Volume 12 Issue 3
Jun.  2022
Turn off MathJax
Article Contents
Akansha Chauhan, Asim Ul Islam, Hridayesh Prakash, Sandhya Singh. Phytochemicals targeting NF-κB signaling:Potential anti-cancer interventions[J]. Journal of Pharmaceutical Analysis, 2022, 12(3): 394-405. doi: 10.1016/j.jpha.2021.07.002
Citation: Akansha Chauhan, Asim Ul Islam, Hridayesh Prakash, Sandhya Singh. Phytochemicals targeting NF-κB signaling:Potential anti-cancer interventions[J]. Journal of Pharmaceutical Analysis, 2022, 12(3): 394-405. doi: 10.1016/j.jpha.2021.07.002

Phytochemicals targeting NF-κB signaling:Potential anti-cancer interventions

doi: 10.1016/j.jpha.2021.07.002
Funds:

The author is thankful to the lab mates Mr. Aniket Angira and Ms. Pooja Singh for their support in completion of this review article.

  • Received Date: Oct. 17, 2020
  • Accepted Date: Jul. 04, 2021
  • Rev Recd Date: Jul. 03, 2021
  • Publish Date: Jul. 06, 2021
  • Nuclear factor κB (NF-κB) is a ubiquitous regulator of the signalome and is indispensable for various biological cell functions. NF-κB consists of five transcription factors that execute both cytoplasmic and nuclear signaling processes in cells. NF-κB is the only signaling molecule that governs both pro- and anti-apoptotic, and pro- and anti-inflammatory responses. This is due to the canonical and non-canonical components of the NF-κB signaling pathway. Together, these pathways orchestrate cancer-related inflammation, hyperplasia, neoplasia, and metastasis. Non-canonical NF-κB pathways are particularly involved in the chemoresistance of cancer cells. In view of its pivotal role in cancer progression, NF-κB represents a potentially significant therapeutic target for modifying tumor cell behavior. Several phytochemicals are known to modulate NF-κB pathways through the stabilization of its inhibitor, IκB, by inhibiting phosphorylation and ubiquitination thereof. Several natural pharmacophores are known to inhibit the nuclear translocation of NF-κB and associated pro-inflammatory responses and cell survival pathways. In view of this and the high degree of specificity exhibited by various phytochemicals for the NF-κB component, we herein present an in-depth overview of these phytochemicals and discuss their mode of interaction with the NF-κB signaling pathways for controlling the fate of tumor cells for cancer-directed interventions.
  • loading
  • R. Sen, D. Baltimore, Inducibility of kappa immunoglobulin enhancer-binding protein Nf-kappa B by a posttranslational mechanism, Cell 47(1986)921-928
    P.A. Baeuerle, D. Baltimore, Activation of DNA-binding activity in an apparently cytoplasmic precursor of the NF-kappa B transcription factor, Cell 53(1988)211-217
    T.D. Gilmore, NF-κB, KBF1, dorsal, and related matters, Cell 62(1990)841-843
    T.D. Gilmore, Introduction to NF-kappaB:players, pathways, perspectives, Oncogene 25(2006)6680-6684
    S.C. Sun, Non-canonical NF-kappaB signaling pathway, Cell Res. 1(2011)71-85
    -gingerol, a major pungent principle of ginger, on phorbol ester-induced inflammation, epidermal ornithine decarboxylase activity and skin tumor promotion in ICR mice, Cancer Lett. 129(1998)139-144
    Hoffmann A, Natoli G, Ghosh G, Transcriptional regulation via the NF-kappaB signaling module, Oncogene 25(2006)6706-6716
    A. Orian, A.L. Schwartz, A. Israel, et al., Structural motifs involved in ubiquitin-mediated processing of the NF-kappaB precursor p105:roles of the Glycine-rich region and a downstream ubiquitination domain, Mol. Cell Biol. 19(1999)3664-3673
    A. Oeckinghaus, S. Ghosh, The NF-B family of transcription factors and its regulation, Cold Spring Harb. Perspect. Biol. 4(2009) a000034
    T. Lawrence, The nuclear factor NF-kappaB pathway in inflammation, Cold Spring Harb.Perspect. Biol. 6(2009) a001651
    N. D. Perkins, Post-translational modifications regulating the activity and function of nuclear factor kappa B pathway, Oncogene 25(51)(2006)6717-6730
    R. Gugasyan, A. Voss, G. Varigos, et al., The transcription factors c-rel and RelA control epidermal development and homeostasis in embryonic and adult skin via distinct mechanisms, Mol. Cell Biol. 24(2004)5733-5745
    M. Baratin, C. Foray, M. Dalod et al., Homeostatic NF-k B signaling in steady-state migratory dendritic cells regulates immune homeostasis and tolerance, Immunity 42(2015)627-639
    A.A. Beg, W.C. Sha, R.T. Bronson, et al., Embryonic lethality and liver degeneration in mice lacking the RelA component of NF-κB, Nature 376(1995)167-170
    L.E. George, M.F. Lokhandwala, M. Asghar, Novel role of NF-κB-p65 in antioxidant homeostasis in human kidney-2 cells, Am. J. Physiol. Ren. Physiol. 302(2012) F1440-F1446
    Elinav E, Nowarski R, Thaiss CA, et al., Inflammation-induced cancer:crosstalk between tumours, immune cells and microorganisms, Nat. Rev. Cancer 13(2013)759-771
    J. Terzic, S. Grivennikov, E. Karin, et al., Inflammation and colon cancer, Gastroenterology 138(2010)2101-2114
    Y. Matsumoto, H. Marusawa, K. Kinoshita, et al., Helicobacter pylori infection triggers aberrant expression of activation-induced cytidine deaminase in gastric epithelium, Nat. Med. 13(2007)470-476
    S. Gerondakis, R. Grumont, R. Gugasyan, et al., Unravelling the complexities of the NF-kappaB signalling pathway using mouse knockout and transgenic models, Oncogene 25(2006)6781-6799
    Forsythe JA, Jiang BH, Iyer NV, et al., Activation of vascular endothelial growth factor gene transcription by hypoxia-inducible factor 1, Mol. Cell. Biol. 16(1996)4604-4613
    S.Y. Huang, C.A. Pettaway, H. Uehara, et al., Blockade of NF-κB activity in human prostate cancer cells is associated with suppression of angiogenesis, invasion, and metastasis, Oncogene 20(2001)4188-4197
    D.C. Guttridge, C. Albanese, J.Y. Reuther, et al., NF-κB controls cell growth and differentiation through transcriptional regulation of cyclin D1, Mol. Cell. Biol. 19(1999)5785-5799
    M. Mu¨ller, A. Morotti, C. Ponzetto, Activation of NF-κB is essential for hepatocyte growth factor-mediated proliferation and tubulogenesis, Mol. Cell. Biol. 22(2002)1060-1072
    J. Witowski, K. Ksiazek, C. Warnecke, et al., Role of mesothelial cell-derived granulocyte colony-stimulating factor in interleukin-17-induced neutrophil accumulation in the peritoneum, Kidney Int. 71(2007)514-525
    J.Q. Feng, L.P. Xing, J.H. Zhang, et al., NF-kappaB specifically activates BMP-2 gene expression in growth plate chondrocytes in vivo and in a chondrocyte cell line in vitro, J. Biol. Chem. 278(2003)29130-29135
    S. Polager, D. Ginsberg, E2F-at the crossroads of life and death, Trends Cell Biol. 18(2008)528-535
    S. Maeda, Y. Hikiba, K. Sakamoto, et al., Ikappa B kinasebeta/nuclear factor-kappaB activation controls the development of liver metastasis by way of interleukin-6 expression, Hepatology 50(2009)1851-1860
    H. Zenan, L. Xiaojun, T. Zhifeng, et al., Possible regulatory role of Snail in NF-κB mediated changes in E-cadherin in gastric cancer, Oncol. Rep. 3(2013)993-1000
    J. Whan Han, B. Gon Lee, Y. Kee Kim, et al., Ergolide, sesquiterpene lactone from Inula britannica, inhibits inducible nitric oxide synthase and cyclo-oxygenase-2 expression in RAW 264.7 macrophages through the inactivation of NF-kappaB, Br. J. Pharmacol. 133(2001)503-512
    S. Kim, M. Karin, Role of TLR2-dependent inflammation in metastatic progression, Ann. N. Y. Acad. Sci. 1217(2011)191-206
    A. Guaadaoui, S. Benaicha, N. Elmajdoub, et al., What is a bioactive compound?A combined definition for a preliminary consensus, Int. J. Nutr. Food Sci. 3(2014)174
    K.K. Park, K.S. Chun, J.M. Lee, et al., Inhibitory effects of
    S.O. Kim, J.K. Kundu, Y.K. Shin, et al., 6] -Gingerol inhibits COX-2 expression by blocking the activation of p38 MAP kinase and NF-kappaB in phorbol ester-stimulated mouse skin, Oncogene 24(2005)2558-2567
    J.K. Kim, Y. Kim, K.M. Na, et al., 6] -Gingerol prevents UVB-induced ROS production and COX-2 expression in vitro and in vivo, Free Radic. Res. 41(2007)603-614
    B. Guenther, C. Christensen, J. Upatnieks, Coherent optical processing:Another approach, IEEE J. Quantum Electron. 15(1979)1348-1362
    B. Salehi, A. Venditti, M. Sharifi-Rad et al., The therapeutic potential of Apigenin, Int. J. Mol. Sci. 6(2019)1305
    Wang J, Liu YT, Xiao L, et al., Anti-inflammatory effects of apigenin in lipopolysaccharide-induced inflammatory in acute lung injury by suppressing COX-2 and NF-kB pathway, Inflammation 37(2014)2085-2090
    Ray B, Bisht S, Maitra A, et al., Neuroprotective and neurorescue effects of a novel polymeric nanoparticle formulation of curcumin (NanoCurcTM) in the neuronal cell culture and animal model:implications for Alzheimer's disease, J. Alzheimers Dis. 23(2011)61-77
    E. Ooko, O. Kadioglu, H.J. Greten, et al., Pharmacogenomic characterization and isobologram analysis of the combination of ascorbic acid and curcumin-two main metabolites of Curcuma longa-in cancer cells, Front. Pharmacol. 8(2017)38
    Y. Abe, S. Hashimoto, T. Horie, Curcumin inhibition of inflammatory cytokine production by human peripheral blood monocytes and alveolar macrophages, Pharmacol. Res. 39(1999)41-47
    C. Buhrmann, A. Mobasheri, F. Busch, et al., Curcumin modulates nuclear factor kappaB (NF-kappaB)-mediated inflammation in human tenocytes in vitro:role of the phosphatidylinositol 3-kinase/Akt pathway, J. Biol. Chem. 286(2011)28556-28566
    D. L. McKay, J.B. Blumberg, The role of tea in human health:an update, J. Am. Coll. Nutr. 1(2002)1-13
    J.K. Kundu, H. K. Na, K. S. Chun et al., Inhibition of Phorbol Ester-Induced COX-2 expression by epigallocatechin gallate in mouse skin and cultured human mammary epithelial cells, J. N. 133(11)(2003)3805S-3810S
    C.C. Wu, M.C. Hsu, C.W. Hsieh, et al., Upregulation of heme oxygenase-1 by Epigallocatechin-3-gallate via the phosphatidylinositol 3-kinase/Akt and ERK pathways, Life Sci. 78(2006)2889-2897
    G.X. Shen, C.J. Xu, R. Hu, et al., Comparison of (-)-epigallocatechin-3-gallate elicited liver and small intestine gene expression profiles between C57BL/6J mice and C57BL/6J/Nrf2(-/-) mice, Pharm. Res. 22(2005)1805-1820
    S.Y. Joo, Y.A. Song, Y.L. Park, et al., Epigallocatechin-3-gallate inhibits LPS-induced NF-κB and MAPK signaling pathways in bone marrow-derived macrophages, Gut Liver 6(2012)188-196
    A.S. Negi, M.P. Darokar, S.K. Chattopadhyay, et al., Synthesis of a novel plant growth promoter from Gallic acid, Bioorg. Med. Chem. Lett. 15(2005)1243-1247
    K.C. Choi, Y.H. Lee, M.G. Jung, et al., Gallic acid suppresses lipopolysaccharide-induced nuclear factor-kappaB signaling by preventing RelA acetylation in A549 lung cancer cells, Mol. Cancer Res. 12(2009)2011-2021
    M.J. Kim, A.R. Seong, J.Y. Yoo, et al., Gallic acid, a histone acetyltransferase inhibitor, suppresses β-amyloid neurotoxicity by inhibiting microglial-mediated neuroinflammation, Mol. Nutr. Food Res. 55(2011)1798-1808
    J. Liggins, L. J. Bluck, S. Runswick, et al., Daidzein and genistein content of fruits and nuts, J. Nutr. Biochem. 6(2000)326-331
    E.J. Seo, N. Fischer, T.Efferth, Phytochemicals as inhibitors of NF-κB for treatment of Alzheimer's disease, Pharmacol. Res. 129(2018)262-273
    X. Zhou, L. Yuan, X. Zhao, et al., Genistein antagonizes inflammatory damage induced by β-amyloid peptide in microglia through TLR4 and NF-κB, Nutrition 30(2014)90-95
    W.C. Hu, G.C. Wang, P.X. Li, et al., Neuroprotective effects of macranthoin G from Eucommia ulmoides against hydrogen peroxide-induced apoptosis in PC12 cells via inhibiting NF-κB activation, Chem. Biol. Interact. 224(2014)108-116
    J. Ock, H.S. Han, S.H. Hong, et al., Obovatol attenuates microglia-mediated neuroinflammation by modulating redox regulation, Br. J. Pharmacol. 159(2010)1646-1662
    M.S. Choi, S.H. Lee, H.S. Cho, et al., Inhibitory effect of obovatol on nitric oxide production and activation of NF-kappaB/MAP kinases in lipopolysaccharide-treated RAW 264.7cells, Eur. J. Pharmacol. 556(2007)181-189
    Y.J. Wang, R.J. Chen, Pterostilbene protection and bladder cancer cells. Cancer. Amsterdam:Elsevier, 2014:271-281
    A.V. Anand David, R. Arulmoli, S. Parasuraman, Overviews of biological importance of quercetin:a bioactive flavonoid, Pharmacogn. Rev. 10(2016)84-89
    M. Endale, S.C. Park, S. Kim, et al., Quercetin disrupts tyrosine-phosphorylated phosphatidylinositol 3-kinase and myeloid differentiation factor-88 association, and inhibits MAPK/AP-1 and IKK/NF-κB-induced inflammatory mediators production in RAW 264.7 cells, Immunobiology 218(2013)1452-1467
    Y. Aqeel, J. Iqbal, R. Siddiqui, et al., Anti-Acanthamoebic properties of resveratrol and demethoxycurcumin, Exp. Parasitol. 132(2012)519-523
    B. Bellaver, D.G. Souza, D.O. Souza, et al., Resveratrol increases antioxidant defenses and decreases proinflammatory cytokines in hippocampal astrocyte cultures from newborn, adult and aged Wistar rats, Toxicol. In Vitro 28(2014)479-484
    H. Capiralla, V. Vingtdeux, H.T. Zhao, et al., Resveratrol mitigates lipopolysaccharide-and Aβ-mediated microglial inflammation by inhibiting the TLR4/NF-κB/STAT signaling cascade, J. Neurochem. 120(2012)461-472
    Z.H. Ren, L. Wang, J.H. Cui, et al., Resveratrol inhibits NF-kB signaling through suppression of p65 and IkappaB kinase activities, Pharmazie 68(2013)689-694
    Y. Zhu, Y.P. Shi, D. Wu, et al., Salidroside protects against hydrogen peroxide-induced injury in cardiac H9c2 cells via PI3K-Akt dependent pathway, DNA Cell Biol. 30(2011)809-819
    C.B. Tan, M. Gao, W.R. Xu, et al., Protective effects of salidroside on endothelial cell apoptosis induced by cobalt chloride, Biol. Pharm. Bull. 32(2009)1359-1363
    J. Gao, H. He, W.J.Jiang, et al., Salidroside ameliorates cognitive impairment in a d-galactose-induced rat model of Alzheimer's disease, Behav. Brain Res. 293(2015)27-33
    J.W. Wang, Y.B. Pan, Y.Q. Cao, et al., Salidroside regulates the expressions of IL-6 and defensins in LPS-activated intestinal epithelial cells through NF-κB/MAPK and STAT3 pathways, Iran J. Basic. Med. Sci. 22(2019)31-37
    C. Saliou, B. Rihn, J. Cillard, et al., Selective inhibition of NF-kappaB activation by the flavonoid hepatoprotector silymarin in HepG2. Evidence for different activating pathways, FEBS Lett. 440(1998)8-12
    W. Lin, R.T. Wu, T. Wu, et al., Sulforaphane suppressed LPS-induced inflammation in mouse peritoneal macrophages through Nrf2 dependent pathway, Biochem. Pharmacol. 76(2008)967-973
    Surh YJ, Na HK, NF-kappaB and Nrf2 as prime molecular targets for chemoprevention and cytoprotection with anti-inflammatory and antioxidant phytochemicals, Genes Nutr. 2(2008)313-317
    F. Bouvier, A. Rahier, B. Camara, Biogenesis, molecular regulation and function of plant isoprenoids, Prog. Lipid Res. 44(2005)357-429
    C.I. Keeling, J. Bohlmann, Genes, enzymes and chemicals of terpenoid diversity in the constitutive and induced defence of conifers against insects and pathogens, New Phytol. 170(2006)657-675
    B. Dinda, S. Debnath, Y. Harigaya, Naturally occurring iridoids. A review. part 1, ChemInform. 38(2007):159-222. DOI: 10.1002/chin.200724256
    Jeong HJ, Koo HN, Na HJ, et al., Inhibition of TNF-alpha and IL-6 production by Aucubin through blockade of NF-kappaB activation RBL-2H3 mast cells, Cytokine 18(2002)252-259
    Y.Y. Chen, S.L. Liu, D.P. Hu, et al., N-methyl-N-nitrosourea-induced retinal degeneration in mice, Exp. Eye Res. 121(2014)102-113
    S.W. Kim, S.C. Choi, E.Y. Choi, et al., Catalposide, a compound isolated from Catalpa ovata, attenuates induction of intestinal epithelial proinflammatory gene expression and reduces the severity of trinitrobenzene sulfonic Acid-induced colitis in mice, Inflamm. Bowel Dis. 10(2004)564-572
    X.J. Lin, Z.X. Peng, C.Q. Su, Potential anti-cancer activities and mechanisms of costunolide and dehydrocostuslactone, Int. J. Mol. Sci. 16(2015)10888-10906
    T.H. Koo, J.H. Lee, Y.J. Park, et al., A sesquiterpene lactone, costunolide, from Magnolia grandiflora inhibits NF-kappa B by targeting I kappa B phosphorylation, Planta Med. 67(2001)103-107
    V.G. Tacias-Pascacio, E. Garcia-Parra, G. Vela-Gutierrez, et al., Genipin as an emergent tool in the design of biocatalysts:mechanism of reaction and applications, Catalysts 9(2019)1035
    Q.S. Wang, Y.Z. Xiang, Y.L. Cui, et al., Dietary blue pigments derived from genipin, attenuate inflammation by inhibiting LPS-induced iNOS and COX-2 expression via the NF-κB inactivation, PLoS One 7(2012) e34122. DOI: 10.1371/journal.pone.0034122
    G. Lyss, A. Knorre, T.J. Schmidt, et al., The anti-inflammatory sesquiterpene lactone helenalin inhibits the transcription factor NF-kappaB by directly targeting p65, J. Biol. Chem. 273(1998)33508-33516
    A.P. Rogerio, E.L. Andrade, D.F. Leite, et al., Preventive and therapeutic anti-inflammatory properties of the sesquiterpene alpha-humulene in experimental airways allergic inflammation, Br. J. Pharmacol. 158(2009)1074-1087
    X.G. Lu, L.B. Zhan, B.G. Feng, et al., Inhibition of growth and metastasis of human gastric cancer implanted in nude mice by d-limonene, World J. Gastroenterol. 10(2004)2140-2144
    C.M. Berchtold, K.S. Chen, S. Miyamoto, et al., Perillyl alcohol inhibits a calcium-dependent constitutive nuclear factor-kappaB pathway, Cancer Res. 65(2005)8558-8566
    G.F. Chi, M.M. Wei, X.X. Xie, et al., Suppression of MAPK and NF-κB pathways by limonene contributes to attenuation of lipopolysaccharide-induced inflammatory responses in acute lung injury, Inflammation 36(2013)501-511
    C.N. Wang, Y.J. Shiao, Y.L. Lin, et al., Nepalolide A inhibits the expression of inducible nitric oxide synthase by modulating the degradation of IkappaB-alpha and IkappaB-beta in C6 glioma cells and rat primary astrocytes, Br. J. Pharmacol. 128(1999)345-356
    A. Saadane, S. Masters, J. DiDonato, et al., Parthenolide inhibits IkappaB kinase, NF-kappaB activation, and inflammatory response in cystic fibrosis cells and mice, Am. J. Respir. Cell Mol. Biol. 36(2007)728-736
    T. Efferth, Willmar Schwabe Award 2006:antiplasmodial and antitumor activity of artemisinin:from bench to bedside, Planta Med. 73(2007)299-309
    E. Aldieri, D. Atragene, L. Bergandi, et al., Artemisinin inhibits inducible nitric oxide synthase and nuclear factor NF-kB activation, FEBS Lett. 552(2003)141-144
    H. Xu, Y. He, X. Yang, et al., Anti-malarial agent artesunate inhibits TNF-alpha-induced production of proinflammatory cytokines via inhibition of NF-kappaB and PI3 kinase/Akt signal pathway in human rheumatoid arthritis fibroblast-like synoviocytes, Rheumatology (Oxford)46(2007)920-926
    M.H. Jang, X.L. Piao, J.M. Kim, et al., Inhibition of cholinesterase and amyloid-beta aggregation by resveratrol oligomers from Vitis amurensis, Phytother. Res. 22(2008)544-549
    Y. Takada, A. Murakami, B.B. Aggarwal, Zerumbone abolishes NF-κB and IκBα kinase activation leading to suppression of antiapoptotic and metastatic gene expression, upregulation of apoptosis, and downregulation of invasion, Oncogene 24(2005)6957-6969
    J.Y. Zhou, F. Di Tang, G.G. Mao et al., Effect of α-pinene on nuclear translocation of NF-kappaB in THP-1 cells, Acta Pharmacol. Sin. 4(2004)480-484
    T.H. Chao, T. Lam, B.G. Vong, et al., A new family of synthetic diterpenes that regulates cytokine synthesis by inhibiting IkappaBalpha phosphorylation, ChemBioChem 6(2005)133-144
    S.C. Huang, C.T. Ho, S.Y. Lin-Shiau, et al., Carnosol inhibits the invasion of B16/F10 mouse melanoma cells by suppressing metalloproteinase-9 through down-regulating nuclear factor-kappa B and c-Jun, Biochem. Pharmacol. 69(2005)221-232
    T.K. Lee, R.T.P. Poon, J.Y. Wo et al., Lupeol suppresses cisplatin-induced nuclear factor-kappaB activation in head and neck squamous cell carcinoma and inhibits local invasion and nodal metastasis in an orthotopic nude mouse model, Cancer Res. 67(18)(2007)8800-8809
    T.C. Hsieh, E.K. Wijeratne, J.Y. Liang, et al., Differential control of growth, cell cycle progression, and expression of NF-kappaB in human breast cancer cells MCF-7, MCF-10A, and MDA-MB-231 by ponicidin and oridonin, diterpenoids from the Chinese herb Rabdosia rubescens, Biochem. Biophys. Res. Commun. 337(2005)224-231
    T. Ikezoe, Y. Yang, K. Bandobashi, et al., Oridonin, a diterpenoid purified from Rabdosia rubescens, inhibits the proliferation of cells from lymphoid malignancies in association with blockade of the NF-kappa B signal pathways, Mol. Cancer Ther. 4(2005)578-586
    S. Alakurtti, T. Makela, S. Koskimies, et al., Pharmacological properties of the ubiquitous natural product betulin, Eur. J. Pharm. Sci. 29(2006)1-13
    L.J. Hofseth, M.J. Wargovich, Inflammation, cancer, and targets of ginseng, J. Nutr. 137(2007)183S-185S
    K. Choi, M. Kim, J. Ryu, et al., Ginsenosides compound K and Rh2 inhibit tumor necrosis factor-α-induced activation of the NF-κB and JNK pathways in human astroglial cells, Neurosci. Lett. 421(2007)37-41
    J.M. Cherng, H.J. Lin, M.S. Hung, et al., Inhibition of nuclear factor kappaB is associated with neuroprotective effects of glycyrrhizic acid on glutamate-induced excitotoxicity in primary neurons, Eur. J. Pharmacol. 547(2006)10-21
    K. Izumi-Nagai, N. Nagai, K. Ohgami, et al., Macular pigment lutein is antiinflammatory in preventing choroidal neovascularization, Arterioscler. Thromb. Vasc. Biol. 27(2007)2555-2562
    R. Marti, S. Rosello, J. Cebolla-Cornejo, Tomato as a source of carotenoids and polyphenols targeted to cancer prevention, Cancers 8(2016)58
    R. Paul, M.K. Mazumder, J. Nath, et al., Lycopene-A pleiotropic neuroprotective nutraceutical:Deciphering its therapeutic potentials in broad spectrum neurological disorders, Neurochem. Int. 140(2020)104823
    M. Kelkel, M. Schumacher, M. Dicato, et al., Antioxidant and anti-proliferative properties of lycopene, Free. Radic. Res. 45(2011)925-940
    W. Lee, S.K. Ku, J.W. Bae, et al., Inhibitory effects of lycopene on HMGB1-mediated pro-inflammatory responses in both cellular and animal models, Food Chem. Toxicol. 50(2012)1826-1833
    J. Marcotorchino, B. Romier, E. Gouranton, et al., Lycopene attenuates LPS-induced TNF-α secretion in macrophages and inflammatory markers in adipocytes exposed to macrophage-conditioned media, Mol. Nutr. Food Res. 56(2012)725-732
    A.K. Sachdeva, K.Chopra, Lycopene abrogates Aβ(1-42)-mediated neuroinflammatory cascade in an experimental model of Alzheimer's disease, J. Nutr. Biochem. 26(2015)736-744
    E.A. Assar, M.C. Vidalle, M. Chopra, et al., Lycopene Acts through inhibition of IκB kinase to suppress NF-κB signaling in human prostate and breast cancer cells, Tumor Biol. 37(2016)9375-9385
    N.M. Kalariya, K.V. Ramana, S.K. Srivastava, et al., Carotenoid derived aldehydes-induced oxidative stress causes apoptotic cell death in human retinal pigment epithelial cells, Exp. Eye Res. 86(2008)70-80
    D. Paris, D. Beaulieu-Abdelahad, C.Bachmeier, et al., Anatabine lowers Alzheimer's Aβ production in vitro and in vivo, Eur. J. Pharmacol. 670(2011)384-391
    D. Paris, D. Beaulieu-Abdelahad, L. Abdullah, et al., Anti-inflammatory activity of anatabine via inhibition of STAT3 phosphorylation, Eur. J. Pharmacol. 698(2013)145-153
    L.Y. Jia, J. Liu, Z. Song, et al., Berberine suppresses amyloid-beta-induced inflammatory response in microglia by inhibiting nuclear factor-kappaB and mitogen-activated protein kinase signalling pathways, J. Pharm. Pharmacol. 64(2012)1510-1521
    Y. Xu, Y. Xue, Y. Wang, et al., Multiple-modulation effects of Oridonin on the production of proinflammatory cytokines and neurotrophic factors in LPS-activated microglia, Int. Immunopharmacol. 9(2009)360-365
    F. Chen, S. Sun, D.C. Kuhn, et al., Tetrandrine inhibits signal-induced NF-kappa B activation in rat alveolar macrophages, Biochem. Biophys. Res. Commun. 231(1997)99-102
    L.J. Ho, T.Y. Juan, P. Chao, et al., Plant alkaloid tetrandrine downregulates IkappaBalpha kinases-IkappaBalpha-NF-kappaB signaling pathway in human peripheral blood T cell, Br. J. Pha. 143(2004)919-927
  • 加载中

Catalog

    通讯作者: 陈斌, bchen63@163.com
    • 1. 

      沈阳化工大学材料科学与工程学院 沈阳 110142

    1. 本站搜索
    2. 百度学术搜索
    3. 万方数据库搜索
    4. CNKI搜索

    Figures(1)

    Article Metrics

    Article views (381) PDF downloads(8) Cited by()
    Proportional views
    Related

    /

    DownLoad:  Full-Size Img  PowerPoint
    Return
    Return