Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
J Natl Cancer Inst ; 106(4): dju012, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24610907

ABSTRACT

BACKGROUND: Asparaginyl endopeptidase (AEP) has been implicated in human cancer development. However, the molecular mechanisms underlying AEP regulation, including the role of pro-AEP activation, remain elusive. METHODS: We investigated the regulation of AEP by TRAF6 and its effects on tumor progression and metastasis in cancer cell lines, murine models, and specimens from patients using biochemical analyses, confocal microscopy, immunoelectron microscopy, and migration-invasion assays. The sera of healthy donors and breast cancer patients were examined by enzyme-linked immunosorbent assay, and a tissue array of 314 breast cancer specimens was assessed for AEP and TRAF6 by immunohistochemistry. Furthermore, the effects of AEP inhibitors or monoclonal antibodies on pulmonary metastasis were evaluated in murine models. The statistical significance between groups was determined using two-tailed Student t tests. RESULTS: We demonstrate that TRAF6 ubiquitinates the proform of AEP through K63-linked polyubiquitin, reversible by USP17, and forms a complex with HSP90α to subsequently promote pro-AEP intracellular stability as well as secretion. Disrupting the interaction between pro-AEP and TRAF6 or inhibiting HSP90α reduced pro-AEP secretion and consequently reduced tumor metastasis. Higher circulating AEP levels were detected in the sera of breast cancer patients, and AEP inhibitors or neutralizing antibodies remarkably decreased tumor metastasis in murine models. Notably, TRAF6 and AEP were overexpressed in human breast neoplasms and correlated with poor prognosis. Patients with low AEP/TRAF6 expression survived for a mean of 111 months (95% confidence interval [CI] = 108 to 115 months), whereas those with high AEP/TRAF6 expression survived for a mean of only 61 months (95% CI = 42 to 79 months; P < .001). CONCLUSIONS: Our study elucidates a novel mechanism of AEP regulation and an alternative oncogenic pathway for TRAF6 in breast cancer, which suggests that AEP and TRAF6 protein levels may have prognostic implications in breast cancer patients. Thus, AEP may serve as a biomarker as well as new therapeutic target.


Subject(s)
Biomarkers, Tumor/metabolism , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cysteine Endopeptidases/metabolism , TNF Receptor-Associated Factor 6/metabolism , Ubiquitination , Animals , Cell Line, Tumor , Disease Progression , Enzyme-Linked Immunosorbent Assay , Female , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Humans , Immunoblotting , Immunohistochemistry , Immunoprecipitation , Mice , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Predictive Value of Tests , Prognosis
2.
Thromb Res ; 133(4): 657-66, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24485401

ABSTRACT

OBJECTIVES: The coagulation protease cascade plays the central requisite role in initiation of arterial atherothrombosis. However, the relative participation of the extrinsic as compared to the intrinsic pathway is incompletely resolved. We have investigated in vivo the relative importance of the extrinsic and intrinsic pathways to define which is more essential to atherothrombosis and therefore the preferable prophylactic therapeutic target. We further addressed which type of plaque associated macrophage population is associated with the thrombotic propensity of atherosclerotic plaques. METHODS: Both photochemical injury and ferric chloride vascular injury models demonstrated arterial thrombosis formation in ApoE deficient mice. We found that direct interference with the extrinsic pathway, but not the intrinsic pathway, markedly diminished the rate of thrombus formation and occlusion of atherosclerotic carotid arteries following experimental challenge. To explore which plaque macrophage subtype may participate in plaque thrombosis in regard to expression tissue factor pathway inhibitor (TFPI), bone marrow derived macrophages of both M and GM phenotypes expressed tissue factor (TF), but the level of TFPI was much greater in M- type macrophages, which exhibited diminished thrombogenic activity, compared to type GM-macrophages. RESULTS AND CONCLUSIONS: Our works support the hypothesis that the TF-initiated and direct extrinsic pathway provides the more significant contribution to arterial plaque thrombogenesis. Activation of the TF driven extrinsic pathway can be influenced by differing colony-stimulating factor influenced macrophage TFPI-1 expression. These results advance our understanding of atherothrombosis and identify potential therapeutic targets associated with the extrinsic pathway and with macrophages populating arterial atherosclerotic plaques.


Subject(s)
Macrophages/metabolism , Thromboplastin/metabolism , Thrombosis/blood , Thrombosis/therapy , Animals , Apolipoproteins E/metabolism , Blood Coagulation/physiology , Female , Humans , Mice , Mice, Inbred C57BL , Thrombosis/prevention & control
3.
FEBS Lett ; 585(4): 671-6, 2011 Feb 18.
Article in English | MEDLINE | ID: mdl-21266177

ABSTRACT

RNA processing is vital for the high fidelity and diversity of eukaryotic transcriptomes and the encoded proteomes. However, control of RNA processing is not fully established. Σ RNA is a class of conserved large non-coding RNAs (murine Hepcarcin; human MALAT-1) up-regulated in carcinomas. Using antisense technology, we identified that RNA post-transcriptional modification is the most significant global function of Σ RNA. Specifically, processing of the pre-mRNAs of genes including Tissue Factor and Endoglin was altered by hydrolysis of Σ RNA/MALAT-1. These results support the hypothesis that Σ RNA/MALAT-1 is a regulatory molecule exerting roles in RNA post-transcriptional modification.


Subject(s)
Carcinoma/metabolism , RNA Processing, Post-Transcriptional , RNA, Neoplasm/metabolism , RNA, Untranslated/metabolism , Alternative Splicing , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Cells, Cultured , Endoglin , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , HeLa Cells , Humans , Nuclear Proteins/metabolism , Oligonucleotide Array Sequence Analysis , Oligonucleotides, Antisense , RNA Precursors/metabolism , RNA, Long Noncoding , RNA, Untranslated/antagonists & inhibitors , RNA, Untranslated/genetics , RNA-Binding Proteins/metabolism , Rats , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Serine-Arginine Splicing Factors , Thromboplastin/genetics , Thromboplastin/metabolism
4.
Proc Natl Acad Sci U S A ; 107(24): 10920-5, 2010 Jun 15.
Article in English | MEDLINE | ID: mdl-20534451

ABSTRACT

Regulation of the actin-myosin cytoskeleton plays a central role in cell migration and cancer progression. Here, we report the discovery of a cytoskeleton-associated kinase, pseudopodium-enriched atypical kinase 1 (PEAK1). PEAK1 is a 190-kDa nonreceptor tyrosine kinase that localizes to actin filaments and focal adhesions. PEAK1 undergoes Src-induced tyrosine phosphorylation, regulates the p130Cas-Crk-paxillin and Erk signaling pathways, and operates downstream of integrin and epidermal growth factor receptors (EGFR) to control cell spreading, migration, and proliferation. Perturbation of PEAK1 levels in cancer cells alters anchorage-independent growth and tumor progression in mice. Notably, primary and metastatic samples from colon cancer patients display amplified PEAK1 levels in 81% of the cases. Our findings indicate that PEAK1 is an important cytoskeletal regulatory kinase and possible target for anticancer therapy.


Subject(s)
Cytoskeleton/metabolism , Neoplasms/metabolism , Protein-Tyrosine Kinases/metabolism , Pseudopodia/metabolism , Actins/metabolism , Animals , Base Sequence , Cell Line , Cell Line, Tumor , Cell Movement , Cell Proliferation , Colonic Neoplasms/metabolism , Computational Biology , DNA Primers/genetics , Female , Focal Adhesions/metabolism , Humans , In Vitro Techniques , Liver Neoplasms/metabolism , Liver Neoplasms/secondary , Mice , Mice, Nude , Neoplasms/etiology , Pancreatic Neoplasms/metabolism , Phosphotyrosine/metabolism , Protein-Tyrosine Kinases/genetics , Proteomics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Signal Transduction , src-Family Kinases/metabolism
5.
Coron Artery Dis ; 21(2): 97-103, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20087175

ABSTRACT

OBJECTIVES: To evaluate tissue factor (TF) expression in vein grafts interposed in the arterial circulation of hypercholesterolemic rabbits. Veins implanted in the arterial circulation of normocholesterolemic rabbits respond by inflammation and infiltration by monocytes with transient TF expression. In a hypercholesterolemic milieu these monocytes may differentiate into macrophages capable of enhanced TF synthesis, which may facilitate hyperplasia and thrombosis. METHODS: Autologous jugular veins interposed in the carotid artery of hypercholesterolemic rabbits were harvested at 1, 2, 4, 6, and 8 weeks after surgery and examined for presence and localization of rabbit TF antigen. Protein extracted from vein segments was evaluated for procoagulant activity by bioassay and for TF protein content by western blotting. RESULTS: Rabbit TF antigen was observed mostly in the subendothelium of vein grafts. Peak TF procoagulant activity observed at 1-2 weeks postsurgery (2.3+/-1.8 pg/mg, P<0.006) declined to 0.9+/-0.5, 0.2+/-0.1, and 0.15+/-0.06 pg/mg at 4, 6, and 8 weeks, respectively (P<0.03). Western blotting showed a time-dependent pattern for rabbit TF protein with prolonged expression peaking at 6 weeks. CONCLUSION: Prolonged expression of biologically active rabbit TF and TF protein were shown within jugular vein grafts of hypercholesterolemic rabbits. This response, reported for the first time and attributed to increased cholesterol levels, may possibly contribute to enhanced hyperplasia. These results suggest that TF expression could serve as another mechanism underlying vein graft failure and that hypercholesterolemia in bypass patients should be treated aggressively beginning within the weeks after surgery.


Subject(s)
Carotid Artery, Common/surgery , Graft Rejection/metabolism , Hypercholesterolemia/metabolism , Jugular Veins/metabolism , Jugular Veins/transplantation , Thromboplastin/metabolism , Animals , Blood Coagulation , Blood Coagulation Tests , Blotting, Western , Cholesterol/metabolism , Disease Models, Animal , Graft Rejection/blood , Graft Rejection/etiology , Hypercholesterolemia/blood , Hypercholesterolemia/complications , Immunohistochemistry , Rabbits , Time Factors , Transplantation, Autologous
6.
Circ Res ; 102(4): 457-64, 2008 Feb 29.
Article in English | MEDLINE | ID: mdl-18174463

ABSTRACT

The serine protease zymogen factor X is converted to its catalytically active form factor Xa by the binary complex of factor VIIa bound to its cell surface receptor tissue factor (TF) or by the intrinsic Xase complex, which consists of active factors VIII (VIIIa), IX (IXa), factor X, and Ca2+. Factor Xa has procoagulant activity by conversion of prothrombin to thrombin and also induces signal transduction, either alone or in the ternary TF:VIIa:factor Xa coagulation initiation complex. Factor Xa cleaves and activates protease activated receptor (PAR)1 or -2, but factor Xa signaling efficiency varies among cell types. We show here that annexin 2 acts as a receptor for factor Xa on the surface of human umbilical vein endothelial cells and that annexin 2 facilitates factor Xa activation of PAR-1 but does not enhance coagulant function of factor Xa. Overexpression of TF abolishes annexin 2 dependence on factor Xa signaling and diminishes binding to cell surface annexin 2, whereas selectively abolishing TF promotes the annexin 2/factor Xa interaction. We propose that annexin 2 serves to regulate factor Xa signaling specifically in the absence of cell surface TF and may thus play physiological or pathological roles when factor Xa is generated in a TF-depleted environment.


Subject(s)
Annexin A2/metabolism , Endothelial Cells/metabolism , Factor Xa/metabolism , Receptor, PAR-1/metabolism , Signal Transduction/physiology , Animals , Blood Coagulation/physiology , Cells, Cultured , Endothelial Cells/cytology , Factor Xa/chemistry , Humans , Immunoprecipitation , Lysine/metabolism , Mice , Mice, Mutant Strains , Protein Structure, Tertiary , Thromboplastin/metabolism , Umbilical Veins/cytology
8.
Cancer Res ; 66(2): 970-80, 2006 Jan 15.
Article in English | MEDLINE | ID: mdl-16424032

ABSTRACT

The tumor microenvironment is notably enriched with a broad spectrum of proteases. The proteolytic specificities of peptide substrates provide modular chemical tools for the rational design of cell-impermeable prodrugs that are specifically activated by proteases extracellularly in the tumor microenvironment. Targeting cell-impermeable prodrug activation to tumor microenvironment will significantly reduce drug toxicity to normal tissues. The activated prodrug attacks both tumor and stroma cells through a "bystander effect" without selectively deleting target-producing cells, therefore further minimizing resistance and toxicity. Here, we showed that legumain, the only asparaginyl endopeptidase of the mammalian genome, is highly expressed by neoplastic, stromal, and endothelial cells in solid tumors. Legumain is present extracellularly in the tumor microenvironment, associated with matrix as well as cell surfaces and functional locally in the reduced pH of the tumor microenvironment. A novel legumain-activated, cell-impermeable doxorubicin prodrug LEG-3 was designed to be activated exclusively in the tumor microenvironment. Upon administration, there is a profound increase of the end-product doxorubicin in nuclei of cells in tumors but little in other tissues. This tumor microenvironment-activated prodrug completely arrested growth of a variety of neoplasms, including multidrug-resistant tumor in vivo and significantly extended survival without evidence of myelosuppression or cardiac toxicity. The tumor microenvironment-activated prodrug design can be extended to other proteases and chemotherapeutic compounds and provides new potentials for the rational development of more effective functionally targeted cancer therapeutics.


Subject(s)
Antibiotics, Antineoplastic/pharmacokinetics , Cysteine Endopeptidases/pharmacology , Doxorubicin/analogs & derivatives , Doxorubicin/pharmacokinetics , Drug Resistance, Multiple , Oligopeptides/pharmacology , Prodrugs/pharmacology , Animals , Cell Nucleus , Cell Proliferation , Colonic Neoplasms/pathology , Doxorubicin/pharmacology , Extracellular Matrix , Fibrosarcoma/pathology , Humans , Mice , Neoplasms , Neuroblastoma/pathology , Permeability , Tumor Cells, Cultured
9.
Cancer Res ; 65(23): 11109-17, 2005 Dec 01.
Article in English | MEDLINE | ID: mdl-16322261

ABSTRACT

We have previously shown that part of the heparin-binding domain of the vascular endothelial growth factor (VEGF), designated HBDt, localizes very selectively to surfaces of the endothelial cells of i.t blood vessels. Here, we have coupled the HBDt to the extracellular domain of tissue factor (TFt), to locally initiate the thrombogenic cascade. In tumor-bearing mice, infusion of this HBDt.TFt results in rapid occlusive thrombosis selective only for tumor microvasculature with resultant infarctive destruction of tumors. We now show that infusion of an optimal combination of this HBDt.TFt and its requisite cofactor (factor VIIa) in tumor models results in significant tumor eradication. Binding studies and confocal microscopy indicate that the target for the HBDt.TFt seems to be a trimolecular complex of chondroitin C sulfate proteoglycan, neuropilin-1, and VEGF receptor-2, overexpressed together only in highly angiogenic sites of the tumor microenvironment. The HBDt.TFt was also colocalized with the trimolecular receptor complex in endothelial sprouts from tumor tissues, and its binding inhibited the growth of such sprouts. In vitro, we show that the HBDt structure has its highest affinity for chondroitin 6 sulfate. We show the potential of this HBDt.TFt as a candidate therapeutic and elucidate its target in vivo.


Subject(s)
Neoplasms, Experimental/blood supply , Neoplasms, Experimental/drug therapy , Recombinant Fusion Proteins/pharmacology , Thrombin/pharmacology , Thromboplastin/pharmacology , Animals , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Heparin/metabolism , Male , Mice , Mice, Inbred BALB C , Necrosis , Neoplasms, Experimental/pathology , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/metabolism , Peptide Fragments/genetics , Peptide Fragments/pharmacokinetics , Peptide Fragments/pharmacology , Protein Structure, Tertiary , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/pharmacokinetics , Spheroids, Cellular , Thrombin/genetics , Thrombin/pharmacokinetics , Thromboplastin/genetics , Thromboplastin/pharmacokinetics , Thrombosis/chemically induced
10.
Arterioscler Thromb Vasc Biol ; 25(8): 1737-43, 2005 Aug.
Article in English | MEDLINE | ID: mdl-15947236

ABSTRACT

OBJECTIVE: To test the hypothesis that Grp78 negatively regulates cell surface tissue factor (TF) procoagulant activity and whether this is mediated by physical interaction. METHODS AND RESULTS: Biopanning with phage-displayed peptidyl libraries has identified peptide probes that bind selectively in vivo to the surface of atherosclerotic plaque endothelium. The highest affinity peptide, EKO130, binds 78-kDa glucose regulated protein (Grp78). Grp78 participates in numerous pathological processes, including the regulation of the coagulation cascade, but the mechanism of Grp78 regulation of coagulation is unknown. To characterize this function, we analyzed the effect of Grp78 on TF-mediated procoagulant activity on murine brain endothelial cells (bEND.3) and macrophage-like (RAW) cells, which are relevant in mediation of atherothrombosis. We show that Grp78 is present on the surface of endothelium and monocyte/macrophage-like cells in atherosclerotic lesions. Inhibition of Grp78 resulted in increased procoagulant activity. We demonstrate that Grp78 negatively regulates procoagulant activity by interacting physically with the TF extracellular domain on the cell surface. CONCLUSIONS: The evidence indicates that Grp78 negatively regulates TF functional activity via direct binding to and functional inhibition of TF. Identification of the mechanism by which Grp78 regulates TF function may advance insight into the pathobiology of atherosclerosis and associated arterial thrombosis.


Subject(s)
Atherosclerosis/immunology , Atherosclerosis/metabolism , Blood Coagulation/physiology , Heat-Shock Proteins/metabolism , Molecular Chaperones/metabolism , Thromboplastin/metabolism , Animals , Atherosclerosis/pathology , Cells, Cultured , Cerebrovascular Circulation , Endoplasmic Reticulum Chaperone BiP , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Factor Xa/metabolism , Foam Cells/cytology , Foam Cells/metabolism , Heat-Shock Proteins/antagonists & inhibitors , Macrophages/cytology , Macrophages/metabolism , Membrane Proteins/metabolism , Mice , Molecular Chaperones/antagonists & inhibitors , Thrombosis/immunology , Thrombosis/metabolism , Thrombosis/pathology
11.
Biochemistry ; 42(36): 10619-26, 2003 Sep 16.
Article in English | MEDLINE | ID: mdl-12962485

ABSTRACT

Tissue factor (TF), the receptor and cofactor for factor VIIa (VIIa) for cellular initiation of the coagulation protease cascade, drives thrombogenesis, inflammation, tumor cell metastasis, and the lethality of severe sepsis. To identify TF surface loci that can selectively inhibit substrate zymogen association and activation, TF(1-218), the extracellular domain, was used as the target for the phage display search. This resulted in selection of 59 clones from a phage gpVIII surface protein-expressed library of constrained combinatorial peptides. Of these, one encoding the peptide Glu-Cys-Leu-Arg-Ser-Val-Val-Thr-Cys on gpVIII most avidly bound TF(1-218), as did the synthetic peptide. Inhibition of binding was selective with an IC(50) of 30 nM for proteolytic activation of factor X by the TF(1-218)-VIIa complex. In contrast, there was no inhibition of factor IX activation. The selective inhibition of only factor X association with TF(1-218) will spare the intrinsic hemostatic pathway while attenuating the extrinsic thrombogenic pathway. This and related peptidyl structures provide the potential for the more precise identification of TF surface loci that mediate selective functional properties of the protein as well as a structural basis for the design of novel molecules for selectively attenuating initiation of the extrinsic limb of the coagulation protease cascade and other functions of TF.


Subject(s)
Blood Coagulation/physiology , Endopeptidases/metabolism , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Thromboplastin/chemistry , Thromboplastin/metabolism , Amino Acid Sequence , Bacteriophage M13/genetics , Bacteriophage M13/metabolism , Binding Sites , Binding, Competitive , Enzyme Precursors/antagonists & inhibitors , Enzyme Precursors/metabolism , Factor IX/metabolism , Factor VIIa/chemistry , Factor VIIa/metabolism , Factor X/metabolism , Humans , Models, Molecular , Peptide Fragments/genetics , Peptide Library , Protein Binding , Protein Structure, Tertiary/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Thermodynamics , Thromboplastin/genetics
12.
Cancer Res ; 62(23): 7118-23, 2002 Dec 01.
Article in English | MEDLINE | ID: mdl-12460934

ABSTRACT

Vascular endothelial growth factor has an exon 7-encoded heparin-binding domain. To explore the expression of complementary ligands on endothelial surfaces in vivo and to assess potential for localization within the vascular tree, we introduced a truncated version of this domain (HBDt) into a modified M13 phage. Despite the small size and trace-level expression, this HBDt endowed the phage with affinity for heparin to which it bound in vitro. It also preferentially and selectively localized the phage in vivo to vascular endothelial surfaces, especially of tumors. Competition assays demonstrated that accumulation and localization of this phage was attributable to expression of the HBDt on the phage surface and sequence comparison suggests its novelty. We propose to use this novel HBDt structure to explore the expression of the ligand glycosaminoglycans within the vascular tree. This structure may facilitate directed delivery of therapeutic molecules.


Subject(s)
Endothelial Growth Factors/metabolism , Endothelium, Vascular/metabolism , Glycosaminoglycans/metabolism , Heparin/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Lymphokines/metabolism , Neoplasms, Experimental/blood supply , Adenocarcinoma/blood supply , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Amino Acid Sequence , Animals , Bacteriophage M13/genetics , Bacteriophage M13/metabolism , Base Sequence , CHO Cells , Colonic Neoplasms/blood supply , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Cricetinae , Endothelial Growth Factors/genetics , Intercellular Signaling Peptides and Proteins/genetics , Lymphokines/genetics , Mammary Neoplasms, Experimental/blood supply , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/metabolism , Mice , Mice, Inbred BALB C , Microscopy, Confocal , Models, Molecular , Molecular Sequence Data , Neoplasms, Experimental/genetics , Neoplasms, Experimental/metabolism , Protein Structure, Tertiary , Rats , Rats, Inbred F344 , Tissue Distribution , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
13.
Cancer Res ; 62(19): 5470-5, 2002 Oct 01.
Article in English | MEDLINE | ID: mdl-12359755

ABSTRACT

Prostate-specific membrane antigen (PSMA), a glutamyl preferring carboxypeptidase, is found in prostate and other carcinomas present on both tumor cells and associated microvascular lining cells. We find that the channel structures delineated by PSMA-expressing cells in human and rat prostate tumors are in functional continuity with the vasculature and thus form part of tumor microvasculature. The PSMA-positive cell-outlined channels are CD31 negative and mutually exclusive of CD31-positive cell-lined channels elsewhere in the tumor consistent with tumor cells adapted to a pseudoendothelial phenotype in vasculogenic mimicry. To assess the functional potential of such PSMA-lined microvasculature to selectively direct infarctive tumor therapy, we coupled the soluble extracellular domain of tissue factor to a PSMA catalytic site inhibitor to create a PSMA-directed selective tumor vascular thrombogen (STVT). This protein induced selective local in vivo infarctive necrosis of the rat Mat Lu prostate tumor when administered i.v. The combined administration of this STVT with low-dose doxorubicin produced a profound tumoricidal effect, resulting in complete eradication of some tumors. This is consistent with the therapeutic potential for a PSMA-directed STVT and expands the potential for selective infarctive ablation of tumors.


Subject(s)
Antigens, Surface , Carboxypeptidases/antagonists & inhibitors , Dipeptides/pharmacology , Prostatic Neoplasms/blood supply , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Binding Sites , Carboxypeptidases/biosynthesis , Catalysis , Dipeptides/administration & dosage , Doxorubicin/administration & dosage , Doxorubicin/pharmacology , Glutamate Carboxypeptidase II , Humans , Infarction/chemically induced , Liposomes , Male , Mice , Mice, Nude , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/enzymology , Rats , Thromboplastin/pharmacology , Thrombosis/chemically induced , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
14.
Atherosclerosis ; 161(1): 35-43, 2002 Mar.
Article in English | MEDLINE | ID: mdl-11882315

ABSTRACT

Circulating monocytes as well as resident monocyte-derived macrophages (MDMs) in the atherosclerotic plaque express tissue factor. We tested the hypothesis that differentiation of monocytes into macrophages increases their tissue factor expression. This was done by isolating monocytes from human blood and culturing them for 24 h or allowing their differentiation into macrophages for 8 days before assay. Tissue factor procoagulant activity was assessed by recalcification clotting assay while Western blotting quantitated tissue factor protein. Immunocytochemistry was applied to localize antigenic tissue factor in cells. The results showed that freshly-isolated monocytes expressed low baseline procoagulant activity (0.0004+/-0.0003 ng rhTF/10(6) cells) which increased 100-fold in 24-h cultured monocytes (0.04+/-0.01 ng/10(6) cells, P<0.0001) and was associated with detectable tissue factor protein by Western blotting. Furthermore, MDMs expressed 25-fold higher procoagulant activity (1.0+/-0.5 ng/10(6) cells, P<0.04) and increased tissue factor protein content compared with 24-h monocytes as corroborated by immunocytochemistry. LPS stimulation increased baseline binding activity of transcription factor kappaB three-fold (P<0.03) and 2.6-fold (P<0.09) in monocytes and in macrophages, respectively. The results demonstrate that differentiation of human monocytes into macrophages in culture enhances their tissue factor expression. The observed tissue factor increase that can be further stimulated by LPS may contribute to thrombogenicity of macrophage-rich atheroma. Activation of the TF-kappaB transduction pathway suggests its potential role in the transcriptional regulation of TF gene in response to acute stimulation in these cells. This cell-culture model may provide the opportunity to evaluate the effect of various interventions on macrophage procoagulant activity.


Subject(s)
Blood Coagulation Factors/metabolism , Macrophages/cytology , Monocytes/cytology , Thromboplastin/biosynthesis , Blotting, Western , Cell Adhesion , Cell Differentiation , Cells, Cultured , Humans , Immunohistochemistry , Lipopolysaccharides/pharmacology , Microscopy, Electron , NF-kappa B/metabolism , Thromboplastin/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...