synthase, inducible Human genes 0.000 description 2
- 101710089543 Nitric oxide synthase, inducible Proteins 0.000 description 2
- 108091034117 Oligonucleotide Proteins 0.000 description 2
- 101150000187 PTGS2 gene Proteins 0.000 description 2
- 108010026552 Proteome Proteins 0.000 description 2
- 206010037211 Psychomotor hyperactivity Diseases 0.000 description 2
- 108010092799 RNA-directed DNA polymerase Proteins 0.000 description 2
- 238000011529 RT qPCR Methods 0.000 description 2
- 102100024735 Resistin Human genes 0.000 description 2
- VYPSYNLAJGMNEJ-UHFFFAOYSA-N Silicium dioxide Chemical compound O=[Si]=O VYPSYNLAJGMNEJ-UHFFFAOYSA-N 0.000 description 2
- 108091008874 T cell receptors Proteins 0.000 description 2
- 102000016266 T-Cell Antigen Receptors Human genes 0.000 description 2
- GLEVLJDDWXEYCO-UHFFFAOYSA-N Trolox Chemical compound O1C(C)(C(O)=O)CCC2=C1C(C)=C(C)C(O)=C2C GLEVLJDDWXEYCO-UHFFFAOYSA-N 0.000 description 2
- 102100033444 Tyrosine-protein kinase JAK2 Human genes 0.000 description 2
- 108010000134 Vascular Cell Adhesion Molecule-1 Proteins 0.000 description 2
- 241000700605 Viruses Species 0.000 description 2
- XJLXINKUBYWONI-DQQFMEOOSA-N [[(2r,3r,4r,5r)-5-(6-aminopurin-9-yl)-3-hydroxy-4-phosphonooxyoxolan-2-yl]methoxy-hydroxyphosphoryl] [(2s,3r,4s,5s)-5-(3-carbamoylpyridin-1-ium-1-yl)-3,4-dihydroxyoxolan-2-yl]methyl phosphate 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-DQQFMEOOSA-N 0.000 description 2
- 230000002159 abnormal effect Effects 0.000 description 2
- 238000009825 accumulation Methods 0.000 description 2
- OIPILFWXSMYKGL-UHFFFAOYSA-N acetylcholine Chemical compound CC(=O)OCC[N+](C)(C)C OIPILFWXSMYKGL-UHFFFAOYSA-N 0.000 description 2
- 230000009471 action Effects 0.000 description 2
- 230000001154 acute effect Effects 0.000 description 2
- FPIPGXGPPPQFEQ-OVSJKPMPSA-N all-trans-retinol Chemical compound OC\C=C(/C)\C=C\C=C(/C)\C=C\C1=C(C)CCCC1(C)C FPIPGXGPPPQFEQ-OVSJKPMPSA-N 0.000 description 2
- 230000003321 amplification Effects 0.000 description 2
- 239000003242 anti bacterial agent Substances 0.000 description 2
- 230000003178 anti-diabetic effect Effects 0.000 description 2
- 229940121363 anti-inflammatory agent Drugs 0.000 description 2
- 239000002260 anti-inflammatory agent Substances 0.000 description 2
- 229940088710 antibiotic agent Drugs 0.000 description 2
- 239000000074 antisense oligonucleotide Substances 0.000 description 2
- 238000013459 approach Methods 0.000 description 2
- 239000006286 aqueous extract Substances 0.000 description 2
- YZXBAPSDXZZRGB-DOFZRALJSA-N arachidonic acid Chemical compound CCCCC\C=C/C\C=C/C\C=C/C\C=C/CCCC(O)=O YZXBAPSDXZZRGB-DOFZRALJSA-N 0.000 description 2
- 239000012131 assay buffer Substances 0.000 description 2
- 208000006673 asthma Diseases 0.000 description 2
- 210000003719 b-lymphocyte Anatomy 0.000 description 2
- 230000001580 bacterial effect Effects 0.000 description 2
- WQZGKKKJIJFFOK-VFUOTHLCSA-N beta-D-glucose Chemical compound OC[C@H]1O[C@@H](O)[C@H](O)[C@@H](O)[C@@H]1O WQZGKKKJIJFFOK-VFUOTHLCSA-N 0.000 description 2
- 230000000975 bioactive effect Effects 0.000 description 2
- 230000036772 blood pressure Effects 0.000 description 2
- 230000004098 cellular respiration Effects 0.000 description 2
- 150000005829 chemical entities Chemical class 0.000 description 2
- 230000019771 cognition Effects 0.000 description 2
- 230000000052 comparative effect Effects 0.000 description 2
- 239000002299 complementary DNA Substances 0.000 description 2
- 210000004748 cultured cell Anatomy 0.000 description 2
- 230000006378 damage Effects 0.000 description 2
- 230000007812 deficiency Effects 0.000 description 2
- 230000001419 dependent effect Effects 0.000 description 2
- 238000001514 detection method Methods 0.000 description 2
- 229960003530 donepezil Drugs 0.000 description 2
- VYFYYTLLBUKUHU-UHFFFAOYSA-N dopamine Chemical compound NCCC1=CC=C(O)C(O)=C1 VYFYYTLLBUKUHU-UHFFFAOYSA-N 0.000 description 2
- 230000003828 downregulation Effects 0.000 description 2
- 229940079593 drug Drugs 0.000 description 2
- 230000002900 effect on cell Effects 0.000 description 2
- 230000002500 effect on skin Effects 0.000 description 2
- 230000002124 endocrine Effects 0.000 description 2
- 210000002889 endothelial cell Anatomy 0.000 description 2
- 230000002708 enhancing effect Effects 0.000 description 2
- 231100000655 enterotoxin Toxicity 0.000 description 2
- 235000011389 fruit/vegetable juice Nutrition 0.000 description 2
- 230000012010 growth Effects 0.000 description 2
- 210000003494 hepatocyte Anatomy 0.000 description 2
- 238000004128 high performance liquid chromatography Methods 0.000 description 2
- 210000003630 histaminocyte Anatomy 0.000 description 2
- JYGXADMDTFJGBT-VWUMJDOOSA-N hydrocortisone Chemical compound O=C1CC[C@]2(C)[C@H]3[C@@H](O)C[C@](C)([C@@](CC4)(O)C(=O)CO)[C@@H]4[C@@H]3CCC2=C1 JYGXADMDTFJGBT-VWUMJDOOSA-N 0.000 description 2
- 230000036737 immune function Effects 0.000 description 2
- 230000006872 improvement Effects 0.000 description 2
- 238000001727 in vivo Methods 0.000 description 2
- CGIGDMFJXJATDK-UHFFFAOYSA-N indomethacin Chemical compound CC1=C(CC(O)=O)C2=CC(OC)=CC=C2N1C(=O)C1=CC=C(Cl)C=C1 CGIGDMFJXJATDK-UHFFFAOYSA-N 0.000 description 2
- 208000015181 infectious disease Diseases 0.000 description 2
- 238000002347 injection Methods 0.000 description 2
- 239000007924 injection Substances 0.000 description 2
- 208000032839 leukemia Diseases 0.000 description 2
- 239000012263 liquid product Substances 0.000 description 2
- 239000003550 marker Substances 0.000 description 2
- 230000004898 mitochondrial function Effects 0.000 description 2
- 230000004048 modification Effects 0.000 description 2
- 238000012986 modification Methods 0.000 description 2
- 230000009456 molecular mechanism Effects 0.000 description 2
- 238000010606 normalization Methods 0.000 description 2
- 238000003199 nucleic acid amplification method Methods 0.000 description 2
- 235000015097 nutrients Nutrition 0.000 description 2
- 229920001542 oligosaccharide Polymers 0.000 description 2
- 150000002482 oligosaccharides Chemical class 0.000 description 2
- 230000036407 pain Effects 0.000 description 2
- 150000002989 phenols Chemical class 0.000 description 2
- 230000026731 phosphorylation Effects 0.000 description 2
- 238000006366 phosphorylation reaction Methods 0.000 description 2
- 230000001766 physiological effect Effects 0.000 description 2
- 210000000229 preadipocyte Anatomy 0.000 description 2
- 210000004986 primary T-cell Anatomy 0.000 description 2
- 230000002035 prolonged effect Effects 0.000 description 2
- 238000011002 quantification Methods 0.000 description 2
- 239000003161 ribonuclease inhibitor Substances 0.000 description 2
- 210000002966 serum Anatomy 0.000 description 2
- 210000002027 skeletal muscle Anatomy 0.000 description 2
- DAEPDZWVDSPTHF-UHFFFAOYSA-M sodium pyruvate Chemical compound [Na+].CC(=O)C([O-])=O DAEPDZWVDSPTHF-UHFFFAOYSA-M 0.000 description 2
- 239000007787 solid Substances 0.000 description 2
- 239000012265 solid product Substances 0.000 description 2
- 241000894007 species Species 0.000 description 2
- 230000009885 systemic effect Effects 0.000 description 2
- XOAAWQZATWQOTB-UHFFFAOYSA-N taurine Chemical compound NCCS(O)(=O)=O XOAAWQZATWQOTB-UHFFFAOYSA-N 0.000 description 2
- 230000001225 therapeutic effect Effects 0.000 description 2
- 230000002103 transcriptional effect Effects 0.000 description 2
- 210000003556 vascular endothelial cell Anatomy 0.000 description 2
- 150000003722 vitamin derivatives Chemical class 0.000 description 2
- QUTFFEUUGHUPQC-ILWYWAAHSA-N (2r,3r,4s,5r)-3,4,5,6-tetrahydroxy-2-[(4-nitro-2,1,3-benzoxadiazol-7-yl)amino]hexanal Chemical compound OC[C@@H](O)[C@@H](O)[C@H](O)[C@H](C=O)NC1=CC=C([N+]([O-])=O)C2=NON=C12 QUTFFEUUGHUPQC-ILWYWAAHSA-N 0.000 description 1
- RUDATBOHQWOJDD-UHFFFAOYSA-N (3beta,5beta,7alpha)-3,7-Dihydroxycholan-24-oic acid Natural products OC1CC2CC(O)CCC2(C)C2C1C1CCC(C(CCC(O)=O)C)C1(C)CC2 RUDATBOHQWOJDD-UHFFFAOYSA-N 0.000 description 1
- RSINTYZGAWHRBE-UHFFFAOYSA-N 1,3-thiazole-4,5-dione Chemical compound O=C1SC=NC1=O RSINTYZGAWHRBE-UHFFFAOYSA-N 0.000 description 1
- FPIPGXGPPPQFEQ-UHFFFAOYSA-N 13-cis retinol Natural products OCC=C(C)C=CC=C(C)C=CC1=C(C)CCCC1(C)C FPIPGXGPPPQFEQ-UHFFFAOYSA-N 0.000 description 1
- 101710154545 16 kDa protein Proteins 0.000 description 1
- RUVJFMSQTCEAAB-UHFFFAOYSA-M 2-[3-[5,6-dichloro-1,3-bis[[4-(chloromethyl)phenyl]methyl]benzimidazol-2-ylidene]prop-1-enyl]-3-methyl-1,3-benzoxazol-3-ium;chloride Chemical compound [Cl-].O1C2=CC=CC=C2[N+](C)=C1C=CC=C(N(C1=CC(Cl)=C(Cl)C=C11)CC=2C=CC(CCl)=CC=2)N1CC1=CC=C(CCl)C=C1 RUVJFMSQTCEAAB-UHFFFAOYSA-M 0.000 description 1
- FAWLNURBQMTKEB-URDPEVQOSA-N 213546-53-3 Chemical compound N([C@@H](C)C(=O)N[C@H](C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](C)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](C)C(=O)N1CCC[C@H]1C(=O)N[C@H](C(=O)N[C@@H](CCC(N)=O)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CCCCN)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CCC(N)=O)C(=O)N[C@@H](CCCCN)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CCSC)C(=O)N1[C@@H](CCC1)C(O)=O)C(C)C)C(C)C)C(=O)[C@@H]1CCCN1C(=O)[C@H](CC(C)C)NC(=O)[C@H](CC(C)C)NC(=O)[C@H](C)NC(=O)[C@@H](NC(=O)[C@H](C)NC(=O)[C@H](C)N)C(C)C FAWLNURBQMTKEB-URDPEVQOSA-N 0.000 description 1
- APIXJSLKIYYUKG-UHFFFAOYSA-N 3 Isobutyl 1 methylxanthine Chemical compound O=C1N(C)C(=O)N(CC(C)C)C2=C1N=CN2 APIXJSLKIYYUKG-UHFFFAOYSA-N 0.000 description 1
- 208000030507 AIDS Diseases 0.000 description 1
- 206010048998 Acute phase reaction Diseases 0.000 description 1
- 102100025672 Angiopoietin-related protein 2 Human genes 0.000 description 1
- 101710085851 Angiopoietin-related protein 2 Proteins 0.000 description 1
- 101710085848 Angiopoietin-related protein 3 Proteins 0.000 description 1
- 206010002556 Ankylosing Spondylitis Diseases 0.000 description 1
- BSYNRYMUTXBXSQ-UHFFFAOYSA-N Aspirin Chemical compound CC(=O)OC1=CC=CC=C1C(O)=O BSYNRYMUTXBXSQ-UHFFFAOYSA-N 0.000 description 1
- 241000894006 Bacteria Species 0.000 description 1
- 102100020683 Beta-klotho Human genes 0.000 description 1
- 101710104526 Beta-klotho Proteins 0.000 description 1
- 241000186000 Bifidobacterium Species 0.000 description 1
- 206010005033 Bladder dilatation Diseases 0.000 description 1
- 108050006947 CXC Chemokine Proteins 0.000 description 1
- 102000019388 CXC chemokine Human genes 0.000 description 1
- 101100205088 Caenorhabditis elegans iars-1 gene Proteins 0.000 description 1
- 241000282472 Canis lupus familiaris Species 0.000 description 1
- 102000014914 Carrier Proteins Human genes 0.000 description 1
- 102000000844 Cell Surface Receptors Human genes 0.000 description 1
- 108010001857 Cell Surface Receptors Proteins 0.000 description 1
- 108010055166 Chemokine CCL5 Proteins 0.000 description 1
- 102000001327 Chemokine CCL5 Human genes 0.000 description 1
- 229920002261 Corn starch Polymers 0.000 description 1
- 108010037462 Cyclooxygenase 2 Proteins 0.000 description 1
- ZZZCUOFIHGPKAK-UHFFFAOYSA-N D-erythro-ascorbic acid Natural products OCC1OC(=O)C(O)=C1O ZZZCUOFIHGPKAK-UHFFFAOYSA-N 0.000 description 1
- MYMOFIZGZYHOMD-UHFFFAOYSA-N Dioxygen Chemical class O=O MYMOFIZGZYHOMD-UHFFFAOYSA-N 0.000 description 1
- 102100025012 Dipeptidyl peptidase 4 Human genes 0.000 description 1
- 206010061818 Disease progression Diseases 0.000 description 1
- 208000016546 Distal 16p11.2 microdeletion syndrome Diseases 0.000 description 1
- 101100393884 Drosophila melanogaster Glut1 gene Proteins 0.000 description 1
- 101710146739 Enterotoxin Proteins 0.000 description 1
- 101710204425 Enterotoxin type B Proteins 0.000 description 1
- 241000283086 Equidae Species 0.000 description 1
- 101000867232 Escherichia coli Heat-stable enterotoxin II Proteins 0.000 description 1
- 108010008165 Etanercept Proteins 0.000 description 1
- 241000282326 Felis catus Species 0.000 description 1
- 102100024802 Fibroblast growth factor 23 Human genes 0.000 description 1
- 102100021066 Fibroblast growth factor receptor substrate 2 Human genes 0.000 description 1
- CITFYDYEWQIEPX-UHFFFAOYSA-N Flavanol Natural products O1C2=CC(OCC=C(C)C)=CC(O)=C2C(=O)C(O)C1C1=CC=C(O)C=C1 CITFYDYEWQIEPX-UHFFFAOYSA-N 0.000 description 1
- 208000010412 Glaucoma Diseases 0.000 description 1
- 208000002705 Glucose Intolerance Diseases 0.000 description 1
- 206010018429 Glucose tolerance impaired Diseases 0.000 description 1
- 241000590002 Helicobacter pylori Species 0.000 description 1
- 101100120063 Homo sapiens FGF21 gene Proteins 0.000 description 1
- 101001051973 Homo sapiens Fibroblast growth factor 23 Proteins 0.000 description 1
- 101000818410 Homo sapiens Fibroblast growth factor receptor substrate 2 Proteins 0.000 description 1
- 101000599951 Homo sapiens Insulin-like growth factor I Proteins 0.000 description 1
- 101001081567 Homo sapiens Insulin-like growth factor-binding protein 1 Proteins 0.000 description 1
- 101001044940 Homo sapiens Insulin-like growth factor-binding protein 2 Proteins 0.000 description 1
- 101000979342 Homo sapiens Nuclear factor NF-kappa-B p105 subunit Proteins 0.000 description 1
- 101000586302 Homo sapiens Oncostatin-M-specific receptor subunit beta Proteins 0.000 description 1
- 101000684208 Homo sapiens Prolyl endopeptidase FAP Proteins 0.000 description 1
- 101000686909 Homo sapiens Resistin Proteins 0.000 description 1
- 208000031226 Hyperlipidaemia Diseases 0.000 description 1
- 206010020710 Hyperphagia Diseases 0.000 description 1
- 206010020751 Hypersensitivity Diseases 0.000 description 1
- 206010020772 Hypertension Diseases 0.000 description 1
- 208000013016 Hypoglycemia Diseases 0.000 description 1
- 101150030450 IRS1 gene Proteins 0.000 description 1
- 101150000070 Igfbp1 gene Proteins 0.000 description 1
- 208000015580 Increased body weight Diseases 0.000 description 1
- 235000000177 Indigofera tinctoria Nutrition 0.000 description 1
- 108010034219 Insulin Receptor Substrate Proteins Proteins 0.000 description 1
- 102100025087 Insulin receptor substrate 1 Human genes 0.000 description 1
- 102000048143 Insulin-Like Growth Factor II Human genes 0.000 description 1
- 108090001117 Insulin-Like Growth Factor II Proteins 0.000 description 1
- 102100037852 Insulin-like growth factor I Human genes 0.000 description 1
- 102000004374 Insulin-like growth factor binding protein 3 Human genes 0.000 description 1
- 108090000965 Insulin-like growth factor binding protein 3 Proteins 0.000 description 1
- 102000004371 Insulin-like growth factor binding protein 5 Human genes 0.000 description 1
- 108090000961 Insulin-like growth factor binding protein 5 Proteins 0.000 description 1
- 102100027636 Insulin-like growth factor-binding protein 1 Human genes 0.000 description 1
- 102000004883 Insulin-like growth factor-binding protein 6 Human genes 0.000 description 1
- 108090001014 Insulin-like growth factor-binding protein 6 Proteins 0.000 description 1
- 102000003814 Interleukin-10 Human genes 0.000 description 1
- 108090000174 Interleukin-10 Proteins 0.000 description 1
- 102000003810 Interleukin-18 Human genes 0.000 description 1
- 108090000171 Interleukin-18 Proteins 0.000 description 1
- 108090000978 Interleukin-4 Proteins 0.000 description 1
- 102000004310 Ion Channels Human genes 0.000 description 1
- 108090000862 Ion Channels Proteins 0.000 description 1
- 241000186660 Lactobacillus Species 0.000 description 1
- 206010049287 Lipodystrophy acquired Diseases 0.000 description 1
- 108010013563 Lipoprotein Lipase Proteins 0.000 description 1
- 108090001030 Lipoproteins Proteins 0.000 description 1
- 102000043136 MAP kinase family Human genes 0.000 description 1
- 108091054455 MAP kinase family Proteins 0.000 description 1
- 108010046938 Macrophage Colony-Stimulating Factor Proteins 0.000 description 1
- 102100028123 Macrophage colony-stimulating factor 1 Human genes 0.000 description 1
- 101710091439 Major capsid protein 1 Proteins 0.000 description 1
- 102000002274 Matrix Metalloproteinases Human genes 0.000 description 1
- 108010000684 Matrix Metalloproteinases Proteins 0.000 description 1
- 102100039364 Metalloproteinase inhibitor 1 Human genes 0.000 description 1
- 101100390675 Mus musculus Fgf15 gene Proteins 0.000 description 1
- 102000008934 Muscle Proteins Human genes 0.000 description 1
- 108010074084 Muscle Proteins Proteins 0.000 description 1
- 101001055320 Myxine glutinosa Insulin-like growth factor Proteins 0.000 description 1
- 108010002998 NADPH Oxidases Proteins 0.000 description 1
- 102000004722 NADPH Oxidases Human genes 0.000 description 1
- 102000015532 Nicotinamide phosphoribosyltransferase Human genes 0.000 description 1
- 108010064862 Nicotinamide phosphoribosyltransferase Proteins 0.000 description 1
- 239000006057 Non-nutritive feed additive Substances 0.000 description 1
- 239000012124 Opti-MEM Substances 0.000 description 1
- 206010033307 Overweight Diseases 0.000 description 1
- 102100036201 Oxygen-dependent coproporphyrinogen-III oxidase, mitochondrial Human genes 0.000 description 1
- 101150053131 PTGER3 gene Proteins 0.000 description 1
- 229940080774 Peroxisome proliferator-activated receptor gamma agonist Drugs 0.000 description 1
- 108010022233 Plasminogen Activator Inhibitor 1 Proteins 0.000 description 1
- 102100039418 Plasminogen activator inhibitor 1 Human genes 0.000 description 1
- 102100038277 Prostaglandin G/H synthase 1 Human genes 0.000 description 1
- 108050003243 Prostaglandin G/H synthase 1 Proteins 0.000 description 1
- 102000004005 Prostaglandin-endoperoxide synthases Human genes 0.000 description 1
- 108090000459 Prostaglandin-endoperoxide synthases Proteins 0.000 description 1
- 102000003923 Protein Kinase C Human genes 0.000 description 1
- 108090000315 Protein Kinase C Proteins 0.000 description 1
- 102100036467 Protein delta homolog 1 Human genes 0.000 description 1
- 101710119301 Protein delta homolog 1 Proteins 0.000 description 1
- 102000000850 Proto-Oncogene Proteins c-rel Human genes 0.000 description 1
- 108010001859 Proto-Oncogene Proteins c-rel Proteins 0.000 description 1
- 201000004681 Psoriasis Diseases 0.000 description 1
- 201000001263 Psoriatic Arthritis Diseases 0.000 description 1
- 208000036824 Psoriatic arthropathy Diseases 0.000 description 1
- 101150109738 Ptger4 gene Proteins 0.000 description 1
- 206010037660 Pyrexia Diseases 0.000 description 1
- 238000002123 RNA extraction Methods 0.000 description 1
- 108700008625 Reporter Genes Proteins 0.000 description 1
- 108091027981 Response element Proteins 0.000 description 1
- 241000283984 Rodentia Species 0.000 description 1
- 101150058068 SLC2A1 gene Proteins 0.000 description 1
- 108091006300 SLC2A4 Proteins 0.000 description 1
- 241000235070 Saccharomyces Species 0.000 description 1
- 206010070834 Sensitisation Diseases 0.000 description 1
- 102100038803 Somatotropin Human genes 0.000 description 1
- 108050007673 Somatotropin Proteins 0.000 description 1
- 241000191967 Staphylococcus aureus Species 0.000 description 1
- OUUQCZGPVNCOIJ-UHFFFAOYSA-M Superoxide Chemical compound [O-][O] OUUQCZGPVNCOIJ-UHFFFAOYSA-M 0.000 description 1
- 206010051379 Systemic Inflammatory Response Syndrome Diseases 0.000 description 1
- 210000000447 Th1 cell Anatomy 0.000 description 1
- 244000269722 Thea sinensis Species 0.000 description 1
- 108010031374 Tissue Inhibitor of Metalloproteinase-1 Proteins 0.000 description 1
- 102000004357 Transferases Human genes 0.000 description 1
- 108090000992 Transferases Proteins 0.000 description 1
- 108700019146 Transgenes Proteins 0.000 description 1
- 108010019530 Vascular Endothelial Growth Factors Proteins 0.000 description 1
- 102000005789 Vascular Endothelial Growth Factors Human genes 0.000 description 1
- 102100023543 Vascular cell adhesion protein 1 Human genes 0.000 description 1
- 208000036142 Viral infection Diseases 0.000 description 1
- 229930003268 Vitamin C Natural products 0.000 description 1
- 208000027418 Wounds and injury Diseases 0.000 description 1
- 238000002835 absorbance Methods 0.000 description 1
- 229960004373 acetylcholine Drugs 0.000 description 1
- 229960001138 acetylsalicylic acid Drugs 0.000 description 1
- 239000002253 acid Substances 0.000 description 1
- 230000003213 activating effect Effects 0.000 description 1
- 239000012190 activator Substances 0.000 description 1
- 230000004658 acute-phase response Effects 0.000 description 1
- 229960002964 adalimumab Drugs 0.000 description 1
- 230000033289 adaptive immune response Effects 0.000 description 1
- 210000000593 adipose tissue white Anatomy 0.000 description 1
- 239000000556 agonist Substances 0.000 description 1
- 210000001552 airway epithelial cell Anatomy 0.000 description 1
- 239000012675 alcoholic extract Substances 0.000 description 1
- 208000026935 allergic disease Diseases 0.000 description 1
- 230000007815 allergy Effects 0.000 description 1
- 108010075843 alpha-2-HS-Glycoprotein Proteins 0.000 description 1
- 102000012005 alpha-2-HS-Glycoprotein Human genes 0.000 description 1
- 239000012491 analyte Substances 0.000 description 1
- 238000010171 animal model Methods 0.000 description 1
- 208000022531 anorexia Diseases 0.000 description 1
- 230000003042 antagnostic effect Effects 0.000 description 1
- 239000005557 antagonist Substances 0.000 description 1
- 235000010208 anthocyanin Nutrition 0.000 description 1
- 229930002877 anthocyanin Natural products 0.000 description 1
- 239000004410 anthocyanin Substances 0.000 description 1
- 150000004636 anthocyanins Chemical class 0.000 description 1
- 230000003712 anti-aging effect Effects 0.000 description 1
- 229940124599 anti-inflammatory drug Drugs 0.000 description 1
- 238000009175 antibody therapy Methods 0.000 description 1
- 229940125708 antidiabetic agent Drugs 0.000 description 1
- 239000000427 antigen Substances 0.000 description 1
- 108091007433 antigens Proteins 0.000 description 1
- 102000036639 antigens Human genes 0.000 description 1
- 229940114079 arachidonic acid Drugs 0.000 description 1
- 235000021342 arachidonic acid Nutrition 0.000 description 1
- 238000003149 assay kit Methods 0.000 description 1
- QVGXLLKOCUKJST-UHFFFAOYSA-N atomic oxygen Chemical compound [O] QVGXLLKOCUKJST-UHFFFAOYSA-N 0.000 description 1
- 230000003305 autocrine Effects 0.000 description 1
- 230000006399 behavior Effects 0.000 description 1
- 239000003613 bile acid Substances 0.000 description 1
- 108091008324 binding proteins Proteins 0.000 description 1
- 230000008827 biological function Effects 0.000 description 1
- 230000031018 biological processes and functions Effects 0.000 description 1
- 230000005540 biological transmission Effects 0.000 description 1
- 238000001574 biopsy Methods 0.000 description 1
- 238000001815 biotherapy Methods 0.000 description 1
- 239000006227 byproduct Substances 0.000 description 1
- 235000014171 carbonated beverage Nutrition 0.000 description 1
- 230000000747 cardiac effect Effects 0.000 description 1
- 210000004413 cardiac myocyte Anatomy 0.000 description 1
- 230000005961 cardioprotection Effects 0.000 description 1
- 230000020411 cell activation Effects 0.000 description 1
- 230000024245 cell differentiation Effects 0.000 description 1
- 230000012292 cell migration Effects 0.000 description 1
- 230000008727 cellular glucose uptake Effects 0.000 description 1
- 230000004635 cellular health Effects 0.000 description 1
- 230000036755 cellular response Effects 0.000 description 1
- 230000004637 cellular stress Effects 0.000 description 1
- 210000003169 central nervous system Anatomy 0.000 description 1
- 235000013339 cereals Nutrition 0.000 description 1
- 239000003795 chemical substances by application Substances 0.000 description 1
- 229940112822 chewing gum Drugs 0.000 description 1
- 235000015218 chewing gum Nutrition 0.000 description 1
- 235000001368 chlorogenic acid Nutrition 0.000 description 1
- OEYIOHPDSNJKLS-UHFFFAOYSA-N choline Chemical compound C[N+](C)(C)CCO OEYIOHPDSNJKLS-UHFFFAOYSA-N 0.000 description 1
- 229960001231 choline Drugs 0.000 description 1
- 210000002932 cholinergic neuron Anatomy 0.000 description 1
- 208000037893 chronic inflammatory disorder Diseases 0.000 description 1
- 230000012085 chronic inflammatory response Effects 0.000 description 1
- 238000010367 cloning Methods 0.000 description 1
- 238000007398 colorimetric assay Methods 0.000 description 1
- 239000012141 concentrate Substances 0.000 description 1
- 208000012696 congenital leptin deficiency Diseases 0.000 description 1
- 239000013068 control sample Substances 0.000 description 1
- 230000001276 controlling effect Effects 0.000 description 1
- 239000008120 corn starch Substances 0.000 description 1
- 230000002596 correlated effect Effects 0.000 description 1
- 239000006071 cream Substances 0.000 description 1
- XUJNEKJLAYXESH-UHFFFAOYSA-N cysteine Natural products SCC(N)C(O)=O XUJNEKJLAYXESH-UHFFFAOYSA-N 0.000 description 1
- 235000018417 cysteine Nutrition 0.000 description 1
- 238000002784 cytotoxicity assay Methods 0.000 description 1
- 231100000263 cytotoxicity test Toxicity 0.000 description 1
- 235000013365 dairy product Nutrition 0.000 description 1
- 206010061428 decreased appetite Diseases 0.000 description 1
- 230000032459 dedifferentiation Effects 0.000 description 1
- 230000002950 deficient Effects 0.000 description 1
- 206010012601 diabetes mellitus Diseases 0.000 description 1
- 230000000378 dietary effect Effects 0.000 description 1
- 235000014113 dietary fatty acids Nutrition 0.000 description 1
- 235000013325 dietary fiber Nutrition 0.000 description 1
- 235000018823 dietary intake Nutrition 0.000 description 1
- 230000029087 digestion Effects 0.000 description 1
- 230000001079 digestive effect Effects 0.000 description 1
- 230000005750 disease progression Effects 0.000 description 1
- 229960003638 dopamine Drugs 0.000 description 1
- 239000003937 drug carrier Substances 0.000 description 1
- 230000004064 dysfunction Effects 0.000 description 1
- 235000013399 edible fruits Nutrition 0.000 description 1
- 230000002526 effect on cardiovascular system Effects 0.000 description 1
- 229940124770 endothelial lipase inhibitor Drugs 0.000 description 1
- 238000005516 engineering process Methods 0.000 description 1
- 239000003623 enhancer Substances 0.000 description 1
- 239000000147 enterotoxin Substances 0.000 description 1
- 210000002919 epithelial cell Anatomy 0.000 description 1
- 239000000686 essence Substances 0.000 description 1
- 229960000403 etanercept Drugs 0.000 description 1
- 230000003090 exacerbative effect Effects 0.000 description 1
- 230000029142 excretion Effects 0.000 description 1
- 229930195729 fatty acid Natural products 0.000 description 1
- 239000000194 fatty acid Substances 0.000 description 1
- 150000004665 fatty acids Chemical class 0.000 description 1
- 230000004634 feeding behavior Effects 0.000 description 1
- 239000000835 fiber Substances 0.000 description 1
- 150000002206 flavan-3-ols Chemical class 0.000 description 1
- 235000011987 flavanols Nutrition 0.000 description 1
- HVQAJTFOCKOKIN-UHFFFAOYSA-N flavonol Natural products O1C2=CC=CC=C2C(=O)C(O)=C1C1=CC=CC=C1 HVQAJTFOCKOKIN-UHFFFAOYSA-N 0.000 description 1
- 150000002216 flavonol derivatives Chemical class 0.000 description 1
- 235000011957 flavonols Nutrition 0.000 description 1
- 235000021554 flavoured beverage Nutrition 0.000 description 1
- 239000012634 fragment Substances 0.000 description 1
- 239000012737 fresh medium Substances 0.000 description 1
- 230000005714 functional activity Effects 0.000 description 1
- 235000020510 functional beverage Nutrition 0.000 description 1
- 230000002496 gastric effect Effects 0.000 description 1
- 230000009229 glucose formation Effects 0.000 description 1
- 230000014101 glucose homeostasis Effects 0.000 description 1
- 230000004153 glucose metabolism Effects 0.000 description 1
- 230000034659 glycolysis Effects 0.000 description 1
- 229940094952 green tea extract Drugs 0.000 description 1
- 235000020688 green tea extract Nutrition 0.000 description 1
- 239000003102 growth factor Substances 0.000 description 1
- 230000009036 growth inhibition Effects 0.000 description 1
- 235000004280 healthy diet Nutrition 0.000 description 1
- 229940037467 helicobacter pylori Drugs 0.000 description 1
- 230000011132 hemopoiesis Effects 0.000 description 1
- 230000002440 hepatic effect Effects 0.000 description 1
- 208000006454 hepatitis Diseases 0.000 description 1
- 231100000283 hepatitis Toxicity 0.000 description 1
- 230000009215 host defense mechanism Effects 0.000 description 1
- 102000050770 human IGFBP2 Human genes 0.000 description 1
- 102000056685 human RETN Human genes 0.000 description 1
- 235000020256 human milk Nutrition 0.000 description 1
- 210000004251 human milk Anatomy 0.000 description 1
- 239000000399 hydroalcoholic extract Substances 0.000 description 1
- 229960000890 hydrocortisone Drugs 0.000 description 1
- 230000007062 hydrolysis Effects 0.000 description 1
- 238000006460 hydrolysis reaction Methods 0.000 description 1
- 238000003384 imaging method Methods 0.000 description 1
- 230000002519 immonomodulatory effect Effects 0.000 description 1
- 210000002865 immune cell Anatomy 0.000 description 1
- 229940097275 indigo Drugs 0.000 description 1
- COHYTHOBJLSHDF-UHFFFAOYSA-N indigo powder Natural products N1C2=CC=CC=C2C(=O)C1=C1C(=O)C2=CC=CC=C2N1 COHYTHOBJLSHDF-UHFFFAOYSA-N 0.000 description 1
- 229960000905 indomethacin Drugs 0.000 description 1
- 230000006698 induction Effects 0.000 description 1
- 230000001524 infective effect Effects 0.000 description 1
- 230000008595 infiltration Effects 0.000 description 1
- 238000001764 infiltration Methods 0.000 description 1
- 210000004969 inflammatory cell Anatomy 0.000 description 1
- 229960000598 infliximab Drugs 0.000 description 1
- 238000001802 infusion Methods 0.000 description 1
- 208000014674 injury Diseases 0.000 description 1
- 230000003993 interaction Effects 0.000 description 1
- 230000005694 interleukin-22 production Effects 0.000 description 1
- 235000015110 jellies Nutrition 0.000 description 1
- 239000008274 jelly Substances 0.000 description 1
- 235000015141 kefir Nutrition 0.000 description 1
- 238000002372 labelling Methods 0.000 description 1
- 229940039696 lactobacillus Drugs 0.000 description 1
- 230000023404 leukocyte cell-cell adhesion Effects 0.000 description 1
- 208000006132 lipodystrophy Diseases 0.000 description 1
- 239000007788 liquid Substances 0.000 description 1
- 230000031142 liver development Effects 0.000 description 1
- 244000144972 livestock Species 0.000 description 1
- 239000006210 lotion Substances 0.000 description 1
- 238000002803 maceration Methods 0.000 description 1
- 238000007726 management method Methods 0.000 description 1
- 210000003622 mature neutrocyte Anatomy 0.000 description 1
- 238000005259 measurement Methods 0.000 description 1
- 230000007246 mechanism Effects 0.000 description 1
- 230000010534 mechanism of action Effects 0.000 description 1
- 230000006371 metabolic abnormality Effects 0.000 description 1
- 230000002503 metabolic effect Effects 0.000 description 1
- 230000037353 metabolic pathway Effects 0.000 description 1
- 239000002207 metabolite Substances 0.000 description 1
- 230000000813 microbial effect Effects 0.000 description 1
- 244000005700 microbiome Species 0.000 description 1
- 239000011785 micronutrient Substances 0.000 description 1
- 235000013369 micronutrients Nutrition 0.000 description 1
- 238000000386 microscopy Methods 0.000 description 1
- 238000013508 migration Methods 0.000 description 1
- 230000003278 mimic effect Effects 0.000 description 1
- 230000005787 mitochondrial ATP synthesis coupled electron transport Effects 0.000 description 1
- 230000002438 mitochondrial effect Effects 0.000 description 1
- 239000003226 mitogen Substances 0.000 description 1
- 150000002772 monosaccharides Chemical class 0.000 description 1
- 210000004400 mucous membrane Anatomy 0.000 description 1
- 201000006417 multiple sclerosis Diseases 0.000 description 1
- 206010028417 myasthenia gravis Diseases 0.000 description 1
- 210000003098 myoblast Anatomy 0.000 description 1
- 210000000107 myocyte Anatomy 0.000 description 1
- 230000017128 negative regulation of NF-kappaB transcription factor activity Effects 0.000 description 1
- 210000005036 nerve Anatomy 0.000 description 1
- 230000001537 neural effect Effects 0.000 description 1
- 210000002569 neuron Anatomy 0.000 description 1
- 239000002858 neurotransmitter agent Substances 0.000 description 1
- 230000003472 neutralizing effect Effects 0.000 description 1
- 210000000440 neutrophil Anatomy 0.000 description 1
- 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 1
- 229940021182 non-steroidal anti-inflammatory drug Drugs 0.000 description 1
- 210000001009 nucleus accumben Anatomy 0.000 description 1
- 238000013116 obese mouse model Methods 0.000 description 1
- 229940125395 oral insulin Drugs 0.000 description 1
- 210000000056 organ Anatomy 0.000 description 1
- 150000007524 organic acids Chemical class 0.000 description 1
- 235000005985 organic acids Nutrition 0.000 description 1
- 230000001590 oxidative effect Effects 0.000 description 1
- 229910052760 oxygen Inorganic materials 0.000 description 1
- 239000001301 oxygen Substances 0.000 description 1
- 230000037324 pain perception Effects 0.000 description 1
- 206010033675 panniculitis Diseases 0.000 description 1
- 230000003076 paracrine Effects 0.000 description 1
- 238000007911 parenteral administration Methods 0.000 description 1
- 230000008506 pathogenesis Effects 0.000 description 1
- 230000001717 pathogenic effect Effects 0.000 description 1
- 230000001575 pathological effect Effects 0.000 description 1
- 230000035699 permeability Effects 0.000 description 1
- 230000009038 pharmacological inhibition Effects 0.000 description 1
- 235000017807 phytochemicals Nutrition 0.000 description 1
- 239000000049 pigment Substances 0.000 description 1
- 239000000419 plant extract Substances 0.000 description 1
- 229930000223 plant secondary metabolite Natural products 0.000 description 1
- 238000007747 plating Methods 0.000 description 1
- 230000008092 positive effect Effects 0.000 description 1
- 230000009819 post translational phosphorylation Effects 0.000 description 1
- 239000002243 precursor Substances 0.000 description 1
- 238000002360 preparation method Methods 0.000 description 1
- 230000002265 prevention Effects 0.000 description 1
- 125000002924 primary amino group Chemical group [H]N([H])* 0.000 description 1
- 230000007112 pro inflammatory response Effects 0.000 description 1
- 230000004647 pro-inflammatory pathway Effects 0.000 description 1
- 230000000529 probiotic effect Effects 0.000 description 1
- 238000012545 processing Methods 0.000 description 1
- 230000035755 proliferation Effects 0.000 description 1
- 230000001681 protective effect Effects 0.000 description 1
- 238000001243 protein synthesis Methods 0.000 description 1
- 230000002797 proteolythic effect Effects 0.000 description 1
- 239000000376 reactant Substances 0.000 description 1
- 238000011084 recovery Methods 0.000 description 1
- 230000008929 regeneration Effects 0.000 description 1
- 238000011069 regeneration method Methods 0.000 description 1
- 238000007634 remodeling Methods 0.000 description 1
- 238000011160 research Methods 0.000 description 1
- 208000023504 respiratory system disease Diseases 0.000 description 1
- 230000000284 resting effect Effects 0.000 description 1
- 229960003471 retinol Drugs 0.000 description 1
- 235000020944 retinol Nutrition 0.000 description 1
- 239000011607 retinol Substances 0.000 description 1
- 238000012216 screening Methods 0.000 description 1
- 230000003248 secreting effect Effects 0.000 description 1
- 230000035945 sensitivity Effects 0.000 description 1
- 230000008313 sensitization Effects 0.000 description 1
- 239000002453 shampoo Substances 0.000 description 1
- 230000035939 shock Effects 0.000 description 1
- 235000021391 short chain fatty acids Nutrition 0.000 description 1
- 150000004666 short chain fatty acids Chemical class 0.000 description 1
- 239000000377 silicon dioxide Substances 0.000 description 1
- 235000009561 snack bars Nutrition 0.000 description 1
- 229940054269 sodium pyruvate Drugs 0.000 description 1
- 238000010186 staining Methods 0.000 description 1
- 230000007863 steatosis Effects 0.000 description 1
- 231100000240 steatosis hepatitis Toxicity 0.000 description 1
- 230000004936 stimulating effect Effects 0.000 description 1
- 239000011550 stock solution Substances 0.000 description 1
- 210000004003 subcutaneous fat Anatomy 0.000 description 1
- 239000000758 substrate Substances 0.000 description 1
- 229940005741 sunflower lecithin Drugs 0.000 description 1
- 230000004083 survival effect Effects 0.000 description 1
- 230000002459 sustained effect Effects 0.000 description 1
- 230000008961 swelling Effects 0.000 description 1
- 210000000225 synapse Anatomy 0.000 description 1
- 210000001258 synovial membrane Anatomy 0.000 description 1
- 210000002437 synoviocyte Anatomy 0.000 description 1
- 230000009897 systematic effect Effects 0.000 description 1
- 239000003826 tablet Substances 0.000 description 1
- 238000004885 tandem mass spectrometry Methods 0.000 description 1
- 230000008685 targeting Effects 0.000 description 1
- 229960003080 taurine Drugs 0.000 description 1
- 235000013616 tea Nutrition 0.000 description 1
- 239000012085 test solution Substances 0.000 description 1
- 238000002560 therapeutic procedure Methods 0.000 description 1
- 230000000699 topical effect Effects 0.000 description 1
- 230000001988 toxicity Effects 0.000 description 1
- 231100000419 toxicity Toxicity 0.000 description 1
- 108091006106 transcriptional activators Proteins 0.000 description 1
- 230000007704 transition Effects 0.000 description 1
- 230000014616 translation Effects 0.000 description 1
- 230000032258 transport Effects 0.000 description 1
- UFTFJSFQGQCHQW-UHFFFAOYSA-N triformin Chemical compound O=COCC(OC=O)COC=O UFTFJSFQGQCHQW-UHFFFAOYSA-N 0.000 description 1
- 239000013638 trimer Substances 0.000 description 1
- 229940046728 tumor necrosis factor alpha inhibitor Drugs 0.000 description 1
- 239000002451 tumor necrosis factor inhibitor Substances 0.000 description 1
- 230000007306 turnover Effects 0.000 description 1
- 230000003827 upregulation Effects 0.000 description 1
- 238000011144 upstream manufacturing Methods 0.000 description 1
- RUDATBOHQWOJDD-UZVSRGJWSA-N ursodeoxycholic acid Chemical compound C([C@H]1C[C@@H]2O)[C@H](O)CC[C@]1(C)[C@@H]1[C@@H]2[C@@H]2CC[C@H]([C@@H](CCC(O)=O)C)[C@@]2(C)CC1 RUDATBOHQWOJDD-UZVSRGJWSA-N 0.000 description 1
- 229960001661 ursodiol Drugs 0.000 description 1
- 230000002792 vascular Effects 0.000 description 1
- 230000008728 vascular permeability Effects 0.000 description 1
- 210000004509 vascular smooth muscle cell Anatomy 0.000 description 1
- 230000024883 vasodilation Effects 0.000 description 1
- 230000009385 viral infection Effects 0.000 description 1
- 235000019154 vitamin C Nutrition 0.000 description 1
- 239000011718 vitamin C Substances 0.000 description 1
- 229940045997 vitamin a Drugs 0.000 description 1
- 230000004584 weight gain Effects 0.000 description 1
- 235000019786 weight gain Nutrition 0.000 description 1
- 239000012224 working solution Substances 0.000 description 1
- 235000013618 yogurt Nutrition 0.000 description 1
Landscapes
- Medicines Containing Plant Substances (AREA)
Abstract
Contemplated compositions and methods are based on a combination of polyphenols common to the Mediterranean diet, which, when combined, reduce the expression of pro-inflammatory signals and senescence-associated genes, and support mitochondrial biogenesis and cellular energy metabolism. Notably, the observed pleiotropic effects not only span multiple signaling pathways, but also exhibit synergistic activity in relation to a number of markers that reduce age-related energy decline, reduce age-related immune decline, and reduce age-related increase or persistence of chronic subacute inflammation. From a different perspective, contemplated compositions represent a harmonious combination of multiple biochemical molecules that form the basis of numerous benefits common in the mediterranean diet.
Description
The present application claims priority from U.S. provisional application Ser. Nos. 63/215,716, filed on 6/28 of 2021, and 63/285,591 filed on 12/3 of 2021, both of which are incorporated herein by reference.
Technical Field
The field of the present invention is compositions and methods for nutritional supplements, in particular to polyphenols and polyphenol mixtures and their use in the alleviation of various conditions associated with aging, such as mild/chronic inflammation, age-related reduced immunity, reduced energy metabolism and/or obesity.
Background
The background description includes information that may be helpful in understanding the present disclosure. It is not an admission that any of the information provided herein is prior art or relevant to the presently claimed invention, or that any publication specifically or implicitly referenced is prior art.
All publications and patent applications herein are incorporated by reference to the same extent as if each individual publication or patent application was specifically and individually indicated to be incorporated by reference. If the definition or usage of a term incorporated by reference does not conform to or is contrary to the definition of that term provided herein, then the definition of that term provided herein applies and the definition of that term as cited does not apply
Vitamins and other isolated nutritional compounds are diverse in species, and the benefits of these compounds are said to include immune support, anti-inflammatory effects, anti-aging effects, cardiac support, and digestive support, among others. Unfortunately, there is relatively little evidence that certain aspects of these so-called benefits are present when these vitamins and other isolated nutritional compounds are ingested. Also, if the nutritional supplement is in the form of an extract or powder of plant parts, various benefits will be advertised, but the actual benefits tend to be poor, or even not demonstrated at all. Furthermore, isolated nutritional compounds, as well as plant extracts and concentrates alone, often do not reflect a healthy diet.
Notably, certain geographic and ethnic diet types are associated with overall health, longevity, and/or physical recovery, and these beneficial effects are indeed well documented and proven. For example, the Mediterranean diet is generally associated with lower cardiovascular risk factors (see, e.g., nutrients 2018,10,379; doi:10.3390/nu 10030379), lower inflammatory and metabolic biomarkers, reduced risk of Alzheimer's disease (see, e.g., J Alzheimer's Dis.2010;22 (2): 483-492.), and reduced of certain inflammatory markers (see, e.g., nutrients 2018,10,62; doi:10.3390/nu 10010062). One common class of ingredients found in such diets are polyphenols, and various studies have been published regarding specific benefits of individual dietary polyphenols (see, e.g., inhibitory Properties of Phenolic Compounds Against Enzymes Linked with Human Diseases: URL: dx.doi.org/10.5772/66844) and selected colored polyphenols (see, e.g., annu.Rev.food Sci.technology 2020.11: 10.1-10.38). However, due to the complexity and number of chemically diverse polyphenols, many studies have focused on only specific biochemical actions of individual polyphenols and such compounds, or provide general epidemiological information, without having molecular features without a more detailed diet.
In an effort to supplement the diet with various polyphenols, various supplements are known. For example, the Vital reactant (Gundry MD) provides a commercially available concentrated polyphenol powder mixture made from a variety of red plant materials to increase energy and promote digestion. Such mixtures advantageously comprise a plurality of chemically different polyphenols. However, the choice of plant material for use as a source of polyphenols does not reflect conventional dietary intake. Similarly, the mixture of the commercially available grape extract, olive extract, pomegranate extract, green tea extract, grapefruit extract, blueberry extract and orange extract sold by Fytexia are provided as antioxidant agents to protect cells from oxidative stress (see, e.g., the oxygenia sold by Fytexia). While the source components of such antioxidant formulations are beneficial in reducing oxidative stress, it has not generally been demonstrated to affect molecular signaling associated with (chronic subacute) inflammation, nfkb signaling, reduced energy metabolism, aging, age-related immune decline, and/or obesity.
Thus, while various nutritional supplements are known in the art, all or almost all of them suffer from various drawbacks. Accordingly, there is a need for compositions and methods that provide improved nutritional supplements, particularly those that have been demonstrated to combat (chronic subacute) inflammation, nfkb signaling, reduced energy metabolism, aging, age-related immune decline, and/or obesity.
Disclosure of Invention
The present inventors have now discovered various compositions and methods of specific combinations of polyphenols and/or polyphenols-rich substances (e.g., extracts and powders) common in food products of the Mediterranean diet that beneficially modulate various molecular characteristics associated with age-related reduced immunity, inflammation, reduced energy metabolism, and/or obesity. Thus, the polyphenols and/or polyphenol-enriched materials presented herein may be advantageously used in a nutritional-based approach to combat inflammation, which may be characterized by one or more of age-related reduced immunity, increased or chronic inflammation, reduced energy metabolism, and/or obesity.
In one aspect of the inventive subject matter, the inventors contemplate a nutritional composition comprising a nutritionally acceptable carrier in combination with a plurality of chemically different polyphenol containing plant materials having red, green, orange and violet colors, and in particularly preferred embodiments, the red, green, orange and violet plant materials have a synergistic effect in reducing proinflammatory cytokine release in human leukocytes.
For example, the red plant material may include apple extract, pomegranate extract, tomato powder, and beet root powder, the green plant material may include olive extract, rosemary extract, green coffee bean extract, and collard powder, the orange plant material may include onion extract, ginger extract, grapefruit extract, and carrot powder, and/or the purple blue plant material may include grape extract, blueberry extract, currant powder, and elderberry powder. From another perspective, the colored plant material may be selected on the basis of, or may be part of, a Mediterranean diet.
In some embodiments, the release of the pro-inflammatory cytokine is the release of at least one, or at least two, or at least three of tumor necrosis factor alpha (TNF-alpha), interleukin-6 (IL-6), prostaglandin E2 (PGE 2), and isoprostadin. In other embodiments, the release of the pro-inflammatory cytokine is the release of tumor necrosis factor alpha (TNF-alpha), interleukin-6 (IL-6), prostaglandin E2 (PGE 2), and isoprostadins.
It is further recognized that the compositions provided herein may also reduce nfkb expression, increase glucose uptake by cells, increase mitochondrial biogenesis in cells, reduce oxidative damage caused by reactive oxygen species, reduce expression of pro-inflammatory adipokines, and/or reduce expression of one or more senescence-associated genes.
Most typically, but not necessarily, the compositions will be formulated for oral administration, either as bulk materials or in single dosage units. For example, a single dosage unit may contain 50mg to 1000mg of the composition and may be formulated as a capsule, a jelly or a bulk powder. It will also be readily appreciated that contemplated compositions may also include vitamins, dietary trace elements or minerals, probiotics, and/or prebiotics. Similarly, contemplated compositions may also include niacin, nicotinamide riboside, nicotinamide mononucleotide, nicotinamide adenine dinucleotide, and/or a nutritionally acceptable CD38 inhibitor.
Thus, contemplated compositions are considered effective in treating or alleviating symptoms associated with inflammatory disorders, metabolic disorders, neurological disorders, cardiovascular disorders, aging, and/or oxidative stress.
Accordingly, the present inventors contemplate a method of supporting the health of a subject, wherein a composition as described herein is administered to an individual. For example, the composition may be administered in an amount effective to treat or reduce symptoms associated with inflammatory disorders, metabolic disorders, neurological disorders, cardiovascular disorders, aging, and/or oxidative stress. Thus, administration may last at least 30 days and/or be administered in daily doses of 50mg to 1000 mg. Thus, contemplated compositions are useful for supporting the health of a subject and healthy aging by orally administering the composition.
Thus, in a further aspect of the inventive subject matter, the inventors also contemplate a method of reducing inflammation in a subject, the method comprising administering to the subject a nutritionally acceptable carrier in combination with a plurality of chemically different polyphenol-containing plant materials having red, green, orange-yellow and violet-blue colors, wherein the combination of plant materials reduces release of at least one pro-inflammatory cytokine in human cells, reduces expression of at least one senescence-associated gene in human cells.
In certain embodiments, the red plant material may include apple extract, pomegranate extract, tomato powder, and beetroot powder, the green plant material may include olive extract, rosemary extract, green coffee bean extract, and collard powder, the orange plant material may include onion extract, ginger extract, grapefruit extract, and carrot powder, and/or the violet plant material may include grape extract, blueberry extract, currant powder, and elderberry powder. Thus, the red, green, orange and violet plant materials are part of the mediterranean diet or may be selected based on the mediterranean diet.
Most typically, the pro-inflammatory cytokine is selected from the group consisting of tumor necrosis factor alpha (TNF-alpha), interleukin-6 (IL-6), prostaglandin E2 (PGE 2), and isoprostadins. In particularly preferred aspects, the red, green, orange and violet plant materials will be present in synergistic amounts that reduce the release of pro-inflammatory cytokines in human cells. . As for the senescence-associated gene, it can be expected that the senescence-associated gene may be selected from HGF, c-fos, p16.sup.INK and p21.
Advantageously, the reduction of inflammation will include a reduction in age-related immunity, a reduction in age-related energy metabolism, a reduction in age-related mitochondrial biogenesis, a reduction in at least one symptom associated with inflammation, and/or a reduction in at least one symptom associated with metabolic syndrome.
Furthermore, it should be appreciated that in at least some embodiments, administration of the composition also advantageously reduces expression of nfkb, increases glucose uptake into cells, increases mitochondrial biogenesis in cells, reduces oxidative damage caused by reactive oxygen species, reduces expression of pro-inflammatory adipokines, and/or increases intracellular ATP.
Preferably, but not necessarily, the administration to the subject includes oral administration of plant material containing a plurality of chemically different polyphenols, e.g., at a dose of about 50mg to 1000mg. If desired, a variety of chemically unique polyphenol containing plant materials can be formulated into tablets, drinks or soft sweets. Furthermore, contemplated materials may also include vitamins, dietary trace elements or minerals, probiotics, and/or prebiotics. Likewise, the combination of plant materials may also comprise niacin, nicotinamide riboside, nicotinamide mononucleotide, nicotinamide adenine dinucleotide, and/or a nutritionally acceptable CD38 inhibitor.
In another aspect of the inventive subject matter, the inventors also contemplate a method of reducing inflammation in a subject, the method comprising the step of administering to the subject a nutritionally acceptable carrier in combination with a plant material having red, green, orange and violet colors comprising a plurality of chemically different polyphenols. Preferably, the red plant material comprises apple extract, pomegranate extract, tomato powder and beet root powder, the green plant material comprises olive extract, rosemary extract, green coffee bean extract and kale powder, the orange plant material comprises onion extract, ginger extract, grapefruit extract, and carrot powder, and/or the violet plant material comprises grape extract, blueberry extract, currant powder and elderberry powder. In such methods, the combination of plant materials will reduce at least one pro-inflammatory cytokine, reduce nfκf signaling, and/or reduce at least one pro-inflammatory adipokine in the subject after administration. Preferably, but not necessarily, the combination of plant materials is a synergistic combination with respect to at least one cytokine reduction.
In another aspect of the inventive subject matter, the inventors contemplate a method of increasing glucose absorption by a cell comprising the step of contacting the cell with a plant material comprising a plurality of chemically distinct polyphenols having red, green, orange and violet colors, wherein the red plant material comprises apple extract, pomegranate extract, tomato powder and beetroot powder, wherein the green plant material comprises olive extract, rosemary extract, green coffee bean extract and collard powder, wherein the orange plant material comprises onion extract, ginger extract, grapefruit extract and carrot powder, and wherein the violet plant material comprises grape extract, blueberry extract, currant powder and elderberry powder. In this method, the combination of plant materials increases glucose uptake by cells after contacting the cells. Notably, as described in more detail below, an increase in glucose uptake by the cells is observed that is similar to or greater than (under otherwise identical conditions) an increase in glucose uptake with a thiazolinedione drug, such as rosiglitazone.
In a further aspect of the inventive subject matter, the inventors also contemplate a method of increasing ATP levels in a cell comprising the step of contacting the cell with a plant material comprising a plurality of chemically distinct polyphenols having red, green, orange and violet colors, wherein the red plant material comprises apple extract, pomegranate extract, tomato powder and beetroot powder, wherein the green plant material comprises olive extract, rosemary extract, green coffee bean extract and collard powder, wherein the orange plant material comprises onion extract, ginger extract, grapefruit extract and carrot powder, and wherein the violet plant material comprises grape extract, blueberry extract, currant powder and elderberry powder. In this method, the combination of plant materials increases the ATP level in the cells (particularly muscle cells) after contacting the cells.
Thus, from a different perspective, the inventors contemplate plant material having red, green, orange and violet colors comprising a plurality of chemically distinct polyphenols for alleviating at least one symptom of inflammation in an individual ingesting the plant material.
Similarly, the inventors also contemplate use of the nutritional compositions described herein to reduce the release of pro-inflammatory cytokines in human leukocytes, reduce nfkb signaling, increase glucose uptake by cells, increase mitochondrial biogenesis in cells, reduce oxidative damage caused by reactive oxygen species, reduce the expression of pro-inflammatory adipokines, and/or reduce the expression of at least one senescence-associated gene.
Various objects, features, aspects and advantages of the present subject matter will become more apparent from the following detailed description of preferred embodiments, along with the accompanying drawings in which like numerals represent like components.
Drawings
FIG. 1 is a graph illustrating exemplary results of the effect of a representative composition on various cytokines in primary human monocytes.
FIG. 2 is a graph illustrating exemplary results of the effect of a representative composition on selected cytokines in primary human monocytes.
FIG. 3 is a graph illustrating exemplary results of the effect of a representative composition on TFN-alpha release in primary human monocytes.
FIG. 4 is a graph illustrating exemplary results of the effect of a representative composition on IL-6 release in primary human monocytes.
FIG. 5 is a graph illustrating PGE in representative compositions versus primary human monocytes 2 Exemplary results of the influence of ReleaseA drawing.
Fig. 6 is a graph illustrating exemplary results of the effect of a representative composition on the release of isoprostadins from primary human monocytes.
FIG. 7 is a graph of exemplary results illustrating the effect of a representative composition on IL-2 synthesis in human primary lymphocytes.
FIG. 8 is a graph of exemplary results illustrating the effect of green sub-mixtures of representative compositions on selected cytokines in primary human monocytes.
FIG. 9 is a graph of exemplary results illustrating the effect of red sub-mixtures of representative compositions on selected cytokines in primary human monocytes.
FIG. 10 is a graph of exemplary results illustrating the effect of orange/yellow sub-mixtures of representative compositions on selected cytokines in primary human monocytes.
FIG. 11 is a graph of exemplary results illustrating the effect of purple/blue sub-mixtures of representative compositions on selected cytokines in primary human monocytes.
FIG. 12 is a graph illustrating exemplary results of the effect of a combined sub-mixture (representative composition) on selected cytokines in primary human monocytes.
FIG. 13 is a graph of exemplary results illustrating the effect of a representative composition on the expression of IL-6mRNA in C2C12 cells.
FIG. 14 is a graph of exemplary results illustrating the effect of representative compositions on TNF- α expression in C2C12 cells.
FIG. 15 is a graph of exemplary results illustrating the effect of a representative composition on NFkB expression in a HEK293 model system.
FIG. 16 is a graph illustrating exemplary results of the effect of a representative composition on glucose uptake by C2C12 muscle cells.
Fig. 17 is a graph illustrating exemplary results of the effect of representative compositions on mitochondrial biogenesis.
Fig. 18 is a graph illustrating exemplary results of the effect of representative compositions on mouse RAW 264.1 macrophage ATP levels.
FIG. 19 is a graph illustrating exemplary results of the effect of representative compositions on the level of ATP in C2C12 muscle cells.
Fig. 20 is a graph illustrating exemplary results of the effect of a representative composition on Reactive Oxygen Species (ROS) in mouse macrophages.
Fig. 21 is a graph illustrating exemplary results of inhibition of selected fat releasing hormone by representative compositions.
Fig. 22 is a graph illustrating exemplary results of stimulation of a first set of selected fat releasing hormones by a representative composition.
Fig. 23 is a graph illustrating exemplary results of inhibition of a second set of selected fat releasing hormones by a representative composition.
FIG. 24 is a graph illustrating exemplary results of the effect of a representative composition on gene expression of selected genes associated with senescence.
FIG. 25 is a graph illustrating exemplary results of the effect of representative compositions on acetylcholinesterase activity.
FIG. 26 is a graph of exemplary results illustrating the effect of representative compositions on macrophage cell viability and metabolism.
Detailed Description
The inventors have now found that a specific combination of polyphenol-containing materials (and polyphenols found therein) can strongly modulate many biomarkers associated with increased or chronic inflammation, nfkb signaling, reduced energy metabolism, aging, age-related immunity decline and/or obesity. Notably, this combination closely tracks the components and polyphenol-containing materials found in the Mediterranean diet (and the polyphenols found therein). In view of these findings, the present inventors therefore contemplate various compositions, compositions and uses in medical foods, even in medicine, for nutritional supplements and other nutritional products.
Based on his extensive studies, as shown in more detail below, the present inventors have now found that specific mixtures of selected plant materials (and polyphenols contained therein) commonly found in the Mediterranean diet can be prepared which mimic the benefits of the Mediterranean diet, such as with inflammation andmodulation of various biomarkers associated with cellular energy is demonstrated. Among other biomarkers, particularly contemplated biomarkers include various pro-inflammatory cytokines and transcription factors (e.g., TNF- α, IL-6, PGE 2 Isoprostadin, nfkb), various adipokines (such as leptin, oncostatin M, resistin, FGF21, HGF, and IL-11), mitochondrial biogenesis, cellular glucose uptake, intracellular ATP, antioxidant capacity as measured by reactive oxygen species, and expression of various senescence-associated genes.
Notably, the inventors have found that the compositions according to the inventive subject matter not only inhibit or reduce the production and secretion of pro-inflammatory cytokines that play a role in different inflammatory pathways, but also down-regulate transcription factors critical to the pro-inflammatory response. It is further unexpected that this modulation of inflammatory components is particularly pronounced and synergistically enhanced when a plurality of chemically distinct polyphenol containing plant materials having red, green, orange and violet colors are combined. Even more surprising results, the inventors have found that combining plant materials containing chemically different polyphenols has a much greater benefit than the anti-inflammatory effect. In addition, as shown in more detail below, the compositions of the present invention have significant activity in increasing glucose uptake and mitochondrial biogenesis by cells, increasing ATP levels, and the like. In addition, the composition also exhibits a significant effect on selected fat-releasing hormones associated with inflammation and energy metabolism. Finally, such compositions also reduce gene expression of various senescence-associated genes. These effects, especially in general, appear to indicate the same or similar beneficial molecular mechanisms as are typically observed in the Mediterranean diet. Thus, without wishing to be bound by a particular theory or hypothesis, the inventors believe that the compositions presented herein represent a potentially harmonious combination of biochemically diverse molecules that form the basis of many of the benefits observed with the Mediterranean diet. In this case, it should also be appreciated that various other diets associated with longevity and healthy aging (blue region diets) may be used as sources of the polyphenol components that will collectively enhance pathways and signals critical to health and longevity.
Preferably, the contemplated mixture belongs to a combination of a plurality (e.g., at least two, at least three, or at least four) of colored plant materials of different colors, in particular plant materials having red, green, orange/yellow and/or violet/blue colors. For example, in one embodiment of such a composition, and as shown in more detail below, from red raw materials including apple extract, pomegranate extract, tomato powder, and beetroot powder; a green source material comprising olive extract, rosemary extract, green coffee bean extract and kale; orange/yellow raw materials including onion extract, ginger extract, grapefruit extract, and carrot; purple/blue raw materials including grape extract, blueberry extract, currant powder and elderberry powder to obtain polyphenol-containing products/extracts; specific components and proportions are described in more detail below. Thus, from a different perspective, contemplated compositions will include a substantial amount of polyphenols below at least two, or at least three, or at least four different polyphenols including organic acids, phenols, flavonols, flavanols, anthocyanins, chlorogenic acids, betabetalains, and the like. It will be readily appreciated that the particular choice of plant material will depend on the (polyphenol) component desired in the plant material and its effect on the particular biological system and/or signal pathway.
Of course, it should also be understood that the plant material may be provided in a variety of forms, including all plant material or portions thereof (e.g., roots, fruits, leaves, etc.) in fresh or dried form, juices or macerations of fresh or dried plant material or plant material portions, as well as aqueous or aqueous/alcoholic extracts and chromatographic fractions of such plant material. Still further, it should be noted that one or more polyphenols of the plant material may even be provided as purified (naturally isolated or synthetic) chemical entities, typically having a chemical purity of at least 90%, or at least 95%, or at least 98%, or at least 99%. However, it should be appreciated that in most embodiments, the plant material will be a complex mixture to provide a desired combination of biological effects on a plurality of different molecular entities (e.g., enzymes, receptors, ion channels), wherein at least some biological effects (e.g., at least one, or at least two, or at least three, etc.) are synergistic. Furthermore, it is expected that biological effects on specific molecular entities will also be complementary in biological function, and in some cases even synergistic. Thus, based on the testing and desired targets described in more detail below, it should be appreciated that the compositions of the present subject matter can be formulated to meet specific needs. However, in particularly preferred aspects, contemplated compositions will inhibit multiple targets (e.g., at least two, at least three, at least four, etc.) in multiple and distinct (e.g., at least two, at least three, at least four, etc.) signaling pathways, preferably associated with inflammation, immune response, and/or energy metabolism.
Thus, from a different perspective, it should be appreciated that the mechanism of action of contemplated compositions is not limited to a single specific function (e.g., antioxidants) or to a specific chemical class (e.g., vitamins), but in fact they are complementary and may cooperate to provide multiple biological activities, spanning different metabolic and signaling pathways, and other important cellular health functions such as mitochondrial biogenesis and glucose turnover. Thus, contemplated compositions and methods target a variety of biological systems, including energy metabolism, immune function, inflammation, and the like. From a different perspective, it is contemplated that compositions and methods will reduce inflammation in subjects ingesting such compositions. As used herein, the term "inflammation" refers to a complex series of physiological events including at least one inflammatory component (e.g., an inflammatory disorder, over-expression of pro-inflammatory cytokines, and/or excessive activity of pro-inflammatory signaling pathways) and a decrease in at least one typical age-related cellular energy or mitochondrial biogenesis, and/or sustained or increased expression of senescence-associated genes. In addition, it is contemplated that the plant material will also provide a variety of micronutrients to support or supplement the function of polyphenols and other colored pigments present in the composition.
Among other benefits, it is to be appreciated that the compositions and methods presented herein may be on a variety of pathways that control pro-inflammatory signalingProvides significant complementary activity and therefore will have a more desirable physiological effect profile. For example, as shown in more detail below, contemplated compositions not only exhibit a significant reduction in proinflammatory cytokine release, but also result in nfkb signaling, PGE 2 Reduction of adipokine and isoprostaglandin release, which are pro-inflammatory mediators that further act in separate signaling pathways. Notably, while the expected sub-mixtures of the compositions do show some effect in modulating the above-described pro-inflammatory mediators, the combination of the sub-mixtures shows a strong synergistic effect in all the above elements. Furthermore, the compositions contemplated herein also exhibit significant antioxidant effects, as determined by the reduction of Reactive Oxidative Species (ROS), which potentially further reduce cellular stress and pro-inflammatory signaling.
It is also notable that contemplated compositions also have a significant impact on cellular energy management, particularly on glucose absorption, mitochondrial biogenesis, and ATP levels. In fact, the contemplated compositions result in improved glucose absorption, which is comparable to, and in some cases even exceeds, that observed for rosiglitazone, a common antidiabetic agent. Advantageously, this increased energy absorption does not result in an increase in free radical production, but contemplated compositions also significantly stimulate mitochondrial biogenesis and an increase in intracellular ATP levels. Thus, the combination of these processes can be seen as counteracting age-related decline in energy metabolism. Still further, regarding age-related decline, the inventors have found that contemplated compositions reduce the expression of many age-related genes.
Thus, it should be appreciated that the compositions contemplated herein may be advantageously used as stand alone products to support various aspects of health and healthy aging, such as support for proper immune function, support for reducing inflammation, and support for glucose levels (especially glucose uptake by cells). From a different perspective, it is to be understood that contemplated compositions may be used to treat or reduce symptoms associated with inflammatory disorders, metabolic disorders, neurological disorders, cardiovascular disorders, aging, and/or oxidative stress. In this context, it should be noted that the term "support" when used in connection with a physiological function or disorder is intended to prevent a decrease in the activity of one or more components or ingredients associated with the physiological function or disorder, at least partially reverse a decrease in the activity of one or more components or ingredients associated with the physiological function or disorder, maintain the normal function of the activity of one or more components or ingredients associated with the bodily function or disorder, prevent abnormal overactivity (or overexpression) of one or more components associated with the physiological function or disorder, and/or at least partially reverse abnormal overactivity (or overexpression) of one or more components associated with the physiological function or disorder. Alternatively, the compositions described herein may also be combined with other nutritional supplements and/or vitamins to provide benefits not available with such supplements or vitamins.
Regarding the potential molecular mechanisms of the various effects observed using contemplated compositions, the inventors investigated various potential targets that may be upregulated by contemplated compounds, including various cytokines, NF- κb, various fat factors, glucose absorption, mitochondrial biogenesis, ATP levels, reactive oxygen species, expression of senescence-associated genes, and acetylcholinesterase.
Interleukin-6: interleukin-6 (IL-6) is a multifunctional cytokine that regulates immune responses, acute phase responses, and hematopoiesis, and may also play a central role in host defense mechanisms. IL-6 is a pleiotropic cytokine produced by a variety of cells. It acts on a wide range of tissues, and according to the nature of target cells, it exerts growth induction, growth inhibition and differentiation effects, respectively. Notably, IL-6 is not composed of normal cells, but its expression is readily induced by a variety of cytokines, bacterial components such as lipopolysaccharide or viral infections. IL-6 is usually produced at the site of inflammation, and IL-6 binds to its soluble receptor sIL-6Rα, which determines the transition from acute to chronic inflammation by altering the nature of leukocyte infiltration (from polymorphonuclear neutrophils to monocytes/macrophages). In addition, IL-6 has stimulatory effects on T cells and B cells, thereby promoting a chronic inflammatory response. Thus, attempts to down-regulate IL-6 and IL-6 signaling are believed to be effective in treating various chronic inflammatory diseases (e.g., rheumatoid arthritis). As described in more detail below, contemplated compositions have a significant impact on IL-6 release and thus may help to alleviate or prevent one or more symptoms common in (chronic and/or subacute) inflammatory diseases.
Tumor necrosis factor α (TNF- α): tumor necrosis factor-alpha (TNF-alpha), produced mainly by activated macrophages, T lymphocytes and Natural Killer (NK) cells, is a central regulator of inflammation, and TNF-alpha antagonists and inhibitors are interesting approaches to the treatment of inflammatory diseases in which TNF-alpha plays an important pathogenic role. Inhibition of TNF has proven to be an effective treatment for patients suffering from rheumatoid arthritis and other forms of inflammatory diseases, including psoriasis, psoriatic arthritis and ankylosing spondylitis, inflammatory bowel disease. In addition, the efficacy of preventing the infective shock and the AIDS is also provided. Current successful biological therapies include etanercept, infliximab, and the fully human monoclonal antibody adalimumab, among others. However, natural TNF inhibitors such as the compositions described herein provide an attractive route due to the higher cost and side effects of currently known synthetic antibody therapies. Indeed, as described in more detail below, contemplated compositions have a significant inhibitory effect on TNF- α secretion and thus may also help to alleviate or prevent one or more symptoms common in various (chronic and/or subacute) inflammatory diseases.
Prostaglandin E 2 (PGE 2 ): prostaglandins (PG) are the major lipid mediators in animals and are synthesized in vivo from arachidonic acid by cyclooxygenase enzymes (COX-1 or COX-2) as rate-limiting enzymes. Prostaglandin E 2 (PGE 2 ) Is the most detected PG in a variety of tissues, exerting a variety of physiological and pathological effects through four receptor subtypes (EP 1-4). Non-steroidal anti-inflammatory drugs, such as aspirin and indomethacin, exert potent anti-inflammatory effects by inhibiting COX activity and the resultant inhibition of PG. PGE (PGE) 2 Regulate acute and chronic inflammation at the molecular and cellular level, as well as autoimmune diseases. PGE (PGE) 2 In one of acute inflammation, fever and painThe effect is considered to be vasodilation of vascular smooth muscle cells. Subsequent additional data indicate that PGE 2 Inducing mast cell activation, thereby enhancing vascular permeability, leading to PGE 2 Induced acute inflammation. In addition, PGE 2 Promoting Th1 cell differentiation, th17 cell proliferation, and Th22 cell production of IL-22, and exacerbating chronic inflammation and various autoimmune diseases. In addition, PGE 2 Activation of mast cells via EP3 receptors causes acute inflammation. PGE (PGE) 2 Chronic inflammation and various autoimmune diseases are also induced by helper T cell 1 (Th 1) differentiation, th17 cell proliferation, and IL-22 production by Th22 cells via EP2 and EP4 receptors. Notably, as shown in more detail below, the inventors have found that the compositions provided herein are useful for PGEs 2 Has remarkable inhibition effect on synthesis and release. Thus, contemplated compositions will again likely help to reduce or prevent one or more symptoms commonly found in various (chronic and/or subacute) inflammatory and autoimmune diseases, as well as downregulate an exacerbated immune response.
Isoprostadin: isoprostadins are COX-2 independent mediators of the pro-inflammatory process and of enhancing pain perception. The isoprostadins can further cause inflammation by increasing neutrophil adhesion to human venous endothelial cells and by increasing endothelial cell permeability to oxidative stress. Thus, contemplated compositions may also be useful in alleviating or preventing one or more symptoms common in various (chronic and/or subacute) inflammatory diseases, particularly symptoms associated with oxidative stress.
Thus, it should be appreciated that the compositions presented herein have unexpected pleiotropic effects on a variety of different pro-inflammatory signaling elements and signaling pathways, potentially resulting in a broad and systematic reduction of a variety of (chronic and/or subacute) inflammatory conditions. More unexpectedly, the inventors found IL-6, TNF- α and PGE 2 Is based on the synergistic effect of the various sub-mixtures present in the compositions described herein (see data below), and the sub-mixture inhibition by itself is less pronounced.
In addition to the effects on individual cytokines as described above, the inventors have also found that the compositions provided herein have significant down-regulation effects on NF- κb. In this case, it should be appreciated that NF-. Kappa.B induces the expression of various pro-inflammatory genes, including genes encoding cytokines, chemokines and COX-2, and is involved in the regulation of inflammatory corpuscles. Furthermore, NF- κb plays a key role in regulating survival, activation and differentiation of innate immune cells and inflammatory T cells, and thus has an impact on both innate and target specific immune responses.
NF- κB is highly activated at inflammatory sites of various diseases and can also induce transcription of adhesion molecules, MMPs and Inducible Nitric Oxide (iNOS). For example, in rheumatoid arthritis, NF-. Kappa.B is overexpressed in inflammatory synovial membranes, whose activity can enhance the growth of inflammatory cells and the production of pro-inflammatory mediators such as IL-1, IL-6, IL-8 and TNF-. Alpha.s. Both p50 and p65 localize to the nucleus of synovial lining cells and to monocytes in the sublining region. Helicobacter pylori-associated gastritis also appears to have increased NF- κB activity in gastric epithelial cells, and the number of NF- κB-positive cells correlates with the extent of gastritis. Also, there is evidence that NF-. Kappa.B activation in inflammatory bowel disease, lamina propria macrophages exhibit activated p50, c-Rel, especially p65. Neurological diseases and inflammation associated with atherosclerosis are also mediated to some extent by NF- κb. In addition, human inflammatory airway diseases are also associated with the expression of cytokines and adhesion molecules. This is associated with NF- κB activation in bronchial biopsies of asthmatic patients. An increase in nuclear localized NF-. Kappa.B activity was observed, particularly in airway epithelial cells, with a large number of pro-inflammatory cytokines, chemokines, iNOS and Cox-2 expressed. Other NF- κb-related diseases include atherosclerosis, multiple sclerosis, asthma, inflammatory bowel disease, and systemic inflammatory response syndrome, among others. Thus, compositions that can modulate NF- κb expression may have significant potential in addressing at least some of the symptoms associated with the above-described inflammatory conditions. Thus, contemplated compositions will again likely help alleviate or prevent one or more symptoms common in a variety of (chronic and/or subacute) inflammatory diseases.
Furthermore, particularly in cases where the Body Mass Index (BMI) of the individual is increased or overweight or obese, as discussed and shown in more detail below, contemplated compositions may have a positive effect on adipokine signaling.
The prevalence of obesity worldwide has raised great attention to studies aimed at understanding adipocyte biology and events occurring in adipose tissue and obese individuals. Interestingly, there is increasing evidence that obesity can lead to chronic low (subacute) inflammation, which can lead to systemic metabolic dysfunction associated with obesity-related diseases. In fact, adipose tissue plays a critical endocrine organ role by releasing a variety of bioactive substances, known as fat-derived secreted factors or fat factors, which in some cases have pro-or anti-inflammatory activity. Disorders in the production or secretion of these adipose factors due to dysfunction of adipose tissue may lead to pathogenesis of obesity-related complications.
Proinflammatory adiposity factor: most of the production of adipokines is up-regulated in the obese state and these pro-inflammatory proteins generally act to promote obesity-related metabolic diseases. In addition to leptin, TNF- α and IL-6, recently identified adipokines that promote inflammation include resistin, retinol binding protein 4 (RbP), lipocalin 2, IL-18, angiopoietin-like protein 2 (ANGPTL 2), CC-chemokine ligand 2 (CCL 2), CXC-chemokine ligand 5 (CXCL 5), and nicotinamide ribonucleoside transferase (NAMPT). It is the up-regulation of these factors (and others) that leads to the development of chronic inflammatory states and to metabolic dysfunction.
Anti-inflammatory fat factor: in addition to the numerous pro-inflammatory adipokines described above, adipose tissue also secretes small amounts of anti-inflammatory factors, such as adiponectin, which has been the focus of research, and sFRP5, recently identified as an adipokine.
The inventors have now found that the compositions provided herein completely inhibit the secretion of IGFBP-1, leptin, oncostatin M and resistin TNF- α induced adipocytes with respect to the secretion of adipokines, and the significance of these adipokines is set forth below.
Leptin: the fat factor leptin is the product of the obese gene (ob; also known as Lep), which was identified in ob/ob mice by positional cloning. Leptin regulates feeding behavior through the central nervous system. Leptin deficient mice (ob/ob mice) exhibit hyperphagia (abnormally increased feeding), obesity, and insulin resistance, and administration of leptin to ob/ob mice can reverse these changes. Administration of leptin to fat-bearing mice (lacking subcutaneous adipose tissue and therefore lower leptin levels) can also ameliorate metabolic abnormalities, including insulin resistance and hyperlipidemia. Leptin has also been shown to be effective in ameliorating metabolic dysfunction in lipodystrophy or congenital leptin deficiency patients. However, leptin levels in blood are positively correlated with fat mass, indicating the onset of leptin resistance, and leptin levels in obese subjects are high but without the expected anorexia response. Leptin is similar in structure to the helical cytokine family, including IL-2 and growth hormone 1, and is believed to have pro-inflammatory activity. In fact, leptin increases monocyte production of TNF and IL-6 by activating JAK2 (Janus kinase 2) -sTAT3 (signal transducer and transcriptional activator 3) pathway and stimulates macrophages to produce CC chemokine ligands (i.e., CCL3, CCL4 and CCL 5). Leptin also stimulates RO production in monocytes, promoting cell proliferation and migration reactions. Leptin levels in serum and adipose tissue are elevated in response to pro-inflammatory stimuli, including pro-inflammatory stimuli of TNF and Lipopolysaccharide (LP). In addition, leptin increases production of TH 1-type cytokines IL-2 and IFNγ and inhibits production of TH 2-type cytokine IL-4 by T cells or monocytes, thereby differentiating T cells into a TH1 cell phenotype. Consistent with these findings, leptin deficiency may protect the liver from injury in a T cell mediated hepatitis model. Thus, leptin is widely recognized as a proinflammatory adipokine.
Oncostatin M (OSM): OSM is a gp130 cytokine with its own specific receptor OSMR, heterodimerizes with gp130 and mediates most OSM actions. OSM has substantial sequence identity to Leukemia Inhibitory Factor (LIF) and can regulate various biological processes such as liver development and regeneration, liver insulin resistance and steatosis, inflammation and cardiomyocyte dedifferentiation and remodeling. In addition, OSM contributes to the inflammatory state during obesity and may be involved in the development of insulin resistance.
Resistin: resistin is a member of the cysteine-rich family of resistin-like molecules (RELm) and is involved in the activation of inflammatory processes. Resistin has been shown to induce insulin resistance in mice, and mice lacking resistin have low blood glucose levels after fasting due to low hepatic glucose production. The antibiotic deficiency of ob/ob mice results in increased obesity, but these severely obese mice have improved glucose tolerance and insulin sensitivity. The ability of resistin to regulate glucose metabolism is associated with activation of cytokine signaling inhibitor 3 (sOCs 3) in adipocytes, sOCs3 being an inhibitor of insulin signaling. Although studies in animal models have consistently shown that resistin promotes insulin resistance, the evidence of this effect in humans is unclear. Resistin exists in two quaternary forms: one is a abundant high molecular weight hexamer and the other is a less abundant but more bioactive trimer, which strongly induces liver insulin resistance. In human monocytes, transcription of the resistin gene (RETN) is induced by pro-inflammatory cytokines including IL-1, IL-6 and TNF, whereas in white adipose tissue it is inhibited by the pparγ agonist rosiglitazone, suggesting that the anti-inflammatory effect of rosiglitazone is mediated in part by attenuation of RETN transcription. Recently, studies in mice lacking endogenous resistance expression in adipocytes but expressing human RETN transgenes in macrophages have shown that the pro-inflammatory properties of macrophage derived resistin contribute to insulin resistance in vivo. The pro-inflammatory properties of resistin in human monocytes are apparent because resistin promotes the expression of TNF and IL-6 by these cells. In addition, resistin enhances leukocyte adhesion by promoting expression of pro-inflammatory adhesion molecules vascular cell adhesion molecule 1 (vCAm 1), intercellular adhesion molecule 1 (ICAm 1), and n-pentamin 3 in vascular endothelial cells, directly antagonizing the anti-inflammatory effects of adiponectin on vascular endothelial cells.
IGFBP-1: insulin-like growth factor binding protein 1 (IBP-1), also known as placental protein 12 (PP 12), is a protein encoded by IGFBP1 gene in humans. The protein binds to insulin-like growth factor (IGF) I and insulin-like growth factor (IGF) II and circulates in plasma. Binding of such proteins prolongs the half-life of IGFs and alters their interaction with cell surface receptors. The IGF system is increasingly involved in the development of cardiovascular diseases. The effects of circulating IGF on the vascular system are largely mediated by IGFBP, which controls its entry into cell surface IGF receptors. IGFBP-1 is considered an acute regulator of IGF bioavailability because it is metabolically regulated by sugar regulating hormones. Post-translational phosphorylation of IGFBP-1 significantly increases its affinity for IGF-I, thus representing a further mechanism for controlling IGF bioavailability.
Furthermore, the inventors have found that in relation to the secretion of adipokines, the compositions provided herein stimulate the secretion of ANGPT-L3, C-reactive protein, endocan, FGF-21, HGF, IGFBP-2, IL-11, RBP4 and n-pentamin 2 in TNF- α induced adipocytes (no observable stimulation in TNF- α induced adipocytes alone), and that the significance of these induced adipokines is as follows.
RBP4: serum RbP is a hepatocyte secretion factor responsible for the systemic transport of retinol (vitamin a). Recently, rbP4 was also found to be secreted by adipocytes and macrophages. RbP4 expression is inversely related to the expression of glucose transporter 4 (GluT 4; also known as sLC2A 4) and administration of recombinant RbP4 to normal mice reduces insulin sensitivity. RbP4 is released by adipocytes and inhibits insulin-induced phosphorylation of insulin receptor substrate 1 (IRs 1) in an autocrine or paracrine manner. These data indicate that RbP4 is an adipose tissue-secreting factor, important for regulation of glucose homeostasis in a model of type 2 diabetes.
ANGPTL3: angiopoietin-like protein 3 (ANGPTL 3) is known for its function as a lipoprotein and endothelial lipase inhibitor. Due to the ability of gene or pharmacologic inhibition of ANGPTL3 to significantly reduce circulating lipoproteins, and the cardioprotection of such inhibition, ANGPTL3 has become an emerging therapeutic target, and both antibody and antisense oligonucleotide (ASO) therapies are being tested clinically. Although antibodies are relatively selective for circulating ANGPTL3, ASOs also deplete intracellular proteins and there is new evidence that ANGPTL3 has cellular autonomous function in the liver. These include regulation of hepatocyte glucose and fatty acid absorption, insulin sensitivity, LDL/VLDL residual absorption, VLDL assembly/secretion, polyunsaturated fatty acids (PUFAs) and PUFA-derived lipid mediator content, and gene expression.
Fibroblast growth factor 21: fibroblast growth factor 21 (FGF 21) is a protein encoded by the FGF21 gene in mammals. The protein encoded by the gene is a member of the Fibroblast Growth Factor (FGF) family, particularly members of the endocrine subfamily, including FGF23 and FGF15/19.FGF21 is a major endogenous agonist of the FGF21 receptor and consists of the co-receptors FGF receptor 1 and β -Klotho. FGF21 is a liver factor, a hormone secreted by the liver, signals through FGF21 receptors in the parahypothalamic nucleus, regulates monosaccharide intake and preference for confectionery, and is associated with reduced dopamine transmission in the nucleus accumbens. FGF21 stimulates glucose uptake by adipocytes, but does not work with other cell types. This effect is an addition of insulin activity. Treatment of adipocytes by FGF21 is associated with phosphorylation of FRS2, a protein that links FGF receptors to the Ras/MAP kinase pathway. Injection of FGF21 in ob/ob mice resulted in an increase in Glut1 in adipose tissue. FGF21 can also protect mice from diet-induced obesity and reduce blood glucose and triglyceride levels in diabetic rodents when the transgenic mice overexpress FGF 21. Treatment of mice with FGF21 resulted in increased energy expenditure, fat utilization and lipid excretion.
Interleukin 11: interleukin 11 (IL-11) is an anti-inflammatory cytokine whose receptor is located on most cell types and tissues throughout the body. The anti-inflammatory properties are mediated by inhibition of cytokine synthesis, and in large part by preventing NF- κb activation. Because adipose tissue synthesizes and secretes cytokines that are involved in the establishment of insulin resistance, and because of the ability of IL-11 to inhibit cytokine synthesis, we began a study that established the signaling pathway that IL-11 initiates in adipose tissue.
Hepatocyte Growth Factor (HGF): obesity and its associated chronic inflammation of adipose tissue can trigger insulin resistance, which is associated with a variety of diseases such as hypertension and atherosclerosis. While wild-type mice fed a High Fat Diet (HFD) for 14 weeks had significantly increased body weight and were accompanied by insulin resistance, HGF transgenic mice prevented weight gain and insulin resistance. Accumulation of macrophages and elevation of inflammatory mediator levels in adipose tissue of HGF-transgenic mice were significantly inhibited compared to wild-type mice. HFD-induced obesity showed an increased response to glucose tolerance testing in wild-type mice treated with HGF neutralizing antibodies. These gain-of-function and loss-of-function studies indicate that HFD-induced elevated HGF levels have protective effects on obesity and insulin resistance.
IGFBP-2: proliferation of adipocyte precursors and differentiation thereof into mature adipocytes contribute to the development of obesity in mammals. IGF-I is a potent mitogen and is an important stimulator of adipocyte differentiation. The biological effects of IGF are closely regulated by the IGF-binding protein family (IGFBP), which mainly plays an inhibitory role. IGFBP-2 is the primary binding protein secreted by differentiated white preadipocytes, indicating its potential role in the development of obesity. Studies in transgenic mice that overexpress human IGFBP-2 under the control of their native promoters have shown that IGFBP-2 overexpression is associated with reduced susceptibility to obesity and increased insulin sensitivity. Although wild-type littermates develop glucose intolerance and elevated blood pressure with age, mice overexpressing IGFBP-2 are protected. Furthermore, IGFBP-2 overexpressed mice are resistant to development of obesity and insulin resistance when fed high fat/high energy diet. This lean phenotype is associated with reduced leptin levels, increased glucose sensitivity and reduced blood pressure compared to wild type animals fed a similar high fat diet. These findings indicate that IGFBP-2 plays an important role in obesity prevention.
Glucose uptake and mitochondrial biogenesis: in the elderly, skeletal muscle protein synthesis may resist insulin anabolism. Insulin resistance is also associated with activation of the muscle proteolytic pathway, which can also lead to muscle loss. In turn, muscle is the primary site of insulin-dependent glucose absorption, and insulin-mediated decrease in the surface area of the glucose absorbing muscle can further exacerbate peripheral insulin resistance, resulting in a vicious circle. Oral insulin sensitizers have been reported to maintain muscle mass, but similar associations with muscle strength have not been investigated. Interestingly, skeletal muscle mitochondrial function is reduced in type 2 diabetes and may be improved by peripheral insulin sensitization. Against this background, the inventors have found that the compositions presented herein have a significant beneficial effect on glucose absorption in muscle cells, as described in more detail below. Furthermore, the benefits of the compositions presented herein extend to improvements in mitochondrial function, particularly an increase in mitochondrial biogenesis, as shown in more detail below. Thus, it should be appreciated that the compositions presented herein not only reduce, even reverse, the age-related decline in glucose absorption into muscle, but also increase mitochondrial biogenesis.
Intracellular ATP levels were also tested to determine if the compositions presented herein could increase energy load in cells. As shown below, intracellular ATP levels do improve, particularly at higher concentrations or doses of the intended composition.
Active oxygen: reactive Oxygen Species (ROS) generally include molecular oxygen derivatives that occur as normal byproducts of aerobic metabolism, and increased formation of different ROS often results in molecular damage ("oxidative stress"). ROS, hydrogen peroxide (H) 2 O 2 ) And superoxide anion radical (O) 2 In physiological levels, can act as redox signalling agents, usually under the control of growth factors and cytokines, produced by more than 40 enzymes, mainly including NADPH oxidase and mitochondrial electron transport chains, however, prolonged signalling or ROS levels will lead to oxidative damage and accumulation of metabolic disorders. Thus, the inventors have undertaken to determine whether the intended composition is capable of counteracting oxidative stress. Notably, in addition to all beneficial functions described above, contemplated compositions exhibit significant reduction in ROS, as shown in more detail below.
Furthermore contemplated compositions are based on the composition of the Mediterranean diet, which is known to be common to the longevity population, the inventors have also undertaken to investigate whether the compositions provided herein would have an effect on senescence-associated genes, and the following exemplary results demonstrate significant downregulation of various senescence-associated genes.
Acetylcholinesterase (AChE) is an enzyme that is mainly present in blood and nerve synapses, and plays an important role in various cognitive processes. Acetylcholinesterase catalyzes the hydrolysis of the neurotransmitter acetylcholine to choline and acetic acid, which is the reaction necessary to restore the resting state after cholinergic neuron activation. Acetylcholinesterase inhibitors (e.g., using donepezil) are important targets for the treatment of alzheimer's disease, with acetylcholinesterase inhibitors being the most common drug used to treat alzheimer's disease. In addition to Alzheimer's disease, acetylcholinesterase inhibitors may also be used to diagnose or treat glaucoma, myasthenia gravis, bladder distension, and other disorders. Thus, acetylcholinesterase inhibitors are believed to have a cognitive-promoting effect. To this end, the inventors also determined whether the compositions provided herein have an inhibitory effect on AChE. Notably, as demonstrated in more detail below, the compositions provided herein have significant inhibitory effects on AChE and thus can significantly improve cognition, as well as other benefits.
In a further contemplated aspect of the inventive subject matter, it is to be understood that the compositions provided herein may be formulated in a variety of forms, and particularly preferred formulations include those in combination with a nutritional or pharmaceutically acceptable carrier, most preferably for oral administration (however, parenteral administration is also expressly contemplated). Thus, contemplated compositions may be formulated as solid or liquid products. . For example, when the contemplated composition is formulated as a solid product, suitable product forms include single dose unit formulations such as capsules, tablets, and powders, while other solid formulations include snack bars, fondants, or other edible products, the composition being coated on the edible product (e.g., on a cereal) or the composition being mixed or layered in the other edible product (e.g., into a chewing gum). In another example, when the contemplated composition is formulated as a liquid product, suitable product forms include flavored and/or carbonated beverages (e.g., tea, juice), functional beverages (e.g., sports or energy beverages), or brewed beverages, or liquid dairy products (e.g., yogurt, kefir).
Thus, contemplated compositions may be provided in bulk as part of an edible or drinkable product, and/or in a single dosage unit for consumption. Most typically, preferably, the contemplated daily dose of the composition (excluding the carrier) is at least 10mg, or at least 50mg, or at least 100mg, or at least 200mg, or at least 300mg, or at least 400mg, or at least 500mg, or at least 750mg, or at least 1000mg, or at least 1500mg. For example, a suitable dosage is 10mg to 50mg, or 50mg to 100mg, or 100mg to 200mg, or 200mg to 400mg, or 300mg to 600mg, or 400mg to 800mg, or 600mg to 1000mg, or 1000mg to 2000mg.
Most typically, contemplated compositions will be administered orally for a prolonged period of time, such as at least 1 week, or at least 2 weeks, or at least 4 weeks, or at least 2 months, or at least 3 months, at least 6 months, or at least 1 year and greater to maintain or support healthy aging. Preferably, the administration is daily, or at least twice a week or at least three times a week. In some embodiments, the contemplated compositions may also be administered topically, typically in the form of a cream, lotion, essence, or elixir or shampoo. In this case, the composition may be a (optionally filtered) aqueous or hydroalcoholic extract, resulting in a clear solution.
As will be readily appreciated, contemplated compositions may also be combined with one or more additional ingredients to impart further desired functionality, and suitable additional ingredients include vitamins (e.g., a single vitamin or vitamin mixture such as a multivitamin mixture), dietary trace elements or minerals (e.g., a single element or mineral, or a mixture of multiple elements or minerals in multiple forms), various specific compounds and mixtures (e.g., prebiotics, human milk oligosaccharides), and/or one or more probiotic microorganisms (e.g., lactobacillus, bifidobacterium, leukotrichia, saccharomyces, etc.), prebiotics (e.g., short chain fatty acids such as butyric acid, secondary bile acids such as (taurine) ursodeoxycholic acid, etc.), dietary fibers (e.g., soluble or insoluble fibers), and/or nutritionally acceptable oligosaccharides (e.g., xylooligosaccharides (XOS) or Fructooligosaccharides (FOS)). Similarly, contemplated compositions may also include niacin, nicotinamide riboside, nicotinamide mononucleotide, nicotinamide adenine dinucleotide, and/or a nutritionally acceptable CD38 inhibitor, thereby increasing intracellular NAD and/or NADP levels (and their reduced forms NADH and NADPH, respectively). Thus, contemplated compositions may also specifically (synergistically) enhance or support cellular metabolism and energy.
Of course, it should be appreciated that compositions according to the inventive subject matter may be administered not only to humans, but also to other non-human mammals, particularly livestock and companion animals (e.g., dogs, cats, horses). The administration is typically between once a day and three times a day (in some cases even more) over a period of at least two days, three days, five days, one week, 2 to 4 weeks, 1 to 3 months, or even more than 3 months. Most typically, the time of administration is sufficient to at least provide symptomatic relief (e.g., pain and swelling associated with inflammation, low energy levels, frequent infections, etc.), or to prophylactically avoid or help reduce the severity of the health condition.
Examples
Representative compositions:
all experiments were performed using defined mixtures of polyphenol containing products/polyphenol containing extracts common in selected mediterranean diets, unless otherwise indicated. The polyphenol-containing product/polyphenol-containing extract is obtained from a raw material having the following color characteristics: red group (sub-mix): apple extract, pomegranate extract, tomato powder, beetroot; green group (sub-mix): olive extract, rosemary extract, green coffee bean extract and kale; orange/yellow group (sub-mixture): onion extract, ginger extract, grapefruit extract, and carrot; and violet/blue group (sub-mixture): grape extract, blueberry extract, currant powder and elderberry powder. Corn starch, silica and sunflower lecithin are used as processing aids. The relative proportions are shown in table 1 below.
TABLE 1
Phytochemical HPLC/MS analysis: HPLC/MS compositional analysis of the above exemplary compositions reveals the following ingredients and ratios, wherein the columns of each of tables 2-8 indicate analyte ID (column 1), chemical entity (column 2), M-H (column 3), RT (column 4), peak intensity (column 5), and MS/MS fragment (column 6):
TABLE 2
TABLE 3 Table 3
TABLE 4 Table 4
TABLE 5
TABLE 6
TABLE 7
TABLE 8
The activity of representative compositions was tested for modulation of various target entities associated with mild/chronic inflammation, age-related immune decline, energy metabolism, aging, reactive oxygen species, acetylcholinesterase activity, obesity and viability, unless specifically stated otherwise, with exemplary activity results shown below.
Cytokines:
in the following experiments, the inventionA series of tests were performed by humans to determine the effect of representative compositions and sub-mixtures on various cytokines. For this purpose, the tested compositions were combined with LPS-treated primary human monocytes and SEB-induced T lymphocytes (helper T cells, CD4 + ) Together to determine a specific response with respect to the selected pro-inflammatory cytokine. Unless otherwise indicated, all test materials were provided as solid powders dissolved in water.
Here, the inventors used primary human monocytes/lymphocytes to test the anti-inflammatory effect of the test article, as described in detail below using the mature and published methods. Human monocytes are one of the major cell types involved in peripheral inflammation, as they are primarily affected by bacterial Lipopolysaccharide (LPS) produced by gram-negative bacteria. In addition, monocyte release of cytokines is also considered to be the first step in the inflammatory cascade, typical cytokines include interleukin-1 (IL-1) beta, IL-6, IL-8, IL-23 and Tumor Necrosis Factor (TNF) alpha, as well as other pro-inflammatory cytokines such as prostaglandin E2 (PGE) 2 ) Certain chemokines such as monocyte chemotactic protein-1 (MCP-1, also known as CCL 2), and macrophage inflammatory protein-1 (MIP-1) as well as free radical and oxidative stress markers such as isoprostadins. While monocytes can also trigger inflammatory cascades in other cell types, such as fibroblasts and the like, T cells respond to cytokine signals and play a central role in the adaptive immune response.
T cells can be easily distinguished from other lymphocytes by the presence of T Cell Receptors (TCRs) on the surface of the T cells. Interleukin-2 (IL-2) is a 15.5kDa to 16kDa protein consisting essentially of activated CD4 + T cells (helper T cells) are produced, for example, by enterotoxin type B, also known as Staphylococcal Enterotoxin B (SEB), an enterotoxin produced by the gram positive bacterium staphylococcus aureus. IL-2 regulates the activity of leukocytes (leukocytes, typically lymphocytes) responsible for immunity. IL-2 is part of the natural response of the human body to microbial infection, as well as part of distinguishing between exotic ("non-self") and "self". The parameters studied are recognized mediators of inflammation, all of which are knownIs associated with inflammation and plays an important role in the inflammatory cascade by inducing pain and inflammation.
Determination of primary human monocyte cytokines: human primary monocytes are isolated (enriched) from the buffy coat of a healthy human donor. Cells were seeded in 24-well plates (about 500000 cells/ml in 1 ml) for ELISA experiments. Cells were incubated with LPS (10 ng/ml) for 24 hours. Representative compositions (5 doses) and dexamethasone were added as anti-inflammatory controls 30 minutes prior to LPS treatment (untreated cells as negative control). After 24 hours, the supernatant was removed, centrifuged, and the required inflammatory parameters, such as MCP-1, IL-8, IL-6, IL-23 and TNF-alpha concentrations, were studied using ELISA (MCP-1, TNF-alpha, IL-6, IL-23 and IL-8, R & D/Biotechne) using the manufacturer's protocol. Each dose was studied 6 times in two to three buffy coats from 2 to 3 different donors (n=2 to 3 per buffy coat, n=6 total).
Determination of primary human monocytes PGE2 and isoprostadins (radical markers): human primary monocytes are isolated (enriched) from the buffy coat of a healthy human donor. Cells were seeded in 24-well plates (about 500000 cells/ml in 1 ml) for EIA experiments. Cells were incubated with LPS (10 ng/ml) for 24 hours. Representative compositions (5 doses) and dexamethasone were added as anti-inflammatory controls 30 minutes prior to LPS treatment (untreated cells as negative control). After 24 hours, the supernatant was removed, centrifuged, and the concentration of isoprostadin and PGE2 was studied using EIAs (from Cayman, distributed by Biomol, hamburg, germany). Study was performed 6 times in each dose on two to three buffy coats from 3 different donors (n=2 to 3 per buffy coat, n=6 total).
Determination of IL-2 in primary T cells: human primary T cells are isolated (enriched) from the buffy coat of a healthy human donor. Cells were seeded in 24-well plates (about 500000 cells/ml in 1 ml) for ELISA experiments. Cells were incubated with SEB (1. Mu.g/ml) for 24 hours. Representative compositions (5 doses) and dexamethasone or hydrocortisone were added as anti-inflammatory controls 30 minutes prior to SEB treatment (untreated cells as negative control). After 24 hours, the supernatant was removed, centrifuged, and the IL-2 concentration was studied using ELISA (Biotechne, wiesbaden, germany) using the manufacturer's protocol (other parameters are also possible). Each dose was studied 6 times in two to three buffy coats from 2 to 3 different donors (n=2 to 3 per buffy coat, n=6 total).
As shown in the data shown in fig. 1-7, representative compositions had a significant effect on a variety of pro-inflammatory markers in LPS-treated primary human monocytes. As shown in the summary of fig. 1, representative compositions effectively and significantly inhibited LPS-induced release of IL-6, TNF- α and PGE2 starting from a low dose of 10 μg/ml. LPS-induced release of the isoprostagliptin (PGF 2-alpha), a free radical marker, was inhibited in high doses of the extract. LPS-induced IL-23 release was not affected, whereas IL-8 and MCP1 levels were slightly increased. The composition enhances LPS-mediated release of IL-1β. The anti-inflammatory agent and positive control dexamethasone effectively exclude all parameters except IL-23. Fig. 2 shows the inhibition of various LPS-induced pro-inflammatory cytokines in primary human monocytes by representative compositions (p < 0.05, p < 0.01, and p < 0.001 relative to LPS control (T-test)). More particularly, fig. 3 shows the inhibition of LPS-induced TNF- α release in primary human monocytes (p < 0.05, p < 0.01, and p < 0.001 relative to LPS control (T-test)) by representative compositions. Fig. 4 shows the inhibition of LPS-induced IL-6 release in primary human monocytes (relative to LPS control (T-test), p < 0.05, p < 0.01 and p < 0.001) by representative compositions. Fig. 5 shows the inhibition of LPS-induced PGE2 release in primary human monocytes (relative to LPS control (T-test), p < 0.05, p < 0.01 and p < 0.001) by representative compositions. Fig. 6 shows the inhibition of LPS-induced release of prostaglandins in primary human monocytes (relative to LPS control (T-test), < p < 0.05, < p < 0.01, and < p < 0.001) by representative compositions.
As can be further seen from the data in fig. 7, representative compositions also have a significant effect on IL-2 synthesis in SEB treated primary human monocytes (primary human T cells). Here, representative compositions significantly inhibited LPS-induced IL-2 at higher doses (e.g., 250 μg/ml). Anti-inflammatory and positive control dexamethasone effectively prevented IL-2 synthesis during the course of the experiment as described above. Fig. 7 illustrates the inhibition of SEB-induced IL-2 release in primary human lymphocytes by representative compositions (p < 0.05, p < 0.01, and p < 0.001 relative to SEB control (T-test)). Notably, it should be appreciated that the representative compositions are effective in attenuating or reducing pro-inflammatory signaling while only moderately or not reducing T cell stimulation at all (which is typically required for antigen-specific immune responses).
To investigate any potential active components in a representative composition, the inventors subsequently conducted experiments as shown above, but using only the specific sub-mixtures as described above. To this end, the inventors tested a red group sub-mixture, a green group sub-mixture, an orange/yellow group sub-mixture, and a violet/blue group sub-mixture, and TNF- α, IL-6, and PGE 2 Typical results for (a) are shown in fig. 8-11 (where p < 0.05, < p < 0.01 and p < 0.001 relative to the LPS control (T-test)). Fig. 8 provides a summary of the green group sub-mixture inhibition data, fig. 9 provides a summary of the red group sub-mixture inhibition data, fig. 10 provides a summary of the orange/yellow group sub-mixture inhibition data, and fig. 11 provides a summary of the violet/blue group sub-mixture inhibition data.
As can be readily seen from the data in fig. 8, inhibition of cytokine release of TNF- α and IL-6 using the green sub-mixture was regulated in a dose dependent manner only, without significant PGE at all concentrations tested 2 Release inhibition. On the other hand, as shown in fig. 9, no significant inhibition of all cytokines tested was observed when using the red sub-mix. However, IL-6 and TNF- α increased in the higher concentration of the red sub-mixture. Similarly, the orange/yellow sub-mixture did not produce significant inhibition or increase in all cytokines tested as shown in fig. 10. As shown in FIG. 11, there was only a slight inhibition of TNF- α in the case of using the purple/blue sub-mixture.
Notably, it can be quite tolerant from FIG. 12 It is readily seen that (where p < 0.05, p < 0.01 and p < 0.001 relative to the LPS control (T-test)) when four sub-blends were used together, TNF- α, IL-6 and PGE were observed 2 Is a strong inhibitory synergy of (a). This synergistic profile is not only unexpected, but also strongly suggests that synergistic combinations can be effective in alleviating the signs and symptoms of various inflammatory conditions (e.g., subacute, chronic, disease-related, allergy-related, etc.) in a variety of different pro-inflammatory pathways. This multi-pathway inhibition is considered to be a significantly more effective and physiologically well-tolerated anti-inflammatory immunomodulation than traditional anti-inflammatory drugs. Furthermore, since all of the ingredients in a representative composition are derived from food products, in many cases (due to targeting a variety of different pathways) inflammation can be reduced in a beneficial manner without the side effects common to conventional anti-inflammatory agents.
To investigate whether representative compositions would also have an effect on other tissues (e.g., muscle cells), the inventors determined the mRNA expression of IL-6 and TNF- α in mouse C2C12 muscle cells (immortalized mouse myoblast cell lines). C2C12 cells are supplied by university of koldo, spain. Cells were maintained in Dulbecco's modified Eagle's medium containing 10% Fetal Bovine Serum (FBS), 4.5mM L-glutamine, 1% penicillin and streptomycin antibiotics. For all experiments, cells were grown to 80% to 90% confluency in 24 well plates.
RNA isolation and quantitative PCR: the cultured cells were incubated with representative compositions (1. Mu.g/ml to 100. Mu.g/ml) at various concentrations dissolved in water for 4 hours. Total RNA was extracted using Universal RNA Kit-roboklon. Use of Moloney mouse leukemia Virus (M-MLV) reverse transcriptase (Promega, mannheim, germany), RNase inhibitor(Promega, mannheim, germany), dNTP master mix (Promega, mannheim, germany) and random hexamer primers (Biomers, germany) were reverse transcribed to cDNA from 1. Mu.g of total RNA. Real-time PCR amplification was performed by qTOWER2.0/2.2 quantitative real-time PCR Thermal Cyclers, anaytik Jena using ORA TM qPCR Green ROX H Mix,2X-highQu GmbH (g Lai Xida mol)Germany). The reaction conditions were 3 minutes at 95℃followed by 40 cycles of 15 seconds at 95℃30 seconds at 50℃and 45 seconds at 72℃each followed by a plate read. Thereafter, at 95℃for 1 minute, at 55℃for one minute, then at 65℃and 95℃under melt profile conditions, the increment was 0.5℃for 5 seconds, and then the final plate reading was performed. Glyceraldehyde 3-phosphate dehydrogenase (GAPDH) was used as an internal control for sample normalization and data quantification was performed using the comparative cycle threshold Ct method.
The effect of representative compositions on IL-6 and TNF- α mRNA levels in C2C12 muscle cells is depicted in the exemplary results of fig. 13 and 14 (p < 0.05, p < 0.01, p < 0.001 in both results). It is apparent that representative compositions were effective and dose-dependently reducing IL-6 and TNF- α mRNA levels starting at a dose of 10 μg/ml. Thus, it should be appreciated that representative compositions have profound effects on the synthesis and secretion of selected cytokines.
NFκB:
In the following experiments, the inventors performed a series of experiments to determine the effect of representative compositions on nfkb, as NF-kb is one of the most important mediators of pro-inflammatory gene expression. For example, the synthesis of various cytokines such as TNF- α, IL-1β, IL-6 and IL-8 is mediated by NF-. Kappa.B, as is the expression of cyclooxygenase 2 (Cox-2).
To determine NF- κB transcriptional activity, the inventors used an assay kit for HEK293t cells that expressed NF- κB (nuclear factor- κ light chain enhancer of activated B cells) and contained a luciferase reporter gene functionally linked to an upstream NF- κB-genetic response element. Thus, quantifying changes in luciferase expression provides a sensitive surrogate measure of changes in NF- κb activation levels. NF-. Kappa.B is a signal transduction dependent transcription factor. NF- κb reporter cell lines have been shown to provide a strong dose-dependent activation response when treated with tnfα or the protein kinase C activator phorbol 12-myristate 13-acetate (PMA). Thus, the assay is particularly suitable for screening test samples to quantify any functional activity they may induce or inhibit NF- κb activity. Here, NF-. Kappa.B transcriptional activity assays (commercially available from INDIGO Biosciences (Biomol)) were performed according to the manufacturer's instructions.
The effect of representative compositions was observed using the assays described above. It is clear from the data in fig. 15 that at low doses (1 μg/ml and 10 μg/ml, possibly the preparation of the test system) PMA-induced NF- κb activation was enhanced, whereas at high doses, starting from a dose of 50 μg/ml, NF- κb activation was strongly and dose-dependently prevented and almost completely inhibited using extracts of 100 μg/ml and 250 μg/ml, (. P < 0.05, (. P < 0.01, (. P < 0.001)). Notably, this inhibition is more potent than the known NF- κb inhibitor QNZ. As indicated above, this NF- κb inhibition was also consistent with the inhibition observed on various pro-inflammatory cytokines and pathways.
Glucose absorption:
in the following experiments, the inventors performed a series of tests to determine the effect of a representative component on energy metabolism, in particular on glucose absorption.
For this, C2C12 cells (5X 10 4 ) Inoculated in 96-well plates and incubated for 24 hours. The medium was then removed and the cells were cultured in OptiMEM with 50. Mu.M 2-NBDG (2- [ N- (7-nitrobenzene-2-oxa-1, 3-diazol-4-yl) amino)]-2-deoxy-D-glucose labelling and treatment with representative composition or positive control rosiglitazone for 24 hours. The medium was removed, wells carefully washed with PBS and incubated in PBS (100. Mu.l/well). Finally, fluorescence was measured according to the manufacturer's instructions.
Fig. 16 is an exemplary result of the effect of representative components on glucose uptake by mouse C2C12 muscle cells (< p <0.05, < p <0.01, < p < 0.001). As can be easily seen from the data in the figures, the effect of the representative composition on glucose absorption is very high and does have an effect comparable to that of the known antidiabetic compound rosiglitazone. Thus, it should be appreciated that the representative compositions have significant antidiabetic therapeutic effects and significantly promote cellular energy.
Mitochondrial biogenesis:
in anticipation of a significant increase in available energy substrates, the inventors conducted a series of experiments to investigate whether the increased energy metabolites could be effectively used for glycolysis and cellular respiration. Since cellular respiration requires mitochondrial activity, the inventors sought to determine the effect of representative compositions on mitochondrial biogenesis in neuronal cells.
To this end, neuro-2a (N2 a) (ATCC, manassas, va., USA) cells were cultured in DMEM supplemented with 10% FBS, 2mM 1-glutamine and 1% (v/v) penicillin/streptomycin. Maintaining the cells at 37deg.C with 5% CO 2 Is in a humid atmosphere.
Measurement of mitochondrial biogenesis. N2a cells were seeded in 96-well plates (3.5X10 per well) 3 Cells), 24 hours later, with an increasing concentration of representative composition (5 doses) in each well for 72 hours. Then Mitotracker Green (100nM;Thermo Fisher Scientific,Waltham,MA,USA) was added to the medium for 30 minutes. Cells were washed with PBS and fresh medium was added. Images were taken and fluorescence measured using a cell imaging system, inticutehd (Essen BioScience, inc., herfordshire, uk). Rosiglitazone served as a positive control.
As can be readily seen from the results of fig. 17, representative compositions significantly increased mitochondrial biogenesis. Unexpectedly, mitochondrial biogenesis using this representative composition was superior to the positive control rosiglitazone in this assay.
ATP:
In the following experiments, the inventors performed a series of tests to determine the effect of representative components on intracellular ATP levels. For this, the inventors used RAW264.1 mouse macrophages and C2C12 mouse myocytes (from Uniklinik Freiburg, germany).
The cells were placed in DMEM complete broth (DMEM complete broth) containing 10% fetal bovine serum and 1% antibiotic penicillin/streptomycin, 37 ℃,5% CO 2 Is maintained in a humid atmosphere. For ATP assays, RAW cells and C2C12 were assayed at 2x10 4 Density of individual cells/wells seeded atIn 96-well plates and in DMEM medium at 37℃in 5% CO 2 Is incubated overnight in a humid atmosphere. Cell cultures were then stimulated with representative compositions at selected concentrations (5 doses, n=4). By passing through2.0 cell viability/ATP assay to determine ATP in cells.
Notably, as shown in fig. 18, only a low dose of 1 μg/ml of the representative ingredient had a modest increase in ATP levels in macrophages (where p <0.05, < p <0.01, < p < 0.001). Other doses were not effective. In C2C12 cells, the inhibition of ATP levels by representative ingredients was weak but dose dependent (see fig. 19, where p <0.05, < p <0.01, < p < 0.001).
Reactive Oxygen Species (ROS):
in the following experiments, the inventors performed a series of experiments to determine the effect of representative components on the antioxidant capacity of cells by measuring Reactive Oxygen Species (ROS) in the macrophage model system.
For this, the inventors used RAW 264.1 mouse macrophages from germany Uniklinik Freiburg. The cells were placed in DMEM complete broth (DMEM complete broth) containing 10% fetal bovine serum and 1% antibiotic penicillin/streptomycin at 37 ℃ and 5% CO 2 Is maintained in a humid atmosphere. Cellular responses were studied in the presence of representative compositions and controls (no compound as negative control, trolox and vitamin C as positive control). From the results of FIG. 19, it is clear that the representative ingredients can effectively reduce H starting from a relatively low dose of 1. Mu.g/ml 2 O 2 Activation of induced ROS levels. Notably, maximum ROS inhibition was achieved using 50 μg/ml of DC, which was comparable in potency to Trolox C, one of the most potent and most commonly used antioxidants.
Fat factor:
in the following experiments, the inventors conducted a series of experiments to determine the effect of representative ingredients on various fat factors associated with obesity and metabolic diseases. More specifically, the inventors used 3T3-L1 cells (preadipocyte fibroblasts) in a standard cell culture model and used a proteome analysis kit to determine the expression levels of a set of expanded adipokines.
Low passage 3T3-L1 cells (ATCC) were cultured in DMEM containing 25mM glucose, 4mM L-glutamine and 1mM sodium pyruvate, 10% (v/v) fetal bovine serum (abbreviated as DMEM/FBS). Cells were plated in 12-well plates at 37℃with 5% (v/v) CO 2 Is cultured in a humidified incubator. Two days after confluence, induced differentiation into adipocytes with a stimulator mixture, 2 μg/mL insulin, 0.5mM isobutyl methylxanthine, 0.25 μΜ dexamethasone and 2 μΜ rosiglitazone (all purchased from Sigma) in DMEM/FBS; . Three days later, the differentiation medium was removed, and the cells were cultured in the post-differentiation medium (DMEM/FBS and 2μg.mL -1 Insulin) for a further 6 to 9 days, with medium changes every two days or as needed. Differentiation was monitored daily by microscopy.
After differentiation, cells were washed with PBS, incubated in complete DMEM medium, and treated with TNF alpha (50 ng/ml) for 24 hours in the absence and presence of representative compositions (10. Mu.g/ml and 100. Mu.g/ml in water). The supernatant is collected and assayed for detection of adipokine and other related soluble mediators. The fat factor and other obesity-related proteins were detected using a semi-quantitative proteome archiver mouse fat factor array kit (R & DSsystem; minneapolis, MN, USA) according to manufacturer's recommendations. The pixel density on the developed X-ray film was collected with a scanner and analyzed using the ImageJ processing and analysis program (NIH; bethesda, MD, USA).
The following fat factors and soluble media were monitored simultaneously: adiponectin, agRP, ANGPT-L3, C-reactive protein, DPPIV, endocan, fetuin A, FGF acid, FGF-21, HGF, ICAM-1, IGF-I, IGF-II, IGFBP-1, IGFBP-2, IGFBP-3, IGFBP-5, IGFBP-6, IL-10, IL-11, leptin, LIF, lipocalin-2, MCP-1, M-CSF, oncostatin M, n-pentameric protein 2, n-pentameric protein 3, pref-1, RAGE, RANTES, RBP4, resistin, serpin E1, TIMP-1, TNF-alpha, and VEGF.
FIGS. 19-22 depict selected effects of representative compositions on obesity-related protein secretion in differentiated 3T3-L1 cells. Most notably, tnfα induced secretion of IGFBP-1, leptin, oncostatin M, and resistin, and such secretion was completely inhibited by the representative composition, as shown in fig. 21. In contrast, as shown in FIGS. 22 and 23, ANGPT-L3, C-reactive protein, endocan, FGF-21, HGF, IGFBP-2, IL-11, RBP4 and n-pentameric protein 2 were significantly induced by representative compositions of TNFα+ but not only TNFα, indicating that synthesis of these proteins was induced by representative compositions.
Aging:
in the following experiments, the inventors performed a series of experiments to determine the effect of representative components on the expression of selected senescence marker genes in Normal Human Dermal Fibroblasts (NHDF).
NHDF cells were purchased from (ATCC, germany). NHDF cells were cultured in Dulbecco's modified Eagle's medium containing 10% Fetal Bovine Serum (FBS), 4.5mM L-glutamine, 1% penicillin and streptomycin antibiotics. NHDF cells wet 5% CO at 37 ℃ 2 And (3) growing in the middle. For all experiments, cells were grown to 80% to 90% confluency in 24 well plates. After 24 hours of cell culture, the medium was changed and NHDF cells were kept for 6 days to age.
Real-time fluorescent quantitative PCR (qPCR) detection was performed on aged normal human dermal fibroblasts. The cultured cells were incubated with representative compositions (1. Mu.g/ml to 100. Mu.g/ml) at various concentrations dissolved in water for 4 hours. Total RNA was extracted using Universal RNA Kit-roboklon. Use of Moloney mouse leukemia Virus (M-MLV) reverse transcriptase (Promega, mannheim, germany), RNase inhibitor(Promega, mannheim, germany), dNTP master mix (Promega, mannheim, germany) and random hexamer primers (Biomers, germany) were reverse transcribed to cDNA from 1. Mu.g of total RNA. Real-time PCR amplification was performed by qTOWER2.0/2.2 quantitative real-time PCR Thermal Cyclers, Anaytik Jena uses ORA TM qPCR Green ROX H Mix,2X-highQu GmbH (gram Lai Xida mol, germany). Primers were designed to amplify the primers encoding HGF, c-fos, p16 INK And p 21. The reaction conditions were 3 minutes at 95℃followed by 40 cycles of 15 seconds at 95℃30 seconds at 50℃and 45 seconds at 72℃each followed by a plate read. Thereafter, at 95℃for 1 minute, at 55℃for one minute, then at 65℃and 95℃under melt profile conditions, the increment was 0.5℃for 5 seconds, and then the final plate reading was performed. Glyceraldehyde 3-phosphate dehydrogenase (GAPDH) was used as an internal control for sample normalization and data quantification was performed using the comparative cycle threshold Ct method.
Notably, it can be seen from FIG. 24 that the representative ingredients strongly inhibited the senescence-associated genes HGF, c-fos, p16 even at very low doses INK And expression of p 21.
Acetylcholinesterase (AChE):
in the following experiments, the inventors performed a series of experiments to determine the effect of a representative ingredient on acetylcholine esters as potential targets for neural function.
For this purpose, the inventors used the amplite colorimetric assay (from AATBioquest) according to the manufacturer's instructions. As negative controls, 40 μl NIC (no inhibitor control) assay buffer, 40 μl AHE solution and 40 μl stock solution of diluted donepezil in assay buffer were pipetted into separate wells of a 96-well plate. For negative control, 10. Mu.l of acetylcholinesterase solution (4 mU/10. Mu.l) of NIC and test solution were added to each well. The control sample and the sample with the representative composition were incubated at room temperature with slow shaking for 15 minutes. To each well 50 μl of acetylcholinesterase working solution was added to make the total acetylcholinesterase assay volume 100 μl/well (t=0 minutes). The reaction was incubated at room temperature for 60 min, protected from light (t=60 min). At time points t=0 min and t=60 min, absorbance was measured at 405nm using a PerkinElmer Victor X52030-0050 multimode plate reader of PerkinElmer (Rodgau, germany).
Exemplary test results are shown in fig. 25, illustrating the effect of representative ingredients on acetylcholinesterase activity. Results are expressed in relative units ± SD (×p <0.05, ×p <0.01, ×p < 0.001). It is readily apparent that the representative ingredients have a significant inhibitory effect on acetylcholinesterase at higher concentrations.
Vitality:
in the following experiments, the inventors performed a series of tests to determine the effect of representative ingredients on macrophage viability. Here, RAW 264.1 mouse macrophages were obtained from Uniklinik Freiburg and cultured in supplemented DMEM medium (DMEM complete medium) containing 10% FBS and 1% antibiotic penicillin/streptomycin at 37℃at 5% CO 2 Is maintained according to standard protocols.
Cytotoxicity assay by plating RAW cells at 2X10 4 Density of individual cells/well was seeded in 96-well plates in DMEM medium at 37 ℃ at 5% CO 2 Is incubated overnight in a humid atmosphere. Cell culture was then stimulated with representative ingredients at selected concentrations (7 doses, n=4), and cell viability was determined by Alamar Blue staining with NaF as a toxicity control. Example results are shown in fig. 26. As can be seen from the results, only high doses of 500. Mu.g/ml and above have a certain effect on cell viability and cell metabolism, whereas lower concentrations have no statistically significant effect on cell viability and cell metabolism.
Other aspects, considerations, and contemplations suitable for use herein are disclosed in U.S. patent No. 11,065,295, which is incorporated herein by reference.
In some embodiments, the numbers expressing quantities of ingredients, properties such as concentration, reaction conditions, and so forth, used to describe and claim certain embodiments of the present invention are to be understood as being modified in some instances by the term "about". Accordingly, in some embodiments, the numerical parameters set forth in the written description and attached claims are approximations that may vary depending upon the desired properties sought to be obtained by the particular embodiment. Recitation of ranges of values herein are merely intended to serve as a shorthand method of referring individually to each separate value falling within the range. Unless otherwise indicated herein, each separate value is incorporated into the specification as if it were individually recited herein.
As used herein, the term "administering" a pharmaceutical or nutraceutical composition refers to the direct and indirect administration of a pharmaceutical or nutraceutical, wherein the direct administration of the pharmaceutical or nutraceutical is typically by a health care professional (e.g., doctor, nurse, dietician, etc.), and wherein the indirect administration comprises the step of providing the pharmaceutical or nutraceutical composition to the health care professional or individual in need thereof for direct administration (e.g., by injection, infusion, oral delivery, topical delivery, etc.). It should further be noted that the term "predicting" or "predicting" a disorder, a susceptibility to disease progression, or a response to an intended treatment is intended to encompass predicting (but not treating or diagnosing) a behavior of the disorder, the susceptibility, and/or the response, including the rate of progression, the rate of improvement, and/or the duration of the subject's condition.
All methods described herein can be performed in any suitable order unless otherwise indicated herein or otherwise clearly contradicted by context. The use of any and all examples, or exemplary language (e.g., "such as") provided with respect to certain embodiments herein, is intended merely to better illuminate the invention and does not pose a limitation on the invention claimed. No language in the specification should be construed as indicating any non-claimed element as essential to the practice of the invention.
As used in the specification herein and throughout the claims that follow, a myriad of words in front of an element may mean more than one unless the context clearly dictates otherwise. Furthermore, as used in the description herein, unless the context clearly dictates otherwise, the meaning of "in … …" includes "in … …" and "on … …". As used herein, unless the context dictates otherwise, the term "coupled to" is intended to include both direct connections (where two elements connected to each other are in contact with each other) and indirect connections (where at least one additional element is located between two elements). Thus, the terms "coupled to" and "coupled to …" are used synonymously.
It will be apparent to those skilled in the art that many more modifications besides those already described are possible without departing from the inventive concepts herein. The inventive subject matter, therefore, is not to be restricted except in the scope of the appended claims. Furthermore, in interpreting both the specification and the claims, all terms should be interpreted in the broadest possible manner consistent with the context. In particular, the terms "comprises" and "comprising" should be interpreted as referring to elements, components, or steps in a non-exclusive manner, indicating that the referenced elements, components, or steps may be present, or utilized, or combined with other elements, components, or steps that are not expressly referenced. When the specification or claims refer to at least one selected from A, B, C … … and N. The text should be interpreted as requiring only one of the elements, not a plus N, or B plus N, etc.
Claim (modification according to treaty 19)
1. A nutritional composition comprising:
a nutritionally acceptable carrier in combination with a plant material having red, green, orange and violet colors comprising a plurality of chemically different polyphenols;
wherein the red plant material, green plant material, orange plant material, and violet plant material are present in synergistic amounts that reduce the release of proinflammatory cytokines in human leukocytes.
2. The composition of claim 1, wherein the red plant material comprises apple extract, pomegranate extract, tomato powder, and beetroot powder, wherein the green plant material comprises olive extract, rosemary extract, green coffee bean extract, and collard powder, wherein the orange plant material comprises onion extract, ginger extract, grapefruit extract, and carrot powder, and/or wherein the violet plant material comprises grape extract, blueberry extract, currant powder, and elderberry powder.
3. The composition of claim 1 or 2, wherein the release of pro-inflammatory cytokines is tumor necrosis factor alpha (TNF-alpha), interleukin-6 (IL-6), prostaglandin E 2 (PGE 2 ) And release of at least one of the isoprostadins.
4. Composition according to claim 1 or 2Wherein the release of the pro-inflammatory cytokine is tumor necrosis factor alpha (TNF-alpha), interleukin-6 (IL-6), prostaglandin E 2 (PGE 2 ) And release of at least two of the isoprostadins.
5. The composition of claim 1 or 2, wherein the release of the pro-inflammatory cytokine is the release of at least three of tumor necrosis factor alpha (TNF-a), interleukin-6 (IL-6), prostaglandin E2 (PGE 2) and isoprostadins.
6. The composition of claim 1 or 2, wherein the release of pro-inflammatory cytokines is tumor necrosis factor alpha (TNF-alpha), interleukin-6 (IL-6), prostaglandin E 2 (PGE 2 ) And release of isoprostadins.
7. The composition of claim 1, wherein the composition further reduces expression of nfkb.
8. The composition of claim 1, wherein the composition further increases glucose uptake by cells.
9. The composition of claim 1, wherein the composition further increases the biogenesis of mitochondria within a cell.
10. The composition of claim 1, wherein the composition further reduces oxidative damage caused by reactive oxygen species.
11. The composition of claim 1, wherein the composition further reduces the expression of a proinflammatory adipokine.
12. The composition of claim 1, wherein the composition further reduces expression of a senescence-associated gene.
13. The composition of claim 1, wherein the composition is formulated for oral administration.
14. The composition of claim 13, wherein the composition is configured in single dosage units.
15. The composition of claim 14, wherein a single dosage unit comprises 50mg to 1000mg of the composition.
16. The composition of claim 14, wherein the single dosage unit is formulated as a capsule, a fudge, or a powder.
17. The composition of claim 1, further comprising vitamins, dietary trace elements or minerals, probiotics and/or prebiotics.
18. The composition of claim 1, further comprising niacin, nicotinamide riboside, nicotinamide mononucleotide, nicotinamide adenine dinucleotide, and/or a nutritionally acceptable CD38 inhibitor.
19. The composition of claim 1, wherein the composition is effective to treat or reduce symptoms associated with inflammatory disorders, metabolic disorders, neurological disorders, cardiovascular disorders, aging, and/or oxidative stress.
20. A method of supporting the health of a subject comprising administering to an individual the composition of claim 1.
21. The method of claim 20, wherein the composition is administered in an amount effective to treat or reduce symptoms associated with inflammatory disorders, metabolic disorders, neurological disorders, cardiovascular disorders, aging, and/or oxidative stress.
22. The method of claim 20 or 21, wherein the composition is administered for at least 30 days and/or at a daily dose of 50mg to 1000 mg.
23. Use of the composition of claim 1 to support the health of a subject by oral administration of the composition.
24. The use of claim 23, wherein the composition is administered in an amount effective to treat or reduce symptoms associated with inflammatory disorders, metabolic disorders, neurological disorders, cardiovascular disorders, aging, and/or oxidative stress.
25. The use according to claim 24 or 25, wherein the composition is administered for at least 30 days and/or at a daily dose of 50mg to 1000 mg.
26. A method for reducing inflammation in a subject, comprising:
administering to the subject a nutritionally acceptable carrier in combination with plant material having red, green, orange and violet colors comprising a plurality of chemically different polyphenols;
wherein the combination of plant materials synergistically reduces the release of at least one pro-inflammatory cytokine in the human cell and reduces the expression of at least one senescence-associated gene in the human cell.
27. The method of claim 26, wherein the red plant material comprises apple extract, pomegranate extract, tomato powder, and beetroot powder, wherein the green plant material comprises olive extract, rosemary extract, green coffee bean extract, and collard powder, wherein the orange plant material comprises onion extract, ginger extract, grapefruit extract, and carrot powder, and/or wherein the violet plant material comprises grape extract, blueberry extract, currant powder, and elderberry powder.
28. The method of claim 26, wherein the red plant material, green plant material, orange plant material, and violet plant material are part of a mediterranean diet.
29. The method of any one of claims 26 to 28, wherein the pro-inflammatory cytokine is selected from tumor necrosis factor alpha (TNF-a), interleukin-6 (IL-6), prostaglandin E 2 (PGE 2 ) And isoprostadins.
30. The method of any one of claims 26 to 28, wherein the red, green, orange and violet plant material are present in a synergistic amount that reduces the release of pro-inflammatory cytokines in human cells.
31. The method of any one of claims 26 to 28, wherein the senescence-associated gene is selected from HGF, c-fos, p16 INK And p21.
32. The method of claim 26, wherein the reduction in inflammation comprises a reduction in age-related immunity, a reduction in age-related energy metabolism, a reduction in age-related mitochondrial biogenesis, a reduction in at least one symptom associated with inflammation, and/or a reduction in at least one symptom associated with metabolic syndrome.
33. The method of claim 26, wherein administration of the composition further reduces expression of nfkb, increases glucose uptake into cells, increases mitochondrial biogenesis in cells, reduces oxidative damage caused by reactive oxygen species, reduces expression of pro-inflammatory adipokines, and/or increases intracellular ATP.
34. The method of claim 26, wherein administering to the subject comprises orally administering a plant material comprising a plurality of chemically distinct polyphenols.
35. The method of claim 34, wherein the plant material comprising the plurality of chemically distinct polyphenols is administered at a dose of about 50mg to 1000 mg.
36. The method of claim 34, wherein the plant material comprising a plurality of chemically different polyphenols is formulated as a tablet, beverage, or soft candy.
37. The method of claim 26, wherein the combination of plant materials further comprises vitamins, dietary trace elements or minerals, probiotics, and/or prebiotics.
38. The method of claim 26, wherein the combination of plant materials further comprises niacin, nicotinamide riboside, nicotinamide mononucleotide, nicotinamide adenine dinucleotide, and/or a nutritionally acceptable CD38 inhibitor.
39. The method of claim 26, wherein the plant material comprising the plurality of chemically distinct polyphenols is applied at least once daily for a period of at least 30 days.
40. A method for reducing inflammation in a subject, comprising:
administering to the subject a nutritionally acceptable carrier in combination with plant material having red, green, orange and violet colors comprising a plurality of chemically different polyphenols;
Wherein the red plant material comprises apple extract, pomegranate extract, tomato powder and beet root powder;
wherein the green plant material comprises olive extract, rosemary extract, green coffee bean extract and kale powder;
wherein the orange plant material comprises onion extract, ginger extract, grapefruit extract, and carrot powder;
wherein the purple blue plant material comprises grape extract, blueberry extract, currant powder and elderberry powder; and
wherein the combination of plant materials synergistically reduces at least one pro-inflammatory cytokine, reduces nfκf signaling, and/or reduces at least one pro-inflammatory adipokine in the subject after administration.
41. The method of claim 40, wherein the combination of plant materials is a synergistic combination with respect to a reduction of at least one cytokine.
42. The method of claim 40 or 41, wherein the combination of plant materials reduces at least one of pro-inflammatory cytokine and nfκf signaling in the subject after administration.
43. The method of claim 40 or 41, wherein the combination of plant materials reduces at least one pro-inflammatory cytokine, nfκf signaling, and at least one pro-inflammatory adipokine in the subject after administration.
44. A method for increasing glucose uptake by a cell, comprising:
contacting the cells with a plant material having red, green, orange and violet colors comprising a plurality of chemically different polyphenols;
wherein the red plant material comprises apple extract, pomegranate extract, tomato powder and beet root powder;
wherein the green plant material comprises olive extract, rosemary extract, green coffee bean extract and kale powder;
wherein the orange plant material comprises onion extract, ginger extract, grapefruit extract, and carrot powder;
wherein the purple blue plant material comprises grape extract, blueberry extract, currant powder and elderberry powder; and
wherein the combination of plant materials increases glucose uptake by the cells upon contact.
45. The method of claim 44, wherein the increased uptake of glucose by the cell is similar to or greater than the increased uptake of glucose with the thiazolidinedione.
46. A method for increasing ATP levels in a cell, comprising:
contacting the cells with a plant material having red, green, orange and violet colors comprising a plurality of chemically different polyphenols;
Wherein the red plant material comprises apple extract, pomegranate extract, tomato powder and beet root powder;
wherein the green plant material comprises olive extract, rosemary extract, green coffee bean extract and kale powder;
wherein the orange plant material comprises onion extract, ginger extract, grapefruit extract, and carrot powder;
wherein the purple blue plant material comprises grape extract, blueberry extract, currant powder and elderberry powder; and
wherein the combination of plant material increases ATP levels in the cells after contact.
47. The method of claim 46, wherein the cells are muscle cells.
48. A plant material having red, green, orange and violet colors comprising a plurality of chemically distinct polyphenols for use in alleviating at least one symptom of inflammation in an individual ingesting the plant material.
49. The use according to claim 48, wherein the red plant material comprises apple extract, pomegranate extract, tomato powder and beetroot powder, wherein the green plant material comprises olive extract, rosemary extract, green coffee bean extract and collard powder, wherein the orange plant material comprises onion extract, ginger extract, grapefruit extract, and carrot powder, and/or wherein the violet plant material comprises grape extract, blueberry extract, currant powder and elderberry powder.
50. Use of the nutritional composition according to claim 1 for reducing the release of pro-inflammatory cytokines in human leukocytes, reducing nfkb signaling, increasing glucose uptake by cells, increasing mitochondrial biogenesis in cells, reducing oxidative damage caused by reactive oxygen species, reducing the expression of pro-inflammatory adipokines and/or reducing the expression of at least one senescence-associated gene.
Claims (51)
1. A nutritional composition comprising:
a nutritionally acceptable carrier in combination with a plant material having red, green, orange and violet colors comprising a plurality of chemically different polyphenols;
wherein the red plant material, green plant material, orange plant material, and violet plant material are present in synergistic amounts that reduce the release of proinflammatory cytokines in human leukocytes.
2. The composition of claim 1, wherein the red plant material comprises apple extract, pomegranate extract, tomato powder, and beetroot powder, wherein the green plant material comprises olive extract, rosemary extract, green coffee bean extract, and collard powder, wherein the orange plant material comprises onion extract, ginger extract, grapefruit extract, and carrot powder, and/or wherein the violet plant material comprises grape extract, blueberry extract, currant powder, and elderberry powder.
3. The composition of claim 1 or 2, wherein the release of pro-inflammatory cytokines is tumor necrosis factor alpha (TNF-alpha), interleukin-6 (IL-6), prostaglandin E 2 (PGE 2 ) And release of at least one of the isoprostadins.
4. The composition of claim 1 or 2, wherein the release of pro-inflammatory cytokines is tumor necrosis factor alpha (TNF-alpha), interleukin-6 (IL-6), prostaglandin E 2 (PGE 2 ) And release of at least two of the isoprostadins.
5. The composition of claim 1 or 2, wherein the release of the pro-inflammatory cytokine is the release of at least three of tumor necrosis factor alpha (TNF-a), interleukin-6 (IL-6), prostaglandin E2 (PGE 2) and isoprostadins.
6. The composition of claim 1 or 2, wherein the release of pro-inflammatory cytokines is tumor necrosis factor alpha (TNF-alpha), interleukin-6 (IL-6), prostaglandin E 2 (PGE 2 ) And the other of the frontRelease of the hormone.
7. The composition of claim 1, wherein the composition further reduces expression of nfkb.
8. The composition of claim 1, wherein the composition further increases glucose uptake by cells.
9. The composition of claim 1, wherein the composition further increases the biogenesis of mitochondria within a cell.
10. The composition of claim 1, wherein the composition further reduces oxidative damage caused by reactive oxygen species.
11. The composition of claim 1, wherein the composition further reduces the expression of a proinflammatory adipokine.
12. The composition of claim 1, wherein the composition further reduces expression of a senescence-associated gene.
13. The composition of claim 1, wherein the composition is formulated for oral administration.
14. The composition of claim 13, wherein the composition is configured in single dosage units.
15. The composition of claim 14, wherein a single dosage unit comprises 50mg to 1000mg of the composition.
16. The composition of claim 14, wherein the single dosage unit is formulated as a capsule, a fudge, or a powder.
17. The composition of claim 1, further comprising vitamins, dietary trace elements or minerals, probiotics and/or prebiotics.
18. The composition of claim 1, further comprising niacin, nicotinamide riboside, nicotinamide mononucleotide, nicotinamide adenine dinucleotide, and/or a nutritionally acceptable CD38 inhibitor.
19. The composition of claim 1, wherein the composition is effective to treat or reduce symptoms associated with inflammatory disorders, metabolic disorders, neurological disorders, cardiovascular disorders, aging, and/or oxidative stress.
20. A method of supporting the health of a subject comprising administering to an individual the composition of claim 1.
21. The method of claim 20, wherein the composition is administered in an amount effective to treat or reduce symptoms associated with inflammatory disorders, metabolic disorders, neurological disorders, cardiovascular disorders, aging, and/or oxidative stress.
22. The method of claim 20 or 21, wherein the composition is administered for at least 30 days and/or at a daily dose of 50mg to 1000 mg.
23. Use of the composition of claim 1 to support the health of a subject by oral administration of the composition.
24. The use of claim 23, wherein the composition is administered in an amount effective to treat or reduce symptoms associated with inflammatory disorders, metabolic disorders, neurological disorders, cardiovascular disorders, aging, and/or oxidative stress.
25. The use according to claim 24 or 25, wherein the composition is administered for at least 30 days and/or at a daily dose of 50mg to 1000 mg.
26. A method for reducing inflammation in a subject, comprising:
administering to the subject a nutritionally acceptable carrier in combination with plant material having red, green, orange and violet colors comprising a plurality of chemically different polyphenols;
Wherein the combination of plant materials reduces the release of at least one pro-inflammatory cytokine in the human cell and reduces the expression of at least one senescence-associated gene in the human cell.
27. The method of claim 26, wherein the red plant material comprises apple extract, pomegranate extract, tomato powder, and beetroot powder, wherein the green plant material comprises olive extract, rosemary extract, green coffee bean extract, and collard powder, wherein the orange plant material comprises onion extract, ginger extract, grapefruit extract, and carrot powder, and/or wherein the violet plant material comprises grape extract, blueberry extract, currant powder, and elderberry powder.
28. The method of claim 26, wherein the red plant material, green plant material, orange plant material, and violet plant material are part of a mediterranean diet.
29. The method of any one of claims 26 to 28, wherein the pro-inflammatory cytokine is selected from tumor necrosis factor alpha (TNF-a), interleukin-6 (IL-6), prostaglandin E 2 (PGE 2 ) And isoprostadins.
30. The method of any one of claims 26 to 28, wherein the red, green, orange and violet plant material are present in a synergistic amount that reduces the release of pro-inflammatory cytokines in human cells.
31. The method of any one of claims 26 to 28, wherein the senescence-associated gene is selected from HGF, c-fos, p16 INK And p21.
32. The method of claim 26, wherein the reduction in inflammation comprises a reduction in age-related immunity, a reduction in age-related energy metabolism, a reduction in age-related mitochondrial biogenesis, a reduction in at least one symptom associated with inflammation, and/or a reduction in at least one symptom associated with metabolic syndrome.
33. The method of claim 26, wherein administration of the composition further reduces expression of nfkb, increases glucose uptake into cells, increases mitochondrial biogenesis in cells, reduces oxidative damage caused by reactive oxygen species, reduces expression of pro-inflammatory adipokines, and/or increases intracellular ATP.
34. The method of claim 26, wherein administering to the subject comprises orally administering a plurality of plant materials comprising chemically different polyphenols.
35. The method of claim 34, wherein the plant material comprising the plurality of chemically distinct polyphenols is administered at a dose of about 50mg to 1000 mg.
36. The method of claim 34, wherein the plant material comprising a plurality of chemically different polyphenols is formulated as a tablet, beverage, or soft candy.
37. The method of claim 26, wherein the combination of plant materials further comprises vitamins, dietary trace elements or minerals, probiotics, and/or prebiotics.
38. The method of claim 26, wherein the combination of plant materials further comprises niacin, nicotinamide riboside, nicotinamide mononucleotide, nicotinamide adenine dinucleotide, and/or a nutritionally acceptable CD38 inhibitor.
39. The method of claim 26, wherein the plant material comprising the plurality of chemically distinct polyphenols is applied at least once daily for a period of at least 30 days.
40. A method for reducing inflammation in a subject, comprising:
administering to the subject a nutritionally acceptable carrier in combination with plant material having red, green, orange and violet colors comprising a plurality of chemically different polyphenols;
wherein the red plant material comprises apple extract, pomegranate extract, tomato powder and beet root powder;
wherein the green plant material comprises olive extract, rosemary extract, green coffee bean extract and kale powder;
wherein the orange plant material comprises onion extract, ginger extract, grapefruit extract, and carrot powder;
Wherein the purple blue plant material comprises grape extract, blueberry extract, currant powder and elderberry powder; and
wherein the combination of plant materials reduces at least one pro-inflammatory cytokine, nfκf signaling, and/or at least one pro-inflammatory adipokine in the subject after administration.
41. The method of claim 40, wherein the combination of plant materials is a synergistic combination with respect to a reduction of at least one cytokine.
42. The method of claim 40 or 41, wherein the combination of plant materials reduces at least one of pro-inflammatory cytokine and nfκf signaling in the subject after administration.
43. The method of claim 40 or 41, wherein the combination of plant materials reduces at least one pro-inflammatory cytokine, nfκf signaling, and at least one pro-inflammatory adipokine in the subject after administration.
44. A method for increasing glucose uptake by a cell, comprising:
contacting the cells with a plant material having red, green, orange and violet colors comprising a plurality of chemically different polyphenols;
wherein the red plant material comprises apple extract, pomegranate extract, tomato powder and beet root powder;
Wherein the green plant material comprises olive extract, rosemary extract, green coffee bean extract and kale powder;
wherein the orange plant material comprises onion extract, ginger extract, grapefruit extract, and carrot powder;
wherein the purple blue plant material comprises grape extract, blueberry extract, currant powder and elderberry powder; and
wherein the combination of plant materials increases glucose uptake by the cells upon contact.
45. The method of claim 44, wherein the increased uptake of glucose by the cell is similar to or greater than the increased uptake of glucose with the thiazolidinedione.
46. The method of claim 44, wherein the step of contacting comprises oral administration.
47. A method for increasing ATP levels in a cell, comprising:
contacting the cells with a plant material having red, green, orange and violet colors comprising a plurality of chemically different polyphenols;
wherein the red plant material comprises apple extract, pomegranate extract, tomato powder and beet root powder;
wherein the green plant material comprises olive extract, rosemary extract, green coffee bean extract and kale powder;
Wherein the orange plant material comprises onion extract, ginger extract, grapefruit extract, and carrot powder;
wherein the purple blue plant material comprises grape extract, blueberry extract, currant powder and elderberry powder; and
wherein the combination of plant material increases ATP levels in the cells after contact.
48. The method of claim 47, wherein the cells are muscle cells.
49. A plant material having red, green, orange and violet colors comprising a plurality of chemically distinct polyphenols for use in alleviating at least one symptom of inflammation in an individual ingesting the plant material.
50. The use according to claim 49, wherein the red plant material comprises apple extract, pomegranate extract, tomato powder, and beetroot powder, wherein the green plant material comprises olive extract, rosemary extract, green coffee bean extract, and collard powder, wherein the orange plant material comprises onion extract, ginger extract, grapefruit extract, and carrot powder, and/or wherein the violet plant material comprises grape extract, blueberry extract, currant powder, and elderberry powder.
51. Use of the nutritional composition according to claim 1 for reducing the release of pro-inflammatory cytokines in human leukocytes, reducing nfkb signaling, increasing glucose uptake by cells, increasing mitochondrial biogenesis in cells, reducing oxidative damage caused by reactive oxygen species, reducing the expression of pro-inflammatory adipokines and/or reducing the expression of at least one senescence-associated gene.
Applications Claiming Priority (4)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
US63/215,716 | 2021-06-28 | ||
US202163285591P | 2021-12-03 | 2021-12-03 | |
US63/285,591 | 2021-12-03 | ||
PCT/US2022/033999 WO2023278181A1 (en) | 2021-06-28 | 2022-06-17 | Compositions and methods to counteract processes associated with inflammation and senescence and to support cellular energy and/or metabolism |
Publications (1)
Publication Number | Publication Date |
---|---|
CN117858630A true CN117858630A (en) | 2024-04-09 |
Family
ID=90542356
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
CN202280057212.6A Pending CN117858630A (en) | 2021-06-28 | 2022-06-17 | Compositions and methods for combating processes associated with inflammation and aging and supporting cellular energy and/or metabolism |
Country Status (1)
Country | Link |
---|---|
CN (1) | CN117858630A (en) |
Citations (2)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US20080260924A1 (en) * | 2007-04-20 | 2008-10-23 | Liang-Rong Chen | Composition Comprising Five Kinds of Processed Fruit or Vegetables |
CN107920561A (en) * | 2015-03-16 | 2018-04-17 | 自然阳光产品公司 | The plant compound of a variety of collaboration antioxidant activities of presentation available for food, dietary supplement, cosmetics and pharmaceutical preparation |
-
2022
- 2022-06-17 CN CN202280057212.6A patent/CN117858630A/en active Pending
Patent Citations (2)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US20080260924A1 (en) * | 2007-04-20 | 2008-10-23 | Liang-Rong Chen | Composition Comprising Five Kinds of Processed Fruit or Vegetables |
CN107920561A (en) * | 2015-03-16 | 2018-04-17 | 自然阳光产品公司 | The plant compound of a variety of collaboration antioxidant activities of presentation available for food, dietary supplement, cosmetics and pharmaceutical preparation |
Non-Patent Citations (1)
Title |
---|
王雪萍等: "原花青素类成分在防治 2 型糖尿病作用机制方面的研究进展", 中国中药杂志, vol. 42, no. 20, 31 October 2017 (2017-10-31), pages 3866 - 3872 * |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
KR101746635B1 (en) | Composition for metabolic diseases comprising Bacteroides acidifaciens | |
KR20120081605A (en) | Method of slowing the aging process by activating sirtuin enzymes with a combination of fucoxanthin and punicic acid | |
KR20210062660A (en) | Probiotic strains for aging, muscle, bone, intestine, hyperlipidemia, skin and brain | |
Derakhshandeh-Rishehri et al. | Effect of honey vinegar syrup on blood sugar and lipid profile in healthy subjects | |
US20230046756A1 (en) | Lactobacillus Acidophilus KBL409 Strain and Use Thereof | |
Li et al. | Naringin ameliorates obesity via stimulating adipose thermogenesis and browning, and modulating gut microbiota in diet-induced obese mice | |
Lin et al. | Quercetin attenuates cisplatin-induced fat loss | |
KR102149185B1 (en) | Enterococcus lactis Wikim0107 strain controlling immune function and ameliorating inflammatory bowel disease and use thereof | |
JP2017507148A (en) | Activated soybean pod fiber | |
US20130059920A1 (en) | Hydroxytyrosol benefits muscle differentiation and muscle contraction and relaxation | |
CN117858630A (en) | Compositions and methods for combating processes associated with inflammation and aging and supporting cellular energy and/or metabolism | |
KR102160424B1 (en) | Composition for preventing, treating or improving obesity comprising Eupatilin as an active ingredient | |
US9895341B2 (en) | Inflammation and immunity treatments | |
Cogorno et al. | Non-alcoholic fatty liver disease: Dietary and nutraceutical approaches | |
KR102650558B1 (en) | Probiotics composition for preventing, improving, or treating inflammatory disease containing Lactobacillus rhamnosus PL60 as an active ingredient | |
US11503851B1 (en) | Compositions and methods to counteract processes associated with inflammation and senescence and to support cellular energy and/or metabolism | |
WO2012074184A1 (en) | Pharmaceutical composition for preventing or treating obesity comprising sphingosine-1-phosphate or a pharmaceutically acceptable salt thereof as an active ingredient | |
Germoush | Antioxidant and anti-inflammatory effects of Padina pavonia and Turbenaria ornate in streptozotocin/nicotinamide diabetic rats. | |
JP2020026396A (en) | GLUT1 expression enhancer | |
KR101572311B1 (en) | A composition for preventing or treating obesity comprising 2-amino-2-norbornanecarboxylic acid | |
WO2013124381A1 (en) | Peltatin for use in the treatment of metabolic disorders | |
Zare Javid et al. | The effect of resveratrol supplementation in adjunct with non-surgical periodontal treatment on blood glucose, triglyceride, periodontal status and some inflammatory markers in type 2 diabetic patients with periodontal disease | |
Javdan et al. | Anti-ocular-inflammatory effects of Salvia hypoleuca extract on rat endotoxin-Induced uveitis | |
JP2017515830A (en) | A composition comprising 7-hydroxymatylesinol | |
KR20200007990A (en) | Composition for preventing, treating or improving obesity comprising Jaceosidin |
Legal Events
Date | Code | Title | Description |
---|---|---|---|
PB01 | Publication | ||
PB01 | Publication | ||
SE01 | Entry into force of request for substantive examination | ||
SE01 | Entry into force of request for substantive examination |