Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
Acta Crystallogr D Struct Biol ; 79(Pt 1): 50-65, 2023 Jan 01.
Article in English | MEDLINE | ID: mdl-36601807

ABSTRACT

It is investigated whether molecular-dynamics (MD) simulations can be used to enhance macromolecular crystallography (MX) studies. Historically, protein crystal structures have been described using a single set of atomic coordinates. Because conformational variation is important for protein function, researchers now often build models that contain multiple structures. Methods for building such models can fail, however, in regions where the crystallographic density is difficult to interpret, for example at the protein-solvent interface. To address this limitation, a set of MD-MX methods that combine MD simulations of protein crystals with conventional modeling and refinement tools have been developed. In an application to a cyclic adenosine monophosphate-dependent protein kinase at room temperature, the procedure improved the interpretation of ambiguous density, yielding an alternative water model and a revised protein model including multiple conformations. The revised model provides mechanistic insights into the catalytic and regulatory interactions of the enzyme. The same methods may be used in other MX studies to seek mechanistic insights.


Subject(s)
Molecular Dynamics Simulation , Proteins , Protein Conformation , Proteins/chemistry , Solvents/chemistry , Crystallography, X-Ray
2.
Proc Natl Acad Sci U S A ; 119(47): e2215420119, 2022 11 22.
Article in English | MEDLINE | ID: mdl-36375071

ABSTRACT

Topological analysis of protein residue networks (PRNs) is a common method that can help to understand the roles of individual residues. Here, we used protein kinase A as a study object and asked what already known functionally important residues can be detected by network analysis. Along several traditional approaches to weight edges in PRNs we used local spatial pattern (LSP) alignment that assigns high weights to edges only if CαCß vectors for the corresponding residues retain their mutual positions and orientation. Our results show that even short molecular dynamic simulations of 10 to 20 ns can give convergent values for betweenness and degree centralities calculated from the LSP-based PRNs. Using these centralities, we were able to clearly distinguish a group of residues that are highly conserved in protein kinases and play important functional and regulatory roles. In comparison, traditional methods based on cross-correlation and linear mutual information were much less efficient for this particular task. These results call for reevaluation of the current methods to generate PRNs.


Subject(s)
Cyclic AMP-Dependent Protein Kinases , Molecular Dynamics Simulation
3.
PLoS Biol ; 20(2): e3001427, 2022 02.
Article in English | MEDLINE | ID: mdl-35192607

ABSTRACT

The 2 major molecular switches in biology, kinases and GTPases, are both contained in the Parkinson disease-related leucine-rich repeat kinase 2 (LRRK2). Using hydrogen-deuterium exchange mass spectrometry (HDX-MS) and molecular dynamics (MD) simulations, we generated a comprehensive dynamic allosteric portrait of the C-terminal domains of LRRK2 (LRRK2RCKW). We identified 2 helices that shield the kinase domain and regulate LRRK2 conformation and function. One helix in COR-B (COR-B Helix) tethers the COR-B domain to the αC helix of the kinase domain and faces its activation loop, while the C-terminal helix (Ct-Helix) extends from the WD40 domain and interacts with both kinase lobes. The Ct-Helix and the N-terminus of the COR-B Helix create a "cap" that regulates the N-lobe of the kinase domain. Our analyses reveal allosteric sites for pharmacological intervention and confirm the kinase domain as the central hub for conformational control.


Subject(s)
Catalytic Domain , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/chemistry , Molecular Dynamics Simulation , Protein Conformation , Allosteric Regulation , Allosteric Site , Deuterium Exchange Measurement/methods , Humans , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/genetics , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/metabolism , Mass Spectrometry/methods , Mutation , Protein Binding
4.
Proc Natl Acad Sci U S A ; 118(23)2021 06 08.
Article in English | MEDLINE | ID: mdl-34088839

ABSTRACT

To explore how pathogenic mutations of the multidomain leucine-rich repeat kinase 2 (LRRK2) hijack its finely tuned activation process and drive Parkinson's disease (PD), we used a multitiered approach. Most mutations mimic Rab-mediated activation by "unleashing" kinase activity, and many, like the kinase inhibitor MLi-2, trap LRRK2 onto microtubules. Here we mimic activation by simply deleting the inhibitory N-terminal domains and then characterize conformational changes induced by MLi-2 and PD mutations. After confirming that LRRK2RCKW retains full kinase activity, we used hydrogen-deuterium exchange mass spectrometry to capture breathing dynamics in the presence and absence of MLi-2. Solvent-accessible regions throughout the entire protein are reduced by MLi-2 binding. With molecular dynamics simulations, we created a dynamic portrait of LRRK2RCKW and demonstrate the consequences of kinase domain mutations. Although all domains contribute to regulating kinase activity, the kinase domain, driven by the DYGψ motif, is the allosteric hub that drives LRRK2 regulation.


Subject(s)
Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/chemistry , Molecular Dynamics Simulation , Amino Acid Motifs , Humans , Hydrogen Deuterium Exchange-Mass Spectrometry , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/genetics , Protein Domains , Protein Transport
5.
PLoS Biol ; 18(12): e3001018, 2020 12.
Article in English | MEDLINE | ID: mdl-33370777

ABSTRACT

When the J-domain of the heat shock protein DnaJB1 is fused to the catalytic (C) subunit of cAMP-dependent protein kinase (PKA), replacing exon 1, this fusion protein, J-C subunit (J-C), becomes the driver of fibrolamellar hepatocellular carcinoma (FL-HCC). Here, we use cryo-electron microscopy (cryo-EM) to characterize J-C bound to RIIß, the major PKA regulatory (R) subunit in liver, thus reporting the first cryo-EM structure of any PKA holoenzyme. We report several differences in both structure and dynamics that could not be captured by the conventional crystallography approaches used to obtain prior structures. Most striking is the asymmetry caused by the absence of the second cyclic nucleotide binding (CNB) domain and the J-domain in one of the RIIß:J-C protomers. Using molecular dynamics (MD) simulations, we discovered that this asymmetry is already present in the wild-type (WT) RIIß2C2 but had been masked in the previous crystal structure. This asymmetry may link to the intrinsic allosteric regulation of all PKA holoenzymes and could also explain why most disease mutations in PKA regulatory subunits are dominant negative. The cryo-EM structure, combined with small-angle X-ray scattering (SAXS), also allowed us to predict the general position of the Dimerization/Docking (D/D) domain, which is essential for localization and interacting with membrane-anchored A-Kinase-Anchoring Proteins (AKAPs). This position provides a multivalent mechanism for interaction of the RIIß holoenzyme with membranes and would be perturbed in the oncogenic fusion protein. The J-domain also alters several biochemical properties of the RIIß holoenzyme: It is easier to activate with cAMP, and the cooperativity is reduced. These results provide new insights into how the finely tuned allosteric PKA signaling network is disrupted by the oncogenic J-C subunit, ultimately leading to the development of FL-HCC.


Subject(s)
Carcinoma, Hepatocellular/genetics , Cyclic AMP-Dependent Protein Kinase RIIbeta Subunit/metabolism , HSP40 Heat-Shock Proteins/metabolism , Adenosine Triphosphate/metabolism , Allosteric Regulation , Carcinoma, Hepatocellular/metabolism , Cryoelectron Microscopy/methods , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinase RIIbeta Subunit/genetics , Cyclic AMP-Dependent Protein Kinase RIIbeta Subunit/ultrastructure , Cyclic AMP-Dependent Protein Kinase RIalpha Subunit/metabolism , Cyclic AMP-Dependent Protein Kinases/genetics , Cyclic AMP-Dependent Protein Kinases/metabolism , HSP40 Heat-Shock Proteins/genetics , HSP40 Heat-Shock Proteins/ultrastructure , Holoenzymes/metabolism , Humans , Liver Neoplasms/genetics , Molecular Dynamics Simulation , Protein Binding , Protein Subunits/metabolism , Recombinant Fusion Proteins/genetics , Scattering, Small Angle , X-Ray Diffraction/methods
6.
Am J Hum Genet ; 107(5): 977-988, 2020 11 05.
Article in English | MEDLINE | ID: mdl-33058759

ABSTRACT

PRKACA and PRKACB code for two catalytic subunits (Cα and Cß) of cAMP-dependent protein kinase (PKA), a pleiotropic holoenzyme that regulates numerous fundamental biological processes such as metabolism, development, memory, and immune response. We report seven unrelated individuals presenting with a multiple congenital malformation syndrome in whom we identified heterozygous germline or mosaic missense variants in PRKACA or PRKACB. Three affected individuals were found with the same PRKACA variant, and the other four had different PRKACB mutations. In most cases, the mutations arose de novo, and two individuals had offspring with the same condition. Nearly all affected individuals and their affected offspring shared an atrioventricular septal defect or a common atrium along with postaxial polydactyly. Additional features included skeletal abnormalities and ectodermal defects of variable severity in five individuals, cognitive deficit in two individuals, and various unusual tumors in one individual. We investigated the structural and functional consequences of the variants identified in PRKACA and PRKACB through the use of several computational and experimental approaches, and we found that they lead to PKA holoenzymes which are more sensitive to activation by cAMP than are the wild-type proteins. Furthermore, expression of PRKACA or PRKACB variants detected in the affected individuals inhibited hedgehog signaling in NIH 3T3 fibroblasts, thereby providing an underlying mechanism for the developmental defects observed in these cases. Our findings highlight the importance of both Cα and Cß subunits of PKA during human development.


Subject(s)
Abnormalities, Multiple/genetics , Cognitive Dysfunction/genetics , Cyclic AMP-Dependent Protein Kinase Catalytic Subunits/genetics , Fingers/abnormalities , Germ-Line Mutation , Heart Septal Defects/genetics , Polydactyly/genetics , Toes/abnormalities , Abnormalities, Multiple/diagnosis , Abnormalities, Multiple/pathology , Adolescent , Adult , Animals , Base Sequence , Cognitive Dysfunction/diagnosis , Cognitive Dysfunction/pathology , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinase Catalytic Subunits/chemistry , Cyclic AMP-Dependent Protein Kinase Catalytic Subunits/deficiency , Female , Fingers/pathology , Gene Expression Regulation, Developmental , Heart Septal Defects/diagnosis , Heart Septal Defects/pathology , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Holoenzymes/chemistry , Holoenzymes/deficiency , Holoenzymes/genetics , Humans , Infant, Newborn , Male , Mice , Models, Molecular , Mosaicism , NIH 3T3 Cells , Pedigree , Polydactyly/diagnosis , Polydactyly/pathology , Protein Structure, Secondary , Toes/pathology
7.
PLoS Pathog ; 16(3): e1008373, 2020 03.
Article in English | MEDLINE | ID: mdl-32150583

ABSTRACT

Lasting protection has long been a goal for malaria vaccines. The major surface antigen on Plasmodium falciparum sporozoites, the circumsporozoite protein (PfCSP), has been an attractive target for vaccine development and most protective antibodies studied to date interact with the central NANP repeat region of PfCSP. However, it remains unclear what structural and functional characteristics correlate with better protection by one antibody over another. Binding to the junctional region between the N-terminal domain and central NANP repeats has been proposed to result in superior protection: this region initiates with the only NPDP sequence followed immediately by NANP. Here, we isolated antibodies in Kymab mice immunized with full-length recombinant PfCSP and two protective antibodies were selected for further study with reactivity against the junctional region. X-ray and EM structures of two monoclonal antibodies, mAb667 and mAb668, shed light on their differential affinity and specificity for the junctional region. Importantly, these antibodies also bind to the NANP repeat region with equal or better affinity. A comparison with an NANP-only binding antibody (mAb317) revealed roughly similar but statistically distinct levels of protection against sporozoite challenge in mouse liver burden models, suggesting that junctional antibody protection might relate to the ability to also cross-react with the NANP repeat region. Our findings indicate that additional efforts are necessary to isolate a true junctional antibody with no or much reduced affinity to the NANP region to elucidate the role of the junctional epitope in protection.


Subject(s)
Antibodies, Monoclonal, Murine-Derived/chemistry , Antibodies, Protozoan/chemistry , Binding Sites, Antibody , Epitopes/chemistry , Plasmodium falciparum/chemistry , Protozoan Proteins/chemistry , Animals , Antibodies, Monoclonal, Murine-Derived/immunology , Antibodies, Protozoan/immunology , Epitopes/immunology , Female , Male , Mice , Mice, Transgenic , Plasmodium falciparum/immunology , Protozoan Proteins/immunology , Structure-Activity Relationship
8.
Biochemistry ; 58(51): 5160-5172, 2019 12 24.
Article in English | MEDLINE | ID: mdl-31794659

ABSTRACT

The inactive state of mitogen-activated protein kinases (MAPKs) adopts an open conformation while the active state exists in a compact form stabilized by phosphorylation. In the active state, eukaryotic kinases undergo breathing motions related to substrate binding and product release that have not previously been detected in the inactive state. However, docking interactions of partner proteins with inactive MAPK kinases exhibit allostery in binding of activating kinases. Interactions at a site distant from the activation loop are coupled to the configuration of the activation loop, suggesting that the inactive state may also undergo concerted dynamics. X-ray crystallographic studies of nonphosphorylated, inactive p38γ reveal differences in domain orientations and active site structure in the two molecules in the asymmetric unit. One molecule resembles an inactive kinase with an open active site. The second molecule has a rotation of the N-lobe that leads to partial compaction of the active site, resulting in a conformation that is intermediate between the inactive open state and the fully closed state of the activated kinase. Although the compact state of apo p38γ displays several of the features of the activated enzyme, it remains catalytically inert. In solution, the kinase fluctuates on a millisecond time scale between the open ground state and a weakly populated excited state that is similar in structure to the compact state observed in the crystal. The nuclear magnetic resonance and crystal structure data imply that interconversion between the open and compact states involves a molecular switch associated with the DFG loop.


Subject(s)
Mitogen-Activated Protein Kinase 12/chemistry , Mitogen-Activated Protein Kinase 12/metabolism , Adenosine Triphosphate/metabolism , Conserved Sequence , Enzyme Activation , Humans , Models, Molecular , Protein Domains
9.
Proc Natl Acad Sci U S A ; 116(33): 16347-16356, 2019 08 13.
Article in English | MEDLINE | ID: mdl-31363049

ABSTRACT

Protein kinase A (PKA) holoenzyme, comprised of a cAMP-binding regulatory (R)-subunit dimer and 2 catalytic (C)-subunits, is the master switch for cAMP-mediated signaling. Of the 4 R-subunits (RIα, RIß, RIIα, RIIß), RIα is most essential for regulating PKA activity in cells. Our 2 RIα2C2 holoenzyme states, which show different conformations with and without ATP, reveal how ATP/Mg2+ functions as a negative orthosteric modulator. Biochemical studies demonstrate how the removal of ATP primes the holoenzyme for cAMP-mediated activation. The opposing competition between ATP/cAMP is unique to RIα. In RIIß, ATP serves as a substrate and facilitates cAMP-activation. The isoform-specific RI-holoenzyme dimer interface mediated by N3A-N3A' motifs defines multidomain cross-talk and an allosteric network that creates competing roles for ATP and cAMP. Comparisons to the RIIß holoenzyme demonstrate isoform-specific holoenzyme interfaces and highlights distinct allosteric mechanisms for activation in addition to the structural diversity of the isoforms.


Subject(s)
Cyclic AMP-Dependent Protein Kinase RIIbeta Subunit/chemistry , Cyclic AMP-Dependent Protein Kinase RIalpha Subunit/chemistry , Cyclic AMP-Dependent Protein Kinases/chemistry , Protein Structure, Quaternary , Adenosine Triphosphate/chemistry , Adenosine Triphosphate/genetics , Allosteric Regulation/genetics , Amino Acid Sequence/genetics , Crystallography, X-Ray , Cyclic AMP/chemistry , Cyclic AMP/genetics , Cyclic AMP-Dependent Protein Kinase RIIbeta Subunit/genetics , Cyclic AMP-Dependent Protein Kinase RIalpha Subunit/genetics , Cyclic AMP-Dependent Protein Kinases/genetics , Gene Expression Regulation, Enzymologic/genetics , Holoenzymes/chemistry , Holoenzymes/genetics , Humans , Protein Binding/genetics , Protein Subunits/chemistry , Protein Subunits/genetics , Signal Transduction/genetics
10.
Proc Natl Acad Sci U S A ; 116(30): 15052-15061, 2019 07 23.
Article in English | MEDLINE | ID: mdl-31285328

ABSTRACT

A dense interplay between structure and dynamics underlies the working of proteins, especially enzymes. Protein kinases are molecular switches that are optimized for their regulation rather than catalytic turnover rates. Using long-simulations dynamic allostery analysis, this study describes an exploration of the dynamic kinase:peptide complex. We have used protein kinase A (PKA) as a model system as a generic prototype of the protein kinase superfamily of signaling enzymes. Our results explain the role of dynamic coupling of active-site residues that must work in coherence to provide for a successful activation or inhibition response from the kinase. Amino acid networks-based community analysis allows us to ponder the conformational entropy of the kinase:nucleotide:peptide ternary complex. We use a combination of 7 peptides that include 3 types of PKA-binding partners: Substrates, products, and inhibitors. The substrate peptides provide for dynamic insights into the enzyme:substrate complex, while the product phospho-peptide allows for accessing modes of enzyme:product release. Mapping of allosteric communities onto the PKA structure allows us to locate the more unvarying and flexible dynamic regions of the kinase. These distributions, when correlated with the structural elements of the kinase core, allow for a detailed exploration of key dynamics-based signatures that could affect peptide recognition and binding at the kinase active site. These studies provide a unique dynamic allostery-based perspective to kinase:peptide complexes that have previously been explored only in a structural or thermodynamic context.


Subject(s)
Adenosine Triphosphate/chemistry , Cyclic AMP-Dependent Protein Kinases/chemistry , Enzyme Inhibitors/chemistry , Magnesium/chemistry , Peptides/chemistry , Adenosine Triphosphate/metabolism , Allosteric Regulation , Allosteric Site , Amino Acid Sequence , Catalytic Domain , Cyclic AMP-Dependent Protein Kinases/metabolism , Enzyme Inhibitors/metabolism , Kinetics , Magnesium/metabolism , Molecular Dynamics Simulation , Peptides/metabolism , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Protein Structure, Tertiary , Substrate Specificity , Thermodynamics
11.
J Am Chem Soc ; 139(32): 11233-11240, 2017 08 16.
Article in English | MEDLINE | ID: mdl-28737940

ABSTRACT

The rate-determining step in the catalytic cycle of E. coli dihydrofolate reductase is tetrahydrofolate (THF) product release, which can occur via an allosteric or an intrinsic pathway. The allosteric pathway, which becomes accessible when the reduced cofactor NADPH is bound, involves transient sampling of a higher energy conformational state, greatly increasing the product dissociation rate as compared to the intrinsic pathway that obtains when NADPH is absent. Although the kinetics of this process are known, the enzyme structure and the THF product conformation in the transiently formed excited state remain elusive. Here, we use side-chain proton NMR relaxation dispersion measurements, X-ray crystallography, and structure-based chemical shift predictions to explore the structural basis of allosteric product release. In the excited state of the E:THF:NADPH product release complex, the reduced nicotinamide ring of the cofactor transiently enters the active site where it displaces the pterin ring of the THF product. The p-aminobenzoyl-l-glutamate tail of THF remains weakly bound in a widened binding cleft. Thus, through transient entry of the nicotinamide ring into the active site, the NADPH cofactor remodels the enzyme structure and the conformation of the THF to form a weakly populated excited state that is poised for rapid product release.


Subject(s)
Escherichia coli K12/enzymology , NADP/metabolism , Tetrahydrofolate Dehydrogenase/metabolism , Tetrahydrofolates/metabolism , Allosteric Regulation , Crystallography, X-Ray , Escherichia coli K12/chemistry , Escherichia coli K12/metabolism , Kinetics , Molecular Docking Simulation , Nuclear Magnetic Resonance, Biomolecular , Protein Conformation , Tetrahydrofolate Dehydrogenase/chemistry
12.
Sci Rep ; 6: 28655, 2016 06 29.
Article in English | MEDLINE | ID: mdl-27353957

ABSTRACT

The intramolecular network structure of a protein provides valuable insights into allosteric sites and communication pathways. However, a straightforward method to comprehensively map and characterize these pathways is not currently available. Here we present an approach to characterize intramolecular network structure using NMR chemical shift perturbations. We apply the method to the mitogen activated protein kinase (MAPK) p38γ. p38γ contains allosteric sites that are conserved among eukaryotic kinases as well as unique to the MAPK family. How these regulatory sites communicate with catalytic residues is not well understood. Using our method, we observe and characterize for the first time information flux between regulatory sites through a conserved kinase infrastructure. This network is accessed, reinforced, and broken in various states of p38γ, reflecting the functional state of the protein. We demonstrate that the approach detects critical junctions in the network corresponding to biologically significant allosteric sites and pathways.


Subject(s)
Mitogen-Activated Protein Kinase 12/chemistry , Nuclear Magnetic Resonance, Biomolecular , Allosteric Regulation , Humans , Mitogen-Activated Protein Kinase 12/genetics , Mitogen-Activated Protein Kinase 12/metabolism , Protein Domains
13.
J Magn Reson ; 246: 31-5, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25063954

ABSTRACT

Non-uniform sampling (NUS) in NMR spectroscopy is a recognized and powerful tool to minimize acquisition time. Recent advances in reconstruction methodologies are paving the way for the use of NUS in quantitative applications, where accurate measurement of peak intensities is crucial. The presence or absence of NUS artifacts in reconstructed spectra ultimately determines the success of NUS in quantitative NMR. The quality of reconstructed spectra from NUS acquired data is dependent upon the quality of the sampling scheme. Here we demonstrate that the best performing sampling schemes make up a very small percentage of the total randomly generated schemes. A scoring method is found to accurately predict the quantitative similarity between reconstructed NUS spectra and those of fully sampled spectra. We present an easy-to-use protocol to batch generate and rank optimal Poisson-gap NUS schedules for use with 2D NMR with minimized noise and accurate signal reproduction, without the need for the creation of synthetic spectra.


Subject(s)
Algorithms , Artifacts , Data Interpretation, Statistical , Magnetic Resonance Spectroscopy/methods , Models, Statistical , Computer Simulation , Reproducibility of Results , Sample Size , Sensitivity and Specificity
14.
Biochemistry ; 53(23): 3767-80, 2014 Jun 17.
Article in English | MEDLINE | ID: mdl-24857522

ABSTRACT

Apomyoglobin folds via sequential helical intermediates that are formed by rapid collapse of the A, B, G, and H helix regions. An equilibrium molten globule with a similar structure is formed near pH 4. Previous studies suggested that the folding intermediates are kinetically trapped states in which folding is impeded by non-native packing of the G and H helices. Fluorescence spectra of mutant proteins in which cysteine residues were introduced at several positions in the G and H helices show differential quenching of W14 fluorescence, providing direct evidence of translocation of the H helix relative to helices A and G in both the kinetic and equilibrium intermediates. Förster resonance energy transfer measurements show that a 5-({2-[(acetyl)amino]ethyl}amino)naphthalene-1-sulfonic acid acceptor coupled to K140C (helix H) is closer to Trp14 (helix A) in the equilibrium molten globule than in the native state, by a distance that is consistent with sliding of the H helix in an N-terminal direction by approximately one helical turn. Formation of an S108C-L135C disulfide prevents H helix translocation in the equilibrium molten globule by locking the G and H helices into their native register. By enforcing nativelike packing of the A, G, and H helices, the disulfide resolves local energetic frustration and facilitates transient docking of the E helix region onto the hydrophobic core but has only a small effect on the refolding rate. The apomyoglobin folding landscape is highly rugged, with several energetic bottlenecks that frustrate folding; relief of any one of the major identified bottlenecks is insufficient to speed progression to the transition state.


Subject(s)
Apoproteins/chemistry , Models, Molecular , Myoglobin/chemistry , Protein Folding , Sperm Whale , Amino Acid Substitution , Animals , Apoproteins/genetics , Apoproteins/metabolism , Cysteine , Cystine , Hydrogen-Ion Concentration , Hydrophobic and Hydrophilic Interactions , Kinetics , Mutagenesis, Site-Directed , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Myoglobin/genetics , Myoglobin/metabolism , Oxidation-Reduction , Protein Conformation , Protein Refolding , Protein Stability , Protein Structure, Secondary , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Thermodynamics
15.
Anal Chem ; 80(14): 5358-66, 2008 Jul 15.
Article in English | MEDLINE | ID: mdl-18522433

ABSTRACT

Biarsenical dyes complexed to tetracysteine motifs have proven to be highly useful fluorescent dyes in labeling specific cellular proteins for microscopic imaging. Their many advantages include membrane permeability, relatively small size, stoichiometric labeling, high affinity, and an assortment of excitation/emission wavelengths. The goal of the current study was to determine whether the biarsenical labeling scheme could be extended to fluorescent detection of analytes in capillary electrophoresis. Recombinant protein or synthesized peptides containing the optimized tetracysteine motif "-C-C-P-G-C-C-" were labeled with biarsenical dyes and then analyzed by micellar electrokinetic capillary chromatography (MEKC). The biarsenical-tetracysteine complex was stable and remained fluorescent under standard MEKC conditions for peptide and protein separations. The detection limit following electrophoresis in a capillary was less than 3 x 10(-20) mol with a simple laser-induced fluorescence system. A mixture of multiple biarsenical-labeled peptides and a protein were easily resolved. Demonstrating that the label did not interfere with bioactivity, a peptide-based enzyme substrate conjugated to the tetracysteine motif and labeled with a biarsenical dye retained its ability to be phosphorylated by the parent kinase. The feasibility of using this label for chemical cytometry experiments was shown by intracellular labeling and subsequent analysis of a recombinant protein possessing the tetracysteine motif expressed in living cells. The extension of the biarsenical-tetracysteine tag to fluorescent labeling of peptides and proteins in chemical separations is a valuable addition to biochemical and cell-based investigations.


Subject(s)
Electrophoresis, Capillary/methods , Fluorescent Dyes/analysis , Peptides/analysis , Amino Acid Motifs , Cysteine/chemistry , Escherichia coli/chemistry , Escherichia coli/cytology , Escherichia coli/genetics , Escherichia coli/metabolism , Fluorescence Resonance Energy Transfer , Lasers , Peptides/chemistry , Phosphorylation , Proteins/analysis , Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Substrate Specificity
16.
Cytometry A ; 71(10): 866-74, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17559133

ABSTRACT

A host of technologies exists for the separation of living, nonadherent cells, with separation decisions typically based on fluorescence or immunolabeling of cells. Methods to separate adherent cells as well as to broaden the range of possible sorting criteria would be of high value and complementary to existing strategies. Cells were cultured on arrays of releasable pallets. The arrays were screened and individual cell(s)/pallets were released and collected. Conventional fluorescence and immunolabeling of cells were compatible with the pallet arrays, as were separations based on gene expression. By varying the size of the pallet and the number of cells cultured on the array, single cells or clonal colonies of cells were isolated from a heterogeneous population. Since cells remained adherent throughout the isolation process, separations based on morphologic characteristics, for example cell shape, were feasible. Repeated measurements of each cell in an array were performed permitting the selection of cells based on their temporal behavior, e.g. growth rate. The pallet array system provides the flexibility to select and collect adherent cells based on phenotypic and temporal criteria and other characteristics not accessible by alternative methods.


Subject(s)
Tissue Array Analysis/methods , Animals , Cell Adhesion/drug effects , Cell Proliferation/drug effects , Cell Separation , Cell Shape/drug effects , Clone Cells , Dimethylpolysiloxanes/pharmacology , Fluorescence , Green Fluorescent Proteins/metabolism , HeLa Cells , Humans , Rats , Time Factors
17.
Article in English | MEDLINE | ID: mdl-16511138

ABSTRACT

HscA is a constitutively expressed Hsp70 that interacts with the iron-sulfur cluster assembly protein IscU. Crystals of a truncated form of HscA (52 kDa; residues 17-505) grown in the presence of an IscU-recognition peptide, WELPPVKI, have been obtained by hanging-drop vapor diffusion using ammonium sulfate as the precipitant. A complete native X-ray diffraction data set was collected from a single crystal at 100 K to a resolution of 2.9 A. The crystal belongs to the orthorhombic space group P2(1)2(1)2(1), with unit-cell parameters a = 158.35, b = 166.15, c = 168.26 A, and contains six molecules per asymmetric unit. Phases were determined by molecular replacement using the nucleotide-binding domain from DnaK and the substrate-binding domain from HscA as models. This is the first reported crystallization of an Hsp70 containing both nucleotide- and substrate-binding domains.


Subject(s)
Escherichia coli Proteins/chemistry , Escherichia coli/metabolism , HSP70 Heat-Shock Proteins/chemistry , Ammonium Sulfate/pharmacology , Crystallization , Crystallography, X-Ray , Models, Molecular , Molecular Chaperones/chemistry , Nucleotides/chemistry , Plasmids/metabolism , Protein Conformation , Protein Structure, Tertiary , Recombinant Proteins/chemistry , X-Ray Diffraction
SELECTION OF CITATIONS
SEARCH DETAIL
...