Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Oncotarget ; 6(18): 16271-86, 2015 Jun 30.
Article in English | MEDLINE | ID: mdl-26009991

ABSTRACT

Interleukin (IL)-24 is a tumor suppressor/cytokine gene that undergoes post-translational modifications (PTMs). Glycosylation and ubiquitination are important for IL-24 protein stabilization and degradation respectively. Little is known about IL-24 protein phosphorylation and its role in IL-24-mediated anti-tumor activities. In this study we conducted molecular studies to determine whether IL-24 phosphorylation is important for IL-24-mediated anti-cancer activity.Human H1299 lung tumor cell line that was stably transfected with a doxycycline (DOX)-inducible (Tet-on) plasmid vector carrying the cDNA of IL-24-wild-type (IL-24wt) or IL-24 with all five phosphorylation sites replaced (IL-24mt) was used in the present study. Inhibition of tumor cell proliferation, cell migration and invasion, and induction of G2/M cell cycle arrest was observed in DOX-induced IL-24wt-expressing cells but not in IL-24mt-expressing cells. Secretion of IL-24mt protein was greatly reduced compared to IL-24wt protein. Further, IL-24wt and IL-24mt proteins markedly differed in their subcellular organelle localization. IL-24wt but not IL-24mt inhibited the AKT/mTOR signaling pathway. SiRNA-mediated AKT knockdown and overexpression of myristolyated AKT protein confirmed that IL-24wt but not IL-24mt mediated its anti-cancer activity by inhibiting the AKT signaling pathway.Our results demonstrate that IL-24 phosphorylation is required for inhibiting the AKT/mTOR signaling pathway and exerting its anti-cancer activities.


Subject(s)
Apoptosis , Carcinoma, Non-Small-Cell Lung/pathology , Cell Movement , Cell Proliferation , Interleukins/metabolism , Lung Neoplasms/pathology , Protein Processing, Post-Translational , Anti-Bacterial Agents/pharmacology , Blotting, Western , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Cycle , Doxycycline/pharmacology , Enzyme-Linked Immunosorbent Assay , Fluorescent Antibody Technique , Humans , Immunoprecipitation , Interleukins/antagonists & inhibitors , Interleukins/genetics , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , Tumor Cells, Cultured
2.
PLoS One ; 10(3): e0122439, 2015.
Article in English | MEDLINE | ID: mdl-25775124

ABSTRACT

BACKGROUND: The stromal cell derived factor (SDF)-1/chemokine receptor (CXCR)-4 signaling pathway plays a key role in lung cancer metastasis and is molecular target for therapy. In the present study we investigated whether interleukin (IL)-24 can inhibit the SDF-1/CXCR4 axis and suppress lung cancer cell migration and invasion in vitro. Further, the efficacy of IL-24 in combination with CXCR4 antagonists was investigated. METHODS: Human H1299, A549, H460 and HCC827 lung cancer cell lines were used in the present study. The H1299 lung cancer cell line was stably transfected with doxycycline-inducible plasmid expression vector carrying the human IL-24 cDNA and used in the present study to determine the inhibitory effects of IL-24 on SDF-1/CXCR4 axis. H1299 and A549 cell lines were used in transient transfection studies. The inhibitory effects of IL-24 on SDF1/CXCR4 and its downstream targets were analyzed by quantitative RT-PCR, western blot, luciferase reporter assay, flow cytometry and immunocytochemistry. Functional studies included cell migration and invasion assays. PRINCIPAL FINDINGS: Endogenous CXCR4 protein expression levels varied among the four human lung cancer cell lines. Doxycycline-induced IL-24 expression in the H1299-IL24 cell line resulted in reduced CXCR4 mRNA and protein expression. IL-24 post-transcriptionally regulated CXCR4 mRNA expression by decreasing the half-life of CXCR4 mRNA (>40%). Functional studies showed IL-24 inhibited tumor cell migration and invasion concomitant with reduction in CXCR4 and its downstream targets (pAKTS473, pmTORS2448, pPRAS40T246 and HIF-1α). Additionally, IL-24 inhibited tumor cell migration both in the presence and absence of the CXCR4 agonist, SDF-1. Finally, IL-24 when combined with CXCR4 inhibitors (AMD3100, SJA5) or with CXCR4 siRNA demonstrated enhanced inhibitory activity on tumor cell migration. CONCLUSIONS: IL-24 disrupts the SDF-1/CXCR4 signaling pathway and inhibits lung tumor cell migration and invasion. Additionally, IL-24, when combined with CXCR4 inhibitors exhibited enhanced anti-metastatic activity and is an attractive therapeutic strategy for lung metastasis.


Subject(s)
Cell Movement/drug effects , Chemokine CXCL12/metabolism , Interleukins/pharmacology , Receptors, CXCR4/metabolism , Signal Transduction/drug effects , Benzylamines , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Movement/genetics , Cyclams , Drug Synergism , Gene Expression , Gene Expression Regulation, Neoplastic , Heterocyclic Compounds/pharmacology , Humans , Interleukins/genetics , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , RNA, Small Interfering , Receptors, CXCR4/antagonists & inhibitors , Receptors, CXCR4/genetics , TOR Serine-Threonine Kinases/metabolism
3.
Mol Pharm ; 11(8): 2720-33, 2014 Aug 04.
Article in English | MEDLINE | ID: mdl-24922589

ABSTRACT

Development of resistance toward anticancer drugs results in ineffective therapy leading to increased mortality. Therefore, overriding resistance and restoring sensitivity to anticancer drugs will improve treatment efficacy and reduce mortality. While numerous mechanisms for drug resistance in cancer have previously been demonstrated, recent studies implicate a role for proteasome and the autophagy regulatory protein P62/SQSTM1 (P62) in contributing to drug resistance. Specifically, reduction in the expression of the ß5 subunit of the proteasome and/or enhanced P62 protein expression is known to contribute to cancer drug resistance such as cisplatin (CDDP) in ovarian cancer cells. Therefore, we hypothesized that restoration of ß5 expression and/or suppression of P62 protein expression in CDDP-resistant ovarian cancer cells will lead to restoration of sensitivity to CDDP and enhanced cell killing. To test our hypothesis we developed a biodegradable multifunctional nanoparticle (MNP) system that codelivered P62siRNA, ß5 plasmid DNA, and CDDP and tested its efficacy in CDDP resistant 2008/C13 ovarian cancer cells. MNP consisted of CDDP loaded polylactic acid nanoparticle as inner core and cationic chitosan (CS) consisting of ionically linked P62siRNA (siP62) and/or ß5 expressing plasmid DNA (pß5) as the outer layer. The MNPs were spherical in shape with a hydrodynamic diameter in the range of 280-350 nm, and demonstrated encapsulation efficiencies of 82% and 78.5% for CDDP and siRNA respectively. MNPs efficiently protected the siRNA and showed superior serum stability compared to naked siRNA as measured by gel retardation and spectrophotometry assays. The MNPs successfully delivered siP62 and pß5 to cause P62 knockdown and restoration of ß5 expression in 2008/C13 cells. Combined delivery of siP62, pß5, and CDDP using the MNPs resulted in a marked reduction in the IC50 value of CDDP in 2008/C13 cells from 125 ± 1.3 µM to 98 ± 0.6 µM (P < 0.05; 21.6% reduction) when compared to the reduction in the IC50 of CDDP observed in cells that had only siP62 delivered (IC50 = 106 ± 1.1 µM; P < 0.05; 15.2% reduction) or pß5 delivered (IC50 = 115 ± 2.8 µM; 8% reduction) via MNPs. Finally, our studies showed that the CDDP resistance index in 2008/C13 cells was reduced from 4.62 for free CDDP to 3.62 for MNP treatment. In conclusion our study results demonstrated the efficacy of our MNP in overcoming CDDP resistance in ovarian cancer cells.


Subject(s)
Chitosan/chemistry , Cisplatin/administration & dosage , Drug Delivery Systems , Lactic Acid/chemistry , Nanoparticles/chemistry , Ovarian Neoplasms/drug therapy , Polymers/chemistry , Antineoplastic Agents/administration & dosage , Cell Line, Tumor , Cell Survival , Cisplatin/chemistry , Drug Resistance, Neoplasm , Female , Fluorescamine/chemistry , Humans , Inhibitory Concentration 50 , Nanomedicine/methods , Ovarian Neoplasms/metabolism , Particle Size , Plasmids/metabolism , Polyesters , Proteasome Endopeptidase Complex/chemistry , RNA, Small Interfering/metabolism , RNA-Binding Proteins/metabolism
4.
PLoS One ; 6(11): e25507, 2011.
Article in English | MEDLINE | ID: mdl-22087216

ABSTRACT

BACKGROUND: The epidermal growth factor receptor (EGFR) is overexpressed in 80% of non-small cell lung cancer (NSCLC) and is associated with poor survival. In recent years, EGFR-targeted inhibitors have been tested in the clinic for NSCLC. Despite the emergence of novel therapeutics and their application in cancer therapy, the overall survival rate of lung cancer patients remains 15%. To develop more effective therapies for lung cancer we have combined the anti-EGFR antibody (Clone 225) as a molecular therapeutic with hybrid plasmonic magnetic nanoparticles (NP) and tested on non-small cell lung cancer (NSCLC) cells. METHODOLOGY/PRINCIPAL FINDINGS: Cell viability was determined by trypan-blue assay. Cellular protein expression was determined by Western blotting. C225-NPs were detected by electron microscopy and confocal microscopy, and EGFR expression using immunocytochemistry. C225-NP exhibited a strong and selective antitumor effect on EGFR-expressing NSCLC cells by inhibiting EGFR-mediated signal transduction and induced autophagy and apoptosis in tumor cells. Optical images showed specificity of interactions between C225-NP and EGFR-expressing NSCLC cells. No binding of C225-NP was observed for EGFR-null NSCLC cells. C225-NP exhibited higher efficiency in induction of cell killing in comparison with the same amount of free C225 antibody in tumor cells with different levels of EGFR expression. Furthermore, in contrast to C225-NP, free C225 antibody did not induce autophagy in cells. However, the therapeutic efficacy of C225-NP gradually approached the level of free antibodies as the amount of C225 antibody conjugated per nanoparticle was decreased. Finally, attaching C225 to NP was important for producing the enhanced tumor cell killing as addition of mixture of free C225 and NP did not demonstrate the same degree of cell killing activity. CONCLUSIONS/SIGNIFICANCE: We demonstrated for the first time the molecular mechanism of C225-NP induced cytotoxic effects in lung cancer cells that are not characteristic for free molecular therapeutics thus increasing efficacy of therapy against NSCLC.


Subject(s)
Antibodies, Neoplasm/therapeutic use , Apoptosis/drug effects , Autophagy/drug effects , Carcinoma, Non-Small-Cell Lung/drug therapy , ErbB Receptors/antagonists & inhibitors , Magnetite Nanoparticles/therapeutic use , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Drug Evaluation, Preclinical , Humans , Neoplasm Proteins/antagonists & inhibitors
5.
J Drug Deliv ; 2011: 465845, 2011.
Article in English | MEDLINE | ID: mdl-21490751

ABSTRACT

Cancer is a major health problem in the world. Advances made in cancer therapy have improved the survival of patients in certain types of cancer. However, the overall five-year survival has not significantly improved in the majority of cancer types. Major challenges encountered in having effective cancer therapy are development of drug resistance by the tumor cells, nonspecific cytotoxicity, and inability to affect metastatic tumors by the chemodrugs. Overcoming these challenges requires development and testing of novel therapies. One attractive cancer therapeutic approach is cancer gene therapy. Several laboratories including the authors' laboratory have been investigating nonviral formulations for delivering therapeutic genes as a mode for effective cancer therapy. In this paper the authors will summarize their experience in the development and testing of a cationic lipid-based nanocarrier formulation and the results from their preclinical studies leading to a Phase I clinical trial for nonsmall cell lung cancer. Their nanocarrier formulation containing therapeutic genes such as tumor suppressor genes when administered intravenously effectively controls metastatic tumor growth. Additional Phase I clinical trials based on the results of their nanocarrier formulation have been initiated or proposed for treatment of cancer of the breast, ovary, pancreas, and metastatic melanoma, and will be discussed.

6.
Lung Cancer (Auckl) ; 1: 23-36, 2010.
Article in English | MEDLINE | ID: mdl-28210104

ABSTRACT

Resistance to chemotherapy drugs is a major problem in cancer treatment. Scientific advances made in the last two decades have resulted in the identification of genes and molecular signaling mechanisms that contribute to drug resistance. This has resulted in a better understanding of the biology of cancer cells and the way these cells adapt or undergo subtle molecular changes thereby protecting themselves from the cytotoxic effects of the anticancer drugs. Based on the knowledge gained to-date new molecularly targeted drugs are being developed and tested in clinical studies, in an attempt to overcome drug resistance and improve drug efficacy. Despite these attempts the overall 5-year survival of patients diagnosed with cancer, such as lung cancer, remains dismal and is less than 15%. It is evident that additional mechanisms contributing to drug resistance exist which are yet to be discovered. It is hoped that identification of new targets and understanding their contribution to drug resistance will provide opportunities for innovative therapies in overcoming drug resistance. In an attempt to broaden our knowledge and understanding on drug resistance we have, in this review article, summarized the most common mechanisms associated with drug resistance in lung cancer.

7.
Cancer Lett ; 254(2): 217-26, 2007 Sep 08.
Article in English | MEDLINE | ID: mdl-17449172

ABSTRACT

Adenovirus-mediated mda-7 (Ad-mda7) gene transfer has been shown to induce apoptosis in various human cancer cells while sparing normal cells. Vitamin E succinate (VES) is also known to exhibit antitumor activity against a number of human cancer cell lines. We hypothesized that a combination of the two agents would produce an enhanced antitumor effect in MDAH2774 human ovarian cancer cells. Treatment of MDAH2774 cells with Ad-mda7 plus VES resulted in enhanced antitumor activity that involved the activation of two apoptotic pathways. Activation of the extrinsic pathway was demonstrated by increased cell-surface Fas expression and cleavage of Bid and caspase-8. Activation of the intrinsic pathway was demonstrated by disruption of mitochondrial potential; and activation of downstream capase-9 and caspase-3 via cytochrome C release. In contrast, the combination of Ad-mda7 plus VES did not show any antitumor activity against normal fibroblasts, indicating selective tumor cell killing. Our in vitro results provide a basis for further preclinical testing of Ad-mda7 plus VES as a potential cancer treatment strategy.


Subject(s)
Apoptosis/drug effects , Cell Survival/drug effects , Interleukins/pharmacology , Ovarian Neoplasms/pathology , Receptors, Virus/physiology , Vitamin E/analogs & derivatives , Adenoviridae/physiology , Cell Line, Tumor , Dose-Response Relationship, Drug , Enterovirus/physiology , Female , Humans , Membrane Potentials , Mitochondrial Membranes/physiology , Tocopherols , Vitamin E/pharmacology
8.
Mol Cancer ; 6: 11, 2007 Feb 02.
Article in English | MEDLINE | ID: mdl-17274815

ABSTRACT

BACKGROUND: Previous studies showed that the human melanoma differentiation-associated gene-7 (mda-7), also known as interleukin-24 (IL-24), has potent antitumor activity against human and murine cancer cells. However, the majority of these studies were limited to in vitro testing. In the present study, we investigated the antitumor activity of mda-7/IL-24 against human ovarian cancer cells both in vitro and in vivo. RESULTS: In vitro, treatment of ovarian cancer cells with an adenoviral vector carrying the mda-7 gene (Ad-mda7) resulted in inhibition of cell proliferation and induction of cell cycle arrest, leading to apoptosis. We did not observe inhibitory activity in Ad-mda7-treated normal cells. In vivo, treatment of subcutaneous tumor xenografts with Ad-mda7 resulted in significant tumor growth inhibition when compared with that in control groups (p < 0.001). Molecular analysis of ovarian tumor tissue lysates treated with Ad-mda7 showed that MDA-7 protein expression was associated with activation of the caspase cascade. CONCLUSION: Our results show that treatment of ovarian cancer cells with mda-7/IL-24 results in growth suppression both in vitro and in vivo.


Subject(s)
Genetic Therapy , Interleukins/metabolism , Interleukins/therapeutic use , Ovarian Neoplasms/pathology , Ovarian Neoplasms/therapy , Adenoviridae/genetics , Animals , Apoptosis , Biomarkers , Caspases/metabolism , Cell Line, Tumor , Cell Proliferation , Female , Humans , Interleukins/genetics , Mice , Mice, Inbred BALB C , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Signal Transduction , Xenograft Model Antitumor Assays
9.
Curr Gene Ther ; 6(1): 73-91, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16475947

ABSTRACT

Despite recent advances in treatment strategies, the overall 5-year survival rate for patients with common epithelial cancers is poor largely because of the difficulty in treating metastatic cancers. Therefore, therapeutic agents are urgently needed that can effectively inhibit both primary epithelial tumors and their metastases. One such agent that has shown promise in preclinical studies is the tumor suppressor/cytokine, melanoma differentiation associated gene-7 also known as interleukin-24 (mda-7/IL-24). Preclinical studies from our and other laboratories have shown that overexpression of MDA-7/IL-24 causes a strong tumor- suppressive effect in many human cancer cells but spares normal cells. This gene therapy also enhances the tumor-suppressive activity of radiotherapy and chemotherapy. Secreted MDA-7 protein that is glycosylated also has been shown to have potent antiangiogenic activity both in vitro and in vivo. Studies examining the immune properties of mda-7 have shown that MDA-7/IL-24 unlike the related IL-10, functions as a Th1 cytokine. Recently, an MDA-7 protein-mediated "bystander effect" on tumor cells has been documented. Building on these findings we successfully completed a Phase I clinical trial of adenovirus-based mda-7 cancer therapy that confirmed the safety of this gene therapy. Phase II trials evaluating the efficacy of mda-7-based gene therapy are warranted. The outcome of such ongoing mda-7-based gene therapy trials will allow us to better understand this therapy's clinical utility.


Subject(s)
Genetic Therapy , Interleukins/genetics , Neoplasms/therapy , Adjuvants, Immunologic/genetics , Clinical Trials as Topic/trends , Combined Modality Therapy , Drug Evaluation, Preclinical/trends , Genetic Therapy/methods , Humans , Interleukins/immunology , Neoplasms/genetics , Neovascularization, Pathologic/genetics
10.
Cancer Gene Ther ; 12(3): 238-47, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15578066

ABSTRACT

We have previously reported that overexpression of the melanoma differentiation-associated gene -7 (mda-7) using a replication-defective adenovirus (Ad-mda7), results in tumor-specific growth suppression and induction of apoptosis in wide variety of cancer cells. In the present study, we investigated the antitumor activity of Ad-mda7 and the underlying mechanism in human prostate cancer cells and normal prostate epithelial cells. Overexpression of MDA-7 induced significant (P=.001) suppression of cell growth and apoptosis in prostate cancer cells (DU 145, LNCaP, and PC-3). In normal prostate epithelial cells (PrEC) some degree of growth inhibition but not apoptosis was observed. However, the inhibitory effects in normal cells were less compared to tumor cells. Growth inhibitory effects were mediated by the intracellular and not by extracellular MDA-7 protein. Molecular effectors that are involved in Ad-mda7-mediated tumor killing included activation of the caspase cascade, and the induction of G2 phase cell cycle arrest through the inhibition of Cdc25C pathway. These results demonstrate the mechanisms by which Ad-mda7 exerts its antitumor activity in human prostate cancer cells. The antitumor activity combined with previously reported antiangiogenic and proimmune properties of Ad-mda7 can serve as a potential therapeutic agent for treatment of primary and disseminated prostate cancer.


Subject(s)
Apoptosis/genetics , Cell Cycle/genetics , Gene Expression Regulation, Neoplastic/drug effects , Genetic Therapy/methods , Interleukins/genetics , Prostatic Neoplasms/therapy , Adenoviridae , Analysis of Variance , Annexin A5/metabolism , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Proliferation , Flow Cytometry , Gene Transfer Techniques , Genes, Tumor Suppressor , Genetic Vectors , Humans , Immunoblotting , Interleukins/pharmacology , Male , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...