Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 54
Filter
1.
Proc Natl Acad Sci U S A ; 114(48): 12791-12796, 2017 11 28.
Article in English | MEDLINE | ID: mdl-29133416

ABSTRACT

Metastasis is a major cause of cancer-related deaths. Approximately 80% of patients with colorectal cancer develop liver metastasis and 20% develop lung metastasis. We found that at different stages of colon cancer, IFNγ secretion from peripheral blood mononuclear cells was decreased compared with healthy controls. The ribosomal S6 kinase (RSK) family of kinases has multiple cellular functions, and we examined their roles in this observed IFNγ decrease. Flow cytometry analysis of wild-type (WT) and RSK2 knockout (KO) mice revealed significantly lower levels of IFNγ in the RSK2 KO mice compared with the WT mice. Since IFNγ is a component of immunity, which contributes to protection against metastatic carcinomas, we conducted a colon cancer liver metastasis experiment. We found significantly greater metastasis in RSK2 KO mice compared with WT mice. Transcription factor T-bet can directly activate Ifnγ gene transcription. In vitro kinase assay results showed that RSK2 phosphorylated T-bet at serines 498 and 502. We show that phosphorylation of T-bet by RSK2 is required for IFNγ expression, because knockdown of RSK2 expression or overexpression of mutant T-bet reduces IFNγ mRNA expression. To verify the function of the phosphorylation sites, we overexpressed a constitutively active mutant T-bet (S498E/S502E) in bone marrow. Mutant T-bet restored the IFNγ mRNA levels and dramatically reduced the metastasis rate in these mice. Overall, these results indicate that phosphorylation of T-bet is required for the inhibition of colon cancer metastasis and growth through a positive regulation of RSK2/T-bet/IFNγ signaling.


Subject(s)
Colonic Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Interferon-gamma/genetics , Liver Neoplasms/genetics , Lung Neoplasms/genetics , Ribosomal Protein S6 Kinases/genetics , T-Box Domain Proteins/genetics , Animals , Bone Marrow Transplantation , Colonic Neoplasms/immunology , Colonic Neoplasms/pathology , Female , Humans , Interferon-gamma/immunology , Isoenzymes/genetics , Isoenzymes/immunology , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/pathology , Liver Neoplasms/immunology , Liver Neoplasms/prevention & control , Liver Neoplasms/secondary , Lung Neoplasms/immunology , Lung Neoplasms/prevention & control , Lung Neoplasms/secondary , Male , Mice , Phosphorylation , Ribosomal Protein S6 Kinases/immunology , Serine/metabolism , Signal Transduction , T-Box Domain Proteins/immunology , Transfection , Whole-Body Irradiation
2.
J Biol Chem ; 289(41): 28192-201, 2014 Oct 10.
Article in English | MEDLINE | ID: mdl-25122774

ABSTRACT

Non-small cell lung cancer (NSCLC) is the most lethal cancer, causing more than 150,000 deaths in the United States in 2013. The receptor tyrosine kinase inhibitors such as gefitinib are not perfect clinical therapeutic agents for NSCLC treatment due to primary or acquired tyrosine kinase inhibitor resistance. Herein, 3,6,2',4',5'-pentahydroxyflavone (36245-PHF) was identified as a multiple kinase inhibitor for NSCLC treatment based on the computational screening of a natural products database. 36245-PHF was shown to inhibit PI3K and Aurora A and B kinases and overcome gefitinib-resistant NSCLC growth. Our data clearly showed that 36245-PHF markedly inhibited anchorage-independent growth of gefitinib-resistant NSCLC cell lines and exerted a substantial chemotherapeutic effect following oral administration in a gefitinib-resistant NSCLC xenograft model. The evidence from three different subsequent methodological approaches, in vitro, ex vivo, and in vivo, all confirmed that 36245-PHF as a multiple protein kinase inhibitor. Overall, we identified 36245-PHF as a multiple protein kinase inhibitor and as a novel therapeutic agent to overcome gefitinib-resistant NSCLC growth, which could provide a new option for clinical NSCLC oral treatment.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , Flavonoids/pharmacology , Gene Expression Regulation, Neoplastic , Lung Neoplasms/drug therapy , Protein Kinase Inhibitors/pharmacology , Quinazolines/pharmacology , Animals , Aurora Kinase A/antagonists & inhibitors , Aurora Kinase A/genetics , Aurora Kinase A/metabolism , Aurora Kinase B/antagonists & inhibitors , Aurora Kinase B/genetics , Aurora Kinase B/metabolism , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Adhesion , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Resistance, Neoplasm/drug effects , Female , Gefitinib , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mice , Mice, Nude , Neoplasm Transplantation , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Signal Transduction , Tumor Burden/drug effects
3.
J Biol Chem ; 287(31): 25881-92, 2012 Jul 27.
Article in English | MEDLINE | ID: mdl-22685297

ABSTRACT

TRAF2 has an important function in mediating the TNF-R signaling pathway toward activation of NF-κB and JNKs. Here we reveal a novel function of TRAF2 in the epidermal growth factor (EGF) signaling pathway. Knockdown of TRAF2 blocked EGF-induced AP-1 activity and anchorage- independent cell transformation. Notably, we showed that EGF induces ribosomal S6 kinase 2 (RSK2) ubiquitination, and knocking down TRAF2 suppresses ubiquitination of RSK2 induced by EGF. We also found that TRAF2 affects RSK2 activity through RSK2 ubiquitination. RSK2 plays a critical role in AP-1 activity mediated through CREB and c-Fos, which regulates anchorage-independent cell transformation. In addition, TRAF2 is overexpressed in colon cancer and required for colon cancer development, suggesting that TRAF2 might be a potential molecular target for cancer prevention and treatment.


Subject(s)
Cyclic AMP Response Element-Binding Protein/metabolism , Epidermal Growth Factor/physiology , Proto-Oncogene Proteins c-fos/metabolism , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , TNF Receptor-Associated Factor 2/metabolism , Animals , Cell Line, Tumor , Cell Proliferation , Cell Transformation, Neoplastic/metabolism , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Female , Gene Knockdown Techniques , Humans , Mice , Mice, Nude , Neoplasm Transplantation , Polyubiquitin/metabolism , Protein Binding , Protein Processing, Post-Translational , Signal Transduction , TNF Receptor-Associated Factor 2/genetics , Transcription Factor AP-1/metabolism , Transcription, Genetic , Tumor Burden , Ubiquitination
4.
J Biol Chem ; 286(9): 6946-54, 2011 Mar 04.
Article in English | MEDLINE | ID: mdl-21183680

ABSTRACT

The ribosomal S6 kinase 2 (RSK2) is a member of the p90 ribosomal S6 kinase (p90RSK) family of proteins and plays a critical role in proliferation, cell cycle, and cell transformation. Here, we report that RSK2 phosphorylates caspase-8, and Thr-263 was identified as a novel caspase-8 phosphorylation site. In addition, we showed that EGF induces caspase-8 ubiquitination and degradation through the proteasome pathway, and phosphorylation of Thr-263 is associated with caspase-8 stability. Finally, RSK2 blocks Fas-induced apoptosis through its phosphorylation of caspase-8. These data provide a direct link between RSK2 and caspase-8 and identify a novel molecular mechanism for caspase-8 modulation by RSK2.


Subject(s)
Apoptosis/physiology , Caspase 8/metabolism , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Signal Transduction/physiology , Ubiquitination/physiology , Cell Division/physiology , HEK293 Cells , HeLa Cells , Humans , Phosphorylation/physiology , Protein Stability , Threonine/metabolism
5.
Carcinogenesis ; 32(5): 779-85, 2011 May.
Article in English | MEDLINE | ID: mdl-21349818

ABSTRACT

In addition to capsaicin, a transient receptor potential channel vanilloid subfamily 1 (TRPV1) agonist, two kinds of antagonists against this receptor are used as therapeutic drugs for pain relief. Indeed, a number of small molecule TRPV1 antagonists are currently undergoing Phase I/II clinical trials to determine their effect on relieving chronic inflammatory pain and migraine headache pain. However, we previously reported that the absence of TRPV1 in mice results in a striking increase in skin carcinogenesis, suggesting that chronic blockade of TRPV1 might increase the risk of tumor development. In this study, we found that a typical TRPV1 antagonist, AMG9810, promotes mouse skin tumor development. The topical application of AMG9810 resulted in a significant increase in the expression level of the epidermal growth factor receptor (EGFR) and its downstream Akt/mammalian target of rapamycin (mTOR)-signaling pathway. This increase was not only observed in AMG9810-treated tumor tissue but was also found in skin tissue treated with AMG9810. In telomerase-immortalized primary human keratinocytes, AMG9810 promoted proliferation that was mediated through the EGFR/Akt/mTOR-signaling pathway. In summary, our data suggest that the TRPV1 antagonist, AMG9810, promotes mouse skin tumorigenesis mediated through EGFR/Akt/mTOR signaling. Thus, the application of this compound for pain relief might increase the risk of skin cancer.


Subject(s)
Acrylamides/toxicity , Bridged Bicyclo Compounds, Heterocyclic/toxicity , ErbB Receptors/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Skin Neoplasms/chemically induced , Skin Neoplasms/pathology , TRPV Cation Channels/antagonists & inhibitors , 9,10-Dimethyl-1,2-benzanthracene/toxicity , Acrylamides/administration & dosage , Animals , Blotting, Western , Bridged Bicyclo Compounds, Heterocyclic/administration & dosage , Carcinogens/toxicity , Cell Proliferation , Cell Transformation, Neoplastic/drug effects , Cells, Cultured , Cocarcinogenesis , Female , Humans , Keratinocytes/cytology , Keratinocytes/drug effects , Keratinocytes/metabolism , Male , Mice , Mice, Hairless , Signal Transduction/drug effects , Skin Neoplasms/metabolism , TRPV Cation Channels/metabolism
6.
J Biol Chem ; 285(50): 39108-16, 2010 Dec 10.
Article in English | MEDLINE | ID: mdl-20921231

ABSTRACT

MST1 (mammalian STE20-like kinase 1) is a serine/threonine kinase that is cleaved and activated by caspases during apoptosis. Overexpression of MST1 induces apoptotic morphological changes such as chromatin condensation, but the mechanism is not clear. Here we show that MST1 induces apoptotic chromatin condensation through its phosphorylation of histone H2AX at Ser-139. During etoposide-induced apoptosis in Jurkat cells, the cleavage of MST1 directly corresponded with strong H2AX phosphorylation. In vitro kinase assay results showed that MST1 strongly phosphorylates histone H2AX. Western blot and kinase assay results with a mutant S139A H2AX confirmed that MST1 phosphorylates H2AX at Ser-139. Direct binding of MST1 and H2AX can be detected when co-expressed in HEK293 cells and was also confirmed by an endogenous immunoprecipitation study. When overexpressed in HeLa cells, both the MST1 full-length protein and the MST1 kinase domain (MST1-NT), but not the kinase-negative mutant (MST1-NT-KN), could induce obvious endogenous histone H2AX phosphorylation. The caspase-3 inhibitor benzyloxycarbonyl-DEVD-fluoromethyl ketone (Z-DEVD-fmk) attenuates phosphorylation of H2AX by MST1 but cannot inhibit MST1-NT-induced histone H2AX phosphorylation, indicating that cleaved MST1 is responsible for H2AX phosphorylation during apoptosis. Histone H2AX phosphorylation and DNA fragmentation were suppressed in MST1 knockdown Jurkat cells after etoposide treatment. Taken together, our data indicated that H2AX is a substrate of MST1, which functions to induce apoptotic chromatin condensation and DNA fragmentation.


Subject(s)
Gene Expression Regulation, Neoplastic , Hepatocyte Growth Factor/metabolism , Histones/metabolism , Proto-Oncogene Proteins/metabolism , Apoptosis , Chromatin/chemistry , DNA Fragmentation , Etoposide/pharmacology , HEK293 Cells , HeLa Cells , Histones/chemistry , Humans , Jurkat Cells , Phosphorylation , Serine/chemistry
7.
FASEB J ; 24(9): 3490-9, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20385620

ABSTRACT

The ribosomal S6 kinase 2 (RSK2) is a well-known serine/threonine kinase and a member of the p90 ribosomal S6 kinase (p90RSK) family of proteins. It is activated downstream of the MEK/ERKs cascade by mitogenic stimuli such as EGF or TPA. Here, we show that RSK2 is activated by treatment with tumor necrosis factor-alpha (TNF-alpha) and directly phosphorylates IkappaBalpha at Ser-32, leading to IkappaBalpha degradation. The phosphorylation of IkappaBalpha promotes the activation and translocation of the nuclear factor-kappaB (NF-kappaB) subunits p65 and p50 to the nucleus. The net result is an increased NF-kappaB activity, which serves as a mechanism for RSK2 blockade of TNF-alpha-induced apoptosis and enhanced cell survival.


Subject(s)
I-kappa B Proteins/metabolism , NF-kappa B/metabolism , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Animals , Caspase 3/metabolism , Caspase 8/metabolism , Cell Line , Cell Line, Tumor , Electrophoretic Mobility Shift Assay , HeLa Cells , Humans , Immunoblotting , Immunoprecipitation , Mice , NF-KappaB Inhibitor alpha , NF-kappa B p50 Subunit/metabolism , Phosphorylation/drug effects , Protein Binding , Protein Transport/drug effects , Transcription Factor RelA/metabolism
8.
Nat Struct Mol Biol ; 12(8): 699-707, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16007099

ABSTRACT

Inactivation of the p16(INK4a) tumor suppressor protein is critical for the development of human cancers, including human melanoma. However, the molecular basis of the protein's inhibitory effect on cancer development is not clear. Here we investigated a possible mechanism for p16(INK4a) inhibition of neoplastic transformation and UV-induced skin cancer. We show that p16(INK4a) suppresses the activity of c-Jun N-terminal kinases (JNKs) and that it binds to the glycine-rich loop of the N-terminal domain of JNK3. Although p16(INK4a) does not affect the phosphorylation of JNKs, its interaction with JNK inhibits c-Jun phosphorylation induced by UV exposure. This, in turn, interferes with cell transformation promoted by the H-Ras-JNK-c-Jun-AP-1 signaling axis.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Melanoma/metabolism , Mitogen-Activated Protein Kinase 10/metabolism , Signal Transduction/physiology , Skin Neoplasms/metabolism , Transcription Factor AP-1/metabolism , Animals , Cell Line, Tumor , Fluorescent Antibody Technique , Genetic Vectors , Glutathione Transferase , Humans , Immunoblotting , Mice , Models, Molecular , Phosphorylation , Protein Binding , Two-Hybrid System Techniques
9.
Mol Cancer Res ; 18(7): 1028-1038, 2020 07.
Article in English | MEDLINE | ID: mdl-32269074

ABSTRACT

The identification of oncogenic biomolecules as drug targets is an unmet need for the development of clinically effective novel anticancer therapies. In this study, we report for the first time that opsin 4/melanopsin (OPN4) plays a critical role in the pathogenesis of non-small cell lung cancer (NSCLC) and is a potential drug target. Our study has revealed that OPN4 is overexpressed in human lung cancer tissues and cells, and is inversely correlated with patient survival probability. Knocking down expression of OPN4 suppressed cells growth and induced apoptosis in lung cancer cells. We have also found that OPN4, a G protein-coupled receptor, interacted with Gα11 and triggered the PKC/BRAF/MEK/ERKs signaling pathway in lung adenocarcinoma cells. Genetic ablation of OPN4 attenuated the multiplicity and the volume of urethane-induced lung tumors in mice. Importantly, our study provides the first report of AE 51310 (1-[(2,5-dichloro-4-methoxyphenyl)sulfonyl]-3-methylpiperidine) as a small-molecule inhibitor of OPN4, suppressed the anchorage-independent growth of lung cancer cells and the growth of patient-derived xenograft tumors in mice. IMPLICATIONS: Overall, this study unveils the role of OPN4 in NSCLC and suggests that targeting OPN4 with small molecules, such as AE 51310 would be interesting to develop novel anticancer therapies for lung adenocarcinoma.


Subject(s)
Adenocarcinoma of Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/drug therapy , MAP Kinase Signaling System/drug effects , Rod Opsins/metabolism , Small Molecule Libraries/administration & dosage , Up-Regulation/drug effects , A549 Cells , Adenocarcinoma of Lung/chemically induced , Adenocarcinoma of Lung/metabolism , Animals , Carcinoma, Non-Small-Cell Lung/chemically induced , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Disease Progression , Gene Expression Regulation, Neoplastic/drug effects , Humans , Lung Neoplasms/chemically induced , Lung Neoplasms/metabolism , Mice , Small Molecule Libraries/pharmacology , Urethane/adverse effects , Xenograft Model Antitumor Assays
10.
Theranostics ; 10(21): 9721-9740, 2020.
Article in English | MEDLINE | ID: mdl-32863956

ABSTRACT

Rationale: Melanoma is an aggressive tumor of the skin and drug resistance is still a major problem in melanoma therapy. Novel targets and effective agents to overcome drug resistant melanoma are urgently needed in clinical therapy. Methods: Gene Expression Omnibus (GEO) database analysis, pathway enrichment analysis, and survival rate analysis were utilized to identify a candidate target. An anchorage-independent cell growth assay, flow cytometry, Western blot, and a xenograft mouse model were used to study the function of Aurora kinase B (AURKB) in both drug-sensitive and drug-resistant melanoma. Next, HI-511, a novel dual-target inhibitor targeting both AURKB and BRAF V600E, was designed and examined by an in vitro kinase assay. Methods as indicated above in addition to a BRAF V600E/PTEN-loss melanoma mouse model were used to demonstrate the effect of HI-511 on melanoma development in vitro and in vivo. Results: AURKB is highly expressed in melanoma and especially in vemurafenib-resistant melanoma and the expression was correlated with patient survival rate. Knocking down AURKB inhibited cell growth and induced apoptosis in melanoma, which was associated with the BRAF/MEK/ERKs and PI3-K/AKT signaling pathways. Importantly, we found that HI-511, a novel dual-target inhibitor against AURKB and BRAF V600E, suppresses both vemurafenib-sensitive and vemurafenib-resistant melanoma growth in vitro and in vivo by inducing apoptosis and mediating the inhibition of the BRAF/MEK/ERKs and PI3K/AKT signaling pathways. Conclusion: AURKB is a potential target for melanoma treatment. HI-511, a novel dual-target inhibitor against both AURKB and BRAF V600E, could achieve durable suppression of melanoma growth, even drug-resistant melanoma growth.


Subject(s)
Aurora Kinase B/metabolism , Drug Resistance, Neoplasm/drug effects , Melanoma/drug therapy , Melanoma/metabolism , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins B-raf/metabolism , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , MAP Kinase Signaling System/drug effects , Mice , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction/drug effects , Vemurafenib/pharmacology
11.
Cancer Res ; 80(19): 4158-4171, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32816906

ABSTRACT

The key functional molecules involved in inflammatory bowel disease (IBD) and IBD-induced colorectal tumorigenesis remain unclear. In this study, we found that the apoptosis repressor with caspase recruitment domain (ARC) protein plays critical roles in IBD. ARC-deficient mice exhibited substantially higher susceptibility to dextran sulfate sodium (DSS)-induced IBD compared with wild-type mice. The inflammatory burden induced in ARC-deficient conditions was inversely correlated with CCL5 and CXCL5 levels in immune cells, especially CD4-positive T cells. Pathologically, ARC expression in immune cells was significantly decreased in clinical biopsy specimens from patients with IBD compared with normal subjects. In addition, ARC levels inversely correlated with CCL5 and CXCL5 levels in human biopsy specimens. ARC interacted with TNF receptor associated factor (TRAF) 6, regulating ubiquitination of TRAF6, which was associated with NF-κB signaling. Importantly, we identified a novel ubiquitination site at lysine 461, which was critical in the function of ARC in IBD. ARC played a critical role in IBD and IBD-associated colon cancer in a bone marrow transplantation model and azoxymethane/DSS-induced colitis cancer mouse models. Overall, these findings reveal that ARC is critically involved in the maintenance of intestinal homeostasis and protection against IBD through its ubiquitination of TRAF6 and subsequent modulation of NF-κB activation in T cells. SIGNIFICANCE: This study uncovers a crucial role of ARC in the immune system and IBD, giving rise to a novel strategy for IBD and IBD-associated colon cancer therapy.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Colorectal Neoplasms/etiology , Inflammatory Bowel Diseases/metabolism , Inflammatory Bowel Diseases/pathology , Muscle Proteins/metabolism , Animals , Apoptosis Regulatory Proteins/chemistry , Apoptosis Regulatory Proteins/genetics , Azoxymethane/toxicity , Bone Marrow Transplantation , CD4-Positive T-Lymphocytes/immunology , Chemokine CCL5/metabolism , Chemokine CXCL5/metabolism , Colitis/chemically induced , Colorectal Neoplasms/chemically induced , Dextran Sulfate/toxicity , Disease Models, Animal , Female , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Jurkat Cells , Male , Mice, Inbred C57BL , Mice, Knockout , Muscle Proteins/chemistry , Muscle Proteins/genetics , Ubiquitination
12.
Cancer Res ; 67(18): 8725-35, 2007 Sep 15.
Article in English | MEDLINE | ID: mdl-17875713

ABSTRACT

The c-jun-NH(2)-kinases (JNK) play a critical role in tumor promoter-induced cell transformation and apoptosis. Here, we showed that the nuclear factor of activated T3 (NFAT3) is phosphorylated by JNK1 or JNK2 at Ser(213) and Ser(217), which are located in the conserved SP motif. The transactivation domain of NFAT3 is found between amino acids (aa) 113 and 260 and includes the phosphorylation targets of JNK1 and JNK2. NFAT3 transactivation activity was suppressed in JNK1(-/-) or JNK2(-/-) mouse embryonic fibroblast (MEF) cells compared with wild-type MEF cells. Moreover, a 3xNFAT-luc reporter gene assay indicated that NFAT3 transcriptional activity was increased in a dose-dependent manner by JNK1 or JNK2. Double mutations at Ser(213) and Ser(217) suppressed NFAT3 transactivation activity; and SP600125, a JNK inhibitor, suppressed NFAT3-induced 3xNFAT-luciferase activity. Knockdown of JNK1 or JNK2 suppressed foci formation in NIH3T3 cells. Importantly, ectopic expression of NFAT3 inhibited AP-1 activity and suppressed foci formation. Furthermore, knockdown of NFAT3 enhanced Ras-JNK1 or JNK2-induced foci formation in NIH3T3 cells. Taken together, these results provided direct evidence for the anti-oncogenic potential of the NFAT3 transcription factor.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Mitogen-Activated Protein Kinase 8/metabolism , Mitogen-Activated Protein Kinase 9/metabolism , NFATC Transcription Factors/metabolism , Transcription Factor AP-1/metabolism , Animals , Cell Line , Fibroblasts/cytology , Fibroblasts/metabolism , Genes, Reporter , Humans , Mice , NFATC Transcription Factors/biosynthesis , NFATC Transcription Factors/genetics , NIH 3T3 Cells , Phosphorylation , Serine/metabolism , Signal Transduction , Transcription Factor AP-1/antagonists & inhibitors , Transcriptional Activation
13.
J Atten Disord ; 23(4): 384-397, 2019 02.
Article in English | MEDLINE | ID: mdl-25646023

ABSTRACT

OBJECTIVE: This study examined the effect of a 12-week table tennis exercise on motor skills and executive functions in children with ADHD. METHOD: Fifteen children with ADHD received the intervention, whereas 15 children with ADHD and 30 typically developing children did not. The Test of Gross Motor Development-2, Stroop, and Wisconsin Card Sorting Test (WCST) were conducted before and after the intervention. RESULTS: After the intervention, the ADHD training group scored significantly higher in the locomotor as well as object-control skills, Stroop Color-Word condition, and WCST total correct performance compared with the ADHD non-training group, and we noted improvements in the locomotor as well as object-control skills, Stroop Color-Word condition, and three aspects of the WCST performances of the ADHD training group over time. CONCLUSION: A 12-week table tennis exercise may have clinical relevance in motor skills and executive functions of children with ADHD.


Subject(s)
Attention Deficit Disorder with Hyperactivity/therapy , Exercise Therapy/methods , Motor Skills/physiology , Child , Child Development/physiology , Executive Function/physiology , Exercise/physiology , Female , Humans , Male , Pilot Projects , Wisconsin Card Sorting Test
14.
EBioMedicine ; 49: 145-156, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31707149

ABSTRACT

BACKGROUND: Barrett's esophagus (BE), a complication of gastroesophageal reflux disease (GERD), predisposes patients to esophageal adenocarcinoma (EAC). Reliable biomarkers for early detection and discovery of potential drug targets are urgently needed for improved BE and EAC patient outcomes. METHODS: Patient biopsy samples were evaluated for COX1/2, and thromboxane A2 synthase (TBXAS) expression. Circulating prostaglandins biosynthesis was determined using enzyme immunoassay kits. Anchorage-independent cell growth assay, crystal violet staining assay, and xenograft experiments were conducted to assess BE and EAC cell growth. A surgical mouse model of reflux (i.e., esophagoduodenostomy) was established and samples were analyzed using an enzyme immunoassay kit, immunohistochemistry, immunoblotting, or RT-PCR. Esophageal biopsy samples (pre- and post-intervention) were obtained from a randomized clinical trial in which participants were administered esomeprazole (40 mg) twice daily in combination with an acetylsalicylic acid (ASA) placebo or 81 or 325 mg ASA for 28 days. Esophageal biopsy specimens before and after the intervention period were analyzed. FINDINGS: COX2 and TBXAS are highly expressed in BE and EAC patients accompanied by a pronounced elevation of circulating TXA2 levels. ASA suppressed BE and EAC growth by targeting the TXA2 pathway. Additionally, biopsies from 49 patients (with similar baseline characteristics) showed that ASA substantially decreased serum TXA2 levels, resulting in reduced inflammation. INTERPRETATION: This study establishes the importance of the COX1/2-driven TXA2 pathway in BE and EAC pathophysiology and lays the groundwork for introducing a TXA2-targeting strategy for EAC prevention and early detection. FUNDING: Hormel Foundation, Exact Sciences, Pentax Medical, Intromedic and National Cancer.


Subject(s)
Adenocarcinoma/drug therapy , Barrett Esophagus/drug therapy , Carcinogenesis/pathology , Cyclooxygenase 1/metabolism , Cyclooxygenase 2/metabolism , Esophageal Neoplasms/drug therapy , Molecular Targeted Therapy , Signal Transduction , Thromboxane A2/metabolism , Adenocarcinoma/blood , Animals , Aspirin/pharmacology , Barrett Esophagus/blood , Carcinogenesis/metabolism , Cell Line, Tumor , Cell Proliferation , Disease Models, Animal , Esophageal Neoplasms/blood , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Humans , Inflammation/pathology , Male , Mice, Inbred C57BL , Middle Aged , STAT3 Transcription Factor/metabolism , Thromboxane A2/blood
15.
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
16.
Mol Carcinog ; 47(10): 797-805, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18381589

ABSTRACT

Targeted molecular cancer therapies can potentially deliver treatment directly to a specific protein or gene to optimize efficacy and reduce adverse side effects often associated with traditional chemotherapy. Key oncoprotein and oncogene targets are rapidly being identified based on their expression, pathogenesis and clinical outcome. One such protein target is cyclooxygenase-2 (COX-2), which is highly expressed in various cancers. Research findings suggest that resveratrol (RSVL; 3,5,4'-trihydroxy-trans-stilbene) demonstrates nonselective COX-2 inhibition. We report herein that RSVL directly binds with COX-2 and this binding is absolutely required for RSVL's inhibition of the ability of human colon adenocarcinoma HT-29 cells to form colonies in soft agar. Binding of COX-2 with RSVL was compared with two RSVL analogues, 3,3',4',5',5-pentahydroxy-trans-stilbene (RSVL-2) or 3,4',5-trimethoxy-trans-stilbene (RSVL-3). The results indicated that COX-2 binds with RSVL-2 more strongly than with RSVL, but does not bind with RSVL-3. RSVL or RSVL-2, but not RSVL-3, inhibited COX-2-mediated PGE(2) production in vitro and ex vivo. HT-29 human colon adenocarcinoma cells express high levels of COX-2 and either RSVL or RSVL-2, but not RSVL-3, suppressed anchorage independent growth of these cells in soft agar. RSVL or RSVL-2 (not RSVL-3) suppressed growth of COX-2(+/+) cells by 60% or 80%, respectively. Notably, cells deficient in COX-2 were unresponsive to RSVL or RSVL-2. These data suggest that the anticancer effects of RSVL or RSLV-2 might be mediated directly through COX-2.


Subject(s)
Cell Transformation, Neoplastic/drug effects , Cyclooxygenase 2/drug effects , Stilbenes/pharmacology , Cell Line, Tumor , Chromatography, Affinity , Humans , Resveratrol , Spectrometry, Fluorescence
17.
Mol Biol Cell ; 16(9): 4437-53, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16000378

ABSTRACT

The ocular lens is the only organ that does not develop spontaneous tumor. The molecular mechanism for this phenomenon remains unknown. Through examination of the signaling pathways mediating stress-induced apoptosis, here we presented evidence to show that different from most other tissues in which the extracellular signal-regulated kinases (ERKs) pathway is generally implicated in mediation of survival signals activated by different factors, the RAF/MEK/ERK signaling pathway alone plays a key role in stress-activated apoptosis of lens epithelial cells. Treatment of N/N1003A cells with calcimycin, a calcium mobilizer, activates the RAF/MEK/ERK pathway through RAS, which is indispensable for the induced apoptosis because inhibition of this pathway by either pharmacological drug or dominant negative mutants greatly attenuates the induced apoptosis. Calcimycin also activates p38 kinase and JNK2, which are not involved in calcium-induced apoptosis. Downstream of ERK activation, p53 is essential. Activation of RAF/MEK/ERK pathway by calcimycin leads to distinct up-regulation of p53. Moreover, overexpression of p53 enhances calcimycin-induced apoptosis, whereas inhibition of p53 expression attenuates calcimycin-induced apoptosis. Up-regulation of p53 directly promotes Bax expression, which changes the integrity of mitochondria, leading to release of cytochrome c, activation of caspase-3 and eventually execution of apoptosis. Overexpression of alphaB-crystallin, a member of the small heat-shock protein family, blocks activation of RAS to inhibit ERK1/2 activation, and greatly attenuates calcimycin-induced apoptosis. Together, our results provide 1) a partial explanation for the lack of spontaneous tumor in the lens, 2) a novel signaling pathway for calcium-induced apoptosis, and 3) a novel antiapoptotic mechanism for alphaB-crystallin.


Subject(s)
Apoptosis/physiology , Calcium Signaling/physiology , Extracellular Signal-Regulated MAP Kinases/physiology , Tumor Suppressor Protein p53/physiology , alpha-Crystallin B Chain/physiology , raf Kinases/physiology , ras Proteins/physiology , Animals , Apoptosis/drug effects , Butadienes/pharmacology , Calcimycin/pharmacology , Caspase 3 , Caspase Inhibitors , Cell Line , Cytochromes c/antagonists & inhibitors , MAP Kinase Kinase Kinase 1/metabolism , MAP Kinase Kinase Kinase 2/metabolism , Nitriles/pharmacology , Rabbits
18.
Cancer Res ; 66(11): 5729-36, 2006 Jun 01.
Article in English | MEDLINE | ID: mdl-16740711

ABSTRACT

The oncoprotein c-Jun is a component of the activator protein-1 transcription factor complex, which is involved in cellular proliferation, transformation, and death. The stabilization of c-Jun is critically important for its function. The phosphorylation of c-Jun by c-Jun NH(2)-terminal kinase 1 and extracellular signal-regulated protein kinases reduces c-Jun ubiquitination resulting in increased stabilization of c-Jun. In this report, we showed that COOH-terminal Src kinase (CSK) binds with and phosphorylates c-Jun at Y26 and Y170. Phosphorylation of c-Jun by CSK, in opposition to c-Jun NH(2)-terminal kinase 1 and extracellular signal-regulated protein kinases, promoted c-Jun degradation and reduced stability. By promoting c-Jun degradation, CSK helps to maintain a low steady-state level of c-Jun, thereby inhibiting activator protein-1 activity and cell transformation caused by c-Jun. These results indicated that this function of CSK controls cell proliferation under normal growth conditions and may have implications for CSK loss of function in carcinogenesis.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Proto-Oncogene Proteins c-jun/metabolism , src-Family Kinases/metabolism , Amino Acid Sequence , Animals , Cell Line, Tumor , HeLa Cells , Humans , Mice , Mitogen-Activated Protein Kinase 8/metabolism , Molecular Sequence Data , NIH 3T3 Cells , Phosphorylation , Protein Binding , Transcription Factor AP-1/metabolism , Ubiquitin/metabolism
19.
Cancer Res ; 66(18): 9260-9, 2006 Sep 15.
Article in English | MEDLINE | ID: mdl-16982771

ABSTRACT

Many beneficial properties have been attributed to (-)-epigallocatechin gallate (EGCG), including chemopreventive, anticarcinogenic, and antioxidant actions. In this study, we investigated the effects of EGCG on the function of glucose-regulated protein 78 (GRP78), which is associated with the multidrug resistance phenotype of many types of cancer cells. Our investigation was directed at elucidating the mechanism of the EGCG and GRP78 interaction and providing evidence about whether EGCG modulates the activity of anticancer drugs through the inhibition of GRP78 function. We found that EGCG directly interacted with GRP78 at the ATP-binding site of protein and regulated its function by competing with ATP binding, resulting in the inhibition of ATPase activity. EGCG binding caused the conversion of GRP78 from its active monomer to the inactive dimer and oligomer forms. Further, we showed that EGCG interfered with the formation of the antiapoptotic GRP78-caspase-7 complex, which resulted in an increased etoposide-induced apoptosis in cancer cells. We also showed that EGCG significantly suppressed the transformed phenotype of breast cancer cells treated with etoposide. Overall, these results strongly suggested that EGCG could prevent the antiapoptotic effect of GRP78, which usually suppresses the caspase-mediated cell death pathways in drug-treated cancer cells, contributing to the development of drug resistance.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Catechin/analogs & derivatives , Etoposide/pharmacology , Heat-Shock Proteins/antagonists & inhibitors , Molecular Chaperones/antagonists & inhibitors , Adenosine Triphosphatases/antagonists & inhibitors , Animals , Apoptosis/drug effects , Apoptosis/physiology , Breast Neoplasms/drug therapy , Caspase 7/metabolism , Caspase Inhibitors , Catechin/metabolism , Catechin/pharmacology , Cell Line, Tumor , Drug Resistance, Neoplasm , Drug Synergism , Endoplasmic Reticulum Chaperone BiP , Heat-Shock Proteins/metabolism , Humans , Mice , Molecular Chaperones/metabolism , Protein Conformation/drug effects , Urinary Bladder Neoplasms/drug therapy , Urinary Bladder Neoplasms/metabolism
20.
Oncogene ; 37(26): 3501-3513, 2018 06.
Article in English | MEDLINE | ID: mdl-29563609

ABSTRACT

Inflammation is a complex biological host reaction to tissue damage, infection and trauma. Extensive study of the inflammatory response has led to the identification of several protein kinases that are essential for signaling and could be potential therapeutic targets. The RSK family of kinases has multiple cellular functions. In our study, we found that RSK2 is a mediator for inflammation signaling and interacts with TRAF6. In vitro kinase assay results indicated that RSK2 strongly phosphorylates TRAF6 at serines 46, 47 and 48. Ectopic overexpression of TRAF6 or knocking down RSK2 expression confirmed that RSK2 is a positive regulator of TRAF6 K63 ubiquitination. TRAF6 is also required for RSK2 ubiquitination. TRAF6 bridges the TNF receptor superfamily and intracellular signaling for the induction of proinflammatory cytokines. We developed a colon inflammation model using RSK2 wild type (WT) and knockout (KO) mice. As expected, F4/80 and CD3 infiltration were significantly upregulated in WT mice compared to RSK2 KO mice. Furthermore, inflammation signaling, including Ikkα/ß, p38 and JNKs, was dramatically upregulated in WT mice. Colon tissue immunoprecipitation results further confirmed that TRAF6 K63 ubiquitination was lower in RSK2 KO mice. Overall, these results indicate that phosphorylation of TRAF6 (S46, 47, 48) by RSK2 is required for TRAF6 K63 ubiquitination and inflammation signaling.


Subject(s)
Colitis/pathology , Colon/pathology , Ribosomal Protein S6 Kinases, 90-kDa/genetics , TNF Receptor-Associated Factor 6/metabolism , Animals , Antigens, Differentiation/metabolism , CD3 Complex/metabolism , Cell Line, Tumor , Colon/immunology , Female , HEK293 Cells , Humans , Inflammation/pathology , Intracellular Signaling Peptides and Proteins , JNK Mitogen-Activated Protein Kinases/metabolism , MAP Kinase Signaling System , Male , Mice , Mice, Inbred BALB C , Mice, Knockout , NF-KappaB Inhibitor alpha/metabolism , Phosphorylation , RAW 264.7 Cells , Ubiquitination/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL