Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
J Infect Dis ; 217(2): 288-297, 2018 01 04.
Article in English | MEDLINE | ID: mdl-29149330

ABSTRACT

Superantigens (SAgs) are extremely potent bacterial toxins, which evoke a virulent immune response, inducing nonspecific T-cell proliferation, rapid cytokine release, and lethal toxic shock, for which there is no effective treatment. We previously developed a small molecule, S101, which potently inhibits proliferating T cells. In a severe mouse model of toxic shock, a single injection of S101 given together with superantigen challenge rescued 100% of the mice. Even when given 2 hours after challenge, S101 rescued 40% of the mice. S101 targets the T-cell receptor, inflammatory response, and actin cytoskeleton pathways. S101 inhibits the aryl hydrocarbon receptor, a ligand-activated transcription factor that is involved in the differentiation of T-helper cells, especially Th17, and regulatory T cells. Our results provide the rationale for developing S101 to treat superantigen-induced toxic shock and other pathologies characterized by T-cell activation and proliferation.


Subject(s)
Immunologic Factors/administration & dosage , Shock, Septic/prevention & control , Shock, Septic/therapy , Superantigens/toxicity , T-Lymphocytes/drug effects , Animals , Disease Models, Animal , Female , Injections, Intravenous , Mice , Mice, Inbred BALB C , Survival Analysis , Treatment Outcome
2.
Proc Natl Acad Sci U S A ; 114(52): 13655-13660, 2017 12 26.
Article in English | MEDLINE | ID: mdl-29229829

ABSTRACT

There is an urgent need for an effective treatment for metastatic prostate cancer (PC). Prostate tumors invariably overexpress prostate surface membrane antigen (PSMA). We designed a nonviral vector, PEI-PEG-DUPA (PPD), comprising polyethylenimine-polyethyleneglycol (PEI-PEG) tethered to the PSMA ligand, 2-[3-(1, 3-dicarboxy propyl)ureido] pentanedioic acid (DUPA), to treat PC. The purpose of PEI is to bind polyinosinic/polycytosinic acid (polyIC) and allow endosomal release, while DUPA targets PC cells. PolyIC activates multiple pathways that lead to tumor cell death and to the activation of bystander effects that harness the immune system against the tumor, attacking nontargeted neighboring tumor cells and reducing the probability of acquired resistance and disease recurrence. Targeting polyIC directly to tumor cells avoids the toxicity associated with systemic delivery. PPD selectively delivered polyIC into PSMA-overexpressing PC cells, inducing apoptosis, cytokine secretion, and the recruitment of human peripheral blood mononuclear cells (PBMCs). PSMA-overexpressing tumors in nonobese diabetic/severe combined immunodeficiency (NOD/SCID) mice with partially reconstituted immune systems were significantly shrunken following PPD/polyIC treatment, in all cases. Half of the tumors showed complete regression. PPD/polyIC invokes antitumor immunity, but unlike many immunotherapies does not need to be personalized for each patient. The potent antitumor effects of PPD/polyIC should spur its development for clinical use.


Subject(s)
Glutamate Carboxypeptidase II/antagonists & inhibitors , Poly I-C/pharmacology , Prostatic Neoplasms/immunology , Prostatic Neoplasms/pathology , Adoptive Transfer , Animals , Antigens, Surface/genetics , Antigens, Surface/metabolism , Bystander Effect , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/genetics , Disease Models, Animal , Gene Expression , Glutamate Carboxypeptidase II/genetics , Glutamate Carboxypeptidase II/metabolism , Humans , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Male , Mice , Poly I-C/chemistry , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/genetics , Protein Binding , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
3.
PLoS One ; 11(9): e0162321, 2016.
Article in English | MEDLINE | ID: mdl-27598772

ABSTRACT

Selective delivery of drugs to tumor cells can increase potency and reduce toxicity. In this study, we describe a novel recombinant chimeric protein, dsRBEC, which can bind polyIC and deliver it selectively into EGFR over-expressing tumor cells. dsRBEC, comprises the dsRNA binding domain (dsRBD) of human PKR (hPKR), which serves as the polyIC binding moiety, fused to human EGF (hEGF), the targeting moiety. dsRBEC shows high affinity towards EGFR and triggers ligand-induced endocytosis of the receptor, thus leading to the selective internalization of polyIC into EGFR over-expressing tumor cells. The targeted delivery of polyIC by dsRBEC induced cellular apoptosis and the secretion of IFN-ß and other pro-inflammatory cytokines. dsRBEC-delivered polyIC is much more potent than naked polyIC and is expected to reduce the toxicity caused by systemic delivery of polyIC.


Subject(s)
Apoptosis/drug effects , Drug Delivery Systems , ErbB Receptors/genetics , Interferon Inducers/pharmacology , Poly I-C/pharmacology , Recombinant Fusion Proteins/genetics , Animals , Cell Line, Tumor , Chemokine CCL5/biosynthesis , Chemokine CCL5/metabolism , Cloning, Molecular , Endocytosis , Epidermal Growth Factor/genetics , Epidermal Growth Factor/metabolism , ErbB Receptors/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Humans , Interferon Inducers/chemistry , Interferon Inducers/metabolism , Interferon-beta/biosynthesis , Interferon-beta/metabolism , MCF-7 Cells , Poly I-C/chemistry , Poly I-C/metabolism , Protein Binding , Protein Domains , Recombinant Fusion Proteins/metabolism , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/metabolism , eIF-2 Kinase/genetics , eIF-2 Kinase/metabolism
4.
Cancer Immunol Res ; 4(8): 688-97, 2016 08.
Article in English | MEDLINE | ID: mdl-27241844

ABSTRACT

The development of targeted therapies that affect multiple signaling pathways and stimulate antitumor immunity is greatly needed. About 20% of patients with breast cancer overexpress HER2. Small molecules and antibodies targeting HER2 convey some survival benefits; however, patients with advanced disease succumb to the disease under these treatment regimens, possibly because HER2 is not completely necessary for the survival of the targeted cancer cells. In the present study, we show that a polyinosine/polycytosine (pIC) HER2-homing chemical vector induced the demise of HER2-overexpressing breast cancer cells, including trastuzumab-resistant cells. Targeting pIC to the tumor evoked a number of cell-killing mechanisms, as well as strong bystander effects. These bystander mechanisms included type I IFN induction, immune cell recruitment, and activation. The HER2-targeted pIC strongly inhibited the growth of HER2-overexpressing tumors in immunocompetent mice. The data presented here could open additional avenues in the treatment of HER2-positive breast cancer. Cancer Immunol Res; 4(8); 688-97. ©2016 AACR.


Subject(s)
Antineoplastic Agents/pharmacology , Neoplasms/immunology , Neoplasms/pathology , Poly I-C/administration & dosage , Receptor, ErbB-2/antagonists & inhibitors , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Cytokines/metabolism , Disease Models, Animal , Female , Gene Expression , Humans , Inflammation Mediators/metabolism , Mice , Neoplasms/drug therapy , Neoplasms/metabolism , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism , Tumor Microenvironment/genetics , Xenograft Model Antitumor Assays
5.
Bioconjug Chem ; 25(9): 1644-54, 2014 Sep 17.
Article in English | MEDLINE | ID: mdl-25121341

ABSTRACT

The delivery of nucleic acids into cells is an attractive approach for cancer therapy. Polyethylenimine (PEI) is among the most efficient nonviral carriers. Recent studies have demonstrated that PEI can be conjugated to targeting ligands, such as epidermal growth factor (EGF) and transferrin (Schaffert et al., 2011; Abourbeh et al., 2012; Ogris et al., 1999). Herein we present a simplified protocol for producing homogeneous preparations of PEGylated linear PEI: LPEI-PEG2k. We generated two well-characterized copolymers, with ratios of LPEI to PEG of 1:1 and 1:3. These copolymers were further conjugated through disulfide bonds to a Her-2 targeting moiety, Her-2 affibody. This reaction yielded two triconjugates that target Her-2 overexpressing tumors. Polyplexes were formed by complexing plasmid DNA with the triconjugates. We characterized the biophysical properties of the conjugates, and found that the triconjugate 1:3 polyplex had lower ζ potential, larger particle size, and more heterogeneous shape than the triconjugate 1:1 polyplex. Triconjugate 1:1 and triconjugate 1:3 polyplexes were highly selective toward cells that overexpress Her-2 receptors, but triconjugate 1:1 polyplex was more efficient at gene delivery. Our studies show that the biophysical and biological properties of the conjugates can be profoundly affected by the ratio of LPEI:PEG2k:ligand. The procedure described here can be adapted to generate a variety of triconjugates, simply by changing the targeting moiety.


Subject(s)
DNA/chemistry , Drug Carriers/chemistry , Polyethylene Glycols/chemistry , Polyethyleneimine/chemistry , Cell Line, Tumor , DNA/genetics , DNA/metabolism , Deoxyribonucleases/metabolism , Humans , Ligands , Molecular Weight , Protein Structure, Tertiary , Receptor, ErbB-2/chemistry , Receptor, ErbB-2/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Sulfhydryl Compounds/chemistry , Transfection
6.
Curr Opin Pharmacol ; 13(4): 504-10, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23648271

ABSTRACT

The understanding that the immune system plays a dual role in cancer progression has led to the recent development of targeted immunotherapies. These treatments, which aim to harness the immune system against cancer, include monoclonal antibodies, immune adjuvants, cell-based therapy and vaccines. Although numerous immune-targeted treatment modalities have entered the clinic, most have shown limited efficacy. The intrinsic heterogeneity and genomic instability of the tumor, coupled with immune suppression induced by both the tumor and its microenvironment, remain the main obstacles to the success of these therapies. We believe that the primary objective of the new generation of therapies must be to reinstate immune surveillance against primary and metastatic tumor cells, while inhibiting the immune suppressive microenvironment. Most probably this will be achieved by combining several treatment modalities. This paper will briefly review current immunotherapies and their promise, as well as the obstacles associated with them.


Subject(s)
Immunotherapy , Neoplasms/therapy , Animals , Antibodies/therapeutic use , Antigens, Neoplasm/immunology , Antineoplastic Agents/therapeutic use , Cancer Vaccines , Humans , Neoplasms/immunology
7.
Nat Commun ; 4: 1403, 2013.
Article in English | MEDLINE | ID: mdl-23360994

ABSTRACT

Noradrenaline can modulate multiple cellular functions important for cancer progression; however, how this single extracellular signal regulates such a broad array of cellular processes is unknown. Here we identify Src as a key regulator of phosphoproteomic signalling networks activated in response to beta-adrenergic signalling in cancer cells. These results also identify a new mechanism of Src phosphorylation that mediates beta-adrenergic/PKA regulation of downstream networks, thereby enhancing tumour cell migration, invasion and growth. In human ovarian cancer samples, high tumoural noradrenaline levels were correlated with high pSrc(Y419) levels. Moreover, among cancer patients, the use of beta blockers was significantly associated with reduced cancer-related mortality. Collectively, these data provide a pivotal molecular target for disrupting neural signalling in the tumour microenvironment.


Subject(s)
Ovarian Neoplasms/enzymology , Ovarian Neoplasms/pathology , Receptors, Adrenergic, beta/metabolism , src-Family Kinases/metabolism , Adrenergic beta-Antagonists/pharmacology , Adrenergic beta-Antagonists/therapeutic use , Animals , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Enzyme Activation/drug effects , Female , Humans , Mice , Models, Molecular , Neoplasm Invasiveness , Neoplasm Metastasis , Norepinephrine/pharmacology , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/mortality , Phosphorylation/drug effects , Phosphoserine/metabolism , Signal Transduction/drug effects , Stress, Physiological/drug effects , Survival Analysis , Tyrosine/metabolism , src-Family Kinases/chemistry
8.
Cancer Res ; 72(22): 5757-66, 2012 Nov 15.
Article in English | MEDLINE | ID: mdl-22986745

ABSTRACT

Melanoma is the deadliest form of skin cancer in which patients with metastatic disease have a 5-year survival rate of less than 10%. Recently, the overexpression of a ß-galactoside binding protein, galectin-3 (LGALS3), has been correlated with metastatic melanoma in patients. We have previously shown that silencing galectin-3 in metastatic melanoma cells reduces tumor growth and metastasis. Gene expression profiling identified the protumorigenic gene autotaxin (ENPP2) to be downregulated after silencing galectin-3. Here we report that galectin-3 regulates autotaxin expression at the transcriptional level by modulating the expression of the transcription factor NFAT1 (NFATC2). Silencing galectin-3 reduced NFAT1 protein expression, which resulted in decreased autotaxin expression and activity. Reexpression of autotaxin in galectin-3 silenced melanoma cells rescues angiogenesis, tumor growth, and metastasis in vivo. Silencing NFAT1 expression in metastatic melanoma cells inhibited tumor growth and metastatic capabilities in vivo. Our data elucidate a previously unidentified mechanism by which galectin-3 regulates autotaxin and assign a novel role for NFAT1 during melanoma progression.


Subject(s)
Galectin 3/deficiency , Melanoma/pathology , NFATC Transcription Factors/biosynthesis , Phosphoric Diester Hydrolases/biosynthesis , Animals , Cell Line, Tumor , Female , Galectin 3/biosynthesis , Galectin 3/genetics , Gene Silencing , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/secondary , Melanoma/blood supply , Melanoma/genetics , Melanoma/metabolism , Mice , Mice, Inbred BALB C , Mice, Nude , NFATC Transcription Factors/genetics , Neoplasm Metastasis , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Phosphoric Diester Hydrolases/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics , Transfection
9.
Cancer Res ; 71(21): 6561-6, 2011 Nov 01.
Article in English | MEDLINE | ID: mdl-22009534

ABSTRACT

Progression of melanoma is dependent on cross-talk between tumor cells and the adjacent microenvironment. The thrombin receptor, protease-activated receptor-1 (PAR-1), plays a key role in exerting this function during melanoma progression. PAR-1 and its activating factors, which are expressed on tumor cells and the surrounding stroma, induce not only coagulation but also cell signaling, which promotes the metastatic phenotype. Several adhesion molecules, cytokines, growth factors, and proteases have recently been identified as downstream targets of PAR-1 and have been shown to modulate interactions between tumor cells and the microenvironment in the process of melanoma growth and metastasis. Inhibiting such interactions by targeting PAR-1 could potentially be a useful therapeutic modality for melanoma patients.


Subject(s)
Melanoma/secondary , Neoplasm Proteins/physiology , Receptor, PAR-1/physiology , Thrombin/physiology , Tumor Microenvironment/physiology , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Blood Coagulation , Cell Adhesion , Cell Movement , Disease Progression , Endothelial Cells/pathology , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/secondary , Lung Neoplasms/therapy , Melanoma/blood , Melanoma/pathology , Melanoma, Experimental/secondary , Melanoma, Experimental/therapy , Mice , Molecular Targeted Therapy , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Neoplastic Cells, Circulating , Phenotype , RNA, Small Interfering/therapeutic use , Receptor, PAR-1/biosynthesis , Receptor, PAR-1/genetics , Signal Transduction/physiology , Stromal Cells/physiology , Xenograft Model Antitumor Assays
10.
Melanoma Res ; 21(4): 357-63, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21738104

ABSTRACT

Gefitinib is an inhibitor of the epidermal growth factor receptor, which is frequently expressed on both choroidal and nonchoroidal melanoma cells. We evaluated the clinical efficacy of gefitinib in patients with metastatic melanoma. Patients with stage IV or unresectable stage III melanoma and Zubrod performance status of less than or equal to 2 were eligible. Previous systemic treatment for metastatic disease was required. The dose of oral gefitinib was 250 mg administered daily, and tumor response was evaluated every 6 weeks. Forty-six patients with nonchoroidal melanoma and six with choroidal melanoma were treated, and 48 were evaluable for response. The median age was 62.5 years. Forty-one patients (79%) had stage M1c disease. There were no drug-related grade 4 or 5 adverse events, and fatigue was the only grade 3 adverse event that occurred in more than 5% of patients. Two patients (4%) had partial responses and 13 patients (27%) had disease stabilization. The two responders had a median duration of response of 10.9 months. The median overall progression-free survival was 1.4 months and the median overall survival was 9.7 months. Among the patients with sufficient tissues obtained before and 6 weeks after starting gefitinib administration, there were no notable trends in the changes of the tumoral expression of p-ERK1/2, p-AKT, PAK1, and serum levels of vascular endothelial growth factor or IL-8 with treatment. We concluded that gefitinib was well tolerated but had minimal clinical efficacy as a single-agent therapy for unselected patients with metastatic melanoma.


Subject(s)
Antineoplastic Agents/therapeutic use , Choroid Neoplasms/drug therapy , Melanoma/drug therapy , Protein Kinase Inhibitors/therapeutic use , Quinazolines/therapeutic use , Administration, Oral , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Choroid Neoplasms/enzymology , Choroid Neoplasms/genetics , Choroid Neoplasms/mortality , Choroid Neoplasms/pathology , Disease-Free Survival , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , ErbB Receptors/metabolism , Female , Gefitinib , Humans , Male , Melanoma/enzymology , Melanoma/genetics , Melanoma/mortality , Melanoma/secondary , Middle Aged , Mutation , Neoplasm Staging , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/adverse effects , Quinazolines/administration & dosage , Quinazolines/adverse effects , Texas , Time Factors , Treatment Outcome , Young Adult
11.
Cancer Res ; 71(10): 3494-504, 2011 May 15.
Article in English | MEDLINE | ID: mdl-21467165

ABSTRACT

The acquisition of the metastatic melanoma phenotype is associated with increased expression of the melanoma cell adhesion molecule MCAM/MUC18 (CD146). However, the mechanism by which MUC18 contributes to melanoma metastasis remains unclear. Herein, we stably silenced MUC18 expression in two metastatic melanoma cell lines, A375SM and C8161, and conducted cDNA microarray analysis. We identified and validated that the transcriptional regulator, inhibitor of DNA binding-1 (Id-1), previously shown to function as an oncogene in several malignancies, including melanoma, was downregulated by 5.6-fold following MUC18 silencing. Additionally, we found that MUC18 regulated Id-1 expression at the transcriptional level via ATF-3, which itself was upregulated by 6.9-fold in our cDNA microarray analysis. ChIP analysis showed increased binding of ATF-3 to the Id-1 promoter after MUC18 silencing. To complement these studies, we rescued the expression of MUC18, which reversed the expression patterns of Id-1 and ATF-3. Moreover, we showed that MUC18 promotes melanoma invasion through Id-1, as overexpression of Id-1 in MUC18-silenced cells resulted in increased MMP-2 expression and activity. To our knowledge, this is the first demonstration that MUC18 is involved in cell signaling regulating the expression of Id-1 and ATF-3, thus contributing to melanoma metastasis.


Subject(s)
Activating Transcription Factor 3/metabolism , CD146 Antigen/metabolism , Gene Expression Regulation, Neoplastic , Inhibitor of Differentiation Protein 1/physiology , Melanoma/metabolism , Animals , Cell Line, Tumor , Chromatin Immunoprecipitation , Disease Progression , Female , Humans , Inhibitor of Differentiation Protein 1/genetics , Melanoma/pathology , Mice , Mice, Inbred BALB C , Neoplasm Metastasis , Neoplasm Transplantation
12.
Oncotarget ; 2(1-2): 8-17, 2011.
Article in English | MEDLINE | ID: mdl-21378407

ABSTRACT

Melanoma remains as the deadliest form of skin cancer with limited and inefficient treatment options available for patients with metastatic disease. Within the last decade, the thrombin receptor, Protease Activated Receptor-1, has been described as an essential gene involved in the progression of human melanoma. PAR-1 is known to activate adhesive, invasive and angiogenic factors to promote melanoma metastasis. It is overexpressed not only in metastatic melanoma cell lines but is also highly expressed in metastatic lesions as compared to primary nevi and normal skin. Recently, PAR-1 has been described to regulate the gap junction protein Connexin 43 and the tumor suppressor gene Maspin to promote the metastatic melanoma phenotype. Herein, we review the role of PAR-1 in the progression of melanoma as well as utilizing PAR-1-regulated genes as potential therapeutic targets for melanoma treatment.


Subject(s)
Melanoma/genetics , Melanoma/pathology , Receptor, PAR-1/physiology , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Disease Progression , Gene Expression Regulation, Neoplastic , Humans , Melanoma/drug therapy , Melanoma/metabolism , Molecular Targeted Therapy , Neoplasm Metastasis , Receptor, PAR-1/genetics , Receptor, PAR-1/metabolism , Skin Neoplasms/drug therapy , Skin Neoplasms/metabolism
13.
Semin Cancer Biol ; 21(2): 83-8, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21147226

ABSTRACT

The molecular changes associated with the transition of melanoma cells from radial growth phase (RGP) to vertical growth phase (VGP) and the metastatic phenotype are not very well defined. However, some of the genes involved in this process and their transcriptional regulation are beginning to be elucidated. For example, the switch from RGP to VGP and the metastatic phenotype is associated with loss of the AP-2α transcription factor. AP-2α regulates the expression of c-KIT, MMP-2, VEGF, and the adhesion molecule MCAM/MUC18. Recently, we reported that AP-2α also regulates two G-protein coupled receptors (GPCRs) PAR-1 and PAFR. In turn, the thrombin receptor, PAR-1, regulates the expression of the gap junction protein Connexin-43 and the tumor suppressor gene Maspin. Activation of PAR-1 also leads to overexpression and secretion of proangiogenic factors such as IL-8, uPA, VEGF, PDGF, as well certain integrins. PAR-1 also cooperates with PAFR to regulate the expression of the MCAM/MUC18 via phosphorylation of CREB. The ligands for these GPCRs, thrombin and PAF, are secreted by stromal cells, emphasizing the importance of the tumor microenvironment in melanoma metastasis. The metastatic phenotype of melanoma is also associated with overexpression and function of CREB/ATF-1. Loss of AP-2α and overexpression of CREB/ATF-1 results in the overexpression of MCAM/MUC18 which by itself contributes to melanoma metastasis by regulating the inhibitor of DNA binding-1 (Id-1). CREB/ATF-1 also regulates the angiogenic factor CYR-61. Our recent data indicate that CREB/ATF-1 regulates the expression of AP-2α, thus, supporting the notion that CREB is an important "master switch" in melanoma progression.


Subject(s)
Melanoma , Tumor Microenvironment , Animals , Cell Adhesion Molecules/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Gene Expression Regulation, Neoplastic , Humans , Melanoma/genetics , Melanoma/pathology , Melanoma/secondary , Neoplasm Metastasis , Receptors, G-Protein-Coupled/metabolism , Transcription Factor AP-2/genetics , Transcription Factor AP-2/metabolism
14.
Proc Natl Acad Sci U S A ; 108(2): 626-31, 2011 Jan 11.
Article in English | MEDLINE | ID: mdl-21187389

ABSTRACT

The thrombin receptor protease activated receptor-1 (PAR-1) is overexpressed in metastatic melanoma cell lines and tumor specimens. Previously, we demonstrated a significant reduction in tumor growth and experimental lung metastasis after PAR-1 silencing via systemic delivery of siRNA encapsulated into nanoliposomes. Gene expression profiling identified a 40-fold increase in expression of Maspin in PAR-1-silenced metastatic melanoma cell lines. Maspin promoter activity was significantly increased after PAR-1 silencing, suggesting that PAR1 negatively regulates Maspin at the transcriptional level. ChIP analyses revealed that PAR-1 decreases binding of Ets-1 and c-Jun transcription factors to the Maspin promoter, both known to activate Maspin transcription. PAR-1 silencing did not affect Ets-1 or c-Jun expression; rather it resulted in increased expression of the chromatin remodeling complex CBP/p300, as well as decreased activity of the CBP/p300 inhibitor p38, resulting in increased binding of Ets-1 and c-Jun to the Maspin promoter and higher Maspin expression. Functionally, Maspin expression reduced the invasive capability of melanoma cells after PAR-1 silencing, which was abrogated after rescuing with PAR-1. Furthermore, tumor growth and experimental lung metastasis was significantly decreased after expressing Maspin in a metastatic melanoma cell line. Moreover, silencing Maspin in PAR-1-silenced cells reverted the inhibition of tumor growth and experimental lung metastasis. Herein, we demonstrate a mechanism by which PAR-1 negatively regulates the expression of the Maspin tumor-suppressor gene in the acquisition of the metastatic melanoma phenotype, thus attributing an alternative function to PAR-1 other than coagulation.


Subject(s)
Gene Expression Regulation, Neoplastic , Genes, Tumor Suppressor , Melanoma/pathology , Receptor, PAR-1/metabolism , Serpins/metabolism , Animals , Chromatin/chemistry , Disease Progression , Humans , Lung Neoplasms/pathology , Mice , Mice, Nude , Neoplasm Metastasis , Phenotype , Proto-Oncogene Protein c-ets-1/metabolism , Proto-Oncogene Proteins c-jun/metabolism , Receptors, G-Protein-Coupled/metabolism , Signal Transduction
15.
PLoS One ; 5(8): e12452, 2010 Aug 27.
Article in English | MEDLINE | ID: mdl-20805990

ABSTRACT

BACKGROUND: The loss of AP-2alpha and increased activity of cAMP-responsive element binding (CREB) protein are two hallmarks of malignant progression of cutaneous melanoma. However, the molecular mechanism responsible for the loss of AP-2alpha during melanoma progression remains unknown. METHODOLOGY/PRINCIPAL FINDINGS: Herein, we demonstrate that both inhibition of PKA-dependent CREB phosphorylation, as well as silencing of CREB expression by shRNA, restored AP-2alpha protein expression in two metastatic melanoma cell lines. Moreover, rescue of CREB expression in CREB-silenced cell lines downregulates expression of AP-2alpha. Loss of AP-2alpha expression in metastatic melanoma occurs via a dual mechanism involving binding of CREB to the AP-2alpha promoter and CREB-induced overexpression of another oncogenic transcription factor, E2F-1. Upregulation of AP-2alpha expression following CREB silencing increases endogenous p21(Waf1) and decreases MCAM/MUC18, both known to be downstream target genes of AP-2alpha involved in melanoma progression. CONCLUSIONS/SIGNIFICANCE: Since AP-2alpha regulates several genes associated with the metastatic potential of melanoma including c-KIT, VEGF, PAR-1, MCAM/MUC18, and p21(Waf1), our data identified CREB as a major regulator of the malignant melanoma phenotype.


Subject(s)
Cyclic AMP Response Element-Binding Protein/metabolism , Gene Expression Regulation, Neoplastic , Melanoma/genetics , Transcription Factor AP-2/genetics , Transcription Factor AP-2/metabolism , Animals , CD146 Antigen/metabolism , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Disease Progression , E2F1 Transcription Factor/metabolism , Humans , Melanoma/metabolism , Melanoma/pathology , Neoplasm Metastasis , Phosphorylation , Promoter Regions, Genetic/genetics , Transcription, Genetic , Up-Regulation
16.
Cancer Res ; 69(16): 6730-7, 2009 Aug 15.
Article in English | MEDLINE | ID: mdl-19679555

ABSTRACT

Protease-activated receptor-1 (PAR-1) is a key player in melanoma metastasis with higher expression seen in metastatic melanoma cell lines and tissue specimens. cDNA microarray and Western blot analyses reveal that the gap junctional intracellular communication molecule connexin 43 (Cx-43), known to be involved in tumor cell diapedesis and attachment to endothelial cells, is significantly decreased after PAR-1 silencing in metastatic melanoma cell lines. Furthermore, Cx-43 promoter activity was significantly inhibited in PAR-1-silenced cells, suggesting that PAR-1 regulates Cx-43 at the transcriptional level. Chromatin immunoprecipitation studies showed a reduction in the binding of SP-1 and AP-1 transcription factors to the promoter of Cx-43. Both transcription factors have been shown previously to be required for maximal Cx-43 promoter activity. These results were corroborated by mutating the AP-1 and SP-1 binding sites resulting in decreased Cx-43 promoter activity in PAR-1-positive cells. Moreover, as Cx-43 has been shown to facilitate arrest of circulating tumor cells at the vascular endothelium, melanoma cell attachment to endothelial cells was significantly decreased in PAR-1-silenced cells, with this effect being abrogated after PAR-1 rescue. Herein, we report that up-regulation of PAR-1 expression, seen in melanoma progression, mediates high levels of Cx-43 expression. As both SP-1 and AP-1 transcription factors act as positive regulators of Cx-43, our data provide a novel mechanism for the regulation of Cx-43 expression by PAR-1. Indeed, Cx-43 expression was restored following PAR-1 rescue in PAR-1-silenced cells. Taken together, our data support the tumor promoting function of Cx-43 in melanoma.


Subject(s)
Connexin 43/genetics , Gene Expression Regulation, Neoplastic , Melanoma/genetics , Melanoma/pathology , Receptor, PAR-1/physiology , Cell Adhesion/drug effects , Cells, Cultured , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelial Cells/physiology , Gene Expression Regulation, Neoplastic/physiology , Humans , Melanoma/metabolism , Neoplasm Metastasis , Promoter Regions, Genetic , Protein Binding , RNA Interference/physiology , RNA, Small Interfering/pharmacology , Receptor, PAR-1/antagonists & inhibitors , Receptor, PAR-1/genetics , Receptor, PAR-1/metabolism , Transcriptional Activation , Transfection
17.
J Biol Chem ; 284(42): 28845-55, 2009 Oct 16.
Article in English | MEDLINE | ID: mdl-19703903

ABSTRACT

The cellular and molecular pathways that regulate platelet activation, blood coagulation, and inflammation are emerging as critical players in cancer progression and metastasis. Here, we demonstrate a novel signaling mechanism whereby protease-activated receptor 1 (PAR1) mediates expression of melanoma cell adhesion molecule MCAM/MUC18 (MUC18), a critical marker of melanoma metastasis, via activation of platelet-activating factor receptor (PAFR) and cAMP-responsive element-binding protein (CREB). We found that PAR1 silencing with small hairpin RNA inhibits MUC18 expression in metastatic melanoma cells by inhibiting CREB phosphorylation, activity, and binding to the MUC18 promoter. We further demonstrate that the PAF/PAFR pathway mediates MUC18 expression downstream of PAR1. Indeed, PAR1 silencing down-regulates PAFR expression and PAF production, PAFR silencing blocks MUC18 expression, and re-expression of PAFR in PAR1-silenced cells rescues MUC18 expression. We further demonstrate that the PAR1-PAFR-MUC18 pathway mediates melanoma cell adhesion to microvascular endothelial cells, transendothelial migration, and metastatic retention in the lungs. Rescuing PAFR expression in PAR1-silenced cells fully restores metastatic phenotype of melanoma, indicating that PAFR plays critical role in the molecular mechanism of PAR1 action. Our results link the two pro-inflammatory G-protein-coupled receptors, PAR1 and PAFR, with the metastatic dissemination of melanoma and suggest that PAR1, PAFR, and MUC18 are attractive therapeutic targets for preventing melanoma metastasis.


Subject(s)
Melanoma/metabolism , Platelet Membrane Glycoproteins/metabolism , Receptor, PAR-1/metabolism , Receptors, G-Protein-Coupled/metabolism , Skin Neoplasms/metabolism , CD146 Antigen/metabolism , CREB-Binding Protein/metabolism , Cell Line, Tumor , Gene Silencing , Humans , Melanoma/pathology , Neoplasm Metastasis , Promoter Regions, Genetic , RNA Interference , RNA, Small Interfering/metabolism , Skin Neoplasms/pathology , Transcription Factors/metabolism
18.
J Biol Chem ; 284(38): 26194-206, 2009 Sep 18.
Article in English | MEDLINE | ID: mdl-19632997

ABSTRACT

Metastatic progression of melanoma is associated with overexpression and activity of cAMP-response element-binding protein (CREB). However, the mechanism by which CREB contributes to tumor progression and metastasis remains unclear. Here, we demonstrate that stably silencing CREB expression in two human metastatic melanoma cell lines, A375SM and C8161-c9, suppresses tumor growth and experimental metastasis. Analysis of cDNA microarrays revealed that CREB silencing leads to increased expression of cysteine-rich protein 61 (CCN1/CYR61) known to mediate adhesion, chemostasis, survival, and angiogenesis. Promoter analysis and chromatin immunoprecipitation assays demonstrated that CREB acts as a negative regulator of CCN1/CYR61 transcription by directly binding to its promoter. Re-expression of CREB in CREB-silenced cells rescued the low CCN1/CYR61 expression phenotype. CCN1/CYR61 overexpression resulted in reduced tumor growth and metastasis and inhibited the activity of matrix metalloproteinase-2. Furthermore, its overexpression decreased melanoma cell motility and invasion through Matrigel, which was abrogated by silencing CCN1/CYR61 in low metastatic melanoma cells. Moreover, a significant decrease in angiogenesis as well as an increase in apoptosis was seen in tumors overexpressing CCN1/CYR61. Our results demonstrate that CREB promotes melanoma growth and metastasis by down-regulating CCN1/CYR61 expression, which acts as a suppressor of melanoma cell motility, invasion and angiogenesis.


Subject(s)
Cyclic AMP Response Element-Binding Protein/metabolism , Cysteine-Rich Protein 61/metabolism , Gene Expression Regulation, Neoplastic , Gene Silencing , Genes, Tumor Suppressor , Melanoma/metabolism , Animals , Cell Line, Tumor , Cyclic AMP Response Element-Binding Protein/genetics , Cysteine-Rich Protein 61/genetics , Gene Expression Regulation, Enzymologic/genetics , Humans , Matrix Metalloproteinase 2/biosynthesis , Matrix Metalloproteinase 2/genetics , Melanoma/genetics , Melanoma/pathology , Mice , Mice, Nude , Neoplasm Metastasis , Neoplasm Transplantation , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Transplantation, Heterologous
19.
Cancer Res ; 68(21): 9078-86, 2008 Nov 01.
Article in English | MEDLINE | ID: mdl-18974154

ABSTRACT

The thrombin receptor [protease-activated receptor-1 (PAR-1)] is overexpressed in highly metastatic melanoma cell lines and in patients with metastatic lesions. Activation of PAR-1 leads to cell signaling and up-regulation of genes involved in adhesion, invasion, and angiogenesis. Herein, we stably silence PAR-1 through the use of lentiviral short hairpin RNA and found significant decreases in both tumor growth (P < 0.01) and metastasis (P < 0.001) of highly metastatic melanoma cell lines in vivo. The use of viruses for therapy is not ideal as it can induce toxic immune responses and possible gene alterations following viral integration. Therefore, we also used systemic delivery of PAR-1 small interfering RNA (siRNA) incorporated into neutral liposomes [1,2-dioleoyl-sn-glycero-3-phosphatidylcholine (DOPC)] to decrease melanoma growth and metastasis in vivo. Significant decreases in tumor growth, weight, and metastatic lung colonies (P < 0.001 for all) were found in mice treated with PAR-1 siRNA-DOPC. The in vivo effects of PAR-1 on invasion and angiogenesis were analyzed via immunohistochemistry. Concomitant decreases in vascular endothelial growth factor, interleukin-8, and matrix metalloproteinase-2 expression levels, as well as decreased blood vessel density (CD31), were found in tumor samples from PAR-1 siRNA-treated mice, suggesting that PAR-1 is a regulator of melanoma cell growth and metastasis by affecting angiogenic and invasive factors. We propose that siRNA incorporated into DOPC nanoparticles could be delivered systemically and used as a new modality for melanoma treatment.


Subject(s)
Cell Division , Melanoma/pathology , Neoplasm Metastasis , RNA, Small Interfering/administration & dosage , Receptor, PAR-1/genetics , Animals , Base Sequence , Cell Line, Tumor , Female , Humans , Immunohistochemistry , Liposomes , Mice , Mice, Nude , Reverse Transcriptase Polymerase Chain Reaction
20.
Cancer Res ; 68(20): 8419-28, 2008 Oct 15.
Article in English | MEDLINE | ID: mdl-18922915

ABSTRACT

Host immunity affects tumor metastasis but the corresponding cellular and molecular mechanisms are not entirely clear. Here, we show that a subset of B lymphocytes (termed B-1 population), but not other lymphocytes, has prometastatic effects on melanoma cells in vivo through a direct heterotypic cell-cell interaction. In the classic B16 mouse melanoma model, one mechanism underlying this phenomenon is a specific up-regulation and subsequent homophilic interaction mediated by the cell surface glycoprotein MUC18 (also known as melanoma cell adhesion molecule). Presence of B-1 lymphocytes in a panel of tumor samples from melanoma patients directly correlates with MUC18 expression in melanoma cells, indicating that the same protein interaction exists in humans. These results suggest a new but as yet unrecognized functional role for host B-1 lymphocytes in tumor metastasis and establish a biochemical basis for such observations. Our findings support the counterintuitive central hypothesis in which a primitive layer of the immune system actually contributes to tumor progression and metastasis in a mouse model and in melanoma patients. Given that monoclonal antibodies against MUC18 are in preclinical development but the reason for their antitumor activity is not well understood, these translational results are relevant in the setting of human melanoma and perhaps of other cancers.


Subject(s)
B-Lymphocyte Subsets/immunology , Melanoma/secondary , Animals , CD146 Antigen/immunology , CD146 Antigen/physiology , Cell Communication , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Humans , Melanoma/immunology , Mice , Phosphorylation
SELECTION OF CITATIONS
SEARCH DETAIL