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

Base de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
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.

2.
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
3.
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.

4.
bioRxiv ; 2023 Nov 14.
Artículo en Inglés | MEDLINE | ID: mdl-38014319

RESUMEN

VEGF-A is a key cytokine in tumor angiogenesis and a major therapeutic target for cancer. VEGF165 is the predominant isoform and 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 VEGF165, it has been proposed that the HBD plays a critical role in the mitogenicity of VEGF165. Integrin αvß3 has been shown to bind to VEGF165, but the role of integrin αvß3 in VEGF165 signaling are unclear. Here we describe that αvß3 specifically bound to the isolated HBD, but not to the NTD. We identified several critical amino acid residues in HBD for integrin binding (Arg-123, Arg-124, Lys-125, Lys-140, Arg-145, and Arg-149) by docking simulation and mutagenesis, and generated full-length VEGF165 that is defective in integrin binding by including mutations in the HBD. The full-length VEGF165 mutant defective in integrin binding (R123A/R124A/K125A/K140A/R145A/R149A) was defective in ERK1/2 phosphorylation, integrin ß3 phosphorylation, and KDR phosphorylation, although the mutation did not affect KDR binding to VEGF165. We propose a model in which VEGF165 induces KDR (through NTD)-VEGF165 (through HBD)-integrin αvß3 ternary complex formation on the cell surface and this process is critically involved in potent mitogenicity of VEGF165.

5.
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
6.
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
7.
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
8.
Anticancer Res ; 43(2): 569-580, 2023 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-36697085

RESUMEN

BACKGROUND/AIM: Itraconazole (ITZ), an antifungal agent, has been reported to have anti-tumor effects in patients with multiple cancer types. We investigated the involvement of tumor-associated macrophages (TAMs) in its tumor-agnostic mechanism. MATERIALS AND METHODS: M1 and M2 macrophages were established from human monocyte leukemia cell line (THP-1) and their phenotypes were determined morphologically. Cell membrane antigens and secreted proteins were evaluated by western blots and enzyme-linked immunosorbent assay, respectively. The proteomic profiling of cells was done by liquid chromatography with tandem mass spectrometry and analyzed. Viability of cervical cancer cells (CaSki) was evaluated after addition of the supernatant of M2 macrophages and during co-culture with M2 macrophages, with or without 10-5 M ITZ. RESULTS: Co-culture of M1 macrophages inhibited the proliferation of CaSki cells (p=0.012), while that of M2 macrophages promoted their proliferation (p<0.0001). After treatment of M2 macrophages with ITZ for 24 h, they changed into M1-like shape with decreased expression of cluster of differentiation 163 (CD163) and chemokine ligand 18 (CCL18). The M1-like shape was maintained for 7 weeks of ITZ treatment and reverted to original after ITZ removal. Proteomic analysis of ITZ treated-M2 macrophages also demonstrated M1-like signature including the elevated levels of tumor necrosis factor (TNF)-related proteins. After treatment with ITZ, both the supernatant of the M2 macrophages and the co-culture with M2 macrophages significantly inhibited the proliferation of CaSki cells (each, p<0.0001). CONCLUSION: ITZ repolarized M2 macrophages to M1 type and suppressed cervical cancer cell growth demonstrating TAM-mediated anti-cancer activity of ITZ.


Asunto(s)
Macrófagos Asociados a Tumores , Neoplasias del Cuello Uterino , Femenino , Humanos , Itraconazol/farmacología , Neoplasias del Cuello Uterino/patología , Proteómica , Macrófagos/metabolismo , Línea Celular Tumoral , Diferenciación Celular
9.
Cells ; 11(19)2022 09 29.
Artículo en Inglés | MEDLINE | ID: mdl-36231020

RESUMEN

Activation of platelet integrin αIIbß3, a key event for hemostasis and thrombus formation, is known to be mediated exclusively by inside-out signaling. We showed that inflammatory chemokines CX3CL1 and CXCL12 in previous studies, and CCL5 in this study, bound to the allosteric binding site (site 2) of vascular integrin αvß3, in addition to the classical ligand binding site (site 1), and allosterically activated integrins independent of inside-out signaling. Since αIIbß3 is exposed to inflammatory chemokines at increased concentrations during inflammation (e.g., cytokine/chemokine storm) and platelet activation, we hypothesized that these chemokines bind to and activate αIIbß3 in an allosteric activation mechanism. We found that these chemokines bound to αIIbß3. Notably, they activated soluble αIIbß3 in 1 mM Ca2+ by binding to site 2. They activated cell-surface αIIbß3 on CHO cells, which lack machinery for inside-out signaling or chemokine receptors, quickly (<1 min) and at low concentrations (1-10 ng/mL) compared to activation of soluble αIIbß3, probably because chemokines bind to cell surface proteoglycans. Furthermore, activation of αIIbß3 by the chemokines was several times more potent than 1 mM Mn2+. We propose that CCL5 and CXCL12 (stored in platelet granules) may allosterically activate αIIbß3 upon platelet activation and trigger platelet aggregation. Transmembrane CX3CL1 on activated endothelial cells may mediate platelet-endothelial interaction by binding to and activating αIIbß3. Additionally, these chemokines in circulation over-produced during inflammation may trigger αIIbß3 activation, which is a possible missing link between inflammation and thrombosis.


Asunto(s)
Integrina alfaVbeta3 , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria , Animales , Quimiocina CCL5 , Cricetinae , Cricetulus , Células Endoteliales/metabolismo , Inflamación , Integrina alfaVbeta3/metabolismo , Ligandos , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/metabolismo , Proteoglicanos , Receptores de Quimiocina
10.
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
11.
Life Sci Alliance ; 5(8)2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35508387

RESUMEN

The sensitivity of phosphorylation site identification by mass spectrometry has improved markedly. However, the lack of kinase-substrate relationship (KSR) data hinders the improvement of the range and accuracy of kinase activity prediction. In this study, we aimed to develop a method for acquiring systematic KSR data on anaplastic lymphoma kinase (ALK) using mass spectrometry and to apply this method to the prediction of kinase activity. Thirty-seven ALK substrate candidates, including 34 phosphorylation sites not annotated in the PhosphoSitePlus database, were identified by integrated analysis of the phosphoproteome and crosslinking interactome of HEK 293 cells with doxycycline-induced ALK overexpression. Furthermore, KSRs of ALK were validated by an in vitro kinase assay. Finally, using phosphoproteomic data from ALK mutant cell lines and patient-derived cells treated with ALK inhibitors, we found that the prediction of ALK activity was improved when the KSRs identified in this study were used instead of the public KSR dataset. Our approach is applicable to other kinases, and future identification of KSRs will facilitate more accurate estimations of kinase activity and elucidation of phosphorylation signals.


Asunto(s)
Proteoma , Transducción de Señal , Quinasa de Linfoma Anaplásico/metabolismo , Células HEK293 , Humanos , Fosforilación , Proteoma/metabolismo
12.
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
13.
ACS Med Chem Lett ; 11(7): 1416-1420, 2020 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-32676148

RESUMEN

Inhibition of hypoxia-inducible factor prolyl hydroxylase domain (HIF-PHD) promotes erythropoietin (EPO) production by stabilizing the HIFα subunit. Thieno[2,3-d]pyrimidine 8 identified based on X-ray crystal structure analysis was optimized to lead to the discovery of pyrazolo[4,3-d]pyrimidine 13 as the lead compound of orally bioavailable HIF-PHD inhibitors. Conversion of the benzyl moiety in 13 gave pyrazolopyrimidine 19 with high solubility and bioavailability, which increased hemoglobin levels in anemic model rats after repeated oral administration. It was shown that pyrazolo[4,3-d]pyrimidine derivatives are promising therapeutic agents for renal anemia through the inhibition of HIF-PHD.

14.
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
15.
Nat Commun ; 11(1): 860, 2020 02 13.
Artículo en Inglés | MEDLINE | ID: mdl-32054864

RESUMEN

Glycosylphosphatidylinositol (GPI)-anchored proteins and glycosphingolipids interact with each other in the mammalian plasma membranes, forming dynamic microdomains. How their interaction starts in the cells has been unclear. Here, based on a genome-wide CRISPR-Cas9 genetic screen for genes required for GPI side-chain modification by galactose in the Golgi apparatus, we report that ß1,3-galactosyltransferase 4 (B3GALT4), the previously characterized GM1 ganglioside synthase, additionally functions in transferring galactose to the N-acetylgalactosamine side-chain of GPI. Furthermore, B3GALT4 requires lactosylceramide for the efficient GPI side-chain galactosylation. Thus, our work demonstrates previously unexpected functional relationships between GPI-anchored proteins and glycosphingolipids in the Golgi. Through the same screening, we also show that GPI biosynthesis in the endoplasmic reticulum (ER) is severely suppressed by ER-associated degradation to prevent GPI accumulation when the transfer of synthesized GPI to proteins is defective. Our data demonstrates cross-talks of GPI biosynthesis with glycosphingolipid biosynthesis and the ER quality control system.


Asunto(s)
Degradación Asociada con el Retículo Endoplásmico , Glicoesfingolípidos/biosíntesis , Glicosilfosfatidilinositoles/biosíntesis , Aciltransferasas/deficiencia , Aciltransferasas/genética , Aciltransferasas/metabolismo , Sistemas CRISPR-Cas , Degradación Asociada con el Retículo Endoplásmico/genética , Galactosiltransferasas/deficiencia , Galactosiltransferasas/genética , Galactosiltransferasas/metabolismo , Técnicas de Inactivación de Genes , Glicoesfingolípidos/genética , Glicosilfosfatidilinositoles/genética , Células HEK293 , Células HeLa , Humanos , Modelos Moleculares , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo
16.
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
17.
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
18.
Dalton Trans ; 48(4): 1161-1165, 2019 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-30543236

RESUMEN

The first phosphido derivative of the bis(bipyridine) ruthenium(ii) fragment, cis-[(bpy)2Ru(PPh2)2] ([RuP2]), has been developed and applied as a P-donor metalloligand to form new Ru-Rh, Ru-Ir and Ru2Cu2 heterometallic complexes. The Ru-Ir hydride complex [([RuP2])IrH(NCMe)3][BF4]2 exhibits significant catalytic activity for (E)-selective semi-hydrogenation of alkynes.

19.
Nat Commun ; 9(1): 405, 2018 01 26.
Artículo en Inglés | MEDLINE | ID: mdl-29374258

RESUMEN

Many eukaryotic proteins are anchored to the cell surface via the glycolipid glycosylphosphatidylinositol (GPI). Mammalian GPIs have a conserved core but exhibit diverse N-acetylgalactosamine (GalNAc) modifications, which are added via a yet unresolved process. Here we identify the Golgi-resident GPI-GalNAc transferase PGAP4 and show by mass spectrometry that PGAP4 knockout cells lose GPI-GalNAc structures. Furthermore, we demonstrate that PGAP4, in contrast to known Golgi glycosyltransferases, is not a single-pass membrane protein but contains three transmembrane domains, including a tandem transmembrane domain insertion into its glycosyltransferase-A fold as indicated by comparative modeling. Mutational analysis reveals a catalytic site, a DXD-like motif for UDP-GalNAc donor binding, and several residues potentially involved in acceptor binding. We suggest that a juxtamembrane region of PGAP4 accommodates various GPI-anchored proteins, presenting their acceptor residue toward the catalytic center. In summary, we present insights into the structure of PGAP4 and elucidate the initial step of GPI-GalNAc biosynthesis.


Asunto(s)
Acetilgalactosamina/química , Glicosilfosfatidilinositoles/química , Aparato de Golgi/metabolismo , N-Acetilgalactosaminiltransferasas/química , Acetilgalactosamina/biosíntesis , Secuencias de Aminoácidos , Animales , Células CHO , Dominio Catalítico , Cricetulus , Cristalografía por Rayos X , Expresión Génica , Vectores Genéticos/química , Vectores Genéticos/metabolismo , Glicosilfosfatidilinositoles/metabolismo , Aparato de Golgi/ultraestructura , Humanos , Ratones , Ratones Noqueados , Modelos Moleculares , Mutación , N-Acetilgalactosaminiltransferasas/genética , N-Acetilgalactosaminiltransferasas/metabolismo , Unión Proteica , Conformación Proteica en Hélice alfa , Conformación Proteica en Lámina beta , Dominios y Motivos de Interacción de Proteínas , Homología Estructural de Proteína , Especificidad por Sustrato , Polipéptido N-Acetilgalactosaminiltransferasa
20.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA