Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
J Med Food ; 25(5): 503-512, 2022 May.
Article in English | MEDLINE | ID: mdl-35561274

ABSTRACT

Salicornia herbacea L. (Chenopodiaceae), an edible salt marsh plant with anti-inflammatory effects, was examined in macrophages and trophoblasts whether it modulates NLRP3 inflammasome activity. Pretreatment and delayed treatment of S. herbacea extract (SHE) in bone marrow-derived macrophages (BMDMs) reduced the activity of NLRP3 inflammasome induced by lipopolysaccharide (LPS) and adenosine triphosphate stimulation and downregulated interleukin (IL)-1ß production. SHE also inhibited pyroptotic cell death, the adaptor molecule apoptosis-associated speck-like protein containing a CARD (ASC), oligomerization, and speck by NLRP3 inflammasome activity in BMDM. Similarly, SHE decreased the mRNA expression of NLRP3, ASC, IL-1ß, and IL-6 in the LPS-stimulated human trophoblast cell line, Swan 71 cells. In addition, SHE inhibited the production of IL-6 and IL-1ß and decreased the expression of cyclooxygenase-2 and prostaglandin E2 in stimulated Swan 71 cells. Finally, 3,5-dicaffeoylquinic acid (3,5-DCQA), one of the components of S. herbacea, inhibited IL-1ß produced by NLRP3 inflammasome activity. In conclusion, SHE downregulated the activity of the NLRP3 inflammasome in macrophages and trophoblasts.


Subject(s)
Chenopodiaceae , Inflammasomes , Caspase 1/metabolism , Caspase 1/pharmacology , Humans , Inflammation/metabolism , Interleukin-1beta/metabolism , Interleukin-6/genetics , Interleukin-6/metabolism , Lipopolysaccharides/pharmacology , Macrophages , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Trophoblasts/metabolism
2.
Am J Reprod Immunol ; 82(4): e13167, 2019 10.
Article in English | MEDLINE | ID: mdl-31295378

ABSTRACT

PROBLEM: Dysregulation of extravillous trophoblast (EVT) invasion leads to pregnancy complications, such as pre-eclampsia, fetal growth restriction, and placenta accreta. The aim of this study was to explore the role of SIRT1 in EVT invasion and its underlying mechanism. METHOD OF STUDY: SIRT1-specific siRNA was transfected into Swan 71 cells, an immortalized first trimester trophoblast cell line. The Boyden chamber invasion assay, the scratch wound healing assay, and cell proliferation assay were performed. The expression levels of epithelial-to-mesenchymal transition (EMT) markers, matrix metalloproteinase-2 (MMP-2), MMP-9, p-Akt, Akt, p-p38MAPK, p38MAPK, p-ERK, ERK, p-JNK, JNK, Fas, and Fas ligand (FasL) were examined by western blot. Tube formation assay was conducted by using Matrigel. RESULTS: SIRT1 knockdown by siRNA significantly enhanced invasion and migration as well as the expression of MMP-2, MMP-9, and EMT markers in Swan 71 cells, but reduced proliferation. The effects of SIRT1 knockdown on invasion, migration, proliferation, and endothelial-like tube formation in Swan 71 cells were reversely regulated by blockade of Akt and p38MAPK signaling. In addition, SIRT1 knockdown markedly promoted colocalization of Swan 71 cells to human umbilical vein endothelial cell (HUVEC) networks and induced reduction in Fas and enhancement of FasL. Conditioned media of SIRT1 knockdown-Swan 71 cells caused reduction in cell proliferation and augmentation of cytotoxicity along with increased Fas expression in HUVECs. CONCLUSION: Our results suggest that SIRT1 may be associated with placental development by controlling EVT invasion and spiral artery remodeling via modulation of EMT, MMP-2, MMP-9, Akt/p38MAPK signaling, and Fas/FasL.


Subject(s)
Neovascularization, Physiologic , Sirtuin 1/physiology , Trophoblasts/physiology , Cell Line , Cell Movement , Cell Proliferation , Chorionic Villi , Epithelial-Mesenchymal Transition , Fas Ligand Protein/physiology , Female , Human Umbilical Vein Endothelial Cells/physiology , Humans , Matrix Metalloproteinase 2/physiology , Matrix Metalloproteinase 9/physiology , Pregnancy , Proto-Oncogene Proteins c-akt/physiology , RNA, Small Interfering , Sirtuin 1/genetics , fas Receptor/physiology , p38 Mitogen-Activated Protein Kinases/metabolism
3.
Am J Reprod Immunol ; 79(2)2018 02.
Article in English | MEDLINE | ID: mdl-29205625

ABSTRACT

PROBLEM: Vitamin D deficiency increases the risk of developing pregnancy-related complications, including preeclampsia and small-for-gestational-age infants. Vitamin D was demonstrated to promote the invasiveness of human extravillous trophoblasts (EVTs). However, whether vitamin D induces the epithelial-mesenchymal transition (EMT) of EVTs remains unclear. Therefore, we investigated whether vitamin D promotes EMT and the related signaling pathways. METHOD OF STUDY: In this study, we performed EMT experiments using JAR cells based on the expression of the mesenchymal markers and vitamin D receptor. JAR cells were treated with calcitriol, the active form of vitamin D. Western blotting was performed to evaluate EMT markers and key molecules of signaling pathways. Invasion assays were conducted. Expression and secretion of MMPs were analyzed by real-time PCR and zymography. RESULTS: Calcitriol significantly enhanced EMT and the invasive capability of JAR cells, along with increased expression and secretion of MMP-2 and MMP-9. Moreover, ERK signaling pathway was activated by calcitriol. The effects of calcitriol were neutralized by ERK signaling blocker. CONCLUSION: Calcitriol facilitated EMT induction and expression of MMPs via ERK signaling pathway, which promoted the invasive capability of EVTs. Further studies are warranted to elucidate the potential application of vitamin D in the prevention of pregnancy complications.


Subject(s)
Calcitriol/pharmacology , Epithelial-Mesenchymal Transition , Placenta/pathology , Pregnancy Complications/prevention & control , Pregnancy , Trophoblasts/physiology , Vitamin D/metabolism , Cell Line , Cell Movement , Female , Humans , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9 , Signal Transduction
4.
Int J Mol Sci ; 18(10)2017 Sep 25.
Article in English | MEDLINE | ID: mdl-28946662

ABSTRACT

Vascular calcification plays a role in the pathogenesis of atherosclerosis, diabetes, and chronic kidney disease; however, the role of apurinic/apyrimidinic endonuclease 1/redox factor-1 (APE1/Ref-1) in inorganic phosphate (Pi)-induced vascular smooth muscle cell (VSMC) calcification remains unknown. In this study, we investigated the possible role of APE1/Ref-1 in Pi-induced VSMC calcification. We observed that Pi decreased endogenous APE1/Ref-1 expression and promoter activity in VSMCs, and that adenoviral overexpression of APE1/Ref-1 inhibited Pi-induced calcification in VSMCs and in an ex vivo organ culture of a rat aorta. However, a redox mutant of APE1/Ref-1(C65A/C93A) did not reduce Pi-induced calcification in VSMCs, suggesting APE1/Ref-1-mediated redox function against vascular calcification. Additionally, APE1/Ref-1 overexpression inhibited Pi-induced intracellular and mitochondrial reactive oxygen species production, and APE1/Ref-1 overexpression resulted in decreased Pi-induced lactate dehydrogenase activity, pro-apoptotic Bax levels, and increased anti-apoptotic Bcl-2 protein levels. Furthermore, APE1/Ref-1 inhibited Pi-induced osteoblastic differentiation associated with alkaline phosphatase activity and inhibited Pi-exposure-induced loss of the smooth muscle phenotype. Our findings provided valuable insights into the redox function of APE1/Ref-1 in preventing Pi-induced VSMC calcification by inhibiting oxidative stress and osteoblastic differentiation, resulting in prevention of altered osteoblastic phenotypes in VSMCs.


Subject(s)
Calcification, Physiologic/genetics , DNA-(Apurinic or Apyrimidinic Site) Lyase/genetics , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/metabolism , Osteoblasts/metabolism , Phenotype , Animals , Apoptosis/drug effects , Apoptosis/genetics , Calcification, Physiologic/drug effects , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cells, Cultured , DNA-(Apurinic or Apyrimidinic Site) Lyase/metabolism , Gene Expression Regulation/drug effects , Gene Silencing , Male , Mitochondria/metabolism , Muscle, Smooth, Vascular/pathology , Mutation , Myocytes, Smooth Muscle/drug effects , Osteoblasts/drug effects , Oxidation-Reduction , Phosphates/metabolism , Phosphates/pharmacology , RNA Interference , Rats , Reactive Oxygen Species/metabolism , Vascular Calcification/genetics , Vascular Calcification/metabolism , Vascular Calcification/pathology
5.
Integr Med Res ; 5(2): 131-139, 2016 Jun.
Article in English | MEDLINE | ID: mdl-28462108

ABSTRACT

BACKGROUND: Ulmus davidiana var. japonica Rehder (UD) has long been used in traditional folk medicine in Asia. This study is designed to investigate the antiadhesive activity of the ethanol extract of UD (UDE) and its underlying mechanisms in cultured endothelial cells. METHODS: The dried root bark of UD was extracted with 80% (v/v) ethanol. The antiadhesive activity of the UDE was investigated in cultured human umbilical vein endothelial cells and human embryonic kidney epithelial 293T (HEK 293T) cells stably transfected with pGL3-vascular cell adhesion molecule (VCAM)-1-luc. Monocyte adhesion in endothelial cells was induced by tumor necrosis factor-alpha (TNF-α), and the protective effects of UDE on monocyte-endothelial cell adhesion, VCAM-1 expression, reactive oxygen species production, and nuclear factor-κB activity were determined. RESULTS: Exposure to UDE at a concentration of 3-30 µg/mL for 24 hours produced no detectable cytotoxicity in human umbilical vein endothelial cells, but it significantly inhibited TNF-α-induced monocyte adhesion and VCAM-1 expression. TNF-α treatment of HEK 293T/VCAM-1-luc cells resulted in increased luciferase activity of the VCAM-1 promoter, which was inhibited by treatment with UDE. Additionally, TNF-α-induced reactive oxygen species generation, nuclear translocation of nuclear factor-κB, and IκBα degradation in human umbilical vein endothelial cells were effectively reduced by treatment with 30 µg/mL of UDE. CONCLUSION: Our results indicated that UDE treatment inhibited TNF-α-induced monocyte adhesion in endothelial cells, suggesting that UD may reduce vascular endothelial inflammation.

6.
BMC Complement Altern Med ; 15: 125, 2015 Apr 22.
Article in English | MEDLINE | ID: mdl-25898017

ABSTRACT

BACKGROUND: Fructus mume (F. mume) has been used as a traditional medicine for many years in Asian countries. The present study was designed to determine the effect of a 70% ethanol extract of F. mume on white matter and hippocampal damage induced by chronic cerebral hypoperfusion. METHODS: Permanent bilateral common carotid artery occlusion (BCCAo) was performed on male Wistar rats to induce chronic cerebral hypoperfusion. Daily oral administration of F. mume (200 mg/kg) was initiated 21 days after BCCAo and continued for 42 days. The experimental groups in this study were divided into three groups: a sham-operated group, a BCCAo group, and a BCCAo group that was administered with the F. mume extract. The activation of glial cells, including microglia and astrocytes, and the levels of myelin basic protein (MBP), inflammatory mediators, Toll-like receptor 4 (TLR4), myeloid differentiation factor 88 (MyD88), and p38 mitogen-activated protein kinase (MAPK) phosphorylation were measured in brains from rats subjected to chronic BCCAo. RESULTS: Our results revealed that F. mume alleviates the reduction in MBP expression caused by chronic BCCAo in the white matter and the hippocampus and significantly attenuates microglial and astrocytic activation induced by chronic BCCAo in the optic tract of white matter. In addition, F. mume treatment reduced the increased expression of cyclooxygenase-2 (COX-2), interleukin-1ß (IL-1ß) and interleukin-6 (IL-6), as well as the activation of TLR4/MyD88 and p38 MAPK signaling, in the hippocampus of rats subjected to chronic BCCAo. CONCLUSION: Taken together, our findings demonstrate that brain injury induced by chronic BCCAo is ameliorated by the anti-inflammatory effects of F. mume via inhibition of MBP degradation, microglial and astrocytic activation, increased inflammatory mediator expression, and activated intracellular signalings, including TLR4 and p38 MAPK, implying that F. mume is potentially an effective therapeutics for the treatment of vascular dementia.


Subject(s)
Brain Ischemia/drug therapy , Brain/drug effects , Dementia, Vascular/metabolism , Inflammation/prevention & control , Prunus , Toll-Like Receptor 4/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Brain/cytology , Brain/metabolism , Brain Ischemia/metabolism , Cytokines/metabolism , Dementia, Vascular/prevention & control , Down-Regulation , Drugs, Chinese Herbal/pharmacology , Drugs, Chinese Herbal/therapeutic use , Hippocampus/drug effects , Hippocampus/metabolism , Inflammation/metabolism , Inflammation Mediators/metabolism , Male , Myelin Basic Protein/metabolism , Phosphorylation/drug effects , Phytotherapy , Rats, Wistar , Signal Transduction/drug effects , White Matter/drug effects , White Matter/metabolism
7.
Diabetes ; 64(1): 117-27, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25114293

ABSTRACT

Triggering receptor expressed on myeloid cells 2 (TREM2) is known to be involved in the anti-inflammatory response and osteoclast development. However, the role of TREM2 in adipogenesis or obesity has not yet been defined. The effect of TREM2 on adipogenesis and obesity was investigated in TREM2 transgenic (TG) mice on a high-fat diet (HFD). To block TREM2 signaling, a neutralizing fusion protein specific for TREM2 (TREM2-Ig) was used. TG mice were much more obese than wild-type mice after feeding with an HFD, independent of the quantity of food intake. These HFD-fed TG mice manifested adipocyte hypertrophy, glucose and insulin resistance, and hepatic steatosis. The expression of adipogenic regulator genes, such as peroxisome proliferator-activated receptor γ and CCAAT/enhancer-binding protein α, was markedly increased in HFD-fed TG mice. Additionally, HFD-fed TG mice exhibited decreased Wnt10b expression and increased GSK-3ß (glycogen synthase kinase-3ß)-mediated ß-catenin phosphorylation. In contrast, the blockade of TREM2 signaling using TREM2-Ig resulted in the inhibition of adipocyte differentiation in vitro and a reduction in body weight in vivo by downregulating the expression of adipogenic regulators. Our data demonstrate that TREM2 promotes adipogenesis and diet-induced obesity by upregulating adipogenic regulators in conjunction with inhibiting the Wnt10b/ß-catenin signaling pathway.


Subject(s)
Adipocytes/metabolism , Adipogenesis/physiology , Membrane Glycoproteins/metabolism , Myeloid Cells/metabolism , Obesity/metabolism , Receptors, Immunologic/metabolism , 3T3-L1 Cells , Adipocytes/cytology , Adipocytes/immunology , Animals , Calorimetry, Indirect , Cell Differentiation/physiology , Diet, High-Fat , Energy Metabolism/physiology , Fibroblasts/cytology , Fibroblasts/immunology , Fibroblasts/metabolism , Insulin Resistance/physiology , Male , Membrane Glycoproteins/genetics , Membrane Glycoproteins/immunology , Mice , Mice, Transgenic , Myeloid Cells/cytology , Myeloid Cells/immunology , Obesity/genetics , Obesity/immunology , Primary Cell Culture , Receptors, Immunologic/genetics , Receptors, Immunologic/immunology , Signal Transduction/physiology , Wnt Signaling Pathway/physiology
8.
BMC Complement Altern Med ; 13: 334, 2013 Nov 26.
Article in English | MEDLINE | ID: mdl-24274593

ABSTRACT

BACKGROUND: The cardiotonic pill (CP) is a herbal medicine composed of Salvia miltiorrhiza (SM), Panax notoginseng (PN), and Dryobalanops aromatica Gaertner (DAG) that is widely used to treat cardiovascular diseases. The present experiment was conducted to examine the effects of CP on white matter and hippocampal damage induced by chronic cerebral hypoperfusion. METHODS: Chronic cerebral hypoperfusion was induced in male Wistar rats by permanent bilateral common carotid artery occlusion (BCCAo). Daily oral administration of CP (200 mg/kg) began 21 days after BCCAo and continued for 42 days. The levels of microglial activation and myelin basic protein (MBP) were measured in the white matter and hippocampus of rats with chronic BCCAo, and the expression levels of mitogen-activated protein kinases (MAPKs) and inflammatory markers such as cyclooxygenase-2, interleukin-1ß, and interleukin-6 were examined. RESULTS: MBP expression was reduced in the white matter and hippocampal regions of rats that received BCCAo. In contrast, reduced levels of MBP were not observed in BCCAo rats given CP treatments. The administration of CP alleviated microglial activation, the alteration of ERK and p38 MAPK signaling, and inflammatory mediator expression in rats with chronic BCCAo. CONCLUSION: These results suggest that CP may have protective effects against chronic BCCAo-induced white matter and hippocampal damage by inhibiting inflammatory processes including microglial activation and proinflammatory mediator expression, and downreguating the hyperphosphorylation of ERK and p38 MAPK signaling.


Subject(s)
Cardiotonic Agents/pharmacology , Carotid Stenosis/drug therapy , Drugs, Chinese Herbal/pharmacology , MAP Kinase Signaling System/drug effects , Mitogen-Activated Protein Kinases/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Analysis of Variance , Animals , Carotid Stenosis/metabolism , Carotid Stenosis/physiopathology , Cyclooxygenase 2/metabolism , Dipterocarpaceae , Hippocampus/drug effects , Hippocampus/metabolism , Interleukin-1beta/metabolism , Interleukin-6/metabolism , Male , Microglia/drug effects , Myelin Basic Protein/metabolism , Panax notoginseng , Phosphorylation/drug effects , Rats , Rats, Wistar , Salvia miltiorrhiza
9.
Toxicol Lett ; 223(2): 154-61, 2013 Nov 25.
Article in English | MEDLINE | ID: mdl-24060340

ABSTRACT

We investigated the cytotoxic effects of formaldehyde (FA) on lymphocytes. FA-exposed mice showed a profound reduction not only in the number of natural killer (NK) cells but also in the expression of NK cell-specific receptors, but these mice did not exhibit decreases in the numbers of T or B lymphocytes. FA exposure also induced decreases in NK cytolytic activity and in the expression of NK cell-associated genes, such as IFN-γ, perforin and CD122. To determine the effect of FA on tumorigenicity, C57BL/6 mice were subcutaneously injected with B16F10 melanoma cells after FA exposure. The mass of the B16F10 tumor and the concentration of extravascular polymorphonuclear leukocytes were greater than those in unexposed tumor-bearing control mice. The number and cytolytic activity of NK cells were also reduced in B16F10 tumor-bearing mice exposed to FA. To determine how FA reduces the NK cell number, NK precursor (pNK) cells were treated with FA, and the differentiation status of the NK cells was analyzed. NK cell differentiation was impaired by FA treatment in a concentration-dependent manner. These findings indicate that FA exposure may promote tumor progression by impairing NK cell function and differentiation.


Subject(s)
Cell Differentiation/drug effects , Formaldehyde/toxicity , Killer Cells, Natural/drug effects , Melanoma, Experimental/pathology , Animals , B-Lymphocytes/drug effects , B-Lymphocytes/metabolism , Bronchoalveolar Lavage , Carcinogenesis/chemically induced , Carcinogenesis/pathology , Cell Survival/drug effects , Female , Killer Cells, Natural/metabolism , Mice , Mice, Inbred C57BL , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism
10.
Int J Mol Med ; 31(1): 179-87, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23165955

ABSTRACT

Enalapril and nifedipine are used as antihypertensive drugs; however, the therapeutic target molecules regulated by enalapril and nifedipine have yet to be fully identified. The aim of this study was to identify novel target genes that are specifically regulated by enalapril and nifedipine in tissues from spontaneously hypertensive rats (SHR) using DNA microarray analysis. We found that administration of SHR with enalapril and nifedipine differentially regulated 33 genes involved in the pathogenesis of cardiovascular diseases. Furthermore, we identified 16 genes that have not previously been implicated in cardiovascular diseases, including interleukin-24 (IL-24). Among them, exogenous administration of IL-24 attenuated the expression of vascular inflammation and hypertension-related genes induced by H2O2 treatment in mouse vascular smooth muscle (MOVAS) cells. This study provides valuable information for the development of novel antihypertensive drugs. In addition, the genes identified may be of use as biomarkers and therapeutic targets for cardiovascular diseases, including hypertension.


Subject(s)
Antihypertensive Agents/administration & dosage , Enalapril/administration & dosage , Myocytes, Smooth Muscle/drug effects , Nifedipine/administration & dosage , Transcriptome , Animals , Blood Pressure/drug effects , Brain/drug effects , Brain/metabolism , Heart/drug effects , Hydrogen Peroxide/toxicity , Hypertension/drug therapy , Inflammation/drug therapy , Interleukins/administration & dosage , Liver/drug effects , Liver/metabolism , Male , Mice , Muscle, Smooth, Vascular/cytology , Oligonucleotide Array Sequence Analysis/methods , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Real-Time Polymerase Chain Reaction , Reproducibility of Results
11.
Biochem Biophys Res Commun ; 428(1): 50-5, 2012 Nov 09.
Article in English | MEDLINE | ID: mdl-23063979

ABSTRACT

Vascular calcification is a hallmark of cardiovascular disease. Interleukin-24 (IL-24) has been known to suppress tumor progression in a variety of human cancers. However, the role of IL-24 in the pathophysiology of diseases other than cancer is unclear. We investigated the role of IL-24 in vascular calcification. IL-24 was applied to a ß-glycerophosphate (ß-GP)-induced rat vascular smooth muscle cell (VSMC) calcification model. In this study, IL-24 significantly inhibited ß-GP-induced VSMC calcification, as determined by von Kossa staining and calcium content. The inhibitory effect of IL-24 on VSMC calcification was due to the suppression of ß-GP-induced apoptosis and expression of calcification and osteoblastic markers. In addition, IL-24 abrogated ß-GP-induced activation of the Wnt/ß-catenin pathway, which plays a key role in the pathogenesis of vascular calcification. The specificity of IL-24 for the inhibition of VSMC calcification was confirmed by using a neutralizing antibody to IL-24. Our results suggest that IL-24 inhibits ß-GP-induced VSMC calcification by inhibiting apoptosis, the expression of calcification and osteoblastic markers, and the Wnt/ ß-catenin pathway. Our study may provide a novel mechanism of action of IL-24 in cardiovascular disease and indicates that IL-24 is a potential therapeutic agent in VSMC calcification.


Subject(s)
Apoptosis , Interleukins/physiology , Vascular Calcification/metabolism , Animals , Biomarkers/metabolism , Cells, Cultured , Glycerophosphates/pharmacology , Humans , Interleukins/antagonists & inhibitors , Interleukins/pharmacology , Male , Muscle, Smooth, Vascular , Osteoblasts/metabolism , Rats , Rats, Sprague-Dawley , Vascular Calcification/chemically induced , Vascular Calcification/pathology , Wnt Proteins/metabolism , beta Catenin/metabolism
12.
Pharmacology ; 90(5-6): 332-41, 2012.
Article in English | MEDLINE | ID: mdl-23052189

ABSTRACT

BACKGROUND/AIM: The abnormal growth of vascular smooth muscle cells (VSMCs) induced by reactive oxygen species (ROS) is considered a major pathogenic process in vascular diseases. Interleukin (IL)-24 specifically inhibits cancer cell growth through the induction of cell cycle arrest and apoptosis. However, the role of IL-24 in ROS-induced VSMC growth has not yet been investigated. METHODS: An MTT assay, gene expression analysis, flow cytometry and a scratch wound healing assay were performed to determine the anti-growth effects of IL-24 in H(2)O(2)-treated mouse vascular aortic smooth muscle (MOVAS) cells. To elucidate the effect of IL-24 on ROS-induced signaling, Western blot analysis was employed. RESULTS: IL-24 inhibited the growth of normal MOVAS cells treated with H(2)O(2) by inducing a cell cycle arrest at the G(0)/G(1) phase through the regulation of p21 and cyclin D1. Furthermore, IL-24 suppressed mRNA expression of vascular endothelial growth factor and platelet-derived growth factor and subsequently decreased the level of cell migration in response to H(2)O(2). Interestingly, IL-24 attenuated the H(2)O(2)-induced ROS production by reducing the mitochondrial H(2)O(2) production and enhancing the expression of antioxidant enzymes. We also showed that the ability of H(2)O(2) to induce the PI3K/Akt and Erk signaling pathways was blocked by IL-24. CONCLUSION: These findings suggest a novel mechanism in which IL-24 suppresses the growth of normal VSMCs by inhibiting H(2)O(2)-induced ROS production through the regulation of mitochondrial ROS production and expression of antioxidant enzymes.


Subject(s)
Interleukins/physiology , Myocytes, Smooth Muscle/physiology , Reactive Oxygen Species/metabolism , Animals , Cell Movement/drug effects , Cells, Cultured , Extracellular Signal-Regulated MAP Kinases/metabolism , Mice , Mitochondria/metabolism , Muscle, Smooth, Vascular/cytology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism
13.
Biomol Ther (Seoul) ; 20(5): 492-8, 2012 Sep.
Article in English | MEDLINE | ID: mdl-24009840

ABSTRACT

Phytochemicals have been known to exhibit potent antioxidant activity. This study examined cytoprotective effects of phytochemicals including quercetin, catechin, caffeic acid, and phytic acid against oxidative damage in SK-Hep-1 cells induced by the oxidative and non-oxidative metabolism of ethanol. Exposure of the cells to excess ethanol resulted in a significant increase in cytotoxicity, reactive oxygen species (ROS) production, lipid hydroperoxide (LPO), and antioxidant enzyme activity. Excess ethanol also caused a reduction in mitochondrial membrane potential (MMP) and the quantity of reduced glutathione (GSH). Co-treatment of cells with ethanol and quercetin, catechin, caffeic acid and phytic acid significantly inhibited oxidative ethanol metabolism-induced cytotoxicity by blocking ROS production. When the cells were treated with ethanol after pretreatment of 4-methylpyrazole (4-MP), increased cytotoxicity, ROS production, antioxidant enzyme activity, and loss of MMP were observed. The addition of quercetin, catechin, caffeic acid and phytic acid to these cells showed suppression of non-oxidative ethanol metabolism-induced cytotoxicity, similar to oxidative ethanol metabolism. These results suggest that quercetin, catechin, caffeic acid and phytic acid have protective effects against ethanol metabolism-induced oxidative insult in SK-Hep-1 cells by blocking ROS production and elevating antioxidant potentials.

SELECTION OF CITATIONS
SEARCH DETAIL
...