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










Publication year range
1.
Immunopharmacol Immunotoxicol ; 43(6): 749-757, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34591732

ABSTRACT

OBJECTIVE: Cerebral infarction (CI) is the mayor reason of death in China. Reperfusion is the only immediate treatment for acute cerebral infarction. However, blood reperfusion recovery may cause ischemia-reperfusion (I/R) injuries. The purpose of this study was to investigate the effects of Tetrandrine (TTD) and 3-n-Butylphthalide (NBP) on cerebral I/R injury. MATERIALS AND METHODS: I/R was used to establish CI model in vivo. TTD was performed to analyze cerebral infarction volume. OGD was applied to establish CI model in vitro. Flow cytometry and TUNEL assays were utilized to determine the cell death. ELISA was conducted to determine the release of cytokines. mRNA and protein expressions were detected using qRT-PCR and western blot. RESULTS: We found that NBP + TTD treatment significantly reduced cerebral infarction volume and inhibited the death of neurons in vivo. Moreover, NBP + TTD treatment suppressed the apoptosis and inflammatory response of neurons in vitro. Additionally, NBP + TTD suppressed the expression of activator transcription factor 2 (ATF2). However, overexpression of ATF2 contributed to the degeneration of neurons. Moreover, ATF2 transcriptionally activated Toll-like receptor 4 (TLR4). NBP + TTD inactivated ATF2/TLR4 signaling. CONCLUSIONS: Taken together, TTD combined with NBP protected against cerebral infarction by inhibiting the inflammatory response and neuronal cell apoptosis via inactivating ATF2/TLR4 signaling pathways. This may provide an alternative for I/R injury.


Subject(s)
Activating Transcription Factor 2/antagonists & inhibitors , Benzofurans/administration & dosage , Benzylisoquinolines/administration & dosage , Brain Ischemia/prevention & control , Reperfusion Injury/prevention & control , Toll-Like Receptor 4/antagonists & inhibitors , Activating Transcription Factor 2/metabolism , Animals , Brain Ischemia/metabolism , Cells, Cultured , Drug Therapy, Combination , Male , Mice , Mice, Inbred C57BL , Neuroprotective Agents/administration & dosage , Reperfusion Injury/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology , Toll-Like Receptor 4/metabolism
2.
Cells ; 10(8)2021 08 12.
Article in English | MEDLINE | ID: mdl-34440842

ABSTRACT

This study aimed to investigate the mechanistic pathway of Naja atra (Taiwan cobra) cardiotoxin 1 (CTX1)-induced death of leukemia cell lines U937 and HL-60. CTX1 increased cytoplasmic Ca2+ and reactive oxygen species (ROS) production, leading to the death of U937 cells. It was found that Ca2+-induced NOX4 upregulation promoted ROS-mediated p38 MAPK phosphorylation, which consequently induced c-Jun and ATF-2 phosphorylation. Using siRNA knockdown, activated c-Jun and ATF-2 were demonstrated to regulate the expression of Fas and FasL, respectively. Suppression of Ca2+-mediated NOX4 expression or ROS-mediated p38 MAPK activation increased the survival of U937 cells exposed to CTX1. FADD depletion abolished CTX1-induced cell death, caspase-8 activation, and t-Bid production, supporting the correlation between the Fas death pathway and CTX1-mediated cytotoxicity. Among the tested N. atra CTX isotoxins, only CTX1 induced Fas and FasL expression. Chemical modification studies revealed that intact Met residues were essential for the activity of CTX1 to upregulate Fas and FasL expression. Taken together, the data in this study indicate that CTX1 induces c-Jun-mediated Fas and ATF-2-mediated FasL transcription by the Ca2+/NOX4/ROS/p38 MAPK axis, thereby activating the Fas death pathway in U937 cells. Furthermore, CTX1 activates Fas/FasL death signaling in the leukemia cell line HL-60.


Subject(s)
Cardiotoxins/pharmacology , Fas Ligand Protein/metabolism , Naja naja/metabolism , Signal Transduction/drug effects , fas Receptor/metabolism , Activating Transcription Factor 2/antagonists & inhibitors , Activating Transcription Factor 2/genetics , Activating Transcription Factor 2/metabolism , Animals , Apoptosis/drug effects , Calcium/metabolism , Cell Line, Tumor , Fas-Associated Death Domain Protein/antagonists & inhibitors , Fas-Associated Death Domain Protein/genetics , Fas-Associated Death Domain Protein/metabolism , Humans , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , JNK Mitogen-Activated Protein Kinases/genetics , JNK Mitogen-Activated Protein Kinases/metabolism , Leukemia/metabolism , Leukemia/pathology , NADPH Oxidase 4/metabolism , Phosphorylation/drug effects , RNA Interference , RNA, Small Interfering/metabolism , Reactive Oxygen Species/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
3.
Inflammation ; 44(1): 91-103, 2021 Feb.
Article in English | MEDLINE | ID: mdl-32789554

ABSTRACT

Activating transcription factor 2(ATF2), a transcription factor belonging to the AP-1 family, plays an important role in inflammation. However, its biological functions and underlying molecular mechanisms in rheumatoid arthritis (RA) remain unclear. Western blot and immunohistochemistry were used to identify the expression of ATF2 and Sprouty2(SPRY2) in RA synovial tissues. SW982 cells were stimulated by TNF-α to establish an in vitro RA fibroblast-like synoviocyte (RA-FLS) model. Transwell and monolayer wound-healing were used to detect cell migration and invasion. RNA interference (si-ATF2) and adenovirus vector (Ad-SPRY2) methods were employed to manipulate ATF2 or SPRY2 expression in SW982 cells. The protein expression and phosphorylation levels in SW982 cells were evaluated by western blot. ATF2 expression and phosphorylation were upregulated in the RA synovial tissues. In RA-FLS model, ATF2 expression and phosphorylation were increased in a time-dependent manner. ATF2 knockdown inhibited the migration and invasion of RA-FLS model, reducing the inflammatory factors, which was consistent with the influence on cell behaviors caused by SPRY2 overexpression. Moreover, SPRY2 overexpression inhibited the TNF-α-induced phosphorylation of ERK and ATF2 in SW982 cells. The high expression and phosphorylation of ATF2 promoted migration and invasion of RA-FLSs. SPRY2 might inhibited the inflammatory responses of RA-FLSs via suppressing ERK-ATF2 pathway.


Subject(s)
Activating Transcription Factor 2/biosynthesis , Arthritis, Rheumatoid/metabolism , Cell Movement/physiology , Fibroblasts/metabolism , Intracellular Signaling Peptides and Proteins/biosynthesis , Membrane Proteins/biosynthesis , Synoviocytes/metabolism , Activating Transcription Factor 2/antagonists & inhibitors , Activating Transcription Factor 2/genetics , Arthritis, Rheumatoid/genetics , Arthritis, Rheumatoid/pathology , Cells, Cultured , Down-Regulation/physiology , Fibroblasts/pathology , Gene Expression Regulation , Humans , Intracellular Signaling Peptides and Proteins/genetics , Membrane Proteins/genetics , Phosphorylation/physiology , Synoviocytes/pathology
4.
Respir Res ; 21(1): 39, 2020 Feb 03.
Article in English | MEDLINE | ID: mdl-32014006

ABSTRACT

BACKGROUND: Recent studies have shown 6'-O-galloylpaeoniflorin (GPF), a nature product extracted from the roots of paeoniflorin exerts anti-oxidant and anti-inflammatory activities. However, the effects of GPF on the proliferation and invasion in non-small cell lung cancer (NSCLC) cells have not been clarified. METHODS: MTT assay was performed to determine the cytotoxicity of GPF treatment on NSCLC cells. Colony formation assay, cell scratch test and transwell assay were performed to determine the proliferation and invasion of NSCLC cells in vitro, respectively. An A549 cell xenograft mouse model was performed to confirm the growth of NSCLC cells in vivo. Western blotting was used to measure the levels of activating transcription factor 2 (ATF2), AMP-activated protein kinase (AMPK) and phosph-AMPK (p-AMPK). Luciferase assay was used to validate the binding of miR-299-5p on the 3' untranslated region (UTR) of ATF2. RESULTS: Administration of GPF (50 or 100 µM) was significantly cytotoxic to A549 cells and H1299 cells, as well as inhibited the clonality, invasion and metastasis of NSCLC cells in vitro. GPF treatment also inhibited the tumor growth of NSCLC cell mouse xenografts in vivo. Exotic expression of miR-299-5p significantly inhibited the growth of NSCLC cells in vitro and in vivo. Downregulation of miR-299-5p expression attenuated the inhibition of the proliferation and metastasis of non-small cell lung cancer cells by GPF treatment. miR-299-5p significantly decreased ATF2 mRNA and protein levels in A549 cells (p < 0.05). Overexpression of ATF2 blocked the inhibitory effect of miR-299-5p on the proliferation and invasiveness of A549 cells. CONCLUSIONS: GPF regulates miR-299-5p/ATF2 axis in A549 cells via the AMPK signalling pathway, thereby inhibiting the proliferation and metastasis of non-small cell lung cancer cells.


Subject(s)
Activating Transcription Factor 2/metabolism , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Carcinoma, Non-Small-Cell Lung/metabolism , Glucosides/pharmacology , Lung Neoplasms/metabolism , MAP Kinase Signaling System/physiology , MicroRNAs/metabolism , Monoterpenes/pharmacology , A549 Cells , Activating Transcription Factor 2/antagonists & inhibitors , Animals , Bridged Bicyclo Compounds, Heterocyclic/therapeutic use , Carcinoma, Non-Small-Cell Lung/pathology , Cell Proliferation/drug effects , Cell Proliferation/physiology , Dose-Response Relationship, Drug , Female , Glucosides/therapeutic use , Humans , Lung Neoplasms/pathology , MAP Kinase Signaling System/drug effects , Male , Mice , Mice, Inbred BALB C , Mice, Nude , MicroRNAs/antagonists & inhibitors , Monoterpenes/therapeutic use , Xenograft Model Antitumor Assays/methods
5.
Cell Biol Int ; 41(6): 599-610, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28318081

ABSTRACT

Pancreatic cancer is one of the leading causes of cancer-related death worldwide. Activating transcription factor 2 (ATF2) is a multifunctional transcription factor, and is implicated in tumor progress, yet its role in pancreatic cancer remains unclear. In the present study, the level of ATF2 in pancreatic cancer tissues and the adjacent non-tumorous tissues was detected by quantitative real-time PCR and Western blot. The roles of ATF2 in the proliferation, cell cycle, and apoptosis of pancreatic cancer cells were investigated through ATF2 silencing, and the effect of ATF2 shRNA on the sensitivity of pancreatic cancer cells to gemcitabine, an anti-tumor drug, was explored. The results of our study showed that the ATF2 level in the pancreatic cancer tissues was higher than that in the adjacent non-tumorous tissues. Silencing of ATF2 was found to inhibit proliferation, arrest cell cycle at G1 phase and induce apoptosis in pancreatic cancer cells. Moreover, ATF2 silencing enhanced gemcitabine-induced growth-inhibition and apoptosis-induction effects in pancreatic cancer cells. In summary, silencing of ATF2 inhibited the growth of pancreatic cancer cells and enhanced the anti-tumor effects of gemcitabine, suggesting that ATF2 plays a pro-survival role in pancreatic cancer. Our results also propose that a high level of ATF2 may serve as a potential biomarker of pancreatic cancer, and that ATF2 may become a potential target for anti-tumor therapy.


Subject(s)
Activating Transcription Factor 2/genetics , Activating Transcription Factor 2/metabolism , Pancreatic Neoplasms/metabolism , Activating Transcription Factor 2/antagonists & inhibitors , Antimetabolites, Antineoplastic/pharmacology , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Biomarkers, Tumor/metabolism , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Drug Resistance, Neoplasm/drug effects , Humans , Pancreas/metabolism , RNA, Small Interfering/pharmacology , Real-Time Polymerase Chain Reaction/methods , Gemcitabine
6.
Cell Physiol Biochem ; 41(2): 623-634, 2017.
Article in English | MEDLINE | ID: mdl-28214831

ABSTRACT

BACKGROUND & AIMS: Our previous studies have provided evidence that p38 mitogen-activated protein kinase (MAPK) is involved in total parenteral nutrition (TPN)-associated complications, but its exact effects and mechanisms have not been fully understood. This study aimed to evaluate the roles of p38 MAPK inhibitor SB203580 in the TPN-induced loss of intestinal barrier function and liver disease. METHODS: A rodent model of TPN was used to analyze the roles of SB203580 in TPN-associated complications.Intestinal barrier function was evaluated by transepithelial electrical resistance (TER) and paracellular permeability in Caco-2 cells. The palmitic acid (PA) was used to induce hepatic lipoapoptosis in vitro. The lipoapoptosis was detected using Caspase-3/7 and lipid staining. RESULTS: In the present study, we showed that SB203580 treatment significantly suppressed TPN-mediated intestinal permeability in rats. SB203580 treatment significantly inhibited IL-1ß-induced an increase in tight junction permeability of Caco-2 cells via repressing the p38/ATF-2 signaling. Unexpectedly, SB203580 treatment enhanced hepatic lipoapoptosis in the model of TPN. Palmitic acid (PA)-induced hepatic lipoapoptosis in human liver cells was significantly augmented by the SB203580 treatment. CONCLUSIONS: We demonstrate that the p38 MAPK inhibitor SB203508 ameliorates intestinal barrier function but promotes hepatic lipoapoptosis in model of TPN.


Subject(s)
Apoptosis/drug effects , Imidazoles/pharmacology , Intestinal Mucosa/drug effects , Pyridines/pharmacology , p38 Mitogen-Activated Protein Kinases/metabolism , Activating Transcription Factor 2/antagonists & inhibitors , Activating Transcription Factor 2/genetics , Activating Transcription Factor 2/metabolism , Animals , Caco-2 Cells , Caspase 3/metabolism , Caspase 7/metabolism , Hepatocytes/cytology , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Interleukin-1beta/pharmacology , Intestinal Mucosa/metabolism , Male , Membrane Potential, Mitochondrial/drug effects , Models, Animal , Palmitic Acid/toxicity , Parenteral Nutrition, Total , Permeability/drug effects , RNA Interference , Rats , Rats, Sprague-Dawley , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors
7.
Oncotarget ; 7(43): 70058-70065, 2016 Oct 25.
Article in English | MEDLINE | ID: mdl-27588402

ABSTRACT

In human cancers, miRNAs are important regulators of multiple cellular processes, and aberrant miRNA expression has been observed, and their alterations contribute to multiple cancer development and progression. Till now, little has been known about the role of miR-204 in human glioblastoma (GBM). In the present study, we used in-vitro assays to investigate the mechanisms of miR-204 in GBM cell lines and 60 cases of GBM tissues. Here, we found that miR-204 expression is downregulated in both GBM cell lines A172, U87 and U251 cells and GBM tissues as compared with NHA cells and normal tissues (all p<0.001). In addition, the ectopic expression of miR-204 suppressed A172 and U87 cell proliferation, migration and invasion. Meanwhile, miR-204 over-expression extremely inhibited the protein expression of ATF2. Notably, the enforced expression of ATF2 in A172 and U87 cells with the over-expression of miR-204 attenuated the inhibitory effects of miR-204 on proliferation, migration and invasion. In conclusion, our findings suggest that miR-204 suppressed cell proliferation, migration and invasion through inhibition of ATF2, thus, miR-204 may function as a useful drug target in the treatment and diagnosis of GBM.


Subject(s)
Activating Transcription Factor 2/antagonists & inhibitors , Brain Neoplasms/prevention & control , Glioblastoma/prevention & control , MicroRNAs/physiology , Activating Transcription Factor 2/physiology , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Movement , Cell Proliferation , Disease Progression , Glioblastoma/pathology , Humans
8.
J Nutr Biochem ; 34: 17-29, 2016 08.
Article in English | MEDLINE | ID: mdl-27162022

ABSTRACT

Benzo(a)pyrene [B(a)P] is an environmental toxicant that alters the steroidogenic profile of testis and induces testicular dysfunction. In the present study, we have investigated the molecular signaling of B(a)P and the ameliorative potential of the natural aryl hydrocarbon receptor (AhR) antagonist and antioxidant, resveratrol, on B(a)P-induced male reproductive toxicity. Studies showed that B(a)P treatment resulted in p38 MAPK activation and increased inducible nitric oxide synthase (iNOS) production along with testicular apoptosis and steroidogenic dysfunction. Resveratrol cotreatment maintained testicular redox potential, increased serum testosterone level and enhanced expression of major testicular steroidogenic proteins (CYPIIA1, StAR, 3ßHSD, 17ßHSD) and prevented subsequent onset of apoptosis. Resveratrol cotreatment resulted inhibition of testicular cytochrome P4501A1 (CYP1A1) expression, which is the major B(a)P metabolizing agent for BPDE-DNA adduct formation. Resveratrol also significantly decreased the B(a)P-induced AhR protein level, its nuclear translocation and subsequent promoter activation, thereby decreased the expression of CYP1A1. Resveratrol also down-regulated B(a)P-induced testicular iNOS production through suppressing the activation of p38 MAPK and ATF2, thus improved the oxidative status of the testis and prevented apoptosis. Our findings cumulatively suggest that resveratrol inhibits conversion of B(a)P into BPDE by modulating the transcriptional regulation of CYP1A1 and acting as an antioxidant thus prevents B(a)P-induced oxidative stress and testicular apoptosis.


Subject(s)
Antioxidants/therapeutic use , Benzo(a)pyrene/antagonists & inhibitors , Dietary Supplements , Environmental Pollutants/antagonists & inhibitors , Infertility, Male/prevention & control , Stilbenes/therapeutic use , Testis/drug effects , Activating Transcription Factor 2/agonists , Activating Transcription Factor 2/antagonists & inhibitors , Activating Transcription Factor 2/genetics , Activating Transcription Factor 2/metabolism , Active Transport, Cell Nucleus/drug effects , Animals , Antioxidants/adverse effects , Apoptosis/drug effects , Basic Helix-Loop-Helix Transcription Factors/agonists , Basic Helix-Loop-Helix Transcription Factors/antagonists & inhibitors , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Benzo(a)pyrene/administration & dosage , Benzo(a)pyrene/toxicity , Cytochrome P-450 CYP1A1/antagonists & inhibitors , Cytochrome P-450 CYP1A1/genetics , Cytochrome P-450 CYP1A1/metabolism , Dietary Supplements/adverse effects , Dose-Response Relationship, Drug , Environmental Pollutants/administration & dosage , Environmental Pollutants/toxicity , Gene Expression Regulation/drug effects , Infertility, Male/chemically induced , Infertility, Male/metabolism , Infertility, Male/pathology , MAP Kinase Signaling System/drug effects , Male , Nitric Oxide Synthase Type II/antagonists & inhibitors , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Oxidative Stress/drug effects , Promoter Regions, Genetic/drug effects , Rats, Wistar , Receptors, Aryl Hydrocarbon/agonists , Receptors, Aryl Hydrocarbon/antagonists & inhibitors , Receptors, Aryl Hydrocarbon/genetics , Receptors, Aryl Hydrocarbon/metabolism , Resveratrol , Stilbenes/adverse effects , Testis/metabolism , Testis/pathology , Testosterone/agonists , Testosterone/antagonists & inhibitors , Testosterone/blood
9.
PLoS One ; 10(5): e0126459, 2015.
Article in English | MEDLINE | ID: mdl-25961580

ABSTRACT

The M2 isoform of pyruvate kinase M2 (PKM2) has been shown to be up-regulated in human skin cancers. To test whether PKM2 may be a target for chemoprevention, shikonin, a natural product from the root of Lithospermum erythrorhizon and a specific inhibitor of PKM2, was used in a chemically-induced mouse skin carcinogenesis study. The results revealed that shikonin treatment suppressed skin tumor formation. Morphological examinations and immunohistochemical staining of the skin epidermal tissues suggested that shikonin inhibited cell proliferation without inducing apoptosis. Although shikonin alone suppressed PKM2 activity, it did not suppress tumor promoter-induced PKM2 activation in the skin epidermal tissues at the end of the skin carcinogenesis study. To reveal the potential chemopreventive mechanism of shikonin, an antibody microarray analysis was performed, and the results showed that the transcription factor ATF2 and its downstream target Cdk4 were up-regulated by chemical carcinogen treatment; whereas these up-regulations were suppressed by shikonin. In a promotable skin cell model, the nuclear levels of ATF2 were increased during tumor promotion, whereas this increase was inhibited by shikonin. Furthermore, knockdown of ATF2 decreased the expression levels of Cdk4 and Fra-1 (a key subunit of the activator protein 1. In summary, these results suggest that shikonin, rather than inhibiting PKM2 in vivo, suppresses the ATF2 pathway in skin carcinogenesis.


Subject(s)
Activating Transcription Factor 2/antagonists & inhibitors , Antineoplastic Agents, Phytogenic/pharmacology , Cell Transformation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic , Naphthoquinones/pharmacology , Skin Neoplasms/drug therapy , 9,10-Dimethyl-1,2-benzanthracene , Activating Transcription Factor 2/genetics , Activating Transcription Factor 2/metabolism , Animals , Apoptosis/drug effects , Carcinogens , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Line , Cell Proliferation/drug effects , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Cyclin-Dependent Kinase 4/genetics , Cyclin-Dependent Kinase 4/metabolism , Epidermis/drug effects , Epidermis/metabolism , Epidermis/pathology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/pathology , Female , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred DBA , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-fos/metabolism , Pyridines , Signal Transduction , Skin Neoplasms/chemically induced , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Thyroid Hormones/genetics , Thyroid Hormones/metabolism , Transcriptional Activation , Thyroid Hormone-Binding Proteins
10.
Environ Toxicol ; 30(2): 205-11, 2015 Feb.
Article in English | MEDLINE | ID: mdl-23836369

ABSTRACT

2-Aminobiphenyls (2-ABP) induces oxidative DNA damage and leads to apoptosis. The precise signaling pathways of inducing apoptosis in vitro are still unknown. This study provides insight into the relationship between 2-ABP-induced apoptosis and the activation of MAPK and downstream transcription factors using pharmacological inhibitors of ERK, p38, and JNK pathways. Results showed that 2-ABP induced the activation of ERK and JNK but not p38. The ERK/JNK pathways downstream transcription factors, c-Jun and ATF-2, were also activated by 2-ABP. The inhibitory effects of ERK inhibitor, U0126, on 2-ABP-induced caspase-3 activity were not detected. However, JNK inhibitor, SP600125, significantly attenuated the caspase-3 activity induced by 2-ABP. The expression of the transcription factors c-Jun and ATF-2 were decreased in 2-ABP treated cells in the presence of ERK/JNK inhibitors, suggesting that the expression of ERK/JNK pathways leads to the downstream activation of c-Jun and ATF-2. N-acetylcysteine, an ROS scavenger, inhibited 2-ABP-induced activation of ERK and JNK, the cell death and caspase-3 activity, which suggested that oxidative stress plays a crucial role in apoptosis through activation of caspase-3 in a ROS/JNK-dependent signaling cascade.


Subject(s)
Aminobiphenyl Compounds/toxicity , Apoptosis/drug effects , MAP Kinase Signaling System/drug effects , Transcription Factors/drug effects , Acetylcysteine/pharmacology , Activating Transcription Factor 2/antagonists & inhibitors , Activating Transcription Factor 2/biosynthesis , Caspase 3/metabolism , Cells, Cultured , DNA Damage , Humans , Phosphorylation , Proto-Oncogene Proteins c-jun/antagonists & inhibitors , Proto-Oncogene Proteins c-jun/biosynthesis , Reactive Oxygen Species/metabolism
11.
Biochem Cell Biol ; 92(1): 53-60, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24471918

ABSTRACT

Adiponectin plays a fundamental role in lipid and carbohydrate metabolism. However, its role in adipocyte differentiation remains controversial. To investigate the effect of gAd on lipid deposition in chicken adipocytes and its related signaling pathways, 200 µg/mL recombinant globular adiponectin, isoproterenol, SB253580, leucine, and rapamycin were used to treat chicken adipocytes. Results demonstrated that gAd increased the expression of endogenous adiponectin and AdipoR1 (P < 0.01); gAd inhibited triglyceride (TG) accumulation in chicken adipocyte and increased the release of free fatty acids (FFA) in medium; gAd decreased the expression of adipogenic marker genes CCAAT/enhancer binding protein alpha (C/EBPα) and fatty acid synthase (FAS), while activating the expression of lipolytic marker gene adipose triglyceride lipase (ATGL) (P < 0.01). Meanwhile, gAd activated the phosphorylation levels of p38 mitogen-activated protein kinase (p38 MAPK) and activating transcription factor 2 (ATF-2), and suppressed the phosphorylation levels of rapamycin (TOR) and p70 S6 Kinase (P < 0.01). In conclusion, the results demonstrate that gAd has an ability to inhibit lipids deposition in chicken adipocyte, which depends on the p38 MAPK/ATF-2 and TOR/p70 S6 Kinase pathways.


Subject(s)
Activating Transcription Factor 2/metabolism , Adipocytes/metabolism , Adiponectin/metabolism , Lipids/biosynthesis , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , TOR Serine-Threonine Kinases/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Activating Transcription Factor 2/antagonists & inhibitors , Adipocytes/cytology , Adipocytes/drug effects , Adiponectin/pharmacology , Animals , Cells, Cultured , Chickens , Phosphorylation , Recombinant Proteins/pharmacology , Ribosomal Protein S6 Kinases, 70-kDa/antagonists & inhibitors , Signal Transduction , TOR Serine-Threonine Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors
12.
PLoS One ; 8(11): e78253, 2013.
Article in English | MEDLINE | ID: mdl-24223142

ABSTRACT

The upstream Gγ-globin cAMP-response element (G-CRE) plays an important role in regulating Gγ-globin expression through binding of ATF2 and its DNA-binding partners defined in this study. ATF2 knockdown resulted in a significant reduction of γ-globin expression accompanied by decreased ATF2 binding to the G-CRE. By contrast, stable ATF2 expression in K562 cells increased γ-globin transcription which was reduced by ATF2 knockdown. Moreover, a similar effect of ATF2 on γ-globin expression was observed in primary erythroid progenitors. To understand the role of ATF2 in γ-globin expression, chromatographically purified G-CRE/ATF2-interacting proteins were subjected to mass spectrometry analysis; major binding partners included CREB1, cJun, Brg1, and histone deacetylases among others. Immunoprecipitation assays demonstrated interaction of these proteins with ATF2 and in vivo GCRE binding in CD34(+) cells undergoing erythroid differentiation which was correlated with γ-globin expression during development. These results suggest synergism between developmental stage-specific recruitments of the ATF2 protein complex and expression of γ-globin during erythropoiesis. Microarray studies in K562 cells support ATF2 plays diverse roles in hematopoiesis and chromatin remodeling.


Subject(s)
Activating Transcription Factor 2/genetics , Erythroid Cells/metabolism , Erythropoiesis/genetics , Gene Expression Regulation, Developmental , RNA, Messenger/genetics , gamma-Globins/genetics , Activating Transcription Factor 2/antagonists & inhibitors , Activating Transcription Factor 2/metabolism , Antigens, CD34/genetics , Antigens, CD34/metabolism , Binding Sites , Cell Differentiation , Cyclic AMP/metabolism , Cyclic AMP Response Element-Binding Protein/genetics , Cyclic AMP Response Element-Binding Protein/metabolism , DNA Helicases/genetics , DNA Helicases/metabolism , Erythroid Cells/cytology , Gene Expression Profiling , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Humans , JNK Mitogen-Activated Protein Kinases/genetics , JNK Mitogen-Activated Protein Kinases/metabolism , K562 Cells , Mass Spectrometry , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Promoter Regions, Genetic , Protein Binding , Protein Interaction Mapping , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction , Transcription Factors/genetics , Transcription Factors/metabolism , gamma-Globins/metabolism
13.
Cell Death Dis ; 2: e237, 2011 Dec 08.
Article in English | MEDLINE | ID: mdl-22158476

ABSTRACT

The IPC-81 cell line is derived from the transplantable BNML model of acute myelogenic leukemia (AML), known to be a reliable predictor of the clinical efficiency of antileukemic agents, like the first-line AML anthracycline drug daunorubicin (DNR). We show here that cAMP acted synergistically with DNR to induce IPC cell death. The DNR-induced death differed from that induced by cAMP by (1) not involving Bim induction, (2) being abrogated by GSK3ß inhibitors, (3) by being promoted by the HSP90/p23 antagonist geldanamycin and truncated p23 and (4) by being insensitive to the CRE binding protein (CREB) antagonist ICER and to cyclin-dependent protein kinase (CDK) inhibitors. In contrast, the apoptosis induced by cAMP correlated tightly with Bim protein expression. It was abrogated by Bim (BCL2L11) downregulation, whether achieved by the CREB antagonist ICER, by CDK inhibitors, by Bim-directed RNAi, or by protein synthesis inhibitor. The forced expression of BimL killed IPC-81(WT) cells rapidly, Bcl2-overexpressing cells being partially resistant. The pivotal role of CREB and CDK activity for Bim transcription is unprecedented. It is also noteworthy that newly developed cAMP analogs specifically activating PKA isozyme I (PKA-I) were able to induce IPC cell apoptosis. Our findings support the notion that AML cells may possess targetable death pathways not exploited by common anti-cancer agents.


Subject(s)
Activating Transcription Factor 2/metabolism , Apoptosis Regulatory Proteins/metabolism , Apoptosis , Cyclic AMP/metabolism , Cyclin-Dependent Kinases/metabolism , Membrane Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Transcription, Genetic , Activating Transcription Factor 2/antagonists & inhibitors , Animals , Antibiotics, Antineoplastic/pharmacology , Apoptosis/drug effects , Apoptosis Regulatory Proteins/antagonists & inhibitors , Apoptosis Regulatory Proteins/genetics , Bcl-2-Like Protein 11 , Benzoquinones/pharmacology , Cell Line, Tumor , Cyclic AMP/analogs & derivatives , Cyclic AMP/pharmacology , Cyclic AMP Response Element Modulator/metabolism , Cyclic AMP-Dependent Protein Kinase Type I/metabolism , Cyclic AMP-Dependent Protein Kinase Type II/metabolism , Cyclin-Dependent Kinases/antagonists & inhibitors , Cyclin-Dependent Kinases/physiology , Daunorubicin/pharmacology , Drug Synergism , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Lactams, Macrocyclic/pharmacology , Leukemia/physiopathology , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/genetics , RNA Interference , Rats
14.
FEBS Lett ; 582(19): 2833-7, 2008 Aug 20.
Article in English | MEDLINE | ID: mdl-18671972

ABSTRACT

Jun-dimerization protein 2 (JDP2) is a member of the activating protein-1 (AP-1) family of transcription factors. JDP2 dimerizes with other AP-1 proteins such as activating transcription factor-2 (ATF2) and Jun to repress transcription from promoters that contain a cyclic AMP-responsive element (CRE). Interferon regulatory factor-2-binding protein-1 (IRF2-BP1), which is reported to be a transcriptional corepressor of IRF2, was isolated as a JDP2-binding protein using an epitope-tagging method. As anticipated from the presence of a RING-finger domain, IRF2-BP1 enhanced the polyubiquitination of JDP2. Moreover, IRF2-BP1 repressed ATF2-mediated transcriptional activation from a CRE-containing promoter.


Subject(s)
Activating Transcription Factor 2/metabolism , Carrier Proteins/metabolism , Repressor Proteins/metabolism , Transcriptional Activation , Ubiquitin-Protein Ligases/metabolism , Ubiquitination , Activating Transcription Factor 2/antagonists & inhibitors , Amino Acid Sequence , Genes, Reporter , HeLa Cells , Humans , Luciferases/genetics , Molecular Sequence Data , Promoter Regions, Genetic , Transcription, Genetic
15.
Int Immunopharmacol ; 8(7): 974-81, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18486908

ABSTRACT

The objective of this study was to assess the effect of ursolic acid, a triterpene on inducing apoptosis in B16F-10 melanoma cells. Treatment of B16F-10 cells with nontoxic concentration of ursolic acid showed the presence of apoptotic bodies and induced DNA fragmentation in a dose depended manner. The apoptotic genes p53 and caspase-3 were found to be upregulated while the anti-apoptotic gene bcl-2 was down regulated in ursolic acid treated cells. The transcription factors NF-kappaBp65, NF-kappaBp50, NF-kappaBc-Rel, c-FOS, ATF-2 and CREB-1 were found to be inhibited significantly (p<0.001) in ursolic acid treated cells compared to untreated control. The pro-inflammatory cytokine production and gene expression of TNF-alpha, IL-1beta, IL-6 and GM-CSF were down regulated in ursolic acid treated cells compared to nontreated B16F-10 metastatic melanoma cells. All these results demonstrate that ursolic acid induce apoptosis via inhibition of NF-kappaB induced bcl-2 mediated anti-apoptotic pathway and subsequent activation of p53 mediated and TNF-alpha induced caspase-3 mediated pro-apoptotic pathways.


Subject(s)
Apoptosis/drug effects , Caspase 3/genetics , Gene Expression Regulation, Neoplastic/drug effects , Genes, bcl-2 , Genes, p53 , Melanoma, Experimental/drug therapy , NF-kappa B/antagonists & inhibitors , Triterpenes/pharmacology , Activating Transcription Factor 2/antagonists & inhibitors , Animals , Cell Line, Tumor , Cyclic AMP Response Element-Binding Protein/antagonists & inhibitors , Cytokines/biosynthesis , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Mice , Ursolic Acid
16.
Mol Carcinog ; 47(3): 172-83, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18095272

ABSTRACT

The cancer preventive action of (-)-epigallocatechin gallate (EGCG), found in green tea, is strongly supported by epidemiology and laboratory research data. However, the mechanism by which EGCG inhibits carcinogenesis and cell transformation is not clear. In this study, we report that EGCG suppressed epidermal growth factor (EGF)-induced cell transformation in JB6 cells. We also found that EGCG inhibited EGF-induced Fyn kinase activity and phosphorylation in vitro and in vivo. Fyn was implicated in the process because EGF-induced JB6 cell transformation was inhibited by small interfering RNA (siRNA)-Fyn-JB6 cells. With an in vitro protein-binding assay, we found that EGCG directly bound with the GST-Fyn-SH2 domain but not the GST-Fyn-SH3 domain. The K(d) value for EGCG binding to the Fyn SH2 domain was 0.367 +/- 0.122 microM and B(max) was 1.35 +/- 0.128 nmol/mg. Compared with control JB6 Cl41 cells, EGF-induced phosphorylation of p38 MAP kinase (p38 MAPK) (Thr180/Tyr182), ATF-2 (Thr71) and signal transducer and activator of transcription 1 (STAT1) (Thr727) was decreased in siRNA-Fyn-JB6 cells. EGCG could inhibit the phosphorylation of p38 MAPK, ATF-2, and STAT1. The DNA binding ability of AP-1, STAT1, and ATF-2 was also decreased in siRNA-Fyn-JB6 cells. Overall, these results demonstrated that EGCG interacted with Fyn and inhibited Fyn kinase activity and thereby regulated EGF-induced cell transformation. Inhibition of Fyn kinase activity is a novel and important mechanism that may be involved in EGCG-induced inhibition of cell transformation.


Subject(s)
Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Antioxidants/pharmacology , Catechin/analogs & derivatives , Cell Transformation, Neoplastic/drug effects , Activating Transcription Factor 2/antagonists & inhibitors , Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , CREB-Binding Protein/metabolism , Catechin/metabolism , Catechin/pharmacology , Cell Survival/drug effects , Cell Transformation, Neoplastic/chemically induced , Dose-Response Relationship, Drug , Epidermal Growth Factor/toxicity , Glutathione Transferase/metabolism , Kinetics , Mice , Phosphorylation/drug effects , Protein Binding , Protein Structure, Tertiary/drug effects , RNA, Small Interfering/pharmacology , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/isolation & purification , Recombinant Fusion Proteins/metabolism , STAT1 Transcription Factor/antagonists & inhibitors , Time Factors , Transcription Factor AP-1/metabolism , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors
17.
Antiviral Res ; 77(2): 153-6, 2008 Feb.
Article in English | MEDLINE | ID: mdl-17961732

ABSTRACT

Arenaviridae is a family of enveloped viruses some of which are capable of causing hemorrhagic fever syndromes in humans. In this report, we demonstrate that treatment of host cells with the tyrosine kinase inhibitor genistein inhibits infection of cells with the New World arenavirus Pichindé (PICV). The greatest degree of inhibition was observed in pre-treated target cells, but modest inhibition of infection was also seen when drug was added to cultures up to 48h after infection. We show that PICV-induced phosphorylation of the activating transcription factor-2 protein (ATF-2) and cyclic adenosine monophosphate response element binding protein (CREB) is inhibited following genistein treatment. Lastly, genistein treatment also inhibited transduction of cells with pseudotyped retrovirus particles expressing envelope proteins of the Old World arenavirus Lassa virus. These results demonstrate that kinase activity is required for arenavirus infection and that therapeutics designed to inhibit kinase activity should be explored.


Subject(s)
Genistein/pharmacology , Pichinde virus/drug effects , Protein Kinase Inhibitors/pharmacology , Activating Transcription Factor 2/antagonists & inhibitors , Activating Transcription Factor 2/metabolism , Animals , Cell Line , Chlorocebus aethiops , Cyclic AMP Response Element-Binding Protein/metabolism , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Lassa virus/drug effects , Nucleoproteins/metabolism , Phosphorylation/drug effects , Vero Cells , Virus Replication/drug effects
18.
Clin Cancer Res ; 13(22 Pt 1): 6769-78, 2007 Nov 15.
Article in English | MEDLINE | ID: mdl-18006779

ABSTRACT

PURPOSE: Sensitize melanomas to apoptosis and inhibit their growth and metastatic potential by compounds that mimic the activities of activating transcription factor 2 (ATF2)-driven peptides. EXPERIMENTAL DESIGN: Small-molecule chemical library consisting of 3,280 compounds was screened to identify compounds that elicit properties identified for ATF2 peptide, including (a) sensitization of melanoma cells to apoptosis, (b) inhibition of ATF2 transcriptional activity, (c) activation of c-Jun NH(2)-terminal kinase (JNK) and c-Jun transcriptional activity, and (d) inhibition of melanoma growth and metastasis in mouse models. RESULTS: Two compounds, celastrol (CSL) and acetyl isogambogic acid, could, within a low micromolar range, efficiently elicit cell death in melanoma cells. Both compounds efficiently inhibit ATF2 transcriptional activities, activate JNK, and increase c-Jun transcriptional activities. Similar to the ATF2 peptide, both compounds require JNK activity for their ability to inhibit melanoma cell viability. Derivatives of CSL were identified as potent inducers of cell death in mouse and human melanomas. CSL and a derivative (CA19) could also efficiently inhibit growth of human and mouse melanoma tumors and reduce the number of lung metastases in syngeneic and xenograft mouse models. CONCLUSIONS: These studies show for the first time the effect of CSL and acetyl isogambogic acid on melanoma. These compounds elicit activities that resemble the well-characterized ATF2 peptide and may therefore offer new approaches for the treatment of this tumor type.


Subject(s)
Chromones/pharmacology , Melanoma/drug therapy , Skin Neoplasms/drug therapy , Triterpenes/pharmacology , Activating Transcription Factor 2/antagonists & inhibitors , Animals , Cell Line, Tumor , Chromones/chemistry , Chromones/therapeutic use , Drug Evaluation, Preclinical , Drug Screening Assays, Antitumor , Humans , MAP Kinase Kinase 4/metabolism , Melanoma/metabolism , Melanoma/pathology , Mice , Pentacyclic Triterpenes , Proto-Oncogene Proteins c-jun/metabolism , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Small Molecule Libraries/chemistry , Transcription, Genetic , Triterpenes/chemistry , Triterpenes/therapeutic use
19.
Blood ; 110(5): 1570-7, 2007 Sep 01.
Article in English | MEDLINE | ID: mdl-17525285

ABSTRACT

Chronic inflammatory diseases often have residual CD8(+) T-cell infiltration despite treatment with systemic corticosteroids, which suggests divergent steroid responses between CD4(+) and CD8(+) cells. To examine steroid sensitivity, dexamethasone (DEX)-induced histone H4 lysine 5 (K5) acetylation and glucocorticoid receptor alpha (GCR alpha) translocation were evaluated. DEX treatment for 6 hours significantly induced histone H4 K5 acetylation in normal CD4(+) cells (P = .001) but not in CD8(+) cells. DEX responses were functionally impaired in CD8(+) compared with CD4(+) cells when using mitogen-activated protein kinase phosphatase (1 hour; P = .02) and interleukin 10 mRNA (24 hours; P = .004) induction as a readout of steroid-induced transactivation. Normal DEX-induced GCR alpha nuclear translocation and no significant difference in GCR alpha and GCR beta mRNA expression were observed in both T-cell types. In addition, no significant difference in SRC-1, p300, or TIP60 expression was found. However, activating transcription factor-2 (ATF2) expression was significantly lower in CD8(+) compared with CD4(+) cells (P = .009). Importantly, inhibition of ATF2 expression by small interfering RNA in CD4(+) cells resulted in inhibition of DEX-induced transactivation in CD4(+) cells. The data indicate refractory steroid-induced transactivation but similar steroid-induced transrepression of CD8(+) cells compared with CD4(+) cells caused by decreased levels of the histone acetyltransferase ATF2.


Subject(s)
Activating Transcription Factor 2/metabolism , Anti-Inflammatory Agents/pharmacology , CD8-Positive T-Lymphocytes/metabolism , Dexamethasone/pharmacology , Receptors, Glucocorticoid/metabolism , Transcriptional Activation/drug effects , Acetylation/drug effects , Activating Transcription Factor 2/antagonists & inhibitors , Activating Transcription Factor 2/immunology , Active Transport, Cell Nucleus/drug effects , Active Transport, Cell Nucleus/immunology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/immunology , Cell Cycle Proteins/biosynthesis , Cell Cycle Proteins/immunology , Cell Nucleus/immunology , Cell Nucleus/metabolism , Dual Specificity Phosphatase 1 , E1A-Associated p300 Protein/biosynthesis , E1A-Associated p300 Protein/immunology , Gene Expression Regulation/drug effects , Gene Expression Regulation/immunology , Histone Acetyltransferases/biosynthesis , Histone Acetyltransferases/immunology , Histone Acetyltransferases/metabolism , Histones/immunology , Histones/metabolism , Humans , Immediate-Early Proteins/biosynthesis , Immediate-Early Proteins/immunology , Interleukin-10/biosynthesis , Interleukin-10/immunology , Lysine Acetyltransferase 5 , Nuclear Receptor Coactivator 1 , Phosphoprotein Phosphatases/biosynthesis , Phosphoprotein Phosphatases/immunology , Protein Phosphatase 1 , Protein Tyrosine Phosphatases/biosynthesis , Protein Tyrosine Phosphatases/immunology , RNA, Messenger/biosynthesis , RNA, Messenger/immunology , RNA, Small Interfering/pharmacology , Receptors, Glucocorticoid/immunology , Time Factors , Transcription Factors/biosynthesis , Transcription Factors/immunology , Transcriptional Activation/immunology
20.
Blood ; 109(10): 4249-57, 2007 May 15.
Article in English | MEDLINE | ID: mdl-17244683

ABSTRACT

Absence of shear stress due to disturbed blood flow at arterial bifurcations and curvatures leads to endothelial dysfunction and proinflammatory gene expression, ultimately resulting in atherogenesis. KLF2 has recently been implicated as a transcription factor involved in mediating the anti-inflammatory effects of flow. We investigated the effect of shear on basal and TNF-alpha-induced genomewide expression profiles of human umbilical vein endothelial cells (HUVECs). Cluster analysis confirmed that shear stress induces expression of protective genes including KLF2, eNOS, and thrombomodulin, whereas basal expression of TNF-alpha-responsive genes was moderately decreased. Promoter analysis of these genes showed enrichment of binding sites for ATF transcription factors, whereas TNF-alpha-induced gene expression was mostly NF-kappaB dependent. Furthermore, human endothelial cells overlying atherosclerotic plaques had increased amounts of phosphorylated nuclear ATF2 compared with endothelium at unaffected sites. In HUVECs, a dramatic reduction of nuclear binding activity of ATF2 was observed under shear and appeared to be KLF2 dependent. Reduction of ATF2 with siRNA potently suppressed basal proinflammatory gene expression under no-flow conditions. In conclusion, we demonstrate that shear stress and KLF2 inhibit nuclear activity of ATF2, providing a potential mechanism by which endothelial cells exposed to laminar flow are protected from basal proinflammatory, atherogenic gene expression.


Subject(s)
Activating Transcription Factor 2/genetics , Atherosclerosis/genetics , Inflammation/genetics , Kruppel-Like Transcription Factors/physiology , Activating Transcription Factor 2/antagonists & inhibitors , Activating Transcription Factor 2/metabolism , Cell Nucleus/metabolism , Cells, Cultured , Cluster Analysis , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Gene Expression Regulation/drug effects , Humans , Phosphorylation , Protein Kinases/metabolism , RNA, Small Interfering/pharmacology , Stress, Mechanical , Time Factors , Transcription, Genetic , Tumor Necrosis Factor-alpha/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...