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.
Biomolecules ; 11(4)2021 03 26.
Artículo en Inglés | MEDLINE | ID: mdl-33810567

RESUMEN

Cancer is an important cause of morbidity and mortality worldwide. Advances in research on the biology of cancer revealed alterations in several key pathways underlying tumorigenesis and provided molecular targets for developing new and improved existing therapies. Syndecan-4, a transmembrane heparan sulfate proteoglycan, is a central mediator of cell adhesion, migration and proliferation. Although several studies have demonstrated important roles of syndecan-4 in cell behavior and its interactions with growth factors, extracellular matrix (ECM) molecules and cytoskeletal signaling proteins, less is known about its role and expression in multiple cancer. The data summarized in this review demonstrate that high expression of syndecan-4 is an unfavorable biomarker for estrogen receptor-negative breast cancer, glioma, liver cancer, melanoma, osteosarcoma, papillary thyroid carcinoma and testicular, kidney and bladder cancer. In contrast, in neuroblastoma and colorectal cancer, syndecan-4 is downregulated. Interestingly, syndecan-4 expression is modulated by anticancer drugs. It is upregulated upon treatment with zoledronate and this effect reduces invasion of breast cancer cells. In our recent work, we demonstrated that the syndecan-4 level was reduced after trastuzumab treatment. Similarly, syndecan-4 levels are also reduced after panitumumab treatment. Together, the data found suggest that syndecan-4 level is crucial for understanding the changes involving in malignant transformation, and also demonstrate that syndecan-4 emerges as an important target for cancer therapy and diagnosis.


Asunto(s)
Neoplasias/patología , Sindecano-4/metabolismo , Antineoplásicos Inmunológicos/farmacología , Antineoplásicos Inmunológicos/uso terapéutico , Apoptosis , Adhesión Celular , Matriz Extracelular/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Interferencia de ARN , Transducción de Señal , Sindecano-4/química , Sindecano-4/genética
2.
Micron ; 137: 102888, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32554186

RESUMEN

The knowledge on how cells interact with microenvironment is particularly important in understanding the interaction of cancer cells with surrounding stroma, which affects cell migration, adhesion, and metastasis. The main cell surface receptors responsible for the interaction with extracellular matrix (ECM) are integrins, however, they are not the only ones. Integrins are accompanied to other molecules such as syndecans. The role of the latter has not yet been fully established. In our study, we would like to answer the question of whether integrins and syndecans, possessing similar functions, share also similar unbinding properties. By using single molecule force spectroscopy (SMFS), we conducted measurements of the unbinding properties of αVß1 and syndecan-4 in the interaction with vitronectin (VN), which, as each ECM protein, possesses two binding sites specific to integrins and syndecans. The unbinding force and the kinetic off rate constant derived from SMFS describe the stability of single molecular complex. Obtained data show one barrier transition for each complex. The proposed model shows that the unbinding of αVß1 from VN proceeds before the unbinding of SDC-4. However, despite different unbinding kinetics, the access to both receptors is needed for cell growth and proliferation.


Asunto(s)
Integrina alfa5beta1/química , Integrina alfa5beta1/metabolismo , Imagen Individual de Molécula/métodos , Sindecano-4/química , Sindecano-4/metabolismo , Adhesión Celular , Línea Celular Tumoral , Matriz Extracelular , Humanos , Integrina alfa5beta1/genética , Sindecano-4/genética , Neoplasias de la Vejiga Urinaria , Vitronectina/metabolismo
3.
Colloids Surf B Biointerfaces ; 193: 111031, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32408257

RESUMEN

Cardiovascular diseases are the leading cause of death around the world according to the World Health Organization. In-stent restenosis is an inflammatory response of the immune system to endovascular stent implantation in atherosclerotic patients. Biocompatible and biodegradable polymers are of great interest in this field in order to limit the side effects of stent treatments. Poly([R,S]-3,3-dimethylmalic acid) (PDMMLA) is a new biodegradable statistical polyester which presents promising properties as a stent coating. In this work, we studied by dynamic tensiometry, the adhesion of extracellular matrix proteins (bovine serum albumin, fibronectin, fibrinogen, and vitronectin) and plasma membrane proteoglycan (syndecan-4) on three PDMMLA derivatives with different hydrophilicity levels. The results show that proteins have different adhesion profiles and affinity on these surfaces. They show similar behavior on the most hydrophilic surface, making hydrophilic, ionic and hydrogen type bonds. Then we compared each protein's individual profile to that of a mixture of all studied proteins. The comparison shows that vitronectin and syndecan-4 are the quantitatively dominating proteins adsorbed by specific interactions. Based on the results from previous studies, this work allowed us to identify the most important PDMMLA surface as a promising biomaterial for bioactive stent-coating.


Asunto(s)
Malatos/química , Polímeros/química , Adsorción , Animales , Bovinos , Fibrinógeno/química , Fibronectinas/química , Estructura Molecular , Tamaño de la Partícula , Polímeros/síntesis química , Albúmina Sérica Bovina/química , Tensión Superficial , Sindecano-4/química , Vitronectina/química
4.
Biophys J ; 117(4): 688-695, 2019 08 20.
Artículo en Inglés | MEDLINE | ID: mdl-31337547

RESUMEN

Dynamic single-molecule force spectroscopy was performed to monitor the unbinding of fibronectin with the proteoglycans syndecan-4 (SDC4) and decorin and to compare this with the unbinding characteristics of α5ß1-integrin. A single energy barrier was sufficient to describe the unbinding of both SDC4 and decorin from fibronectin, whereas two barriers were observed for the dissociation of α5ß1-integrin from fibronectin. The outer (high-affinity) barriers in the interactions of fibronectin with α5ß1-integrin and SDC4 are characterized by larger barrier heights and widths and slower dissociation rates than those of the inner (low-affinity) barriers in the interactions of fibronectin with α5ß1-integrin and decorin. These results indicate that SDC4 and (ultimately) α5ß1-integrin have the ability to withstand deformation in their interactions with fibronectin, whereas the decorin-fibronectin interaction is considerably more brittle.


Asunto(s)
Decorina/metabolismo , Fibronectinas/metabolismo , Integrina alfa5beta1/metabolismo , Sindecano-4/metabolismo , Sitios de Unión , Decorina/química , Fibronectinas/química , Humanos , Integrina alfa5beta1/química , Unión Proteica , Sindecano-4/química , Termodinámica
5.
PLoS One ; 14(5): e0214737, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31091226

RESUMEN

OBJECTIVE: The endothelial glycocalyx constitutes part of the endothelial barrier but its degradation leaves endothelial cells exposed to transmigrating cells and circulating mediators that can damage the barrier or promote intercellular gaps. Syndecan proteins are key components of the endothelial glycocalyx and are shed during disease states where expression and activity of proteases such as thrombin are elevated. We tested the ability of thrombin to cleave the ectodomains of syndecans and whether the products could act directly on endothelial cells to alter barrier function. APPROACH AND RESULTS: Using transmission electron microscopy, we illustrated the presence of glycocalyx in human lung microvasculature. We confirmed expression of all syndecan subtypes on the endothelial surface of agarose-inflated human lungs. ELISA and western blot analysis suggested that thrombin can cleave syndecan-3/-4 ectodomains to produce fragments. In vivo, syndecan-3 ectodomain fragments increased extravasation of albumin-bound Evans blue in mouse lung, indicative of plasma protein leakage into the surrounding tissue. Syndecan-3/-4 ectodomain fragments decreased transendothelial electrical resistance, a measure of cell-cell adhesive barrier integrity, in a manner sensitive to a Rho kinase inhibitor. These effects were independent of glycosylation and thrombin receptor PAR1. Moreover, these cleavage products caused rapid VE-cadherin-based adherens junction disorganization and increased F-actin stress fibers, supporting their direct effect on endothelial paracellular permeability. CONCLUSIONS: We suggest that thrombin can cleave syndecan-3/4 ectodomain into fragments which interact with endothelial cells causing paracellular hyperpermeability. This may have important implications in the pathogenesis of vascular dysfunction during sepsis or thrombotic disease states where thrombin is activated.


Asunto(s)
Sindecano-3/metabolismo , Sindecano-4/metabolismo , Trombina/metabolismo , Animales , Antígenos CD/metabolismo , Cadherinas/metabolismo , Endotelio Vascular/citología , Endotelio Vascular/metabolismo , Femenino , Glicocálix/metabolismo , Células Endoteliales de la Vena Umbilical Humana , Humanos , Pulmón/metabolismo , Pulmón/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Microscopía Fluorescente , Microvasos/citología , Permeabilidad , Dominios Proteicos , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Sindecano-3/química , Sindecano-3/genética , Sindecano-4/química , Sindecano-4/genética , Quinasas Asociadas a rho/antagonistas & inhibidores , Quinasas Asociadas a rho/metabolismo
6.
J Biol Chem ; 294(22): 8717-8731, 2019 05 31.
Artículo en Inglés | MEDLINE | ID: mdl-30967474

RESUMEN

Costameres are signaling hubs at the sarcolemma and important contact points between the extracellular matrix and cell interior, sensing and transducing biomechanical signals into a cellular response. The transmembrane proteoglycan syndecan-4 localizes to these attachment points and has been shown to be important in the initial stages of cardiac remodeling, but its mechanistic function in the heart remains insufficiently understood. Here, we sought to map the cardiac interactome of syndecan-4 to better understand its function and downstream signaling mechanisms. By combining two different affinity purification methods with MS analysis, we found that the cardiac syndecan-4 interactome consists of 21 novel and 29 previously described interaction partners. Nine of the novel partners were further validated to bind syndecan-4 in HEK293 cells (i.e. CAVIN1/PTRF, CCT5, CDK9, EIF2S1, EIF4B, MPP7, PARVB, PFKM, and RASIP). We also found that 19 of the 50 interactome partners bind differently to syndecan-4 in the left ventricle lysate from aortic-banded heart failure (ABHF) rats compared with SHAM-operated animals. One of these partners was the well-known mechanotransducer muscle LIM protein (MLP), which showed direct and increased binding to syndecan-4 in ABHF. Nuclear translocation is important in MLP-mediated signaling, and we found less MLP in the nuclear-enriched fractions from syndecan-4-/- mouse left ventricles but increased nuclear MLP when syndecan-4 was overexpressed in a cardiomyocyte cell line. In the presence of a cell-permeable syndecan-4-MLP disruptor peptide, the nuclear MLP level was reduced. These findings suggest that syndecan-4 mediates nuclear translocation of MLP in the heart.


Asunto(s)
Núcleo Celular/metabolismo , Ventrículos Cardíacos/metabolismo , Proteínas con Dominio LIM/metabolismo , Proteínas Musculares/metabolismo , Sindecano-4/metabolismo , Animales , Línea Celular , Células HEK293 , Insuficiencia Cardíaca/metabolismo , Insuficiencia Cardíaca/patología , Humanos , Proteínas con Dominio LIM/química , Ratones , Ratones Noqueados , Proteínas Musculares/química , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Dominios PDZ , Mapas de Interacción de Proteínas , Transporte de Proteínas , Proteínas de Unión al ARN/química , Proteínas de Unión al ARN/metabolismo , Ratas , Ratas Wistar , Transducción de Señal , Sindecano-4/química , Sindecano-4/genética
7.
Biomaterials ; 187: 81-92, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30308478

RESUMEN

Biomaterials are a powerful platform for directing cellular behaviour. Herein, we employed a biomimetic strategy to synthesize a low-fouling polymer functionalized with nano-scale clusters of ligands that bind both integrin and syndecan-4 receptors, as both receptor types are critical in focal adhesion signalling and mechanotransduction. Our results demonstrate that the presence of both ligand types synergistically increases the adhesion of human umbilical vein endothelial cells (more than a two fold increase after 4 h) and increases the rate of surface endothelialization compared to surfaces functionalized with only one ligand type. Additionally, we observe that the mixed population of ligands regulates endothelial cell migration, likely due to improved focal adhesion formation as observed through confocal microscopy. Furthermore, we illustrate that only endothelial cells cultured on these mixed ligand surfaces exhibit the appropriate morphological changes - elongation and alignment in the direction of flow - when exposed to laminar shear flow, and neither of the individual ligands alone is sufficient. These results illustrate that both receptor types must be engaged for optimum cell-material interactions and are mandatory for appropriate mechanotransduction. The results presented in this manuscript will be critical for the development of next generation biomedical devices and tissue engineering scaffolds.


Asunto(s)
Integrinas/química , Nanopartículas/química , Oligopéptidos/química , Sindecano-4/química , Resinas Acrílicas/química , Materiales Biocompatibles/química , Adhesión Celular , Movimiento Celular , Proliferación Celular , Células Endoteliales de la Vena Umbilical Humana/citología , Células Endoteliales de la Vena Umbilical Humana/fisiología , Humanos , Integrinas/metabolismo , Ligandos , Mecanotransducción Celular , Unión Proteica , Sindecano-4/metabolismo
8.
PLoS One ; 12(11): e0187094, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29121646

RESUMEN

The small GTPases of the Rho family comprising RhoA, Rac1 and Cdc42 function as molecular switches controlling several essential biochemical pathways in eukaryotic cells. Their activity is cycling between an active GTP-bound and an inactive GDP-bound conformation. The exchange of GDP to GTP is catalyzed by guanine nucleotide exchange factors (GEFs). Here we report a novel regulatory mechanism of Rac1 activity, which is controlled by a phosphomimetic (Ser179Glu) mutant of syndecan-4 (SDC4). SDC4 is a ubiquitously expressed transmembrane, heparan sulfate proteoglycan. In this study we show that the Ser179Glu mutant binds strongly Tiam1, a Rac1-GEF reducing Rac1-GTP by 3-fold in MCF-7 breast adenocarcinoma cells. Mutational analysis unravels the PDZ interaction between SDC4 and Tiam1 is indispensable for the suppression of the Rac1 activity. Neither of the SDC4 interactions is effective alone to block the Rac1 activity, on the contrary, lack of either of interactions can increase the activity of Rac1, therefore the Rac1 activity is the resultant of the inhibitory and stimulatory effects. In addition, SDC4 can bind and tether RhoGDI1 (GDP-dissociation inhibitor 1) to the membrane. Expression of the phosphomimetic SDC4 results in the accumulation of the Rac1-RhoGDI1 complex. Co-immunoprecipitation assays (co-IP-s) reveal that SDC4 can form complexes with RhoGDI1. Together, the regulation of the basal activity of Rac1 is fine tuned and SDC4 is implicated in multiple ways.


Asunto(s)
Mutación/genética , Sindecano-4/genética , Sindecano-4/metabolismo , Proteína 1 de Invasión e Inducción de Metástasis del Linfoma-T/química , Proteína 1 de Invasión e Inducción de Metástasis del Linfoma-T/metabolismo , Proteína de Unión al GTP rac1/metabolismo , Secuencia de Aminoácidos , Humanos , Células MCF-7 , Modelos Biológicos , Dominios PDZ , Unión Proteica , Proteína Quinasa C-alfa/metabolismo , Sindecano-4/química , Quinasas p21 Activadas/metabolismo , Inhibidor alfa de Disociación del Nucleótido Guanina rho/metabolismo
9.
Stem Cells ; 35(2): 522-531, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27662820

RESUMEN

Endothelial progenitor cells (EPCs) are a subtype of bone marrow-derived progenitor cells. Stromal cell-derived factor 1 (SDF-1)-mediated EPC mobilization from bone marrow to areas of ischemia plays an important role in angiogenesis. Previous studies have reported that advanced glycation endproducts (AGEs), which are important mediators of diabetes-related vascular pathology, may impair EPC migration and homing, but the mechanism is unclear. Syndecan-4 (synd4) is a ubiquitous heparan sulfate proteoglycan receptor on the cell surface, involved in SDF-1-dependent cell migration. The extracellular domain of synd4 (ext-synd4) is shed in the context of acute inflammation, but the shedding of ext-synd4 in response to AGEs is undefined. Here we investigated changes in ext-synd4 on EPCs in response to AGEs, focusing on the influence of impaired synd4 signaling on EPC migration and homing. We found decreased full length and increased residue of synd4 in cells incubated with AGEs, with concomitant increase in the soluble fragment of ext-synd4 in the cell medium. EPCs from patients with type 2 diabetes expressed less ext-synd4 as assessed by Western blotting. Flow cytometry analysis showed less ext-synd4 on circulating CD34+ peripheral blood mononuclear cells, of which EPCs form a subgroup. We then explored the role of synd4 in EPC migration and homing. Impaired migration of synd4-deficient EPCs was observed by a 2D-chemotaxis slide. Furthermore, poor homing of synd4-/- EPCs was observed in a mouse model of lower limb ischemia. This study demonstrates that the shedding of synd4 from EPCs plays a key role in AGE-mediated dysfunction of EPC migration and homing. Stem Cells 2017;35:522-531.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Células Progenitoras Endoteliales/metabolismo , Células Progenitoras Endoteliales/patología , Productos Finales de Glicación Avanzada/farmacología , Sindecano-4/metabolismo , Animales , Antígenos de Neoplasias/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patología , Células Progenitoras Endoteliales/efectos de los fármacos , Humanos , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/metabolismo , Ratones Endogámicos C57BL , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Dominios Proteicos , Especies Reactivas de Oxígeno/metabolismo , Sindecano-4/química , Sindecano-4/deficiencia
10.
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
11.
J Biol Chem ; 290(43): 26103-13, 2015 Oct 23.
Artículo en Inglés | MEDLINE | ID: mdl-26350464

RESUMEN

The α6ß4 integrin is known to associate with receptor tyrosine kinases when engaged in epithelial wound healing and in carcinoma invasion and survival. Prior work has shown that HER2 associates with α6ß4 integrin and syndecan-1 (Sdc1), in which Sdc1 engages the cytoplasmic domain of the ß4 integrin subunit allowing HER2-dependent motility and carcinoma cell survival. In contrast, EGFR associates with Sdc4 and the α6ß4 integrin, and EGFR-dependent motility depends on cytoplasmic engagement of ß4 integrin with Sdc4. However, how HER2 and EGFR assimilate into a complex with the syndecans and integrin, and why kinase capture is syndecan-specific has remained unknown. In the present study, we demonstrate that HER2 is captured via a site, comprised of amino acids 210-240, in the extracellular domain of human Sdc1, and EGFR is captured via an extracellular site comprised of amino acids 87-131 in human Sdc4. Binding assays using purified recombinant proteins demonstrate that the interaction between the EGFR family members and the syndecans is direct. The α3ß1 integrin, which is responsible for the motility of the cells, is captured at these sites as well. Peptides based on the interaction motifs in Sdc1 and Sdc4, called synstatins (SSTN210-240 and SSTN87-131) competitively displace the receptor tyrosine kinase and α3ß1 integrin from the syndecan with an IC50 of 100-300 nm. The syndecans remain anchored to the α6ß4 integrin via its cytoplasmic domain, but the activation of cell motility is disrupted. These novel SSTN peptides are potential therapeutics for carcinomas that depend on these HER2- and EGFR-coupled mechanisms for their invasion and survival.


Asunto(s)
Movimiento Celular , Receptores ErbB/metabolismo , Integrina alfa3beta1/metabolismo , Integrina alfa6beta4/metabolismo , Receptor ErbB-2/metabolismo , Sindecano-1/metabolismo , Sindecano-4/metabolismo , Secuencia de Aminoácidos , Animales , Sitios de Unión , Línea Celular , Células Epiteliales/metabolismo , Humanos , Datos de Secuencia Molecular , Homología de Secuencia de Aminoácido , Sindecano-1/química , Sindecano-4/química
12.
PLoS One ; 10(6): e0129288, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26068620

RESUMEN

The cell surface proteoglycan syndecan-4 has been reported to be crucial for muscle differentiation, but the molecular mechanisms still remain to be fully understood. During in vitro differentiation of bovine muscle cells immunocytochemical analyses showed strong labelling of syndecan-4 intracellularly, in close proximity with Golgi structures, in membranes of intracellular vesicles and finally, in the nuclear area including the nuclear envelope. Chase experiments showed that syndecan-4 was internalized from the plasma membrane during this process. Furthermore, when syndecan-4 was knocked down by siRNA more myotubes were formed, and the expression of myogenic transcription factors, ß1-integrin and actin was influenced. However, when bovine muscle cells were treated with a cell-penetrating peptide containing the cytoplasmic region of syndecan-4, myoblast fusion and thus myotube formation was blocked, both in normal cells and in syndecan-4 knock down cells. Altogether this suggests that the cytoplasmic domain of syndecan-4 is important in regulation of myogenesis. The internalization of syndecan-4 from the plasma membrane during muscle differentiation and the nuclear localization of syndecan-4 in differentiated muscle cells may be part of this regulation, and is a novel aspect of syndecan biology which merits further studies.


Asunto(s)
Diferenciación Celular , Membrana Celular/metabolismo , Desarrollo de Músculos , Fibras Musculares Esqueléticas/metabolismo , Sindecano-4/metabolismo , Secuencia de Aminoácidos , Animales , Bovinos , Células Cultivadas , Datos de Secuencia Molecular , Fibras Musculares Esqueléticas/citología , Estructura Terciaria de Proteína , Transporte de Proteínas , Sindecano-4/química , Sindecano-4/genética
13.
Biochem Biophys Res Commun ; 463(4): 762-7, 2015 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-26100207

RESUMEN

Syndesmos, nucleoside diphosphate linked moiety X (nudix)-type motif 16-like 1 (Nudt16l1), is evolutionarily divergent from the Nudt16 family. Syndesmos, which is co-localized with syndecan-4 cytoplasmic domain (Syn4(cyto)) in focal contacts, interacts with various cell adhesion adaptor proteins to control cell signaling. We determined the X-ray crystal structure of syndesmos; it is composed of seven α-helices and seven ß-strands. Although syndesmos has a molecular topology similar to that of nudix hydrolase proteins, the structure of the nudix motif differs from that of X29. The dimeric interface of syndesmos is composed of α-helix 4, 7 and ß-strand 2, 7, which primarily form hydrophobic interactions. The binding interaction between syndesmos and syn4(cyto) was characterized as a low-affinity interaction (Kd = 62 µM) by surface plasmon resonance (SPR) and nuclear magnetic resonance (NMR). The NMR resonances of Lys (177, 178, 179), Gly182, and Ser183 in the C1 region and Lys193 and Lys194 in the V region of syndecan-4 are perturbed upon syndesmos binding. Our results provide structural insight into the molecular function of syndesmos in the regulation of cell signaling via binding to syndecan-4.


Asunto(s)
Pirofosfatasas/metabolismo , Sindecano-4/metabolismo , Secuencia de Aminoácidos , Animales , Cristalografía por Rayos X , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Ratones , Modelos Moleculares , Datos de Secuencia Molecular , Estructura Molecular , Unión Proteica , Pirofosfatasas/química , Homología de Secuencia de Aminoácido , Resonancia por Plasmón de Superficie , Sindecano-4/química
14.
J Biol Chem ; 290(27): 16943-53, 2015 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-25979339

RESUMEN

Syndecans, a family of transmembrane heparansulfate proteoglycans, are known to interact through their transmembrane domains to form non-covalently linked homodimers, a process essential for their individual functions. Because all syndecan transmembrane domains are highly conserved and thus might mediate interactions between different members of the syndecan family, we investigated syndecan interactions in detail. All recombinant syndecan-2 and -4 protein variants containing the transmembrane domain formed not only sodium dodecyl sulfate (SDS)-resistant homodimers but also SDS-resistant heterodimers. Biochemical and structural data revealed that recombinant syndecan-2 and -4 formed intermolecular interactions in vitro, and the GXXXG motif in transmembrane domain mediated this interaction. When exogenously expressed in rat embryonic fibroblasts, syndecan-2 interacted with syndecan-4 and vice versa. Furthermore, bimolecular fluorescence complementation-based assay demonstrated specific hetero-molecular interactions between syndecan-2 and -4, supporting hetero-oligomer formation of syndecans in vivo. Interestingly, hetero-oligomerization significantly reduced syndecan-4-mediated cellular processes such as protein kinase Cα activation and protein kinase Cα-mediated cell adhesion as well as syndecan-2-mediated tumorigenic activities in colon cancer cells such as migration and anchorage-independent growth. Taken together, these data provide evidence that hetero-oligomerization produces distinct syndecan functions and offer insights into the underlying signaling mechanisms of syndecans.


Asunto(s)
Sindecano-2/química , Sindecano-2/metabolismo , Sindecano-4/química , Sindecano-4/metabolismo , Secuencias de Aminoácidos , Animales , Dimerización , Fibroblastos/química , Fibroblastos/metabolismo , Unión Proteica , Ratas , Sindecano-2/genética , Sindecano-4/genética
15.
J Biol Chem ; 290(9): 5772-82, 2015 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-25572401

RESUMEN

The syndecans are a type of cell surface adhesion receptor that initiates intracellular signaling events through receptor clustering mediated by their highly conserved transmembrane domains (TMDs). However, the exact function of the syndecan TMD is not yet fully understood. Here, we investigated the specific regulatory role of the syndecan-2 TMD. We found that syndecan-2 mutants in which the TMD had been replaced with that of syndecan-4 were defective in syndecan-2-mediated functions, suggesting that the TMD of syndecan-2 plays one or more specific roles. Interestingly, syndecan-2 has a stronger tendency to form sodium dodecyl sulfate (SDS)-resistant homodimers than syndecan-4. Our structural studies showed that a unique phenylalanine residue (Phe(167)) enables an additional molecular interaction between the TMDs of the syndecan-2 homodimer. The presence of Phe(167) was correlated with a higher tendency toward oligomerization, and its replacement with isoleucine significantly reduced the SDS-resistant dimer formation and cellular functions of syndecan-2 (e.g. cell migration). Conversely, replacement of isoleucine with phenylalanine at this position in the syndecan-4 TMD rescued the defects observed in a mutant syndecan-2 harboring the syndecan-4 TMD. Taken together, these data suggest that Phe(167) in the TMD of syndecan-2 endows the protein with specific functions. Our work offers new insights into the signaling mediated by the TMD of syndecan family members.


Asunto(s)
Isoleucina/genética , Mutación Missense , Fenilalanina/genética , Sindecano-2/genética , Secuencia de Aminoácidos , Animales , Membrana Celular/metabolismo , Células Cultivadas , Células HCT116 , Células HEK293 , Humanos , Immunoblotting , Isoleucina/química , Isoleucina/metabolismo , Microscopía Confocal , Datos de Secuencia Molecular , Fenilalanina/química , Fenilalanina/metabolismo , Multimerización de Proteína , Estructura Terciaria de Proteína , Ratas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Homología de Secuencia de Aminoácido , Dodecil Sulfato de Sodio/química , Dodecil Sulfato de Sodio/metabolismo , Sindecano-2/química , Sindecano-2/metabolismo , Sindecano-4/química , Sindecano-4/genética , Sindecano-4/metabolismo
16.
J Biol Chem ; 289(44): 30318-30332, 2014 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-25202019

RESUMEN

Epithelial cells are highly dependent during wound healing and tumorigenesis on the α6ß4 integrin and its association with receptor tyrosine kinases. Previous work showed that phosphorylation of the ß4 subunit upon matrix engagement depends on the matrix receptor syndecan (Sdc)-1 engaging the cytoplasmic domain of the ß4 integrin and coupling of the integrin to human epidermal growth factor receptor-2 (HER2). In this study, HER2-dependent migration activated by matrix engagement is compared with migration stimulated by EGF. We find that whereas HER2-dependent migration depends on Sdc1, EGF-dependent migration depends on a complex consisting of human epidermal growth factor receptor-1 (HER1, commonly known as EGFR), α6ß4, and Sdc4. The two syndecans recognize distinct sites at the extreme C terminus of the ß4 integrin cytoplasmic domain. The binding motif in Sdc1 is QEEXYX, composed in part by its syndecan-specific variable (V) region and in part by the second conserved (C2) region that it shares with other syndecans. A cell-penetrating peptide containing this sequence competes for HER2-dependent epithelial migration and carcinoma survival, although it is without effect on the EGFR-stimulated mechanism. ß4 mutants bearing mutations specific for Sdc1 and Sdc4 recognition act as dominant negative mutants to block cell spreading or cell migration that depends on HER2 or EGFR, respectively. The interaction of the α6ß4 integrin with the syndecans appears critical for it to be utilized as a signaling platform; migration depends on α3ß1 integrin binding to laminin 332 (LN332; also known as laminin 5), whereas antibodies that block α6ß4 binding are without effect. These findings indicate that specific syndecan family members are likely to have key roles in α6ß4 integrin activation by receptor tyrosine kinases.


Asunto(s)
Movimiento Celular , Supervivencia Celular , Integrina alfa6beta4/metabolismo , Sindecano-1/metabolismo , Sindecano-4/metabolismo , Secuencia de Aminoácidos , Moléculas de Adhesión Celular/metabolismo , Línea Celular Tumoral , Citoplasma/metabolismo , Receptores ErbB/metabolismo , Receptores ErbB/fisiología , Humanos , Integrina alfa6beta4/química , Integrina alfa6beta4/genética , Datos de Secuencia Molecular , Mutación Missense , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Receptor ErbB-2/fisiología , Transducción de Señal , Sindecano-1/química , Sindecano-4/química , Kalinina
17.
Nat Commun ; 5: 4886, 2014 Sep 12.
Artículo en Inglés | MEDLINE | ID: mdl-25216363

RESUMEN

Cancer cell adhesion to the vascular endothelium is a critical step of tumour metastasis. Endothelial surface molecule Thy-1 (CD90) is implicated in the metastatic process through its interactions with integrins and syndecans. However, how Thy-1 supports cell-cell adhesion in a dynamic mechanical environment is not known. Here we show that Thy-1 supports ß1 integrin- and syndecan-4 (Syn4)-mediated contractility-dependent mechanosignalling of melanoma cells. At the single-molecule level, Thy-1 is capable of independently binding α5ß1 integrin and syndecan-4 (Syn4) receptors. However, in the presence of both α5ß1 and Syn4, the two receptors bind cooperatively to Thy-1, to form a trimolecular complex. This trimolecular complex displays a unique phenomenon we coin 'dynamic catch', characterized by abrupt bond stiffening followed by the formation of catch bonds, where force prolongs the bond lifetime. Thus, we reveal a new class of trimolecular interactions where force strengthens the synergistic binding of two co-receptors and modulates downstream mechanosignalling.


Asunto(s)
Integrina alfa5beta1/química , Integrina beta1/química , Sindecano-4/química , Antígenos Thy-1/química , Fenómenos Biomecánicos , Adhesión Celular , Línea Celular Tumoral , Expresión Génica , Humanos , Integrina alfa5beta1/genética , Integrina alfa5beta1/metabolismo , Integrina beta1/genética , Integrina beta1/metabolismo , Células K562 , Mecanotransducción Celular , Unión Proteica , Pliegue de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Sindecano-4/genética , Sindecano-4/metabolismo , Resistencia a la Tracción , Antígenos Thy-1/genética , Antígenos Thy-1/metabolismo
19.
Structure ; 21(3): 342-54, 2013 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-23395182

RESUMEN

PDZ (PSD-95/Dlg/ZO-1) domains are protein-protein interaction modules often regulated by ligand phosphorylation. Here, we investigated the specificity, structure, and dynamics of Tiam1 PDZ domain/ligand interactions. We show that the PDZ domain specifically binds syndecan1 (SDC1), phosphorylated SDC1 (pSDC1), and SDC3 but not other syndecan isoforms. The crystal structure of the PDZ/SDC1 complex indicates that syndecan affinity is derived from amino acids beyond the four C-terminal residues. Remarkably, the crystal structure of the PDZ/pSDC1 complex reveals a binding pocket that accommodates the phosphoryl group. Methyl relaxation experiments of PDZ/SCD1 and PDZ/pSDC1 complexes reveal that PDZ-phosphoryl interactions dampen dynamic motions in a distal region of the PDZ domain by decoupling them from the ligand-binding site. Our data are consistent with a selection model by which specificity and phosphorylation regulate PDZ/syndecan interactions and signaling events. Importantly, our relaxation data demonstrate that PDZ/phospho-ligand interactions regulate protein dynamics and their coupling to distal sites.


Asunto(s)
Factores de Intercambio de Guanina Nucleótido/química , Modelos Moleculares , Sindecano-1/química , Secuencia de Aminoácidos , Sitios de Unión , Simulación por Computador , Cristalografía por Rayos X , Humanos , Ligandos , Datos de Secuencia Molecular , Dominios PDZ , Fosforilación , Unión Proteica , Estructura Secundaria de Proteína , Relación Estructura-Actividad , Sindecano-2/química , Sindecano-3/química , Sindecano-4/química , Proteína 1 de Invasión e Inducción de Metástasis del Linfoma-T
20.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA