Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
Int J Mol Sci ; 23(19)2022 Oct 01.
Article in English | MEDLINE | ID: mdl-36232909

ABSTRACT

Tau hyperphosphorylation has been linked directly to the formation of toxic neurofibrillary tangles (NFTs) in tauopathies, however, prior to NFT formation, the sequence of pathological events involving tau phosphorylation remains unclear. Here, the effect of glycogen synthase kinase 3ß (GSK3ß) on tau pathology was examined independently for each step of transcellular propagation; namely, tau intracellular aggregation, release, cellular uptake and seeding activity. We find that overexpression of GSK3ß-induced phosphorylated 0N4R tau led to a higher level of tau oligomerization in SH-SY5Y neuroblastoma cells than wild type 0N4R, as determined by several orthogonal assays. Interestingly, the presence of GSK3ß also enhanced tau release. Further, we demonstrated that cells endocytosed more monomeric tau protein when pre-phosphorylated by GSK3ß. Using an extracellular vesicle (EVs)-assisted tau neuronal delivery system, we show that exosomal GSK3ß-phosphorylated tau, when added to differentiated SH-SY5Y cells, induced more efficient tau transfer, showing much higher total tau levels and increased tau aggregate formation as compared to wild type exosomal tau. The role of a primary tau phosphorylation site targeted by microtubule-affinity regulating kinases (MARKs), Ser262, was tested by pseudo-phosphorylation using site-directed mutagenesis to aspartate (S262D). S262D tau overexpression significantly enhanced tau release and intracellular tau accumulation, which were concurrent with the increase of pathological states of tau, as determined by immunodetection. Importantly, phosphorylation-induced tau accumulation was augmented by co-transfecting S262D tau with GSK3ß, suggesting a possible interplay between Ser262 phosphorylation and GSK3ß activity in tau pathology. Lastly, we found that pre-treatment of cells with amyloid-ß (Aß) further tau phosphorylation and accumulation when Ser262 pre-phosphorylation was present, suggesting that S262 may be a primary mediator of Aß-induced tau toxicity. These findings provide a potential therapeutic target for treating tau-related disorders by targeting specific phospho-tau isoforms and further elucidate the GSK3ß-mediated pathological seeding mechanisms.


Subject(s)
Neuroblastoma , tau Proteins , Amyloid beta-Peptides/metabolism , Aspartic Acid/metabolism , Glycogen Synthase Kinase 3 beta/genetics , Glycogen Synthase Kinase 3 beta/metabolism , Humans , Neuroblastoma/genetics , Neuroblastoma/pathology , Phosphorylation , tau Proteins/genetics , tau Proteins/metabolism
2.
Molecules ; 27(11)2022 May 31.
Article in English | MEDLINE | ID: mdl-35684466

ABSTRACT

BACKGROUND: Membrane cholesterol dysregulation has been shown to alter the activity of the adenosine A2A receptor (A2AR), a G protein-coupled receptor, thereby implicating cholesterol levels in diseases such as Alzheimer's and Parkinson's. A limited number of A2AR crystal structures show the receptor interacting with cholesterol, as such molecular simulations are often used to predict cholesterol interaction sites. METHODS: Here, we use experimental methods to determine whether a specific interaction between amino acid side chains in the cholesterol consensus motif (CCM) of full length, wild-type human A2AR, and cholesterol modulates activity of the receptor by testing the effects of mutational changes on functional consequences, including ligand binding, G protein coupling, and downstream activation of cyclic AMP. RESULTS AND CONCLUSIONS: Our data, taken with previously published studies, support a model of receptor state-dependent binding between cholesterol and the CCM, whereby cholesterol facilitates both G protein coupling and downstream signaling of A2AR.


Subject(s)
Adenosine , Receptor, Adenosine A2A , Adenosine/pharmacology , Adenosine A2 Receptor Agonists/pharmacology , Carrier Proteins , Cholesterol/metabolism , Cyclic AMP/metabolism , Humans , Receptor, Adenosine A2A/metabolism
3.
Biomedicines ; 9(2)2021 Feb 14.
Article in English | MEDLINE | ID: mdl-33672982

ABSTRACT

Tauopathies represent a group of neurodegenerative diseases including Alzheimer's disease (AD) that are characterized by the deposition of filamentous tau aggregates in the brain. The pathogenesis of tauopathies starts from the formation of toxic 'tau seeds' from hyperphosphorylated tau monomers. The presence of specific phosphorylation sites and heat shock protein 90 facilitates soluble tau protein aggregation. Transcellular propagation of pathogenic tau into synaptically connected neuronal cells or adjacent glial cells via receptor-mediated endocytosis facilitate disease spread through the brain. While neuroprotective effects of glial cells-including phagocytotic microglial and astroglial phenotypes-have been observed at the early stage of neurodegeneration, dysfunctional neuronal-glial cellular communication results in a series of further pathological consequences as the disease progresses, including abnormal axonal transport, synaptic degeneration, and neuronal loss, accompanied by a pro-inflammatory microenvironment. Additionally, the discovery of microtubule-associated protein tau (MAPT) gene mutations and the strongest genetic risk factor of tauopathies-an increase in the presence of the ε2 allele of apolipoprotein E (ApoE)-provide important clues to understanding tau pathology progression. In this review, we describe the crucial signaling pathways and diverse cellular contributors to the progression of tauopathies. A systematic understanding of disease pathogenesis provides novel insights into therapeutic targets within altered signaling pathways and is of great significance for discovering effective treatments for tauopathies.

4.
Biochim Biophys Acta Biomembr ; 1861(4): 760-767, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30629951

ABSTRACT

Cholesterol has been shown to modulate the activity of multiple G Protein-coupled receptors (GPCRs), yet whether cholesterol acts through specific interactions, indirectly via modifications to the membrane, or via both mechanisms is not well understood. High-resolution crystal structures of GPCRs have identified bound cholesterols; based on a ß2-adrenergic receptor (ß2AR) structure bound to cholesterol and the presence of conserved amino acids in class A receptors, the cholesterol consensus motif (CCM) was identified. Here in mammalian cells expressing the adenosine A2A receptor (A2AR), ligand dependent production of cAMP is reduced following membrane cholesterol depletion with methyl-beta-cyclodextrin (MßCD), indicating that A2AR signaling is dependent on cholesterol. In contrast, ligand binding is not dependent on cholesterol depletion. All-atom molecular simulations suggest that cholesterol interacts specifically with the CCM when the receptor is in an active state, but not when in an inactive state. Taken together, the data support a model of receptor state-dependent binding between cholesterol and the CCM, which could facilitate both G-protein coupling and downstream signaling of A2AR.


Subject(s)
Cholestyramine Resin , Molecular Dynamics Simulation , Receptor, Adenosine A2A , Signal Transduction , Cholestyramine Resin/chemistry , Cholestyramine Resin/metabolism , HEK293 Cells , Humans , Receptor, Adenosine A2A/chemistry , Receptor, Adenosine A2A/metabolism , beta-Cyclodextrins/chemistry
5.
Biotechnol J ; 12(1)2017 Jan.
Article in English | MEDLINE | ID: mdl-27933725

ABSTRACT

Recombinant proteins offer many therapeutic advantages unavailable in traditional small molecule drugs, but the need for cellular versus chemical synthesis complicates production. Avenues for producing therapeutic biologics are continuously expanding, and developments in biochemistry, cell biology, and bioengineering fuel new discoveries that promise safer, more efficient, and cheaper drugs for consumers. Numerous approaches to express recombinant proteins exist, but Escherichia coli, Saccharomyces cerevisiae, and mammalian systems (e.g. Chinese hamster ovary cells, CHO) are the most widely utilized. Improvements to production in these hosts have focused on novel expression cassettes, cell line modifications, engineering secretion pathways, and media design. Here, we describe recent developments for improving protein production in E. coli, S. cerevisiae, and CHO systems and compare recent advancements to previous knowledge in the field. With the expanding importance and prevalence of protein therapeutics, these improvements will serve as the framework for future discoveries.


Subject(s)
Cell Engineering/methods , Escherichia coli/genetics , Protein Engineering/methods , Saccharomyces cerevisiae/genetics , Animals , Apoptosis/physiology , Cell Line , Culture Media/chemistry , Culture Media/pharmacology , Escherichia coli/metabolism , Humans , Mammals , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Saccharomyces cerevisiae/metabolism
6.
SLAS Discov ; 22(2): 166-175, 2017 02.
Article in English | MEDLINE | ID: mdl-27577981

ABSTRACT

Ligand binding plays a fundamental role in stimulating the downstream signaling of membrane receptors. Here, ligand-binding kinetics of the full-length human adenosine A2A receptor (A2AR) reconstituted in detergent micelles were measured using a fluorescently labeled ligand via fluorescence anisotropy. Importantly, to optimize the signal-to-noise ratio, these experiments were conducted in the ligand depletion regime. In the ligand depletion regime, the assumptions used to determine analytical solutions for one-site binding models for either one or two ligands in competition are no longer valid. We therefore implemented a numerical solution approach to analyze kinetic binding data as experimental conditions approach the ligand depletion regime. By comparing the results from the numerical and the analytical solutions, we highlight the ligand-receptor ratios at which the analytical solution begins to lose predictive accuracy. Using the numerical solution approach, we determined the kinetic rate constants of the fluorescent ligand, FITC-APEC, and those for three unlabeled ligands using competitive association experiments. The association and dissociation rate constants of the unlabeled ligands determined from the competitive association experiments were then independently validated using competitive dissociation data. Based on this study, a numerical solution is recommended to determine kinetic ligand-binding parameters for experiments conducted in the ligand-depletion regime.


Subject(s)
Ligands , Protein Transport/genetics , Receptor, Adenosine A2A/chemistry , Binding Sites , Binding, Competitive , Fluorescence Polarization , Humans , Kinetics , Protein Binding , Receptor, Adenosine A2A/metabolism
7.
Protein Expr Purif ; 124: 62-7, 2016 08.
Article in English | MEDLINE | ID: mdl-27241126

ABSTRACT

The adenosine A2A receptor (A2AR) is a much-studied class A G protein-coupled receptor (GPCR). For biophysical studies, A2AR is commonly purified in a detergent mixture of dodecylmaltoside (DDM), 3-(3-cholamidopropyl) dimethylammoniopropane sulfonate (CHAPS), and cholesteryl hemisuccinate (CHS). Here we studied the effects of CHAPS on the ligand binding activity and stability of wild type, full-length human A2AR. We also tested the cholesterol requirement for maintaining the active conformation of the receptor when solubilized in detergent micelles. To this end, the receptor was purified using DDM, DDM/CHAPS, or the short hydrocarbon chain lipid 1,2-dihexanoyl-sn-glycero-3-phosphocholine (DHPC, di-6:0PC). After solubilization in DDM, DDM/CHAPS, or DHPC micelles, although A2AR was found to retain its native-like fold, its binding ability was significantly compromised compared to DDM or DDM/CHAPS with CHS. It therefore appears that although cholesterol is not needed for A2AR to retain a native-like, α-helical conformation, it may be a critical component for high affinity ligand binding. Further, this result suggests that the conformational differences between the active and inactive protein may be so subtle that commonly used spectroscopic methods are unable to differentiate between the two forms, highlighting the need for activity measurements. The studies presented in this paper also underline the importance of the protein's purification history; i.e., detergents that interact with the protein during purification affect the ligand binding properties of the receptor in an irreversible manner.


Subject(s)
Cholesterol Esters/chemistry , Cholic Acids/chemistry , Phospholipid Ethers/chemistry , Receptor, Adenosine A2A/chemistry , Receptor, Adenosine A2A/isolation & purification , Recombinant Proteins/chemistry , Humans , Protein Structure, Secondary
8.
Protein Eng Des Sel ; 27(5): 157-67, 2014 May.
Article in English | MEDLINE | ID: mdl-24722670

ABSTRACT

Non-native protein aggregation is a prevalent problem occurring in many biotechnological manufacturing processes and can compromise the biological activity of the target molecule or induce an undesired immune response. Additionally, some non-native aggregation mechanisms lead to amyloid fibril formation, which can be associated with debilitating diseases. For natively folded proteins, partial or complete unfolding is often required to populate aggregation-prone conformational states, and therefore one proposed strategy to mitigate aggregation is to increase the free energy for unfolding (ΔGunf) prior to aggregation. A computational design approach was tested using human γD crystallin (γD-crys) as a model multi-domain protein. Two mutational strategies were tested for their ability to reduce/increase aggregation rates by increasing/decreasing ΔGunf: stabilizing the less stable domain and stabilizing the domain-domain interface. The computational protein design algorithm, RosettaDesign, was implemented to identify point variants. The results showed that although the predicted free energies were only weakly correlated with the experimental ΔGunf values, increased/decreased aggregation rates for γD-crys correlated reasonably well with decreases/increases in experimental ΔGunf, illustrating improved conformational stability as a possible design target to mitigate aggregation. However, the results also illustrate that conformational stability is not the sole design factor controlling aggregation rates of natively folded proteins.


Subject(s)
Protein Engineering/methods , Protein Multimerization , gamma-Crystallins/chemistry , gamma-Crystallins/genetics , Humans , Models, Molecular , Mutation , Protein Stability , Protein Structure, Tertiary , Protein Unfolding/drug effects , Thermodynamics
9.
Biochemistry ; 50(5): 628-39, 2011 Feb 08.
Article in English | MEDLINE | ID: mdl-21184609

ABSTRACT

γD crystallin is a natively monomeric eye-lens protein that is associated with hereditary juvenile cataract formation. It is an attractive model system as a multidomain Greek-key protein that aggregates through partially folded intermediates. Point mutations M69Q and S130P were used to test (1) whether the protein-design algorithm RosettaDesign would successfully predict mutants that are resistant to aggregation when combined with informatic sequence-based predictors of peptide aggregation propensity and (2) how the mutations affected relative unfolding free energies (ΔΔG(un)) and intrinsic aggregation propensity (IAP). M69Q was predicted to have ΔΔG(un) ≫ 0, without significantly affecting IAP. S130P was predicted to have ΔΔG(un) ∼ 0 but with reduced IAP. The stability, conformation, and aggregation kinetics in acidic solution were experimentally characterized and compared for the variants and wild-type (WT) protein using circular dichroism and intrinsic fluorescence spectroscopy, calorimetric and chemical unfolding, thioflavin-T binding, chromatography, static laser light scattering, and kinetic modeling. Monomer secondary and tertiary structures of both variants were indistinguishable from WT, while ΔΔG(un) > 0 for M69Q and ΔΔG(un) < 0 for S130P. Surprisingly, despite being the least conformationally stable, S130P was the most resistant to aggregation, indicating a significant decrease of its IAP compared to WT and M69Q.


Subject(s)
Point Mutation , gamma-Crystallins/chemistry , gamma-Crystallins/genetics , Amino Acid Sequence , Circular Dichroism , Computer-Aided Design , Humans , Kinetics , Molecular Sequence Data , Protein Conformation , Protein Folding , Protein Stability , Thermodynamics , gamma-Crystallins/metabolism
10.
Biotechnol Bioeng ; 104(1): 20-9, 2009 Sep 01.
Article in English | MEDLINE | ID: mdl-19415776

ABSTRACT

Heterologous protein expression can easily overwhelm a cell's capacity to properly fold protein, initiating the unfolded protein response (UPR), and resulting in a loss of protein expression. In the current model of the UPR, the chaperone BiP modulates the activation of the UPR due to its interactions with the signaling protein Ire1p and newly synthesized proteins. In this research, 4-4-20 scFv variants were generated by rational design to alter BiP binding to newly synthesized scFv proteins or via directed evolution aimed at improved secretion. Interestingly, the predicted BiP binding ability did not correlate significantly with the UPR. However, pulse-chase analysis of scFv fate revealed that mutants with a decreased ER residence time were more highly secreted, indicating that improved protein folding was more likely the cause for improved secretion. In fact, decreased secretion correlated with increased binding by BiP, as determined by co-immune precipitation studies. This suggests that the algorithm is not useful for in vivo prediction of variants, and that in vivo screens are more effective for finding variants with improved properties.


Subject(s)
Antibodies/metabolism , Recombinant Proteins/metabolism , Saccharomyces/metabolism , Antibodies/genetics , Fungal Proteins/metabolism , HSP70 Heat-Shock Proteins/metabolism , Immunoprecipitation/methods , Models, Molecular , Protein Binding , Protein Folding , Protein Structure, Tertiary , Protein Transport , Recombinant Proteins/genetics
11.
Biochem J ; 419(3): 595-602, 2009 May 01.
Article in English | MEDLINE | ID: mdl-19196242

ABSTRACT

TSP (P22 tailspike protein) is a well-established model system for studying the folding and assembly of oligomeric proteins, and previous studies have documented both in vivo and in vitro folding intermediates using this protein. Especially important is the C-terminus of TSP, which plays a critical role in the assembly and maturation of the protrimer intermediate to its final trimeric form. In the present study, we show that by grafting the C-terminus of TSP on to the monomeric MBP (maltose-binding protein), the resulting chimaera (MBP-537) is a trimeric protein. Moreover, Western blot studies (using an anti-TSP antibody) indicate that the TSP C-terminus in the MBP-537 chimaera has the same conformation as the native TSP. The oligomerization of the MBP-537 chimaera appears to involve hydrophobic interactions and a refolding sequence, both of which are analogous to the native TSP. These results underscore the importance of the TSP C-terminus in the assembly of the mature trimer and demonstrate its potential utility as a model to study the folding and assembly of the TSP C-terminus in isolation.


Subject(s)
Bacteriophage P22/chemistry , Protein Multimerization , Viral Tail Proteins/chemistry , Antibodies/immunology , Blotting, Western , Carrier Proteins/metabolism , Centrifugation , Electrophoresis, Polyacrylamide Gel , Glycoside Hydrolases , Hydrophobic and Hydrophilic Interactions , Maltose-Binding Proteins , Mutant Proteins/metabolism , Mutation/genetics , Protein Folding , Protein Structure, Secondary , Protein Structure, Tertiary , Recombinant Fusion Proteins/metabolism , Viral Tail Proteins/immunology , Viral Tail Proteins/metabolism
12.
Biophys J ; 95(8): 3535-58, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18641066

ABSTRACT

A top-down approach to mechanistic modeling of biological systems is presented and exemplified with the development of a hypothesis-driven mathematical model for single-chain antibody fragment (scFv) folding in Saccharomyces cerevisiae by mediators BiP and PDI. In this approach, model development starts with construction of the most basic mathematical model--typically consisting of predetermined or newly-elucidated biological behavior motifs--capable of reproducing desired biological behaviors. From this point, mechanistic detail is added incrementally and systematically, and the effects of each addition are evaluated. This approach follows the typical progression of experimental data availability in that higher-order, lumped measurements are often more prevalent initially than specific, mechanistic ones. It also necessarily provides the modeler with insight into the structural requirements and performance capabilities of the resulting detailed mechanistic model, which facilitates further analysis. The top-down approach to mechanistic modeling identified three such requirements and a branched dependency-degradation competition motif critical for the scFv folding model to reproduce experimentally observed scFv folding dependencies on BiP and PDI and increased production when both species are overexpressed and promoted straightforward prediction of parameter dependencies. It also prescribed modification of the guiding hypothesis to capture BiP and PDI synergy.


Subject(s)
Antibodies/chemistry , Antibodies/metabolism , Immunoglobulin Variable Region/chemistry , Immunoglobulin Variable Region/metabolism , Models, Biological , Protein Folding , Computational Biology , Endoplasmic Reticulum Chaperone BiP , Heat-Shock Proteins/metabolism , Kinetics , Molecular Chaperones/metabolism , Protein Disulfide-Isomerases/metabolism , Saccharomyces cerevisiae/enzymology , Saccharomyces cerevisiae/metabolism
13.
Protein Expr Purif ; 58(2): 342-55, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18249008

ABSTRACT

Heterotrimeric G proteins relay signals from G protein-coupled receptors (GPCRs) to the interior of the cell. The signaling cascades induced by G protein activation control a wide range of cellular processes. The alpha subunit is believed to determine which G protein couples to each GPCR, and is the primary determinant of the type of signal transmitted. Several members of the G(alpha) family have been expressed in active form in Escherichia coli. However, production levels of these proteins are limited: in most cases only approximately 10% of total G(alpha) protein expressed is active; the rest accumulates in inclusion bodies. Although G(ialpha) has been readily expressed in soluble form (to 10 mg/L), other alpha subunits are minimally soluble, and many are exclusively expressed to inclusion bodies. Previous efforts to solubilize and refold G(alpha) from inclusion bodies have not been successful. Here we did a thorough study of the characteristics of G(alpha) subunits (human G(ialpha(1)), human G(salpha(short)), human G(11alpha) and human G(talpha(cone))), solubilized and purified from inclusion bodies. We find that we can obtain soluble protein both by on-column and rapid-dilution techniques. Comparison to native, soluble G(ialpha) expressed from E. coli showed that although the refolded G(alpha) subunits were soluble and retained partial alpha-helicity characteristic of the native, folded G(alpha) subunit, they did not bind GDP or GTP as effectively as native protein. We conclude that the refolded G(ialpha) protein has a native-like secondary structure, but is predominantly in a molten globular state.


Subject(s)
Escherichia coli/metabolism , GTP-Binding Protein alpha Subunits/chemistry , Inclusion Bodies/metabolism , Circular Dichroism , GTP-Binding Protein alpha Subunits/isolation & purification , Humans , Protein Folding , Protein Structure, Tertiary , Spectrophotometry, Ultraviolet
14.
Biotechnol Prog ; 23(3): 540-7, 2007.
Article in English | MEDLINE | ID: mdl-17397185

ABSTRACT

G protein-coupled receptors (GPCRs) are an important, medically relevant class of integral membrane proteins. Laboratories throughout all disciplines of science devote time and energy into developing practical methods for the discovery, isolation, and characterization of these proteins. Since the crystal structure of rhodopsin was solved 6 years ago, the race to determine high-resolution structures of more GPCRs has gained momentum. Since certain GPCRs are currently produced at sufficient levels for X-ray crystallography trials, it is speculated that heterologous expression of GPCRs may no longer be a bottleneck in obtaining crystal structures. This Review focuses on the current approaches in heterologous expression of GPCRs and explores the problems associated with obtaining crystal structures from GPCRs expressed in different systems. Although milligram amounts of certain GPCRs are attainable, the majority of GPCRs are still either produced at very low levels or not at all. Developing reliable expression techniques for GPCRs is still a major priority for the structural characterization of GPCRs.


Subject(s)
Crystallography, X-Ray/methods , Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/metabolism , Animals , Escherichia coli/genetics , Gene Expression , Receptors, G-Protein-Coupled/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Yeasts/genetics
15.
Biotechnol Prog ; 23(2): 364-9, 2007.
Article in English | MEDLINE | ID: mdl-17315887

ABSTRACT

Although manipulation of the endoplasmic reticulum (ER) folding environment in the yeast Saccharomyces cerevisiae has been shown to increase the secretory productivity of recombinant proteins, the cellular interactions and processes of native enzymes and chaperones such as protein disulfide isomerase (PDI) are still unclear. Previously, we reported that overexpression of the ER chaperone PDI enabled up to a 3-fold increase in secretion levels of the Pyrococcus furiosus beta-glucosidase in the yeast S. cerevisiae. This result was surprising since beta-glucosidase contains only one cysteine per monomer and no disulfide bonds. Two possible mechanisms were proposed: PDI either forms a transient disulfide bond with the lone cysteine residue of the nascent beta-glucosidase during the folding and assembly process or acts as a chaperone to aid in proper folding. To discern between the two mechanisms, the single cysteine residue was mutated to serine, and the secretion of the two protein variants was determined. The serine mutant still showed increased secretion in vivo when PDI levels were elevated. When the folding bottleneck is removed by increasing expression temperatures to 37 degrees C rather than 30 degrees C, PDI no longer has an improvement on secretion. These results suggest that, unexpectedly, PDI acts in a chaperone-like capacity or possibly cooperates with the cell's folding or degradation mechanisms regardless of whether the protein is redox-active.


Subject(s)
Protein Disulfide-Isomerases/genetics , Protein Disulfide-Isomerases/metabolism , Protein Engineering/methods , Pyrococcus furiosus/physiology , Saccharomyces cerevisiae/physiology , beta-Glucosidase/genetics , beta-Glucosidase/metabolism , Genetic Enhancement/methods , Oxidation-Reduction , Recombinant Proteins/metabolism
16.
Extremophiles ; 11(1): 179-89, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17072686

ABSTRACT

To elucidate determinants of thermostability and folding pathways of the intrinsically stable proteins from extremophilic organisms, we are studying beta-glucosidase from Pyrococcus furiosus. Using fluorescence and circular dichroism spectroscopy, we have characterized the thermostability of beta-glucosidase at 90 degrees C, the lowest temperature where full unfolding is achieved with urea. The chemical denaturation profile reveals that this homotetrameric protein unfolds at 90 degrees C with an overall DeltaG degrees of approximately 20 kcal mol(-1). The high temperatures needed to chemically denature P. furiosus beta-glucosidase and the large DeltaG degrees of unfolding at high temperatures shows this to be one of the most stable proteins yet characterized. Unfolding proceeds via a three-state pathway that includes a stable intermediate species. Stability of the native and intermediate forms is concentration dependent, and we have identified a dimeric assembly intermediate using high temperature native gel electrophoresis. Based on this data, we have developed a model for the denaturation of beta-glucosidase in which the tetramer dissociates to partially folded dimers, followed by the coupled dissociation and denaturation of the dimers to unfolded monomers. The extremely high stability is thus derived from a combination of oligomeric interactions and subunit folding.


Subject(s)
Archaeal Proteins/chemistry , Pyrococcus furiosus/enzymology , beta-Glucosidase/chemistry , Archaeal Proteins/genetics , Archaeal Proteins/metabolism , Circular Dichroism , Cloning, Molecular , Dimerization , Enzyme Stability , Guanidine/chemistry , Models, Molecular , Protein Denaturation , Protein Folding , Protein Structure, Quaternary , Protein Subunits , Recombinant Proteins/chemistry , Spectrometry, Fluorescence , Temperature , Urea/chemistry , beta-Glucosidase/genetics , beta-Glucosidase/metabolism
17.
Protein Expr Purif ; 52(2): 348-55, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17166740

ABSTRACT

G-protein coupled receptors (GPCRs) are seven transmembrane helical proteins involved in cell signaling and response. They are targets for many existing therapeutic agents, and numerous drug discovery efforts. Production of large quantities of these receptors for drug screening and structural biology remains challenging. To address this difficulty, we sought to express genes for several human GPCRs in Escherichia coli. For most of the receptors, expression was poor, and was not markedly improved even in strains designed to compensate for differences in codon bias between human and E. coli genes. However, the gene for human NK(1) receptor (hNK(1)R) was expressed in large quantities as inclusion bodies in E. coli. The inclusion bodies were not soluble in chemical denaturants such as guanidine chloride or urea, but were soluble in ionic detergents such as SDS, and the zwitterionic detergent fos-choline. Using immobilized metal affinity chromatography, we purified milligram amounts of hNK(1)R. Although inactive in ligand-binding assays, purified hNK(1)R in fos-choline micelles appeared to have a high content of alpha-helix, and was well-behaved in solution. Thus this protein is suitable for additional biophysical characterization and refolding studies.


Subject(s)
Gene Expression/physiology , Receptors, G-Protein-Coupled/metabolism , Receptors, Neurokinin-1/metabolism , Cloning, Molecular , Escherichia coli/genetics , Escherichia coli/metabolism , Humans , Ligands , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/isolation & purification , Receptors, Neurokinin-1/chemistry , Receptors, Neurokinin-1/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
18.
Protein Sci ; 15(7): 1791-3, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16751612

ABSTRACT

Each chain of the native trimeric P22 tailspike protein has eight cysteines that are reduced and buried in its hydrophobic core. However, disulfide bonds have been observed in the folding pathway and they are believed to play a critical role in the registration of the three chains. Interestingly, in the presence of sodium dodecyl sulfate (SDS) only monomeric chains, rather than disulfide-linked oligomers, have been observed from a mixture of folding intermediates. Here we show that when the oligomeric folding intermediates were separated from the monomer by native gel electrophoresis, the reduction of intermolecular disulfide bonds did not occur in the subsequent second-dimension SDS-gel electrophoresis. This result suggests that when tailspike monomer is present in free solution with SDS, the partially unfolded tailspike monomer can facilitate the reduction of disulfide bonds in the tailspike oligomers.


Subject(s)
Disulfides/chemistry , Sodium Dodecyl Sulfate/pharmacology , Viral Tail Proteins/chemistry , Dimerization , Electrophoresis, Polyacrylamide Gel , Glycoside Hydrolases , Protein Folding
19.
Protein Expr Purif ; 46(2): 204-11, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16289981

ABSTRACT

The yeast Saccharomyces cerevisiae was used to express a medically relevant G-protein coupled receptor (GPCR), the human adenosine (A2a) receptor, with a C-terminal green fluorescent protein (GFP) fusion tag. In prior studies, we established an expression system for A2a-GFP. Here, we quantified the total A2a-GFP expression levels by correlating GFP levels as detected by fluorescence and densitometry to A2a-GFP molecules overexpressed in the system. We also quantified A2a-GFP functional levels by classical radioligand binding assays. Approximately, 120,000 functional A2a-GFP molecules per cell were present on the plasma membrane as determined by radioligand binding. Using whole cell GFP fluorescence, 340,000 A2a-GFP molecules per cell were detected; approximately 70% of those molecules were plasma membrane localized, as determined by using confocal microscopy analysis. These results show that a significant portion of the total expressed protein is functional. In addition, the quick and inexpensive whole cell fluorescence appears to provide a good approximation of functional receptor numbers for this case. Importantly, the amount of functionally expressed A2a-GFP per culture ( approximately 4 mg/L) is among the highest reported for any GPCR in any expression system.


Subject(s)
Receptor, Adenosine A2A/biosynthesis , Recombinant Fusion Proteins/biosynthesis , Saccharomyces cerevisiae/growth & development , Green Fluorescent Proteins/biosynthesis , Green Fluorescent Proteins/genetics , Humans , Receptor, Adenosine A2A/genetics , Recombinant Fusion Proteins/genetics , Saccharomyces cerevisiae/genetics
20.
Protein Sci ; 14(9): 2333-43, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16081648

ABSTRACT

The P22 tailspike protein folds by forming a folding competent monomer species that forms a dimeric, then a non-native trimeric (protrimer) species by addition of folding competent monomers. We have found three residues, R549, R563, and D572, which play a critical role in both the stability of the native tailspike protein and assembly and maturation of the protrimer. King and colleagues reported previously that substitution of R563 to glutamine inhibited protrimer formation. We now show that the R549Q and R563K variants significantly delay the protrimer-to-trimer transition both in vivo and in vitro. Previously, variants that destabilize intermediates have shown wild-type chemical stability. Interestingly, both the R549Q and R563K variants destabilize the tailspike trimer in guanidine denaturation studies, indicating that they represent a new class of tailspike folding variants. R549Q has a midpoint of unfolding at 3.2M guanidine, compared to 5.6M for the wild-type tailspike protein, while R563K has a midpoint of unfolding of 1.8 M. R549Q and R563K also denature over a broader pH range than the wild-type tailspike protein and both proteins have increased sensitivity to pH during refolding, suggesting that both residues are involved in ionic interactions. Our model is that R563 and D572 interact to stabilize the adjacent turn, aiding the assembly of the dimer and protrimer species. We believe that the interaction between R563 and D572 is also critical following assembly of the protrimer to properly orient D572 in order to form a salt bridge with R549 during protrimer maturation.


Subject(s)
Viral Tail Proteins/chemistry , Amino Acid Substitution , Amino Acids/chemistry , Amino Acids/genetics , Glycoside Hydrolases , Hydrogen-Ion Concentration , Mutagenesis, Site-Directed , Protein Conformation , Protein Denaturation , Protein Folding , Viral Tail Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...