Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
J Transl Med ; 9: 59, 2011 May 12.
Article in English | MEDLINE | ID: mdl-21569399

ABSTRACT

BACKGROUND: Implantation and growth of metastatic cancer cells at distant organs is promoted by inflammation-dependent mechanisms. A hepatic melanoma metastasis model where a majority of metastases are generated via interleukin-18-dependent mechanisms was used to test whether anti-inflammatory properties of resveratrol can interfere with mechanisms of metastasis. METHODS: Two experimental treatment schedules were used: 1) Mice received one daily oral dose of 1 mg/kg resveratrol after cancer cell injection and the metastasis number and volume were determined on day 12. 2) Mice received one daily oral dose of 1 mg/kg resveratrol along the 5 days prior to the injection of cancer cells and both interleukin-18 (IL-18) concentration in the hepatic blood and microvascular retention of luciferase-transfected B16M cells were determined on the 18th hour. In vitro, primary cultured hepatic sinusoidal endothelial cells were treated with B16M-conditioned medium to mimic their in vivo activation by tumor-derived factors and the effect of resveratrol on IL-18 secretion, on vascular cell adhesion molecule-1 (VCAM-1) expression and on tumor cell adhesion were studied. The effect of resveratrol on melanoma cell activation by IL-18 was also studied. RESULTS: Resveratrol remarkably inhibited hepatic retention and metastatic growth of melanoma cells by 50% and 75%, respectively. The mechanism involved IL-18 blockade at three levels: First, resveratrol prevented IL-18 augmentation in the blood of melanoma cell-infiltrated livers. Second, resveratrol inhibited IL-18-dependent expression of VCAM-1 by tumor-activated hepatic sinusoidal endothelium, preventing melanoma cell adhesion to the microvasculature. Third, resveratrol inhibited adhesion- and proliferation-stimulating effects of IL-18 on metastatic melanoma cells through hydrogen peroxide-dependent nuclear factor-kappaB translocation blockade on these cells. CONCLUSIONS: These results demonstrate multiple sites for therapeutic intervention using resveratrol within the prometastatic microenvironment generated by tumor-induced hepatic IL-18, and suggest a remarkable effect of resveratrol in the prevention of inflammation-dependent melanoma metastasis in the liver.


Subject(s)
Inflammation/prevention & control , Interleukin-18/metabolism , Liver Neoplasms/pathology , Melanoma/prevention & control , Melanoma/secondary , Stilbenes/therapeutic use , Animals , Cell Adhesion/drug effects , Cell Proliferation/drug effects , Endothelium/drug effects , Endothelium/metabolism , Endothelium/pathology , Inflammation/complications , Liver/blood supply , Liver/drug effects , Liver/pathology , Liver Neoplasms/complications , Melanoma/complications , Melanoma/metabolism , Melanoma, Experimental/complications , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Microvessels/drug effects , Microvessels/pathology , Models, Biological , Neoplasm Transplantation , Resveratrol , Stilbenes/pharmacology , Tumor Microenvironment/drug effects , Vascular Cell Adhesion Molecule-1/metabolism
2.
Cancer Metastasis Rev ; 25(3): 417-34, 2006 Sep.
Article in English | MEDLINE | ID: mdl-17001512

ABSTRACT

Interleukin-18 (IL-18, interferon [IFN]-gamma-inducing factor) is a proinflammatory cytokine converted to a biologically active molecule by interleukin (IL)-1beta converting enzyme (caspase-1). A wide range of normal and cancer cell types can produce and respond to IL-18 through a specific receptor (IL-18R) belonging to the toll-like receptor family. The activity of IL-18 is regulated by IL-18-binding protein (IL-18bp), a secreted protein possessing the ability to neutralize IL-18 and whose blood level is affected by renal function and is induced by IFNgamma. IL-18 plays a central role in inflammation and immune response, contributing to the pathogenesis and pathophysiology of infectious and inflammatory diseases. Because immune-stimulating effects of IL-18 have antineoplastic properties, IL-18 has been proposed as a novel adjuvant therapy against cancer. However, IL-18 increases in the blood of the majority of cancer patients and has been associated with disease progression and, in some cancer types, with metastatic recurrence risk and poor clinical outcome and survival. Under experimental conditions, cancer cells can also escape immune recognition, increase their adherence to the microvascular wall and even induce production of angiogenic and tumor growth-stimulating factors via IL-18-dependent mechanism. This is particularly visible in melanoma cells. Thus, the role of IL-18 in cancer progression and metastasis remains controversial. This review examines the clinical correlations and biological effects of IL-18 during cancer development and highlights recent experimental insights into prometastatic and proangiogenic effects of IL-18 and the use of IL-18bp against cancer progression.


Subject(s)
Interleukin-18/pharmacology , Interleukin-18/physiology , Neoplasms/immunology , Neoplasms/pathology , Tumor Escape , Animals , Cell Line, Tumor , Disease Progression , Humans , Interleukin-18/genetics , Mice , Neoplasms/blood supply , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/pathology
3.
Cancer Res ; 64(1): 304-10, 2004 Jan 01.
Article in English | MEDLINE | ID: mdl-14729638

ABSTRACT

We investigated effects of endostatin (ES) in the prometastatic microenvironment of inflammation occurring during the microvascular phase of cancer cell infiltration in the liver. We used a model of intrasplenic injection of B16 melanoma (B16M) cells leading to hepatic metastasis through vascular cell adhesion molecule-(VCAM-1)-mediated capillary arrest of cancer cells via interleukin-18 (IL-18)-dependent mechanism. We show that administration of 50 mg/kg recombinant human (rh) ES 30 min before B16M, plus repetition of same dose for 3 additional days decreased metastasis number by 60%. A single dose of rhES before B16M injection reduced hepatic microvascular retention of luciferase-transfected B16M by 40% and inhibited hepatic production of tumor necrosis factor alpha (TNF-alpha) and IL-18 and VCAM-1 expression by hepatic sinusoidal endothelia (HSE). Consistent with these data, rhES inhibited VCAM-1-dependent B16M cell adhesion to primary cultured HSE receiving B16M conditioned medium, and it abolished the HSE cell production of TNF-alpha and IL-18 induced by tumor-derived vascular endothelial cell growth factor (VEGF). rhES abrogated recombinant murine VEGF-induced tyrosine phosphorylation of KDR/flk-1 receptor in HSE cells, preventing the proinflammatory action of tumor-derived VEGF on HSE. rhES also abolished hepatic production of TNF-alpha, microvascular retention of luciferase-transfected B16M, and adhesion of B16M cells to isolated HSE cells, all of them induced in mice given 5 micro g/kg recombinant murine VEGF for 18 h. This capillary inflammation-deactivating capability constitutes a nonantiangiogenic antitumoral action of endostatin that decreases cancer cell arrest within liver microvasculature and prevents metastases promoted by proinflammatory cytokines induced by VEGF.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Endostatins/pharmacology , Liver Neoplasms/blood supply , Liver Neoplasms/secondary , Melanoma, Experimental/blood supply , Microcirculation/drug effects , Animals , Disease Models, Animal , Humans , Liver Neoplasms/prevention & control , Male , Mice , Mice, Inbred C57BL , Recombinant Proteins/pharmacology , Tumor Cells, Cultured , Vascular Cell Adhesion Molecule-1/analysis
4.
Hepatology ; 37(3): 674-85, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12601365

ABSTRACT

Myofibroblasts infiltrate malignant liver tumors, although their pathogenic implications are unclear. Immunohistochemical detection of alpha-smooth muscle actin, glial fibrillary acidic protein (GFAP), and CD31 and CD34 expression was used to analyze the contribution of myofibroblasts to angiogenesis in hepatic metastasis produced by intrasplenically-injected B16 melanoma (B16M). Because activated hepatic stellate cells (HSCs) are oxygen-sensing myofibroblasts producing vascular endothelial growth factor (VEGF), the effect of B16M and human A375 melanoma supernatants on VEGF production by immortalized rat HSC line T6 and primary cultured human HSCs also was studied under an hypoxic atmosphere mimicking a tumor microenvironment. Myofibroblast infiltration preceded endothelium recruitment in avascular micrometastasis and generated specific stroma for sinusoidal-type and portal-type angiogeneses. Thereafter, myofibroblasts and endothelial cells colocalized within both angiogenic patterns and their numerical densities correlated with metastasis development. Myofibroblasts often were GFAP-positive, suggesting an HSC origin. Melanoma supernatants stimulated VEGF messenger RNA and protein synthesis by HSCs. These effects were potentiated by hypoxia. VEGF up-regulation was accompanied by increased expression of cyclooxygenase type 2 (COX-2) and PGE2 synthesis. HSC production of VEGF decreased under COX-2 inhibition, whereas it was increased by exogenous PGE2. The high VEGF expression in HSCs induced by melanoma factors and hypoxia resulted in mitogenic, antiapoptotic, and motogenic stimulation of both murine hepatic sinusoidal endothelium and human umbilical vein endothelium. In conclusion, temporal and positional relationships evolve between myofibroblast and endothelium recruitment during metastasis development. Mechanistically, hypoxic induction of VEGF in tumor-activated HSCs may create a proangiogenic microenvironment, facilitating endothelial cell recruitment and survival during hepatic metastasis transition from an avascular to a vascular stage.


Subject(s)
Liver Neoplasms/secondary , Liver/pathology , Melanoma, Experimental/pathology , Neovascularization, Pathologic , Animals , Apoptosis , Cell Division , Cell Hypoxia , Cell Line , Cell Movement , Cyclooxygenase 2 , Endothelial Growth Factors/analysis , Endothelial Growth Factors/pharmacology , Endothelium, Vascular/pathology , Glial Fibrillary Acidic Protein/analysis , Intercellular Signaling Peptides and Proteins/analysis , Intercellular Signaling Peptides and Proteins/pharmacology , Isoenzymes/analysis , Liver Neoplasms/pathology , Lymphokines/analysis , Lymphokines/pharmacology , Male , Mice , Mice, Inbred C57BL , Neoplasm Transplantation , Prostaglandin-Endoperoxide Synthases/analysis , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
5.
Cancer Res ; 63(2): 491-7, 2003 Jan 15.
Article in English | MEDLINE | ID: mdl-12543807

ABSTRACT

We studied the role of endogenous interleukin (IL)-18 in hepatic metastasis by blocking this cytokine using the naturally occurring IL-18 binding protein (IL-18BP). A single i.p. dose of IL-18BP given 30 min before intrasplenic injection of murine B16 melanoma (B16M) cells reduced the number of hepatic metastatic foci by 75% and metastatic volume by 80%. Same treatment reduced the intrahepatic retention of luciferase-transfected B16M by 50% and abolished VCAM-1 up-regulation in the hepatic microvasculature, as assessed by reverse transcription-PCR, Western blot, and immunohistochemistry. Twelve hours after IL-18BP, hepatic sinusoidal endothelium (HSE) cells were isolated, and adhesion of B16M cells to these cultured HSE cells was reduced to the level of vehicle-treated mice. IL-18BP treatment of mice with established micrometastases resulted in a 25% decrease in metastasis number and 40% decrease in metastasis volume, suggesting inhibition of endogenous growth factors. Indeed, the addition of IL-18BP to normal HSE abolished the release of melanoma cell growth factor(s) induced by B16M. IL-18 promoted the in vitro growth of B16M and human melanoma cells, which was IL-1 dependent. These data demonstrate a significant role of endogenous IL-18 on hepatic metastasis by up-regulating melanoma cell adhesion to HSE cells and tumor growth, implicating a possible antimetastatic benefit of neutralizing IL-18.


Subject(s)
Endothelium, Vascular/metabolism , Glycoproteins/pharmacology , Liver Neoplasms, Experimental/prevention & control , Liver Neoplasms, Experimental/secondary , Melanoma, Experimental/drug therapy , Melanoma, Experimental/secondary , Animals , Cell Adhesion/drug effects , Cell Division/drug effects , Endothelial Growth Factors/metabolism , Endothelial Growth Factors/pharmacology , Humans , Intercellular Signaling Peptides and Proteins , Interleukin-1/physiology , Interleukin-18/antagonists & inhibitors , Interleukin-18/blood , Interleukin-18/pharmacology , Liver Neoplasms, Experimental/pathology , Male , Melanoma, Experimental/pathology , Melanoma, Experimental/prevention & control , Mice , Mice, Inbred C57BL , Recombinant Proteins/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...