Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
J Proteome Res ; 22(3): 768-789, 2023 03 03.
Artículo en Inglés | MEDLINE | ID: mdl-36763541

RESUMEN

Phosphorylation-dependent signal transduction plays an important role in regulating the functions and fate of skeletal muscle cells. Central players in the phospho-signaling network are the protein kinases AKT, S6K, and RSK as part of the PI3K-AKT-mTOR-S6K and RAF-MEK-ERK-RSK pathways. However, despite their functional importance, knowledge about their specific targets is incomplete because these kinases share the same basophilic substrate motif RxRxxp[ST]. To address this, we performed a multifaceted quantitative phosphoproteomics study of skeletal myotubes following kinase inhibition. Our data corroborate a cross talk between AKT and RAF, a negative feedback loop of RSK on ERK, and a putative connection between RSK and PI3K signaling. Altogether, we report a kinase target landscape containing 49 so far unknown target sites. AKT, S6K, and RSK phosphorylate numerous proteins involved in muscle development, integrity, and functions, and signaling converges on factors that are central for the skeletal muscle cytoskeleton. Whereas AKT controls insulin signaling and impinges on GTPase signaling, nuclear signaling is characteristic for RSK. Our data further support a role of RSK in glucose metabolism. Shared targets have functions in RNA maturation, stability, and translation, which suggests that these basophilic kinases establish an intricate signaling network to orchestrate and regulate processes involved in translation.


Asunto(s)
Fosfatidilinositol 3-Quinasas , Proteínas Proto-Oncogénicas c-akt , Fibras Musculares Esqueléticas/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/fisiología , Proteínas Quinasas S6 Ribosómicas 90-kDa , Proteínas Quinasas S6 Ribosómicas 70-kDa
2.
Commun Biol ; 3(1): 253, 2020 05 22.
Artículo en Inglés | MEDLINE | ID: mdl-32444788

RESUMEN

The PI3K/Akt pathway promotes skeletal muscle growth and myogenic differentiation. Although its importance in skeletal muscle biology is well documented, many of its substrates remain to be identified. We here studied PI3K/Akt signaling in contracting skeletal muscle cells by quantitative phosphoproteomics. We identified the extended basophilic phosphosite motif RxRxxp[S/T]xxp[S/T] in various proteins including filamin-C (FLNc). Importantly, this extended motif, located in a unique insert in Ig-like domain 20 of FLNc, is doubly phosphorylated. The protein kinases responsible for this dual-site phosphorylation are Akt and PKCα. Proximity proteomics and interaction analysis identified filamin A-interacting protein 1 (FILIP1) as direct FLNc binding partner. FILIP1 binding induces filamin degradation, thereby negatively regulating its function. Here, dual-site phosphorylation of FLNc not only reduces FILIP1 binding, providing a mechanism to shield FLNc from FILIP1-mediated degradation, but also enables fast dynamics of FLNc necessary for its function as signaling adaptor in cross-striated muscle cells.


Asunto(s)
Proteínas Portadoras/metabolismo , Proteínas del Citoesqueleto/metabolismo , Filaminas/metabolismo , Fibras Musculares Esqueléticas/metabolismo , Fosfoproteínas/metabolismo , Proteoma/metabolismo , Secuencias de Aminoácidos , Células HEK293 , Humanos , Desarrollo de Músculos , Fibras Musculares Esqueléticas/citología , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , Unión Proteica , Proteolisis , Proteoma/análisis , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal
3.
ACS Synth Biol ; 8(7): 1679-1684, 2019 07 19.
Artículo en Inglés | MEDLINE | ID: mdl-31185174

RESUMEN

Subcellular localization of signal molecules is a hallmark in organizing the signaling network. OpEn-Tag is a modular optogenetic endomembrane targeting toolbox that allows alteration of the localization and therefore the activity of signaling processes with the spatiotemporal resolution of optogenetics. OpEn-Tag is a two-component system employing (1) a variety of targeting peptides fused to and thereby dictating the localization of mCherry-labeled cryptochrome 2 binding protein CIBN toward distinct endomembranes and (2) the cytosolic, fluorescence-labeled blue light photoreceptor cryptochrome 2 as a customizable building block that can be fused to proteins of interest. The combination of OpEn-Tag with growth factor stimulation or the use of two membrane anchor sequences allows investigation of multilayered signal transduction processes as demonstrated here for the protein kinase AKT.


Asunto(s)
Proteínas Portadoras/metabolismo , Criptocromos/metabolismo , Transducción de Señal/fisiología , Línea Celular Tumoral , Células HeLa , Humanos , Luz , Optogenética/métodos , Células Fotorreceptoras/metabolismo , Células Fotorreceptoras/fisiología , Proteínas Proto-Oncogénicas c-akt/metabolismo
4.
Commun Biol ; 2: 15, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30652127

RESUMEN

Optogenetic approaches have gathered momentum in precisely modulating and interrogating cellular signalling and gene expression. The use of optogenetics on the outer cell surface to interrogate how cells receive stimuli from their environment, however, has so far not reached its full potential. Here we demonstrate the development of an optogenetically regulated membrane receptor-ligand pair exemplified by the optically responsive interaction of an integrin receptor with the extracellular matrix. The system is based on an integrin engineered with a phytochrome-interacting factor domain (OptoIntegrin) and a red light-switchable phytochrome B-functionalized matrix (OptoMatrix). This optogenetic receptor-ligand pair enables light-inducible and -reversible cell-matrix interaction, as well as the controlled activation of downstream mechanosensory signalling pathways. Pioneering the application of optogenetic switches in the extracellular environment of cells, this OptoMatrix-OptoIntegrin system may serve as a blueprint for rendering matrix-receptor interactions amendable to precise control with light.


Asunto(s)
Matriz Extracelular/metabolismo , Integrina alfaVbeta3/metabolismo , Optogenética/métodos , Arabidopsis/metabolismo , Proteínas de Arabidopsis/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Matriz Extracelular/efectos de la radiación , Células HEK293 , Células HeLa , Humanos , Luz , Células MCF-7 , Fitocromo B/metabolismo , Plásmidos/genética , Conformación Proteica/efectos de la radiación , Transducción de Señal/efectos de la radiación , Transfección
5.
Cell Signal ; 42: 176-183, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-29074139

RESUMEN

Focal adhesion kinase (FAK) integrates signaling from integrins, growth factor receptors and mechanical stress to control cell adhesion, motility, survival and proliferation. Here, we developed a single-component, photo-activatable FAK, termed optoFAK, by using blue light-induced oligomerization of cryptochrome 2 (CRY2) to activate FAK-CRY2 fusion proteins. OptoFAK functions uncoupled from physiological stimuli and activates downstream signaling rapidly and reversibly upon blue light exposure. OptoFAK stimulates SRC creating a positive feedback loop on FAK activation, facilitating phosphorylation of paxillin and p130Cas in adherent cells. In detached cells or in mechanically stressed adherent cells, optoFAK is autophosphorylated upon exposure to blue light, however, downstream signaling is hampered indicating that the accessibility to these substrates is disturbed. OptoFAK may prove to be a useful tool to study the biological function of FAK in growth factor and integrin signaling, tension-mediated focal adhesion maturation or anoikis and could additionally serve as test system for kinase inhibitors.


Asunto(s)
Criptocromos/metabolismo , Retroalimentación Fisiológica , Quinasa 1 de Adhesión Focal/metabolismo , Optogenética/métodos , Transducción de Señal , Adhesión Celular , Proteína Sustrato Asociada a CrK/genética , Proteína Sustrato Asociada a CrK/metabolismo , Criptocromos/genética , Quinasa 1 de Adhesión Focal/genética , Regulación de la Expresión Génica , Células HEK293 , Células HeLa , Humanos , Luz , Paxillin/genética , Paxillin/metabolismo , Fosforilación , Plásmidos/química , Plásmidos/metabolismo , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Estrés Mecánico , Transfección
6.
Sci Adv ; 3(8): e1700475, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28819643

RESUMEN

Spatiotemporal control is a common mechanism that modulates activity and function of signal transducers in the signaling network. We identified acetylation of CNK1 (connector enhancer of kinase suppressor of Ras-1) as a late step in the activation of CNK1 signaling, accompanied with prolonged stimulation of extracellular signal-regulated kinase (ERK). We identified the acetyltransferase CREB (cyclic adenosine 3',5'-monophosphate response element-binding protein)-binding protein and the deacetylase SIRT2 (sirtuin type 2) as novel binding partners of CNK1, modulating the acetylation state of CNK1. Acetylation of CNK1 at position Lys414 located in the pleckstrin homology domain drives membrane localization of CNK1 in growth factor-stimulated cells. Inhibition of ERK signaling abolishes CNK1 acetylation. Cosmic database search identified CNK1 mutants at position Arg426 near the acetylation site in several human tumor types. These mutants show constitutive acetylation and membrane localization. CNK1 mutants substituting Arg426, the acetylation mimetic mutant CNK1-K414Q, and membrane-anchored CNK1 mutants all interact with the protein kinase CRAF and stimulate ERK-dependent cell proliferation and cell migration. In RAS-transformed cells, CNK1 is acetylated and membrane-bound and drives cell proliferation. Thus, growth factor-stimulated ERK signaling induces CNK1 acetylation, and acetylated CNK1 promotes ERK signaling, demonstrating a novel function of CNK1 as positive feedback regulator of the RAF/MEK (mitogen-activated protein kinase kinase)/ERK pathway. In addition, acetylation of CNK1 is an important step in oncogenic signaling, promoting cell proliferation and migration.


Asunto(s)
Membrana Celular/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Transducción de Señal , Quinasas raf/metabolismo , Acetilación , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Técnica del Anticuerpo Fluorescente , Humanos , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Péptidos y Proteínas de Señalización Intracelular/química , Péptidos y Proteínas de Señalización Intracelular/genética , Modelos Biológicos , Mutación , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Transporte de Proteínas , Proteínas ras/metabolismo
7.
Methods Mol Biol ; 1596: 257-270, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28293892

RESUMEN

Optogenetic approaches enable the control of biological processes in a time- and space-resolved manner. These light-based methods are noninvasive and by using light as sole activator minimize side effects in contrast to chemical inducers. Here, we provide a protocol for the targeted control of the activity of protein kinases in mammalian cells based on the photoreceptor cryptochrome 2 (CRY2) of Arabidopsis thaliana and its interaction partner CIB1. Blue light (450 nm)-induced binding of CRY2 to CIB1 allows the recruitment of a chimeric cytosolic protein kinase AKT1 to the plasma membrane accompanied with stimulation of its kinase activity. This protocol comprises the transient and stable implementation of the light-regulated system into mammalian cells and its stimulation by blue light-emitting diodes (450 nm) irradiation as well as analysis of the light-activated AKT1.


Asunto(s)
Proteínas de Arabidopsis/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Criptocromos/metabolismo , Proteínas Quinasas/metabolismo , Animales , Arabidopsis/metabolismo , Membrana Celular/metabolismo , Células HEK293 , Humanos , Luz , Células MCF-7 , Mamíferos/metabolismo , Optogenética/métodos , Canales de Potasio/metabolismo , Unión Proteica/fisiología , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/metabolismo
8.
Mol Cell Proteomics ; 16(3): 346-367, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28028127

RESUMEN

The Z-disc is a protein-rich structure critically important for the development and integrity of myofibrils, which are the contractile organelles of cross-striated muscle cells. We here used mouse C2C12 myoblast, which were differentiated into myotubes, followed by electrical pulse stimulation (EPS) to generate contracting myotubes comprising mature Z-discs. Using a quantitative proteomics approach, we found significant changes in the relative abundance of 387 proteins in myoblasts versus differentiated myotubes, reflecting the drastic phenotypic conversion of these cells during myogenesis. Interestingly, EPS of differentiated myotubes to induce Z-disc assembly and maturation resulted in increased levels of proteins involved in ATP synthesis, presumably to fulfill the higher energy demand of contracting myotubes. Because an important role of the Z-disc for signal integration and transduction was recently suggested, its precise phosphorylation landscape further warranted in-depth analysis. We therefore established, by global phosphoproteomics of EPS-treated contracting myotubes, a comprehensive site-resolved protein phosphorylation map of the Z-disc and found that it is a phosphorylation hotspot in skeletal myocytes, underscoring its functions in signaling and disease-related processes. In an illustrative fashion, we analyzed the actin-binding multiadaptor protein filamin C (FLNc), which is essential for Z-disc assembly and maintenance, and found that PKCα phosphorylation at distinct serine residues in its hinge 2 region prevents its cleavage at an adjacent tyrosine residue by calpain 1. Fluorescence recovery after photobleaching experiments indicated that this phosphorylation modulates FLNc dynamics. Moreover, FLNc lacking the cleaved Ig-like domain 24 exhibited remarkably fast kinetics and exceedingly high mobility. Our data set provides research community resource for further identification of kinase-mediated changes in myofibrillar protein interactions, kinetics, and mobility that will greatly advance our understanding of Z-disc dynamics and signaling.


Asunto(s)
Fibras Musculares Esqueléticas/metabolismo , Mioblastos/citología , Proteína Quinasa C/metabolismo , Proteómica/métodos , Sarcómeros/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Diferenciación Celular , Línea Celular , Estimulación Eléctrica , Filaminas/metabolismo , Ratones , Mioblastos/metabolismo , Fosforilación , Mapas de Interacción de Proteínas
9.
Biochim Biophys Acta Mol Cell Res ; 1864(1): 89-100, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-27769899

RESUMEN

Scaffold proteins are hubs for the coordination of intracellular signaling networks. The scaffold protein CNK1 promotes several signal transduction pathway. Here we demonstrate that sterile motif alpha (SAM) domain-dependent oligomerization of CNK1 stimulates CNK1-mediated signaling in growth factor-stimulated cells. We identified Ser22 located within the SAM domain as AKT-dependent phosphorylation site triggering CNK1 oligomerization. Oligomeric CNK1 increased the affinity for active AKT indicating a positive AKT feedback mechanism. A CNK1 mutant lacking the SAM domain and the phosphorylation-defective mutant CNK1S22A antagonizes oligomerization and prevents CNK1-driven cell proliferation and matrix metalloproteinase 14 promoter activation. The phosphomimetic mutant CNK1S22D constitutively oligomerizes and stimulates CNK1 downstream signaling. Searching the COSMIC database revealed Ser22 as putative target for oncogenic activation of CNK1. Like the phosphomimetic mutant CNK1S22D, the oncogenic mutant CNK1S22F forms clusters in serum-starved cells comparable to clusters of CNK1 in growth factor-stimulated cells. CNK1 clusters induced by activating Ser22 mutants correlate with enhanced cell invasion and binding to and activation of ADP ribosylation factor 1 associated with tumor formation. Mutational analysis indicate that EGF-triggered phosphorylation of Thr8 within the SAM domain prevents AKT binding and antagonizes CNK1-mediated AKT signaling. Our findings reveal SAM domain-dependent oligomerization by AKT as switch for CNK1 activation.


Asunto(s)
Retroalimentación Fisiológica , Regulación Neoplásica de la Expresión Génica , Péptidos y Proteínas de Señalización Intracelular/genética , Proteínas Proto-Oncogénicas c-akt/genética , Motivo alfa Estéril , Adhesión Celular , Movimiento Celular , Proliferación Celular , Bases de Datos Genéticas , Células HEK293 , Células HeLa , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Metaloproteinasa 14 de la Matriz/genética , Metaloproteinasa 14 de la Matriz/metabolismo , Imitación Molecular , Mutación , Fosforilación , Regiones Promotoras Genéticas , Multimerización de Proteína , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal
10.
Sci Rep ; 6: 38155, 2016 11 30.
Artículo en Inglés | MEDLINE | ID: mdl-27901111

RESUMEN

Scaffold proteins such as the multidomain protein CNK1 orchestrate the signalling network by integrating and controlling the underlying pathways. Using an optogenetic approach to stimulate CNK1 uncoupled from upstream effectors, we identified selective clusters of CNK1 that either stimulate RAF-MEK-ERK or AKT signalling depending on the light intensity applied. OptoCNK1 implemented in MCF7 cells induces differentiation at low light intensity stimulating ERK activity whereas stimulation of AKT signalling by higher light intensity promotes cell proliferation. CNK1 clustering in response to increasing EGF concentrations revealed that CNK1 binds to RAF correlating with ERK activation at low EGF dose. At higher EGF dose active AKT binds to CNK1 and phosphorylates and inhibits RAF. Knockdown of CNK1 protects CNK1 from this AKT/RAF crosstalk. In C2 skeletal muscle cells CNK1 expression is induced with the onset of differentiation. Hence, AKT-bound CNK1 counteracts ERK stimulation in differentiated but not in proliferating cells. Ectopically expressed CNK1 facilitates C2 cell differentiation and knockdown of CNK1 impaired the transcriptional network underlying C2 cell differentiation. Thus, CNK1 expression, CNK1 clustering and the thereto related differential signalling processes decide on proliferation and differentiation in a cell type- and cell stage-dependent manner by orchestrating AKT and RAF signalling.


Asunto(s)
Diferenciación Celular/genética , Proliferación Celular/genética , Péptidos y Proteínas de Señalización Intracelular/genética , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-raf/genética , Animales , Línea Celular , Células HEK293 , Células HeLa , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Células MCF-7 , Ratones , Optogenética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas c-raf/metabolismo , Interferencia de ARN , Transducción de Señal/genética
11.
Sci Rep ; 6: 23713, 2016 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-27025703

RESUMEN

Here, we applied optoRAF, an optogenetic tool for light-controlled clustering and activation of RAF proteins that mimics the natural occurring RAS-mediated dimerization. This versatile tool allows studying the effect on BRAF and CRAF homodimer- as well as heterodimer-induced RAF signaling. Vemurafenib and dabrafenib are two clinically approved inhibitors for BRAF that efficiently suppress the kinase activity of oncogenic BRAF (V600E). However in wild-type BRAF expressing cells, BRAF inhibitors can exert paradoxical activation of wild-type CRAF. Using optoRAF, vemurafenib was identified as paradoxical activator of BRAF and CRAF homo- and heterodimers. Dabrafenib enhanced activity of light-stimulated CRAF at low dose and inhibited CRAF signaling at high dose. Moreover, dabrafenib increased the protein level of CRAF proteins but not of BRAF proteins. Increased CRAF levels correlate with elevated RAF signaling in a dabrafenib-dependent manner, independent of light activation.


Asunto(s)
Antineoplásicos/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas B-raf/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-raf/antagonistas & inhibidores , Ensayos de Selección de Medicamentos Antitumorales , Inducción Enzimática/efectos de la radiación , Células HEK293 , Células HeLa , Humanos , Imidazoles/farmacología , Indoles/farmacología , Optogenética , Oximas/farmacología , Fosforilación , Procesamiento Proteico-Postraduccional , Proteínas Proto-Oncogénicas B-raf/genética , Proteínas Proto-Oncogénicas B-raf/metabolismo , Proteínas Proto-Oncogénicas c-raf/genética , Proteínas Proto-Oncogénicas c-raf/metabolismo , Transducción de Señal , Sulfonamidas/farmacología , Vemurafenib
12.
Biochim Biophys Acta ; 1854(6): 632-40, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25219547

RESUMEN

The prostate-specific G-protein-coupled receptor 1 (PSGR1) is an olfactory receptor specifically expressed in the prostate gland. PSGR1 expression is elevated both in benign prostatic hyperplasia tissue and in prostate cancer. Stimulation of PSGR1 by the odorant ß-ionone leads to an increase in the intracellular Ca(2+) concentration, activation of mitogen-activated protein (MAP) kinases and a decrease in prostate cancer cell proliferation. To further extend our knowledge about PSGR1 signaling in prostate cancer cells, we performed a quantitative phosphoproteomics study using stable isotope labeling by amino acids in cell culture and mass spectrometry. We report 51 differentially regulated phosphorylation sites in 24 proteins with functions in cytoskeletal remodeling, signaling and ion transport. Activation of PSGR1 evoked an increase in intracellular pH mediated by the sodium/hydrogen exchanger NHE1. Furthermore, we report the protein tyrosine kinase Pyk2 as a central effector of PSGR1 signaling cascades in LNCaP cells. Our data show that phosphorylation of p38 MAP kinase is triggered by Pyk2. In addition, we confirmed dephosphorylation of the tumor suppressor protein N-myc downstream regulated gene 1 (NDRG1) at Ser330 downstream of Pyk2. Since NDRG1 impacts oncogenic signaling pathways interfering with tumor progression, we suggest that the Pyk2-NDRG1 axis is possibly involved in conveying the anti-proliferative effect of ß-ionone in prostate cancer cells. This article is part of a Special Issue entitled: Medical Proteomics.


Asunto(s)
Quinasa 2 de Adhesión Focal/metabolismo , Sistema de Señalización de MAP Quinasas , Proteínas de Neoplasias/metabolismo , Fosfoproteínas/metabolismo , Neoplasias de la Próstata/metabolismo , Receptores Odorantes/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Quinasa 2 de Adhesión Focal/genética , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Masculino , Proteínas de Neoplasias/genética , Norisoprenoides/farmacología , Fosfoproteínas/genética , Fosforilación/efectos de los fármacos , Fosforilación/genética , Neoplasias de la Próstata/genética , Receptores Odorantes/genética , Proteínas Quinasas p38 Activadas por Mitógenos/genética , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
13.
ChemMedChem ; 9(7): 1458-62, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24668962

RESUMEN

PDZ (PSD-95, Dlg, ZO-1) domains are ubiquitous interaction modules that are involved in many cellular signal transduction pathways. Interference with PDZ-mediated protein-protein interactions has important implications in disease-related signaling processes. For this reason, PDZ domains have gained attention as potential targets for inhibitor design and, in the long run, drug development. Herein we report the development of small molecules to probe the function of the PDZ domain from human AF6 (ALL1-fused gene from chromosome 6), which is an essential component of cell-cell junctions. These compounds bind to AF6 PDZ with substantially higher affinity than the peptide (Ile-Gln-Ser-Val-Glu-Val) derived from its natural ligand, EphB2. In intact cells, the compounds inhibit the AF6-Bcr interaction and interfere with epidermal growth factor (EGF)-dependent signaling.


Asunto(s)
Cinesinas/antagonistas & inhibidores , Miosinas/antagonistas & inhibidores , Bibliotecas de Moléculas Pequeñas/química , Secuencia de Aminoácidos , Sitios de Unión , Humanos , Cinesinas/metabolismo , Ligandos , Simulación del Acoplamiento Molecular , Miosinas/metabolismo , Dominios PDZ , Péptidos/química , Péptidos/metabolismo , Dominios y Motivos de Interacción de Proteínas , Receptor EphB2/química , Transducción de Señal/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas/metabolismo , Relación Estructura-Actividad
14.
ACS Synth Biol ; 3(5): 280-5, 2014 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-24090449

RESUMEN

Light-dependent dimerization is the basis for recently developed noninvasive optogenetic tools. Here we present a novel tool combining optogenetics with the control of protein kinase activity to investigate signal transduction pathways. Mediated by Arabidopsis thaliana photoreceptor cryptochrome 2, we activated the protein kinase C-RAF by blue light-dependent dimerization, allowing for decoupling from upstream signaling events induced by surface receptors. The activation by light is fast, reversible, and not only time but also dose dependent as monitored by phosphorylation of ERK1/2. Additionally, light-activated C-RAF controls serum response factor-mediated gene expression. Light-induced heterodimerization of C-RAF with a kinase-dead mutant of B-RAF demonstrates the enhancing role of B-RAF as a scaffold for C-RAF activity, which leads to the paradoxical activation of C-RAF found in human cancers. This optogenetic tool enables reversible control of protein kinase activity in signal duration and strength. These properties can help to shed light onto downstream signaling processes of protein kinases in living cells.


Asunto(s)
Optogenética/métodos , Proteínas Quinasas , Transducción de Señal , Biología Sintética/métodos , Proteínas de Arabidopsis , Criptocromos , Células HEK293 , Humanos , Fosforilación/genética , Fosforilación/efectos de la radiación , Proteínas Quinasas/genética , Proteínas Quinasas/metabolismo , Proteínas Quinasas/efectos de la radiación , Multimerización de Proteína/genética , Multimerización de Proteína/efectos de la radiación , Proteínas Proto-Oncogénicas c-raf/genética , Proteínas Proto-Oncogénicas c-raf/metabolismo , Proteínas Proto-Oncogénicas c-raf/efectos de la radiación , Transducción de Señal/genética , Transducción de Señal/efectos de la radiación
15.
Sci Rep ; 3: 2716, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24056365

RESUMEN

The precise manipulation of growth factor signaling is central to the progress of tissue engineering. Methods for direct time-resolved activation of signaling pathways through controlled receptor dimerization have been reported; however, these suffer from the risks associated with gene transfer. Here we present an alternative gene transfer-free approach in the form of a protein switch featuring pharmacologically controlled ON-OFF regulation of growth factor activity. The reversible operation of the switch enables stimulation of target processes within a defined period of time. The protein switch provides a means for both studying and manipulating signaling processes, and is thus believed to be a valuable tool for basic research as well as tissue engineering and biomedical applications.


Asunto(s)
Transducción de Señal , Factor A de Crecimiento Endotelial Vascular/metabolismo , Secuencia de Aminoácidos , Movimiento Celular , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Datos de Secuencia Molecular , Multimerización de Proteína , Proteínas Recombinantes/metabolismo , Factor A de Crecimiento Endotelial Vascular/química , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
16.
Biochim Biophys Acta ; 1813(11): 1971-7, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21320536

RESUMEN

FoxO transcription factors mediate anti-proliferative and pro-apoptotic signals and act as tumor suppressors in cancer. Posttranslational modifications including phosphorylation and acetylation regulate FoxO activity by a cytoplasmic-nuclear shuttle mechanism. Scaffold proteins coordinating signaling pathways in time and space play a critical role in this process. CNK1 acts as a scaffold protein in several signaling pathways controlling the function of FoxO proteins. An understanding of CNK1 and other scaffolds in the FoxO signaling network will provide insights how to release the tumor suppressor function of FoxO as a possibility to block oncogenic pathways. This article is part of a Special Issue entitled: P13K-AKT-FoxO axis in cancer and aging.


Asunto(s)
Factores de Transcripción Forkhead/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Animales , Apoptosis , Proteína Forkhead Box O1 , Humanos , Modelos Biológicos
17.
Mol Cancer Res ; 8(3): 395-406, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20197385

RESUMEN

Hallmarks of cancer cells are uncontrolled proliferation, evasion of apoptosis, angiogenesis, cell invasion, and metastasis, which are driven by oncogenic activation of signaling pathways. Herein, we identify the scaffold protein CNK1 as a mediator of oncogenic signaling that promotes invasion in human breast cancer and cervical cancer cells. Downregulation of CNK1 diminishes the invasiveness of cancer cells and correlates with reduced expression of matrix metalloproteinase 9 (MMP-9) and membrane-type 1 MMP (MT1-MMP). Ectopic expression of CNK1 elevates MT1-MMP promoter activity in a NF-kappaB-dependent manner. Moreover, CNK1 cooperates with the NF-kappaB pathway, but not with the extracellular signal-regulated protein kinase pathway, to promote cell invasion. Mechanistically, CNK1 regulates the alternative branch of the NF-kappaB pathway because knockdown of CNK1 interferes with processing of NF-kappaB2 p100 to p52 and its localization to the nucleus. In agreement with this, the invasion of CNK1-depleted cells is less sensitive to RelB downregulation compared with the invasion of control cells. Moreover, CNK1-dependent MT1-MMP promoter activation is blocked by RelB siRNA. Thus, CNK1 is an essential mediator of an oncogenic pathway involved in invasion of breast and cervical cancer cells and is therefore a putative target for cancer therapy.


Asunto(s)
Regulación Neoplásica de la Expresión Génica/genética , Péptidos y Proteínas de Señalización Intracelular/genética , FN-kappa B/genética , Invasividad Neoplásica/genética , Transducción de Señal/genética , Transporte Activo de Núcleo Celular/genética , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Carcinoma/genética , Carcinoma/metabolismo , Regulación hacia Abajo/genética , Proteínas de la Matriz Extracelular/genética , Proteínas de la Matriz Extracelular/metabolismo , Femenino , Células HeLa , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Metaloproteinasa 1 de la Matriz/genética , Metaloproteinasa 1 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/genética , Metaloproteinasa 9 de la Matriz/metabolismo , FN-kappa B/metabolismo , Subunidad p52 de NF-kappa B/genética , Subunidad p52 de NF-kappa B/metabolismo , ARN Interferente Pequeño/genética , Factor de Transcripción ReIB/genética , Factor de Transcripción ReIB/metabolismo , Neoplasias del Cuello Uterino/genética , Neoplasias del Cuello Uterino/metabolismo
18.
Mol Cancer Res ; 7(10): 1635-44, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19825990

RESUMEN

The noncoding RNA miR-125b has been described to reduce ErbB2 protein expression as well as proliferation and migration of cancer cell lines. As additional target of miR-125b, we identified the c-raf-1 mRNA by sequence analysis. We designed a short hairpin-looped oligodeoxynucleotide (ODN) targeted to the same 3' untranslated region of c-raf-1 mRNA as miR-125b. The fully complementary ODN antisense strand is linked to a second strand constituting a partially double-stranded structure of the ODN. Transfection of the c-raf-1-specific ODN (ODN-Raf) in a breast cancer cell line reduced the protein levels of C-Raf, ErbB2, and their downstream effector cyclin D1 similar to miR-125b. MiR-125b as well as ODN-Raf showed no effect on the c-raf-1 mRNA level in contrast to small interfering RNA. Unlike miR-125b, ODN-Raf induced a cytopathic effect. This may be explained by the structural properties of ODN-Raf, which can form G-tetrads. Thus, the short hairpin-looped ODN-Raf, targeting the same region of c-raf-1 as miR-125b, is a multifunctional molecule reducing the expression of oncoproteins and stimulating cell death. Both features may be useful to interfere with tumor growth.


Asunto(s)
Neoplasias de la Mama/genética , Carcinoma/genética , MicroARNs/genética , Oligodesoxirribonucleótidos Antisentido/genética , Proteínas Proto-Oncogénicas c-raf/antagonistas & inhibidores , Transfección/métodos , Neoplasias de la Mama/fisiopatología , Neoplasias de la Mama/terapia , Carcinoma/fisiopatología , Carcinoma/terapia , Muerte Celular/genética , Línea Celular Tumoral , Proliferación Celular , Supervivencia Celular/genética , Regulación hacia Abajo/genética , Femenino , Regulación Neoplásica de la Expresión Génica/genética , Terapia Genética/métodos , Humanos , Secuencias Invertidas Repetidas/genética , Oligodesoxirribonucleótidos Antisentido/síntesis química , Proteínas Proto-Oncogénicas c-raf/genética , Proteínas Proto-Oncogénicas c-raf/metabolismo , ARN Mensajero/metabolismo , Receptor ErbB-2/genética
19.
Mol Cell Biol ; 28(2): 642-55, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18039857

RESUMEN

c-Src tyrosine kinase controls proliferation, cell adhesion, and cell migration and is highly regulated. A novel regulatory mechanism to control c-Src function that has recently been identified involves the C-terminal amino acid sequence Gly-Glu-Asn-Leu (GENL) of c-Src as ligand for PDZ domains. Herein, we determined the biological relevance of this c-Src regulation in human breast epithelial cells. The intact GENL sequence maintained c-Src in an inactive state in starved cells and restricted c-Src functions that might lead to metastatic transformation under normal growth conditions. c-Src with a C-terminal Leu/Ala mutation in GENL (Src-A) promoted the activation and translocation of cortactin and focal adhesion kinase and increased the motility and persistence of cell migration on the basement membrane. Src-A promoted increased extracellular proteolytic activity, and in acinar cultures, it led to the escape of cells through the basement membrane into the surrounding matrix. We ascribe the regulatory function of C-terminal Leu to the role of GENL in modulating c-Src activity downstream of cell matrix adhesion. We propose that the C terminus of c-Src via its GENL sequence presents a mechanism that restricts c-Src in epithelia and prevents progression toward an invasive phenotype.


Asunto(s)
Movimiento Celular , Células Epiteliales/citología , Células Epiteliales/metabolismo , Dominios PDZ , Proteínas Proto-Oncogénicas pp60(c-src)/química , Proteínas Proto-Oncogénicas pp60(c-src)/metabolismo , Sitios de Unión , Células de la Médula Ósea/citología , Células de la Médula Ósea/enzimología , Cadherinas/metabolismo , Adhesión Celular , Línea Celular , Polaridad Celular , Forma de la Célula , Transformación Celular Neoplásica , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Regulación Enzimológica de la Expresión Génica , Humanos , Ligandos , Invasividad Neoplásica , Fosforilación , Proteínas Proto-Oncogénicas pp60(c-src)/genética , Cicatrización de Heridas
20.
Chembiochem ; 8(18): 2302-7, 2007 Dec 17.
Artículo en Inglés | MEDLINE | ID: mdl-17973281

RESUMEN

Transient macromolecular complexes are often formed by protein-protein interaction domains (e.g., PDZ, SH2, SH3, WW), which are often regulated (positively or negatively) by phosphorylation. To address the in vitro analysis of PDZ domain regulation by such phosphorylation, we improved the inverted peptide method. This method is based on standard SPOT synthesis, followed by inversion of the peptide under acidic conditions to generate the free C termini necessary for PDZ domain ligand recognition. The benefit of the newly introduced acidic conditions is the preservation of the incorporated phosphate group during peptide synthesis. Furthermore, the improved method is more robust and shows an increased signal-to-noise ratio. As representative examples, we used the AF6, ERBIN, and SNA1 (alpha-1-syntrophin) PDZ domains to analyze the influence of ligand-position-dependent phosphorylation. We could clearly demonstrate severe down-regulation by phosphorylation of the PDZ ligand position -2 (<50 %) and slightly less at position -1 ( approximately 50 %). These results are specific and reproducible for all three PDZ domains. Finally, we confirmed the influence of negative regulation by using the protein kinase BCR as the AF6 PDZ domain ligand. For the first time, this approach allows the SPOT synthesis technique to be used to screen large libraries of phosphorylated peptides in vitro. This should ultimately help in the identification of phosphorylation-dependent regulation mechanisms in vivo.


Asunto(s)
Análisis por Micromatrices/métodos , Péptidos/síntesis química , Regulación de la Expresión Génica , Estructura Molecular , Biblioteca de Péptidos , Fosforilación , Unión Proteica , Estructura Terciaria de Proteína
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA