Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
J Bone Miner Res ; 17(7): 1180-9, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12102052

ABSTRACT

Decorin (dcn) and biglycan (bgn), two members of the family of small leucine-rich proteoglycans (SLRPs), are the predominant proteoglycans expressed in skin and bone, respectively. Targeted disruption of the dcn gene results in skin laxity and fragility, whereas disruption of the bgn gene results in reduced skeletal growth and bone mass leading to generalized osteopenia, particularly in older animals. Here, we report that bgn deficiency leads to structural abnormality in collagen fibrils in bone, dermis, and tendon, and to a "subclinical" cutaneous phenotype with thinning of the dermis but without overt skin fragility. A comparative ultrastructural study of different tissues from bgn- and dcn-deficient mice revealed that bgn and dcn deficiency have similar effects on collagen fibril structure in the dermis but not in bone. Ultrastructural and phenotypic analysis of newly generated bgn/dcn double-knockout (KO) mice revealed that the effects of dcn and bgn deficiency are additive in the dermis and synergistic in bone. Severe skin fragility and marked osteopenia characterize the phenotype of double-KO animals in which progeroid changes are observed also in the skin. Ultrastructural analysis of bone collagen fibrils in bone of double-KO mice reveals a complete loss of the basic fibril geometry with the emergence of marked "serrated fibril" morphology. The phenotype of the double-KO animal mimics directly the rare progeroid variant of human Ehlers-Danlos syndrome (EDS), in which skin fragility, progeroid changes in the skin (reduced hypodermis), and osteopenia concur as a result of impaired glycosaminoglycan (GAG) linking to bgn and dcn core proteins. Our data show that changes in collagen fibril morphology reminiscent of those occurring in the varied spectrum of human EDS are induced by both bgn deficiency and den deficiency in mice. The effects of an individual SLRP deficiency are tissue specific, and the expression of a gross phenotype depends on multiple variables including level of expression of individual SLRPs in different tissues and synergisms between different SLRPs (and likely other macromolecules) in determining matrix structure and functional properties.


Subject(s)
Bone and Bones/pathology , Collagen/metabolism , Connective Tissue/pathology , Ehlers-Danlos Syndrome/pathology , Proteoglycans/physiology , Animals , Biglycan , Decorin , Ehlers-Danlos Syndrome/etiology , Extracellular Matrix Proteins , Male , Mice , Mice, Knockout , Phenotype , Proteoglycans/genetics , Skin/pathology
2.
Biochem J ; 359(Pt 1): 153-63, 2001 Oct 01.
Article in English | MEDLINE | ID: mdl-11563979

ABSTRACT

Perlecan, a widespread heparan sulphate (HS) proteoglycan, is directly involved in the storing of angiogenic growth factors, mostly members of the fibroblast growth factor (FGF) gene family. We have previously shown that antisense targeting of the perlecan gene causes a reduced growth and responsiveness to FGF7 [also known as keratinocyte growth factor (KGF)] in human cancer cells, and that the perlecan protein core interacts specifically with FGF7. In the present paper, we have investigated human colon carcinoma cells in which the perlecan gene was disrupted by targeted homologous recombination. After screening over 1000 clones, we obtained two clones heterozygous for the null mutation with no detectable perlecan, indicating that the other allele was non-functioning. The perlecan-deficient cells grew more slowly, did not respond to FGF7 with or without the addition of heparin, and were less tumorigenic than control cells. Paradoxically, the perlecan-deficient cells displayed increased FGF7 surface binding. However, the perlecan protein core was required for functional activation of the KGF receptor and downstream signalling. Because heparin could not substitute for perlecan, the HS chains are not critical for FGF7-mediated signalling in this cell system. These results provide the first genetic evidence that the perlecan protein core is a molecular entity implicated in FGF7 binding and activation of its receptor.


Subject(s)
Colonic Neoplasms/metabolism , Fibroblast Growth Factors/metabolism , Heparan Sulfate Proteoglycans/physiology , Neoplasms, Experimental/pathology , Receptors, Fibroblast Growth Factor/metabolism , Animals , Blotting, Northern , Blotting, Western , Cell Division/drug effects , DNA, Antisense/pharmacology , Fibrinolytic Agents/pharmacology , Fibroblast Growth Factor 1/metabolism , Fibroblast Growth Factor 7 , Gene Targeting , Heparin/pharmacology , Humans , Mice , Mice, Nude , Neoplasms, Experimental/metabolism , Protein Binding , Receptor, Fibroblast Growth Factor, Type 2 , Recombinant Proteins/metabolism , Signal Transduction , Transplantation, Heterologous , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/metabolism , Tumor Cells, Cultured/transplantation
3.
J Biol Chem ; 275(45): 35153-61, 2000 Nov 10.
Article in English | MEDLINE | ID: mdl-10942781

ABSTRACT

The leucine-rich proteoglycan decorin interacts with the epidermal growth factor receptor and triggers a signaling pathway that leads to growth suppression. We find that decorin causes a functional inactivation of the oncogenic ErbB2 protein in breast carcinoma cells. Upon de novo expression of decorin, the ErbB2 protein is reduced by approximately 40%, whereas its degree of tyrosyl phosphorylation is almost completely abrogated. Both co-culture experiments or experiments with recombinant decorin demonstrate an initial induction of ErbB2 tyrosine kinase, followed by a profound and long-lasting down-regulation of its activity. This leads to growth inhibition and cytodifferentiation of mammary tumor cells and a concurrent suppression of their tumorigenic potential in vivo. These decorin-mediated effects appear to involve the activation of ErbB4, which in turn would block the phosphorylation of heterodimers containing either ErbB2 or ErbB3. These results provide an explanation for the heightened decorin levels around invasive carcinomas and suggest that decorin may function as a natural antagonist of neoplastic cells enriched in ErbB2.


Subject(s)
Down-Regulation , Proteoglycans/physiology , Receptor, ErbB-2/metabolism , Animals , Blotting, Northern , Blotting, Western , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Cell Adhesion , Cell Cycle , Cell Differentiation , Cell Division , Cell Separation , Coculture Techniques , DNA, Complementary/metabolism , Decorin , Dose-Response Relationship, Drug , Enzyme Activation , ErbB Receptors/metabolism , Extracellular Matrix Proteins , Flow Cytometry , Humans , Immunohistochemistry , Kinetics , Leucine/chemistry , Mice , Phenotype , Phosphorylation , Precipitin Tests , Proteoglycans/pharmacology , Receptor, ErbB-3/metabolism , Receptor, ErbB-4 , Recombinant Proteins/metabolism , Signal Transduction , Time Factors , Transfection , Tumor Cells, Cultured , Tyrosine/metabolism
4.
J Biol Chem ; 275(42): 32879-87, 2000 Oct 20.
Article in English | MEDLINE | ID: mdl-10913155

ABSTRACT

The small leucine-rich proteoglycan decorin interacts with the epidermal growth factor receptor (EGFR) and triggers a signaling cascade that leads to elevation of endogenous p21 and growth suppression. We demonstrate that decorin causes a sustained down-regulation of the EGFR. Upon stable expression of decorin, the EGFR number is reduced by approximately 40%, without changes in EGFR expression. However, EGFR phosphorylation is nearly completely abolished. Concurrently, decorin attenuates the EGFR-mediated mobilization of intracellular calcium and blocks the growth of tumor xenografts by down-regulating the EGFR kinase in vivo. Thus, decorin acts as an autocrine and paracrine regulator of tumor growth and could be utilized as an effective anti-cancer agent.


Subject(s)
Calcium Signaling/physiology , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/therapy , Down-Regulation/physiology , ErbB Receptors/genetics , Proteoglycans/physiology , Adenosine Triphosphate/pharmacology , Animals , Calcium/metabolism , Calcium Signaling/drug effects , Cell Division , Decorin , Down-Regulation/drug effects , Epidermal Growth Factor/pharmacology , ErbB Receptors/metabolism , Extracellular Matrix Proteins , Female , Humans , Mice , Mice, Nude , Phosphorylation , Proteoglycans/genetics , Proteoglycans/pharmacology , Recombinant Proteins/pharmacology , Transfection , Transforming Growth Factor beta/antagonists & inhibitors , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
5.
Lab Invest ; 80(12): 1869-80, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11140699

ABSTRACT

Decorin is a small leucine-rich proteoglycan that interacts with several matrix molecules, including various types of collagen and growth factors, and suppresses the growth of neoplastic cells by an epidermal growth factor (EGF) receptor-mediated pathway. Decorin is abundantly expressed in the periodontal connective tissues during development and tissue maintenance. In periodontal disease, which is one of the most common diseases in the human kind, the level of decorin is decreased in the periodontal connective tissue. Abnormal expression of decorin may also associate with certain inherited disorders that involve increased susceptibility to severe periodontal disease in the early childhood. Therefore, we investigated the periodontal tissues of mice with targeted disruption of the decorin gene. Gross and microscopic analyses showed that decorin-deficient mice appeared to have normal tooth development and eruption, and there were no signs of periodontal disease. However, electron microscopic analysis revealed abnormal morphology and organization of the collagen fibrils in the periodontal ligament. The number of periodontal ligament fibroblasts in the decorin-deficient mice was also increased about two-fold as compared with the wild-type mice. In cell culture, ectopic overexpression of decorin in NIH 3T3 fibroblasts or decorin added exogenously to periodontal fibroblasts suppressed cell growth. However, blocking the EGF receptor tyrosine kinase activity did not prevent the decorin-elicited growth suppression in periodontal fibroblasts. Additionally, decorin did not induce a marked increase in the relative expression of p21 mRNA in periodontal fibroblasts. Therefore, decorin appeared to regulate growth of normal periodontal fibroblasts by a mechanism distinct from that reported for neoplastic cells. The findings demonstrate that decorin plays a role in the organization of collagen fibrils and regulates cell proliferation in the periodontal ligament.


Subject(s)
Aging/physiology , Periodontal Ligament/cytology , Periodontal Ligament/physiology , Proteoglycans/physiology , 3T3 Cells , Animals , Cell Division/drug effects , Decorin , Enzyme Inhibitors/pharmacology , Extracellular Matrix Proteins , Female , Male , Mice , Mice, Knockout , Microscopy, Electron , Microscopy, Electron, Scanning , Periodontal Ligament/ultrastructure , Proteoglycans/deficiency , Proteoglycans/genetics , Quinazolines , Recombinant Proteins/metabolism , Transfection , Transforming Growth Factor beta/antagonists & inhibitors , Tyrphostins/pharmacology
6.
Proc Natl Acad Sci U S A ; 96(6): 3092-7, 1999 Mar 16.
Article in English | MEDLINE | ID: mdl-10077642

ABSTRACT

Ectopic expression of decorin in a wide variety of transformed cells results in growth arrest and the inability to generate tumors in nude mice. This process is caused by a decorin-mediated activation of the epidermal growth factor receptor, which leads to a sustained induction of endogenous p21(WAF1/CIP1) (the cyclin-dependent kinase inhibitor p21) and growth arrest. However, mice harboring a targeted disruption of the decorin gene do not develop spontaneous tumors. To test the role of decorin in tumorigenesis, we generated mice lacking both decorin and p53, an established tumor-suppressor gene. Mice lacking both genes showed a faster rate of tumor development and succumbed almost uniformly to thymic lymphomas within 6 months [mean survival age (T50) approximately 4 months]. Mice harboring one decorin allele and no p53 gene developed the same spectrum of tumors as the double knockout animals, but had a survival rate similar to the p53 null animals (T50 approximately 6 months). Ectopic expression of decorin in thymic lymphoma cells isolated from double mutant animals markedly suppressed their colony-forming ability. When these lymphoma cells were cocultured with fibroblasts derived from either wild-type or decorin null embryos, the cells grew faster in the absence of decorin. Moreover, exogenous decorin proteoglycan or its protein core significantly retarded their growth in vitro. These results indicate that the lack of decorin is permissive for lymphoma tumorigenesis in a mouse model predisposed to cancer and suggest that germ-line mutations in decorin and p53 may cooperate in the transformation of lymphocytes and ultimately lead to a more aggressive phenotype by shortening the tumor latency.


Subject(s)
Gene Expression Regulation, Neoplastic , Lymphoma, T-Cell/genetics , Lymphoma, T-Cell/pathology , Proteoglycans/genetics , Tumor Suppressor Protein p53/genetics , Animals , Cell Division/genetics , Cell Line , Decorin , Extracellular Matrix Proteins , Germ-Line Mutation , Mice , Mice, Knockout , Neoplasms, Experimental/genetics , Neoplasms, Experimental/pathology
7.
J Biol Chem ; 274(8): 4489-92, 1999 Feb 19.
Article in English | MEDLINE | ID: mdl-9988678

ABSTRACT

Ectopic expression of decorin induces profound cytostatic effects in transformed cells with diverse histogenetic backgrounds. The mechanism of action has only recently begun to be elucidated. Exogenous decorin activates the epidermal growth factor (EGF) receptor, thereby triggering a signaling cascade that leads to phosphorylation of mitogen-activated protein (MAP) kinase, induction of p21, and growth suppression. In this study we demonstrate a direct interaction of decorin with the EGF receptor. Binding of decorin induces dimerization of the EGF receptor and rapid and sustained phosphorylation of MAP kinase in squamous carcinoma cells. In a cell-free system, decorin induces autophosphorylation of purified EGF receptor by activating the receptor tyrosine kinase and can also act as a substrate for the EGF receptor kinase itself. Using radioligand binding assays we show that both immobilized and soluble decorin bind to the EGF receptor ectodomain or to purified EGF receptor. The binding is mediated by the protein core and has relatively low affinity (Kd approximately 87 nM). Thus, decorin should be considered as a novel biological ligand for the EGF receptor, an interaction that could regulate cell growth during remodeling and cancer growth.


Subject(s)
ErbB Receptors/metabolism , Proteoglycans/metabolism , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Decorin , Dimerization , Enzyme Activation , Extracellular Matrix Proteins , Ligands , Phosphorylation , Protein Binding , Tumor Cells, Cultured
8.
J Clin Invest ; 102(8): 1599-608, 1998 Oct 15.
Article in English | MEDLINE | ID: mdl-9788974

ABSTRACT

Perlecan, a ubiquitous heparan sulfate proteoglycan, possesses angiogenic and growth-promoting attributes primarily by acting as a coreceptor for basic fibroblast growth factor (FGF-2). In this report we blocked perlecan expression by using either constitutive CMV-driven or doxycycline- inducible antisense constructs. Growth of colon carcinoma cells was markedly attenuated upon obliteration of perlecan gene expression and these effects correlated with reduced responsiveness to and affinity for mitogenic keratinocyte growth factor (FGF-7). Exogenous perlecan effectively reconstituted the activity of FGF-7 in the perlecan-deficient cells. Moreover, soluble FGF-7 specifically bound immobilized perlecan in a heparan sulfate-independent manner. In both tumor xenografts induced by human colon carcinoma cells and tumor allografts induced by highly invasive mouse melanoma cells, perlecan suppression caused substantial inhibition of tumor growth and neovascularization. Thus, perlecan is a potent inducer of tumor growth and angiogenesis in vivo and therapeutic interventions targeting this key modulator of tumor progression may improve cancer treatment.


Subject(s)
DNA, Antisense/therapeutic use , Fibroblast Growth Factors , Heparan Sulfate Proteoglycans , Heparitin Sulfate/genetics , Neoplasms, Experimental/drug therapy , Neovascularization, Pathologic/drug therapy , Proteoglycans/genetics , Animals , Carcinoma/drug therapy , Colonic Neoplasms/drug therapy , Fibroblast Growth Factor 10 , Fibroblast Growth Factor 2/metabolism , Fibroblast Growth Factor 7 , Gene Expression , Growth Substances/metabolism , Heparitin Sulfate/biosynthesis , Heparitin Sulfate/metabolism , Humans , Melanoma, Experimental/drug therapy , Mice , Neoplasm Transplantation , Protein Binding , Proteoglycans/biosynthesis
9.
Cancer Res ; 55(16): 3495-9, 1995 Aug 15.
Article in English | MEDLINE | ID: mdl-7627953

ABSTRACT

The Wnt-5A gene codes for a secreted cysteine-rich growth factor that mediates cell to cell signaling via a paracrine mechanism during development and ontogeny. We have recently determine the genomic organization and chromosomal mapping of the human Wnt-5A, and observed distinct patterns of expression in developing human embryos. In this report, we have performed a detailed expression analysis of 100 adult human tissues and tumors and 10 human cell lines. Our data show a widespread expression of Wnt-5A in adult tissues and cells, and aberrant mRNA levels in lungs, breast, and prostate carcinomas and in melanomas. The up-regulation of Wnt-5A in human malignancy was not due to either gene rearrangement or amplification. These findings document an abnormal expression of this growth factor in malignancy and implicate Wnt-5A in the genesis of human cancer.


Subject(s)
Gene Expression Regulation, Neoplastic , Growth Substances/genetics , Proto-Oncogene Proteins/genetics , Genes , Humans , In Vitro Techniques , RNA, Messenger/genetics , RNA, Neoplasm/genetics , Tissue Distribution , Tumor Cells, Cultured , Wnt Proteins , Wnt-5a Protein
10.
Mol Cell Biochem ; 145(1): 61-8, 1995 Apr 12.
Article in English | MEDLINE | ID: mdl-7544867

ABSTRACT

Perlecan is a modular heparan sulfate proteoglycan that harbors five domains with homology to the low density lipoprotein receptor, epidermal growth factor, laminin and neural cell adhesion molecule. Using a monoclonal antibody directed against the laminin-like domain of perlecan, we have recently shown that perlecan is widely expressed in all lymphoreticular systems. To investigate further this observation we have studied the expression of perlecan in two human leukemic cell lines. Using reverse transcriptase-PCR, ribonuclease protection assay, and metabolic labeling we detected significant perlecan expression in the multipotential cell line K562, originally derived from a patient with chronic myelogenous leukemia. In contrast, the promyelocytic cell line HL-60 expressed perlecan at barely detectable levels. These results were intriguing because the K562 cells do not assemble or produce a classical basement membrane. Following induction with either sodium butyrate or the phorbol diester 12-0-tetradecanoylphorbol-13-acetate (TPA), K562 and HL-60 differentiate into early progenitor cells with erythroid or megakaryocytic properties, respectively. Following treatment of K562 and HL-60 cells with either of these agents, perlecan expression was markedly increased in K562 cells. In contrast, we could detect perlecan protein synthesis in HL-60 cells only at very low levels, even after induction with TPA or sodium butyrate. Collectively, these results indicate that perlecan is actively synthesized by bone marrow derived cells and suggest that this proteoglycan may play a role in hematopoietic cell differentiation.


Subject(s)
Butyrates/pharmacology , Heparan Sulfate Proteoglycans , Heparitin Sulfate/biosynthesis , Leukemia, Erythroblastic, Acute/metabolism , Proteoglycans/biosynthesis , Tetradecanoylphorbol Acetate/pharmacology , Up-Regulation , Base Sequence , Butyric Acid , Cell Differentiation , Chondroitinases and Chondroitin Lyases/metabolism , Glyceraldehyde-3-Phosphate Dehydrogenases/biosynthesis , Glyceraldehyde-3-Phosphate Dehydrogenases/genetics , Heparitin Sulfate/genetics , Heparitin Sulfate/metabolism , Humans , Immunoblotting , Leukemia, Promyelocytic, Acute/metabolism , Molecular Sequence Data , Polysaccharide-Lyases/metabolism , Proteoglycans/chemistry , Proteoglycans/genetics , Proteoglycans/metabolism , RNA/analysis , Tumor Cells, Cultured
11.
Genomics ; 18(2): 249-60, 1993 Nov.
Article in English | MEDLINE | ID: mdl-8288227

ABSTRACT

The highly conserved Wnt genes belong to a widely distributed family of presumptive signaling molecules that have been implicated not only in the regulation of normal pattern formation during embryogenesis and differentiation of cell lineages, but also in oncogenic events. All of the known vertebrate Wnt genes encode for 38- to 43-kDa cysteine-rich putative glycoproteins, which have features typical of secreted growth factors: a hydrophobic signal sequence, a conserved asparagine-linked oligosaccharide consensus sequence, and 22 conserved cysteine residues whose relative spacing is maintained. In this study, we report the cloning and sequencing of several overlapping cDNAs encoding approximately 4.1 kb of the human homologue of Wnt-5A. The mature protein contained 343 residues (M(r) approximately 38,000 excluding any post-translational modifications) with a > 93% homology to the reported sequences of other Wnt-5A proteins (> 99% homologous to mouse Wnt-5A). This protein maintained certain features--a hydrophobic signal sequence, the Wnt-1 family "signature sequence" (CKCHGvSGSC), and a number of other conserved amino acid residues: 24 cysteine residues, 4 asparagine-linked oligosaccharide consensus sequences, and a tyrosine sulfation site--that have been found in all other Wnt-5A proteins. Reverse transcriptase PCR analysis of RNA from a variety of human embryonic, neonatal, and adult cells and/or tissues showed that human Wnt-5A expression was detected only in neonatal heart and lung. It may be relevant, however, that the 3'-untranslated region contained numerous AT-rich motifs that could be involved in the rapid degradation of mRNA. Finally, using a combination of Southern blotting, PCR amplification, and in situ hybridization, the human Wnt-5A (WNT5A) gene was mapped to chromosome 3p14-p21.


Subject(s)
Chromosomes, Human, Pair 3 , Proto-Oncogene Proteins/genetics , Proto-Oncogenes , Amino Acid Sequence , Animals , Base Sequence , Chromosome Mapping , Cloning, Molecular , DNA , Humans , Hybrid Cells , Molecular Sequence Data , Polymerase Chain Reaction , Proto-Oncogene Mas , Sequence Homology, Amino Acid , Wnt Proteins , Wnt-5a Protein
12.
Genomics ; 15(1): 146-60, 1993 Jan.
Article in English | MEDLINE | ID: mdl-8432526

ABSTRACT

Decorin is a chondroitin/dermatan sulfate proteoglycan expressed by most vascular and avascular connective tissues and, because of its ability to interact with collagen and growth factors, has been implicated in the control of matrix assembly and cellular growth. To understand the molecular mechanisms involved in regulating its tissue expression, we have isolated a number of genomic clones encoding the complete decorin gene. The human decorin gene spans over 38 kb of continuous DNA sequence and contains eight exons and very large introns, two of which are 5.4 and > 13.2 kb. We have discovered two alternatively spliced leader exons, exons Ia and Ib, in the 5' untranslated region. These exons were identified by cloning and sequencing cDNAs obtained by polymerase chain reaction amplification of a fibroblast cDNA library. Using Northern blotting or reverse transcriptase PCR, we detected the two leader exons in a variety of mRNAs isolated from human cell lines and tissues. Interestingly, sequences highly (74-87%) homologous to exons Ia and Ib are found in the 5' untranslated region of avian and bovine decorin, respectively. This high degree of conservation among species suggests regulatory functions for these leader exons. In the 3' untranslated region there are several polyadenylation sites, and at least two of these sites could give rise to the transcripts of approximately 1.6 and approximately 1.9 kb, typically detected in a variety of tissues and cells. Using a genomic clone as the labeled probe and in situ hybridization of human metaphase chromosomes, we have mapped the decorin gene to the discrete region of human chromosome 12q23. This study provides the molecular basis for discerning the transcriptional control of the decorin gene and offers the opportunity to investigate genetic disorders linked to this important human gene.


Subject(s)
Alternative Splicing , Chromosomes, Human, Pair 12 , Exons , Introns , Proteoglycans/genetics , Amino Acid Sequence , Animals , Base Sequence , Blotting, Northern , Cattle , Chickens , Chromosome Mapping , Cloning, Molecular , Cricetinae , DNA , Decorin , Extracellular Matrix Proteins , Humans , Hybrid Cells , In Situ Hybridization , Molecular Sequence Data , Poly A/metabolism , Polymerase Chain Reaction , Promoter Regions, Genetic , Sequence Homology, Nucleic Acid
SELECTION OF CITATIONS
SEARCH DETAIL
...