Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
ACS Chem Biol ; 19(2): 497-505, 2024 02 16.
Artículo en Inglés | MEDLINE | ID: mdl-38270585

RESUMEN

The chemogenetic control of cellular protein stability using degron tags is a powerful experimental strategy in biomedical research. However, this technique requires permanent fusion of the degron to a target protein, which may interfere with the proper function of the protein. Here, we report a peptide fragment from the carboxyl terminus of ubiquitin as a cleavable linker that exhibits the slow but efficient cleavage of a degron tag via cellular deubiquitinating enzymes (DUBs). We designed a fusion protein consisting of a cleavable linker and a destabilizing domain (DD), which conditionally controls the expression and release of a target protein in a ligand-induced state, allowing the free unmodified protein to perform its function. Insertion of an AGIA epitope at the carboxyl terminus of the linker made space for the DUBs to access the site to assist the cleavage reaction when the amino terminus of the target protein caused steric hindrance. The developed system, termed a cleavable degron using ubiquitin-derived linkers (c-DUB), provides robust and tunable regulation of target proteins in their native forms. The c-DUB system is a useful tool for the regulation of proteins that have terminal sites that are essential for the proper localization and function. In addition, a mechanistic investigation using proximity labeling showed that DUBs associate with the refolded DD to reverse ubiquitination, suggesting a cellular surveillance system for distinguishing the refolded DD from misfolded proteins. The c-DUB method may benefit from this machinery so that DUBs subsequently cleave the neighboring linker.


Asunto(s)
Degrones , Ubiquitina , Ubiquitina/metabolismo , Proteínas/metabolismo , Ubiquitinación , Péptidos/metabolismo
2.
ACS Chem Biol ; 17(8): 2024-2030, 2022 08 19.
Artículo en Inglés | MEDLINE | ID: mdl-35839076

RESUMEN

cAMP is a ubiquitous second messenger with many functions in diverse organisms. Current cAMP sensors, including Föster resonance energy transfer (FRET)-based and single-wavelength-based sensors, allow for real time visualization of this small molecule in cultured cells and in some cases in vivo. Nonetheless the observation of cAMP in living animals is still difficult, typically requiring specialized microscopes and ex vivo tissue processing. Here we used ligand-dependent protein stabilization to create a new cAMP sensor. This sensor allows specific and sensitive detection of cAMP in living zebrafish embryos, which may enable new understanding of the functions of cAMP in living vertebrates.


Asunto(s)
Técnicas Biosensibles , AMP Cíclico , Animales , AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Transferencia Resonante de Energía de Fluorescencia , Ligandos , Pez Cebra/metabolismo
3.
Science ; 372(6537)2021 04 02.
Artículo en Inglés | MEDLINE | ID: mdl-33795428

RESUMEN

T cell exhaustion limits immune responses against cancer and is a major cause of resistance to chimeric antigen receptor (CAR)-T cell therapeutics. Using murine xenograft models and an in vitro model wherein tonic CAR signaling induces hallmark features of exhaustion, we tested the effect of transient cessation of receptor signaling, or rest, on the development and maintenance of exhaustion. Induction of rest through enforced down-regulation of the CAR protein using a drug-regulatable system or treatment with the multikinase inhibitor dasatinib resulted in the acquisition of a memory-like phenotype, global transcriptional and epigenetic reprogramming, and restored antitumor functionality in exhausted CAR-T cells. This work demonstrates that rest can enhance CAR-T cell efficacy by preventing or reversing exhaustion, and it challenges the notion that exhaustion is an epigenetically fixed state.


Asunto(s)
Dasatinib/farmacología , Epigénesis Genética , Inmunoterapia Adoptiva , Receptores Quiméricos de Antígenos/metabolismo , Linfocitos T/inmunología , Animales , Línea Celular Tumoral , Citotoxicidad Inmunológica , Regulación hacia Abajo , Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Epigenoma , Femenino , Factor Nuclear 1-alfa del Hepatocito/metabolismo , Proteínas del Grupo de Alta Movilidad/metabolismo , Humanos , Memoria Inmunológica , Activación de Linfocitos , Factor de Unión 1 al Potenciador Linfoide/metabolismo , Masculino , Ratones , Neoplasias Experimentales/terapia , Dominios Proteicos , Estabilidad Proteica , Receptores Quiméricos de Antígenos/química , Receptores Quiméricos de Antígenos/inmunología , Transducción de Señal , Linfocitos T/metabolismo , Transcripción Genética , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Nat Commun ; 7: 11689, 2016 05 27.
Artículo en Inglés | MEDLINE | ID: mdl-27229621

RESUMEN

The accumulation of protein aggregates is a common pathological hallmark of many neurodegenerative diseases. However, we do not fully understand how aggregates are formed or the complex network of chaperones, proteasomes and other regulatory factors involved in their clearance. Here, we report a chemically controllable fluorescent protein that enables us to rapidly produce small aggregates inside living cells on the order of seconds, as well as monitor the movement and coalescence of individual aggregates into larger structures. This method can be applied to diverse experimental systems, including live animals, and may prove valuable for understanding cellular responses and diseases associated with protein aggregates.


Asunto(s)
Proteínas Fluorescentes Verdes/metabolismo , Agregado de Proteínas , Agregación Patológica de Proteínas , Proteína 1A de Unión a Tacrolimus/metabolismo , Animales , Sitios de Unión/genética , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Línea Celular Tumoral , Proteínas Fluorescentes Verdes/química , Proteínas Fluorescentes Verdes/genética , Células HEK293 , Humanos , Ratones , Microscopía Confocal , Microscopía Fluorescente , Células 3T3 NIH , Proteína 1A de Unión a Tacrolimus/química , Proteína 1A de Unión a Tacrolimus/genética , Imagen de Lapso de Tiempo/métodos
5.
Mol Syst Biol ; 11(4): 804, 2015 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-25908733

RESUMEN

Numerous molecular components have been identified that regulate the directed migration of eukaryotic cells toward sources of chemoattractant. However, how the components of this system are wired together to coordinate multiple aspects of the response, such as directionality, speed, and sensitivity to stimulus, remains poorly understood. Here we developed a method to shape chemoattractant gradients optically and analyze cellular chemotaxis responses of hundreds of living cells per well in 96-well format by measuring speed changes and directional accuracy. We then systematically characterized migration and chemotaxis phenotypes for 285 siRNA perturbations. A key finding was that the G-protein Giα subunit selectively controls the direction of migration while the receptor and Gß subunit proportionally control both speed and direction. Furthermore, we demonstrate that neutrophils chemotax persistently in response to gradients of fMLF but only transiently in response to gradients of ATP. The method we introduce is applicable for diverse chemical cues and systematic perturbations, can be used to measure multiple cell migration and signaling parameters, and is compatible with low- and high-resolution fluorescence microscopy.


Asunto(s)
Quimiotaxis , Rayos Ultravioleta , Adenosina Trifosfato/farmacología , Línea Celular Tumoral , Factores Quimiotácticos/farmacología , Quimiotaxis/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Diseño de Equipo , Fluoresceínas/análisis , Colorantes Fluorescentes/análisis , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/fisiología , Humanos , Procesamiento de Imagen Asistido por Computador , Leucemia Mieloide Aguda/patología , Microscopía Fluorescente/métodos , N-Formilmetionina Leucil-Fenilalanina/farmacología , Proteínas de Neoplasias/fisiología , Neutrófilos/citología , Neutrófilos/efectos de los fármacos , Fotoquímica , ARN Interferente Pequeño/farmacología , Imagen de Lapso de Tiempo
6.
J Clin Invest ; 123(8): 3600-13, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23867624

RESUMEN

Dysfunctional bone morphogenetic protein receptor-2 (BMPR2) signaling is implicated in the pathogenesis of pulmonary arterial hypertension (PAH). We used a transcriptional high-throughput luciferase reporter assay to screen 3,756 FDA-approved drugs and bioactive compounds for induction of BMPR2 signaling. The best response was achieved with FK506 (tacrolimus), via a dual mechanism of action as a calcineurin inhibitor that also binds FK-binding protein-12 (FKBP12), a repressor of BMP signaling. FK506 released FKBP12 from type I receptors activin receptor-like kinase 1 (ALK1), ALK2, and ALK3 and activated downstream SMAD1/5 and MAPK signaling and ID1 gene regulation in a manner superior to the calcineurin inhibitor cyclosporine and the FKBP12 ligand rapamycin. In pulmonary artery endothelial cells (ECs) from patients with idiopathic PAH, low-dose FK506 reversed dysfunctional BMPR2 signaling. In mice with conditional Bmpr2 deletion in ECs, low-dose FK506 prevented exaggerated chronic hypoxic PAH associated with induction of EC targets of BMP signaling, such as apelin. Low-dose FK506 also reversed severe PAH in rats with medial hypertrophy following monocrotaline and in rats with neointima formation following VEGF receptor blockade and chronic hypoxia. Our studies indicate that low-dose FK506 could be useful in the treatment of PAH.


Asunto(s)
Receptores de Proteínas Morfogenéticas Óseas de Tipo II/metabolismo , Células Endoteliales/fisiología , Hipertensión Pulmonar/tratamiento farmacológico , Tacrolimus/farmacología , Animales , Apoptosis , Proteína Morfogenética Ósea 4/fisiología , Receptores de Proteínas Morfogenéticas Óseas de Tipo II/genética , Hipoxia de la Célula , Línea Celular Tumoral , Proliferación Celular , Células Endoteliales/efectos de los fármacos , Endotelio Vascular/patología , Endotelio Vascular/fisiopatología , Ensayos Analíticos de Alto Rendimiento , Humanos , Hipertensión Pulmonar/metabolismo , Hipertensión Pulmonar/patología , Proteína 1 Inhibidora de la Diferenciación/genética , Proteína 1 Inhibidora de la Diferenciación/metabolismo , Masculino , Ratones , Ratones Noqueados , Microvasos/patología , Neointima/tratamiento farmacológico , Neointima/metabolismo , Neointima/patología , Arteria Pulmonar/patología , Ratas , Ratas Sprague-Dawley , Transducción de Señal , Proteínas Smad/metabolismo , Proteína 1A de Unión a Tacrolimus/metabolismo
7.
Cancer Cell ; 24(1): 30-44, 2013 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-23770012

RESUMEN

Most deaths from breast cancer result from tumor recurrence, but mechanisms underlying tumor relapse are largely unknown. We now report that Par-4 is downregulated during tumor recurrence and that Par-4 downregulation is necessary and sufficient to promote recurrence. Tumor cells with low Par-4 expression survive therapy by evading a program of Par-4-dependent multinucleation and apoptosis that is otherwise engaged following treatment. Low Par-4 expression is associated with poor response to neoadjuvant chemotherapy and an increased risk of relapse in patients with breast cancer, and Par-4 is downregulated in residual tumor cells that survive neoadjuvant chemotherapy. Our findings identify Par-4-induced multinucleation as a mechanism of cell death in oncogene-addicted cells and establish Par-4 as a negative regulator of breast cancer recurrence.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/fisiología , Neoplasias de la Mama/etiología , Recurrencia Local de Neoplasia/etiología , Animales , Proteínas Reguladoras de la Apoptosis/antagonistas & inhibidores , Neoplasias de la Mama/tratamiento farmacológico , Proteínas Quinasas Dependientes de Calcio-Calmodulina/fisiología , Miosinas Cardíacas/metabolismo , Proteínas Quinasas Asociadas a Muerte Celular , Regulación hacia Abajo , Femenino , Humanos , Ratones , Cadenas Ligeras de Miosina/metabolismo , Fosforilación , Receptor ErbB-2/análisis , Proteína p53 Supresora de Tumor/fisiología
8.
Proc Natl Acad Sci U S A ; 110(21): 8567-72, 2013 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-23650381

RESUMEN

Interactions among neighboring cells underpin many physiological processes ranging from early development to immune responses. When these interactions do not function properly, numerous pathologies, including infection and cancer, can result. Molecular imaging technologies, especially optical imaging, are uniquely suited to illuminate complex cellular interactions within the context of living tissues in the body. However, no tools yet exist that allow the detection of microscopic events, such as two cells coming into close proximity, on a global, whole-animal scale. We report here a broadly applicable, longitudinal strategy for probing interactions among cells in living subjects. This approach relies on the generation of bioluminescent light when two distinct cell populations come into close proximity, with the intensity of the optical signal correlating with relative cellular location. We demonstrate the ability of this reporter strategy to gauge cell-cell proximity in culture models in vitro and then evaluate this approach for imaging tumor-immune cell interactions using a murine breast cancer model. In these studies, our imaging strategy enabled the facile visualization of features that are otherwise difficult to observe with conventional imaging techniques, including detection of micrometastatic lesions and potential sites of tumor immunosurveillance. This proximity reporter will facilitate probing of numerous types of cell-cell interactions and will stimulate the development of similar techniques to detect rare events and pathological processes in live animals.


Asunto(s)
Comunicación Celular/inmunología , Genes Reporteros , Vigilancia Inmunológica , Neoplasias Mamarias Experimentales/inmunología , Modelos Biológicos , Animales , Línea Celular Tumoral , Femenino , Células HEK293 , Humanos , Neoplasias Mamarias Experimentales/genética , Neoplasias Mamarias Experimentales/patología
9.
Cell Rep ; 2(1): 89-100, 2012 Jul 26.
Artículo en Inglés | MEDLINE | ID: mdl-22840400

RESUMEN

In neurons, transmembrane proteins are targeted to dendrites in vesicles that traffic solely within the somatodendritic compartment. How these vesicles are retained within the somatodendritic domain is unknown. Here, we use a novel pulse-chase system, which allows synchronous release of exogenous transmembrane proteins from the endoplasmic reticulum to follow movements of post-Golgi transport vesicles. Surprisingly, we found that post-Golgi vesicles carrying dendritic proteins were equally likely to enter axons and dendrites. However, once such vesicles entered the axon, they very rarely moved beyond the axon initial segment but instead either halted or reversed direction in an actin and Myosin Va-dependent manner. In contrast, vesicles carrying either an axonal or a nonspecifically localized protein only rarely halted or reversed and instead generally proceeded to the distal axon. Thus, our results are consistent with the axon initial segment behaving as a vesicle filter that mediates the differential trafficking of transport vesicles.


Asunto(s)
Transporte Axonal/fisiología , Dendritas/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Vesículas Transportadoras/metabolismo , Vesículas Transportadoras/fisiología , Animales , Transporte Biológico/fisiología , Células Cultivadas , Dendritas/ultraestructura , Embrión de Mamíferos , Modelos Biológicos , Miosinas/metabolismo , Ratas , Ratas Sprague-Dawley , Análisis de la Célula Individual , Imagen de Lapso de Tiempo , Distribución Tisular
10.
J Am Chem Soc ; 134(9): 3942-5, 2012 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-22332638

RESUMEN

Methods to rapidly and reversibly perturb the functions of specific proteins are desirable tools for studies of complex biological processes. We have demonstrated an experimental strategy to regulate the intracellular concentration of any protein of interest by using an engineered destabilizing protein domain and a cell-permeable small molecule. Destabilizing domains have general utility to confer instability to a wide range of proteins including integral transmembrane proteins. This study reports a destabilizing domain system based on the ligand binding domain of the estrogen receptor that can be regulated by one of two synthetic ligands, CMP8 or 4-hydroxytamoxifen.


Asunto(s)
Receptores de Estrógenos/química , Animales , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Sitios de Unión/efectos de los fármacos , Humanos , Ligandos , Proteínas Luminiscentes/química , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Ratones , Modelos Moleculares , Estructura Molecular , Mutación , Células 3T3 NIH , Ingeniería de Proteínas , Receptores de Estrógenos/genética , Receptores de Estrógenos/metabolismo , Relación Estructura-Actividad , Tamoxifeno/análogos & derivados , Tamoxifeno/química , Tamoxifeno/farmacología
11.
PLoS One ; 7(1): e30177, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22276156

RESUMEN

The analysis of dynamic events in the tumor microenvironment during cancer progression is limited by the complexity of current in vivo imaging models. This is coupled with an inability to rapidly modulate and visualize protein activity in real time and to understand the consequence of these perturbations in vivo. We developed an intravital imaging approach that allows the rapid induction and subsequent depletion of target protein levels within human cancer xenografts while assessing the impact on cell behavior and morphology in real time. A conditionally stabilized fluorescent E-cadherin chimera was expressed in metastatic breast cancer cells, and the impact of E-cadherin induction and depletion was visualized using real-time confocal microscopy in a xenograft avian embryo model. We demonstrate the assessment of protein localization, cell morphology and migration in cells undergoing epithelial-mesenchymal and mesenchymal-epithelial transitions in breast tumors. This technique allows for precise control over protein activity in vivo while permitting the temporal analysis of dynamic biophysical parameters.


Asunto(s)
Neoplasias de la Mama/metabolismo , Cadherinas/metabolismo , Animales , Aves/embriología , Línea Celular Tumoral , Diagnóstico por Imagen , Transición Epitelial-Mesenquimal/efectos de los fármacos , Femenino , Humanos , Microscopía Confocal , Microscopía Fluorescente , Morfolinas/farmacocinética , Morfolinas/farmacología , Trasplante Heterólogo , Vimentina/metabolismo
12.
Nat Chem Biol ; 7(8): 531-7, 2011 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-21725303

RESUMEN

The ability to rapidly regulate the functions of specific proteins in living cells is a valuable tool for biological research. Here we describe a new technique by which the degradation of a specific protein is induced by a small molecule. A protein of interest is fused to a ligand-induced degradation (LID) domain, resulting in the expression of a stable and functional fusion protein. The LID domain is comprised of the FK506- and rapamycin-binding protein (FKBP) and a 19-amino-acid degron fused to the C terminus of FKBP. In the absence of the small molecule Shield-1, the degron is bound to the FKBP fusion protein and the protein is stable. When present, Shield-1 binds tightly to FKBP, displacing the degron and inducing rapid and processive degradation of the LID domain and any fused partner protein. Structure-function studies of the 19-residue peptide showed that a 4-amino-acid sequence within the peptide is responsible for degradation.


Asunto(s)
Desnaturalización Proteica , Proteínas de Unión a Tacrolimus/química , Secuencia de Aminoácidos , Animales , Proteínas Bacterianas , Dominio Catalítico , Línea Celular , Clonación Molecular , Regulación de la Expresión Génica/fisiología , Proteínas Luminiscentes , Ratones , Modelos Moleculares , Morfolinas/farmacología , Péptidos/metabolismo , Estructura Terciaria de Proteína , Proteínas de Unión a Tacrolimus/genética , Proteínas de Unión a Tacrolimus/metabolismo
13.
J Biol Chem ; 286(13): 11307-13, 2011 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-21262969

RESUMEN

Chemical control of protein secretion using a small molecule approach provides a powerful tool to optimize tissue engineering strategies by regulating the spatial and temporal dimensions that are exposed to a specific protein. We placed fibroblast growth factor 2 (FGF-2) under conditional control of a small molecule and demonstrated greater than 50-fold regulation of FGF-2 release as well as tunability, reversibility, and functionality in vitro. We then applied conditional control of FGF-2 secretion to a cell-based, skeletal tissue engineering construct consisting of adipose stem cells (ASCs) on a biomimetic scaffold to promote bone formation in a murine critical-sized calvarial defect model. ASCs are an easily harvested and abundant source of postnatal multipotent cells and have previously been demonstrated to regenerate bone in critical-sized defects. These results suggest that chemically controlled FGF-2 secretion can significantly increase bone formation by ASCs in vivo. This study represents a novel approach toward refining protein delivery for tissue engineering applications.


Asunto(s)
Adipocitos , Factor 2 de Crecimiento de Fibroblastos/biosíntesis , Curación de Fractura , Fracturas Craneales/terapia , Cráneo/lesiones , Trasplante de Células Madre , Células Madre/metabolismo , Adipocitos/metabolismo , Adipocitos/trasplante , Animales , Factor 2 de Crecimiento de Fibroblastos/genética , Masculino , Ratones , Ratones Desnudos , Ingeniería de Tejidos , Andamios del Tejido , Trasplante Homólogo
15.
Nat Med ; 14(10): 1123-7, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18836461

RESUMEN

Conditional control of protein function in vivo offers great potential for deconvoluting the roles of individual proteins in complicated systems. We recently developed a method in which a small protein domain, termed a destabilizing domain, confers instability to fusion protein partners in cultured cells. Instability is reversed when a cell-permeable small molecule binds this domain. Here we describe the use of this system to regulate protein function in living mammals. We show regulation of secreted proteins and their biological activity with conditional secretion of an immunomodulatory cytokine, resulting in tumor burden reduction in mouse models. Additionally, we use this approach to control the function of a specific protein after systemic delivery of the gene that encodes it to a tumor, suggesting uses for enhancing the specificity and efficacy of targeted gene-based therapies. This method represents a new strategy to regulate protein function in living organisms with a high level of control.


Asunto(s)
Neoplasias Experimentales/tratamiento farmacológico , Proteínas de Unión a Tacrolimus/química , Proteínas de Unión a Tacrolimus/fisiología , Animales , Células HCT116 , Humanos , Interleucina-2/metabolismo , Ratones , Ratones SCID , Trasplante de Neoplasias , Neoplasias Experimentales/inmunología , Estructura Terciaria de Proteína , Trasplante Heterólogo , Factor de Necrosis Tumoral alfa/metabolismo
16.
Science ; 314(5804): 1458-61, 2006 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-17095657

RESUMEN

Many signaling, cytoskeletal, and transport proteins have to be localized to the plasma membrane (PM) in order to carry out their function. We surveyed PM-targeting mechanisms by imaging the subcellular localization of 125 fluorescent protein-conjugated Ras, Rab, Arf, and Rho proteins. Out of 48 proteins that were PM-localized, 37 contained clusters of positively charged amino acids. To test whether these polybasic clusters bind negatively charged phosphatidylinositol 4,5-bisphosphate [PI(4,5)P2] lipids, we developed a chemical phosphatase activation method to deplete PM PI(4,5)P2. Unexpectedly, proteins with polybasic clusters dissociated from the PM only when both PI(4,5)P2 and phosphatidylinositol 3,4,5-trisphosphate [PI(3,4,5)P3] were depleted, arguing that both lipid second messengers jointly regulate PM targeting.


Asunto(s)
Membrana Celular/metabolismo , GTP Fosfohidrolasas/metabolismo , Fosfatidilinositol 4,5-Difosfato/metabolismo , Fosfatos de Fosfatidilinositol/metabolismo , Factores de Ribosilacion-ADP/química , Factores de Ribosilacion-ADP/metabolismo , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , GTP Fosfohidrolasas/química , Células HeLa , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Cinética , Ratones , Datos de Secuencia Molecular , Células 3T3 NIH , Sistemas de Mensajero Secundario , Transducción de Señal , Electricidad Estática , Proteínas de Unión al GTP rab/química , Proteínas de Unión al GTP rab/metabolismo , Proteínas ras/química , Proteínas ras/metabolismo , Proteínas de Unión al GTP rho/metabolismo
17.
J Am Chem Soc ; 127(13): 4715-21, 2005 Apr 06.
Artículo en Inglés | MEDLINE | ID: mdl-15796538

RESUMEN

Rapamycin is an important immunosuppressant, a possible anticancer therapeutic, and a widely used research tool. Essential to its various functions is its ability to bind simultaneously to two different proteins, FKBP and mTOR. Despite its widespread use, a thorough analysis of the interactions between FKBP, rapamycin, and the rapamycin-binding domain of mTOR, FRB, is lacking. To probe the affinities involved in the formation of the FKBP.rapamycin.FRB complex, we used fluorescence polarization, surface plasmon resonance, and NMR spectroscopy. Analysis of the data shows that rapamycin binds to FRB with moderate affinity (K(d) = 26 +/- 0.8 microM). The FKBP12.rapamycin complex, however, binds to FRB 2000-fold more tightly (K(d) = 12 +/- 0.8 nM) than rapamycin alone. No interaction between FKBP and FRB was detected in the absence of rapamycin. These studies suggest that rapamycin's ability to bind to FRB, and by extension to mTOR, in the absence of FKBP is of little consequence under physiological conditions. Furthermore, protein-protein interactions at the FKBP12-FRB interface play a role in the stability of the ternary complex.


Asunto(s)
Proteínas Quinasas/química , Sirolimus/química , Proteínas de Unión a Tacrolimus/química , Unión Competitiva , Polarización de Fluorescencia , Cinética , Modelos Moleculares , Resonancia Magnética Nuclear Biomolecular , Unión Proteica , Proteínas Quinasas/metabolismo , Estructura Terciaria de Proteína , Sirolimus/metabolismo , Sirolimus/farmacología , Resonancia por Plasmón de Superficie , Serina-Treonina Quinasas TOR , Proteína 1A de Unión a Tacrolimus/química , Proteína 1A de Unión a Tacrolimus/metabolismo , Proteínas de Unión a Tacrolimus/metabolismo
18.
J Org Chem ; 69(25): 8810-20, 2004 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-15575762

RESUMEN

As part of investigations into cell cycle checkpoint inhibitors, an asymmetric synthesis of the antimitotic natural product, ustiloxin D, has been completed. A salen-Al-catalyzed aldol reaction was employed to construct a chiral oxazoline 9 (99% yield, 98% ee) that served the dual purpose of installing the necessary 1,2-amino alcohol functionality as well as providing an efficient synthon for the requisite methylamino group at C9. The chiral aryl-alkyl ether was assembled using a Pd-catalyzed asymmetric allylic alkylation that notably delivered a product with stereochemistry opposite to that predicted by precedent. The linear tetrapeptide was subsequently cyclized to produce ustiloxin D. The mechanistic origin of the allylic alkylation selectivity was further investigated, and a working hypothesis for the origin of the observed stereoselectivity has been proposed.


Asunto(s)
Antineoplásicos/síntesis química , Micotoxinas/síntesis química , Péptidos/síntesis química , Alquilación , Modelos Moleculares , Conformación Molecular , Péptidos Cíclicos
19.
Biochemistry ; 43(18): 5406-13, 2004 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-15122906

RESUMEN

Small molecules can be discovered or engineered to bind tightly to biologically relevant proteins, and these molecules have proven to be powerful tools for both basic research and therapeutic applications. In many cases, detailed biophysical analyses of the intermolecular binding events are essential for improving the activity of the small molecules. These interactions can often be characterized as straightforward bimolecular binding events, and a variety of experimental and analytical techniques have been developed and refined to facilitate these analyses. Several investigators have recently synthesized heterodimeric molecules that are designed to bind simultaneously with two different proteins to form ternary complexes. These heterodimeric molecules often display compelling biological activity; however, they are difficult to characterize. The bimolecular interaction between one protein and the heterodimeric ligand (primary dissociation constant) can be determined by a number of methods. However, the interaction between that protein-ligand complex and the second protein (secondary dissociation constant) is more difficult to measure due to the noncovalent nature of the original protein-ligand complex. Consequently, these heterodimeric compounds are often characterized in terms of their activity, which is an experimentally dependent metric. We have developed a general quantitative mathematical model that can be used to measure both the primary (protein + ligand) and secondary (protein-ligand + protein) dissociation constants for heterodimeric small molecules. These values are largely independent of the experimental technique used and furthermore provide a direct measure of the thermodynamic stability of the ternary complexes that are formed. Fluorescence polarization and this model were used to characterize the heterodimeric molecule, SLFpYEEI, which binds to both FKBP12 and the Fyn SH2 domain, demonstrating that the model is useful for both predictive as well as ex post facto analytical applications.


Asunto(s)
Modelos Químicos , Oligopéptidos/química , Oligopéptidos/metabolismo , Proteínas de Unión a Tacrolimus/química , Proteínas de Unión a Tacrolimus/metabolismo , Unión Competitiva , Dimerización , Polarización de Fluorescencia , Concentración 50 Inhibidora , Cinética , Ligandos , Unión Proteica , Proteínas Proto-Oncogénicas/química , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-fyn , Dominios Homologos src
20.
Mol Cell ; 12(6): 1615-24, 2003 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-14690613

RESUMEN

We have developed a general method of making conditional alleles that allows the rapid and reversible regulation of specific proteins. A mouse line was produced in which proteins encoded by the endogenous glycogen synthase kinase-3 beta (GSK-3beta) gene are fused to an 89 amino acid tag, FRB*. FRB* causes the destabilization of GSK-3beta, producing a severe loss-of-function allele. In the presence of C20-MaRap, a highly specific, nontoxic, cell-permeable small molecule, GSK-3betaFRB* binds to the ubiquitously expressed FKBP12 protein. This interaction stabilizes GSK-3betaFRB* and restores both protein levels and activity. C20-MaRap-mediated stabilization is rapidly reversed by the addition of an FKBP12 binding competitor molecule. This technology may be applied to a wide range of FRB*-tagged mouse genes while retaining their native transcriptional control. Inducible stabilization could be valuable for many developmental and physiological studies and for drug target validation.


Asunto(s)
Alelos , Glucógeno Sintasa Quinasa 3/genética , Ratones Transgénicos , Animales , Antifúngicos/química , Antifúngicos/metabolismo , Células Cultivadas , Cisteína Endopeptidasas/metabolismo , Dimerización , Embrión de Mamíferos/fisiología , Estabilidad de Enzimas , Femenino , Fibroblastos/citología , Fibroblastos/metabolismo , Eliminación de Gen , Genes Reporteros , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Ratones , Estructura Molecular , Complejos Multienzimáticos/metabolismo , Embarazo , Complejo de la Endopetidasa Proteasomal , Estructura Terciaria de Proteína , Proteínas Recombinantes de Fusión/metabolismo , Sirolimus/química , Sirolimus/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA