Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 3 de 3
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Pediatr Transplant ; 25(2): e13885, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33131194

RESUMEN

Donor-specific antibody (DSA) is an independent risk factor for antibody-mediated rejection (ABMR) and graft loss. The C1q assay differentiates complement from non-complement-binding DSA and C1q-binding DSA may lead to poor allograft survival. Our aim was to characterize the type of DSA seen in pediatric kidney transplant recipients and to determine whether complement binding DSA was associated with inferior graft survival.This was a single-center retrospective study of 48 children who were transplanted between 2009 and 2016. DSA were monitored using Luminex single antigen beads. A negative crossmatch was required to proceed with transplantation. The median follow-up time was 4.9 (3.4, 7.9) years. The median age was 12 (5.7, 15.4) years. DSA developed in 27/48 (56.3%), while C1q-binding DSA developed in 17/27 (63%). There were no significant differences between DSA negative, C1q-binding DSA, and C1q negative DSA, with regard to the number of HLA-ABDR (P  =  .09) or HLA-DQ mismatches alone (P  =  .16). For both C1q negative and C1q-binding DSA, DQ was the most common target of the DSA (19/27; 70.4%). C1q-binding DSA was associated with a significantly higher frequency of biopsy proven rejection (76.5%) when compared to C1q negative (10%) and DSA negative (14.3%); P  =  .001. Graft loss was seen in 6 (12.5%), all of whom had C1q-binding DSA (P  =  .004). C1q-binding DSA was most commonly directed to DQ antigens. C1q-binding DSA was associated with increased rejection and graft loss. Monitoring for C1q-binding DSA may risk stratify recipients and guide physician management.


Asunto(s)
Aloinjertos/inmunología , Complemento C1q/inmunología , Rechazo de Injerto/inmunología , Supervivencia de Injerto/inmunología , Antígenos HLA/inmunología , Isoanticuerpos/inmunología , Trasplante de Riñón , Adolescente , Biomarcadores/sangre , Niño , Preescolar , Femenino , Estudios de Seguimiento , Rechazo de Injerto/sangre , Rechazo de Injerto/diagnóstico , Antígenos HLA/sangre , Humanos , Isoanticuerpos/sangre , Estimación de Kaplan-Meier , Modelos Logísticos , Masculino , Evaluación de Resultado en la Atención de Salud , Estudios Retrospectivos , Medición de Riesgo , Factores de Riesgo , Donantes de Tejidos
2.
RNA Biol ; 15(2): 251-260, 2018 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-29168431

RESUMEN

Nucleolin (NCL) is an abundant stress-responsive, RNA-binding phosphoprotein that controls gene expression by regulating either mRNA stability and/or translation. NCL binds to the AU-rich element (ARE) in the 3'UTR of target mRNAs, mediates miRNA functions in the nearby target sequences, and regulates mRNA deadenylation. However, the mechanism by which NCL phosphorylation affects these functions and the identity of the deadenylase involved, remain largely unexplored. Earlier we demonstrated that NCL phosphorylation is vital for cell cycle progression and proliferation, whereas phosphorylation-deficient NCL at six consensus CK2 sites confers dominant-negative effect on proliferation by increasing p53 expression, possibly mimicking cellular DNA damage conditions. In this study, we show that NCL phosphorylation at those CK2 consensus sites in the N-terminus is necessary to induce deadenylation upon oncogenic stimuli and UV stress. NCL-WT, but not hypophosphorylated NCL-6/S*A, activates poly (A)-specific ribonuclease (PARN) deadenylase activity. We further demonstrate that NCL interacts directly with PARN, and under non-stress conditions also forms (a) complex (es) with factors that regulate deadenylation, such as p53 and the ARE-binding protein HuR. Upon UV stress, the interaction of hypophosphorylated NCL-6/S*A with these proteins is favored. As an RNA-binding protein, NCL interacts with PARN deadenylase substrates such as TP53 and BCL2 mRNAs, playing a role in their downregulation under non-stress conditions. For the first time, we show that NCL phosphorylation offers specificity to its protein-protein, protein-RNA interactions, resulting in the PARN deadenylase regulation, and hence gene expression, during cellular stress responses.


Asunto(s)
Quinasa de la Caseína II/metabolismo , Activación Enzimática , Exorribonucleasas/metabolismo , Fosfoproteínas/metabolismo , Proteínas de Unión al ARN/metabolismo , Línea Celular , Regulación de la Expresión Génica/efectos de la radiación , Humanos , Fosfoproteínas/química , Fosforilación , Proteínas Proto-Oncogénicas c-bcl-2/genética , Proteínas de Unión al ARN/química , Estrés Fisiológico , Proteína p53 Supresora de Tumor/genética , Rayos Ultravioleta/efectos adversos , Nucleolina
3.
PLoS One ; 9(10): e109858, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25313645

RESUMEN

Nucleolin (NCL) is a major nucleolar phosphoprotein that has pleiotropic effects on cell proliferation and is elevated in a variety of tumors. NCL is highly phosphorylated at the N-terminus by two major kinases: interphase casein kinase 2 (CK2) and mitotic cyclin-dependent kinase 1 (CDK1). Earlier we demonstrated that a NCL-mutant that is partly defective in undergoing phosphorylation by CK2 inhibits chromosomal replication through its interactions with Replication Protein A, mimicking the cellular response to DNA damage. We further delineated that the N-terminus of NCL associates with Hdm2, the most common E3 ubiquitin ligase of p53. We reported that NCL antagonizes Hdm2 to stabilize p53 and stimulates p53 transcriptional activity. Although NCL-phosphorylation by CK2 and ribosomal DNA transcription are closely coordinated during interphase, the role of NCL phosphorylation in regulating cell proliferation remains unexplored. We have therefore engineered unique human cells that specifically induce expression of NCL-wild type (WT) or a phosphorylation-deficient NCL-mutant, 6/S*A where all the six CK2 consensus serine sites residing in the N-terminus NCL were mutated to alanine. Here we show that this NCL-mutant is defective in undergoing phosphorylation by CK2. We also demonstrate that NCL-phosphorylation by CK2 is required through the S-phase progression in cell cycle and hence proliferation. Induced expression of NCL with mutated CK2 phosphorylation sites stabilizes p53, results in higher expression of Bcl2 (B-cell lymphoma 2) homology 3 (BH3)-only apoptotic markers and causes a dominant-negative effect on cell viability. Our unique cellular system thus provides the first evidential support to delineate phospho-specific functions of NCL on cell proliferation.


Asunto(s)
Proliferación Celular , Fosfoproteínas/genética , Proteínas de Unión al ARN/genética , Sustitución de Aminoácidos , Apoptosis , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/metabolismo , Quinasa de la Caseína II/metabolismo , Línea Celular Tumoral , Expresión Génica , Genes Dominantes , Semivida , Humanos , Fosfoproteínas/metabolismo , Fosforilación , Procesamiento Proteico-Postraduccional , Estabilidad Proteica , Proteínas de Unión al ARN/metabolismo , Activación Transcripcional , Proteína p53 Supresora de Tumor/metabolismo , Nucleolina
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...