Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Methods Mol Biol ; 1513: 83-100, 2017.
Article in English | MEDLINE | ID: mdl-27807832

ABSTRACT

Tumor necrosis factor-α-inducible protein 8 (TNFAIP8) is the first discovered oncogenic and an anti-apoptotic member of a conserved TNFAIP8 or TIPE family of proteins. TNFAIP8 mRNA is induced by NF-kB, and overexpression of TNFAIP8 has been correlated with poor prognosis in many cancers. Downregulation of TNFAIP8 expression has been associated with decreased pulmonary colonization of human tumor cells, and enhanced sensitivities of tumor xenografts to radiation and docetaxel. Here we have investigated the effects of depletion of TNFAIP8 on the mRNA, microRNA and protein expression profiles in prostate and breast cancers and melanoma. Depending on the tumor cell type, knockdown of TNFAIP8 was found to be associated with increased mRNA expression of several antiproliferative and apoptotic genes (e.g., IL-24, FAT3, LPHN2, EPHA3) and fatty acid oxidation gene ACADL, and decreased mRNA levels of oncogenes (e.g., NFAT5, MALAT1, MET, FOXA1, KRAS, S100P, OSTF1) and glutamate transporter gene SLC1A1. TNFAIP8 knockdown cells also exhibited decreased expression of multiple onco-proteins (e.g., PIK3CA, SRC, EGFR, IL5, ABL1, GAP43), and increased expression of the orphan nuclear receptor NR4A1 and alpha 1 adaptin subunit of the adaptor-related protein complex 2 AP2 critical to clathrin-mediated endocytosis. TNFAIP8-centric molecules were found to be predominately implicated in the hypoxia-inducible factor-1α (HIF-1α) signaling pathway, and cancer and development signaling networks. Thus TNFAIP8 seems to regulate the cell survival and cancer progression processes in a multifaceted manner. Future validation of the molecules identified in this study is likely to lead to new subset of molecules and functional determinants of cancer cell survival and progression.


Subject(s)
Apoptosis Regulatory Proteins/genetics , Breast Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Melanoma/genetics , Prostatic Neoplasms/genetics , Proteomics/methods , Amino Acid Sequence , Apoptosis Regulatory Proteins/antagonists & inhibitors , Apoptosis Regulatory Proteins/metabolism , Breast Neoplasms/diagnosis , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Survival , Disease Progression , Excitatory Amino Acid Transporter 3/genetics , Excitatory Amino Acid Transporter 3/metabolism , Female , Humans , Male , Melanoma/diagnosis , Melanoma/metabolism , Melanoma/pathology , NF-kappa B/genetics , NF-kappa B/metabolism , Oncogene Proteins/genetics , Oncogene Proteins/metabolism , Prognosis , Prostatic Neoplasms/diagnosis , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction
2.
Int J Cancer ; 133(1): 31-42, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23280553

ABSTRACT

TNFAIP8 is a NF-κB-inducible, oncogenic molecule. Previous "promoter array" studies have identified differential methylation and regulation of TNFAIP8 in prostate epithelial and cancer cell lines. Here we demonstrate that TNFAIP8 expression is induced by androgen in hormone-responsive LNCaP prostate cancer cells. In athymic mice bearing hormone-refractory PC-3 prostate tumor xenografts, intravenous treatment with a liposomal formulation of TNFAIP8 antisense oligonucleotide (LE-AS5) caused reduced expression of TNFAIP8 in tumor tissues, and a combination of LE-AS5 and radiation or docetaxel treatment resulted in significant inhibition of PC-3 tumor growth as compared to single agents. The immunohistochemical evaluation of TNFAIP8 expression revealed correlation of both cytoplasmic and nuclear TNFAIP8 overexpression with high grade prostatic adenocarcinomas, while nuclear overexpression was found to be an independent predictor of disease recurrence controlling for tumor grade. Increased nuclear TNFAIP8 expression was statistically significantly associated with a 2.44 fold (95 % confidence interval: 1.01-5.91) higher risk of prostate cancer recurrence. Mechanistically, TNFAIP8 seems to function as a scaffold (or adaptor) protein. In the antibody microarray analysis of proteins associated with the TNFAIP8 immune-complex, we have identified Karyopherin alpha2 as a novel binding partner of nuclear TNFAIP8 in PC-3 cells. The Ingenuity Pathway Analysis of the TNFAIP8 interacting proteins suggested that TNFAIP8 influences cancer progression pathways and networks involving integrins and matrix metalloproteinases. Taken together, present studies demonstrate that TNFAIP8 is a novel therapeutic target in prostate cancer, and indicate a potential relationship of the nuclear trafficking of TNFAIP8 with adverse outcomes in a subset of prostate cancer patients.


Subject(s)
Adenocarcinoma/drug therapy , Adenocarcinoma/radiotherapy , Antineoplastic Agents/therapeutic use , Apoptosis Regulatory Proteins/metabolism , Biomarkers, Tumor/metabolism , Oligonucleotides, Antisense/therapeutic use , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/radiotherapy , Taxoids/therapeutic use , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Animals , Apoptosis Regulatory Proteins/genetics , Cell Line, Tumor , Chemotherapy, Adjuvant , Disease Progression , Docetaxel , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , Immunohistochemistry , Liposomes , Male , Mice , Mice, Nude , Neoplasm Grading , Oligonucleotides, Antisense/chemical synthesis , Predictive Value of Tests , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Protein Array Analysis , Radiotherapy, Adjuvant , Transplantation, Heterologous , Up-Regulation
3.
Methods Mol Biol ; 480: 65-83, 2009.
Article in English | MEDLINE | ID: mdl-19085118

ABSTRACT

By virtue of their potential to selectively silence oncogenic molecules in cancer cells, antisense oligonucleotides (ASO) and small interfering RNAs (siRNAs) are powerful tools for development of tailored anti-cancer drugs. The clinical benefit of ASO/siRNA therapeutic is, however, hampered due to poor pharmacokinetics and biodistribution, and suboptimal suppression of the target in tumor tissues. Raf-1 protein serine/threonine kinase is a druggable signaling molecule in cancer therapy. Our laboratory has developed cationic liposomes for systemic delivery of raf ASO (LErafAON) and raf siRNA (LErafsiRNA) to human tumor xenografts grown in athymic mice. LErafAON is also the first ASO containing liposomal drug tested in humans. In this article, we primarily focus on a modified formulation of systemically delivered cationic liposomes containing raf antisense oligonucleotide (md-LErafAON). The cationic liposomes were prepared using dimyristoyl 1,2-diacyl-3-trimethylammonium-propane (DMTAP), phosphatidylcholine (PC), and cholesterol (CHOL). The toxicology, pharmacokinetics, biodistribution, target selectivity, and anti-tumor efficacy studies of md-LErafAON were conducted in mice. We demonstrate that md-LErafAON is the next generation of systemically delivered and well-tolerated antisense therapeutic suitable for clinical evaluation.


Subject(s)
Drug Evaluation, Preclinical , Nanoparticles , Oligonucleotides, Antisense/administration & dosage , RNA, Small Interfering/administration & dosage , Animals , Base Sequence , Cations , Gene Silencing , Humans , Liposomes , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Oligonucleotides, Antisense/chemistry , Oligonucleotides, Antisense/pharmacokinetics , RNA, Small Interfering/pharmacokinetics , Tissue Distribution
4.
Int J Mol Med ; 19(1): 29-39, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17143545

ABSTRACT

Raf-1 protein serine/threonine kinase plays an important role in ERK signal transduction pathway of cell survival and proliferation. Raf-induced transcriptional changes are dependent on phosphorylation/activation of ERK. However, regulation of phospho-ERK (p-ERK) via Raf transcriptome is as yet unknown. We report the initial characterization of BRCC3, a novel gene discovered previously by mRNA expression profiling in MDA-MB 231 human breast cancer cells treated with Raf antisense oligonucleotide. BRCC3 is localized at human chromosome 5q12.1. BRCC3 open reading frame consists of 529 amino acids, coding for an approximate 60-kDa predominantly membrane-associated protein. Expression levels of BRCC3 mRNA and protein are high during G2/M phase of the cell cycle in breast cancer cells. Treatment of MDA-MB 231 cells with Raf-1 siRNA resulted in decreased expression of Raf-1, BRCC3 and p-ERK, but not B-Raf. Transient or stable expression of the epitope-tagged BRCC3 cDNA was associated with increased p-ERK in three different cell lines. Consistently, BRCC3 siRNA treatment of MDA-MB 231 cells caused decreased expression of BRCC3 and p-ERK. Furthermore, exogenous BRCC3 expression was associated with a delay in etoposide-induced cell death and an increase in cell proliferation. These findings demonstrate that BRCC3 is a novel effector of Raf-1, and implicate a role of BRCC3 in modulation of p-ERK, cell survival and proliferation.


Subject(s)
Cell Cycle Proteins/genetics , Cell Cycle Proteins/physiology , Extracellular Signal-Regulated MAP Kinases/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Amino Acid Sequence , Animals , COS Cells , Cell Cycle , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Proliferation , Cell Survival , Chlorocebus aethiops , Chromosome Mapping , Deubiquitinating Enzymes , HeLa Cells , Humans , Molecular Sequence Data , Proto-Oncogene Proteins c-raf/metabolism , RNA, Small Interfering , Transfection
5.
Int J Oncol ; 29(4): 861-7, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16964381

ABSTRACT

Raf-1 serine/threonine protein kinase plays an important role in cell growth, differentiation and cell survival. Recent reports using c-raf-1 gene-knockouts have observed MEK/ERK independent functions of Raf-1 in cell survival and protection from apoptosis. Raf-1 has also been shown to be involved in counteracting specific apoptotic pathways by restraining caspase activation, although the precise mechanism is unknown. XIAP is a potent inhibitor of apoptosis that blocks both the mitochondria and death receptor mediated pathways of apoptosis by directly binding to and inhibiting the initiator and effector caspases. In our efforts to understand the mechanism by which Raf-1 inhibits caspase activation, we discovered a novel interaction between Raf-1 and XIAP. In this study, we describe the physical interaction between Raf-1 and XIAP in vitro and in vivo in mammalian cells. We also demonstrate that Raf-1 phosphorylates XIAP in vitro and in vivo. Additionally, Raf-1 prevents XIAP degradation in response to different apoptotic triggers. Our studies identify XIAP as a new substrate of Raf-1 and provide potentially important insight into mechanisms underlying Raf-1 effects on cell survival.


Subject(s)
Proto-Oncogene Proteins c-raf/metabolism , X-Linked Inhibitor of Apoptosis Protein/metabolism , Apoptosis , Humans , Immunoprecipitation , Proto-Oncogene Proteins c-raf/analysis , Tumor Cells, Cultured , X-Linked Inhibitor of Apoptosis Protein/analysis
6.
Int J Mol Med ; 17(3): 457-63, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16465392

ABSTRACT

Raf-1 protein serine-threonine kinase plays an important role in cell growth, proliferation, and cell survival. Previously, we and others have demonstrated that antisense raf oligonucleotide-mediated inhibition of Raf-1 expression leads to tumor growth arrest, radiosensitization and chemosensitization in vivo. Raf-1 inhibition is also associated with apoptotic cell death. In this study, we inhibited Raf-1 using an antisense raf oligonucleotide (AS-raf-ODN) to identify downstream targets of Raf-1 using microarray gene expression analysis. Treatment of MDA-MB-231 breast cancer cells with 250 nM AS-raf-ODN led to significant inhibition of Raf-1 protein (75.2 +/- 9.6%) and c-raf-1 mRNA levels (86.2 +/- 3.3%) as compared to untreated control cells. The lipofectin control or mismatch oligonucleotide had no effect on Raf-1 expression. To determine the changes in gene expression profiles that were due to inhibition of Raf-1, we simultaneously compared the gene expression patterns in AS-raf-ODN treated cells with untreated control cells and cells treated with lipofectin alone or MM-ODN. A total of 17 genes (4 upregulated and 13 down-regulated) including c-raf-1 were identified that were altered after AS-raf-ODN treatment. Functional clustering analysis revealed genes involved in apoptosis (Bcl-XL), cell adhesion (paxillin, plectin, Rho GDIalpha, CCL5), metabolism (GM2A, SLC16A3, PYGB), signal transduction (protein kinase C nu), and transcriptional regulation (HMGA1), and membrane-associated genes (GNAS, SLC16A3). Real-time PCR, Northern analysis and Western analysis confirmed the microarray findings. Our study provides insight into Raf-1 related signaling pathways and a model system to identify potential target genes.


Subject(s)
Breast Neoplasms/genetics , Gene Expression Profiling , Proto-Oncogene Proteins c-raf/antagonists & inhibitors , Blotting, Northern , Blotting, Western , Breast Neoplasms/enzymology , Breast Neoplasms/pathology , Cell Line, Tumor , Cells, Cultured , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Oligonucleotide Array Sequence Analysis , Oligonucleotides, Antisense/pharmacology , Proto-Oncogene Proteins c-raf/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction
7.
Mol Ther ; 13(5): 947-55, 2006 May.
Article in English | MEDLINE | ID: mdl-16455304

ABSTRACT

SCC-S2/GG2-1/NDED (approved gene symbol TNFAIP8) is a transcription factor NF-kappaB-inducible, antiapoptotic, and oncogenic molecule. In this study, we examined the role of SCC-S2 in invasion and experimental metastasis. We demonstrate that expression of SCC-S2 cDNA in MDA-MB 435 human breast cancer cells is associated with enhanced invasion in vitro and increased frequency of pulmonary colonization of tumor cells in athymic mice. Systemic treatment of athymic mice with a cationic liposomal formulation of SCC-S2 antisense oligo led to decreased incidence of pulmonary metastasis and inhibition of SCC-S2 expression in vivo. Antisense inhibition of endogenous SCC-S2 expression correlated with decreased expression of VEGF receptor-2 in tumor cells and human lung microvascular endothelial cells and loss of endothelial cell viability. In addition, downregulation of SCC-S2 expression in tumor cells was associated with decreased expression of known metastasis-related molecules MMP-1 and MMP-9. These results demonstrate a novel role for SCC-S2 in tumor progression, involving multiple effectors, and provide a basis for SCC-S2-targeted cancer gene therapy.


Subject(s)
Breast Neoplasms/pathology , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Metalloproteases/metabolism , Proteins/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism , Animals , Apoptosis Regulatory Proteins , Cell Line, Tumor , Female , Humans , Liposomes , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Lung Neoplasms/secondary , Matrix Metalloproteinase 1/genetics , Matrix Metalloproteinase 1/metabolism , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Metalloproteases/genetics , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Invasiveness , Neoplasm Metastasis/drug therapy , Neoplasm Transplantation , Oligonucleotides, Antisense/therapeutic use , Proteins/genetics , Transplantation, Heterologous , Vascular Endothelial Growth Factor Receptor-2/genetics
8.
Int J Oncol ; 24(5): 1181-8, 2004 May.
Article in English | MEDLINE | ID: mdl-15067340

ABSTRACT

Raf-1 protein kinase plays an important role in cell growth, proliferation and cell survival. We have previously described the use of liposome-entrapped antisense raf oligonucleotide (LErafAON) to inhibit Raf-1 expression resulting in tumor growth inhibition and radiosensitization. The present study was undertaken to evaluate the chemosensitization effects of LErafAON in combination with doxorubicin or paclitaxel on a panel of human tumor xenografts. LErafAON (25.0 mg/kg i.v. x 10) displayed significant antitumor activity (P<0.05) when administered as a single agent in prostate (PC-3), lung (A549) and breast (MDA-MB 231) carcinoma models. Doxorubicin (1.0-4.0 mg/kg i.v. per week x 3) and paclitaxel (1.0-4.0 mg/kg i.v. on alternate days x 3) were administered as single agents at non-toxic doses that led to only minimal to moderate antitumor activity. However, a combination of LErafAON with doxorubicin or paclitaxel led to significantly enhanced antitumor activity in all the tumor types tested (PC-3, P<0.03; A549, P<0.035; MDA-MB 231, P<0.045) as compared with LErafAON alone or chemotherapeutic agents alone treated groups. This effect of chemosensitization appeared to be sequence-specific because a mismatch control oligonucleotide continued to show significant tumor growth. Additionally, no inhibition in Raf-1 expression in MDA-MB 231 tumor tissue was observed with mismatch oligonucleotide treatment. On the other hand, LErafAON treatment led to >75% inhibition of Raf-1 expression in tumor tissue. These preclinical observations support the use of LErafAON in combination with chemotherapeutic agents to improve the treatment of human cancers.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/drug therapy , Lung Neoplasms/drug therapy , Prostatic Neoplasms/drug therapy , Proto-Oncogene Proteins c-raf/genetics , Xenograft Model Antitumor Assays/methods , Animals , Breast Neoplasms/genetics , Doxorubicin/administration & dosage , Female , Humans , Liposomes , Lung Neoplasms/genetics , Male , Mice , Mice, Nude , Oligonucleotides, Antisense/administration & dosage , Paclitaxel/administration & dosage , Prostatic Neoplasms/genetics , Proto-Oncogene Proteins c-raf/metabolism , Tumor Cells, Cultured/transplantation
SELECTION OF CITATIONS
SEARCH DETAIL
...