Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters











Publication year range
1.
Front Biosci ; 13: 2293-8, 2008 Jan 01.
Article in English | MEDLINE | ID: mdl-17981711

ABSTRACT

During the last decade, sphingolipid deregulation, namely the balance between the pro-apoptotic molecule ceramide and the anti-apoptotic sphingolipid sphingosine-1-phosphate, has emerged as an important factor in cancer pathology and resistance to therapy. Thus, our research has been focused on developing drugs that are able to restore normal sphingolipid balance, precisely through increasing the levels of ceramide and decreasing sphingosine-1-phosphate. Particularly, inhibition of the ceramide metabolizing enzyme acid ceramidase, whose over-expression in cancer cells has been implicated in resistance to treatment, is proving to be an efficient and promising strategy. In this review, we consider our recent work with acid ceramidase inhibitors, in combination with radiation or gene therapy as a sensitizer that enhance cancer therapy.


Subject(s)
Enzyme Inhibitors/pharmacology , Galactosylgalactosylglucosylceramidase/antagonists & inhibitors , Genetic Therapy/methods , Neoplasms/drug therapy , Neoplasms/radiotherapy , Neoplasms/therapy , Animals , Antineoplastic Agents/therapeutic use , Chicken anemia virus/genetics , Fas Ligand Protein/metabolism , Genetic Vectors/metabolism , Humans , Neoplasms/metabolism
2.
Mol Ther ; 15(7): 1259-63, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17426710

ABSTRACT

Head and neck squamous cell cancers (HNSCC) are particularly aggressive and are resistant to many forms of treatment. Ceramide metabolism has been shown to play an important role in cancer progression and cancer resistance to therapy in many tumor models, including HNSCC. Here, we study the role of the ceramide-metabolizing enzyme acid ceramidase (AC) in therapeutic responses in HNSCC. First, we show that AC is over-expressed in 70% of head and neck squamous cell tumors compared with normal tissues, suggesting that this enzyme may play an important role in facilitating HNSCC growth. Next, comparison of three HNSCC cell lines with low, medium, and high levels of AC reveals an inverse correlation between the levels of AC and their response to exogenous C-6-ceramide. Furthermore, over-expression of AC in SCC-1 cells increased resistance to Fas-induced cell killing. Conversely, down-regulation of AC using specific AC small interfering RNA (siRNA) sensitized the SCC-1 cancer cell line to Fas-induced apoptosis. Finally, we show that the AC inhibitor LCL 204 can sensitize HNSCC cell lines to Fas-induced apoptosis both in vitro and in a xenograft model in vivo, suggesting that the combination of FasL gene therapy and LCL 204 may become a new treatment option for advanced-stage head and neck cancer.


Subject(s)
Enzyme Inhibitors/pharmacology , Fas Ligand Protein/genetics , Fas Ligand Protein/metabolism , Galactosylgalactosylglucosylceramidase/antagonists & inhibitors , Galactosylgalactosylglucosylceramidase/metabolism , Genetic Therapy , Head and Neck Neoplasms/metabolism , Animals , Cell Line, Tumor , Cell Survival , Ceramides/metabolism , Down-Regulation , Female , Gene Expression Regulation, Neoplastic , Head and Neck Neoplasms/genetics , Humans , Mice , Mice, Nude , RNA, Small Interfering/genetics , Sensitivity and Specificity , Xenograft Model Antitumor Assays
3.
Mol Ther ; 14(5): 627-36, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16926120

ABSTRACT

The potential anti-tumor agent Apoptin activates apoptosis in many human cancers and transformed cell lines, but is believed to be less potent in primary cells. Although caspase 3 is activated during apoptin-induced apoptosis, the mechanism of tumor cell killing remains elusive. We now show that apoptin-mediated cell death involves modulation of the sphingomyelin-ceramide pathway. Treating cells with Ad-GFPApoptin resulted in increased ceramide accumulation and enhanced expression of acid sphingomyelinase (ASMase) with a concomitant increase in ASMase activity and decreased sphingomyelin. Using confocal microscopy, ASMase, normally present in the endosomal/lysosomal compartment, was observed to translocate to the cell's periphery. Cotreatment of Ad-GFPApoptin-infected cells with the ASMase inhibitor desipramine (2.5 muM) attenuated (30%; P<0.01) apoptin-induced cell death. Apoptin was also able to induce a significant decline in sphingosine content by inhibition of ceramide deacylation through down-regulation of acid ceramidase at the protein level. Supporting the role of ceramide in apoptin action, treatment of cells with the combination of an exogenous cell-permeable ceramide analog (C6-ceramide) and Ad-GFPApoptin infection yielded a significant increase (P<0.01) in apoptosis over either treatment modality alone. Together, these data suggest that apoptin modulates ceramide/sphingolipid metabolism as part of its mechanism of action.


Subject(s)
Apoptosis , Capsid Proteins/metabolism , Sphingolipids/metabolism , Adenoviridae/genetics , Apoptosis/drug effects , Capsid Proteins/genetics , Cell Line, Tumor , Cell Membrane/metabolism , Ceramides/biosynthesis , Desipramine/pharmacology , Down-Regulation , Endosomes/metabolism , Galactosylgalactosylglucosylceramidase/metabolism , Gene Expression , Genes, Reporter/genetics , Humans , Lysosomes/metabolism , Male , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Protein Transport , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sphingomyelin Phosphodiesterase/metabolism , Up-Regulation
4.
Mol Ther ; 13(2): 357-65, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16226492

ABSTRACT

Pancreatic adenocarcinoma (PC) is an aggressive malignancy resistant to standard treatment modalities. Previously, we have reported that cancer-associated Sm-like protein (CaSm) contributes to the neoplastic transformation of PC. In this study, we utilized a recently established preclinical model of PC to determine if molecular targeting of CaSm can serve as the basis for a novel PC therapy. In a subcutaneous tumor model, intratumoral administration of an adenoviral vector encoding CaSm antisense RNA (Ad-alphaCaSm) significantly inhibited Panc02 tumor growth. Furthermore, in a metastatic tumor model, systemic administration of Ad-alphaCaSm resulted in a significant decrease in the number of hepatic metastases and increased survival time. We assessed the efficiency of in vivo delivery and observed significant levels of vector transduction in tissues containing PC, as well as a bystander effect that was amplifying the efficacy of CaSm gene therapy. This bystander effect was also active in vitro and was shown to be at least partially independent of host-related mechanisms. We conclude that CaSm antisense gene therapy is an effective novel therapy for PC and that the antitumor efficacy is dependent on both direct and bystander mechanisms.


Subject(s)
Bystander Effect/genetics , Genetic Therapy/methods , Pancreatic Neoplasms/therapy , Proto-Oncogene Proteins/administration & dosage , Proto-Oncogene Proteins/genetics , RNA, Antisense/therapeutic use , Adenoviridae/genetics , Animals , Cell Line , Cell Line, Tumor , Drug Evaluation, Preclinical , Female , Genetic Vectors/administration & dosage , Genetic Vectors/therapeutic use , Liver Neoplasms, Experimental/secondary , Liver Neoplasms, Experimental/therapy , Liver Neoplasms, Experimental/virology , Mice , Mice, Inbred C57BL , Pancreatic Neoplasms/virology , Proto-Oncogene Proteins/metabolism , RNA-Binding Proteins , Transduction, Genetic
5.
Mol Ther ; 11(3): 363-72, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15727932

ABSTRACT

We have recently shown that the cancer-associated Sm-like protein (CaSm) is overexpressed in human pancreatic adenocarcinoma (PC). However, the role of CaSm in the process of neoplastic transformation remains unclear. To define further the role of CaSm in PC transformation, we have established a murine model based on the murine pancreatic cancer cell lines Panc02 and Panc03. CaSm is overexpressed in the aggressive Panc02 cells and expressed at much lower levels in the more indolent Panc03 cells. Up-regulation of CaSm in Panc03 cells increased in vitro proliferation and anchorage-independent growth and promoted subcutaneous tumor establishment and growth in syngeneic mice. Conversely, adenoviral down-regulation of CaSm in Panc02 led to significant inhibition of cellular proliferation and anchorage-independent growth in vitro and complete abolition of tumor growth and metastasis in vivo. Up-regulation of CaSm in NIH3T3 resulted in loss of contact inhibition and increased soft agar colony formation in vitro. The requirement for CaSm overexpression for neoplastic transformation confirms the concept that CaSm is a critical oncogene and potential target for molecular intervention. Furthermore, establishment of the murine clinically relevant model of pancreatic metastases provides a framework for the generation of preclinical data to support the development of novel molecular therapies targeting CaSm.


Subject(s)
Adenocarcinoma/genetics , Disease Models, Animal , Pancreatic Neoplasms/genetics , Proto-Oncogene Proteins/genetics , Adenocarcinoma/metabolism , Animals , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Mice , Pancreatic Neoplasms/metabolism , Proto-Oncogene Proteins/metabolism , RNA-Binding Proteins , Time Factors , Up-Regulation
6.
Cancer Gene Ther ; 12(1): 12-8, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15514684

ABSTRACT

Previous investigations have revealed that bladder cancer cells are generally resistant to Fas-mediated apoptosis by conventional Fas agonists. However, the ability of these cell lines to undergo Fas-mediated apoptosis may have been underappreciated. As a result, we investigated the in vitro efficacy of Fas ligand gene therapy for bladder cancer. Three human bladder cancer lines (T24, J82, and 5637) were treated with the conventional Fas agonist CH-11, a monoclonal antibody to the Fas receptor. Cells were also treated with a replication-deficient adenovirus containing a modified murine Fas ligand gene fused to green fluorescent protein (GFP), AdGFPFasL. A virus containing the GFP gene alone was used to control for viral toxicity (AdGFP). Cell death was quantified using a tetrazolium-based (MTS) assay. Cells were also evaluated by Western blotting to evaluate poly (ADP-ribose) polymerase, caspase 8, and caspase 9 cleavage and by flow cytometry to determine the presence of coxsackie/adenovirus receptor (CAR). These studies confirmed bladder cancer resistance to cell death by the anti-Fas monoclonal antibody CH-11. This resistance was overcome with AdGFPFasL at a multiplicity of infection (MOI) of 1000 achieving over 80% cell death in all cell lines. Furthermore, greater than 80% cell death was evident in 5637 cells treated with low-dose AdGFPFasL (MOI=10). 5637 cells expressed significantly higher levels of surface CAR than J82 or T24 cells (P<.05). AdGFPFasL is cytotoxic to bladder cancer cells that would otherwise be considered Fas resistant, supporting its in vivo potential. Enhanced sensitivity to AdGFPFasL may be in part due to increased cell surface CAR levels.


Subject(s)
Apoptosis/genetics , Genetic Therapy , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/therapy , Adenoviridae , Antibodies/pharmacology , Antibodies, Monoclonal , Antigens, Surface , Calcium-Binding Proteins , Eye Proteins , Fas Ligand Protein , Genetic Vectors , Humans , Ligands , Lipoproteins , Membrane Glycoproteins , Recoverin , Tumor Cells, Cultured , fas Receptor
7.
Future Oncol ; 1(1): 115-23, 2005 Feb.
Article in English | MEDLINE | ID: mdl-16555981

ABSTRACT

Gene therapy has been in a continuous evolutionary process since the first approved trial occurred in 1990 at the National Institute of Health. In the USA, as of March 2004, there were 619 approved gene therapy/transfer protocols and 405 of these were for cancer treatment. Another 294 trials are in progress worldwide, with most concentrated in Europe. However, cancer gene therapy is in its relative infancy when compared with the well-established use of chemo-radiotherapy for treating cancer. As the field develops it is becoming clear that using gene therapy in conjunction with established chemo-radiotherapy approaches is yielding the best results. This concept shall be reviewed in the context of the status of the field, and a future direction based on a combination of gene therapy with small molecule modification of sphingolipid metabolism shall be discussed.


Subject(s)
Adenoviridae , Carcinoma, Squamous Cell/therapy , Genetic Therapy , Head and Neck Neoplasms/therapy , Prostatic Neoplasms/therapy , Amidohydrolases/antagonists & inhibitors , Ceramidases , Ceramides/metabolism , Cytosine Deaminase/genetics , Fas Ligand Protein , Genes, p53 , Humans , Male , Membrane Glycoproteins/genetics , Thymidine Kinase/genetics , Tumor Necrosis Factors/genetics , Viral Vaccines
8.
Arthritis Rheum ; 50(5): 1566-77, 2004 May.
Article in English | MEDLINE | ID: mdl-15146427

ABSTRACT

OBJECTIVE: Aberrant transforming growth factor beta (TGFbeta) signaling has been implicated in the pathogenesis of scleroderma (systemic sclerosis [SSc]), but the contribution of specific components in this pathway to SSc fibroblast phenotype remains unclear. This study was undertaken to delineate the role of TGFbeta receptor type I (TGFbetaRI) and TGFbetaRII in collagen overexpression by SSc fibroblasts. METHODS: Primary dermal fibroblasts from SSc patients and healthy adults were studied (n = 10 matched pairs). Adenoviral vectors were generated for TGFbetaRI (AdTGFbetaRI), TGFbetaRII (AdTGFbetaRII), and kinase-deficient TGFbetaRII (AdDeltakRII). TGFbetaRI basal protein levels were analyzed by (35)S-methionine labeling/immunoprecipitation and immunohistochemistry. Type I collagen and TGFbetaRII basal protein levels were analyzed by Western blot and newly secreted collagen by (3)H-proline incorporation assay. RESULTS: Analysis of endogenous TGFbetaRI and TGFbetaRII protein levels revealed that SSc TGFbetaRI levels were increased 1.7-fold (P = 0.008; n = 7) compared with levels in healthy controls, while TGFbetaRII levels were decreased by 30% (P = 0.03; n = 7). This increased TGFbetaRI:TGFbetaRII ratio correlated with SSc collagen overexpression. To determine the consequences of altered TGFbetaRI:TGFbetaRII ratio on collagen expression, healthy fibroblasts were transduced with AdTGFbetaRI or AdTGFbetaRII. Forced expression of TGFbetaRI in the range corresponding to elevated SSc TGFbetaRI levels increased basal collagen expression in a dose-dependent manner, while similar TGFbetaRII overexpression had no effect, although transduction of fibroblasts at higher multiplicities of infection led to a marked reduction of basal collagen levels. Blockade of TGFbeta signaling via AdDeltakRII resulted in approximately 50% inhibition of basal collagen levels in healthy fibroblasts and in 5 of 9 SSc cell lines. A subset of SSc fibroblasts (4 of 9 cell lines) was resistant to this treatment. SSc fibroblasts with the highest levels of TGFbetaRI were the least responsive to collagen inhibition via DeltakRII. CONCLUSION: This study indicates that an increased TGFbetaRI:TGFbetaRII ratio may underlie aberrant TGFbeta signaling in SSc and contribute to elevated basal collagen production, which is insensitive to TGFbeta signaling blockade via DeltakRII.


Subject(s)
Activin Receptors, Type I/genetics , Activin Receptors, Type I/metabolism , Collagen/biosynthesis , Receptors, Transforming Growth Factor beta/genetics , Receptors, Transforming Growth Factor beta/metabolism , Scleroderma, Systemic/genetics , Scleroderma, Systemic/metabolism , Adult , Aged , Dermis/pathology , Female , Fibroblasts/metabolism , Fibroblasts/pathology , Humans , Male , Middle Aged , Mutagenesis , Phenotype , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Receptor, Transforming Growth Factor-beta Type I , Receptor, Transforming Growth Factor-beta Type II , Scleroderma, Systemic/pathology , Signal Transduction/physiology
9.
Cancer Gene Ther ; 10(11): 814-22, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14605667

ABSTRACT

Brain tumors (BTs) are among the most malignant forms of human cancer. Unfortunately, current treatments are often ineffective and produce severe side effects. Cytotoxic gene therapy is an alternative treatment strategy, with the potential advantages of reduced toxicity to normal brain tissue. Apoptosis-inducing "death ligands" Fas ligand and TNF-related apoptosis-inducing ligand (TRAIL) are genes with substantial cytotoxic activity in susceptible tumor cells. Here, we compared the effectiveness of Ad vector-mediated delivery of Fas ligand-green fluorescent protein (FasL-GFP) fusion protein, human TRAIL, and both genes simultaneously. We examined a panel of 13 cell lines (eight derived from primary isolates) for susceptibility to Ad5-based vector infection and for sensitivity to FasL- and TRAIL-mediated apoptosis. All cell lines were efficiently transduced, but, as expected, varied in their sensitivity to ligand-induced apoptosis. Generally, sensitivity to FasL-GFP correlated with cell surface FasR levels, but no such correlation was seen for TRAIL and its functional receptors, DR4 and DR5. The vector expressing both FasL-GFP and TRAIL was more effective than either of the single-gene vectors at comparable transduction levels, and it was effective against a broader range of cell lines. In five cell lines, coexpression resulted in apoptosis levels greater than those predicted for strictly additive activity of the two death ligands. We believe that Ad vector-mediated delivery of multiple death ligands may be developed as a potential BT therapy, either alone or in conjunction with surgical resection of the primary tumor.


Subject(s)
Apoptosis/genetics , Brain Neoplasms/pathology , Genetic Therapy/methods , Glioma/pathology , Intracellular Signaling Peptides and Proteins , Membrane Glycoproteins/genetics , Tumor Necrosis Factor-alpha/genetics , Adenoviridae/genetics , Apoptosis Regulatory Proteins , Astrocytoma/genetics , Astrocytoma/pathology , Astrocytoma/therapy , Brain Neoplasms/genetics , Brain Neoplasms/therapy , CASP8 and FADD-Like Apoptosis Regulating Protein , Carrier Proteins/metabolism , Caspase 8 , Caspases/metabolism , Cell Line , Fas Ligand Protein , Genetic Vectors/genetics , Glioma/genetics , Glioma/therapy , Green Fluorescent Proteins , Humans , Luminescent Proteins/genetics , Protein Isoforms , Receptors, TNF-Related Apoptosis-Inducing Ligand , Receptors, Tumor Necrosis Factor/genetics , Receptors, Tumor Necrosis Factor/metabolism , TNF-Related Apoptosis-Inducing Ligand , Transduction, Genetic
10.
Cancer Gene Ther ; 10(4): 330-9, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12679806

ABSTRACT

Inducing Fas-mediated apoptosis in prostate cancer (PCa) is a promising new therapeutic approach with the potential to overcome delivery issues currently problematic in cancer gene therapy. We have previously demonstrated that a Fas Ligand (FasL) expressing adenovirus (AdGFPFasL(TET)) was able to induce Fas-mediated apoptosis in a panel of PCa cell lines regardless of their Fas-sensitivity as determined by the agonistic Fas antibody CH-11. We now report that AdGFPFasL(TET)-infected cells produce apoptotic bodies and cellular debris that continues to elicit FasL-mediated bystander killing in uninfected neighboring cells. Using light microscopy, we demonstrate that AdGFPFasL(TET)-infected cells release apoptotic bodies and cellular debris into the local environment and that this material will induce bystander killing in Jurkat, PPC-1, and PC-3 target cells, but not in DU145 and K-562 cells. The bystander killing mechanism is mediated through Fas/FasL interaction because it is significantly inhibited if target cells are pretreated with the pan spectrum caspase inhibitor Z-VAD-FMK or the Fas neutralizing antibody ZB-4. Coincubation of PPC-1 target cells with apoptotic bodies and cellular debris (effector material) induce nearly complete target cell killing at a ratio of 1:1 target to effector. Collectively, these data indicate that AdGFPFasL(TET)-infected PCa cells release apoptotic and cellular debris capable of inducing bystander killing in PCa and supports the development of FasL as a gene therapy agent.


Subject(s)
Adenoviridae/genetics , Genetic Therapy , Membrane Glycoproteins/genetics , Prostatic Neoplasms/therapy , Apoptosis , Bystander Effect , Caspase Inhibitors , Cell Line, Tumor , Fas Ligand Protein , Genetic Vectors , Humans , Jurkat Cells , Male , Membrane Glycoproteins/metabolism , Microscopy, Fluorescence , Prostatic Neoplasms/pathology
12.
Cancer Biol Ther ; 1(4): 401-6, 2002.
Article in English | MEDLINE | ID: mdl-12432255

ABSTRACT

Although DU145 prostate cancer cells are resistant to exogenously applied Fas agonist CH-11 (anti-Fas monoclonal antibody), Fas-resistance can be overcome using a FasL expressing adenovirus (AdGFPFasL(TET)) [Hyer et al., Molecular Therapy, 2000; 2:348-58 (ref.12)]. The purpose of this study was to try to understand why DU145 cells are resistant to CH-11 and determine the signaling pathway utilized by AdGFPFasL(TET) to induce apoptosis in these Fas-resistant cells. Using immunoblot analysis, we show that AdGFPFasL(TET) is capable of initiating the classic Fas-mediated apoptotic pathway in DU145 cells, which includes activation of caspases-8, -3, -7, and -9, BID cleavage, cytochrome c release from mitochondria, and PARP cleavage. In contrast, CH-11 binds to Fas, but is unable to transmit the death signal beyond the plasma membrane suggesting a block at the DISC (death inducing signaling complex). The anti-apoptotic protein c-FLIP (cellular Flice-like inhibitory protein), which has been shown to inhibit Fas-mediated apoptosis at the DISC, was down-regulated following AdGFPFasL(TET) treatment prompting us to investigate its role in inhibiting CH-11-induced cell death. Using c-FLIP anti-sense oligonucleotides to down-regulate c-FLIP we sensitized DU145 cells to CH-11-induced apoptosis. These data suggest that c-FLIP may play a critical role in regulating Fas-mediated apoptosis in prostate cancer cells and that modulation of c-FLIP may enhance Fas signaling based therapies.


Subject(s)
Apoptosis , Carrier Proteins/metabolism , Down-Regulation , Intracellular Signaling Peptides and Proteins , Prostatic Neoplasms/metabolism , fas Receptor/metabolism , BH3 Interacting Domain Death Agonist Protein , Blotting, Western , CASP8 and FADD-Like Apoptosis Regulating Protein , Caspase 3 , Caspase 7 , Caspase 8 , Caspase 9 , Caspases/metabolism , Cell Death , Cell Line , Cell Separation , Cytochrome c Group/metabolism , Enzyme Activation , Fas Ligand Protein , Flow Cytometry , Humans , Immunoblotting , Jurkat Cells , Male , Membrane Glycoproteins/metabolism , Mitochondria/metabolism , Oligonucleotides, Antisense/pharmacology , Prostatic Neoplasms/pathology , Signal Transduction , Time Factors , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL