Antioxidant activity
|
Antioxidative effect of flavonoids in biological system is related to:
-
-
ability to scavenge ROS including singlet oxygen (1 O2), hydroxyl radical (OH●), hydrogen peroxide (H2O2), superoxide anions (O2−●), perhydroxy radical (HO2●), lipid radical (LO●), and lipid peroxy radical (LOO●)
-
-
ability to scavenge nitric reactive radical (HOONO, NO, NO3, and others)
-
-
suppression of oxidative enzymes;
-
-
chelation of metal ions (Fe2+, Cu2+, Zn2+ and, Mg2+)
-
-
increased activity and protection of antioxidant enzymes
-
-
synergistic action with other antioxidants
-
-
inhibition of Nrf2 degradation and increase in transcriptional activity of protective genes such as antioxidant proteins and phase II detoxification enzymes
|
Inhibition of
nitrosation and
nitration
|
-
-
flavonoids directly react with ONOO− or scavenge ·OH and ·NO2 generated by ONOO−, thus blocking the nitration reaction
-
-
peroxynitrite scavenging and further inhibition of tyrosine nitration, DNA strand breaks, and oxidation of low-density lipoproteins
|
Reduction in
iron ions
|
Iron reduction has multiple anticancer actions, including:
-
-
depriving neoplastic cells of a key required nutrient,
-
-
producing an antiangiogenic effect due to decreased ferritin
-
-
inhibition of the formation of 8-hydroxydeoxyguanosine in vivo
-
-
influencing cell cycle regulation at multiple sites
-
-
tumor suppressor genes may have specific vulnerability to the iron-catalyzed Fenton reaction
-
-
decreasing prooxidant activity of iron and other metal ions
|
Anti-inflammatory
effects
|
Anti-inflammatory effect of flavonoids is related to:
Antioxidant activity
Regulation of inflammatory cells
Regulation of proinflammatory enzymes
Modulation of proinflammatory mediators
Modulation of pro-inflammatory gene expression
Inhibition of the Toll-like receptor 4 (TLR-4) signaling pathway |
Antimutagenic mechanisms
|
Anti-mutagenic effect of flavonoids is related to:
Extracellular mechanisms
-
-
inhibition of mutagen uptake
-
-
inhibition of endogenous mutagens (inhibition of nitrosation; modification of the intestinal flora)
-
-
formation of complexes with mutagens and/or their deactivation
-
-
preferable absorption of protective compounds
Cellular mechanisms
-
-
inhibition of or competition with mutagens (ROS scavenging; protection of DNA nucleophilic sites)
-
-
trapping and detoxification in non-target cells
-
-
modification of transmembrane transport
-
-
altered function of xenobiotic metabolizing enzymes (inhibition of promutagen activation; activation of detoxification pathways)
-
-
modulation of DNA metabolism and repair
-
-
enhancement of apoptosis
-
-
maintenance of genomic stability
|
Process of
detoxification
by fibers
|
|
Enzyme
inhibition
|
Enzyme inhibition effect of flavonoids is related to:
-
-
cyclooxygenase-2
-
-
inducible nitric oxide synthase
-
-
xanthine oxidase
-
-
phase I enzyme (P450 enzymes, block activation of carcinogens; CYP1A1, CYP1A2, CYP1B1, CYP2E1, CYP3A4, CYP19)
|
Enzyme
induction and
enhanced
detoxification
|
Enzymes included in this reaction are:
-
-
sulfotransferases (SULT1A1, SULT1A3, SULT1E1)
-
-
UDP-glucuronosyltransferases (UGT, UGT1A1)
-
-
quinone reductase (QR)
-
-
acetyltransferases
Effect is related to:
-
-
inhibition of organic anion transporters participating in the uptake of nephrotoxic compounds by flavonoids and their phase II metabolites (sulfates, glucuronides)
-
-
xenobiotic uptake and efflux by solute carrier transporters; organic anion transporting polypeptides (OATPs), organic anion and cation transporters (OATs and OCTs, respectively) and ATP-binding cassette (ABC) transporters
|
Inhibition of
signal transduction
pathways
|
Regulate the signal transduction pathways including:
-
-
NF-κB signaling
-
-
PI3K/Akt/mTOR signaling
-
-
Hedgehog signaling
-
-
Akt, MAPKs, p53
-
-
androgen receptor (AR), and estrogen receptor (ER) pathways
-
-
STAT signaling
-
-
AP-1 signaling
-
-
Notch-1 signaling
-
-
Wnt/β catenin signaling
-
-
Insulin-like growth factor (IGF) signaling
-
-
NF-E2-related factor 2/antioxidant response element (Nrf2/ARE) pathway
|
Inhibition of
cell
proliferation
|
Suppression of cancer stem cells self-renewal, progenitor formation, and clonal growth
Telomerase inhibition
The disruption of microtubules in mitosis
Inhibition of topoisomerase I and II
Proteasome inhibition
Cell cycle inhibition
-
-
by suppressing expression of cyclin A, cyclin B, Cdk2
-
-
by enhancing the p21 and p27 levels
-
-
by inhibiting activity of hTERT in tumor cells
Inactivation of prooxidative enzymes
Inhibition of ornithine decarboxylase and polyamines synthesis
Modulation of signal transduction pathways Inhibition of various protein kinases:
-
-
protein tyrosine kinase (PTK),
-
-
cAMP-dependent protein kinase (PKA),
-
-
phosphoinositide 3-kinase (PI3K),
-
-
protein kinase C (PKC),
-
-
mitogen-activating protein kinases (MAPK),
-
-
cyclin-dependent kinases (CDKs),
-
-
p34cdcz kinase
-
-
focal adhesion kinase (FAK)
Inhibition of DNA synthesis enzymes
|
Induction of cell
differentiation
|
|
Inhibition of
oncogene expression
|
|
Induction of tumor
suppressor gene
expression
|
-
-
modulation of p53, Rh, Bcl-2, p21, p27, BRCA1, BRCA2, RhoB
|
Induction of
cell-cycle arrest
|
-
-
quercetin exerts growth inhibitory effect on human colon cancer via related 17 kDa protein, which blocks cell transition from G0/G1 into the S phase of the cycle
-
-
curcumin induces cell cycle arrest in various cell types, preferentially in G2/M phase
|
Induction of
apoptosis
|
By inhibition of heat shock proteins
Topoisomerase-mediated apoptosis
Mitochondrial toxin-mediated apoptosis
Oxidative stress-induced apoptosis
Other mechanisms for inducing apoptosis
-
-
upregulation of Bax and p21
-
-
elevation of intracellular cAMP to high levels with cAMP analogs, adenylate cyclase activators, or phosphodiesterase inhibitors.
-
-
increase in Ca2+ levels (endonuclease activation) and wild-type p53 overexpression.
-
-
upregulation of TRAIL-R1 and TRAIL-R2
-
-
downregulation of p53 and antiapoptotic proteins (Bcl2, IAP, c-FLIP, Akt)
|
Enhancement of
immune functions
and surveillance
|
-
-
increased activity of macrophages, B, T, and NK cells
-
-
upregulation of toll-like receptors (TLR2 and TLR4)
-
-
inhibition of M1 to M2 macrophage polarization
-
-
triggering Immunogenic cell death effect (ICD)
-
-
downregulation of the PD-L1 expression through JAK-STAT and NF-κB pathways
|
Antiangiogenesis
|
-
-
inhibition of EGF, TGFα, bFGF, VEGF, VEGFR, MMPs, claudin, β-catenin, COX2
|
Overcoming
resistance
to cancer therapy
|
-
-
inhibition of P-gp (transcriptional downregulation of MDR-1; direct high-affinity binding to nucleotide-binding domain (NBD); ATPase inhibition; nucleotide hydrolysis; energy-dependent drug interactions with membranes enriched in transporters)
-
-
inhibition of CYPs
-
-
induction of apoptosis
-
-
inhibition of NF-κB
-
-
inhibition of glycolysis by inhibitors of glycolytic enzymes
|
Interaction with cellular drug transport systems
|
-
-
inhibition of P-gp pump, MRP1, BCRP
-
-
competition with glucose for transmembrane transport
|
Inhibition of
NF-κB
|
Inhibition of NF-κB leads to negative effects:
-
-
viability (Survivin, Bcl-2, Bcl-xL, c-FLIP, c-IAP, XIAP)
-
-
proliferation (Cyclin D1, CDK, c-Myc, COX-2, IL-1, IL-6, TNF)
-
-
invasion (ELAM-1, ICAM-1, MMP, urokinase-type plasminogen activator (u-PA), VCAM-1)
-
-
angiogenesis (angiopoietin, VEGF)
-
-
metastasis (CXCR4)
|
Inhibition of
cell adhesion
and invasion
|
-
-
by inhibiting matrix metalloproteinase (MMP2, MMP9)
-
-
by inhibiting intercellular adhesion molecule (ICAM; mainly ICAM-1)
-
-
by inhibiting cysteine proteases and serine proteases such as u-PA and u-PAR
-
-
by stimulating cell-cell communication in transformed cells, decreasing malignancy
-
-
modulation of the platelet function
-
-
downregulation of Rac1, CXCR4, HIF-1α
-
-
inhibition of NF-κB and Akt signaling pathways
|
Inhibitors of
metastasis
|
-
-
altered expression of oncogenes and tumor suppressors
-
-
reduced migration and adhesion of cells
-
-
reduced adhesion to laminin substrate and ability of invasion or migration
-
-
suppression of cells migration by downregulating cell motility-related genes Rac1 and VASP
-
-
EMMPRIN reduction via the PTEN/Akt/HIF-1α signaling pathway
-
-
reduced cell motility due to downregulation of MMP-2 and MMP-9
-
-
upregulation of tissue inhibitor of metalloproteinase
-
-
inhibition of hypoxia-inducible genes involved in invasion/migration, such as uPAR, ADM, and MMP2
-
-
decreased expression of CXCR4, through the NF-κB suppression
-
-
inhibition of TNF-α-induced migration and EMT through Akt/NF-κB pathway inhibition
-
-
inhibition of M1 to M2 macrophage polarization
-
-
inhibition of EMT-inducing transcription factors (Snail, Zeba, and/or Twist)
|
Disruption of
tumor cell
glycolytic
metabolism
|
-
-
reduction in glucose consumption and lactate production
-
-
inhibition of expression of HIF-1 and its target genes (GLUT1, HKII, and VEGF)
-
-
modulation of the expression of glucose transporters (especially GLUT1 and GLUT4)
-
-
inhibition of glycolysis by inhibitors of glycolytic enzymes hexokinase 2 (HK2), phosphofructokinase (PFK), muscle isozyme pyruvate kinase M2, (PKM2), and lactate dehydrogenase A (LDHA)
-
-
inhibition of lactate transport through monocarboxylate transporters (MCTs)
|
Receptor binding
|
High concentrations could modulate receptor or enzyme activity in vivo:
|
Prevention of
DNA adduct
formation or
DNA intercalation
|
-
-
free radicals scavenging
-
-
protection against DNA adduct formation
-
-
protection against chromosome aberration
-
-
inhibition of topoisomerase enzymes (accumulation of DNA breaks and mutations without covalent binding to DNA)
-
-
prevention of carcinogenesis by N-nitrosamines due to increased glutathione-S-transferase
-
-
protection against H2O2-induced DNA damage by inhibiting DNA strand breaks
-
-
protection against 8-hydroxy-2′-deoxyguanosine (8-OHdG) generation and downregulation of nuclear phospho histone H2AX expression
-
-
protection against high glucose-induced DNA fragmentation, chromatin condensation, and hypodiploid DNA
-
-
protection against oxidative DNA damage (intercalation into the DNA duplex and reaction with free radicals)
-
-
helical stabilization by low flavonoid concentration
-
-
helix opening by high flavonoid concentration
-
-
interaction with telomerase sequences and stabilization of the G-quadruplex structure
-
-
reactivation of p53 and DNA repair
|
Regulation of
steroid
hormone
metabolism
|
-
-
binding to the androgen receptor (AR) and estrogen receptor
-
-
prevention of estrogen action in promoting growth of certain tumors
-
-
decrease in estrogen biosynthesis through aromatase inhibition
|
Effects on
biomarkers in
tumor promotion
|
-
-
reduction in ROS, xanthine oxidase, NADPH oxidase, and peroxidase
-
-
reduction in PKC, calcium release, and calcium canal activation
-
-
reduction in ornithine decarboxylase, and polyamine biosynthesis
-
-
reduction in cyclooxygenase I and II, arachidonic acid metabolism, prostaglandins, and thromboxanes
-
-
reduction in MAPK cascades
-
-
inhibition of AP-1 (TPA-response element) and NFκB (IκB kinase, PKC, ROS)
-
-
downregulation of c-Jun, c-Fos, c-Myc, Bax, and Cdks (inhibiting oncogene activation)
-
-
modulation of p53, Rh, Bcl-2, p21, p27
|
Prooxidative effect
|
Prooxidative effect of flavonoids is related to:
-
-
in the presence of O2, transition metals catalyze phenolic redox cycling and formation of reactive oxygen species (ROS) and phenoxyl radicals that can damage DNA, lipids, and other biological targets
-
-
autoxidation of flavonols with pyrogallol or catechol B rings in the presence of transition metals increase production of ROS and accelerate oxidation of low-density lipoproteins during the propagation phase
-
-
peroxidase-catalyzed oxidation of phenol B ring-containing flavonoids to pro-oxidant phenoxyl radicals
-
-
peroxidase-mediated oxidation of catechol B ring-containing flavonoids results in the formation of semiquinone- and quinone-type metabolites
-
-
prooxidant phenoxyl radicals cause mitochondrial toxicity
|
Antibacterial,
antiviral, and
antiparasitic activity
|
-
-
reduced tumor formation related to many viruses and bacteria capable to induce inflammation and tumor including: Epstein–Barr virus (EBV), human immunodeficiency virus (HIV) infection, chronic infection with Hepatitis B (HBV) and C (HCV) virus, human T cell leukemia virus type 1 (HTLV-I)
-
-
Helicobacter pylori (stomach cancer and MALT-lymphoma)
-
-
Opisthorchis viverrini infection (bile duct, a rare kind of adenocarcinoma)
-
-
Schisostoma infection—Schistosomiasis (bladder and colon)
-
-
Herpes simplex virus type 1 and 2 infection (human cervical cancer)
|
The main mechanisms
of antiviral,
antibacterial, and
antiparasitic action
|
-
-
inhibition of growth and adhesion to mucosal cells
-
-
decrease in gastric concentration
-
-
enhanced IgA response to the virus
-
-
improvement of mucosal barrier function
-
-
suppression of proinflammatory cytokine release
|
Beneficial effect
on microbiota
activity
|
Direct effects of flavonoids are related to:
-
-
positive effect on microbiota
-
-
reduced production of deleterious endotoxins
-
-
positive effect on the production of beneficial short-chain fatty acids (SCFA)
-
-
systemic effect on glucose homeostasis, lipid, and energy metabolism
-
-
prevention of the harmful effects of food, e.g., oxidants and pharmacological insults
-
-
inhibition of hydrolytic enzymes, e.g., pancreatic lipase, amylase
-
-
alleviation of intestinal permeabilization and associated paracellular transport of endotoxins that can initiate local/systemic inflammation
-
-
modulation of the secretion of gut hormones by enteroendocrine cells, which have local effects and systemically modulate energy and metabolic homeostasis
-
-
modulation of the GI tract immune system, e.g., Paneth cells (PC)
-
-
neutralization of dietary carcinogens
Indirect effects of microbiota on cancer are related to:
-
-
beneficial effect of microbiota on inhibition of tumor initiation
-
-
beneficial effect of microbiota on prevention and cure of colon cancer
-
-
beneficial effect of microbiota on immune system
-
-
beneficial effect of microbiota in the protection of the damaged immune system after chemo- and radiotherapy
|