Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
1.
Nature ; 555(7698): 678-682, 2018 03 29.
Artículo en Inglés | MEDLINE | ID: mdl-29562234

RESUMEN

Histidine phosphorylation, the so-called hidden phosphoproteome, is a poorly characterized post-translational modification of proteins. Here we describe a role of histidine phosphorylation in tumorigenesis. Proteomic analysis of 12 tumours from an mTOR-driven hepatocellular carcinoma mouse model revealed that NME1 and NME2, the only known mammalian histidine kinases, were upregulated. Conversely, expression of the putative histidine phosphatase LHPP was downregulated specifically in the tumours. We demonstrate that LHPP is indeed a protein histidine phosphatase. Consistent with these observations, global histidine phosphorylation was significantly upregulated in the liver tumours. Sustained, hepatic expression of LHPP in the hepatocellular carcinoma mouse model reduced tumour burden and prevented the loss of liver function. Finally, in patients with hepatocellular carcinoma, low expression of LHPP correlated with increased tumour severity and reduced overall survival. Thus, LHPP is a protein histidine phosphatase and tumour suppressor, suggesting that deregulated histidine phosphorylation is oncogenic.


Asunto(s)
Histidina/metabolismo , Pirofosfatasa Inorgánica/metabolismo , Neoplasias Hepáticas/enzimología , Neoplasias Hepáticas/patología , Proteínas Supresoras de Tumor/metabolismo , Animales , Carcinoma Hepatocelular/enzimología , Carcinoma Hepatocelular/patología , Modelos Animales de Enfermedad , Humanos , Pirofosfatasa Inorgánica/deficiencia , Pirofosfatasa Inorgánica/genética , Masculino , Ratones , Fosforilación , Proteómica , Análisis de Supervivencia , Serina-Treonina Quinasas TOR/metabolismo , Carga Tumoral , Proteínas Supresoras de Tumor/deficiencia , Proteínas Supresoras de Tumor/genética
2.
Mol Cell ; 63(3): 457-69, 2016 08 04.
Artículo en Inglés | MEDLINE | ID: mdl-27453048

RESUMEN

Whereas phosphorylation of serine, threonine, and tyrosine is exceedingly well characterized, the role of histidine phosphorylation in mammalian signaling is largely unexplored. Here we show that phosphoglycerate mutase family 5 (PGAM5) functions as a phosphohistidine phosphatase that specifically associates with and dephosphorylates the catalytic histidine on nucleoside diphosphate kinase B (NDPK-B). By dephosphorylating NDPK-B, PGAM5 negatively regulates CD4(+) T cells by inhibiting NDPK-B-mediated histidine phosphorylation and activation of the K(+) channel KCa3.1, which is required for TCR-stimulated Ca(2+) influx and cytokine production. Using recently developed monoclonal antibodies that specifically recognize phosphorylation of nitrogens at the N1 (1-pHis) or N3 (3-pHis) positions of the imidazole ring, we detect for the first time phosphoisoform-specific regulation of histidine-phosphorylated proteins in vivo, and we link these modifications to TCR signaling. These results represent an important step forward in studying the role of histidine phosphorylation in mammalian biology and disease.


Asunto(s)
Linfocitos T CD4-Positivos/enzimología , Activación de Linfocitos , Proteínas Mitocondriales/metabolismo , Fosfoproteínas Fosfatasas/metabolismo , Animales , Linfocitos T CD4-Positivos/inmunología , Señalización del Calcio , Citocinas/metabolismo , Predisposición Genética a la Enfermedad , Enfermedad Injerto contra Huésped/enzimología , Enfermedad Injerto contra Huésped/genética , Enfermedad Injerto contra Huésped/inmunología , Células HEK293 , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Histidina , Humanos , Mediadores de Inflamación/metabolismo , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/metabolismo , Células Jurkat , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Mitocondriales/genética , Nucleósido-Difosfato Quinasa/metabolismo , Fenotipo , Fosfoproteínas Fosfatasas/deficiencia , Fosfoproteínas Fosfatasas/genética , Fosforilación , Interferencia de ARN , Receptores de Antígenos de Linfocitos T/metabolismo , Factores de Tiempo , Transfección
3.
Proc Natl Acad Sci U S A ; 118(6)2021 02 09.
Artículo en Inglés | MEDLINE | ID: mdl-33547238

RESUMEN

In 2015, monoclonal antibodies (mAbs) that selectively recognize the 1-pHis or 3-pHis isoforms of phosphohistidine were developed by immunizing rabbits with degenerate Ala/Gly peptides containing the nonhydrolyzable phosphohistidine (pHis) analog- phosphotriazolylalanine (pTza). Here, we report structures of five rabbit mAbs bound to cognate pTza peptides: SC1-1 and SC50-3 that recognize 1-pHis, and their 3-pHis-specific counterparts, SC39-4, SC44-8, and SC56-2. These cocrystal structures provide insights into the binding modes of the pTza phosphate group that are distinct for the 1- and 3-pHis mAbs with the selectivity arising from specific contacts with the phosphate group and triazolyl ring. The mode of phosphate recognition in the 3-pHis mAbs recapitulates the Walker A motif, as present in kinases. The complementarity-determining regions (CDRs) of four of the Fabs interact with the peptide backbone rather than peptide side chains, thus conferring sequence independence, whereas SC44-8 shows a proclivity for binding a GpHAGA motif mediated by a sterically complementary CDRL3 loop. Specific hydrogen bonding with the triazolyl ring precludes recognition of pTyr and other phosphoamino acids by these mAbs. Kinetic binding experiments reveal that the affinity of pHis mAbs for pHis and pTza peptides is submicromolar. Bound pHis mAbs also shield the pHis peptides from rapid dephosphorylation. The epitope-paratope interactions illustrate how these anti-pHis antibodies are useful for a wide range of research techniques and this structural information can be utilized to improve the specificity and affinity of these antibodies toward a variety of pHis substrates to understand the role of histidine phosphorylation in healthy and diseased states.


Asunto(s)
Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/inmunología , Histidina/análogos & derivados , Péptidos/química , Péptidos/inmunología , Secuencia de Aminoácidos , Animales , Reacciones Cruzadas/inmunología , Histidina/química , Histidina/inmunología , Fragmentos Fab de Inmunoglobulinas/química , Isomerismo , Cinética , Fosfatos/metabolismo , Conejos , Relación Estructura-Actividad
4.
J Biol Chem ; 286(17): 14830-41, 2011 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-21362625

RESUMEN

Caveolin (Cav) proteins in the plasma membrane have numerous binding partners, but the determinants of these interactions are poorly understood. We show here that Cav-3 has a small ubiquitin-like modifier (SUMO) consensus motif (ΨKX(D/E, where Ψ is a hydrophobic residue)) near the scaffolding domain and that Cav-3 is SUMOylated in a manner that is enhanced by the SUMO E3 ligase PIASy (protein inhibitor of activated STAT-y). Site-directed mutagenesis revealed that the consensus site lysine is the preferred SUMOylation site but that mutation of all lysines is required to abolish SUMOylation. Co-expression of a SUMOylation-deficient mutant of Cav-3 with ß-adrenergic receptors (ßARs) alters the expression level of ß(2)ARs but not ß(1)ARs following agonist stimulation, thus implicating Cav-3 SUMOylation in the mechanisms for ß(2)AR but not ß(1)AR desensitization. Expression of endothelial nitric-oxide synthase (NOS3) was not altered by the SUMOylation-deficient mutant. Thus, SUMOylation is a covalent modification of caveolins that influence the regulation of certain signaling partners.


Asunto(s)
Caveolina 3/metabolismo , Proteínas Inhibidoras de STAT Activados/metabolismo , Sumoilación/fisiología , Línea Celular , Humanos , Lisina , Proteínas de Unión a Poli-ADP-Ribosa , Receptores Adrenérgicos beta 2 , Receptores Acoplados a Proteínas G/metabolismo , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo
5.
J Mol Graph Model ; 89: 234-241, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30921557

RESUMEN

In drug discovery, structural knowledge of a target enables structure-based design approaches and thereby reduces the time and labor required to develop a therapy. Whilst molecular graphics frameworks coupled with computational analysis are now ubiquitous tools for the structural and computational biologist, sharing the detailed visualization and derived structural information with non-expert users still presents a challenge. Here we describe an intuitive virtual world for viewing, manipulating, and modifying chemical and macromolecular structures in a fully immersive and collaborative 3D environment. By reducing the barriers to viewing and interacting with structural data, structural analysis can be democratized to a general scientist, which in turn fosters novel collaboration, ideas, and findings in structural biology and structure-based drug discovery.


Asunto(s)
Descubrimiento de Drogas , Relación Estructura-Actividad Cuantitativa , Realidad Virtual , Sitios de Unión , Biología Computacional , Diseño de Fármacos , Humanos , Ligandos , Unión Proteica , Proteína Serina-Treonina Quinasa 2 de Interacción con Receptor/antagonistas & inhibidores , Proteína Serina-Treonina Quinasa 2 de Interacción con Receptor/química , Programas Informáticos , Interfaz Usuario-Computador
6.
Elife ; 52016 08 19.
Artículo en Inglés | MEDLINE | ID: mdl-27542194

RESUMEN

KCa2.1, KCa2.2, KCa2.3 and KCa3.1 constitute a family of mammalian small- to intermediate-conductance potassium channels that are activated by calcium-calmodulin. KCa3.1 is unique among these four channels in that activation requires, in addition to calcium, phosphorylation of a single histidine residue (His358) in the cytoplasmic region, by nucleoside diphosphate kinase-B (NDPK-B). The mechanism by which KCa3.1 is activated by histidine phosphorylation is unknown. Histidine phosphorylation is well characterized in prokaryotes but poorly understood in eukaryotes. Here, we demonstrate that phosphorylation of His358 activates KCa3.1 by antagonizing copper-mediated inhibition of the channel. Furthermore, we show that activated CD4(+) T cells deficient in intracellular copper exhibit increased KCa3.1 histidine phosphorylation and channel activity, leading to increased calcium flux and cytokine production. These findings reveal a novel regulatory mechanism for a mammalian potassium channel and for T-cell activation, and highlight a unique feature of histidine versus serine/threonine and tyrosine as a regulatory phosphorylation site.


Asunto(s)
Cobre/metabolismo , Inhibidores Enzimáticos/metabolismo , Histidina/metabolismo , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/antagonistas & inhibidores , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/metabolismo , Animales , Linfocitos T CD4-Positivos/inmunología , Células Cultivadas , Citocinas/metabolismo , Humanos , Ratones , Nucleósido-Difosfato Quinasa/metabolismo , Técnicas de Placa-Clamp , Fosforilación
7.
Mol Pharmacol ; 71(2): 508-18, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16968809

RESUMEN

We have developed a new assay for measuring epidermal growth factor receptor (EGFR) activation using the bioluminescence resonance energy transfer (BRET) technology, which directly measures the recruitment of signaling proteins to activated EGFR. Our results demonstrate that EGFR BRET assays precisely measure the pharmacology and signaling properties of EGFR expressed in human embryonic kidney 293T cells. EGFR BRET assays are highly sensitive to known EGFR ligands [pEC50 of epidermal growth factor (EGF)=10.1+/-0.09], consistent with previous pharmacological methods for measuring EGFR activation. We applied EGFR BRET assays to study the characteristics of somatic EGFR mutations that were recently identified in lung cancer. In agreement with recent reports, we detected constitutively active mutant EGFR isoforms, which predominantly signal through the phosphatidylinositol-3-kinase/Akt pathway. The EGFR inhibitors Iressa or Tarceva are severalfold more potent in inhibiting constitutive activity of mutant EGFR isoforms compared with wild-type EGFR. Notable, our results reveal that most of the mutant EGFR isoforms tested were significantly impaired in their response to EGF. The highest level of constitutive activity and nearly complete loss of epidermal growth factor responsiveness was detected in isoforms that carry the activating mutation L858R and the secondary resistance mutation T790M. In summary, our study reveals that somatic mutations in EGFR quantitatively differ in pharmacology and signaling properties, which suggest the possibility of differential clinical responsiveness to treatment with EGFR inhibitors. Furthermore, we demonstrate that the EGFR BRET assays are a useful tool to study the pharmacology of ligand-induced interaction between EGFR and signaling pathway-specifying adapter proteins.


Asunto(s)
Receptores ErbB/metabolismo , Mediciones Luminiscentes/métodos , Transducción de Señal , Línea Celular , Resistencia a Medicamentos/genética , Receptores ErbB/análisis , Receptores ErbB/genética , Transferencia Resonante de Energía de Fluorescencia , Humanos , Proteínas Luminiscentes , Neoplasias Pulmonares/genética , Mutación , Fosfatidilinositol 3-Quinasas/metabolismo , Isoformas de Proteínas , Proteínas Proto-Oncogénicas c-akt/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA