Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Biomed Res Int ; 2022: 8077058, 2022.
Article in English | MEDLINE | ID: mdl-35757465

ABSTRACT

Purpose: Dioscorea nipponica Makino (DNM) is a traditional herb with multiple medicinal functions. This study is aimed at exploring the therapeutic effects of DNM on asthma and the underlying mechanisms involving RKIP-mediated MAPK signaling pathway. Methods: An ovalbumin-induced asthma model was established in mice, which was further administrated with DNM and/or locostatin (RKIP inhibitor). ELISA was performed to detect the serum titers of OVA-IgE and OVA-IgG1, bronchoalveolar lavage fluid (BALF) levels of inflammation-related biomarkers, and tissue levels of oxidative stress-related biomarkers. The expression of RKIP was measured by quantitative real-time PCR, Western blot, immunohistochemistry, and immunofluorescence. HE staining was used to observe the pathological morphology of lung tissues. The protein expression of MAPK pathway-related proteins was detected by Western blot. Results: Compared with the controls, the model mice exhibited significantly higher serum titers of OVA-IgE and OVA-IgG1, BALF levels of IL-6, IL-8, IL-13, TGF-ß1, and MCP-1, tissue levels of MDA and ROS, lower BALF levels of IL-10 and IFN-γ, and tissue level of GSH. DNM relieved the allergic inflammatory response and oxidative stress in the model mice. DNM also recovered the downregulation of RKIP and the pathological injury of lung tissues in asthma mice. In addition, the Raf-1/MEK/MAPK/ERK pathway in the model mice was blocked by DNM. Silencing of RKIP by locostatin weakened the relieving effects of DNM on asthma through activating the Raf-1/MEK/MAPK/ERK pathway. Conclusion: DNM relieves asthma via blocking the Raf-1/MEK/MAPK/ERK pathway that mediated by RKIP upregulation.


Subject(s)
Asthma , Dioscorea , MAP Kinase Signaling System , Plant Extracts , Animals , Asthma/chemically induced , Asthma/drug therapy , Asthma/metabolism , Bronchoalveolar Lavage Fluid , Cytokines/metabolism , Dioscorea/chemistry , Disease Models, Animal , Immunoglobulin E/metabolism , Immunoglobulin G/metabolism , MAP Kinase Signaling System/drug effects , Mice , Mice, Inbred BALB C , Mitogen-Activated Protein Kinase Kinases/metabolism , Ovalbumin , Phosphatidylethanolamine Binding Protein/metabolism , Plant Extracts/pharmacology , Proto-Oncogene Proteins c-raf/metabolism
2.
Gynecol Endocrinol ; 38(12): 1136-1146, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36592742

ABSTRACT

BACKGROUND: NeiyiKangfu tablets (NYKF) are widely used clinically for the treatment of endometriosis (EMS), whose mechanism of action has been extensively studied. Researchers have found that NYKF may control the development of ectopic lesions by inhibiting angiogenesis and inflammatory cytokine secretion. Nevertheless, NYKF's mechanism of action remains unclear. METHODS: In the present study, the function of NYKF in the progression of EMS and the associated underlying mechanism was investigated by in vivo and in vitro experiments. EMS model mice were treated with NYKF and the pro-inflammatory factors and apoptosis of ectopic endometrium as well as RAF/mitogen-activated protein kinase kinase (MEK)/extracellular signal-regulated kinase (ERK) signaling activation were assessed. In addition, human endometriosis-derived immortalized entopic stromal (hEM15A) cells transfected with or without RAF kinase inhibitor protein (RKIP)-small-interfering RNA (siRNA) were also treated with NYKF and the proliferation, migration, apoptosis, and RAF/MEK/ERK signaling activation were measured by Cell Counting Kit-8 (CCK-8), flow cytometry, Transwell, and western blot, respectively. RESULTS: Results showed that NYKF increased the expression of RKIP, inhibited RAF/MEK/ERK signaling activation, and induced apoptosis while inhibiting proliferation and migration both in EMS mice and hEM15A cells. RKIP knockdown could inhibit the effect of NYKF treatment, leading to the activation of RAF/MEK/ERK signaling and the proliferation and migration of hEM15A cells. CONCLUSIONS: In conclusion, these results suggest that NYKF treatment promotes apoptosis and inhibits proliferation and migration in EMS by inhibiting the RAF/MEK/ERK signaling pathway by targeting RKIP.


Subject(s)
Drugs, Chinese Herbal , Endometriosis , Extracellular Signal-Regulated MAP Kinases , Phosphatidylethanolamine Binding Protein , Animals , Female , Humans , Mice , Endometriosis/drug therapy , Extracellular Signal-Regulated MAP Kinases/metabolism , MAP Kinase Signaling System , Mitogen-Activated Protein Kinase Kinases/metabolism , Mitogen-Activated Protein Kinase Kinases/pharmacology , Phosphatidylethanolamine Binding Protein/drug effects , Phosphatidylethanolamine Binding Protein/metabolism , Phosphatidylethanolamine Binding Protein/pharmacology , Signal Transduction
3.
Crit Rev Oncog ; 26(2): 27-37, 2021.
Article in English | MEDLINE | ID: mdl-34347971

ABSTRACT

Drug resistance, which is often of a multiple type, can be defined as the ability of cancer cells to obtain resistance to both conventional and novel chemotherapy agents. It remains a major problem to solve in cancer therapy. The mechanisms of resistance are multifactorial, and in our cellular models of acute myeloid leukemia, hepatocellular carcinoma, and triple-negative breast cancer, it involves the NF-κB pathway. In our opinion, multitarget molecules can be considered as privileged compounds capable of attacking and reversing the resistant phenotype. In the phenomena of both innate and acquired drug resistance that we have been studying since 1998 to today and up to 2016 under the guidance of Professor Natale D'Alessandro, more strictly pharmacological factors are certainly involved. These factors include P-glycoprotein and biological factors such as inhibitory proteins; apoptosis; the Raf-1 kinase inhibitor protein, an important tumor suppressor and metastasis inhibitor, which enhances drug-induced apoptosis of cancer cells; and Yin Yang, a transcription factor involved in drug resistance.


Subject(s)
Antineoplastic Agents , Drug Resistance, Neoplasm , Neoplasms , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Humans , NF-kappa B/metabolism , Neoplasms/drug therapy , Neoplasms/genetics , Phosphatidylethanolamine Binding Protein/metabolism , YY1 Transcription Factor/metabolism
4.
Int J Med Sci ; 15(14): 1658-1666, 2018.
Article in English | MEDLINE | ID: mdl-30588189

ABSTRACT

In a previous study, we reported the positive effects of extremely low frequency electromagnetic field (ELF-MF) exposure on Alzheimer's disease (AD) rats; however, the underlying mechanism remains unclear. In addition, we found that Raf-1 kinase inhibitor protein (RKIP) was downregulated by microwave exposure in the rat hippocampus. Our hypothesis was that RKIP-mediated NF-κB pathway signaling is involved in the effect of ELF-MF on the AD rat. In this study, D-galactose intraperitoneal (50 mg/kg/d for 42 d) and Aß25-35 hippocampal (5 µL/unilateral, bilateral, single-dose) injection were implemented to establish an AD rat model. Animals were exposed to 50 Hz and 400 µT ELF-MF for 60 continuous days. The spatial memory ability of the rat was then tested using the Morris water maze. Protein expression and interaction were detected by western blotting and co-immunoprecipitation for RKIP-mediated NF-κB pathway factors. The results showed that ELF-MF exposure partially improved the cognitive disorder, upregulated the levels of RKIP, TAK1, and the RKIP/TAK1 interaction, but downregulated p-IKK levels in AD rats. These results indicated that RKIP-mediated NF-κB pathway signaling plays an important role in the ELF-MF exposure-mediated improvements in the AD rat. Our study suggested that ELF-MF exposure might have a potential therapeutic value for AD. Further in depth studies are required in the future.


Subject(s)
Alzheimer Disease/therapy , Hippocampus/metabolism , Magnetic Field Therapy/methods , NF-kappa B/metabolism , Phosphatidylethanolamine Binding Protein/metabolism , Alzheimer Disease/chemically induced , Alzheimer Disease/pathology , Amyloid beta-Peptides/administration & dosage , Amyloid beta-Peptides/toxicity , Animals , Behavior, Animal , Disease Models, Animal , Down-Regulation , Galactose/administration & dosage , Galactose/toxicity , Humans , Male , Maze Learning , Peptide Fragments/administration & dosage , Peptide Fragments/toxicity , Rats , Rats, Wistar , Signal Transduction , Treatment Outcome
5.
Crit Rev Oncog ; 23(5-6): 307-320, 2018.
Article in English | MEDLINE | ID: mdl-30311562

ABSTRACT

The role of autophagy in the pathogenesis of various cancers has been well documented in many reports. Autophagy in cancer cells regulates cell proliferation, viability, invasion, epithelial-to-mesenchymal transition (EMT), metastasis, and responses to chemotherapeutic and immunotherapeutic treatment strategies. These manifestations are the result of various regulatory gene products that govern autophagic, biochemical, and molecular mechanisms. In several human cancer cell models, the presence of a dysregulated circuit-namely, NFκB/SNAIL/YY1/RKIP/PTEN-that plays a major role in the regulation of tumor cell unique characteristics just listed for autophagy-regulated activities. Accordingly, the autophagic mechanism and the dysregulated circuit in cancer cells share many of the same properties and activities. Thus, it has been hypothesized that there must exist a biochemical/molecular link between the two. The present review describes the link and the association of each gene product of the dysregulated circuit with the autophagic mechanism and delineates the presence of crosstalk. Crosstalk between autophagy and the dysregulated circuit is significant and has important implications in the development of targeted therapies aimed at either autophagy or the dysregulated gene products in cancer cells.


Subject(s)
Autophagy , Gene Expression Regulation, Neoplastic , Neoplasms/etiology , Neoplasms/metabolism , Signal Transduction , Animals , Autophagy/genetics , Biomarkers , Humans , Molecular Targeted Therapy , NF-kappa B/genetics , NF-kappa B/metabolism , Neoplasms/drug therapy , Neoplasms/pathology , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Phosphatidylethanolamine Binding Protein/genetics , Phosphatidylethanolamine Binding Protein/metabolism , Signal Transduction/drug effects , Snail Family Transcription Factors/genetics , Snail Family Transcription Factors/metabolism , YY1 Transcription Factor/genetics , YY1 Transcription Factor/metabolism
6.
Curr Mol Med ; 18(9): 630-639, 2018.
Article in English | MEDLINE | ID: mdl-30608040

ABSTRACT

BACKGROUND: Overexpression of MDA-9/Syntenin occurs in multiple human cancer cell lines and is associated with higher grade of tumor classification, invasiveness and metastasis. In some cases, its role in cancer biology depends on relationships between MDA-9/Syntenin and NF-κB. OBJECTIVE: This study aims to analyze the presence of a regulation loop like that between MDA-9/Syntenin - NF-κB - RKIP in human liver carcinoma. METHODS: Transient transfection was performed with siRNA anti-MDA-9/Syntenin. Expression of different factors was evaluated by Real time-PCR and Western blotting, while NF-κB activation by TransAM assay. Invasion capacity was analyzed by Matrigel Invasion Assay and the effects of agents on cell viability were examined by MTS assay. RESULTS: We have examined basal expression of MDA-9/Syntenin in three cell lines of human liver carcinoma (HA22T/VGH, Hep3B and HepG2). In all cell lines there was an inverse relationship between MDA-9/Syntenin and RKIP expression levels, and a positive correlation between MDA-9/Syntenin expression and NF-κB activation levels. By silencing with a siRNA anti-MDA-9/Syntenin we observed in all cell lines a very strong increase of RKIP at mRNA level. Interestingly, in all cell lines, inhibition of MDA- 9/Syntenin expression induced NF-κB downregulation and contemporary a reduction in invasion ability MMP-2 dependent. Finally, we showed a good additive effect of MDA- 9/Syntenin siRNA when associated with Curcumin or Doxorubicin on cell growth inhibition. CONCLUSION: Our data confirm the key role of MDA-9/Syntenin in HCC biology. The presence of a regulation loop among MDA-9/Syntenin, NF-κB and RKIP provide new pharmacological approaches.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Carcinoma, Hepatocellular , Liver Neoplasms , NF-kappa B/metabolism , Neoplasm Proteins/metabolism , Phosphatidylethanolamine Binding Protein/metabolism , Syntenins/metabolism , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Curcumin/pharmacology , Doxorubicin/pharmacology , Hep G2 Cells , Humans , Liver Neoplasms/drug therapy , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , NF-kappa B/genetics , Neoplasm Invasiveness , Neoplasm Proteins/genetics , Phosphatidylethanolamine Binding Protein/genetics , Signal Transduction/drug effects , Signal Transduction/genetics , Syntenins/genetics
7.
Invest New Drugs ; 35(1): 115-123, 2017 02.
Article in English | MEDLINE | ID: mdl-27726093

ABSTRACT

Photodynamic therapy (PDT) has drawn considerable attention for its efficacy against certain types of cancers. It shows however limits in the case of deep cancers, favoring tumor recurrence under suboptimal conditions. More insight into the molecular mechanisms of PDT-induced cytotoxicity and cytoprotection is essential to extend and strengthen this therapeutic modality. As PDT induces iNOS/NO in both tumor and microenvironment, we examined the role of nitric oxide (NO) in cytotoxicity and cytoprotection. Our findings show that NO mediates its cellular effects by acting on the NF-κB/YY1/RKIP loop, which controls cell growth and apoptosis. The cytoprotective effect of PDT-induced NO is observed at low NO levels, which activate the pro-survival/anti-apoptotic NF-κB and YY1, while inhibiting the anti-survival/pro-apoptotic and metastasis suppressor RKIP. In contrast, high PDT-induced NO levels inhibit NF-κB and YY1 and induce RKIP, resulting in significant anti-tumor activity. These findings reveal a critical role played by NO in PDT and suggest that the use of bifunctional PDT agents composed of a photosensitizer and a NO-donor could enhance the photo-treatment effect. A successful application of NO in anticancer therapy requires control of its concentration in the target tissue. To address this issue we propose as PDT agent, a bimolecular conjugate called DR2, composed of a photosensitizer (Pheophorbide a) and a non-steroidal anti-androgen molecule capable of releasing NO under the exclusive control of light. The mechanism of action of DR2 in prostate cancer cells is reported and discussed.


Subject(s)
Chlorophyll/analogs & derivatives , Nitric Oxide Donors/pharmacology , Photochemotherapy , Photosensitizing Agents/pharmacology , Prostatic Neoplasms/drug therapy , Triazenes/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Chlorophyll/pharmacology , Humans , Male , NF-kappa B/metabolism , Nitric Oxide/metabolism , Phosphatidylethanolamine Binding Protein/metabolism , Prostatic Neoplasms/metabolism , YY1 Transcription Factor/metabolism
8.
Int Immunopharmacol ; 42: 130-138, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27912149

ABSTRACT

A flavone was isolated from Origanum vulgare and identified as didymin (O. vulgare didymin, OVD). The protective effect and mechanism of OVD on acute liver injury was then assessed in vivo and in vitro. Our results showed that OVD significantly alleviated CCl4-induced liver injury in mice and markedly decreased serum ALT and AST activities. OVD treatment significantly reduced CYP2E1 activity, lipid peroxidation level, ROS generation, NO production and pro-inflammatory cytokines (such as TNF-α, IL-6 and IL-1ß) in liver tissues and RAW 264.7 cells, but enhanced the hepatic antioxidative enzymes activities. Further study showed that OVD significantly inhibited the NF-κB and MAPK pathways. Interestingly, OVD notably enhanced Raf kinase inhibitor protein (RKIP) expression, and the effects of OVD on histological changes, oxidative stress and inflammation was largely abolished by the RKIP specific inhibitor locostatin. Our findings indicate that OVD can ameliorate CCl4-induced liver injury, which may be ascribed to its radical scavenging action, antioxidant activity, and modulation of MAPK and NF-κB signaling pathways.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Chemical and Drug Induced Liver Injury/drug therapy , Flavonoids/therapeutic use , Glycosides/therapeutic use , Origanum , Phosphatidylethanolamine Binding Protein/metabolism , Alanine Transaminase/blood , Animals , Aspartate Aminotransferases/blood , Carbon Tetrachloride , Cytochrome P-450 CYP2E1/metabolism , Cytokines/metabolism , Humans , Inflammation Mediators/metabolism , Lipid Peroxidation/drug effects , Male , Mice , Mice, Inbred ICR , Oxazolidinones/pharmacology , Phosphatidylethanolamine Binding Protein/antagonists & inhibitors , RAW 264.7 Cells , Reactive Oxygen Species/metabolism
9.
Redox Biol ; 6: 311-317, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26319434

ABSTRACT

Photodynamic therapy (PDT) against cancer has gained attention due to the successful outcome in some cancers, particularly those on the skin. However, there have been limitations to PDT applications in deep cancers and, occasionally, PDT treatment resulted in tumor recurrence. A better understanding of the underlying molecular mechanisms of PDT-induced cytotoxicity and cytoprotection should facilitate the development of better approaches to inhibit the cytoprotective effects and also augment PDT-mediated cytotoxicity. PDT treatment results in the induction of iNOS/NO in both the tumor and the microenvironment. The role of NO in cytotoxicity and cytoprotection was examined. The findings revealed that NO mediates its effects by interfering with a dysregulated pro-survival/anti-apoptotic NF-κB/Snail/YY1/RKIP loop which is often expressed in cancer cells. The cytoprotective effect of PDT-induced NO was the result of low levels of NO that activates the pro-survival/anti-apoptotic NF-κB, Snail, and YY1 and inhibits the anti-survival/pro-apoptotic and metastasis suppressor RKIP. In contrast, PDT-induced high levels of NO result in the inhibition of NF-kB, Snail, and YY1 and the induction of RKIP, all of which result in significant anti-tumor cytotoxicity. The direct role of PDT-induced NO effects was corroborated by the use of the NO inhibitor, l-NAME, which reversed the PDT-mediated cytotoxic and cytoprotective effects. In addition, the combination of the NO donor, DETANONOate, and PDT potentiated the PDT-mediated cytotoxic effects. These findings revealed a new mechanism of PDT-induced NO effects and suggested the potential therapeutic application of the combination of NO donors/iNOS inducers and PDT in the treatment of various cancers. In addition, the study suggested that the combination of PDT with subtoxic cytotoxic drugs will result in significant synergy since NO has been shown to be a significant chemo-immunosensitizing agent to apoptosis.


Subject(s)
Gene Expression Regulation, Neoplastic , Neoplasms/drug therapy , Nitric Oxide/pharmacology , Photochemotherapy/methods , Photosensitizing Agents/therapeutic use , Apoptosis/drug effects , Cell Line, Tumor , Humans , Light , NF-kappa B/genetics , NF-kappa B/metabolism , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Nitric Oxide/metabolism , Nitric Oxide Donors/metabolism , Organ Specificity , Phosphatidylethanolamine Binding Protein/genetics , Phosphatidylethanolamine Binding Protein/metabolism , Signal Transduction , Snail Family Transcription Factors , Transcription Factors/genetics , Transcription Factors/metabolism , YY1 Transcription Factor/genetics , YY1 Transcription Factor/metabolism
10.
PLoS One ; 8(2): e56829, 2013.
Article in English | MEDLINE | ID: mdl-23451095

ABSTRACT

Rituximab is the first line drug to treat non Hodgkin's lymphoma (B-NHL) alone or in combination with chemotherapy. However, 30-40% of B-NHL patients are unresponsive to rituximab or resistant after therapy. Human phosphatidylethanolamine-binding protein 4 (hPEBP4) is a novel member of PEBP family and functions as an anti-apoptotic molecule. In this study, we found hPEBP4 to be expressed in up to 90% of B-cell lymphoma patients, but in only 16.7% of normal lymph nodes. Interestingly, hPEBP4 overexpression inhibited rituximab-mediated complement dependent cytotoxicity (R-CDC) and antibody-dependent cell-mediated cytotoxicity (ADCC) in B-NHL cells while downregulation of hPEBP4 augmented the therapeutic efficacy of rituximab both in vitro and in vivo. Furthermore, hPEBP4 silencing sensitized the primary B-acute lymphocytic leukemia (B-ALL) cells to R-CDC. During rituximab-mediated complement dependent cytotoxicity, hPEBP4 was recruited to the cell membrane in a PE-binding domain dependent manner and inhibited R-CDC induced calcium flux and reactive oxygen species (ROS) generation. These events contributed to the decrease of cell death induced by R-CDC in B-cell lymphomas. Meanwhile, hPEBP4 knockdown potentiated the chemosensitization of the rituximab in B-cell lymphoma cells by regulating the expression of Bcl-xl, Cycline E, p21(waf/cip1) and p53 and the activation of caspase-3 and caspase-9. Considering that hPEBP4 conferred cellular resistance to rituximab treatment and was preferentially expressed in lymphoma tissue, it could be a potential valuable target for adjuvant therapy for B-cell lymphoma.


Subject(s)
Antibodies, Monoclonal, Murine-Derived/pharmacology , Antibodies, Monoclonal, Murine-Derived/therapeutic use , Lymphoma, B-Cell/drug therapy , Phosphatidylethanolamine Binding Protein/metabolism , Animals , Apoptosis/drug effects , Apoptosis/genetics , Cell Line, Tumor , Drug Resistance, Neoplasm/genetics , Female , Gene Silencing , Humans , Immunohistochemistry , In Vitro Techniques , Lymphoma, B-Cell/genetics , Lymphoma, B-Cell/metabolism , Mice , Mice, Inbred BALB C , Mice, Nude , Microscopy, Confocal , Phosphatidylethanolamine Binding Protein/genetics , Reactive Oxygen Species/metabolism , Rituximab
11.
Lung Cancer ; 74(2): 197-205, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21529991

ABSTRACT

INTRODUCTION: Lung cancer is one of the most lethal tumors and, although standard chemotherapy produces clinical response, there has been little improvement in prognosis. Therefore, research effort has focused on target-specific agents, such as sorafenib, which blocks both the RAF/MEK/ERK signalling pathways and receptors involved in neovascularization and tumor progression, including VEGFR-2 and c-Kit. We investigated whether sorafenib would be synergistic with gemcitabine against NSCLC cell lines. MATERIALS AND METHODS: Human lung cancer cells A549, CALU-1, CALU-6, H23 and HCC 827 were treated with sorafenib and gemcitabine, alone or in combination, and the cytotoxicity was assessed with CellTiter 96 Non-radioactive cell proliferation kit. Cell cycle and apoptosis were investigated with flow cytometry and fluorescence microscopy, respectively. Moreover, the effects of drugs on Akt (S473), c-Kit (Y823) and ERK (pTpY185/187) phosphorylation were studied with ELISA. Finally, quantitative PCR analysis was performed to assess whether sorafenib and gemcitabine modulated the expression of genes related to drug activity. RESULTS: Gemcitabine and sorafenib synergistically interacted on the inhibition of cell proliferation, and assessment of apoptosis demonstrated that drug associations increased the apoptotic index. Sorafenib reduced c-Kit and ERK activation and gemcitabine inhibited Akt phosphorylation. Moreover quantitative PCR showed that sorafenib modulated the expression of targets related to gemcitabine activity, while gemcitabine induced the expression of RKIP. CONCLUSIONS: These data demonstrate that sorafenib and gemcitabine synergistically interact against NSCLC cells, through suppression of Akt, c-Kit and ERK phosphorylation, induction of apoptosis and modulation of dCK, RRM1, RRM2 and RKIP gene expression. The association between traditional cytotoxic agents with new target-specific agents, such as sorafenib, is a challenge for both clinical and preclinical future investigations in lung cancer treatments.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Apoptosis , Carcinoma, Non-Small-Cell Lung/drug therapy , Gene Expression Regulation, Neoplastic , Lung Neoplasms/drug therapy , MAP Kinase Signaling System , Apoptosis/drug effects , Benzenesulfonates/administration & dosage , Benzenesulfonates/pharmacology , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/physiopathology , Cell Growth Processes/drug effects , Cell Line, Tumor , Deoxycytidine/administration & dosage , Deoxycytidine/analogs & derivatives , Drug Synergism , Gene Expression Regulation, Neoplastic/drug effects , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Lung Neoplasms/physiopathology , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/genetics , Neovascularization, Pathologic , Niacinamide/analogs & derivatives , Phenylurea Compounds , Phosphatidylethanolamine Binding Protein/genetics , Phosphatidylethanolamine Binding Protein/metabolism , Phosphorylation/drug effects , Proto-Oncogene Proteins c-kit/antagonists & inhibitors , Pyridines/administration & dosage , Pyridines/pharmacology , Receptors, Vascular Endothelial Growth Factor/antagonists & inhibitors , Sorafenib , raf Kinases/antagonists & inhibitors , Gemcitabine
12.
J Appl Toxicol ; 31(8): 730-40, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21437922

ABSTRACT

Hexavalent chromium (CrVI) is considered to be a risk factor in the formation of human cancer. Raf kinase inhibitor protein (RKIP), Rho-GDIα, galectin, c-Myc and p53 play important roles in cancer formation. The purpose of this study was to determine if Cr(VI) induces the formation of gastrointestinal cancer. We explored the expression of RKIP, Rho-GDIα, galectin, c-Myc and p53 in the colon and stomach in rats exposed to chromium (CrVI). Thirty Wistar rats were divided into six groups which were chronically fed with 250, 500, 750, 1000 and 1250 ppm Na(2) Cr(2) O(7) and water for 60 days. The level of Cr(VI) was determined by electrothermal atomic absorption spectrometry. The expression of RKIP, Rho-GDIα, galectin, c-Myc and p53 of stomach and colon was measured by western blot. The gene expression of RKIP, Rho-GDIα, galectin, c-Myc and p53 of the stomach and colon was determined by RT-PCR. The results showed that the expression of p53 and Rho-GDIα was decreased in the stomach and colon of rats with Cr(VI) treatment. The expression of RKIP was decreased in the stomach and colon of rats treated with high-dose Cr(VI). The expression of c-Myc and gelectin-1 was increased in the stomach and colon of rats with Cr(VI) treatment. We concluded that the anomalous expression of RKIP, Rho-GDIα, galectin, c-Myc and p53 might be a dangerous index of cancer formation in the stomach and colon of rats with Cr(VI) exposure.


Subject(s)
Chromium/toxicity , Galectins/genetics , Gastrointestinal Tract/drug effects , Guanine Nucleotide Dissociation Inhibitors/genetics , Phosphatidylethanolamine Binding Protein/genetics , Proto-Oncogene Proteins c-myc/genetics , Tumor Suppressor Protein p53/genetics , Animals , Apoptosis/drug effects , Blotting, Western , Galectins/metabolism , Gastrointestinal Tract/pathology , Gene Expression Regulation , Guanine Nucleotide Dissociation Inhibitors/metabolism , Male , Phosphatidylethanolamine Binding Protein/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Rats , Rats, Wistar , Spectrophotometry, Atomic , Tumor Suppressor Protein p53/metabolism , rho Guanine Nucleotide Dissociation Inhibitor alpha , rho-Specific Guanine Nucleotide Dissociation Inhibitors
13.
OMICS ; 15(5): 267-72, 2011 May.
Article in English | MEDLINE | ID: mdl-21332389

ABSTRACT

The transcription factor Yin Yang 1 (YY1) can favor several aspects of tumorigenesis. In turn, Raf-1 Kinase Inhibitor Protein (RKIP) inhibits the oncogenic activities of MAPK and NF-κB pathways and promotes drug-induced apoptosis. Mutual influences between YY1 and RKIP may exist, and there are already separate evidences that relevant increases in YY1 and reductions in RKIP occur in hepatocellular carcinoma (HCC). However, the levels of the two factors have never been concomitantly examined in HCC. We evaluated by RT-PCR the mRNA levels of YY1, YY1AP, RKIP, and survivin in 35 clinical HCCs (91% HCV-related), in their adjacent cirrhotic tissues and in 6 healthy livers. Immunohistochemical analyses were also performed. The ratio of YY1 to RKIP mRNA was constantly profoundly inverted in the tumors compared with the adjacent nontumoral tissues. A similar result occurred frequently at protein level. Hyperactivation of YY1 in tumors was corroborated by its nuclear localization and the finding that in the tumors there were also increases in YY1AP, a YY1 coactivator not expressed in normal liver, and in survivin, as a possible target of YY1. The frequent alteration in the YY1-RKIP balance might represent a marker of malignant progression and be exploited for therapeutic interventions in HCC.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/metabolism , Phosphatidylethanolamine Binding Protein/metabolism , YY1 Transcription Factor/metabolism , Adult , Aged , Aged, 80 and over , Carcinoma, Hepatocellular/enzymology , Carcinoma, Hepatocellular/genetics , Cell Cycle Proteins , Female , Gene Expression Regulation, Neoplastic , Humans , Inhibitor of Apoptosis Proteins/genetics , Liver/metabolism , Liver Cirrhosis/genetics , Liver Cirrhosis/metabolism , Liver Neoplasms/enzymology , Liver Neoplasms/genetics , Male , Middle Aged , Nuclear Proteins/genetics , Phosphatidylethanolamine Binding Protein/genetics , RNA, Messenger/genetics , Survivin , Transcription Factors/genetics , YY1 Transcription Factor/genetics
14.
Nitric Oxide ; 24(1): 1-7, 2011 Jan 01.
Article in English | MEDLINE | ID: mdl-20933602

ABSTRACT

Several studies have implicated the role of Nitric Oxide (NO) in the regulation of tumor cell behavior and have shown that NO either promotes or inhibits tumorigenesis. These conflicting findings have been resolved, in part, by the levels of NO used such that low levels promote tumor growth and high levels inhibit tumor growth. Our studies have focused on the use of high levels of NO provided primarily by the NO donor, DETANONOate. We have shown that treatment of resistant tumor cells with DETANONOate sensitizes them to apoptosis by both chemotherapeutic drugs and cytotoxic immunotherapeutic ligands. The underlying mechanisms by which NO sensitizes tumor cells to apoptosis were shown to be regulated, in part, by NO-mediated inhibition of the NF-κB survival/anti-apoptotic pathways and downstream of NF-κB by inhibition of the transcription factor Yin Yang 1 (YY1). In addition to NO-induced sensitization to apoptosis, we have also shown that NO induced the expression of the metastasis-suppressor/immunosurveillance cancer gene product, Raf-1 kinase inhibitor protein (RKIP). Overexpression of RKIP mimics NO in tumor cells-induced sensitization to apoptosis. The induction of RKIP by NO was the result of the inhibition of the RKIP repressor, Snail, downstream of NF-κB. These findings established the presence of a dysregulated NF-κB/Snail/YY1/ RKIP circuitry in resistance and that treatment with NO modifies this loop in tumor cells in favor of the inhibition of tumor cell survival and the response to cytotoxic drugs. Noteworthy, the NF-κB/Snail/YY1/RKIP loop consists of gene products that regulate the epithelial to mesenchymal transition (EMT) and, thus, tumor metastasis. Hence, we have found that treatment of metastatic cancer cell lines with DETANONOate inhibited the EMT phenotype, through both the inhibition of the metastasis-inducers, NF-κB and Snail and the induction of the metastasis-suppressor, RKIP. Altogether, the above findings establish, for the first time, the dual role of high levels of NO in the sensitization of tumor cells to apoptotic stimuli as well as inhibition of EMT. Hence, NO donors may be considered as novel potential therapeutic agents with dual roles in the treatment of patients with refractory cancer and in the prevention of the initiation of the metastatic cascade via EMT.


Subject(s)
Neoplasms/drug therapy , Nitric Oxide Donors/pharmacology , Nitroso Compounds/pharmacology , Animals , Cell Transformation, Neoplastic , Drug Resistance, Neoplasm , Drug Synergism , Humans , NF-kappa B/metabolism , Neoplasms/metabolism , Nitric Oxide Donors/pharmacokinetics , Nitroso Compounds/pharmacokinetics , Phosphatidylethanolamine Binding Protein/metabolism , Signal Transduction/drug effects , Snail Family Transcription Factors , Transcription Factors/metabolism , YY1 Transcription Factor/metabolism
15.
Crit Rev Oncog ; 16(3-4): 211-26, 2011.
Article in English | MEDLINE | ID: mdl-22248055

ABSTRACT

The majority of cancers develop genetic and molecular strategies to resist conventional cytotoxic therapies as well as escape from the host's immune surveillance. These events lead to tumor persistence and spread through activation of the epithelial to mesenchymal transition (EMT) program and metastasis. Expression profiling analysis has revealed various tumor metastasis-inducing and metastasis-suppressing genes that regulate the metastatic process and maintain the microenvironment of the tumor cells. EMT in cancer entails the molecular reprogramming and phenotypic changes that characterize the conversion of immobile cancer epithelial cells to motile mesenchymal cells. A hallmark of EMT is the loss of E-cadherin, the epithelial homotypic adhesion molecule, and gain of mesenchymal markers such as vimentin and fibronectin. The molecular mechanisms underlying the initiation of EMT consist, in part, in the constitutive activation of survival signaling pathways such as the nuclear factor (NF)-κB pathway. The NF-κB pathway has been implicated directly in the regulation of EMT and indirectly through the transcription and expression of several gene products that participate in the EMT cascade, such as Snail, the metastasis-inducer and E-cadherin suppressor transcription factor. In turn, Snail represses the metastasis-suppressor gene product Raf-kinase inhibitor protein (RKIP) that inhibits both the Raf-1/MEK/ERK and NF-κB survival pathways implicated in EMT. Consequently, tumor cells normally exhibit a dysregulated NF-κB/Snail/RKIP circuitry that is intimately involved in the initiation of EMT and maintenance of drug resistance. Additional deregulated gene products in this circuit, such as the metastasis-suppressor phosphatase and tensin homologue (PTEN; repressed by Snail) and the putative-metastasis inducer Yin Yang (YY) 1 (target of NF-κB) also have been associated in the regulation of EMT. We recently have investigated the direct role of YY1 in the regulation of EMT. This review discusses the molecular regulation of EMT in cancer cells through the activity of the dysregulated NF-κB/Snail/ YY1/PTEN/RKIP circuitry. In addition, we propose YY1 as a potential novel prognostic biomarker for EMT and a therapeutic target for both the prevention of metastasis and the reversal of resistance.


Subject(s)
Epithelial-Mesenchymal Transition , Neoplasms/pathology , Oncogenes , Humans , NF-kappa B/metabolism , Neoplasms/metabolism , PTEN Phosphohydrolase/metabolism , Phosphatidylethanolamine Binding Protein/metabolism , Snail Family Transcription Factors , Transcription Factors/metabolism , YY1 Transcription Factor/metabolism
16.
Cell Res ; 18(4): 452-7, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18379591

ABSTRACT

Cells have a multitude of controls to maintain their integrity and prevent random switching from one biological state to another. Raf Kinase Inhibitory Protein (RKIP), a member of the phosphatidylethanolamine binding protein (PEBP) family, is representative of a new class of modulators of signaling cascades that function to maintain the "yin yang" or balance of biological systems. RKIP inhibits MAP kinase (Raf-MEK-ERK), G protein-coupled receptor (GPCR) kinase and NFkappaB signaling cascades. Because RKIP targets different kinases dependent upon its state of phosphorylation, RKIP also acts to integrate crosstalk initiated by multiple environmental stimuli. Loss or depletion of RKIP results in disruption of the normal cellular stasis and can lead to chromosomal abnormalities and disease states such as cancer. Since RKIP and the PEBP family have been reviewed previously, the goal of this analysis is to provide an update and highlight some of the unique features of RKIP that make it a critical player in the regulation of cellular signaling processes.


Subject(s)
Neoplasm Metastasis , Phosphatidylethanolamine Binding Protein/metabolism , Signal Transduction , Tumor Suppressor Proteins/metabolism , Animals , Humans , Mitosis , Phosphatidylethanolamine Binding Protein/chemistry , Tumor Suppressor Proteins/chemistry , raf Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL