Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
Add more filters










Publication year range
1.
Theranostics ; 10(9): 4116-4133, 2020.
Article in English | MEDLINE | ID: mdl-32226543

ABSTRACT

Background: CUB domain-containing protein 1 (CDCP1) is a cell surface receptor regulating key signalling pathways in malignant cells. CDCP1 has been proposed as a molecular target to abrogate oncogenic signalling pathways and specifically deliver anti-cancer agents to tumors. However, the development of CDCP1-targeting agents has been questioned by its frequent proteolytic processing which was thought to result in shedding of the CDCP1 extracellular domain limiting its targetability. In this study, we investigated the relevance of targeting CDCP1 in the context of pancreatic ductal adenocarcinoma (PDAC) and assess the impact of CDCP1 proteolysis on the effectiveness of CDCP1 targeting agents. Methods: The involvement of CDCP1 in PDAC progression was assessed by association analysis in several PDAC cohorts and the proteolytic processing of CDCP1 was evaluated in PDAC cell lines and patient-derived cells. The consequences of CDCP1 proteolysis on its targetability in PDAC cells was assessed using immunoprecipitation, immunostaining and biochemical assays. The involvement of CDCP1 in PDAC progression was examined by loss-of-function in vitro and in vivo experiments employing PDAC cells expressing intact or cleaved CDCP1. Finally, we generated antibody-based imaging and therapeutic agents targeting CDCP1 to demonstrate the feasibility of targeting this receptor for detection and treatment of PDAC tumors. Results: High CDCP1 expression in PDAC is significantly associated with poorer patient survival. In PDAC cells proteolysis of CDCP1 does not always result in the shedding of CDCP1-extracellular domain which can interact with membrane-bound CDCP1 allowing signal transduction between the different CDCP1-fragments. Targeting CDCP1 impairs PDAC cell functions and PDAC tumor growth independently of CDCP1 cleavage status. A CDCP1-targeting antibody is highly effective at delivering imaging radionuclides and cytotoxins to PDAC cells allowing specific detection of tumors by PET/CT imaging and superior anti-tumor effects compared to gemcitabine in in vivo models. Conclusion: Independent of its cleavage status, CDCP1 exerts oncogenic functions in PDAC and has significant potential to be targeted for improved radiological staging and treatment of this cancer. Its elevated expression by most PDAC tumors and lack of expression by normal pancreas and other major organs, suggest that targeting CDCP1 could benefit a significant proportion of PDAC patients. These data support the further development of CDCP1-targeting agents as personalizable tools for effective imaging and treatment of PDAC.


Subject(s)
Antigens, Neoplasm/metabolism , Carcinoma, Pancreatic Ductal/metabolism , Cell Adhesion Molecules/metabolism , Pancreatic Neoplasms/metabolism , Animals , Biomarkers, Tumor/metabolism , Carcinoma, Pancreatic Ductal/therapy , Cell Line, Tumor , Disease Progression , Gene Expression Regulation, Neoplastic , Humans , Mice , Pancreatic Neoplasms/therapy , Precision Medicine , Proteolysis
2.
Oncogene ; 37(14): 1815-1829, 2018 04.
Article in English | MEDLINE | ID: mdl-29348457

ABSTRACT

Latent transforming growth factor ß (TGFß)-binding proteins (LTBPs) are important for the secretion, activation, and function of mature TGFß, especially so in cancer cell physiology. However, specific roles of the LTBPs remain understudied in the context of the primary tumor microenvironment. Herein, we investigated the role of LTBP3 in the distinct processes involved in cancer metastasis. By using three human tumor cell lines of different tissue origin (epidermoid HEp-3 and prostate PC-3 carcinomas and HT-1080 fibrosarcoma) and several metastasis models conducted in both mammalian and avian settings, we show that LTBP3 is involved in the early dissemination of primary cancer cells, namely in the intravasation step of the metastatic cascade. Knockdown of LTBP3 in all tested cell lines led to significant inhibition of tumor cell intravasation, but did not affect primary tumor growth. LTBP3 was dispensable in the late steps of carcinoma cell metastasis that follow tumor cell intravasation, including vascular arrest, extravasation, and tissue colonization. However, LTBP3 depletion diminished the angiogenesis-inducing potential of HEp-3 cells in vivo, which was restorable by exogenous delivery of LTBP3 protein. A similar compensatory approach rescued the dampened intravasation of LTBP3-deficient HEp-3 cells, suggesting that LTBP3 regulates the induction of the intravasation-supporting angiogenic vasculature within developing primary tumors. Using our recently developed microtumor model, we confirmed that LTBP3 loss resulted in the development of intratumoral vessels with an abnormal microarchitecture incompatible with efficient intravasation of HEp-3 carcinoma cells. Collectively, these findings demonstrate that LTBP3 represents a novel oncotarget that has distinctive functions in the regulation of angiogenesis-dependent tumor cell intravasation, a critical process during early cancer dissemination. Our experimental data are also consistent with the survival prognostic value of LTBP3 expression in early-stage head and neck squamous cell carcinomas, further indicating a specific role for LTBP3 in cancer progression toward metastatic disease.


Subject(s)
Latent TGF-beta Binding Proteins/physiology , Neoplasms/genetics , Neoplasms/pathology , Animals , Cell Line, Tumor , Chick Embryo , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Latent TGF-beta Binding Proteins/antagonists & inhibitors , Latent TGF-beta Binding Proteins/genetics , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplasms/mortality , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/pathology , RNA, Small Interfering/pharmacology , Survival Analysis
3.
Cell Rep ; 19(3): 601-616, 2017 04 18.
Article in English | MEDLINE | ID: mdl-28423322

ABSTRACT

Intravasation, active entry of cancer cells into the circulation, is often considered to be a relatively late event in tumor development occurring after stromal invasion. Here, we provide evidence that intravasation can be initiated early during tumor development and proceed in parallel to or independent of tumor invasion into surrounding stroma. By applying direct and unbiased intravasation-scoring methods to two histologically distinct human cancer types in live-animal models, we demonstrate that intravasation takes place almost exclusively within the tumor core, involves intratumoral vasculature, and does not involve vasculotropic cancer cells invading tumor-adjacent stroma and migrating along tumor-converging blood vessels. Highlighting an additional role for EGFR in cancer, we find that EGFR is required for the development of an intravasation-sustaining intratumoral vasculature. Intratumoral localization of intravasation supports the notion that overt metastases in cancer patients could be initiated much earlier during cancer progression than appreciated within conventional clinical tumor staging systems.


Subject(s)
Cell Movement , Neoplasms/pathology , Animals , Cell Line, Tumor , Chick Embryo , Disease Models, Animal , Ear/pathology , ErbB Receptors/metabolism , Green Fluorescent Proteins/metabolism , Humans , Hypoxia/pathology , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, SCID , Neoplasm Invasiveness , Neoplasm Metastasis , Neovascularization, Pathologic , Permeability , Stromal Cells/pathology
4.
Methods Mol Biol ; 1430: 283-98, 2016.
Article in English | MEDLINE | ID: mdl-27172961

ABSTRACT

The mechanisms governing the development of angiogenic blood vessels, which not only deliver the nutrients to growing tumors but also provide the conduits for tumor cell dissemination, are still not fully resolved. The model systems based on the grafting of human tumor cells onto the chorioallantoic membrane (CAM) of the chick embryo offer several advantages to study complex processes underlying tumor angiogenesis and tumor cell dissemination. In particular, the CAM model described here allows for investigation of multiple microtumors as independent entities, thereby greatly facilitating quantification and statistical analyses of tumor neovascularization and cancer spreading. This CAM microtumor system was designed specifically to measure the level of tumor cell intravasation in combination with quantitative analyses of the microarchitecture and permeability of the intratumoral angiogenic blood vessels. By using this newly established microtumor model we have demonstrated the functional involvement of tumor matrix metalloproteinase-1 (MMP-1) and epidermal growth factor receptor (EGFR) in regulating the development of a distinct angiogenic vasculature capable of sustaining tumor cell intravasation and metastasis.


Subject(s)
Chorioallantoic Membrane/blood supply , Chorioallantoic Membrane/pathology , Neovascularization, Pathologic/metabolism , Animals , Capillary Permeability , Cell Line, Tumor , Chick Embryo , Chorioallantoic Membrane/metabolism , ErbB Receptors/metabolism , Humans , Matrix Metalloproteinase 1/metabolism , Neoplasm Invasiveness , Neoplasm Transplantation
5.
Neoplasia ; 17(8): 634-49, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26408256

ABSTRACT

Many malignant characteristics of cancer cells are regulated through pathways induced by the tyrosine kinase activity of the epidermal growth factor receptor (EGFR). Herein, we show that besides directly affecting the biology of cancer cells per se, EGFR also regulates the primary tumor microenvironment. Specifically, our findings demonstrate that both the expression and signaling activity of EGFR are required for the induction of a distinct intratumoral vasculature capable of sustaining tumor cell intravasation, a critical rate-limiting step in the metastatic cascade. An intravasation-sustaining mode of intratumoral angiogenic vessels depends on high levels of tumor cell EGFR and the interplay between EGFR-regulated production of interleukin 8 by tumor cells, interleukin-8-induced influx of tumor-infiltrating neutrophils delivering their unique matrix metalloproteinase-9, and neutrophil matrix metalloproteinase-9-dependent release of the vascular permeability and endothelial growth factor, VEGF. Our data indicate that through VEGF-mediated disruption of endothelial layer integrity and increase of intratumoral vasculature permeability, EGFR activity significantly facilitates active intravasation of cancer cells. Therefore, this study unraveled an important but overlooked function of EGFR in cancer, namely, its ability to create an intravasation-sustaining microenvironment within the developing primary tumor by orchestrating several interrelated processes required for the initial steps of cancer metastasis through vascular routes. Our findings also suggest that EGFR-targeted therapies might be more effective when implemented in cancer patients with early-staged primary tumors containing a VEGF-dependent angiogenic vasculature. Accordingly, early EGFR inhibition combined with various anti-VEGF approaches could synergistically suppress tumor cell intravasation through inhibiting the highly permeable angiogenic vasculature induced by EGFR-overexpressing aggressive cancer cells.


Subject(s)
Chorioallantoic Membrane/blood supply , ErbB Receptors/metabolism , Neoplasms/metabolism , Neovascularization, Pathologic/metabolism , Animals , Blotting, Western , Cell Line, Tumor , Chick Embryo , Chorioallantoic Membrane/metabolism , Chorioallantoic Membrane/pathology , ErbB Receptors/genetics , Gene Expression Regulation, Neoplastic , Humans , Matrix Metalloproteinase 9/metabolism , Microscopy, Fluorescence , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplasm Transplantation , Neoplasms/blood supply , Neoplasms/genetics , Neovascularization, Pathologic/genetics , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Tumor Microenvironment/genetics , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
6.
Matrix Biol ; 44-46: 94-112, 2015.
Article in English | MEDLINE | ID: mdl-25912949

ABSTRACT

Metastasis is a distinct stage of cancer progression that requires the development of angiogenic blood vessels serving as conduits for tumor cell dissemination. An accumulated body of evidence indicates that metastasis-supporting neovasculature should possess certain structural characteristics allowing for the process of tumor cell intravasation, an active entry of cancer cells into the vessel interior. It appears that the development of tumor vessels with lumens of a distinctive size and support of these vessels by a discontinuous pericyte coverage constitute critical microarchitectural requirements to: (a) provide accessible points for vessel wall penetration by primary tumor cells; (b) provide enough lumen space for a tumor cell or cell aggregate upon intravasation; and (c) allow for sufficient rate of blood flow to carry away intravasated cells from the primary tumor to the next, proximal or distal site. This review will primarily focus on the functional roles of matrix metalloproteinases (MMPs), which catalytically trigger the development of an intravasation-sustaining neovasculature at the early stages of tumor growth and are also required for the maintenance of a metastasis-supporting state of blood vessels at later stages of cancer progression.


Subject(s)
Matrix Metalloproteinases/metabolism , Neoplasms/pathology , Neovascularization, Pathologic/enzymology , Animals , Humans , Neoplasm Metastasis , Neoplasms/blood supply , Neoplasms/metabolism , Vascular Endothelial Growth Factor A/metabolism
7.
Biochem J ; 465(2): 259-70, 2015 Jan 15.
Article in English | MEDLINE | ID: mdl-25360794

ABSTRACT

Gelatinase B/matrix metalloproteinase-9 (MMP-9) (EC 3.4.24.35) cleaves many substrates and is produced by most cell types as a zymogen, proMMP-9, in complex with the tissue inhibitor of metalloproteinases-1 (TIMP-1). Natural proMMP-9 occurs as monomers, homomultimers and heterocomplexes, but our knowledge about the overall structure of proMMP-9 monomers and multimers is limited. We investigated biochemical, biophysical and functional characteristics of zymogen and activated forms of MMP-9 monomers and multimers. In contrast with a conventional notion of a dimeric nature of MMP-9 homomultimers, we demonstrate that these are reduction-sensitive trimers. Based on the information from electrophoresis, AFM and TEM, we generated a 3D structure model of the proMMP-9 trimer. Remarkably, the proMMP-9 trimers possessed a 50-fold higher affinity for TIMP-1 than the monomers. In vivo, this finding was reflected in a higher extent of TIMP-1 inhibition of angiogenesis induced by trimers compared with monomers. Our results show that proMMP-9 trimers constitute a novel structural and functional entity that is differentially regulated by TIMP-1.


Subject(s)
Enzyme Precursors/chemistry , Matrix Metalloproteinase 9/chemistry , Models, Molecular , Multiprotein Complexes/chemistry , Tissue Inhibitor of Metalloproteinase-1/chemistry , Enzyme Precursors/genetics , Enzyme Precursors/metabolism , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Microscopy, Atomic Force , Microscopy, Electron, Transmission , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Multiprotein Complexes/ultrastructure , Tissue Inhibitor of Metalloproteinase-1/genetics , Tissue Inhibitor of Metalloproteinase-1/metabolism
8.
Neoplasia ; 16(10): 771-88, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25379015

ABSTRACT

According to established notion, one of the major angiogenesis-inducing factors, pro-matrix metalloproteinase-9 (proMMP-9), is supplied to the tumor microenvironment by tumor-associated macrophages (TAMs). Accumulated evidence, however, indicates that tumor-associated neutrophils (TANs) are also critically important for proMMP-9 delivery, especially at early stages of tumor development. To clarify how much angiogenic proMMP-9 is actually contributed by TAMs and TANs, we quantitatively evaluated TAMs and TANs from different tumor types, including human xenografts and syngeneic murine tumors grown in wild-type and Mmp9-knockout mice. Whereas host MMP-9 competence was required for full angiogenic potential of both normal and tumor-associated leukocytes, direct comparisons of neutrophils versus macrophages and TANs versus TAMs demonstrated that macrophages and TAMs secrete 40- to 50-fold less proMMP-9 than the same numbers of neutrophils or TANs. Correspondingly, the levels of MMP-9-mediated in vivo angiogenesis induced by neutrophils and TANs substantially exceeded those induced by macrophages and TAMs. MMP-9-delivering TANs were also required for development of metastasis-supporting intratumoral vasculature, characterized by ≥ 11-µm size lumens and partial coverage with stabilizing pericytes. Importantly, MMP-9-producing TAMs exhibit M2-skewed phenotype but do not express tissue inhibitor of metalloproteinases-1 (TIMP-1), a novel characteristic allowing them to secrete TIMP-1-free, neutrophil-like MMP-9 zymogen unencumbered by its natural inhibitor. Together, our findings support the notion whereby TANs, capable of immediate release of their pre-stored cargo, are the major contributors of highly angiogenic MMP-9, whereas tumor-influxing precursors of macrophages require time to differentiate, polarize into M2-skewed TAMs, shut down their TIMP-1 expression, and only then, initiate relatively low-level production of TIMP-free MMP-9 zymogen.


Subject(s)
Angiogenesis Inducing Agents/metabolism , Matrix Metalloproteinase 9/metabolism , Neutrophils/pathology , Tumor Microenvironment/physiology , Animals , Cell Line, Tumor , Humans , Macrophages/enzymology , Macrophages/pathology , Male , Matrix Metalloproteinase 9/genetics , Mice, Inbred C57BL , Mice, Knockout , Neovascularization, Pathologic , Neutrophils/metabolism , Prostatic Neoplasms/blood supply , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/pathology , Tissue Inhibitor of Metalloproteinase-1/metabolism , Xenograft Model Antitumor Assays
9.
Cell Commun Signal ; 12: 61, 2014 Sep 25.
Article in English | MEDLINE | ID: mdl-25248616

ABSTRACT

BACKGROUND: INPP4B and PTEN dual specificity phosphatases are frequently lost during progression of prostate cancer to metastatic disease. We and others have previously shown that loss of INPP4B expression correlates with poor prognosis in multiple malignancies and with metastatic spread in prostate cancer. RESULTS: We demonstrate that de novo expression of INPP4B in highly invasive human prostate carcinoma PC-3 cells suppresses their invasion both in vitro and in vivo. Using global gene expression analysis, we found that INPP4B regulates a number of genes associated with cell adhesion, the extracellular matrix, and the cytoskeleton. Importantly, de novo expressed INPP4B suppressed the proinflammatory chemokine IL-8 and induced PAK6. These genes were regulated in a reciprocal manner following downregulation of INPP4B in the independently derived INPP4B-positive LNCaP prostate cancer cell line. Inhibition of PI3K/Akt pathway, which is highly active in both PC-3 and LNCaP cells, did not reproduce INPP4B mediated suppression of IL-8 mRNA expression in either cell type. In contrast, inhibition of PKC signaling phenocopied INPP4B-mediated inhibitory effect on IL-8 in either prostate cancer cell line. In PC-3 cells, INPP4B overexpression caused a decline in the level of metastases associated BIRC5 protein, phosphorylation of PKC, and expression of the common PKC and IL-8 downstream target, COX-2. Reciprocally, COX-2 expression was increased in LNCaP cells following depletion of endogenous INPP4B. CONCLUSION: Taken together, we discovered that INPP4B is a novel suppressor of oncogenic PKC signaling, further emphasizing the role of INPP4B in maintaining normal physiology of the prostate epithelium and suppressing metastatic potential of prostate tumors.


Subject(s)
Phosphoric Monoester Hydrolases/metabolism , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Protein Kinase C/metabolism , Animals , Cell Line, Tumor , Cell Movement , Cell Proliferation , Cyclooxygenase 2/metabolism , Gene Expression Regulation, Neoplastic , HEK293 Cells , Humans , Indoles/pharmacology , Inhibitor of Apoptosis Proteins/metabolism , Interleukin-8/genetics , Male , Maleimides/pharmacology , Mice, SCID , Neoplasm Invasiveness , Phosphoric Monoester Hydrolases/genetics , Protein Kinase C/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , RNA, Small Interfering/genetics , Survivin , p21-Activated Kinases/genetics
10.
Blood ; 122(25): 4054-67, 2013 Dec 12.
Article in English | MEDLINE | ID: mdl-24174628

ABSTRACT

A proangiogenic function of tissue-infiltrating monocytes/macrophages has long been attributed to their matrix metalloproteinase-9 zymogen (proMMP-9). Herein, we evaluated the capacity of human monocytes, mature M0 macrophages, and M1- and M2-polarized macrophages to induce proMMP-9-mediated angiogenesis. Only M2 macrophages induced angiogenesis at levels comparable with highly angiogenic neutrophils previously shown to release their proMMP-9 in a unique form, free of tissue inhibitor of metalloproteinases-1 (TIMP-1). Macrophage differentiation was accompanied by induction of low-angiogenic, TIMP-1-encumbered proMMP-9. However, polarization toward the M2, but not the M1 phenotype, caused a substantial downregulation of TIMP-1 expression, resulting in production of angiogenic, TIMP-deficient proMMP-9. Correspondingly, the angiogenic potency of M2 proMMP-9 was lost after its complexing with TIMP-1, whereas TIMP-1 silencing in M0/M1 macrophages rendered them both angiogenic. Similar to human cells, murine bone marrow-derived M2 macrophages also shut down their TIMP-1 expression and produced proMMP-9 unencumbered by TIMP-1. Providing proof that angiogenic capacity of murine M2 macrophages depended on their TIMP-free proMMP-9, Mmp9-null M2 macrophages were nonangiogenic, although their TIMP-1 was severely downregulated. Our study provides a unifying molecular mechanism for high angiogenic capacity of TIMP-free proMMP-9 that would be uniquely produced in a pathophysiological microenvironment by influxing neutrophils and/or M2 polarized macrophages.


Subject(s)
Cell Differentiation/physiology , Enzyme Precursors/metabolism , Macrophages/metabolism , Matrix Metalloproteinase 9/metabolism , Neovascularization, Physiologic/physiology , Tissue Inhibitor of Metalloproteinase-1/metabolism , Animals , Chick Embryo , Down-Regulation/physiology , Enzyme Precursors/genetics , Humans , Macrophages/cytology , Matrix Metalloproteinase 9/genetics , Mice , Mice, Mutant Strains , Neutrophils/cytology , Neutrophils/metabolism , Tissue Inhibitor of Metalloproteinase-1/genetics
11.
Cancer Res ; 73(14): 4196-211, 2013 Jul 15.
Article in English | MEDLINE | ID: mdl-23687338

ABSTRACT

Intravasation, the active entry of primary tumor cells into the vasculature, remains the least studied step in the metastatic cascade. Protease-mediated escape and stromal invasion of tumor cells represent widely accepted processes leading up to the intravasation step. However, molecular factors that contribute directly to tumor cell vascular penetration have not been identified. In this study, the in vivo role of the collagenolytic protease, MMP-1, in cancer cell intravasation and metastasis was analyzed by using a highly disseminating variant of human HEp3 epidermoid carcinoma, HEp3-hi/diss. Although naturally acquired or experimentally induced MMP-1 deficiency substantially suppressed HEp3-hi/diss intravasation, supplementation of recombinant MMP-1 to MMP-1-silenced primary tumors restored their impaired vascular dissemination. Surprisingly, abrogation of MMP-1 production and activity did not significantly affect HEp3-hi/diss migration or matrix invasion, suggesting noncollagenolytic mechanisms underlying MMP-1-dependent cell intravasation. In support of such noncollagenolytic mechanisms, MMP-1 silencing in HEp3-hi/diss cells modulated the microarchitecture and integrity of the angiogenic vasculature in a novel microtumor model. Concomitantly, MMP-1 deficiency led to decreased levels of intratumoral vascular permeability, tumor cell intravasation, and metastatic dissemination. Taking advantage of PAR1 deficiency of HEp3-hi/diss cells, we further show that endothelial PAR1 is a putative nontumor-cell/nonmatrix target, activation of which by carcinoma-produced MMP-1 regulates endothelial permeability and transendothelial migration. The inhibitory effects of specific PAR1 antagonists in live animals have also indicated that the mechanisms of MMP-1-dependent vascular permeability in tumors involve endothelial PAR1 activation. Together, our findings mechanistically underscore the contribution of a tumor MMP-1/endothelial PAR1 axis to actual intravasation events manifested by aggressive carcinoma cells.


Subject(s)
Carcinoma, Squamous Cell/enzymology , Matrix Metalloproteinase 1/metabolism , Receptor, PAR-1/metabolism , Animals , Capillary Permeability , Carcinoma, Squamous Cell/blood supply , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Cell Movement/physiology , Chick Embryo , Endothelium/blood supply , Endothelium/enzymology , Endothelium/metabolism , Endothelium/pathology , Humans , Matrix Metalloproteinase 1/deficiency , Matrix Metalloproteinase 1/genetics , Neoplasm Invasiveness , Neoplasm Metastasis , Neovascularization, Pathologic/enzymology , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Receptor, PAR-1/deficiency , Receptor, PAR-1/genetics
12.
J Biomed Biotechnol ; 2012: 564259, 2012.
Article in English | MEDLINE | ID: mdl-23097597

ABSTRACT

Plasmin, one of the most potent and reactive serine proteases, is involved in various physiological processes, including embryo development, thrombolysis, wound healing and cancer progression. The proteolytic activity of plasmin is tightly regulated through activation of its precursor, plasminogen, only at specific times and in defined locales as well as through inhibition of active plasmin by its abundant natural inhibitors. By exploiting the plasminogen activating system and overexpressing distinct components of the plasminogen activation cascade, such as pro-uPA, uPAR and plasminogen receptors, malignant cells can enhance the generation of plasmin which in turn, modifies the tumor microenvironment to sustain cancer progression. While plasmin-mediated degradation and modification of extracellular matrix proteins, release of growth factors and cytokines from the stroma as well as activation of several matrix metalloproteinase zymogens, all have been a focus of cancer research studies for decades, the ability of plasmin to cleave transmembrane molecules and thereby to generate functionally important cleaved products which induce outside-in signal transduction, has just begun to receive sufficient attention. Herein, we highlight this relatively understudied, but important function of the plasmin enzyme as it is generated de novo at the interface between cross-talking cancer and host cells.


Subject(s)
Cell Communication/physiology , Cell Membrane/metabolism , Fibrinolysin/metabolism , Models, Biological , Animals , Humans
13.
PLoS One ; 7(10): e46576, 2012.
Article in English | MEDLINE | ID: mdl-23056350

ABSTRACT

After neoplastic cells leave the primary tumor and circulate, they may extravasate from the vasculature and colonize tissues to form metastases. ß1 integrins play diverse roles in tumorigenesis and tumor progression, including extravasation. In blood cells, activation of ß1 integrins can be regulated by "inside-out" signals leading to extravasation from the circulation into tissues. However, a role for inside-out ß1 activation in tumor cell metastasis is uncertain. Here we show that ß1 integrin activation promotes tumor metastasis and that activated ß1 integrin may serve as a biomarker of metastatic human melanoma. To determine whether ß1 integrin activation can influence tumor cell metastasis, the ß1 integrin subunit in melanoma and breast cancer cell lines was stably knocked down with shRNA and replaced with wild-type or constitutively-active ß1. When tumor cells expressing constitutively-active ß1 integrins were injected intravenously into chick embryos or mice, they demonstrated increased colonization of the liver when compared to cells expressing wild-type ß1 integrins. Rescue expression with mutant ß1 integrins revealed that tumor cell extravasation and hepatic colonization required extracellular ligand binding to ß1 as well as ß1 interaction with talin, an intracellular mediator of integrin activation by the Rap1 GTPase. Furthermore, shRNA-mediated knock down of talin reduced hepatic colonization by tumor cells expressing wild-type ß1, but not constitutively-active ß1. Overexpression in tumor cells of the tumor suppressor, Rap1GAP, inhibited Rap1 and ß1 integrin activation as well as hepatic colonization. Using an antibody that detects activated ß1 integrin, we found higher levels of activated ß1 integrins in human metastatic melanomas compared to primary melanomas, suggesting that activated ß1 integrin may serve as a biomarker of invasive tumor cells. Altogether, these studies establish that inside-out activation of ß1 integrins promotes tumor cell extravasation and colonization, suggesting diagnostic and therapeutic approaches for targeting of ß1 integrin signaling in neoplasia.


Subject(s)
Integrin beta1/metabolism , Neoplasm Metastasis , Neoplasms/pathology , Cell Line, Tumor , Humans , Neoplasms/metabolism
14.
Mol Cancer Res ; 10(12): 1532-43, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23038812

ABSTRACT

Data accumulated over the latest two decades have established that the serine protease urokinase-type plasminogen activator (uPA) is a potential therapeutic target in cancer. When designing inhibitors of the proteolytic activity of serine proteases, obtaining sufficient specificity is problematic, because the topology of the proteases' active sites are highly similar. In an effort to generate highly specific uPA inhibitors with new inhibitory modalities, we isolated uPA-binding RNA aptamers by screening a library of 35 nucleotides long 2'-fluoro-pyrimidine RNA molecules using a version of human pro-uPA lacking the epidermal growth factor-like and kringle domains as bait. One pro-uPA-binding aptamer sequence, referred to as upanap-126, proved to be highly specific for human uPA. Upanap-126 delayed the proteolytic conversion of human pro-uPA to active uPA, but did not inhibit plasminogen activation catalyzed by two-chain uPA. The aptamer also inhibited the binding of pro-uPA to uPAR and the binding of vitronectin to the preformed pro-uPA/uPAR complex, both in cell-free systems and on cell surfaces. Furthermore, upanap-126 inhibited human tumor cell invasion in vitro in the Matrigel assay and in vivo in the chick embryo assay of cell escape from microtumors. Finally, upanap-126 significantly reduced the levels of tumor cell intravasation and dissemination in the chick embryo model of spontaneous metastasis. Together, our findings show that usage of upanap-126 represents a novel multifunctional mechanistic modality for inhibition of uPA-dependent processes involved in tumor cell spread.


Subject(s)
Aptamers, Nucleotide/genetics , Aptamers, Nucleotide/pharmacology , Prostatic Neoplasms/pathology , Prostatic Neoplasms/therapy , Urokinase-Type Plasminogen Activator/antagonists & inhibitors , Urokinase-Type Plasminogen Activator/genetics , Animals , Cell Line, Tumor , Cell-Free System , Chick Embryo , Epidermal Growth Factor/genetics , Epidermal Growth Factor/metabolism , HEK293 Cells , Humans , Male , Molecular Targeted Therapy , Neoplasm Invasiveness , Plasminogen/genetics , Plasminogen/metabolism , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Serine Proteases/genetics , Serine Proteases/metabolism , Urokinase-Type Plasminogen Activator/metabolism
16.
Neoplasia ; 13(9): 806-21, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21969814

ABSTRACT

Urokinase-type plasminogen activator (uPA) and plasmin have long been implicated in cancer progression. However, the precise contributions of the uPA/plasmin system to specific steps involved in cancer cell dissemination have not been fully established. Herein, we have used a highly disseminating variant of the human PC-3 prostate carcinoma cell line, PC-hi/diss, as a prototype of aggressive carcinomas to investigate the mechanisms whereby pro-uPA activation and uPA-generated plasmin functionally contribute to specific stages of metastasis. The PC-hi/diss cells secrete and activate significant amounts of pro-uPA, leading to efficient generation of plasmin in solution and at the cell surface. In a mouse orthotopic xenograft model, treatment with the specific pro-uPA activation-blocking antibody mAb-112 significantly inhibited local invasion and distant metastasis of the PC-hi/diss cells. To mechanistically examine the uPA/plasmin-mediated aspects of tumor cell dissemination, the anti-pro-uPA mAb-112 and the potent serine protease inhibitor, aprotinin, were used in parallel in a number of in vivo assays modeling various rate-limiting steps in early metastatic spread. Our findings demonstrate that, by generating plasmin, activated tumor-derived uPA facilitates early stages of PC-hi/diss dissemination, specifically the escape from the primary tumor and tumor cell intravasation. Moreover, through a series of in vitro and in vivo analyses, we suggest that PC-hi/diss-invasive escape and dissemination may be enhanced by cleavage of stromal fibronectin by uPA-generated plasmin. Together, our findings point to inhibition of pro-uPA activation at the apex of the uPA/plasmin cascade as a therapy-valid approach to control onset of tumor escape and ensuing metastatic spread.


Subject(s)
Fibrinolysin/metabolism , Neoplasm Metastasis , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Urokinase-Type Plasminogen Activator/metabolism , Animals , Antibodies, Monoclonal/immunology , Aprotinin/pharmacology , Cell Line, Tumor , Cell Movement , Chick Embryo , Enzyme Activation , Fibronectins/metabolism , Humans , Male , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Invasiveness , Serine Proteinase Inhibitors/pharmacology , Transplantation, Heterologous , Tumor Escape , Urokinase-Type Plasminogen Activator/immunology
17.
Am J Pathol ; 179(3): 1455-70, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21741942

ABSTRACT

Tumor-associated neutrophils contribute to neovascularization by supplying matrix metalloproteinase-9 (MMP-9), a protease that has been genetically and biochemically linked to induction of angiogenesis. Specific roles of inflammatory neutrophils and their distinct proMMP-9 in the coordinate regulation of tumor angiogenesis and tumor cell dissemination, however, have not been addressed. We demonstrate that the primary tumors formed by highly disseminating variants of human fibrosarcoma and prostate carcinoma recruit elevated levels of infiltrating MMP-9-positive neutrophils and concomitantly exhibit enhanced levels of angiogenesis and intravasation. Specific inhibition of neutrophil influx by interleukin 8 (IL-8) neutralization resulted in the coordinated diminishment of tumor angiogenesis and intravasation, both of which were rescued by purified neutrophil proMMP-9. However, if neutrophil proMMP-9, naturally devoid of tissue inhibitor of metalloproteinases (TIMP), was delivered in complex with TIMP-1 or in a mixture with TIMP-2, the protease failed to rescue the inhibitory effects of anti-IL8 therapy, indicating that the TIMP-free status of proMMP-9 is critical for facilitating tumor angiogenesis and intravasation. Our findings directly link tumor-associated neutrophils and their TIMP-free proMMP-9 with the ability of aggressive tumor cells to induce the formation of new blood vessels that serve as conduits for tumor cell dissemination. Thus, treatment of cancers associated with neutrophil infiltration may benefit from specific targeting of neutrophil MMP-9 at early stages to prevent ensuing tumor angiogenesis and tumor metastasis.


Subject(s)
Enzyme Precursors/metabolism , Matrix Metalloproteinase 9/metabolism , Neovascularization, Pathologic/enzymology , Neutrophils/metabolism , Tissue Inhibitor of Metalloproteinases/physiology , Animals , Cell Line, Tumor , Chick Embryo , Fibrosarcoma/blood supply , Fibrosarcoma/metabolism , Fibrosarcoma/secondary , Humans , Interleukin-8/pharmacology , Male , Mice , Mice, Nude , Mice, SCID , Neoplasm Invasiveness , Prostatic Neoplasms/blood supply , Prostatic Neoplasms/metabolism
18.
Biochim Biophys Acta ; 1803(1): 103-20, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19800930

ABSTRACT

A number of extensive reviews are available discussing the roles of MMPs in various aspects of cancer progression from benign tumor formation to overt cancer present with deadly metastases. This review will focus specifically on the evidence functionally linking the MMPs and tumor-induced angiogenesis in various in vivo models. Emphasis has been placed on the cellular origin of the MMPs in tumor tissue, the requirement of proMMP activation and the resulting proteolytic activity for the induction and progression of tumor angiogenesis, and the pleiotropic roles for some of the MMPs. The functional mechanisms of the angiogenic MMPs are discussed as well as their catalytic detection in complex biological systems. In addition, the contribution of active MMPs to metastatic spread and establishment of secondary metastasis will be discussed in view of the findings indicating that MMPs are involved in the preparation of pre-metastatic niches. Finally, the most recent evidence, indicating the pro-metastatic consequences of anti-angiogenic therapies employing MMP inhibitors will be presented as examples highlighting possible outcomes of interfering with the pleiotropic nature of the MMP functionality.


Subject(s)
Matrix Metalloproteinases/metabolism , Neoplasms/blood supply , Neoplasms/enzymology , Neovascularization, Pathologic/enzymology , Animals , Enzyme Activation , Humans , Neoplasms/pathology , Neovascularization, Pathologic/therapy , Protein Processing, Post-Translational , Stem Cells/cytology
19.
Clin Exp Metastasis ; 26(8): 1033-47, 2009.
Article in English | MEDLINE | ID: mdl-19842048

ABSTRACT

Increased metastatic and angiogenic potentials of aggressive human colon carcinoma cells were verified in independent chick embryo models by comparing in vivo highly metastatic SW620 colon carcinoma cell line with its isogenic, non-metastatic SW480 cell variant. In the experimental metastasis model, both cell types rapidly arrested in the chorioallantoic membrane (CAM) vasculature as demonstrated by quantitative PCR and immunohistochemistry. Live cell imaging also indicated that both SW620 and SW480 cells efficiently extravasated from the CAM capillary system. However, only few SW480 cells were present in the CAM tissue after 24-48 h. In contrast, the numbers of SW620 cells increased exponentially, indicating proliferative and survival advantages of metastatic colon carcinoma cells in vivo. Multicellular SW620 foci were identified in close proximity to CAM blood vessels. A positive correlation between increased metastatic ability and VEGF-expression of colon carcinoma SW620 cells was demonstrated by the substantial inhibitory effects of anti-VEGF treatment on the levels of metastatic colonization and density of blood vessels adjacent to tumor cell foci. Furthermore, the chick embryo angiogenesis model confirmed high levels of VEGF-dependent angiogenesis induced by SW620 cells, but not SW480 cells. Thus, chick embryo experimental metastasis and CAM angiogenesis models appear to coordinately reflect critical features of advanced colon carcinomas, i.e., the acquisition of enhanced survival and increased angiogenic potentials, both constituting critical determinants of colon cancer progression. The use of rapid and quantitative chick embryo models might provide alternative approaches to conventional mammalian model systems for screening anti-cancer agents.


Subject(s)
Colonic Neoplasms/blood supply , Colonic Neoplasms/pathology , Disease Models, Animal , Neoplasm Metastasis , Neovascularization, Pathologic , Animals , Cell Line, Tumor , Chick Embryo , Chorioallantoic Membrane/blood supply , Humans , Vascular Endothelial Growth Factor A/metabolism
20.
Am J Pathol ; 175(4): 1638-52, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19729488

ABSTRACT

To analyze the process of tumor cell intravasation, we used the human tumor-chick embryo spontaneous metastasis model to select in vivo high (PC-hi/diss) and low (PC-lo/diss) disseminating variants from the human PC-3 prostate carcinoma cell line. These variants dramatically differed in their intravasation and dissemination capacities in both chick embryo and mouse spontaneous metastasis models. Concomitant with enhanced intravasation, PC-hi/diss exhibited increased angiogenic potential in avian and murine models. PC-hi/diss angiogenesis and intravasation were dependent on increased secretion of vascular endothelial growth factor (VEGF), since treating developing tumors with a function-blocking anti-VEGF antibody simultaneously inhibited both processes without affecting primary tumor growth. PC-hi/diss cells were also more migratory and invasive, suggestive of heightened ability to escape from primary tumors due to matrix-degrading activity. Consistent with this suggestion, PC-hi/diss cells produced more of the serine protease urokinase-type plasminogen activator (uPA) as compared with PC-lo/diss. The functional role of uPA in PC-hi/diss dissemination was confirmed by inhibition of invasion, angiogenesis, and intravasation with specific function-blocking antibodies that prevented uPA activation and blocked uPA activity. These processes were similarly sensitive to aprotinin, a potent inhibitor of serine proteases, including uPA-generated plasmin. Thus, our comparison of the PC-3 intravasation variants points to key roles for the uPA-plasmin system in PC-hi/diss intravasation, possibly via (1) promoting tumor cell matrix invasion and (2) facilitating development of VEGF-dependent angiogenic blood vessels.


Subject(s)
Neovascularization, Pathologic/pathology , Prostatic Neoplasms/blood supply , Prostatic Neoplasms/pathology , Urokinase-Type Plasminogen Activator/metabolism , Vascular Endothelial Growth Factor A/metabolism , Animals , Cell Adhesion , Cell Line, Tumor , Cell Movement , Chick Embryo , Disease Models, Animal , Humans , Male , Mice , Neoplasm Invasiveness , Neoplasm Metastasis , Vascular Endothelial Growth Factor A/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL
...