Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 76
Filtrar
1.
Blood ; 142(22): 1909-1917, 2023 11 30.
Artículo en Inglés | MEDLINE | ID: mdl-37738558

RESUMEN

Sialic acids occupy the terminal position of glycan chains and have the potential to influence the antigenicity of glycoproteins (GP). The polymorphisms of human platelet alloantigens (HPA)-3 and HPA-9, located near the C-terminus of the extracellular domain of platelet membrane GPIIb, are adjacent to sialyl-core 1 O-glycans emanating from serines 845 and 847. Whether the nearby O-glycans affect the antigenicity of HPA-9b or influence the binding of anti-HPA-9b alloantibodies in clinically significant cases of neonatal alloimmune thrombocytopenia is unknown. To address this issue, we generated a series of O-glycan mutant HPA-9 allele-specific induced pluripotent stem cell lines, differentiated them to megakaryocytes (MKs), and examined their ability to bind HPA-9b-specific alloantibodies. We found that both wild-type MKs treated with neuraminidase and those genetically modified to lack the sialidases ST3GAL1 and ST3GAL2 dramatically increased anti-HPA-9b alloantibody binding, indicating that the HPA-9b epitope is partially masked by terminal sialic acids on nearby O-glycans of GPIIb. Interestingly, mutating the serine residues that carry these glycan chains to alanine actually reduced the binding of anti-HPA-9b alloantibodies, indicating that these 2 O-glycan chains contribute to the presentation of the HPA-9b epitope-perhaps by stabilizing the conformation of the GP in this region. Collectively, our data suggest that detection of anti-HPA-9b alloantibodies may be enhanced through the use of HPA-9b-specific MKs that have been genetically altered to lack nearby terminal sialic acid residues but retain the glycan chains to which they are attached.


Asunto(s)
Antígenos de Plaqueta Humana , Recién Nacido , Humanos , Glicoproteína IIb de Membrana Plaquetaria , Ácido N-Acetilneuramínico , Isoanticuerpos , Glicoproteínas , Polisacáridos , Epítopos
2.
Blood ; 140(20): 2146-2153, 2022 11 17.
Artículo en Inglés | MEDLINE | ID: mdl-35881848

RESUMEN

Fetal/neonatal alloimmune thrombocytopenia (FNAIT) is a life-threatening bleeding disorder caused by maternal alloantibodies directed against paternally inherited human platelet alloantigens (HPAs) present on the surface of fetal and neonatal platelets. There are currently no approved therapies for the prevention of FNAIT. We report herein the ability of 2 human HPA-1a-specific therapeutic candidates, one a polyclonal, and the other a monoclonal antibody, to prevent alloimmunization in a novel preclinical mouse model of FNAIT. Both antibody preparations effected the rapid and complete elimination of HPA-1a+ platelets from circulation and prevented the development of HPA-1a alloantibodies. HPA-1a- female mice treated prophylactically with anti-HPA-1a antibody prior to exposure to HPA-1a+ platelets gave birth to HPA-1a+/- pups with significantly improved platelet counts and no bleeding symptoms. These preclinical data establish both the potential and threshold exposure targets for prophylactic treatment with HPA-1a-specific antibodies for the prevention of FNAIT in humans.


Asunto(s)
Antígenos de Plaqueta Humana , Trombocitopenia Neonatal Aloinmune , Embarazo , Humanos , Femenino , Ratones , Animales , Trombocitopenia Neonatal Aloinmune/prevención & control , Isoanticuerpos , Integrina beta3 , Atención Prenatal , Feto
3.
Arterioscler Thromb Vasc Biol ; 42(2): 193-204, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34937389

RESUMEN

OBJECTIVE: PECAM-1 (platelet endothelial cell adhesion molecule 1) is a 130 kDa member of the immunoglobulin (Ig) gene superfamily that is expressed on the surfaces of platelets and leukocytes and concentrated at the intercellular junctions of confluent endothelial cell monolayers. PECAM-1 Ig domains 1 and 2 (IgD1 and IgD2) engage in homophilic interactions that support a host of vascular functions, including support of leukocyte transendothelial migration and the maintenance of endothelial junctional integrity. The recently solved crystal structure of PECAM-1 IgD1 and IgD2 revealed a number of intermolecular interfaces predicted to play important roles in stabilizing PECAM-1/PECAM-1 homophilic interactions and in formation and maintenance of endothelial cell-cell contacts. We sought to determine whether the protein interfaces implicated in the crystal structure reflect physiologically important interactions. Approach and Results: We assessed the impact of single amino acid substitutions at the interfaces between opposing PECAM-1 molecules on homophilic binding and endothelial cell function. Substitution of key residues within the IgD1-IgD1 and IgD1-IgD2 interfaces but not those within the smaller IgD2-IgD2 interface, markedly disrupted PECAM-1 homophilic binding and its downstream effector functions, including the ability of PECAM-1 to localize at endothelial cell-cell borders, mediate the formation of endothelial tubes, and restore endothelial barrier integrity. CONCLUSIONS: Taken together, these results validate the recently described PECAM-1 IgD1/IgD2 crystal structure by demonstrating that specific residues visualized within the IgD1-IgD1 and IgD1-IgD2 interfaces of opposing molecules in the crystal are required for functionally important homophilic interactions. This information can now be exploited to modulate functions of PECAM-1 in vivo.


Asunto(s)
Células Endoteliales/metabolismo , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Adhesión Celular , Comunicación Celular , Células Endoteliales/citología , Células Endoteliales de la Vena Umbilical Humana , Humanos , Modelos Moleculares , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/análisis , Unión Proteica
4.
J Cell Mol Med ; 25(19): 9340-9349, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34547166

RESUMEN

Despite the recent advancements in transfusion medicine, red blood cell (RBC) alloimmunization remains a challenge for multiparous women and chronically transfused patients. At times, diagnostic laboratories depend on difficult-to-procure rare reagent RBCs for the identification of different alloantibodies in such subjects. We have addressed this issue by developing erythroblasts with custom phenotypes (Rh null, GPB null and Kx null/Kell low) using CRISPR/Cas9 gene-editing of a human induced pluripotent stem cell (hiPSC) parent line (OT1-1) for the blood group system genes: RHAG, GYPB and XK. Guide RNAs were cloned into Cas9-puromycin expression vector and transfected into OT1-1. Genotyping was performed to select puromycin-resistant hiPSC KOs. CRISPR/Cas9 gene-editing resulted in the successful generation of three KO lines, RHAG KO, GYPB KO and XK KO. The OT1-1 cell line, as well as the three KO hiPSC lines, were differentiated into CD34+ CD41+ CD235ab+ hematopoietic progenitor cells (HPCs) and subsequently to erythroblasts. Native OT1-1 erythroblasts were positive for the expression of Rh, MNS, Kell and H blood group systems. Differentiation of RHAG KO, GYPB KO and XK KO resulted in the formation of Rh null, GPB null and Kx null/Kell low erythroblasts, respectively. OT1-1 as well as the three KO erythroblasts remained positive for RBC markers-CD71 and BAND3. Erythroblasts were mostly at the polychromatic/ orthochromatic stage of differentiation. Up to ~400-fold increase in erythroblasts derived from HPCs was observed. The availability of custom erythroblasts generated from CRISPR/Cas9 gene-edited hiPSC should be a useful addition to the tools currently used for the detection of clinically important red cell alloantibodies.


Asunto(s)
Sistemas CRISPR-Cas , Diferenciación Celular , Linaje de la Célula , Eritroblastos/metabolismo , Edición Génica , Células Madre Pluripotentes Inducidas/metabolismo , Biomarcadores , Antígenos de Grupos Sanguíneos/genética , Antígenos de Grupos Sanguíneos/metabolismo , Línea Celular , Eritroblastos/citología , Técnicas de Silenciamiento del Gen , Hematopoyesis , Histocitoquímica , Humanos , Inmunofenotipificación , Células Madre Pluripotentes Inducidas/citología , ARN Guía de Kinetoplastida/genética
5.
Blood ; 134(22): e1-e8, 2019 11 28.
Artículo en Inglés | MEDLINE | ID: mdl-31697836

RESUMEN

Human platelet membrane glycoprotein polymorphisms can be immunogenic in man and are frequently the cause of clinically important immune reactions responsible for disorders such as neonatal alloimmune thrombocytopenia. Platelets from individuals carrying rare polymorphisms are often difficult to obtain, making diagnostic testing and transfusion of matched platelets challenging. In addition, class I HLA antibodies frequently present in maternal sera interfere with the detection of platelet-reactive alloantibodies. Detection of alloantibodies to human platelet antigen 3 (HPA-3) and HPA-9 is especially challenging, in part because of the presence of cell type-specific glycans situated near the polymorphic amino acid that together form the alloepitope. To overcome these limitations, we generated a series of HLA class I-negative blood group O induced pluripotent stem cell (iPSC) lines that were gene edited to sequentially convert their endogenous HPA-3a alloantigenic epitope to HPA-3b, and HPA-9a to HPA-9b. Subjecting these cell lines, upon differentiation into CD41+/CD42b+ human megakaryocytes (MKs), to flow cytometric detection of suspected anti-HPA-3 and HPA-9 alloantisera revealed that the HPA-3a-positive MKs specifically reacted with HPA-3a patient sera, whereas the HPA-3b MKs lost reactivity with HPA-3a patient sera while acquiring reactivity to HPA-3b patient sera. Importantly, HPA-9b-expressing MKs specifically reacted with anti-HPA-9b-suspected patient samples that had been undetectable using conventional techniques. The provision of specialized iPSC-derived human MKs expressing intact homozygous glycoprotein alloantigens on the cell surface that carry the appropriate endogenous carbohydrate moieties should greatly enhance detection of clinically important and rare HPA-specific alloantibodies that, to date, have resisted detection using current methods.


Asunto(s)
Antígenos de Plaqueta Humana/inmunología , Ingeniería Celular , Células Madre Pluripotentes Inducidas/inmunología , Isoanticuerpos/inmunología , Megacariocitos/inmunología , Antígenos de Plaqueta Humana/genética , Antígenos de Plaqueta Humana/metabolismo , Citometría de Flujo , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Isoanticuerpos/sangre , Megacariocitos/metabolismo
6.
Proc Natl Acad Sci U S A ; 115(7): E1550-E1559, 2018 02 13.
Artículo en Inglés | MEDLINE | ID: mdl-29386381

RESUMEN

There is a growing appreciation for the contribution of platelets to immunity; however, our knowledge mostly relies on platelet functions associated with vascular injury and the prevention of bleeding. Circulating immune complexes (ICs) contribute to both chronic and acute inflammation in a multitude of clinical conditions. Herein, we scrutinized platelet responses to systemic ICs in the absence of tissue and endothelial wall injury. Platelet activation by circulating ICs through a mechanism requiring expression of platelet Fcγ receptor IIA resulted in the induction of systemic shock. IC-driven shock was dependent on release of serotonin from platelet-dense granules secondary to platelet outside-in signaling by αIIbß3 and its ligand fibrinogen. While activated platelets sequestered in the lungs and leaky vasculature of the blood-brain barrier, platelets also sequestered in the absence of shock in mice lacking peripheral serotonin. Unexpectedly, platelets returned to the blood circulation with emptied granules and were thereby ineffective at promoting subsequent systemic shock, although they still underwent sequestration. We propose that in response to circulating ICs, platelets are a crucial mediator of the inflammatory response highly relevant to sepsis, viremia, and anaphylaxis. In addition, platelets recirculate after degranulation and sequestration, demonstrating that in adaptive immunity implicating antibody responses, activated platelets are longer lived than anticipated and may explain platelet count fluctuations in IC-driven diseases.


Asunto(s)
Anafilaxia/inmunología , Complejo Antígeno-Anticuerpo/inmunología , Plaquetas/inmunología , Serotonina/inmunología , Choque Séptico/inmunología , Adulto , Anafilaxia/sangre , Anafilaxia/genética , Animales , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Activación Plaquetaria , Recuento de Plaquetas , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/genética , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/inmunología , Receptores de IgG/genética , Receptores de IgG/inmunología , Choque Séptico/sangre , Choque Séptico/genética , Adulto Joven
7.
Genesis ; 58(2): e23346, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31729819

RESUMEN

Platelet endothelial cell adhesion molecule 1 (PECAM-1) is an adhesion and signaling receptor that is expressed on endothelial and hematopoietic cells and plays important roles in angiogenesis, vascular permeability, and regulation of cellular responsiveness. To better understanding the tissue specificity of PECAM-1 functions, we generated mice in which PECAM1, the gene encoding PECAM-1, could be conditionally knocked out. A targeting construct was created that contains loxP sites flanking PECAM1 exons 1 and 2 and a neomycin resistance gene flanked by flippase recognition target (FRT) sites that was positioned upstream of the 3' loxP site. The targeting construct was electroporated into C57BL/6 embryonic stem (ES) cells, and correctly targeted ES cells were injected into C57BL/6 blastocysts, which were implanted into pseudo-pregnant females. Resulting chimeric animals were bred with transgenic mice expressing Flippase 1 (FLP1) to remove the FRT-flanked neomycin resistance gene and mice heterozygous for the floxed PECAM1 allele were bred with each other to obtain homozygous PECAM1 flox/flox offspring, which expressed PECAM-1 at normal levels and had no overt phenotype. PECAM1 flox/flox mice were bred with mice expressing Cre recombinase under the control of the SRY-box containing gene 2 (Sox2Cre) promoter to delete the floxed PECAM1 allele in offspring (Sox2Cre;PECAM1 del/WT ), which were crossbred to generate Sox2Cre; PECAM1 del/del offspring. Sox2Cre; PECAM1 del/del mice recapitulated the phenotype of conventional global PECAM-1 knockout mice. PECAM1 flox/flox mice will be useful for studying distinct roles of PECAM-1 in tissue specific contexts and to gain insights into the roles that PECAM-1 plays in blood and vascular cell function.


Asunto(s)
Técnicas de Inactivación de Genes/métodos , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/genética , Animales , Integrasas/metabolismo , Ratones , Ratones Endogámicos C57BL , Células Madre Embrionarias de Ratones/metabolismo , Factores de Transcripción SOXB1/genética , Factores de Transcripción SOXB1/metabolismo
8.
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
9.
Blood ; 127(6): 675-80, 2016 Feb 11.
Artículo en Inglés | MEDLINE | ID: mdl-26634302

RESUMEN

Human platelet alloantigens (HPAs) reside on functionally important platelet membrane glycoproteins and are caused by single nucleotide polymorphisms in the genes that encode them. Antibodies that form against HPAs are responsible for several clinically important alloimmune bleeding disorders, including fetal and neonatal alloimmune thrombocytopenia and posttransfusion purpura. The HPA-1a/HPA-1b alloantigen system, also known as the Pl(A1)/Pl(A2) polymorphism, is the most frequently implicated HPA among whites, and a single Leu33Pro amino acid polymorphism within the integrin ß3 subunit is responsible for generating the HPA-1a/HPA-1b alloantigenic epitopes. HPA-1b/b platelets, like those bearing other low-frequency platelet-specific alloantigens, are relatively rare in the population and difficult to obtain for purposes of transfusion therapy and diagnostic testing. We used CRISPR/Cas9 (clustered regularly interspaced short palindromic repeats/CRISPR associated protein 9) gene-editing technology to transform Leu33 (+) megakaryocytelike DAMI cells and induced pluripotent stem cells (iPSCs) to the Pro33 allotype. CD41(+) megakaryocyte progenitors derived from these cells expressed the HPA-1b (Pl(A2)) alloantigenic epitope, as reported by diagnostic NciI restriction enzyme digestion, DNA sequencing, and western blot analysis using HPA-1b-specific human maternal alloantisera. Application of CRISPR/Cas9 technology to genetically edit this and other clinically-important HPAs holds great potential for production of designer platelets for diagnostic, investigative, and, ultimately, therapeutic use.


Asunto(s)
Antígenos de Plaqueta Humana/genética , Proteínas Asociadas a CRISPR/genética , Proteínas Asociadas a CRISPR/inmunología , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Isoantígenos/genética , Antígenos de Plaqueta Humana/inmunología , Secuencia de Bases , Células Cultivadas , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas/genética , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas/inmunología , Epítopos/genética , Epítopos/inmunología , Humanos , Integrina beta3/genética , Integrina beta3/inmunología , Isoanticuerpos/genética , Isoanticuerpos/inmunología , Isoantígenos/inmunología , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/genética , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/metabolismo , Glicoproteínas de Membrana Plaquetaria/metabolismo , Polimorfismo de Nucleótido Simple
10.
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
11.
Arterioscler Thromb Vasc Biol ; 36(8): 1517-24, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27283740

RESUMEN

OBJECTIVE: Fetal/neonatal alloimmune thrombocytopenia is a severe bleeding disorder, which can result in intracranial hemorrhage (ICH), leading to death or neurological sequelae. In whites, maternal anti-human platelet antigen-1a (HPA-1a) antibodies are responsible for the majority of cases. No predictive factors for ICH are available to guide prophylactic treatment during pregnancy. In this study, we investigated antibodies from mothers with ICH-positive fetal/neonatal alloimmune thrombocytopenia and with ICH-negative fetal/neonatal alloimmune thrombocytopenia to identify serological and functional differences between the groups. APPROACH AND RESULTS: In an antigen capture assay, we observed a stronger binding of +ICH antibodies to endothelial cell (EC)-derived αvß3. By absorption experiments, we subsequently identified anti-HPA-1a antibodies of anti-αvß3 specificity in the +ICH but not in the -ICH cohort. Only the anti-αvß3 subtype, but not the anti-ß3 subtype, induced EC apoptosis of HPA-1a-positive ECs by caspase-3/7 activation, and mediated by reactive oxygen species. In addition, only the anti-αvß3 subtype, but not the anti-ß3 subtype, interfered with EC adhesion to vitronectin and with EC tube formation. CONCLUSIONS: We conclude that the composition of the anti-HPA-1a antibody subtype(s) of the mother may determine whether ICH occurs. Analysis of anti-HPA-1a antibodies of the anti-αvß3 subtype in maternal serum has potential in the diagnostic prediction of ICH development and may allow for modification of prophylactic treatment in fetal/neonatal alloimmune thrombocytopenia.


Asunto(s)
Autoanticuerpos/inmunología , Células Endoteliales/inmunología , Integrina alfaVbeta3/inmunología , Hemorragias Intracraneales/inmunología , Trombocitopenia Neonatal Aloinmune/inmunología , Animales , Especificidad de Anticuerpos , Antígenos de Plaqueta Humana/inmunología , Antígenos de Plaqueta Humana/metabolismo , Apoptosis , Autoanticuerpos/metabolismo , Células CHO , Estudios de Casos y Controles , Caspasa 3/metabolismo , Caspasa 7/metabolismo , Adhesión Celular , Cricetulus , Células Endoteliales/metabolismo , Células Endoteliales/patología , Femenino , Edad Gestacional , Células Endoteliales de la Vena Umbilical Humana/inmunología , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Células Endoteliales de la Vena Umbilical Humana/patología , Humanos , Recién Nacido , Integrina alfaVbeta3/genética , Integrina alfaVbeta3/metabolismo , Integrina beta3 , Hemorragias Intracraneales/metabolismo , Hemorragias Intracraneales/patología , Masculino , Intercambio Materno-Fetal , Neovascularización Fisiológica , Embarazo , Especies Reactivas de Oxígeno/metabolismo , Trombocitopenia Neonatal Aloinmune/metabolismo , Trombocitopenia Neonatal Aloinmune/patología , Transfección
12.
Curr Opin Hematol ; 23(3): 253-9, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-27055047

RESUMEN

PURPOSE OF REVIEW: The purpose of this article is to describe the function of the vascular cell adhesion and signaling molecule, platelet/endothelial cell adhesion molecule-1 (PECAM-1), in endothelial cells, with special emphasis on its role in maintaining and restoring the vascular permeability barrier following disruption of the endothelial cell junction. RECENT FINDINGS: In addition to its role as an inhibitory receptor in circulating platelets and leukocytes, PECAM-1 is highly expressed at endothelial cell-cell junctions, where it functions as an adhesive stress-response protein to both maintain endothelial cell junctional integrity and speed restoration of the vascular permeability barrier following inflammatory or thrombotic challenge. SUMMARY: Owing to the unique ability of antibodies that bind the membrane proximal region of the extracellular domain to trigger conformational changes leading to affinity modulation and homophilic adhesion strengthening, PECAM-1 might be an attractive target for treating vascular permeability disorders.


Asunto(s)
Células Endoteliales/fisiología , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Humanos
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.
Blood ; 122(15): 2732-42, 2013 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-23926302

RESUMEN

Sitosterolemia is a rare, autosomal recessive disease caused by mutations in the adenosine triphosphate-binding cassette transporter genes ABCG5 or ABCG8 that result in accumulation of xenosterols in the body. Clinical manifestations include tendon xanthomas, premature coronary artery disease, hemolytic anemia, macrothrombocytopenia, and bleeding. Although the effect of sterol accumulation on the predisposition for atherosclerosis is evident, how xenosterol accumulation leads to defects in platelet physiology is unknown. Sitosterolemia induced in Abcg5- and Abcg8-deficient mice fed a high plant sterol diet resulted in accumulation of free sterols in platelet plasma membranes, leading to hyperactivatable platelets characterized by constitutive binding of fibrinogen to its αIIbß3 integrin receptor, internalization of the αIIbß3 complex, generation of platelet-derived microparticles, and changes in the quantity and subcellular localization of filamin. The latter was associated with macrothrombocytopenia, shedding of GPIbα, impaired platelet adhesion to von Willebrand factor, and inability to form stable thrombi. Plasma levels of soluble GPIbα were strongly correlated with plasma sitosterol levels in samples from human sitosterolemic patients, implicating a similar mechanism of sterol-induced platelet passivation in the human disease. Intercalation of plant sterols into the plasma membrane therefore results in dysregulation of multiple platelet activation pathways, leading to macrothrombocytopenia and bleeding.


Asunto(s)
Plaquetas/fisiología , Hemorragia/sangre , Hipercolesterolemia/sangre , Enfermedades Intestinales/sangre , Errores Innatos del Metabolismo Lipídico/sangre , Fitosteroles/efectos adversos , Activación Plaquetaria/fisiología , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 5 , Transportador de Casete de Unión a ATP, Subfamilia G, Miembro 8 , Transportadoras de Casetes de Unión a ATP/genética , Animales , Modelos Animales de Enfermedad , Fibrinógeno/metabolismo , Filaminas/metabolismo , Hemorragia/genética , Humanos , Hipercolesterolemia/genética , Enfermedades Intestinales/genética , Errores Innatos del Metabolismo Lipídico/genética , Lipoproteínas/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo , Fitosteroles/sangre , Fitosteroles/genética , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/genética
15.
Blood ; 121(10): 1858-67, 2013 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-23264598

RESUMEN

The integrin family is composed of a series of 24 αß heterodimer transmembrane adhesion receptors that mediate cell-cell and cell-extracellular matrix interactions. Adaptor molecules bearing immunoreceptor tyrosine-based activation motifs (ITAMs) have recently been shown to cooperate with specific integrins to increase the efficiency of transmitting ligand-binding-induced signals into cells. In human platelets, Fc receptor γ-chain IIa (FcγRIIa) has been identified as an ITAM-bearing transmembrane receptor responsible for mediating "outside-in" signaling through αIIbß3, the major adhesion receptor on the platelet surface. To explore the importance of FcγRIIa in thrombosis and hemostasis, we subjected FcγRIIa-negative and FcγRIIa-positive murine platelets to a number of well-accepted models of platelet function. Compared with their FcγRIIa-negative counterparts, FcγRIIa-positive platelets exhibited increased tyrosine phosphorylation of Syk and phospholipase Cγ2 and increased spreading upon interaction with immobilized fibrinogen, retracted a fibrin clot faster, and showed markedly enhanced thrombus formation when perfused over a collagen-coated flow chamber under conditions of arterial and venous shear. They also displayed increased thrombus formation and fibrin deposition in in vivo models of vascular injury. Taken together, these data establish FcγRIIa as a physiologically important functional conduit for αIIbß3-mediated outside-in signaling, and suggest that modulating the activity of this novel integrin/ITAM pair might be effective in controlling thrombosis.


Asunto(s)
Plaquetas/metabolismo , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/metabolismo , Receptores de IgG/fisiología , Receptores Inmunológicos/metabolismo , Transducción de Señal , Trombosis/etiología , Tirosina/metabolismo , Animales , Arteriolas/metabolismo , Arteriolas/patología , Fibrina/metabolismo , Fibrinógeno/metabolismo , Hemostasis/fisiología , Humanos , Técnicas In Vitro , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Agregación Plaquetaria , Recuento de Plaquetas , Trombosis/metabolismo , Trombosis/patología , Venas/metabolismo , Venas/patología
16.
Blood ; 122(3): 321-7, 2013 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-23645838

RESUMEN

Fetal/neonatal alloimmune thrombocytopenia (FNAIT) is often caused by maternal alloantibodies against the human platelet antigen (HPA)-1a, which opsonizes fetal platelets (PLTs). Subsequent PLT destruction is mediated via the Fc part of the alloantibodies. The monoclonal antibody (mAb) SZ21 binds to the HPA-1a epitope and inhibits the binding of maternal alloantibodies. However, it also promotes complement activation and phagocytosis. Deglycosylation of antibodies abrogates the Fc-related effector functions. We modified the N-glycan of SZ21 by endoglycosidase F. The in vivo transplacental transport of N-glycan-modified (NGM)-SZ21 was not impaired. When injected into pregnant mice, both native-SZ21 and NGM-SZ21 were transported equally into fetal circulation (8.9% vs 8.7%, respectively, P = .58). Neither the binding properties of NGM-SZ21 to HPA-1a in surface plasmon resonance, nor the inhibition of anti-HPA-1a-induced PLT phagocytosis, were affected by N-glycan modification. NGM-SZ21 prevented PLT destruction induced by maternal anti-HPA-1a antibodies in vivo in a mouse model (PLT clearance after 5 hours; 18% vs 62%, in the presence or absence of NGM-SZ21, respectively, P = .013). Deglycosylation of SZ21 abrogates Fc-effector functions without interfering with placental transport or the ability to block anti-HPA-1a binding. Humanized, deglycosylated anti-HPA-1a mAbs may represent a novel treatment strategy to prevent anti-HPA-1a-mediated PLT destruction in FNAIT.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Antígenos de Plaqueta Humana/inmunología , Plaquetas/inmunología , Isoanticuerpos/inmunología , Terapia Molecular Dirigida , Trombocitopenia Neonatal Aloinmune/sangre , Trombocitopenia Neonatal Aloinmune/inmunología , Animales , Anticuerpos Monoclonales/farmacología , Transporte Biológico/efectos de los fármacos , Modelos Animales de Enfermedad , Femenino , Glicosilación/efectos de los fármacos , Humanos , Integrina beta3 , Manosil-Glicoproteína Endo-beta-N-Acetilglucosaminidasa/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones SCID , Fagocitosis/efectos de los fármacos , Fagocitosis/inmunología , Placenta/efectos de los fármacos , Placenta/metabolismo , Polisacáridos/inmunología , Embarazo , Unión Proteica/efectos de los fármacos , Trombocitopenia Neonatal Aloinmune/tratamiento farmacológico
17.
Cell Tissue Res ; 355(3): 607-19, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24435645

RESUMEN

PECAM-1 (also known as CD31) is a cellular adhesion and signaling receptor comprising six extracellular immunoglobulin (Ig)-like homology domains, a short transmembrane domain and a 118 amino acid cytoplasmic domain that becomes serine and tyrosine phosphorylated upon cellular activation. PECAM-1 expression is restricted to blood and vascular cells. In circulating platelets and leukocytes, PECAM-1 functions largely as an inhibitory receptor that, via regulated sequential phosphorylation of its cytoplasmic domain, limits cellular activation responses. PECAM-1 is also highly expressed at endothelial cell intercellular junctions, where it functions as a mechanosensor, as a regulator of leukocyte trafficking and in the maintenance of endothelial cell junctional integrity. In this review, we will describe (1) the functional domains of PECAM-1 and how they contribute to its barrier-enhancing properties, (2) how the physical properties of PECAM-1 influence its subcellular localization and its ability to influence endothelial cell barrier function, (3) various stimuli that initiate PECAM-1 signaling and/or function at the endothelial junction and (4) cross-talk of PECAM-1 with other junctional molecules, which can influence endothelial cell function.


Asunto(s)
Adhesión Celular/fisiología , Células Endoteliales/citología , Células Endoteliales/metabolismo , Uniones Intercelulares/fisiología , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Animales , Humanos , Uniones Intercelulares/metabolismo
18.
Blood ; 119(12): 2906-13, 2012 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-22174152

RESUMEN

Interaction between the cytoplasmic domain of GPIbα with its cytoskeletal binding partner, filamin, is a major determinant of platelet size, and deficiency of either protein results in macrothrombocytopenia. To clarify the mechanism by which GPIbα-filamin interactions regulate platelet production, we manipulated the expression levels of filamin and GPIb in cultured embryonic stem cells (ESCs) that were subsequently differentiated into platelets. Knocking down filamins A and B resulted in the production of ESC-derived proplatelets with abnormally large swellings and proplatelet shafts that generated giant platelets in culture. Large platelets could also be generated by overexpressing GPIbα in ESCs, or by overexpressing in vivo a transgene encoding a chimeric protein containing the cytoplasmic domain of GPIbα. To identify the mechanism by which the GPIb:filamin ratio regulates platelet size, we manipulated filamin and GPIbα levels in HEK293T cells and examined the effects of overexpressing either protein on their ability to traffic to the cell periphery. Accumulation of either protein within the endoplasmic reticulum resulted in trapping of the other. Taken together, these data demonstrate that coordinated expression of GPIbα and filamin is required for efficient trafficking of either protein to the cell surface, and for production of normal-sized platelets.


Asunto(s)
Plaquetas/citología , Plaquetas/metabolismo , Diferenciación Celular/fisiología , Proteínas Contráctiles/metabolismo , Glicoproteínas de Membrana/metabolismo , Proteínas de Microfilamentos/metabolismo , Trombopoyesis/fisiología , Animales , Filaminas , Citometría de Flujo , Humanos , Immunoblotting , Inmunoprecipitación , Ratones , Ratones Transgénicos , Complejo GPIb-IX de Glicoproteína Plaquetaria , Transporte de Proteínas/fisiología
19.
Arterioscler Thromb Vasc Biol ; 33(2): 275-84, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23202369

RESUMEN

OBJECTIVE: The principle role of the vascular endothelium is to present a semi-impermeable barrier to soluble factors and circulating cells, while still permitting the passage of leukocytes from the bloodstream into the tissue. The process of diapedesis involves the selective disruption of endothelial cell junctions, which could compromise vascular integrity. It is therefore somewhat surprising that neutrophil transmigration does not significantly impair endothelial barrier function. We examined whether neutrophils might secrete factors that promote vascular integrity during the latter stages of neutrophil transmigration, in particular, the role of neutrophil serine proteinase 3 (PR3). METHODS AND RESULTS: Endothelial cells were treated with PR3 either in its soluble form or in a complex form with cell surface NB1. We observed that PR3 mediated the enhancement of endothelial cell junctional integrity and that this required its proteolytic activity, as well as endothelial cell expression of the protease-activated receptor-2. Importantly, PR3 suppressed the vascular permeability changes and disruption of junctional proteins induced by the action of protease-activated receptor-1 agonists. CONCLUSIONS: These findings establish the potential for neutrophil-derived PR3 to play a role in reestablishing vascular integrity after leukocyte transmigration and in protecting endothelial cells from protease-activated receptor-1-induced permeability changes that occur during thrombotic and inflammatory events.


Asunto(s)
Permeabilidad Capilar , Células Endoteliales de la Vena Umbilical Humana/enzimología , Mieloblastina/metabolismo , Neutrófilos/enzimología , Receptor PAR-2/metabolismo , Migración Transendotelial y Transepitelial , Señalización del Calcio , Técnicas de Cocultivo , Proteínas Ligadas a GPI/metabolismo , Células HEK293 , Humanos , Isoantígenos/metabolismo , Mieloblastina/genética , Interferencia de ARN , Receptor PAR-1/genética , Receptor PAR-1/metabolismo , Receptores de Superficie Celular/metabolismo , Factores de Tiempo , Transfección , Proteína de Unión al GTP rac1/metabolismo
20.
J Immunol ; 188(5): 2419-26, 2012 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-22266279

RESUMEN

Neutrophil transmigration requires the localization of neutrophils to endothelial cell junctions, in which receptor-ligand interactions and the action of serine proteases promote leukocyte diapedesis. NB1 (CD177) is a neutrophil-expressed surface molecule that has been reported to bind proteinase 3 (PR3), a serine protease released from activated neutrophils. PR3 has demonstrated proteolytic activity on a number of substrates, including extracellular matrix proteins, although its role in neutrophil transmigration is unknown. Recently, NB1 has been shown to be a heterophilic binding partner for the endothelial cell junctional protein, PECAM-1. Disrupting the interaction between NB1 and PECAM-1 significantly inhibits neutrophil transendothelial cell migration on endothelial cell monolayers. Because NB1 interacts with endothelial cell PECAM-1 at cell junctions where transmigration occurs, we considered that NB1-PR3 interactions may play a role in aiding neutrophil diapedesis. Blocking Abs targeting the heterophilic binding domain of PECAM-1 significantly inhibited transmigration of NB1-positive neutrophils through IL-1ß-stimulated endothelial cell monolayers. PR3 expression and activity were significantly increased on NB1-positive neutrophils following transmigration, whereas neutrophils lacking NB1 demonstrated no increase in PR3. Finally, using selective serine protease inhibitors, we determined that PR3 activity facilitated transmigration of NB1-positive neutrophils under both static and flow conditions. These data demonstrate that PR3 contributes in the selective recruitment of the NB1-positive neutrophil population.


Asunto(s)
Isoantígenos/biosíntesis , Mieloblastina/fisiología , Neutrófilos/enzimología , Neutrófilos/inmunología , Receptores de Superficie Celular/biosíntesis , Migración Transendotelial y Transepitelial/inmunología , Activación Enzimática/genética , Activación Enzimática/inmunología , Proteínas Ligadas a GPI/biosíntesis , Proteínas Ligadas a GPI/genética , Proteínas Ligadas a GPI/fisiología , Regulación de la Expresión Génica/inmunología , Células Endoteliales de la Vena Umbilical Humana , Humanos , Isoantígenos/genética , Isoantígenos/fisiología , Mieloblastina/biosíntesis , Mieloblastina/genética , Neutrófilos/citología , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA