Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Int J Med Sci ; 18(16): 3759-3767, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34790051

RESUMEN

Background: YKL-40, a secreted glycoprotein, has a role in promoting tumor angiogenesis through syndecan-1 receptor. Syndecan-4 is a member of syndecan family. However, the effects of YKL-40 on migration and tube formation of human umbilical vein cells (HUVECs) mediated by syndecan-4 receptor are unknown. Materials and methods: HUVECs were transfected with lentivirus encoding syndecan-4 short hairpin (sh) RNAs (lenti-synd4 shRNAs) and the efficiency of transfection was measured using qRT-PCR and western blotting. The effects of recombinant protein of YKL-40 on migration and angiogenesis of HUVECs adjusted by syndecan-4 were determined by wound healing and tube formation assay. The expressions of protein kinase Cα (PKCα) and extracellular signal regulated kinases (ERKs) 1 and 2 (ERK1/2) in HUVECs were measured using western blotting. Results: The mRNA and protein expression of syndecan-4 were significantly decreased in HUVECs successfully transfected with lenti-synd4 shRNAs. Lenti-synd4 shRNAs remarkably inhibited the migration and tube formation of HUVECs stimulated by recombinant protein of YKL-40. The levels of PKCα and ratio of p-ERK1/2 to ERK1/2 in HUVECs were also decreased by down-regulating syndecan-4. Conclusion: The effects of YKL-40 on migration and tube formation of HUVECs are partly inhibited by knock-downing syndecan-4 through suppressing PKCα and ERK1/2 signaling pathways.


Asunto(s)
Proteína 1 Similar a Quitinasa-3/fisiología , Células Endoteliales de la Vena Umbilical Humana/fisiología , Neovascularización Fisiológica/genética , Sindecano-4/fisiología , Movimiento Celular/efectos de los fármacos , Movimiento Celular/genética , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Células Cultivadas , Proteína 1 Similar a Quitinasa-3/antagonistas & inhibidores , Células HEK293 , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Humanos , Neovascularización Fisiológica/efectos de los fármacos , ARN Interferente Pequeño/farmacología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Sindecano-4/antagonistas & inhibidores
2.
Int J Biol Sci ; 16(5): 766-776, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32071547

RESUMEN

Syndecan-4 is a member of the polysaccharide syndecan family and plays a vital role in intervertebral disc development. Several studies have demonstrated the positive relationship between syndecan-4 expression and intervertebral disc degeneration. However, the detailed molecular mechanism by which syndecan-4 affects the degeneration of nucleus pulposus cells (NPCs) remains unclear. In this study, cell viability was determined by CCK-8 assay, mRNA level was determined by qPCR, and protein expression was determined by western blot. Molecular interaction was determined by chromatin immunoprecipitation assay. A rabbit intervertebral disc degeneration model was established to test for syndecan in vivo. We found that the morphology and viability of NPCs were not affected by the expression of syndecan-4 in the long term. While the NPC function were affected, which results in the degeneration of intervertebral disc. Syndecan-4 overexpression promoted the degeneration of NPCs. Syndecan-4 also activated the JNK signaling pathway and downstream p53 pathways, and promoted degeneration. Inhibition of the JNK pathway, which down-regulated p53 expression, alleviated the degeneration. In an in vivo study, syndecan-4 siRNA injection stopped the development of rabbit disc degeneration, and even created a reverse effect, in which JNK/p53 played a role. Syndecan-4 may be a novel therapeutic target for intervertebral disc degeneration via suppressing the JNK/p53 pathway.


Asunto(s)
Degeneración del Disco Intervertebral/metabolismo , Núcleo Pulposo/metabolismo , Sindecano-4/fisiología , Agrecanos/genética , Agrecanos/metabolismo , Animales , Western Blotting , Inmunoprecipitación de Cromatina , Humanos , Degeneración del Disco Intervertebral/genética , Degeneración del Disco Intervertebral/patología , Sistema de Señalización de MAP Quinasas/genética , Sistema de Señalización de MAP Quinasas/fisiología , Imagen por Resonancia Magnética , Núcleo Pulposo/patología , Reacción en Cadena de la Polimerasa , Unión Proteica , Proteínas Proto-Oncogénicas c-mdm2/genética , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Conejos , Transducción de Señal/genética , Transducción de Señal/fisiología , Sindecano-4/genética , Sindecano-4/metabolismo
3.
Ann Biomed Eng ; 48(1): 357-366, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31520333

RESUMEN

The lipid membrane of endothelial cells plays a pivotal role in maintaining normal circulatory system functions. To investigate the response of the endothelial cell membrane to changes in vascular conditions, an atomistic model of the lipid membrane interspersed with Syndecan-4 core protein was established based on experimental observations and a series of molecular dynamics simulations were undertaken. The results show that flow results in continuous deformation of the lipid membrane, and the degree of membrane deformation is not in monotonic relationship with the environmental changes (either the changes in blood velocity or the alteration of the core protein configuration). An explanation for such non-monotonic relationship is provided, which agrees with previous experimental results. The elevation of the lipid membrane surface around the core protein of the endothelial glycocalyx was also observed, which can be mainly attributed to the Coulombic interactions between the biomolecules therein. The present study demonstrates that the blood flow can deform the lipid membrane directly via the interactions between water molecules and lipid membrane atoms thereby affecting mechanosensing; it also presents an additional force transmission pathway from the flow to the lipid membrane via the glycocalyx core protein, which complements previous mechanotransduction hypothesis.


Asunto(s)
Membrana Celular/fisiología , Células Endoteliales/fisiología , Sindecano-4/fisiología , Simulación de Dinámica Molecular , Flujo Sanguíneo Regional , Agua/fisiología
4.
Sci Rep ; 9(1): 10175, 2019 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-31308409

RESUMEN

Extravillous trophoblast (EVT) invasion is an essential component of human placentation. Poor EVT invasion is associated with obstetrical complications including preeclampsia. Integration of cues from the extracellular environment is required for directional EVT invasion, but how EVTs coordinate responses to these cues is not well understood. Syndecan-4 (SDC4) is a transmembrane heparan sulfate proteoglycan that binds to, and modulates the activity of, many extracellular proteins implicated in placental development. Therefore, we determined the functional importance of SDC4 for EVT invasion. We found that SDC4 is expressed by a first trimester EVT line (HTR8), and in EVTs in placenta throughout pregnancy, with higher expression during early pregnancy than at term. Higher expression was also observed in placentas from preeclampsia compared to normotensive pregnancies. SDC4-deficient HTR8 EVTs exhibited reduced migration and Matrigel-based invasion, both under basal conditions and following exposure to basic fibroblast growth factor and heparin-binding epidermal growth factor. SDC4-deficient HTR8 EVTs also showed reduced protein kinase C-alpha (PKCα) and AKT phosphorylation. SDC4 directly bound to activated PKCα in EVTs, and inhibition of PKCα decreased EVT invasion and migration. Our findings reveal an essential role of SDC4 as a regulator of EVT motility, in part through coordination of PKCα activation.


Asunto(s)
Placenta/metabolismo , Placentación/fisiología , Sindecano-4/metabolismo , Adulto , Línea Celular , Movimiento Celular/fisiología , Femenino , Edad Gestacional , Humanos , Preeclampsia/metabolismo , Embarazo , Primer Trimestre del Embarazo , Proteína Quinasa C/metabolismo , Transducción de Señal , Sindecano-4/fisiología , Trofoblastos/metabolismo
5.
Nat Commun ; 10(1): 1562, 2019 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-30952866

RESUMEN

The proteoglycan Syndecan-2 (Sdc2) has been implicated in regulation of cytoskeleton organization, integrin signaling and developmental angiogenesis in zebrafish. Here we report that mice with global and inducible endothelial-specific deletion of Sdc2 display marked angiogenic and arteriogenic defects and impaired VEGFA165 signaling. No such abnormalities are observed in mice with deletion of the closely related Syndecan-4 (Sdc4) gene. These differences are due to a significantly higher 6-O sulfation level in Sdc2 versus Sdc4 heparan sulfate (HS) chains, leading to an increase in VEGFA165 binding sites and formation of a ternary Sdc2-VEGFA165-VEGFR2 complex which enhances VEGFR2 activation. The increased Sdc2 HS chains 6-O sulfation is driven by a specific N-terminal domain sequence; the insertion of this sequence in Sdc4 N-terminal domain increases 6-O sulfation of its HS chains and promotes Sdc2-VEGFA165-VEGFR2 complex formation. This demonstrates the existence of core protein-determined HS sulfation patterns that regulate specific biological activities.


Asunto(s)
Neovascularización Fisiológica/genética , Sindecano-2/fisiología , Factor A de Crecimiento Endotelial Vascular/fisiología , Animales , Ratones , Dominios Proteicos , Retina/crecimiento & desarrollo , Análisis de Secuencia de Proteína , Sindecano-2/genética , Sindecano-2/metabolismo , Sindecano-4/genética , Sindecano-4/metabolismo , Sindecano-4/fisiología , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/fisiología
6.
Cell Mol Life Sci ; 76(5): 865-871, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-30465083

RESUMEN

Prior to the cytokinesis, the cell-matrix interactions should be disrupted, and the mitotic cells round up. Prerequisite of mitosis, the centrosomes duplicate, spindle fibers are generated and move away from each other to opposite sides of the cells marking the cell poles. Later, an invagination in the plasma membrane is formed a few minutes after anaphase. This furrow ingression is driven by a contractile actomyosin ring, whose assembly is regulated by RhoA GTPase. At the completion of cytokinesis, the two daughter cells are still connected by a thin intercellular bridge, which is subjected to abscission, as the terminal step of cytokinesis. Here, it is overviewed, how syndecan-4, a transmembrane, heparan sulfate proteoglycan, can contribute to these processes in a phosphorylation-dependent manner.


Asunto(s)
División Celular , Proteoglicanos de Heparán Sulfato/fisiología , Actinas/química , Animales , Citocinesis , Humanos , Mitosis , Sindecano-4/fisiología , Proteína de Unión al GTP rhoA/fisiología
7.
Sci Rep ; 6: 36818, 2016 11 10.
Artículo en Inglés | MEDLINE | ID: mdl-27830760

RESUMEN

The PDZ domain-containing scaffold protein, syntenin-1, binds to the transmembrane proteoglycan, syndecan-4, but the molecular mechanism/function of this interaction are unknown. Crystal structure analysis of syntenin-1/syndecan-4 cytoplasmic domains revealed that syntenin-1 forms a symmetrical pair of dimers anchored by a syndecan-4 dimer. The syndecan-4 cytoplasmic domain is a compact intertwined dimer with a symmetrical clamp shape and two antiparallel strands forming a cavity within the dimeric twist. The PDZ2 domain of syntenin-1 forms a direct antiparallel interaction with the syndecan-4 cytoplasmic domain, inhibiting the functions of syndecan-4 such as focal adhesion formation. Moreover, C-terminal region of syntenin-1 reveals an essential role for enhancing the molecular homodimerization. Mutation of key syntenin-1 residues involved in the syndecan-4 interaction or homodimer formation abolishes the inhibitory function of syntenin-1, as does deletion of the homodimerization-related syntenin-1 C-terminal domain. Syntenin-1, but not dimer-formation-incompetent mutants, rescued the syndecan-4-mediated inhibition of migration and pulmonary metastasis by B16F10 cells. Therefore, we conclude that syntenin-1 negatively regulates syndecan-4 function via oligomerization and/or syndecan-4 interaction, impacting cytoskeletal organization and cell migration.


Asunto(s)
Sindecano-4/química , Sinteninas/química , Secuencia de Aminoácidos , Animales , Línea Celular Tumoral , Movimiento Celular , Cristalografía por Rayos X , Humanos , Metástasis Linfática , Melanoma Experimental/metabolismo , Melanoma Experimental/secundario , Ratones , Ratones Endogámicos C57BL , Modelos Moleculares , Unión Proteica , Conformación Proteica en Hélice alfa , Dominios y Motivos de Interacción de Proteínas , Multimerización de Proteína , Ratas , Transducción de Señal , Neoplasias Cutáneas/metabolismo , Neoplasias Cutáneas/patología , Sindecano-4/fisiología , Sinteninas/fisiología
8.
Nat Commun ; 7: 11848, 2016 06 09.
Artículo en Inglés | MEDLINE | ID: mdl-27279133

RESUMEN

Retinoic acid-inducible gene I (RIG-I) plays important roles in pathogen recognition and antiviral signalling transduction. Here we show that syndecan-4 (SDC4) is a RIG-I-interacting partner identified in a yeast two-hybrid screen. We find that SDC4 negatively regulates the RIG-I-mediated antiviral signalling in a feedback-loop control manner. The genetic evidence obtained by using knockout mice further emphasizes this biological role of SDC4 in antiviral signalling. Mechanistically, we show that SDC4 interacts with both RIG-I and deubiquitinase CYLD via its carboxyl-terminal intracellular region. SDC4 likely promotes redistribution of RIG-I and CYLD in a perinuclear pattern post viral infection, and thus enhances the RIG-I-CYLD interaction and potentiates the K63-linked deubiquitination of RIG-I. Collectively, our findings uncover a mechanism by which SDC4 antagonizes the activation of RIG-I in a CYLD-mediated deubiquitination-dependent process, thereby balancing antiviral signalling to avoid deleterious effects on host cells.


Asunto(s)
Cisteína Endopeptidasas/fisiología , Inmunidad Innata/genética , Proteínas de la Membrana/fisiología , Proteínas del Tejido Nervioso/fisiología , Sindecano-4/fisiología , Animales , Cisteína Endopeptidasas/genética , Cisteína Endopeptidasas/metabolismo , Enzima Desubiquitinante CYLD , Inmunidad Innata/fisiología , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Noqueados , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Receptores de Superficie Celular , Transducción de Señal , Sindecano-4/genética , Sindecano-4/metabolismo , Ubiquitinación
9.
Bioconjug Chem ; 27(4): 1119-30, 2016 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-27019270

RESUMEN

Arginine-rich cell-penetrating peptides (CPPs) such as Tat and oligoarginine peptides have been widely used as carriers for intracellular delivery of bioactive molecules. Despite accumulating evidence for involvement of endocytosis in the cellular uptake of arginine-rich CPPs, the primary cell-surface receptors for these peptide carriers that would initiate endocytic processes leading to intracellular delivery of bioactive cargoes have remained poorly understood. Our previous attempt to identify membrane receptors for octa-arginine (R8) peptide, one of the representative arginine-rich CPPs, using the photo-cross-linking probe bearing a photoreactive diazirine was not successful due to considerable amounts of cellular proteins nonspecifically bound to the affinity beads. To address this issue, here we developed a photo-cross-linking probe in which a cleavable linker of a diazobenzene moiety was employed to allow selective elution of cross-linked proteins by reducing agent-mediated cleavage. We demonstrated that introduction of the diazobenzene moiety into the photoaffinity probe enables efficient purification of cross-linked proteins with significant reduction of nonspecific binding proteins, leading to successful identification of 17 membrane-associated proteins that would interact with R8 peptide. RNAi-mediated knockdown experiments in combination with the pharmacological inhibitors revealed that, among the proteins identified, syndecan-4, one of the heparan sulfate proteoglycans, is an endogenous membrane-associated receptor for the cellular uptake of R8 peptide via clathrin-mediated endocytosis. This syndecan-4-dependent pathway was also involved in the intracellular delivery of bioactive proteins mediated by R8 peptide. These results reveal that syndecan-4 is a primary cell-surface target for R8 peptide that allows intracellular delivery of bioactive cargo molecules via clathrin-mediated endocytosis.


Asunto(s)
Arginina/metabolismo , Endocitosis/fisiología , Sindecano-4/metabolismo , Humanos , Proteínas de la Membrana/metabolismo , Sindecano-4/fisiología
10.
Biochim Biophys Acta ; 1853(1): 201-12, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25449226

RESUMEN

Tissue transglutaminase (TG2) is a multifunctional protein cross-linking enzyme that has been implicated in apoptotic cell clearance but is also important in many other cell functions including cell adhesion, migration and monocyte to macrophage differentiation. Cell surface-associated TG2 regulates cell adhesion and migration, via its association with receptors such as syndecan-4 and ß1 and ß3 integrins. Whilst defective apoptotic cell clearance has been described in TG2-deficient mice, the precise role of TG2 in apoptotic cell clearance remains ill-defined. Our work addresses the role of macrophage extracellular TG2 in apoptotic cell corpse clearance. Here we reveal TG2 expression and activity (cytosolic and cell surface) in human macrophages and demonstrate that inhibitors of protein crosslinking activity reduce macrophage clearance of dying cells. We show also that cell-impermeable TG2 inhibitors significantly inhibit the ability of macrophages to migrate and clear apoptotic cells through reduced macrophage recruitment to, and binding of, apoptotic cells. Association studies reveal TG2-syndecan-4 interaction through heparan sulphate side chains, and knockdown of syndecan-4 reduces cell surface TG2 activity and apoptotic cell clearance. Furthermore, inhibition of TG2 activity reduces crosslinking of CD44, reported to augment AC clearance. Thus our data define a role for TG2 activity at the surface of human macrophages in multiple stages of AC clearance and we propose that TG2, in association with heparan sulphates, may exert its effect on AC clearance via a mechanism involving the crosslinking of CD44.


Asunto(s)
Apoptosis , Proteínas de Unión al GTP/fisiología , Receptores de Hialuranos/fisiología , Macrófagos/fisiología , Sindecano-4/fisiología , Transglutaminasas/fisiología , Comunicación Celular , Movimiento Celular , Células Cultivadas , Humanos , Proteína Glutamina Gamma Glutamiltransferasa 2
11.
J Am Soc Nephrol ; 25(5): 1013-27, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24357671

RESUMEN

Transglutaminase type 2 (TG2) is an extracellular matrix crosslinking enzyme with a pivotal role in kidney fibrosis. The interaction of TG2 with the heparan sulfate proteoglycan syndecan-4 (Sdc4) regulates the cell surface trafficking, localization, and activity of TG2 in vitro but remains unstudied in vivo. We tested the hypothesis that Sdc4 is required for cell surface targeting of TG2 and the development of kidney fibrosis in CKD. Wild-type and Sdc4-null mice were subjected to unilateral ureteric obstruction and aristolochic acid nephropathy (AAN) as experimental models of kidney fibrosis. Analysis of renal scarring by Masson trichrome staining, kidney hydroxyproline levels, and collagen immunofluorescence demonstrated progressive fibrosis associated with increases in extracellular TG2 and TG activity in the tubulointerstitium in both models. Knockout of Sdc-4 reduced these effects and prevented AAN-induced increases in total and active TGF-ß1. In wild-type mice subjected to AAN, extracellular TG2 colocalized with Sdc4 in the tubular interstitium and basement membrane, where TG2 also colocalized with heparan sulfate chains. Heparitinase I, which selectively cleaves heparan sulfate, completely abolished extracellular TG2 in normal and diseased kidney sections. In conclusion, the lack of Sdc4 heparan sulfate chains in the kidneys of Sdc4-null mice abrogates injury-induced externalization of TG2, thereby preventing profibrotic crosslinking of extracellular matrix and recruitment of large latent TGF-ß1. This finding suggests that targeting the TG2-Sdc4 interaction may provide a specific interventional strategy for the treatment of CKD.


Asunto(s)
Proteínas de Unión al GTP/metabolismo , Nefritis Intersticial/prevención & control , Nefroesclerosis/etiología , Insuficiencia Renal Crónica/complicaciones , Insuficiencia Renal Crónica/metabolismo , Sindecano-4/deficiencia , Sindecano-4/genética , Transglutaminasas/metabolismo , Uveítis/prevención & control , Animales , Ácidos Aristolóquicos , Fibrosis , Proteínas de Unión al GTP/antagonistas & inhibidores , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Nefritis Intersticial/metabolismo , Nefritis Intersticial/patología , Nefroesclerosis/metabolismo , Nefroesclerosis/patología , Proteína Glutamina Gamma Glutamiltransferasa 2 , Insuficiencia Renal Crónica/patología , Sindecano-4/fisiología , Factor de Crecimiento Transformador beta1/metabolismo , Transglutaminasas/antagonistas & inhibidores , Obstrucción Ureteral/metabolismo , Obstrucción Ureteral/patología , Uveítis/metabolismo , Uveítis/patología
12.
Mol Cell Biochem ; 375(1-2): 115-30, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23212449

RESUMEN

Syndecan-4 (S4) is a cell membrane-associated heparan sulfate proteoglycan that forms oligomers in muscle satellite cells. The S4 oligomers activate protein kinase Cα (PKCα) through the S4 cytoplasmic domain and may regulate the activation of ras homolog gene family member A (RhoA), a signal transduction molecule down-stream of PKCα which is thought to influence cell migration. However, little is known about the function of the S4 cytoplasmic domain in satellite cell migration and RhoA activation. The objective of the current study was to determine the function of S4 and its cytoplasmic domain in cell migration and RhoA activation. To study the objective, clones of S4 and S4 without the cytoplasmic domain (S4C) were used in overexpression studies, and small interference RNAs targeting S4 or RhoA were used in knockdown studies. Satellite cell migration was increased by S4 overexpression, but decreased by the knockdown or deletion of the S4 cytoplasmic domain. The RhoA protein was activated by the overexpression of S4, but not with the deletion of the S4 cytoplasmic domain. The treatment of Rho activator II or the knockdown of RhoA also modulated satellite cell migration. Finally, co-transfection (S4 overexpression and RhoA knockdown) and rescue (the knockdown of S4 and the treatment with Rho activator II) studies demonstrated that S4-mediated satellite cell migration was regulated through the activation of RhoA. The cytoplasmic domain of S4 is required for cell migration and RhoA activation which will affect muscle fiber formation.


Asunto(s)
Proteínas Aviares/fisiología , Movimiento Celular , Células Satélite del Músculo Esquelético/fisiología , Sindecano-4/fisiología , Proteína de Unión al GTP rhoA/metabolismo , Animales , Proteínas Aviares/química , Células Cultivadas , Activación Enzimática , Técnicas de Silenciamiento del Gen , Masculino , Proteína Quinasa C-alfa/metabolismo , Estructura Terciaria de Proteína , ARN Interferente Pequeño/genética , Sindecano-4/química , Turquía , Proteína de Unión al GTP rhoA/genética
13.
Arthritis Rheum ; 65(3): 743-52, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23233348

RESUMEN

OBJECTIVE: Syndecan 4, a heparan sulfate proteoglycan, has been associated with osteoarthritis. The present study was undertaken to analyze the functional role of syndecan 4 in endochondral ossification of mouse embryos and in adult fracture repair, which, like osteoarthritis, involves an inflammatory component. METHODS: Sdc4 promoter activity was analyzed in Sdc4(-/-) lacZ-knockin mice, using ß-galactosidase staining. Endochondral ossification in embryos from embryonic day 16.5 was assessed by histologic and immunohistologic staining. Bone fracture repair was analyzed in femora of adult mice on days 7 and 14 postfracture. To evaluate Sdc2 and Sdc4 gene expression with and without tumor necrosis factor α (TNFα) and Wnt-3a stimulation, quantitative real-time polymerase chain reaction was performed. RESULTS: In Sdc4(-/-) lacZ-knockin animals, syndecan 4 promoter activity was detectable at all stages of chondrocyte differentiation, and Sdc4 deficiency inhibited chondrocyte proliferation. Aggrecan turnover in the uncalcified cartilage of the epiphysis was decreased transiently in vivo, but this did not lead to a growth phenotype at birth. In contrast, among adult mice, fracture healing was markedly delayed in Sdc4(-/-) animals and was accompanied by increased callus formation. Blocking of inflammation via anti-TNFα treatment during fracture healing reduced these changes in Sdc4(-/-) mice to levels observed in wild-type controls. We analyzed the differences between the mild embryonic and the severe adult phenotype, and found a compensatory up-regulation of syndecan 2 in the developing cartilage of Sdc4(-/-) mice that was absent in adult tissue. Stimulation of chondrocytes with Wnt-3a in vitro led to increased expression of syndecan 2, while stimulation with TNFα resulted in up-regulation of syndecan 4 but decreased expression of syndecan 2. TNFα stimulation reduced syndecan 2 expression and increased syndecan 4 expression even in the presence of Wnt-3a, suggesting that inflammation has a strong effect on the regulation of syndecan expression. CONCLUSION: Our results demonstrate that syndecan 4 is functionally involved in endochondral ossification and that its loss impairs fracture healing, due to inhibition of compensatory mechanisms under inflammatory conditions.


Asunto(s)
Desarrollo Óseo/fisiología , Fracturas del Fémur/fisiopatología , Curación de Fractura/fisiología , Sindecano-4/fisiología , Animales , Diferenciación Celular/fisiología , Condrocitos/citología , Condrocitos/fisiología , Femenino , Fémur/citología , Fémur/embriología , Fémur/fisiología , Placa de Crecimiento/citología , Placa de Crecimiento/embriología , Placa de Crecimiento/fisiología , Inflamación/fisiopatología , Operón Lac/genética , Masculino , Ratones , Ratones Noqueados , Osteogénesis/fisiología , Embarazo , Regiones Promotoras Genéticas/fisiología , ARN Mensajero/metabolismo , Sindecano-2/genética , Sindecano-2/fisiología , Sindecano-4/genética , Tibia/citología , Tibia/embriología , Tibia/fisiología
14.
Sci Signal ; 5(223): ra36, 2012 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-22569333

RESUMEN

Fibroblast growth factor 2 (FGF2) induces endothelial cell migration and angiogenesis through two classes of receptors: receptor tyrosine kinases, such as FGF receptor 1 (FGFR1), and heparan sulfate proteoglycans, such as syndecan 4 (S4). We examined the distinct contributions of FGFR1 and S4 in shaping the endothelial response to FGF2. S4 determined the kinetics and magnitude of FGF2-induced mitogen-activated protein kinase (MAPK) signaling by promoting the macropinocytosis of the FGFR1-S4-FGF2 signaling complex. Internalization of the S4 receptor complex was independent of clathrin and dynamin, proceeded from lipid raft-enriched membranes, and required activation of the guanosine triphosphatases RhoG and Rab5. Genetic knockout of S4, disruption of S4 function, or inhibition of Rab5 led to increased endocytosis and MAPK signaling. These data define the mechanism by which FGFR1 and S4 coordinate downstream signaling upon FGF2 stimulation: FGFR1 initiates MAPK signaling, whereas S4-dependent FGFR1 macropinocytosis modulates the kinetics of MAPK activation. Our studies identify S4 as a regulator of MAPK signaling and address the question of how distinct classes of FGFRs individually contribute to signal transduction in endothelial cells.


Asunto(s)
Endotelio/metabolismo , Pinocitosis/fisiología , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/metabolismo , Transducción de Señal/fisiología , Sindecano-4/fisiología , Endotelio/citología , Células HeLa , Humanos , Sistema de Señalización de MAP Quinasas
15.
Arterioscler Thromb Vasc Biol ; 32(2): 378-85, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22155451

RESUMEN

OBJECTIVE: Syndecan 4 (Sdc4) modulates signal transduction and regulates activity of protein channels. Sdc4 is essential for the regulation of cellular permeability. We hypothesized that Sdc4 may regulate transient receptor potential canonical 6 (TRPC6) channels, a determinant of glomerular permeability, in a RhoA/Rho-associated protein kinase-dependent manner. METHODS AND RESULTS: Sdc4 knockout (Sdc4(-/-)) mice showed increased glomerular filtration rate and ameliorated albuminuria under baseline conditions and after bovine serum albumin overload (each P<0.05). Using reverse transcription-polymerase chain reaction and immunoblotting, Sdc4(-/-) mice showed reduced TRPC6 mRNA by 79% and TRPC6 protein by 82% (each P<0.05). Sdc4(-/-) mice showed an increased RhoA activity by 87% and increased phosphorylation of ezrin in glomeruli by 48% (each P<0.05). Sdc4 knockdown in cultured podocytes reduced TRPC6 gene expression and reduced the association of TRPC6 with plasma membrane and TRPC6-mediated calcium influx and currents. Sdc4 knockdown inactivated negative regulatory protein Rho GTPase activating protein by 33%, accompanied by a 41% increase in RhoA activity and increased phosphorylation of ezrin (P<0.05). Conversely, overexpression of Sdc4 reduced RhoA activity and increased TRPC6 protein and TRPC6-mediated calcium influx and currents. CONCLUSIONS: Our results establish a previously unknown function of Sdc4 for regulation of TRPC6 channels and support the role of Sdc4 for the regulation of glomerular permeability.


Asunto(s)
Podocitos/fisiología , Transducción de Señal/fisiología , Sindecano-4/fisiología , Canales Catiónicos TRPC/fisiología , Proteínas de Unión al GTP rho/fisiología , Quinasas Asociadas a rho/fisiología , Animales , Calcio/fisiología , Membrana Celular/fisiología , Células Cultivadas , Tasa de Filtración Glomerular/fisiología , Corteza Renal/citología , Ratones , Ratones Noqueados , Modelos Animales , Podocitos/citología , Sindecano-4/deficiencia , Sindecano-4/genética , Canal Catiónico TRPC6 , Proteína de Unión al GTP rhoA
16.
PLoS One ; 6(12): e28302, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22164265

RESUMEN

Sustained pressure overload leads to compensatory myocardial hypertrophy and subsequent heart failure, a leading cause of morbidity and mortality. Further unraveling of the cellular processes involved is essential for development of new treatment strategies. We have investigated the hypothesis that the transmembrane Z-disc proteoglycan syndecan-4, a co-receptor for integrins, connecting extracellular matrix proteins to the cytoskeleton, is an important signal transducer in cardiomyocytes during development of concentric myocardial hypertrophy following pressure overload. Echocardiographic, histochemical and cardiomyocyte size measurements showed that syndecan-4(-/-) mice did not develop concentric myocardial hypertrophy as found in wild-type mice, but rather left ventricular dilatation and dysfunction following pressure overload. Protein and gene expression analyses revealed diminished activation of the central, pro-hypertrophic calcineurin-nuclear factor of activated T-cell (NFAT) signaling pathway. Cardiomyocytes from syndecan-4(-/-)-NFAT-luciferase reporter mice subjected to cyclic mechanical stretch, a hypertrophic stimulus, showed minimal activation of NFAT (1.6-fold) compared to 5.8-fold increase in NFAT-luciferase control cardiomyocytes. Accordingly, overexpression of syndecan-4 or introducing a cell-permeable membrane-targeted syndecan-4 polypeptide (gain of function) activated NFATc4 in vitro. Pull-down experiments demonstrated a direct intracellular syndecan-4-calcineurin interaction. This interaction and activation of NFAT were increased by dephosphorylation of serine 179 (pS179) in syndecan-4. During pressure overload, phosphorylation of syndecan-4 was decreased, and association between syndecan-4, calcineurin and its co-activator calmodulin increased. Moreover, calcineurin dephosphorylated pS179, indicating that calcineurin regulates its own binding and activation. Finally, patients with hypertrophic myocardium due to aortic stenosis had increased syndecan-4 levels with decreased pS179 which was associated with increased NFAT activation. In conclusion, our data show that syndecan-4 is essential for compensatory hypertrophy in the pressure overloaded heart. Specifically, syndecan-4 regulates stretch-induced activation of the calcineurin-NFAT pathway in cardiomyocytes. Thus, our data suggest that manipulation of syndecan-4 may provide an option for therapeutic modulation of calcineurin-NFAT signaling.


Asunto(s)
Calcineurina/metabolismo , Hipertrofia/metabolismo , Miocardio/patología , Factores de Transcripción NFATC/metabolismo , Sindecano-4/fisiología , Animales , Estenosis de la Válvula Aórtica/patología , Calmodulina/metabolismo , Membrana Celular/metabolismo , Células HEK293 , Humanos , Hipertrofia/patología , Hipertrofia Ventricular Izquierda/patología , Luciferasas/metabolismo , Ratones , Ratones Transgénicos , Modelos Biológicos , Fosforilación , Transducción de Señal , Sindecano-4/genética
17.
Dev Cell ; 21(4): 681-93, 2011 Oct 18.
Artículo en Inglés | MEDLINE | ID: mdl-21982645

RESUMEN

Cell migration during wound healing requires adhesion receptor turnover to enable the formation and disassembly of cell-extracellular matrix contacts. Although recent advances have improved our understanding of integrin trafficking pathways, it is not known how extracellular ligand engagement controls receptor dynamics. Using atomic force microscopy, we have measured cell avidity for fibronectin and defined a mechanism for the outside-in regulation of α(5)ß(1)-integrin. Surprisingly, adhesive strength was attenuated by the syndecan-4-binding domain of fibronectin due to a rapid triggering of α(5)ß(1)-integrin endocytosis. Association of syndecan-4 with PKCα was found to trigger RhoG activation and subsequent dynamin- and caveolin-dependent integrin uptake. Like disruption of syndecan-4 or caveolin, gene disruption of RhoG in mice was found to retard closure of dermal wounds due to a migration defect of the fibroblasts and keratinocytes of RhoG null mice. Thus, this syndecan-4-regulated integrin endocytic pathway appears to play a key role in tissue repair.


Asunto(s)
Caveolina 1/fisiología , Movimiento Celular , GTP Fosfohidrolasas/fisiología , Sindecano-4/fisiología , Cicatrización de Heridas , Animales , Adhesión Celular , Células Cultivadas , Endocitosis , Matriz Extracelular , Fibroblastos/citología , Fibroblastos/metabolismo , Fibronectinas/metabolismo , Integrina alfa5beta1/metabolismo , Queratinocitos/citología , Queratinocitos/metabolismo , Ratones , Microscopía de Fuerza Atómica , Proteína Quinasa C-alfa/metabolismo , Transducción de Señal , Proteínas de Unión al GTP rho
18.
FEBS J ; 278(24): 4704-16, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21902810

RESUMEN

Tissue transglutaminase (TG2) is a ubiquitously expressed member of the transglutaminase family of Ca(2+)-dependent crosslinking enzymes. Unlike other family members, TG2 is a multifunctional protein, which has several other well documented enzymatic and non-enzymatic functions. A significant body of evidence accumulated over the last decade reveals multiple and complex activities of this protein on the cell surface and in the extracellular matrix (ECM), including its role in the regulation of cell-ECM interactions and outside-in signaling by several types of transmembrane receptors. Moreover, recent findings indicate a dynamic regulation of the levels and functions of extracellular TG2 by several complementary mechanisms. This review summarizes and assesses recent research into the emerging functions and regulation of extracellular TG2.


Asunto(s)
Matriz Extracelular/metabolismo , Proteínas de Unión al GTP/fisiología , Transglutaminasas/fisiología , Adhesión Celular/fisiología , Reactivos de Enlaces Cruzados/metabolismo , Cisteína/metabolismo , Activación Enzimática , Fibronectinas/metabolismo , Proteínas de Unión al GTP/metabolismo , Humanos , Integrinas/metabolismo , Proteína-5 Relacionada con Receptor de Lipoproteína de Baja Densidad/fisiología , Proteína-6 Relacionada a Receptor de Lipoproteína de Baja Densidad/fisiología , Macrófagos/fisiología , Metaloproteinasas de la Matriz Asociadas a la Membrana/metabolismo , Trasplante de Células Madre Mesenquimatosas , Conformación Proteica/efectos de los fármacos , Proteína Glutamina Gamma Glutamiltransferasa 2 , Transporte de Proteínas/fisiología , Receptores de Factores de Crecimiento/fisiología , Transducción de Señal/fisiología , Sindecano-4/fisiología , Transglutaminasas/metabolismo , beta Catenina/fisiología
19.
Cardiovasc Res ; 92(1): 123-31, 2011 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-21632883

RESUMEN

AIMS: Myocardial infarction (MI) results in acute impairment of left ventricular (LV) function through the initial development of cardiomyocyte death and subsequent progression of LV remodelling. The expression of syndecan-4 (Sdc4), a transmembrane proteoglycan, is up-regulated after MI, but its function in the heart remains unknown. Here, we characterize the effects of Sdc4 deficiency in murine myocardial ischaemia and permanent infarction. METHODS AND RESULTS: Targeted deletion of Sdc4 (Sdc4(-/-)) leads to increased myocardial damage after ischaemic-reperfusion injury due to enhanced cardiomyocyte apoptosis associated with reduced activation of extracellular signal-regulated kinase in cardiomyocytes in vitro and in vivo. After ischaemic-reperfusion injury and permanent infarction, we observed an increase in cardiomyocyte area, nuclear translocation of nuclear factor of activated T cells (NFAT), and transcription of the NFAT target rcan1.4 in wild-type mice. NFAT pathway activation was enhanced in Sdc4(-/-) mice. In line with the in vivo data, NFAT activation and hypertrophy occurs in isolated cardiomyocytes with reduced Sdc4 expression during phenylephrine stimulation in vitro. Despite the initially increased myocardial damage, echocardiography revealed improved LV geometry and function in Sdc4(-/-) mice 7 days after MI. CONCLUSION: Interception of the Sdc4 pathway enhances infarct expansion and hypertrophic remodelling during early infarct healing in ischaemic-reperfusion injury and permanent infarction mouse models and exerts net beneficial effects on LV function.


Asunto(s)
Apoptosis , Daño por Reperfusión Miocárdica/prevención & control , Miocardio/patología , Factores de Transcripción NFATC/fisiología , Transducción de Señal/fisiología , Sindecano-4/fisiología , Remodelación Ventricular , Animales , Cardiomegalia/etiología , Células Cultivadas , Ratones , Ratones Endogámicos C57BL , Infarto del Miocardio/patología , Miocitos Cardíacos/patología , Ratas , Ratas Sprague-Dawley , Sindecano-4/deficiencia , Función Ventricular Izquierda , Proteínas Quinasas p38 Activadas por Mitógenos/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...