2,458 results on '"Protein Unfolding"'
Search Results
2. Protein Unfolding in Freeze Frames: Intermediate States are Revealed by Variable-Temperature Ion Mobility–Mass Spectrometry
- Author
-
Jakub Ujma, Jacquelyn Jhingree, Emma Norgate, Rosie Upton, Xudong Wang, Florian Benoit, Bruno Bellina, and Perdita Barran
- Subjects
Ions ,Protein Conformation ,Ubiquitin ,Solvents ,Temperature ,Proteins ,Mass Spectrometry ,Protein Unfolding ,Analytical Chemistry - Abstract
The gas phase is an idealized laboratory for the study of protein structure, from which it is possible to examine stable and transient forms of mass-selected ions in the absence of bulk solvent. With ion mobility-mass spectrometry (IM-MS) apparatus built to operate at both cryogenic and elevated temperatures, we have examined conformational transitions that occur to the monomeric proteins: ubiquitin, lysozyme, and α-synuclein as a function of temperature and in source activation. We rationalize the experimental observations with a temperature-dependent framework model and comparison to known conformers. Data from ubiquitin show unfolding transitions that proceed through diverse and highly elongated intermediate states, which converge to more compact structures. These findings contrast with data obtained from lysozyme─a protein where (un)-folding plasticity is restricted by four disulfide linkages, although this is alleviated in its reduced form. For structured proteins, collision activation of the protein ions in-source enables subsequent "freezing" or thermal annealing of unfolding intermediates, whereas disordered proteins restructure substantially at 250 K even without activation, indicating that cold denaturation can occur without solvent. These data are presented in the context of a toy model framework that describes the relative occupancy of the available conformational space.
- Published
- 2022
3. Thermal stability enhancement: Fundamental concepts of protein engineering strategies to manipulate the flexible structure
- Author
-
Mahdie Rahban, Samaneh Zolghadri, Najmeh Salehi, Faizan Ahmad, Thomas Haertlé, Nasrollah Rezaei-Ghaleh, Lindsay Sawyer, and Ali Akbar Saboury
- Subjects
Protein Stability ,Structural Biology ,Temperature ,Proteins ,General Medicine ,Protein Engineering ,Molecular Biology ,Biochemistry ,Protein Unfolding - Abstract
Increasing the temperature by just a few degrees may lead to structural perturbation or unfolding of the protein and consequent loss of function. The concepts of flexibility and rigidity are fundamental for understanding the relationships between function, structure and stability. Protein unfolding can often be triggered by thermal fluctuations with flexible residues usually on the protein surface. Therefore, identification and knowledge of the effect of modification to flexible regions in protein structures are required for efficient protein engineering and the rational design of thermally stable proteins. The most flexible regions in protein are loops, hence their rigidification is one of the effective strategies for increasing thermal stability. Directed evolution or rational design by computational prediction can also lead to the generation of thermally stable proteins. Computational protein design has been improved significantly in recent years and has successfully produced de novo stable backbone structures with optimized sequences and functions. This review discusses intramolecular and intermolecular interactions that determine the protein structure, and the strategies utilized in the mutagenesis of mesophilic proteins to stabilize and improve the functional characteristics of biocatalysts by describing efficient techniques and strategies to rigidify flexible loops at appropriate positions in the structure of the protein.
- Published
- 2022
4. Disruption of Proteostasis by Natural Products and Synthetic Compounds That Induce Pervasive Unfolding of Proteins: Therapeutic Implications
- Author
-
Nuria Vilaboa, Juan Antonio Lopez, Marco de Mesa, Clara Escudero-Duch, Natalie Winfield, Melanie Bayford, and Richard Voellmy
- Subjects
Drug Discovery ,Pharmaceutical Science ,Molecular Medicine ,proteostasis inhibition ,proteostasis disruption ,protein unfolding ,protein aggregation ,IHSF ,celastrol ,withaferin A ,multiple myeloma 1 - Abstract
Exposure of many cancer cells, including multiple myeloma cells, to cytotoxic concentrations of natural products celastrol and withaferin A or synthetic compounds of the IHSF series resulted in denaturation of a luciferase reporter protein. Proteomic analysis of detergent-insoluble extract fractions from HeLa-derived cells revealed that withaferin A, IHSF058 and IHSF115 caused denaturation of 915, 722 and 991 of 5132 detected cellular proteins, respectively, of which 440 were targeted by all three compounds. Western blots showed that important fractions of these proteins, in some cases approaching half of total protein amounts, unfolded. Relatively indiscriminate covalent modification of target proteins was observed; 1178 different proteins were modified by IHSF058. Further illustrating the depth of the induced proteostasis crisis, only 13% of these proteins detectably aggregated, and 79% of the proteins that aggregated were not targets of covalent modification. Numerous proteostasis network components were modified and/or found in aggregates. Proteostasis disruption caused by the study compounds may be more profound than that mediated by proteasome inhibitors. The compounds act by a different mechanism that may be less susceptible to resistance development. Multiple myeloma cells were particularly sensitive to the compounds. Development of an additional proteostasis-disrupting therapy of multiple myeloma is suggested.
- Published
- 2023
- Full Text
- View/download PDF
5. Nanopore Current Enhancements Lack Protein Charge Dependence and Elucidate Maximum Unfolding at Protein’s Isoelectric Point
- Author
-
Y. M. Nuwan D. Y. Bandara, Nasim Farajpour, and Kevin J. Freedman
- Subjects
Nanopores ,Colloid and Surface Chemistry ,Protein Conformation ,CRISPR-Associated Protein 9 ,Isoelectric Point ,General Chemistry ,Electroosmosis ,Hydrogen-Ion Concentration ,Biochemistry ,Catalysis ,Protein Unfolding - Abstract
Protein sequencing, as well as protein fingerprinting, has gained tremendous attention in the electrical sensing realm of solid-state nanopores and is challenging due to fast translocations and the use of high molar electrolytes. Despite providing an appreciable signal-to-noise ratio, high electrolyte concentrations can have adverse effects on the native protein structure. Herein, we present a thorough investigation of low electrolyte sensing conditions across a broad pH and voltage range generating conductive pulses (CPs) irrespective of protein net charge. We used Cas9 as the model protein and demonstrated that unfolding is noncooperative, represented by the gradual elongation or stretching of the protein, and sensitive to both the applied voltage and pH (i.e., charge state). The magnitude of unfolding and the isoelectric point (pI) of Cas9 was found to be correlated and a critical factor in our experiments. Electroosmotic flow (EOF) was always aligned with the transit direction, whereas electrophoretic force (EPF) was either reinforcing (pHpI) or opposing (pHpI) the protein's movement, which led to slower translocations at higher pH values. Further exploration of higher pH values led to slowing down of protein with30% of the population being slower than 0.5 ms. Our results would be critical for protein sensing at very low electrolytes and to retard their translocation speed without resorting to high-bandwidth equipment.
- Published
- 2022
6. Metal Ion Binding Induces Local Protein Unfolding and Destabilizes Human Carbonic Anhydrase II
- Author
-
Kayla D. McConnell, Nicholas C. Fitzkee, and Joseph P. Emerson
- Subjects
Ions ,Inorganic Chemistry ,Zinc ,Binding Sites ,Coordination Complexes ,Humans ,Calorimetry ,Physical and Theoretical Chemistry ,Carbonic Anhydrase Inhibitors ,Carbonic Anhydrase II ,Article ,Copper ,Protein Unfolding - Abstract
Human carbonic anhydrase II (HCA) is a robust metalloprotein and an excellent biological model system to study the thermodynamics of metal ion coordination. Apo-HCA binds one zinc ion or two copper ions. We studied these binding processes at five temperatures (15–35 °C) using isothermal titration calorimetry, yielding thermodynamic parameters corrected for pH and buffer effects. We then sought to identify binding-induced structural changes. Our data suggest that binding at the active site organizes 6–8 residues; however, copper binding near the N-terminus results in a net unfolding of 6–7 residues. This surprising destabilization was confirmed using circular dichroism and protein stability measurements. Metal binding induced unfolding may represent an important regulatory mechanism, but it may be easily missed by NMR and X-ray crystallography. Thus, in addition to highlighting a highly novel binding-induced unfolding event, we demonstrate the value of calorimetry for studying the structural implications of metal binding.
- Published
- 2022
7. Monitoring protein unfolding transitions by NMR-spectroscopy
- Author
-
Matthias Dreydoppel, Jochen Balbach, and Ulrich Weininger
- Subjects
Protein Denaturation ,Protein Folding ,Magnetic Resonance Spectroscopy ,Proteins ,Thermodynamics ,Nuclear Magnetic Resonance, Biomolecular ,Biochemistry ,Spectroscopy ,Protein Unfolding - Abstract
NMR-spectroscopy has certain unique advantages for recording unfolding transitions of proteins compared e.g. to optical methods. It enables per-residue monitoring and separate detection of the folded and unfolded state as well as possible equilibrium intermediates. This allows a detailed view on the state and cooperativity of folding of the protein of interest and the correct interpretation of subsequent experiments. Here we summarize in detail practical and theoretical aspects of such experiments. Certain pitfalls can be avoided, and meaningful simplification can be made during the analysis. Especially a good understanding of the NMR exchange regime and relaxation properties of the system of interest is beneficial. We show by a global analysis of signals of the folded and unfolded state of GB1 how accurate values of unfolding can be extracted and what limits different NMR detection and unfolding methods. E.g. commonly used exchangeable amides can lead to a systematic under determination of the thermodynamic protein stability. We give several perspectives of how to deal with more complex proteins and how the knowledge about protein stability at residue resolution helps to understand protein properties under crowding conditions, during phase separation and under high pressure.
- Published
- 2022
8. Engineering shape memory and morphing protein hydrogels based on protein unfolding and folding
- Author
-
Bian, Qingyuan, Fu, Linglan, and Li, Hongbin
- Subjects
Protein Conformation, alpha-Helical ,Protein Folding ,animal structures ,Biomaterials - proteins ,genetic structures ,Polymers ,Science ,ComputingMethodologies_IMAGEPROCESSINGANDCOMPUTERVISION ,General Physics and Astronomy ,Protein Engineering ,Article ,General Biochemistry, Genetics and Molecular Biology ,GeneralLiterature_MISCELLANEOUS ,Biopolymers ,Elastic Modulus ,Amino Acid Sequence ,reproductive and urinary physiology ,Protein Unfolding ,ComputingMethodologies_COMPUTERGRAPHICS ,Multidisciplinary ,fungi ,technology, industry, and agriculture ,Proteins ,Hydrogels ,General Chemistry ,Elastomers ,Wettability ,Protein Conformation, beta-Strand ,Gels and hydrogels ,psychological phenomena and processes - Abstract
Engineering shape memory/morphing materials have achieved considerable progress in polymer-based systems with broad potential applications. However, engineering protein-based shape memory/morphing materials remains challenging and under-explored. Here we report the design of a bilayer protein-based shape memory/morphing hydrogel based on protein folding-unfolding mechanism. We fabricate the protein-bilayer structure using two tandem modular elastomeric proteins (GB1)8 and (FL)8. Both protein layers display distinct denaturant-dependent swelling profiles and Young’s moduli. Due to such protein unfolding-folding induced changes in swelling, the bilayer hydrogels display highly tunable and reversible bidirectional bending deformation depending upon the denaturant concentration and layer geometry. Based on these programmable and reversible bending behaviors, we further utilize the protein-bilayer structure as hinge to realize one-dimensional to two-dimensional and two-dimensional to three-dimensional folding transformations of patterned hydrogels. The present work will offer new inspirations for the design and fabrication of novel shape morphing materials., Engineering shape memory and morphing materials achieved considerable progress in polymer-based systems, but protein-based shape memory and morphing materials remain less investigated. Here, the authors report the engineering of protein-based shape memory and morphing hydrogels using protein folding-unfolding as a general mechanism to trigger shape morphing in protein-bilayer structures.
- Published
- 2022
9. Protein Unfolding-Thermodynamic Perspectives and Unfolding Models
- Author
-
Seelig, Joachim and Seelig, Anna
- Subjects
Inorganic Chemistry ,Organic Chemistry ,General Medicine ,Physical and Theoretical Chemistry ,protein unfolding ,differential scanning calorimetry ,Zimm–Bragg theory ,cold denaturation ,free energy ,Molecular Biology ,Spectroscopy ,Catalysis ,Computer Science Applications - Abstract
We review the key steps leading to an improved analysis of thermal protein unfolding. Thermal unfolding is a dynamic cooperative process with many short-lived intermediates. Protein unfolding has been measured by various spectroscopic techniques that reveal structural changes, and by differential scanning calorimetry (DSC) that provides the heat capacity change Cp(T). The corresponding temperature profiles of enthalpy ΔH(T), entropy ΔS(T), and free energy ΔG(T) have thus far been evaluated using a chemical equilibrium two-state model. Taking a different approach, we demonstrated that the temperature profiles of enthalpy ΔH(T), entropy ΔS(T), and free energy ΔG(T) can be obtained directly by a numerical integration of the heat capacity profile Cp(T). DSC thus offers the unique possibility to assess these parameters without resorting to a model. These experimental parameters now allow us to examine the predictions of different unfolding models. The standard two-state model fits the experimental heat capacity peak quite well. However, neither the enthalpy nor entropy profiles (predicted to be almost linear) are congruent with the measured sigmoidal temperature profiles, nor is the parabolic free energy profile congruent with the experimentally observed trapezoidal temperature profile. We introduce three new models, an empirical two-state model, a statistical–mechanical two-state model and a cooperative statistical-mechanical multistate model. The empirical model partially corrects for the deficits of the standard model. However, only the two statistical–mechanical models are thermodynamically consistent. The two-state models yield good fits for the enthalpy, entropy and free energy of unfolding of small proteins. The cooperative statistical–mechanical multistate model yields perfect fits, even for the unfolding of large proteins such as antibodies.
- Published
- 2023
10. Collision-Induced Unfolding of Native-like Protein Ions Within a Trapped Ion Mobility Spectrometry Device
- Author
-
Nicholas B. Borotto, Kemi E. Osho, Talitha Kamakamakamae Richards, and Katherine A. Graham
- Subjects
Ions ,Structural Biology ,Ion Mobility Spectrometry ,Proteins ,Spectroscopy ,Protein Unfolding - Abstract
Native mass spectrometry and collision-induced unfolding (CIU) workflows continue to grow in utilization due to their ability to rapidly characterize protein conformation and stability. To perform these experiments, the instrument must be capable of collisionally activating ions prior to ion mobility spectrometry (IMS) analyses. Trapped ion mobility spectrometry (TIMS) is an ion mobility implementation that has been increasingly adopted due to its inherently high resolution and reduced instrumental footprint. In currently deployed commercial instruments, however, typical modes of collisional activation do not precede IMS analysis, and thus, the instruments are incapable of performing CIU. In this work, we expand on a recently developed method of activating protein ions within the TIMS device and explore its analytical utility toward the unfolding of native-like protein ions. We demonstrate the unfolding of native-like ions of ubiquitin, cytochrome C, β-lactoglobulin, and carbonic anhydrase. These ions undergo extensive unfolding upon collisional activation. Additionally, the improved resolution provided by the TIMS separation uncovers previously obscured unfolding complexity.
- Published
- 2021
11. The intrinsic amyloidogenic propensity of cofilin-1 is aggravated by Cys-80 oxidation: A possible link with neurodegenerative diseases
- Author
-
Vibha Kaushik, Daniela Brünnert, Suneel Kateriya, Pankaj Goyal, Phulwanti Kumari Sharma, Eva-Maria Hanschmann, Bibin G. Anand, and Karunakar Kar
- Subjects
Cofilin 1 ,Models, Molecular ,Amyloid ,In silico ,Biophysics ,Amyloidogenic Proteins ,macromolecular substances ,medicine.disease_cause ,Protein Aggregation, Pathological ,environment and public health ,Biochemistry ,Protein structure ,Alzheimer Disease ,Partial loss ,Actin dynamics ,medicine ,Humans ,Computer Simulation ,Amino Acid Sequence ,Cysteine ,Propensity Score ,Molecular Biology ,Actin ,Protein Unfolding ,Sequence Homology, Amino Acid ,Protein Stability ,Chemistry ,Neurodegenerative Diseases ,Cell Biology ,Cell biology ,Mutation ,Oxidation-Reduction ,Oxidative stress - Abstract
Cofilin-1, an actin dynamizing protein, forms actin-cofilin rods, which is one of the major events that exacerbates the pathophysiology of amyloidogenic diseases. Cysteine oxidation in cofilin-1 under oxidative stress plays a crucial role in the formation of these rods. Others and we have reported that cofilin-1 possesses a self-oligomerization property in vitro and in vivo under physiological conditions. However, it remains elusive if cofilin-1 itself forms amyloid-like structures. We, therefore, hypothesized that cofilin-1 might form amyloid-like assemblies, with a potential to intensify the pathophysiology of amyloid-linked diseases. We used various in silico and in vitro techniques and examined the amyloid-forming propensity of cofilin-1. The study confirms that cofilin-1 possesses an intrinsic tendency of aggregation and forms amyloid-like structures in vitro. Further, we studied the effect of cysteine oxidation on the stability and structural features of cofilin-1. Our data show that oxidation at Cys-80 renders cofilin-1 unstable, leading to a partial loss of protein structure. The results substantiate our hypothesis and establish a strong possibility that cofilin-1 aggregation might play a role in cofilin-mediated pathology and the progression of several amyloid-linked diseases.
- Published
- 2021
12. Conformational scanning of individual EF-hand motifs of calcium sensor protein centrin-1
- Author
-
Kumarasamy Thangaraj, Regur Phanindranath, Digumarthi V. S. Sudhakar, and Yogendra Sharma
- Subjects
Cell division ,Calmodulin ,Biophysics ,chemistry.chemical_element ,Cell Cycle Proteins ,Cooperativity ,Calcium ,Affinity binding ,Biochemistry ,Troponin C ,Genes, Reporter ,Humans ,Magnesium ,EF Hand Motifs ,Molecular Biology ,Protein Unfolding ,biology ,Chemistry ,EF hand ,Calcium-Binding Proteins ,Tryptophan ,Cell Biology ,Centrin ,biology.protein - Abstract
Centrin-1, a Ca2+ sensor protein of the centrin family is a crucial player for cell division in eukaryotes and plays a key role in the microtubule organising centre. Despite being regarded as a calcium sensor with a matched structure to calmodulin/troponin C, the protein undergoes mild changes in conformation and binds Ca2+ with moderate affinity. We present an in-depth analysis of the Ca2+ sensing by individual EF-hand motifs of centrin-1 and address unsolved questions of the rationales for moderate affinity and conformational transitions of the protein. Employing the more sensitive approach of Trp scanning of individual EF-hand motif, we have undertaken an exhaustive investigation of Ca2+ binding to individual EF-hand motifs, named EF1 to EF4. All four EF-hand motifs of centrin-1 are structural as all of them bind both Ca2+ and Mg2+. EF1 and EF4 are the most flexible sites as they undergo drastic conformational changes following Ca2+ binding, whereas EF3 responds to Ca2+ minimally. On the other hand, EF2 moves towards the protein surface upon binding Ca2+. The independent filling mode of Ca2+ to EF-hand motifs and lack of intermotif communication explain the lack of cooperativity of binding, thus constraining centrin-1 to a moderate affinity binding protein. Thus, centrin-1 is distinct from other calcium sensors such as calmodulin.
- Published
- 2021
13. Using Intrinsic Fluorescence to Measure Protein Stability Upon Thermal and Chemical Denaturation
- Author
-
Nathalia, Varejão and David, Reverter
- Subjects
Protein Denaturation ,Protein Stability ,Circular Dichroism ,Tryptophan ,Protein Unfolding - Abstract
Understanding how point mutations affect the performance of protein stability has been the focus of several studies all over the years. Intrinsic fluorescence is commonly used to follow protein unfolding since during denaturation, progressive redshifts on tryptophan fluorescence emission are observed. Since the unfolding process (achieved by chemical or physical denaturants) can be considered as two-state N➔D, it is possible to utilize the midpoint unfolding curves (fU = 50%) as a parameter to evaluate if the mutation destabilizes wild-type protein. The idea is to determine the [D]
- Published
- 2022
14. Linking Gas-Phase and Solution-Phase Protein Unfolding via Mobile Proton Simulations
- Author
-
Charles Eldrid, Tristan Cragnolini, Aisha Ben-Younis, Junjie Zou, Daniel P. Raleigh, and Konstantinos Thalassinos
- Subjects
Ions ,Protein Conformation ,Proteins ,Protons ,Molecular Dynamics Simulation ,Analytical Chemistry ,Protein Unfolding - Abstract
Native mass spectrometry coupled to ion mobility (IM-MS) combined with collisional activation (CA) of ions in the gas phase (
- Published
- 2022
15. Snapshots of urea-induced early structural changes and unfolding of an ankyrin repeat protein at atomic resolution
- Author
-
Mukund Sudharsan Medur Gurushankar, Somavally Dalvi, and Prasanna Venkatraman
- Subjects
Protein Denaturation ,Protein Folding ,X-Ray Diffraction ,Protein Conformation ,Urea ,Molecular Biology ,Biochemistry ,Ankyrin Repeat ,Protein Unfolding - Abstract
Protein folding and unfolding is a complex process, underscored by the many proteotoxic diseases associated with misfolded proteins. Mapping pathways from a native structure to an unfolded protein or vice versa, identifying the intermediates, and defining the role of sequence and structure en route remain outstanding problems in the field. It is even more challenging to capture the events at atomistic resolution. X-ray diffraction has so far been used to understand how urea interacts with and unfolds two stable globular proteins. Here, we present the case study on PSMD10
- Published
- 2022
16. Dry Molten Globule-Like Intermediates in Protein Folding, Function, and Disease
- Author
-
Nirbhik Acharya and Santosh Kumar Jha
- Subjects
Protein Folding ,Protein Conformation ,Circular Dichroism ,Materials Chemistry ,Physical and Theoretical Chemistry ,Surfaces, Coatings and Films ,Protein Unfolding - Abstract
The performance of a protein depends on its correct folding to the final functional native form. Hence, understanding the process of protein folding has remained an important field of research for the scientific community for the past five decades. Two important intermediate states, namely, wet molten globule (WMG) and dry molten globule (DMG), have emerged as critical milestones during protein folding-unfolding reactions. While much has been discussed about WMGs as a common unfolding intermediate, the evidence for DMGs has remained elusive owing to their near-native features, which makes them difficult to probe using global structural probes. This Review puts together the available literature and new evidence on DMGs to give a broader perspective on the universality of DMGs and discuss their significance in protein folding, function, and disease.
- Published
- 2022
17. Volumetric Properties of the Transition State Ensemble for Protein Folding
- Author
-
Samvel Avagyan and George I. Makhatadze
- Subjects
Protein Folding ,Materials Chemistry ,Proteins ,Physical and Theoretical Chemistry ,Molecular Dynamics Simulation ,Surfaces, Coatings and Films ,Protein Unfolding - Abstract
Understanding how high hydrostatic pressure affects biomacromolecular interaction is important for deciphering the molecular mechanisms by which organisms adapt to live at the bottom of the ocean. The relative effect of hydrostatic pressure on the rates of folding/unfolding reactions is defined by the volumetric properties of the transition state ensemble relative to the folded and unfolded states. All-atom structure-based molecular dynamics simulations combined with quantitative computational protocol to compute volumes from three-dimensional coordinates allow volumetric mapping of protein folding landscape. This, is turn, provides qualitative understanding of the effects of hydrostatic pressure on energy landscape of proteins. The computational results for six different proteins are directly benchmark against experimental data and show an excellent agreement. Both experiments and computation show that the transition-state ensemble volume appears to be in-between the folded and unfolded state volumes, and thus the hydrostatic pressure accelerates protein unfolding.
- Published
- 2022
18. Unfolding and identification of membrane proteins in situ
- Author
-
Zhongjie Ye, Nicola Galvanetto, Arin Marchesi, Simone Mortal, Sourav Maity, Alessandro Laio, Vincent Torre, and Molecular Biophysics
- Subjects
Proteomics ,atomic force microscopy ,General Immunology and Microbiology ,General Neuroscience ,Lipid Bilayers ,Membrane Proteins ,Bayes Theorem ,General Medicine ,Microscopy, Atomic Force ,Settore BIO/09 - Fisiologia ,single-molecule force spectroscopy ,General Biochemistry, Genetics and Molecular Biology ,Settore FIS/03 - Fisica della Materia ,protein identification ,molecular biophysics ,structural biology ,rat ,Protein Unfolding - Abstract
Single-molecule force spectroscopy (SMFS) uses the cantilever tip of an atomic force microscope (AFM) to apply a force able to unfold a single protein. The obtained force-distance curve encodes the unfolding pathway, and from its analysis it is possible to characterize the folded domains. SMFS has been mostly used to study the unfolding of purified proteins, in solution or reconstituted in a lipid bilayer. Here, we describe a pipeline for analyzing membrane proteins based on SMFS, which involves the isolation of the plasma membrane of single cells and the harvesting of force-distance curves directly from it. We characterized and identified the embedded membrane proteins combining, within a Bayesian framework, the information of the shape of the obtained curves, with the information from mass spectrometry and proteomic databases. The pipeline was tested with purified/reconstituted proteins and applied to five cell types where we classified the unfolding of their most abundant membrane proteins. We validated our pipeline by overexpressing four constructs, and this allowed us to gather structural insights of the identified proteins, revealing variable elements in the loop regions. Our results set the basis for the investigation of the unfolding of membrane proteins in situ, and for performing proteomics from a membrane fragment.
- Published
- 2022
19. DAXX represents a new type of protein-folding enabler
- Author
-
Guixin Zhu, Ronen Marmorstein, Xiaolu Yang, Liming Tao, JiaBei Lin, Trisha Agrawal, Sixiang Yu, Liangqian Huang, and James Shorter
- Subjects
Models, Molecular ,Protein Folding ,Protein Conformation ,Cells ,Protein domain ,Protein aggregation ,Protein Aggregation, Pathological ,Cell Line ,Evolution, Molecular ,Protein Aggregates ,Protein structure ,Death-associated protein 6 ,Protein Domains ,Animals ,Humans ,Proteostasis Deficiencies ,Protein Unfolding ,Multidisciplinary ,biology ,Chemistry ,Proto-Oncogene Proteins c-mdm2 ,Cell biology ,Chaperone (protein) ,Mutation ,biology.protein ,Unfolded protein response ,Protein folding ,Tumor Suppressor Protein p53 ,Co-Repressor Proteins ,Function (biology) ,Molecular Chaperones - Abstract
Protein quality control systems are crucial for cellular function and organismal health. At present, most known protein quality control systems are multicomponent machineries that operate via ATP-regulated interactions with non-native proteins to prevent aggregation and promote folding1, and few systems that can broadly enable protein folding by a different mechanism have been identified. Moreover, proteins that contain the extensively charged poly-Asp/Glu (polyD/E) region are common in eukaryotic proteomes2, but their biochemical activities remain undefined. Here we show that DAXX, a polyD/E protein that has been implicated in diverse cellular processes3–10, possesses several protein-folding activities. DAXX prevents aggregation, solubilizes pre-existing aggregates and unfolds misfolded species of model substrates and neurodegeneration-associated proteins. Notably, DAXX effectively prevents and reverses aggregation of its in vivo-validated client proteins, the tumour suppressor p53 and its principal antagonist MDM2. DAXX can also restore native conformation and function to tumour-associated, aggregation-prone p53 mutants, reducing their oncogenic properties. These DAXX activities are ATP-independent and instead rely on the polyD/E region. Other polyD/E proteins, including ANP32A and SET, can also function as stand-alone, ATP-independent molecular chaperones, disaggregases and unfoldases. Thus, polyD/E proteins probably constitute a multifunctional protein quality control system that operates via a distinctive mechanism. A protein chaperone system is identified that consists of proteins with poly-Asp/Glu sequence, and may have an important role in diseases characterized by protein aggregation.
- Published
- 2021
20. A Continuum Model for the Unfolding of von Willebrand Factor
- Author
-
Mehrdad Massoudi, Wei-Tao Wu, James F. Antaki, Mansur Zhussupbekov, and Rodrigo Méndez Rojano
- Subjects
congenital, hereditary, and neonatal diseases and abnormalities ,Conformational change ,Continuum (design consultancy) ,Biomedical Engineering ,FOS: Physical sciences ,Models, Biological ,Quantitative Biology - Quantitative Methods ,Article ,Von Willebrand factor ,hemic and lymphatic diseases ,von Willebrand Factor ,Von Willebrand disease ,medicine ,Physics - Biological Physics ,Hemostatic function ,Quantitative Methods (q-bio.QM) ,Protein Unfolding ,biology ,Chemistry ,Fluid Dynamics (physics.flu-dyn) ,Thrombosis ,Physics - Fluid Dynamics ,medicine.disease ,Simple shear ,Shear (sheet metal) ,Shear rate ,Biological Physics (physics.bio-ph) ,FOS: Biological sciences ,Biophysics ,biology.protein ,circulatory and respiratory physiology - Abstract
von Willebrand Factor is a mechano-sensitive protein circulating in blood that mediates platelet adhesion to subendothelial collagen and platelet aggregation at high shear rates. Its hemostatic function and thrombogenic effect, as well as susceptibility to enzymatic cleavage, are regulated by a conformational change from a collapsed globular state to a stretched state. Therefore, it is essential to account for the conformation of the vWF multimers when modeling vWF-mediated thrombosis or vWF degradation. We introduce a continuum model of vWF unfolding that is developed within the framework of our multi-constituent model of platelet-mediated thrombosis. The model considers two interconvertible vWF species corresponding to the collapsed and stretched conformational states. vWF unfolding takes place via two regimes: tumbling in simple shear and strong unfolding in flows with dominant extensional component. These two regimes were demonstrated in a Couette flow between parallel plates and an extensional flow in a cross-slot geometry. The vWF unfolding model was then verified in several microfluidic systems designed for inducing high-shear vWF-mediated thrombosis and screening for von Willebrand Disease. The model predicted high concentration of stretched vWF in key regions where occlusive thrombosis was observed experimentally. Strong unfolding caused by the extensional flow was limited to the center axis or middle plane of the channels, whereas vWF unfolding near the channel walls relied upon the shear tumbling mechanism. The continuum model of vWF unfolding presented in this work can be employed in numerical simulations of vWF-mediated thrombosis or vWF degradation in complex geometries. However, extending the model to 3-D arbitrary flows and turbulent flows will pose considerable challenges.
- Published
- 2021
21. Control of Nanoscale In Situ Protein Unfolding Defines Network Architecture and Mechanics of Protein Hydrogels
- Author
-
Matt D. G. Hughes, Lorna Dougan, Benjamin S. Hanson, Najet Mahmoudi, Sophie Cussons, and David J. Brockwell
- Subjects
In situ ,Circular dichroism ,General Physics and Astronomy ,Biocompatible Materials ,02 engineering and technology ,010402 general chemistry ,01 natural sciences ,Article ,hierarchical biomechanics ,Rheology ,protein hydrogels ,General Materials Science ,Bovine serum albumin ,Nanoscopic scale ,biology ,Chemistry ,protein unfolding ,General Engineering ,Hydrogels ,Serum Albumin, Bovine ,Mechanics ,biomimetic and bioinspired materials ,021001 nanoscience & nanotechnology ,0104 chemical sciences ,Covalent bond ,Self-healing hydrogels ,Unfolded protein response ,biology.protein ,0210 nano-technology ,biomaterials - Abstract
Hierarchical assemblies of proteins exhibit a wide-range of material properties that are exploited both in nature and by artificially by humankind. However, little is understood about the importance of protein unfolding on the network assembly, severely limiting opportunities to utilize this nanoscale transition in the development of biomimetic and bioinspired materials. Here we control the force lability of a single protein building block, bovine serum albumin (BSA), and demonstrate that protein unfolding plays a critical role in defining the architecture and mechanics of a photochemically cross-linked native protein network. The internal nanoscale structure of BSA contains "molecular reinforcement" in the form of 17 covalent disulphide "nanostaples", preventing force-induced unfolding. Upon addition of reducing agents, these nanostaples are broken rendering the protein force labile. Employing a combination of circular dichroism (CD) spectroscopy, small-angle scattering (SAS), rheology, and modeling, we show that stapled protein forms reasonably homogeneous networks of cross-linked fractal-like clusters connected by an intercluster region of folded protein. Conversely,iin situ/iprotein unfolding results in more heterogeneous networks of denser fractal-like clusters connected by an intercluster region populated by unfolded protein. In addition, gelation-induced protein unfolding and cross-linking in the intercluster region changes the hydrogel mechanics, as measured by a 3-fold enhancement of the storage modulus, an increase in both the loss ratio and energy dissipation, and markedly different relaxation behavior. By controlling the protein's ability to unfold through nanoscale (un)stapling, we demonstrate the importance ofiin situ/iunfolding in defining both network architecture and mechanics, providing insight into fundamental hierarchical mechanics and a route to tune biomaterials for future applications.
- Published
- 2021
22. Statistical Learning from Single-Molecule Experiments: Support Vector Machines and Expectation–Maximization Approaches to Understanding Protein Unfolding Data
- Author
-
Farkhad Maksudov, Lee K. Jones, and Valeri Barsegov
- Subjects
Models, Molecular ,Motivation ,Quantitative Biology::Biomolecules ,Support Vector Machine ,010304 chemical physics ,Computer science ,Data classification ,Force spectroscopy ,Proteins ,Experimental data ,010402 general chemistry ,01 natural sciences ,Standard deviation ,0104 chemical sciences ,Surfaces, Coatings and Films ,Support vector machine ,Margin (machine learning) ,0103 physical sciences ,Expectation–maximization algorithm ,Materials Chemistry ,Force dynamics ,Physical and Theoretical Chemistry ,Algorithm ,Protein Unfolding - Abstract
Single-molecule force spectroscopy has become a powerful tool for the exploration of dynamic processes that involve proteins; yet, meaningful interpretation of the experimental data remains challenging. Owing to low signal-to-noise ratio, experimental force-extension spectra contain force signals due to nonspecific interactions, tip or substrate detachment, and protein desorption. Unravelling of complex protein structures results in the unfolding transitions of different types. Here, we test the performance of Support Vector Machines (SVM) and Expectation Maximization (EM) approaches in statistical learning from dynamic force experiments. When the output from molecular modeling in silico (or other studies) is used as a training set, SVM and EM can be applied to understand the unfolding force data. The maximal margin or maximum likelihood classifier can be used to separate experimental test observations into the unfolding transitions of different types, and EM optimization can then be utilized to resolve the statistics of unfolding forces: weights, average forces, and standard deviations. We designed an EM-based approach, which can be directly applied to the experimental data without data classification and division into training and test observations. This approach performs well even when the sample size is small and when the unfolding transitions are characterized by overlapping force ranges.
- Published
- 2021
23. HU-AB simulacrum: Fusion of HU-B and HU-A into HU-B-A, a functional analog of the Escherichia coli HU-AB heterodimer
- Author
-
Archit Gupta, Kanika Arora, Purnananda Guptasarma, and Bhishem Thakur
- Subjects
Protein Conformation, alpha-Helical ,0301 basic medicine ,Protein Folding ,Stereochemistry ,Recombinant Fusion Proteins ,Population ,Biophysics ,medicine.disease_cause ,Biochemistry ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,medicine ,Urea ,Nucleoid ,education ,Molecular Biology ,Escherichia coli ,Histidine ,Protein Unfolding ,chemistry.chemical_classification ,education.field_of_study ,Chemistry ,Escherichia coli Proteins ,DNA ,Cell Biology ,Protein engineering ,Amino acid ,DNA-Binding Proteins ,030104 developmental biology ,030220 oncology & carcinogenesis ,Thermodynamics ,Protein Multimerization ,Dimerization ,Linker - Abstract
In enteric bacteria such as Escherichia coli, there are two homologs of the DNA-binding nucleoid associated protein (NAP) known as HU. The two homologs are known as HU-A and HU-B, and exist either in the form of homodimers (HU-AA, or HU-BB) or as heterodimers (HU-AB), with different propensities to form higher-order oligomers. The three different dimeric forms dominate different stages of bacterial growth, with the HU-AB heterodimer dominating cultures in the stationary phase. Due to similarities in their properties, and the facile equilibrium that exists between the dimeric forms, the dimers are difficult to purify away from each other. Although HU-AA and HU-BB can be purified through extensive ion-exchange chromatography, reestablishment of equilibrium interferes with the purification of the HU-AB heterodimer (which constitutes ∼90% of any population with equal numbers of HU-B and HU-A chains). Here, we report the creation of a functional analog of HU-AB that does not appear to partition to generate any minority populations of HU-AA or HU-BB. The analog was constructed through genetic fusion of the HU-B and HU-A chains into a single polypeptide (HU-B-A) with a glycine/serine-rich linker of 11 amino acids separating HU-B from HU-A, and a histidine tag at the N-terminus of HU-B. HU-B-A folds to bind 4-way junction DNA, and displays a significant tendency to form dimers (i.e., analogs of HU tetramers), and a higher thermodynamic stability than HU-BB or HU-AA, thus explaining why it dominates mixtures of HU-B and HU-A chains.
- Published
- 2021
24. Thermodynamic destabilization of azurin by four different tetramethylguanidinium amino acid ionic liquids
- Author
-
Gurvir Singh, Timothy D. Vaden, Hunter Gogoj, Isabella DeStefano, Roshani Patel, Gabriella DeStefano, Austin K. Clark, Keertana S. Jonnalagadda, Chun Wu, Gregory A. Caputo, Nicholas Paradis, and Aashka Y. Patel
- Subjects
Anions ,Entropy ,Ionic Liquids ,Methylguanidine ,02 engineering and technology ,Molecular Dynamics Simulation ,Biochemistry ,Protein Structure, Secondary ,Serine ,03 medical and health sciences ,chemistry.chemical_compound ,Azurin ,Structural Biology ,Cations ,Side chain ,Transition Temperature ,Amino Acids ,Threonine ,Molecular Biology ,Protein Unfolding ,030304 developmental biology ,chemistry.chemical_classification ,0303 health sciences ,Protein Stability ,Chemistry ,Imidazoles ,General Medicine ,Hydrogen-Ion Concentration ,021001 nanoscience & nanotechnology ,Amino acid ,Crystallography ,Ionic liquid ,Chemical stability ,0210 nano-technology ,Hydrophobic and Hydrophilic Interactions ,Entropy (order and disorder) - Abstract
The thermal unfolding of the copper redox protein azurin was studied in the presence of four different amino acid-based ionic liquids (ILs), all of which have tetramethylguanidium as cation. The anionic amino acid includes two with alcohol side chains, serine and threonine, and two with carboxylic acids, aspartate and glutamate. Control experiments showed that amino acids alone do not significantly change protein stability and pH changes anticipated by the amino acid nature have only minor effects on the protein. With the ILs, the protein is destabilized and the melting temperature is decreased. The two ILs with alcohol side chains strongly destabilize the protein while the two ILs with acid side chains have weaker effects. Unfolding enthalpy (ΔHunf°) and entropy (ΔSunf°) values, derived from fits of the unfolding data, show that some ILs increase ΔHunf°while others do not significantly change this value. All ILs, however, increase ΔSunf°. MD simulations of both the folded and unfolded protein conformations in the presence of the ILs provide insight into the different IL-protein interactions and how they affect the ΔHunf° values. The simulations also confirm that the ILs increase the unfolded state entropies which can explain the increased ΔSunf° values.
- Published
- 2021
25. Assessing the Functional and Structural Stability of the Met80Ala Mutant of Cytochrome c in Dimethylsulfoxide
- Author
-
Giulia Di Rocco, Antonio Ranieri, Marco Borsari, Marco Sola, Carlo Augusto Bortolotti, and Gianantonio Battistuzzi
- Subjects
cytochrome c ,Chemistry (miscellaneous) ,protein unfolding ,Organic Chemistry ,Drug Discovery ,Molecular Medicine ,Pharmaceutical Science ,cytochrome c, dimethylsulfoxide, electron transfer, protein unfolding ,Physical and Theoretical Chemistry ,dimethylsulfoxide ,electron transfer ,Analytical Chemistry - Abstract
The Met80Ala variant of yeast cytochrome c is known to possess electrocatalytic properties that are absent in the wild type form and that make it a promising candidate for biocatalysis and biosensing. The versatility of an enzyme is enhanced by the stability in mixed aqueous/organic solvents that would allow poorly water-soluble substrates to be targeted. In this work, we have evaluated the effect of dimethylsulfoxide (DMSO) on the functionality of the Met80Ala cytochrome c mutant, by investigating the thermodynamics and kinetics of electron transfer in mixed water/DMSO solutions up to 50% DMSO v/v. In parallel, we have monitored spectroscopically the retention of the main structural features in the same medium, focusing on both the overall protein structure and the heme center. We found that the organic solvent exerts only minor effects on the redox and structural properties of the mutant mostly as a result of the modification of the dielectric constant of the solvent. This would warrant proper functionality of this variant also under these potentially hostile experimental conditions, that differ from the physiological milieu of cytochrome c.
- Published
- 2022
- Full Text
- View/download PDF
26. Termodinaminiai parametrai baltymų-ligandų modelinėse sistemose: jungimosi tūrio ir sąveikos energijos tyrimas
- Author
-
Skvarnavičius, Gediminas and Petrauskas, Vytautas
- Subjects
Protein-ligand binding ,Protein unfolding ,carbonic anhydrases ,electrostatic interaction ,polymer-surfactant interaction - Abstract
Understanding protein-ligand binding thermodynamic parameters is fundamental for rational drug design. However, despite the importance, due to the complexity of protein-ligand systems, areas of this field, including volume change due to protein-ligand binding (Vb) and ionic interaction, are left largely unexplored. This thesis researches the volumetric characteristics of protein-ligand binding in carbonic anhydrase (CA)-sulfonamide model systems. CA XIII unfolding pathway using guanidinium hydrochloride (GdmHCl) was determined for the first time. The relationship between melting pressure (Pm) and GdmHCl concentration was evaluated for CA I, CA II, and CA XIII. A concept of fluorescent pressure shift assay technique using different GdmHCl and ligand concentrations for strong ligand Vb determination was proposed. The high-pressure NMR technique was employed to determine Vb values of primary sulfonamide inhibitor binding to CA I and CA II. A method for determining changes in volume upon protein-ligand binding from a single series of NMR spectra was described for the first time.A model system consisting of oppositely charged amino acid homopolymers and surfactants was used to determine the contributions of chemical modifications and ionic interaction toward protein-ligand binding. Contributions of alkyl chain and head group make-up towards enthalpy and heat capacity of interactions were determined for the first time in an oppositely charged poly(amino acid)- surfactant system.
- Published
- 2022
27. Analysis of Biologics Molecular Descriptors towards Predictive Modelling for Protein Drug Development Using Time-Gated Raman Spectroscopy
- Author
-
Jaakko Itkonen, Leo Ghemtio, Daniela Pellegrino, Pia J. Jokela (née Heinonen), Henri Xhaard, and Marco G. Casteleijn
- Subjects
PCA ,in-line measurement ,pharmaceutical proteins ,protein unfolding ,DLS ,Raman spectroscopy ,Pharmaceutical Science ,biologics ,K-means clustering ,tryptophan fluorescence ,CD - Abstract
Pharmaceutical proteins, compared to small molecular weight drugs, are relatively fragile molecules, thus necessitating monitoring protein unfolding and aggregation during production and post-marketing. Currently, many analytical techniques take offline measurements, which cannot directly assess protein folding during production and unfolding during processing and storage. In addition, several orthogonal techniques are needed during production and market surveillance. In this study, we introduce the use of time-gated Raman spectroscopy to identify molecular descriptors of protein unfolding. Raman spectroscopy can measure the unfolding of proteins in-line and in real-time without labels. Using K-means clustering and PCA analysis, we could correlate local unfolding events with traditional analytical methods. This is the first step toward predictive modeling of unfolding events of proteins during production and storage.
- Published
- 2022
- Full Text
- View/download PDF
28. Inherent Minor Conformer of
- Author
-
Adi, Yahalom, Hadassa, Shaked, Sharon, Ruthstein, and Jordan H, Chill
- Subjects
Magnetic Resonance Spectroscopy ,Bacterial Proteins ,Type III Secretion Systems ,Bordetella pertussis ,Molecular Chaperones ,Protein Unfolding - Abstract
The pathogen
- Published
- 2022
29. Tuning Protein Hydrogel Mechanics through Modulation of Nanoscale Unfolding and Entanglement in Postgelation Relaxation
- Author
-
Matt D. G. Hughes, Sophie Cussons, Najet Mahmoudi, David J. Brockwell, and Lorna Dougan
- Subjects
General Engineering ,General Physics and Astronomy ,Proteins ,General Materials Science ,Hydrogels ,Biocompatible Materials ,Rheology ,Protein Unfolding - Abstract
Globular folded proteins are versatile nanoscale building blocks to create biomaterials with mechanical robustness and inherent biological functionality due to their specific and well-defined folded structures. Modulating the nanoscale unfolding of protein building blocks during network formation (in situ protein unfolding) provides potent opportunities to control the protein network structure and mechanics. Here, we control protein unfolding during the formation of hydrogels constructed from chemically cross-linked maltose binding protein using ligand binding and the addition of cosolutes to modulate protein kinetic and thermodynamic stability. Bulk shear rheology characterizes the storage moduli of the bound and unbound protein hydrogels and reveals a correlation between network rigidity, characterized as an increase in the storage modulus, and protein thermodynamic stability. Furthermore, analysis of the network relaxation behavior identifies a crossover from an unfolding dominated regime to an entanglement dominated regime. Control of in situ protein unfolding and entanglement provides an important route to finely tune the architecture, mechanics, and dynamic relaxation of protein hydrogels. Such predictive control will be advantageous for future smart biomaterials for applications which require responsive and dynamic modulation of mechanical properties and biological function.
- Published
- 2022
30. Sequence of Events during Peptide Unbinding from RNase S: A Complete Experimental Description
- Author
-
Jeannette Ruf, Claudio Zanobini, Brankica Jankovic, David Buhrke, Peter Hamm, Olga Bozovic, University of Zurich, and Hamm, Peter
- Subjects
Protein Conformation, alpha-Helical ,10120 Department of Chemistry ,0301 basic medicine ,Light ,RNase P ,Binding pocket ,FOS: Physical sciences ,Sequence (biology) ,Peptide ,010402 general chemistry ,01 natural sciences ,03 medical and health sciences ,chemistry.chemical_compound ,Ribonucleases ,540 Chemistry ,Moiety ,General Materials Science ,Physics - Biological Physics ,Amino Acid Sequence ,Physical and Theoretical Chemistry ,Protein Unfolding ,chemistry.chemical_classification ,030102 biochemistry & molecular biology ,Chemistry ,Biomolecules (q-bio.BM) ,Helicity ,2500 General Materials Science ,0104 chemical sciences ,Intrinsically Disordered Proteins ,Kinetics ,Quantitative Biology - Biomolecules ,Azobenzene ,Biological Physics (physics.bio-ph) ,FOS: Biological sciences ,Biophysics ,Peptides ,1606 Physical and Theoretical Chemistry ,Azo Compounds ,Protein Binding - Abstract
The photo-triggered unbinding of the intrinsically disordered S-peptide from the RNase S complex is studied with the help of transient IR spectroscopy, covering a wide range of time scales from 100 ps to 10 ms. To that end, an azobenzene moiety has been linked to the S-peptide in a way that its helicity is disrupted by light, thereby initiating its complete unbinding. The full sequence of events is observed, starting from unfolding of the helical structure of the S-peptide on a 20 ns timescale while still being in the binding pocket of the S-protein, S-peptide unbinding after 300 microseconds, and the structural response of the S-protein after 3 ms. With regard to the S-peptide dynamics, the binding mechanism can be classified as an induced fit, while the structural response of the S-protein is better described as conformational selection.
- Published
- 2021
31. Elevated temperatures accelerate the formation of toxic amyloid fibrils of hen egg‐white lysozyme
- Author
-
Zili Feng, Yu Bai, and Ying Li
- Subjects
Amyloid ,Circular dichroism ,Veterinary medicine ,Fibril ,hen egg‐white lysozyme ,prion ,amyloid fibrils ,chemistry.chemical_compound ,Microscopy, Electron, Transmission ,SF600-1100 ,Animals ,MTT assay ,Viability assay ,Cytotoxicity ,General Veterinary ,Circular Dichroism ,protein unfolding ,Temperature ,Original Articles ,Congo red ,chemistry ,Biophysics ,Muramidase ,Original Article ,Thioflavin ,Lysozyme ,elevated temperatures - Abstract
The formation of amyloid fibrils is critical for neurodegenerative diseases. Some physiochemical conditions can promote the conversion of proteins from soluble globular shapes into insoluble well‐organized amyloid fibrils. The aim of this study was to investigate the effect of temperatures on amyloid fibrils formation in vitro using the protein model of hen egg‐white lysozyme (HEWL). The HEWL fibrils were prepared at temperatures of 37, 45, 50 and 57°C in glycine solution of pH 2.2. Under transmission electron microscopy, we found the well‐organized HEWL amyloid fibrils at temperatures of 45, 50 and 57°C after 10 days of incubation. Thioflavin T and Congo red florescence assays confirmed that the formation and growth of HEWL fibrils displayed a temperature‐dependent increase, and 57°C produced the most amounts. Meanwhile, the surface hydrophobicity of aggregates was greatly increased by ANS binding assay, and β‐sheet contents by circular dichroism analysis were increased by 17.8%, 22.0% and 34.9%, respectively. Furthermore, the HEWL fibrils formed at 57°C caused significant cytotoxicity in SH‐SY5Y cells after 48 hr exposure, and the cell viability determined by MTT assay was decreased, with 81.35 ± 0.29% for 1 μM, 61.45 ± 2.62% for 2 μM, and 11.58 ± 0.39% (p, Elevated temperatures of 45−57°C (over physiological 37°C) increased protein misfolding and the formation and growth of amyloid using prion‐like HEWL protein.
- Published
- 2021
32. Cyclic Ion Mobility–Collision Activation Experiments Elucidate Protein Behavior in the Gas Phase
- Author
-
Dale Cooper-Shepherd, Konstantinos Thalassinos, Rehana Akter, Kevin Giles, Aisha Ben-Younis, Michael Morris, Daniel P. Raleigh, Jakub Ujma, Charles Eldrid, Hannah M. Britt, Symeon Kalfas, Tristan Cragnolini, and Nick Tomczyk
- Subjects
Cytochrome ,Ion-mobility spectrometry ,ion-mobility mass spectrometry ,Peptide ,010402 general chemistry ,Mass spectrometry ,01 natural sciences ,Dissociation (chemistry) ,Mass Spectrometry ,Ion ,Structural Biology ,Application Note ,Ion Mobility Spectrometry ,Animals ,Humans ,Horses ,Spectroscopy ,chemistry.chemical_classification ,Ions ,biology ,Tandem ,Protein dynamics ,protein unfolding ,010401 analytical chemistry ,Cytochromes c ,Baculoviral IAP Repeat-Containing 3 Protein ,0104 chemical sciences ,chemistry ,biology.protein ,Biophysics ,Gases - Abstract
Ion mobility coupled to mass spectrometry (IM-MS) is widely used to study protein dynamics and structure in the gas phase. Increasing the energy with which the protein ions are introduced to the IM cell can induce them to unfold, providing information on the comparative energetics of unfolding between different proteoforms. Recently, a high-resolution cyclic IM-mass spectrometer (cIM-MS) was introduced, allowing multiple, consecutive tandem IM experiments (IMn) to be carried out. We describe a tandem IM technique for defining detailed protein unfolding pathways and the dynamics of disordered proteins. The method involves multiple rounds of IM separation and collision activation (CA): IM-CA-IM and CA-IM-CA-IM. Here, we explore its application to studies of a model protein, cytochrome C, and dimeric human islet amyloid polypeptide (hIAPP), a cytotoxic and amyloidogenic peptide involved in type II diabetes. In agreement with prior work using single stage IM-MS, several unfolding events are observed for cytochrome C. IMn-MS experiments also show evidence of interconversion between compact and extended structures. IMn-MS data for hIAPP shows interconversion prior to dissociation, suggesting that the certain conformations have low energy barriers between them and transition between compact and extended forms.
- Published
- 2021
33. pH induced conformational alteration in human peroxiredoxin 6 might be responsible for its resistance against lysosomal pH or high temperature
- Author
-
Laishram Rajendrakumar Singh, Sunaina Hotumalani, Rimpy Kaur Chowhan, and Hamidur Rahaman
- Subjects
0301 basic medicine ,Protein moonlighting ,Hot Temperature ,Science ,Article ,Supramolecular assembly ,03 medical and health sciences ,Structure-Activity Relationship ,Fluorescence Resonance Energy Transfer ,Humans ,Protein Structure, Quaternary ,Conformational isomerism ,Cellular compartment ,Protein Unfolding ,chemistry.chemical_classification ,Multidisciplinary ,030102 biochemistry & molecular biology ,biology ,Chemistry ,Circular Dichroism ,Substrate (chemistry) ,Hydrogen Peroxide ,Compartmentalization (psychology) ,Hydrogen-Ion Concentration ,Molecular conformation ,Cytosol ,030104 developmental biology ,Enzyme ,Spectrometry, Fluorescence ,biology.protein ,Biophysics ,Chromatography, Gel ,Thermodynamics ,Medicine ,Calcium ,Lysosomes ,Oxidoreductases ,Peroxidase ,Peroxiredoxin VI - Abstract
Peroxiredoxin 6 (Prdx6), the ubiquitously expressed enzyme belonging to the family of peroxidases, namely, peroxiredoxins, exhibits a unique feature of functional compartmentalization within cells. Whereas, the enzyme localized in cytosol shows glutathione peroxidase activity, its lysosomal counterpart performs calcium independent phospholipase A2 (aiPLA2) activity. Like any true moonlighting protein, these two activities of Prdx6 are mutually exclusive of each other as a function of the pH of the cellular compartments. Differential substrate preference at different pH (i.e. peroxidised phospholipids at neutral pH and reduced phospholipids at acidic pH) is considered to be the reason for this behavior. To gain insight into the pH-induced structural–functional interplay we have systematically evaluated conformational variations, thermodynamic stability of the protein and quaternary state of the conformers at both pH 7.0 and 4.0. Our findings suggest that change in pH allows alterations in native states of Prdx6 at pH 7.0 and 4.0 such that the changes make the protein resistant to thermal denaturation at low pH.
- Published
- 2021
34. Small-Molecule Degraders beyond PROTACs—Challenges and Opportunities
- Author
-
Johanna M Kastl, Gareth M. Davies, Eleanor Godsman, and Geoffrey A. Holdgate
- Subjects
Proteomics ,0301 basic medicine ,Proteasome Endopeptidase Complex ,Ubiquitin-Protein Ligases ,High-throughput screening ,Computational biology ,Protein degradation ,Ligands ,01 natural sciences ,Biochemistry ,Analytical Chemistry ,Small Molecule Libraries ,03 medical and health sciences ,Drug Discovery ,Humans ,Molecular Targeted Therapy ,Protein Unfolding ,biology ,010405 organic chemistry ,Chemistry ,Drug discovery ,Ubiquitination ,Ligand (biochemistry) ,Small molecule ,High-Throughput Screening Assays ,0104 chemical sciences ,Ubiquitin ligase ,Eukaryotic Cells ,030104 developmental biology ,Proteostasis ,Proteolysis ,biology.protein ,Molecular Medicine ,Target protein ,Hydrophobic and Hydrophilic Interactions ,Protein Processing, Post-Translational ,Protein Binding ,Biotechnology - Abstract
Targeted protein degradation (TPD) is a recent strategy, utilizing the cell's proteostasis machinery to deplete specific proteins. This represents a paradigm shift in early drug discovery, away from occupancy-driven to event-driven mechanisms.Recent efforts have focused on the development of proteolysis-targeting chimeras (PROTACs). These heterobifunctional molecules combine a target-specific binding moiety linked to an E3 ligase ligand and trigger selective ubiquitination of the target protein, marking it for proteasomal degradation. While these molecules can be highly efficacious, they generally have unfavorable physicochemical properties due to their large size.In contrast, smaller molecules that induce degradation could represent an attractive, simple option to overcoming the limitations of both traditional modulators and PROTACs. These molecules may have a range of mechanisms: recruitment of an E3 ligase (molecular glues), introduction of hydrophobic areas, or inducing local unfolding, each of which triggers degradation.We recently completed a high-throughput screen of 111,000 compounds in a cellular HiBiT assay in an effort to identify such molecules. Preliminary analysis indicates that we have been able to identify alternative small-molecule degraders. We highlight methods for triage, characterization, selectivity, and mode of action. In summary, we believe that these types of small-molecule degraders, which may possibly have more acceptable physicochemical properties than the inherently larger heterobifunctional molecules, are an exciting approach for inducing TPD, and we illustrate that a general screening approach can be successful in identifying useful start points for developing such molecules.
- Published
- 2021
35. Activation of von Willebrand factor via mechanical unfolding of its discontinuous autoinhibitory module
- Author
-
Emily R. Legan, Emma-Ruoqi Xu, Michael C. Berndt, Nicholas A Arce, Jonas Emsley, Alexander K. Brown, Wenpeng Cao, Renhao Li, Moriah Simone Wilson, and X. Frank Zhang
- Subjects
0301 basic medicine ,Models, Molecular ,congenital, hereditary, and neonatal diseases and abnormalities ,Platelet Aggregation ,Protein Conformation ,Science ,Shear force ,General Physics and Astronomy ,030204 cardiovascular system & hematology ,In Vitro Techniques ,Crystallography, X-Ray ,General Biochemistry, Genetics and Molecular Biology ,Article ,03 medical and health sciences ,0302 clinical medicine ,Von Willebrand factor ,Single-molecule biophysics ,Protein Domains ,Tensile Strength ,hemic and lymphatic diseases ,von Willebrand Factor ,Humans ,Platelet ,Protein Unfolding ,Multidisciplinary ,biology ,Chemistry ,Blood proteins ,Protein Stability ,Antibodies, Monoclonal ,General Chemistry ,Single-Domain Antibodies ,Single Molecule Imaging ,Biomechanical Phenomena ,030104 developmental biology ,Ristocetin ,Hemostasis ,Mutation ,Biophysics ,biology.protein ,cardiovascular system ,Caplacizumab ,circulatory and respiratory physiology - Abstract
Von Willebrand factor (VWF) activates in response to shear flow to initiate hemostasis, while aberrant activation could lead to thrombosis. Above a critical shear force, the A1 domain of VWF becomes activated and captures platelets via the GPIb-IX complex. Here we show that the shear-responsive element controlling VWF activation resides in the discontinuous autoinhibitory module (AIM) flanking A1. Application of tensile force in a single-molecule setting induces cooperative unfolding of the AIM to expose A1. The AIM-unfolding force is lowered by truncating either N- or C-terminal AIM region, type 2B VWD mutations, or binding of a ristocetin-mimicking monoclonal antibody, all of which could activate A1. Furthermore, the AIM is mechanically stabilized by the nanobody that comprises caplacizumab, the only FDA-approved anti-thrombotic drug to-date that targets VWF. Thus, the AIM is a mechano-regulator of VWF activity. Its conformational dynamics may define the extent of VWF autoinhibition and subsequent activation under force., Von Willebrand factor (VWF) is a large glycoprotein in the blood secreted from endothelial cells lining the blood vessel and activation of VWF leads to formation of VWF-platelet complexes or thrombi. Here authors use single-molecule force measurement, X-ray crystallography and functional measurements to monitor the activation of VWF via mechanical unfolding of the autoinhibitory module (AIM).
- Published
- 2021
36. Reconstruction of ARNT PAS-B Unfolding Pathways by Steered Molecular Dynamics and Artificial Neural Networks
- Author
-
Alessandro Pandini, Arianna Fornili, Stefano Motta, Laura Bonati, Motta, S, Pandini, A, Fornili, A, and Bonati, L
- Subjects
Physics ,PAS protein ,Aryl hydrocarbon receptor nuclear translocator ,010304 chemical physics ,Basic helix-loop-helix ,Artificial neural network ,Protein Conformation ,Atomic force microscopy ,Folding (DSP implementation) ,Molecular Dynamics Simulation ,Microscopy, Atomic Force ,01 natural sciences ,Article ,Computer Science Applications ,CHIM/02 - CHIMICA FISICA ,Molecular dynamics ,Self Organizing Maps ,0103 physical sciences ,Basic Helix-Loop-Helix Transcription Factors ,Neural Networks, Computer ,Physical and Theoretical Chemistry ,Biological system ,Protein Unfolding - Abstract
© 2021 The Authors. Several experimental studies indicated that large conformational changes, including partial domain unfolding, have a role in the functional mechanisms of the basic helix loop helix Per/ARNT/SIM (bHLH-PAS) transcription factors. Recently, single-molecule atomic force microscopy (AFM) revealed two distinct pathways for the mechanical unfolding of the ARNT PAS-B. In this work we used steered molecular dynamics simulations to gain new insights into this process at an atomistic level. To reconstruct and classify pathways sampled in multiple simulations, we designed an original approach based on the use of self-organizing maps (SOMs). This led us to identify two types of unfolding pathways for the ARNT PAS-B, which are in good agreement with the AFM findings. Analysis of average forces mapped on the SOM revealed a stable conformation of the PAS-B along one pathway, which represents a possible structural model for the intermediate state detected by AFM. The approach here proposed will facilitate the study of other signal transmission mechanisms involving the folding/unfolding of PAS domains. National Institutes of Environmental Health Sciences (R01-ES007685); Leverhulme Trust (RPG-2017-222).
- Published
- 2021
37. Potential involvement of environmental triggers in protein aggregation with mercuric chloride as a model
- Author
-
Charuvila T. Aravindakumar, Manjumol Mathew, Divyalakshmi T.V, and Usha K. Aravind
- Subjects
Amyloid ,Ovalbumin ,Molecular Conformation ,02 engineering and technology ,Environment ,Protein aggregation ,Microscopy, Atomic Force ,Spectrum Analysis, Raman ,Biochemistry ,Phase Transition ,Metal ,Protein Aggregates ,03 medical and health sciences ,chemistry.chemical_compound ,symbols.namesake ,Protein structure ,Structural Biology ,Metals, Heavy ,Phase (matter) ,Molecular Biology ,Protein Unfolding ,030304 developmental biology ,0303 health sciences ,Quenching (fluorescence) ,Proteins ,Mercury ,General Medicine ,Hydrogen-Ion Concentration ,021001 nanoscience & nanotechnology ,Amorphous solid ,Kinetics ,Monomer ,chemistry ,visual_art ,Mercuric Chloride ,visual_art.visual_art_medium ,Biophysics ,symbols ,0210 nano-technology ,Raman spectroscopy - Abstract
Heavy metal based toxicity has a direct relation with the perturbation of protein structure. We have investigated the progressive unfolding of ovalbumin, in the presence of increasing concentration mercury (0–6.25 μM) using different spectroscopic techniques. Formation of amorphous aggregate has been observed at the physiological pH. Initial addition of HgCl2 resulted in the association of monomers to oligomers that proceeded to non-fibrillar aggregates on further addition. The sigmoidal curve obtained from the Stern-Volmer plot clearly divided into three stage transition. A strong lag phase is observed indicating the time dependence for the association of competent monomers. The second stage was resolved into non-cooperative binding. These results match very well with the data from atomic force microscopy and the free energy change observed in the regions. Raman spectroscopic studies indicated toxic antiparallel β-sheets structure. Time dependent atomic force microscopy study revealed the off-pathway nature of amorphous aggregates. At molten globular state, similar quenching behaviour is observed. The atomic force microscopy images clearly indicate at pH 2.2 the initiation of fibril formation occurs at lower concentration of HgCl2 itself. Our results revealed the conformation switch of ovalbumin upon the contact of an environmental toxin and its possible way of toxicity.
- Published
- 2021
38. Probing Folded Proteins and Intact Protein Complexes by Desorption Electrospray Ionization Mass Spectrometry
- Author
-
Josephine Bunch and Bin Yan
- Subjects
Protein Folding ,Spectrometry, Mass, Electrospray Ionization ,010402 general chemistry ,Mass spectrometry ,01 natural sciences ,Spectral line ,Ion ,Hemoglobins ,chemistry.chemical_compound ,Protein structure ,Tetramer ,Structural Biology ,Non-covalent interactions ,Animals ,Humans ,Horses ,Spectroscopy ,Protein Unfolding ,chemistry.chemical_classification ,Desorption electrospray ionization ,Chromatography ,biology ,Myoglobin ,Ubiquitin ,Cytochrome c ,010401 analytical chemistry ,Cytochromes c ,Proteins ,0104 chemical sciences ,Solvent ,chemistry ,biology.protein ,Biophysics ,Cattle ,Hemoglobin ,Methanol ,Ammonium acetate - Abstract
Native mass spectrometry (MS) enables the study of intact proteins as well as noncovalent protein-protein and protein-ligand complexes in their biological state. In this work, we present the application of a Waters desorption electrospray ionization (DESI) source with a prototype spray emitter for rapid surface measurements of folded and native protein structures. A comparison of DESI spray solvent shows that adding 50% methanol to 200 mM ammonium acetate solution does not reduce its performance in preserving folded protein structures. Instead, improved signal-to-noise (S/N) ratio is obtained, and less adducted peaks are detected by using this uncommon native MS solvent system. The standard DESI design with an inlet tube allows optimization of sampling temperature conditions to improve desolvation and therefore S/N ratio. Furthermore, tuning the inlet temperature enables the control and study of unfolding behavior of proteins from surface samples. The optimized condition for native DESI has been applied to several selected proteins and protein complexes with the molecular weight ranging from 8.6 to 66.4 kDa. Ions of folded proteins with narrow charge state distribution (CSD), or peaks showing noncovalent-bond-assembled intact protein complexes, are observed in the spectra. Evidence for the structural refolding of denatured proteins and protein complexes sampled with native solvent highlights the need for care when interpreting DESI native MS data, particularly for proteins with stable native structures.
- Published
- 2021
39. Nucleocapsid protein preferentially binds the stem-loop of duplex/quadruplex hybrid that unfolds the quadruplex structure
- Author
-
Siming Yuan, Mingxi Ou, Na Zhang, Yangzhong Liu, Taotao Zou, Yu Wang, Bei Cao, and Yaping Sheng
- Subjects
Models, Molecular ,Catalysis ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Materials Chemistry ,heterocyclic compounds ,Thermal stability ,Binding site ,Protein Unfolding ,030304 developmental biology ,0303 health sciences ,Binding Sites ,Hydrogen bond ,Metals and Alloys ,General Chemistry ,Nucleocapsid Proteins ,Stem-loop ,Surfaces, Coatings and Films ,Electronic, Optical and Magnetic Materials ,G-Quadruplexes ,Crystallography ,chemistry ,Duplex (building) ,Ceramics and Composites ,030217 neurology & neurosurgery ,DNA - Abstract
NCp7 protein binds the duplex/quadruplex hybrid structure, which decreases the thermal stability of DNA and unfolds the G-quadruplex structure. Interestingly, the duplex in the stem-loop region is the more favorable binding site of NCp7. The NCp7 binding twists the top G-tetrad, weakens hydrogen bonding and causes K+ ejection, hence disrupting the G4 structure.
- Published
- 2021
40. Bioinformatics analysis of correlation between protein function and intrinsic disorder
- Author
-
Goran Vinterhalter, Jovana Kovačević, Vladimir N. Uversky, and Gordana Pavlović-Lažetić
- Subjects
Jaccard index ,Protein Conformation ,02 engineering and technology ,Computational biology ,Biochemistry ,Correlation ,03 medical and health sciences ,Annotation ,Similarity (network science) ,Sequence Analysis, Protein ,Structural Biology ,Databases, Protein ,Molecular Biology ,Protein Unfolding ,030304 developmental biology ,Mathematics ,0303 health sciences ,Sequence ,Computational Biology ,Proteins ,Molecular Sequence Annotation ,General Medicine ,Function (mathematics) ,021001 nanoscience & nanotechnology ,Term (time) ,Proteome ,0210 nano-technology - Abstract
The correlation of molecular function and protein intrinsic disorder is an important aspect of understanding the relationship between function, sequence and structure. This research was inspired by statistical correlation evaluation method described by Xie et al. (J Proteome Res 6 (2007) 1882-1898, reference study), where the authors analyzed the relationship between structure and function of proteins from Swiss-Prot database and where these functions were described with Swiss-Prot function keywords. In this research, we investigated whether the conclusions from the reference study stand for another dataset with richer functional annotation. We used CAFA3 challenge training dataset where the function was described with terms from Gene Ontology (GO terms). In order to compare the results with the previous work, we associated the GO terms with the corresponding Swiss-Prot function keywords. The results were compared with the reference study by first repeating the analysis with Swiss-Prot function keywords and then by GO terms. We used PONDR VSL2b disorder predictor to label over 66,000 CAFA3 proteins as putatively disordered or ordered. Out of 186 Swiss-Prot keywords (belonging to molecular function type) with more than 20 annotated proteins, we found 47 to be highly order related and 44 highly disorder related. Using the same dataset and annotation constraints, out of 1781 GO term (belonging to molecular function type), we found 746 to be highly order related and 564 highly disorder related. GO term results are presented as interactive graphs displaying complex hierarchical structure of Gene Ontology. Comparison of two functional annotations, GO and Swiss-Prot keywords, showed consistent results in cases when it was possible to map a Swiss-Prot keyword to a corresponding GO term. Because of the small number of such cases, we propose a new method for deriving the missing mappings between Swiss-Prot keywords and GO terms with the highest likelihood by measuring similarity (Jaccard index) between sets of protein annotated by different functions. Comparison with results from the reference study revealed prevalence of binding related functions (disorder related) in the current dataset even though the same functions were not present in previous results.
- Published
- 2021
41. Disaggregation of Amyloid-β Plaques by a Local Electric Field Generated by a Vertical Nanowire Electrode Array
- Author
-
Hye S. Lee, Heon Jin Choi, Jukwan Na, Jaesuk Sung, Jaejun Lee, Jun Shik Choi, Juyoung Kwon, Hyo Jung Lee, and Yong Beom Lim
- Subjects
Protein Conformation, alpha-Helical ,In situ ,Circular dichroism ,Amyloid beta-Peptides ,Materials science ,Nanowires ,Circular Dichroism ,Nanowire ,Protein Aggregates ,Electricity ,Alzheimer Disease ,Electric field ,Electrode ,Electrode array ,Biophysics ,Humans ,Thermodynamics ,Protein Conformation, beta-Strand ,General Materials Science ,Cyclic voltammetry ,Science, technology and society ,Electrodes ,Protein Unfolding - Abstract
The aggregation and accumulation of amyloid-β (Aβ) peptides is a characteristic pathology for Alzheimer's disease (AD). Although noninvasive therapies involving stimulation by electric field (EF) have been reported, the efficiency of Aβ disaggregation needs to be further improved for this strategy to be used in clinical settings. In this study, we show that an electrode based on a vertical nanowire electrode array (VNEA) is far more superior to a typical flat-type electrode in disaggregating Aβ plaques. The enhanced disaggregation efficiency of VNEA is due to the formation of high-strength local EF between the nanowires, as verified by in silico and empirical evidence. Compared with those of the flat electrode, the simulation data revealed that 19.8-fold and 8.8-fold higher EFs are generated above and between the nanowires, respectively. Moreover, empirical cyclic voltammetry data demonstrated that VNEA had a 2.7-fold higher charge capacity than the flat electrode; this is associated with the higher surface area of VNEA. The conformational transition of Aβ peptides between the β-sheet and α-helix could be sensitively monitored in real time by the newly designed in situ circular dichroism instrument. This highly efficient EF-configuration of VNEA will lower the stimulation power for disaggregating the Aβ plaques, compared to that of other existing field-mediated modulation systems. Considering the complementary metal-oxide-semiconductor-compatibility and biocompatible strength of the EF for perturbing the Aβ aggregation, our study could pave the way for the potential use of electric stimulation devices for in vivo therapeutic application as well as scientific studies for AD.
- Published
- 2020
42. Cryo-EM structure of the inhibited (10S) form of myosin II
- Author
-
Shixin Yang, Prince Tiwari, Raul Padron, Osamu Sato, Roger Craig, Mitsuo Ikebe, and Kyounghwan Lee
- Subjects
Models, Molecular ,Turkeys ,Protein Conformation ,Cryo-electron microscopy ,macromolecular substances ,Plasma protein binding ,Article ,Protein filament ,Motor protein ,03 medical and health sciences ,0302 clinical medicine ,Protein structure ,Myosin ,Animals ,Myocyte ,Phosphorylation ,Actin ,Protein Unfolding ,030304 developmental biology ,Myosin Type II ,0303 health sciences ,Binding Sites ,Multidisciplinary ,Chemistry ,Cryoelectron Microscopy ,Muscle, Smooth ,Mutation ,Biophysics ,030217 neurology & neurosurgery ,Protein Binding - Abstract
Myosin II is the motor protein that enables muscle cells to contract and nonmuscle cells to move and change shape1. The molecule has two identical heads attached to an elongated tail, and can exist in two conformations: 10S and 6S, named for their sedimentation coefficients2,3. The 6S conformation has an extended tail and assembles into polymeric filaments, which pull on actin filaments to generate force and motion. In 10S myosin, the tail is folded into three segments and the heads bend back and interact with each other and the tail3–7, creating a compact conformation in which ATPase activity, actin activation and filament assembly are all highly inhibited7,8. This switched-off structure appears to function as a key energy-conserving storage molecule in muscle and nonmuscle cells9–12, which can be activated to form functional filaments as needed13—but the mechanism of its inhibition is not understood. Here we have solved the structure of smooth muscle 10S myosin by cryo-electron microscopy with sufficient resolution to enable improved understanding of the function of the head and tail regions of the molecule and of the key intramolecular contacts that cause inhibition. Our results suggest an atomic model for the off state of myosin II, for its activation and unfolding by phosphorylation, and for understanding the clustering of disease-causing mutations near sites of intramolecular interaction. High-resolution cryo-electron microscopy structure of smooth muscle myosin II in the inhibited state enables increased understanding of the functions of the head and tail regions in regulation of myosin activity and the pathological mechanisms of disease mutations.
- Published
- 2020
43. The interaction of Naphthol Yellow S (NYS) with pepsin: Insights from spectroscopic to molecular dynamics studies
- Author
-
Fatemeh Hashemi-Shahraki, Sadegh Farhadian, and Behzad Shareghi
- Subjects
Circular dichroism ,Swine ,02 engineering and technology ,Molecular Dynamics Simulation ,Biochemistry ,Protein Structure, Secondary ,Fluorescence spectroscopy ,03 medical and health sciences ,chemistry.chemical_compound ,Molecular dynamics ,symbols.namesake ,Naphthalenesulfonates ,Structural Biology ,Enzyme Stability ,Animals ,Spectroscopy ,Molecular Biology ,Protein Unfolding ,030304 developmental biology ,0303 health sciences ,Hydrogen bond ,Circular Dichroism ,General Medicine ,021001 nanoscience & nanotechnology ,Binding constant ,Pepsin A ,Naphthol yellow S ,Molecular Docking Simulation ,Kinetics ,Crystallography ,Spectrometry, Fluorescence ,chemistry ,symbols ,Thermodynamics ,Spectrophotometry, Ultraviolet ,van der Waals force ,0210 nano-technology - Abstract
The effects of Naphthol Yellow S (NYS) on the structure and activity of pepsin were carried out using ultraviolet-visible (UV-Vis) spectroscopy, intrinsic fluorescence spectroscopy, circular dichroism (CD), thermal stability, kinetic techniques, as well as molecular docking, and Molecular dynamic simulations (MD) technique. The experimental results from fluorescence spectroscopy showed that the changes in pepsin's tertiary structure were caused by NYS binding. The apparent binding constant Ka, the number of the binding sites, and thermodynamic parameters were computed at three different temperatures. Thermodynamic results revealed that NYS interacts with pepsin spontaneously by hydrogen bond and Van der Waals forces. The result of the circular dichroism spectral suggests the secondary structural changes. An increase in the content of the β-sheet and β-turn structure was shown. Kinetic parameters revealed that NYS inhibited the activity of pepsin by the mixed model. The Molecular dynamic (MD) and docking simulations supported experimental findings. The main interactions between NYS and pepsin are hydrogen bonds and Van der Waals Forces. As a result, NYS could be considered as an inhibitor with adverse effects on pepsin structure and function.
- Published
- 2020
44. Multi-domain unfolding of the Fab fragment of a humanized anti-cocaine mAb characterized by non-reducing SDS-PAGE
- Author
-
Andrew B. Norman and Terence L. Kirley
- Subjects
0301 basic medicine ,medicine.drug_class ,Biophysics ,Antibodies, Monoclonal, Humanized ,Monoclonal antibody ,Biochemistry ,Article ,Immunoglobulin Fab Fragments ,03 medical and health sciences ,0302 clinical medicine ,Cocaine ,Protein Domains ,Fab Fragments ,medicine ,Molecular Biology ,Polyacrylamide gel electrophoresis ,Protein Unfolding ,Cocaine binding ,biology ,Chemistry ,Fragment (computer graphics) ,Cell Biology ,Native Polyacrylamide Gel Electrophoresis ,Multi domain ,030104 developmental biology ,030220 oncology & carcinogenesis ,biology.protein ,Antibody ,Small molecule binding - Abstract
Monoclonal antibodies and their fragments are widely used for research and therapy. Fab fragments are useful since they retain antigen binding specificity, but being smaller proteins, are better able to penetrate biological compartments and tumors, and do not induce Fc-dependent immunological system activation. Our laboratory developed an anti-cocaine mAb (named h2E2) for the treatment of cocaine use disorders, which is currently in advanced pre-clinical development. We are also interested in the Fab fragment of this mAb for potential therapy of acute cocaine overdose. Previously, we previously showed that this mAb and its F(ab’)(2) and Fab fragments undergo discrete domain unfolding, as detected by non-reducing SDS-PAGE, and that ligand-induced protein thermal stabilization can be quantitated utilizing differential scanning fluorimetry in the absence of SDS. Here, we demonstrate that multiple Fab protein gel bands observed using non-reducing SDS-PAGE in the presence and absence of cocaine and its metabolites can be explained and interpreted based on the differential stabilization of two unfolding domains in the Fab fragment. The variable domain is stabilized by ligands against SDS unfolding, while the constant domain is not. This data and its interpretation are also supported by differential scanning fluorimetry data for the Fab fragment in SDS. It is likely that these non-reducing SDS-PAGE results and the gel band domain unfolding model will be applicable to other small molecule binding antibodies. Thus, non-reducing SDS-PAGE is a widely available and simple method for assessing domain stability and multi-step unfolding of Fab fragments.
- Published
- 2020
45. High Energy Channeling and Malleable Transition States: Molecular Dynamics Simulations and Free Energy Landscapes for the Thermal Unfolding of Protein U1A and 13 Mutants
- Author
-
Na Le Dang, Anne M. Baranger, and David L. Beveridge
- Subjects
Protein Folding ,Molecular dynamics ,protein unfolding ,transition state ,U1A ,Proteins ,Reproducibility of Results ,Thermodynamics ,Molecular Dynamics Simulation ,Molecular Biology ,Biochemistry ,Protein Structure, Secondary - Abstract
The spliceosome protein U1A is a prototype case of the RNA recognition motif (RRM) ubiquitous in biological systems. The in vitro kinetics of the chemical denaturation of U1A indicate that the unfolding of U1A is a two-state process but takes place via high energy channeling and a malleable transition state, an interesting variation of typical two-state behavior. Molecular dynamics (MD) simulations have been applied extensively to the study of two-state unfolding and folding of proteins and provide an opportunity to obtain a theoretical account of the experimental results and a molecular model for the transition state ensemble. We describe herein all-atom MD studies including explicit solvent of up to 100 ns on the thermal unfolding (UF) of U1A and 13 mutants. Multiple MD UF trajectories are carried out to ensure accuracy and reproducibility. A vector representation of the MD unfolding process in RMSD space is obtained and used to calculate a free energy landscape for U1A unfolding. A corresponding MD simulation and free energy landscape for the protein CI2, well known to follow a simple two state folding/unfolding model, is provided as a control. The results indicate that the unfolding pathway on the MD calculated free energy landscape of U1A shows a markedly extended transition state compared with that of CI2. The MD results support the interpretation of the observed chevron plots for U1A in terms of a high energy, channel-like transition state. Analysis of the MDUF structures shows that the transition state ensemble involves microstates with most of the RRM secondary structure intact but expanded by ~14% with respect to the radius of gyration. Comparison with results on a prototype system indicates that the transition state involves an ensemble of molten globule structures and extends over the region of ~1–35 ns in the trajectories. Additional MDUF simulations were carried out for 13 U1A mutants, and the calculated φ-values show close accord with observed results and serve to validate our methodology.
- Published
- 2022
46. Characterization of full-length p53 aggregates and their kinetics of formation
- Author
-
Linda Julian, Jason C. Sang, Yunzhao Wu, Georg Meisl, Jack H. Brelstaff, Alyssa Miller, Matthew R. Cheetham, Michele Vendruscolo, Tuomas P.J. Knowles, Francesco Simone Ruggeri, Clare Bryant, Susana Ros, Kevin M. Brindle, and David Klenerman
- Subjects
Kinetics ,Protein Aggregates ,Mutation ,Biophysics ,Tumor Suppressor Protein p53 ,Genes, p53 ,Protein Unfolding - Abstract
Mutations in the TP53 gene are common in cancer with the R248Q missense mutation conferring an increased propensity to aggregate. Previous p53 aggregation studies showed that, at micromolar concentrations, protein unfolding to produce aggregation-prone species is the rate-determining step. Here we show that, at physiological concentrations, aggregation kinetics of insect cell-derived full-length wild-type p53 and p53R248Q are determined by a nucleation-growth model, rather than formation of aggregation-prone monomeric species. Self-seeding, but not cross-seeding, increases aggregation rate, confirming the aggregation process as rate determining. p53R248Q displays enhanced aggregation propensity due to decreased solubility and increased aggregation rate, forming greater numbers of larger amorphous aggregates that disrupt lipid bilayers and invokes an inflammatory response. These results suggest that p53 aggregation can occur under physiological conditions, a rate enhanced by R248Q mutation, and that aggregates formed can cause membrane damage and inflammation that may influence tumorigenesis.
- Published
- 2022
47. von Willebrand Factor unfolding mediates platelet deposition in a model of high-shear thrombosis
- Author
-
Mansur Zhussupbekov, Rodrigo Méndez Rojano, Wei-Tao Wu, and James F. Antaki
- Subjects
Blood Platelets ,congenital, hereditary, and neonatal diseases and abnormalities ,Biophysics ,Fluid Dynamics (physics.flu-dyn) ,FOS: Physical sciences ,Thrombosis ,Physics - Fluid Dynamics ,Hemostatics ,von Willebrand Diseases ,Biological Physics (physics.bio-ph) ,hemic and lymphatic diseases ,von Willebrand Factor ,Humans ,Physics - Biological Physics ,Protein Unfolding ,circulatory and respiratory physiology - Abstract
Thrombosis under high-shear conditions is mediated by the mechanosensitive blood glycoprotein von Willebrand Factor (vWF). vWF unfolds in response to strong flow gradients and facilitates rapid recruitment of platelets in flowing blood. While the thrombogenic effect of vWF is well recognized, its conformational response in complex flows has largely been omitted from numerical models of thrombosis. We recently presented a continuum model for the unfolding of vWF, where we represented vWF transport and its flow-induced conformational change using convection-diffusion-reaction equations. Here, we incorporate the vWF component into our multi-constituent model of thrombosis, where the local concentration of stretched vWF amplifies the deposition rate of free-flowing platelets and reduces the shear cleaning of deposited platelets. We validate the model using three benchmarks: in vitro model of atherothrombosis, a stagnation point flow, and the PFA-100, a clinical blood test commonly used for screening for von Willebrand Disease (vWD). The simulations reproduced the key aspects of vWF-mediated thrombosis observed in these experiments, such as the thrombus location, thrombus growth dynamics, and the effect of blocking platelet-vWF interactions. The PFA-100 simulations closely matched the reported occlusion times for normal blood and several hemostatic deficiencies, namely, thrombocytopenia, vWD Type 1, and vWD Type 3. Overall, the multi-constituent model of thrombosis presented in this work enables macro-scale 3-D simulations of thrombus formation in complex geometries over a wide range of shear rates and accounts for qualitative and quantitative hemostatic deficiencies in patient blood. The results also demonstrate the utility of the continuum model of vWF unfolding that could be adapted to other numerical models of thrombosis.
- Published
- 2022
48. Structure of proteins under pressure: Covalent binding effects of biliverdin on β-lactoglobulin
- Author
-
Simeon Minić, Burkhard Annighöfer, Arnaud Hélary, Laïla Sago, David Cornu, Annie Brûlet, Sophie Combet, Laboratoire Léon Brillouin (LLB - UMR 12), Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-Université Paris-Saclay-Centre National de la Recherche Scientifique (CNRS), LLB - Matière molle et biophysique (MMB), Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-Université Paris-Saclay-Centre National de la Recherche Scientifique (CNRS)-Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-Université Paris-Saclay-Centre National de la Recherche Scientifique (CNRS), LLB - Infrastructures et développement (INFRA), and Institut de Biologie Intégrative de la Cellule (I2BC)
- Subjects
[SDV.BBM.BP]Life Sciences [q-bio]/Biochemistry, Molecular Biology/Biophysics ,[SDV.BBM.BS]Life Sciences [q-bio]/Biochemistry, Molecular Biology/Structural Biology [q-bio.BM] ,Biliverdine ,Biophysics ,Animals ,[SDV.BBM]Life Sciences [q-bio]/Biochemistry, Molecular Biology ,Cattle ,Cysteine ,Lactoglobulins ,Article ,Protein Unfolding - Abstract
International audience; High pressure (HP) is a particularly powerful tool to study protein folding/unfolding, revealing subtle structural rearrangements. Bovine β-lactoglobulin (BLG), a protein of interest in food science, exhibits a strong propensity to bind various bioactive molecules. We probed the effects of the binding of biliverdin (BV), a tetrapyrrole linear chromophore, on the stability of BLG under pressure, by combining in situ HP-small-angle neutron scattering (SANS) and HP-UV absorption spectroscopy. Although BV induces a slight destabilization of BLG during HP-induced unfolding, a ligand excess strongly prevents BLG oligomerization. Moreover, at SANS resolution, an excess of BV induces the complete recovery of the protein “native” 3D structure after HP removal, despite the presence of the BV covalently bound adduct. Mass spectrometry highlights the crucial role of cysteine residues in the competitive and protective effects of BV during pressure denaturation of BLG through SH/S-S exchange.
- Published
- 2022
49. Modelling Protein Plasticity: The Example of Frataxin and Its Variants
- Author
-
Simone Botticelli, Giovanni La Penna, Germano Nobili, Giancarlo Rossi, Francesco Stellato, and Silvia Morante
- Subjects
frataxin ,Organic Chemistry ,Settore FIS/07 ,Pharmaceutical Science ,high-performance computing ,Analytical Chemistry ,Friedreich Ataxia ,Chemistry (miscellaneous) ,Iron-Binding Proteins ,Drug Discovery ,molecular statistics ,iron homeostasis ,cancer ,Humans ,Thermodynamics ,Molecular Medicine ,Physical and Theoretical Chemistry ,Protein Unfolding - Abstract
Frataxin (FXN) is a protein involved in storage and delivery of iron in the mitochondria. Single-point mutations in the FXN gene lead to reduced production of functional frataxin, with the consequent dyshomeostasis of iron. FXN variants are at the basis of neurological impairment (the Friedreich’s ataxia) and several types of cancer. By using altruistic metadynamics in conjunction with the maximal constrained entropy principle, we estimate the change of free energy in the protein unfolding of frataxin and of some of its pathological mutants. The sampled configurations highlight differences between the wild-type and mutated sequences in the stability of the folded state. In partial agreement with thermodynamic experiments, where most of the analyzed variants are characterized by lower thermal stability compared to wild type, the D104G variant is found with a stability comparable to the wild-type sequence and a lower water-accessible surface area. These observations, obtained with the new approach we propose in our work, point to a functional switch, affected by single-point mutations, of frataxin from iron storage to iron release. The method is suitable to investigate wide structural changes in proteins in general, after a proper tuning of the chosen collective variable used to perform the transition.
- Published
- 2022
- Full Text
- View/download PDF
50. A DNA repair-independent role for alkyladenine DNA glycosylase in alkylation-induced unfolded protein response
- Author
-
Larissa Milano, Clara F. Charlier, Rafaela Andreguetti, Thomas Cox, Eleanor Healing, Marcos P. Thomé, Ruan M. Elliott, Leona D. Samson, Jean-Yves Masson, Guido Lenz, João Antonio P. Henriques, Axel Nohturfft, and Lisiane B. Meira
- Subjects
X-Box Binding Protein 1 ,Multidisciplinary ,Alkylation ,DNA Repair ,Brain Neoplasms ,Endoplasmic Reticulum Stress ,female genital diseases and pregnancy complications ,DNA Glycosylases ,Mice ,polycyclic compounds ,Animals ,Humans ,Glioblastoma ,Protein Unfolding - Abstract
Alkylating agents damage DNA and proteins and are widely used in cancer chemotherapy. While cellular responses to alkylation-induced DNA damage have been explored, knowledge of how alkylation affects global cellular stress responses is sparse. Here, we examined the effects of the alkylating agent methylmethane sulfonate (MMS) on gene expression in mouse liver, using mice deficient in alkyladenine DNA glycosylase (Aag), the enzyme that initiates the repair of alkylated DNA bases. MMS induced a robust transcriptional response in wild-type liver that included markers of the endoplasmic reticulum (ER) stress/unfolded protein response (UPR) known to be controlled by XBP1, a key UPR effector. Importantly, this response is significantly reduced in the Aag knockout. To investigate how AAG affects alkylation-induced UPR, the expression of UPR markers after MMS treatment was interrogated in human glioblastoma cells expressing different AAG levels. Alkylation induced the UPR in cells expressing AAG; conversely, AAG knockdown compromised UPR induction and led to a defect in XBP1 activation. To verify the requirements for the DNA repair activity of AAG in this response, AAG knockdown cells were complemented with wild-type Aag or with an Aag variant producing a glycosylase-deficient AAG protein. As expected, the glycosylase-defective Aag does not fully protect AAG knockdown cells against MMS-induced cytotoxicity. Remarkably, however, alkylation-induced XBP1 activation is fully complemented by the catalytically inactive AAG enzyme. This work establishes that, besides its enzymatic activity, AAG has noncanonical functions in alkylation-induced UPR that contribute to cellular responses to alkylation.
- Published
- 2022
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.