Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Cell Physiol Biochem ; 26(3): 471-82, 2010.
Article in English | MEDLINE | ID: mdl-20798532

ABSTRACT

BACKGROUND: Several secondary metabolites from herbal nutrient products act as weak estrogens (phytoestrogens), competing with endogenous estrogen for binding to the estrogen receptors and inhibiting steroid converting enzymes. However, it is still unclear whether these compounds elicit estrogen dependent transcription of genes at physiological concentrations. METHODS: We compare the effects of physiological concentrations (100 nM) of the two phytoestrogens Enterolactone and Quercetin and the suspected phytoestrogen Curcumin on gene expression in the breast cancer cell line MCF7 with the effects elicited by 17-beta-estradiol (E2). RESULTS: All three phytocompounds have weak effects on gene transcription; most of the E2 genes respond to the phytoestrogens in the same direction though to a much lesser extent and in the order Curcumin > Quercetin > Enterolactone. Gene regulation induced by these compounds was low for genes strongly induced by E2 and similar to the latter for genes only weakly regulated by the classic estrogen. Of interest with regard to the treatment of menopausal symptoms, the survival factor Birc5/survivin and the oncogene MYBL1 are strongly induced by E2 but only marginally by phytoestrogens. CONCLUSION: This approach demonstrates estrogenic effects of putative phytoestrogens at physiological concentrations and shows, for the first time, estrogenic effects of Curcumin.


Subject(s)
Curcumin/pharmacology , Phytoestrogens/pharmacology , Transcription, Genetic/drug effects , 4-Butyrolactone/analogs & derivatives , 4-Butyrolactone/pharmacology , Cell Line, Tumor , Estradiol/pharmacology , Gene Expression Profiling/standards , Humans , Inhibitor of Apoptosis Proteins , Lignans/pharmacology , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Quercetin/pharmacology , Reference Values , Survivin , Trans-Activators/genetics , Trans-Activators/metabolism
2.
Cancer Lett ; 287(1): 33-43, 2010 Jan 01.
Article in English | MEDLINE | ID: mdl-19553003

ABSTRACT

The Interferon-inducible gene, IFI16 has been implicated in the control of cell growth, apoptosis, angiogenesis and immunomodulation. In a previous study we demonstrated that restoring levels of IFI16 in a head and neck squamous cell carcinoma (HNSCC)-derived cell line, HNO136, reduced its growth in vitro accompanied by a marked increase in doxorubicin-induced apoptosis. To evaluate the ability of IFI16 to inhibit in vivo tumorigenesis of HNO136 cells and to characterize the molecular mechanisms responsible for its anti-tumor activity, IFI16 expression on cell growth was evaluated by an in vivo tumorigenicity assay. After excision, tumors were subjected to morphometric and immunohistochemical analyses with markers of apoptosis, angiogenesis, and inflammation. Restoring IFI16 expression significantly reduced the in vivo tumorigenesis of HNO136, decreased tumor vascularization and increased areas of tumor necrosis. Further analysis revealed that IFI16 expression triggered apoptosis of tumor cells, as evaluated using TUNEL assay. Finally, restoring IFI16 protein to HNO136 cells increased CD45+ inflammatory cell infiltration of the tumor burden, predominantly consisting of CD68/CD14 positive macrophages. In accordance with our previous in vitro experiments, this study demonstrates for the first time that IFI16 exerts in vivo anti-tumoral activity by promoting apoptosis of tumor cells, by inhibiting neo-vascularisation, and by increasing the recruitment of macrophages through the release of chemotactic factors.


Subject(s)
Carcinoma, Squamous Cell/prevention & control , Head and Neck Neoplasms/prevention & control , Nuclear Proteins/physiology , Phosphoproteins/physiology , Animals , Antigens, CD/analysis , Antigens, Differentiation, Myelomonocytic/analysis , Carcinoma, Squamous Cell/immunology , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Head and Neck Neoplasms/immunology , Head and Neck Neoplasms/pathology , Humans , Immunohistochemistry , Lipopolysaccharide Receptors/analysis , Mice , Necrosis , Nuclear Proteins/analysis , Nuclear Proteins/genetics , Phosphoproteins/analysis , Phosphoproteins/genetics
3.
Cancer ; 110(9): 2007-11, 2007 Nov 01.
Article in English | MEDLINE | ID: mdl-17823911

ABSTRACT

BACKGROUND: Leukemias are dependent on Akt/NF-kappaB activation and angiogenesis. METHODS: The antiangiogenic Akt/NF-kappaB inhibitor xanthohumol (XN) has in vitro activity against acute and chronic myelogenous leukemia cell lines (AML, CML) and fresh samples from patients were investigated. RESULTS: Inhibition of cell proliferation is associated with induction of apoptosis and reduced VEGF secretion. Decreased cell invasion, metalloprotease production, and adhesion to endothelial cells observed in the presence of XN could prevent in vivo life-threatening complications of leukostasis and tissue infiltration. CONCLUSIONS: As endothelial cells and hematopoietic cells are mutually correlated in their development and growth, targeting both tumor cells and endothelial cells with agents possessing cytotoxic and antiangiogenic activities may lead to synergistic antitumor effects interrupting a reciprocal stimulatory loop between leukemia and endothelial cells.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Cell Proliferation/drug effects , Hematologic Neoplasms , Neovascularization, Pathologic/drug therapy , Propiophenones/pharmacology , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Blotting, Western , Cell Adhesion/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Dose-Response Relationship, Drug , Flavonoids , Humans , NF-kappa B/antagonists & inhibitors , NF-kappa B/drug effects , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/drug effects
4.
Clin Exp Metastasis ; 24(7): 485-93, 2007.
Article in English | MEDLINE | ID: mdl-17653825

ABSTRACT

Ocular tumors such as retinoblastoma and uveal melanoma have devastating effects on vision. Patients with uveal melanoma also have low 5-year survival rates, thus new therapeutic modalities are necessary. As both retinoblastoma and uveal melanoma are highly vascular, we tested application of a gene transduction approach with a potent TH1 cytokine also endowed with strong anti-angiogenic activity, Interleukin-12 (IL-12). Gene transfer into murine 99E1 uveal melanoma-like cells, while having no effects on growth in vitro, essentially blocked subcutaneous tumor growth in vivo without evident signs of toxicity. Orthotopic intraocular injection resulted in invasive tumors that destroyed ocular architecture by the control cells while the IL-12 transduced cells rarely formed tumors. Histological analysis revealed highly invasive and angiogenic tumor growth in the controls and poorly vascularized tumors in the presence of IL-12. The tumor repression effect could be reproduced by a systemic anti-angiogenic effect, where controlateral injection of IL-12 expressing cells strongly repressed growth in tumors formed by parental 99E1 cells. This was associated with significantly lowered tumor vessel densities, a trend toward lower VEGF levels in the lesion, and significantly decreased NK cells in the parental tumors exposed to systemic IL-12. Taken together, our data suggest that IL-12 gene transfer can provide anti-angiogenic effects without toxicity and may be particularly suited for therapy of vascularized ocular tumors.


Subject(s)
Gene Transfer Techniques , Interleukin-12/genetics , Melanoma/blood supply , Melanoma/therapy , Neovascularization, Pathologic/therapy , Transduction, Genetic , Uveal Neoplasms/blood supply , Uveal Neoplasms/therapy , Animals , Cell Proliferation , Female , Mice , Mice, Nude , Neoplasm Transplantation , Skin Neoplasms/therapy , Transfection , Tumor Cells, Cultured
5.
Cell Physiol Biochem ; 19(1-4): 137-52, 2007.
Article in English | MEDLINE | ID: mdl-17310108

ABSTRACT

Dissemination of metastatic cells probably occurs long before diagnosis of the primary tumor. Metastasis during early phases of carcinogenesis in high risk patients is therefore a potential prevention target. The plant polyphenol Curcumin has been proposed for dietary prevention of cancer. We therefore examined its effects on the human breast cancer cell line MDA-MB-231 in vitroand in a mouse metastasis model. Curcumin strongly induces apoptosis in MDA-MB-231 cells in correlation with reduced activation of the survival pathway NFkappaB, as a consequence of diminished IotakappaB and p65 phosphorylation. Curcumin also reduces the expression of major matrix metalloproteinases (MMPs) due to reduced NFkappa B activity and transcriptional downregulation of AP-1. NFkappa B/p65 silencing is sufficient to downregulate c-jun and MMP expression. Reduced NFkappa B/AP-1 activity and MMP expression lead to diminished invasion through a reconstituted basement membrane and to a significantly lower number of lung metastases in immunodeficient mice after intercardiac injection of 231 cells (p=0.0035). 68% of Curcumin treated but only 17% of untreated animals showed no or very few lung metastases, most likely as a consequence of down-regulation of NFkappa B/AP-1 dependent MMP expression and direct apoptotic effects on circulating tumor cells but not on established metastases. Dietary chemoprevention of metastases appears therefore feasible.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/pathology , Curcumin/pharmacology , Flavonoids/pharmacology , Lung Neoplasms/prevention & control , Neoplasm Metastasis/prevention & control , Phenols/pharmacology , Animals , Apoptosis , Breast Neoplasms/drug therapy , Cell Line, Tumor , Cell Survival , Female , Forkhead Transcription Factors/genetics , Humans , I-kappa B Proteins/metabolism , Lung Neoplasms/secondary , Matrix Metalloproteinases/metabolism , Mice , Mice, SCID , NF-kappa B/metabolism , Neoplasm Invasiveness , Phosphorylation , Polyphenols , Transcription Factor AP-1/metabolism , Transfection , Xenograft Model Antitumor Assays/methods
6.
J Immunol ; 178(2): 1122-35, 2007 Jan 15.
Article in English | MEDLINE | ID: mdl-17202376

ABSTRACT

IFNs are highly pleiotropic cytokines also endowed with marked antiangiogenic activity. In this study, the mRNA expression profiles of endothelial cells (EC) exposed in vitro to IFN-alpha, IFN-beta, or IFN-gamma were determined. We found that in HUVEC as well as in other EC types 175 genes were up-regulated (>2-fold increase) by IFNs, including genes involved in the host response to RNA viruses, inflammation, and apoptosis. Interestingly, 41 genes showed a >5-fold higher induction by IFN-alpha in EC compared with human fibroblasts; among them, the gene encoding the angiostatic chemokine CXCL11 was selectively induced by IFN-alpha in EC along with other genes associated with angiogenesis regulation, including CXCL10, TRAIL, and guanylate-binding protein 1. These transcriptional changes were confirmed and extended by quantitative PCR analysis and ELISA; whereas IFN-alpha and IFN-beta exerted virtually identical effects on transcriptome modulation, a differential gene regulation by type I and type II IFN emerged, especially as far as quantitative aspects were concerned. In vivo, IFN-alpha-producing tumors overexpressed murine CXCL10 and CXCL11, guanylate-binding protein 1, and TRAIL, with evidence of CXCL11 production by tumor-associated EC. Overall, these findings improve our understanding of the antiangiogenic effects of IFNs by showing that these cytokines trigger an antiangiogenic transcriptional program in EC. Moreover, we suggest that quantitative differences in the magnitude of the transcriptional activation of IFN-responsive genes could form the basis for cell-specific transcriptional signatures.


Subject(s)
Endothelial Cells/drug effects , Endothelial Cells/metabolism , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Interferons/pharmacology , Animals , Cells, Cultured , Chemokine CXCL10 , Chemokine CXCL11 , Chemokines, CXC/metabolism , Gene Expression , Gene Expression Profiling , Humans , Kinetics , Male , Mice , Oligonucleotide Array Sequence Analysis , RNA, Messenger/genetics , Receptors, CXCR3 , Receptors, Chemokine/metabolism , Skin/blood supply , Skin/drug effects , Skin/metabolism , TNF-Related Apoptosis-Inducing Ligand/metabolism , Umbilical Cord/metabolism
7.
Carcinogenesis ; 28(2): 404-13, 2007 Feb.
Article in English | MEDLINE | ID: mdl-16952909

ABSTRACT

Several natural compounds, especially plant products and dietary constituents, are able to exhibit 'angiopreventive' (anti-angiogenic chemoprevention) activities both in vitro and in vivo. Deguelin is a rotenoid of the flavonoid family with chemopreventive activities able to decrease tumor incidence in animal models for lung, colon, mammary and skin carcinogenesis through Akt inhibition. Here we show that deguelin belongs to the 'angiopreventive' molecules and provide evidence for molecular events associated with its anti-angiogenic properties. The data show that deguelin inhibits HUVE cells growth by inducing cell-cycle arrest in the G0/G1 phase and in the absence of apoptosis. Growth arrest is associated with induction of p21 and p53 and decreased survivin levels. Deguelin also interferes with several points in the angiogenic process, including inhibition of endothelial cell migration, invasion and metalloprotease production, and potently inhibits in vivo angiogenesis and vascular tumor growth. In addition to Akt, the nuclear factor-kappaB (NF-kappaB) kinase pathway, which plays a critical role in the regulation of inflammation, vascular homeostasis and angiogenesis, was also repressed by deguelin even in the presence of inflammatory stimuli such as tumor necrosis factor-alpha (TNF-alpha). These findings reveal a new therapeutic potential for deguelin in angioprevention and anti-angiogenic therapy.


Subject(s)
Angiogenesis Inhibitors/pharmacology , NF-kappa B/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Rotenone/analogs & derivatives , Base Sequence , Cells, Cultured , DNA Primers , Enzyme-Linked Immunosorbent Assay , Homeostasis , Humans , Polymerase Chain Reaction , Proto-Oncogene Proteins p21(ras)/metabolism , Rotenone/pharmacology , Sarcoma, Kaposi/blood supply , Sarcoma, Kaposi/pathology , Tumor Suppressor Protein p53/metabolism , Up-Regulation
8.
Circ Res ; 100(1): 61-9, 2007 Jan 05.
Article in English | MEDLINE | ID: mdl-17138942

ABSTRACT

Nutlin-3, a nongenotoxic activator of the p53 pathway, dose-dependently (range 0.1 to 10 micromol/L) inhibited the formation of capillaries in an in vivo matrigel assay, as well as the formation of capillary-like structures in an in vitro coculture system composed of endothelial cells surrounded by fibroblasts. In contrast to the chemotherapeutic agent doxorubicin, nutlin-3 showed no induction of apoptosis in vitro either in the cocultures or in isolated vascular endothelial cells, even when used at the highest concentration (10 micromol/L). However, treatment with pharmacological inhibitors of the nuclear factor kappaB and phosphatidylinositol 3-kinase/Akt pathways sensitized endothelial cells to nutlin-3-induced apoptosis. Although nutlin-3 and doxorubicin induced a comparable p53 accumulation in endothelial cells, nutlin-3 was significantly more efficient than doxorubicin in upregulating the p53 target genes CDKN1A/p21, MDM2, and GDF-15, as well as in inhibiting cell cycle progression. However, the predominant in vitro effect of nutlin-3 was its strong antimigratory activity observed at concentrations significantly lower (0.1 micromol/L) than those required to inhibit endothelial cell cycle progression. Taken together, our data suggest that the antiangiogenic activity of nutlin-3 observed in vivo was mainly attributable to inhibition of endothelial cell migration, to some extent attributable to cell cycle arrest, and to a lesser extent attributable to induction of apoptosis.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Imidazoles/pharmacology , Piperazines/pharmacology , Proto-Oncogene Proteins c-mdm2/antagonists & inhibitors , Animals , Apoptosis/drug effects , Capillaries/drug effects , Cell Cycle/drug effects , Cell Movement/drug effects , Cells, Cultured , Endothelial Cells/cytology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelial Cells/physiology , Humans , Mice , Mice, Inbred C57BL , NF-kappa B/antagonists & inhibitors , Neovascularization, Physiologic/drug effects , Phosphoinositide-3 Kinase Inhibitors , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Tumor Suppressor Protein p53/metabolism
9.
FASEB J ; 20(3): 527-9, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16403733

ABSTRACT

Xanthohumol (XN), the principal flavonoid of the hop plant (Humulus lupulus L.) and a constituent of beer, has been suggested to have potential cancer chemopreventive activities. We have observed that most cancer chemopreventive agents show antiangiogenic properties in vitro and in vivo, a concept we termed "angioprevention." Here we show for the first time that XN can inhibit growth of a vascular tumor in vivo. Histopathology and in vivo angiogenesis assays indicated that tumor angiogenesis inhibition was involved. Further, we show the mechanisms for its inhibition of angiogenesis in vivo and related endothelial cell activities in vitro. XN repressed both the NF-kappaB and Akt pathways in endothelial cells, indicating that components of these pathways are major targets in the molecular mechanism of XN. Moreover, using in vitro analyses, we show that XN interferes with several points in the angiogenic process, including inhibition of endothelial cell invasion and migration, growth, and formation of a network of tubular-like structures. Our results suggest that XN can be added to the expanding list of antiangiogenic chemopreventive drugs whose potential in cancer prevention and therapy should be evaluated.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Endothelial Cells/drug effects , Humulus/chemistry , NF-kappa B/antagonists & inhibitors , Propiophenones/pharmacology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , 3T3 Cells/metabolism , Administration, Oral , Animals , Apoptosis/drug effects , Cell Division/drug effects , Cell Line, Transformed/drug effects , Cell Movement/drug effects , Chemotaxis/drug effects , Collagen , Culture Media, Conditioned/pharmacology , Drug Combinations , Drug Screening Assays, Antitumor , Endothelial Cells/metabolism , Endothelial Cells/ultrastructure , Endothelium, Vascular/drug effects , Flavonoids , Humans , I-kappa B Proteins/metabolism , Laminin , Mice , Mice, Nude , Morphogenesis/drug effects , NF-KappaB Inhibitor alpha , Neoplasm Transplantation , Phosphorylation/drug effects , Protein Processing, Post-Translational/drug effects , Protein Transport/drug effects , Proteoglycans , Sarcoma, Kaposi/blood supply , Sarcoma, Kaposi/drug therapy , Sarcoma, Kaposi/pathology
10.
Mutat Res ; 591(1-2): 198-211, 2005 Dec 11.
Article in English | MEDLINE | ID: mdl-16084531

ABSTRACT

The anti-oxidants N-acetyl-l-cysteine (NAC) and (-)-epigallocatechin-3-gallate (EGCG) inhibit tumor vascularization by reducing endothelial cell migration and invasion in a similar, non additive and non synergistic manner but do not alter the growth of human umbilical vein endothelial cells. Here we address the effects of the two chemopreventive drugs on endothelial cell signaling by means of expression profiling and real-time PCR validation. We identify a series of angiogenesis related genes that are similarly regulated by the two drugs. Anti-oxidant treated endothelial cells show gene expression profiles compatible with a less activated, less apoptosis prone and less migratory phenotype. The anti-oxidants affect expression of several components of the TNFalpha response pathway including downstream genes that are regulated in the opposite direction in the absence of the inflammatory cytokine. The interference with the TNFalpha pathway is reflected by reduced NFkappaB activation in anti-oxidants treated cells but the compounds are not able to contrast TNFalpha mediated activation of NFkappaB. The chemopreventive action of these compounds thus relies on a reduction of basal levels of endothelial cell activation. Down-regulation of the TNFalpha responsive pro-metastatic, pro-inflammatory genes, urokinase plasminogen activator and selectin E, further implies anti-metastatic effects for these drugs.


Subject(s)
Acetylcysteine/pharmacology , Anticarcinogenic Agents/pharmacology , Catechin/analogs & derivatives , Endothelial Cells/drug effects , Endothelial Cells/physiology , Free Radical Scavengers/pharmacology , Angiogenesis Inhibitors/pharmacology , Apoptosis/physiology , Catechin/pharmacology , Cell Movement/drug effects , Cells, Cultured , Endothelial Cells/cytology , Gene Expression Profiling , Gene Expression Regulation , Humans , NF-kappa B/metabolism , Oligonucleotide Array Sequence Analysis , RNA, Messenger/metabolism , Reproducibility of Results , Tumor Necrosis Factor-alpha/metabolism
11.
Clin Cancer Res ; 11(12): 4610-9, 2005 Jun 15.
Article in English | MEDLINE | ID: mdl-15958647

ABSTRACT

PURPOSE: Tumor growth appears to be an angiogenesis-dependent process. N-(4-hydroxyphenyl)retinamide (fenretinide; 4HPR) has been found to inhibit and/or prevent tumor growth under diverse conditions. Although 4HPR is antiangiogenic, the molecular mechanisms of this effect remain largely unknown. EXPERIMENTAL DESIGN: Endothelial cells were treated with 4HPR in vitro to study the effects on migration, invasion, and organization, as well as gene expression by microarray and quantitative PCR studies. In vivo angiogenesis was evaluated in the Matrigel model. RESULTS: 4HPR treatment substantially modified the biological activities of endothelial cells, repressing their capacity to migrate, invade, and organize into capillary-like structures. The inhibition of invasion induced by 4HPR was also associated with decreased activities of the metalloproteases matrix metalloproteinase-2 and CD13/APN. Using oligonucleotide microarrays, we observed that bone morphogenetic protein-2 and macrophage inhibitory cytokine-1, two multifunctional cytokines of the transforming growth factor-beta family that regulate the growth, differentiation, apoptosis, and matrix accumulation of a variety of cells, are up-regulated in vitro by 4HPR. Both these molecules specifically inhibited endothelial cell growth, migration, and invasion in vitro and suppressed angiogenesis in the Matrigel plug assay in vivo. Blocking antibodies to bone morphogenetic protein-2 were able to reverse the suppressive effects of 4HPR in vitro and in vivo. CONCLUSIONS: These data support the conclusion that 4HPR inhibits tumor growth by repression of new vessel growth and identify novel points of regulation of angiogenesis in transforming growth factor-beta family proteins.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Bone Morphogenetic Proteins/genetics , Fenretinide/pharmacology , Membrane Proteins/genetics , Transforming Growth Factor beta/genetics , Animals , Bone Morphogenetic Protein 2 , Bone Morphogenetic Proteins/metabolism , Cell Line , Cell Movement/drug effects , Cell Proliferation/drug effects , Collagen , Culture Media, Conditioned/pharmacology , Dose-Response Relationship, Drug , Drug Combinations , Endothelial Cells/cytology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelium, Vascular/drug effects , Endothelium, Vascular/growth & development , Endothelium, Vascular/metabolism , Enzyme-Linked Immunosorbent Assay , Gene Expression/drug effects , Growth Differentiation Factor 15 , Humans , Laminin , Membrane Proteins/metabolism , Metalloproteases/metabolism , Mice , Morphogenesis/drug effects , NIH 3T3 Cells , Oligonucleotide Array Sequence Analysis , Proteoglycans , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Transforming Growth Factor beta/metabolism , Umbilical Veins/cytology
12.
Biochem Pharmacol ; 68(12): 2359-66, 2004 Dec 15.
Article in English | MEDLINE | ID: mdl-15548382

ABSTRACT

Artesunate (ART) is a semi-synthetic derivative of the sesquiterpene artemisinin used for the second line therapy of malaria infections with Plasmodium falciparum. ART also inhibits growth of many transformed cell lines. In the present investigation, we show that ART inhibited the growth of normal human umbilical endothelial cells and of KS-IMM cells that we have established from a Kaposi's sarcoma lesion obtained from a renal transplant patient. The growth inhibitory activity correlated with the induction of apoptosis in KS-IMM cells. Apoptosis was not observed in normal endothelial cells, which, however, showed drastically increased cell doubling times upon ART treatment. ART strongly reduced angiogenesis in vivo in terms of vascularization of Matrigel plugs injected subcutaneously into syngenic mice. We conclude that ART represents a promising candidate drug for the treatment of the highly angiogenic Kaposi's sarcoma. As a low-cost drug, it might be of particular interest for areas of Kaposi's sarcoma endemics. ART could be useful for the prevention of tumor angiogenesis.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Antineoplastic Agents/therapeutic use , Artemisinins/therapeutic use , Sarcoma, Kaposi/drug therapy , Sesquiterpenes/therapeutic use , Animals , Antimalarials/therapeutic use , Apoptosis , Artesunate , Collagen/drug effects , Disease Models, Animal , Drug Combinations , Laminin/drug effects , Male , Metalloproteases/metabolism , Mice , Mice, Inbred C57BL , Mice, Nude , Neoplasm Transplantation , Neovascularization, Physiologic/drug effects , Proteoglycans/drug effects , Xenograft Model Antitumor Assays
13.
J Neuroimmunol ; 148(1-2): 146-53, 2004 Mar.
Article in English | MEDLINE | ID: mdl-14975595

ABSTRACT

Alpha-lipoic acid (alpha-LA) is a neuroprotective metabolic antioxidant that has been shown to cross the blood brain barrier. We tested whether alpha-LA is capable to prevent MOG35-55-induced experimental autoimmune encephalomyelitis (EAE), an established model of multiple sclerosis (MS). Daily oral administration of alpha-LA, starting at the time of immunization, significantly prevented EAE progression as compared to control mice. This was associated with a reduction of CNS infiltrating T cells and macrophages as well as decreased demyelination. We then tested alpha-LA in a therapeutic protocol aimed at suppressing EAE after its onset. Intraperitoneal (i.p.), but not oral, administration of alpha-LA significantly prevented disease progression when compared to vehicle-treated controls. Similarly, we observed significant reduction of demyelination and inflammatory infiltration. This clinical effect was not due to an impairment of MOG35-55 recognition by encephalitogenic T cells. In contrast, MOG-specific T cells showed a decreased production of IFNgamma and IL-4, suggesting an immunosuppressive activity on both Th1 and Th2 cytokines. In addition, alpha-LA inhibited the proteolytic activity of MMP2 and MMP9 only at very high doses. Our data indicate that alpha-LA can effectively interfere with the autoimmune reaction associated with EAE through mechanisms other than its antioxidant activity and supports further studies on the use of alpha-LA as a potential therapy for MS.


Subject(s)
Antioxidants/therapeutic use , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/prevention & control , Thioctic Acid/therapeutic use , Analysis of Variance , Animals , Cell Line, Tumor , Dose-Response Relationship, Drug , Encephalomyelitis, Autoimmune, Experimental/chemically induced , Female , Fibrosarcoma/drug therapy , Glycoproteins , Immunization , Interferon-gamma/metabolism , Interleukin-4/metabolism , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Inbred C57BL , Mycobacterium tuberculosis , Myelin-Oligodendrocyte Glycoprotein , Peptide Fragments , Spinal Cord/drug effects , Spinal Cord/pathology , T-Lymphocytes/drug effects
14.
Cancer Gene Ther ; 11(1): 73-80, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14681728

ABSTRACT

The activity of matrix metalloproteinases (MMPs) is a universal feature of cellular invasion, tumor angiogenesis and metastasis, which is counterbalanced and regulated by the natural tissue inhibitors of MMPs (Timps). Here we show that Timp1 gene transfer delivered by an adeno-associated virus (AAV) vector inhibits tumor growth in a murine xenotransplant model. A human Kaposi's sarcoma cell line, forming highly vascularized tumors in vivo and having a high natural permissivity to AAV gene transfer, was transduced to express the Timp1 cDNA. AAV-Timp1-transduced cells secreted high levels of Timp1 that inhibited MMP2 and MMP9 gelatinolytic activity. Following subcutaneous inoculation in nude mice, the AAV-Timp1-transduced cells showed significantly reduced tumor growth when compared to control AAV-LacZ-transduced cells. In addition, direct intratumoral injection of AAV-Timp1 into pre-existing tumors significantly impaired the further expansion of the tumor mass. Histological analyses showed that the AAV-Timp1-transduced tumors had limited development of vascular structures and extensive areas of cell death, suggesting that Timp1 overexpression had an antiangiogenic effect. To further support this conclusion, we demonstrated that AAV-Timp1 transduction significantly reduced endothelial cell migration and the invasion of a Matrigel barrier and strongly inhibited angiogenesis in the chick chorioallantoic membrane assay. These results indicate that transfer and overexpression of the Timp1 gene is a promising therapeutic strategy to target tumor-associated angiogenesis in cancer gene therapy.


Subject(s)
Dependovirus/genetics , Genetic Therapy , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/therapy , Sarcoma, Kaposi/blood supply , Sarcoma, Kaposi/pathology , Tissue Inhibitor of Metalloproteinase-1/genetics , Tissue Inhibitor of Metalloproteinase-1/therapeutic use , Animals , Basement Membrane/pathology , Cell Division , Cell Line, Tumor , Cell Movement , Dependovirus/physiology , Endothelial Cells/pathology , Gene Transfer Techniques , Humans , Mice , Neoplasm Invasiveness , Neoplasm Transplantation , Sarcoma, Kaposi/genetics , Sarcoma, Kaposi/therapy , Tissue Inhibitor of Metalloproteinase-1/metabolism
15.
Mol Cell Endocrinol ; 193(1-2): 109-20, 2002 Jul 31.
Article in English | MEDLINE | ID: mdl-12161010

ABSTRACT

The first exon of the human androgen receptor (AR) contains a translated CAG (poly-glutamine) repeat. The repeat length is polymorphic in the normal population ranging from 8 to 35 repeats. Expansions to over 40 repeats lead to spinal bulbar muscular atrophy (SBMA), a late onset neurodegenerative disease. The repeat is located between the two parts of a bipartite amino-terminal transactivation function and the repeat length, also within in the normal range, is inversely correlated to the transactivation power of the receptor. P160 type co-activators bind more strongly to shorter repeats. A correlation between AR CAG repeat length and total risk, age at diagnosis, recurrence after surgery and aggressive growth has been reported for tumors of classical androgen target tissues. In the prostate, where androgens exert a mitogenic effect, the cancer risk increases with decreasing AR-CAG repeat length. In contrast, in the breast, where the hormone probably acts as anti-mitogen, a higher risk and earlier onset of breast cancer has been reported for carriers of BRCA1 mutations who also have long CAG repeats in the receptor gene. Somatic alterations during carcinogenesis appear to be frequent in endometrial and in colon cancer. In the endometrium the AR CAG repeat prevalently undergoes expansions consistent with the putative protective function of androgens in this tissue. Frequent repeat reductions during colon carcinogenesis would be consistent with a mitogenic effect of androgens. Analysis of AR protein expression by Western blot reveals expression of the AR in healthy and neoplastic colon tissues. Normal mucosa of the colon expresses both AR-isoforms of 110 and 87 kDa, while the tumor samples have lost the expression of the 110-kDa isoform. The 87-kDa isoform is devoid of the amino-terminal portion of the receptor molecule that also contains the poly-glutamine tract. The temporal and causal relation between isoform switch and somatic repeat reductions during colon carcinogenesis is as yet unclear, but the two events could both enhance p160 mediated androgen signaling. The recent finding that smad3 interacts with the AR in a way similar to p160 links the AR to TGFbeta signaling. Interruption of this signaling pathway is a frequent event in colon carcinogenesis.


Subject(s)
Neoplasms/genetics , Receptors, Androgen/genetics , Trinucleotide Repeats/physiology , Breast Neoplasms/etiology , Breast Neoplasms/genetics , Colonic Neoplasms/etiology , Colonic Neoplasms/genetics , Endometrial Neoplasms/etiology , Endometrial Neoplasms/genetics , Female , Humans , Male , Neoplasms/etiology , Polymorphism, Genetic , Prostatic Neoplasms/etiology , Prostatic Neoplasms/genetics
16.
Endothelium ; 9(1): 3-10, 2002.
Article in English | MEDLINE | ID: mdl-12901356

ABSTRACT

Tumor growth, local invasion, and metastatic dissemination are dependent on the formation of new microvessels. The process of angiogenesis is regulated by a balance between pro-angiogenic and anti-angiogenic factors, and the shift to an angiogenic phenotype (the "angiogenic switch") is a key event in tumor progression. The use of anti-angiogenic agents to restore this balance represents a promising approach to cancer treatment. Known physiological inhibitors include trombospondin, several interleukins, and the proteolytic break-down products of several proteins. Angiostatin, an internal fragment of plasminogen, is one of the more potent of this latter class of angiogenesis inhibitors. Like endostatin, another anti-angiogenic peptide derived from collagen XVIII, angiostatin can induce tumor vasculature regression, leading to a complete cessation of tumor growth. Inhibitors of angiogenesis target normal endothelial cells, therefore the development of resistance to these drugs is unlikely. The efficacy of angiostatin has been demonstrated in animal models for many different types of solid tumors. Anti-angiogenic cancer therapy with angiostatin requires prolonged administration of the peptide. The production of the functional polypeptides is expensive and technical problems related to physical properties and purity are frequently encountered. Gene transfer represents an alternative method to deliver angiostatin. Gene therapy has the potential to produce the therapeutic agent in high concentrations in a local area for a sustained period, thereby avoiding the problems encountered with long-term administration of recombinant proteins, monoclonal antibodies, or anti-angiogenic drugs. In this review we compare the different gene therapy strategies that have been applied to angiostatin, with special regard to their ability to provide sufficient angiostatin at the target site.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Genetic Therapy/methods , Neoplasms/therapy , Neovascularization, Pathologic/therapy , Peptide Fragments/therapeutic use , Plasminogen/therapeutic use , Angiostatins , Animals , Genetic Vectors , Humans , Neoplasms/blood supply , Neoplasms/drug therapy , Neovascularization, Pathologic/drug therapy , Peptide Fragments/genetics , Plasminogen/genetics , Recombinant Proteins/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...