51
|
Song Y, Nestor KE, McFarland DC, Velleman SG. Effect of glypican-1 covalently attached chains on turkey myogenic satellite cell proliferation, differentiation, and fibroblast growth factor 2 responsiveness. Poult Sci 2010; 89:123-34. [PMID: 20008810 DOI: 10.3382/ps.2009-00325] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Glypican-1 is a cell membrane heparan sulfate proteoglycan that is composed of a core protein and covalently attached glycosaminoglycan (GAG) chains and N-linked glycosylated (N-glycosylated) chains. The glypican-1 GAG chains are required for cell differentiation and responsiveness to fibroblast growth factor 2 (FGF2). The role of glypican-1 N-glycosylated chains in regulating cell activities has not been reported. The objective of the current study was to investigate the role of glypican-1 N-glycosylated chains and the interaction between N-glycosylated and GAG chains in turkey myogenic satellite cell proliferation, differentiation, and FGF2 responsiveness. The wild-type turkey glypican-1 and turkey glypican-1 with mutated GAG chain attachment sites were cloned into the pCMS-EGFP mammalian expression vector and were used as templates to generate glypican-1 N-glycosylated 1-chain and no-chain mutants with or without GAG chains by site-directed mutagenesis. The wild-type glypican-1 and all glypican-1 N-glycosylated 1-chain and no-chain mutants with or without GAG chains were transfected into turkey myogenic satellite cells. Cell proliferation, differentiation, and FGF2 responsiveness were measured. The overexpression of glypican-1 N-glycosylated 1-chain and no-chain mutants without GAG chains increased cell proliferation and differentiation compared with the wild-type glypican-1 but not the glypican-1 N-glycosylated mutants with GAG chains attached. Cells overexpressing glypican-1 N-glycosylated mutants with or without GAG chains increased cell responsiveness to FGF2 compared with wild-type glypican-1. These data suggest that glypican-1 N-glycosylated chains and GAG chains are critical in regulating turkey myogenic satellite cell proliferation, differentiation, and responsivness to FGF2.
Collapse
Affiliation(s)
- Y Song
- Department of Animal Sciences, Ohio Agricultural Research and Development Center, The Ohio State University, Wooster 44691, USA
| | | | | | | |
Collapse
|
52
|
Friedl A. Proteoglycans: master modulators of paracrine fibroblast-carcinoma cell interactions. Semin Cell Dev Biol 2009; 21:66-71. [PMID: 19931629 DOI: 10.1016/j.semcdb.2009.11.013] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2009] [Accepted: 11/13/2009] [Indexed: 11/19/2022]
Abstract
Reciprocal interactions between tumor and stromal cells govern carcinoma growth and progression. Signaling functions between these cell types in the tumor microenvironment are largely carried out by secreted growth factors and cytokines. This review discusses how proteoglycans, which are abundantly present in normal and neoplastic tissues, modulate paracrine growth factor signaling events. General principles of proteoglycan involvement in paracrine signaling include stromal induction, core protein processing by proteases and growth factor binding via proteoglycan glycosaminoglycan chains or core protein domains.
Collapse
Affiliation(s)
- Andreas Friedl
- Department of Pathology and Laboratory Medicine, 6051 Wisconsin Institutes for Medical Research, University of Wisconsin-Madison, 1111 Highland Ave, Madison, WI 53705-2275, United States.
| |
Collapse
|
53
|
Ng A, Wong M, Viviano B, Erlich JM, Alba G, Pflederer C, Jay PY, Saunders S. Loss of glypican-3 function causes growth factor-dependent defects in cardiac and coronary vascular development. Dev Biol 2009; 335:208-15. [PMID: 19733558 PMCID: PMC2763964 DOI: 10.1016/j.ydbio.2009.08.029] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2009] [Revised: 08/17/2009] [Accepted: 08/31/2009] [Indexed: 12/13/2022]
Abstract
Glypican-3 (Gpc3) is a heparan sulfate proteoglycan (HSPG) expressed widely during vertebrate development. Loss-of-function mutations cause Simpson-Golabi-Behmel syndrome (SGBS), a rare and complex congenital overgrowth syndrome with a number of associated developmental abnormalities including congenital heart disease. We found that Gpc3-deficient mice display a high incidence of congenital cardiac malformations like ventricular septal defects, common atrioventricular canal and double outlet right ventricle. In addition we observed coronary artery fistulas, which have not been previously reported in SGBS. Coronary artery fistulas are noteworthy because little is known about the molecular basis of this abnormality. Formation of the coronary vascular plexus in Gpc3-deficient embryos was delayed compared to wild-type, and consistent with GPC3 functioning as a co-receptor for fibroblast growth factor-9 (FGF9), we found a reduction in Sonic Hedgehog (Shh) mRNA expression and signaling in embryonic mutant hearts. Interestingly, we found an asymmetric reduction in SHH signaling in cardiac myocytes, as compared with perivascular cells, resulting in excessive coronary artery formation in the Gpc3-deficient animals. We hypothesize that the excessive development of coronary arteries over veins enables the formation of coronary artery fistulas. This work has broad significance to understanding the genetic basis of coronary development and potentially to molecular mechanisms relevant to revascularization following ischemic injury to the heart.
Collapse
MESH Headings
- Animals
- Coronary Vessel Anomalies/embryology
- Coronary Vessel Anomalies/genetics
- Coronary Vessel Anomalies/pathology
- Coronary Vessels/embryology
- Coronary Vessels/pathology
- Fistula/pathology
- Glypicans/genetics
- Glypicans/metabolism
- Heart/anatomy & histology
- Heart/embryology
- Heart Defects, Congenital/embryology
- Heart Defects, Congenital/pathology
- Hedgehog Proteins/genetics
- Hedgehog Proteins/metabolism
- Humans
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Myocytes, Cardiac/cytology
- Myocytes, Cardiac/metabolism
- Patched Receptors
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Receptors, Cell Surface/genetics
- Receptors, Cell Surface/metabolism
- Signal Transduction/physiology
Collapse
Affiliation(s)
- Ann Ng
- Department of Pediatrics, Washington University School of Medicine and St. Louis Children's Hospital, St. Louis, MO 63110
- Department of Biology, Washington University, St. Louis, MO 63130
| | - Michelle Wong
- Department of Pediatrics, Washington University School of Medicine and St. Louis Children's Hospital, St. Louis, MO 63110
| | - Beth Viviano
- Department of Pediatrics, Washington University School of Medicine and St. Louis Children's Hospital, St. Louis, MO 63110
| | - Jonathan M. Erlich
- Department of Pediatrics, Washington University School of Medicine and St. Louis Children's Hospital, St. Louis, MO 63110
| | - George Alba
- Department of Pediatrics, Washington University School of Medicine and St. Louis Children's Hospital, St. Louis, MO 63110
- Department of Biology, Washington University, St. Louis, MO 63130
| | - Camila Pflederer
- Department of Pediatrics, Washington University School of Medicine and St. Louis Children's Hospital, St. Louis, MO 63110
| | - Patrick Y. Jay
- Department of Pediatrics, Washington University School of Medicine and St. Louis Children's Hospital, St. Louis, MO 63110
- Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110
| | - Scott Saunders
- Department of Pediatrics, Washington University School of Medicine and St. Louis Children's Hospital, St. Louis, MO 63110
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110
| |
Collapse
|
54
|
Martínez A, Spencer ML, Brethauer U, Grez P, Marchesani FJ, Rojas IG. Reduction of syndecan-1 expression during lip carcinogenesis. J Oral Pathol Med 2009; 38:580-3. [DOI: 10.1111/j.1600-0714.2009.00761.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
55
|
Molica S, Vitelli G, Mirabelli R, Digiesu G, Giannarelli D, Cuneo A, Ribatti D, Vacca A. Serum levels of syndecan-1 in B-cell chronic lymphocytic leukemia: Correlation with the extent of angiogenesis and disease-progression risk in early disease. Leuk Lymphoma 2009; 47:1034-40. [PMID: 16840194 DOI: 10.1080/10428190500470358] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Syndecan-1 is a transmembrane proteoglycan generally not expressed in mature B-cell neoplasias like chronic lymphocytic leukemia (CLL). Moreover, information dealing with the evaluation of soluble syndecan-1 in CLL are lacking. We measured syndecan-1 concentrations in serum drawn at the time of diagnosis from 67 B-cell CLL patients (Binet stage A, 46; stage B, 7; stage C, 14). For this purpose a syndecan-1 enzyme-linked immunosorbent assay (ELISA, Diaclone, Besancon, France) was used. Detectable levels of syndecan-1 were found in all patients, although serum concentrations were significantly lower in CLL patients in comparison to age- and sex-matched controls (P = 0.02; Mann-Whitney test). No correlation was found with Binet clinical stages (P = 0.796), beta2-microglobulin (P = 0.923), hemoglobin level (P = 0.605), platelet count (P = 0.992) and lymphocyte doubling time (P = 0.709). Only an association with absolute peripheral blood lymphocytosis (PBL) (P = 0.01) and LDH (P = 0.05) could be detected. Serum levels of syndecan-1 did not parallel those of several angiogenic cytokines such as vascular endothelial growth factor (VEGF) (P = 0.963), basic fibroblastic growth factor (FGF-2) (P = 0.216), angiogenin (P = 0.478), metalloproteinase-9 (MMP-9) (P = 0.125) as well as bone marrow (BM) microvessel density (P = 0.110). The same applied with adhesion molecules such as CD54 (P = 0.233), CD108 (P = 0.799), CD44 (P = 0.816) and CD31 (P = 0.508). Interestingly, the inverse correlation (r = -0.4967; P = 0.03) between serum concentrations of syndecan-1 and plasma levels of stromal derived growth factor-1 (SDF-1) is in keeping with the different function, respectively, pro- and anti-apoptotic, of these molecules. In 46 Binet stage A patients, serum levels of syndecan-1 were further evaluated as a dichotomous variable with respect to progression-free survival (PFS), an end-point surrogate for overall survival in early B-cell CLL. The best separation of curves was seen with a cut-off point at the median value of syndecan-1 (i.e. 36.5 pg/ml). Median PFS was not reached in the patient group with low syndecan-1, compared to a median of 34 months observed in the remaining patients (P = 0.018; HR = 0.208; 95% CI = 0.115 - 0.816). At the multivariate analysis performed including variables significant in the univariate analysis [i.e. PBL (P = 0.03) and syndecan-1 (P = 0.01)], only syndecan-1 retained a trend of significance (P = 0.08). Despite the pro-angiogenic activity of syndecan-1 which mediates FGF-2 binding and activity, no correlation with either angiogenic cytokines or the extent of BM angiogenesis was found in CLL. The inverse correlation with plasma levels of SDF-1 suggests an involvement in the processes leading to apoptosis. Finally, our results highlight the involvement of syndecan-1 in the mechanisms of disease-progression of early CLL.
Collapse
Affiliation(s)
- Stefano Molica
- Medical Oncology, Azienda Ospedaliera Pugliese-Ciaccio, Catanzaro, Italy.
| | | | | | | | | | | | | | | |
Collapse
|
56
|
Olivares GH, Carrasco H, Aroca F, Carvallo L, Segovia F, Larraín J. Syndecan-1 regulates BMP signaling and dorso-ventral patterning of the ectoderm during early Xenopus development. Dev Biol 2009; 329:338-49. [PMID: 19303002 DOI: 10.1016/j.ydbio.2009.03.007] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2008] [Revised: 03/06/2009] [Accepted: 03/06/2009] [Indexed: 10/21/2022]
Abstract
Extracellular regulation of growth factor signaling is a key event for embryonic patterning. Heparan sulfate proteoglycans (HSPG) are among the molecules that regulate this signaling during embryonic development. Here we study the function of syndecan1 (Syn1), a cell-surface HSPG expressed in the non-neural ectoderm during early development of Xenopus embryos. Overexpression of Xenopus Syn1 (xSyn1) mRNA is sufficient to reduce BMP signaling, induce chordin expression and rescue dorso-ventral patterning in ventralized embryos. Experiments using chordin morpholinos established that xSyn1 mRNA can inhibit BMP signaling in the absence of chordin. Knockdown of xSyn1 resulted in a reduction of BMP signaling and expansion of the neural plate with the concomitant reduction of the non-neural ectoderm. Overexpression of xSyn1 mRNA in xSyn1 morphant embryos resulted in a biphasic effect, with BMP being inhibited at high concentrations and activated at low concentrations of xSyn1. Interestingly, the function of xSyn1 on dorso-ventral patterning and BMP signaling is specific for this HSPG. In summary, we report that xSyn1 regulates dorso-ventral patterning of the ectoderm through modulation of BMP signaling.
Collapse
Affiliation(s)
- Gonzalo H Olivares
- Center for Aging and Regeneration, Center for Cell Regulation and Pathology, MIFAB, Faculty of Biological Sciences, P. Universidad Católica de Chile, Alameda 340, Santiago, Chile
| | | | | | | | | | | |
Collapse
|
57
|
Nikolova V, Koo CY, Ibrahim SA, Wang Z, Spillmann D, Dreier R, Kelsch R, Fischgräbe J, Smollich M, Rossi LH, Sibrowski W, Wülfing P, Kiesel L, Yip GW, Götte M. Differential roles for membrane-bound and soluble syndecan-1 (CD138) in breast cancer progression. Carcinogenesis 2009; 30:397-407. [PMID: 19126645 DOI: 10.1093/carcin/bgp001] [Citation(s) in RCA: 153] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The heparan sulfate proteoglycan syndecan-1 (Sdc1) modulates cell proliferation, adhesion, migration and angiogenesis. Proteinase-mediated shedding converts Sdc1 from a membrane-bound coreceptor into a soluble effector capable of binding the same ligands. In breast carcinomas, Sdc1 overexpression correlates with poor prognosis and an aggressive phenotype. To distinguish between the roles of membrane-bound and shed forms of Sdc1 in breast cancer progression, human MCF-7 breast cancer cells were stably transfected with plasmids overexpressing wild-type (WT), constitutively shed and uncleavable forms of Sdc1. Overexpression of WT Sdc1 increased cell proliferation, whereas overexpression of constitutively shed Sdc1 decreased proliferation. Fibroblast growth factor-2-mediated mitogen-activated protein kinase signaling was reduced following small-interfering RNA (siRNA)-mediated knockdown of Sdc1 expression. Constitutively, membrane-bound Sdc1 inhibited invasiveness, whereas soluble Sdc1 promoted invasion of MCF-7 cells into matrigel matrices. The latter effect was reversed by the matrix metalloproteinase inhibitors N-isobutyl-N-(4-methoxyphenylsufonyl) glycyl hydroxamic acid and tissue inhibitor of metalloproteinase (TIMP)-1. Affymetrix microarray analysis identified TIMP-1, Furin and urokinase-type plasminogen activator receptor as genes differentially regulated in soluble Sdc1-overexpressing cells. Endogenous TIMP-1 expression was reduced in cells overexpressing soluble Sdc1 and increased in those overexpressing the constitutively membrane-bound Sdc1. Moreover, E-cadherin protein expression was downregulated in cells overexpressing soluble Sdc1. Our results suggest that the soluble and membrane-bound forms of Sdc1 play different roles at different stages of breast cancer progression. Proteolytic conversion of Sdc1 from a membrane-bound into a soluble molecule marks a switch from a proliferative to an invasive phenotype, with implications for breast cancer diagnostics and potential glycosaminoglycan-based therapies.
Collapse
Affiliation(s)
- Viktoriya Nikolova
- Department of Gynecology and Obstetrics, University Hospital Münster, Münster, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
58
|
Mendes-de-Aguiar CBN, Costa-Silva B, Alvarez-Silva M, Tasca CI, Trentin AG. Thyroid hormone mediates syndecan expression in rat neonatal cerebellum. Cell Mol Neurobiol 2008; 28:795-801. [PMID: 18219570 DOI: 10.1007/s10571-008-9260-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2007] [Accepted: 01/04/2008] [Indexed: 10/22/2022]
Abstract
Thyroid hormone (T(3)) plays an essential role in the central nervous system development. Astrocytes mediate many of the T(3) effects in the growth and differentiation of cerebellum. In culture, T(3) induces cerebellar astrocytes to secrete growth factors, mainly FGF(2), and alters the expression and organization of the extracellular matrix (ECM) proteins, laminin, and fibronectin. In addition, T(3)-treated astrocytes promote neuronal differentiation. In this study, we have investigated whether other ECM molecules, such as syndecans, are involved in T(3) action. Thus, we analyzed the expression of syndecans (1-4) by RT-PCR in astrocyte cultures from cerebellum, cortex, and hippocampus of newborn rats. Our results showed that syndecans (1-4) are expressed in astrocytes of cerebellum and cortex, whereas in hippocampus only syndecans 2 and 4 were detected. Semi-quantitative RT-PCR analysis revealed the reduced expression of syndecans 1, 2, and 4, and increased expression of syndecan 3 in hypothyroid cerebellum, when compared to the euthyroid tissue. Furthermore, we observed a reduced expression of syndecans 2 and 3 in T(3)-treated cerebellar astrocytes, when compared to control cultures. This balance of proteoglycans may be involved in T(3) action mediated by FGF(2) signaling, possibly affecting the formation of the trimeric signaling receptor complex composed by syndecan/FGF/FGF-receptor (FGFR), which is essential for FGFR dimerization, activation, and subsequent cell signaling.
Collapse
Affiliation(s)
- Cláudia Beatriz Nedel Mendes-de-Aguiar
- Departamento de Biologia Celular, Embriologia e Genética, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Campus Universitário, Trindade, 88040-900, Florianópolis, SC, Brasil
| | | | | | | | | |
Collapse
|
59
|
Capurro MI, Xu P, Shi W, Li F, Jia A, Filmus J. Glypican-3 inhibits Hedgehog signaling during development by competing with patched for Hedgehog binding. Dev Cell 2008; 14:700-11. [PMID: 18477453 DOI: 10.1016/j.devcel.2008.03.006] [Citation(s) in RCA: 264] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2007] [Revised: 11/16/2007] [Accepted: 03/10/2008] [Indexed: 12/14/2022]
Abstract
Loss-of-function mutations in glypican-3 (GPC3), one of the six mammalian glypicans, causes the Simpson-Golabi-Behmel overgrowth syndrome (SGBS), and GPC3 null mice display developmental overgrowth. Because the Hedgehog signaling pathway positively regulates body size, we hypothesized that GPC3 acts as an inhibitor of Hedgehog activity during development. Here, we show that GPC3 null embryos display increased Hedgehog signaling and that GPC3 inhibits Hedgehog activity in cultured mouse embryonic fibroblasts. In addition, we report that GPC3 interacts with high affinity with Hedgehog but not with its receptor, Patched, and that GPC3 competes with Patched for Hedgehog binding. Furthermore, GPC3 induces Hedgehog endocytosis and degradation. Surprisingly, the heparan sulfate chains of GPC3 are not required for its interaction with Hedgehog. We conclude that GPC3 acts as a negative regulator of Hedgehog signaling during mammalian development and that the overgrowth observed in SGBS patients is, at least in part, the consequence of hyperactivation of the Hedgehog signaling pathway.
Collapse
Affiliation(s)
- Mariana I Capurro
- Division of Molecular and Cell Biology, Sunnybrook Research Institute and Department of Medical Biophysics, University of Toronto, 2075 Bayview Avenue, Toronto, Ontario M4N3M5, Canada
| | | | | | | | | | | |
Collapse
|
60
|
Mezzano V, Cabrera D, Vial C, Brandan E. Constitutively activated dystrophic muscle fibroblasts show a paradoxical response to TGF-beta and CTGF/CCN2. J Cell Commun Signal 2008; 1:205-17. [PMID: 18600480 DOI: 10.1007/s12079-008-0018-2] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2008] [Accepted: 02/09/2008] [Indexed: 12/20/2022] Open
Abstract
Transforming growth factor beta (TGF-beta) and connective tissue growth factor (CTGF) have been described to induce the production of extracellular matrix (ECM) proteins and have been reported to be increased in different fibrotic disorders. Skeletal muscle fibrosis is a common feature of Duchenne muscular dystrophy (DMD). The mdx mouse diaphragm is a good model for DMD since it reproduces the muscle degenerative and fibrotic changes. Fibronectin (FN) and proteoglycans (PG) are some of the ECM proteins upregulated in dystrophic conditions. In view of understanding the fibrotic process involved in DMD we have isolated fibroblasts from dystrophic mdx diaphragms. Here we report that regardless of the absence of degenerative myofibers, adult mdx diaphragm fibroblasts show increased levels of FN and condroitin/dermatan sulfate PGs synthesis. Fibroblasts isolated from non fibrotic tissue, such as 1 week old mice diaphragms or skin, do not present elevated FN levels. Furthermore, mdx fibroblast conditioned media is able to stimulate FN synthesis in control fibroblasts. Autocrine TGF-beta signaling was unaltered in mdx cells. When control fibroblasts are exposed to TGF-beta and CTGF, FN increases as expected. Paradoxically, in mdx cells it decreases in a concentration dependent manner and this decrease is not due to a downregulation of FN synthesis. According to this data we hypothesize that a pathological environment is able to reprogram fibroblasts into an activated phenotype which can be maintained through generations.
Collapse
Affiliation(s)
- Valeria Mezzano
- Centro de Regulación Celular y Patología, Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, MIFAB, Pontificia Universidad Católica de Chile, Santiago, Chile
| | | | | | | |
Collapse
|
61
|
|
62
|
Denef C. Paracrinicity: the story of 30 years of cellular pituitary crosstalk. J Neuroendocrinol 2008; 20:1-70. [PMID: 18081553 PMCID: PMC2229370 DOI: 10.1111/j.1365-2826.2007.01616.x] [Citation(s) in RCA: 85] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2007] [Revised: 07/30/2007] [Accepted: 08/13/2007] [Indexed: 11/27/2022]
Abstract
Living organisms represent, in essence, dynamic interactions of high complexity between membrane-separated compartments that cannot exist on their own, but reach behaviour in co-ordination. In multicellular organisms, there must be communication and co-ordination between individual cells and cell groups to achieve appropriate behaviour of the system. Depending on the mode of signal transportation and the target, intercellular communication is neuronal, hormonal, paracrine or juxtacrine. Cell signalling can also be self-targeting or autocrine. Although the notion of paracrine and autocrine signalling was already suggested more than 100 years ago, it is only during the last 30 years that these mechanisms have been characterised. In the anterior pituitary, paracrine communication and autocrine loops that operate during fetal and postnatal development in mammals and lower vertebrates have been shown in all hormonal cell types and in folliculo-stellate cells. More than 100 compounds have been identified that have, or may have, paracrine or autocrine actions. They include the neurotransmitters acetylcholine and gamma-aminobutyric acid, peptides such as vasoactive intestinal peptide, galanin, endothelins, calcitonin, neuromedin B and melanocortins, growth factors of the epidermal growth factor, fibroblast growth factor, nerve growth factor and transforming growth factor-beta families, cytokines, tissue factors such as annexin-1 and follistatin, hormones, nitric oxide, purines, retinoids and fatty acid derivatives. In addition, connective tissue cells, endothelial cells and vascular pericytes may influence paracrinicity by delivering growth factors, cytokines, heparan sulphate proteoglycans and proteases. Basement membranes may influence paracrine signalling through the binding of signalling molecules to heparan sulphate proteoglycans. Paracrine/autocrine actions are highly context-dependent. They are turned on/off when hormonal outputs need to be adapted to changing demands of the organism, such as during reproduction, stress, inflammation, starvation and circadian rhythms. Specificity and selectivity in autocrine/paracrine interactions may rely on microanatomical specialisations, functional compartmentalisation in receptor-ligand distribution and the non-equilibrium dynamics of the receptor-ligand interactions in the loops.
Collapse
Affiliation(s)
- C Denef
- Laboratory of Cell Pharmacology, University of Leuven, Medical School, Leuven, Belgium.
| |
Collapse
|
63
|
Zhang X, Liu C, Nestor KE, McFarland DC, Velleman SG. The effect of glypican-1 glycosaminoglycan chains on turkey myogenic satellite cell proliferation, differentiation, and fibroblast growth factor 2 responsiveness. Poult Sci 2007; 86:2020-8. [PMID: 17704393 DOI: 10.1093/ps/86.9.2020] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The glypicans are a family of cell-surface heparan sulfate proteoglycans consisting of a core protein covalently attached with glycosaminoglycans (GAG). Only glypican-1 is expressed in skeletal muscle and increases in expression during myoblast differentiation. Previous studies have suggested that glypican-1 influences fibroblast growth factor 2 (FGF2) signaling pathway by its heparan sulfate chains. Fibroblast growth factor 2 is a potent stimulator of muscle cell proliferation and an intense inhibitor of differentiation. To investigate the functional contribution of each GAG chain attachment site, a turkey glypican-1 full length cDNA (1,650 bp, Gen-Bank accession number AY551002) was cloned into the pCMS-EGFP vector and mutated at 2 or all 3 potential GAG attachment sites at Ser(483), Ser(485), and Ser(487) to obtain 1-chain and no-chain mutants, respectively. The unmutated glypican-1, 1-chain, and no-chain mutants, and the pCMS-EGFP vector without an insert were transfected into turkey myogenic satellite cells. The transfected cell cultures were assayed for cell proliferation, differentiation, and FGF2 responsiveness. The overexpression of glypican-1 increased FGF2 responsiveness during proliferation compared with the 1-chain, no-chain mutants, and the pCMS-EGFP vector without an insert, but there was no significant interaction between FGF2 and glypican-1. The overexpression of glypican-1 also increased differentiation but did not affect proliferation when compared with the 1-chain, no-chain mutants, and the pCMS-EGFP vector without an insert. To support the overexpression data, glypican-1 expression was reduced using a small interfering RNA against turkey glypican-1. Inhibition of glypican-1 expression decreased myogenic satellite cell proliferation, differentiation, and FGF2 responsiveness during proliferation. These data indicate that glypican-1 function requires the GAG chain attachment sites for myogenic satellite cell FGF2 responsiveness during proliferation and to affect the process of differentiation.
Collapse
Affiliation(s)
- X Zhang
- Department of Animal Sciences, Ohio Agricultural Research and Development Center, The Ohio State University, Wooster Ohio 44691, USA
| | | | | | | | | |
Collapse
|
64
|
Abstract
Muscle cell proliferation, migration, adhesion, and fusion are processes involved with the formation of multinucleated myotubes that will further differentiate into mature muscle fibers. The process of muscle fiber development is nearly complete at the time of hatch. Muscle growth during the embryonic period of development is characterized by an increase in myoblast cell number through hyperplasia. Posthatch muscle fiber growth occurs through muscle fiber enlargement by the process of hypertrophy, which results from the recruitment of satellite cell nuclei. Hyperplasia and hypertrophy are regulated by factors extrinsic to the cell. These extrinsic elements include growth factors and the extracellular matrix. The growth factors, hepatocyte growth factor, fibroblast growth factor 2, transforming growth factor-beta, insulin-like growth factor, and myostatin stimulate or inhibit myoblast and satellite cell proliferation and differentiation. Some of these growth factors like fibroblast growth factor 2 must interact with a low affinity extracellular matrix macromolecule to bind to their high affinity receptor necessary for cell signaling. It is probable that the expression of extracellular matrix proteins involved in growth factor signaling will affect muscle growth properties during hyperplasia and hypertrophy. As signaling pathways associated with muscle growth mechanisms are further understood, the poultry industry may find it beneficial to include the expression of key genes in their selection strategies.
Collapse
Affiliation(s)
- S G Velleman
- Department of Animal Sciences, Ohio Agricultural Research and Development Center, The Ohio State University, Wooster 44691, USA.
| |
Collapse
|
65
|
Parker CJ. The pathophysiology of paroxysmal nocturnal hemoglobinuria. Exp Hematol 2007; 35:523-33. [PMID: 17379062 DOI: 10.1016/j.exphem.2007.01.046] [Citation(s) in RCA: 78] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2006] [Revised: 01/18/2007] [Accepted: 01/19/2007] [Indexed: 12/12/2022]
Abstract
The molecular basis of PNH is known. Somatic mutation of the X-chromosome gene PIGA accounts for deficiency of glycosyl phosphatidylinositol-anchored proteins (GPI-AP) on affected hematopoietic stem cells and their progeny. However, neither mutant PIGA nor the consequent deficiency of GPI-AP provides a direct explanation for the clonal outgrowth of the mutant stem cells. Therefore, PNH differs from malignant myelopathies in which clonal expansion is directly attributable to a specific, monogenetic event (e.g., t(9;22) in CML) that bestows a growth/survival advantage upon the affected cell. Multiple, discrete PIGA mutant clones are present in many patients, suggesting that a selection pressure that favors the PNH phenotype (i.e., GPI-AP deficiency) was applied to the bone marrow. The nature of this putative selection pressure, however, is speculative, as is the basis of clonal expansion. In many patients, the majority of hematopoiesis is derived from PIGA mutant stem cells. Yet clonal expansion is limited (nonmalignant), and the contribution of the mutant clones to hematopoiesis may remain stable for decades. Understanding the basis of clonal selection and expansion will not only delineate further the pathophysiology of PNH but also provide new insights into stem cell biology and suggest novel therapeutic strategies for enhancing marrow function.
Collapse
Affiliation(s)
- Charles J Parker
- Division of Hematology and Bone Marrow Transplant, University of Utah School of Medicine and the George E. Whalen Department of Veterans Affairs Medical Center, 500 Foothill Boulevard, Salt Lake City, UT 84114, USA.
| |
Collapse
|
66
|
Mochizuki M, Philp D, Hozumi K, Suzuki N, Yamada Y, Kleinman HK, Nomizu M. Angiogenic activitiy of syndecan-binding laminin peptide AG73 (RKRLQVQLSIRT). Arch Biochem Biophys 2007; 459:249-55. [PMID: 17286955 DOI: 10.1016/j.abb.2006.12.026] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2006] [Revised: 12/11/2006] [Accepted: 12/12/2006] [Indexed: 11/18/2022]
Abstract
The AG73 peptide (RKRLQVQLSIRT, mouse laminin alpha 1 chain 2719-2730) promotes cell adhesion and tumor metastasis, and interacts with transmembrane syndecan proteoglycans. Here, we demonstrate AG73 peptide angiogenic activity using in vitro, ex vivo, and in vivo models. AG73 induced murine endothelial cell (SVEC4-10) tube formation on Cultrex Basement Membrane Extract (Cultrex BME) and stimulated sprouting of aortic rings. None of the homologous sequences from the laminin alpha2, alpha3, alpha4, or alpha5 chains was as active as AG73 in promoting sprouting formation. AG73 also mediated angiogenesis in the chick chorioallantonic membrane (CAM) assay. Using subcutaneously injected Cultrex BME supplemented with AG73, we observed a large angiogenic response. Furthermore, AG73-conjugated to a chitosan membrane promoted a strong angiogenic response in the CAM assay. These results indicate that the AG73 peptide is a potent syndecan-binding angiogenesis stimulator and may be useful for therapeutic application to treat ischemic injuries.
Collapse
Affiliation(s)
- Mayumi Mochizuki
- Tokyo University of Pharmacy and Life Science, 1432-1 Horinouchi, Hachioji, Tokyo 192-0392, Japan
| | | | | | | | | | | | | |
Collapse
|
67
|
Pazos MDC, Ricci R, Simioni AR, Lopes CC, Tedesco AC, Nader HB. Putative role of heparan sulfate proteoglycan expression and shedding on the proliferation and survival of cells after photodynamic therapy. Int J Biochem Cell Biol 2007; 39:1130-41. [PMID: 17416540 DOI: 10.1016/j.biocel.2007.02.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2006] [Revised: 01/22/2007] [Accepted: 02/08/2007] [Indexed: 01/09/2023]
Abstract
INTRODUCTION Photodynamic therapy is based on the selective retention of a photosensitizer by highly proliferating cells and its activation with light at the appropriate wavelength. This combination generates reactive oxygen species that ultimately kill the cells. Some cells, however, may survive photodynamic therapy and the interaction of these cells with the extracellular matrix has profound effect in tumor biology. The knowledge of photodynamic therapy action on the extracellular matrix has not been fully explored. It has been focused mainly on integrins, matrix metalloproteinases and on growth factors and immunological mediators. Other important molecules involved in the regulation of many cell processes are the glycosaminoglycans, polymers of disaccharide units, present on the cell surface and in the extracellular matrix. In most cases, the glycosaminoglycans occur as proteoglycans. AIMS The purpose of the present investigation is to evaluate heparan sulfate proteoglycan expression and shedding, and its relation to the survival of the remaining cells, after a liposomal-AlClPc based photodynamic treatment. MATERIALS A wild-type endothelial cell derived from rabbit aorta and its counterpart transfected with EJ-ras oncogene were used. RESULTS Both cell lines presented augmented heparan sulfate proteoglycan syndecan-4 mRNA expression, augmented synthesis of heparan sulfate chains and increased shedding. Also, the formation of stress fibers on the border of the cells and the arrest in G(1) phase of the cell cycle was observed. CONCLUSIONS These results show that surviving cells after photodynamic therapy exhibit changes in their morphology and cell processes that differ from that of non-treated cells, and these changes are probably hindering the cells from resuming normal proliferation.
Collapse
|
68
|
Alexopoulou AN, Multhaupt HAB, Couchman JR. Syndecans in wound healing, inflammation and vascular biology. Int J Biochem Cell Biol 2006; 39:505-28. [PMID: 17097330 DOI: 10.1016/j.biocel.2006.10.014] [Citation(s) in RCA: 233] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2006] [Revised: 10/20/2006] [Accepted: 10/23/2006] [Indexed: 01/24/2023]
Abstract
Syndecans are heparan sulphate proteoglycans consisting of a type I transmembrane core protein modified by heparan sulphate and sometimes chondroitin sulphate chains. They are major proteoglycans of many organs including the vasculature, along with glypicans and matrix proteoglycans. Heparan sulphate chains have potential to interact with a wide array of ligands, including many growth factors, cytokines, chemokines and extracellular matrix molecules relevant to growth regulation in vascular repair, hypoxia, angiogenesis and immune cell function. This is consistent with the phenotypes of syndecan knock-out mice, which while viable and fertile, show deficits in tissue repair. Furthermore, there are potentially important changes in syndecan distribution and function described in a variety of human vascular diseases. The purpose of this review is to describe syndecan structure and function, consider the role of syndecan core proteins in transmembrane signalling and also their roles as co-receptors with other major classes of cell surface molecules. Current debates include potential redundancy between syndecan family members, the significance of multiple heparan sulphate interactions, regulation of the cytoskeleton and cell behaviour and the switch between promoter and inhibitor of important cell functions, resulting from protease-mediated shedding of syndecan ectodomains.
Collapse
Affiliation(s)
- Annika N Alexopoulou
- Division of Biomedical Sciences, Imperial College London, Sir Alexander Fleming Building, Exhibition Road, London SW7 2AZ, UK
| | | | | |
Collapse
|
69
|
Wilsie LC, Gonzales AM, Orlando RA. Syndecan-1 mediates internalization of apoE-VLDL through a low density lipoprotein receptor-related protein (LRP)-independent, non-clathrin-mediated pathway. Lipids Health Dis 2006; 5:23. [PMID: 16945147 PMCID: PMC1592478 DOI: 10.1186/1476-511x-5-23] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2006] [Accepted: 08/31/2006] [Indexed: 01/20/2023] Open
Abstract
Background Triacylglyerol-rich very low density lipoprotein (VLDL) particles are the primary carriers of fatty acids in the circulation and as such serve as a rich energy source for peripheral tissues. Receptor-mediated uptake of these particles is dependent upon prior association with apolipoprotein E (apoE-VLDL) and is brought about by cell surface heparan sulfate proteoglycans (HSPG) in some cell types and by the low density lipoprotein receptor-related protein (LRP) in others. Although LRP's role in apoE-VLDL uptake has been well studied, the identity of the HSPG family member that mediates apoE-VLDL uptake has not been established. We investigated if syndecan-1 (Syn-1), a transmembrane cell surface HSPG, is able to mediate the internalization of apoE-VLDL and examined the relationship between Syn-1 and LRP toward apoE-VLDL uptake. For this study, we used a human fibroblast cell line (GM00701) that expresses large amounts of LRP, but possesses no LDL receptor activity to eliminate its contributions toward apoE-VLDL uptake. Results Although LRP in these cells is fully active as established by substantial α2macroglobulin binding and internalization, uptake of apoE-VLDL is absent. Expression of human Syn-1 cDNA restored apoE-VLDL binding and uptake by these cells. Competition for this uptake with an LRP ligand-binding antagonist had little or no effect, whereas co-incubation with heparin abolished apoE-VLDL internalization. Depleting Syn-1 expressing cells of K+, to block clathrin-mediated endocytosis, showed no inhibition of Syn-1 internalization of apoE-VLDL. By contrast, treatment of cells with nystatin to inhibit lipid raft function, prevented the uptake of apoE-VLDL by Syn-1. Conclusion These data demonstrate that Syn-1 is able to mediate apoE-VLDL uptake in human fibroblasts with little or no contribution from LRP and that the endocytic path taken by Syn-1 is clathrin-independent and relies upon lipid raft function. These data are consistent with previous studies demonstrating Syn-1 association with lipid raft domains.
Collapse
Affiliation(s)
- Larissa C Wilsie
- Department of Biochemistry and Molecular Biology, University of New Mexico, School of Medicine, MSC08 4670 1 University of New Mexico, Albuquerque, New Mexico, 87131, USA
| | - Amanda M Gonzales
- Department of Biochemistry and Molecular Biology, University of New Mexico, School of Medicine, MSC08 4670 1 University of New Mexico, Albuquerque, New Mexico, 87131, USA
| | - Robert A Orlando
- Department of Biochemistry and Molecular Biology, University of New Mexico, School of Medicine, MSC08 4670 1 University of New Mexico, Albuquerque, New Mexico, 87131, USA
| |
Collapse
|
70
|
Su G, Meyer K, Nandini CD, Qiao D, Salamat S, Friedl A. Glypican-1 is frequently overexpressed in human gliomas and enhances FGF-2 signaling in glioma cells. THE AMERICAN JOURNAL OF PATHOLOGY 2006; 168:2014-26. [PMID: 16723715 PMCID: PMC1606624 DOI: 10.2353/ajpath.2006.050800] [Citation(s) in RCA: 122] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Signaling by fibroblast growth factor 2 (FGF-2), an autocrine stimulator of glioma growth, is regulated by heparan sulfate proteoglycans (HSPGs) via a ternary complex with FGF-2 and the FGF receptor (FGFR). To characterize glioma growth signaling, we examined whether altered HSPGs contribute to loss of growth control in gliomas. In a screen of five human glioma cell lines, U118 and U251 cell HSPGs activated FGF-2 signaling via FGFR1c. The direct comparison of U251 glioma cells with normal astrocyte HSPGs demonstrated that the glioma HSPGs had a significantly elevated ability to promote FGF-2-dependent mitogenic signaling via FGFR1c. This enhanced activity correlated with a higher level of overall sulfation, specifically the abundance of 2S- and 6S-containing disaccharides. Glioma cell expression of the cell-surface HSPG glypican-1 closely mirrored the FGF-2 coactivator activity. Furthermore, forced expression of glypican-1 in (glypican-1-deficient) U87 glioma cells enhanced their FGF-2 response. Immunohistochemical analysis revealed a highly significant overexpression of glypican-1 in human astrocytoma and oligodendroglioma samples compared with nonneoplastic gliosis. In summary, these observations suggest that altered HSPGs contribute to enhanced signaling of FGF-2 via FGFR1c in gliomas with glypican-1 playing a significant role in this mitogenic pathway.
Collapse
Affiliation(s)
- Gui Su
- University of Wisconsin-Madison, Department of Pathology and Laboratory Medicine, Clinical Sciences Center K4/850, 600 Highland Ave., Madison, WI 53792-8550, USA
| | | | | | | | | | | |
Collapse
|
71
|
Viklund L, Vorontsova N, Henttinen T, Salmivirta M. Syndecan-1 regulates FGF8b responses in S115 mammary carcinoma cells. Growth Factors 2006; 24:151-7. [PMID: 16801134 DOI: 10.1080/08977190600699426] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
In murine mammary carcinoma cells Shionogi 115 (S115) testosterone induces phenotypical transformation which is largely due to expression of fibroblast growth factor (FGF) 8b. Concomitantly, the expression of the cell surface heparan sulfate proteoglycan syndecan-1 is down-regulated. However, if syndecan-1 expression is maintained by transfection with a testosterone-driven syndecan-1 construct, transformation does not occur. Here we have investigated how the down-regulation of syndecan-1 expression in testosterone-treated S115 cells and the high level of expression in syndecan-1 transfected cells influence the cellular responses toward FGF8b. Our results show that high level of syndecan-1 is associated with a decreased magnitude and duration of the FGF8b induced Erk phosphorylation. This effect was observed regardless whether the cells were stimulated directly with exogenous FGF8b or with testosterone to induce autocrine FGF8b production. Moreover, syndecan-1 transfected cells did not respond to FGF8b stimulation by increase in the intracellular free calcium, whereas untransfected cells displayed a rapid (10 s) induction. These data suggest that, in S115 cells, syndecan-1 acts as a modulator of FGF8b signaling that can limit cellular responses to FGF receptor activation. The decreased levels of syndecan-1 expression and upregulation of the FGF signaling system seen in many cancers may contribute to the proliferation of the malignant cells in vivo.
Collapse
Affiliation(s)
- Leif Viklund
- Turku Centre for Biotechnology, University of Turku and Abo Akademi University, Finland
| | | | | | | |
Collapse
|
72
|
Liu C, McFarland DC, Nestor KE, Velleman SG. Differential expression of membrane-associated heparan sulfate proteoglycans in the skeletal muscle of turkeys with different growth rates. Poult Sci 2006; 85:422-8. [PMID: 16553270 DOI: 10.1093/ps/85.3.422] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Heparan sulfate proteoglycans (HSPG) are key components of the cell membrane and extracellular matrix of skeletal muscle cells. Two major groups of membrane-associated HSPG found in skeletal muscle are syndecans (SYN) and glypicans (GPC), both of which can regulate growth factor activities and, thus, modulate cell proliferation and differentiation. In the current study, the mRNA expression of a group of membrane-associated HSPG (SYN 2 through 4 and GPC 1) was investigated in embryonic pectoralis major muscle [embryonic days (ED) 14, 16, 18, 20, 22, 24] and myogenic satellite cells isolated from males of a turkey genetic line selected for increased 16-wk BW (F line) and an unselected randombred control (RBC2 line) from which the F line was developed. The mRNA expression was measured by a real-time quantitative PCR approach. The SYN 2 and SYN 4 expression exhibited a similar pattern during embryonic p. major muscle development, which remained constant from ED 14 to ED 22 and declined sharply from ED 22 to ED 24 to a very low level. In contrast, the SYN 3 and GPC 1 expression showed a continuous decline from ED 14 to ED 24. The F line had higher SYN 2 (ED 14, 18, 20, 22), SYN 3 (ED 22), and SYN 4 (ED 22) expression than the RBC2 line. In myogenic satellite cells, initiating differentiation resulted in a decrease in SYN 2 expression and an increase in GPC 1 expression. Both SYN 3 and SYN 4 expression stayed almost constant through both the proliferation and differentiation stages. The proliferating satellite cells from the F line displayed higher SYN 4 expression than those from the RBC2 line. Collectively, the results from the current study suggest that membrane-associated HSPG are differentially expressed in both embryonic p. major muscle tissue and satellite cells isolated from F-line and RBC2-line male turkeys, implying their distinct roles in myogenesis and differing influence on muscle growth properties.
Collapse
Affiliation(s)
- C Liu
- Department of Animal Sciences, Ohio Agricultural Research and Development Center, The Ohio State University, Wooster 44691, USA
| | | | | | | |
Collapse
|
73
|
Maeda T, Desouky J, Friedl A. Syndecan-1 expression by stromal fibroblasts promotes breast carcinoma growth in vivo and stimulates tumor angiogenesis. Oncogene 2006; 25:1408-12. [PMID: 16247452 DOI: 10.1038/sj.onc.1209168] [Citation(s) in RCA: 109] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The induction of the cell surface heparan sulfate proteoglycan syndecan-1 (Sdc1) in stromal fibroblasts is observed in more than 70% of human breast carcinomas. Using a coculture model, we have recently shown that stromal cell-derived Sdc1 stimulates carcinoma cell proliferation in vitro, and that this activity requires Sdc1 glycanation. In the present study, we investigated the effect of stromal cell Sdc1 on breast carcinoma growth in vivo. MDA-MB-231 human breast carcinoma cells were inoculated into the flanks of athymic nude mice either alone, or as mixed suspensions with Sdc1-transfected or mock-transfected 3T3 mouse fibroblasts. The mixed tumors showed an intimate association between carcinoma cells and stromal fibroblasts and histologically closely resembled poorly differentiated human breast carcinomas. The presence of fibroblasts led to significantly accelerated tumor growth, which was further augmented (88% increase) by forced expression of stromal Sdc1. The hyperemic macroscopic appearance of tumors containing Sdc1-positive stromal cells contrasted with pale tumors developing in the presence of mock-transfected fibroblasts, which prompted us to examine tumor microvessels. Stromal Sdc1 expression was associated with a significantly elevated microvessel density (36% increase) and a larger vessel area (153% increase). To evaluate the relevance of this finding in human breast cancer, the relationship between stromal Sdc1 and tumor vascularity was also examined in a tissue array containing 207 human breast carcinoma samples. Similar to the xenografts, stromal Sdc1 expression correlated with both vessel density (P=0.013) and total vessel area (P=0.0026). In conclusion, stromal fibroblast-derived Sdc1 stimulates breast carcinoma growth and angiogenesis in vivo.
Collapse
Affiliation(s)
- T Maeda
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI 53792, USA
| | | | | |
Collapse
|
74
|
Salminen-Mankonen H, Säämänen AM, Jalkanen M, Vuorio E, Pirilä L. Syndecan-1 expression is upregulated in degenerating articular cartilage in a transgenic mouse model for osteoarthritis. Scand J Rheumatol 2006; 34:469-74. [PMID: 16393771 DOI: 10.1080/03009740500304338] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
OBJECTIVE Mice heterozygous for the Del1 transgene locus with a short deletion mutation in the type II collagen gene develop early-onset degenerative changes in the knee joints that progress to end-stage osteoarthritis by the age of 12-15 months. This study focuses on the expression and distribution of syndecan-1, a cell-surface heparan sulfate proteoglycan, during the development of osteoarthritic cartilage degeneration, to better understand its role in this disease. METHODS Northern analyses of total RNA extracted from knee joints of transgenic Del1 mice and their nontransgenic controls were used to monitor changes in syndecan-1 mRNA levels during development, growth, ageing, and cartilage degeneration. Immunohistochemistry was used to study the distribution of syndecan-1 in the knee joints at different stages of cartilage degeneration. RESULTS Syndecan-1 mRNA was present in knee joints throughout life, with the highest mRNA levels in ageing knee joints. In Del1 mice, a transient upregulation of syndecan-1 mRNA synthesis was observed at the age of 6 months coinciding with early stages of cartilage degeneration and a period of attempted repair. Immunostaining for syndecan-1 was most intense in chondrocytes of superficial and intermediate zones of articular cartilage adjacent to defect areas. Chondrocyte clusters also stained strongly for syndecan-1. CONCLUSION The present temporospatial expression data on upregulation of syndecan-1 in articular cartilage during early stages of cartilage degeneration suggest that this molecule is involved in the attempted repair of cartilage fibrillations. Combined with the known role of syndecan-1 during skeletal development and wound healing, this interesting finding warrants further validation.
Collapse
|
75
|
Gutierrez J, Osses N, Brandan E. Changes in secreted and cell associated proteoglycan synthesis during conversion of myoblasts to osteoblasts in response to bone morphogenetic protein-2: role of decorin in cell response to BMP-2. J Cell Physiol 2006; 206:58-67. [PMID: 15920756 DOI: 10.1002/jcp.20428] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Proteoglycans have been identified within the extracellular matrices (ECM) of bone and are known to play a role in ECM assembly, mineralization, and bone formation. Bone morphogenetic protein-2 (BMP-2) specifically converts the differentiation pathway of C2C12 myoblasts into that of osteoblast lineage cells. Microarray analyses of the mouse myoblast cell line C2C12 and its differentiation into osteoblastic cells in response to BMP-2 have suggested the up-regulation of several proteoglycan species, although there is a lack of biochemical evidence for this response. In this study we have biochemically analyzed and characterized the proteoglycan populations that are induced in C2C12 cells upon osteoblastic differentiation produced by BMP-2. An important and specific increase in the synthesis of secreted decorin was observed in BMP-2-treated cells, as compared to untreated myoblasts and myoblasts induced to differentiate into myotubes. Decorin was seen to contain larger glycosaminoglycan (GAG) chains in induced than in non-induced cells. BMP-2 also produced an augment in the synthesis of different heparan sulfate proteoglycans such syndecan-2, - 3, glypican, and perlecan in detergent-soluble and non-soluble cellular fractions. We also examined whether the evident changes induced by BMP-2 in secreted decorin could have a functional role. BMP-2 signaling dependent as well as induction of alkaline phosphatase (ALP) activity was diminished in decorin null myoblasts compared to wild type myoblats although cell surface level of BPM-2 receptors was unchanged. These results are the first biochemical evidence and analysis for the effect of BMP-2 on the synthesis of proteoglycan during osteogenic conversion of myoblasts and suggest a role for decorin in cell response to BMP-2.
Collapse
Affiliation(s)
- Jaime Gutierrez
- Centro de Regulación Celular y Patología, Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, MIFAB, P. Universidad Católica de Chile, Santiago, Chile
| | | | | |
Collapse
|
76
|
Carrasco H, Olivares GH, Faunes F, Oliva C, Larraín J. Heparan sulfate proteoglycans exert positive and negative effects in Shh activity. J Cell Biochem 2006; 96:831-8. [PMID: 16149075 DOI: 10.1002/jcb.20586] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Hedgehog (Hh) proteins are morphogens involved in short- and long-range effects during early embryonic development. Genetic analysis in fly and vertebrate embryos showed that heparan sulfate proteoglycans (HSPGs) are required for Hh transport and signaling. To further understand how HSPGs regulate Sonic hedgehog (Shh), we performed experiments using cell culture and biochemical assays. When the synthesis of HSPGs was reduced, a decrease in Shh activity was observed. Contrary to that, addition of a peptide that competes the binding of Shh to HSPGs resulted in augmentation of Shh activity. From these results, we concluded that HSPGs exert positive and negative effects in Shh activity. This dual effect correlates with the finding that Shh interacts preferentially with two HSPGs. The current model for the role of HSPGs in Shh diffusion is discussed in view of our findings.
Collapse
Affiliation(s)
- Héctor Carrasco
- Department of Cell and Molecular Biology, Center for Cell Regulation and Pathology, Millennium Nucleus in Developmental Biology, Faculty of Biological Sciences, P. Universidad Católica de Chile, Alameda 340, Santiago, Chile
| | | | | | | | | |
Collapse
|
77
|
Murakami K, Namikawa K, Shimizu T, Shirasawa T, Yoshida S, Kiyama H. Nerve injury induces the expression of EXT2, a glycosyltransferase required for heparan sulfate synthesis. Neuroscience 2006; 141:1961-9. [PMID: 16784821 DOI: 10.1016/j.neuroscience.2006.05.026] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2006] [Revised: 05/11/2006] [Accepted: 05/11/2006] [Indexed: 11/25/2022]
Abstract
Heparan sulfate proteoglycans, which bear long chains of heparan sulfate glycosaminoglycan, play significant roles during embryogenesis, including the formation of the CNS. However, their involvement in nerve regeneration has not yet been clarified. Here, we found that the mRNA expression of EXT2, one of the crucial enzymes for heparan sulfate-glycosaminoglycan synthesis, was markedly up-regulated in injured hypoglossal motor neurons after axotomy. In addition, immunohistochemical staining with an antibody specific for heparan sulfate-glycosaminoglycan chains demonstrated increased expression of heparan sulfate-glycosaminoglycan chains in the injured nucleus. Furthermore, the mRNA expressions of glypican-1 and syndecan-1, which are both well-known heparan sulfate proteoglycans, were prominently up-regulated in injured motor neurons. These results suggest that the biosynthesis of heparan sulfate chains promoted by EXT2 is activated in injured motor neurons, and that glypican-1 and syndecan-1 are potent candidates for heparan sulfate proteoglycans involved in peripheral nerve regeneration.
Collapse
Affiliation(s)
- K Murakami
- Department of Anatomy and Neurobiology, Osaka City University Graduate School of Medicine, 1-4-3 Asahimachi, Abenoku, Osaka 545-8585, Japan
| | | | | | | | | | | |
Collapse
|
78
|
Broekelmann TJ, Kozel BA, Ishibashi H, Werneck CC, Keeley FW, Zhang L, Mecham RP. Tropoelastin Interacts with Cell-surface Glycosaminoglycans via Its COOH-terminal Domain. J Biol Chem 2005; 280:40939-47. [PMID: 16192266 DOI: 10.1074/jbc.m507309200] [Citation(s) in RCA: 124] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Using a biochemical and cell biological approach, we have identified a cell interaction site at the carboxyl terminus of tropoelastin. Cell interactions with the COOH-terminal sequence are not through the elastin-binding protein (EBP67) because neither VGVAPG-like peptides nor galactoside sugars altered adhesion. Our results also show that cell adhesion to tropoelastin is not promoted by integrins. Through the use of mutant Chinese hamster ovary cell lines defective in glycosaminoglycan biosynthesis, as well as competition studies and enzymatic removal of specific cell-surface glycosaminoglycans, the tropoelastin-binding moieties on the cell surface were identified as heparan and chondroitin sulfate-containing glycosaminoglycans, with heparan sulfate being greatly preferred. Heparin affinity chromatography combined with cell adhesion assays identified the last 17 amino acids as the sequence element at the carboxyl terminus of tropoelastin responsible for the adhesive activity.
Collapse
Affiliation(s)
- Thomas J Broekelmann
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | | | | | | | | | | | | |
Collapse
|
79
|
Ding K, Lopez-Burks M, Sánchez-Duran JA, Korc M, Lander AD. Growth factor-induced shedding of syndecan-1 confers glypican-1 dependence on mitogenic responses of cancer cells. ACTA ACUST UNITED AC 2005; 171:729-38. [PMID: 16286510 PMCID: PMC2171561 DOI: 10.1083/jcb.200508010] [Citation(s) in RCA: 100] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The cell surface heparan sulfate proteoglycan (HSPG) glypican-1 is up-regulated by pancreatic and breast cancer cells, and its removal renders such cells insensitive to many growth factors. We sought to explain why the cell surface HSPG syndecan-1, which is also up-regulated by these cells and is a known growth factor coreceptor, does not compensate for glypican-1 loss. We show that the initial responses of these cells to the growth factor FGF2 are not glypican dependent, but they become so over time as FGF2 induces shedding of syndecan-1. Manipulations that retain syndecan-1 on the cell surface make long-term FGF2 responses glypican independent, whereas those that trigger syndecan-1 shedding make initial FGF2 responses glypican dependent. We further show that syndecan-1 shedding is mediated by matrix metalloproteinase-7 (MMP7), which, being anchored to cells by HSPGs, also causes its own release in a complex with syndecan-1 ectodomains. These results support a specific role for shed syndecan-1 or MMP7-syndecan-1 complexes in tumor progression and add to accumulating evidence that syndecans and glypicans have nonequivalent functions in vivo.
Collapse
Affiliation(s)
- Kan Ding
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697
| | | | | | | | | |
Collapse
|
80
|
Wang D, Anderson JC, Gladson CL. The role of the extracellular matrix in angiogenesis in malignant glioma tumors. Brain Pathol 2005; 15:318-26. [PMID: 16389944 PMCID: PMC8095805 DOI: 10.1111/j.1750-3639.2005.tb00117.x] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Angiogenesis is a promising target for the development of effective strategies for the treatment of malignant brain tumors in that it has the potential to starve large tumors and prevent the regrowth of residual margins. Two critical steps in angiogenesis, the proliferation of activated endothelial cells and their migration into the perivascular space (sprouting), require adherence of the endothelial cells to the extracellular matrix (ECM). Thus, the availability of the appropriate ligands within the ECM contributes to the regulation of angiogenesis. In addition, several components of the ECM can act through other mechanisms to further promote angiogenesis or inhibit it. Current evidence suggests that the regulation of angiogenesis is a dynamic process in which the endothelial cells can promote angiogenesis by secreting proteases that remodel the ECM, tumor cells can further promote angiogenesis by secreting ECM components and actively remodeling their environment, and stromal cells may respond to angiogenesis associated with tumors and inflammatory reactions by secreting inhibitory molecules. Here, we provide a critical review of the protein and proteoglycan components of the ECM that have been implicated in angiogenesis with an emphasis on their role in promoting or inhibiting angiogenesis in brain tumors.
Collapse
Affiliation(s)
- Dongyan Wang
- Department of Pathology, Division of Neuropathology, University of Alabama at Birmingham
| | - Joshua C. Anderson
- Department of Pathology, Division of Neuropathology, University of Alabama at Birmingham
| | - Candece L. Gladson
- Department of Pathology, Division of Neuropathology, University of Alabama at Birmingham
| |
Collapse
|
81
|
Capurro MI, Xiang YY, Lobe C, Filmus J. Glypican-3 promotes the growth of hepatocellular carcinoma by stimulating canonical Wnt signaling. Cancer Res 2005; 65:6245-54. [PMID: 16024626 DOI: 10.1158/0008-5472.can-04-4244] [Citation(s) in RCA: 366] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Glypican-3 (GPC3) is a heparan sulfate proteoglycan that is bound to the cell membrane by a glycosyl-phosphatidylinositol anchor. GPC3 is expressed by most hepatocellular carcinomas but not by normal hepatocytes and benign liver lesions. We report here that GPC3 stimulates the in vitro and in vivo growth of hepatocellular carcinoma cells by increasing autocrine/paracrine canonical Wnt signaling. Co-immunoprecipitation experiments showed that GPC3 is able to form complexes with Wnts, and cell-binding assays indicated that GPC3-expressing cells have an increased capacity to bind Wnt. Collectively, these results suggest that GPC3 stimulates Wnt activity by facilitating the interaction of this polypeptide with its signaling receptors. Surprisingly, in contrast to the current model that proposes that Wnt-glypican binding is mediated by the heparan sulfate chains, we found that the nonglycanated GPC3 core protein can form complexes with Wnts. Furthermore, we showed that the glycosaminoglycan chains are not required for the stimulatory effect on Wnt signaling and hepatocellular carcinoma growth.
Collapse
Affiliation(s)
- Mariana I Capurro
- Division of Molecular and Cell Biology, Sunnybrook and Women's College Health Sciences Centre and Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | | | | | | |
Collapse
|
82
|
Rege TA, Fears CY, Gladson CL. Endogenous inhibitors of angiogenesis in malignant gliomas: nature's antiangiogenic therapy. Neuro Oncol 2005; 7:106-21. [PMID: 15831230 PMCID: PMC1871889 DOI: 10.1215/s115285170400119x] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Angiogenesis is necessary for tumor growth beyond a volume of approximately 2 mm(3). This observation, along with the accessibility of tumor vessels to therapeutic targeting, has resulted in a research focus on inhibitors of angiogenesis. A number of endogenous inhibitors of angiogenesis are found in the body. Some of these are synthesized by specific cells in different organs, and others are created by extracellular proteolytic cleavage of plasma-derived or extracellular matrix-localized proteins. In this review, we focus on angiostatin, endostatin, PEX, pigment epithelial-derived factor, and thrombospondin (TSP)-1 and -2, either because these molecules are expressed in malignant glioma biopsies or because animal studies in malignant glioma models have suggested that their therapeutic administration could be efficacious. We review the known mechanisms of action, potential receptors, expression in glioma biopsy samples, and studies testing their potential therapeutic efficacy in animal models of malignant glioma. Two conclusions can be made regarding the mechanisms of action of these inhibitors: (1) Several of these inhibitors appear to mediate their antiangiogenic effect through multiple protein-protein interactions that inhibit the function of proangiogenic molecules rather than through a specific receptor-mediated signaling event, and (2) TSP-1 and TSP-2 appear to mediate their antiangiogenic effect, at least in part, through a specific receptor, CD36, which initiates the antiangiogenic signal. Although not proven in gliomas, evidence suggests that expression of specific endogenous inhibitors of angiogenesis in certain organs may be part of a host antitumor response. The studies reviewed here suggest that new antiangiogenic therapies for malignant gliomas offer exciting promise as nontoxic, growth-inhibitory agents.
Collapse
Affiliation(s)
| | | | - Candece L. Gladson
- Address correspondence to Candece L. Gladson, The University of Alabama at Birmingham, LHRB 567, 701 South 19th Street, Birmingham, AL 35294, USA (
)
| |
Collapse
|
83
|
Minniti AN, Labarca M, Hurtado C, Brandan E. Caenorhabditis elegans syndecan (SDN-1) is required for normal egg laying and associates with the nervous system and the vulva. J Cell Sci 2005; 117:5179-90. [PMID: 15456854 DOI: 10.1242/jcs.01394] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
In Caenorhabditis elegans, the identification of many enzymes involved in the synthesis and modification of glycosaminoglycans (GAGs), essential components of proteoglycans, has attained special attention in recent years. Mutations in all the genes that encode for GAG biosynthetic enzymes show defects in the development of the vulva, specifically in the invagination of the vulval epithelium. Mutants for certain heparan sulfate modifying enzymes present axonal and cellular guidance defects in specific neuronal classes. Although most of the enzymes involved in the biosynthesis and modification of heparan sulfate have been characterized in C. elegans, little is known regarding the core proteins to which these GAGs covalently bind in proteoglycans. A single syndecan homologue (sdn-1) has been identified in the C. elegans genome through sequence analysis. In the present study, we show that C. elegans synthesizes sulfated proteoglycans, seen as three distinct species in western blot analysis. In the sdn-1 (ok449) deletion mutant allele we observed the lack of one species, which corresponds to a 50 kDa product after heparitinase treatment. The expression of sdn-1 mRNA and sequencing revealed that sdn-1 (ok449) deletion mutants lack two glycosylation sites. Hence, the missing protein in the western blot analysis probably corresponds to SDN-1. In addition, we show that SDN-1 localizes to the C. elegans nerve ring, nerve cords and to the vulva. SDN-1 is found specifically phosphorylated in nerve ring neurons and in the vulva, in both wild-type worms and sdn-1 (ok449) deletion mutants. These mutants show a defective egg-laying phenotype. Our results show for the first time, the identification, localization and some functional aspects of syndecan in the nematode C. elegans.
Collapse
Affiliation(s)
- Alicia N Minniti
- Centro de Regulación Celular y Patología, Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, MIFAB, P. Universidad Católica de Chile, Casilla 114-D, Santiago
| | | | | | | |
Collapse
|
84
|
Liu X, McFarland DC, Nestor KE, Velleman SG. Developmental regulated expression of syndecan-1 and glypican in pectoralis major muscle in turkeys with different growth rates. Dev Growth Differ 2005; 46:37-51. [PMID: 15008853 DOI: 10.1111/j.1440-169x.2004.00728.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Heparan sulfate proteoglycans, syndecan-1 and glypican, are low-affinity receptors for fibroblast growth factor 2 (FGF2). Since FGF2 stimulates skeletal muscle cell proliferation but inhibits differentiation, differences in syndecan-1 and glypican expression might affect muscle development and growth by changing the intensity of FGF2 signaling. In the present study, the pectoralis major muscle from 14 to 24-day-old-embryos, and from 1 to 16-week-old birds from a turkey line (F) selected for increased 16-week bodyweight and its genetic control line (RBC2), were used to address how syndecan-1 and glypican are expressed during skeletal muscle formation. The expression of syndecan-1 and glypican was measured by semiquantitative reverse transcription polymerase chain reaction. For males, the F-line embryos expressed more syndecan-1 (days 14 and 16) and glypican (days 14 and 18) than the RBC2 line. Similar line differences for males were observed during posthatch development. The male embryos from both lines expressed more syndecan-1 at days 18 through 22 and more glypican at days 20 and 22 than the corresponding females. The temporal and spatial distribution of syndecan-1 and glypican was detected by in situ hybridization. Syndecan-1 was identified in all muscle fibers at all embryonic stages studied, whereas the glypican was detected from embryonic day 18. The data from the current study provided new information about the expression of syndecan-1 and glypican as it relates to skeletal muscle growth properties.
Collapse
Affiliation(s)
- Xiaosong Liu
- Department of Animal Sciences, The Ohio State University/Ohio Agricultural Research and Development Center, Wooster, OH, USA
| | | | | | | |
Collapse
|
85
|
Andersen NF, Standal T, Nielsen JL, Heickendorff L, Borset M, Sørensen FB, Abildgaard N. Syndecan-1 and angiogenic cytokines in multiple myeloma: correlation with bone marrow angiogenesis and survival. Br J Haematol 2005; 128:210-7. [PMID: 15638855 DOI: 10.1111/j.1365-2141.2004.05299.x] [Citation(s) in RCA: 75] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Angiogenesis is a complex process involved in the proliferation and metastasis of malignant tumours, and partly triggered by the secretion of various angiogenic factors by tumour cells or cells in the stromal environment. We investigated the correlation between bone marrow angiogenesis, estimated as microvessel density (MVD), and interleukin-6 (IL-6), basic fibroblastic growth factor (bFGF), hepatocyte growth factor (HGF) and syndecan-1 in 67 patients with newly diagnosed multiple myeloma, and evaluated the prognostic value of these parameters. Circulating levels of IL-6, bFGF, HGF and syndecan-1 were significantly higher in patients than in controls. Moreover, in patients, bone marrow levels of bFGF, HGF and syndecan-1 were higher than peripheral blood levels. Positive correlations were found between MVD and syndecan-1 blood levels (r = 0.33, P = 0.017), syndecan-1 bone marrow levels (r = 0.49, P = 0.046) and HGF blood levels (r = 0.36, P = 0.008) respectively. High MVD and high blood levels of IL-6, HGF and syndecan-1 were predictive of a shorter survival. In a multivariate survival analysis MVD and blood levels of IL-6 retained independent prognostic significance, while in a survival analysis without MVD the peripheral blood levels of HGF and syndecan-1 were strong independent prognostic factors.
Collapse
Affiliation(s)
- Niels Frost Andersen
- Department of Haematology, Aarhus University Hospital, Aarhus Hospital, Aarhus, Denmark.
| | | | | | | | | | | | | |
Collapse
|
86
|
Rauch BH, Millette E, Kenagy RD, Daum G, Fischer JW, Clowes AW. Syndecan-4 is required for thrombin-induced migration and proliferation in human vascular smooth muscle cells. J Biol Chem 2005; 280:17507-11. [PMID: 15731100 DOI: 10.1074/jbc.m410848200] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Thrombin is a mitogen and chemoattractant for vascular smooth muscle cells (SMCs) and may contribute to vascular lesion formation. We have previously shown that human SMCs, when stimulated with thrombin, release basic fibroblast growth factor (bFGF), causing phosphorylation of FGF receptor-1 (FGFR-1). Treatment with bFGF-neutralizing antibodies (anti-bFGF) or heparin inhibits thrombin-induced DNA synthesis. We concluded that thrombin may stimulate entry into the cell cycle via bFGF release and FGFR-1 activation. In the present study, we demonstrate a requirement for not only FGFR-1 but also syndecan-4, a transmembrane heparan-sulfate proteoglycan. Inhibition of syndecan-4 expression using small interfering RNA (siRNA) resulted in reduced DNA synthesis by human SMCs after stimulation with thrombin (10 nmol/liter). Anti-bFGF antibody, which inhibits DNA synthesis in control cells, had no inhibitory effect when syndecan-4 expression was reduced by siRNA. Thrombin- or bFGF-induced SMC migration, determined in Boyden chamber assays, was reduced in cells treated with syndecan-4 or FGFR-1 siRNA or by anti-bFGF. Thrombin induced phosphorylation of extracellular signal-regulated kinase (ERK) 1/2 in a biphasic pattern. Although thrombin-mediated ERK phosphorylation at 5 min was not affected by syndecan-4 or FGFR-1 siRNA, ERK phosphorylation at later time points was reduced. We conclude that thrombin-released bFGF binds to syndecan-4 and FGFR-1, which is required for thrombin-induced mitogenesis and migration.
Collapse
MESH Headings
- Aorta/cytology
- Blotting, Western
- Cell Movement
- Cell Proliferation
- Cells, Cultured
- DNA/metabolism
- Dose-Response Relationship, Drug
- Extracellular Signal-Regulated MAP Kinases/metabolism
- Fibroblast Growth Factor 2/metabolism
- Gene Silencing
- Heparin/metabolism
- Humans
- Immunoprecipitation
- Membrane Glycoproteins/physiology
- Mitogen-Activated Protein Kinase 1/metabolism
- Mitogen-Activated Protein Kinase 3/metabolism
- Muscle, Smooth, Vascular/cytology
- Myocytes, Smooth Muscle/cytology
- Phosphorylation
- Proteoglycans/physiology
- RNA, Small Interfering/metabolism
- Receptor Protein-Tyrosine Kinases/metabolism
- Receptor, Fibroblast Growth Factor, Type 1
- Receptors, Fibroblast Growth Factor/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Syndecan-4
- Thrombin/metabolism
- Time Factors
Collapse
Affiliation(s)
- Bernhard H Rauch
- University of Washington School of Medicine, Department of Surgery, Seattle, Washington 98195-6410, USA.
| | | | | | | | | | | |
Collapse
|
87
|
Vanlandschoot P, Van Houtte F, Serruys B, Leroux-Roels G. The arginine-rich carboxy-terminal domain of the hepatitis B virus core protein mediates attachment of nucleocapsids to cell-surface-expressed heparan sulfate. J Gen Virol 2005; 86:75-84. [PMID: 15604433 DOI: 10.1099/vir.0.80580-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Binding of hepatitis B virus nucleocapsids to mouse B cells leads to production of nucleocapsid-specific antibodies, class II presentation of peptides and the generation of T helper-1 immunity. This T-cell-independent activation of B cells is thought to result from cross-linking of cell-surface immunoglobulin molecules, if these contain a specific motif in the framework region 1-complementarity determining region 1 junction. In the present study, it was observed that nucleocapsids bound to different B-cell lines, an interaction that was not dependent on cell-surface-expressed immunoglobulins. Furthermore, binding to several non-B-cell lines was observed. Capsids that lacked the carboxy-terminal protamine-like domains did not bind to cells. Treatment of nucleocapsids with ribonucleases enhanced the attachment of nucleocapsids to cells. Various soluble glycosaminoglycans inhibited attachment of nucleocapsids, while treatment of cells with heparinase I also reduced binding. These observations demonstrated that the arginine-rich protamine-like regions of the core proteins are responsible for the attachment of nucleocapsids to glycosaminoglycans expressed on the plasma membranes of cells.
Collapse
Affiliation(s)
- Peter Vanlandschoot
- Virus Host Interactions Unit, Center for Vaccinology, Department of Clinical Biology, Microbiology and Immunology, Ghent University Hospital, De Pintelaan 185, 9000 Ghent, Belgium
| | - Freya Van Houtte
- Virus Host Interactions Unit, Center for Vaccinology, Department of Clinical Biology, Microbiology and Immunology, Ghent University Hospital, De Pintelaan 185, 9000 Ghent, Belgium
| | - Benedikte Serruys
- Virus Host Interactions Unit, Center for Vaccinology, Department of Clinical Biology, Microbiology and Immunology, Ghent University Hospital, De Pintelaan 185, 9000 Ghent, Belgium
| | - Geert Leroux-Roels
- Virus Host Interactions Unit, Center for Vaccinology, Department of Clinical Biology, Microbiology and Immunology, Ghent University Hospital, De Pintelaan 185, 9000 Ghent, Belgium
| |
Collapse
|
88
|
Millette E, Rauch BH, Defawe O, Kenagy RD, Daum G, Clowes AW. Platelet-Derived Growth Factor-BB–Induced Human Smooth Muscle Cell Proliferation Depends on Basic FGF Release and FGFR-1 Activation. Circ Res 2005; 96:172-9. [PMID: 15625285 DOI: 10.1161/01.res.0000154595.87608.db] [Citation(s) in RCA: 90] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
We have shown that the G protein–coupled receptor (GPCR) agonists, thrombin and Factor Xa, stimulate smooth muscle cell (SMC) proliferation through transactivation of the EGF receptor (EGFR) or the FGF receptor (FGFR), both of which are tyrosine kinase receptors. In the present study, we investigated whether platelet-derived growth factor (PDGF), a tyrosine kinase receptor agonist, might transactivate another tyrosine kinase receptor to induce SMC proliferation. Because heparin inhibits PDGF-mediated proliferation in human SMCs, we investigated whether the heparin-binding growth factor basic fibroblast growth factor (bFGF) and one of its receptors, FGFR-1, play a role in the response of human arterial SMCs to PDGF-BB. PDGF-BB induced the release of bFGF and sustained phosphorylation of FGFR-1 (30 minutes to 6 hours). A bFGF-neutralizing antibody inhibited PDGF-BB–mediated phosphorylation of FGFR-1, DNA synthesis, and cell proliferation. In the presence of bFGF antibody, PDGF-BB–induced early activation of ERK (0 to 60 minutes) was not affected, whereas late ERK activation (2 to 4 hours) was reduced. When FGFR-1 expression was suppressed using small interfering RNA (siRNA), ERK activation was reduced at late, but not early, time points after PDGF-BB stimulation. Addition of bFGF antibody to cells treated with siRNA to FGFR-1 had no further effect on ERK activation. Our results provide support for a novel mechanism by which PDGF-BB induces the release of bFGF and activation of FGFR-1 followed by the sustained activation of ERK and proliferation of human SMCs.
Collapse
MESH Headings
- Aorta, Abdominal
- Becaplermin
- Cell Division/drug effects
- Cell Movement/drug effects
- Cells, Cultured/drug effects
- Cells, Cultured/metabolism
- Chromones/pharmacology
- DNA Replication/drug effects
- Enzyme Activation/drug effects
- Fibroblast Growth Factor 2/metabolism
- Fibroblast Growth Factor 2/pharmacology
- Fibroblast Growth Factor 2/physiology
- Flavonoids/pharmacology
- Heparin/pharmacology
- Humans
- Indoles/pharmacology
- MAP Kinase Kinase 1/metabolism
- MAP Kinase Kinase 2/metabolism
- Maleimides/pharmacology
- Mitogen-Activated Protein Kinase 1/metabolism
- Mitogen-Activated Protein Kinase 3/metabolism
- Morpholines/pharmacology
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Phosphorylation/drug effects
- Platelet-Derived Growth Factor/pharmacology
- Protein Kinase Inhibitors/pharmacology
- Protein Processing, Post-Translational/drug effects
- Protein Serine-Threonine Kinases/metabolism
- Proto-Oncogene Proteins/metabolism
- Proto-Oncogene Proteins c-akt
- Proto-Oncogene Proteins c-sis
- RNA, Small Interfering/pharmacology
- Receptor Protein-Tyrosine Kinases/genetics
- Receptor Protein-Tyrosine Kinases/physiology
- Receptor, Fibroblast Growth Factor, Type 1
- Receptors, Fibroblast Growth Factor/genetics
- Receptors, Fibroblast Growth Factor/physiology
- Recombinant Proteins/pharmacology
- Tyrphostins/pharmacology
Collapse
Affiliation(s)
- Esther Millette
- University of Washington School of Medicine, Department of Surgery, Box 356410, 1959 NE Pacific St, Seattle, WA 98195-6410, USA.
| | | | | | | | | | | |
Collapse
|
89
|
Forsten-Williams K, Chua CC, Nugent MA. The kinetics of FGF-2 binding to heparan sulfate proteoglycans and MAP kinase signaling. J Theor Biol 2004; 233:483-99. [PMID: 15748910 DOI: 10.1016/j.jtbi.2004.10.020] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2004] [Revised: 10/04/2004] [Accepted: 10/14/2004] [Indexed: 11/20/2022]
Abstract
Binding of growth factors to specific cell surface receptors is the first step in initiating cell signaling cascades that ultimately result in diverse activities such as proliferation, differentiation, and apoptosis. Dimerization and phosphorylation of tyrosine kinase transmembrane receptors is the typical paradigm for this activation but, for many growth factors, cell surface interactions are not limited to a single receptor type. In particular, heparin-binding growth factors, such as fibroblast growth factor-2 (FGF-2), bind to heparan sulfate proteoglycans (HSPG) on the cell surface and within the extracellular matrix (ECM), and these molecules have been viewed as accessory co-receptors serving to facilitate tyrosine kinase receptor binding. Recent studies, however, have indicated that HSPG can directly participate in signal transduction in response to FGF-2 binding. Thus, in the present study, we used mathematical modeling to examine whether the kinetics of formation of the various FGF-2 bound complexes on the cell surface correlate with the activation of the downstream mediators of FGF-2 response, Erk1/2. We find that FGF-2 binding to its receptor correlates well with Erk1/2 activation and that HSPG can modulate this response through its ability to stabilize these ligand receptor complexes. Moreover, we also observed that FGF-2 binding to HSPG correlates strongly with Erk1/2 activation under conditions where there is a loss of receptor activity, and we demonstrate that the relative amounts of signaling and non-signaling HSPG on the cell surface, as well as the presence of competing HSPG in the ECM, can impact the signal potential via this pathway. Thus, the selective regulation of specific HSPG might provide a mechanism for fine tuned modulation of heparin-binding growth factor signaling in cells where signal intensity and duration could direct cellular response toward growth, migration or differentiation.
Collapse
Affiliation(s)
- Kimberly Forsten-Williams
- Department of Chemical Engineering and Virginia Tech-Wake Forest University School of Biomedical Engineering and Sciences, Virginia Polytechnic Institute & State University, 141 Randolph Hall, Blacksburg, VA 24061, USA.
| | | | | |
Collapse
|
90
|
Leivonen M, Lundin J, Nordling S, von Boguslawski K, Haglund C. Prognostic Value of Syndecan-1 Expression in Breast Cancer. Oncology 2004; 67:11-8. [PMID: 15459490 DOI: 10.1159/000080280] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2003] [Accepted: 12/18/2003] [Indexed: 11/19/2022]
Abstract
OBJECTIVE Syndecan-1 is a cell surface heparan sulphate proteoglycan which participates in cell proliferation, cell migration and cell-matrix interactions. Epithelial syndecan-1 expression is reduced in several malignant tumours, but in breast and pancreatic cancer, increased expression has also been described. Loss of epithelial syndecan-1 has been associated with poor prognosis in some forms of cancer, but previous findings in breast cancer have been contradictory. The objective of this study was to evaluate the prognostic value of the immunohistochemical expression of syndecan-1 in a series of 200 patients with invasive breast cancer with a median follow-up of 17 years. METHODS Formalin-fixed paraffin-embedded specimens were stained using a monoclonal antibody against syndecan-1. RESULTS Syndecan-1 was expressed in the epithelium in 61% and in the stroma in 67% of the tumours. Epithelial syndecan-1 expression was associated with negative oestrogen receptor (ER) status (p < 0.01), and stromal syndecan-1 expression with positive ER status (p = 0.02). The breast cancer-specific 10-year overall survival for patients with epithelial syndecan-1 expression was 65%, compared with 82% for those with loss of epithelial expression (p = 0.02). Ten-year survival was 66% for those expressing stromal syndecan-1 and 83% for those lacking stromal expression (p = 0.15). Patients with both epithelial and stromal expression had a 10-year survival of only 56%, compared to 78% in patients with other expression pattern combinations (p < 0.002). In Cox multivariate analysis, only axillary involvement and tumour size were significant predictors of breast cancer-specific survival. CONCLUSION Concomitant expression of syndecan-1 in both epithelium and stroma may be a predictor of unfavourable prognosis in breast cancer, and in contrast with previous studies, loss of epithelial syndecan-1 was associated with a more favourable prognosis.
Collapse
Affiliation(s)
- Marja Leivonen
- Department of Surgery, Peijas Hospital, Helsinki University Central Hospital, Vantaa, Finland.
| | | | | | | | | |
Collapse
|
91
|
Nakano K, Okada Y, Saito K, Tanaka Y. Induction of RANKL expression and osteoclast maturation by the binding of fibroblast growth factor 2 to heparan sulfate proteoglycan on rheumatoid synovial fibroblasts. ACTA ACUST UNITED AC 2004; 50:2450-8. [PMID: 15334457 DOI: 10.1002/art.20367] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
OBJECTIVE Rheumatoid arthritis (RA) is characterized by progressive joint destruction. The aim of this study was to clarify the relevance of RA synovial fibroblasts (RASFs) and fibroblast growth factor 2 (FGF-2), which is produced abundantly by RASFs, to the osteoclastogenesis and bone resorption in RA. METHODS Synovial fibroblasts were prepared from the synovial tissues of 10 patients with active RA and 7 patients with osteoarthritis (OA). The expression of RANKL, intercellular adhesion molecule 1 (ICAM-1), FGF receptor 1 (FGFR-1), and heparan sulfate proteoglycan (HSPG) on synovial fibroblasts was measured by FACScan. Osteoclast formation in cocultures of RASFs and peripheral blood mononuclear cells (PBMCs) was evaluated by tartrate-resistant acid phosphatase staining and a pit-formation assay using dentin slices. RESULTS FGF-2 induced the expression of both RANKL and ICAM-1 on RASFs more so than on OA synovial fibroblasts (OASFs). FGF-2-induced up-regulation of RANKL and ICAM-1 was inhibited by anti-FGF-2 antibody. Although FGFR-1 was equally expressed on RASFs and OASFs, HSPG was highly expressed on RASFs. Up-regulation of RANKL by FGF-2 on RASFs was diminished by the removal of heparan sulfate with heparitinase. Osteoclast formation from PBMCs induced by RASFs was inhibited by the addition of either heparitinase, anti-ICAM-1 antibody, anti-FGF-2 antibody, or osteoprotegerin. FGF-2-induced RANKL on RASFs and osteoclast formation were suppressed by an inhibitor of ERK. CONCLUSION FGF-2 was transferred to FGFR-1 through binding to HSPG, which is characteristically expressed on RASFs, resulting in RANKL- and ICAM-1-mediated maturation of osteoclasts via ERK activation. Thus, we propose that FGF-2 not only augments the proliferation of RASFs, but also is involved in osteoclast maturation, which leads to bone destruction in RA.
Collapse
Affiliation(s)
- Kazuhisa Nakano
- University of Occupational and Environmental Health, Kitakyushu, Japan
| | | | | | | |
Collapse
|
92
|
Filla MS, David G, Weinreb RN, Kaufman PL, Peters DM. Distribution of syndecans 1-4 within the anterior segment of the human eye: expression of a variant syndecan-3 and matrix-associated syndecan-2. Exp Eye Res 2004; 79:61-74. [PMID: 15183101 DOI: 10.1016/j.exer.2004.02.010] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2003] [Accepted: 02/20/2004] [Indexed: 11/23/2022]
Abstract
Control of the actomyosin network plays a role in regulating the movement of aqueous humor through the anterior segment of the eye. Receptors that could control its activity are unknown. In this study, we show that all four members of the syndecan family, which can regulate the actomyosin network, are present within the anterior segment. In both sections of human anterior segments and cultures of human trabecular meshwork (HTM), Schlemm's canal (HSC) and the ciliary muscle (HCM) cells from the anterior segment, syndecans-3 and -4 were the predominant family members. They were widely distributed throughout the anterior segment. Syndecan-3 within the anterior segment was a novel, recently described variant 55 kDa form. Low levels of syndecans-1 and -2 were also observed in situ and in all three cultures. Their expression was weaker and more localized than that observed for syndecans-3 and -4. Staining for syndecan-1 in HCM cultures was variable. In HTM and HSC cultures, syndecan-2 also co-distributed with fibronectin, laminin and type IV collagen suggesting that it was shed and associated with the extracellular matrix. Western blots supported this idea and showed syndecan-2 ectodomains in lysates from anterior segments.
Collapse
Affiliation(s)
- Mark S Filla
- Department of Ophthalmology & Visual Sciences, University of Wisconsin, Madison, USA
| | | | | | | | | |
Collapse
|
93
|
Santin AD, Zhan F, Bellone S, Palmieri M, Cane S, Gokden M, Roman JJ, O'Brien TJ, Tian E, Cannon MJ, Shaughnessy J, Pecorelli S. Discrimination between uterine serous papillary carcinomas and ovarian serous papillary tumours by gene expression profiling. Br J Cancer 2004; 90:1814-24. [PMID: 15208622 PMCID: PMC2409747 DOI: 10.1038/sj.bjc.6601791] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
High-grade ovarian serous papillary cancer (OSPC) and uterine serous papillary carcinoma (USPC) represent two histologically similar malignancies characterised by markedly different biological behavior and response to chemotherapy. Understanding the molecular basis of these differences may significantly refine differential diagnosis and management, and may lead to the development of novel, more specific and more effective treatment modalities for OSPC and USPC. We used an oligonucleotide microarray with probe sets complementary to >10 000 human genes to determine whether patterns of gene expression may differentiate OSPC from USPC. Hierarchical cluster analysis of gene expression in OSPC and USPC identified 116 genes that exhibited >two-fold differences (P<0.05) and that readily distinguished OSPC from USPC. Plasminogen activator inhibitor (PAI-2) was the most highly overexpressed gene in OSPC when compared to USPC, while c-erbB2 was the most strikingly overexpressed gene in USPC when compared to OSPC. Overexpression of the c-erbB2 gene and its expression product (i.e., HER-2/neu receptor) was validated by quantitative RT-PCR as well as by flow cytometry on primary USPC and OSPC, respectively. Immunohistochemical staining of serous tumour samples from which primary OSPC and USPC cultures were derived as well as from an independent set of 20 clinical tissue samples (i.e., 10 OSPC and 10 USPC) further confirmed HER-2/neu as a novel molecular diagnostic and therapeutic marker for USPC. Gene expression fingerprints have the potential to predict the anatomical site of tumour origin and readily identify the biologically more aggressive USPC from OSPC. A therapeutic strategy targeting HER-2/neu may be beneficial in patients harbouring chemotherapy-resistant USPC.
Collapse
Affiliation(s)
- A D Santin
- Department of Obstetrics & Gynecology, Division of Gynecologic Oncology, University of Arkansas for Medical Sciences, UAMS Medical Center, 4301 West Markham, Little Rock, AR 72205-7199, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
94
|
Elenius V, Götte M, Reizes O, Elenius K, Bernfield M. Inhibition by the soluble syndecan-1 ectodomains delays wound repair in mice overexpressing syndecan-1. J Biol Chem 2004; 279:41928-35. [PMID: 15220342 DOI: 10.1074/jbc.m404506200] [Citation(s) in RCA: 82] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Wound repair is a tightly regulated process stimulated by proteases, growth factors, and chemokines, which are modulated by heparan sulfate. To characterize further the role of the heparan sulfate proteoglycan syndecan-1 in wound repair, we generated mice overexpressing syndecan-1 (Snd/Snd) and studied dermal wound repair. Wound closure, reepithelialization, granulation tissue formation, and remodeling were delayed in Snd/Snd mice. Soluble syndecan-1 was increased, and shedding was prolonged in wounds from Snd/Snd mice. Excess syndecan-1 increased the elastolytic activity of wound fluids. Additionally, cells in the granulation tissue and keratinocytes at wound edges showed markedly reduced proliferation rates in Snd/Snd mice. Skin grafting experiments between Snd/Snd and control mice indicated that the slower growth rate was mainly due to a soluble factor in the Snd/Snd mouse skin. Syndecan-1 immunodepletion and further degradation experiments identified syndecan-1 ectodomain as a dominant negative inhibitor of cell proliferation. These studies indicate that shed syndecan-1 ectodomain may enhance proteolytic activity and inhibit cell proliferation during wound repair.
Collapse
Affiliation(s)
- Varpu Elenius
- Department of Pediatrics, Children's Hospital and Harvard Medical School, Boston, Massachusetts 02115, USA.
| | | | | | | | | |
Collapse
|
95
|
Velleman SG, Liu X, Coy CS, McFarland DC. Effects of Syndecan-1 and Glypican on Muscle Cell Proliferation and Differentiation: Implications for Possible Functions During Myogenesis. Poult Sci 2004; 83:1020-7. [PMID: 15206631 DOI: 10.1093/ps/83.6.1020] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The heparan sulfate proteoglycans, syndecan-1 and glypican, are low-affinity receptors for fibroblast growth factor 2 (FGF2). Because FGF2 is a potent stimulator of skeletal muscle cell proliferation and a strong inhibitor of differentiation, it is likely that changes in syndecan-1 and glypican expression will affect myogenesis as both, in part, regulate FGF-dependent signaling. In the current study, expression vector constructs containing either syndecan-1 or glypican were transfected into turkey myogenic satellite cells resulting in the overexpression of these genes. The amount of expression of each of these genes was measured by semiquantitative reverse transcriptase polymerase chain reaction. The satellite cell cultures overexpressing syndecan-1 were unable to fuse to form multinucleated myotubes after differentiation was induced. The syndecan-1-transfected cells maintained a rounded morphology typical of cells during proliferation. In contrast, the satellite cells transfected with glypican formed larger myotubes. These results suggest that both syndecan-1 and glypican play pivotal, but different, roles in both muscle cell proliferation and differentiation.
Collapse
Affiliation(s)
- S G Velleman
- Department of Animal Sciences, Ohio Agricultural Research and Development Center, The Ohio State University, Wooster, Ohio 44691, USA.
| | | | | | | |
Collapse
|
96
|
Vincent T, Mechti N. IL-6 regulates CD44 cell surface expression on human myeloma cells. Leukemia 2004; 18:967-75. [PMID: 15014527 DOI: 10.1038/sj.leu.2403333] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Multiple myeloma (MM) is a progressive B-lineage neoplasia characterized by the accumulation of slow proliferative malignant plasma cells in the bone marrow compartment where the microenvironment seems to be favorable for their growth and survival. Heparan sulfate proteoglycans such as syndecan-1 and CD44 are thought to play a central role in the survival signals provided by these bone marrow survival niches, which require complex interactions between myeloma cells, extracellular matrix, stromal cells and soluble factors. In this report, we demonstrate that interleukin-6 (IL-6), the main survival and growth factor for myeloma cells, strongly increases CD44 gene expression. In addition, we show that IL-6 modulates CD44 RNA alternative splicing and induces the overexpression of all CD44 variant exons. Finally, we show that IL-6-induced CD44 cell surface molecules have a functional polarized membrane distribution. As IL-6 secretion induced from bone marrow stromal cells by myeloma cells is partly mediated through direct cell-to-cell interaction involving CD44 adhesion molecules, our findings suggest that a CD44/IL-6 amplification loop plays a crucial role in myeloma cell survival.
Collapse
Affiliation(s)
- T Vincent
- INSERM Unité U475, Montpellier Cedex, France
| | | |
Collapse
|
97
|
Bloechlinger S, Karchewski LA, Woolf CJ. Dynamic changes in glypican-1 expression in dorsal root ganglion neurons after peripheral and central axonal injury. Eur J Neurosci 2004; 19:1119-32. [PMID: 15016071 DOI: 10.1111/j.1460-9568.2004.03262.x] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Glypican-1, a glycosyl phosphatidyl inositol (GPI)-anchored heparan sulphate proteoglycan expressed in the developing and mature cells of the central nervous system, acts as a coreceptor for diverse ligands, including slit axonal guidance proteins, fibroblast growth factors and laminin. We have examined its expression in primary sensory dorsal root ganglion (DRG) neurons and spinal cord after axonal injury. In noninjured rats, glypican-1 mRNA and protein are constitutively expressed at low levels in lumbar DRGs. Sciatic nerve transection results in a two-fold increase in mRNA and protein expression. High glypican-1 expression persists until the injured axons reinnervate their peripheral targets, as in the case of a crushed nerve. Injury to the central axons of DRG neurons by either a dorsal column injury or a dorsal root transection also up-regulates glypican-1, a feature that differs from most DRG axonal injury-induced genes, whose regulation changes only after peripheral and not central axonal injury. After axonal injury, the cellular localization of glypican-1 changes from a nuclear pattern restricted to neurons in noninjured DRGs, to the cytoplasm and membrane of injured neurons, as well as neighbouring non-neuronal cells. Sciatic nerve transection also leads to an accumulation of glypican-1 in the proximal nerve segment of injured axons. Glypican-1 is coexpressed with robo 2 and its up-regulation after axonal injury may contribute to an altered sensitivity to axonal growth or guidance cues.
Collapse
Affiliation(s)
- Stefan Bloechlinger
- Department of Anaesthesia and Critical Care, Massachusetts General Hospital and Harvard Medical School, MGH-East, 149 13th Street, Rm 4309, Charlestown, MA 02129, USA
| | | | | |
Collapse
|
98
|
Yuan K, Hong TM, Chen JJW, Tsai WH, Lin MT. Syndecan-1 up-regulated by ephrinB2/EphB4 plays dual roles in inflammatory angiogenesis. Blood 2004; 104:1025-33. [PMID: 15126321 DOI: 10.1182/blood-2003-09-3334] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
EphrinB2 and EphB4, its cognate receptor, are important in the vascular development of the mouse embryo. Their roles in human inflammatory angiogenesis, however, are not well understood. By examining hyperinflammatory lesions, we saw that ephrinB2 was predominantly expressed in macrophage-like cells and EphB4 in small venules. Because macrophages usually transmigrate through postcapillary venules during inflammation, we wanted to explore the downstream effects of EphB4 after binding to ephrinB2. By using cDNA microarray technique and following reverse transcriptase-polymerase chain reaction (RT-PCR), we found that syntenin and syndecan-1 were up-regulated in EphB4-positive endothelial cells dose dependently and time dependently after stimulation with preclustered ephrinB2. In vitro, ephrinB2 suppressed the angiogenic effects of basic fibroblast growth factor (bFGF) on EphB4-positive endothelial cells, partially due to syndecan-1's competition with fibroblast growth factor receptor (FGFR) for bFGF. However, ephrinB2 exhibited angiogenic effects in vivo, possibly due to an inflammation-associated enzyme-heparanase. The enzymes could convert the inhibitory effect of ephrinB2 on EphB4-positive endothelial cells to an activating effect by removing poorly sulfated side chains of up-regulated syndecan-1 ectodomain. Depending on the presence of heparanases, the roles of syndecan-1 may be opposite in different physiological settings.
Collapse
Affiliation(s)
- Kuo Yuan
- Institute of Medical Sciences, Tzu Chi University, No. 701, Jung-Yang Rd, Section 3, Hualien, Taiwan 970
| | | | | | | | | |
Collapse
|
99
|
Maeda T, Alexander CM, Friedl A. Induction of syndecan-1 expression in stromal fibroblasts promotes proliferation of human breast cancer cells. Cancer Res 2004; 64:612-21. [PMID: 14744776 DOI: 10.1158/0008-5472.can-03-2439] [Citation(s) in RCA: 107] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Infiltrating carcinomas characteristically elicit a reactive stromal response, and accumulating evidence indicates that tumor stroma fibroblasts reciprocally promote tumor development and growth. The cell surface heparan sulfate proteoglycan, syndecan-1 (Sdc1), is thought to function as a coreceptor for growth factor and extracellular matrix interactions, and Sdc1 expression is induced in reactive stromal cells in both mice and man. Mice with a targeted mutation in Sdc1 show reduced tumor development in response to oncogene expression and altered responses to other pathological stimuli that are associated with the induction of stromal Sdc1. Here, we test the hypothesis that Sdc1 is required for the growth-promoting activities of reactive stroma. We found that when highly invasive carcinoma cells (MDA-MB-231) were placed in contact with mouse embryonic fibroblasts (MEFs) in a coculture model, Sdc1 expression was induced. Sdc1 was not induced by less invasive or normal cell lines (T47D and NMuMG). Furthermore, the growth of MDA-MB-231 cells was enhanced by 42% when cocultured with Sdc1+/+ MEFs compared with Sdc1-/- MEFs. When T47D cells were cocultured with fibroblasts that expressed transfected Sdc1, these Sdc1-positive fibroblasts stimulated growth of the breast epithelial cells by 85% compared with untransfected controls. The growth-promoting effect was completely abolished when fibroblasts were transfected with mutant Sdc1 lacking heparan sulfate attachment sites. In conclusion, we have demonstrated that a growth-promoting loop exists between breast cancer cells and their stroma that depends on the activity of glycanated Sdc1.
Collapse
Affiliation(s)
- Takashi Maeda
- Pathology and Laboratory Medicine Service, Department of Veterans Affairs Medical Center, Madison, Wisconsin, USA
| | | | | |
Collapse
|
100
|
Abstract
Transforming growth factor-beta (TGF-beta) has multiple functions including increasing extracellular matrix deposition in fibrosis. It functions through a complex family of cell surface receptors that mediate downstream signaling. We report here that a transmembrane heparan sulfate proteoglycan, syndecan-2 (S2), can regulate TGF-beta signaling. S2 protein increased in the renal interstitium in diabetes and regulated TGF-beta-mediated increased matrix deposition in vitro. Transfection of renal papillary fibroblasts with S2 or a S2 construct that has a truncated cytoplasmic domain (S2DeltaS) promoted TGF-beta binding and S2 core protein ectodomain directly bound TGF-beta. Transfection with S2 increased the amounts of type I and type II TGF-beta receptors (TbetaRI and TbetaRII), whereas S2DeltaS was much less effective. In contrast, S2DeltaS dramatically increased the level of type III TGF-beta receptor (TbetaRIII), betaglycan, whereas S2 resulted in a decrease. Syndecan-2 specifically co-immunoprecipitated with betaglycan but not with TbetaRI or TbetaRII. This is a novel mechanism of control of TGF-beta action that may be important in fibrosis.
Collapse
Affiliation(s)
- Ligong Chen
- Department of Cell Biology, University of Alabama at Birmingham, Birmingham, Alabama 35294, USA
| | | | | |
Collapse
|