Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 104
Filter
1.
Cell Death Discov ; 2: 16003, 2016.
Article in English | MEDLINE | ID: mdl-27551498

ABSTRACT

Apoptosis culminates in secondary necrosis due to lack of ATP. Cancer stem cells form spheres after apoptosis by evoking the blebbishield emergency program. Hence, determining how blebbishields avoid secondary necrosis is crucial. Here we demonstrate that N-Myc and VEGFR2 control transformation from blebbishields, during which oligomers of K-Ras, p27, BAD, Bax, and Bak boost glycolysis to avoid secondary necrosis. Non-apoptotic cancer cells also utilize oligomers to boost glycolysis, which differentiates the glycolytic function of oligomers from their apoptotic action. Smac mimetic in combination with TNF-α or TRAIL but not in combination with FasL abrogates transformation from blebbishields by inducing secondary necrosis. Thus blebbishield-mediated transformation is dependent on glycolysis, and Smac mimetics represent potential candidates to abrogate the blebbishield emergency program.

2.
Oncogene ; 35(33): 4312-20, 2016 08 18.
Article in English | MEDLINE | ID: mdl-26725326

ABSTRACT

MicroRNAs (miRNAs) are small RNA molecules that affect cellular processes by controlling gene expression. Recent studies have shown that hypoxia downregulates Drosha and Dicer, key enzymes in miRNA biogenesis, causing a decreased pool of miRNAs in cancer and resulting in increased tumor growth and metastasis. Here we demonstrate a previously unrecognized mechanism by which hypoxia downregulates Dicer. We found that miR-630, which is upregulated under hypoxic conditions, targets and downregulates Dicer expression. In an orthotopic mouse model of ovarian cancer, delivery of miR-630 using 1,2-dioleoyl-sn-glycero-3-phosphocholine (DOPC) nanoliposomes resulted in increased tumor growth and metastasis, and decreased Dicer expression. Treatment with the combination of anti-miR-630 and anti-vascular endothelial growth factor antibody in mice resulted in rescue of Dicer expression and significantly decreased tumor growth and metastasis. These results indicate that targeting miR-630 is a promising approach to overcome Dicer deregulation in cancer. As demonstrated in the study, use of DOPC nanoliposomes for anti-miR delivery serves as a better alternative approach to cell line-based overexpression of sense or antisense miRNAs, while avoiding potential in vitro selection effects. Findings from this study provide a new understanding of miRNA biogenesis downregulation observed under hypoxia and suggest therapeutic avenues to target this dysregulation in cancer.


Subject(s)
Cell Hypoxia , DEAD-box RNA Helicases/genetics , Gene Expression Regulation, Neoplastic , MicroRNAs/physiology , Neoplasms/etiology , Ribonuclease III/genetics , Animals , Cell Line, Tumor , Disease Progression , Female , Humans , Liposomes , Mice , MicroRNAs/antagonists & inhibitors , Neoplasms/therapy , Ovarian Neoplasms/metabolism , Phosphatidylcholines/administration & dosage , Vascular Endothelial Growth Factor A/antagonists & inhibitors
3.
Oncogene ; 35(20): 2675-80, 2016 05 19.
Article in English | MEDLINE | ID: mdl-26119932

ABSTRACT

It is well known that specific signal transduction inhibitors rarely suffice as anti-cancer agents. In most cases, tumors possess primary drug resistance due to their inherent heterogeneity, or acquire drug resistance due to genomic instability and acquisition of mutations. Here we expand our previous study of the novel compound, NT157, and show that it acts as a dual-targeting agent that invokes the blockage of two signal transduction pathways that are central to the development and maintenance of multiple human cancers. We show that NT157 targets not only IGF1R-IRS1/2, as previously reported, but also the Stat3 signaling pathway and demonstrates remarkable anti-cancer characteristics in A375 human melanoma cells and in a metastatic melanoma model in mice.


Subject(s)
Antineoplastic Agents/pharmacology , Melanoma/drug therapy , Molecular Targeted Therapy/methods , Pyrogallol/analogs & derivatives , Receptors, Somatomedin/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Sulfonamides/pharmacology , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Drug Discovery , Humans , Melanoma/pathology , Neoplasm Invasiveness , Pyrogallol/pharmacology , Pyrogallol/therapeutic use , Receptor, IGF Type 1 , Sulfonamides/therapeutic use
4.
Minerva Med ; 101(3): 149-62, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20562803

ABSTRACT

Changes in the adhesive properties of neoplastic cells, as well as intracellular signaling mediated by cell surface adhesion molecules, play essential roles in the development and progression of cancer. In this review, we summarize the recent progress in understanding the biology of cell adhesion molecules (CAMs) as well as their clinical significance as prognostic biomarkers or as potential therapeutic targets in a variety of malignancies.


Subject(s)
Cell Adhesion Molecules/physiology , Cell Adhesion/physiology , Neoplasm Proteins/physiology , Neoplasms/pathology , Antineoplastic Agents/therapeutic use , Cadherins/physiology , Cell Communication/physiology , Disease Progression , Humans , Immunoglobulins/physiology , Integrins/physiology , Neoplasms/drug therapy , Selectins/physiology
5.
Br J Cancer ; 99(5): 734-40, 2008 Sep 02.
Article in English | MEDLINE | ID: mdl-18728664

ABSTRACT

Metastatic melanoma cells express a number of protein tyrosine kinases (PTKs) that are considered to be targets for imatinib. We conducted a phase II trial of imatinib in patients with metastatic melanoma expressing at least one of these PTKs. Twenty-one patients whose tumours expressed at least one PTK (c-kit, platelet-derived growth factor receptors, c-abl, or abl-related gene) were treated with 400 mg of imatinib twice daily. One patient with metastatic acral lentiginous melanoma, containing the highest c-kit expression among all patients, had dramatic improvement on positron emission tomographic scan at 6 weeks and had a partial response lasting 12.8 months. The responder had a substantial increase in tumour and endothelial cell apoptosis at 2 weeks of treatment. Imatinib was fairly well tolerated: no patient required treatment discontinuation because of toxicity. Fatigue and oedema were the only grade 3 or 4 toxicities that occurred in more than 10% of the patients. Imatinib at the studied dose had minimal clinical efficacy as a single-agent therapy for metastatic melanoma. However, based on the characteristics of the responding tumour in our study, clinical activity of imatinib, specifically in patients with melanoma with certain c-kit aberrations, should be examined.


Subject(s)
Antineoplastic Agents/therapeutic use , Melanoma/drug therapy , Piperazines/therapeutic use , Pyrimidines/therapeutic use , Skin Neoplasms/drug therapy , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/adverse effects , Base Sequence , Benzamides , DNA Primers , Disease Progression , Female , Humans , Imatinib Mesylate , Male , Melanoma/blood supply , Melanoma/diagnostic imaging , Melanoma/secondary , Middle Aged , Piperazines/adverse effects , Positron-Emission Tomography , Pyrimidines/adverse effects , Skin Neoplasms/blood supply , Skin Neoplasms/diagnostic imaging , Skin Neoplasms/pathology , Treatment Outcome
6.
Oncogene ; 26(28): 4049-58, 2007 Jun 14.
Article in English | MEDLINE | ID: mdl-17224907

ABSTRACT

Activator protein-2 (AP-2) is a transcription factor that regulates proliferation and differentiation in mammalian cells and has been implicated in the acquisition of the metastatic phenotype in several types of cancer. Herein, we examine the role of AP-2alpha in colon cancer progression. We provide evidence for the lack of AP-2alpha expression in the late stages of colon cancer cells. Re-expression of the AP-2alpha gene in the AP-2alpha-negative SW480 colon cancer cells suppressed their tumorigenicity following orthotopic injection into the cecal wall of nude mice. The inhibition of tumor growth could be attributed to the increased expression of E-cadherin and decreased expression and activity of matrix-metalloproteinase-9 (MMP-9) in the transfected cells, as well as a substantial loss of their in vitro invasive properties. Conversely, targeting constitutive expression of AP-2alpha in AP-2-positive KM12C colon cancer cells with small interfering RNA resulted in an increase in their invasive potential, downregulation of E-cadherin and increased expression of MMP-9. In SW480 cells, re-expression of AP-2alpha resulted in a fourfold increase in the activity of E-cadherin promoter, and a 5-14-fold decrease in the activity of MMP-9 promoter, indicating transcriptional regulation of these genes by AP-2alpha. Chromatin immunoprecipitation assay showed that re-expressed AP-2alpha directly binds to the promoter of E-cadherin, where it has been previously reported to act as a transcriptional activator. Furthermore, chromatin immunoprecipitation assay revealed AP-2alpha binding to the MMP-9 promoter, which ensued by decreased binding of transcription factor Sp-1 and changes in the recruitment of transcription factors to a distal AP-1 element, thus, contributing to the overall downregulation of MMP-9 promoter activity. Collectively, our data provide evidence that AP-2alpha acts as a tumor suppressor gene in colon cancer..


Subject(s)
Cadherins/physiology , Colonic Neoplasms/metabolism , Matrix Metalloproteinase 9/physiology , Transcription Factor AP-2/physiology , Animals , Cell Line, Tumor , Colonic Neoplasms/enzymology , Colonic Neoplasms/pathology , Down-Regulation , Humans , Mice , Mice, Nude , Transcription Factor AP-2/genetics , Transfection , Up-Regulation
7.
Oncogene ; 26(12): 1739-47, 2007 Mar 15.
Article in English | MEDLINE | ID: mdl-16964286

ABSTRACT

Loss of the metastasis suppressor gene, KiSS-1 has been strongly correlated to the progression of metastases in numerous types of cancers. The mechanism through which KiSS-1 is lost during metastasis, however, is still not completely known. Previous studies have shown that genetic material on human chromosome 6q16.3-q23 is essential for KiSS-1 expression in normal tissues. Additionally, microcell-mediated transfer of this chromosome in cancerous tissue results in rescued expression of KiSS-1 and reduced metastatic phenotype. Here, we show that loss of Sp1-coactivator protein DRIP-130, which is encoded by human chromosome 6q16.3-q23, results in reduced KiSS-1 promoter activation in highly malignant melanoma cells. Co-expression of Sp1 and DRIP-130 not only rescues KiSS-1 expression, but also induces an inhibition of the invasive and migratory behavior in highly metastatic melanoma cells, similar to the overexpression of KiSS-1 metastasis suppressor gene in those cells. Furthermore, we demonstrate that KiSS-1 expression is regulated by Sp1 elements within the first 100-bp region of the KiSS-1 promoter and that targeted deletion of a single GC-rich region spanning -93 to -58 interrupts Sp1- and DRIP-130-modulated transcriptional control of KiSS-1 expression. Our results thus suggest that DRIP-130 is a key regulator in KiSS-1 transactivation in normal tissue, and that the loss of DRIP-130 expression, as a result of the gross loss of human chromosome 6q16.3-q23, provokes increased tumor metastasis.


Subject(s)
Gene Expression Regulation, Neoplastic/physiology , Melanoma/genetics , Sp1 Transcription Factor/physiology , Trans-Activators/physiology , Transcription, Genetic/physiology , Tumor Suppressor Proteins/genetics , Base Sequence , Blotting, Western , Cell Line, Tumor , DNA Primers , Electrophoretic Mobility Shift Assay , Humans , Kisspeptins , Mediator Complex , Melanoma/pathology , Neoplasm Metastasis , Promoter Regions, Genetic , Reverse Transcriptase Polymerase Chain Reaction
8.
Gene Ther ; 14(4): 316-23, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17024104

ABSTRACT

Melanoma patients with metastases have a very low survival rate and limited treatment options. Therefore, the targeting of melanoma cells when they begin to invade and metastasize would be beneficial. An adhesion molecule that is upregulated at the vertical growth phase is the melanoma cell adhesion molecule (MCAM/MUC18). MUC18 is expressed in late primary and metastatic melanoma with little or no expression on normal melanocytes. We utilized the alphavirus-based DNA plasmid, SINCp, encoding murine MUC18 (SINCp c-muMUC18) for vaccination against B16F10 murine melanoma cells expressing murine MUC18. This vaccine effectively protected mice from lethal challenges with melanoma-expressing murine MUC18 in both primary and metastatic tumor models. Vaccination against MUC18 elicited effective humoral and CD8+ T-cell immune responses against melanoma. We propose that targeting molecules important in tumor invasion may be useful in the design of future strategies for the prevention and treatment of melanoma.


Subject(s)
Genetic Therapy/methods , Melanoma/therapy , Skin Neoplasms/therapy , Vaccines, DNA/administration & dosage , Alphavirus/genetics , Animals , CD146 Antigen/genetics , Cell Line, Tumor , DNA Fragmentation , Flow Cytometry , Immunotherapy/methods , Lymphocyte Activation , Melanoma/immunology , Melanoma/secondary , Mice , Mice, Inbred C57BL , Neoplasm Invasiveness/immunology , Neoplasms, Experimental , Skin Neoplasms/immunology , T-Lymphocytes, Cytotoxic/immunology
9.
J Sci Med Sport ; 7(4): 454-64, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15712502

ABSTRACT

In this study, the relationship between mental training with biofeedback and swimmers' performance was investigated. The Wingate five-step approach was used as a mental preparation technique for enhancing the performance among 16-18 year-old pre-elite swimmers. Participants (n = 40) were randomly assigned to one of two conditions: (a) experimental--regular training plus the Wingate 5-step mental training program (adapted for swimming), and (b) control--regular training plus relaxing activities. After a baseline measurement, participants were tested on running and swimming five times during a 10-week period. Results indicated that the experimental group improved its performance over time on both running and swimming, with improvement being most substantial during transformation and realisation (steps 4 and 5). In contrast, the control group remained relatively stable on both dependent measures. Results are discussed in reference to previous work on the 5-step approach, including several methodological and theoretical aspects that are particularly relevant to the use of such interventions with other athletic tasks and populations.


Subject(s)
Biofeedback, Psychology/methods , Physical Education and Training/methods , Swimming/education , Swimming/psychology , Task Performance and Analysis , Adolescent , Female , Humans , Male , Running/physiology , Running/psychology , Swimming/physiology
10.
Apoptosis ; 8(4): 371-6, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12815280

ABSTRACT

We recently demonstrated that tumor necrosis factor alpha activates caspase 6, which in turn cleaves transcription factor AP-2 alpha. We mapped the cleavage site at 19 amino acids from the N-terminus at the sequence aspartate-argenine-histidine-aspartate (DRHD). Mutating aspartic acid at position 19 abrogated the cleavage site. From these observations, we hypothesized that the DRHD peptide could act as a caspase 6 inhibitor. To test this hypothesis, the peptide zAsp(Ome)-Arg-His-Asp(Ome)-fluoromethyl ketone (zDRHDfmk) was synthesized. Here we show that zDRHDfmk inhibits TNFalpha-induced caspase 6 activity and apoptosis in breast cancer cells. When compared to other caspase inhibitors, zDRHDfmk inhibited caspase 6 activity more effectively than the general caspase inhibitor zVal-Ala-Lys(Ome)-fluoromethy ketone (zVADfmk) or the caspase 6 inhibitor zVal-Glu-Ile-Asp-(Ome)-fluoromethyl ketone (zVEIDfmk). However, it was less effective in inhibiting TNFalpha-induced apoptosis than zVADfmk or zVEIDfmk, presumably because caspase 6 is only one of at least three effector caspases, the others being caspase 3 and 7, that are active during caspase-dependent apoptosis. The discovery of this sequence-based caspase 6 inhibitor provides a new tool for studying caspase 6. More importantly, it could be used, in combination with other agents, as a drug to inhibit apoptosis in neurodegenrative diseases such as Alzheimer's, Parkinson and amyotrophic lateral sclerosis.


Subject(s)
Caspase Inhibitors , Caspases/metabolism , Cysteine Proteinase Inhibitors/pharmacology , Oligopeptides/metabolism , Oligopeptides/pharmacology , Adaptor Protein Complex 2/metabolism , Amino Acid Sequence , Animals , Caspase 6 , Caspase 8 , Caspase 9 , Cell Line, Tumor , Cysteine Proteinase Inhibitors/chemistry , Humans , Oligopeptides/chemistry , Tumor Necrosis Factor-alpha/metabolism
11.
Cell Death Differ ; 10(5): 558-69, 2003 May.
Article in English | MEDLINE | ID: mdl-12728254

ABSTRACT

Several studies have shown that matrix metalloproteases (MMPs) promote tumor growth, invasion, and metastasis. Consequently, MMP inhibitors have been developed as a new class of anticancer drugs, many of which are in clinical trials. The exact mechanism of the antineoplastic activity of MMP antagonists is unknown. To investigate the mechanism, we hypothesized that MMP inhibitors enhance the actions of apoptosis-inducing agents. To test this hypothesis, we treated breast, melanoma, leukemia, osteosarcoma, and normal breast epithelial cells with (2R)-2-[(4-biphenylsulfonyl)amino]-3-phenylproprionic acid (compound 5a), an organic inhibitor of MMP-2/MMP-9, alone or in combination with TNFalpha or other apoptotic agents. FACS analysis showed that 5a interacted synergistically with ligands of the TNF receptor superfamily, including TNFalpha and TNF receptor-like apoptosis-inducing ligand (TRAIL), and with a Fas-cross-linking antibody (CH11), UV, paclitaxel, thapsigargin, and staurosporin, to induce apoptosis in a cell-type-specific manner. Other MMP inhibitors did not synergize with TNFalpha. Compound 5a did not act directly on the mitochondrion or via changes in protein synthesis. Instead, the mechanism requires ligand-receptor interaction and caspase 8 activation. Investigation of the effect of 5a on tumor growth in vivo revealed that continuous treatment of subcutaneous melanoma with a combination of 5a plus TRAIL reduced tumor growth and angiogenesis in nude mice. Our data demonstrate that 5a possesses a novel proapoptotic function, thus providing an alternative mechanism for its antineoplastic action. These observations have important implications for combination cancer therapy.


Subject(s)
Apoptosis/drug effects , Enzyme Inhibitors/pharmacology , Matrix Metalloproteinase Inhibitors , Phenylalanine/pharmacology , Sulfonamides/pharmacology , Animals , Apoptosis/physiology , Apoptosis Regulatory Proteins , Caspase 8 , Caspase 9 , Caspases/drug effects , Caspases/metabolism , Cytokines/pharmacology , Enzyme Activation/drug effects , Humans , Intracellular Membranes/drug effects , Intracellular Membranes/physiology , Ligands , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Membrane Glycoproteins/pharmacology , Membrane Potentials/drug effects , Mice , Mice, Inbred BALB C , Mice, Nude , Mitochondria/drug effects , Mitochondria/enzymology , Mitochondria/physiology , Neoplasm Transplantation , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/pathology , Phenylalanine/analogs & derivatives , Receptors, Tumor Necrosis Factor/metabolism , TNF-Related Apoptosis-Inducing Ligand , Time Factors , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/enzymology , Tumor Necrosis Factor-alpha/pharmacology , Xenograft Model Antitumor Assays
12.
Clin Cancer Res ; 7(12): 4086-95, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11751506

ABSTRACT

PURPOSE: Prostate cancer progression is associated with deregulation of genes like E-cadherin, p21/WAF1, MMP-2, VEGF, and IGF-binding protein, 3 and 5, all of which are target genes for the transcription factor activator protein 2alpha (AP-2alpha). We, therefore, hypothesize that the development/progression of prostate cancer is associated with changes in the expression of AP-2alpha. EXPERIMENTAL DESIGN: We used immunofluorescent staining to assess the presence of AP-2alpha in normal, benign, and malignant human prostate tissues and to correlate its expression with tumor grade and stage. RESULTS: We found that although AP-2alpha was expressed in normal prostate epithelium, it was not expressed in 30 prostate cancer specimens of different Gleason scores. Moreover, AP-2alpha protein was present in the luminal cell layer but not in the basal cell layer of the normal epithelium, which indicated that the loss of AP-2alpha staining in the prostate cancer specimens was not attributable to a lack of AP-2alpha-expressing cells. Further analysis demonstrated the presence of AP-2alpha in 2 (40%) of 5 atrophic normal epithelium, in 4 (24%) of 17 cases of benign prostatic hyperplasia, and in 2 (13%) of 13 cases of high-grade prostatic intraepithelial neoplasia. Loss or reduction in AP-2alpha expression was also observed in LNCaP, LNCaP-LN3, and PC3M-LN4 cell lines. CONCLUSIONS: Our data demonstrate that AP-2alpha expression is associated with normal luminal differentiation and that a loss of AP-2alpha expression occurs early in the development of prostate adenocarcinoma. Loss of AP-2alpha may lead to deregulation in AP-2alpha target genes that normally regulate cellular growth and differentiation.


Subject(s)
Biomarkers, Tumor/analysis , Cell Differentiation/physiology , DNA-Binding Proteins/analysis , Prostatic Neoplasms/pathology , Transcription Factors/analysis , Carcinoma/pathology , Disease Progression , Epithelial Cells/pathology , Fluorescent Antibody Technique , Humans , Immunohistochemistry , Keratins/analysis , Male , Prostate/chemistry , Prostate-Specific Antigen/analysis , Prostatectomy , Prostatic Hyperplasia/pathology , Prostatic Neoplasms/genetics , Reference Values , Transcription Factor AP-2 , Tumor Cells, Cultured
13.
Crit Rev Immunol ; 21(1-3): 275-86, 2001.
Article in English | MEDLINE | ID: mdl-11642609

ABSTRACT

Activating transcription factor-1 (ATF-1) and cAMP-responsive element (CRE)-binding protein (CREB) have been implicated in cAMP and Ca2+-induced transcriptional activation. The expression of the transcription factors ATF-1 and CREB is upregulated in metastatic melanoma cells. However, how overexpression of ATF-1/CREB contributes to the acquisition of the metastatic phenotype is unclear. Previously we demonstrated that quenching of CREB activity in metastatic melanoma cells by means of a dominant-negative form of CREB (KCREB) led to a decrease in their tumorigenicity and metastatic potential in nude mice. We identified two mechanisms that explain how overexpression of CREB/ATF-1 contributes to the metastatic phenotype. The first is one in which CREB/ATF-1 play an essential role in invasion by regulating the CRE-dependent expression of the metalloproteinase MMP-2 and the adhesion molecule MCAM/MUC18 genes. In the second mechanism, CREB and ATF-1 act as survival factors for human melanoma cells. Here, the effect of disrupting ATF-1 activity was investigated using intracellular expression of an inhibitory anti-ATF-1 single chain antibody fragment (ScFv). Intracellular expression of ScFv anti-ATF-1 in MeWo melanoma cells caused significant reduction in CRE-dependent promoter activation. In addition, expression of ScFv anti-ATF-1 in melanoma cells suppressed their tumorigenicity and metastatic potential in nude mice. ScFv anti-ATF-1 rendered the melanoma cells susceptible to thapsigargin-induced apoptosis in vitro and caused massive apoptosis in tumors transplanted subcutaneously into nude mice, confirming that AFT-1/CREB act as survival factors for human melanoma cells. These studies demonstrate the potential usage of ScFv anti-ATF-1 as an inhibitor of tumor growth and metastasis of solid tumors in vivo.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Cyclic AMP Response Element-Binding Protein/antagonists & inhibitors , DNA-Binding Proteins , Immunoglobulin Fragments/therapeutic use , Melanoma/therapy , Transcription Factors/antagonists & inhibitors , Activating Transcription Factor 1 , Apoptosis , Cyclic AMP Response Element-Binding Protein/physiology , Humans , Matrix Metalloproteinase 2/genetics , Melanoma/etiology , Melanoma/pathology , Transcription Factors/physiology
14.
Mol Cell Biol ; 21(15): 4856-67, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11438643

ABSTRACT

Several reports have linked activating protein 2alpha (AP-2alpha) to apoptosis, leading us to hypothesize that AP-2alpha is a substrate for caspases. We tested this hypothesis by examining the effects of tumor necrosis factor alpha (TNF-alpha) on the expression of AP-2 in breast cancer cells. Here, we provide evidence that TNF-alpha downregulates AP-2alpha and AP-2gamma expression posttranscriptionally during TNF-alpha-induced apoptosis. Both a general caspase antagonist (zVADfmk) and a caspase 6-preferred antagonist (zVEIDfmk) inhibited TNF-alpha-induced apoptosis and AP-2alpha downregulation. In vivo tests showed that AP-2alpha was cleaved by caspases ahead of the DNA fragmentation phase of apoptosis. Recombinant caspase 6 cleaved AP-2alpha preferentially, although caspases 1 and 3 also cleaved it, albeit at 50-fold or higher concentrations. Activated caspase 6 was detected in TNF-alpha-treated cells, thus confirming its involvement in AP-2alpha cleavage. All three caspases cleaved AP-2alpha at asp(19) of the sequence asp-arg-his-asp (DRHD(19)). Mutating D(19) to A(19) abrogated AP-2alpha cleavage by all three caspases. TNF-alpha-induced cleavage of AP-2alpha in vivo led to AP-2alpha degradation and loss of DNA-binding activity, both of which were prevented by pretreatment with zVEIDfmk. AP-2alpha degradation but not cleavage was inhibited in vivo by PS-431 (a proteasome antagonist), suggesting that AP-2alpha is degraded subsequent to cleavage by caspase 6 or caspase 6-like enzymes. Cells transfected with green fluorescent protein-tagged mutant AP-2alpha are resistant to TNF-alpha-induced apoptosis, further demonstrating the link between caspase-mediated cleavage of AP-2alpha and apoptosis. This is the first report to demonstrate that degradation of AP-2alpha is a critical event in TNF-alpha-induced apoptosis. Since the DRHD sequence in vertebrate AP-2 is widely conserved, its cleavage by caspases may represent an important mechanism for regulating cell survival, proliferation, differentiation, and apoptosis.


Subject(s)
Breast Neoplasms/pathology , Caspases/metabolism , Cysteine Endopeptidases/metabolism , DNA-Binding Proteins/metabolism , Multienzyme Complexes/metabolism , Transcription Factors/metabolism , Tumor Necrosis Factor-alpha/metabolism , Amino Acid Chloromethyl Ketones/pharmacology , Amino Acid Sequence , Animals , Apoptosis , Base Sequence , Blotting, Western , Breast Neoplasms/metabolism , Caspase 6 , Cell Death , DNA/metabolism , DNA Fragmentation , Dose-Response Relationship, Drug , Down-Regulation , Enzyme Inhibitors/pharmacology , Flow Cytometry , Green Fluorescent Proteins , Humans , In Situ Nick-End Labeling , Luminescent Proteins/metabolism , Molecular Sequence Data , Mutagenesis, Site-Directed , Mutation , Peptides/pharmacology , Proteasome Endopeptidase Complex , RNA Processing, Post-Transcriptional , Recombinant Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Transcription Factor AP-2 , Transcription, Genetic , Transfection , Tumor Cells, Cultured
15.
Oncogene ; 20(26): 3363-75, 2001 Jun 07.
Article in English | MEDLINE | ID: mdl-11423987

ABSTRACT

We have previously demonstrated that the transition of melanoma to the metastatic phenotype is associated with a loss of expression of the transcription factor AP-2. To further investigate the role of AP-2 in the progression of human melanoma, we attempted to inactivate AP-2 in primary cutaneous SB-2 melanoma cells by using a dominant-negative AP-2, or AP-2B, gene. AP-2B is an alternatively spliced AP-2 variant capable of inhibiting AP-2 trans-activator function. Stable transfection of primary cutaneous melanoma SB-2 cells with the dominant-negative AP-2B gene was confirmed by RT--PCR and Northern blot analyses. Electromobility shift assay using nuclear extracts from these cell lines demonstrated decreased functional binding of AP-2B-transfected cells to the AP-2 consensus binding sequence compared with neo-transfected controls. In addition, CAT activity driven by a construct containing the AP-2 consensus binding sequence was downregulated in the AP-2B transfected cells, indicating AP-2 activity was quenched in the transfected cells. Orthotopic (subcutaneous) injection of the dominant-negative (AP-2B)-transfected cell lines into nude mice increased their tumorigenicity compared to control neo-transfected cells. The AP-2B-transfected cells displayed an increase in MMP-2 expression (by Northern blot) and MMP-2 activity (by zymography), which resulted in an increase in invasiveness through Matrigel-coated filters. The AP-2B-transfected tumors also displayed an increase in MMP-2 expression, microvessel density, and angiogenesis in vivo. These results demonstrate that inactivation of AP-2 contributes to the progression of melanoma, at least partially via deregulation of the MMP-2 gene.


Subject(s)
DNA-Binding Proteins/physiology , Genes, Dominant , Melanoma, Experimental/pathology , Melanoma/pathology , Skin Neoplasms/pathology , Transcription Factors/physiology , Alternative Splicing , Animals , Antigens, Neoplasm/analysis , Blotting, Northern , Cell Movement , Collagen , Culture Media , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Drug Combinations , Genes, Reporter , Humans , Laminin , Lung Neoplasms/blood supply , Lung Neoplasms/enzymology , Lung Neoplasms/secondary , Lymphatic Metastasis , Male , Matrix Metalloproteinase 2/biosynthesis , Matrix Metalloproteinase 2/genetics , Melanoma/genetics , Melanoma, Experimental/blood supply , Melanoma, Experimental/enzymology , Melanoma, Experimental/secondary , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplasm Transplantation , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/pathology , Platelet Endothelial Cell Adhesion Molecule-1/analysis , Proteoglycans , Reverse Transcriptase Polymerase Chain Reaction , Skin Neoplasms/genetics , Transcription Factor AP-2 , Transcription Factors/deficiency , Transcription Factors/genetics , Transfection
16.
Pigment Cell Res ; 14(2): 78-85, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11310795

ABSTRACT

The molecular changes associated with the transition of melanoma cells from radial growth phase (RGP) to vertical growth phase [(VGP), metastatic phenotype] are not very well defined. We previously demonstrated that expression of the cell-surface adhesion molecule MCAM/MUC18 correlates directly with the metastatic potential of human melanoma cells. In addition, the progression of human melanoma towards the metastatic phenotype is associated with loss of expression of the tyrosine-kinase receptor c-KIT. In this review, I will summarize our recent studies demonstrating that the expression of both genes is regulated by the AP-2 transcription factor. Moreover, we have observed a loss of AP-2 expression in metastatic melanoma cells. Re-expression of AP-2 in the highly metastatic A375SM cells decreased their tumorigenicity and inhibited their metastatic potential in nude mice. MCAM/MUC18 mRNA and protein expression was significantly down-regulated while c-KIT expression was up-regulated in the AP-2-transfected cells. To further investigate the role of AP-2 in the progression of human melanoma, we attempted to inactivate AP-2 in primary cutaneous melanoma by using a dominant-negative AP-2, or the AP-2B gene. Expression of AP-2B in SB-2 cells augmented their tumorigenicity in nude mice, and upregulated MMP-2 expression and activity. As AP-2 also regulates other genes that are involved in the progression of human melanoma such as E-cadherin, p21/WAF-1, HER2/neu, Bcl-2, FAS/APO-1, IGF-R-1, VEGF and the thrombin receptor (PAR-1), we therefore propose that loss of AP-2 is a crucial event in the development of malignant melanoma. In addition, the transition of melanoma cells from RGP to VGP is also associated with over-expression of the transcription factors CREB and ATF-1. The notion that the balance between AP-2 and CREB/ATF-1 expression determines the progression of melanoma cells towards the metastatic phenotype will be discussed.


Subject(s)
Antigens, CD , DNA-Binding Proteins/physiology , Gene Expression Regulation, Neoplastic , Melanoma/genetics , Melanoma/pathology , Membrane Glycoproteins , Neural Cell Adhesion Molecules , Transcription Factors/physiology , Animals , Antigens, Surface/genetics , Antigens, Surface/metabolism , CD146 Antigen , Humans , Melanoma/drug therapy , Neoplasm Metastasis/genetics , Proto-Oncogene Proteins c-kit/genetics , Proto-Oncogene Proteins c-kit/metabolism , Transcription Factor AP-2
17.
Mol Cell Biochem ; 212(1-2): 19-28, 2000 Sep.
Article in English | MEDLINE | ID: mdl-11108132

ABSTRACT

The purpose of this study was to determine the role of CREB and its associated proteins in melanoma progression. We used MeWo human melanoma cells transfected with a dominant negative construct of CREB, KCREB. KCREB has a mutation in its DNA-binding domain and can not bind the CRE element. Expression of KCREB yields proper heterodimerization with CREB and its associated proteins, but the proteins associated with KCREB do not confer the same degree of transcriptional activity as they would in the case of wild-type CREB. Here, we demonstrate that expression of KCREB in MeWo melanoma cells leads to a decrease in their tumorigenicity and metastatic potential in nude mice. We identified two mechanisms that explain at least partially this effect of KCREB. The first, is one in which CREB and its associated proteins play an essential role in invasion. We showed that the invasive properties of KCREB-transfected MeWo cells were reduced due to the downregulation of the CRE-dependent expression of the type IV collagenase MMP-2 and the adhesion molecule MCAM/MUC18. In the second mechanism, CREB and its associated proteins act as survival factors for human melanoma cells. Here we demonstrated that expression of KCREB in MeWo cells rendered them susceptible to apoptosis induced by thapsigargin, which in turn increased the intracellular level of Ca2+. Thapsigargin induced CREB and ATF-1 phosphorylation and activated CRE-dependent transcription in MeWo cells. Collectively, our data demonstrate that CREB and its associated proteins play an important role in tumor growth and metastasis of human melanoma.


Subject(s)
Cyclic AMP Response Element-Binding Protein/metabolism , Melanoma/pathology , Transcription Factors/metabolism , Animals , Apoptosis , Cell Division , Cyclic AMP Response Element-Binding Protein/chemistry , Cyclic AMP Response Element-Binding Protein/genetics , Genes, Reporter , Humans , Male , Melanoma/genetics , Melanoma/metabolism , Mice , Mice, Nude , Neoplasm Invasiveness , Neoplasm Metastasis , Transfection , Transplantation, Heterologous , Tumor Cells, Cultured
18.
Oncogene ; 19(22): 2721-30, 2000 May 18.
Article in English | MEDLINE | ID: mdl-10851072

ABSTRACT

Activating transcription factor-1 (ATF-1) and cAMP-responsive element (CRE)-binding protein (CREB) have been implicated in cAMP and Ca2+-induced transcriptional activation. The expression of the transcription factors CREB and ATF-1 is upregulated in metastatic melanoma cells. However, how overexpression of ATF-1/CREB contributes to the acquisition of the metastatic phenotype remains unclear. Here, the effect of disrupting ATF-1 activity was investigated using intracellular expression of an inhibitory anti-ATF-1 single chain antibody fragment (ScFv). Intracellular expression of ScFv anti-ATF-1 in MeWo melanoma cells caused significant reduction in CRE-dependent promoter activation. In addition, expression of ScFv anti-ATF-1 in melanoma cells suppressed their tumorigenicity and metastatic potential in nude mice. ScFv anti-ATF-1 rendered the melanoma cells susceptible to thapsigargin-induced apoptosis in vitro and caused massive apoptosis in tumors transplanted subcutaneously into nude mice, suggesting that ATF-1 and its associated proteins act as survival factor for human melanoma cells. This is the first report to demonstrate the potential of ScFv anti-ATF-1 as an inhibitor of tumor growth and metastasis of solid tumor in vivo. Oncogene (2000).


Subject(s)
DNA-Binding Proteins , Gene Expression Regulation, Neoplastic , Immunoglobulin Fragments/genetics , Immunoglobulin Variable Region/genetics , Melanoma/genetics , Melanoma/pathology , Transcription Factors/genetics , Transcription Factors/immunology , Activating Transcription Factor 1 , Animals , Gene Expression Regulation, Neoplastic/immunology , Humans , Immunoglobulin Fragments/immunology , Melanoma/immunology , Mice , Neoplasm Metastasis/genetics , Recombinant Proteins/genetics , Transcription Factors/antagonists & inhibitors , Transcription, Genetic , Tumor Cells, Cultured
19.
Clin Cancer Res ; 6(5): 2104-19, 2000 May.
Article in English | MEDLINE | ID: mdl-10815938

ABSTRACT

Interleukin 8 (IL-8) is mitogenic and chemotactic for endothelial cells. Within a neoplasm, IL-8 is secreted by inflammatory and neoplastic cells. The highly metastatic PC-3M-LN4 cell line overexpresses IL-8 relative to the poorly metastatic PC-3P cell line. We evaluated whether IL-8 expression by human prostate cancer growing within the prostate of athymic nude mice regulates tumor angiogenesis, growth, and metastasis. PC-3P cells were transfected with the full-length sense IL-8 cDNA, whereas PC-3M-LN4 cells were transfected with the full-sequence antisense IL-8 cDNA. Control cells were transfected with the neomycin resistance gene (Neo). In vitro, sense-transfected PC-3P cells overexpressed IL-8-specific mRNA and protein, which resulted in up-regulation of matrix metalloproteinase 9 (MMP-9) mRNA, and collagenase activity, resulting in increased invasion through Matrigel. After antisense transfection of the PC-3M-LN4 cells, IL-8 and MMP-9 expression, collagenase activity, and invasion were markedly reduced relative to controls. After orthotopic implantation, the sense-transfected PC-3P cells were highly tumorigenic and metastatic, with significantly increased neovascularity and IL-8 expression compared with either PC-3P cells or controls. Antisense transfection significantly reduced the expression of IL-8 and MMP-9 and tumor-induced neovascularity, resulting in inhibition of tumorigenicity and metastasis. These results demonstrate that IL-8 expression regulates angiogenesis in prostate cancer, in part by induction of MMP-9 expression, and subsequently regulates the growth and metastasis of human prostate cancer.


Subject(s)
Interleukin-8/genetics , Prostatic Neoplasms/genetics , Androgens/physiology , Animals , Blotting, Northern , Chloramphenicol O-Acetyltransferase/genetics , Chloramphenicol O-Acetyltransferase/metabolism , Collagenases/metabolism , Endothelial Growth Factors/genetics , Endothelial Growth Factors/metabolism , Fibroblast Growth Factor 2/genetics , Fibroblast Growth Factor 2/metabolism , Gene Expression Regulation, Neoplastic , Humans , Immunochemistry , In Situ Hybridization , Interleukin-8/metabolism , Lymphatic Metastasis , Lymphokines/genetics , Lymphokines/metabolism , Male , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Invasiveness , Neovascularization, Pathologic , Promoter Regions, Genetic/genetics , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transfection , Tumor Cells, Cultured , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
20.
Cancer Res ; 60(8): 2290-9, 2000 Apr 15.
Article in English | MEDLINE | ID: mdl-10786697

ABSTRACT

Interleukin 8 (IL-8) is mitogenic and chemotactic for endothelial cells. Within a neoplasm, IL-8 is secreted by inflammatory and neoplastic cells. The highly tumorigenic and highly metastatic human transitional cell carcinoma (TCC) cell line 253J B-V overexpresses IL-8 relative to the nontumorigenic and nometastatic 253J-P cell line. To determine whether IL-8 expression regulates tumorigenicity and metastasis in human TCC, 253J B-V cells were transfected with the full-sequence antisense (AS) cDNA for IL-8, whereas 253J-P cells were transfected with the full-length IL-8 cDNA, and control cells for each were transfected with the neomycin resistance (Neo) gene. In vitro, sense-transfected 253J-P cells overexpressed IL-8-specific mRNA and protein, whereas both of these were markedly reduced in AS-IL-8-transfected 253J B-V cells relative to controls. Moreover, sense-transfected cells showed up-regulation in matrix metalloproteinase type 9 mRNA, collagenase activity, and increased invasiveness through Matrigel-coated filters, whereas these measures were lower in AS-transfected cells relative to controls. After implantation into the bladders of athymic nude mice, the sense-transfected 253J-P cells acquired increased tumorigenicity and metastasis, whereas the AS-transfected cells significantly inhibited tumorigenicity and metastases in the 253J B-V cell lines. This effect was accompanied by reduced IL-8 expression and microvessel density. These studies demonstrate that IL-8 expression enhances angiogenic activity through the induction of matrix metalloproteinase type 9 and subsequently regulates the tumorigenesis and production of spontaneous metastases of human TCC.


Subject(s)
Carcinoma, Transitional Cell/pathology , Interleukin-8/metabolism , Lymphatic Metastasis , Neovascularization, Pathologic , Urinary Bladder Neoplasms/pathology , Animals , Carcinoma, Transitional Cell/blood supply , Carcinoma, Transitional Cell/metabolism , Carcinoma, Transitional Cell/secondary , Collagen/metabolism , Collagenases/metabolism , Drug Combinations , Endothelial Growth Factors/genetics , Endothelial Growth Factors/metabolism , Fibroblast Growth Factor 2/genetics , Fibroblast Growth Factor 2/metabolism , Gene Expression Regulation, Neoplastic , Humans , Interleukin-8/genetics , Laminin/metabolism , Lymphokines/genetics , Lymphokines/metabolism , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Nude , Neoplasm Invasiveness , Neoplasm Transplantation , Promoter Regions, Genetic/genetics , Proteoglycans/metabolism , RNA Stability , RNA, Antisense/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured , Urinary Bladder Neoplasms/blood supply , Urinary Bladder Neoplasms/metabolism , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
SELECTION OF CITATIONS
SEARCH DETAIL