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

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
EMBO Rep ; 23(7): e54352, 2022 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-35603428

RESUMEN

Glycosylphosphatidylinositols (GPIs) are glycolipids that anchor many proteins (GPI-APs) on the cell surface. The core glycan of GPI precursor has three mannoses, which in mammals, are all modified by ethanolamine-phosphate (EthN-P). It is postulated that EthN-P on the third mannose (EthN-P-Man3) is the bridge between GPI and the protein and the second (EthN-P-Man2) is removed after GPI-protein attachment. However, EthN-P-Man2 may not be always transient, as mutations of PIGG, the enzyme that transfers EthN-P to Man2, result in inherited GPI deficiencies (IGDs), characterized by neuronal dysfunctions. Here, we show that EthN-P on Man2 is the preferential bridge in some GPI-APs, among them, the Ect-5'-nucleotidase and Netrin G2. We find that CD59, a GPI-AP, is attached via EthN-P-Man2 both in PIGB-knockout cells, in which GPI lacks Man3, and with a small fraction in wild-type cells. Our findings modify the current view of GPI anchoring and provide a mechanistic basis for IGDs caused by PIGG mutations.


Asunto(s)
Glicosilfosfatidilinositoles , Manosa , Animales , Etanolaminas/metabolismo , Proteínas Ligadas a GPI/genética , Glicosilfosfatidilinositoles/genética , Glicosilfosfatidilinositoles/metabolismo , Mamíferos/metabolismo , Manosa/metabolismo , Fosfatos
2.
J Biol Chem ; 296: 100399, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33571526

RESUMEN

CD40L is a member of the TNF superfamily that participates in immune cell activation. It binds to and signals through several integrins, including αvß3 and α5ß1, which bind to the trimeric interface of CD40L. We previously showed that several integrin ligands can bind to the allosteric site (site 2), which is distinct from the classical ligand-binding site (site 1), raising the question of if CD40L activates integrins. In our explorations of this question, we determined that integrin α4ß1, which is prevalently expressed on the same CD4+ T cells as CD40L, is another receptor for CD40L. Soluble (s)CD40L activated soluble integrins αvß3, α5ß1, and α4ß1 in cell-free conditions, indicating that this activation does not require inside-out signaling. Moreover, sCD40L activated cell-surface integrins in CHO cells that do not express CD40. To learn more about the mechanism of binding, we determined that sCD40L bound to a cyclic peptide from site 2. Docking simulations predicted that the residues of CD40L that bind to site 2 are located outside of the CD40L trimer interface, at a site where four HIGM1 (hyper-IgM syndrome type 1) mutations are clustered. We tested the effect of these mutations, finding that the K143T and G144E mutants were the most defective in integrin activation, providing support that this region interacts with site 2. We propose that allosteric integrin activation by CD40L also plays a role in CD40L signaling, and defective site 2 binding may be related to the impaired CD40L signaling functions of these HIGM1 mutants.


Asunto(s)
Ligando de CD40/metabolismo , Integrina alfa4beta1/metabolismo , Integrina alfa5beta1/metabolismo , Integrina alfaVbeta3/metabolismo , Receptores de Superficie Celular/química , Linfocitos T/metabolismo , Sitio Alostérico , Animales , Ligando de CD40/inmunología , Línea Celular , Cricetinae , Humanos , Integrina alfa4beta1/inmunología , Integrina alfa5beta1/inmunología , Integrina alfaVbeta3/inmunología , Simulación del Acoplamiento Molecular , Unión Proteica , Receptores de Superficie Celular/metabolismo , Transducción de Señal , Linfocitos T/inmunología
3.
J Biol Chem ; 295(22): 7789-7798, 2020 05 29.
Artículo en Inglés | MEDLINE | ID: mdl-32321762

RESUMEN

Prion diseases are transmissible, lethal neurodegenerative disorders caused by accumulation of the aggregated scrapie form of the prion protein (PrPSc) after conversion of the cellular prion protein (PrPC). The glycosylphosphatidylinositol (GPI) anchor of PrPC is involved in prion disease pathogenesis, and especially sialic acid in a GPI side chain reportedly affects PrPC conversion. Thus, it is important to define the location and structure of the GPI anchor in human PrPC Moreover, the sialic acid linkage type in the GPI side chain has not been determined for any GPI-anchored protein. Here we report GPI glycan structures of human PrPC isolated from human brains and from brains of a knock-in mouse model in which the mouse prion protein (Prnp) gene was replaced with the human PRNP gene. LC-electrospray ionization-MS analysis of human PrPC from both biological sources indicated that Gly229 is the ω site in PrPC to which GPI is attached. Gly229 in human PrPC does not correspond to Ser231, the previously reported ω site of Syrian hamster PrPC We found that ∼41% and 28% of GPI anchors in human PrPCs from human and knock-in mouse brains, respectively, have N-acetylneuraminic acid in the side chain. Using a sialic acid linkage-specific alkylamidation method to discriminate α2,3 linkage from α2,6 linkage, we found that N-acetylneuraminic acid in PrPC's GPI side chain is linked to galactose through an α2,3 linkage. In summary, we report the GPI glycan structure of human PrPC, including the ω-site amino acid for GPI attachment and the sialic acid linkage type.


Asunto(s)
Glicosilfosfatidilinositoles , Ácido N-Acetilneuramínico , Proteínas PrPC , Proteínas Priónicas , Animales , Conformación de Carbohidratos , Glicosilfosfatidilinositoles/química , Glicosilfosfatidilinositoles/genética , Glicosilfosfatidilinositoles/metabolismo , Humanos , Masculino , Mesocricetus , Ratones , Ratones Noqueados , Ácido N-Acetilneuramínico/química , Ácido N-Acetilneuramínico/genética , Ácido N-Acetilneuramínico/metabolismo , Proteínas PrPC/química , Proteínas PrPC/genética , Proteínas PrPC/metabolismo , Enfermedades por Prión/genética , Enfermedades por Prión/metabolismo , Enfermedades por Prión/patología , Proteínas Priónicas/química , Proteínas Priónicas/genética , Proteínas Priónicas/metabolismo
4.
J Immunol ; 203(5): 1383-1391, 2019 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-31331973

RESUMEN

CD40L plays a major role in immune response and is a major therapeutic target for inflammation. Integrin α5ß1 and CD40 simultaneously bind to CD40L. It is unclear if α5ß1 and CD40 work together in CD40/CD40L signaling or how α5ß1 binds to CD40L. In this article, we describe that the integrin-binding site of human CD40L is predicted to be located in the trimeric interface by docking simulation. Mutations in the predicted integrin-binding site markedly reduced the binding of α5ß1 to CD40L. Several CD40L mutants defective in integrin binding were defective in NF-κB activation and B cell activation and suppressed CD40L signaling induced by wild-type CD40L; however, they still bound to CD40. These findings suggest that integrin α5ß1 binds to monomeric CD40L through the binding site in the trimeric interface of CD40L, and this plays a critical role in CD40/CD40L signaling. Integrin αvß3, a widely distributed vascular integrin, bound to CD40L in a KGD-independent manner, suggesting that αvß3 is a new CD40L receptor. Several missense mutations in CD40L that induce immunodeficiency with hyper-IgM syndrome type 1 (HIGM1) are clustered in the integrin-binding site of the trimeric interface. These HIGM1 CD40L mutants were defective in binding to α5ß1 and αvß3 (but not to CD40), suggesting that the defect in integrin binding may be a causal factor of HIGM1. These findings suggest that α5ß1 and αvß3 bind to the overlapping binding site in the trimeric interface of monomeric CD40L and generate integrin-CD40L-CD40 ternary complex. CD40L mutants defective in integrins have potential as antagonists of CD40/CD40L signaling.


Asunto(s)
Antígenos CD40/metabolismo , Ligando de CD40/metabolismo , Integrina alfa5beta1/metabolismo , Integrina alfaVbeta3/metabolismo , Transducción de Señal/fisiología , Animales , Sitios de Unión/fisiología , Células CHO , Línea Celular , Línea Celular Tumoral , Cricetulus , Células HEK293 , Humanos , Síndrome de Inmunodeficiencia con Hiper-IgM Tipo 1/metabolismo , Células K562 , Mutación/fisiología , Unión Proteica/fisiología
5.
FASEB J ; 33(8): 9131-9141, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31116572

RESUMEN

Proper control of cell migration is critically important in many biologic processes, such as wound healing, immune surveillance, and development. Much progress has been made in the initiation of cell migration; however, little is known about termination and sometimes directional reversal. During active cell migration, as in wound healing, development, and immune surveillance, the integrin expression profile undergoes drastic changes. Here, we uncovered the extensive regulatory and even opposing roles of integrins in directional cell migration in electric fields (EFs), a potentially important endogenous guidance mechanism. We established cell lines that stably express specific integrins and determined their responses to applied EFs with a high throughput screen. Expression of specific integrins drove cells to migrate to the cathode or to the anode or to lose migration direction. Cells expressing αMß2, ß1, α2, αIIbß3, and α5 migrated to the cathode, whereas cells expressing ß3, α6, and α9 migrated to the anode. Cells expressing α4, αV, and α6ß4 lost directional electrotaxis. Manipulation of α9 molecules, one of the molecular directional switches, suggested that the intracellular domain is critical for the directional reversal. These data revealed an unreported role for integrins in controlling stop, go, and reversal activity of directional migration of mammalian cells in EFs, which might ensure that cells reach their final destination with well-controlled speed and direction.-Zhu, K., Takada, Y., Nakajima, K., Sun, Y., Jiang, J., Zhang, Y., Zeng, Q., Takada, Y., Zhao, M. Expression of integrins to control migration direction of electrotaxis.


Asunto(s)
Movimiento Celular/fisiología , Integrinas/fisiología , Animales , Células CHO , Movimiento Celular/genética , Cricetulus , Electricidad , Colorantes Fluorescentes , Humanos , Cadenas alfa de Integrinas/genética , Cadenas alfa de Integrinas/fisiología , Integrinas/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Taxia/fisiología , Imagen de Lapso de Tiempo , Transcriptoma
6.
Biochem J ; 475(4): 723-732, 2018 02 16.
Artículo en Inglés | MEDLINE | ID: mdl-29301984

RESUMEN

Leukocyte arrest on the endothelial cell surface during leukocyte extravasation is induced by rapid integrin activation by chemokines. We recently reported that fractalkine induces integrin activation without its receptor CX3CR1 through binding to the allosteric site (site 2) of integrins. Peptides from site 2 bound to fractalkine and suppressed integrin activation by fractalkine. We hypothesized that this is not limited to membrane-bound fractalkine. We studied whether stromal cell-derived factor-1 (SDF1), another chemokine that plays a critical role in leukocyte arrest, activates integrins through binding to site 2. We describe here that (1) SDF1 activated soluble integrin αvß3 in cell-free conditions, suggesting that SDF1 can activate αvß3 without CXCR4; (2) site 2 peptide bound to SDF1, suggesting that SDF1 binds to site 2; (3) SDF1 activated integrins αvß3, α4ß1, and α5ß1 on CHO cells (CXCR4-negative) and site 2 peptide suppressed the activation; (4) A CXCR4 antagonist AMD3100 did not affect the site 2-mediated integrin activation by SDF1; (5) Cell-surface integrins were fully activated in 1 min (much faster than activation of soluble αvß3) and the activation lasted at least for 1 h. We propose that the binding of SDF1 to cell-surface proteoglycan facilitates the allosteric activation process; (6) Mutations in the predicted site 2-binding site in SDF1 suppressed integrin activation. These results suggest that SDF1 (e.g. presented on proteoglycans) can rapidly activate integrins in an allosteric manner by binding to site 2 in the absence of CXCR4. The allosteric integrin activation by SDF1 is a novel target for drug discovery.


Asunto(s)
Quimiocina CXCL12/química , Integrinas/química , Receptores CXCR4/química , Sitio Alostérico , Animales , Sitios de Unión , Células CHO , Sistema Libre de Células , Quimiocina CX3CL1/química , Quimiocina CX3CL1/genética , Quimiocina CXCL12/genética , Cricetulus , Humanos , Integrinas/genética , Simulación del Acoplamiento Molecular , Mutación , Unión Proteica , Receptores CXCR4/genética , Transducción de Señal/genética
7.
J Biol Chem ; 292(49): 20067-20075, 2017 12 08.
Artículo en Inglés | MEDLINE | ID: mdl-29030430

RESUMEN

There is a strong link between integrins and interleukin-1ß (IL-1ß), but the specifics of the role of integrins in IL-1ß signaling are unclear. We describe that IL-1ß specifically bound to integrins αvß3 and α5ß1. The E128K mutation in the IL1R-binding site enhanced integrin binding. We studied whether direct integrin binding is involved in IL-1ß signaling. We compared sequences of IL-1ß and IL-1 receptor antagonist (IL1RN), which is an IL-1ß homologue but has no agonistic activity. Several surface-exposed Lys residues are present in IL-1ß, but not in IL1RN. A disulfide linkage is present in IL1RN, but is not in IL-1ß because of natural C117F mutation. Substitution of the Lys residues to Glu markedly reduced integrin binding of E128K IL-1ß, suggesting that the Lys residues mediate integrin binding. The Lys mutations reduced, but did not completely abrogate, agonistic action of IL-1ß. We studied whether the disulfide linkage plays a role in agonistic action of IL-1ß. Reintroduction of the disulfide linkage by the F117C mutation did not affect agonistic activity of WT IL-1ß, but effectively reduced the remaining agonistic activity of the Lys mutants. Also, deletion of the disulfide linkage in IL1RN by the C116F mutation did not make it agonistic. We propose that the direct binding to IL-1ß to integrins is primarily important for agonistic IL-1ß signaling, and that the disulfide linkage indirectly affects signaling by blocking conformational changes induced by weak integrin binding to the Lys mutants. The integrin-IL-1ß interaction is a potential target for drug discovery.


Asunto(s)
Integrina alfa5beta1/metabolismo , Integrina alfaVbeta3/metabolismo , Integrinas/metabolismo , Interleucina-1beta/metabolismo , Animales , Células CHO , Cricetulus , Disulfuros/farmacología , Humanos , Interleucina-1beta/genética , Células MCF-7 , Mutación , Unión Proteica , Transducción de Señal
8.
Biochem J ; 474(4): 589-596, 2017 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-27993971

RESUMEN

Tetraspanins play important roles in normal (e.g. cell adhesion, motility, activation, and proliferation) and pathological conditions (e.g. metastasis and viral infection). Tetraspanins interact with integrins and regulate integrin functions, but the specifics of tetraspanin-integrin interactions are unclear. Using co-immunoprecipitation with integrins as a sole method to detect interaction between integrins and full-length tetraspanins, it has been proposed that the variable region (helices D and E) of the extracellular-2 (EC2) domain of tetraspanins laterally associates with a non-ligand-binding site of integrins. We describe that, using adhesion assays, the EC2 domain of CD81, CD9, and CD151 bound to integrin αvß3, and this binding was suppressed by cRGDfV, a specific inhibitor of αvß3, and antibody 7E3, which is mapped to the ligand-binding site of ß3. We also present evidence that the specificity loop of ß3 directly bound to the EC2 domains. This suggests that the EC2 domains specifically bind to the classical ligand-binding site of αvß3. αvß3 was a more effective receptor for the EC2 domains than the previously known tetraspanin receptors α3ß1, α4ß1, and α6ß1. Docking simulation predicted that the helices A and B of CD81 EC2 bind to the RGD-binding site of αvß3. Substituting Lys residues at positions 116 and 144/148 of CD81 EC2 in the predicted integrin-binding interface reduced the binding of CD81 EC2 to αvß3, consistent with the docking model. These findings suggest that, in contrast with previous models, the ligand-binding site of integrin αvß3, a new tetraspanin receptor, binds to the constant region (helices A and B) of the EC2 domain.


Asunto(s)
Integrina alfaVbeta3/química , Tetraspanina 24/química , Tetraspanina 28/química , Tetraspanina 29/química , Secuencia de Aminoácidos , Animales , Anticuerpos Monoclonales/química , Sitios de Unión , Células CHO , Clonación Molecular , Cricetulus , Escherichia coli/genética , Escherichia coli/metabolismo , Expresión Génica , Integrina alfaVbeta3/genética , Integrina alfaVbeta3/inmunología , Cinética , Simulación del Acoplamiento Molecular , Unión Proteica , Conformación Proteica en Hélice alfa , Conformación Proteica en Lámina beta , Dominios y Motivos de Interacción de Proteínas , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Tetraspanina 24/genética , Tetraspanina 24/inmunología , Tetraspanina 28/genética , Tetraspanina 28/inmunología , Tetraspanina 29/genética , Tetraspanina 29/inmunología
9.
J Biol Chem ; 290(1): 259-71, 2015 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-25398877

RESUMEN

Integrins are activated by signaling from inside the cell (inside-out signaling) through global conformational changes of integrins. We recently discovered that fractalkine activates integrins in the absence of CX3CR1 through the direct binding of fractalkine to a ligand-binding site in the integrin headpiece (site 2) that is distinct from the classical RGD-binding site (site 1). We propose that fractalkine binding to the newly identified site 2 induces activation of site 1 though conformational changes (in an allosteric mechanism). We reasoned that site 2-mediated activation of integrins is not limited to fractalkine. Human secreted phospholipase A2 type IIA (sPLA2-IIA), a proinflammatory protein, binds to integrins αvß3 and α4ß1 (site 1), and this interaction initiates a signaling pathway that leads to cell proliferation and inflammation. Human sPLA2-IIA does not bind to M-type receptor very well. Here we describe that sPLA2-IIA directly activated purified soluble integrin αvß3 and transmembrane αvß3 on the cell surface. This activation did not require catalytic activity or M-type receptor. Docking simulation predicted that sPLA2-IIA binds to site 2 in the closed-headpiece of αvß3. A peptide from site 2 of integrin ß1 specifically bound to sPLA2-IIA and suppressed sPLA2-IIA-induced integrin activation. This suggests that sPLA2-IIA activates αvß3 through binding to site 2. sPLA2-IIA also activated integrins α4ß1 and α5ß1 in a site 2-mediated manner. We recently identified small compounds that bind to sPLA2-IIA and suppress integrin-sPLA2-IIA interaction (e.g. compound 21 (Cmpd21)). Cmpd21 effectively suppressed sPLA2-IIA-induced integrin activation. These results define a novel mechanism of proinflammatory action of sPLA2-IIA through integrin activation.


Asunto(s)
Fosfolipasas A2 Grupo II/química , Integrina alfa4beta1/química , Integrina alfaVbeta3/química , Receptores de Vitronectina/química , Secuencia de Aminoácidos , Animales , Sitios de Unión , Células CHO , Cricetulus , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Escherichia coli/genética , Escherichia coli/metabolismo , Regulación de la Expresión Génica , Fosfolipasas A2 Grupo II/antagonistas & inhibidores , Fosfolipasas A2 Grupo II/genética , Fosfolipasas A2 Grupo II/metabolismo , Humanos , Inflamación/genética , Inflamación/metabolismo , Inflamación/patología , Integrina alfa4beta1/genética , Integrina alfa4beta1/metabolismo , Integrina alfaVbeta3/genética , Integrina alfaVbeta3/metabolismo , Células K562 , Modelos Moleculares , Simulación del Acoplamiento Molecular , Datos de Secuencia Molecular , Péptidos/síntesis química , Péptidos/química , Unión Proteica , Receptores de Vitronectina/genética , Receptores de Vitronectina/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Alineación de Secuencia , Transducción de Señal
10.
J Biol Chem ; 288(5): 3059-69, 2013 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-23243309

RESUMEN

Integrin αvß3 plays a role in insulin-like growth factor 1 (IGF1) signaling (integrin-IGF1 receptor (IGF1R) cross-talk) in non-transformed cells in anchorage-dependent conditions. We reported previously that IGF1 directly binds to αvß3 and induces αvß3-IGF1-IGF1R ternary complex formation in these conditions. The integrin-binding defective IGF1 mutant (R36E/R37E) is defective in inducing ternary complex formation and IGF signaling, whereas it still binds to IGF1R. We studied if IGF1 can induce signaling in anchorage-independent conditions in transformed Chinese hamster ovary cells that express αvß3 (ß3-CHO) cells. Here we describe that IGF1 signals were more clearly detectable in anchorage-independent conditions (polyHEMA-coated plates) than in anchorage-dependent conditions. This suggests that IGF signaling is masked by signals from cell-matrix interaction in anchorage-dependent conditions. IGF signaling required αvß3 expression, and R36E/R37E was defective in inducing signals in polyHEMA-coated plates. These results suggest that αvß3-IGF1 interaction, not αvß3-extracellular matrix interaction, is essential for IGF signaling. Inhibitors of IGF1R, Src, AKT, and ERK1/2 did not suppress αvß3-IGF-IGF1R ternary complex formation, suggesting that activation of these kinases are not required for ternary complex formation. Also, mutations of the ß3 cytoplasmic tail (Y747F and Y759F) that block ß3 tyrosine phosphorylation did not affect IGF1R phosphorylation or AKT activation. We propose a model in which IGF1 binding to IGF1R induces recruitment of integrin αvß3 to the IGF-IGF1R complex and then ß3 and IGF1R are phosphorylated. It is likely that αvß3 should be together with the IGF1-IGF1R complex for triggering IGF signaling.


Asunto(s)
Factor I del Crecimiento Similar a la Insulina/metabolismo , Integrina alfaVbeta3/metabolismo , Complejos Multiproteicos/metabolismo , Receptor IGF Tipo 1/metabolismo , Transducción de Señal , Familia-src Quinasas/metabolismo , Animales , Células CHO , Adhesión Celular , Inhibición de Contacto , Cricetinae , Activación Enzimática , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Humanos , Integrina alfaVbeta3/química , Modelos Biológicos , Mutación/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , Unión Proteica , Estructura Terciaria de Proteína , Familia-src Quinasas/antagonistas & inhibidores
11.
J Biol Chem ; 288(27): 19593-603, 2013 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-23696648

RESUMEN

Insulin-like growth factor-1 (IGF1) is a major therapeutic target for cancer. We recently reported that IGF1 directly binds to integrins (αvß3 and α6ß4) and induces ternary complex formation (integrin-IGF1-IGF1 receptor (IGF1R)) and that the integrin binding-defective mutant of IGF1 (R36E/R37E) is defective in signaling and ternary complex formation. These findings predict that R36E/R37E competes with WT IGF1 for binding to IGF1R and inhibits IGF signaling. Here, we described that excess R36E/R37E suppressed cell viability increased by WT IGF1 in vitro in non-transformed cells. We studied the effect of R36E/R37E on viability and tumorigenesis in cancer cell lines. We did not detect an effect of WT IGF1 or R36E/R37E in cancer cells under anchorage-dependent conditions. However, under anchorage-independent conditions, WT IGF1 enhanced cell viability and induced signals, whereas R36E/R37E did not. Notably, excess R36E/R37E suppressed cell viability and signaling induced by WT IGF1 under anchorage-independent conditions. Using cancer cells stably expressing WT IGF1 or R36E/R37E, we determined that R36E/R37E suppressed tumorigenesis in vivo, whereas WT IGF1 markedly enhanced it. R36E/R37E suppressed the binding of WT IGF1 to the cell surface and the subsequent ternary complex formation induced by WT IGF1. R36E/R37E suppressed activation of IGF1R by insulin. WT IGF1, but not R36E/R37E, induced ternary complex formation with the IGF1R/insulin receptor hybrid. These findings suggest that 1) IGF1 induces signals under anchorage-independent conditions and that 2) R36E/R37E acts as a dominant-negative inhibitor of IGF1R (IGF1 decoy). Our results are consistent with a model in which ternary complex formation is critical for IGF signaling.


Asunto(s)
Sustitución de Aminoácidos , Transformación Celular Neoplásica/efectos de los fármacos , Factor I del Crecimiento Similar a la Insulina/farmacología , Mutación Missense , Receptor IGF Tipo 1/metabolismo , Transducción de Señal/efectos de los fármacos , Animales , Línea Celular Tumoral , Supervivencia Celular , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Humanos , Insulina/genética , Insulina/metabolismo , Factor I del Crecimiento Similar a la Insulina/genética , Factor I del Crecimiento Similar a la Insulina/metabolismo , Integrinas , Ratones , Modelos Biológicos , Células 3T3 NIH , Unión Proteica , Estructura Cuaternaria de Proteína , Receptor IGF Tipo 1/genética , Transducción de Señal/genética
12.
J Immunol ; 189(12): 5809-19, 2012 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-23125415

RESUMEN

The membrane-bound chemokine fractalkine (FKN, CX3CL1) on endothelial cells plays a role in leukocyte trafficking. The chemokine domain (FKN-CD) is sufficient for inducing FKN signaling (e.g., integrin activation), and FKN-CD binds to its receptor CX3CR1 on leukocytes. Whereas previous studies suggest that FKN-CD does not directly bind to integrins, our docking simulation studies predicted that FKN-CD directly interacts with integrin α(v)ß(3). Consistent with this prediction, we demonstrated that FKN-CD directly bound to α(v)ß(3) and α(4)ß(1) at a very high affinity (K(D) of 3.0 × 10(-10) M to α(v)ß(3) in 1 mM Mn(2+)). Also, membrane-bound FKN bound to integrins α(v)ß(3) and α(4)ß(1), suggesting that the FKN-CD/integrin interaction is biologically relevant. The binding site for FKN-CD in α(v)ß(3) was similar to those for other known α(v)ß(3) ligands. Wild-type FKN-CD induced coprecipitation of integrins and CX3CR1 in U937 cells, suggesting that FKN-CD induces ternary complex formation (CX3CR1, FKN-CD, and integrin). Based on the docking model, we generated an integrin-binding defective FKN-CD mutant (the K36E/R37E mutant). K36E/R37E was defective in ternary complex formation and integrin activation, whereas K36E/R37E still bound to CX3CR1. These results suggest that FKN-CD binding to CX3CR1 is not sufficient for FKN signaling, and that FKN-CD binding to integrins as coreceptors and the resulting ternary complex formation are required for FKN signaling. Notably, excess K36E/R37E suppressed integrin activation induced by wild-type FKN-CD and effectively suppressed leukocyte infiltration in thioglycollate-induced peritonitis. These findings suggest that K36E/R37E acts as a dominant-negative CX3CR1 antagonist and that FKN-CD/integrin interaction is a novel therapeutic target in inflammatory diseases.


Asunto(s)
Quimiocina CX3CL1/genética , Quimiocina CX3CL1/metabolismo , Integrina alfa4beta1/metabolismo , Integrina alfaVbeta3/metabolismo , Receptores de Citocinas/antagonistas & inhibidores , Receptores de Citocinas/metabolismo , Receptores del VIH/antagonistas & inhibidores , Receptores del VIH/metabolismo , Animales , Células CHO , Receptor 1 de Quimiocinas CX3C , Quimiocinas/metabolismo , Simulación por Computador , Cricetinae , Cricetulus , Humanos , Células K562 , Mutación/genética , Unión Proteica/genética , Unión Proteica/inmunología , Estructura Terciaria de Proteína/genética , Transducción de Señal/genética , Transducción de Señal/inmunología , Células U937
13.
Front Cell Dev Biol ; 12: 1347616, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38803393

RESUMEN

VEGF-A is a key cytokine in tumor angiogenesis and a major therapeutic target for cancer. VEGF165 is the predominant isoform of VEGF-A, and it is the most potent angiogenesis stimulant. VEGFR2/KDR domains 2 and 3 (D2D3) bind to the N-terminal domain (NTD, residues 1-110) of VEGF165. Since removal of the heparin-binding domain (HBD, residues 111-165) markedly reduced the mitogenic activity of the growth factor, it has been proposed that the HBD plays a critical role in the mitogenicity of VEGF165. Here, we report that αvß3 specifically bound to the isolated VEGF165 HBD but not to VEGF165 NTD. Based on docking simulation and mutagenesis, we identified several critical amino acid residues within the VEGF165 HBD required for αvß3 binding, i.e., Arg123, Arg124, Lys125, Lys140, Arg145, and Arg149. We discovered that VEGF165 HBD binds to the KDR domain 1 (D1) and identified that Arg123 and Arg124 are critical for KDR D1 binding by mutagenesis, indicating that the KDR D1-binding and αvß3-binding sites overlap in the HBD. Full-length VEGF165 mutant (R123A/R124A/K125A/K140A/R145A/R149A) defective in αvß3 and KDR D1 binding failed to induce ERK1/2 phosphorylation, integrin ß3 phosphorylation, and KDR phosphorylation and did not support proliferation of endothelial cells, although the mutation did not affect the KDR D2D3 interaction with VEGF165. Since ß3-knockout mice are known to show enhanced VEGF165 signaling, we propose that the binding of KDR D1 to the VEGF165 HBD and KDR D2D3 binding to the VEGF165 NTD are critically involved in the potent mitogenicity of VEGF165. We propose that binding competition between KDR and αvß3 to the VEGF165 HBD endows integrin αvß3 with regulatory properties to act as a negative regulator of VEGF165 signaling.

14.
Cells ; 13(4)2024 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-38391921

RESUMEN

FGF9 is a potent mitogen and survival factor, but FGF9 protein levels are generally low and restricted to a few adult organs. Aberrant expression of FGF9 usually results in cancer. However, the mechanism of FGF9 action has not been fully established. Previous studies showed that FGF1 and FGF2 directly bind to integrin αvß3, and this interaction is critical for signaling functions (FGF-integrin crosstalk). FGF1 and FGF2 mutants defective in integrin binding were defective in signaling, whereas the mutants still bound to FGFR suppressed angiogenesis and tumor growth, indicating that they act as antagonists. We hypothesize that FGF9 requires direct integrin binding for signaling. Here, we show that docking simulation of the interaction between FGF9 and αvß3 predicted that FGF9 binds to the classical ligand-binding site of αvß3. We show that FGF9 bound to integrin αvß3 and generated FGF9 mutants in the predicted integrin-binding interface. An FGF9 mutant (R108E) was defective in integrin binding, activating FRS2α and ERK1/2, inducing DNA synthesis, cancer cell migration, and invasion in vitro. R108E suppressed DNA synthesis and activation of FRS2α and ERK1/2 induced by WT FGF9 (dominant-negative effect). These findings indicate that FGF9 requires direct integrin binding for signaling and that R108E has potential as an antagonist to FGF9 signaling.


Asunto(s)
Integrina alfaVbeta3 , Mitógenos , Integrina alfaVbeta3/metabolismo , Ligandos , Factor 1 de Crecimiento de Fibroblastos , Factor 2 de Crecimiento de Fibroblastos , ADN
15.
J Biol Chem ; 287(15): 12491-500, 2012 Apr 06.
Artículo en Inglés | MEDLINE | ID: mdl-22351760

RESUMEN

Integrin αvß3 plays a role in insulin-like growth factor-1 (IGF1) signaling (integrin-IGF1 receptor (IGF1R) cross-talk). The specifics of the cross-talk are, however, unclear. In a current model, "ligand occupancy" of αvß3 (i.e. the binding of extracellular matrix proteins) enhances signaling induced by IGF1 binding to IGF1R. We recently reported that IGF1 directly binds to αvß3 and induces αvß3-IGF1-IGF1R ternary complex formation. Consistently, the integrin binding-defective IGF1 mutant (R36E/R37E) is defective in inducing ternary complex formation and IGF signaling, but it still binds to IGF1R. Like αvß3, integrin α6ß4 is overexpressed in many cancers and is implicated in cancer progression. Here, we discovered that α6ß4 directly bound to IGF1, but not to R36E/R37E. Grafting the ß4 sequence WPNSDP (residues 167-172), which corresponds to the specificity loop of ß3, to integrin ß1 markedly enhanced IGF1 binding to ß1, suggesting that the WPNSDP sequence is involved in IGF1 recognition. WT IGF1 induced α6ß4-IGF1-IGF1R ternary complex formation, whereas R36E/R37E did not. When cells were attached to matrix, exogenous IGF1 or α6ß4 expression had little or no effect on intracellular signaling. When cell-matrix adhesion was reduced (in poly(2-hydroxyethyl methacrylate-coated plates), IGF1 induced intracellular signaling and enhanced cell survival in an α6ß4-dependent manner. Also IGF1 enhanced colony formation in soft agar in an α6ß4-dependent manner. These results suggest that IGF binding to α6ß4 plays a major role in IGF signaling in anchorage-independent conditions, which mimic the in vivo environment, and is a novel therapeutic target.


Asunto(s)
Factor I del Crecimiento Similar a la Insulina/metabolismo , Integrina alfa6beta4/metabolismo , Receptor Cross-Talk , Receptor IGF Tipo 1/metabolismo , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , Sitios de Unión , Células CHO , Adhesión Celular , Técnicas de Cultivo de Célula , Cricetinae , Expresión Génica , Humanos , Factor I del Crecimiento Similar a la Insulina/fisiología , Integrina alfa6beta4/química , Integrina alfa6beta4/genética , Ratones , Datos de Secuencia Molecular , Complejos Multiproteicos/metabolismo , Unión Proteica , Transducción de Señal
16.
Bioorg Med Chem Lett ; 23(1): 340-5, 2013 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-23164706

RESUMEN

Increased concentrations of secreted phospholipase A2 type IIA (sPLA2-IIA), have been found in the synovial fluid of patients with rheumatoid arthritis. It has been shown that sPLA2-IIA specifically binds to integrin αvß3, and initiates a signaling pathway that leads to cell proliferation and inflammation. Therefore, the interaction between integrin and sPLA2-IIA could be a potential therapeutic target for the treatment of proliferation or inflammation-related diseases. Two one-bead-one-compound peptide libraries were constructed and screened, and seven target hits were identified. Herein we report the identification, synthesis, and biological testing of two pyrazolylthiazole-tethered peptide hits and their analogs. Biological assays showed that these compounds were able to suppress the sPLA2-IIA-integrin interaction and sPLA2-IIA-induced migration of monocytic cells and that the blockade of the sPLA2-IIA-integrin binding was specific to sPLA2-IIA and not to the integrin.


Asunto(s)
Fosfolipasas A2 Grupo II/metabolismo , Integrina alfaVbeta3/metabolismo , Secuencia de Aminoácidos , Sitios de Unión , Fosfolipasas A2 Grupo II/antagonistas & inhibidores , Integrina alfaVbeta3/antagonistas & inhibidores , Simulación del Acoplamiento Molecular , Biblioteca de Péptidos , Péptidos/química , Péptidos/farmacología , Unión Proteica/efectos de los fármacos , Estructura Terciaria de Proteína , Relación Estructura-Actividad , Tiazoles/química
17.
Cells ; 12(15)2023 07 31.
Artículo en Inglés | MEDLINE | ID: mdl-37566056

RESUMEN

CD40L is expressed in activated T cells, and it plays a major role in immune response and is a major therapeutic target for inflammation. High IgM syndrome type 1 (HIGM1) is a congenital functional defect in CD40L/CD40 signaling due to defective CD40L. CD40L is also stored in platelet granules and transported to the surface upon platelet activation. Platelet integrin αIIbß3 is known to bind to fibrinogen and activation of αIIbß3 is a key event that triggers platelet aggregation. Also, the KGD motif is critical for αIIbß3 binding and the interaction stabilizes thrombus. Previous studies showed that CD40L binds to and activates integrins αvß3 and α5ß1 and that HIGM1 mutations are clustered in the integrin-binding sites. However, the specifics of CD40L binding to αIIbß3 were unclear. Here, we show that CD40L binds to αIIbß3 in a KGD-independent manner using CD40L that lacks the KGD motif. Two HIGM1 mutants, S128E/E129G and L155P, reduced the binding of CD40L to the classical ligand-binding site (site 1) of αIIbß3, indicating that αIIbß3 binds to the outer surface of CD40L trimer. Also, CD40L bound to the allosteric site (site 2) of αIIbß3 and allosterically activated αIIbß3 without inside-out signaling. Two HIMG1 mutants, K143T and G144E, on the surface of trimeric CD40L suppressed CD40L-induced αIIbß3 activation. These findings suggest that CD40L binds to αIIbß3 in a manner different from that of αvß3 and α5ß1 and induces αIIbß3 activation. HIGM1 mutations are clustered in αIIbß3 binding sites in CD40L and are predicted to suppress thrombus formation and immune responses through αIIbß3.


Asunto(s)
Síndrome de Inmunodeficiencia con Hiper-IgM Tipo 1 , Trombosis , Humanos , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/metabolismo , Ligando de CD40/genética , Ligando de CD40/metabolismo , Sitio Alostérico , Sitios de Unión , Mutación/genética , Integrina alfa5beta1/metabolismo
18.
Life Sci Alliance ; 6(7)2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37184585

RESUMEN

Recognition of integrins by CD62P has not been reported and this motivated a docking simulation using integrin αvß3 as a target. We predicted that the C-type lectin domain of CD62P functions as a potential integrin ligand and observed that it specifically bound to soluble ß3 and ß1 integrins. Known inhibitors of the interaction between CD62P-PSGL-1 did not suppress the binding, whereas the disintegrin domain of ADAM-15, a known integrin ligand, suppressed recognition by the lectin domain. Furthermore, an R16E/K17E mutation in the predicted integrin-binding interface located outside of the glycan-binding site within the lectin domain, strongly inhibited CD62P binding to integrins. In contrast, the E88D mutation that strongly disrupts glycan binding only slightly affected CD62P-integrin recognition, indicating that the glycan and integrin-binding sites are distinct. Notably, the lectin domain allosterically activated integrins by binding to the allosteric site 2. We conclude that CD62P-integrin binding may function to promote a diverse set of cell-cell adhesive interactions given that ß3 and ß1 integrins are more widely expressed than PSGL-1 that is limited to leukocytes.


Asunto(s)
Adhesión Celular , Integrina alfaVbeta3 , Lectinas Tipo C , Selectina-P , Dominios Proteicos , Lectinas Tipo C/química , Humanos , Animales , Células CHO , Cricetulus , Selectina-P/química , Selectina-P/genética , Selectina-P/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Ligandos , Mutación , Integrina alfaVbeta3/metabolismo , Glicoproteínas de Membrana/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas ADAM/metabolismo , Unión Proteica , Sitio Alostérico , Comunicación Celular
19.
Cells ; 12(18)2023 09 13.
Artículo en Inglés | MEDLINE | ID: mdl-37759488

RESUMEN

Integrins were originally identified as receptors for extracellular matrix (ECM) and cell-surface molecules (e.g., VCAM-1 and ICAM-1). Later, we discovered that many soluble growth factors/cytokines bind to integrins and play a critical role in growth factor/cytokine signaling (growth factor-integrin crosstalk). We performed a virtual screening of protein data bank (PDB) using docking simulations with the integrin headpiece as a target. We showed that several growth factors (e.g., FGF1 and IGF1) induce a integrin-growth factor-cognate receptor ternary complex on the surface. Growth factor/cytokine mutants defective in integrin binding were defective in signaling functions and act as antagonists of growth factor signaling. Unexpectedly, several growth factor/cytokines activated integrins by binding to the allosteric site (site 2) in the integrin headpiece, which is distinct from the classical ligand (RGD)-binding site (site 1). Since 25-hydroxycholesterol, a major inflammatory mediator, binds to site 2, activates integrins, and induces inflammatory signaling (e.g., IL-6 and TNFα secretion), it has been proposed that site 2 is involved in inflammatory signaling. We showed that several inflammatory factors (CX3CL1, CXCL12, CCL5, sPLA2-IIA, and P-selectin) bind to site 2 and activate integrins. We propose that site 2 is involved in the pro-inflammatory action of these proteins and a potential therapeutic target. It has been well-established that platelet integrin αIIbß3 is activated by signals from the inside of platelets induced by platelet agonists (inside-out signaling). In addition to the canonical inside-out signaling, we showed that αIIbß3 can be allosterically activated by inflammatory cytokines/chemokines that are stored in platelet granules (e.g., CCL5, CXCL12) in the absence of inside-out signaling (e.g., soluble integrins in cell-free conditions). Thus, the allosteric activation may be involved in αIIbß3 activation, platelet aggregation, and thrombosis. Inhibitory chemokine PF4 (CXCL4) binds to site 2 but did not activate integrins, Unexpectedly, we found that PF4/anti-PF4 complex was able to activate integrins, indicating that the anti-PF4 antibody changed the phenotype of PF4 from inhibitory to inflammatory. Since autoantibodies to PF4 are detected in vaccine-induced thrombocytopenic thrombosis (VIPP) and autoimmune diseases (e.g., SLE, and rheumatoid arthritis), we propose that this phenomenon is related to the pathogenesis of these diseases. P-selectin is known to bind exclusively to glycans (e.g., sLex) and involved in cell-cell interaction by binding to PSGL-1 (CD62P glycoprotein ligand-1). Unexpectedly, through docking simulation, we discovered that the P-selectin C-type lectin domain functions as an integrin ligand. It is interesting that no one has studied whether P-selectin binds to integrins in the last few decades. The integrin-binding site and glycan-binding site were close but distinct. Also, P-selectin lectin domain bound to site 2 and allosterically activated integrins.


Asunto(s)
Comunicación Celular , Selectina-P , Regulación Alostérica , Ligandos , Péptidos y Proteínas de Señalización Intercelular , Factores Inmunológicos , Citocinas , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria
20.
bioRxiv ; 2023 Dec 02.
Artículo en Inglés | MEDLINE | ID: mdl-38076804

RESUMEN

FGF9 is a potent mitogen and survival factor, but FGF9 protein level is generally low and restricted to a few adult organs. Aberrant expression of FGF9 usually results in cancer. However, the mechanism of FGF9 action has not been fully established. Previous studies showed that FGF1 and FGF2 directly bind to integrin αvß3 and this interaction is critical for signaling functions (FGF-integrin crosstalk). FGF1 and FGF2 mutants defective in integrin binding were defective in signaling, whereas the mutants still bound to FGFR, and suppressed angiogenesis and tumor growth, indicating that they act as antagonists. We hypothesize that FGF9 requires direct integrin binding for signaling. Here we show that docking simulation of interaction between FGF9 and αvß3 predicted that FGF9 binds to the classical ligand-binding site of αvß3. We showed that FGF9 actually bound to integrin αvß3, and generated an FGF9 mutants in the predicted integrin-binding interface. An FGF9 mutant (R108E) was defective in integrin binding, activating FRS2α and ERK1/2, inducing DNA synthesis, cancer cell migration, and invasion in vitro. R108E suppressed DNA synthesis induced by WT FGF9 and suppressed DNA synthesis and activation of FRS2α and ERK1/2 induced by WT FGF9 (dominant-negative effect). These findings indicate that FGF9 requires direct integrin binding for signaling and that R108E has potential as an antagonist to FGF9 signaling.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA