Skip to main content
. 2021 Apr 12;12:633447. doi: 10.3389/fphar.2021.633447

TABLE 1.

Bioactivities of the main components extracted from C. officinalis.

Components Bioactivities Diseases/Tissues Targets (In vitro and In vivo) Effects References
Morroniside Anti- inflammation Osteoarthritis In vitro: IL-1β treated primary mouse chondrocytes; in vivo: destabilization of the medial meniscus-treated C57BL/6J mice. Inhibit NF-κB signaling and proinflammatory NLRP3 expression; down-regulate the expressions of MMP-13 and caspase-1; promote collagen type II and cartilage matrix synthesis. (Yu et al., 2021)
Colitis In vitro: LPS treated colorectal cancer cell; in vivo: DSS induced acute colitis mouse model. Increase the expression of tight junction proteins; decrease the expressions of pro-inflammatory cytokines; suppress the phosphorylation of STAT3 and NF-κB. (Yuan et al., 2020)
Anti-oxidation Neuropathic pain In vitro: H2O2 treated microglial N9 cells, HEK293 cells and HEK293T cells; in vivo: neuropathic pain rat model (L5, L6 spinal nerves ligation). Active GLP-1 receptors. (Xu et al., 2017)
Anti-apoptosis Diabetic cardiomyopathy In vitro: primary cultured rat cardiomyocytes. Down-regulate the expressions of ROS, caspase-3 and bax; up-regulate the expression of bcl-2. (Pi et al., 2017)
Anti-diabetes Diabetic osteoporosis In vitro: primary cultured rat BMSCs; in vivo: DM rats model (single intraperitoneal injection of 60 mg/kg STZ, SD rats). Promote osteogenic differentiation of BMSCs; up-regulate the activation and expression of Glo1; down-regulate AGEs formation and RAGE expression. (Sun et al., 2020)
Renal tissue In vitro: AGEs treated mesangial cells. Inhibit the secretion of ECM major components (LN, FN, and Col-IV) induced by AGEs; inhibit the expressions of RAGE, p38MAPK, NF-κB, and TGF-β induced by AGEs. (Lv et al., 2016b)
Liver In vitro: serum-free DMEM with 50mmol/L glucose treated HepG2 cells; in vivo: DM mice model (single intraperitoneal injection of 60 mg/kg STZ). Promote glucose uptake; decrease FBG levels in DM mice. (He et al., 2016)
Liver In vivo: db/db mice. Decrease serum glucose; decrease ROS and lipid peroxidation in liver tissue; down-regulate the expressions of NOX4, P22phox, SREBP-1, SREBP-2, Nrf2, HO-1, NF-κB, COX-2, iNOS, MCP-1, bax and Cytochrome C in liver tissue; up-regulate GSH/GSSG ratio. (Park et al., 2011a; Park et al., 2009)
Renal tissue In vivo: db/db mice; STZ (50 mg/kg)-induced DM rats. Decrease serum TC; decrease renal lipid peroxidation and ROS levels; down-regulate renal SREBP-1, SREBP-2, NF-κB, COX-2 and iNOS expressions; up-regulate GSH/GSSG ratio. (Park et al., 2010b; Yokozawa et al., 2010)
Renal tissue In vivo: STZ (50 mg/kg)-induced DM rats. Decrease serum glucose, BUN and urinary protein levels; elevate serum albumin and total protein; reduce glycosylated protein and lipid peroxidation; down-regulate RAGE, HO-1. (Yokozawa et al., 2008)
Renal tissue In vitro: AGEs treated rat mesangial cells. Inhibit AGE-induced mesangial cells proliferation and cell cycle; down-regulate ROS and MDA; up-regulate SOD and GSH. (Xu et al., 2006)
1,6-α-glucans Anti-atherosclerosis Atherosclerosis In vitro: oxidized-LDL treated RAW264.7 macrophages; in vivo: ApoE−/− mice fed with HFD. Reduce ox-LDL induced cholesterol levels and inhibit the foam cell formation in RAW264.7 cells; reduce aortic atherosclerotic lesion area in ApoE−/− mice fed with HFD; decrease serum LDL, TC levels, MDA activity, and CD36, SR-A1 expressions; increase SOD activity. (Zhang et al., 2020)
Loganin Anti- inflammation Colitis In vitro: LPS treated colorectal cancer cell; in vivo: DSS-induced acute colitis mouse model. Increase the expression of tight junction proteins; decrease the expressions of pro-inflammatory cytokines; suppress the phosphorylation of STAT3 and NF-κB. (Yuan et al., 2020)
Ulcerative colitis In vivo: DSS-induced ulcerative colitis mouse model. Down-regulate the expressions of IL-6, TNF-α, IL-1β, MCP-1, CXCL10, COX-2 and Sirt1; inhibit the acetylation of NF-κB; reduce macrophage M1 polarization. (Liu et al., 2020b)
Anti-diabetes Renal tissue In vitro: AGEs treated podocytes; in vivo: KK-Ay mice. Decrease FBG, Scr and BUN levels; increase serum insulin level; alleviate podocyte loss and apoptosis; activate RAGE/p38 MAPK/NF-κB and RAGE/NOX4/NF-κB pathways in podocytes. (Chen et al., 2020)
Liver In vitro: serum-free DMEM with 50mmol/L glucose treated HepG2 cells; in vivo: DM mice model (single intraperitoneal injection of 60 mg/kg STZ). Promote glucose uptake; decrease FBG levels in DM mice; down-regulate MDA level and Aldose reductase activity. (He et al., 2016)
DM In vitro: kinetic and molecular docking studies. Inhibit aldose reductase activity. (Lee et al., 2015)
Renal tissue In vitro: high glucose (30mmol/L) treated HBZY-1 mesangial cells. Inhibit the expression of FN and IL-6. (Ma et al., 2014)
Renal tissue In vitro: high glucose (27.5mmolL) treated HK-2 cells; in vivo: DM rats model (single intraperitoneal injection of 45 mg/kg STZ). Inhibit CTGF expression in vitro and in vivo. (Jiang et al., 2012)
Liver In vivo: db/db mice. Decrease serum glucose and elevate serum leptin; inhibit ROS and lipid peroxidation in the serum and liver; down-regulate the expressions of NOX4 and p22phox; down-regulate the expressions of NF-κB, COX-2, iNOS and MCP-1. (Park et al., 2011b)
Hepatic and renal tissue In vivo: db/db mice. Decrease serum glucose, TG, LDL/VLDL and increase serum HDL; elevate GSH/GSSG ratio; up-regulate PPARα expression; down-regulate SREBP-1, SREBP-2 expressions; inhibit AGEs formation and RAGE expression in hepatic and renal tissues. (Yamabe et al., 2010)
Renal tissue In vitro: AGEs treated rat mesangial cells. Inhibit AGEs-induced mesangial cells proliferation and cell cycle; down-regulate ROS and MDA; up-regulate SOD and GSH. (Xu et al., 2006)
Anti-cholinesterase Alzheimer's disease In vivo: scopolamine (1 mg/kg, s.c.)-induced amnesic mice. Mitigate scopolamine-induced memory deficits in passive avoidance test and Morris water maze test; inhibit acetylcholinesterase activity in the mouse hippocampus. (Lee et al., 2009)
Ursolic acid Anti-diabetes Liver In vitro: serum-free DMEM with 50mmol/L glucose treated HepG2 cells; in vivo: DM mice model(single intraperitoneal injection of 60 mg/kg STZ). Inhibit α-glucosidase activity; promote glucose uptake; decrease FBG level in DM mice; down-regulate MDA and Aldose reductase activity, up-regulate SOD activity. (He et al., 2016)
Anti-inflammation Colitis In vitro: LPS-treated peritoneal macrophages; in vivo: TNBS-induced colitis mice. Inhibit phosphorylation of IRAK1, TAK1, IKKβ, and IkappaBalpha; inhibit the activation of NF-κB and MAPKs; inhibit IL-1β, IL-6, TNF-α, COX-2 and iNOS expression as well as PGE2 and NO levels; inhibit LPS bind to TLR4 on immune cells. (Jang et al., 2014)
Inhibit mucin secretion Airway diseases In vitro: EGF (25 ng/mL) or PMA (10 ng/mL) treated NCI-H292 cells (the human pulmonary mucoepidermoid carcinoma cell line). Inhibit MUC5AC mucin gene expression and mucin protein production. (Cho et al., 2011)
Antioxidation Inner ear diseases In vitro: H2O2-treated HEI-OC1 auditory cells. Reduce lipid peroxidation; up-regulate the activities of CAT and GPX. (Yu et al., 2009)
Oleanolic Acid Anti-proliferation Benign prostatic hyperplasia In vitro: Human BPH-1 cells; in vivo: BPH rat model (male rats injected with testosterone propionate). Decrease prostate weight and prostate epithelial thickness; reduce serum DHT and 5α-reductase mRNA levels; down-regulate protein expressions of bcl-2, bcl-xL and PCNA; down-regulate the cell cycle markers CdK4-cyclin D1 and CdK2-cyclin E. (Cheon et al., 2020)
Inhibit mucin secretion Airway diseases In vitro: EGF (25 ng/mL) or PMA (10 ng/mL) treated NCI-H292 cells. Inhibit MUC5AC mucin gene expression and mucin protein production. (Cho et al., 2011)
Anti-diabetes DM In vivo: intraperitoneal inject into the fasting Wistar rats. Enhance the release of ACh from nerve terminals, stimulate muscarinic M(3) receptors in the pancreatic β cells and augment the insulin release. (Hsu et al., 2006)
Cornuside Anti-cholinesterase Alzheimer's disease In vitro experiments. Inhibit ChEs and BACE1 by interacting with both the catalytic active sites and the peripheral anionic sites. (Bhakta et al., 2017)
Vasorelaxant activity In vitro: isometric vascular tone of phenylephrine-contracted thoracic aortae; cGMP production in HUVECs. Dilates vascular smooth muscle via endothelium-dependent NO/cGMP signaling. (Kang et al., 2007)
Polymeric proanthocyanidins Anti-cholinesterase Alzheimer's disease In vitro experiments. Inhibit ChEs and BACE1 by interacting with both the catalytic active sites and the peripheral anionic sites. (Bhakta et al., 2017)
Anti-diabetes DM In vivo: male Wistar rats with normal blood glucose. Inhibit α-glucosidase activity; improve the oral sucrose tolerance, and inhibit the rise in the plasma glucose levels in normal rats. (Park et al., 2011c)
1,2,3-tri-O-galloyl-beta-D-glucose Anti-cholinesterase Alzheimer's disease In vitro experiments. Inhibit ChEs and BACE1 by interacting with both the catalytic active sites and the peripheral anionic sites. (Bhakta et al., 2017)
Anti-diabetes DM In vitro experiments. Inhibit the formation of AGEs and RLAR, inhibit AGE-BSA cross-linking. Lee et al. (2011)
1,2,3,6-tetra-O-galloyl-beta-D-glucose Anti-cholinesterase Alzheimer's disease In vitro experiments. Inhibit ChEs and BACE1 by interacting with both the catalytic active sites and the peripheral anionic sites. (Bhakta et al., 2017)
Anti-diabetes DM In vitro experiments. Inhibit the formation of AGEs and RLAR, inhibit AGE-BSA cross-linking; prevent the opacity of lenses. (Lee et al., 2011)
1,2,6-tri-O-galloyl-beta-D-glucose Anti-diabetes Diabetes mellitus(DM) In vitro experiments. Inhibit the formation of AGEs and RLAR, inhibit AGE-BSA cross-linking; prevent the opacity of lenses. (Lee et al., 2011)
1,2,4,6-tetra-O-galloyl-bta-D-glucose Anti-diabetes Diabetes mellitus(DM) In vitro experiments. Inhibit the formation of AGEs and RLAR, inhibit AGE-BSA cross-linking; prevent the opacity of lenses. (Lee et al., 2011)
1,2,3,4,6-penta-O-galloyl-beta-Dglucose Anti-diabetes Diabetes mellitus(DM) In vitro experiments. Inhibit the formation of AGEs and RLAR, inhibit AGE-BSA cross-linking; prevent the opacity of lenses. (Lee et al., 2011)
Tellimagrandin I Anti-cholinesterase Alzheimer's disease In vitro experiments. Inhibit ChEs and BACE1 by interacting with both the catalytic active sites and the peripheral anionic sites. (Bhakta et al., 2017)
Tellimagrandin II Anti-cholinesterase Alzheimer's disease In vitro experiments. Inhibit ChEs and BACE1 by interacting with both the catalytic active sites and the peripheral anionic sites. (Bhakta et al., 2017)
Anti-diabetes DM In vitro experiments. Inhibit the formation of AGEs and RLAR, inhibit AGE-BSA cross-linking; prevent the opacity of lenses. (Lee et al., 2011)
Isoterchebinc Anti-cholinesterase Alzheimer's disease In vitro experiments. inhibit ChEs and BACE1 by interacting with both the catalytic active sites and the peripheral anionic sites. (Bhakta et al., 2017)
7-O-Galloyl-D-sedoheptulose Anti-diabetes Hepatic tissue In vivo: db/db mice. Decrease serum glucose, leptin, insulin, TNF-α, IL-6, resistin, ALT, AST; reduce hepatic AGEs, RAGE and ROS; reduce hepatic p-ERK1/2, p-cJNK, NF-κB, AP-1, MCP-1, ICAM-1, TNF-α, and IL-6. (Park et al., 2015b)
DM In vitro: kinetic and molecular docking studies. inhibit aldose reductase activity (Lee et al., 2015)
Adipose tissue In vivo: db/db mice. Decrease serum glucose, leptin, insulin, C-peptide, resistin, TNF-α, IL-6, TG, TC, HDL, VLDL/LDL, ROS and TBARS; up-regulate serum adiponectin; decrease adipose tissue lipid, ROS, TBARS contents and SREBP-1, NF-κB, COX-2, iNOS, MCP-1, ICAM-1, p-cJNK, AP-1, TGF-β1, bax, cytochrome c , caspase-3 expressions; elevate PPARα, PPARγ, and β-cell lymphoma 2 in the adipose tissue. (Park et al., 2013)
Renal tissue In vivo: db/db mice. Decrease serum insulin, Cr and BUN levels; down-regulate serum TNF-α, IL-6 and ROS; down-regulate renal ROS, TBARS, NF-κBp65, NOX4, p22phox, COX-2, iNOS, bax and Cytochrome C; up-regulate GSH/GSSG ratio. (Park et al., 2012)
Hepatic and renal tissue In vivo: db/db mice. Down-regulated the expression of SREBP-1; inhibit AGEs formation and RAGE expression in hepatic and renal tissues. (Park et al., 2010a)
Malic acid Anti-platelet In vitro: platelet viability assay and platelet adhesion assay. Inhibit platelet aggregation and prevent platelet adhesion. (Zhang et al., 2013)
Succinic acid Anti-platelet In vitro: platelet viability assay and platelet adhesion assay. Inhibit platelet aggregation and prevent platelet adhesion. (Zhang et al., 2013)
Citric acid Anti-platelet In vitro: platelet viability assay and platelet adhesion assay. Inhibit platelet aggregation and prevent platelet adhesion. (Zhang et al., 2013)
7-O-butylmorroniside Neuroprotection Neurodegenerative diseases In vitro: glutamate-treated HT22 hippocampal cells. Elevate the cell viability (MTT) of glutamate-treated HT22 hippocampal cells. (Jeong et al., 2012)
5-hydroxymethylfurfuralc (processed C. officinalis) Anti-diabetes Vascular system In vitro: high glucos treated HUVECs. Inhibit HUVECs apoptosis induced by high glucose; reduce ROS and superoxide; down-regulate the expressions of IL-8, JNK1 and JNK2/3; up-regulated the expression of p-Akt. (Cao et al., 2013)
Antioxidation Acute liver injury In vitro: H2O2-treated human vein epidermal cell ; in vivo: CCL4-induced acute liver injury mice. Protect human vein epidermal cell against H2O2; decrease ALT, AST in acute liver injury in mice. (Ding et al., 2008)

Notes: Abbreviations of table 1.

NLRP3: NOD-like receptors three; MMP: matrix metalloprotein; STAT: signal transducer and activator of transcription; GLP: glucagon-like peptide-1; BMSCs: bone marrow mesenchymal stem cells; Glo1: glyoxalase-1; FBG: fasting blood glucose; SREBP: sterol-regulatory element binding proteins; GSSG: oxidized glutathione; HFD: high-fat diet; SR-A1: type A1 scavenger receptor; LPS: lipopolysaccharide; DSS: dextran sodium sulfate; TNF-α: tumor necrosis factor-α; CXCL10: CXC chemokine ligand-10; CTGF: connective tissue growth factor; TNBS: 2,4,6-trinitrobenzenesulfonic acid; IRAK: interleukin one receptor-associated kinase; IKKβ: inhibitor of nuclear factor kappaB kinase subunit β; PGE2: prostaglandin E2; TLR4: toll-like receptor four; EGF: epidermal growth factor; PMA: phorbol 12-myristate 13-acetate; GPX: glutathione peroxidase; DHT: dihydrotestosterone; PCNA: proliferating cell nuclear antigen; HUVECs: human umbilical vein endothelial cells; RLAR: rat lens aldose reductase; AP-1: activator protein-1; TBARS: thiobarbituric acid-reactive substance.