Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 943
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Annu Rev Biochem ; 86: 97-122, 2017 06 20.
Article in English | MEDLINE | ID: mdl-28489421

ABSTRACT

A healthy proteome is essential for cell survival. Protein misfolding is linked to a rapidly expanding list of human diseases, ranging from neurodegenerative diseases to aging and cancer. Many of these diseases are characterized by the accumulation of misfolded proteins in intra- and extracellular inclusions, such as amyloid plaques. The clear link between protein misfolding and disease highlights the need to better understand the elaborate machinery that manages proteome homeostasis, or proteostasis, in the cell. Proteostasis depends on a network of molecular chaperones and clearance pathways involved in the recognition, refolding, and/or clearance of aberrant proteins. Recent studies reveal that an integral part of the cellular management of misfolded proteins is their spatial sequestration into several defined compartments. Here, we review the properties, function, and formation of these compartments. Spatial sequestration plays a central role in protein quality control and cellular fitness and represents a critical link to the pathogenesis of protein aggregation-linked diseases.


Subject(s)
Aging/metabolism , Molecular Chaperones/metabolism , Neurodegenerative Diseases/metabolism , Protein Aggregation, Pathological/metabolism , Proteostasis Deficiencies/metabolism , Aging/genetics , Aging/pathology , Amyloidogenic Proteins/chemistry , Amyloidogenic Proteins/genetics , Amyloidogenic Proteins/metabolism , Cell Compartmentation , Gene Expression Regulation , Humans , Molecular Chaperones/genetics , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/pathology , Prion Proteins/chemistry , Prion Proteins/genetics , Prion Proteins/metabolism , Protein Aggregation, Pathological/genetics , Protein Aggregation, Pathological/pathology , Protein Biosynthesis , Protein Conformation , Protein Folding , Protein Refolding , Proteolysis , Proteostasis Deficiencies/genetics , Proteostasis Deficiencies/pathology
2.
Nature ; 590(7847): 666-670, 2021 02.
Article in English | MEDLINE | ID: mdl-33442061

ABSTRACT

A non-enveloped virus requires a membrane lesion to deliver its genome into a target cell1. For rotaviruses, membrane perforation is a principal function of the viral outer-layer protein, VP42,3. Here we describe the use of electron cryomicroscopy to determine how VP4 performs this function and show that when activated by cleavage to VP8* and VP5*, VP4 can rearrange on the virion surface from an 'upright' to a 'reversed' conformation. The reversed structure projects a previously buried 'foot' domain outwards into the membrane of the host cell to which the virion has attached. Electron cryotomograms of virus particles entering cells are consistent with this picture. Using a disulfide mutant of VP4, we have also stabilized a probable intermediate in the transition between the two conformations. Our results define molecular mechanisms for the first steps of the penetration of rotaviruses into the membranes of target cells and suggest similarities with mechanisms postulated for other viruses.


Subject(s)
Capsid Proteins/chemistry , Capsid Proteins/ultrastructure , Cryoelectron Microscopy , Protein Refolding , Rotavirus/metabolism , Rotavirus/ultrastructure , Virus Internalization , Animals , Antigens, Viral/metabolism , Capsid Proteins/genetics , Capsid Proteins/metabolism , Cell Line , Cell Membrane/chemistry , Cell Membrane/metabolism , Cell Membrane/ultrastructure , Disulfides/chemistry , Disulfides/metabolism , Models, Molecular , Mutant Proteins/chemistry , Mutant Proteins/genetics , Mutant Proteins/metabolism , Mutant Proteins/ultrastructure , Mutation , Protein Conformation , RNA-Binding Proteins/metabolism , Rotavirus/chemistry , Rotavirus/physiology , Viral Nonstructural Proteins/metabolism , Virion/chemistry , Virion/metabolism , Virion/ultrastructure
3.
Proc Natl Acad Sci U S A ; 121(19): e2403049121, 2024 May 07.
Article in English | MEDLINE | ID: mdl-38691587

ABSTRACT

Molecular chaperones assist in protein refolding by selectively binding to proteins in their nonnative states. Despite progress in creating artificial chaperones, these designs often have a limited range of substrates they can work with. In this paper, we present molecularly imprinted flexible polymer nanoparticles (nanoMIPs) designed as customizable biomimetic chaperones. We used model proteins such as cytochrome c, laccase, and lipase to screen polymeric monomers and identify the most effective formulations, offering tunable charge and hydrophobic properties. Utilizing a dispersed phase imprinting approach, we employed magnetic beads modified with destabilized whole-protein as solid-phase templates. This process involves medium exchange facilitated by magnetic pulldowns, resulting in the synthesis of nanoMIPs featuring imprinted sites that effectively mimic chaperone cavities. These nanoMIPs were able to selectively refold denatured enzymes, achieving up to 86.7% recovery of their activity, significantly outperforming control samples. Mechanistic studies confirmed that nanoMIPs preferentially bind denatured rather than native enzymes, mimicking natural chaperone interactions. Multifaceted analyses support the functionality of nanoMIPs, which emulate the protective roles of chaperones by selectively engaging with denatured proteins to inhibit aggregation and facilitate refolding. This approach shows promise for widespread use in protein recovery within biocatalysis and biomedicine.


Subject(s)
Molecular Chaperones , Nanoparticles , Polymers , Protein Denaturation , Nanoparticles/chemistry , Molecular Chaperones/chemistry , Molecular Chaperones/metabolism , Polymers/chemistry , Protein Refolding , Protein Folding , Cytochromes c/chemistry , Cytochromes c/metabolism , Laccase/chemistry , Laccase/metabolism , Lipase/chemistry , Lipase/metabolism
4.
Nature ; 578(7794): 317-320, 2020 02.
Article in English | MEDLINE | ID: mdl-31996849

ABSTRACT

The ability to reverse protein aggregation is vital to cells1,2. Hsp100 disaggregases such as ClpB and Hsp104 are proposed to catalyse this reaction by translocating polypeptide loops through their central pore3,4. This model of disaggregation is appealing, as it could explain how polypeptides entangled within aggregates can be extracted and subsequently refolded with the assistance of Hsp704,5. However, the model is also controversial, as the necessary motor activity has not been identified6-8 and recent findings indicate non-processive mechanisms such as entropic pulling or Brownian ratcheting9,10. How loop formation would be accomplished is also obscure. Indeed, cryo-electron microscopy studies consistently show single polypeptide strands in the Hsp100 pore11,12. Here, by following individual ClpB-substrate complexes in real time, we unambiguously demonstrate processive translocation of looped polypeptides. We integrate optical tweezers with fluorescent-particle tracking to show that ClpB translocates both arms of the loop simultaneously and switches to single-arm translocation when encountering obstacles. ClpB is notably powerful and rapid; it exerts forces of more than 50 pN at speeds of more than 500 residues per second in bursts of up to 28 residues. Remarkably, substrates refold while exiting the pore, analogous to co-translational folding. Our findings have implications for protein-processing phenomena including ubiquitin-mediated remodelling by Cdc48 (or its mammalian orthologue p97)13 and degradation by the 26S proteasome14.


Subject(s)
Endopeptidase Clp/metabolism , Escherichia coli Proteins/metabolism , Heat-Shock Proteins/metabolism , Peptides/chemistry , Peptides/metabolism , Protein Aggregates , Adenosine Triphosphatases/chemistry , Adenosine Triphosphatases/metabolism , Arabidopsis Proteins/chemistry , Arabidopsis Proteins/metabolism , Endopeptidase Clp/chemistry , Escherichia coli Proteins/chemistry , Fluorescence , Heat-Shock Proteins/chemistry , Kinetics , Molecular Chaperones/chemistry , Molecular Chaperones/metabolism , Optical Tweezers , Proteasome Endopeptidase Complex/metabolism , Protein Multimerization , Protein Refolding , Ubiquitin/metabolism
5.
Mol Cell ; 68(1): 76-88.e6, 2017 Oct 05.
Article in English | MEDLINE | ID: mdl-28943314

ABSTRACT

Chromatin remodelers catalyze dynamic packaging of the genome by carrying out nucleosome assembly/disassembly, histone exchange, and nucleosome repositioning. Remodeling results in evenly spaced nucleosomes, which requires probing both sides of the nucleosome, yet the way remodelers organize sliding activity to achieve this task is not understood. Here, we show that the monomeric Chd1 remodeler shifts DNA back and forth by dynamically alternating between different segments of the nucleosome. During sliding, Chd1 generates unstable remodeling intermediates that spontaneously relax to a pre-remodeled position. We demonstrate that nucleosome sliding is tightly controlled by two regulatory domains: the DNA-binding domain, which interferes with sliding when its range is limited by a truncated linking segment, and the chromodomains, which play a key role in substrate discrimination. We propose that active interplay of the ATPase motor with the regulatory domains may promote dynamic nucleosome structures uniquely suited for histone exchange and chromatin reorganization during transcription.


Subject(s)
DNA-Binding Proteins/genetics , DNA/genetics , Histones/genetics , Nucleosomes/chemistry , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae/genetics , Transcription, Genetic , Amino Acid Sequence , Animals , Binding Sites , Chromatin Assembly and Disassembly , Cloning, Molecular , DNA/chemistry , DNA/metabolism , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Histones/chemistry , Histones/metabolism , Models, Molecular , Nucleosomes/metabolism , Plasmids/chemistry , Plasmids/metabolism , Protein Binding , Protein Conformation, alpha-Helical , Protein Interaction Domains and Motifs , Protein Multimerization , Protein Refolding , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/metabolism , Substrate Specificity , Xenopus laevis/genetics , Xenopus laevis/metabolism
6.
Proc Natl Acad Sci U S A ; 119(33): e2205278119, 2022 08 16.
Article in English | MEDLINE | ID: mdl-35951650

ABSTRACT

Lambdoid bacteriophage Q proteins are transcription antipausing and antitermination factors that enable RNA polymerase (RNAP) to read through pause and termination sites. Q proteins load onto RNAP engaged in promoter-proximal pausing at a Q binding element (QBE) and adjacent sigma-dependent pause element to yield a Q-loading complex, and they translocate with RNAP as a pausing-deficient, termination-deficient Q-loaded complex. In previous work, we showed that the Q protein of bacteriophage 21 (Q21) functions by forming a nozzle that narrows and extends the RNAP RNA-exit channel, preventing formation of pause and termination RNA hairpins. Here, we report atomic structures of four states on the pathway of antitermination by the Q protein of bacteriophage λ (Qλ), a Q protein that shows no sequence similarity to Q21 and that, unlike Q21, requires the transcription elongation factor NusA for efficient antipausing and antitermination. We report structures of Qλ, the Qλ-QBE complex, the NusA-free pre-engaged Qλ-loading complex, and the NusA-containing engaged Qλ-loading complex. The results show that Qλ, like Q21, forms a nozzle that narrows and extends the RNAP RNA-exit channel, preventing formation of RNA hairpins. However, the results show that Qλ has no three-dimensional structural similarity to Q21, employs a different mechanism of QBE recognition than Q21, and employs a more complex process for loading onto RNAP than Q21, involving recruitment of Qλ to form a pre-engaged loading complex, followed by NusA-facilitated refolding of Qλ to form an engaged loading complex. The results establish that Qλ and Q21 are not structural homologs and are solely functional analogs.


Subject(s)
Bacteriophage lambda , Escherichia coli Proteins , Protein Refolding , Transcription Termination, Genetic , Transcriptional Elongation Factors , Viral Proteins , Bacteriophage lambda/genetics , Cryoelectron Microscopy , DNA-Directed RNA Polymerases/chemistry , Escherichia coli Proteins/chemistry , Protein Conformation , Transcriptional Elongation Factors/chemistry , Viral Proteins/chemistry
7.
Proc Natl Acad Sci U S A ; 119(48): e2210536119, 2022 11 29.
Article in English | MEDLINE | ID: mdl-36417429

ABSTRACT

The journey by which proteins navigate their energy landscapes to their native structures is complex, involving (and sometimes requiring) many cellular factors and processes operating in partnership with a given polypeptide chain's intrinsic energy landscape. The cytosolic environment and its complement of chaperones play critical roles in granting many proteins safe passage to their native states; however, it is challenging to interrogate the folding process for large numbers of proteins in a complex background with most biophysical techniques. Hence, most chaperone-assisted protein refolding studies are conducted in defined buffers on single purified clients. Here, we develop a limited proteolysis-mass spectrometry approach paired with an isotope-labeling strategy to globally monitor the structures of refolding Escherichia coli proteins in the cytosolic medium and with the chaperones, GroEL/ES (Hsp60) and DnaK/DnaJ/GrpE (Hsp70/40). GroEL can refold the majority (85%) of the E. coli proteins for which we have data and is particularly important for restoring acidic proteins and proteins with high molecular weight, trends that come to light because our assay measures the structural outcome of the refolding process itself, rather than binding or aggregation. For the most part, DnaK and GroEL refold a similar set of proteins, supporting the view that despite their vastly different structures, these two chaperones unfold misfolded states, as one mechanism in common. Finally, we identify a cohort of proteins that are intransigent to being refolded with either chaperone. We suggest that these proteins may fold most efficiently cotranslationally, and then remain kinetically trapped in their native conformations.


Subject(s)
Escherichia coli Proteins , Protein Refolding , Proteome , Cytosol/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Heat-Shock Proteins/metabolism , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , Proteome/metabolism
8.
J Cell Sci ; 135(6)2022 03 15.
Article in English | MEDLINE | ID: mdl-35237814

ABSTRACT

Osp94 (also known as HSPA4L or HSPH3), a member of the Hsp110/Sse1 family of heat-shock proteins, has a longer C-terminus than found in Hsc70/Hsp70 family proteins, composed of the loop region with a partial substrate-binding domain (SBD) ß (L), and the SBDα and the C-terminal extension (H), but the functions of these domains are poorly understood. Here, we found that Osp94 suppressed heat-induced aggregation of luciferase (Luc). Osp94-bound heat-inactivated Luc was reactivated in the presence of rabbit reticulocyte lysate (RRL) and/or a combination of Hsc70 and Hsp40 (also known as HSPA8 and DNAJB1, respectively). Targeted deletion mutagenesis revealed that the SBDß and H domains of Osp94 are critical for protein disaggregation and RRL-mediated refolding. Reactivation of Hsp90-bound heat-inactivated Luc was abolished in the absence of RRL but compensated for by PA28α (also known as PSME1), a proteasome activator. Interestingly, the LH domain also reactivated heat-inactivated Luc, independently of PA28α. Biotin-tag cross-linking experiments indicated that the LH domain and PA28α interact with Luc bound by Hsp90 during refolding. A chimeric protein in which the H domain was exchanged for PA28α also mediated disaggregation and reactivation of heat-inactivated Luc. These results indicate that Osp94 acts as a holdase, and that the C-terminal region plays a PA28α-like role in the refolding of unfolded proteins.


Subject(s)
HSP40 Heat-Shock Proteins , HSP70 Heat-Shock Proteins , Animals , Family , HSC70 Heat-Shock Proteins/metabolism , HSP40 Heat-Shock Proteins/genetics , HSP40 Heat-Shock Proteins/metabolism , HSP70 Heat-Shock Proteins/genetics , HSP70 Heat-Shock Proteins/metabolism , Heat-Shock Proteins/metabolism , Protein Refolding , Rabbits
9.
Protein Expr Purif ; 221: 106504, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38782082

ABSTRACT

Reteplase (recombinant plasminogen activator, rPA) is a mutant non-glycosylated tissue-type plasminogen activator (tPA) containing 355 amino acids with longer half-life and promising thrombolytic activity than its original counterpart, full length tPA. In this study, we aimed to produce and optimize the purification process of recombinant tissue-type plasminogen activator (tPA) known as Reteplase (rPA). Reteplase cDNA synthesized from total mRNA isolated from human placenta was PCR amplified, cloned into a pET-28a(+) E. coli expression vector and expressed in Rosetta-gami 2 E. coli (NovagenⓇ) host. rPA was expressed as an inclusion body in E. coli and its biological activity was achieved after single step solubilization, purification and refolding. We exploited the strategy of Slow Refolding using Gradual Dialysis (SRGD) in which a refolding buffer containing glutathione oxidized (1 mM GSSG) and glutathione reduced (3 mM GSH) and pH 9.0 was used. Using the SRGD method, we were able to successfully obtain the protein in its active form. We obtained 4.26 mg of active refolded protein from a 50 mL culture that was scaled up in a bioreactor. The purity and homogeneity of rPA was evaluated by SDS-PAGE, Western blotting and mass spectrometry. Circular dichroism spectroscopy was conducted to evaluate the refolding and stability of the refolded rPA in comparison to reference standard rPA. The thrombolytic potential of rPA was assessed by fibrin plate assay and In Vitro clot lysis assay. The presented protocol offers a viable approach for enhancing both the yield and refolding efficiency of reteplase, potentially resulting in an increase in yield.


Subject(s)
Escherichia coli , Protein Refolding , Recombinant Proteins , Tissue Plasminogen Activator , Tissue Plasminogen Activator/genetics , Tissue Plasminogen Activator/chemistry , Tissue Plasminogen Activator/isolation & purification , Tissue Plasminogen Activator/biosynthesis , Escherichia coli/genetics , Escherichia coli/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Recombinant Proteins/biosynthesis , Humans , Gene Expression , Fibrinolytic Agents/chemistry , Fibrinolytic Agents/isolation & purification , Cloning, Molecular
10.
Anal Bioanal Chem ; 416(12): 3019-3032, 2024 May.
Article in English | MEDLINE | ID: mdl-38573344

ABSTRACT

Inclusion bodies (IBs) are protein aggregates formed as a result of overexpression of recombinant protein in E. coli. The formation of IBs is a valuable strategy of recombinant protein production despite the need for additional processing steps, i.e., isolation, solubilization and refolding. Industrial process development of protein refolding is a labor-intensive task based largely on empirical approaches rather than knowledge-driven strategies. A prerequisite for knowledge-driven process development is a reliable monitoring strategy. This work explores the potential of intrinsic tryptophan and tyrosine fluorescence for real-time and in situ monitoring of protein refolding. In contrast to commonly established process analytical technology (PAT), this technique showed high sensitivity with reproducible measurements for protein concentrations down to 0.01 g L - 1 . The change of protein conformation during refolding is reflected as a shift in the position of the maxima of the tryptophan and tyrosine fluorescence spectra as well as change in the signal intensity. The shift in the peak position, expressed as average emission wavelength of a spectrum, was correlated to the amount of folding intermediates whereas the intensity integral correlates to the extent of aggregation. These correlations were implemented as an observation function into a mechanistic model. The versatility and transferability of the technique were demonstrated on the refolding of three different proteins with varying structural complexity. The technique was also successfully applied to detect the effect of additives and process mode on the refolding process efficiency. Thus, the methodology presented poses a generic and reliable PAT tool enabling real-time process monitoring of protein refolding.


Subject(s)
Inclusion Bodies , Protein Refolding , Spectrometry, Fluorescence , Inclusion Bodies/chemistry , Inclusion Bodies/metabolism , Spectrometry, Fluorescence/methods , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Tryptophan/chemistry , Escherichia coli/metabolism , Escherichia coli/chemistry , Tyrosine/chemistry , Fluorescence , Protein Folding
11.
Nature ; 563(7730): 209-213, 2018 11.
Article in English | MEDLINE | ID: mdl-30232455

ABSTRACT

Tc toxins secrete toxic enzymes into host cells using a unique syringe-like injection mechanism. They are composed of three subunits, TcA, TcB and TcC. TcA forms the translocation channel and the TcB-TcC heterodimer functions as a cocoon that shields the toxic enzyme. Binding of the cocoon to the channel triggers opening of the cocoon and translocation of the toxic enzyme into the channel. Here we show in atomic detail how the assembly of the three components activates the toxin. We find that part of the cocoon completely unfolds and refolds into an alternative conformation upon binding. The presence of the toxic enzyme inside the cocoon is essential for its subnanomolar binding affinity for the TcA subunit. The enzyme passes through a narrow negatively charged constriction site inside the cocoon, probably acting as an extruder that releases the unfolded protein with its C terminus first into the translocation channel.


Subject(s)
Bacterial Toxins/chemistry , Bacterial Toxins/metabolism , Cryoelectron Microscopy , Multiprotein Complexes/ultrastructure , Photorhabdus/ultrastructure , Protein Refolding , Protein Unfolding , ADP Ribose Transferases/chemistry , ADP Ribose Transferases/metabolism , ADP Ribose Transferases/ultrastructure , Bacterial Toxins/biosynthesis , Cytotoxins/biosynthesis , Cytotoxins/chemistry , Cytotoxins/metabolism , Models, Biological , Models, Molecular , Multiprotein Complexes/biosynthesis , Multiprotein Complexes/chemistry , Multiprotein Complexes/metabolism , Photorhabdus/chemistry , Protein Conformation , Protein Transport
12.
Proc Natl Acad Sci U S A ; 118(32)2021 08 10.
Article in English | MEDLINE | ID: mdl-34362850

ABSTRACT

DegP is an oligomeric protein with dual protease and chaperone activity that regulates protein homeostasis and virulence factor trafficking in the periplasm of gram-negative bacteria. A number of oligomeric architectures adopted by DegP are thought to facilitate its function. For example, DegP can form a "resting" hexamer when not engaged to substrates, mitigating undesired proteolysis of cellular proteins. When bound to substrate proteins or lipid membranes, DegP has been shown to populate a variety of cage- or bowl-like oligomeric states that have increased proteolytic activity. Though a number of DegP's substrate-engaged structures have been robustly characterized, detailed mechanistic information underpinning its remarkable oligomeric plasticity and the corresponding interplay between these dynamics and biological function has remained elusive. Here, we have used a combination of hydrodynamics and NMR spectroscopy methodologies in combination with cryogenic electron microscopy to shed light on the apo-DegP self-assembly mechanism. We find that, in the absence of bound substrates, DegP populates an ensemble of oligomeric states, mediated by self-assembly of trimers, that are distinct from those observed in the presence of substrate. The oligomeric distribution is sensitive to solution ionic strength and temperature and is shifted toward larger oligomeric assemblies under physiological conditions. Substrate proteins may guide DegP toward canonical cage-like structures by binding to these preorganized oligomers, leading to changes in conformation. The properties of DegP self-assembly identified here suggest that apo-DegP can rapidly shift its oligomeric distribution in order to respond to a variety of biological insults.


Subject(s)
Heat-Shock Proteins/chemistry , Heat-Shock Proteins/metabolism , Periplasmic Proteins/chemistry , Periplasmic Proteins/metabolism , Serine Endopeptidases/chemistry , Serine Endopeptidases/metabolism , Cryoelectron Microscopy , Dynamic Light Scattering , Heat-Shock Proteins/genetics , Molecular Chaperones/chemistry , Molecular Chaperones/metabolism , Mutation , Nuclear Magnetic Resonance, Biomolecular/methods , Osmolar Concentration , Periplasmic Proteins/genetics , Protein Domains , Protein Refolding , Serine Endopeptidases/genetics , Temperature
13.
Molecules ; 29(11)2024 May 22.
Article in English | MEDLINE | ID: mdl-38893310

ABSTRACT

The human paraoxonase 2 (PON2) is the oldest member of a small family of arylesterase and lactonase enzymes, representing the first line of defense against bacterial infections and having a major role in ROS-associated diseases such as cancer, cardiovascular diseases, neurodegeneration, and diabetes. Specific Post-Translational Modifications (PTMs) clustering nearby two residues corresponding to pon2 polymorphic sites and their impact on the catalytic activity are not yet fully understood. Thus, the goal of the present study was to develop an improved PON2 purification protocol to obtain a higher amount of protein suitable for in-depth biochemical studies and biotechnological applications. To this end, we also tested several compounds to stabilize the active monomeric form of the enzyme. Storing the enzyme at 4 °C with 30 mM Threalose had the best impact on the activity, which was preserved for at least 30 days. The catalytic parameters against the substrate 3-Oxo-dodecanoyl-Homoserine Lactone (3oxoC12-HSL) and the enzyme ability to interfere with the biofilm formation of Pseudomonas aeruginosa (PAO1) were determined, showing that the obtained enzyme is well suited for downstream applications. Finally, we used the purified rPON2 to detect, by the direct molecular fishing (DMF) method, new putative PON2 interactors from soluble extracts of HeLa cells.


Subject(s)
Aryldialkylphosphatase , Proteomics , Aryldialkylphosphatase/metabolism , Aryldialkylphosphatase/chemistry , Humans , Proteomics/methods , Protein Refolding , Pseudomonas aeruginosa/enzymology , Enzyme Stability , Biofilms , Protein Processing, Post-Translational
14.
Protein Expr Purif ; 208-209: 106278, 2023 08.
Article in English | MEDLINE | ID: mdl-37094772

ABSTRACT

MMP-2 has been reported as the most validated target for cancer progression and deserves further investigation. However, due to the lack of methods for obtaining large amounts of highly purified and bioactive MMP-2, identifying specific substrates and developing specific inhibitors of MMP-2 remains extremely difficult. In this study, the DNA fragment coding for pro-MMP-2 was inserted into plasmid pET28a in an oriented manner, and the resulting recombinant protein was effectively expressed and led to accumulation as inclusion bodies in E. coli. This protein was easy to purify to near homogeneity by the combination of common inclusion bodies purification procedure and cold ethanol fractionation. Then, our results of gelatin zymography and fluorometric assay revealed that pro-MMP-2 at least partially restored its natural structure and enzymatic activity after renaturation. We obtained approximately 11 mg refolded pro-MMP-2 protein from 1 L LB broth, which was higher than other strategies previously reported. In conclusion, a simple and cost-effective procedure for obtaining high amounts of functional MMP-2 was developed, which would contribute to the progress of studies on the gamut of biological action of this important proteinase. Furthermore, our protocol should be appropriate for the expression, purification, and refolding of other bacterial toxic proteins.


Subject(s)
Escherichia coli , Matrix Metalloproteinase 2 , Escherichia coli/metabolism , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 2/chemistry , Recombinant Proteins/chemistry , Bacterial Proteins/metabolism , Inclusion Bodies/chemistry , Protein Folding , Protein Refolding
15.
Nature ; 543(7645): 443-446, 2017 03 16.
Article in English | MEDLINE | ID: mdl-28241148

ABSTRACT

Loss of proteostasis underlies ageing and neurodegeneration characterized by the accumulation of protein aggregates and mitochondrial dysfunction. Although many neurodegenerative-disease-associated proteins can be found in mitochondria, it remains unclear how mitochondrial dysfunction and protein aggregation could be related. In dividing yeast cells, protein aggregates that form under stress or during ageing are preferentially retained by the mother cell, in part through tethering to mitochondria, while the disaggregase Hsp104 helps to dissociate aggregates and thereby enables refolding or degradation of misfolded proteins. Here we show that, in yeast, cytosolic proteins prone to aggregation are imported into mitochondria for degradation. Protein aggregates that form under heat shock contain both cytosolic and mitochondrial proteins and interact with the mitochondrial import complex. Many aggregation-prone proteins enter the mitochondrial intermembrane space and matrix after heat shock, and some do so even without stress. Timely dissolution of cytosolic aggregates requires the mitochondrial import machinery and proteases. Blocking mitochondrial import but not proteasome activity causes a marked delay in the degradation of aggregated proteins. Defects in cytosolic Hsp70s leads to enhanced entry of misfolded proteins into mitochondria and elevated mitochondrial stress. We term this mitochondria-mediated proteostasis mechanism MAGIC (mitochondria as guardian in cytosol) and provide evidence that it may exist in human cells.


Subject(s)
Cytosol/metabolism , Homeostasis , Mitochondria/metabolism , Protein Aggregates/physiology , Protein Folding , Proteins/chemistry , Proteins/metabolism , Saccharomyces cerevisiae , Cell Line , Cytosol/drug effects , HSP70 Heat-Shock Proteins/metabolism , Heat-Shock Response , Humans , Mitochondria/drug effects , Mitochondrial Proteins/chemistry , Mitochondrial Proteins/metabolism , Peptide Hydrolases/metabolism , Proteasome Endopeptidase Complex/metabolism , Protein Refolding , Protein Stability , Protein Transport/drug effects , Proteolysis , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/metabolism
16.
Protein Expr Purif ; 191: 106007, 2022 03.
Article in English | MEDLINE | ID: mdl-34728367

ABSTRACT

Metacaspases are known to have a fundamental role in apoptosis-like, a programmed cellular death (PCD) in plants, fungi, and protozoans. The last includes several parasites that cause diseases of great interest to public health, mostly without adequate treatment and included in the neglected tropical diseases category. One of them is Trypanosoma cruzi which causes Chagas disease and has two metacaspases involved in its PCD: TcMCA3 and TcMCA5. Their roles seemed different in PCD, TcMCA5 appears as a proapoptotic protein negatively regulated by its C-terminal sequence, while TcMCA3 is described as a cell cycle regulator. Despite this, the precise role of TcMCA3 and TcMCA5 and their atomic structures remain elusive. Therefore, developing methodologies to allow investigations of those metacaspases is relevant. Herein, we produced full-length and truncated versions of TcMCA5 and applied different strategies for their folded recombinant production from E. coli inclusion bodies. Biophysical assays probed the efficacy of the production method in providing a high yield of folded recombinant TcMCA5. Moreover, we modeled the TcMCA5 protein structure using experimental restraints obtained by XLMS. The experimental design for novel methods and the final protocol provided here can guide studies with other metacaspases. The production of TcMCA5 allows further investigations as protein crystallography, HTS drug discovery to create potential therapeutic in the treatment of Chagas' disease and in the way to clarify how the PCD works in the parasite.


Subject(s)
Caspases/chemistry , Protein Refolding , Protozoan Proteins/chemistry , Trypanosoma cruzi/enzymology , Caspases/genetics , Protein Domains , Protozoan Proteins/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Trypanosoma cruzi/genetics
17.
Protein Expr Purif ; 197: 106110, 2022 09.
Article in English | MEDLINE | ID: mdl-35577182

ABSTRACT

Interferon alpha-2b (IFNα-2b) is an essential cytokine widely used in hepatitis and cancer treatment. This paper presents a novel protocol for purification and efficient refolding of recombinant interferon alpha-2b (IFNα-2b) expressed as insoluble inclusion bodies in Escherichia coli. Purification of IFNα-2b from solubilized inclusion bodies was performed by solvent extraction using 2-butanol. Refolding conditions were optimized using the response surface method (RSM). Under optimized conditions, refolding yield of solvent-extracted IFNα-2b was 1.5 fold higher than refolding yield of unpurified IFNα-2b. High-concentration protein refolding was carried out by pulse-fed method, and refolding yield of 75% was achieved at a protein concentration of 300 µg ml-1. Under optimized conditions, 259.16 mg of purified IFNα-2b with the biological activity of 2.4 × 108 IU mg-1 was achieved per liter of bacterial culture. The developed protocol provides an efficient production process of high-quality IFNα-2b suitable for research and pharmaceutical applications.


Subject(s)
Escherichia coli , Inclusion Bodies , Escherichia coli/genetics , Escherichia coli/metabolism , Inclusion Bodies/metabolism , Interferon alpha-2/metabolism , Protein Refolding , Recombinant Proteins , Solvents
18.
Biomacromolecules ; 23(9): 3860-3865, 2022 09 12.
Article in English | MEDLINE | ID: mdl-36030420

ABSTRACT

We have reported that ureido polymers exhibit upper critical solution temperature (UCST)-type phase behavior in solution, which is the opposite of lower critical solution temperature (LCST)-type behavior. Furthermore, UCST-type ureido polymers undergo liquid-liquid phase separation (LLPS) upon cooling rather than the liquid-solid phase transition of the typical LCST-type polymers. In this study, ureido polymers with hydrophobic groups were prepared to evaluate the effects of cooling-induced LLPS of UCST-type polymers on refolding of proteins. When protein was heated with a ureido polymer functionalized with undecyl groups, aggregation of the protein was prevented. Subsequent cooling incubation resulted in the spontaneous release of the protein from the polymer. The released protein had enzymatic activity, suggesting that the protein refolded properly. Interestingly, efficient refolding was observed when the solution of the UCST-type ureido polymer and protein was incubated at around the phase separation temperature of the polymer, implying that cooling-induced LLPS of the polymer enhanced the release of the protein. Additionally, by centrifugation at 4 °C, the refolded protein was readily separated from the ureido polymers, which precipitated upon cooling.


Subject(s)
Polymers , Proteins , Hydrophobic and Hydrophilic Interactions , Phase Transition , Polymers/chemistry , Protein Refolding , Proteins/chemistry , Temperature
19.
Microb Cell Fact ; 21(1): 164, 2022 Aug 17.
Article in English | MEDLINE | ID: mdl-35978337

ABSTRACT

BACKGROUND: Native-like secondary structures and biological activity have been described for proteins in inclusion bodies (IBs). Tertiary structure analysis, however, is hampered due to the necessity of mild solubilization conditions. Denaturing reagents used for IBs solubilization generally lead to the loss of these structures and to consequent reaggregation due to intermolecular interactions among exposed hydrophobic domains after removal of the solubilization reagent. The use of mild, non-denaturing solubilization processes that maintain existing structures could allow tertiary structure analysis and increase the efficiency of refolding. RESULTS: In this study we use a variety of biophysical methods to analyze protein structure in human growth hormone IBs (hGH-IBs). hGH-IBs present native-like secondary and tertiary structures, as shown by far and near-UV CD analysis. hGH-IBs present similar λmax intrinsic Trp fluorescence to the native protein (334 nm), indicative of a native-like tertiary structure. Similar fluorescence behavior was also obtained for hGH solubilized from IBs and native hGH at pH 10.0 and 2.5 kbar and after decompression. hGH-IBs expressed in E. coli were extracted to high yield and purity (95%) and solubilized using non-denaturing conditions [2.4 kbar, 0.25 M arginine (pH 10), 10 mM DTT]. After decompression, the protein was incubated at pH 7.4 in the presence of the glutathione-oxidized glutathione (GSH-GSSG) pair which led to intramolecular disulfide bond formation and refolded hGH (81% yield). CONCLUSIONS: We have shown that hGH-IBs present native-like secondary and tertiary structures and that non-denaturing methods that aim to preserve them can lead to high yields of refolded protein. It is likely that the refolding process described can be extended to different proteins and may be particularly useful to reduce the pH required for alkaline solubilization.


Subject(s)
Human Growth Hormone , Inclusion Bodies , Humans , Escherichia coli/genetics , Escherichia coli/metabolism , Human Growth Hormone/metabolism , Inclusion Bodies/metabolism , Protein Refolding , Protein Structure, Secondary , Protein Structure, Tertiary , Recombinant Proteins/biosynthesis , Solubility
20.
Nature ; 539(7629): 448-451, 2016 11 17.
Article in English | MEDLINE | ID: mdl-27783598

ABSTRACT

The Hsp70 system is a central hub of chaperone activity in all domains of life. Hsp70 performs a plethora of tasks, including folding assistance, protection against aggregation, protein trafficking, and enzyme activity regulation, and interacts with non-folded chains, as well as near-native, misfolded, and aggregated proteins. Hsp70 is thought to achieve its many physiological roles by binding peptide segments that extend from these different protein conformers within a groove that can be covered by an ATP-driven helical lid. However, it has been difficult to test directly how Hsp70 interacts with protein substrates in different stages of folding and how it affects their structure. Moreover, recent indications of diverse lid conformations in Hsp70-substrate complexes raise the possibility of additional interaction mechanisms. Addressing these issues is technically challenging, given the conformational dynamics of both chaperone and client, the transient nature of their interaction, and the involvement of co-chaperones and the ATP hydrolysis cycle. Here, using optical tweezers, we show that the bacterial Hsp70 homologue (DnaK) binds and stabilizes not only extended peptide segments, but also partially folded and near-native protein structures. The Hsp70 lid and groove act synergistically when stabilizing folded structures: stabilization is abolished when the lid is truncated and less efficient when the groove is mutated. The diversity of binding modes has important consequences: Hsp70 can both stabilize and destabilize folded structures, in a nucleotide-regulated manner; like Hsp90 and GroEL, Hsp70 can affect the late stages of protein folding; and Hsp70 can suppress aggregation by protecting partially folded structures as well as unfolded protein chains. Overall, these findings in the DnaK system indicate an extension of the Hsp70 canonical model that potentially affects a wide range of physiological roles of the Hsp70 system.


Subject(s)
HSP70 Heat-Shock Proteins/chemistry , HSP70 Heat-Shock Proteins/metabolism , Protein Folding , Adenosine Triphosphate/metabolism , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Models, Biological , Optical Tweezers , Protein Aggregates , Protein Binding , Protein Conformation , Protein Denaturation , Protein Refolding , Protein Stability , Single Molecule Imaging , Substrate Specificity
SELECTION OF CITATIONS
SEARCH DETAIL