Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
2.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-851423

ABSTRACT

Objective System evolution relationship and molecular identification method of the germplasm resources of Lycoris aurea from different regions was analyzed based on the sequence of psbA-trnH chloroplast gene. Methods DNA samples of 52 L. aurea populations were extracted from 15 provinces or cities in China. The psbA-trnH sequences of the populations were amplified by PCR, and the purified PCR products were sequenced and analyzed by Mega 5.0 software etc. Results The length of psbA-trnH sequences were 544-656 bp, and GC content of them was 35.8%-37.0%, and the genetic distances among the populations were 0-0.009 47. There were 33 variable (polymorphic) sites, including nine parsimony informative sites and 18 singleton variable sites and six insertion/deletion gaps. Ten haplotypes (H) were identified. Values of haplotype diversity (Hd) and nucleotide diversity (π) were 0.749 and 0.002 63, respectively. The genetic diversity of the populations of L. aurea were very high. In the maximum parsimony phylogenetic tree, 52 populations of L. aurea were clustered into four branches, which was almost consistent with their geographical distributions. Conclusion The genetic variation of L. aurea populations from different regions is significant and the psbA-trnH sequence could be used as a molecular evidence for identifying the germplasm resources of L. aurea from different regions. There is very obvious regional characteristics in evolution for germplasm resources of L. aurea in China.

3.
J Biol Chem ; 282(18): 13326-33, 2007 May 04.
Article in English | MEDLINE | ID: mdl-17347152

ABSTRACT

When shed from the cell surface, the heparan sulfate proteoglycan syndecan-1 can facilitate the growth, angiogenesis, and metastasis of tumors. Here we report that tumor cell expression of heparanase, an enzyme known to be a potent promoter of tumor progression and metastasis, regulates both the level and location of syndecan-1 within the tumor microenvironment by enhancing its synthesis and subsequent shedding from the tumor cell surface. Heparanase regulation of syndecan-1 is detected in both human myeloma and breast cancer cell lines. This regulation requires the presence of active enzyme, because mutated forms of heparanase lacking heparan sulfate-degrading activity failed to influence syndecan-1 expression or shedding. Removal of heparan sulfate from the cell surface using bacterial heparitinase dramatically accelerated syndecan-1 shedding, suggesting that the effects of heparanase on syndecan-1 expression by tumor cells may be due, at least in part, to enzymatic removal or reduction in the size of heparan sulfate chains. Animals bearing tumors formed from cells expressing high levels of heparanase or animals transgenic for heparanase expression exhibited elevated levels of serum syndecan-1 as compared with controls, indicating that heparanase regulation of syndecan-1 expression and shedding can occur in vivo and impact cancer progression and perhaps other pathological states. These results reveal a new mechanism by which heparanase promotes an aggressive tumor phenotype and suggests that heparanase and syndecan-1 act synergistically to fine tune the tumor microenvironment and ensure robust tumor growth.


Subject(s)
Breast Neoplasms/enzymology , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Glucuronidase/biosynthesis , Multiple Myeloma/enzymology , Neovascularization, Pathologic/enzymology , Syndecan-1/metabolism , Animals , Bacterial Proteins/pharmacology , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Female , Gene Expression Regulation, Enzymologic/genetics , Gene Expression Regulation, Neoplastic/genetics , Glucuronidase/genetics , Humans , Mice , Mice, Transgenic , Multiple Myeloma/genetics , Multiple Myeloma/pathology , Neoplasm Metastasis , Neoplasms, Experimental/enzymology , Neoplasms, Experimental/genetics , Neoplasms, Experimental/pathology , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/pathology , Polysaccharide-Lyases/pharmacology
4.
J Biol Chem ; 282(4): 2363-73, 2007 Jan 26.
Article in English | MEDLINE | ID: mdl-17121850

ABSTRACT

Heparan sulfate proteoglycans (HSPGs) function as a co-receptor for heparin-binding growth factors, such as fibroblast growth factors (FGFs) and heparin-bound epidermal growth factor (HB-EGF). The HS side chain of HSPGs can be cleaved by HPR1 (heparanase-1), an endoglycosidase that is overexpressed in many types of malignancies. In the present study, we demonstrated that HPR1 expression in pancreatic adenocarcinomas inversely correlated with the presence of heparan sulfate (HS) in the basement membrane. In vitro cell culture study revealed that cell surface HS levels inversely correlated with HPR1 activity in five pancreatic cancer cell lysates and their conditioned media. Heparin and PI-88, two HPR1 inhibitors, were able to increase cell surface HS levels in PANC-1 cells in a dose-dependent manner. The ability of HPR1 to degrade cell surface HS was confirmed by showing that cell surface HS levels were increased in HT1080 cells stably transfected with the HPR1 antisense gene but was decreased in the cells overexpressing HPR1. Further studies showed that PI-88 and heparin were able to stimulate PANC-1 cell proliferation in the absence or presence of exogenous FGF2, whereas exogenous HPR1 was able to inhibit PANC-1 cell proliferation in a dose-dependent manner. Modulation of PANC-1 cell proliferation by HPR1 or HPR1 inhibitors corresponded with the inhibition or activation of the mitogen-activated protein kinase. Our results suggest that HPR1 expressed in pancreatic adenocarcinomas can suppress the proliferation of pancreatic tumor cells in response to the growth factors that require HSPGs as their co-receptors.


Subject(s)
Adenoma/metabolism , Fibroblast Growth Factor 2/metabolism , Glucuronidase/metabolism , Heparan Sulfate Proteoglycans/metabolism , Pancreatic Neoplasms/metabolism , Adenoma/pathology , Cell Line, Tumor , Cell Membrane/metabolism , Cell Proliferation , DNA, Antisense , Glucuronidase/genetics , Humans , Immunohistochemistry , Male , Pancreatic Neoplasms/pathology , Signal Transduction , Transfection
5.
Curr Med Chem ; 13(18): 2101-11, 2006.
Article in English | MEDLINE | ID: mdl-16918340

ABSTRACT

Heparanase is an endo-beta-D-glucuronidase that degrades heparan sulfate glycosaminoglycan side chains of the proteoglycans in extracellular matrix and basement membrane. Heparanase enzymatic activity is important in the promotion of tumor angiogenesis, primary tumor growth, invasion, and metastasis. Expression of heparanase in many tumor types conversely correlates with prognosis. Much progress has been made in studying the regulation of heparanase expression, processing and activation. The interaction between heparanase and its substrate heparan sulfate has been well characterized. The fact that heparanase was identified as the single predominant heparan sulfate-degrading enzyme in human cancer sparked considerable interest in developing heparanase inhibitors for potential therapeutic applications. Recent progress in drug development led to several classes of heparanase inhibitors, including chemically modified natural products, small molecule inhibitors, and antibodies. Some of these inhibitors have demonstrated potent activities to inhibit tumor angiogenesis, tumor progress, or tumor metastasis. A leading compound, PI-88, is currently being evaluated in clinical phase II trials in patients with melanoma, liver, or lung cancers. This review summarizes the recent progress in heparanase biochemical research and the development of heparanase antagonists as novel anti-cancer therapeutics.


Subject(s)
Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Glucuronidase/antagonists & inhibitors , Glucuronidase/physiology , Neoplasms/drug therapy , Biomarkers/analysis , Drug Design , Glucuronidase/analysis , Humans , Neoplasm Metastasis , Neoplasms/pathology , Neovascularization, Pathologic , Nitrogen/chemistry , Oxygen/chemistry , Sulfates/chemistry
6.
J Biomed Mater Res A ; 78(4): 792-7, 2006 Sep 15.
Article in English | MEDLINE | ID: mdl-16741978

ABSTRACT

A family of biopolymers engineered to protect and stabilize heparin binding growth factors (HBGFs) show remarkable properties as wound healing agents in several in vivo tissue repair models to the extend that damaged tissues would recover almost its initial aspect and properties. These polymers where named RGTA for regenerating agents and proposed to act in vivo by enhancing the bioavailability of HBGFs at the site of the injury. To provide support for this hypothesis, we studied interaction of RGTA with FGF-2, taken as the paradigm of HBGFs, and its high- and low-affinity receptors as well as its ability to inhibit heparanase activity. We show that RGTA is comparable to heparin as it favors FGF-2 binding to FGFR-1 and FGF-2 dimerization and potentiates FGF-2-induced mitogenic activity. Furthermore, we show that RGTA inhibits the release of FGF-2 from its extracellular matrix storage sites by heparanase. Our data provide new evidence to support that RGTA may act in vivo both by enhancing HBGF activity and preserving HBGF availability by protecting the matrix low affinity heparan sulfates from rapid heparanase degradation.


Subject(s)
Heparin/pharmacology , Molecular Mimicry , Polymers/pharmacology , Animals , CHO Cells , Cattle , Cells, Cultured , Cricetinae , Dextrans/pharmacology , Dimerization , Extracellular Matrix/drug effects , Extracellular Matrix/metabolism , Fibroblast Growth Factor 2/chemistry , Fibroblast Growth Factor 2/metabolism , Glucuronidase/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism
7.
Biochem Biophys Res Commun ; 345(1): 438-45, 2006 Jun 23.
Article in English | MEDLINE | ID: mdl-16682007

ABSTRACT

Compelling evidence suggest that vascular endothelial growth factor (VEGF) and its receptors, especially receptor 2 (VEGFR2, or kinase insert domain-containing receptor, KDR), play a critical role in angiogenesis under both physiological and pathological conditions, including cancer and angiogenic retinopathies such as age-related macular degeneration (AMD). To this end, inhibition of angiogenesis with antagonists to either VEGF or KDR has yielded significant therapeutic efficacy both in preclinical studies in animal models and in clinical trials in patients with cancer and AMD. We previously reported the identification of a high affinity, fully human anti-KDR antibody fragment, 1121B Fab, through a highly stringent affinity maturation process with a Fab originally isolated from a naïve human antibody phage display library. In this study, we demonstrate that 1121B Fab is able to strongly block KDR/VEGF interaction, resulting in potent inhibition of an array of biological activities of VEGF, including activation of the receptor and its signaling pathway, intracellular calcium mobilization, and migration and proliferation of endothelial cells. Taken together, our data lend strong support to the further development of 1121B Fab fragment as an anti-angiogenesis agent in both cancer and angiogenic retinopathies.


Subject(s)
Antibodies, Monoclonal/immunology , Endothelial Cells/immunology , Immunoglobulin Fab Fragments/immunology , Vascular Endothelial Growth Factor A/immunology , Vascular Endothelial Growth Factor Receptor-2/immunology , Animals , Cell Movement/immunology , Cell Proliferation , Cells, Cultured , Humans , Swine
8.
Bioorg Med Chem Lett ; 16(2): 404-8, 2006 Jan 15.
Article in English | MEDLINE | ID: mdl-16246551

ABSTRACT

A novel class of N-(4-{[4-(1H-benzoimidazol-2-yl)-arylamino]-methyl}-phenyl)-benzamides are described as inhibitors of the endo-beta-glucuronidase heparanase. Among them are N-(4-{[4-(1H-benzoimidazol-2-yl)-phenylamino]-methyl}-phenyl)-3-bromo-4-methoxy-benzamide (15h), and N-(4-{[5-(1H-benzoimidazol-2-yl)-pyridin-2-ylamino]-methyl}- phenyl)-3-bromo-4-methoxy-benzamide (23) which displayed good heparanase inhibitory activity (IC(50) 0.23-0.29 microM), with the latter showing oral exposure in mice.


Subject(s)
Benzamides/pharmacology , Benzimidazoles/pharmacology , Enzyme Inhibitors/pharmacology , Glucuronidase/antagonists & inhibitors , Administration, Oral , Animals , Benzamides/administration & dosage , Benzamides/chemistry , Benzimidazoles/administration & dosage , Benzimidazoles/chemistry , Carbohydrate Conformation , Carbohydrate Sequence , Drug Design , Drug Evaluation, Preclinical , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/chemistry , In Vitro Techniques , Mice , Models, Animal , Molecular Sequence Data , Molecular Structure , Molecular Weight , Structure-Activity Relationship
9.
Bioorg Med Chem Lett ; 16(2): 409-12, 2006 Jan 15.
Article in English | MEDLINE | ID: mdl-16246560

ABSTRACT

A novel class of 1-[4-(1H-benzoimidazol-2-yl)-phenyl]-3-[4-(1H-benzoimidazol-2-yl)-phenyl]-ureas are described as potent inhibitors of heparanase. Among them are 1,3-bis-[4-(1H-benzoimidazol-2-yl)-phenyl]-urea (7a) and 1,3-bis-[4-(5,6-dimethyl-1H-benzoimidazol-2-yl)-phenyl]-urea (7d), which displayed good heparanase inhibitory activity (IC(50) 0.075-0.27 microM). Compound 7a showed good efficacy in a B16 metastasis model.


Subject(s)
Carbanilides/pharmacology , Enzyme Inhibitors/pharmacology , Glucuronidase/antagonists & inhibitors , Lung Neoplasms/drug therapy , Melanoma, Experimental/drug therapy , Animals , Carbanilides/chemical synthesis , Carbanilides/classification , Cell Line, Tumor , Drug Design , Drug Screening Assays, Antitumor , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/classification , In Vitro Techniques , Melanoma, Experimental/secondary , Mice , Molecular Structure , Molecular Weight , Neoplasm Metastasis/drug therapy , Structure-Activity Relationship
10.
Cancer Res ; 65(13): 5778-84, 2005 Jul 01.
Article in English | MEDLINE | ID: mdl-15994953

ABSTRACT

Heparanase is an enzyme that cleaves heparan sulfate and through this activity promotes tumor growth, angiogenesis, invasion, and metastasis in several tumor types. In human breast cancer patients, heparanase expression is associated with sentinel lymph node metastases. However, the precise role of heparanase in the malignant progression of breast cancer is unknown. To examine this, a variant of MDA-MB-231 cells was transfected with the cDNA for human heparanase (HPSE cells) or with vector alone as a control (NEO cells). Transfection produced a 6-fold increase in heparanase activity in HPSE cells relative to NEO cells. When injected into the mammary fat pads of severe combined immunodeficient mice, the tumors formed by HPSE cells initially grow significantly faster than the tumors formed by NEO cells. The rapid growth is due in part to increased angiogenesis, as microvessel densities are substantially elevated in primary HPSE tumors compared with NEO tumors. Although metastases to bones are not detected, surprisingly vigorous bone resorption is stimulated in animals bearing tumors formed by the HPSE cells. These animals have high serum levels of the C-telopeptide derived from type I collagen as well as significant elevation of the active form of tartrate-resistant acid phosphatase (TRAP)-5b. In contrast, in animals having a high tumor burden of Neo cells, the serum levels of C-telopeptide and TRAP-5b never increase above the levels found before tumor injection. Consistent with these findings, histologic analysis for TRAP-expressing cells reveals extensive osteoclastogenesis in animals harboring HPSE tumors. In vitro osteoclastogenesis assays show that the osteoclastogenic activity of HPSE cell conditioned medium is significantly enhanced beyond that of NEO conditioned medium. This confirms that a soluble factor or factors that stimulate osteoclastogenesis are specifically produced when heparanase expression is elevated. These factors exert a distal effect resulting in resorption of bone and the accompanying enrichment of the bone microenvironment with growth-promoting factors that may nurture the growth of metastatic tumor cells. This novel role for heparanase as a promoter of osteolysis before tumor metastasis suggests that therapies designed to block heparanase function may disrupt the early progression of bone-homing tumors.


Subject(s)
Adenocarcinoma/enzymology , Bone Resorption/enzymology , Breast Neoplasms/enzymology , Glucuronidase/biosynthesis , Adenocarcinoma/pathology , Adenocarcinoma/secondary , Animals , Bone Neoplasms/secondary , Breast Neoplasms/pathology , Cell Line, Tumor , Female , Humans , Mice , Mice, SCID , Neoplasm Transplantation , Transplantation, Heterologous
11.
Blood ; 105(3): 1303-9, 2005 Feb 01.
Article in English | MEDLINE | ID: mdl-15471949

ABSTRACT

Although widespread skeletal dissemination is a critical step in the progression of myeloma, little is known regarding mechanisms that control metastasis of this cancer. Heparanase-1 (heparanase), an enzyme that cleaves heparan sulfate chains, is expressed at high levels in some patients with myeloma and promotes metastasis of some tumor types (eg, breast, lymphoma). Using a severe combined immunodeficient (SCID) mouse model, we demonstrate that enhanced expression of heparanase by myeloma cells dramatically up-regulates their spontaneous metastasis to bone. This occurs from primary tumors growing subcutaneously and also from primary tumors established in bone. Interestingly, tumors formed by subcutaneous injection of cells metastasize not only to bone, but also to other sites including spleen, liver, and lung. In contrast, tumors formed by injection of cells directly into bone exhibit a restricted pattern of metastasis that includes dissemination of tumor to other bones but not to extramedullary sites. In addition, expression of heparanase by myeloma cells (1) accelerates the initial growth of the primary tumor, (2) increases whole-body tumor burden as compared with controls, and (3) enhances both the number and size of microvessels within the primary tumor. These studies describe a novel experimental animal model for examining the spontaneous metastasis of bone-homing tumors and indicate that heparanase is a critical determinant of myeloma dissemination and growth in vivo.


Subject(s)
Bone and Bones/pathology , Cell Division/physiology , Glucuronidase/metabolism , Multiple Myeloma/pathology , Neoplasm Metastasis/pathology , DNA, Complementary/analysis , Glucuronidase/genetics , Humans , Recombinant Proteins , Transfection , Tumor Cells, Cultured
12.
J Cell Sci ; 118(Pt 1): 253-64, 2005 Jan 01.
Article in English | MEDLINE | ID: mdl-15615789

ABSTRACT

Crucial events in myogenesis rely on the highly regulated spatiotemporal distribution of cell surface heparan sulfate proteoglycans to which are associated growth factors, thus creating a specific microenvironment around muscle cells. Most growth factors involved in control of myoblast growth and differentiation are stored in the extracellular matrix through interaction with specific sequences of glycosaminoglycan oligosaccharides, mainly heparan sulfate (HS). Different HS subspecies revealed by specific antibodies, have been shown to provide spatiotemporal regulation during muscle development. We have previously shown that glycosaminoglycan (GAG) mimetics called RGTA (ReGeneraTing Agent), stimulate muscle precursor cell growth and differentiation. These data suggest an important role of GAGs during myogenesis; however, little is yet known about the different species of GAGs synthesized during myogenesis and their metabolic regulation. We therefore quantified GAGs during myogenesis of C2.7 cells and show that the composition of GAG species was modified during myogenic differentiation. In particular, HS levels were increased during this process. In addition, the GAG mimetic RGTA, which stimulated both growth and differentiation of C2.7 cells, increased the total amount of GAG produced by these cells without significantly altering their rate of sulfation. RGTA treatment further enhanced HS levels and changed its sub-species composition. Although mRNA levels of the enzymes involved in HS biosynthesis were almost unchanged during myogenic differentiation, heparanase mRNA levels decreased. RGTA did not markedly alter these levels. Here we show that the effects of RGTA on myoblast growth and differentiation are in part mediated through an alteration of GAG species and provide an important insight into the role of these molecules in normal or pathologic myogenic processes.


Subject(s)
Glycosaminoglycans/chemical synthesis , Glycosaminoglycans/pharmacology , Muscle Development , Myoblasts/drug effects , Cell Differentiation/drug effects , Cell Line , Cell Proliferation/drug effects , Chromatography, High Pressure Liquid , Enzyme-Linked Immunosorbent Assay , Glycosaminoglycans/analysis , Glycosaminoglycans/chemistry , Heparitin Sulfate/biosynthesis , Immunohistochemistry , Molecular Structure , Muscle, Skeletal/cytology , Myoblasts/cytology , Polymerase Chain Reaction , RNA, Messenger/analysis , RNA, Messenger/metabolism
13.
J Cell Sci ; 117(Pt 13): 2745-56, 2004 Jun 01.
Article in English | MEDLINE | ID: mdl-15150322

ABSTRACT

In this report we utilize a novel antagonist antibody to the human VEGFR-3 to elucidate the role of this receptor in in vitro tubular morphogenesis of bovine and human endothelial cells (EC cells) induced by VEGF-C. The antibody hF4-3C5 was obtained by panning a human phage display library on soluble human VEGFR-3. The binding affinity constant of hF4-3C5 significantly exceeds that of the interaction of VEGFR-3 with VEGF-C. hF4-3C5 strongly inhibits the binding of soluble VEGFR-3 to immobilized VEGF-C and abolishes the VEGF-C-mediated mitogenic response of cells that expresses a chimeric human VEGFR-3-cFMS receptor. In fluorescence experiments, hF4-3C5 reactivity is observed with human lymphatic endothelial cells (LECs) and human umbilical vein endothelial cells (HUVECs). Binding of hF4-3C5 shows that about half of bovine aortic endothelial (BAE) cells express VEGFR-3 and cells in this subpopulation are primarily responsible for the chemotactic response to the mature form of VEGF-C (VEGF-C(DeltaNDeltaC)). This response was strongly inhibited by the addition of hF4-3C5. In vitro tube formation by BAE cells induced by VEGF-C(DeltaNDeltaC) was reduced by greater than 60% by hF4-3C5 whereas the response to VEGF(165) was unaffected. Addition of hF4-3C5 together with an antagonist antibody to VEGFR-2 completely abolished the response to VEGF-C(DeltaNDeltaC). Similar results were obtained with HUVECs. Together, these findings point to a role for VEGFR-3 in vascular tubular morphogenesis and highlight the utility of hF4-3C5 as a tool for the investigation of the biology of VEGFR-3.


Subject(s)
Antibodies, Monoclonal/metabolism , Endothelium, Vascular/growth & development , Morphogenesis , Vascular Endothelial Growth Factor C/metabolism , Vascular Endothelial Growth Factor Receptor-3/antagonists & inhibitors , Animals , Aorta/cytology , Cattle , Cell Division/drug effects , Chemotaxis/drug effects , Clone Cells , Culture Media, Conditioned/pharmacology , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Fluorescent Antibody Technique, Indirect , Humans , Lymphatic System/cytology , Mice , Microscopy, Fluorescence , NIH 3T3 Cells , Peptide Library , Recombinant Proteins/metabolism , Umbilical Veins/cytology , Vascular Endothelial Growth Factor C/pharmacology , Vascular Endothelial Growth Factor Receptor-3/metabolism
14.
Cancer Res ; 63(24): 8749-56, 2003 Dec 15.
Article in English | MEDLINE | ID: mdl-14695190

ABSTRACT

Heparanase is an enzyme that cleaves heparan sulfate chains of proteoglycans, and its expression has been associated with increased growth, metastasis, and angiogenesis of some tumors. Because myeloma tumor cells express high levels of the syndecan-1 heparan sulfate proteoglycan and because these tumors grow as highly vascularized aggregates within the bone marrow, we analyzed the activity, expression, and function of heparanase in myeloma patients. Analysis of heparanase activity in the plasma isolated from bone marrow biopsies of 100 patients reveals 86 positive for heparanase activity and 14 negative. The bone marrow samples can be further divided into three categories of heparanase activity, high activity (42 patients), low activity (44 patients), and negative (14 patients). In contrast to the bone marrow plasma, levels of heparanase activity in peripheral blood plasma of 29 myeloma patients were either negative or low, suggesting that in multiple myeloma, heparanase functions in the local microenvironment of the bone marrow and its activity is not significantly elevated systemically. Immunohistochemistry reveals that patients with high levels of heparanase activity often have tumor cells with intense staining for the enzyme. Interestingly, a marked heterogeneity among tumor cells was noted, with clusters of heavily stained cells surrounded by cells with weak or negative staining for heparanase. Analysis of microvessel density reveals a strikingly higher concentration of vessels in patients with high heparanase activity (78.96 vessels/mm(2)) as compared with patients negative for heparanase activity (25.03 vessels/mm(2)). When human myeloma cells transfected with the cDNA for heparanase are implanted in severe combined immunodeficient (SCID) mice, the resulting tumors exhibited a significantly higher microvessel density than did tumors established with control cells. Thus, expression of heparanase appears to play a direct role in enhancing microvessel density in these myeloma tumors. Because heparanase is known to stimulate angiogenesis, and because high microvessel density is associated with poor prognosis in myeloma, we conclude that heparanase expression likely plays an important role in regulating the growth and progression of myeloma, and that therapies designed to block heparanase activity may aid in controlling this cancer.


Subject(s)
Glucuronidase/metabolism , Multiple Myeloma/blood supply , Multiple Myeloma/enzymology , Animals , Bone Marrow/enzymology , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Membrane Glycoproteins/biosynthesis , Membrane Glycoproteins/genetics , Mice , Mice, SCID , Multiple Myeloma/genetics , Multiple Myeloma/pathology , Neovascularization, Pathologic/enzymology , Neovascularization, Pathologic/pathology , Proteoglycans/biosynthesis , Proteoglycans/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Syndecan-1 , Syndecans
15.
Cancer Res ; 63(22): 7733-41, 2003 Nov 15.
Article in English | MEDLINE | ID: mdl-14633698

ABSTRACT

Heparanase is an endo-beta-glucuronidase responsible for the cleavage of heparan sulfate, participating in extracellular matrix degradation and remodeling. Traditionally, heparanase activity was well correlated with the metastatic potential of a large number of tumor-derived cell types. More recently, heparanase up-regulation was detected in essentially all human tumors examined, correlating, in some cases, with poor postoperative survival and increased tumor vascularity. The role of heparanase in primary tumor progression is, however, poorly understood. Here, we overexpressed the human heparanase gene in a human glioma cell line, U87. We found that heparanase overexpression induces cell invasion, as might be expected. Surprisingly, elevated heparanase expression levels correlated with decreased proliferation rates and increased cell spreading and formation of a tight monolayer rather than large cell aggregates. This phenotypic appearance was accompanied by beta1-integrin activation, FAK and Akt phosphorylation, and Rac activation. In a xenograft tumor model, relatively moderate heparanase expression levels significantly enhanced tumor development and tumor vascularity, whereas high heparanase expression levels inhibited tumor growth. These results indicate that heparanase activates signal transduction pathways and, depending on its expression levels, may modulate tumor progression.


Subject(s)
Glioma/enzymology , Glioma/pathology , Glucuronidase/physiology , Animals , Cell Adhesion/physiology , Cell Division/physiology , Cell Line, Tumor , Cell Movement/physiology , Chickens , Glioma/genetics , Glucuronidase/biosynthesis , Glucuronidase/genetics , Humans , Integrin beta1/metabolism , Integrin beta1/physiology , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Invasiveness , Neoplasm Transplantation , Transfection , Transplantation, Heterologous
16.
Biochem Biophys Res Commun ; 308(4): 885-91, 2003 Sep 05.
Article in English | MEDLINE | ID: mdl-12927802

ABSTRACT

Heparanase is an endo-beta-D-glucuronidase involved in cleavage of heparan sulfate residues and hence participates in extracellular matrix degradation and remodeling. The heparanase cDNA encodes for a polypeptide of 543 amino acids that appears as a approximately 65 kDa band in SDS-PAGE analysis. The protein undergoes a proteolytic cleavage that is likely to occur at two potential cleavage sites, Glu(109)-Ser(110) and Gln(157)-Lys(158), yielding an 8 kDa polypeptide at the N-terminus, a 50 kDa polypeptide at the C-terminus, and a 6 kDa linker polypeptide that resides in-between. The active form of heparanase has long been thought to be a 50 kDa polypeptide isolated from cells and tissues. However, attempts to obtain heparanase activity after expression of the 50 kDa polypeptide failed, suggesting that the N-terminal region is important for heparanase enzymatic activity. It has been hypothesized that heterodimer formation between the 8 and 50 kDa heparanase subunits is important for heparanase enzymatic activity. By individually or co-expressing the 8 and 50 kDa heparanase subunits in mammalian cells, we demonstrate specific association between the heparanase subunits by means of co-immunoprecipitation and pull-down experiments. Moreover, a region in the 50 kDa heparanase subunit that mediates interaction with the 8 kDa subunit was identified. Altogether, our results clearly indicate that heterodimer formation is necessary and sufficient for heparanase enzymatic activity in mammalian cells.


Subject(s)
Glucuronidase/metabolism , Amino Acids/chemistry , Binding Sites , Blotting, Western , Cell Line , Cloning, Molecular , DNA, Complementary/metabolism , Dimerization , Electrophoresis, Polyacrylamide Gel , Glucuronidase/chemistry , Glutathione Transferase/metabolism , Heparitin Sulfate/chemistry , Humans , Immunoblotting , Peptides/chemistry , Plasmids/metabolism , Precipitin Tests , Protein Structure, Tertiary , Transfection , Tumor Cells, Cultured
17.
Protein Expr Purif ; 26(3): 425-31, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12460766

ABSTRACT

Heparanase is an endoglucuronidase that plays an important role in tumor invasion and metastasis. A full-length heparanase gene was cloned from a mouse embryo cDNA library and determined to encode a protein of 535 amino acids that is 77% identical to human heparanase. The full-length mouse gene was stably expressed in NS0 myeloma cells. The recombinant mouse heparanase protein was purified to homogeneity from cell lysates by a combination of Con-A affinity chromatography, heparin affinity chromatography, and size exclusion chromatography. The purified protein consisted of a non-covalent heterodimer of 50- and 8-kDa polypeptides, similar to the human homolog. The protein was enzymatically active in assays using radiolabeled ECM and heparan sulfate as substrates. The maximum heparanase activity was observed at acidic conditions; however, significant activity was also detected at neutral pH. The enzymatic activity of mouse heparanase was blocked by known heparanase inhibitors.


Subject(s)
Glucuronidase/genetics , Glucuronidase/isolation & purification , Amino Acid Sequence , Animals , Base Sequence , Chromatography, Affinity , Cloning, Molecular , DNA, Complementary/genetics , Electrophoresis, Polyacrylamide Gel , Enzyme Inhibitors/pharmacology , Gene Expression , Glucuronidase/antagonists & inhibitors , Glucuronidase/chemistry , Heparitin Sulfate/metabolism , Humans , Hydrogen-Ion Concentration , Mice , Molecular Sequence Data , Molecular Weight , Sequence Homology , Tumor Cells, Cultured
18.
J Cell Biochem ; 85(2): 357-68, 2002.
Article in English | MEDLINE | ID: mdl-11948691

ABSTRACT

Co-expression of NRP1 and (VEGFR-2) KDR on the surface of endothelial cells (EC) enhances VEGF165 binding to KDR and EC chemotaxis in response to VEGF165. Overexpression of NRP1 by prostate tumor cells in vivo results in increased tumor angiogenesis and growth. We investigated the molecular mechanisms underlying NRP1-mediated angiogenesis by analyzing the association of NRP1 and KDR. An intracellular complex containing NRP1 and KDR was immunoprecipitated from EC by anti-NRP1 antibodies only in the presence of VEGF165. In contrast, VEGF121, which does not bind to NRP1, did not support complex formation. Complexes containing VEGF165, NRP1, and KDR were also formed in an intercellular fashion by co-culture of EC expressing KDR only, with cells expressing NRP1 only, for example, breast carcinoma cells. VEGF165 also mediated the binding of a soluble NRP1 dimer to cells expressing KDR only, confirming the formation of such complexes. Furthermore, the formation of complexes containing KDR and NRP1 markedly increased 125I-VEGF165 binding to KDR. Our results suggest that formation of a ternary complex of VEGF165, KDR, and NRP1 potentiates VEGF165 binding to KDR. These complexes are formed on the surface of EC and in a juxtacrine manner via association of tumor cell NRP1 and EC KDR.


Subject(s)
Endothelial Growth Factors/metabolism , Endothelium, Vascular/metabolism , Lymphokines/metabolism , Nerve Tissue Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Cell Surface/metabolism , Receptors, Growth Factor/metabolism , Receptors, Mitogen/metabolism , Affinity Labels , Binding, Competitive , Cells, Cultured , Cross-Linking Reagents , Humans , Macromolecular Substances , Models, Molecular , Neuropilin-1 , Phosphorylation , Precipitin Tests , Protein Binding , Protein Isoforms/metabolism , Receptors, Vascular Endothelial Growth Factor , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Signal Transduction , Tyrosine/metabolism , Umbilical Veins/metabolism , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...