Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
1.
Nature ; 618(7963): 159-168, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-37225977

RESUMEN

Harnessing the potential beneficial effects of kinase signalling through the generation of direct kinase activators remains an underexplored area of drug development1-5. This also applies to the PI3K signalling pathway, which has been extensively targeted by inhibitors for conditions with PI3K overactivation, such as cancer and immune dysregulation. Here we report the discovery of UCL-TRO-1938 (referred to as 1938 hereon), a small-molecule activator of the PI3Kα isoform, a crucial effector of growth factor signalling. 1938 allosterically activates PI3Kα through a distinct mechanism by enhancing multiple steps of the PI3Kα catalytic cycle and causes both local and global conformational changes in the PI3Kα structure. This compound is selective for PI3Kα over other PI3K isoforms and multiple protein and lipid kinases. It transiently activates PI3K signalling in all rodent and human cells tested, resulting in cellular responses such as proliferation and neurite outgrowth. In rodent models, acute treatment with 1938 provides cardioprotection from ischaemia-reperfusion injury and, after local administration, enhances nerve regeneration following nerve crush. This study identifies a chemical tool to directly probe the PI3Kα signalling pathway and a new approach to modulate PI3K activity, widening the therapeutic potential of targeting these enzymes through short-term activation for tissue protection and regeneration. Our findings illustrate the potential of activating kinases for therapeutic benefit, a currently largely untapped area of drug development.


Asunto(s)
Regeneración Nerviosa , Humanos , Neoplasias/tratamiento farmacológico , Regeneración Nerviosa/efectos de los fármacos , Isoformas de Proteínas/agonistas , Transducción de Señal/efectos de los fármacos , Fosfatidilinositol 3-Quinasa Clase I/química , Fosfatidilinositol 3-Quinasa Clase I/efectos de los fármacos , Cardiotónicos/farmacología , Animales , Biocatálisis/efectos de los fármacos , Conformación Proteica/efectos de los fármacos , Neuritas/efectos de los fármacos , Daño por Reperfusión/prevención & control , Compresión Nerviosa , Proliferación Celular/efectos de los fármacos
2.
3.
Biochim Biophys Acta Mol Basis Dis ; 1869(2): 166601, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36442790

RESUMEN

BACKGROUND: Development of adult T-cell leukemia/lymphoma (ATL) involves human T-cell leukemia virus type 1 (HTLV-1) infection and accumulation of somatic mutations. The most frequently mutated gene in ATL (36 % of cases) is phospholipase C gamma1 (PLCG1). PLCG1 is also frequently mutated in other T-cell lymphomas. However, the functional consequences of the PLCG1 mutations in cancer cells have not been characterized. METHODS: We compared the activity of the wild-type PLCγ1 with that of a mutant carrying a hot-spot mutation of PLCγ1 (S345F) observed in ATL, both in cells and in cell-free assays. To analyse the impact of the mutation on cellular properties, we quantified cellular proliferation, aggregation, chemotaxis and apoptosis by live cell-imaging in an S345F+ ATL-derived cell line (KK1) and a KK1 cell line in which we reverted the mutation to the wild-type sequence using CRISPR/Cas9 and homology-directed repair. FINDINGS: The PLCγ1 S345F mutation results in an increase of basal PLC activity in vitro and in different cell types. This higher basal activity is further enhanced by upstream signalling. Reversion of the S345F mutation in the KK1 cell line resulted in reduction of the PLC activity, lower rates of proliferation and aggregation, and a marked reduction in chemotaxis towards CCL22. The PLCγ1-pathway inhibitors ibrutinib and ritonavir reduced both the PLC activity and the tested functions of KK1 cells. INTERPRETATION: Consistent with observations from clinical studies, our data provide direct evidence that activated variants of the PLCγ1 enzyme contribute to the properties of the malignant T-cell clone in ATL. FUNDING: MRC (UK) Project Grant (P028160).


Asunto(s)
Virus Linfotrópico T Tipo 1 Humano , Leucemia-Linfoma de Células T del Adulto , Fosfolipasa C gamma , Adulto , Humanos , Leucemia-Linfoma de Células T del Adulto/genética , Mutación , Fosfolipasa C gamma/genética
4.
Sci Adv ; 8(25): eabp9688, 2022 06 24.
Artículo en Inglés | MEDLINE | ID: mdl-35749497

RESUMEN

PLCγ enzymes are autoinhibited in resting cells and form key components of intracellular signaling that are also linked to disease development. Insights into physiological and aberrant activation of PLCγ require understanding of an active, membrane-bound form, which can hydrolyze inositol-lipid substrates. Here, we demonstrate that PLCγ1 cannot bind membranes unless the autoinhibition is disrupted. Through extensive molecular dynamics simulations and experimental evidence, we characterize membrane binding by the catalytic core domains and reveal previously unknown sites of lipid interaction. The identified sites act in synergy, overlap with autoinhibitory interfaces, and are shown to be critical for the phospholipase activity in cells. This work provides direct evidence that PLCγ1 is inhibited through obstruction of its membrane-binding surfaces by the regulatory region and that activation must shift PLCγ1 to a conformation competent for membrane binding. Knowledge of the critical sites of membrane interaction extends the mechanistic framework for activation, dysregulation, and therapeutic intervention.


Asunto(s)
Lípidos , Transducción de Señal , Dominio Catalítico
5.
Mol Neurodegener ; 16(1): 22, 2021 04 06.
Artículo en Inglés | MEDLINE | ID: mdl-33823896

RESUMEN

The central role of the resident innate immune cells of the brain (microglia) in neurodegeneration has become clear over the past few years largely through genome-wide association studies (GWAS), and has rapidly become an active area of research. However, a mechanistic understanding (gene to function) has lagged behind. That is now beginning to change, as exemplified by a number of recent exciting and important reports that provide insight into the function of two key gene products - TREM2 (Triggering Receptor Expressed On Myeloid Cells 2) and PLCγ2 (Phospholipase C gamma2) - in microglia, and their role in neurodegenerative disorders. In this review we explore and discuss these recent advances and the opportunities that they may provide for the development of new therapies.


Asunto(s)
Enfermedad de Alzheimer/inmunología , Células del Tejido Conectivo/metabolismo , Linfocitos/metabolismo , Glicoproteínas de Membrana/fisiología , Microglía/metabolismo , Células Mieloides/metabolismo , Fosfolipasa C gamma/fisiología , Receptores Inmunológicos/fisiología , Transducción de Señal/fisiología , Edad de Inicio , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/prevención & control , Humanos , Metabolismo de los Lípidos , Glicoproteínas de Membrana/química , Microglía/fisiología , Modelos Moleculares , Mutación , Fosfolipasa C gamma/química , Fosfolipasa C gamma/genética , Conformación Proteica , Dominios Proteicos , Mapeo de Interacción de Proteínas , Receptores Inmunológicos/química , Homología de Secuencia de Aminoácido , Relación Estructura-Actividad
6.
J Clin Immunol ; 40(7): 987-1000, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32671674

RESUMEN

Autoinflammatory diseases (AIDs) were first described as clinical disorders characterized by recurrent episodes of seemingly unprovoked sterile inflammation. In the past few years, the identification of novel AIDs expanded their phenotypes toward more complex clinical pictures associating vasculopathy, autoimmunity, or immunodeficiency. Herein, we describe two unrelated patients suffering since the neonatal period from a complex disease mainly characterized by severe sterile inflammation, recurrent bacterial infections, and marked humoral immunodeficiency. Whole-exome sequencing detected a novel, de novo heterozygous PLCG2 variant in each patient (p.Ala708Pro and p.Leu845_Leu848del). A clear enhanced PLCγ2 activity for both variants was demonstrated by both ex vivo calcium responses of the patient's B cells to IgM stimulation and in vitro assessment of PLC activity. These data supported the autoinflammation and PLCγ2-associated antibody deficiency and immune dysregulation (APLAID) diagnosis in both patients. Immunological evaluation revealed a severe decrease of immunoglobulins and B cells, especially class-switched memory B cells, with normal T and NK cell counts. Analysis of bone marrow of one patient revealed a reduced immature B cell fraction compared with controls. Additional investigations showed that both PLCG2 variants activate the NLRP3-inflammasome through the alternative pathway instead of the canonical pathway. Collectively, the evidences here shown expand APLAID diversity toward more severe phenotypes than previously reported including dominantly inherited agammaglobulinemia, add novel data about its genetic basis, and implicate the alternative NLRP3-inflammasome activation pathway in the basis of sterile inflammation.


Asunto(s)
Agammaglobulinemia/diagnóstico , Agammaglobulinemia/genética , Enfermedades Autoinflamatorias Hereditarias/diagnóstico , Enfermedades Autoinflamatorias Hereditarias/genética , Mutación , Fosfolipasa C gamma/genética , Adolescente , Agammaglobulinemia/terapia , Autoinmunidad/genética , Biomarcadores , Caspasa 1/metabolismo , Niño , Citocinas/metabolismo , Análisis Mutacional de ADN , Femenino , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Enfermedades Autoinflamatorias Hereditarias/terapia , Humanos , Inflamasomas/metabolismo , Masculino , Linaje , Fenotipo , Fosfolipasa C gamma/química , Fosfolipasa C gamma/metabolismo , Relación Estructura-Actividad
7.
Int J Mol Sci ; 21(9)2020 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-32370101

RESUMEN

Selective FGFR inhibitors such as infigratinib (BGJ398) and erdafitinib (JNJ-42756493) have been evaluated in clinical trials for cancers with FGFR3 molecular alterations, particularly in urothelial carcinoma patients. However, a substantial proportion of these patients (up to 50%) display intrinsic resistance to these drugs and receive minimal clinical benefit. There is thus an unmet need for alternative therapeutic strategies to overcome primary resistance to selective FGFR inhibitors. In this study, we demonstrate that cells expressing cancer-associated activating FGFR3 mutants and the FGFR3-TACC3 fusion showed primary resistance to infigratinib in long-term colony formation assays in both NIH-3T3 and urothelial carcinoma models. We find that expression of these FGFR3 molecular alterations resulted in elevated constitutive Src activation compared to wildtype FGFR3 and that cells co-opted this pathway as a means to achieve intrinsic resistance to infigratinib. Targeting the Src pathway with low doses of the kinase inhibitor dasatinib synergistically sensitized multiple urothelial carcinoma lines harbouring endogenous FGFR3 alterations to infigratinib. Our data provide preclinical rationale that supports the use of dasatinib in combination with selective FGFR inhibitors as a means to overcome intrinsic drug resistance in the salvage therapy setting in urothelial cancer patients with FGFR3 molecular alterations.


Asunto(s)
Inhibidores de Proteínas Quinasas/farmacología , Receptor Tipo 3 de Factor de Crecimiento de Fibroblastos/metabolismo , Transducción de Señal/efectos de los fármacos , Neoplasias Urológicas/metabolismo , Familia-src Quinasas/metabolismo , Animales , Línea Celular Tumoral , Humanos , Ratones , Proteínas Asociadas a Microtúbulos/genética , Terapia Molecular Dirigida , Células 3T3 NIH , Proteínas de Fusión Oncogénica/genética , Proteínas de Fusión Oncogénica/metabolismo , Inhibidores de Proteínas Quinasas/uso terapéutico , Neoplasias Urológicas/tratamiento farmacológico , Neoplasias Urológicas/etiología , Neoplasias Urológicas/patología
8.
EBioMedicine ; 51: 102607, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31918402

RESUMEN

BACKGROUND: PLCγ enzymes are key nodes in cellular signal transduction and their mutated and rare variants have been recently implicated in development of a range of diseases with unmet need including cancer, complex immune disorders, inflammation and neurodegenerative diseases. However, molecular nature of activation and the impact and dysregulation mechanisms by mutations, remain unclear; both are critically dependent on comprehensive characterization of the intact PLCγ enzymes. METHODS: For structural studies we applied cryo-EM, cross-linking mass spectrometry and hydrogen-deuterium exchange mass spectrometry. In parallel, we compiled mutations linked to main pathologies, established their distribution and assessed their impact in cells and in vitro. FINDINGS: We define structure of a complex containing an intact, autoinhibited PLCγ1 and the intracellular part of FGFR1 and show that the interaction is centred on the nSH2 domain of PLCγ1. We define the architecture of PLCγ1 where an autoinhibitory interface involves the cSH2, spPH, TIM-barrel and C2 domains; this relative orientation occludes PLCγ1 access to its substrate. Based on this framework and functional characterization, the mechanism leading to an increase in PLCγ1 activity for the largest group of mutations is consistent with the major, direct impact on the autoinhibitory interface. INTERPRETATION: We reveal features of PLCγ enzymes that are important for determining their activation status. Targeting such features, as an alternative to targeting the PLC active site that has so far not been achieved for any PLC, could provide new routes for clinical interventions related to various pathologies driven by PLCγ deregulation. FUND: CR UK, MRC and AstaZeneca.


Asunto(s)
Mutación/genética , Fosfolipasa C gamma/química , Fosfolipasa C gamma/genética , Humanos , Modelos Moleculares , Proteínas Mutantes/química , Proteínas Mutantes/genética , Fosfolipasa C gamma/ultraestructura , Unión Proteica , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/metabolismo
9.
Biophys J ; 115(1): 31-45, 2018 07 03.
Artículo en Inglés | MEDLINE | ID: mdl-29972810

RESUMEN

Phosphatidylinositol phospholipase Cγ (PLCγ) is an intracellular membrane-associated second-messenger signaling protein activated by tyrosine kinases such as fibroblast growth factor receptor 1. PLCγ contains the regulatory γ-specific array (γSA) comprising a tandem Src homology 2 (SH2) pair, an SH3 domain, and a split pleckstrin homology domain. Binding of an activated growth factor receptor to γSA leads to Tyr783 phosphorylation and consequent PLCγ activation. Several disease-relevant mutations in γSA have been identified; all lead to elevated phospholipase activity. In this work, we describe an allosteric mechanism that connects the Tyr783 phosphorylation site to the nSH2-cSH2 junction and involves dynamic interactions between the cSH2-SH3 linker and cSH2. Molecular dynamics simulations of the tandem SH2 protein suggest that Tyr783 phosphorylation is communicated to the nSH2-cSH2 junction by modulating cSH2 binding to sections of the cSH2-SH3 linker. NMR chemical shift perturbation analyses for designed tandem SH2 constructs reveal combined fast and slow dynamic processes that can be attributed to allosteric communication involving these regions of the protein, establishing an example in which complex N-site exchange can be directly inferred from 1H,15N-HSQC spectra. Furthermore, in tandem SH2 and γSA constructs, molecular dynamics and NMR results show that the Arg687Trp mutant in PLCγ1 (equivalent to the cancer mutation Arg665Trp in PLCγ2) perturbs the dynamic allosteric pathway. This combined experimental and computational study reveals a rare example of multistate kinetics involved in a dynamic allosteric process that is modulated in the context of a disease-relevant mutation. The allosteric influences and the weakened binding of the cSH2-SH3 linker to cSH2 should be taken into account in any more holistic investigation of PLCγ regulation.


Asunto(s)
Simulación de Dinámica Molecular , Mutación , Neoplasias/genética , Resonancia Magnética Nuclear Biomolecular , Fosfolipasa C gamma/química , Fosfolipasa C gamma/metabolismo , Regulación Alostérica , Fosfolipasa C gamma/genética , Fosforilación , Dominios Homologos src
10.
Biomol NMR Assign ; 12(2): 231-235, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29582384

RESUMEN

Fibroblast growth factors receptors (FGFR) are transmembrane protein tyrosine kinases involved in many cellular process, including growth, differentiation and angiogenesis. Dysregulation of FGFR enzymatic activity is associated with developmental disorders and cancers; therefore FGFRs have become attractive targets for drug discovery, with a number of agents in late-stage clinical trials. Here, we present the backbone resonance assignments of FGFR3 tyrosine kinase domain in the ligand-free form and in complex with the canonical FGFR kinase inhibitor PD173074. Analysis of chemical shift changes upon inhibitor binding highlights a characteristic pattern of allosteric network perturbations that is of relevance for future drug discovery activities aimed at development of conformationally-selective FGFR inhibitors.


Asunto(s)
Apoproteínas/química , Apoproteínas/metabolismo , Resonancia Magnética Nuclear Biomolecular , Pirimidinas/metabolismo , Receptor Tipo 3 de Factor de Crecimiento de Fibroblastos/química , Receptor Tipo 3 de Factor de Crecimiento de Fibroblastos/metabolismo , Apoproteínas/antagonistas & inhibidores , Humanos , Unión Proteica , Dominios Proteicos , Pirimidinas/farmacología , Receptor Tipo 3 de Factor de Crecimiento de Fibroblastos/antagonistas & inhibidores
11.
Structure ; 26(3): 446-458.e8, 2018 03 06.
Artículo en Inglés | MEDLINE | ID: mdl-29478821

RESUMEN

Receptor tyrosine kinase FGFR3 is involved in many signaling networks and is frequently mutated in developmental disorders and cancer. The Hsp90/Cdc37 chaperone system is essential for function of normal and neoplastic cells. Here we uncover the mechanistic inter-relationships between these proteins by combining approaches including NMR, HDX-MS, and SAXS. We show that several disease-linked mutations convert FGFR3 to a stronger client, where the determinant underpinning client strength involves an allosteric network through the N-lobe and at the lobe interface. We determine the architecture of the client kinase/Cdc37 complex and demonstrate, together with site-specific information, that binding of Cdc37 to unrelated kinases induces a common, extensive conformational remodeling of the kinase N-lobe, beyond localized changes and interactions within the binary complex. As further shown for FGFR3, this processing by Cdc37 deactivates the kinase and presents it, in a specific orientation established in the complex, for direct recognition by Hsp90.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Chaperoninas/metabolismo , Proteínas HSP90 de Choque Térmico/metabolismo , Mutación , Receptor Tipo 3 de Factor de Crecimiento de Fibroblastos/química , Receptor Tipo 3 de Factor de Crecimiento de Fibroblastos/metabolismo , Sitio Alostérico , Humanos , Modelos Moleculares , Resonancia Magnética Nuclear Biomolecular , Unión Proteica , Conformación Proteica , Receptor Tipo 3 de Factor de Crecimiento de Fibroblastos/genética , Dispersión del Ángulo Pequeño , Difracción de Rayos X
12.
Sci Rep ; 7: 39841, 2017 01 03.
Artículo en Inglés | MEDLINE | ID: mdl-28045057

RESUMEN

Protein kinases share significant structural similarity; however, structural features alone are insufficient to explain their diverse functions. Thus, bridging the gap between static structure and function requires a more detailed understanding of their dynamic properties. For example, kinase activation may occur via a switch-like mechanism or by shifting a dynamic equilibrium between inactive and active states. Here, we utilize a combination of FRET and molecular dynamics (MD) simulations to probe the activation mechanism of the kinase domain of Fibroblast Growth Factor Receptor (FGFR). Using genetically-encoded, site-specific incorporation of unnatural amino acids in regions essential for activation, followed by specific labeling with fluorescent moieties, we generated a novel class of FRET-based reporter to monitor conformational differences corresponding to states sampled by non phosphorylated/inactive and phosphorylated/active forms of the kinase. Single molecule FRET analysis in vitro, combined with MD simulations, shows that for FGFR kinase, there are populations of inactive and active states separated by a high free energy barrier resulting in switch-like activation. Compared to recent studies, these findings support diversity in features of kinases that impact on their activation mechanisms. The properties of these FRET-based constructs will also allow further studies of kinase dynamics as well as applications in vivo.


Asunto(s)
Transferencia Resonante de Energía de Fluorescencia/métodos , Simulación de Dinámica Molecular , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/química , Imagen Individual de Molécula/métodos , Sustitución de Aminoácidos , Humanos , Fosforilación , Dominios Proteicos , Procesamiento Proteico-Postraduccional , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/genética , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/metabolismo
13.
Oncotarget ; 7(17): 24252-68, 2016 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-26992226

RESUMEN

Frequent genetic alterations discovered in FGFRs and evidence implicating some as drivers in diverse tumors has been accompanied by rapid progress in targeting FGFRs for anticancer treatments. Wider assessment of the impact of genetic changes on the activation state and drug responses is needed to better link the genomic data and treatment options. We here apply a direct comparative and comprehensive analysis of FGFR3 kinase domain variants representing the diversity of point-mutations reported in this domain. We reinforce the importance of N540K and K650E and establish that not all highly activating mutations (for example R669G) occur at high-frequency and conversely, that some "hotspots" may not be linked to activation. Further structural characterization consolidates a mechanistic view of FGFR kinase activation and extends insights into drug binding. Importantly, using several inhibitors of particular clinical interest (AZD4547, BGJ-398, TKI258, JNJ42756493 and AP24534), we find that some activating mutations (including different replacements of the same residue) result in distinct changes in their efficacy. Considering that there is no approved inhibitor for anticancer treatments based on FGFR-targeting, this information will be immediately translatable to ongoing clinical trials.


Asunto(s)
Benzamidas/farmacología , Biomarcadores de Tumor/genética , Transformación Celular Neoplásica/patología , Mutación , Neoplasias/genética , Piperazinas/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Pirazoles/farmacología , Receptor Tipo 3 de Factor de Crecimiento de Fibroblastos/genética , Animales , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Transformación Celular Neoplásica/efectos de los fármacos , Transformación Celular Neoplásica/genética , Humanos , Ratones , Células 3T3 NIH , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Fosforilación/efectos de los fármacos , Transducción de Señal/efectos de los fármacos
14.
Mol Cell Proteomics ; 15(2): 715-25, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26604261

RESUMEN

AMPylation of mammalian small GTPases by bacterial virulence factors can be a key step in bacterial infection of host cells, and constitutes a potential drug target. This posttranslational modification also exists in eukaryotes, and AMP transferase activity was recently assigned to HYPE Filamentation induced by cyclic AMP domain containing protein (FICD) protein, which is conserved from Caenorhabditis elegans to humans. In contrast to bacterial AMP transferases, only a small number of HYPE substrates have been identified by immunoprecipitation and mass spectrometry approaches, and the full range of targets is yet to be determined in mammalian cells. We describe here the first example of global chemoproteomic screening and substrate validation for HYPE-mediated AMPylation in mammalian cell lysate. Through quantitative mass-spectrometry-based proteomics coupled with novel chemoproteomic tools providing MS/MS evidence of AMP modification, we identified a total of 25 AMPylated proteins, including the previously validated substrate endoplasmic reticulum (ER) chaperone BiP (HSPA5), and also novel substrates involved in pathways of gene expression, ATP biosynthesis, and maintenance of the cytoskeleton. This dataset represents the largest library of AMPylated human proteins reported to date and a foundation for substrate-specific investigations that can ultimately decipher the complex biological networks involved in eukaryotic AMPylation.


Asunto(s)
Proteínas Portadoras/metabolismo , AMP Cíclico/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas de Unión al GTP Monoméricas/metabolismo , Proteómica , Secuencia de Aminoácidos/efectos de los fármacos , Animales , Caenorhabditis elegans/genética , Caenorhabditis elegans/microbiología , Proteínas Portadoras/genética , Retículo Endoplásmico/genética , Retículo Endoplásmico/metabolismo , Chaperón BiP del Retículo Endoplásmico , Interacciones Huésped-Patógeno , Humanos , Infecciones/genética , Infecciones/microbiología , Proteínas de la Membrana/genética , Proteínas de Unión al GTP Monoméricas/genética , Nucleotidiltransferasas , Procesamiento Proteico-Postraduccional/genética , Espectrometría de Masas en Tándem , Transferasas/metabolismo
15.
Adv Biol Regul ; 60: 6-13, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26482290

RESUMEN

In vitro and in vivo imaging of protein tyrosine kinase activity requires minimally invasive, molecularly precise optical probes to provide spatiotemporal mechanistic information of dimerization and complex formation with downstream effectors. We present here a construct with genetically encoded, site-specifically incorporated, bioorthogonal reporter that can be selectively labelled with exogenous fluorogenic probes to monitor the structure and function of fibroblast growth factor receptor (FGFR). GyrB.FGFR1KD.TC contains a coumermycin-induced artificial dimerizer (GyrB), FGFR1 kinase domain (KD) and a tetracysteine (TC) motif that enables fluorescent labelling with biarsenical dyes FlAsH-EDT2 and ReAsH-EDT2. We generated bimolecular system for time-resolved FRET (TR-FRET) studies, which pairs FlAsH-tagged GyrB.FGFR1KD.TC and N-terminal Src homology 2 (nSH2) domain of phospholipase Cγ (PLCγ), a downstream effector of FGFR1, fused to mTurquoise fluorescent protein (mTFP). We demonstrated phosphorylation-dependent TR-FRET readout of complex formation between mTFP.nSH2 and GyrB.FGFR1KD.TC. By further application of TR-FRET, we also demonstrated formation of the GyrB.FGFR1KD.TC homodimer by coumermycin-induced dimerization. Herein, we present a spectroscopic FRET approach to facilitate and propagate studies that would provide structural and functional insights for FGFR and other tyrosine kinases.


Asunto(s)
Transferencia Resonante de Energía de Fluorescencia/métodos , Fosfolipasa C gamma/química , Fosfolipasa C gamma/metabolismo , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/química , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/metabolismo , Dimerización , Humanos , Fosfolipasa C gamma/genética , Unión Proteica , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/genética
16.
EBioMedicine ; 2(3): 194-204, 2015 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-26097890

RESUMEN

Fibroblast growth factor receptors (FGFRs) are recognized therapeutic targets in cancer. We here describe insights underpinning the impact of mutations on FGFR1 and FGFR3 kinase activity and drug efficacy, using a combination of computational calculations and experimental approaches including cellular studies, X-ray crystallography and biophysical and biochemical measurements. Our findings reveal that some of the tested compounds, in particular TKI258, could provide therapeutic opportunity not only for patients with primary alterations in FGFR but also for acquired resistance due to the gatekeeper mutation. The accuracy of the computational methodologies applied here shows a potential for their wider application in studies of drug binding and in assessments of functional and mechanistic impacts of mutations, thus assisting efforts in precision medicine.

17.
Structure ; 22(12): 1831-1843, 2014 Dec 02.
Artículo en Inglés | MEDLINE | ID: mdl-25435325

RESUMEN

Protein AMPylation, the transfer of AMP from ATP to protein targets, has been recognized as a new mechanism of host-cell disruption by some bacterial effectors that typically contain a FIC-domain. Eukaryotic genomes also encode one FIC-domain protein,HYPE, which has remained poorly characterized.Here we describe the structure of human HYPE, solved by X-ray crystallography, representing the first structure of a eukaryotic FIC-domain protein. We demonstrate that HYPE forms stable dimers with structurally and functionally integrated FIC-domains and with TPR-motifs exposed for protein-protein interactions. As HYPE also uniquely possesses a transmembrane helix, dimerization is likely to affect its positioning and function in the membrane vicinity. The low rate of auto AMPylation of the wild-type HYPE could be due to autoinhibition, consistent with the mechanism proposed for a number of putative FIC AMPylators. Our findings also provide a basis to further consider possible alternative cofactors of HYPE and distinct modes of target-recognition.


Asunto(s)
Proteínas Portadoras/metabolismo , Proteínas de la Membrana/metabolismo , Modelos Moleculares , Cristalografía por Rayos X , Humanos , Nucleotidiltransferasas , Conformación Proteica , Estructura Terciaria de Proteína , Relación Estructura-Actividad
18.
Trends Biochem Sci ; 39(12): 603-11, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25456276

RESUMEN

The surge in genetic and genomic investigations over the past 5 years has resulted in many discoveries of causative variants relevant to disease pathophysiology. Although phospholipase C (PLC) enzymes have long been recognized as important components in intracellular signal transmission, it is only recently that this approach highlighted their role in disease development through gain-of-function mutations. In this review we describe the new findings that link the PLCγ family to immune disorders and cancer, and illustrate further efforts to elucidate the molecular mechanisms that underpin their dysfunction.


Asunto(s)
Enfermedades del Sistema Inmune/metabolismo , Modelos Biológicos , Modelos Moleculares , Neoplasias/metabolismo , Fosfolipasa C gamma/metabolismo , Sistemas de Mensajero Secundario , Animales , Diglicéridos/metabolismo , Humanos , Enfermedades del Sistema Inmune/inmunología , Inositol 1,4,5-Trifosfato/metabolismo , Mutación , Neoplasias/genética , Fosfolipasa C gamma/química , Fosfolipasa C gamma/genética , Conformación Proteica
19.
Nat Genet ; 46(4): 376-379, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24633157

RESUMEN

Angiosarcoma is an aggressive malignancy that arises spontaneously or secondarily to ionizing radiation or chronic lymphoedema. Previous work has identified aberrant angiogenesis, including occasional somatic mutations in angiogenesis signaling genes, as a key driver of angiosarcoma. Here we employed whole-genome, whole-exome and targeted sequencing to study the somatic changes underpinning primary and secondary angiosarcoma. We identified recurrent mutations in two genes, PTPRB and PLCG1, which are intimately linked to angiogenesis. The endothelial phosphatase PTPRB, a negative regulator of vascular growth factor tyrosine kinases, harbored predominantly truncating mutations in 10 of 39 tumors (26%). PLCG1, a signal transducer of tyrosine kinases, encoded a recurrent, likely activating p.Arg707Gln missense variant in 3 of 34 cases (9%). Overall, 15 of 39 tumors (38%) harbored at least one driver mutation in angiogenesis signaling genes. Our findings inform and reinforce current therapeutic efforts to target angiogenesis signaling in angiosarcoma.


Asunto(s)
Hemangiosarcoma/tratamiento farmacológico , Hemangiosarcoma/genética , Neovascularización Patológica/tratamiento farmacológico , Fosfolipasa C gamma/genética , Proteínas Tirosina Fosfatasas Clase 3 Similares a Receptores/genética , Análisis de Varianza , Secuencia de Bases , Exoma/genética , Células Endoteliales de la Vena Umbilical Humana , Humanos , Datos de Secuencia Molecular , Mutación/genética , Neovascularización Patológica/genética , Interferencia de ARN , Análisis de Secuencia de ARN , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores
20.
Structure ; 20(12): 2062-75, 2012 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-23063561

RESUMEN

Multidomain proteins incorporating interaction domains are central to regulation of cellular processes. The elucidation of structural organization and mechanistic insights into many of these proteins, however, remain challenging due to their inherent flexibility. Here, we describe the organization and function of four interaction domains in PLCγ1 using a combination of structural biology and biochemical approaches. Intramolecular interactions within the regulatory region center on the cSH2 domain, the only domain that also interacts with the PLC-core. In the context of fibroblast growth-factor receptor signaling, the coordinated involvement of nSH2 and cSH2 domains mediates efficient phosphorylation of PLCγ1 resulting in the interruption of an autoinhibitory interface by direct competition and, independently, dissociation of PLCγ1 from the receptor. Further structural insights into the autoinhibitory surfaces provide a framework to interpret gain-of-function mutations in PLCγ isoforms linked to immune disorders and illustrate a distinct mechanism for regulation of PLC activity by common interaction domains.


Asunto(s)
Modelos Moleculares , Fosfolipasa C gamma/química , Secuencias de Aminoácidos , Sustitución de Aminoácidos , Animales , Dominio Catalítico , Línea Celular , Cristalografía por Rayos X , Activación Enzimática , Humanos , Fosfatos de Inositol/química , Cinética , Mutagénesis Sitio-Dirigida , Resonancia Magnética Nuclear Biomolecular , Fosfolipasa C gamma/genética , Fosfolipasa C gamma/metabolismo , Fosforilación , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Procesamiento Proteico-Postraduccional , Estructura Secundaria de Proteína , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/metabolismo , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/fisiología , Transducción de Señal , Sus scrofa , Termodinámica
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...