refolding kinetics
Recently Published Documents


TOTAL DOCUMENTS

41
(FIVE YEARS 4)

H-INDEX

18
(FIVE YEARS 0)

2021 ◽  
Author(s):  
Katharina F Hohmann ◽  
Anja Blümler ◽  
Alexander Heckel ◽  
Boris Fürtig

Abstract In bacteria RNA gene regulatory elements refold dependent on environmental clues between two or more long-lived conformational states each associated with a distinct regulatory state. The refolding kinetics are strongly temperature-dependent and especially at lower temperatures they reach timescales that are biologically not accessible. To overcome this problem, RNA chaperones have evolved. However, the precise molecular mechanism of how these proteins accelerate RNA refolding reactions remains enigmatic. Here we show how the RNA chaperone StpA of Escherichia coli leads to an acceleration of a bistable RNA’s refolding kinetics through the selective destabilization of key base pairing interactions. We find in laser assisted real-time NMR experiments on photocaged bistable RNAs that the RNA chaperone leads to a two-fold increase in refolding rates at low temperatures due to reduced stability of ground state conformations. Further, we can show that upon interaction with StpA, base pairing interactions in the bistable RNA are modulated to favor refolding through the dominant pseudoknotted transition pathway. Our results shed light on the molecular mechanism of the interaction between RNA chaperones and bistable RNAs and are the first step into a functional classification of chaperones dependent on their biophysical mode of operation.


Biomedicines ◽  
2021 ◽  
Vol 9 (10) ◽  
pp. 1395
Author(s):  
Colleen Kelly ◽  
Matthew J. Gage

While protein refolding has been studied for over 50 years since the pioneering work of Christian Anfinsen, there have been a limited number of studies correlating results between chemical, thermal, and mechanical unfolding. The limited knowledge of the relationship between these processes makes it challenging to compare results between studies if different refolding methods were applied. Our current work compares the energetic barriers and folding rates derived from chemical, thermal, and mechanical experiments using an immunoglobulin-like domain from the muscle protein titin as a model system. This domain, I83, has high solubility and low stability relative to other Ig domains in titin, though its stability can be modulated by calcium. Our experiments demonstrated that the free energy of refolding was equivalent with all three techniques, but the refolding rates exhibited differences, with mechanical refolding having slightly faster rates. This suggests that results from equilibrium-based measurements can be compared directly but care should be given comparing refolding kinetics derived from refolding experiments that used different unfolding methods.


2021 ◽  
Vol 105 (6) ◽  
pp. 2243-2260
Author(s):  
Jan Niklas Pauk ◽  
Janani Raju Palanisamy ◽  
Julian Kager ◽  
Krisztina Koczka ◽  
Gerald Berghammer ◽  
...  

Abstract Overexpression of recombinant proteins in Escherichia coli results in misfolded and non-active protein aggregates in the cytoplasm, so-called inclusion bodies (IB). In recent years, a change in the mindset regarding IBs could be observed: IBs are no longer considered an unwanted waste product, but a valid alternative to produce a product with high yield, purity, and stability in short process times. However, solubilization of IBs and subsequent refolding is necessary to obtain a correctly folded and active product. This protein refolding process is a crucial downstream unit operation—commonly done as a dilution in batch or fed-batch mode. Drawbacks of the state-of-the-art include the following: the large volume of buffers and capacities of refolding tanks, issues with uniform mixing, challenging analytics at low protein concentrations, reaction kinetics in non-usable aggregates, and generally low re-folding yields. There is no generic platform procedure available and a lack of robust control strategies. The introduction of Quality by Design (QbD) is the method-of-choice to provide a controlled and reproducible refolding environment. However, reliable online monitoring techniques to describe the refolding kinetics in real-time are scarce. In our view, only monitoring and control of re-folding kinetics can ensure a productive, scalable, and versatile platform technology for re-folding processes. For this review, we screened the current literature for a combination of online process analytical technology (PAT) and modeling techniques to ensure a controlled refolding process. Based on our research, we propose an integrated approach based on the idea that all aspects that cannot be monitored directly are estimated via digital twins and used in real-time for process control. Key points • Monitoring and a thorough understanding of refolding kinetics are essential for model-based control of refolding processes. • The introduction of Quality by Design combining Process Analytical Technology and modeling ensures a robust platform for inclusion body refolding.


2020 ◽  
Author(s):  
Soham Chakraborty ◽  
Deep Chaudhuri ◽  
Dyuti Chaudhuri ◽  
Vihan Singh ◽  
Souradeep Banerjee ◽  
...  

AbstractStudies of free energy, kinetics or elasticity are common to most disciplines of science. Detailed quantification of these properties demands number of specialized technologies. Furthermore, monitoring ‘perturbation’ in any of these properties, in presence of external stimuli (protein/DNA/drugs/nanoparticles etc.), requires multiple experiments. However, none of these available technologies can monitor these perturbations simultaneously in real time on the very same molecule in a single shot experiment.Here we present real-time microfluidics-magnetic tweezers technology with the unique advantage of tracking a single protein dynamics for hours, in absence of any significant drift, with the flexibility of changing physical environment in real time. Remarkable stability of this technique allows us to quantify five molecular properties (unfolding kinetics, refolding kinetics, conformational change, chain flexibility, and ΔG for folding/unfolding), and most importantly, their dynamic perturbation upon interacting with salt on the same protein molecule from a single experiment. We observe salt reshapes the energy landscape by two specific ways: increasing the refolding kinetics and decreasing the unfolding kinetics, which is characterized as mean first passage time. Importantly, from the same trajectory, we calculated the flexibility of the protein polymer, which changes with salt concentration and can be explained by our modified ‘electrolyte FJC model’. The correlation between ΔG, kinetics and polymer elasticity strongly argues for a stiffness driven energy landscape of proteins. Having the advantage of sub nanometer resolution, this methodology will open new exciting window to study proteins – one such examples is demonstrated in this article: electrolyte driven conformational fluctuation under force, which was not studied before.


2018 ◽  
Author(s):  
Huafeng Xu

AbstractThe heat shock protein 70 (Hsp70) chaperones, vital to the proper folding of proteins inside cells, consume ATP and require cochaperones in assisting protein folding. It is unclear whether Hsp70 can utilize the free energy from ATP hydrolysis to fold a protein into a native state that is thermodynamically unstable in the chaperone-free equilibrium. Here we present a model of Hsp70-mediated protein folding, which predicts that Hsp70, as a result of differential stimulation of ATP hydrolysis by its Hsp40 cochaperone, dissociates faster from a substrate in fold-competent conformations than from one in misfolding-prone conformations, thus elevating the native concentration above and suppressing the misfolded concentration below their respective equilibrium values. Previous models would not make or imply these predictions, which are experimentally testable. Our model quantitatively reproduces experimental refolding kinetics, predicts how modulations of the Hsp70/Hsp40 chaperone system affect protein folding, and suggests new approaches to regulating cellular protein quality.


2013 ◽  
Vol 104 (2) ◽  
pp. 310a
Author(s):  
Jens Herzog ◽  
Tim R. Leonard ◽  
Azim Jinha ◽  
Walter Herzog
Keyword(s):  

2012 ◽  
Vol 27 ◽  
pp. 343-348 ◽  
Author(s):  
Catarina S. H. Jesus ◽  
Daniela C. Vaz ◽  
Maria J. M. Saraiva ◽  
Rui M. M. Brito

Transthyretin (TTR) is a homotetrameric protein implicated in several amyloid diseases. The mechanism by which TTR is converted into elongated fibrillar assemblies has been extensively investigated, and numerous studies showed that dissociation of the native tetrameric structure into partially unfolded monomeric species precedes amyloid formation. The small differences observed in the crystal structures of different TTR variants, as well as the thermodynamics and kinetics of tetramer dissociation, do not seem to completely justify the amyloidogenic potential of different TTR variants. With this in mind, we have studied the refolding kinetics of WT-TTR and its most common amyloidogenic variant V30M-TTR, monitoring changes in intrinsic tryptophan fluorescence at different urea and protein concentrations. Our results demonstrate that thein vitrorefolding mechanisms of WT- and V30M-TTR are similar, involving a dimeric intermediate. However, there are large differences in the refolding rate constants for the two variants, specially close to physiological conditions. Interestingly, tetramer formation occurs at a much slower rate in the amyloidogenic variant V30M-TTR than in WT-TTR, which in thein vivosetting may promote the accumulation of monomeric species in the extracellular environment, resulting in higher susceptibility for aggregation and amyloid formation instead of spontaneous refolding.


Sign in / Sign up

Export Citation Format

Share Document