Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Plant Dis ; 98(10): 1450, 2014 Oct.
Article in English | MEDLINE | ID: mdl-30703965

ABSTRACT

In the spring of 2014, a survey of viral diseases on wheat (Triticum aestivum L.) was carried out in Hebei Province, China. The samples with virus-like symptoms of dwarfing and flag leaf yellowing were collected in Zhaoxian, Quyang, Anxin, and Luannan. To reproduce the viral symptoms and confirm whether the unknown virus was transmitted by insect vectors, the nymphs of aviruliferous planthopper (Laodelphax striatellus Fallen, Homoptera: Delphacidae) were transferred onto diseased wheat from the field for a 3-day acquisition access period and a 10-day incubation on fresh wheat seedlings, and then were exposed to 2- to 3-leaf stage wheat seedlings of wheat variety Shixin828 for a 3-day inoculation access period. The infected wheat plants developed mosaic symptoms on the young leaves at 7 days post inoculation (dpi), and followed with severe symptoms including stunting, chlorotic spots, and striation along the veins of leaves at around 14 dpi. The infection symptoms were same as in the field but distinct from wheat infected with Rice black streaked dwarf virus (RBSDV) or Northern cereal mosaic virus (NCMV). For further confirmation, total RNA was extracted from the symptomatic wheat leaves, and NCMV specific primers, NCMV-PF/NCMV-PR (5'-ATGGATAAGAAAGCAAGTGGA-3'/5'-TTAAAAGTCGGCATACGGGTC-3') and RBSDV specific primers, S10-F/S10-R (5'-TTACCCAACATCACGCAACT-3'/5'-GAGCAGGAACTTCACGACAAC-3') were used for amplification of sequences of phosphoprotein and coat protein genes, respectively. Neither RBSDV nor NCMV were present in the symptomatic tissue according to the RT-PCR assay (4). Tissues derived from symptomatic wheat leaves were fixed and embedded in Spurr's resin and used for ultra-thin sectioning and transmission electron microscopy observations, revealing large amounts of Rhabdovirus-like particles in the cytoplasm. The identified particles were about 315 to 353 × 46 to 57 nm, similar in size to Barley yellow striate mosaic virus (BYSMV), a member of the genus Cytorhabdovirus reported from Italy (2). The specific primer pair (5'-ACTAAGGGGGTACTCCGACC-3' and 5'-CTGATCTGCTTTGAGGGGCA-3') was designed based on the reported polymerase (L) gene sequence of BYSMV isolate Zanjan-1 (GenBank Accession No. FJ665628) (1), and used for the BYSMV detection by RT-PCR. A single bright band of the expected size (~500 bp) was obtained from total RNA extracted from the plants exhibiting symptoms in the greenhouse. No such band was amplified from asymptomatic plants, while 15 out of 23 field samples also produced the same 500-bp products in RT-PCR. PCR products from three virus-positive field samples were sequenced directly and the sequences were submitted to GenBank (KM052176, KM052177, and KM052178). BLAST search showed that the sequences shared 96 to 97% nucleotide identity with the polymerase L gene sequence of BYSMV isolate Zanjan-1, whereas only 73 to 75% identity with NCMV (AB030277 and GU985153) (1,3,5). To our knowledge, this is the first report of BYSMV occurrence on wheat in China. References: (1) R. Almasi et al. J. Phytopathol. 158:351, 2010. (2) A. Appiano et al. Cytol. 6:105, 1974. (3) H. C. Chen et al. Sci. Agric. Sinica 3:64, 1980. (4) X. F. Duan et al. Acta Phytopathol. Sinica 40:337, 2010. (5) F. Tanno et al. Arch. Virol. 145:1373, 2000.

2.
FASEB J ; 14(15): 2477-85, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11099465

ABSTRACT

We have previously demonstrated that halofuginone, a low molecular weight quinazolinone alkaloid, is a potent inhibitor of collagen alpha1(I) and matrix metalloproteinase 2 (MMP-2) gene expression. Halofuginone also effectively suppresses tumor progression and metastasis in mice. These results together with the well-documented role of extracellular matrix (ECM) components and matrix degrading enzymes in formation of new blood vessels led us to investigate the effect of halofuginone on the angiogenic process. In a variety of experimental system, representing sequential events in the angiogenic cascade, halofuginone treatment resulted in profound inhibitory effect. Among these are the abrogation of endothelial cell MMP-2 expression and basement membrane invasion, capillary tube formation, and vascular sprouting, as well as deposition of subendothelial ECM. The most conclusive anti-angiogenic activity of halofuginone was demonstrated in vivo (mouse corneal micropocket assay) by showing a marked inhibition of basic fibroblast growth factor (bFGF) -induced neovascularization in response to systemic administration of halofuginone, either i.p. or in the diet. The ability of halofuginone to interfere with key events in neovascularization, together with its oral bioavailability and safe use as an anti-parasitic agent, make it a promising drug for further evaluation in the treatment of a wide range of diseases associated with pathological angiogenesis.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Neovascularization, Physiologic/drug effects , Quinazolines/pharmacology , Animals , Aorta/drug effects , Capillaries/drug effects , Cattle , Dose-Response Relationship, Drug , Endothelium, Vascular/drug effects , Endothelium, Vascular/enzymology , Fibroblast Growth Factor 2/pharmacology , Matrix Metalloproteinase 2/biosynthesis , Piperidines , Quinazolinones , Rats
3.
FASEB J ; 14(15): 2532-9, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11099472

ABSTRACT

Neuropilin-1 (NRP1) is a VEGF(165) and semaphorin receptor expressed by vascular endothelial cells (EC) and tumor cells. The function of NRP1 in tumor cells is unknown. NRP1 was overexpressed in Dunning rat prostate carcinoma AT2.1 cells using a tetracycline-inducible promoter. Concomitant with increased NRP1 expression in response to a tetracycline homologue, doxycycline (Dox), basal cell motility, and VEGF(165) binding were increased three- to fourfold in vitro. However, induction of NRP1 did not affect tumor cell proliferation. When rats injected with AT2.1/NRP1 tumor cells were fed Dox, NRP1 synthesis was induced in vivo and AT2.1 cell tumor size was increased 2.5- to 7-fold in a 3-4 wk period compared to controls. The larger tumors with induced NRP1 expression were characterized by markedly increased microvessel density, increased proliferating EC, dilated blood vessels, and notably less tumor cell apoptosis compared to noninduced controls. It was concluded that NRP1 expression results in enlarged tumors associated with substantially enhanced tumor angiogenesis.


Subject(s)
Neovascularization, Pathologic , Nerve Tissue Proteins/biosynthesis , Prostatic Neoplasms/blood supply , Receptors, Cell Surface/biosynthesis , Animals , Cell Movement , Doxorubicin/pharmacology , Endothelial Growth Factors/metabolism , Lymphokines/metabolism , Male , Neuropilin-1 , Prostatic Neoplasms/pathology , Rats , Receptor Protein-Tyrosine Kinases/biosynthesis , Receptors, Growth Factor/biosynthesis , Receptors, Vascular Endothelial Growth Factor , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
4.
Proc Natl Acad Sci U S A ; 97(6): 2573-8, 2000 Mar 14.
Article in English | MEDLINE | ID: mdl-10688880

ABSTRACT

Neuropilin-1 (NRP1) is a 130-kDa transmembrane receptor for semaphorins, mediators of neuronal guidance, and for vascular endothelial growth factor 165 (VEGF(165)), an angiogenesis factor. A 2.2-kb truncated NRP1 cDNA was cloned that encodes a 644-aa soluble NRP1 (sNRP1) isoform containing just the a/CUB and b/coagulation factor homology extracellular domains of NRP1. sNRP1 is secreted by cells as a 90-kDa protein that binds VEGF(165), but not VEGF(121). It inhibits (125)I-VEGF(165) binding to endothelial and tumor cells and VEGF(165)-induced tyrosine phosphorylation of KDR in endothelial cells. The 3' end of sNRP1 cDNA contains a unique, 28-bp intron-derived sequence that is absent in full-length NRP1 cDNA. Using a probe corresponding to this unique sequence, sNRP1 mRNA could be detected by in situ hybridization differentially from full-length NRP1 mRNA, for example, in cells of liver, kidney, skin, and breast. Analysis of blood vessels in situ showed that NRP1, but not sNRP1, was expressed. sNRP1 was functional in vivo. Unlike control tumors, tumors of rat prostate carcinoma cells expressing recombinant sNRP1 were characterized by extensive hemorrhage, damaged vessels, and apoptotic tumor cells. These results demonstrate the existence of a naturally occurring, soluble NRP1 that is expressed differently from intact NRP1 and that appears to be a VEGF(165) antagonist.


Subject(s)
Antineoplastic Agents/metabolism , Endothelial Growth Factors/metabolism , Lymphokines/metabolism , Nerve Tissue Proteins/metabolism , Animals , Apoptosis , Blotting, Northern , CHO Cells , Cloning, Molecular , Cricetinae , DNA, Complementary/metabolism , Endothelial Growth Factors/antagonists & inhibitors , Humans , Introns , Liver/metabolism , Lymphokines/antagonists & inhibitors , Male , Molecular Sequence Data , Neoplasm Transplantation , Nerve Tissue Proteins/genetics , Neuropilin-1 , Phosphorylation , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Protein Binding/drug effects , Protein Isoforms , Rats , Recombinant Proteins/metabolism , Tissue Distribution , Tumor Cells, Cultured , Tyrosine/metabolism , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
5.
Cancer Metastasis Rev ; 19(1-2): 29-37, 2000.
Article in English | MEDLINE | ID: mdl-11191060

ABSTRACT

Neuropilin is both a receptor for semaphorins, which are mediators of neuronal guidance, and for VEGF, an angiogenesis factor. While the function of neuropilin in the nervous system has been characterized, its role in angiogenesis is only beginning to be elucidated. This article reviews some of the structural and functional features of neuropilin and presents the experimental evidence showing that it is a mediator of angiogenesis. In particular, we show that neuropilin and its soluble isoforms regulate tumor angiogenesis and subsequent tumor growth.


Subject(s)
Neovascularization, Pathologic/physiopathology , Neovascularization, Physiologic/physiology , Nerve Tissue Proteins/physiology , Animals , Endothelial Growth Factors/physiology , Humans , Lymphokines/physiology , Membrane Glycoproteins/physiology , Neuropilin-1 , Receptor Protein-Tyrosine Kinases/physiology , Receptors, Cell Surface/physiology , Receptors, Growth Factor/physiology , Receptors, Vascular Endothelial Growth Factor , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
6.
Int J Cancer ; 83(3): 424-31, 1999 Oct 29.
Article in English | MEDLINE | ID: mdl-10495437

ABSTRACT

Heparanase activity correlates with the metastatic potential of tumor cells. Moreover, the anti-metastatic effect of non-anti-coagulant species of heparin and certain sulfated polysaccharides was attributed to their heparanase-inhibiting activity. We investigated the effect of a chemically sulfated polysaccharide (laminarin), consisting primarily of beta-1,3 glucan (sodium laminarin), and of synthetic phosphorothioate oligodeoxynucleotides, primarily phosphorothioate homopolymer of cytidine (SdC28), on heparanase activity and tumor metastasis. Investigation of the ability of tumor cells to degrade heparan sulfate in intact extracellular matrix revealed that heparanase activity expressed by B16-BL6 mouse melanoma cells and 13762 MAT rat mammary adenocarcinoma cells was effectively inhibited by LS (50% inhibition at 0.2-1 microgram/ml), but there was no inhibition by sodium laminarin up to a concentration of 50 microgram/ml. Complete inhibition of the melanoma heparanase was obtained in the presence of 0.1 microM SdC28. A single i.p. injection of laminarin sulfate, but not of sodium laminarin, before i.v. inoculation of the melanoma or breast-carcinoma cells inhibited the extent of lung colonization by the tumor cells by 80 to 90%. Similar inhibition was exerted by 0.1 microM SdC28. At the effective concentrations, both compounds had a small effect on proliferation of the tumor cells and on growth of the primary tumors in vivo. These results further emphasize the involvement of heparanase in tumor metastasis and the potential clinical application of diverse heparanase-inhibiting molecules such as sulfated polysaccharides and synthetic polyanionic molecules.


Subject(s)
Antineoplastic Agents/pharmacology , Enzyme Inhibitors/pharmacology , Glucuronidase , Glycoside Hydrolases/antagonists & inhibitors , Neoplasm Metastasis/prevention & control , Oligodeoxyribonucleotides/pharmacology , Polysaccharides/pharmacology , Thionucleotides/pharmacology , Animals , Cell Adhesion/drug effects , Cell Division/drug effects , Female , Heparin/pharmacology , Lung Neoplasms/secondary , Melanoma, Experimental/secondary , Mice , Mice, Inbred C57BL , Rats
7.
Cancer Res ; 59(16): 4111-8, 1999 Aug 15.
Article in English | MEDLINE | ID: mdl-10463616

ABSTRACT

We have previously demonstrated that halofuginone, a widely used alkaloid coccidiostat, is a potent inhibitor of collagen alpha1(I) and matrix metalloproteinase 2 gene expression. Halofuginone also suppresses extracellular matrix deposition and cell proliferation. We investigated the effect of halofuginone on transplantable and chemically induced mouse bladder carcinoma. In both systems, oral administration of halofuginone resulted in a profound anticancerous effect, even when the treatment was initiated at advanced stages of tumor development. Although halofuginone failed to prevent proliferative preneoplastic alterations in the bladder epithelium, it inhibited further progression of the chemically induced tumor into a malignant invasive stage. Histological examination and in situ analysis of the tumor tissue revealed a marked decrease in blood vessel density and in both collagen alpha1(I) and H19 gene expression. H19 is regarded as an early marker of bladder carcinoma. The antiangiogenic effect of halofuginone was also demonstrated by inhibition of microvessel formation in vitro. We attribute the profound antitumoral effect of halofuginone to its combined inhibition of the tumor stromal support, vascularization, invasiveness, and cell proliferation.


Subject(s)
Antineoplastic Agents/pharmacology , Neovascularization, Pathologic/drug therapy , Protein Synthesis Inhibitors/pharmacology , Quinazolines/pharmacology , Urinary Bladder Neoplasms/drug therapy , Animals , Antineoplastic Agents/therapeutic use , Cell Division/drug effects , Humans , Male , Mice , Neoplasm Transplantation , Piperidines , Protein Synthesis Inhibitors/therapeutic use , Quinazolines/therapeutic use , Quinazolinones , Stromal Cells/drug effects , Stromal Cells/pathology , Tumor Cells, Cultured , Urinary Bladder Neoplasms/blood supply , Urinary Bladder Neoplasms/pathology
8.
J Cell Biol ; 146(1): 233-42, 1999 Jul 12.
Article in English | MEDLINE | ID: mdl-10402473

ABSTRACT

Neuropilin-1 (NRP1) is a receptor for two unrelated ligands with disparate activities, vascular endothelial growth factor-165 (VEGF165), an angiogenesis factor, and semaphorin/collapsins, mediators of neuronal guidance. To determine whether semaphorin/collapsins could interact with NRP1 in nonneuronal cells, the effects of recombinant collapsin-1 on endothelial cells (EC) were examined. Collapsin-1 inhibited the motility of porcine aortic EC (PAEC) expressing NRP1 alone; coexpressing KDR and NRP1 (PAEC/KDR/NRP1), but not parental PAEC; or PAEC expressing KDR alone. The motility of PAEC expressing NRP1 was inhibited by 65-75% and this inhibition was abrogated by anti-NRP1 antibody. In contrast, VEGF165 stimulated the motility of PAEC/KDR/NRP1. When VEGF165 and collapsin-1 were added simultaneously to PAEC/KDR/NRP1, dorsal root ganglia (DRG), and COS-7/NRP1 cells, they competed with each other in EC motility, DRG collapse, and NRP1-binding assays, respectively, suggesting that the two ligands have overlapping NRP1 binding sites. Collapsin-1 rapidly disrupted the formation of lamellipodia and induced depolymerization of F-actin in an NRP1-dependent manner. In an in vitro angiogenesis assay, collapsin-1 inhibited the capillary sprouting of EC from rat aortic ring segments. These results suggest that collapsin-1 can inhibit EC motility as well as axon motility, that these inhibitory effects on motility are mediated by NRP1, and that VEGF165 and collapsin-1 compete for NRP1-binding sites.


Subject(s)
Cell Movement/drug effects , Endothelial Growth Factors/pharmacology , Endothelium, Vascular/cytology , Glycoproteins/pharmacology , Lymphokines/pharmacology , Nerve Tissue Proteins/metabolism , Actins/metabolism , Animals , Aorta/cytology , Aorta/drug effects , Aorta/growth & development , Binding, Competitive , Cell Line , Cytoskeleton/drug effects , Endothelial Growth Factors/metabolism , Endothelium, Vascular/drug effects , Endothelium, Vascular/growth & development , Endothelium, Vascular/metabolism , Ganglia, Spinal/drug effects , Glycoproteins/metabolism , Humans , In Vitro Techniques , Lymphokines/metabolism , Neovascularization, Physiologic/drug effects , Nerve Tissue Proteins/genetics , Neuropilin-1 , Pseudopodia/drug effects , Rats , Rats, Inbred Lew , Receptor Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Growth Factor/genetics , Receptors, Growth Factor/metabolism , Receptors, Vascular Endothelial Growth Factor , Semaphorin-3A , Swine , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
9.
Mol Pathol ; 51(1): 21-5, 1998 Feb.
Article in English | MEDLINE | ID: mdl-9624415

ABSTRACT

AIMS/BACKGROUND: To study the expression of the H19 gene in hepatocellular carcinoma. H19 is an imprinted, maternally expressed gene, which is tightly linked, both physically and functionally, to the paternally expressed insulin-like growth factor 2 (IGF II). IGF II is known to be involved in liver carcinogenesis. H19 was first discovered in the fetal mouse liver to be under the same regulatory genes as alpha fetoprotein (alpha FP), a widely used tumour marker for hepatocellular carcinoma. METHODS: Using both radioactive and non-radioactive in situ hybridisation, the expression of the H19 gene was compared with the presence of alpha FP, as demonstrated by immunohistochemistry, in 18 cases of hepatocellular carcinoma. RESULTS: H19 expression was present in 13 of 18 cases, whereas staining for alpha FP was positive in only nine of 18 cases. Concordance was found in 12 of 18 tumours (66.7%). In general, the staining pattern for H19 was more diffuse than the immunohistochemical staining for alpha FP. CONCLUSIONS: The addition of a non-radioactive in situ hybridisation assay for H19 RNA to the panel of tumour markers used for the histopathological and cytological diagnosis of hepatocellular carcinoma might be useful.


Subject(s)
Biomarkers, Tumor/analysis , Carcinoma, Hepatocellular/genetics , Genomic Imprinting , Liver Neoplasms/genetics , RNA, Neoplasm/analysis , Adult , Aged , Carcinoma, Hepatocellular/metabolism , Female , Humans , Immunoenzyme Techniques , In Situ Hybridization/methods , Liver Neoplasms/metabolism , Male , Middle Aged , alpha-Fetoproteins/metabolism
10.
Cell ; 92(6): 735-45, 1998 Mar 20.
Article in English | MEDLINE | ID: mdl-9529250

ABSTRACT

Vascular endothelial growth factor (VEGF), a major regulator of angiogenesis, binds to two receptor tyrosine kinases, KDR/Flk-1 and Flt-1. We now describe the purification and the expression cloning from tumor cells of a third VEGF receptor, one that binds VEGF165 but not VEGF121. This isoform-specific VEGF receptor (VEGF165R) is identical to human neuropilin-1, a receptor for the collapsin/semaphorin family that mediates neuronal cell guidance. When coexpressed in cells with KDR, neuropilin-1 enhances the binding of VEGF165 to KDR and VEGF165-mediated chemotaxis. Conversely, inhibition of VEGF165 binding to neuropilin-1 inhibits its binding to KDR and its mitogenic activity for endothelial cells. We propose that neuropilin-1 is a novel VEGF receptor that modulates VEGF binding to KDR and subsequent bioactivity and therefore may regulate VEGF-induced angiogenesis.


Subject(s)
Endothelial Growth Factors/metabolism , Endothelium, Vascular/chemistry , Lymphokines/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Antigens, Surface/genetics , Antigens, Surface/metabolism , Cell Line/chemistry , Cell Line/cytology , Cell Line/metabolism , Chemotaxis/physiology , Cloning, Molecular , Endothelial Growth Factors/chemistry , Endothelium, Vascular/cytology , Exons/physiology , Gene Expression , Humans , Isomerism , Lymphokines/chemistry , Molecular Sequence Data , Neovascularization, Physiologic/physiology , Neuropilin-1 , RNA, Messenger/metabolism , Receptor Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/isolation & purification , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Growth Factor/genetics , Receptors, Growth Factor/isolation & purification , Receptors, Growth Factor/metabolism , Receptors, Mitogen/genetics , Receptors, Mitogen/isolation & purification , Receptors, Mitogen/metabolism , Receptors, Vascular Endothelial Growth Factor , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Umbilical Cord/cytology , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
11.
J Am Soc Nephrol ; 8(11): 1688-97, 1997 Nov.
Article in English | MEDLINE | ID: mdl-9355071

ABSTRACT

Proliferation of mesangial cells (MC) is a key feature in the pathogenesis of numerous renal diseases involving the glomerulus. Heparin, one of several compounds capable of suppressing MC proliferation, did not prove beneficial in the treatment of human glomerular diseases. In a search for a superior antiproliferative agent, a synthetic polyaromatic "heparin mimicking" compound (RG-13577, polymer of 4-hydroxyphenoxy acetic acid, M(r) approximately 5800), previously reported to inhibit the proliferation of vascular smooth muscle cells, was applied. RG-13577 exhibits approximately 1% of the anticoagulant activity of heparin and is nontoxic in animal experiments. Proliferation of primary rat MC was almost completely inhibited in the presence of 10 to 25 micrograms/ml RG-13577, and 50% inhibition was obtained at 1 to 5 micrograms/ml RG-13577. The cells resumed their normal growth rate after removal of RG-13577 from the culture medium. Under the same conditions, heparin exerted only a small inhibitors effect. RG-13577 inhibited signaling (i.e., tyrosine phosphorylation) and MC proliferation induced by both basic fibroblast growth factor and platelet-derived growth factor. RG-13577 binds to a naturally produced extracellular matrix, and the bound molecule retained its antiproliferative effect toward MC. 14C-Labeled RG-13577 also binds to cultured MC in a specific and saturable manner. Binding of 14C-RG-13577 was reduced by 80 to 90% in the presence of excess unlabeled RG-13577, apolipoprotein E, or lactoferrin, but there was no effect with heparin. Furthermore, the antiproliferative effect of RG-13577 was abolished in the presence of lactoferrin. It is proposed that compound RG-13577 inhibits MC proliferation through neutralization of growth-promoting factors, primarily heparin-binding growth factors, and possibly through binding to specific cell surface receptors, most likely the LDL receptor-related protein. RG-13577 and related polyanionic compounds may be applied to inhibit MC proliferation in glomerular diseases.


Subject(s)
Glomerular Mesangium/cytology , Glomerular Mesangium/drug effects , Phenoxyacetates/pharmacology , Polymers/pharmacology , Animals , Binding Sites , Cattle , Cell Division/drug effects , Cells, Cultured , Chemistry, Pharmaceutical , Extracellular Matrix/metabolism , Fibroblast Growth Factors/drug effects , Heparin/chemistry , Heparin/pharmacology , Humans , Molecular Weight , Phenoxyacetates/chemistry , Phosphorylation/drug effects , Polymers/chemistry , Rats , Rats, Wistar , Reference Values , Sensitivity and Specificity , Tyrosine/chemistry
12.
Circulation ; 95(7): 1853-62, 1997 Apr 01.
Article in English | MEDLINE | ID: mdl-9107173

ABSTRACT

BACKGROUND: Proliferation of smooth muscle cells (SMCs) of the arterial wall in response to local injury is an important factor in vascular proliferative disorders. Among the growth factors that promote SMC proliferation is basic fibroblast growth factor (bFGF), which is characterized by a high affinity for heparin and is associated with heparan sulfate on cell surfaces and extracellular matrices. We investigated whether heparin can displace endogenous active bFGF from the lumenal surface of blood vessels, whether bFGF is preferentially bound to injured blood vessels, and whether a synthetic, polyanionic, heparin-mimicking compound (RG-13577) can prevent sequestration of bFGF by the vessel wall. METHODS AND RESULTS: Injured and noninjured saphenous vein segments were perfused with or without heparin, in the absence or presence of 125I-bFGF and/or RG-13577 (a polymer of 4-hydroxyphenoxy acetic acid). Heparin displaced bFGF from the lumenal surface of the vein, and the released bFGF stimulated proliferation of SMCs. Likewise, systemic administration of heparin during open heart surgery resulted in a marked increase in plasma bFGF levels. Injured veins sequestered 125I-bFGF to a much higher extent than noninjured vein segments, both in the absence and presence of heparin. This sequestration was inhibited by compound RG-13577. CONCLUSIONS: Despite its beneficial effects, heparin may displace active bFGF, which subsequently may be preferentially deposited on injured vessel walls, thus contributing to the pathogenesis of restenosis. This effect may be prevented by a synthetic heparin-mimicking compound.


Subject(s)
Endothelium, Vascular/metabolism , Fibroblast Growth Factor 2/metabolism , Heparin/pharmacology , Receptors, Fibroblast Growth Factor/drug effects , Saphenous Vein/injuries , 3T3 Cells , Animals , Binding, Competitive , Cardiopulmonary Bypass , Cattle , Cell Division/drug effects , Fibroblast Growth Factor 2/pharmacology , Heparin Lyase , Heparitin Sulfate/metabolism , Humans , Mice , Muscle, Smooth, Vascular/pathology , Perfusion , Phenoxyacetates/pharmacology , Polymers/pharmacology , Polysaccharide-Lyases/pharmacology , Receptors, Fibroblast Growth Factor/metabolism , Recombinant Proteins/pharmacology , Saphenous Vein/drug effects , Saphenous Vein/metabolism
13.
J Clin Invest ; 99(7): 1565-75, 1997 Apr 01.
Article in English | MEDLINE | ID: mdl-9120000

ABSTRACT

Heparan sulfate (HS) proteoglycans play a key role in cell proliferation induced by basic fibroblast growth factor (FGF-2) and other heparin-binding growth factors. To modulate the involvement of HS, we have used a synthetic, nonsulfated polyanionic aromatic compound (RG-13577) that mimics functional features of heparin/HS. FGF-2-stimulated proliferation of vascular endothelial cells was markedly inhibited in the presence of 5-10 microg/ml compound RG-13577 (poly-4-hydroxyphenoxy acetic acid; Mr approximately 5 kD). Direct interaction between RG-13577 and FGF-2 was demonstrated by the ability of the former to compete with heparin on binding to FGF-2. RG-13577 inhibited FGF-2 binding to soluble- and cell surface-FGF receptor 1 (FGFR1). Unlike heparin, RG-13577 alone failed to mediate dimerization of FGF-2. Moreover, it abrogated heparin-mediated dimerization of FGF-2 and FGFR1, as well as FGF-2 mitogenic activity in HS-deficient F32 lymphoid cells. The antiproliferative effect of compound RG-13577 was associated with abrogation of FGF-2-induced tyrosine phosphorylation of FGFR1 and of cytoplasmic proteins involved in FGF-2 signal transduction, such as p90 and mitogen-activated protein kinase. A more effective inhibition of tyrosine phosphorylation was obtained after removal of the cell surface HS by heparinase. In contrast, tyrosine phosphorylation of an approximately 200-kD protein was stimulated by RG-13577, but not by heparin or FGF-2. RG-13577 prevented microvessel outgrowth from rat aortic rings embedded in a collagen gel. Development of nontoxic polyanionic compounds may provide an effective strategy to inhibit FGF-2-induced cell proliferation associated with angiogenesis, arteriosclerosis, and restenosis.


Subject(s)
Neovascularization, Physiologic/drug effects , Phenoxyacetates/pharmacology , Polymers/pharmacology , Receptor Protein-Tyrosine Kinases/drug effects , Receptors, Fibroblast Growth Factor/drug effects , 3T3 Cells , Animals , Cattle , Cells, Cultured , DNA/biosynthesis , Endothelium, Vascular/drug effects , Fibroblast Growth Factor 2/metabolism , Mice , Phosphorylation , Rats , Rats, Sprague-Dawley , Receptor Protein-Tyrosine Kinases/metabolism , Receptor, Fibroblast Growth Factor, Type 2 , Receptors, Fibroblast Growth Factor/metabolism , Tyrosine/metabolism
14.
Arterioscler Thromb Vasc Biol ; 17(1): 194-202, 1997 Jan.
Article in English | MEDLINE | ID: mdl-9012656

ABSTRACT

Proliferation of vascular smooth muscle cells (SMCs) and accumulation of extracellular matrix (ECM) components within the arterial wall in response to local injury are important etiologic factors in vascular proliferative disorders such as arteriosclerosis and restenosis after angioplasty. Fibrillar and nonfibrillar collagens are major constituents of the ECM that modulate cell shape and proliferative responses and thereby contribute to the pathogenesis of intimal hyperplasia. Halofuginone, an anticoccidial quinoazolinone derivative, inhibits collagen type I gene expression. We investigated the effect of halofuginone on (1) proliferation of bovine aortic endothelial cells and SMCs derived from the same specimen and maintained in vitro, (2) ECM deposition and collagen type I synthesis and gene expression, and (3) injury-induced intimal hyperplasia in vivo. DNA synthesis and proliferation of vascular SMCs in response to serum or basic fibroblast growth factor were abrogated in the presence of as little as 0.1 microgram/mL halofuginone; this inhibition was reversible upon removal of the compound. Under the same conditions, halofuginone exerted a relatively small antiproliferative effect on the respective vascular endothelial cells. Halofuginone also inhibited the synthesis and deposition of ECM components by vascular SMCs as indicated both by a substantial reduction in the amount of sulfated proteoglycans and collagen type I synthesis and gene expression. Local administration of halofuginone in the rabbit ear model of crush injury-induced arterial intimal hyperplasia resulted in a 50% reduction in intimal thickening as measured by a morphometric analysis of the neointima/media ratio. The differential inhibitory effect of halofuginone on vascular SMCs versus endothelial cells, its inhibition of ECM deposition and collagen type I synthesis, and its ability to attenuate injury-induced intimal hyperplasia may place halofuginone alone or in combination with other antiproliferative compounds as a potential candidate for prevention of arterial stenosis and accelerated atherosclerosis.


Subject(s)
Arteries/pathology , Collagen/biosynthesis , Muscle, Smooth, Vascular/drug effects , Quinazolines/pharmacology , Tunica Intima/pathology , Animals , Cattle , Cell Division/drug effects , Cells, Cultured , Collagen/drug effects , Humans , Hyperplasia , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Piperidines , Quinazolinones , Rabbits , Tunica Intima/drug effects , Tunica Intima/metabolism
15.
Kidney Int ; 52(6): 1561-9, 1997 Dec.
Article in English | MEDLINE | ID: mdl-9407501

ABSTRACT

Mesangial cell proliferation, increased deposition of collagen, and expansion of the mesangial extracellular matrix (ECM) are key features in the development of mesangioproliferative diseases. Halofuginone, a low molecular weight anti-coccidial quinoazolinone derivative, inhibits collagen type alpha 1(I) gene expression and synthesis. We investigated the effect of halofuginone on both normal and SV40 transformed mesangial cell proliferation, collagen synthesis, and ECM deposition. Proliferation of both cell types was almost completely inhibited in the presence of 50 ng/ml halofuginone. The cells were arrested in the late G1 phase of the cell cycle and resumed their normal growth rate following removal of the compound from the culture medium. The antiproliferative effect of halofuginone was associated with inhibition of tyrosine phosphorylation of cellular proteins. Similar results were obtained whether the mesangial cells were seeded on regular tissue culture plastic or in close contact with a naturally produced ECM resembling their local environment in vivo. Halofuginone also inhibited synthesis and deposition of ECM by mesangial cells as indicated by a substantial reduction in 14C-glycine and Na2(35)SO4 incorporation into the ECM, and by the inhibition of collagen type I synthesis and gene expression. It is proposed that by inhibiting collagen type I synthesis and matrix deposition, halofuginone exerts a potent antiproliferative effect that may be applied to inhibit mesangial cell proliferation and matrix expansion in a variety of chronic progressive glomerular diseases.


Subject(s)
Extracellular Matrix/metabolism , Glomerular Mesangium/cytology , Glomerular Mesangium/metabolism , Protein Synthesis Inhibitors/pharmacology , Quinazolines/pharmacology , Animals , Carbon Radioisotopes , Cell Division/drug effects , Cells, Cultured , Collagen/biosynthesis , Collagen/genetics , Collagen/metabolism , Extracellular Matrix/drug effects , Gene Expression/drug effects , Piperidines , Proline/metabolism , Proline/pharmacology , Quinazolinones , Rats , Rats, Wistar , Sulfates/metabolism , Sulfates/pharmacology , Tritium
16.
Proc Natl Acad Sci U S A ; 93(14): 7069-74, 1996 Jul 09.
Article in English | MEDLINE | ID: mdl-8692946

ABSTRACT

Recent studies have demonstrated the existence of a soluble fibroblast growth factor (FGF) receptor type 1 (FGFR1) extracellular domain in the circulation and in vascular basement membranes. However, the process of FGFR1 ectodomain release from the plasma membrane is not known. Here we report that the 72-kDa gelatinase A (matrix metalloproteinase type 2, MMP2) can hydrolyze the Val368-Met369 peptide bond of the FGFR1 ectodomain, eight amino acids upstream of the transmembrane domain, thus releasing the entire extracellular domain. Similar results were obtained regardless of whether FGF was first bound to the receptor or not. The action of MMP2 abolished binding of FGF to an immobilized recombinant FGFR1 ectodomain fusion protein and to Chinese hamster ovary cells overexpressing FGFR1 The released recombinant FGFR1 ectodomain was able to bind FGF after MMP2 cleavage, suggesting that the cleaved soluble receptor maintained its FGF binding capacity. The activity of MMP2 could not be reproduced by the 92-kDa gelatinase B (MMP9) and was inhibited by tissue inhibitor of metalloproteinase type 2. These studies demonstrate that FGFR1 may be a specific target for MMP2 on the cell surface, yielding a soluble FGF receptor that may modulate the mitogenic and angiogenic activities of FGF.


Subject(s)
Gelatinases/metabolism , Metalloendopeptidases/metabolism , Receptor Protein-Tyrosine Kinases , Receptors, Fibroblast Growth Factor/chemistry , Receptors, Fibroblast Growth Factor/metabolism , Amino Acid Sequence , Animals , CHO Cells , Cell Membrane/metabolism , Cricetinae , Enzyme Inhibitors/pharmacology , Fibroblast Growth Factor 1/metabolism , Fibroblast Growth Factor 2/metabolism , HeLa Cells , Humans , Matrix Metalloproteinase 2 , Methionine , Mice , Molecular Sequence Data , Proteins/pharmacology , Receptor, Fibroblast Growth Factor, Type 1 , Recombinant Fusion Proteins/metabolism , Recombinant Proteins/pharmacology , Substrate Specificity , Tissue Inhibitor of Metalloproteinase-2 , Transfection , Valine
17.
Cancer Metastasis Rev ; 15(2): 177-86, 1996 Jun.
Article in English | MEDLINE | ID: mdl-8842489

ABSTRACT

Heparan sulfate proteoglycans (HSPGs) are ubiquitous macromolecules associated with the cell surface and extracellular matrix (ECM) of a wide range of cells of vertebrate and invertebrate tissues [1, 2]. The basic HSPG structure consists of a protein core to which several linear heparan sulfate (HS) chains are covalently attached. The polysaccharide chains are typically composed of repeating hexuronic and D-glucosamine disaccharide units that are substituted to a varying extent with N- and O-linked sulfate moieties and N-linked acetyl groups [1, 2]. Beside serving as a scaffold for the attachment of various ECM components (e.g., collagen, laminin, fibronectin), the binding of HS to certain proteins has been suggested to induce a conformational change which may lead to the exposure of novel reactive determinants or conversely stabilize an inert protein configuration [1-4]. Of particular significance is the interaction of HS with fibroblast growth factors (FGFs), mediating their sequestration, stabilization and high affinity receptor binding and signaling [3-7]. Cellular responses to FGFs may hence be modulated by metabolic inhibitors of HS synthesis and sulfation, HS-degrading enzymes, and synthetic mimetics of heparin/HS. In the present review we focus on the involvement of HS in basic FGF (bFGF) receptor binding and mitogenic activity and its modulation by species of heparin, HS, and synthetic polyanionic 'heparin-mimicking' compounds. The results are discussed in relation to the current thoughts on the dual involvement of low and high affinity receptor sites in the growth promoting and angiogenic activities of bFGF and other heparin-binding growth factors.


Subject(s)
Fibroblast Growth Factor 2/physiology , Heparitin Sulfate/physiology , Animals , Cell Division/physiology , Extracellular Matrix/physiology , Heparin/physiology , Humans , Proteoglycans/physiology
18.
J Biol Chem ; 271(9): 4879-86, 1996 Mar 01.
Article in English | MEDLINE | ID: mdl-8617759

ABSTRACT

The growth promoting activity of the subendothelial extracellular matrix (ECM) is attributed to sequestration of basic fibroblast growth factor (bFGF) by heparan sulfate proteoglycans and its regulated release by heparin-like molecules and heparan sulfate (HS) degrading enzymes. HS is also involved in bFGF receptor binding and activation. The present study focuses on the growth promoting activity and bFGF binding capacity of sulfate-depleted ECM. Corneal endothelial cells (EC) maintained in the presence of chlorate, an inhibitor of phosphoadenosine phosphosulfate synthesis, produced ECM containing 10-15% of the sulfate normally present in ECM. Incorporation of sulfate into HS was reduced by more than 90%. Binding of 125I-bFGF to sulfate-depleted ECM was reduced by 50-60% and only about 10% of the ECM-bound bFGF was accessible to release by heparin. Incubation of 125I-bFGF on top of native ECM resulted in dimerization of the ECM-bound bFGF, but there was a markedly reduced binding and dimerization of bFGF on sulfate-depleted ECM. ECM produced in the presence of chlorate contained a nearly 10-fold less endogenous bFGF as compared to native ECM and exerted little or no mitogenic activity toward vascular EC and 3T3 fibroblasts. In other studies, we investigated the interaction between chlorate-treated vascular EC and either native or sulfate-depleted ECM. Exogenous heparin stimulated the proliferation of chlorate-treated EC seeded on native ECM, suggesting its interaction with ECM-bound bFGF and subsequent presentation to high affinity cell surface receptors. On the other hand, heparin had no effect on chlorate-treated cells seeded in contact with sulfate-depleted ECM or regular tissue culture plastic. Altogether, the present experiments indicate that heparan sulfate proteoglycans associated with the cell surface and ECM act in concert to regulate the bioavailability and growth promoting activity of bFGF. While HS in the subendothelial ECM functions primarily in sequestration of bFGF in the vicinity of responsive cells, HS on cell surfaces is playing a more active role in displacing the ECM-bound bFGF and its subsequent presentation to high affinity signal transducing receptors.


Subject(s)
Cell Division/drug effects , Endothelium, Corneal/physiology , Endothelium, Vascular/metabolism , Fibroblast Growth Factor 2/metabolism , Fibroblast Growth Factor 2/pharmacology , 3T3 Cells , Animals , Cattle , Endothelium, Corneal/cytology , Endothelium, Corneal/drug effects , Extracellular Matrix/physiology , Fibroblast Growth Factor 2/chemistry , Heparin/pharmacology , Humans , Iodine Radioisotopes , Macromolecular Substances , Mice , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Signal Transduction , Sulfates/metabolism , Sulfur Radioisotopes
19.
J Autoimmun ; 8(5): 741-50, 1995 Oct.
Article in English | MEDLINE | ID: mdl-8579728

ABSTRACT

The extravasation of activated T lymphocytes through blood vessel walls and their migration to inflammatory loci are associated with secretion of extracellular matrix (ECM)-degrading enzymes, such as heparanase, which degrades heparan sulfate (HS) moieties of the ECM. The HS-degrading activity of heparanase was found to be inhibited by HS and heparin. Since induction of experimental autoimmune encephalomyelitis (EAE) requires extravasation and migration of autoimmune T cells, degradation of ECM by heparanase is expected to be involved in induction of the disease. Herein, we examined whether laminarin sulfate, a polysulfated polysaccharide (PSS) isolated from the cell walls of seaweeds and subjected to chemical sulfation, could inhibit ECM degradation by mammalian heparanase, and could prevent EAE. PSS was a more potent inhibitor of heparanase-mediated degradation of ECM than heparin. In-vivo, PSS, injected once a week, inhibited the severity of actively-induced EAE in rats. However, inhibition of EAE was not due to an overall suppression of autoimmune T cells, since PSS enhanced the proliferation of myelin basic protein (MBP)-specific, encephalitogenic T cells. PSS-activated autoimmune T cells, but not MBP-activated cells, failed to induce EAE in recipient rats. Moreover, rats injected with PSS-activated T cells were resistant to induction of EAE by anti-MBP CD4+ T cells. Thus, PSS may have potential clinical applications in the treatment of autoimmune diseases.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/drug therapy , Glucuronidase , Glycoside Hydrolases/antagonists & inhibitors , Lymphocyte Activation/drug effects , Polysaccharides/therapeutic use , T-Lymphocytes/immunology , Animals , Encephalomyelitis, Autoimmune, Experimental/enzymology , Encephalomyelitis, Autoimmune, Experimental/immunology , Female , Rats , Rats, Inbred F344 , Rats, Inbred Lew , Species Specificity , T-Lymphocytes/drug effects , T-Lymphocytes/enzymology
20.
J Cell Physiol ; 164(3): 482-90, 1995 Sep.
Article in English | MEDLINE | ID: mdl-7650058

ABSTRACT

Heparin and heparin-like molecules may function, apart from their effect on hemostasis, as regulators of cell growth and neovascularization. We investigated whether similar effects are exerted by laminarin sulfate, an unrelated polysulfated saccharide isolated from the cell wall of seaweed and composed of chemically O-sulfated beta-(1,3)-linked glucose residues. Laminarin sulfate exhibits about 30% of the anticoagulant activity of heparin and is effective therapeutically in the prevention and treatment of cerebrovascular diseases. We characterized the effect of laminarin sulfate on interaction of the heparin-binding angiogenic factor, basic fibroblast growth factor (bFGF), with a naturally produced subendothelial extra-cellular matrix (ECM) and with cell surface receptor sites. Laminarin sulfate (1-2 micrograms/ml) inhibited the binding of bFGF to ECM and to the surface of vascular smooth muscle cells (SMC) in a manner similar to that observed with heparin. Likewise, laminarin sulfate efficiently displaced both ECM- and cell-bound bFGF at concentrations as low as 1 microgram/ml. Both laminarin sulfate and heparin efficiently induced restoration of bFGF receptor binding in xylosyltransferase-deficient CHO cell mutants defective in initiation of glycosaminoglycan synthesis. Moreover, laminarin sulfate elicited bFGF receptor activation and mitogenic response in heparan sulfate (HS)-deficient, cytokine-dependent lymphoid cells. These results indicate that laminarin sulfate effectively replaced the need for heparin and HS in the induction of bFGF receptor binding and signaling. In other experiments, laminarin sulfate was found to inhibit the proliferation of vascular SMC in a manner similar to that observed with heparin. These effects of laminarin sulfate may have potential clinical applications in diverse situations such as wound healing, angiogenesis, and atherosclerosis.


Subject(s)
Cell Division/drug effects , Fibroblast Growth Factor 2/pharmacology , Growth Inhibitors , Heparin/pharmacology , Muscle, Smooth, Vascular/cytology , Polysaccharides/pharmacology , Receptors, Fibroblast Growth Factor/metabolism , Animals , Cattle , Cells, Cultured , Extracellular Matrix/metabolism , Fibroblast Growth Factor 2/metabolism , In Vitro Techniques
SELECTION OF CITATIONS
SEARCH DETAIL
...