Skip to main content
. 2020 May 30;7(6):31. doi: 10.3390/medicines7060031

Table 4.

Anti-inflammatory effects of the selected flavonoids.

Flavonoid Model Mechanisms
Biochanin A In vitro: cytokine release from keratinocytes and HMEC-1 endothelial cells in serum from patients with Behçet’s disease [41]
In vitro: LPS-induced inflammation in HUVED cells [42]

In vivo: focal cerebral ischemia–reperfusion model [43]
In vitro: LPS-induced pro-inflammatory responses in murine BV2 microglial cells [44]
In vitro: LPS-induced inflammatory cytokines and mediators production in murine BV2 microglial cells [45]
In vivo: LPS/GalN-induced liver injury [46]


Ex vivo: interleukin-1β-induced catabolic inflammation through the modulation of NFκB cellular signaling in primary rat chondrocytes [47]
In vitro and in vivo: LPS-induced damage of dopaminergic neurons [48]
In vivo: cisplatin induced acute kidney injury in mice [49]
In vivo: ritonavir induced hepatotoxicity [50]
In vivo: transient coronary ligation in Sprague-Dawley rats [51]
In vivo: LPS-induced acute lung injury in mice [52]
In vitro: LPS-induced NO production, LPS-induced IKK activity, LPS-induced phosphorylation of IκBα and p38 MAPK [53]
In vitro: CCl4-induced hepatotoxicity in rats [54]

In vivo: Sprague-Dawley rat subarachnoid hemorrhage [55]
In vitro: barrier function of intestinal epithelial CaCo-2/TC-7 cells via TEER measurements [56]

In vitro: LPS-stimulated macrophages [57]

In vivo: focal cerebral ischemia established by middle cerebral artery occlusion [58]
↓IL-8


↓IL-8, TNF-α, VCAM-1, ICAM-1, E-selection
↑PPAR-γ
↓IL-8, TNF-α, P38 expression

↓IL-1β, TNF-α, NO, phosphorylation of JNK, ERK and p38
↓IL-1β, TNF-α, NO, PGE2, NF-κB
↑PPAR-γ

IL-1β, TNF-α, ALT, AST, MDA, TXNIP, NLRP3 inflammasome
↑SOD, GPx, catalase, HO-1, Nrf2
↓IL-1β, TNF-α, IL-6, IL-1α, INFγ, IL- 2, GM-CSF, fractalkine, MCP-1, MIP-3α, LIX

↓IL-1β, TNF-α, IL-6, phosphorylation of JNK, ERK and p38,
↓IL-1β, TNF-α, caspase-3, p53 protein

↓IL-1β, IL-6
↑IL-10

↓IL-1β, IL-18, IL-6, TNF-α
IL-1β, IL-6, TNF-α, TLR4/NF-κB
↑PPAR-γ

IL-6, TNF-α
PPAR-γ, PPAR-α
iNOS, COX2, TNF-α

sTNFR1, TNF-α, NF-κB, ERK, tyrosine phosphorylation
↑SOD, GSH-Px, HO-1, Nrf2
↓iNOS, phosphorylation of IκBα and p38 MAPK
↓TLR/NF-κB
Prunetin In vitro: barrier function of intestinal epithelial CaCo-2/TC-7 cells via TEER measurements [56]
In vitro: LPS-stimulated RAW 264.7 macrophage [59]
In vivo: LPS-induced septic shock [59]
In vitro: LPS-induced in- flammatory response and MUC5AC expression [60]
↓sTNFR1, TNF-α, NF-κB, ERK, tyrosine phosphorylation
↓iNOS, PGE2, COX2, NF-κB, p38, IL-1β, TNF-α
IL-1β, TNF-α
IL-8, IL-6, MUC5AC, TLR4/MyD88
Daidzein In vitro: LPS-stimulated macrophages [57]
In vivo: angiotensin II-induced AAA [61]

In vivo: 5-fluorouracil-induced intestinal mucositis [62]
In vivo: cisplatin-induced kidney injury [63]


In vivo: ischemia/reperfusion injury-induced neurological function deficits in Sprague-Dawley [64]
↓IL-6
↓IL-1β, TNF-α, NF-κB, iNOS, COX-2, p38MAPK, TGF-β1
↓IL-1β, IL-6, TNF-α, NO, COX-2

↓IL-6, TNF-α, MDA, NO, COX-2, MAPK
↑SOD, GSH

↓TNF-α, NF-κB subunit p65
Genistein In vitro: LPS-stimulated macrophages [57]

In vitro: homocysteine-induced endothelial cell inflammation [65]
In vivo: cyclophosphamide - induced hepatotoxicity [66]
In vivo: LPS-induced microglial activation in murine BV2 microglial cell line and primary microglial culture [67]
In vivo: imiquimod- induced psoriasis-like lesions in mice [68]
In vivo: DSS-induced murine colitis [69]

In vivo: NASH mouse model [70]
In vivo: chronic sleep deprivation [71]


In vitro: barrier function of intestinal epithelial CaCo-2/TC-7 cells via TEER measurements [56]
In vivo: mouse model of periodontitis [72]

In vivo: high-fat high-fructose diet-induced NASH rats [73]
In vitro: angiotensin II-stimulated CRP and MMP-9 expression in VSMC [74]
↓IL-6, TNF-α
PPAR-γ, PPAR-α
NF-κB subunit p65, IL-6, ICAM-1

↓IL-1β, COX-2, MPO

↓IL-1β, IL-6, COX-2, iNOS, TNF-α, NF-κB, MAPK

↓IL-1β, IL-6, IL-8, TNF-α, IL-17, IL-23, CCL2, NF-κB, VEGFA
↓IL-1β, IL-18, TNF-α, MPO, NLRP3 inflammasome
↓IL-6, TNF-α,
↓IL-1β, IL-6, COX-2, iNOS, TNF-α, NF-κB p65
↑HO-1, Nrf2
↓sTNFR1, tyrosine phosphorylation

↓TNF-α, COX-2, Nos2, ICAM-1, MMP-2, MMP-9
↓TNF-α, NF-κB

↓p-ERK1/2, p-p38, NF-κB
↑PPAR-γ,
Rutin In vivo: HMGB1-induced inflammation and CLP-induced sepsis model [75]
In vivo: LPS-induced acute endotoxemic kidney injury in C57BL/6 mice [76]
In vivo: NaF-induced neurotoxicity [77]
In vivo: HgCl2-induced nephrotoxicity [78]
In vivo: HgCl2-induced hepatotoxicity [79]

In vitro: PMA-induced neutrophil stimulation [80]
↓TLR 4, RAGE, p38 MAPK, VCAM-1, ICAM-1, ERK1/2, NF-κB
↓TLR 4, COX-2, TNF-α, IL-6, SIRT1, NF-κB

↓IL-1β, IL-6, TNF-α
↓IL-1β, IL-33, TNF-α, NF-κB, Bcl-3
↓IL-1β, TNF-α, NF-κB, Bcl-3, Bcl-2, Bax, p53, p38 MAPK, caspase-3
↓NO, TNF-α, MPO
Taxifolin In vitro: osteoclastogenesis [81]
In vivo: and ovariectomy-induced osteoporosis [81]
In vivo: osteolysis model [82]
In vitro: on IgE/Ag-stimulated mast cells including BMMCs [83]
In vivo: acetaminophen-induced liver injury [84]
↓AKT, RANKL
↓TNF-α, IL-1β, NF-κB, MAPK, NFATc1, MMP-9, cathepsin K, TRAP
↓MAPK, p38, ERK, JNK; RANKL, NF-κB
↓LTC4, IL-6, COX-2, TNF-α, NF-κB

↓ inhibiting metabolic activation mediated by CYP450 enzymes