Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 72
Filtrar
1.
Biomed Mater ; 13(6): 065013, 2018 10 02.
Artículo en Inglés | MEDLINE | ID: mdl-30277888

RESUMEN

Engineered adipose tissues are developed for their use as substitutes for tissue replacement in reconstructive surgery. To ensure a timely perfusion of the grafted substitutes, different strategies can be used such as the incorporation of an endothelial component. In this study, we engineered human adipose tissue substitutes comprising of functional adipocytes as well as a natural extracellular matrix using the self-assembly approach, without the use of exogenous scaffolding elements. Human microvascular endothelial cells (hMVECs) were incorporated during tissue production in vitro and we hypothesized that their presence would favor the early connection with the host vascular network translating into functional enhancement after implantation into nude mice in comparison to the substitutes that were not enriched in hMVECs. In vitro, no significant differences were observed between the substitutes in terms of histological aspects. After implantation, both groups presented numerous adipocytes and an abundant matrix in addition to the presence of host capillaries within the grafts. The substitutes thickness and volume were not significantly different between groups over the short-term time course of 14 days (d). For the microvascularized adipose tissues, human CD31 staining revealed a human capillary network connecting with the host microvasculature as early as 3 d after grafting. The detection of murine red blood cells within human CD31+ structures confirmed the functionality of the human capillary network. By analyzing the extent of the global vascularization achieved, a tendency towards increased total capillary network surface and volume was revealed for prevascularized tissues over 14 d. Therefore, applying this strategy on thicker reconstructed adipose tissues with rate-limiting oxygen diffusion might procure added benefits and prove useful to provide voluminous substitutes for patients suffering from adipose tissue loss or defects.


Asunto(s)
Tejido Adiposo/metabolismo , Prótesis Vascular , Células Endoteliales/citología , Ingeniería de Tejidos/métodos , Adipocitos/citología , Adulto , Animales , Capilares/metabolismo , Medios de Cultivo Condicionados , Eritrocitos/metabolismo , Matriz Extracelular/metabolismo , Femenino , Humanos , Técnicas In Vitro , Masculino , Ratones , Ratones Desnudos , Microcirculación , Neovascularización Fisiológica , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/química , Células del Estroma/citología
2.
Bioconjug Chem ; 29(1): 56-66, 2018 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-29200285

RESUMEN

The conjugation of antibodies to drugs and drug carriers improves delivery to target tissues. Widespread implementation and effective translation of this pharmacologic strategy awaits the development of affinity ligands capable of a defined degree of modification and highly efficient bioconjugation without loss of affinity. To date, such ligands are lacking for the targeting of therapeutics to vascular endothelial cells. To enable site-specific, click-chemistry conjugation to therapeutic cargo, we used the bacterial transpeptidase, sortase A, to attach short azidolysine containing peptides to three endothelial-specific single chain antibody fragments (scFv). While direct fusion of a recognition motif (sortag) to the scFv C-terminus generally resulted in low levels of sortase-mediated modification, improved reaction efficiency was observed for one protein, in which two amino acids had been introduced during cloning. This prompted insertion of a short, semi-rigid linker between scFv and sortag. The linker significantly enhanced modification of all three proteins, to the extent that unmodified scFv could no longer be detected. As proof of principle, purified, azide-modified scFv was conjugated to the antioxidant enzyme, catalase, resulting in robust endothelial targeting of functional cargo in vitro and in vivo.


Asunto(s)
Química Clic/métodos , Anticuerpos de Cadena Única/química , Anticuerpos de Cadena Única/farmacocinética , Secuencia de Aminoácidos , Aminoaciltransferasas/metabolismo , Animales , Proteínas Bacterianas/metabolismo , Catalasa/metabolismo , Línea Celular , Cisteína Endopeptidasas/metabolismo , Humanos , Molécula 1 de Adhesión Intercelular/administración & dosificación , Molécula 1 de Adhesión Intercelular/química , Molécula 1 de Adhesión Intercelular/metabolismo , Ratones , Ratones Endogámicos C57BL , Modelos Moleculares , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/administración & dosificación , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/química , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/farmacocinética , Anticuerpos de Cadena Única/administración & dosificación , Anticuerpos de Cadena Única/metabolismo , Distribución Tisular
3.
J Biol Chem ; 292(48): 19639-19655, 2017 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-28974577

RESUMEN

Platelet endothelial cell adhesion molecule 1 (PECAM-1) is a cell adhesion protein involved in the regulation of cell adhesion and migration. Interestingly, several PECAM-1-deficient hematopoietic cells exhibit impaired chemotactic responses to stromal cell-derived factor 1 (SDF-1), a chemokine essential for B lymphopoiesis and bone marrow myelopoiesis. However, whether PECAM-1 is involved in SDF-1-regulated chemotaxis is unknown. We report here that SDF-1 induces tyrosine phosphorylation of PECAM-1 at its immunoreceptor tyrosine-based inhibition motifs in several hematopoietic cell lines via the Src family kinase Lyn, Bruton's tyrosine kinase, and JAK2 and that inhibition of these kinases reduced chemotaxis. Overexpression and knockdown of PECAM-1 enhanced and down-regulated, respectively, SDF-1-induced Gαi-dependent activation of the PI3K/Akt/mTORC1 pathway and small GTPase Rap1 in hematopoietic 32Dcl3 cells, and these changes in activation correlated with chemotaxis. Furthermore, pharmacological or genetic inhibition of the PI3K/Akt/mTORC1 pathway or Rap1, respectively, revealed that these pathways are independently activated and required for SDF-1-induced chemotaxis. When coexpressed in 293T cells, PECAM-1 physically associated with the SDF-1 receptor CXCR4. Moreover, PECAM-1 overexpression and knockdown reduced and enhanced SDF-1-induced endocytosis of CXCR4, respectively. Furthermore, when expressed in 32Dcl3 cells, an endocytosis-defective CXCR4 mutant, CXCR4-S324A/S325A, could activate the PI3K/Akt/mTORC1 pathway as well as Rap1 and induce chemotaxis in a manner similar to PECAM-1 overexpression. These findings suggest that PECAM-1 enhances SDF-1-induced chemotaxis by augmenting and prolonging activation of the PI3K/Akt/mTORC1 pathway and Rap1 and that PECAM-1, at least partly, exerts its activity by inhibiting SDF-1-induced internalization of CXCR4.


Asunto(s)
Quimiocina CXCL12/fisiología , Leucocitos/fisiología , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/fisiología , Animales , Células de la Médula Ósea/metabolismo , Línea Celular , Ratones , Fosforilación , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/química , Proteínas Tirosina Quinasas/metabolismo , Receptores CXCR4/metabolismo , Tirosina/metabolismo
4.
Biol Cell ; 109(8): 312-321, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28643869

RESUMEN

BACKGROUND INFORMATION: Vascular endothelial cells (ECs) are a well-known cell system used in the study of mechanobiology. Using cultured ECs, we found that platelet EC adhesion molecule 1 (PECAM-1, CD31), a cell adhesion protein localised to regions of EC-EC contact, was rapidly tyrosine phosphorylated in ECs exposed to shear or cyclic stretch. Src-homology 2 domain-containing protein tyrosine phosphatase 2 (SHP2) binds phosphorylated PECAM-1 and activates the extracellular signal-regulated kinase1/2 (ERK1/2) signalling cascade, a known flow-activated signalling pathway. RESULTS: Although PECAM-1 tyrosine phosphorylation is characterised in ECs exposed to fluid shear stress, it is less well demonstrated in the cells stretched cyclically. Thus, we first show that PECAM-1 is tyrosine-phosphorylated in ECs cyclically stretched. We hypothesise that when an external force is applied to a monolayer of ECs, the force is directly transmitted to PECAM-1 which is then stretched and phosphorylation sites in its cytoplasmic domain are exposed and phosphorylated. This hypothesis requires the presence of any stretchable structure within the PECAM-1 cytoplasmic domain. Force spectroscopy measurements were performed with a construct containing cytoplasmic PECAM-1 domains inserted between I27 motifs, a recombinant string of the structural elements from titin. This strategy allowed us to identify the events in which a single molecule is being pulled and to detect the unravelling of the cytoplasmic domain of PECAM-1 by force. The response by PECAM-1 to mechanical loading was heterogeneous but with magnitudes as high as or higher than the naturally force bearing I27 domains. CONCLUSIONS: The PECAM-1 cytoplasmic domain has a structure that can be unfolded by externally applied force and this unfolding of PECAM-1 may be necessary for its phosphorylation, the first step of PECAM-1 mechanosignalling. SIGNIFICANCE: When EC monolayers are mechanically stimulated, the PECAM-1 found at EC contacts is phosphorylated. We have proposed that under these conditions, the cytoplasmic domain of PECAM-1 is unfolded, which then exposes a phosphorylation site, allowing it to be accessed. The stretch induced unfolding is essential to this model of PECAM-1 mechanosignalling. In this study, we investigate whether the cytoplasmic domain of PECAM-1 has a stretchable structure, and the results are in line with our hypothesis.


Asunto(s)
Aorta/metabolismo , Endotelio Vascular/metabolismo , Mecanotransducción Celular/fisiología , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Tirosina/metabolismo , Animales , Aorta/citología , Bovinos , Células Cultivadas , Endotelio Vascular/citología , Mutación , Fosforilación , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/química , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/genética , Transducción de Señal
5.
Sci Rep ; 6: 38655, 2016 12 13.
Artículo en Inglés | MEDLINE | ID: mdl-27958302

RESUMEN

Cell adhesion involved in signal transduction, tissue integrity and pathogen infection is mainly mediated by cell adhesion molecules (CAM). One CAM member, platelet-endothelial-cell adhesion molecule-1 (PECAM-1), plays an important role in tight junction among endothelia cells, leukocyte trafficking, and immune response through its homophilic and heterophilic binding patterns. Both kinds of interactions, which lead to endogenous and exogenous signal transmission, are derived from extracellular immunoglobulin-like (IgL) domains and cytoplasmic immunoreceptor tyrosine-based inhibitory motifs (ITIMs) of PECAM-1. To date, the mechanism of trans-homophilic interaction of PECAM-1 remains unclear. Here, we present the crystal structure of PECAM-1 IgL1-2 trans-homo dimer. Both IgL 1 and 2 adopt the classical Ig domain conformation comprised of two layers of ß-sheets possessing antiparallel ß-strands with each being anchored by a pair of cysteines forming a disulfide bond. The dimer interface includes hydrophobic and hydrophilic interactions. The Small-Angle X-ray Scattering (SAXS) envelope of PECAM-1 IgL1-6 supported such a dimer formation in solution. Cell adhesion assays on wildtype and mutant PECAM-1 further characterized the structural determinants in cell junction and communication.


Asunto(s)
Molécula-1 de Adhesión Celular Endotelial de Plaqueta/química , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Adhesión Celular , Línea Celular , Análisis Mutacional de ADN , Glicosilación , Humanos , Modelos Moleculares , Unión Proteica , Multimerización de Proteína , Dispersión del Ángulo Pequeño , Soluciones , Relación Estructura-Actividad , Difracción de Rayos X
6.
J Biol Chem ; 291(50): 26216-26225, 2016 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-27793989

RESUMEN

Platelet Endothelial Cell Adhesion Molecule 1 (PECAM-1) is a major component of the endothelial cell intercellular junction. Previous studies have shown that PECAM-1 homophilic interactions, mediated by amino-terminal immunoglobulin homology domain 1, contribute to maintenance of the vascular permeability barrier and to its re-establishment following inflammatory or thrombotic insult. PECAM-1 glycans account for ∼30% of its molecular mass, and the newly solved crystal structure of human PECAM-1 immunoglobulin homology domain 1 reveals that a glycan emanating from the asparagine residue at position 25 (Asn-25) is located within the trans homophilic-binding interface, suggesting a role for an Asn-25-associated glycan in PECAM-1 homophilic interactions. In support of this possibility, unbiased molecular docking studies revealed that negatively charged α2,3 sialic acid moieties bind tightly to a groove within the PECAM-1 homophilic interface in an orientation that favors the formation of an electrostatic bridge with positively charged Lys-89, mutation of which has been shown previously to disrupt PECAM-1-mediated homophilic binding. To verify the contribution of the Asn-25 glycan to endothelial barrier function, we generated an N25Q mutant form of PECAM-1 that is not glycosylated at this position and examined its ability to contribute to vascular integrity in endothelial cell-like REN cells. Confocal microscopy showed that although N25Q PECAM-1 concentrates normally at cell-cell junctions, the ability of this mutant form of PECAM-1 to support re-establishment of a permeability barrier following disruption with thrombin was significantly compromised. Taken together, these data suggest that a sialic acid-containing glycan emanating from Asn-25 reinforces dynamic endothelial cell-cell interactions by stabilizing the PECAM-1 homophilic binding interface.


Asunto(s)
Comunicación Celular/fisiología , Células Endoteliales/metabolismo , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Polisacáridos/metabolismo , Sustitución de Aminoácidos , Línea Celular , Células Endoteliales/citología , Humanos , Mutación Missense , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/química , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/genética , Polisacáridos/química , Polisacáridos/genética , Ácidos Siálicos/química , Ácidos Siálicos/genética , Ácidos Siálicos/metabolismo , Trombina/química , Trombina/genética , Trombina/metabolismo
7.
Anticancer Res ; 36(9): 4449-58, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27630281

RESUMEN

BACKGROUND: The putative functions of the cellular prion protein (PrP(c)) are believed to be associated with cell signaling, differentiation, survival, and cancer progression. With respect to cancer development and progression, elevations and mutations of PrP(c) expression have been shown to increase the risk for malignancy and metastasis in breast and colorectal cancer. Since both natural supplements and direct regulation of PrP(c) expression contribute to inhibition of cancer progression and growth, we hypothesized that knockdown of PrP(c) could lead to an enhanced synergic effect on the inhibition of cancer growth by fucoidan. MATERIALS AND METHODS: PrP(c) expression was suppressed in HT29 human colon cancer cells by utilizing small-interfering RNA (si-PRNP), and cells were subsequently used to study the antiproliferative and anticancer effects of fucoidan treatment of HT29 human colon cancer cells. RESULTS: Fucoidan treatment significantly inhibited growth and reduced cyclin and cyclin-dependent kinase (CDK) expression in HT29 colon cancer cells. Furthermore, silencing PrP(c) expression with si-PRNP amplified the fucoidan-induced changes in cell proliferation, apoptosis, and migration. Intraperitoneal injection of si-PRNP with fucoidan reduced proliferation and tumor volume in Balb/c nude mice. This enhanced antitumor efficacy was associated with decreased angiogenesis. CONCLUSION: Combination of fucoidan with silencing of PrP(c) has a synergic effect on the inhibition of HT29 colon cancer cell growth. Furthermore, we provide evidence for the therapeutic application of PrP(c) silencing with other anticancer drugs for cancer.


Asunto(s)
Antineoplásicos/química , Silenciador del Gen , Polisacáridos/química , Proteínas Priónicas/genética , Animales , Apoptosis , Caspasa 3/metabolismo , Ciclo Celular , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Neoplasias del Colon/tratamiento farmacológico , Células HT29 , Humanos , Inmunohistoquímica , Masculino , Ratones , Ratones Endogámicos BALB C , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/química , Proteínas Priónicas/metabolismo , Antígeno Nuclear de Célula en Proliferación/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo
8.
Int J Biochem Cell Biol ; 77(Pt A): 102-108, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27270333

RESUMEN

Platelet endothelial cell adhesion molecule 1 (PECAM-1) is a cell surface receptor widely distributed on endothelium and hematopoietic-derived cells, and maintain the integrity of the blood vessels. PECAM-1 is widely recognized as an endothelial cell marker. The homophilic interaction through its extracellular domain of PECAM-1 (soluble PECAM-1, or sPECAM-1) is important to its functions. However, structural details for such dimerization are not fully understood. Here we report the production of recombinant sPECAM-1 in large quantity by Drosophila expression system and the small-angle X-ray diffraction (SAXS) study. The recombinant sPECAM-1 was found to form one population of dimer, but not oligomer, and was able to bind to heparin immobilized on a chip in surface plasmon resonance imaging (SPRi) binding experiments. The results of SAXS demonstrated that sPECAM-1 formed a symmetric homodimer of Ω-shape in solution, and each protomer adopted an extended conformation. The dimer is mediated through the intermolecular interactions through the first N-terminal domain (D1) of sPECAM-1. This model provides new structural information for its homophilic interaction mechanism.


Asunto(s)
Molécula-1 de Adhesión Celular Endotelial de Plaqueta/química , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Multimerización de Proteína , Animales , Biomarcadores/química , Biomarcadores/metabolismo , Línea Celular , Endotelio/metabolismo , Espacio Extracelular/metabolismo , Regulación de la Expresión Génica , Heparina/metabolismo , Humanos , Modelos Moleculares , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/genética , Dominios Proteicos , Estructura Cuaternaria de Proteína , Solubilidad
9.
Blood ; 127(8): 1052-61, 2016 Feb 25.
Artículo en Inglés | MEDLINE | ID: mdl-26702061

RESUMEN

Platelet endothelial cell adhesion molecule-1 (PECAM-1) is a 130-kDa member of the immunoglobulin gene superfamily (IgSF) that is present on the surface of circulating platelets and leukocytes, and highly expressed at the junctions of confluent endothelial cell monolayers. PECAM-1-mediated homophilic interactions, known to be mediated by its 2 amino-terminal immunoglobulin homology domains, are essential for concentrating PECAM-1 at endothelial cell intercellular junctions, where it functions to facilitate diapedesis, maintain vascular integrity, and transmit survival signals into the cell. Given the importance of PECAM-1-mediated homophilic interactions in mediating each of these cell physiological events, and to reveal the nature and orientation of the PECAM-1-PECAM-1 homophilic-binding interface, we undertook studies aimed at determining the crystal structure of the PECAM-1 homophilic-binding domain, which is composed of amino-terminal immunoglobulin homology domains 1 and 2 (IgD1 and IgD2). The crystal structure revealed that both IgD1 and IgD2 exhibit a classical IgSF fold, having a ß-sandwich topology formed by 2 sheets of antiparallel ß strands stabilized by the hallmark disulfide bond between the B and F strands. Interestingly, despite previous assignment to the C2 class of immunoglobulin-like domains, the structure of IgD1 reveals that it actually belongs to the I2 set of IgSF folds. Both IgD1 and IgD2 participate importantly in the formation of the trans homophilic-binding interface, with a total buried interface area of >2300 Å(2). These and other unique structural features of PECAM-1 allow for the development of an atomic-level model of the interactions that PECAM-1 forms during assembly of endothelial cell intercellular junctions.


Asunto(s)
Células Endoteliales/metabolismo , Uniones Intercelulares/metabolismo , Modelos Moleculares , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/química , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Secuencia de Aminoácidos , Animales , Adhesión Celular/fisiología , Cristalografía por Rayos X , Electroforesis en Gel de Poliacrilamida , Humanos , Ratones , Datos de Secuencia Molecular , Unión Proteica/fisiología , Estructura Terciaria de Proteína , Especificidad de la Especie , Transfección
10.
Cell Signal ; 28(3): 117-124, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26706435

RESUMEN

Endothelial dysfunction, characterized by impaired activation of endothelial nitric oxide (NO) synthase (eNOS) and ensued decrease of NO production, is a common mechanism of various cardiovascular pathologies, including hypertension and atherosclerosis. Laminar blood flow-mediated specific signaling cascades modulate vascular endothelial cells (ECs) structure and functions. We have previously shown that flow-stimulated Gab1 (Grb2-associated binder-1) tyrosine phosphorylation mediates eNOS activation in ECs, which in part confers laminar flow atheroprotective action. However, the molecular mechanisms whereby flow regulates Gab1 tyrosine phosphorylation and its downstream signaling events remain unclear. Here we show that platelet endothelial cell adhesion molecule-1 (PECAM1), a key molecule in an endothelial mechanosensing complex, specifically mediates Gab1 tyrosine phosphorylation and its downstream Akt and eNOS activation in ECs upon flow rather than hepatocyte growth factor (HGF) stimulation. Small interfering RNA (siRNA) targeting PECAM1 abolished flow- but not HGF-induced Gab1 tyrosine phosphorylation and Akt, eNOS activation as well as Gab1 membrane translocation. Protein-tyrosine phosphatase SHP2, which has been shown to interact with Gab1, was involved in flow signaling and HGF signaling, as SHP2 siRNA diminished the flow- and HGF-induced Gab1 tyrosine phosphorylation, membrane localization and downstream signaling. Pharmacological inhibition of PI3K decreased flow-, but not HGF-mediated Gab1 phosphorylation and membrane localization as well as eNOS activation. Finally, we observed that flow-mediated Gab1 and eNOS phosphorylation in vivo induced by voluntary wheel running was reduced in PECAM1 knockout mice. These results demonstrate a specific role of PECAM1 in flow-mediated Gab1 tyrosine phosphorylation and eNOS signaling in ECs.


Asunto(s)
Fosfoproteínas/metabolismo , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Transducción de Señal , Proteínas Adaptadoras Transductoras de Señales , Animales , Cromonas/farmacología , Factor de Crecimiento de Hepatocito/genética , Factor de Crecimiento de Hepatocito/metabolismo , Factor de Crecimiento de Hepatocito/farmacología , Células Endoteliales de la Vena Umbilical Humana , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Morfolinas/farmacología , Óxido Nítrico Sintasa de Tipo III/metabolismo , Fosforilación/efectos de los fármacos , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/química , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 11/antagonistas & inhibidores , Proteína Tirosina Fosfatasa no Receptora Tipo 11/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 11/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/farmacología , Transducción de Señal/efectos de los fármacos , Tirosina/metabolismo
11.
Zhong Nan Da Xue Xue Bao Yi Xue Ban ; 40(7): 723-8, 2015 Jul.
Artículo en Chino | MEDLINE | ID: mdl-26267683

RESUMEN

OBJECTIVE: To explore the effect of Fuzheng Huayu (FZHY) recipe on the fenestration of capillarization in liver sinusoidal endothelial cells (LSECs).
 METHODS: Ten Sprague Dawley (SD) rats were fed with 0.46 g/kg FZHY powder by intragastric administration. Two hours later, a second gavage were given to the rats. The serum from rat heart at 1 hour after second gavage was collected (FZHY group, n=10). Another ten SD rats was administrated with distilled water through the same process and served as the control (control group, n=10). The serum from both groups were separately diluted with Dulbecco minimum essential medium (DMEM) for 10% and served as the culture medium for LSECs. At the different conditions, the vWF and CD31 expressions were detected by immunocytochemistry and Western blot, while the changes of LSECs fenestrae structure were observed under scanning electron microscopy.
 RESULTS: 1) Immunocytochemistry and Western blot showed that the vWF and CD31 protein levels were lower in LSECs in the FZHY group than those in the control group. The gray levels of vWF and CD31 protein were 0.548±0.020 and 0.262±0.010 in the FZHY group, and 0.845±0.090 and 0.383±0.010 in the control group respectively, with statistical significant difference (t=5.18, 9.61, both P<0.05). 2) The results from scanning electron microscopy showed that the fenestration of LSECs was closed and almost lost in the control group, but many fenestra appeared in the LSECs in the FZHY group.
 CONCLUSION: FZHY recipe can suppress the expression of vWF and CD31, increase the fenestrae on the LSECs surface and reverse the capillarization of LSECs.


Asunto(s)
Medicamentos Herbarios Chinos/farmacología , Células Endoteliales/efectos de los fármacos , Hígado/citología , Animales , Células Endoteliales/citología , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/química , Ratas , Ratas Sprague-Dawley , Factor de von Willebrand/química
12.
ACS Nano ; 9(7): 6785-93, 2015 Jul 28.
Artículo en Inglés | MEDLINE | ID: mdl-26153796

RESUMEN

Nanocarriers (NCs) coated with antibodies (Abs) to extracellular epitopes of the transmembrane glycoprotein PECAM (platelet endothelial cell adhesion molecule-1/CD31) enable targeted drug delivery to vascular endothelial cells. Recent studies revealed that paired Abs directed to adjacent, yet distinct epitopes of PECAM stimulate each other's binding to endothelial cells in vitro and in vivo ("collaborative enhancement"). This phenomenon improves targeting of therapeutic fusion proteins, yet its potential role in targeting multivalent NCs has not been addressed. Herein, we studied the effects of Ab-mediated collaborative enhancement on multivalent NC spheres coated with PECAM Abs (Ab/NC, ∼180 nm diameter). We found that PECAM Abs do mutually enhance endothelial cell binding of Ab/NC coated by paired, but not "self" Ab. In vitro, collaborative enhancement of endothelial binding of Ab/NC by paired Abs is modulated by Ab/NC avidity, epitope selection, and flow. Cell fixation, but not blocking of endocytosis, obliterated collaborative enhancement of Ab/NC binding, indicating that the effect is mediated by molecular reorganization of PECAM molecules in the endothelial plasmalemma. The collaborative enhancement of Ab/NC binding was affirmed in vivo. Intravascular injection of paired Abs enhanced targeting of Ab/NC to pulmonary vasculature in mice by an order of magnitude. This stimulatory effect greatly exceeded enhancement of Ab targeting by paired Abs, indicating that '"collaborative enhancement"' effect is even more pronounced for relatively large multivalent carriers versus free Abs, likely due to more profound consequences of positive alteration of epitope accessibility. This phenomenon provides a potential paradigm for optimizing the endothelial-targeted nanocarrier delivery of therapeutic agents.


Asunto(s)
Plaquetas/metabolismo , Epítopos/inmunología , Nanosferas/química , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Animales , Membrana Celular/metabolismo , Femenino , Células Endoteliales de la Vena Umbilical Humana , Ratones , Ratones Endogámicos C57BL , Nanosferas/metabolismo , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/química , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/inmunología , Unión Proteica
13.
J Biol Chem ; 289(30): 20836-44, 2014 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-24936065

RESUMEN

PECAM-1 is a 130-kDa member of the immunoglobulin (Ig) superfamily that is expressed on the surface of platelets and leukocytes, and at the intracellular junctions of confluent endothelial cell monolayers. Previous studies have shown that PECAM-1/PECAM-1 homophilic interactions play a key role in leukocyte transendothelial migration, in allowing PECAM-1 to serve as a mechanosensory complex in endothelial cells, in its ability to confer cytoprotection to proapoptotic stimuli, and in maintaining endothelial cell junctional integrity. To examine the adhesive properties of full-length PECAM-1 in a native lipid environment, we purified it from platelets and assembled it into phospholipid nanodiscs. PECAM-1-containing nanodiscs retained not only their ability to bind homophilically to PECAM-1-expressing cells, but exhibited regulatable adhesive interactions that could be modulated by ligands that bind membrane- proximal Ig Domain 6. This property was exploited to enhance the rate of barrier restoration in endothelial cell monolayers subjected to inflammatory challenge. The finding that the adhesive properties of PECAM-1 are regulatable suggests novel approaches for controlling endothelial cell migration and barrier function in a variety of vascular permeability disorders.


Asunto(s)
Anticuerpos/farmacología , Permeabilidad Capilar/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Enfermedades Vasculares/metabolismo , Anticuerpos/inmunología , Permeabilidad Capilar/inmunología , Movimiento Celular/inmunología , Células HEK293 , Células Endoteliales de la Vena Umbilical Humana/química , Humanos , Membranas Artificiales , Fosfolípidos/química , Fosfolípidos/inmunología , Fosfolípidos/metabolismo , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/química , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/inmunología , Estructura Terciaria de Proteína , Enfermedades Vasculares/inmunología , Enfermedades Vasculares/patología
14.
ACS Nano ; 8(5): 4996-5009, 2014 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-24720540

RESUMEN

The tumor microenvironment plays an important role in the tumor's progression and metastasis. Therefore, successful alteration of this delicate setting against the tumor's favor can open a window for therapeutic efficacy. We have developed a modality to bring about treatment-induced alterations in the tumor microenvironment by employing the synergistic effects between two drugs. Co-delivery of rapamycin (RAPA), an mTOR inhibitor that may offer notable therapy through antiangiogenic activity, alongside cisplatin can foster significant potency as RAPA sensitizes A375 melanoma cells to cisplatin therapy through microenvironment modulation. However, encapsulation of these drugs into poly(lactic-co-glycolic acid) (PLGA) NPs was inefficient due to the incompatibility between the two free drugs and the polymer matrix. Here, we show cisplatin can be made hydrophobic by coating a nanoprecipitate (cores) of the drug with dioleoylphosphatidic acid (DOPA). These DOPA coated cisplatin cores are compatible with PLGA and can be coencapsulated in PLGA NPs alongside RAPA at a molar ratio to promote synergistic antitumor activity. The presence of the cisplatin cores significantly improved the encapsulation of RAPA into PLGA NPs. Furthermore, PLGA NPs containing both cisplatin cores and RAPA induced significant apoptosis on A375-luc human melanoma cells in vitro. Additionally, they inhibited the growth of A375-luc melanoma in a xenograft tumor model through modulation of the tumor vasculature and permitted enhanced penetration of NPs into the tumor.


Asunto(s)
Antineoplásicos/administración & dosificación , Cisplatino/administración & dosificación , Neoplasias/tratamiento farmacológico , Sirolimus/administración & dosificación , Animales , Apoptosis , Línea Celular Tumoral , Cromatografía Líquida de Alta Presión , Cisplatino/química , Dihidroxifenilalanina/química , Progresión de la Enfermedad , Portadores de Fármacos , Humanos , Etiquetado Corte-Fin in Situ , Ácido Láctico/química , Melanoma/tratamiento farmacológico , Ratones , Nanopartículas/química , Nanotecnología/métodos , Metástasis de la Neoplasia , Trasplante de Neoplasias , Neoplasias/patología , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/química , Ácido Poliglicólico/química , Copolímero de Ácido Poliláctico-Ácido Poliglicólico , Neoplasias Cutáneas/tratamiento farmacológico , Solventes/química , Microambiente Tumoral
15.
Mol Immunol ; 58(2): 206-13, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24388971

RESUMEN

SAP is a small cytosolic adaptor protein expressed in hematopoietic lineages whose main function is to regulate intracellular signaling pathways induced by the triggering of members of the SLAM receptor family. In this paper, we have identified the adhesion molecule PECAM-1 as a new partner for SAP in a conditional yeast two-hybrid screen. PECAM-1 is an immunoglobulin-like molecule expressed by endothelial cells and leukocytes, which possesses both pro- and anti-inflammatory properties. However, little is known about PECAM-1 functions in T cells. We show that SAP directly and specifically interacts with the cytosolic tyrosine 686 of PECAM-1. We generated different T-like cell lines in which SAP or PECAM-1 are expressed or down modulated and we demonstrate that a diminished SAP expression correlates with a diminished PECAM-1-mediated adhesion. Although SAP has mainly been shown to associate with SLAM receptors, we evidence here that SAP is a new actor downstream of PECAM-1.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular/metabolismo , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Linfocitos T/fisiología , Secuencia de Aminoácidos , Secuencia de Bases , Adhesión Celular , Línea Celular , Células HeLa , Humanos , Péptidos y Proteínas de Señalización Intracelular/química , Células Jurkat , Datos de Secuencia Molecular , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/química , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Proteína Asociada a la Molécula de Señalización de la Activación Linfocitaria
16.
Microvasc Res ; 89: 1-6, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23851046

RESUMEN

Patients with nephrotic syndrome (NS) carry a high risk of venous thromboembolism (VTE) due to the abnormalities in coagulation and fibrinolysis. Although massive urine protein loss is considered to trigger the cascade of hypercoagulation, the exact nature of VTE in NS patients still remains obscure, especially in some cases when VTE occurs far before the presence of nephrotic proteinuria. Recent findings illustrate that loss of local glomerular homeostasis, like disturbance of cytokine profiles in endothelial cells or aberrant cellular crosstalks in glomerulus, is sufficient to initiate the development of thrombotic disease in glomerulonephropathy. Emerging data have highlighted the glomerular endothelial cell as a key regulator of local homeostasis, which might mediate the haemostatic derangement in the beginning of glomerular disease by expression of numerous prothrombotic factors and result in the subsequent predilection of VTE in NS. As the glomerulus-derived circulating factors are all collected and flushed into the renal vein directly, it is reasonable to suggest that increased release of glomerulus-derived thrombotic regulators, particularly from endothelial cells, may play a significant role in the highest proclivity for the renal vein as the site of thrombosis in NS. In this review, we thus discuss the current understandings of thromboembolism in NS with focus on how the glomerular endothelial cell involves in the pathogenesis of VTE, which may help to increase our understandings in the anti-thrombotic therapy for patients with NS.


Asunto(s)
Células Endoteliales/citología , Síndrome Nefrótico/fisiopatología , Trombosis/fisiopatología , Animales , Coagulación Sanguínea , Micropartículas Derivadas de Células/metabolismo , Fibrinólisis , Homeostasis , Humanos , Riñón/metabolismo , Glomérulos Renales/patología , Óxido Nítrico/química , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/química , Proteinuria/diagnóstico , Riesgo , Tromboembolia/patología , Trombosis/patología
17.
Contrast Media Mol Imaging ; 8(5): 393-401, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23740809

RESUMEN

An increasing amount of studies have provided evidence for vascular remodeling, for example, angiogenesis, after cerebral ischemia, which may play a significant role in post-stroke brain plasticity and recovery. Molecular imaging can provide unique in vivo whole-brain information on alterations in the expression of specific endothelial markers. A possible target for molecular magnetic resonance imaging (MRI) of post-stroke (neo)vascularization is platelet endothelial cell adhesion molecule-1 (PECAM-1). Here we describe significantly increased PECAM-1 mRNA levels in ipsilesional brain tissue at 6 h, 24 h and 3 days after transient middle cerebral artery occlusion in mice, and elevated PECAM-1 staining throughout the lesion at 3, 7 and 21 days post-stroke. The potential of micron-sized particles of iron oxide (MPIO) conjugated with PECAM-1-targeted antibodies, that is, αPECAM-1-MPIO, to expose stroke-induced PECAM-1 upregulation with molecular MRI was assessed. In vitro studies demonstrated that PECAM-1-expressing brain endothelial cells could be effectively labeled with αPECAM-1-MPIO, giving rise to a fourfold increase in MRI relaxation rate R2. Injection of near-infrared fluorescent dye-labeled αPECAM-1 showed target specificity and dose efficiency of the antibody for detection of brain endothelial cells at 3 days post-stroke. However, in vivo molecular MRI at 3 and 7 days after stroke revealed no αPECAM-1-MPIO-based contrast enhancement, which was corroborated by the absence of αPECAM-1-MPIO in post mortem brain tissue. This indicates that this molecular MRI approach, which has been proven successful for in vivo detection of other types of cell adhesion molecules, is not invariably effective for MRI-based assessment of stroke-induced alterations in expression of cerebrovascular markers.


Asunto(s)
Medios de Contraste/administración & dosificación , Compuestos Férricos/administración & dosificación , Angiografía por Resonancia Magnética , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Accidente Cerebrovascular/diagnóstico por imagen , Animales , Isquemia Encefálica/patología , Medios de Contraste/química , Compuestos Férricos/química , Regulación de la Expresión Génica , Humanos , Infarto de la Arteria Cerebral Media/patología , Ratones , Imagen Molecular , Neovascularización Fisiológica , Tamaño de la Partícula , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/biosíntesis , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/química , Radiografía , Accidente Cerebrovascular/patología
18.
Curr Biol ; 23(11): 1024-30, 2013 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-23684974

RESUMEN

Fluid shear stress (FSS) from blood flow acting on the endothelium critically regulates vascular morphogenesis, blood pressure, and atherosclerosis. FSS applied to endothelial cells (ECs) triggers signaling events including opening of ion channels, activation of signaling pathways, and changes in gene expression. Elucidating how ECs sense flow is important for understanding both normal vascular function and disease. EC responses to FSS are mediated in part by a junctional mechanosensory complex consisting of VE-cadherin, PECAM-1, and VEGFR2. Previous work suggested that flow increases force on PECAM-1, which initiates signaling. Deletion of PECAM-1 blocks responses to flow in vitro and flow-dependent vascular remodeling in vivo. To understand this process, we developed and validated FRET-based tension sensors for VE-cadherin and PECAM-1 using our previously developed FRET tension biosensor. FRET measurements showed that in static culture, VE-cadherin in cell-cell junctions bears significant myosin-dependent tension, whereas there was no detectable tension on VE-cadherin outside of junctions. Onset of shear stress triggered a rapid (<30 s) decrease in tension across VE-cadherin, which paralleled a decrease in total cell-cell junctional tension. Flow triggered a simultaneous increase in tension across junctional PECAM-1, while nonjunctional PECAM-1 was unaffected. Tension on PECAM-1 was mediated by flow-stimulated association with vimentin. These data confirm the prediction that shear increases force on PECAM-1. However, they also argue against the current model of passive transfer of force through the cytoskeleton to the junctions, showing instead that flow triggers cytoskeletal remodeling, which alters forces across the junctional receptors.


Asunto(s)
Antígenos CD/metabolismo , Cadherinas/metabolismo , Movimiento Celular , Adhesiones Focales/metabolismo , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Estrés Mecánico , Animales , Antígenos CD/química , Antígenos CD/genética , Técnicas Biosensibles , Cadherinas/química , Cadherinas/genética , Línea Celular , Citoesqueleto/metabolismo , Células Endoteliales/metabolismo , Humanos , Ratones , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/química , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/genética , Transducción de Señal
19.
Int J Nanomedicine ; 7: 3769-86, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22888233

RESUMEN

This paper presents the synthesis of aqueous cadmium sulfide (CdS) quantum dots (QDs) and silica-encapsulated CdS QDs by reverse microemulsion method and utilized as targeted bio-optical probes. We report the role of CdS as an efficient cell tag with fluorescence on par with previously documented cadmium telluride and cadmium selenide QDs, which have been considered to impart high levels of toxicity. In this study, the toxicity of bare QDs was efficiently quenched by encapsulating them in a biocompatible coat of silica. The toxicity profile and uptake of bare CdS QDs and silica-coated QDs, along with the CD31-labeled, silica-coated CdS QDs on human umbilical vein endothelial cells and glioma cells, were investigated. The effect of size, along with the time-dependent cellular uptake of the nanomaterials, has also been emphasized. Enhanced, high-specificity imaging toward endothelial cell lines in comparison with glioma cells was achieved with CD31 antibody-conjugated nanoparticles. The silica-coated nanomaterials exhibited excellent biocompatibility and greater photostability inside live cells, in addition to possessing an extended shelf life. In vivo biocompatibility and localization study of silica-coated CdS QDs in medaka fish embryos, following direct nanoparticle exposure for 24 hours, authenticated the nanomaterials' high potential for in vivo imaging, augmented with superior biocompatibility. As expected, CdS QD-treated embryos showed 100% mortality, whereas the silica-coated QD-treated embryos stayed viable and healthy throughout and after the experiments, devoid of any deformities. We provide highly cogent and convincing evidence for such silica-coated QDs as a model nanoparticle in practice, to achieve in vitro and in vivo precision targeted imaging.


Asunto(s)
Compuestos de Cadmio/química , Nanopartículas/química , Imagen Óptica/métodos , Puntos Cuánticos , Compuestos de Selenio/química , Dióxido de Silicio/química , Animales , Compuestos de Cadmio/toxicidad , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Embrión no Mamífero/efectos de los fármacos , Glioma/tratamiento farmacológico , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Humanos , Ensayo de Materiales , Nanopartículas/toxicidad , Oryzias , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/química , Compuestos de Selenio/toxicidad , Dióxido de Silicio/toxicidad , Espectrometría de Fluorescencia , Análisis Espectral/métodos
20.
Platelets ; 23(5): 331-5, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22035359

RESUMEN

Platelet endothelial cell adhesion molecule-1 (PECAM-1), an immunoreceptor tyrosine-based inhibitory motif containing receptor, plays diverse and apparently contradictory roles in regulating the response of platelets to stimuli; inhibiting platelet response to immunoreceptor tyrosine-based activation motif and G protein-coupled receptor signalling following stimulation with collagen, adenosine diphosphate, and thrombin, as well as enhancing integrin outside-in signalling. These dual, and opposing, roles suggest an important and complex role for PECAM-1 in orchestrating platelet response to vascular damage. Indeed, during thrombus formation, the influence of PECAM-1 on the multiple signalling pathways combines leading to a relatively large inhibitory effect on thrombus formation.


Asunto(s)
Plaquetas/metabolismo , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/sangre , Plaquetas/citología , Humanos , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/química , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA