Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Acta Crystallogr E Crystallogr Commun ; 72(Pt 5): 704-8, 2016 May 01.
Article in English | MEDLINE | ID: mdl-27308023

ABSTRACT

In the structures of the two title coumarin derivatives, C14H12O6, (1), and C20H16O6, (2), one with acetate and the other with pent-4-ynoate substituents, both the coumarin rings are almost planar. In (1), both acetate substituents are significantly rotated out of the coumarin plane to minimize steric repulsions. One acetate substituent is disordered over two equivalent conformations, with occupancies of 0.755 (17) and 0.245 (17). In (2), there are two pent-4-ynoate substituents, the C C group of one being disordered over two positions with occupancies of 0.55 (2) and 0.45 (2). One of the pent-4-ynoate substituents is in an extended conformation, while the other is in a bent conformation. In this derivative, the planar part of both pent-4-ynoate substituents deviate from the coumarin plane. The packing of (1) is dominated by π-π stacking involving the coumarin rings and weak C-H⋯O contacts link the parallel stacks in the [101] direction. In contrast, in (2) the packing is dominated by R 2 (2)(24) hydrogen bonds, involving the acidic sp H atom and the oxo O atom, which link the mol-ecules into centrosymmetric dimers. The bent conformation of one of the pent-4-ynoate substituents prevents the coumarin rings from engaging in π-π stacking.

3.
J Biol Chem ; 282(34): 24866-72, 2007 Aug 24.
Article in English | MEDLINE | ID: mdl-17603093

ABSTRACT

The ability to regulate the function of specific proteins using cell-permeable molecules can be a powerful method for interrogating biological systems. To bring this type of "chemical genetic" control to a wide range of proteins, we recently developed an experimental system in which the stability of a small protein domain expressed in mammalian cells depends on the presence of a high affinity ligand. This ligand-dependent stability is conferred to any fused partner protein. The FK506- and rapamycin-binding protein (FKBP12) has been the subject of extensive biophysical analyses, including both kinetic and thermodynamic studies of the wild-type protein as well as dozens of mutants. The goal of this study was to determine if the thermodynamic stabilities (DeltaDeltaG(U-F)) of various amino acid substitutions within a given protein are predictive for engineering additional ligand-dependent destabilizing domains. We used FKBP12 as a model system and found that in vitro thermodynamic stability correlates weakly with intracellular degradation rates of the mutants and that the ability of a given mutation to destabilize the protein is context-dependent. We evaluated several new FKBP12 ligands for their ability to stabilize these mutants and found that a cell-permeable molecule called Shield-1 is the most effective stabilizing ligand. We then performed an unbiased microarray analysis of NIH3T3 cells treated with various concentrations of Shield-1. These studies show that Shield-1 does not elicit appreciable cellular responses.


Subject(s)
Protein Engineering/methods , Animals , Bacterial Proteins/chemistry , Biophysics/methods , Humans , Kinetics , Ligands , Luminescent Proteins/chemistry , Mice , Models, Molecular , Mutation , NIH 3T3 Cells , Oligonucleotide Array Sequence Analysis , Protein Binding , Tacrolimus Binding Protein 1A/chemistry , Thermodynamics
4.
Cell ; 126(5): 995-1004, 2006 Sep 08.
Article in English | MEDLINE | ID: mdl-16959577

ABSTRACT

Rapid and reversible methods for perturbing the function of specific proteins are desirable tools for probing complex biological systems. We have developed a general technique to regulate the stability of specific proteins in mammalian cells using cell-permeable, synthetic molecules. We engineered mutants of the human FKBP12 protein that are rapidly and constitutively degraded when expressed in mammalian cells, and this instability is conferred to other proteins fused to these destabilizing domains. Addition of a synthetic ligand that binds to the destabilizing domains shields them from degradation, allowing fused proteins to perform their cellular functions. Genetic fusion of the destabilizing domain to a gene of interest ensures specificity, and the attendant small-molecule control confers speed, reversibility, and dose-dependence to this method. This general strategy for regulating protein stability should enable conditional perturbation of specific proteins with unprecedented control in a variety of experimental settings.


Subject(s)
Gene Expression Regulation , Morpholines/metabolism , Proteasome Endopeptidase Complex/metabolism , Recombinant Fusion Proteins/metabolism , Tacrolimus Binding Protein 1A/genetics , Tacrolimus Binding Proteins/genetics , Animals , Ligands , Luminescent Proteins/genetics , Mice , Mutation , NIH 3T3 Cells , Phenotype , Protein Binding , Protein Structure, Tertiary , Tacrolimus Binding Protein 1A/chemistry , Tacrolimus Binding Proteins/chemistry , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...