Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters










Database
Language
Publication year range
1.
FASEB J ; 21(14): 3928-36, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17622568

ABSTRACT

In light of the involvement of tumor-associated macrophages (TAM) in the promotion of tumor growth and metastasis, strategies to prevent TAM recruitment within the tumor microenvironment are currently under investigation. The recent observation that angiostatin reduces macrophage infiltration in an atherosclerosis model prompted our laboratory to further explore the use of human plasminogen angiostatin (hK1-3) protein as a macrophage modulatory agent. We demonstrate that hK1-3 blocks migration of murine peritoneal macrophages (91% decrease, P<0.00005) and human monocytes (85% decrease, P<0.05) in vitro. Cell viability of hK1-3-treated cells is not affected, as determined by fluorochrome-labeled inhibitors of caspase-propidium iodide (FLICA/PI) flow cytometry analysis. Furthermore, confocal microscopy of phalloidin-stained cells reveals that hK1-3 leads to disruption of actin filopodia/lamellipodia in human monocytes and induces distinct podosome accumulation in mature differentiated macrophages. Paradoxically, we observed a 3.5-fold increase in secretion and a 3- to 5.5-fold increase in gelatinolytic activity of macrophage-produced matrix metalloproteinase-9, which we suggest is a cellular response to compensate for the dominant static effect of hK1-3 on actin. We also demonstrate that hK1-3 induces the phosphorylation of extracellular signal-regulated kinase (ERK1/2) in human monocytes. hK1-3-mediated macrophage immobilization has the potential to be exploited therapeutically in pathological conditions associated with cellular hypoxia, such as cancer and atherosclerosis.


Subject(s)
Actins/antagonists & inhibitors , Angiostatins/physiology , Cell Migration Inhibition/physiology , Cytoskeleton/metabolism , Macrophages, Peritoneal/metabolism , Monocytes/metabolism , Actins/metabolism , Animals , Cells, Cultured , Cytoskeleton/pathology , Humans , Kringles/physiology , Macrophages, Peritoneal/pathology , Mice , Mice, Inbred C57BL , Monocytes/pathology , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Peptide Fragments/physiology
2.
Mol Cancer Ther ; 6(2): 441-9, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17308045

ABSTRACT

Proteolytic processing of human plasminogen generates potent antiangiogenic peptides such as angiostatin. The plasminogen kringle 5 (K5) domain, which is distinct from angiostatin, possesses potent antiangiogenic properties on its own, which can be exploited in cancer therapy. It has been recently observed that antiangiogenic agents promote leukocyte-vessel wall interaction as part of their antitumor effect. Although we have previously shown that K5 suppresses cancer growth in tumor xenograft models, its modulation of inflammation in experimental mice with intact immune systems is unknown. To determine whether K5 possesses immune proinflammatory properties, we investigated the effects of K5 in an immune competent model of breast cancer and observed that tumor rejection is substantially reduced in nonobese diabetic/severe combined immunodeficient and BALB/c nude when compared with wild-type BALB/c mice, suggesting an important role for T-lymphoid cells in the antitumor effect of K5. Tumor explant analysis shows that K5 enhances tumor recruitment of CD3(+) lymphoid cells, in particular, the NKT phenotype. We also observed a significant decrease in tumor-associated microvessel length and density consistent with antiangiogenic activity. Histologic analysis of K5 tumors also revealed a robust neutrophilic infiltrate, which may be explained by the neutrophil chemotactic activity of K5 as well as its ability to promote CD64 up-regulation within the CD11b(+) adhesive neutrophil population. In sum, our findings confirm that the K5 protein acts as a potent angiostatic agent and possesses a novel proinflammatory role via its ability to recruit tumor-associated neutrophils and NKT lymphocytes, leading to a potent antitumor response.


Subject(s)
Angiogenesis Inhibitors/metabolism , Mammary Neoplasms, Experimental/therapy , Peptide Fragments/physiology , Plasminogen/physiology , Signal Transduction , Adenocarcinoma/blood supply , Adenocarcinoma/immunology , Adenocarcinoma/therapy , Animals , CD3 Complex/metabolism , Collagen/metabolism , Disease Progression , Drug Combinations , Genes, MHC Class I/physiology , Humans , Immunity, Cellular , Inflammation/metabolism , Laminin/metabolism , Lymphocytes/immunology , Lymphocytes/metabolism , Lymphocytes, Tumor-Infiltrating/pathology , Mammary Neoplasms, Experimental/blood supply , Mammary Neoplasms, Experimental/immunology , Mice , Mice, Inbred BALB C , Mice, Nude , Neovascularization, Pathologic/prevention & control , Neutrophils/metabolism , Proteoglycans/metabolism , Retroviridae , Survival Rate , Xenograft Model Antitumor Assays
3.
Cancer Res ; 65(18): 8359-65, 2005 Sep 15.
Article in English | MEDLINE | ID: mdl-16166313

ABSTRACT

Angiostatin, a well-characterized angiostatic agent, is a proteolytic cleavage product of human plasminogen encompassing the first four kringle structures. The fifth kringle domain (K5) of human plasminogen is distinct from angiostatin and has been shown, on its own, to act as a potent endothelial cell inhibitor. We propose that tumor-targeted K5 cDNA expression may act as an effective therapeutic intervention as part of a cancer gene therapy strategy. In this study, we provide evidence that eukaryotically expressed His-tagged human K5 cDNA (hK5His) is exported extracellularly and maintains predicted disulfide bridging conformation in solution. Functionally, hK5His protein produced by retrovirally engineered human U87MG glioma cells suppresses in vitro migration of both human umbilical vein endothelial cells and human macrophages. Subcutaneous implantation of Matrigel-embedded hK5His-producing glioma cells in nonobese diabetic/severe combined immunodeficient mice reveals that hK5His induces a marked reduction in blood vessel formation and significantly suppresses the recruitment of tumor-infiltrating CD45+ Mac3+ Gr1- macrophages. Therapeutically, we show in a nude mouse orthotopic brain cancer model that tumor-targeted K5 expression is capable of effectively suppressing glioma growth and promotes significant long-term survival (>120 days) of test animals. These data suggest that plasminogen K5 acts as a novel two-pronged anticancer agent, mediating its inhibitory effect via its action on host-derived endothelial cells and tumor-associated macrophages, resulting in a potent, clinically relevant antitumor effect.


Subject(s)
Angiogenesis Inhibitors/genetics , Brain Neoplasms/blood supply , Brain Neoplasms/therapy , Glioma/blood supply , Glioma/therapy , Macrophages/pathology , Peptide Fragments/genetics , Plasminogen/genetics , Amino Acid Sequence , Angiogenesis Inhibitors/biosynthesis , Angiogenesis Inhibitors/chemistry , Animals , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Movement/genetics , DNA, Complementary/genetics , Disease Progression , Female , Genetic Therapy/methods , Glioma/genetics , Glioma/pathology , Humans , Mice , Mice, Inbred NOD , Mice, Nude , Mice, SCID , Molecular Sequence Data , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/pathology , Neovascularization, Pathologic/therapy , Peptide Fragments/biosynthesis , Peptide Fragments/chemistry , Plasminogen/biosynthesis , Plasminogen/chemistry , Protein Conformation , Protein Engineering , Retroviridae/genetics , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Transfection , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...