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










Publication year range
1.
Int J Mol Sci ; 23(22)2022 Nov 13.
Article in English | MEDLINE | ID: mdl-36430486

ABSTRACT

Fulminant hepatitis is characterized by rapid and massive immune-mediated liver injury. Dosage-sensitive sex reversal-adrenal hypoplasia congenita critical region on the X chromosome, gene 1 (DAX1; NR0B1) represses the transcription of various genes. Here, we determine whether DAX1 serves as a regulator of inflammatory liver injury induced by concanavalin A (ConA). C57BL/6J (WT), myeloid cell-specific Dax1 knockout (MKO), and hepatocyte-specific Dax1 knockout (LKO) mice received single intravenous administration of ConA. Histopathological changes in liver and plasma alanine aminotransferase and aspartate aminotransferase levels in Dax1 MKO mice were comparable with those in WT mice following ConA administration. Unlike Dax1 MKO mice, Dax1 LKO mice were greatly susceptible to ConA-induced liver injury, which was accompanied by enhanced infiltration of immune cells, particularly CD4+ and CD8+ T cells, in the liver. Factors related to T-cell recruitment, including chemokines and adhesion molecules, significantly increased following enhanced and prolonged phosphorylation of NF-κB p65 in the liver of ConA-administered Dax1 LKO mice. This is the first study to demonstrate that hepatocyte-specific DAX1 deficiency exacerbates inflammatory liver injury via NF-κB p65 activation, thereby causing T-cell infiltration by modulating inflammatory chemokines and adhesion molecules. Our results suggest DAX1 as a therapeutic target for fulminant hepatitis treatment.


Subject(s)
CD8-Positive T-Lymphocytes , Massive Hepatic Necrosis , Mice , Animals , NF-kappa B , Mice, Inbred C57BL , Hepatocytes , Signal Transduction , Concanavalin A/toxicity , CD4-Positive T-Lymphocytes
2.
Int J Mol Sci ; 23(19)2022 Oct 04.
Article in English | MEDLINE | ID: mdl-36233086

ABSTRACT

Acetaminophen (APAP) is a widely used analgesic and antipyretic drug, but its overdose can cause acute liver failure. The dosage-sensitive sex reversal adrenal hypoplasia congenita critical region on the X chromosome, gene 1 (DAX-1, NR0B1), is an orphan nuclear receptor that acts as a transcriptional co-repressor of various genes. In this study, we identified the role of DAX-1 in APAP-induced liver injury using hepatocyte-specific Dax-1 knockout (Dax-1 LKO) mice. Mouse primary hepatocytes were used as a comparative in vitro study. APAP overdose led to decreased plasma alanine aminotransferase and aspartate aminotransferase levels in Dax-1 LKO mice compared to C57BL/6J (WT) controls, accompanied by reduced liver necrosis. The expression of the genes encoding the enzymes catalyzing glutathione (GSH) synthesis and metabolism and antioxidant enzymes was increased in the livers of APAP-treated Dax-1 LKO mice. The rapid recovery of GSH levels in the mitochondrial fraction of APAP-treated Dax-1 LKO mice led to reduced reactive oxygen species levels, resulting in the inhibition of the prolonged JNK activation. The hepatocyte-specific DAX-1 deficiency increased the protein expression of nuclear factor erythroid 2-related factor 2 (Nrf2) compared with WT controls after APAP administration. These results indicate that DAX-1 deficiency in hepatocytes protects against APAP-induced liver injury by Nrf2-regulated antioxidant defense.


Subject(s)
Antipyretics , Chemical and Drug Induced Liver Injury , DAX-1 Orphan Nuclear Receptor , NF-E2-Related Factor 2 , Acetaminophen/toxicity , Alanine Transaminase/metabolism , Animals , Antioxidants/metabolism , Aspartate Aminotransferases/metabolism , Chemical and Drug Induced Liver Injury/genetics , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/prevention & control , Co-Repressor Proteins/metabolism , DAX-1 Orphan Nuclear Receptor/genetics , Glutathione/metabolism , Hepatocytes/metabolism , Liver/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Orphan Nuclear Receptors/metabolism , Reactive Oxygen Species/metabolism
3.
Anticancer Res ; 35(2): 785-94, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25667458

ABSTRACT

BACKGROUND/AIM: Endocrine therapies that inhibit oestrogen receptor (ER)-α signaling are the most common and effective treatment for ER-α-positive breast cancer. The present study aimed to elucidate the mechanisms by which down-regulation of serum- and glucocorticoid-inducible protein kinase-1 (SGK1) expression confers tamoxifen resistance in breast cancer. MATERIALS AND METHODS: SGK1 expression and the cytotoxic effects of combinatorial 4-hydroxy-tamoxifen (4-OHT) treatment with SGK1 overexpression were investigated by immunoblotting, bromodeoxyuridine incorporation, and soft agar assay. RESULTS: We showed that PIN1 down-regulates SGK1 expression through interaction with and ubiquitination of SGK1. PIN1 silencing in MCF7 cells increased SGK1 expression. In tamoxifen-resistant human breast cancer, immunohistochemical staining analysis showed an inverse correlation between SGK1 expression and severity of tamoxifen resistance. Importantly, 4-OHT in combination with overexpression of SGK1 increased cleavage of poly-(ADP-ribose) polymerase and DNA fragmentation to inhibit clonogenic growth of tamoxifen-resistant MCF7 (TAMR-MCF7) cells. CONCLUSION: We suggest that PIN1-mediated SGK1 ubiquitination is a major regulator of tamoxifen-resistant breast cancer cell growth and survival.


Subject(s)
Immediate-Early Proteins/metabolism , Peptidylprolyl Isomerase/metabolism , Protein Serine-Threonine Kinases/metabolism , Tamoxifen/pharmacology , Base Sequence , Cell Line, Tumor , DNA Primers , Drug Resistance, Neoplasm , Enzyme Stability , Female , Humans , NIMA-Interacting Peptidylprolyl Isomerase , Real-Time Polymerase Chain Reaction
4.
Mol Carcinog ; 54(6): 440-8, 2015 Jun.
Article in English | MEDLINE | ID: mdl-24265246

ABSTRACT

Pin1, a conserved eukaryotic Peptidyl-prolyl cis/trans isomerase, has profound effects on numerous key-signaling molecules, and its deregulation contributes to disease, particularly cancer. Although Pin1-mediated prolyl isomerization is an essential and novel regulatory mechanism for protein phosphorylation, little is known about the upstream signaling pathway(s) that regulates Pin1 activity. Here, we identify MAP3K-related serine-threonine kinase (the gene encoding COT/Tpl2) as a kinase responsible for phosphorylation of Pin1 Ser16. COT interacts with and phosphorylates Pin1 on Ser16. Consequently, Pin1 Ser16 phosphorylation by COT increases cyclin D1 abundance and enhances tumorigenecity of MCF7 cells. In contrast, depletion of COT in MCF7 cells leads to downregulation of Pin1 Ser16 phosphorylation, which subsequently decrease cyclin D1 levels, inhibiting tumorigenecity of MCF7 cells. In a xenograft model, treatment of TKI, a COT inhibitor, and Juglone, a Pin1 inhibitor, abrogates tumor growth. In human breast cancer patients, immunohistochemical staining shows that Pin1 pSer16 levels are positively correlated with COT levels, providing strong evidence for an essential role of the COT/Pin1 axis in conveying oncogenic signals to promote aggressiveness in human breast cancer.


Subject(s)
Breast Neoplasms/metabolism , Carcinogenesis/metabolism , MAP Kinase Kinase Kinases/metabolism , Peptidylprolyl Isomerase/metabolism , Proto-Oncogene Proteins/metabolism , Animals , Breast/metabolism , Breast/pathology , Breast Neoplasms/pathology , Carcinogenesis/pathology , Female , HEK293 Cells , Humans , MCF-7 Cells , Mice, Inbred BALB C , NIMA-Interacting Peptidylprolyl Isomerase , Peptidylprolyl Isomerase/chemistry , Phosphorylation , Protein Structure, Tertiary
5.
Anticancer Res ; 34(3): 1409-16, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24596388

ABSTRACT

BACKGROUND/AIM: Clinical trials have shown efficacy of the anti-HER2 monoclonal antibody trastuzumab in metastatic breast cancer patients. The aim of the present study was to elucidate the mechanisms by which up-regulation of fatty acid synthase (FAS) expression confers resistance to trastuzumab in HER2-positive breast cancers. MATERIALS AND METHODS: The expression of FAS as well as the cytotoxic effects of combinatorial treatment of trastuzumab and juglone was investigated by immunoblotting, BrdU incorporation, TUNEL assay, and soft agar assay. RESULTS: Pin1 enhanced EGF-induced SREBP1c promoter activity, resulting in the induction of FAS expression in BT474 cells. In contrast, juglone, a potent Pin1 inhibitor, significantly enhanced trastuzumab-induced FAS down-regulation and cell death in BT474 cells. Furthermore, trastuzumab, when used in combination with gene silencing or chemical inhibition of Pin1, increased cleaved poly(ADP-ribose) polymerase and DNA fragmentation to increase trastuzumab sensitivity. CONCLUSION: Pin1-mediated FAS overexpression is a major regulator of trastuzumab-resistant breast cancer growth and survival.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Fatty Acid Synthases/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Peptidylprolyl Isomerase/metabolism , Animals , Apoptosis/drug effects , Blotting, Western , Breast Neoplasms/pathology , Cell Adhesion/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/pathology , Epidermal Growth Factor/pharmacology , Female , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , NIMA-Interacting Peptidylprolyl Isomerase , Peptidylprolyl Isomerase/antagonists & inhibitors , Peptidylprolyl Isomerase/genetics , Promoter Regions, Genetic/genetics , RNA, Small Interfering/genetics , Sterol Regulatory Element Binding Protein 1/metabolism , Trastuzumab , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
6.
Carcinogenesis ; 35(6): 1352-61, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24517997

ABSTRACT

Interleukin-22 (IL-22), one of the cytokines secreted by T-helper 17 (Th17) cells, binds to a class II cytokine receptor containing an IL-22 receptor 1 (IL-22R1) and IL-10R2 and influences a variety of immune reactions. IL-22 has also been shown to modulate cell cycle and proliferation mediators such as extracellular signal-regulated kinase (ERK) and c-Jun N-terminal kinase (JNK), but little is known about the underlying molecular mechanisms of IL-22 in tumorigenesis. In this paper, we propose that IL-22 has a crucial role to play in controlling epithelial cell proliferation and tumorigenesis in the breast. IL-22 increased MAP3K8 phosphorylation through IL-22R1, followed by the induction of MEK-ERK, JNK-c-Jun, and STAT3 signaling pathways. Furthermore, IL-22-IL-22R1 signaling pathway activated activator protein-1 and HER2 promoter activity. In addition, Pin1 was identified as a key positive regulator for the phosphorylation-dependent MEK, c-Jun and STAT3 activity induced by IL-22. Pin1(-/-) mouse embryonic fibroblasts (MEF) exhibited significantly a decrease in IL-22-induced MEK1/2, c-Jun, and STAT3 phosphorylation compared with Pin1(+/+) MEF. In addition, a knockdown of Pin1 prevented phosphorylation induced by IL-22. The in vivo chorioallantoic membrane assay also showed that IL-22 increased tumor formation of JB6 Cl41 cells. Moreover, the knockdown of MAP3K8 and Pin1 attenuated tumorigenicity of MCF7 cells. Consistent with these observations, IL-22 levels positively correlate with MAP3K8 and Pin1 expression in human breast cancer. Overall, our findings point to a critical role for the IL-22-induced MAP3K8 signaling pathway in promoting cancer-associated inflammation in the tumor microenvironment.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Transformation, Neoplastic/metabolism , Epithelial Cells/metabolism , Epithelial Cells/pathology , Interleukins/metabolism , MAP Kinase Kinase Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Animals , Breast Neoplasms/genetics , Cell Line , Cell Transformation, Neoplastic/drug effects , Disease Models, Animal , Enzyme Activation/drug effects , Epithelial Cells/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Heterografts , Humans , Interleukins/pharmacology , MAP Kinase Signaling System/drug effects , Mice , Mitogen-Activated Protein Kinases/metabolism , NIMA-Interacting Peptidylprolyl Isomerase , Peptidylprolyl Isomerase/genetics , Peptidylprolyl Isomerase/metabolism , Proto-Oncogene Proteins c-jun/metabolism , STAT3 Transcription Factor/metabolism , Transcription Factor AP-1/metabolism , Interleukin-22
7.
FASEB J ; 27(11): 4606-18, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23934277

ABSTRACT

Pin1, a conserved eukaryotic peptidyl-prolyl cis/trans isomerase, has profound effects on numerous key-signaling molecules, and its deregulation contributes to disease, particularly cancer. Although Pin1-mediated prolyl isomerization of protein servers as a regulatory switch in signaling pathways, the significance of proline isomerase activity in chromatin modifying complex remains unclear. Here, we identify Pin1 as a key negative regulator for suppressor of variegation 3-9 homologue 1 (SUV39H1) stability, a major methyltransferase responsible for histone H3 trimethylation on Lys9 (H3K9me3). Pin1 interacts with SUV39H1 in a phosphorylation-dependent manner and promotes ubiquitination-mediated degradation of SUV39H1. Consequently, Pin1 reduces SUV39H1 abundance and suppresses SUV39H1 ability to induce H3K9me3. In contrast, depletion of Pin1 in cancer cells leads to elevated SUV39H1 expression, which subsequently increases H3K9me3, inhibiting tumorigenecity of cancer cells. In a xenograft model with 4T1 metastatic mouse breast carcinoma cells, Pin1 overexpression increases tumor growth, whereas SUV39H1 overexpression abrogates it. In human breast cancer patients, immunohistochemical staining shows that Pin1 levels are negatively correlated with SUV39H1 as well as H3K9me3 levels. Thus, Pin1-mediated reduction of SUV39H1 stability contributes to convey oncogenic signals for aggressiveness of human breast cancer, suggesting that Pin1 may be a promising drug target for anticancer therapy.


Subject(s)
Breast Neoplasms/metabolism , Carcinogenesis/metabolism , Carcinoma/metabolism , Methyltransferases/metabolism , Peptidylprolyl Isomerase/metabolism , Repressor Proteins/metabolism , Adult , Aged , Animals , Breast Neoplasms/pathology , Carcinoma/pathology , Female , HEK293 Cells , Histones/metabolism , Humans , MCF-7 Cells , Methylation , Methyltransferases/genetics , Mice , Mice, Inbred BALB C , Middle Aged , NIMA-Interacting Peptidylprolyl Isomerase , Peptidylprolyl Isomerase/genetics , Protein Processing, Post-Translational , Repressor Proteins/genetics
8.
Biol Pharm Bull ; 36(9): 1466-73, 2013.
Article in English | MEDLINE | ID: mdl-23748899

ABSTRACT

Diarylureas and diarylamides derivatives are reported to have antitumor activity. Encouraged by the interesting antiproliferative activity of diarylurea and diarylamide derivatives, we synthesized a new series of diarylureas and diarylamides containing pyrrolo[3,2-c]pyridine scaffold. In this study, we demonstrate that a N-(3-(4-benzamido-1H-pyrrolo[3,2-c]pyridin-1-yl)phenyl)-4-morpholino-3-(trifluoromethyl)benzamide, KIST101029, inhibits neoplastic cell transformation induced by insulin-like growth factor 1 (IGF-1) in mouse epidermal JB6 Cl41 cells. The KIST101029 compound inhibited mitogen-activated protein kinase/extracellular signal-regulated kinase kinases (MEK), c-jun N-terminal kinases (JNK), and mechanistic target of rapamycin (mTOR) signaling pathways induced by IGF-1 in JB6 Cl41 cells, resulting in the inhibition of c-fos and c-jun transcriptional activity. In addition, the KIST101029 inhibited the associated activator protein-1 (AP-1) transactivation activity and cell transformation induced by IGF-1 in JB6 Cl41 cells. Consistent with these observations, in vivo chorioallantoic membrane assay also showed that the KIST101029 inhibited IGF-1-induced tumorigenicity of JB6 Cl41 cells. Importantly, KIST101029 suppressed the colony formation of A375 cells in soft agar. Taken together, these results indicate that a KIST101029 might exert chemopreventive effects through the inhibition of phosphorylation of MAPK and mTOR signaling pathway.


Subject(s)
Antineoplastic Agents/pharmacology , Benzamides/pharmacology , Cell Transformation, Neoplastic/drug effects , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-raf/antagonists & inhibitors , Animals , Cell Line , Cell Proliferation/drug effects , Cell Survival/drug effects , Cell Transformation, Neoplastic/chemically induced , Cell Transformation, Neoplastic/metabolism , Chickens , Chorioallantoic Membrane , Insulin-Like Growth Factor I , Mice , Proto-Oncogene Proteins c-raf/metabolism , Transcription Factor AP-1/metabolism
9.
Carcinogenesis ; 34(2): 341-50, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23125217

ABSTRACT

Inflammatory conditions elicited by extrinsic environmental factors promote malignant transformation, tumor growth and metastasis. Although the role of T cells in cancer promotion has been examined, little is known about the underlying molecular mechanisms of interleukin-17 A (IL-17A), a proinflammatory cytokine produced by activated CD4(+) memory T cells, in carcinogenesis. Here, we report that IL-17A induces neoplastic transformation of JB6 Cl41 cells through activation of tumor progression locus 2 (TPL2). IL-17A dose- and time-dependently increases TPL2 phosphorylation in JB6 Cl41 cells through IL-17A receptor. IL-17A activates mitogen-activated protein kinase/extracellular signal-regulated kinase kinases, c-jun N-terminal kinases and STAT3 signaling pathways, which are inhibited by a TPL2 kinase inhibitor (TKI). Furthermore, IL-17A activates c-fos and c-jun promoter activity, resulting in increased activator protein-1 (AP-1) activity. When small interfering RNA of IL-17A receptor (IL-17R), IL-17A and TPL2 were introduced into JB6 Cl41 cells, respectively, IL-17A-induced AP-1 activity was significantly decreased compared with control cells. Similarly, TPL2 inhibition suppressed AP-1 activity induced by IL-17A. The knockdown of IL-17R and TKI treatment in JB6 Cl41 cells resulted in decreased IL-17A-induced cell transformation. The in vivo chorioallantoic membrane assay also showed that IL-17A increased tumor formation of JB6 Cl41 cells, whereas TKI inhibited the tumorigenesis promoted by IL-17A. Consistent with these observations, knockdown of IL-17A and/or inhibition of TPL2 attenuated tumorigenicity of human breast cancer MCF7 cells. Together, our findings point to a critical role for the IL-17A-induced TPL2 signaling pathway in supporting cancer-associated inflammation in the tumor microenvironment. Therapeutic approaches that target this pathway may, therefore, effectively inhibit carcinogenesis.


Subject(s)
Cell Transformation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Interleukin-17/pharmacology , MAP Kinase Kinase Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Transcription Factor AP-1/metabolism , Animals , Apoptosis , Blotting, Western , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Carcinoma, Ductal, Breast/drug therapy , Carcinoma, Ductal, Breast/metabolism , Carcinoma, Ductal, Breast/pathology , Cell Adhesion , Cell Proliferation , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Chickens , Chorioallantoic Membrane , Epidermal Cells , Epidermis/drug effects , Epidermis/metabolism , Female , Humans , Immunoenzyme Techniques , JNK Mitogen-Activated Protein Kinases/metabolism , MAP Kinase Kinase Kinases/antagonists & inhibitors , MAP Kinase Kinase Kinases/genetics , Mitogen-Activated Protein Kinases/metabolism , Phosphorylation/drug effects , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-fos/metabolism , RNA, Small Interfering/genetics , Receptors, Interleukin-17/metabolism , Signal Transduction , Transcriptional Activation
10.
Mol Carcinog ; 52(10): 813-23, 2013 Oct.
Article in English | MEDLINE | ID: mdl-22549912

ABSTRACT

Pin1 is a unique regulator that catalyzes the conversion of a specific phospho-Ser/Thr-Pro-containing motif in target proteins. Herein, we identified AMP-activated protein kinase (AMPK) as a Pin1-binding protein. Pin1 wild-type, but not Pin1 mutant at serine 16 (S16A), associated with AMPK. Reciprocally, the constitutively active form of AMPK (AMPK-CA), but not the dominant negative form of AMPK (AMPK-DN), interacted with Pin1 wild type. In addition, mutation of Ser176 site in AMPK led to a significant loss of binding between AMPK and Pin1. Ablation of the Pin1 gene in MEFs enhanced AMPK phosphorylation induced by AICAR. Pin1 overexpression in Pin1(-/-) MEFs and SK-HEP-1 cells attenuated AMPK phosphorylation induced by EGF, whereas gene knockdown of Pin1 by siRNA enhanced it. The association between Pin1 and AMPK was increased by EGF, leading to their interaction with protein phosphatase-2A (PP2A). Furthermore, Pin1 increased the PP2A activity induced by EGF. In addition, AMPK-WT and AMPK-CA, but not AMPK-DN, inhibited EGF-induced neoplastic cell transformation of JB6 Cl41 cells and tumorigenicity of SK-HEP-1 cells. The overexpression of Pin1 in JB6 Cl41 cells and SK-HEP-1 cells attenuated the inhibitory effect of AMPK in EGF-induced neoplastic cell transformation of JB6 Cl41 and tumorigenicity of SK-HEP-1 cells, respectively. Taken together, these results indicate that Pin1 plays a pivotal role in EGF-induced carcinogenesis through downregulation of AMPK activity in hepatocarcinoma cells.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Carcinoma, Hepatocellular/pathology , Cell Transformation, Neoplastic/pathology , Gene Expression Regulation, Neoplastic , Liver Neoplasms/pathology , Peptidylprolyl Isomerase/physiology , Protein Phosphatase 2/metabolism , AMP-Activated Protein Kinases/antagonists & inhibitors , Animals , Apoptosis , Blotting, Western , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Cell Adhesion , Cell Movement , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cells, Cultured , Down-Regulation , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Glutathione Transferase/metabolism , Humans , Immunoprecipitation , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Mice , Mice, Knockout , NIMA-Interacting Peptidylprolyl Isomerase , Phosphorylation , Protein Binding , Two-Hybrid System Techniques
11.
Biol Pharm Bull ; 34(10): 1553-8, 2011.
Article in English | MEDLINE | ID: mdl-21963494

ABSTRACT

Rhizochalin is a two-headed sphingolipid-like compound isolated from the sponge Rhizochalina incrustata. It has been reported that rhizocalin and its derivates have a chemopreventive and chemotherapeutic effect. However, the molecular mechanism of these effects is not understood. Here, we demonstrate that aglycon of rhizochalin (AglRhz) from the Rhizochalina incrustata induces AMP-activated protein kinase (AMPK) phosphorylation, and thereby inhibits mammalian target of rapamycin (mTOR)-p70S6 kinase-extracellular signal-regulated kinase (ERK) signaling and activator protein 1 (AP-1) activity via phosphorylation of Raptor in HT-29 cells. In addition, AglRhz induced activation of caspase-3 and poly(ADP-ribose) polymerase (PARP), and DNA fragmentation in HT-29 cells, leads to induction of apoptosis as well as suppression of tumorigenicity of HT-29 cells. Notably, AglRhz inhibits insulin-like growth factor (IGF)-1-induced AP-1 activity and cell transformation in JB6 Cl41 cells. Overall, our findings identify AMPK as an important target protein for mediating the anti-tumor properties of AglRhz in HT-29 colon cancer cells and have important implication for sponges, the most important marine source, in colon cancer.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Antineoplastic Agents/pharmacology , Chemoprevention , Colonic Neoplasms/drug therapy , Fatty Alcohols/pharmacology , Glycosphingolipids/pharmacology , AMP-Activated Protein Kinases/genetics , Animals , Antineoplastic Agents/metabolism , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Caspase 3/metabolism , Cell Transformation, Neoplastic/drug effects , Colonic Neoplasms/enzymology , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , DNA Fragmentation/drug effects , Drug Evaluation, Preclinical , Extracellular Signal-Regulated MAP Kinases/metabolism , Fatty Alcohols/metabolism , Fatty Alcohols/therapeutic use , Glycosphingolipids/chemistry , Glycosphingolipids/metabolism , Glycosphingolipids/therapeutic use , HT29 Cells , Humans , Mice , Mitogen-Activated Protein Kinases/metabolism , Molecular Targeted Therapy , Oceans and Seas , Phosphorylation , Phytotherapy , Plant Preparations/chemistry , Plant Preparations/isolation & purification , Plant Preparations/pharmacology , Poly(ADP-ribose) Polymerases/metabolism , Porifera , Protein Serine-Threonine Kinases/metabolism , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , Tetrazolium Salts , Thiazoles , Tumor Cells, Cultured
12.
Mol Nutr Food Res ; 55(4): 594-605, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21462327

ABSTRACT

SCOPE: Capsaicin is a cancer-suppressing agent. The aim of our study was to determine the effect of capsaicin on tumor invasion and migration; the possible mechanisms involved in this inhibition were investigated in human fibrosarcoma cells. METHODS AND RESULTS: We employed invasion, migration and gelatin zymography assays to characterize the effect of capsaicin on HT-1080 cells. Transient transfection assays and immunoblot analysis were performed to study its molecular mechanisms of action. Capsaicin inhibited the epidermal growth factor (EGF)-induced activation of matrix metalloproteinase (MMP)-9 and MMP-2, and further inhibited cell invasion and migration. Capsaicin decreased the EGF-induced expression of MMP-9, MMP-2, and MT1-MMP, but did not alter TIMP-1 and TIMP-2 levels. Capsaicin suppressed EGF-induced c-Jun and c-Fos nuclear translocation, and also abrogated the EGF-induced phosphorylation of EGF receptor (EGFR), focal adhesion kinase (FAK), protein kinase C (PKC), phosphatidylinositol 3-Kinase (PI3K)/Akt, extracellular regulated kinase (ERK)1/2, and JNK1/2, an upstream modulator of AP-1. Furthermore, the EGFR inhibitor inhibited EGF-induced MMP-9 expression, as well as AP-1 activity and cell migration. CONCLUSION: Capsaicin inhibited the EGF-induced invasion and migration of human fibrosarcoma cells via EGFR-dependent FAK/Akt, PKC/Raf/ERK, p38 mitogen-activated protein kinase (MAPK), and AP-1 signaling, leading to the down-regulation of MMP-9 expression. These results indicate the role of capsaicin as a potent anti-metastatic agent, which can markedly inhibit the metastatic and invasive capacity of fibrosarcoma cells.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Capsaicin/pharmacology , Down-Regulation/drug effects , Epidermal Growth Factor/antagonists & inhibitors , Fibrosarcoma/drug therapy , Matrix Metalloproteinase 9/metabolism , Signal Transduction/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Nucleus/drug effects , ErbB Receptors/metabolism , Fibrosarcoma/metabolism , Fibrosarcoma/pathology , Gene Expression Regulation, Neoplastic/drug effects , Hormone Antagonists/pharmacology , Humans , Matrix Metalloproteinase 14/genetics , Matrix Metalloproteinase 14/metabolism , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/genetics , Neoplasm Invasiveness/prevention & control , Phosphorylation/drug effects , Protein Transport/drug effects , RNA, Messenger/metabolism , Transcription Factor AP-1/metabolism
13.
Toxicol Lett ; 203(1): 9-19, 2011 May 30.
Article in English | MEDLINE | ID: mdl-21354279

ABSTRACT

Piperine is a major component of black pepper, Piper nigrum Linn, used widely in traditional medicine. Several previous studies reported that piperine possesses various beneficial biological activities including antioxidant, anti-tumor and anti-inflammation properties. In the present study, we investigated the inhibitory effects of piperine on tumor invasion and migration and the possible mechanisms involved using human fibrosarcoma HT-1080 cells. We found that piperine suppresses PMA-enhanced matrix metalloproteinase-9 (MMP-9) expression at the protein, mRNA, and transcriptional levels through the suppression of NF-κB and AP-1 activation without changing the level of tissue inhibitor of metalloproteinase (TIMP)-1. Piperine also inhibits PMA-enhanced membrane-type 1 MMP expression without changing the level of tissue inhibitor of metalloproteinase (TIMP)-1 and TIMP-2. Piperine inhibited PMA-induced NF-κB and c-Jun nuclear translocation, which are upstream of PMA-induced MMP-9 expression and invasion. Furthermore, piperine strongly repressed the PMA-induced phosphorylation of ERK, which are dependent on the PKCα pathway. In conclusion, we demonstrated that the anti-invasive effects of piperine may occur through inhibition of PKCα and ERK phosphorylation and reduction of NF-κB and AP-1 activation, leading to down-regulation of MMP-9 expression. Thus, piperine has potential as a potent anti-cancer drug in therapeutic strategies for fibrosarcoma metastasis.


Subject(s)
Alkaloids/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , Benzodioxoles/pharmacology , Cell Movement/drug effects , Fibrosarcoma/enzymology , Matrix Metalloproteinase 9/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Piperidines/pharmacology , Polyunsaturated Alkamides/pharmacology , Protein Kinase C-alpha/metabolism , Tetradecanoylphorbol Acetate/pharmacology , Cell Line, Tumor , Dose-Response Relationship, Drug , Down-Regulation , Fibrosarcoma/genetics , Fibrosarcoma/pathology , Gene Expression Regulation, Enzymologic/drug effects , Humans , Matrix Metalloproteinase 9/genetics , NF-kappa B/genetics , NF-kappa B/metabolism , Neoplasm Invasiveness , Phosphorylation , Proto-Oncogene Proteins c-jun/metabolism , RNA, Messenger/metabolism , Signal Transduction/drug effects , Time Factors , Transcription Factor AP-1/genetics , Transcription Factor AP-1/metabolism , Transcription, Genetic/drug effects , Transfection
14.
Carcinogenesis ; 32(4): 545-53, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21216846

ABSTRACT

Phenolic constituents of virgin olive oil are reported to have antitumor activity. However, the underlying molecular mechanisms and specific target proteins of virgin olive oil remain to be elucidated. Here, we report that dialdehydic form of decarboxymethyl ligstroside aglycone (p-HPEA-EDA), a phenolic compound of virgin olive oil, inhibits tumor promoter-induced cell transformation in JB6 Cl41 cells and suppress cyclooxygenase-2 (COX-2) and tumorigenicity by adenosine monophosphate-activated protein kinase (AMPK) activation in HT-29 cells. p-HPEA-EDA inhibited 12-O-tetradecanoylphorbol-13-acetate (TPA)-induced phosphorylation of extracellular signal-regulated kinases 1/2 and p90RSK in JB6 Cl41 cells, resulting in the inhibition of cell proliferation, activator protein-1 transactivation and cell transformation promoted by TPA. Moreover, p-HPEA-EDA strongly inhibited the cell viability and COX-2 expression by activation of AMPK activity in HT-29 cells, resulted from depletion of intracellular adenosine triphosphate. p-HPEA-EDA-induced activation of caspase-3 and poly-adenosine diphosphate-ribose polymerase, phosphorylation of p53 (Ser15) and DNA fragmentation in HT-29 cells, leading to apoptosis. Importantly, p-HPEA-EDA suppressed the colony formation of HT-29 cells in soft agar. In contrast, Compound C, an AMPK inhibitor, and Z-DEVD-FMK, a caspase-3 inhibitor, blocked the p-HPEA-EDA-inhibited colony formation in HT-29 cells. In vivo chorioallantoic membrane assay also showed that p-HPEA-EDA-inhibited tumorigenicity of HT-29 cells. These findings revealed that targeted activation of AMPK and inhibition of COX-2 expression by p-HPEA-EDA contribute to the chemopreventive and chemotherapeutic potential of virgin olive oil against colon cancer cells.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Aldehydes/pharmacology , Antineoplastic Agents/pharmacology , Cell Transformation, Neoplastic/drug effects , Phenols/pharmacology , Plant Oils/chemistry , Apoptosis/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Cyclooxygenase 2/analysis , Cyclopentane Monoterpenes , Enzyme Activation/drug effects , Humans , Olive Oil , Tetradecanoylphorbol Acetate , Tumor Suppressor Protein p53/metabolism
15.
Food Chem Toxicol ; 49(1): 93-9, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20934476

ABSTRACT

Anthocyanins of the purple sweet potato exhibit antioxidant and hepatoprotective activities via a multitude of biochemical mechanisms. However, the signaling pathways involved in the actions of anthocyanin-induced antioxidant enzymes against chronic liver injury are not fully understood. We examined whether an anthocyanin fraction (AF) from purple sweet potato may prevent dimethylnitrosamine (DMN)-induced liver injury by inducing antioxidants via nuclear erythroid 2-related factor 2 (Nrf2) pathways and by reducing inflammation. Treatment with AF attenuated the DMN-induced increased serum alanine aminotransferase and aspartate aminotransferase activities. It also prevented the formation of hepatic malondialdehyde and the depletion of glutathione and maintained normal glutathione-S-transferase (GST) activity in the livers of DMN-intoxicated rats. Furthermore, AF increased the expression of Nrf2, NADPH:quinine oxidoreductase-1, heme oxygenase-1, and GSTα, which were reduced by DMN, and decreased the expression of cyclooxygenase-2 and inducible nitric oxide synthase. An increase in the nuclear translocation of nuclear factor kappa B (NF-κB) was observed in the DMN-induced liver injury group, but AF inhibited this translocation. Taken together, these results demonstrate that AF increases the expression of antioxidant enzymes and Nrf2 and at the same time decreases the expression of inflammatory mediators in DMN-induced liver injury. These data imply that AF induces antioxidant defense via the Nrf2 pathway and reduces inflammation via NF-κB inhibition.


Subject(s)
Anthocyanins/pharmacology , Cyclooxygenase 2/metabolism , Dimethylnitrosamine/antagonists & inhibitors , Ipomoea batatas/chemistry , Liver/drug effects , NF-E2-Related Factor 2/physiology , Nitric Oxide Synthase Type II/metabolism , Animals , Anthocyanins/isolation & purification , Blotting, Western , Dimethylnitrosamine/toxicity , Glutathione/metabolism , Glutathione Transferase/metabolism , Lipid Peroxidation , Liver/enzymology , Liver/metabolism , Male , NF-kappa B/metabolism , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction
16.
J Dermatol Sci ; 61(1): 23-31, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21112745

ABSTRACT

BACKGROUND: Ultraviolet (UV) irradiation causes major changes in skin connective tissues as a result of the degradation of collagen, a major structural component of the extracellular matrix. This process is likely mediated by matrix metalloproteinases (MMPs). Such changes in collagenous skin tissues have been suggested to be causes of cutaneous aging and skin cancer. OBJECTIVE: We investigated the protective effects of apigenin and luteolin on immortalized human keratinocytes (HaCaT) against UVA damage. We then explored the inhibitory effects of apigenin and luteolin on UVA-induced MMP-1 and investigated the molecular mechanism underlying those effects. METHODS: HaCaT cells were treated with apigenin and luteolin for the indicated times followed by irradiation with UVA. Those effects were assessed by semi-quantitative PCR, Western blotting and enzymic activity assays. RESULTS: These two compounds, at concentrations of 1-5µM, increased the viability of, and inhibited ROS production in HaCaT cells exposed to UVA irradiation. Pre-treatment of HaCaT cells with apigenin and luteolin also inhibited UVA-induced production of the collagenases MMP-1. They also suppressed UVA-induced expression of c-Jun and c-Fos and the phosphorylation of three MAP kinases, upstream modulators of AP-1. Furthermore, the same two flavonoids decreased the UVA-induced influx of Ca(2+) into HaCaT cells and the phosphorylation of Ca(2+)/calmodulin-dependent kinases (CaMKs). CONCLUSION: The results indicate that apigenin and luteolin inhibited UVA-induced collagenolytic MMP-1 production by interfering with Ca(2+)-dependent MAPKs and AP-1 signaling. They may thus be potentially useful in the prevention and treatment of skin photoaging.


Subject(s)
Apigenin/pharmacology , Keratinocytes/metabolism , Luteolin/pharmacology , MAP Kinase Signaling System/drug effects , Matrix Metalloproteinase 1/metabolism , Mitogen-Activated Protein Kinases/metabolism , Transcription Factor AP-1/metabolism , Ultraviolet Rays/adverse effects , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cell Survival/drug effects , Cell Survival/radiation effects , Cells, Cultured , Collagenases/metabolism , Enzyme Activation/drug effects , Genes, fos/drug effects , Genes, jun/drug effects , Humans , Keratinocytes/drug effects , Keratinocytes/enzymology , Keratinocytes/radiation effects , MAP Kinase Signaling System/radiation effects , Matrix Metalloproteinase 1/drug effects , Phosphorylation , Reactive Oxygen Species/metabolism , Signal Transduction
17.
Toxicol Lett ; 198(2): 200-9, 2010 Oct 05.
Article in English | MEDLINE | ID: mdl-20599481

ABSTRACT

In this study, we determined the effects of a novel chlorogenic acid, 3-caffeoyl, 4-dicaffeoylquinic acid (CDCQ) isolated from Salicornia herbacea, on tumor invasion and migration in human fibrosarcoma HT-1080 cells and investigated the possible mechanism(s) involved. CDCQ reduced the phorbol myristate acetate (PMA)-induced activation of matrix metalloproteinase (MMP)-9 and MMP-2 and inhibited cell invasion and migration. CDCQ suppressed PMA-induced expression of MMP-9 mRNA and protein by suppressing the transcription factor AP-1, without changing the level of tissue inhibitor of metalloproteinase (TIMP)-1. CDCQ-inhibited PMA-induced MMP-2 expression by suppressing membrane-type 1 MMP (MT1-MMP), but did not alter the TIMP-2 level. CDCQ also inhibited the PMA-induced nuclear translocation of c-Jun and c-Fos, which are upstream of PMA-induced MMP-9 expression. Furthermore, CDCQ strongly repressed PMA-induced phosphorylation of ERK, p38 MAPK, and JNK, which are dependent on the PKCdelta pathway. In conclusion, we demonstrated that the anti-invasive effects of CDCQ occur through the inhibition of AP-1 and signaling pathways involving PKCdelta and three MAPKs, leading to the downregulation of MMP-9 expression. Thus, CDCQ is an effective anti-metastatic agent that functions by downregulating MMP-9 gene expression.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Chenopodiaceae/chemistry , Chlorogenic Acid/analogs & derivatives , Gene Expression/drug effects , Matrix Metalloproteinase 9/genetics , Protein Kinase C-delta/metabolism , Antineoplastic Agents, Phytogenic/isolation & purification , Blotting, Western , Cell Line, Tumor , Cell Movement/drug effects , Cell Survival/drug effects , Chlorogenic Acid/isolation & purification , Chlorogenic Acid/pharmacology , Humans , Reverse Transcriptase Polymerase Chain Reaction
18.
Biochem Pharmacol ; 79(12): 1714-26, 2010 Jun 15.
Article in English | MEDLINE | ID: mdl-20152819

ABSTRACT

Dihydroartemisinin (DHA), a semi-synthetic derivative of artemisinin, has recently been shown to possess antitumor activity in various cancer cells. However, the effects of DHA in preventing the invasion of cancer cells have not been studied. In the present study, we investigated the inhibitory effects of DHA on tumor invasion and migration and the possible mechanisms involved using human fibrosarcoma HT-1080 cells. DHA reduced PMA-induced activation of MMP-9 and MMP-2 and further inhibited cell invasion and migration. DHA suppressed PMA-enhanced expression of MMP-9 protein, mRNA, and transcriptional activity through suppressing NF-kappaB and AP-1 activation without changing the level of tissue inhibitor of metalloproteinase (TIMP)-1. DHA also reduced PMA-enhanced MMP-2 expression by suppressing membrane-type 1 MMP (MT1-MMP), but did not alter TIMP-2 levels. DHA-inhibited PMA-induced NF-kappaB and c-Jun nuclear translocation, which are upstream of PMA-induced MMP-9 expression and invasion. Furthermore, DHA strongly repressed the PMA-induced phosphorylation of Raf/ERK and JNK, which are dependent on the PKCalpha pathway. In conclusion, we demonstrated that the anti-invasive effects of DHA may occur through inhibition of PKCalpha/Raf/ERK and JNK phosphorylation and reduction of NF-kappaB and AP-1 activation, leading to down-regulation of MMP-9 expression. The data presented show that DHA is an effective anti-metastatic agent that functions by down-regulating MMP-9 gene expression.


Subject(s)
Artemisinins/pharmacology , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , NF-kappa B/antagonists & inhibitors , Protein Kinase C-alpha/antagonists & inhibitors , Transcription Factor AP-1/antagonists & inhibitors , raf Kinases/antagonists & inhibitors , Artemisinins/chemistry , Cell Line, Tumor , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , MAP Kinase Kinase 4/antagonists & inhibitors , MAP Kinase Kinase 4/genetics , MAP Kinase Kinase 4/metabolism , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Matrix Metalloproteinase Inhibitors , Mitogen-Activated Protein Kinase Kinases/genetics , Mitogen-Activated Protein Kinase Kinases/metabolism , Molecular Structure , NF-kappa B/genetics , NF-kappa B/metabolism , Protein Kinase C-alpha/metabolism , Signal Transduction/drug effects , Transcription Factor AP-1/genetics , Transcription Factor AP-1/metabolism , raf Kinases/genetics , raf Kinases/metabolism
19.
Mol Nutr Food Res ; 54(7): 977-90, 2010 Jul.
Article in English | MEDLINE | ID: mdl-19943262

ABSTRACT

Matrix metalloproteinase (MMP) plays an important role in the invasion and metastasis of cancer cells. The inhibitory effects of bergamottin, a cytochrome P450 inhibitor from Citrus paradis (grapefruit), on tumor invasion and migration and the possible mechanisms involved in this inhibition were investigated in human fibrosarcoma HT-1080 cells. Bergamottin reduced phorbol-12-myristate-13-acetate (PMA)-induced activation of MMP-9 and MMP-2 and further inhibited cell invasion and migration. Bergamottin suppressed PMA-enhanced expression of MMP-9 protein, mRNA and transcription activity levels through suppression of nuclear factor-kappaB (NF-kappaB) activation without changing the tissue inhibitor of metalloproteinase 1 level. Bergamottin also reduced PMA-enhanced MMP-2 expression through suppression of membrane-type 1 MMP, but did not alter tissue inhibitor of metalloproteinase 2 levels. Bergamottin inhibited PMA-induced NF-kappaB nuclear translocation and IkappaBalpha degradation, which are upstream of PMA-induced MMP-9 expression and invasion. Furthermore, bergamottin strongly repressed the PMA-induced phosphorylation of p38 mitogen-activated protein kinase and c-Jun N-terminal kinase (JNK), which are dependent on the protein kinase C-delta pathway. In conclusion, we demonstrated that the anti-invasive effects of bergamottin might occur through inhibition of protein kinase C-delta, p38 mitogen-activated protein kinase, and JNK phosphorylation and reduction of NF-kappaB activation, leading to downregulation of MMP-9 expression. These results suggest that the suppression of MMP expression contributes, at least in part, to the antitumor activity of bergamottin.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Carcinogens/toxicity , Furocoumarins/pharmacology , Matrix Metalloproteinase 9/metabolism , Signal Transduction/drug effects , Tetradecanoylphorbol Acetate/analogs & derivatives , Carcinogens/antagonists & inhibitors , Cell Line, Tumor , Cell Movement/drug effects , Cytochrome P-450 Enzyme Inhibitors , Down-Regulation/drug effects , Fibrosarcoma/drug therapy , Fibrosarcoma/metabolism , Fibrosarcoma/pathology , Humans , I-kappa B Proteins/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/genetics , NF-KappaB Inhibitor alpha , NF-kappa B/metabolism , Neoplasm Invasiveness/prevention & control , Osmolar Concentration , Phosphorylation/drug effects , Protein Kinase C-delta/metabolism , Protein Transport/drug effects , RNA, Messenger/metabolism , Tetradecanoylphorbol Acetate/antagonists & inhibitors , Tetradecanoylphorbol Acetate/toxicity , p38 Mitogen-Activated Protein Kinases/metabolism
20.
Chem Biol Interact ; 181(3): 366-76, 2009 Oct 30.
Article in English | MEDLINE | ID: mdl-19647727

ABSTRACT

Salicornia herbacea has been used as a folk medicine for disorders such as constipation, obesity, diabetes, and cancer. Recent studies have shown that S. herbacea has antioxidative, anti-inflammatory, immunomodulatory, antihyperglycemic, and antihyperlipidemic activities. In the present work, we investigated the protective effects of the chlorogenic acid derivative, 3-caffeoyl, 4-dihydrocaffeoyl quinic acid (CDCQ), which was isolated from S. herbacea, against tert-butyl hydroperoxide (t-BHP)-induced hepatotoxicity in Hepa1c1c7 cells. Pretreatment of Hepa1c1c7 cells with CDCQ significantly reduced t-BHP-induced generation of ROS, caspase-3 activation, and subsequent cell death. Also, CDCQ up-regulated heme oxygenase-1 (HO-1) expression, which conferred cytoprotection against oxidative injury induced by t-BHP. Moreover, CDCQ-induced nuclear translocation of the transcription factor NF-E2-related factor 2 (Nrf2), which is upstream of CDCQ-induced HO-1 expression, and PI3K/Akt activation, a pathway that is involved in induced Nrf2 nuclear translocation. Taken together, these results suggest that the protective effects of CDCQ against t-BHP-induced hepatotoxicity may be due, at least in part, to its ability to scavenge ROS and to regulate the antioxidant enzyme HO-1 via the PI3K/Akt-Nrf2 signaling pathways.


Subject(s)
Antioxidants/pharmacology , Chenopodiaceae/chemistry , Chlorogenic Acid/analogs & derivatives , Oxidative Stress , tert-Butylhydroperoxide/toxicity , Blotting, Western , Caspase 3/metabolism , Cell Line , Chlorogenic Acid/pharmacology , Humans , Immunohistochemistry , In Situ Nick-End Labeling , NF-E2-Related Factor 2/genetics , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , RNA, Small Interfering , Reactive Oxygen Species/metabolism , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...