Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 267
Filter
1.
Nat Chem Biol ; 17(11): 1148-1156, 2021 11.
Article in English | MEDLINE | ID: mdl-34556859

ABSTRACT

The unfolded protein response (UPR) homeostatically matches endoplasmic reticulum (ER) protein-folding capacity to cellular secretory needs. However, under high or chronic ER stress, the UPR triggers apoptosis. This cell fate dichotomy is promoted by differential activation of the ER transmembrane kinase/endoribonuclease (RNase) IRE1α. We previously found that the RNase of IRE1α can be either fully activated or inactivated by ATP-competitive kinase inhibitors. Here we developed kinase inhibitors, partial antagonists of IRE1α RNase (PAIRs), that partially antagonize the IRE1α RNase at full occupancy. Biochemical and structural studies show that PAIRs promote partial RNase antagonism by intermediately displacing the helix αC in the IRE1α kinase domain. In insulin-producing ß-cells, PAIRs permit adaptive splicing of Xbp1 mRNA while quelling destructive ER mRNA endonucleolytic decay and apoptosis. By preserving Xbp1 mRNA splicing, PAIRs allow B cells to differentiate into immunoglobulin-producing plasma cells. Thus, an intermediate RNase-inhibitory 'sweet spot', achieved by PAIR-bound IRE1α, captures a desirable conformation for drugging this master UPR sensor/effector.


Subject(s)
Adenosine Triphosphate/pharmacology , Endoribonucleases/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Adenosine Triphosphate/chemistry , Endoribonucleases/metabolism , Humans , Models, Molecular , Molecular Structure , Protein Kinase Inhibitors/chemistry , Protein Serine-Threonine Kinases/metabolism , Protein Unfolding/drug effects
2.
Mol Pharmacol ; 101(2): 95-105, 2022 02.
Article in English | MEDLINE | ID: mdl-34866045

ABSTRACT

Folding-deficient mutants of solute carrier 6 (SLC6) family members have been linked to human diseases. The serotonin transporter [(SERT)/SLC6A4] is an important drug target in the treatment of depression, anxiety, and obsessive-compulsive disorders and-with structural information in several conformational states-one of the best understood transporters. Here, we surmised that thermal unfolding offered a glimpse on the folding energy landscape of SLC6 transporters. We carried out molecular dynamic (MD) simulations to understand the mechanistic basis for enhanced and reduced stability, respectively, of the thermostabilized variant SERT-Y110A/I291A/T439S, which had previously been used for crystallization of human SERT in the outward-facing state, and of the folding-deficient SERT-P601A/G602A. We also examined the hydrophobic mismatch caused by the absence of cholesterol to explore the contribution of cholesterol to protein stability. When compared with wild type SERT, the thermodynamic and kinetic stability of SERT-Y110A/I291A/T439S was enhanced. In the other instances, changes in these two components were not correlated: the mutations in SERT-P601A/G602A led to a drop in thermodynamic but an increase in kinetic stability. The divergence was even more pronounced after cholesterol depletion, which reduced thermodynamic stability but increased the kinetic stability of wild type SERT to a level comparable to that of SERT-Y110A/I291A/T439S. We conclude that the low cholesterol content of the endoplasmic reticulum facilitates progression of the folding trajectory by reducing the energy difference between folding intermediates and the native state. SIGNIFICANCE STATEMENT: Point mutations in solute carrier 6 (SLC6) family members cause folding diseases. The serotonin transporter [(SERT)/SLC6A4] is a target for antidepressants and the best understood SLC6. This study produced molecular dynamics simulations and examined thermal unfolding of wild type and mutant SERT variants to understand their folding energy landscape. In the folding-deficient SERT-P012A/G602A, changes in kinetic and thermodynamic stability were not correlated. Similarly, cholesterol depletion lowered thermodynamic but enhanced kinetic stability. These observations allow for rationalizing the action of pharmacochaperones.


Subject(s)
Cholesterol/metabolism , Mutation/genetics , Protein Unfolding/drug effects , Serotonin Plasma Membrane Transport Proteins/genetics , Serotonin Plasma Membrane Transport Proteins/metabolism , Thermodynamics , Antidepressive Agents/metabolism , Antidepressive Agents/pharmacology , Genetic Variation/genetics , HEK293 Cells , Humans , Kinetics , Molecular Dynamics Simulation , Protein Binding/physiology , Protein Stability/drug effects , Protein Structure, Secondary , Serotonin Plasma Membrane Transport Proteins/chemistry
3.
J Am Chem Soc ; 143(45): 19137-19148, 2021 11 17.
Article in English | MEDLINE | ID: mdl-34739240

ABSTRACT

The assembly and maturation of human immunodeficiency virus type 1 (HIV-1) require proteolytic cleavage of the Gag polyprotein. The rate-limiting step resides at the junction between the capsid protein CA and spacer peptide 1, which assembles as a six-helix bundle (6HB). Bevirimat (BVM), the first-in-class maturation inhibitor drug, targets the 6HB and impedes proteolytic cleavage, yet the molecular mechanisms of its activity, and relatedly, the escape mechanisms of mutant viruses, remain unclear. Here, we employed extensive molecular dynamics (MD) simulations and free energy calculations to quantitatively investigate molecular structure-activity relationships, comparing wild-type and mutant viruses in the presence and absence of BVM and inositol hexakisphosphate (IP6), an assembly cofactor. Our analysis shows that the efficacy of BVM is directly correlated with preservation of 6-fold symmetry in the 6HB, which exists as an ensemble of structural states. We identified two primary escape mechanisms, and both lead to loss of symmetry, thereby facilitating helix uncoiling to aid access of protease. Our findings also highlight specific interactions that can be targeted for improved inhibitor activity and support the use of MD simulations for future inhibitor design.


Subject(s)
Anti-HIV Agents/metabolism , HIV-1/chemistry , Succinates/metabolism , Triterpenes/metabolism , gag Gene Products, Human Immunodeficiency Virus/metabolism , Molecular Dynamics Simulation , Mutation , Phytic Acid/metabolism , Protein Conformation, alpha-Helical/drug effects , Protein Unfolding/drug effects , gag Gene Products, Human Immunodeficiency Virus/chemistry , gag Gene Products, Human Immunodeficiency Virus/genetics
4.
Nucleic Acids Res ; 47(4): 1637-1652, 2019 02 28.
Article in English | MEDLINE | ID: mdl-30649466

ABSTRACT

The DNA binding domain (DBD) of the tumor suppressor p53 is the site of several oncogenic mutations. A subset of these mutations lowers the unfolding temperature of the DBD. Unfolding leads to the exposure of a hydrophobic ß-strand and nucleates aggregation which results in pathologies through loss of function and dominant negative/gain of function effects. Inspired by the hypothesis that structural changes that are associated with events initiating unfolding in DBD are likely to present opportunities for inhibition, we investigate the dynamics of the wild type (WT) and some aggregating mutants through extensive all atom explicit solvent MD simulations. Simulations reveal differential conformational sampling between the WT and the mutants of a turn region (S6-S7) that is contiguous to a known aggregation-prone region (APR). The conformational properties of the S6-S7 turn appear to be modulated by a network of interacting residues. We speculate that changes that take place in this network as a result of the mutational stress result in the events that destabilize the DBD and initiate unfolding. These perturbations also result in the emergence of a novel pocket that appears to have druggable characteristics. FDA approved drugs are computationally screened against this pocket.


Subject(s)
DNA-Binding Proteins/chemistry , Mutant Proteins/chemistry , Small Molecule Libraries/chemistry , Tumor Suppressor Protein p53/chemistry , DNA-Binding Proteins/genetics , Drug Evaluation, Preclinical/methods , Humans , Hydrophobic and Hydrophilic Interactions/drug effects , Models, Molecular , Molecular Dynamics Simulation , Mutant Proteins/genetics , Mutation/genetics , Protein Conformation/drug effects , Protein Domains/drug effects , Protein Domains/genetics , Protein Unfolding/drug effects , Tumor Suppressor Protein p53/genetics
5.
Int J Mol Sci ; 22(7)2021 Mar 30.
Article in English | MEDLINE | ID: mdl-33808390

ABSTRACT

When combined with NMR spectroscopy, high hydrostatic pressure is an alternative perturbation method used to destabilize globular proteins that has proven to be particularly well suited for exploring the unfolding energy landscape of small single-domain proteins. To date, investigations of the unfolding landscape of all-ß or mixed-α/ß protein scaffolds are well documented, whereas such data are lacking for all-α protein domains. Here we report the NMR study of the unfolding pathways of GIPC1-GH2, a small α-helical bundle domain made of four antiparallel α-helices. High-pressure perturbation was combined with NMR spectroscopy to unravel the unfolding landscape at three different temperatures. The results were compared to those obtained from classical chemical denaturation. Whatever the perturbation used, the loss of secondary and tertiary contacts within the protein scaffold is almost simultaneous. The unfolding transition appeared very cooperative when using high pressure at high temperature, as was the case for chemical denaturation, whereas it was found more progressive at low temperature, suggesting the existence of a complex folding pathway.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Magnetic Resonance Spectroscopy/methods , Protein Unfolding/drug effects , Humans , Kinetics , Models, Molecular , Protein Conformation/drug effects , Protein Conformation, alpha-Helical/physiology , Protein Denaturation , Protein Domains , Temperature , Thermodynamics
6.
Biochemistry ; 59(31): 2870-2881, 2020 08 11.
Article in English | MEDLINE | ID: mdl-32786888

ABSTRACT

The properties of enzymes packaged within the coat protein shell of virus-like particles (VLPs) were studied to provide a comprehensive assessment of such factors. Such entrainment did not seem to perturb enzyme function, but it did significantly enhance enzyme stability against several denaturing stimuli such as heat, organic solvents, and chaotropic agents. This improvement in performance was found to be general and independent of the number of independent subunits required and of the number of catalytically active enzymes packaged. Packaged enzymes were found by measurements of intrinsic tryptophan fluorescence to retain some of their native folded structure even longer than their catalytic activity, suggesting that protein folding is a significant component of the observed catalytic benefits. While we are unable to distinguish between kinetic and thermodynamic effects - including inhibition of enzyme unfolding, acceleration of refolding, and biasing of folding equilibria - VLP packaging appears to represent a useful general strategy for the stabilization of enzymes that operate on diffusible substrates and products.


Subject(s)
Enzymes/metabolism , Leviviridae , Virion/metabolism , Biocatalysis , Enzyme Stability/drug effects , Enzymes/chemistry , Protein Unfolding/drug effects , Solvents/pharmacology
7.
Biochemistry ; 59(39): 3650-3659, 2020 10 06.
Article in English | MEDLINE | ID: mdl-32924445

ABSTRACT

Misfolding of Cu, Zn superoxide dismutase (SOD1) variants may lead to protein aggregation and ultimately amyotrophic lateral sclerosis (ALS). The mechanism and protein conformational changes during this process are complex and remain unclear. To study SOD1 variant aggregation at the molecular level and in solution, we chemically induced aggregation of a mutant variant (G93A SOD1) with trifluoroethanol (TFE) and used both native mass spectrometry (MS) to analyze the intact protein and fast photochemical oxidation of proteins (FPOP) to characterize the structural changes induced by TFE. We found partially unfolded G93A SOD1 monomers prior to oligomerization and identified regions of the N-terminus, C-terminus, and strands ß5, ß6 accountable for the partial unfolding. We propose that exposure of hydrophobic interfaces of these unstructured regions serves as a precursor to aggregation. Our results provide a possible mechanism and molecular basis for ALS-linked SOD1 misfolding and aggregation.


Subject(s)
Protein Aggregates/drug effects , Protein Unfolding/drug effects , Superoxide Dismutase/chemistry , Trifluoroethanol/pharmacology , Humans , Mass Spectrometry , Models, Molecular , Protein Conformation/drug effects , Protein Footprinting , Spectrometry, Mass, Electrospray Ionization
8.
Biochemistry ; 59(31): 2849-2858, 2020 08 11.
Article in English | MEDLINE | ID: mdl-32667811

ABSTRACT

The sterile α motif, also called the SAM domain, is known to form homo or heterocomplexes that modulate diverse biological functions through the regulation of specific protein-protein interactions. The MAPK pathway of budding yeast Saccharomyces cerevisiae is comprised of a three-tier kinase system akin to mammals. The MAPKKK Ste11 protein of yeast contains a homodimer SAM domain, which is critical for transmitting cues to the downstream kinases. The structural stability of the dimeric Ste11 SAM is maintained by hydrophobic and ionic interactions at the interfacial amino acids. The urea-induced equilibrium-unfolding process of the Ste11 SAM domain is cooperative without evidence of any intermediate states. The native-state H/D exchange under subdenaturing conditions is a useful method for the detection of intermediate states of proteins. In the present study, we investigated the effect of ionic strength on the conformational stability of the dimer using the H/D exchange experiments. The hydrogen exchange behavior of the Ste11 dimer under physiological salt concentrations reveals two partially unfolded metastable intermediate states, which may be generated by a sequential and cooperative unfolding of the five helices present in the domain. These intermediates appear to be significant for the reversible unfolding kinetics via hydrophobic collapse. In contrast, higher ionic concentrations eliminate this cooperative interactions that stabilize the pairs of helices.


Subject(s)
Deuterium Exchange Measurement , MAP Kinase Kinase Kinases/chemistry , Models, Molecular , Nuclear Magnetic Resonance, Biomolecular , Protein Multimerization/drug effects , Saccharomyces cerevisiae Proteins/chemistry , Sodium Chloride/pharmacology , Dose-Response Relationship, Drug , Enzyme Stability/drug effects , Hydrogen Deuterium Exchange-Mass Spectrometry , Protein Structure, Quaternary , Protein Unfolding/drug effects , Urea/pharmacology
9.
J Biol Chem ; 294(45): 16942-16952, 2019 11 08.
Article in English | MEDLINE | ID: mdl-31582563

ABSTRACT

The chemistry of protein-ligand binding is the basis of virtually every biological process. Ligand binding can be essential for a protein to function in the cell by stabilizing or altering the conformation of a protein, particularly for partially or completely unstructured proteins. However, the mechanisms by which ligand binding impacts disordered proteins or influences the role of disorder in protein folding is not clear. To gain insight into this question, the mechanism of folding induced by the binding of a Pro-rich peptide ligand to the SH3 domain of phosphatidylinositol 3-kinase unfolded in the presence of urea has been studied using kinetic methods. Under strongly denaturing conditions, folding was found to follow a conformational selection (CS) mechanism. However, under mildly denaturing conditions, a ligand concentration-dependent switch in the mechanism was observed. The folding mechanism switched from being predominantly a CS mechanism at low ligand concentrations to being predominantly an induced fit (IF) mechanism at high ligand concentrations. The switch in the mechanism manifests itself as an increase in the reaction flux along the IF pathway at high ligand concentrations. The results indicate that, in the case of intrinsically disordered proteins too, the folding mechanism is determined by the concentration of the ligand that induces structure formation.


Subject(s)
Protein Unfolding , Kinetics , Ligands , Phosphatidylinositol 3-Kinases/chemistry , Phosphatidylinositol 3-Kinases/metabolism , Protein Binding/drug effects , Protein Unfolding/drug effects , Urea/pharmacology , src Homology Domains
10.
Biochem Biophys Res Commun ; 526(1): 122-127, 2020 05 21.
Article in English | MEDLINE | ID: mdl-32199617

ABSTRACT

Overloading of the saturated fatty acid (SFA) palmitate induces cardiomyocyte death. The purpose of this study is to elucidate signaling pathways contributing to palmitate-induced cardiomyocyte death. Palmitate-induced cardiomyocyte death was induced in Toll-like receptor 2/4 double-knockdown cardiomyocytes to a similar extent as wild-type cardiomyocytes, while cardiomyocyte death was canceled out by triacsin C, a long-chain acyl-CoA synthetase inhibitor. These results indicated that palmitate induced cytotoxicity after entry and conversion into palmitoyl-CoA. Palmitoyl-CoA is not only degraded by mitochondrial oxidation but also taken up as a component of membrane phospholipids. Palmitate overloading causes cardiomyocyte membrane fatty acid (FA) saturation, which is associated with the activation of endoplasmic reticulum (ER) unfolded protein response (UPR) signaling. We focused on the ER UPR signaling as a possible mechanism of cell death. Palmitate loading activates the UPR signal via membrane FA saturation, but not via unfolded protein overload in the ER since the chemical chaperone 4-phenylbutyrate failed to suppress palmitate-induced ER UPR. The mammalian UPR relies on three ER stress sensors named inositol requiring enzyme-1 (IRE1), PKR-like endoplasmic reticulum kinase (PERK), and activating transcription factor 6 (ATF6). Palmitate loading activated only IRE1 and PERK. Knockdown of PERK did not affect palmitate-induced cardiomyocyte death, while knockdown of IRE1 suppressed palmitate-induced cardiomyocyte death. However, knockdown of X-box binding protein 1 (XBP1), the downstream effector of IRE1, did not affect palmitate-induced cardiomyocyte death. These results were validated by pharmacological inhibitor experiments. In conclusion, we identified that palmitate-induced cardiomyocyte death was triggered by IRE1-mediated signaling independent of XBP1.


Subject(s)
Membrane Proteins/metabolism , Myocytes, Cardiac/pathology , Palmitic Acid/toxicity , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , X-Box Binding Protein 1/metabolism , Animals , Animals, Newborn , Cell Death/drug effects , Cells, Cultured , Endoplasmic Reticulum/drug effects , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Protein Unfolding/drug effects , Rats , Signal Transduction/drug effects
11.
NMR Biomed ; 33(10): e4367, 2020 10.
Article in English | MEDLINE | ID: mdl-32656956

ABSTRACT

The aim of this study was to investigate possible sodium triple-quantum (TQ) signal dependence on pH variation and protein unfolding which may happen in vivo. The model system, composed of bovine serum albumin (BSA), was investigated over a wide pH range of 0.70 to 13.05 and during urea-induced unfolding. In both experimental series, the sodium and BSA concentration were kept constant so that TQ signal changes solely arose from an environmental change. The experiments were performed using unique potential to detect weak TQ signals by implementing a TQ time proportional phase increment pulse sequence. At a pH of 0.70, in which case the effect of the negatively charged groups was minimized, the minimum TQ percentage relative to single-quantum of 1.34% ± 0.05% was found. An increase of the pH up to 13.05 resulted in an increase of the sodium TQ signal by 225%. Urea-induced unfolding of BSA, without changes in pH, led to a smaller increase in the sodium TQ signal of up to 40%. The state of BSA unfolding was verified by fluorescence microscopy. Results of both experiments were well fitted by sigmoid functions. Both TQ signal increases were in agreement with an increase of the availability of negatively charged groups. The results point to vital contributions of the biochemical environment to the TQ MR signals. The sodium TQ signal in vivo could be a valuable biomarker of cell viability, and therefore possible effects of pH and protein unfolding need to be considered for a proper interpretation of changes in sodium TQ signals.


Subject(s)
Magnetic Resonance Spectroscopy , Signal Processing, Computer-Assisted , Sodium/chemistry , Animals , Cattle , Fluorescence , Hydrogen-Ion Concentration , Phantoms, Imaging , Protein Conformation , Protein Unfolding/drug effects , Serum Albumin, Bovine/chemistry , Serum Albumin, Bovine/metabolism , Static Electricity , Urea/pharmacology
12.
Mol Pharm ; 17(8): 2971-2986, 2020 08 03.
Article in English | MEDLINE | ID: mdl-32687367

ABSTRACT

Coformulations containing two therapeutic monoclonal antibodies (mAbs) could offer various benefits like enhanced therapeutic efficacy and better patient compliance. However, there are very few published studies on coformulations and binary mixtures of mAbs. It remains unclear to what extent mAbs with different physicochemical properties can be combined in solution without detrimental effects on protein stability. Here, we present a study including six model mAbs of the IgG1 subclass that are commercially available. In silico and biophysical characterization shows that the proteins have different physicochemical properties. Thus, their combinations represent various scenarios for coformulation development. We prepared all possible binary mixtures of the six mAbs and determined several biophysical parameters that are assessed during early-stage protein drug product development. The measured biophysical parameters are indicative of the conformational protein stability (inflection points of the thermal protein unfolding transitions) and the colloidal protein stability (aggregation onset temperatures and interaction parameter kD from dynamic light scattering). Remarkably, all 15 binary mAb mixtures do not exhibit biophysical parameters that indicate inferior conformational or colloidal stability compared to the least stable mAb in the mixture. Our findings suggest that the coformulation of some therapeutic monoclonal antibodies of the IgG1 subclass could be possible in a straightforward way as severe detrimental effects on the stability of these proteins in binary mixtures were not observed.


Subject(s)
Antibodies, Monoclonal/chemistry , Pharmaceutical Preparations/chemistry , Biophysics/methods , Immunoglobulin G/chemistry , Protein Stability/drug effects , Protein Unfolding/drug effects
13.
Eur Biophys J ; 49(3-4): 267-277, 2020 May.
Article in English | MEDLINE | ID: mdl-32356119

ABSTRACT

The stability of Debaryomyces nepalensis NCYC 3413 xylose reductase, a homodimeric enzyme recombinantly expressed and purified from E. coli Rosetta cells, was studied at different pH ranging from 5.0 to 10.0. Deactivation kinetics at different pH were studied by analyzing residual activity of the recombinant enzyme over time at 40 °C whereas conformational changes and stability dependence were investigated by using circular dichroism and differential scanning calorimetry. Four osmolytes viz. glycerol, sucrose, trehalose and sorbitol were explored for their effect on the deactivation and melting temperatures of the enzyme under neutral and extreme pH conditions. The enzyme was found to be catalytically and structurally stable at pH 7.0 with half-life of 250 min and a melting temperature of 50 °C. It was found that alteration in both secondary and tertiary structures caused enzyme deactivation in acidic pH while increased deactivation rates at alkaline pH was attributed to the variation of tertiary structure over time. Estimated thermodynamic parameters also showed that the enzyme stability was highest at neutral pH with ΔH of 348 kcal/mole and ΔG40 of 9.53 kcal/mole. All four osmolytes were effective in enhancing enzyme stability by several folds at extreme pH with sorbitol being the most efficient, which increased enzyme half-life by 11-fold at pH 10.0 and 8-fold at pH 5.0.


Subject(s)
Aldehyde Reductase/chemistry , Osmosis/drug effects , Protein Unfolding/drug effects , Saccharomycetales/enzymology , Enzyme Stability/drug effects , Hydrogen-Ion Concentration , Thermodynamics
14.
Phys Chem Chem Phys ; 22(35): 19779-19786, 2020 Sep 16.
Article in English | MEDLINE | ID: mdl-32844828

ABSTRACT

Ionic liquids (ILs) are gaining attention as protein stabilizers and refolding additives. However, varying degrees of success with this approach motivates the need to better understand fundamental IL-protein interactions. A combination of experiment and simulation is used to investigate the thermal unfolding of lysozyme in the presence of two imidazolium-based ILs (1-ethyl-3-methylimidazolium ethylsulfate, [EMIM][EtSO4] and 1-ethyl-3-methylimidazolium diethylphosphate, [EMIM][Et2PO4]). Both ILs reduce lysozyme melting temperature Tm, but more gradually than strong denaturants. [EMIM][Et2PO4] lowers lysozyme Tm more readily than [EMIM][EtSO4], as well as requiring less energy to unfold the protein, as determined by the calorimetric enthalpy ΔH. Intrinsic fluorescence measurements indicate that both ILs bind to tryptophan residues in a dynamic mode, and furthermore, molecular dynamics simulations show a high density of [EMIM]+ near lysozyme's Trp62 residue. For both ILs approximately half of the [EMIM]+ cations near Trp62 show perfect alignment of their respective rings. The [EMIM]+ cations, having a "local" effect in binding to tryptophan, likely perturb a critically important Arg-Trp-Arg bridge through favorable π-π and cation-π interactions. Simulations show that the anions, [EtSO4]- and [Et2PO4]-, interact in a "global" manner with lysozyme, due to this protein's strong net positive charge. The anions also determine the local distribution of ions surrounding the protein. [Et2PO4]- is found to have a closer first coordination shell around the protein and stronger Coulomb interactions with lysozyme than [EtSO4]-, which could explain why the former anion is more destabilizing. Patching of ILs to the protein surface is also observed, suggesting there is no universal IL solvent for proteins, and highlighting the complexity of the IL-protein environment.


Subject(s)
Ionic Liquids/chemistry , Muramidase/chemistry , Protein Unfolding/drug effects , Animals , Chickens , Imidazoles/chemistry , Molecular Dynamics Simulation , Organophosphates/chemistry , Protein Stability/drug effects , Thermodynamics , Transition Temperature/drug effects
15.
Nature ; 510(7503): 172-175, 2014 Jun 05.
Article in English | MEDLINE | ID: mdl-24899312

ABSTRACT

Previous studies have established that the folding, structure and function of membrane proteins are influenced by their lipid environments and that lipids can bind to specific sites, for example, in potassium channels. Fundamental questions remain however regarding the extent of membrane protein selectivity towards lipids. Here we report a mass spectrometry approach designed to determine the selectivity of lipid binding to membrane protein complexes. We investigate the mechanosensitive channel of large conductance (MscL) from Mycobacterium tuberculosis and aquaporin Z (AqpZ) and the ammonia channel (AmtB) from Escherichia coli, using ion mobility mass spectrometry (IM-MS), which reports gas-phase collision cross-sections. We demonstrate that folded conformations of membrane protein complexes can exist in the gas phase. By resolving lipid-bound states, we then rank bound lipids on the basis of their ability to resist gas phase unfolding and thereby stabilize membrane protein structure. Lipids bind non-selectively and with high avidity to MscL, all imparting comparable stability; however, the highest-ranking lipid is phosphatidylinositol phosphate, in line with its proposed functional role in mechanosensation. AqpZ is also stabilized by many lipids, with cardiolipin imparting the most significant resistance to unfolding. Subsequently, through functional assays we show that cardiolipin modulates AqpZ function. Similar experiments identify AmtB as being highly selective for phosphatidylglycerol, prompting us to obtain an X-ray structure in this lipid membrane-like environment. The 2.3 Å resolution structure, when compared with others obtained without lipid bound, reveals distinct conformational changes that re-position AmtB residues to interact with the lipid bilayer. Our results demonstrate that resistance to unfolding correlates with specific lipid-binding events, enabling a distinction to be made between lipids that merely bind from those that modulate membrane protein structure and/or function. We anticipate that these findings will be important not only for defining the selectivity of membrane proteins towards lipids, but also for understanding the role of lipids in modulating protein function or drug binding.


Subject(s)
Membrane Lipids/metabolism , Membrane Lipids/pharmacology , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Protein Folding/drug effects , Ammonia/metabolism , Apoproteins/chemistry , Apoproteins/metabolism , Aquaporins/chemistry , Aquaporins/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Cardiolipins/chemistry , Cardiolipins/metabolism , Cardiolipins/pharmacology , Cation Transport Proteins/chemistry , Cation Transport Proteins/metabolism , Crystallography, X-Ray , Escherichia coli/chemistry , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Ion Channels/chemistry , Ion Channels/metabolism , Lipid Bilayers/chemistry , Mass Spectrometry , Membrane Lipids/chemistry , Models, Molecular , Mycobacterium tuberculosis/chemistry , Phosphatidylglycerols/chemistry , Phosphatidylglycerols/metabolism , Phosphatidylglycerols/pharmacology , Protein Conformation/drug effects , Protein Stability/drug effects , Protein Unfolding/drug effects , Substrate Specificity
16.
Chem Biodivers ; 17(10): e2000495, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32865330

ABSTRACT

Human pancreatic tumor cells such as PANC-1 are known for their ability to tolerate nutrient starvation and thrive under the hypovascular tumor microenvironment, a phenomenon termed as 'austerity'. A search of agents that preferentially inhibit the cancer cell viability under the starvation condition without toxicity in the nutrient-rich condition is a promising approach in anticancer drug discovery. In this study, a triterpene lactone, 3ß-hydroxy-13,28-epoxyurs-11-en-28-one (ursenolide), isolated from a Callistemon citrinus extract has shown strong preferential cytotoxicity against PANC-1 cells under nutrient starvation with PC50 value of 0.4 µm. Ursenolide-induced rounding of PANC-1 cell morphology followed by rupture of the cell membrane leading to cell death. In a real-time cell migration study, ursenolide was found to inhibit PANC-1 cell migration significantly. Mechanistically, it inhibited GRP78 and GRP94 under the starvation condition suggesting inhibition of unfolded protein response (UPR), an adaptive process of cell survival during starvation. It also inhibited the phosphorylation of the key survival protein Akt and mTOR. Overall results suggested that ursenolide is a potential anticancer agent against pancreatic cancer.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Heat-Shock Proteins/antagonists & inhibitors , Lactones/pharmacology , Membrane Glycoproteins/antagonists & inhibitors , Myrtaceae/chemistry , Pancreatic Neoplasms/drug therapy , Triterpenes/pharmacology , Antineoplastic Agents, Phytogenic/chemistry , Antineoplastic Agents, Phytogenic/isolation & purification , Cell Proliferation/drug effects , Cell Survival/drug effects , Drug Screening Assays, Antitumor , Endoplasmic Reticulum Chaperone BiP , Heat-Shock Proteins/metabolism , Humans , Lactones/chemistry , Lactones/isolation & purification , Membrane Glycoproteins/metabolism , Molecular Conformation , Pancreatic Neoplasms/pathology , Protein Unfolding/drug effects , Triterpenes/chemistry , Triterpenes/isolation & purification , Tumor Cells, Cultured
17.
J Am Chem Soc ; 141(20): 8327-8338, 2019 05 22.
Article in English | MEDLINE | ID: mdl-31042030

ABSTRACT

For HIV to become infectious, any new virion produced from an infected cell must undergo a maturation process that involves the assembly of viral polyproteins Gag and Gag-Pol at the membrane surface. The self-assembly of these viral proteins drives formation of a new viral particle as well as the activation of HIV protease, which is needed to cleave the polyproteins so that the final core structure of the virus will properly form. Molecules that interfere with HIV maturation will prevent any new virions from infecting additional cells. In this manuscript, we characterize the unique mechanism by which a mercaptobenzamide thioester small molecule (SAMT-247) interferes with HIV maturation via a series of selective acetylations at highly conserved cysteine and lysine residues in Gag and Gag-Pol polyproteins. The results provide the first insights into how acetylation can be utilized to perturb the process of HIV maturation and reveal a new strategy to limit the infectivity of HIV.


Subject(s)
Anti-HIV Agents/pharmacology , Benzamides/pharmacology , HIV/drug effects , Protein Unfolding/drug effects , Virus Assembly/drug effects , gag Gene Products, Human Immunodeficiency Virus/drug effects , Acetylation , Amino Acid Sequence , Cell Line , Cysteine/chemistry , Fusion Proteins, gag-pol/chemistry , Fusion Proteins, gag-pol/drug effects , Humans , Lysine/chemistry , gag Gene Products, Human Immunodeficiency Virus/chemistry
18.
Langmuir ; 35(30): 9732-9739, 2019 07 30.
Article in English | MEDLINE | ID: mdl-31282164

ABSTRACT

Fibrinogen adsorption plays a key role in important biological processes, such as blood coagulation and foreign body reaction, which determine the biocompatibility of a material. Fibrinogen conformation on a surface is one of the main factors triggering these processes. Understanding the conformational dynamics of fibrinogen molecules adsorbed on solid surfaces is, therefore, of great interest in biomedicine and may contribute to the development of new biomaterials. In this work, unfolding of fibrinogen molecules adsorbed on a model surface (highly oriented pyrolytic graphite modified with an oligoglycine-hydrocarbon graphite modifier) is directly visualized using time-lapse atomic force microscopy. A gradual transformation of native-like fibrinogen molecules into fibrillar structures is observed at a timescale of several minutes. This transformation is accompanied by a decrease in molecular height from 4-5 to 1-2 nm. Independent unfolding of different fibrinogen domains is demonstrated. The obtained results provide a new, direct insight into the unfolding of individual fibrinogen molecules on a surface and give new opportunities for the development of graphite-based biosensors and biomaterials.


Subject(s)
Fibrinogen/chemistry , Graphite/chemistry , Graphite/pharmacology , Microscopy, Atomic Force , Protein Unfolding/drug effects , Surface Properties
19.
Eur Biophys J ; 48(4): 341-348, 2019 May.
Article in English | MEDLINE | ID: mdl-30929094

ABSTRACT

[Formula: see text]-Lactamases (penicillinases) facilitate bacterial resistance to antibiotics and are excellent theoretical and experimental models in protein structure, dynamics and evolution. Bacillus licheniformis exo-small penicillinase (ESP) is a Class A [Formula: see text]-lactamase with three tryptophan residues located one in each of its two domains and one in the interface between domains. The conformational landscape of three well-characterized ESP Trp[Formula: see text]Phe mutants was characterized in equilibrium unfolding experiments by measuring tryptophan fluorescence, far-UV CD, activity, hydrodynamic radius, and limited proteolysis. The Trp[Formula: see text]Phe substitutions had little impact on the native conformation, but changed the properties of the partially folded states populated at equilibrium. The results were interpreted in the framework of modern theories of protein folding.


Subject(s)
Bacillus licheniformis/enzymology , Protein Folding , beta-Lactamases/chemistry , Models, Molecular , Protein Domains , Protein Unfolding/drug effects , Urea/pharmacology
20.
Arch Biochem Biophys ; 658: 66-76, 2018 11 15.
Article in English | MEDLINE | ID: mdl-30261166

ABSTRACT

We studied the structure, function and thermodynamic properties for the unfolding of the Triosephosphate isomerase (TIM) from Zea mays (ZmTIM). ZmTIM shows a catalytic efficiency close to the diffusion limit. Native ZmTIM is a dimer that dissociates upon dilution into inactive and unfolded monomers. Its thermal unfolding is irreversible with a Tm of 61.6 ±â€¯1.4 °C and an activation energy of 383.4 ±â€¯11.5 kJ mol-1. The urea-induced unfolding of ZmTIM is reversible. Transitions followed by catalytic activity and spectroscopic properties are monophasic and superimposable, indicating that ZmTIM unfolds/refolds in a two-state behavior with an unfolding ΔG°(H20) = 99.8 ±â€¯5.3 kJ mol-1. This contrasts with most other studied TIMs, where folding intermediates are common. The three-dimensional structure of ZmTIM was solved at 1.8 Å. A structural comparison with other eukaryotic TIMs shows a similar number of intramolecular and intermolecular interactions. Interestingly the number of interfacial water molecules found in ZmTIM is lower than those observed in most TIMs that show folding intermediates. Although with the available data, there is no clear correlation between structural properties and the number of equilibrium intermediates in the unfolding of TIM, the identification of such structural properties should increase our understanding of folding mechanisms.


Subject(s)
Plant Proteins/chemistry , Triose-Phosphate Isomerase/chemistry , Zea mays/enzymology , Catalysis , Crystallography, X-Ray , Humans , Protein Conformation , Protein Stability , Protein Unfolding/drug effects , Temperature , Urea/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL