Table 1.
Study Type | Subjects | Dose | Effect | Ref. |
---|---|---|---|---|
In vitro | Splenic lymphocytes, CTLs and LAKs | 25–50 μM | Suppresses mitogen-, IL-2-, and alloantigen-induced proliferation of splenic lymphocytes; development of antigen-specific CTLs; LAK cells were less sensitive. | [10] |
In vitro | T lymphocytes and Macrophages | 1–20 µM | Suppresses: T cells proliferation and secretion of IFN-γ and IL-4; B cells proliferation and production of IgG1 and IgG2a isotypes; IL-1, IL-6, TNF-α. Enhances: IL-10; down-regulates the expression of CD28 on CD4+ T cells and of CD80 on macrophages. | [13] |
In vitro | NK92 cell line | 1.5 µM | Enhances perforin expression and cytotoxic activity acting via NKG2D-dependent JNK and ERK-1/2 pathways. | [12] |
Ex vivo In vivo |
Splenocytes C57BL/6 and BALB/c mice |
25–75 µM 4 mg/kg, i.p. |
Suppresses the CD4+CD25+ subsets; downregulated secretion of TGF-β. Enhances IFN-γ expression in CD8+ T cells. |
[15] |
In vitro | RAW 264.7 cell line and BV-2 cell line | 50 μM | Suppresses IL-6, M-CSF, MCP-1, MCP-5, CD54, IL-1ra, IL-27, and TNF-α in both cell lines. Inhibits the TLR4/NF-κB/STAT signaling cascade |
[20] |
In vivo | NOD mice were given | 250 mg/kg | Decreases in expression of CCR-6. Inhibits CD11b+F4/80hi macrophages. It reduces CCR6+ IL-17-producing cells and CD11b+F4/80hi in the pancreas. It reduces migration of splenocytes toward media containing CCL20. Prevents type 1 diabetes in NOD mice. | [32] |
In vitro | U-937 Jurkat HeLa and H4 cells lines |
0.5–25 μM | Suppresses TNF-induced phosphorylation and nuclear translocation of the p65 subunit of NFκ B, and NFκ B-dependent reporter gene transcription. It suppresses TNF-induced NFκ B activation. Blocks NFκ B activation induced by PMA, LPS, H2O2, and okadaic acid. Suppresses AP-1. Inhibits the TNF-induced activation of MEK and JNK. Abrogates TNF-induced cytotoxicity and caspase activation. Suppresses ROI generation and lipid peroxidation. | [37] |
In vitro | Bone-derived cell cultures and MC3T3-E1 cell lines | 5 μM | Inhibits RANKL-induced acetylation and nuclear translocation of NFκ B. Induces Sirt1-p300 association in bone-derived and preosteoblastic cells, leading to deacetylation of RANKL-induced NFκ B, inhibition of NFκ B transcriptional activation, and osteoclastogenesis. It activates the bone transcription factors Cbfa-1 and Sirt1 and induces the formation of Sirt1-Cbfa-1 complexes. It regulates the balance between the osteoclastic versus osteoblastic activity. It could exert a therapeutic potential for treating osteoporosis and rheumatoid arthritis-related bone loss. | [38] |
In vitro | MH7A cell lines | 100 μM | Induces MH7A cell apoptosis by activating caspase-9 and the effector caspase-3, reduces Bcl-XL expression, allowing cytochrome c release from the mitochondria into the cytosol, in a sirtuin 1-dependent manner. It could suppress hyperplasia of synovial cells, a critical factor of rheumatoid arthritis. | [40] |
In vitro | RAW264.7 and HEK 293T cell lines | 30, 50, 75, 100 μM | Inhibits TRIF signaling in the TLR3 and TLR4 pathway by targeting TANK-binding kinase 1 and RIP1 in TRIF complex. Modulates TLR-derived signaling and inflammatory target gene expression. It could alter susceptibility to microbial infection and chronic inflammatory diseases. | [46] |
In vitro | RAW 264.7 cell line | 50 μM | Suppresses LPS-induced TRAF6 expression and ubiquitination, attenuates the LPS-induced TLR4–TRAF6, MAPK, and AKT pathways. It could exert anti-inflammatory effects. | [47] |
In vitro | Mouse bone-marrow cells J774 cell line |
5 μM | Inhibits the accumulation of acetylated α-tubulin and suppressing NLRP3-inflammasome assembly. It prevents the NLRP3-related inflammatory diseases. | [53] |
In vitro | AR42J cell line | 10–100 μM | It decreases CD14 and IRAK1 expression and increases the p38 MAPK protein phosphorylation. It exerts antioxidant properties either by a Myd88-dependent way not involving IRAK1 or by a TRIF dependent pathway. | [55] |
In vitro | RAW 264.7 THP-1 HUVEC cell lines and PBLs |
6.25–12.5–25–50 μM 3.13–6.25–12.5–25 μM 10–20–30 μM 6.25–12.5–25 μM |
Modulates many mediators of the inflammatory response. Its effects are context-dependent, influencing chemokines and cytokines in opposite ways in different cells. | [58] |
In vitro | Macrophages | 2.5 μM | Suppresses LPS-induced phosphorylation of FoxO3a. Blocks the LPS-induced PI3K-AKT pathway and affects FoxO3a phosphorylation. Inhibits Nox1 and MCP-1 expression. Could modulate the activations of important macrophage functions associated with atherosclerosis. | [61] |
In vitro | TPH1 cell line | 25 μM | Promotes apoA-1 and HDL-mediated efflux, downregulates oxLDL uptake, and diminishes foam cell formation. Regulates expression of the cholesterol metabolizing enzyme CYP27A1, and helps cholesterol elimination. | [62] |
In vitro | TPH1 cell line | 2.5 μM | Inhibits foam cells formation by regulating the expression of the inflammatory cytokine, MCP-1, and by activating the AMPK-Sirt1-PPAR signaling pathway. | [63] |
In vitro | Granulocytes Monocytes RAW 264.7 cell line |
5–100 μM | Inhibits oxidative burst and CD11b expression in granulocytes and monocytes. Inhibits the production of NO and PGE2, but did not reduce iNOS, TNFα, or IL-1β gene expression in LPS-stimulated RAW 264.7. Induces NRf2 nuclear translocation and reduced miR-146a expression in LPS treated RAW 264.7. |
[64] |
In vitro | Human rheumatoid arthritis synovial fibroblasts | 20 μM | Suppresses the bradykinin-induced COX-2/PGE2. Inhibits the phosphorylation and acetylation of p65, c-Jun, and Fos and reduces the binding to the COX-2 promoter, thereby attenuated the COX-2 expression. Could be used for inflammatory arthritis therapy. | [65] |
In vivo In vitro |
C3H/He mice Splenocytes |
1.5 mg/Kg 1.25–2.5–5 μM |
Reprograms M-2 phenotype (TAM) countering the immunosuppressive and tumor progressive influences of TAM. | [83] |
In vitro | M2 polarization of human monocyte derived macrophages | 20 μM | Decreases STAT3. It inhibits F4/80 positive expressing cells and M2 polarization in the tumors. | [86] |
In vivo | C3H/He mice | 0.5, 1 and 1.5 mg/kg | Reduces Tregs (CD4 + CD25 + Foxp3 + cells) and the production of TGF-β. Increases IFN-γ-expressing CD8 + T cells. Upregulates IFN-γ production and enhances the cytotoxicity of splenocytes against FM3A tumor cells. | [97] |
In vitro In vivo |
T cell C57/BL6 and DBA1 mice |
0.5 μM or 25 μM 25 mg/kg |
Upregulates Sirt1 expression. Decreases c-Jun acetylation and its translocation. Reduces the incidence and severity of collagen-induced arthritis in mice. |
[100] |
In vivo | Wistar rats | 100 mg kg-1 ml | Downregulates PKC9 level in T lymphocytes | [101] |
In vivo | C57BL/6 mice | HFD supplemented with 0.06% resveratrol |
Activates the PI3K and Sirt1 signaling transduction. Activates the Nrf2-regulated adaptive response. Increases the CD3+CD4+/CD3+CD8+ subsets percentages and the Tregs. Maintains glucose homeostasis alleviating inflammation. |
[104] |
In vitro | PBMCs | 0.625–2.5–5–10 μM | Modulates the functional activities of both T and NK effector cells, with stimulation at low concentrations and suppression at high concentrations. Affects cytokine-production by activated CD41 and CD81 T cells. | [114] |
In vitro | KHYG-1, NKL, NK-92, and NK-YS cell lines | 3.125–6.25–12.5– 25–50 μM |
Suppresses STAT3 and inhibits JAK2 phosphorylation. Induces downregulation of the anti-apoptotic proteins MCL1 and survivin. Induces apoptotic and antiproliferative activities of L-asparaginase against KHYG-1, NKL and NK-92 cells. | [115] |
In vitro | Human NKs Jurkat cell line |
0.5−50 μM | At high concentration promotes apoptosis of NK cells and of Jurkat cells. At low concentration increases the NK cells cytotoxicity via up-regulating the expression of NKG2D and IFN-γ. |
[116] |
In vitro | KG-1a cells PBMCs |
25–100 μM | Inhibits KG-1a cell growth but has the least growth-inhibition effect PBMCs. Makes KG-1a cells susceptible to CIKs-mediated cytolysis correlated with an increase in cell-surface expression of NKG2D ligands and DR4, coupled with a downregulation of cell-surface expression of DcR1. |
[13] |
In vitro | DU145, and PC3 cells | 5–30 μM | Induces apoptosis in prostate cancer cells. Downregulates Bcl-2, Bcl-XL, and surviving. Upregulates Bax, Bak, PUMA, Noxa, and Bim, TRAIL-R1/DR4 and TRAIL-R2/DR5 expression. Activates caspase-3 and caspase-9 and induces apoptosis. |
[119] |
In vitro | cell lines LU120 cell line | 25–100 μM | Decreases STAT3 and NF-κB activation. Suppresses expression of cFLIP and Bcl-xL proteins and increases sensitivity to exogenous TRAIL in DR5-positive melanomas. In combination with TRAIL it could have a significant efficacy in the treatment of human melanomas. |
[121] |
In vitro | HL60 T47D cell line |
32 μM | Induces cell death mediated by intracellular caspases Dose-dependent increase in proteolytic cleavage of caspase substrate PARP. Enhances CD95L expression on both HL60 cells T47D breast carcinoma cells. |
[123] |
In vivo In vitro |
BALB/c or C57BL/6 mice tBregs |
20 or 50 mg/mouse 12.5 mM |
Inhibits lung metastasis in mice. Inactivates Stat3, preventing the generation and function of tBregs, including expression of TGF-β. It reduces antitumor effector immune responses by disabling tBreg-induced conversion of Foxp3+ Tregs. Could control cancer escape-promoting tBregs/Tregs without nonspecific inactivation of effector immune cells. | [131] |
In vivo | MRL/lpr mice BJAB B cells |
20 mg kg−1 per day | Increases the expression of FcγRIIB in B cells. Decreases serum autoantibody titers in MRL/lpr mice. The upregulation of FcγRIIB causes an increase of Sirt1 protein and deacetylation of p65 NF-κB. Reduces plasma cells in MRL/lpr mice, leading to improvement of nephritis and prolonged survival. |
[132] |
In vivo | BALB/c mice | 20 mg/kg | Reduces proteinuria, immunoglobulin deposition in kidney, and in serum in pristane-induced lupus mice. Inhibits CD69 and CD71 expression on CD4+ T cells and CD4+ T cell proliferation. Induces CD4+ T cell apoptosis, and decreased CD4 IFNc+ Th1 cells and the ratio of Th1/Th2 cells in vitro. Inhibits antibody production and proliferation of B cells in vitro. |
[134] |
Abbreviations: AKT, protein kinase B; AMPK, AMP-activated protein kinase; AP-1, activator protein 1; apoA-1, apolipoprotein (Apo) A-I; Bax, Bcl-2-associated X protein; Bak, Bcl-2 homologous antagonist killer; Bcl-2, B-cell lymphoma; Bcl-xL, B-cell lymphoma-extra-large; Bim, Bcl-2-like 11; Cbfa-1, core-binding factor a1; CCL20, chemokine (C-C motif) ligand 20; CCR 6 chemokine (C-C motif) receptor 6; cFLIP, cellular FLICE-inhibitory protein; CIKs, cytokine-induced killer cells; COX-2, cyclooxygenase-2; CTLs, cytotoxic T lymphocytes; CYP27A1, cytochrome P450 27-hydroxylase; DR, death receptor; DcR1, decoy receptor 1; ERK1/2, extracellular signal–regulated kinases; FcγRIIB, Fc gamma receptor IIb; FoxO3a, forkhead box O3A; Foxp3, forkhead box P3; HDL, high-density lipoprotein cholesterol; HFD, high-fat diet; IFN-γ interferon-gamma; IL, interleukin; iNOS, inducible nitric oxide synthase; IRAK1, interleukin-1 receptor-associate kinase 1; JAK2, janus activated kinase; JNK, c-Jun N-terminal kinase; LAKs, lymphokine activated killer cells; LPS, lipopolysaccharide; MAPK, mitogen-activated protein kinase; M-CSF, macrophage colony stimulating factor; MCP, monocyte chemoattractant protein; MEK, mitogen-activated protein kinase; Myd88, myeloid differentiation factor 88; NK, natural killer; NLRP3, NOD-like receptor family pyrin domain containing 3; NKG2D, natural killer group 2 member D; Nrf2, nuclear factor (erythroid-derived 2)-related factor-2; NF-κB, nuclear factor-kappa B; NOD, nucleotide oligomerization domain; PARP poly (ADP-ribose) polymerase; PBMCs, peripheral blood mononuclear cells; PGE2, prostaglandin E2; PI3K, phosphoinositide 3-kinase; PKCϑ, protein kinase c-delta; PMA, phorbol 12-myristate13-acetate; PPAR, peroxisome proliferator-activated receptors; PUMA, p53 upregulated modulator of apoptosis; RANKL, receptor activator of nuclear factor kB ligand; RIP, receptor interacting protein; ROI, reactive oxygen intermediate, Sirt1, Sirtuin-1; STAT, signal transducer and activator; TAMs, tumor associated macrophages; TANK, TRAF family member-associated NF-κB activator; tBregs, TGFβ-expressing regulatory B cells; Tregs, regulatory T cells; TRAF6, tumor necrosis factor receptor-associated factor 6; TRAIL, tumor necrosis factor-related apoptosis-inducing ligand; TNF-related apoptosis inducing ligand; TRIF, TIR-domain-containing adapter-inducing interferon; TLR, toll-like receptor; TGF-β, transforming growth factor beta; TNF-α, tumor necrosis alpha.