Generic placeholder image

CNS & Neurological Disorders - Drug Targets

Editor-in-Chief

ISSN (Print): 1871-5273
ISSN (Online): 1996-3181

Review Article

Bioavailability and Pharmaco-therapeutic Potential of Luteolin in Overcoming Alzheimer’s Disease

Author(s): Fahad Ali and Yasir Hasan Siddique*

Volume 18, Issue 5, 2019

Page: [352 - 365] Pages: 14

DOI: 10.2174/1871527318666190319141835

Price: $65

Abstract

Luteolin is a naturally occurring, yellow crystalline flavonoid found in numerous dietary supplements we frequently have in our meals. Studies in the last 2 decades have revealed its therapeutic potential to reduce the Alzheimer’s disease (AD) symptoms in various in vitro and in vivo models. The anti-Alzheimer’s potential of luteolin is attributed to its ability to suppress Aβ as well as tau aggregation or promote their disaggregation, down-regulate the expression of COX-2, NOS, MMP-9, TNF-α, interleukins and chemokines, reduce oxidative stress by scavenging ROS, modulate the activities of transcription factors CREB, cJun, Nrf-1, NF-κB, p38, p53, AP-1 and β-catenine and inhibiting the activities of various protein kinases. In several systems, luteolin has been described as a potent antioxidant and anti-inflammatory agent. In addition, we have also discussed about the bio-availability of the luteolin in the plasma. After being metabolized luteolin persists in plasma as glucuronides and sulphate-conjugates. Human clinical trials indicated no dose limiting toxicity when administered at a dose of 100 mg/day. Improvements in the formulations and drug delivery systems may further enhance the bioavailability and potency of luteolin. The current review describes in detail the data supporting these studies.

Keywords: Luteolin, Alzheimer's disease, oxidative stress, bioavailability, ROS, pharmaco-therapeutic.

Graphical Abstract
[1]
López-Lázaro M. Distribution and biological activities of the flavonoid luteolin. Mini Rev Med Chem 2009; 9: 31-59.
[2]
Nile SH, Keum YS, Nile AS, Jalde SS, Patel RV. Antioxidant, anti‐inflammatory, and enzyme inhibitory activity of natural plant flavonoids and their synthesized derivatives. J Biochem Mol Toxicol 2018; 32e22002
[3]
Shan XU, Cheng J, Chen KL, Liu YM, Juan LI. Comparison of lipoxygenase, cyclooxygenase, xanthine oxidase inhibitory effects and cytotoxic activities of selected flavonoids DES Tech Trans Environ Earth Sci 2017.
[4]
Kwon SM, Kim S, Song NJ, et al. Antiadipogenic and proosteogenic effects of luteolin, a major dietary flavone, are mediated by the induction of DnaJ (Hsp40) Homolog, Subfamily B, Member 1. J Nutr Biochem 2016; 30: 24-32.
[5]
Burton MD, Rytych JL, Amin R, Johnson RW. Dietary luteolin reduces proinflammatory microglia in the brain of senescent mice. Rejuv Res 2016; 19: 286-92.
[6]
Leyva-López N, Gutierrez-Grijalva EP, Ambriz-Perez DL, Heredia JB. Flavonoids as cytokine modulators: A possible therapy for inflammation-related diseases. Int J Mol Sci 2016; 17: 921.
[7]
Lin Y, Shi R, Wang X, Shen HM. Luteolin, a flavonoid with potential for cancer prevention and therapy. Curr Cancer Drug Targets 2008; 8(7): 634-46.
[8]
Faggio C, Sureda A, Morabito S, et al. Flavonoids and platelet aggregation: A brief review. Eur J Pharmacol 2017; 807: 91-101.
[9]
Rafacho BP, Stice CP, Liu C, Greenberg AS, Ausman LM, Wang XD. Inhibition of diethylnitrosamine-initiated alcohol-promoted hepatic inflammation and precancerous lesions by flavonoid luteolin is associated with increased sirtuin 1 activity in mice. Hepatobiliary Surg Nutr 2015; 4: 124.
[10]
Liu G, Zhang Y, Liu C, et al. Luteolin alleviates alcoholic liver disease induced by chronic and binge ethanol feeding in mice1–3. J Nutr 2014; 144: 1009-15.
[11]
Domitrović R, Jakovac H, Tomac J, Šain I. Liver fibrosis in mice induced by carbon tetrachloride and its reversion by luteolin. Toxicol Appl Pharm 2009; 241: 311-21.
[12]
Rodriguez-Ramiro I, Vauzour D, Minihane AM. Polyphenols and non-alcoholic fatty liver disease: Impact and mechanisms. P Nutr Soc 2016; 75: 47-60.
[13]
Wang GG, Lu XH, Li W, Zhao X, Zhang C. Protective effects of luteolin on diabetic nephropathy in STZ-induced diabetic rats. Evid-Based Compl Alt 2011; 2011323171
[14]
Rungsung S, Singh TU, Rabha DJ, et al. Luteolin attenuates acute lung injury in experimental mouse model of sepsis. Cytokine 2018; 11: 333-43.
[15]
Tan X, Jin P, Feng L, et al. Protective effect of luteolin on cigarette smoke extractinduced cellular toxicity and apoptosis in normal human bronchial epithelial cells via the Nrf2 pathway. Oncol Rep 2014; 31: 1855-62.
[16]
Luo Y, Shang P, Li D. Luteolin: A flavonoid that has multiple cardio-protective effects and its molecular mechanisms. Front Pharmacol 2017; 8: 692.
[17]
Basu A, Das AS, Majumder M, Mukhopadhyay R. Antiatherogenic roles of dietary flavonoids chrysin, quercetin, and luteolin. J Cardiovasc Pharm 2016; 68: 89-96.
[18]
Liu Y, Tian X, Gou L, Sun L, Ling X, Yin X. Luteolin attenuates diabetes-associated cognitive decline in rats. Brain Res Bull 2013; 94: 23-9.
[19]
Liu Y, Shi B, Li Y, Zhang H. Protective effect of luteolin against renal ischemia/reperfusion injury via modulation of pro-inflammatory cytokines, oxidative stress and apoptosis for possible benefit in kidney transplant. Int J Exp Clin Res 2017; 23: 5720.
[20]
Ayoobi F, Moghadam-Ahmadi A, Amiri H, et al. Achillea millefolium is beneficial as an add-on therapy in patients with multiple sclerosis: A randomized placebo-controlled clinical trial. Phytomedicine 2018; 52: 89-97.
[21]
Wu Y, Jiang X, Yang K, et al. Inhibition of α-Synuclein contributes to the ameliorative effects of dietary flavonoids luteolin on arsenite-induced apoptotic cell death in the dopaminergic PC12 cells. Toxicol Mech Method 2017; 27: 598-608.
[22]
Oliveira AM, Cardoso SM, Ribeiro M, Seixas RS, Silva AM, Rego AC. Protective effects of 3-alkyl luteolin derivatives are mediated by Nrf2 transcriptional activity and decreased oxidative stress in Huntington’s disease mouse striatal cells. Neurochem Int 2015; 91: 1-2.
[23]
Theoharides TC. Luteolin as a therapeutic option for multiple sclerosis. J Neuroinflammation 2009; 6: 29.
[24]
Crupi R, Impellizzeri D, Bruschetta G, et al. Co-ultramicronized palmitoylethanolamide/luteolin promotes neuronal regeneration after spinal cord injury. Front Pharmacol 2016; 7: 47.
[25]
Shen XF, Teng Y, Sha KH, et al. Dietary flavonoid luteolin attenuates uropathogenic Escherichia. Coli invasion of the urinary bladder. Biofactors 2016; 42(6): 674-85.
[26]
Li J, Li X, Xu W, et al. Antifibrotic effects of luteolin on hepatic stellate cells and liver fibrosis by targeting AKT/mTOR/p70S6K and TGFβ/Smad signalling pathways. Liver Int 2015; 35: 1222-33.
[27]
Peng M, Watanabe S, Chan KW, et al. Luteolin restricts dengue virus replication through inhibition of the proprotein convertase furin. Antiviral Res 2017; 143: 176-85.
[28]
Fan W, Qian S, Qian P, Li X. Antiviral activity of luteolin against Japanese encephalitis virus. Virus Res 2016; 220: 112-6.
[29]
Bai L, Nong Y, Shi Y, et al. Luteolin inhibits hepatitis B virus replication through extracellular signal-regulated kinase-mediated down-regulation of hepatocyte nuclear factor 4α expression. Mol Pharm 2015; 13: 568-77.
[30]
Hytti M, Piippo N, Korhonen E, Honkakoski P, Kaarniranta K, Kauppinen A. Fisetin and luteolin protect human retinal pigment epithelial cells from oxidative stress-induced cell death and regulate inflammation. Sci Rep 2015; 5: 17645.
[31]
Hanneken A, Lin FF, Johnson J, Maher P. Flavonoids protect human retinal pigment epithelial cells from oxidative-stress–induced death. Invest Opthalmol Vis Sci 2006; 47: 3164-77.
[32]
Ozay Y, Guzel S, Erdogdu IH, et al. Evaluation of the wound healing properties of luteolin ointments on excision and incision wound models in diabetic and non-diabetic rats. Rec Nat Prod 2018; 12(4)
[33]
Nash LA, Sullivan PJ, Peters SJ, Ward WE. Rooibos flavonoids, orientin and luteolin, stimulate mineralization in human osteoblasts through the Wnt pathway. Mol Nutr Food Res 2015; 59: 443-53.
[34]
Jones RS, Parker MD, Morris ME. Quercetin, morin, luteolin, and phloretin are dietary flavonoid inhibitors of monocarboxylate transporter 6. Mol Pharm 2017; 14: 2930-6.
[35]
Ali F, Rahul NF, Jyoti S, Siddique YH. Health functionality of apigenin: A review. Int J Food Prop 2017; 20: 1197-238.
[36]
Rao PS, Satelli A, Moridani M, Jenkins M, Rao US. Luteolin induces apoptosis in multidrug resistant cancer cells without affecting the drug transporter function: Involvement of cell line‐specific apoptotic mechanisms. Int J Cancer 2012; 130(11): 2703-14.
[37]
Tsilioni I, Taliou A, Francis K, Theoharides TC. Children with autism spectrum disorders, who improved with a luteolin-containing dietary formulation, show reduced serum levels of TNF and IL-6. Transl Psychiatry 2015; 5e647
[38]
Siddique YH, Ali F. Protective effect of nordihydroguaiaretic acid (NDGA) on the transgenic Drosophila model of Alzheimer’s disease. Chem Biol Interact 2017; 269: 59-66.
[39]
Siddique YH, Beg T, Jyoti S. Protective effect of kaempferol on the transgenic Drosophila model of Alzheimer’s disease. CNS Neurol Disord Drug Targets 2018; 17(6): 421-9.
[40]
Fatima A, Khanam S, Rahul R, et al. Protective effect of tangeritin in transgenic Drosophila model of Parkinson’s disease. Front Biosci Elite 2016; 9: 44-53.
[41]
Siddique YH, Naz F, Jyoti S, Ali F, Fatima A, Khanam S. Protective effect of Geraniol on the transgenic Drosophila model of Parkinson’s disease. Environ Toxicol Pharmacol 2016; 43: 225-31.
[42]
Yasuda MT, Fujita K, Hosoya T, Imai S, Shimoi K. Absorption and metabolism of luteolin and its glycosides from the extract of Chrysanthemum morifolium flowers in rats and Caco-2 cells. J Agric Food Chem 2015; 63: 7693-9.
[43]
Shimoi K, Okada H, Furugori M, et al. Intestinal absorption of luteolin and luteolin 7‐O‐β‐glucoside in rats and humans. FEBS Lett 1998; 438: 220-4.
[44]
Shimoi K, Saka N, Nozawa R, et al. Deglucuronidation of a flavonoid, luteolin monoglucuronide, during inflammation. Drug Metab Dispos 2001; 29(12): 1521-4.
[45]
Kure A, Nakagawa K, Kondo M, et al. Metabolic fate of luteolin in rats: Its relationship to anti-inflammatory effect. J Agric Food Chem 2016; 64: 4246-54.
[46]
Li LP, Wu XD, Chen ZJ, et al. Interspecies difference of luteolin and apigenin after oral administration of Chrysanthemum morifolium extract and prediction of human pharmacokinetics. Int J Pharma Sci 2013; 68: 195-200.
[47]
Lu X, Sun D, Chen Z, et al. Evaluation of hepatic clearance and drug-drug interactions of luteolin and apigenin by using primary cultured rat hepatocytes. Int J Pharma Sci 2011; 66: 600-5.
[48]
Chen T, Li LP, Lu XY, Jiang HD, Zeng S. Absorption and excretion of luteolin and apigenin in rats after oral administration of Chrysanthemum morifolium extract. J Agric Food Chem 2007; 55: 273-7.
[49]
Wilsher NE, Arroo RR, Matsoukas MT, Tsatsakis AM, Spandidos DA, Androutsopoulos VP. Cytochrome P450 CYP1 metabolism of hydroxylated flavones and flavonols: Selective bioactivation of luteolin in breast cancer cells. Food Chem Toxicol 2017; 110: 383-94.
[50]
Boersma MG, van der Woude H, Bogaards, et al. Regioselectivity of phase II metabolism of luteolin and quercetin by UDP-glucuronosyl transferases. Chem Res Toxicol 2002; 15: 662-70.
[51]
Zhou P, Li LP, Luo SQ, Jiang HD, Zeng S. Intestinal absorption of luteolin from peanut hull extract is more efficient than that from individual pure luteolin. J Agric Food Chem 2007; 56: 296-300.
[52]
Chaaban H, Ioannou I, Chebil L, et al. Effect of heat processing on thermal stability and antioxidant activity of six flavonoids. J Food Process Preserv 2017; 41e13203
[53]
Naso LG, Lezama L, Valcarcel M, et al. Bovine serum albumin binding, antioxidant and anticancer properties of an oxidovanadium (IV) complex with luteolin. J Inorg Biochem 2016; 157: 80-93.
[54]
Roy S, Mallick S, Chakraborty T, et al. Synthesis, characterisation and antioxidant activity of luteolin-vanadium (II) complex. Food Chem 2015; 173: 1172-8.
[55]
Kasprzak MM, Erxleben A, Ochocki J. Properties and applications of flavonoid metal complexes. RSC Adv 2015; 5: 45853-77.
[56]
Brown EJ, Khodr H, Hider CR, Rice-Evans CA. Structural dependence of flavonoid interactions with Cu2+ ions: Implications for their antioxidant properties. Biochem J 1998; 330: 1173-8.
[57]
Jullian C, Cifuentes C, Alfaro M, et al. Spectroscopic characterization of the inclusion complexes of luteolin with native and derivatized β-cyclodextrin. Bioorg Med Chem 2010; 18: 5025-31.
[58]
Li J, Wang X, Zhang T, et al. A review on phospholipids and their main applications in drug delivery systems. Asian J Pharm Sci 2015; 10: 81-98.
[59]
Bonifácio BV, da Silva PB, dos Santos Ramos MA, Negri KM, Bauab TM, Chorilli M. Nanotechnology-based drug delivery systems and herbal medicines: A review. Int J Nanomed 2014; 9: 1.
[60]
Saraf S. Applications of novel drug delivery system for herbal formulations. Fitoterapia 2010; 81: 680-9.
[61]
Huang M, Su E, Zheng F, Tan C. Encapsulation of flavonoids in liposomal delivery systems: The case of quercetin, kaempferol and luteolin. Food Funct 2017; 8: 3198-208.
[62]
Oppenheim RC. Nanoparticulate drug delivery systems based on gelatin and albumin. In: Guiot P, Ed.; Polymeric nanoparticles and microspheres. CRC Press, USA, 2018; 8: 1-27
[63]
Walters A, Phillips E, Zheng R, Biju M, Kuruvilla T. Evidence for neuroinflammation in Alzheimer’s disease. Prog Neurol Psychiatry 2016; 20: 25-31.
[64]
Kumar A, Singh A. A review on Alzheimer’s disease pathophysiology and its management: An update. Pharmacol Rep 2015; 67: 195-203.
[65]
Barnard ND, Bush AI, Ceccarelli A, et al. Dietary and lifestyle guidelines for the prevention of Alzheimer’s disease. Neurobiol Aging 2014; 35: S74-8.
[66]
Faden AI, Loane DJ. Chronic neurodegeneration after traumatic brain injury: Alzheimer disease, chronic traumatic encephalopathy, or persistent neuroinflammation? Neurotherapeutics 2015; 12: 143-50.
[67]
Sharp DJ, Scott G, Leech R. Network dysfunction after traumatic brain injury. Nat Rev Neurol 2014; 10: 156.
[68]
Van Cauwenberghe C, Van Broeckhoven C, Sleegers K. The genetic landscape of Alzheimer disease: Clinical implications and perspectives. Genet Med 2016; 18: 421.
[69]
Giri M, Zhang M, Lü Y. Genes associated with Alzheimer’s disease: an overview and current status. Clin Interv Aging 2016; 11: 665.
[70]
Lambert JC, Ibrahim-Verbaas CA, Harold D, et al. Meta-analysis of 74,046 individuals identifies 11 new susceptibility loci for Alzheimer’s disease. Nat Genet 2013; 45: 1452.
[71]
Cox PA, Davis DA, Mash DC, Metcalf JS, Banack SA. Dietary exposure to an environmental toxin triggers neurofibrillary tangles and amyloid deposits in the brain Proc Biol Sci 2016; 283(1823): 20152397.
[72]
Chin-Chan M, Navarro-Yepes J, Quintanilla-Vega B. Environmental pollutants as risk factors for neurodegenerative disorders: Alzheimer and Parkinson diseases. Front Cell Neurosci 2015; 9: 124.
[73]
Yegambaram M, Manivannan BG, Beach TU, Halden R. Role of environmental contaminants in the etiology of Alzheimer’s disease: A review. Curr Alzheimer Res 2015; 12: 116-46.
[74]
Tamano H, Takeda A. Is interaction of amyloid β-peptides with metals involved in cognitive activity? Metallomics 2015; 7: 1205-12.
[75]
Jiang XW, Lu HY, Xu Z, et al. In silico analyses for key genes and molecular genetic mechanism in epilepsy and alzheimer’s disease. CNS Neurol Disord Drug Targets 2018; 17: 608-17.
[76]
Blacher E, Dadali T, Bespalko A, et al. Alzheimer’s disease pathology is attenuated in a CD 38‐deficient mouse model. Ann Neurol 2015; 78: 88-103.
[77]
Singh A, Hasan A, Tiwari S, Pandey LM. Therapeutic advancement in alzheimer disease: New hopes on the horizon? CNS Neurol Disord Drug Targets 2018; 17: 571-89.
[78]
Ambrose GO, Afees OJ, Nwamaka NC, et al. Selection of Luteolin as a potential antagonist from molecular docking analysis of EGFR mutant. Bioinformatics 2018; 14: 241-7.
[79]
Yadav AK, Thakur J, Prakash OM, Khan F, Saikia D, Gupta MM. Screening of flavonoids for antitubercular activity and their structure–activity relationships. Med Chem Res 2013; 22: 2706-16.
[80]
Lipinski CA. Lead-and drug-like compounds: The rule-of-five revolution. Drug Discov Today Technol 2004; 1: 337-41.
[81]
Leeson P. Drug discovery: Chemical beauty contest. Nature 2012; 481(7382): 455.
[82]
Lien EJ, Ren S, Bui HH, Wang R. Quantitative structure-activity relationship analysis of phenolic antioxidants. Free Radic Biol Med 1999; 26: 285-94.
[83]
Kumar A, Nisha CM, Silakari C, et al. Current and novel therapeutic molecules and targets in Alzheimer’s disease. J Formos Med Assoc 2016; 115: 3-10.
[84]
Fu X, Zhang J, Guo L, et al. Protective role of luteolin against cognitive dysfunction induced by chronic cerebral hypoperfusion in rats. Pharmacol Biochem Behav 2014; 126: 122-30.
[85]
Xu B, Li XX, He GR, et al. Luteolin promotes long-term potentiation and improves cognitive functions in chronic cerebral hypoperfused rats. Eur J Pharmacol 2010; 627: 99-105.
[86]
Yao ZH, Yao XL, Zhang Y, Zhang SF, Hu JC. Luteolin could improve cognitive dysfunction by inhibiting neuroinflammation. Neurochem Res 2018; 43: 806-20.
[87]
Sawmiller D, Li S, Shahaduzzaman M, Smith AJ, et al. Luteolin reduces Alzheimer’s disease pathologies induced by traumatic brain injury. Int J Mol Sci 2014; 15: 895-904.
[88]
Wang H, Wang H, Cheng H, Che Z. Ameliorating effect of luteolin on memory impairment in an Alzheimer’s disease model. Mol Med Rep 2016; 13: 4215-20.
[89]
Yu TX, Zhang P, Guan Y, Wang M, Zhen MQ. Protective effects of luteolin against cognitive impairment induced by infusion of Aβ peptide in rats. Int J Clin Exp Path 2015; 8: 6740.
[90]
Liu R, Gao M, Qiang GF, et al. The anti-amnesic effects of luteolin against amyloid β25–35 peptide-induced toxicities in mice involve the protection of neurovascular unit. Neuroscience 2009; 162: 1232-43.
[91]
Omri AE, Han J, Kawada K, Abdrabbah MB, Isoda H. Luteolin enhances cholinergic activities in PC12 cells through ERK1/2 and PI3K/Akt pathways. Brain Res 2012; 1437: 16-25.
[92]
Rezai‐Zadeh K, Douglas Shytle R, Bai Y, et al. Flavonoid‐mediated presenilin‐1 phosphorylation reduces Alzheimer’s disease β‐amyloid production. J Cell Mol Med 2009; 13: 574-88.
[93]
Tang Y, Le W. Differential roles of M1 and M2 microglia in neurodegenerative diseases. Mol Neurobiol 2016; 53: 1181-94.
[94]
Koenigsknecht-Talboo J, Landreth GE. Microglial phagocytosis induced by fibrillar β-amyloid and IgGs are differentially regulated by proinflammatory cytokines. J Neurol 2005; 25: 8240-9.
[95]
Churches QI, Caine J, Cavanagh K, et al. Naturally occurring polyphenolic inhibitors of amyloid beta aggregation. Bioorg Med Chem Lett 2014; 24: 3108-12.
[96]
Lee BI, Suh YS, Chung YJ, Yu K, Park CB. Shedding Light on Alzheimer’s β-Amyloidosis: Photosensitized methylene blue inhibits self-assembly of β-Amyloid peptides and disintegrates their aggregates. Sci Rep 2017; 7: 7523.
[97]
Liu R, Meng F, Zhang L, et al. Luteolin isolated from the medicinal plant Elsholtzia rugulosa (Labiatae) prevents copper-mediated toxicity in β-amyloid precursor protein Swedish mutation overexpressing SH-SY5Y cells. Molecules 2011; 16: 2084-96.
[98]
Choi SM, Kim BC, Cho YH, et al. Effects of flavonoid compounds on β-amyloid-peptide-induced neuronal death in cultured mouse cortical neurons. Chonnam Med J 2014; 50: 45-51.
[99]
Zhang JX, Xing JG, Wang LL, Jiang HL, Guo SL, Liu R. Luteolin Inhibits Fibrillary β-Amyloid1-40-Induced Inflammation in a Human Blood-Brain Barrier Model by Suppressing the p38 MAPK-Mediated NF-κB Signaling Pathways. Molecules 2017; 22: 334.
[100]
Cheng HY, Hsieh MT, Tsai FS, et al. Neuroprotective effect of luteolin on amyloid β protein (25-35)‐induced toxicity in cultured rat cortical neurons. Phytother Res 2010; 24: S102-8.
[101]
Jang S, Kelley KW, Johnson RW. Luteolin reduces IL-6 production in microglia by inhibiting JNK phosphorylation and activation of AP-1. Proc Natl Acad Sci U S A 2008; 105: 7534-9.
[102]
Paterniti I, Cordaro M, Campolo M, et al. Neuroprotection by association of palmitoylethanolamide with luteolin in experimental Alzheimer’s disease models: The control of neuroinflammation. CNS Neurol Disord Drug Targets 2014; 13: 1530-41.
[103]
Khan H, Amin S, Kamal MA, Patel S. Flavonoids as acetylcholinesterase inhibitors: Current therapeutic standing and future prospects. Biomed Pharmacother 2018; 101: 860-70.
[104]
Omar SH, Scott CJ, Hamlin AS, Obied HK. Biophenols: enzymes (β-secretase, Cholinesterases, histone deacetylase and tyrosinase) inhibitors from olive (Olea europaea L). Fitoterapia 2018; 128: 118-29.
[105]
Uriarte-Pueyo I, Calvo I. Flavonoids as acetylcholinesterase inhibitors. Curr Med Chem 2011; 18: 5289-302.
[106]
Choi SH, Hur JM, Yang EJ, et al. Song KS. β-Secretase (BACE1) inhibitors from Perilla frutescens var. acuta. Arch Pharm Res 2008; 31: 183-7.
[107]
Nantakornsuttanan N, Thuphairo K, Kukreja RK, Charoenkiatkul S, Suttisansanee U. Anti-cholinesterase inhibitory activities of different varieties of chili peppers extracts. Int Food Res J 2016; 23: 1953.
[108]
Zheng N, Yuan P, Li C, Wu J, Huang J. Luteolin reduces BACE1 expression through NF-κB and estrogen receptor mediated pathways in HEK293 and SH-SY5Y Cells. J Alzheimers Dis 2015; 45: 659-71.
[109]
Ogunruku OO, Oboh G, Passamonti S, Tramer F, Boligon AA. Capsicum annuum var. grossum (Bell Pepper) Inhibits β-Secretase Activity and β-Amyloid1–40 Aggregation. J Med Food 2017; 20: 124-30.
[110]
Lin TY, Lu CW, Chang CC, Huang SK, Wang SJ. Luteolin inhibits the release of glutamate in rat cerebrocortical nerve terminals. J Agric Food Chem 2011; 59: 8458-66.
[111]
Liu F, Xu K, Xu Z, et al. The small molecule luteolin inhibits N-acetyl-α-galactosaminyltransferases and reduces mucin-type O-glycosylation of amyloid precursor protein. J Biol Chem 2017; 292: 21304-19.
[112]
Wang XX, Tan MS, Yu JT, Tan L. Matrix metalloproteinases and their multiple roles in Alzheimer’s disease. BioMed Res Int 2014; 2014908636
[113]
Tahanian E, Sanchez LA, Shiao TC, Roy R, Annabi B. Flavonoids targeting of IκB phosphorylation abrogates carcinogen-induced MMP-9 and COX-2 expression in human brain endothelial cells. Drug Des Devel Ther 2011; 5: 299-309.
[114]
Kim JK, Kang KA, Ryu YS, et al. Induction of endoplasmic reticulum stress via reactive oxygen species mediated by luteolin in melanoma cells. Anticancer Res 2016; 36(5): 2281-9.
[115]
Choi AY, Choi JH, Yoon H, et al. Luteolin induces apoptosis through endoplasmic reticulum stress and mitochondrial dysfunction in Neuro-2a mouse neuroblastoma cells. Eur J Pharmacol 2011; 668: 115-26.
[116]
Park SH, Park HS, Lee JH, et al. Induction of endoplasmic reticulum stress-mediated apoptosis and non-canonical autophagy by luteolin in NCI-H460 lung carcinoma cells. Food Chem Toxicol 2013; 56: 100-9.
[117]
Gerakis Y, Hetz C. Emerging roles of ER stress in the etiology and pathogenesis of Alzheimer’s disease. FEBS J 2018; 285: 995-1011.
[118]
Salminen A, Kauppinen A, Suuronen T, Kaarniranta K, Ojala J. ER stress in Alzheimer’s disease: A novel neuronal trigger for inflammation and Alzheimer’s pathology. J Neuroinflammation 2009; 6: 41.
[119]
Kim S, Chin YW, Cho J. Protection of cultured cortical neurons by luteolin against oxidative damage through inhibition of apoptosis and induction of heme oxygenase-1. Biol Pharm Bull 2017; 40: 256-65.
[120]
Zhu L, Bi W, Lu D, Zhang C, Shu X, Lu D. Luteolin inhibits SH-SY5Y cell apoptosis through suppression of the nuclear transcription factor-κB, mitogenactivated protein kinase and protein kinase B pathways in lipopolysaccharide-stimulated cocultured BV2 cells. Exp Ther Med 2014; 7: 1065-70.
[121]
Wu PS, Yen JH, Kou MC, Wu MJ. Luteolin and apigenin attenuate 4-hydroxy-2-nonenal-mediated cell death through modulation of UPR, Nrf2-ARE and MAPK pathways in PC12 cells. PLoS One 2015; 10e0130599
[122]
Bandaruk Y, Mukai R, Terao J. Cellular uptake of quercetin and luteolin and their effects on monoamine oxidase-A in human neuroblastoma SH-SY5Y cells. Toxicol Rep 2014; 1: 639-49.
[123]
Lamy S, Moldovan PL, Saad AB, Annabi B. Biphasic effects of luteolin on interleukin-1β-induced cyclooxygenase-2 expression in glioblastoma cells. Mol Cell Res 2015; 1853: 126-35.
[124]
Wszelaki N, Melzig MF. Additive protective effects of luteolin and pyruvate against 6-hydroxydopamine and 3-hydroxykynurenine induced neurotoxicity in SH-SY5Y cells. Pharmacology 2013; 4: 369.
[125]
Guo DJ, Li F, Yu PH, Chan SW. Neuroprotective effects of luteolin against apoptosis induced by 6-hydroxydopamine on rat pheochromocytoma PC12 cells. Pharm Biol 2013; 51: 190-6.
[126]
Lin P, Tian XH, Yi YS, Jiang WS, Zhou YJ, Cheng WJ. Luteolininduced protection of H2O2induced apoptosis in PC12 cells and the associated pathway. Mol Med Rep 2015; 12: 7699-704.
[127]
Dirscherl K, Karlstetter M, Ebert S, et al. Luteolin triggers global changes in the microglial transcriptome leading to a unique anti-inflammatory and neuroprotective phenotype. J Neurol 2010; 7: 3.
[128]
Zhu LH, Bi W, Qi RB, Wang HD, Lu DX. Luteolin inhibits microglial inflammation and improves neuron survival against inflammation. Int J Neurol 2011; 121: 329-36.
[129]
Zhu LH, Bi W, Qi RB, et al. Luteolin reduces primary hippocampal neurons death induced by neuroinflammation. Neurol Res 2011; 33: 927-34.
[130]
Zhou F, Chen S, Xiong J, Li Y, Qu L. Luteolin reduces zinc-induced tau phosphorylation at Ser262/356 in an ROS-dependent manner in SH-SY5Y cells. Biol Trace Elem Res 2012; 149: 273-9.
[131]
Zhou F, Qu L, Lv K, et al. Luteolin protects against reactive oxygen species‐mediated cell death induced by zinc toxicity via the PI3K-Akt-NF‐κB-ERK‐dependent pathway. J Neurosci Res 2011; 89: 1859-68.
[132]
Hu LW, Yen JH, Shen YT, Wu KY, Wu MJ. Luteolin modulates 6-hydroxydopamine-induced transcriptional changes of stress response pathways in PC12 cells. PLoS One 2014; 9e97880
[133]
Kao TK, Ou YC, Lin SY, et al. Luteolin inhibits cytokine expression in endotoxin/cytokine-stimulated microglia. J Nutr Biochem 2011; 22: 612-24.
[134]
Wruck CJ, Claussen M, Fuhrmann G, et al. Luteolin protects rat PC 12 and C6 cells against MPP+ induced toxicity via an ERK dependent Keapl-Nrf2-ARE pathway. In: Gerlach M, Deckert J, Double K, Koutsilieri E, Eds. Neuropsychiatric Disorders An Integrative Approach, J Neural Transm Suppl. Springer Vienna 2007; Vol. 72: pp. 57-67.
[135]
Lin CW, Wu MJ, Liu IY, Su JD, Yen JH. Neurotrophic and cytoprotective action of luteolin in PC12 cells through ERK-dependent induction of Nrf2-driven HO-1 expression. J Agric Food Chem 2010; 58: 4477-86.
[136]
Bernardo J, Ferreres F, Gil-Izquierdo Á, Valentao P, Andrade PB. Medicinal species as MTDLs: Turnera diffusa Willd. Ex Schult inhibits CNS enzymes and delays glutamate excitotoxicity in SH-SY5Y cells via oxidative damage. Food Chem Toxicol 2017; 106: 466-76.
[137]
Bajpai VK, Alam MB, Ju MK, et al. Antioxidant mechanism of polyphenol-rich Nymphaea nouchali leaf extract protecting DNA damage and attenuating oxidative stress-induced cell death via Nrf2-mediated heme-oxygenase-1 induction coupled with ERK/p38 signaling pathway. Biomed Pharmacother 2018; 103: 1397-407.
[138]
Liu Y, Huang J, Zheng X, et al. Luteolin, a natural flavonoid, inhibits methylglyoxal induced apoptosis via the mTOR/4E-BP1 signaling pathway. Sci Rep 2017; 7: 7877.
[139]
Byun EB, Cho EJ, Kim YE, Kim WS, Byun EH. Neuroprotective effect of polysaccharide separated from Perilla frutescens Britton var. acuta Kudo against H2O2-induced oxidative stress in HT22 hippocampus cells. Biosci Biotech Biochem 2018; 2: 1-5.
[140]
Huang S, Meng N, Liu Z, et al. Neuroprotective effects of Taraxacum officinale wigg. extract on glutamate-induced oxidative stress in HT22 Cells via HO-1/Nrf2 pathways. Nutrient 2018; 10: 926.
[141]
Moniruzzaman M, Chin YW, Cho J. HO-1 dependent antioxidant effects of ethyl acetate fraction from Physalis alkekengi fruit ameliorates scopolamine-induced cognitive impairments. Cell Stress 2018; 15: 1-0.
[142]
Chalatsa I, Arvanitis DA, Mikropoulou EV, et al. Beneficial effects of sideritis scardica and cichorium spinosum against amyloidogenic pathway and tau misprocessing in alzheimer’s disease neuronal cell culture models. J Alzheimer Dis 2018; pp. 1-4.
[143]
Lin TY, Lu CW, Wang SJ. Luteolin protects the hippocampus against neuron impairments induced by kainic acid in rats. Neurotoxicology 2016; 55: 48-57.
[144]
Dabo S, Maillard P, Rodriguez MC, et al. Inhibition of the inflammatory response to stress by targeting interaction between PKR and its cellular activator PACT. Sci Rep 2017; 7: 16129.
[145]
Jang S, Dilger RN, Johnson RW. Luteolin inhibits microglia and alters hippocampal-dependent spatial working memory in aged Mice-3. J Nutr 2010; 140: 1892-8.
[146]
Xu J, Wang H, Lu X, et al. Posttraumatic administration of luteolin protects mice from traumatic brain injury: Implication of autophagy and inflammation. Brain Res 2014; 1582: 237-46.
[147]
Ali F, Jyoti S, Naz F, et al. Therapeutic potential of luteolin in transgenic Drosophila model of Alzheimer’s disease. Neurosci Lett 2018; 23(692): 90-9.

Rights & Permissions Print Cite
© 2024 Bentham Science Publishers | Privacy Policy