IYCr crystallization series\(\def\hfill{\hskip 5em}\def\hfil{\hskip 3em}\def\eqno#1{\hfil {#1}}\)

Journal logoSTRUCTURAL BIOLOGY
COMMUNICATIONS
ISSN: 2053-230X

Protein stability: a crystallographer's perspective

CROSSMARK_Color_square_no_text.svg

aStanford ChEM-H, Macromolecular Structure Knowledge Center, Stanford University, Shriram Center, 443 Via Ortega, Room 097, MC5082, Stanford, CA 94305-4125, USA, bLaboratory of Cell and Molecular Biology, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), National Institutes of Health (NIH), Building 8, Room 1A03, 8 Center Drive, Bethesda, MD 20814, USA, cDepartment of Forensic Crystallography, k.-k. Hofkristallamt, 91 Audrey Place, Vista, CA 92084, USA, and dDepartment of Genetic Epidemiology, Medical University of Innsbruck, Schöpfstrasse 41, A-6020 Innsbruck, Austria
*Correspondence e-mail: mdeller@stanford.edu

Edited by H. M. Einspahr, Lawrenceville, USA (Received 27 November 2015; accepted 21 December 2015; online 26 January 2016)

Protein stability is a topic of major interest for the biotechnology, pharmaceutical and food industries, in addition to being a daily consideration for academic researchers studying proteins. An understanding of protein stability is essential for optimizing the expression, purification, formulation, storage and structural studies of proteins. In this review, discussion will focus on factors affecting protein stability, on a somewhat practical level, particularly from the view of a protein crystallographer. The differences between protein conformational stability and protein compositional stability will be discussed, along with a brief introduction to key methods useful for analyzing protein stability. Finally, tactics for addressing protein-stability issues during protein expression, purification and crystallization will be discussed.

1. Introduction

The main purpose of this review is to introduce the reader to the concepts of protein stability from the viewpoint of a structural biologist, a structural biologist being defined as a scientist who determines the detailed molecular structure of a protein using methods such as crystallography, NMR spectroscopy or cryo-EM. Particular emphasis will be given to crystallographic techniques, as protein stability, or the lack thereof, represents a substantial challenge in the crystallization of many proteins. Protein stability is a wide-ranging topic including aspects of physical chemistry, thermodynamics, entropy, computational chemistry, protein folding and dynamics. For the purposes of this review, many of the computational and theoretical aspects are skipped over and the reader is referred to other excellent reviews on this topic (Compiani & Capriotti, 2013[Compiani, M. & Capriotti, E. (2013). Computational and theoretical methods for protein folding. Biochemistry, 52, 8601-8624.]; Lazaridis & Karplus, 2002[Lazaridis, T. & Karplus, M. (2002). Thermodynamics of protein folding: a microscopic view. Biophys. Chem. 100, 367-395.]).

Stability is the potential of a pattern to survive over time, and therefore is integral to our understanding of biological systems and their evolution (Schrödinger, 1945[Schrödinger, E. (1945). What is Life? The Physical Aspect of the Living Cell. New York: McMillan.]). Clearly, the exact meaning of a `pattern' for a protein molecule is somewhat vague, but we know that processes such as protein unfolding, denaturation, degradation, conformational change, enzymatic modification and proteolytic cleavage may transform this `pattern'. These transformations are generally considered, or analyzed, with respect to the integrity of the primary and conformational structure of the fully folded protein. Additionally, protein stability means different things to different people. For example, a pharmacologist, biotechnologist or food scientist may primarily consider the half-life of a protein's activity as a measure of its stability. However, a protein chemist or a structural biologist may concern themselves with changes in the primary, secondary, tertiary or quaternary structure of a protein as a measure of its stability. Again, for the purposes of this review, we will focus on the structural aspects of protein stability and will refer the reader to other excellent reviews on protein stability from a pharmacological and biotechnological perspective (Hall, 2014[Hall, M. P. (2014). Biotransformation and in vivo stability of protein biotherapeutics: impact on candidate selection and pharmacokinetic profiling. Drug Metab. Dispos. 42, 1873-1880.]).

We will first discuss protein stability as a fundamental prerequisite for crystallization (§[link]2) and then some important aspects of stability on a higher, structural level (see §[link]3). At this stage it is important to discuss the differences between thermo­dynamic protein stability and conformational protein disorder, especially given some of the unique parameters that structural biologists use to describe and analyze their structures. For example, NMR spectroscopists often report root-mean-squared deviation (r.m.s.d.) values between their ensemble structures, whereas crystallographers report B factors as a measure of the positional uncertainty in a given protein crystal structure model. Both of these parameters represent displacements and disorder within a structure and can be reflective of the level of conformational stability. We will then discuss some important factors to consider when expressing and purifying proteins for structural studies. Structural genomics efforts have alleviated many of the bottlenecks of a traditional structure-determination pipeline, but researchers are still all too aware of the difficulties of expressing and purifying challenging protein targets. Careful consideration of the primary structure, construct design, expression conditions and hosts cells can all be used to mitigate many of the protein-stability issues observed during expression and purification (see §[link]4). We will then discuss some common methods used to analyze protein stability, with a focus on methods routinely used to asses protein stability, including protein melting temperature analysis (Tm), NMR and cryo-EM (see §[link]5).

2. Stability is a fundamental prerequisite for crystallization

Biomolecular crystallization can be described as the self-organization of macromolecules into a translationally periodic arrangement with long-range order. In order to achieve this goal optimally, the moieties within each asymmetric unit of a crystal should be of the same kind and of the same shape. If a protein cannot form such stable entities per se, a fundamental and primary requirement for crystallization is not met, and no effort to find suitable thermodynamic and kinetic conditions will lead to crystals of such a protein construct (Fig. 1[link]a).

[Figure 1]
Figure 1
Factors influencing protein stability. (a) Protein compositional stability and conformational stability as key determining factors for successful crystallization. The stability properties of the protein determine whether the process of crystal formation is possible. Thermodynamics establish the necessary conditions for crystallization, and the kinetics and dynamics of the processes determine whether a possible scenario actually becomes reality. Only if all of the parameters are satisfied will crystal formation proceed. Figure adapted from Rupp (2015[Rupp, B. (2015). Origin and use of crystallization phase diagrams. Acta Cryst. F71, 247-260.]). (b) The marginal net stability of a folded protein is highlighted with respect to the contributing factors; the overwhelming lack of conformational stability is only marginally balanced by the contribution of van der Waals (VdW), hydrogen-bonding (H-bonds) and hydrophobic forces. Figure adapted from https://bit.ly/1L921Oi .

It is important to note that from a crystallization perspective, there are at least two major flavors of protein stability: compositional stability and conformational stability (Table 1[link]). The crystallographer must carefully assess both types of stability in order to enable crystallization of the target protein.

Table 1
Compositional stability versus conformational stability: some important questions to ask when embarking on the crystallization of a protein and some factors to investigate for problem proteins

Global types of protein stability Key questions to ask when trying to crystallize a protein Required answers for successful crystallization Factors to investigate if the answer is `No'
Compositional stability Is the chemical makeup of the protein well defined? Yes Check amino-acid sequence. Check for PTMs, especially proteolysis. Purify protein more. Carry out more rigorous bioanalytical methods such as mass spectrometry and light scattering.
Does the protein have a high level of chemical homogeneity? Yes
Is the protein chemically stable in the crystallization conditions? Yes Use customized or less harsh crystallization screens. Explore different temperatures for screening.
Is the protein stable over the course of the crystallization experiment? Yes
Conformational stability Are there minimal disordered regions in the protein? Yes Redesign expression constructs to engineer out disordered or dynamic regions. Identify stabilizing protein partners or ligands.
Does the protein have a minimal content of domains or regions that undergo dynamic variability over time? Yes

2.1. Compositional stability

During the processes of crystallization it is essential to maintain the same species within the crystallization experiment; there needs to be some form of compositional stability. On a simple level this means that the protein molecules must have the same chemical makeup. The chemical homogeneity of a sample can often be determined using mass spectrometry or an SDS–PAGE gel. Compositional homogeneity is typically compromised by post-translational modifications, such as glycosylation and proteolysis, which can affect the primary structure of the protein molecules and generate compositional variability (see §3.1[link]). Because protein crystallization takes time, the primary requirement for compositional stability must be maintained over a period of time, and preferably within a reasonable range of environmental conditions. It is important to note that there is no such thing as absolute stability. For example, a protein that is compositionally stable enough to produce a single band on an SDS–PAGE gel may still not be stable enough over the timeframe of a crystallization experiment.

2.2. Conformational stability

Assuming that the protein sample has a degree of compositional homogeneity, it will still likely not crystallize unless it possesses conformational stability. A large number of proteins fall into the category of conformationally disordered proteins displaying little or no conformational order (Longhi et al., 2010[Longhi, S., Lieutaud, P. & Canard, B. (2010). Conformational disorder. Methods Mol. Biol. 609, 307-325.]). A protein with substantial disordered regions, or separate domains exhibiting dynamic variability, will be less likely to self-organize into a crystal. This can be the case even if the sample has perfect compositional stability. The strict requirement for limited conformational variability is a unique problem that a crystallographer faces when trying to crystallize a protein sample. The problem is confounded by the fact that the conformation of flexible regions of a protein is a context-driven property. For example, conformations may be quite different in the cellular context, in an NMR tube or in a macromolecular crystal. While structural methods can be used to probe conformational homogeneity, it is important to realise that the results are only meaningful within the context and conditions of that particular method (see §5[link]). For example, analysis of conformational stability and dynamics is often limited using crystallographic methods as the crystal packing can hinder such movements. In these cases NMR solution methods can provide complementary information.

Structures determined using X-ray crystallography provide limited information regarding the dynamics of the protein structure. Nonetheless, some dynamics information is included in the atomic model in the form of the atomic displacement parameter (ADP) or B factor. The B factor is expressed in units of Å2 and is essentially a statistical measure describing the probability of finding an atom at that particular mean position in the structure (Willis & Pryor, 1975[Willis, B. T. M. & Pryor, A. W. (1975). Thermal Vibrations in Crystallography. Cambridge University Press.]). If the B factor of a particular atom is high then it suggests that the certainty of finding the atom at that position in the structure is low. Atoms in regions of high B factor can be displaced as a result of dynamic disorder of the polypeptide chain or as a result of short-range or long-range disorder within the crystal. Such flexible or dynamic regions can often be identified in a crystal structure and engineered out at the cloning stage to produce protein samples with better conformational stability (see §§[link]4.1 and [link]6.1). Not only do such modifications result in better protein stability during expression and purification, but they also increase the probability that the molecules will pack within the crystal lattice in a more orderly fashion. As a consequence, such efforts often result in better diffracting crystals and higher-resolution X-ray data.

Comparison of ensembles of structures, as typically generated by NMR spectroscopy, can be used to provide a measure analogous to the crystallographic B factor in the form of a root-mean-squared deviation (r.m.s.d.) between corresponding atoms of the ensemble members. This measure can be used to assess the flexibility, dynamics, disorder or stereochemical variability across a set of structural models. The r.m.s.d. value is complementary to the crystallographic B factor and because the structure is in solution it is not perturbed or influenced by crystal packing (Sikic & Carugo, 2009[Sikic, K. & Carugo, O. (2009). CARON - average RMSD of NMR structure ensembles. Bioinformation, 4, 132-133.]).

A large class of proteins, referred to as intrinsically dis­ordered proteins (IDPs), contain significant levels of conformational disorder and, in some cases, have no discernable three-dimensional structure at all. It is estimated that ∼40% of all human proteins contain at least one disordered segment and ∼25% are completely disordered (Uversky & Dunker, 2010[Uversky, V. N. & Dunker, A. K. (2010). Understanding protein non-folding. Biochim. Biophys. Acta, 1804, 1231-1264.]). These proteins have largely been avoided by the crystallographic community owing to the expected difficulties in crystallizing them. However, NMR techniques have been central to unraveling how these unstructured proteins function. Such studies have led to a paradigm shift in our understanding of protein structure and function (Wright & Dyson, 1999[Wright, P. E. & Dyson, H. J. (1999). Intrinsically unstructured proteins: re-assessing the protein structure-function paradigm. J. Mol. Biol. 293, 321-331.]). Traditional theory dictates that proteins function by adopting a rigid, preformed structure that binds to a target ligand or protein in a fashion analogous to a lock and key. However, NMR studies on IDP proteins such as CREB, p53 and 14-3-3 have revealed that these disordered regions allow plasticity and flexibility and often only form structure upon binding of the partner protein (Oldfield et al., 2008[Oldfield, C. J., Meng, J., Yang, J. Y., Yang, M. Q., Uversky, V. N. & Dunker, A. K. (2008). Flexible nets: disorder and induced fit in the associations of p53 and 14-3-3 with their partners. BMC Genomics, 9, S1.]; Sugase et al., 2007[Sugase, K., Dyson, H. J. & Wright, P. E. (2007). Mechanism of coupled folding and binding of an intrinsically disordered protein. Nature (London), 447, 1021-1025.]; Mujtaba et al., 2004[Mujtaba, S., He, Y., Zeng, L., Yan, S., Plotnikova, O., Sachchidanand, Sanchez, R., Zeleznik-Le, N. J., Ronai, Z. & Zhou, M.-M. (2004). Structural mechanism of the bromodomain of the coactivator CBP in p53 transcriptional activation. Mol. Cell, 13, 251-263.]). These so-called `hub proteins' are capable of interacting with many different protein partners in a context-sensitive manner, and this is only possible as a result of the plasticity and initial lack of conformational stability. High-resolution crystal structures of complexes of these vital `hub proteins' will be essential for understanding their role in human diseases such as Parkinson's disease and Alzheimer's disease (Wang et al., 2011[Wang, J., Cao, Z., Zhao, L. & Li, S. (2011). Novel strategies for drug discovery based on intrinsically disordered proteins (IDPs). Int. J. Mol. Sci. 12, 3205-3219.]). Although the poor conformational stability of these proteins poses challenges for the protein crystallographer, in a cellular context IDPs offer many advantages over more traditional single-function folded proteins, including the ability to bind to many different protein partners (Liu & Huang, 2014[Liu, Z. & Huang, Y. (2014). Advantages of proteins being disordered. Protein Sci. 23, 539-550.]).

In addition to IDPs, many proteins contain aggregation-prone regions (APRs) that typically contain a run of 5–15 amino acids with a propensity for forming extended β-sheet structures. For example, APR segments are observed in β2-microglobulin and are responsible for aggregation into amyloid fibers in diseases such as amyloidosis (De Baets et al., 2014[De Baets, G., Schymkowitz, J. & Rousseau, F. (2014). Predicting aggregation-prone sequences in proteins. Essays Biochem. 56, 41-52.]). Another group of proteins referred to as intrinsically insoluble proteins (IIPs) are completely insoluble and cannot be refolded in traditional buffer solutions (Goyal et al., 2015[Goyal, S., Qin, H., Lim, L. & Song, J. (2015). Insoluble protein characterization by circular dichroism (CD) spectroscopy and nuclear magnetic resonance (NMR). Methods Mol. Biol. 1258, 371-385.]; Liu & Song, 2009[Liu, J. & Song, J. (2009). Insights into protein aggregation by NMR characterization of insoluble SH3 mutants solubilized in salt-free water. PLoS One, 4, e7805.]). For example, naturally occurring mutants of SH3, such as V22-SH3, are insoluble in the presence of ions, but they can be resurrected and solubilized in pure water, allowing further study of the unstructured proteins in solution using NMR spectroscopy (Liu & Song, 2009[Liu, J. & Song, J. (2009). Insights into protein aggregation by NMR characterization of insoluble SH3 mutants solubilized in salt-free water. PLoS One, 4, e7805.]).

3. Stability of the protein on a structural level

One simple way of conceptualizing protein stability from a structural perspective is to consider stability at each level of protein structure: primary structure, secondary structure, tertiary structure and quaternary structure (Table 2[link]). Protein stability with respect to each of the structural levels will now be discussed in turn. Wherever possible, we will emphasize aspects of particular importance to the structural biologist, with a particular focus on protein crystallization.

Table 2
Common measures of protein stability

Definitions of protein stability at each structural level are shown along with common methods used to analyze the degree of stability. Asterisks denote the relative merits of the three main structure-determination techniques, with five asterisks denoting the optimal method. For example, NMR solution methods are often more favorable for studying dynamic processes and quaternary states as they are not influenced by crystal packing.

        Relative merits of structural methods
Structural level Definition of stability Example biochemical processes or features Common methods Crystallography NMR EM
Primary Change of amino-acid sequence or modification of amino acids PTM Proteolysis Protein splicing Half-life analysis SDS–PAGE Mass spectrometry Eastern and Western blotting ***** **** *
Secondary Change of α-helix, β-sheet and loop content Secondary-structure formation Racemization Aromatic side-chain interactions Ligands Circular dichroism (CD) Synchrotron-radiation CD UV-CD FT-IR 2D-IR Deuterium-exchange mass spectromety (DXMS) ***** ***** *
Tertiary Change of overall fold or protein conformation Hydrogen bonding Hydrophobic interactions Conformational change Disulfide bonding Topology ITC DSC Thermofluor **** **** **
Quaternary Change in oligomeric state Protein–protein interactions Oligomerization Size-exclusion chromatography Native gel electrophoresis * ***** *****

3.1. Primary structure

The primary structure of the protein, or the sequence of the amino acids in the polypeptide chain, can be modified in several ways by post-translational modifications (PTMs). PTMs result in alteration of the structure and function of a protein and for this reason are central to any discussion of protein stability. As discussed above (see §[link]2), PTMs can affect both compositional stability, as the modifications may be non-uniform or incomplete, and also conformational stability, as the modifications may affect protein disorder and dynamics. This is illustrated by glycoproteins, which are often not uniformly glycosylated at all possible glycosylation sites, therefore leading to compositional heterogeneity. Furthermore, complex hydrocarbon chains tend to have a greater degree of conformational freedom. This conformational freedom results in an increase in disorder on the protein surface, while at the same time shielding polar or charged residues on the protein surface required for intermolecular crystal contact formation (see §[link]6.6). Although the heterogeneity of glycosylation tends to impair crystallization, its variability can have important functional implications for a protein.

PTMs are the result of many different changes to the primary structure of a protein, including proteolytic processing, protein splicing and the addition of other functional groups to the amino acids. PTMs are often used for targeting of the protein to a specific region of the cell or modification of the activity or specificity of an enzyme. For example, functional groups such as myristate, palmitate, isoprenoid and glycosylphosphatidylinositol (GPI) are often attached to the protein and used for targeting of the protein to the membrane (Chatterjee & Mayor, 2001[Chatterjee, S. & Mayor, S. (2001). The GPI-anchor and protein sorting. Cell. Mol. Life Sci. 58, 1969-1987.]). Other functional groups such as carboxylate (Walker et al., 2001[Walker, C. S., Shetty, R. P., Clark, K., Kazuko, S. G., Letsou, A., Olivera, B. M. & Bandyopadhyay, P. K. (2001). On a potential global role for vitamin K-dependent γ-carboxylation in animal systems: evidence for a γ-glutamyl carboxylase in Drosophila. J. Biol. Chem. 276, 7769-7774.]), ethanolamine phospho­glycerol (Whiteheart et al., 1989[Whiteheart, S. W., Shenbagamurthi, P., Chen, L., Cotter, R. J. & Hart, G. W. (1989). Murine elongation factor 1 alpha (EF-1 alpha) is posttranslationally modified by novel amide-linked ethanolamine-phosphoglycerol moieties. Addition of ethanolamine-phosphoglycerol to specific glutamic acid residues on EF-1 alpha. J. Biol. Chem. 264, 14334-14341.]) and hypusine (Park et al., 2010[Park, M. H., Nishimura, K., Zanelli, C. F. & Valentini, S. R. (2010). Functional significance of eIF5A and its hypusine modification in eukaryotes. Amino Acids, 38, 491-500.]) can be added to proteins to regulate their activity. Additionally, larger peptides and proteins can also be covalently added to proteins, including ubiquitin (Komander & Rape, 2012[Komander, D. & Rape, M. (2012). The ubiquitin code. Annu. Rev. Biochem. 81, 203-229.]), SUMO (Hay, 2005[Hay, R. T. (2005). SUMO. Mol. Cell, 18, 1-12.]), ISG15 (Malakhova et al., 2003[Malakhova, O. A., Yan, M., Malakhov, M. P., Yuan, Y., Ritchie, K. J., Kim, K. I., Peterson, L. F., Shuai, K. & Zhang, D.-E. (2003). Protein ISGylation modulates the JAK-Stat signaling pathway. Genes Dev. 17, 455-460.]), PUP (Striebel et al., 2014[Striebel, F., Imkamp, F., Özcelik, D. & Weber-Ban, E. (2014). Pupylation as a signal for proteasomal degradation in bacteria. Biochim. Biophys. Acta, 1843, 103-113.]) and NEDD (Rabut & Peter, 2008[Rabut, G. & Peter, M. (2008). Function and regulation of protein neddylation. EMBO Rep. 9, 969-976.]). Of the 821 182 proteins that were experimentally analyzed by Khoury and coworkers, the top ten observed PTMs are phosphorylation (58383), acetylation (6751), N-linked glycosylation (5526), amidation (2844), hydroxylation (1619), methylation (1523), O-linked glycosylation (1133), ubiquitylation (878), pyrrolidone carboxylic acid (826) and sulfation (504) (https://bit.ly/1jdfXR8 ; Khoury et al., 2011[Khoury, G. A., Baliban, R. C. & Floudas, C. A. (2011). Proteome-wide post-translational modification statistics: frequency analysis and curation of the Swiss-Prot database. Sci. Rep. 1, 90.]). Key methods used to analyze and identify changes at the primary-structure level include mass spectrometry and Eastern and Western blots (Liu et al., 2014[Liu, Z.-Q., Mahmood, T. & Yang, P.-C. (2014). Western blot: technique, theory and trouble shooting. N. Am. J. Med. Sci. 6, 160.]; Towbin et al., 1979[Towbin, H., Staehelin, T. & Gordon, J. (1979). Electrophoretic transfer of proteins from polyacrylamide gels to nitrocellulose sheets: procedure and some applications. Proc. Natl Acad. Sci. USA, 76, 4350-4354.]; Table 2[link]).

It is important to note that many PTMs play a role in stabilizing proteins, particular with respect to the half-life and turnover of the protein within the cell. For example, PTMs such as ubiquitination target proteins to the proteasome for degradation and recycling, therefore directly affect the half-life of the protein and its stability within the cell (Komander & Rape, 2012[Komander, D. & Rape, M. (2012). The ubiquitin code. Annu. Rev. Biochem. 81, 203-229.]). A myriad of other PTMs exist, including acyl­ation, alkylation, arginylation, butyrylation, malonylation, ADP-ribosylation, iodination, oxidation, succinylation, S-nitrosylation, S-glutathionylation and glycosylation. Currently, just under 500 PTMs have been identified in the SWISS-PROT and TrEMBL databases (for a full list, see https://bit.ly/1P6Rbj3 ). All of these modifications play a role in the structure and the function of the target protein. However, some PTMs, such as proteolytic cleavage and protein splicing, significantly influence protein structure at the primary level and can lead to drastic changes in compositional stability.

Protein splicing occurs in proteins called inteins (or protein introns), which are a large class of self-cleaving proteins found in all domains of life (Paulus, 2000[Paulus, H. (2000). Protein splicing and related forms of protein autoprocessing. Annu. Rev. Biochem. 69, 447-496.]; Novikova et al., 2014[Novikova, O., Topilina, N. & Belfort, M. (2014). Enigmatic distribution, evolution, and function of inteins. J. Biol. Chem. 289, 14490-14497.]). One of the first examples identified was the VMA1 protein, a yeast vacuolar membrane H+-ATPase, which was shown to undergo protein splicing (Hirata et al., 1990[Hirata, R., Ohsumk, Y., Nakano, A., Kawasaki, H., Suzuki, K. & Anraku, Y. (1990). Molecular structure of a gene, VMA1, encoding the catalytic subunit of H+-translocating adenosine triphosphatase from vacuolar membranes of Saccharomyces cerevisiae. J. Biol. Chem. 265, 6726-6733.]). Protein splicing is a naturally occurring process analogous to the splicing of introns from RNA. A precursor polypeptide is processed into a mature and functional protein. The intein is autocatalytically excised from the precursor protein and the flanking exteins are ligated together, producing two new polypetides (Mills et al., 2014[Mills, K. V., Johnson, M. A. & Perler, F. B. (2014). Protein splicing: how inteins escape from precursor proteins. J. Biol. Chem. 289, 14498-14505.]). Inteins are of great importance for the stability of proteins, but they are also of interest from a protein-engineering perspective (Aranko et al., 2014[Aranko, A. S., Wlodawer, A. & Iwai, H. (2014). Nature's recipe for splitting inteins. Protein Eng. Des. Sel. 27, 263-271.]). For example, inteins can be used for the preparation of isotope-labeled proteins for NMR spectroscopy, for site-specific fluorescent labeling and as self-cleaving affinity purification tags such as cSAT and intein-CDB (commercially available as the IMPACT system from NEB; Chong et al., 1997[Chong, S., Mersha, F. B., Comb, D. G., Scott, M. E., Landry, D., Vence, L. M., Perler, F. B., Benner, J., Kucera, R. B., Hirvonen, C. A., Pelletier, J. J., Paulus, H. & Xu, M.-Q. (1997). Single-column purification of free recombinant proteins using a self-cleavable affinity tag derived from a protein splicing element. Gene, 192, 271-281.]; Volkmann & Iwaï, 2010[Volkmann, G. & Iwaï, H. (2010). Protein trans-splicing and its use in structural biology: opportunities and limitations. Mol. Biosyst. 6, 2110-2121.]; Lin et al., 2015[Lin, Z., Zhao, Q., Zhou, B., Xing, L. & Xu, W. (2015). Cleavable self-aggregating tags (cSAT) for protein expression and purification. Methods Mol. Biol. 1258, 65-78.]; see §[link]4.1).

3.2. Secondary structure

Protein secondary structure is the localized three-dimensional structure of the polypeptide chain. Secondary structure can be described in terms of the pattern of hydrogen bonding between amide H atoms and carbonyl O atoms of the backbone (Pauling et al., 1951[Pauling, L., Corey, R. B. & Branson, H. R. (1951). The structure of proteins: two hydrogen-bonded helical configurations of the polypeptide chain. Proc. Natl Acad. Sci. USA, 37, 205-211.]) or by the stereochemistry adopted by the polypeptide backbone (Ramachandran et al., 1963[Ramachandran, G. N., Ramakrishnan, C. & Sasisekharan, V. (1963). Stereochemistry of polypeptide chain configurations. J. Mol. Biol. 7, 95-99.]). On a somewhat simplified level, the primary driving forces behind the formation of secondary structure, and in turn tertiary structure, are hydrogen bonding and hydrophobic interaction (Pace, Scholtz et al., 2014[Pace, C. N., Scholtz, J. M. & Grimsley, G. R. (2014). Forces stabilizing proteins. FEBS Lett. 588, 2177-2184.]; see §[link]3.3).

The α-helix is the predominant type of secondary structure, accounting for approximately one-third of all secondary-structure elements (Stickle et al., 1992[Stickle, D. F., Presta, L. G., Dill, K. A. & Rose, G. D. (1992). Hydrogen bonding in globular proteins. J. Mol. Biol. 226, 1143-1159.]). Analysis of the first crystal structures suggested that certain residues including alanine, leucine and glutamate are found frequently in α-helices. In contrast, other residues such as proline, glycine and aspartic acid are found less frequently (Davies, 1964[Davies, D. R. (1964). A correlation between amino acid composition and protein structure. J. Mol. Biol. 9, 605-609.]; Prothero, 1966[Prothero, J. W. (1966). Correlation between the distribution of amino acids and alpha helices. Biophys. J. 6, 367-370.]; Guzzo, 1965[Guzzo, A. V. (1965). The influence of amino acid sequence on protein structure. Biophys. J. 5, 809-822.]). This information has been used to develop many algorithms for the prediction of protein secondary structure, including the popular Chou and Fasman method (Chou & Fasman, 1974[Chou, P. Y. & Fasman, G. D. (1974). Conformational parameters for amino acids in helical, β-sheet, and random coil regions calculated from proteins. Biochemistry, 13, 211-222.]). Secondary-structure propensity data have been expanded using mutagenesis data, and tables of α-helical (Pace & Scholtz, 1998[Pace, C. N. & Scholtz, J. M. (1998). Biophys. J. 75, 422-427.]) and β-sheet (Smith et al., 1994[Smith, C. K., Withka, J. M. & Regan, L. (1994). A thermodynamic scale for the β-sheet forming tendencies of the amino acids. Biochemistry, 33, 5510-5517.]) propensity have been compiled.

One overwhelming consensus of the amino-acid propensity rules is the destabilizing effect that proline has on the α-helix (ΔΔG of 3.16 kcal mol−1 cf. alanine at 0 kcal mol−1; Pace & Scholtz, 1998[Pace, C. N. & Scholtz, J. M. (1998). Biophys. J. 75, 422-427.]; see §[link]3.3). This destabilization is a result of the missing backbone amide H atom, which prevents proline from participating in stabilizing hydrogen bonding. Additionally, the bulky cyclic side chain of proline results in a ∼30% kink in the α-helix backbone as a result of steric hindrance (Richardson, 1981[Richardson, J. S. (1981). The anatomy and taxonomy of protein structure. Adv. Protein Chem. 34, 167-339.]; Yun et al., 1991[Yun, R. H., Anderson, A. & Hermans, J. (1991). Proline in α-helix: stability and conformation studied by dynamics simulation. Proteins, 10, 219-228.]). Glycine has the next lowest propensity for forming α-helices as a result of enhanced conformational flexibility upon folding to form an α-helix (Hermans et al., 1992[Hermans, J., Anderson, A. G. & Yun, R. H. (1992). Differential helix propensity of small apolar side chains studied by molecular dynamics simulations. Biochemistry, 31, 5646-5653.]). It is important to note that many of these secondary-structure propensities are highly context-dependent. For example, proline occurs widely in the transmembrane helices of integral membrane proteins and has been shown to have a stabilizing effect on α-helices in such environments (Li et al., 1996[Li, S.-C., Goto, N. K., Williams, K. A. & Deber, C. M. (1996). α-Helical, but not β-sheet, propensity of proline is determined by peptide environment. Proc. Natl Acad. Sci. USA, 93, 6676-6681.]).

Clearly, such findings are in support of the hypothesis that the stability of the folded protein is largely dictated by the amino-acid composition and, as such, the primary structure results in a unique, kinetic minimum of free energy as first suggested by Anfinsen (1973[Anfinsen, C. B. (1973). Principles that govern the folding of protein chains. Science, 181, 223-230.]). These simple principles have been expanded into complex algorithms that can be used to design both stable α-helices and β-sheets (Jiménez, 2014[Jiménez, M. A. (2014). Design of monomeric water-soluble β-hairpin and β-sheet peptides. Methods Mol. Biol. 1216, 15-52.]; Yakimov et al., 2014[Yakimov, A., Rychkov, G. & Petukhov, M. (2014). De novo design of stable α-helices. Methods Mol. Biol. 1216, 1-14.]). Furthermore, comparative modeling can be used to design proteins with a greater degree of thermal stability, and similar models can be used to predict the crystallizability of a protein (Olson et al., 2015[Olson, M. A., Zabetakis, D., Legler, P. M., Turner, K. B., Anderson, G. P. & Goldman, E. R. (2015). Fusion to a highly stable consensus albumin binding domain allows for tunable pharmacokinetics. Protein Eng. Des. Sel. 28, 395-402.]; Smialowski & Frishman, 2010[Smialowski, P. & Frishman, D. (2010). Protein crystallizability. Methods Mol. Biol. 609, 385-400.]; see §[link]6).

3.3. Tertiary structure

The tertiary structure of a protein is the overall shape, or fold, adopted by the polypeptide chain. Many factors affect the process of protein folding, including conformational and compositional stability, cellular environment including temperature and pH, primary and secondary structure, solvation, hydrogen bonding, salt bridges, hydrophobic effects, van der Waals (vdW) forces, ligand binding, cofactor binding, ion binding, chaperones and PTMs, to name just a few.

The conformational stability of the polypeptide chain results in a significant entropic penalty (−TΔS >> 0), and under normal cellular conditions a folded protein is only marginally stable (∼10 kcal mol−1 for a 10 kDa protein; Fig. 1[link]b). In order to overcome this entropic penalty, all of the other factors influencing protein folding must outweigh the loss of conformational entropy (Dill, 1990[Dill, K. A. (1990). Dominant forces in protein folding. Biochemistry, 29, 7133-7155.]). A series of studies by Pace and coworkers have recently quantified some of these influences (Pace et al., 2011[Pace, C. N., Fu, H., Fryar, K. L., Landua, J., Trevino, S. R., Shirley, B. A., Hendricks, M. M., Iimura, S., Gajiwala, K., Scholtz, J. M. & Grimsley, G. R. (2011). Contribution of hydrophobic interactions to protein stability. J. Mol. Biol. 408, 514-528.]; Pace, Fu et al., 2014[Pace, C. N., Fu, H. et al. (2014). Contribution of hydrogen bonds to protein stability. Protein Sci. 23, 652-661.]; Pace, Scholtz et al., 2014[Pace, C. N., Scholtz, J. M. & Grimsley, G. R. (2014). Forces stabilizing proteins. FEBS Lett. 588, 2177-2184.]). These studies suggest that the hydrophobic effect contributes ∼60% to the stability of the protein and hydrogen bonding contributes ∼40% (Pace et al., 2011[Pace, C. N., Fu, H., Fryar, K. L., Landua, J., Trevino, S. R., Shirley, B. A., Hendricks, M. M., Iimura, S., Gajiwala, K., Scholtz, J. M. & Grimsley, G. R. (2011). Contribution of hydrophobic interactions to protein stability. J. Mol. Biol. 408, 514-528.]). Specifically, the burial of a single methyl group contributes ∼1.1 kcal mol−1 to net protein stability and loss of its conformational entropy contributes ∼2.4 kcal mol−1 to net protein instability (Pace et al., 2011[Pace, C. N., Fu, H., Fryar, K. L., Landua, J., Trevino, S. R., Shirley, B. A., Hendricks, M. M., Iimura, S., Gajiwala, K., Scholtz, J. M. & Grimsley, G. R. (2011). Contribution of hydrophobic interactions to protein stability. J. Mol. Biol. 408, 514-528.]). The net contribution of hydrogen bonding to overall protein stability is also ∼1.1 kcal mol−1 and is largely independent of the size of the protein (Stickle et al., 1992[Stickle, D. F., Presta, L. G., Dill, K. A. & Rose, G. D. (1992). Hydrogen bonding in globular proteins. J. Mol. Biol. 226, 1143-1159.]; Pace, Scholtz et al., 2014[Pace, C. N., Scholtz, J. M. & Grimsley, G. R. (2014). Forces stabilizing proteins. FEBS Lett. 588, 2177-2184.]). However, in contrast, hydrophobic interactions typically contribute less to the stability of small proteins (Pace et al., 2011[Pace, C. N., Fu, H., Fryar, K. L., Landua, J., Trevino, S. R., Shirley, B. A., Hendricks, M. M., Iimura, S., Gajiwala, K., Scholtz, J. M. & Grimsley, G. R. (2011). Contribution of hydrophobic interactions to protein stability. J. Mol. Biol. 408, 514-528.]; Pace, Fu et al., 2014[Pace, C. N., Fu, H. et al. (2014). Contribution of hydrogen bonds to protein stability. Protein Sci. 23, 652-661.]).

The stability of the protein fold is of particular interest for the design of thermally stable proteins for industrial uses such as biofuel production and as proteases for laundry detergents. Thermophilic organisms such as Thermotoga maritima, which thrives in hot deep-sea vents in the Sargasso Sea, require proteins that maintain fold and structure under such extreme conditions. The study of these thermophilic proteins suggests that the protein structures are similar to their mesophilic counterparts and thermal stability is inferred by subtle changes in the amino-acid composition. On comparing thermo­philic proteins with their mesophilic counterparts, certain patterns are observed including an increase in the number of salt bridges, an increase in hydrophobicity and an increase in the number of aromatic residues (Dekker et al., 1991[Dekker, K., Yamagata, H., Sakaguchi, K. & Udaka, S. (1991). Xylose (glucose) isomerase gene from the thermophile Thermus thermophilus: cloning, sequencing, and comparison with other thermostable xylose isomerases. J. Bacteriol. 173, 3078-3083.]; Tanner et al., 1996[Tanner, J. J., Hecht, R. M. & Krause, K. L. (1996). Determinants of enzyme thermostability observed in the molecular structure of Thermus aquaticus D-glyceraldehyde-3-phosphate dehydrogenase at 2.5 Å resolution. Biochemistry, 35, 2597-2609.]; Zhou et al., 2008[Zhou, X.-X., Wang, Y.-B., Pan, Y.-J. & Li, W.-F. (2008). Differences in amino acids composition and coupling patterns between mesophilic and thermophilic proteins. Amino Acids, 34, 25-33.]; Fields et al., 2015[Fields, P. A., Dong, Y., Meng, X. & Somero, G. N. (2015). Adaptations of protein structure and function to temperature: there is more than one way to `skin a cat'. J. Exp. Biol. 218, 1801-1811.]; Somero, 2004[Somero, G. N. (2004). Adaptation of enzymes to temperature: searching for basic `strategies'. Comp. Biochem. Physiol. B Biochem. Mol. Biol. 139, 321-333.]).

In contrast to the IDPs discussed above (see §[link]2.2), the stability of the protein tertiary structure is often considered to be essential for the maintenance of protein function. However, many proteins undergo an overall change of protein fold as part of their mechanism of action. For example, serine protease inhibitors (serpins) undergo a transformation from a long-term stable native form (S, stressed) into a more stable folded form (R, relaxed) upon interaction with the proteinase (Whisstock & Bottomley, 2006[Whisstock, J. C. & Bottomley, S. P. (2006). Molecular gymnastics: serpin structure, folding and misfolding. Curr. Opin. Struct. Biol. 16, 761-768.]; Whisstock et al., 2000[Whisstock, J. C., Skinner, R., Carrell, R. W. & Lesk, A. M. (2000). Conformational changes in serpins. I. The native and cleaved conformations of α1-antitrypsin. J. Mol. Biol. 296, 685-699.]). These structural rearrangements include the insertion of a loop into the center of a core β-sheet or the insertion of a β-strand to form a domain-swapped dimer that can initiate polymerization (Mottonen et al., 1992[Mottonen, J., Strand, A., Symersky, J., Sweet, R. M., Danley, D. E., Geoghegan, K. F., Gerard, R. D. & Goldsmith, E. J. (1992). Structural basis of latency in plasminogen activator inhibitor-1. Nature (London), 355, 270-273.]; Yamasaki et al., 2008[Yamasaki, M., Li, W., Johnson, D. J. & Huntington, J. A. (2008). Crystal structure of a stable dimer reveals the molecular basis of serpin polymerization. Nature (London), 455, 1255-1258.]; see Fig. 2[link]c). Large conformational changes such as this are commonplace and are observed in many proteins including influenza virus hemagglutinin, lymphotactin, Mad2 spindle checkpoint protein and chloride intracellular channel 1 (CLIC1; Bryan & Orban, 2010[Bryan, P. N. & Orban, J. (2010). Proteins that switch folds. Curr. Opin. Struct. Biol. 20, 482-488.]). Therefore, it is important that any discussion on stability carefully considers the mechanism of the protein under study, as some protein folds are designed to be inherently unstable.

[Figure 2]
Figure 2
Matrix of examples of protein stability and disorder. (a) Examples of proteins with high conformational stability include the protein–protein destabilizing compound cyclosporin in complex with calcineurin and cyclophilin (Huai et al., 2002[Huai, Q., Kim, H.-Y., Liu, Y., Zhao, Y., Mondragon, A., Liu, J. O. & Ke, H. (2002). Crystal structure of calcineurin-cyclophilin-cyclo­sporin shows common but distinct recognition of immunophilin-drug complexes. Proc. Natl Acad. Sci. USA, 99, 12037-12042.]) and (b) the protein–protein stabilizing drug Tafamidis in combination with transthyretin (Bulawa et al., 2012[Bulawa, C. E., Connelly, S., Devit, M., Wang, L., Weigel, C., Fleming, J. A., Packman, J., Powers, E. T., Wiseman, R. L., Foss, T. R., Wilson, I. A., Kelly, J. W. & Labaudinière, R. (2012). Tafamidis, a potent and selective transthyretin kinetic stabilizer that inhibits the amyloid cascade. Proc. Natl Acad. Sci. 109, 9629-9634.]). (c) Examples of proteins with low conformational stability include the serpins, which undergo large changes in fold and oligomerization state (Yamasaki et al., 2008[Yamasaki, M., Li, W., Johnson, D. J. & Huntington, J. A. (2008). Crystal structure of a stable dimer reveals the molecular basis of serpin polymerization. Nature (London), 455, 1255-1258.]), and (d) intrinsically disordered proteins (IDPs) such as the tumor suppressor protein p53 (Mujtaba et al., 2004[Mujtaba, S., He, Y., Zeng, L., Yan, S., Plotnikova, O., Sachchidanand, Sanchez, R., Zeleznik-Le, N. J., Ronai, Z. & Zhou, M.-M. (2004). Structural mechanism of the bromodomain of the coactivator CBP in p53 transcriptional activation. Mol. Cell, 13, 251-263.]).

3.4. Quaternary structure

Quaternary structure is the arrangement of the folded protein subunits into a multi-subunit complex. The stability of such complexes is of importance for the regulation of allostery and cooperativity, which often results from the conformational changes within individual polypeptide chains. One of the classic models used to describe allosteric transitions in proteins is the Monod–Wyman–Changeux (MWC) model (Monod et al., 1965[Monod, J., Wyman, J. & Changeux, J.-P. (1965). On the nature of allosteric transitions: a plausible model. J. Mol. Biol. 12, 88-118.]). In this model proteins may exist in one of two states: tense (T) and relaxed (R). One of the key features of this model is that ligands may bind to either the T or R state with equal affinity, but if the R state is preferred then the affinity will be increased. However, if binding to the T state is favored then the affinity is decreased and the substance is described as an allosteric modulator. One of the best-studied examples is hemoglobin, with the R state representing deoxyhemoglobin and the T state representing oxyhemo­globin (Brunori, 2014[Brunori, M. (2014). Variations on the theme: allosteric control in hemoglobin. FEBS J. 281, 633-643.]; Ronda et al., 2013[Ronda, L., Bruno, S. & Bettati, S. (2013). Tertiary and quaternary effects in the allosteric regulation of animal hemoglobins. Biochim. Biophys. Acta, 1834, 1860-1872.]). As discussed above for IDPs and metastable proteins (see §§[link]2.2 and [link]3.3), it is not sufficient to consider protein stability in isolation from function. Many proteins undergo large conformational changes involving both secondary and tertiary structure, and each state may have a different conformational or compositional stability (Fig. 2[link]).

Modulation of the quaternary structure, and more specifically the protein–protein interactions responsible for quaternary structure, has long been a goal for the pharmaceutical industry. Such efforts present considerable challenges as a result of the large surface areas involved. Small molecules that destabilize protein–protein interactions have been demonstrated, but stabilizing examples are somewhat scarce (Giordanetto et al., 2014[Giordanetto, F., Schäfer, A. & Ottmann, C. (2014). Stabilization of protein-protein interactions by small molecules. Drug Discov. Today, 19, 1812-1821.]; Wells & McClendon, 2007[Wells, J. A. & McClendon, C. L. (2007). Reaching for high-hanging fruit in drug discovery at protein-protein interfaces. Nature (London), 450, 1001-1009.]). Destabilizing examples include the Abbott drug Navitoclax, which destabilizes the interaction between the anti-apoptotic protein Bcl-2 and Bad/Bid/Bak (Oltersdorf et al., 2005[Oltersdorf, T. et al. (2005). An inhibitor of Bcl-2 family proteins induces regression of solid tumours. Nature (London), 435, 677-681.]), and the Roche drug Nutlin-3, which inhibits the interaction between the tumor suppressor p53 and MDM2 (Secchiero et al., 2011[Secchiero, P., Bosco, R., Celeghini, C. & Zauli, G. (2011). Recent advances in the therapeutic perspectives of nutlin-3. Curr. Pharm. Des. 17, 569-577.]). Examples of compounds that stabilize protein–protein interactions include natural products such as cyclosporin A, which stabilizes the interaction between calcineurin and cyclophin (Huai et al., 2002[Huai, Q., Kim, H.-Y., Liu, Y., Zhao, Y., Mondragon, A., Liu, J. O. & Ke, H. (2002). Crystal structure of calcineurin-cyclophilin-cyclo­sporin shows common but distinct recognition of immunophilin-drug complexes. Proc. Natl Acad. Sci. USA, 99, 12037-12042.]), and the drug Tafamidis, which binds to a pocket at the interface of the transthyretin dimer (Bulawa et al., 2012[Bulawa, C. E., Connelly, S., Devit, M., Wang, L., Weigel, C., Fleming, J. A., Packman, J., Powers, E. T., Wiseman, R. L., Foss, T. R., Wilson, I. A., Kelly, J. W. & Labaudinière, R. (2012). Tafamidis, a potent and selective transthyretin kinetic stabilizer that inhibits the amyloid cascade. Proc. Natl Acad. Sci. 109, 9629-9634.]) (see Figs. 2[link]a and 2[link]b, respectively). In the case of Tafamidis, stabilization of the dimerized form of transthyretin prevents the aggregation and misfolding which has been shown to be the mechanism leading to transthyretin amyloid­osis diseases such as peripheral neuropathy.

4. Protein stability during protein expression and purification

The stability of the protein during the course of expression and purification is often an issue. In order to obtain sufficient quantities of protein for crystallization screening, formulation, vaccine development or therapeutic use, it is essential that intact, stable and folded protein is produced. Many proteins are unstable, unfolded or proteolytically cleaved under the conditions used for protein expression; again it is important to emphasize that these factors can lead to poor stability on both the conformational and compositional levels (see §[link]2 and Table 1[link]). Factors giving rise to poor protein stability during expression and purification may include the primary structure of the protein, the construction of the recombinant expression plasmid, the temperature and the expression medium used and the toxicity of the protein to the host organism (Table 3[link]). Therefore, there are many factors to test during expression and purification, and combinatorial design approaches are often used, in combination with high throughput methods to find the appropriate combination of conditions (Papaneophytou & Kontopidis, 2014[Papaneophytou, C. P. & Kontopidis, G. (2014). Statistical approaches to maximize recombinant protein expression in Escherichia coli: a general review. Protein Expr. Purif. 94, 22-32.]). The use of advanced genetic engineering methods to modify both cells and expression plasmids is covered in more detail in reviews such as Sørensen & Mortensen (2005[Sørensen, H. P. & Mortensen, K. K. (2005). Advanced genetic strategies for recombinant protein expression in Escherichia coli. J. Biotechnol. 115, 113-128.]).

Table 3
Methods for improving protein stability during expression and purification

  Construct design Expression conditions
  Removal of degradation-prone and protease sites Optimized codon usage Truncations and point mutations Chaperones and co-expression Affinity tags for improved solubility
Examples N-end rule, PEST sequences and specific protease sites Synthetic gene synthesis offered by many companies Stabilizing mutations for membrane proteins Coexpression and fusion vectors with DnaK and GroEL GST, MBP, Halo tags
Key references Bachmair et al. (1986[Bachmair, A., Finley, D. & Varshavsky, A. (1986). In vivo half-life of a protein is a function of its amino-terminal residue. Science, 234, 179-186.]), Rogers et al. (1986[Rogers, S., Wells, R. & Rechsteiner, M. (1986). Amino acid sequences common to rapidly degraded proteins: the PEST hypothesis. Science, 234, 364-368.]), Spiegel et al. (2015[Spiegel, H., Schinkel, H., Kastilan, R., Dahm, P., Boes, A., Scheuermayer, M., Chudobová, I., Maskus, D., Fendel, R., Schillberg, S., Reimann, A. & Fischer, R. (2015). Optimization of a multi-stage, multi-subunit malaria vaccine candidate for the production in Pichia pastoris by the identification and removal of protease cleavage sites. Biotechnol. Bioeng. 112, 659-667.]) Daniel et al. (2015[Daniel, E., Onwukwe, G. U., Wierenga, R. K., Quaggin, S. E., Vainio, S. J. & Krause, M. (2015). ATGme: open-source web application for rare codon identification and custom DNA sequence optimization. BMC Bioinformatics, 16, 303.]), Kane (1995[Kane, J. F. (1995). Effects of rare codon clusters on high-level expression of heterologous proteins in Escherichia coli. Curr. Opin. Biotechnol. 6, 494-500.]) Gräslund et al. (2008[Gräslund, S., Sagemark, J., Berglund, H., Dahlgren, L. G., Flores, A., Hammarström, M., Johansson, I., Kotenyova, T., Nilsson, M., Nordlund, P. & Weigelt, J. (2008). The use of systematic N- and C-terminal deletions to promote production and structural studies of recombinant proteins. Protein Expr. Purif. 58, 210-221.]), Klock et al. (2008[Klock, H. E., Koesema, E. J., Knuth, M. W. & Lesley, S. A. (2008). Combining the polymerase incomplete primer extension method for cloning and mutagenesis with microscreening to accelerate structural genomics efforts. Proteins, 71, 982-994.]) Kyratsous & Panagiotidis (2012[Kyratsous, C. A. & Panagiotidis, C. A. (2012). Heat-shock protein fusion vectors for improved expression of soluble recombinant proteins in Escherichia coli. Methods Mol. Biol. 824, 109-129.]), Kyratsous et al. (2009[Kyratsous, C. A., Silverstein, S. J., DeLong, C. R. & Panagiotidis, C. A. (2009). Chaperone-fusion expression plasmid vectors for improved solubility of recombinant proteins in Escherichia coli. Gene, 440, 9-15.]) Walls & Loughran (2011[Walls, D. & Loughran, S. T. (2011). Tagging recombinant proteins to enhance solubility and aid purification. Methods Mol. Biol. 681, 151-175.]), Kapust & Waugh (1999[Kapust, R. B. & Waugh, D. S. (1999). Escherichia coli maltose-binding protein is uncommonly effective at promoting the solubility of polypeptides to which it is fused. Protein Sci. 8, 1668-1674.])
  Expression conditions Host cells
  Cofactors and ligands Low-temperature expression Codon-optimized cells Reduced-toxicity and reduced-protease cells
Examples Metal ions and cofactors essential for folding, also other stabilizing cofactors Cold-shock, low-temperature inducible promoters such as pCold E. coli Rosetta E. coli BL21 Star and pLysS
Key references Leibly et al. (2012[Leibly, D. J., Nguyen, T. N., Kao, L. T., Hewitt, S. N., Barrett, L. K. & Van Voorhis, W. C. (2012). Stabilizing additives added during cell lysis aid in the solubilization of recombinant proteins. PLoS One, 7, e52482.]) Qing et al. (2004[Qing, G., Ma, L.-C., Khorchid, A., Swapna, G. V. T., Mal, T. K., Takayama, M. M., Xia, B., Phadtare, S., Ke, H., Acton, T., Montelione, G. T., Ikura, M. & Inouye, M. (2004). Cold-shock induced high-yield protein production in Escherichia coli. Nature Biotechnol. 22, 877-882.]), Vasina & Baneyx (1997[Vasina, J. A. & Baneyx, F. (1997). Expression of aggregation-prone recombinant proteins at low temperatures: a comparative study of the Escherichia coli cspA and tac promoter systems. Protein Expr. Purif. 9, 211-218.]) Tegel et al. (2010[Tegel, H., Tourle, S., Ottosson, J. & Persson, A. (2010). Increased levels of recombinant human proteins with the Escherichia coli strain Rosetta(DE3). Protein Expr. Purif. 69, 159-167.]) Studier (1991[Studier, F. W. (1991). Use of bacteriophage T7 lysozyme to improve an inducible T7 expression system. J. Mol. Biol. 219, 37-44.])

4.1. Construct design, sequence, expression tags and protein stability

The design of the expression construct is one of the primary decisions that a structural biologist must make to ensure the efficient expression and purification of the target protein. For example, the compositional stability of a protein can be severely affected by the presence of protease cleavage sites within the target protein. These sites can result in cleavage of the target protein by endogenous proteases produced by the expression host. Proteolysis can be mitigated by the removal of cleavage sites from the expression construct during recombinant assembly. For example, such an approach was used to remove two protease sites from a malarial vaccine candidate that was proteolytically degraded by endogenous KEX2 protease during expression. Removal of these sites enabled the production of full-length protein on a large scale (Spiegel et al., 2015[Spiegel, H., Schinkel, H., Kastilan, R., Dahm, P., Boes, A., Scheuermayer, M., Chudobová, I., Maskus, D., Fendel, R., Schillberg, S., Reimann, A. & Fischer, R. (2015). Optimization of a multi-stage, multi-subunit malaria vaccine candidate for the production in Pichia pastoris by the identification and removal of protease cleavage sites. Biotechnol. Bioeng. 112, 659-667.]). Additionally, the primary structures of some proteins are inherently unstable, with unusually short half-lives. For example, proteins containing sequences rich in proline, glutamate, serine and threonine (PESTs) often have half-lives of less than 2 h (Rogers et al., 1986[Rogers, S., Wells, R. & Rechsteiner, M. (1986). Amino acid sequences common to rapidly degraded proteins: the PEST hypothesis. Science, 234, 364-368.]). It is suggested that PEST sequences target the protein for intracellular degradation via the proteasome machinery (Spencer et al., 2004[Spencer, M. L., Theodosiou, M. & Noonan, D. J. (2004). NPDC-1, a novel regulator of neuronal proliferation, is degraded by the ubiquitin/proteasome system through a PEST degradation motif. J. Biol. Chem. 279, 37069-37078.]) or via more traditional proteolysis pathways utilizing calpain (Shumway et al., 1999[Shumway, S. D., Maki, M. & Miyamoto, S. (1999). The PEST domain of IκBα is necessary and sufficient for in vitro degradation by μ-calpain. J. Biol. Chem. 274, 30874-30881.]). Furthermore, the N-end rule is a strong predictor of protein half-life in vivo. For example, if the N-terminal residue of a protein is methionine, serine, alanine, glycine, threonine, valine or proline the half-life is stabilized (>20 h). In contrast, if the N-terminal residue is phenylalanine, aspartate, lysine or arginine the half-life of the protein is destabilized (<3 min; Bachmair et al., 1986[Bachmair, A., Finley, D. & Varshavsky, A. (1986). In vivo half-life of a protein is a function of its amino-terminal residue. Science, 234, 179-186.]). Proteins destabilized in this way are targeted for degradation via the ubiquitination pathway; therefore, the N-end rule is of no concern when using bacterial expression systems. It is important to note that the N-terminal residue is `masked' by the inclusion of an N-terminal affinity purification tag, as is typically used in most laboratories today.

In addition to the half-life stabilizing effects of affinity purification tags, they are also of considerable interest for improving the solubility of a target protein (Amarasinghe & Jin, 2015[Amarasinghe, C. & Jin, J.-P. (2015). The use of affinity tags to overcome obstacles in recombinant protein expression and purification. Protein Pept. Lett. 22, 885-892.]; Wood, 2014[Wood, D. W. (2014). New trends and affinity tag designs for recombinant protein purification. Curr. Opin. Struct. Biol. 26, 54-61.]). This is particularly true for maltose-binding protein (MBP), which has a strong effect in solubilizing the protein to which it is attached. MBP has also been shown to promote the correct folding of the protein target to which it is attached, suggesting that it can act as a form of molecular chaperone (Kapust & Waugh, 1999[Kapust, R. B. & Waugh, D. S. (1999). Escherichia coli maltose-binding protein is uncommonly effective at promoting the solubility of polypeptides to which it is fused. Protein Sci. 8, 1668-1674.]). Other chaperones that can be used to assist protein stability and folding during expression include DnaK and GroEL (Kyratsous & Panagiotidis, 2012[Kyratsous, C. A. & Panagiotidis, C. A. (2012). Heat-shock protein fusion vectors for improved expression of soluble recombinant proteins in Escherichia coli. Methods Mol. Biol. 824, 109-129.]). Some proteins are simply not stable in the cytoplasm and they can be redirected to other compartments of the host cell using an affinity tag. For example, the pMal vector system (NEB) incorporates the malE signal sequence and can be used to direct the protein of interest across the plasma membrane and into the periplasm. This has the added advantage of keeping the target protein away from cytoplasmic proteases during subsequent purification steps, thus further enhancing the compositional stability.

Self-cleaving affinity purification tags can be applied to carefully control the compositional stability. Traditional affinity-tag removal procedures often use proteases, such as thrombin or factor Xa, which can result in nonspecific degradation of the target protein. However, the use of highly specific self-cleaving tags prevents this issue as exogenous protease addition is not required. In addition to the self-cleaving intein-based tags discussed above (see §[link]3.1), it has also been reported that nickel ions can be used to cleave an affinity tag. In the example of the GmSPI-2 inhibitor structure the peptide bond preceding the serine or threonine residue in the (S/T)XHZ motif was cleaved by nickel ions (Kopera et al., 2014[Kopera, E., Bal, W., Lenarčič Živkovič, M., Dvornyk, A., Kludkiewicz, B., Grzelak, K., Zhukov, I., Zagórski-Ostoja, W., Jaskolski, M. & Krzywda, S. (2014). Atomic resolution structure of a protein prepared by non-enzymatic His-tag removal. Crystallographic and NMR study of GmSPI-2 inhibitor. PLoS One, 9, e106936.]; Krężel et al., 2010[Krężel, A., Kopera, E., Protas, A. M., Poznański, J., Wysłouch-Cieszyńska, A. & Bal, W. (2010). Sequence-specific NiII-dependent peptide bond hydrolysis for protein engineering. Combinatorial library determination of optimal sequences. J. Am. Chem. Soc. 132, 3355-3366.]).

4.2. Expression conditions and protein stability

In order to ensure proper protein folding and stability, it is essential that the expression host is provided with the necessary prosthetic groups, cofactors and ligands as required by the target protein. Many of these are provided by the expression medium and are scavenged by the host cells during the course of expression. However, some cofactors and prosthetic groups cannot be synthesized by the host cell and others are not available in sufficient quantities. For example, heme incorporation is often low when heme-containing proteins are expressed in Escherichia coli. In these cases the expression medium must be supplemented with δ-aminolevulinate in order to achieve satisfactory levels of heme incorporation (Kery et al., 1995[Kery, V., Elleder, D. & Kraus, J. P. (1995). δ-Aminolevulinate increases heme saturation and yield of human cystathionine β-synthase expressed in Escherichia coli. Arch. Biochem. Biophys. 316, 24-29.]). Additionally, the solubility of the protein can also be significantly improved during expression by the addition of additives such as trehalose, glycine betaine, mannitol, L-arginine, potassium citrate, CuCl2, proline, xylitol, NDSB 201, CTAB and K2PO4 (Leibly et al., 2012[Leibly, D. J., Nguyen, T. N., Kao, L. T., Hewitt, S. N., Barrett, L. K. & Van Voorhis, W. C. (2012). Stabilizing additives added during cell lysis aid in the solubilization of recombinant proteins. PLoS One, 7, e52482.]; see §[link]6.2 for more on buffer screening).

Varying the temperature at which the expression is carried out can also be used to control protein stability and solubility. For example, cold-shock induction systems such as pCold (Takara/Clontech) can be used to improve the overall stability of the target protein (Qing et al., 2004[Qing, G., Ma, L.-C., Khorchid, A., Swapna, G. V. T., Mal, T. K., Takayama, M. M., Xia, B., Phadtare, S., Ke, H., Acton, T., Montelione, G. T., Ikura, M. & Inouye, M. (2004). Cold-shock induced high-yield protein production in Escherichia coli. Nature Biotechnol. 22, 877-882.]). As an added benefit, at lower temperatures, cell proliferation is halted and the expression of endogenous proteins such as proteases is reduced. Therefore, the target protein is further protected from degradation and purity is improved. To assist in low-temperature expression, cold-adapted E. coli cells, for example ArcticExpress (Agilent Technologies), have been developed. These cells co-express the cold-adapted chaperonins Cpn10 and Cpn60 from the psychrophilic bacterium Oleispira antartica (see §[link]4.3 for more on host-cell selection).

4.3. Host cells and protein stability

The choice of the host cells that are used for the expression of recombinant proteins has an important influence on protein stability. For example, the protein may be toxic to the host cell or the protein may be cleaved by endogenous proteases made by the cell. Clearly, such issues can be a primary source of compositional instability in proteins. Toxicity can be controlled by tight regulation of the expression level using promoters that respond in a concentration-dependent manner to the inducer. Examples of tightly controlled expression vectors include pBAD, which responds to the inducer L-arabinose (Guzman et al., 1995[Guzman, L. M., Belin, D., Carson, M. J. & Beckwith, J. (1995). Tight regulation, modulation, and high-level expression by vectors containing the arabinose PBAD promoter. J. Bacteriol. 177, 4121-4130.]). The background expression of proteases (and proteins in general) can also be controlled using plasmids that express T7 lysozyme, such as pLysS. T7 lysozyme is a natural inhibitor of T7 RNA polymerase, the promoter utilized in the pET vector system, and can be used to reduce background levels of protein expression (Studier, 1991[Studier, F. W. (1991). Use of bacteriophage T7 lysozyme to improve an inducible T7 expression system. J. Mol. Biol. 219, 37-44.]). Background levels of protease activity can also be reduced using OmpT bacterial strains, which do not express the outer membrane aspartyl protease. Such systems are commercially available as BL21 Star strains of E. coli (Invitrogen/ThermoFisher Scientific).

Host-cell selection is particularly important when expressing mammalian proteins in bacterial cells, as the codon usage between the organisms is different. For example, the AGA codon for arginine is particularly rare in E. coli and can result in premature chain termination, frame-shifting and incorrect amino-acid insertion (Calderone et al., 1996[Calderone, T. L., Stevens, R. D. & Oas, T. G. (1996). High-level misincorporation of lysine for arginine at AGA codons in a fusion protein expressed in Escherichia coli. J. Mol. Biol. 262, 407-412.], Kane, 1995[Kane, J. F. (1995). Effects of rare codon clusters on high-level expression of heterologous proteins in Escherichia coli. Curr. Opin. Biotechnol. 6, 494-500.]). This issue can be addressed in a number of ways, including the generation of a synthetic gene reflecting the codon usage of the host organism or by co-transformation of the host with a plasmid that can provide the tRNA of the missing codons (e.g. CodonPlus, Stratagene and pRARE; EMD Millipore/Novagen; Dieci et al., 2000[Dieci, G., Bottarelli, L., Ballabeni, A. & Ottonello, S. (2000). tRNA-assisted overproduction of eukaryotic ribosomal proteins. Protein Expr. Purif. 18, 346-354.]; Fu et al., 2007[Fu, W., Lin, J. & Cen, P. (2007). 5-Aminolevulinate production with recombinant Escherichia coli using a rare codon optimizer host strain. Appl. Microbiol. Biotechnol. 75, 777-782.]). Competent E. coli BL21 cells containing the pRARE plasmid are commercially available under the trade name Rosetta. These cells have been used to optimize the expression of many human proteins in E. coli. For example, the Swedish Human Protein Atlas project has been successful in improving both the level of expression and the purity of proteins using the Rosetta E. coli strain (Tegel et al., 2010[Tegel, H., Tourle, S., Ottosson, J. & Persson, A. (2010). Increased levels of recombinant human proteins with the Escherichia coli strain Rosetta(DE3). Protein Expr. Purif. 69, 159-167.]).

Finally, mammalian expression systems such as Chinese hamster ovary (CHO) cells (Fischer et al., 2015[Fischer, S., Handrick, R. & Otte, K. (2015). The art of CHO cell engineering: a comprehensive retrospect and future perspectives. Biotechnol. Adv. 33, 1878-1896.]) and human embryonic kidney cells (e.g. HEK 293T and 293F; Nettleship et al., 2015[Nettleship, J. E., Watson, P. J., Rahman-Huq, N., Fairall, L., Posner, M. G., Upadhyay, A., Reddivari, Y., Chamberlain, J. M., Kolstoe, S. E., Bagby, S., Schwabe, J. W. & Owens, R. J. (2015). Transient expression in HEK 293 cells: an alternative to E. coli for the production of secreted and intracellular mammalian proteins. Methods Mol. Biol. 1258, 209-222.]) are often essential for the expression of mammalian or human proteins (for a discussion of the merits of the various expression systems, see Brondyk, 2009[Brondyk, W. H. (2009). Selecting an appropriate method for expressing a recombinant protein. Methods Enzymol. 463, 131-147.]). In addition to addressing the codon-usage issue, expression in mammalian cells is often required to ensure that PTMs are correctly added and the protein is correctly folded and active (see §[link]6.6 for a discussion of GnTI and lec1 glycosylation-deficient mammalian cells). Alternatively, insect cells such as Spodoptera frugiperda (e.g. Sf9 and Sf21) and Trichoplusia ni can be used (Altmann et al., 1999[Altmann, F., Staudacher, E., Wilson, I. B. & März, L. (1999). Insect cells as hosts for the expression of recombinant glycoproteins. Glycoconj. J. 16, 109-123.]; Jarvis, 2009[Jarvis, D. L. (2009). Baculovirus-insect cell expression systems. Methods Enzymol. 463, 191-222.]).

5. Key techniques for determining protein stability

The relative merits of the three main structural methods for assessing protein stability are shown in Table 2[link]. Given the solid-state nature of protein crystallography it is often difficult to crystallize dynamic and disordered proteins, and for this reason NMR spectroscopy has been one of the main tools used to study IDPs such as p53 and CREB (Brutscher et al., 2015[Brutscher, B., Felli, I. C., Gil-Caballero, S., Hošek, T., Kümmerle, R., Piai, A., Pierattelli, R. & Sólyom, Z. (2015). NMR methods for the study of instrinsically disordered proteins structure, dynamics, and interactions: general overview and practical guidelines. Adv. Exp. Med. Biol. 870, 49-122.]; Dunker & Oldfield, 2015[Dunker, A. K. & Oldfield, C. J. (2015). Back to the future: nuclear magnetic resonance and bioinformatics studies on intrinsically disordered proteins. Adv. Exp. Med. Biol. 870, 1-34.]; Mujtaba et al., 2004[Mujtaba, S., He, Y., Zeng, L., Yan, S., Plotnikova, O., Sachchidanand, Sanchez, R., Zeleznik-Le, N. J., Ronai, Z. & Zhou, M.-M. (2004). Structural mechanism of the bromodomain of the coactivator CBP in p53 transcriptional activation. Mol. Cell, 13, 251-263.]; Fig. 2[link]d). NMR is extremely useful for assessing both secondary and tertiary structure in dynamic and disordered systems (see §[link]5.1). The higher resolution of crystal structures make them a particularly attractive method for determining changes at the primary-structure level. For example, uncharacterized PTMs, such as glycosylation, can often be interpreted directly from the electron-density maps if the experimental data are of sufficiently high resolution. Similarly, given sufficiently high-resolution maps, cryo-EM can be a powerful technique for determining protein stability and dynamics at the quaternary level (see §[link]5.2).

5.1. Nuclear magnetic resonance (NMR)

NMR spectroscopy is a powerful method for the determination of the stability of proteins in solution (Bieri et al., 2011[Bieri, M., Kwan, A. H., Mobli, M., King, G. F., Mackay, J. P. & Gooley, P. R. (2011). Macromolecular NMR spectroscopy for the non-spectroscopist: beyond macromolecular solution structure determination. FEBS J. 278, 704-715.]; Kwan et al., 2011[Kwan, A. H., Mobli, M., Gooley, P. R., King, G. F. & Mackay, J. P. (2011). Macromolecular NMR spectroscopy for the non-spectroscopist. FEBS J. 278, 687-703.]). The method is highly complementary to X-ray structure analysis, but given its ability to analyze structures in the solution state it is of tremendous value for assessing protein conformational stability (Krishnan & Rupp, 2012[Krishnan, V. V. & Rupp, B. (2012). Macromolecular structure determination: comparison of X-ray crystallography and NMR spectroscopy. eLS, doi:10.1002/9780470015902.a0002716.pub2.]).

The fact that NMR can readily distinguish between folded and unfolded proteins, and detect the presence of disordered and unstructured regions, makes it inherently useful as a diagnostic tool for crystallization experiments (Fig. 3[link]a). Modern instruments can extract this information with minimal sample requirement (∼10 nM) and a simple one-dimensional proton NMR spectrum can provide information on the conformational stability of the macromolecule. Specifically, as a result of the principal inverse relation between spin–spin relaxation time and the peak width, large soluble aggregates will not yield an interpretable high-resolution NMR spectrum. For non-aggregated protein samples that yield usable one-dimensional NMR spectra, good discrimination in the backbone amide region below 8.3 p.p.m., as well as peaks at around ∼1 p.p.m., are indicative of folded protein (Rehm et al., 2002[Rehm, T., Huber, R. & Holak, T. A. (2002). Application of NMR in structural proteomics: screening for proteins amenable to structural analysis. Structure, 10, 1613-1618.]). Furthermore, two-dimensional heteronuclear single-quantum coherence (HSQC) NMR spectra can be used to analyze the difference between folded and unstructured protein (Fig. 3[link]a) and also to compare apoprotein and ligand-bound complexes (Figs. 3[link]b and 3[link]c). Such a two-dimensional spectrum maps the backbone amide groups according to their 1H and 15N resonance frequencies. This method necessitates the production of 15N-labeled protein and requires larger amounts of sample compared with the more qualitative one-dimensional spectral analysis (Zhao et al., 2004[Zhao, Q., Frederick, R., Seder, K., Thao, S., Sreenath, H., Peterson, F., Volkman, B. F., Markley, J. L. & Fox, B. G. (2004). Production in two-liter beverage bottles of proteins for NMR structure determination labeled with either 15N- or 13C-15N. J. Struct. Funct. Genomics, 5, 87-93.]).

[Figure 3]
Figure 3
HSQC spectrum of folded, unstructured and apo and ligand-bound proteins. (a) Two-dimensional 1H–15N heteronuclear single-quantum coherence (HSQC) NMR spectrum showing the distinct discrimination in the region below 8.3 p.p.m. in ω1 identifying a folded protein (red, sharp peak contours) compared with the wide and unresolved peaks for disordered protein sample (blue contours). Image courtesy of Simon Colebrook, Department of Biochemistry, Oxford University and Joanne Nettleship, Oxford Protein Production facility. (b) HSQC spectrum of apo and ligand-bound protein. The two-dimensional 1H–15N HSQC NMR spectrum of bacterial methionine aminopeptidase (bMAP) with (right) and without (left) a tightly bound novel inhibitor (Evdokimov et al., 2007[Evdokimov, A. G. et al. (2007). Serendipitous discovery of novel bacterial methionine aminopeptidase inhibitors. Proteins, 66, 538-546.]). Note the drastic improvement in the discrimination of the spectrum for the bMAP–ligand complex compared with the apoprotein. The crystals of the bMAP–ligand complex diffracted to 0.9 Å resolution. Image courtesy of Artem Evdokimov, Procter & Gamble Pharmaceuticals, Mason, Ohio, USA. Figure adapted from Rupp (2015[Rupp, B. (2015). Origin and use of crystallization phase diagrams. Acta Cryst. F71, 247-260.]).

One of the main benefits of NMR methods is that the effect of environmental conditions, such as pH, temperature or ligand binding, can be readily varied and studied in a near-native solution state. Additionally, the nondestructive nature of NMR spectroscopy means that the samples can also be used for subsequent crystallization experiments, and high-throughput structure-determination facilities often combine NMR screening with crystallization experiments.

5.2. Electron microscopy

The high conformational heterogeneity of proteins, especially of large multi-domain protein–protein complexes, can often hinder crystallization efforts. Mutational variants and different combinations of protein partners may need to be screened for suitability for crystallization. To facilitate this, electron microscopy (EM) can be used to directly visualize the sample and assess the level of heterogeneity. In the best-case scenario three-dimensional cryo-EM reconstructions can be generated, but this can be time-consuming and often requires substantial efforts in screening for suitable data-collection parameters. However, the generation of raw images of negatively stained particles is straightforward and requires very little protein (typically <10 µg). This method is already routinely practiced by EM practitioners to screen for good samples to move forward for cryo-EM analysis, but has recently been adapted for crystallography. However, the negative stain (e.g. uranyl formate) may introduce artifacts or otherwise disrupt the protein sample. Additionally, the protein (or protein–protein complex) needs to be relatively large (>150 kDa) in order to be imaged. If these caveats can be overcome then simple negative staining can be a powerful technique for providing a low-resolution glimpse of the protein sample especially for rapid large-scale screening purposes. Furthermore, simple image analysis of the particles can be used to produce a two-dimensional class average and quantify the conformational classes within the protein sample. Thus, EM can guide the screening of protein constructs for further structural studies, such as crystallization or full three-dimensional cryo-EM reconstructions (Pugach et al., 2015[Pugach, P. et al. (2015). A native-like SOSIP.664 trimer based on an HIV-1 subtype B env gene. J. Virol. 89, 3380-3395.]; Julien et al., 2015[Julien, J.-P. et al. (2015). Design and structure of two HIV-1 clade C SOSIP.664 trimers that increase the arsenal of native-like Env immunogens. Proc. Natl Acad. Sci. USA, 112, 11947-11952.]). It is important to note that protein crystallography generally works well with protein complexes of <150 kDa, which makes these two techniques highly complementary.

With technological innovations pushing its data sets beyond 3 Å resolution, cryo-EM techniques are becoming an increasingly useful tool for screening of both protein conformational stability and compositional stability. Examples of recent studies using EM to assess protein stability and dynamics include the structural transitions of αβ-tubulin (Alushin et al., 2014[Alushin, G. M., Lander, G. C., Kellogg, E. H., Zhang, R., Baker, D. & Nogales, E. (2014). High-resolution microtubule structures reveal the structural transitions in αβ-tubulin upon GTP hydrolysis. Cell, 157, 1117-1129.]) and the stability of the HBV capsid protein (Selzer et al., 2015[Selzer, L., Kant, R., Wang, J. C., Bothner, B. & Zlotnick, A. (2015). Hepatitis B virus core protein phosphorylation sites affect capsid stability and transient exposure of the C-terminal domain. J. Biol. Chem. 290, 28584-28593]).

5.3. Spectroscopic methods

Spectroscopic methods are primarily used to assess the stability of the protein at the level of secondary structure. Secondary structure can be analyzed using a variety of spectroscopic methods including circular dichroism (CD), Fourier transform infrared (FT-IR) and Raman spectroscopy (Pelton & McLean, 2000[Pelton, J. T. & McLean, L. R. (2000). Spectroscopic methods for analysis of protein secondary structure. Anal. Biochem. 277, 167-176.]). CD spectroscopy, or spectropolarimetry, of proteins is carried out in the far-ultraviolet range (170–250 nm) and has seen a recent resurgence in usage as a result of the development of synchrotron-radiation circular dichroism (SRCD), which can operate at lower wavelengths (Whitmore & Wallace, 2008[Whitmore, L. & Wallace, B. A. (2008). Protein secondary structure analyses from circular dichroism spectroscopy: methods and reference databases. Biopolymers, 89, 392-400.]; Wallace & Janes, 2010[Wallace, B. A. & Janes, R. W. (2010). Synchrotron radiation circular dichroism (SRCD) spectroscopy: an enhanced method for examining protein conformations and protein interactions. Biochem. Soc. Trans. 38, 861-873.]). SRCD data are now publicly available in a central repository at the Protein Circular Dichroism Data Bank (PCDDB), which currently contains 529 entries (Whitmore et al., 2011[Whitmore, L., Woollett, B., Miles, A. J., Klose, D. P., Janes, R. W. & Wallace, B. A. (2011). PCDDB: the Protein Circular Dichroism Data Bank, a repository for circular dichroism spectral and metadata. Nucleic Acids Res. 39, D480-D486.]; https://bit.ly/1OrNzrP ). Using this technique, a twin minimum in the ellipticity spectrum at 208 and 222 nm is produced by α-helical content, whereas a single minimum at 204 or 217 nm is suggestive of random-coil or β-sheet content, respectively. Example uses of UV-CD for assessing protein stability include a study of the plant membrane protein MBP-b6. The percentages of helix, sheet, turn and unordered secondary structure were determined and it was demonstrated that n-dodecyl-β-D-maltoside and Triton X-100 both preserved the correct secondary structure, whereas sodium dodecyl sulfate was shown to disrupt the secondary structure (Surma et al., 2014[Surma, M. A., Szczepaniak, A. & Króliczewski, J. (2014). Comparative studies on detergent-assisted apocytochrome b6 reconstitution into liposomal bilayers monitored by zetasizer instruments. PLoS One, 9, e111341.]).

Infrared spectroscopy can also be used to identify secondary-structure content (Barth, 2007[Barth, A. (2007). Infrared spectroscopy of proteins. Biochim. Biophys. Acta, 1767, 1073-1101.]). This technique is used to analyze changes in the bond oscillation of amide groups which result from differences in the hydrogen-bonding pattern. Refinements of this technique include femtosecond two-dimensional infrared spectroscopy (2D-IR), which is more sensitive in detecting structural differences (Demirdöven et al., 2004[Demirdöven, N., Cheatum, C. M., Chung, H. S., Khalil, M., Knoester, J. & Tokmakoff, A. (2004). Two-dimensional infrared spectroscopy of antiparallel β-sheet secondary structure. J. Am. Chem. Soc. 126, 7981-7990.]).

5.4. Melting-temperature analysis

Assessment of protein stability, particularly thermodynamic stability, is of tremendous value for the crystallization of both soluble and membrane proteins. The global thermal stability of a protein can be represented by its thermal denaturation midpoint or melting temperature (Tm). Methods used for the determination of Tm include Thermofluor (also known as differential scanning fluorimetry; DSF; Boivin et al., 2013[Boivin, S., Kozak, S. & Meijers, R. (2013). Optimization of protein purification and characterization using Thermofluor screens. Protein Expr. Purif. 91, 192-206.]; Ericsson et al., 2006[Ericsson, U. B., Hallberg, B. M., DeTitta, G. T., Dekker, N. & Nordlund, P. (2006). Thermofluor-based high-throughput stability optimization of proteins for structural studies. Anal. Biochem. 357, 289-298.]; Reinhard et al., 2013[Reinhard, L., Mayerhofer, H., Geerlof, A., Mueller-Dieckmann, J. & Weiss, M. S. (2013). Optimization of protein buffer cocktails using Thermofluor. Acta Cryst. F69, 209-214.]) and differential scanning calorimetry (DSC; Sanchez-Ruiz, 1995[Sanchez-Ruiz, J. M. (1995). Differential scanning calorimetry of proteins. Subcell. Biochem. 24, 133-176.]; Privalov & Dragan, 2007[Privalov, P. L. & Dragan, A. I. (2007). Microcalorimetry of biological macromolecules. Biophys. Chem. 126, 16-24.]; Bruylants et al., 2005[Bruylants, G., Wouters, J. & Michaux, C. (2005). Differential scanning calorimetry in life science: thermodynamics, stability, molecular recognition and application in drug design. Curr. Med. Chem. 12, 2011-2020.]).

The Tm is often determined using the Thermofluor technique using a reporter dye such as SYPRO Orange or 1-anilinonaphthalene-8-sulfonate (ANS). The reporter dye undergoes a change in fluorescence properties upon binding to the hydrophobic core of the unfolded protein (Semisotnov et al., 1991[Semisotnov, G. V., Rodionova, N. A., Razgulyaev, O. I., Uversky, V. N., Gripas', A. F. & Gilmanshin, R. I. (1991). Study of the `molten globule' intermediate state in protein folding by a hydrophobic fluorescent probe. Biopolymers, 31, 119-128.]). A major advantage of the technique is that very little protein is required (typically <<1 mg) and the only equipment required is a readily available qPCR machine. One interesting application of this technique is the detection of ligand binding to proteins of unknown function. For example, by screening a library of 3000 compounds this technique was used to determine that an essential gene from Streptococcus pneumoniae was a nucleoside diphospho-keto-sugar aminotransferase (Carver et al., 2005[Carver, T. E. et al. (2005). Decrypting the biochemical function of an essential gene from Streptococcus pneumoniae using Thermofluor technology. J. Biol. Chem. 280, 11704-11712.]). Additionally, Thermofluor techniques can be used for the quantitation of protein–protein interactions. This has been used to analyze the stabilizing effect of maltose-binding protein on ankyrin-repeat proteins via the production of a series of alanine mutants to probe the interaction (Layton & Hellinga, 2011[Layton, C. J. & Hellinga, H. W. (2011). Quantitation of protein-protein interactions by thermal stability shift analysis. Protein Sci. 20, 1439-1450.]). Using such techniques, it is possible to screen hundreds of protein truncations (see §[link]6.1) and buffer conditions (see §[link]6.2) on a high-throughput scale (Boivin et al., 2013[Boivin, S., Kozak, S. & Meijers, R. (2013). Optimization of protein purification and characterization using Thermofluor screens. Protein Expr. Purif. 91, 192-206.]; Ristic et al., 2015[Ristic, M., Rosa, N., Seabrook, S. A. & Newman, J. (2015). Formulation screening by differential scanning fluorimetry: how often does it work? Acta Cryst. F71, 1359-1364.]; Seabrook & Newman, 2013[Seabrook, S. A. & Newman, J. (2013). High-throughput thermal scanning for protein stability: making a good technique more robust. ACS Comb. Sci. 15, 387-392.]; Reinhard et al., 2013[Reinhard, L., Mayerhofer, H., Geerlof, A., Mueller-Dieckmann, J. & Weiss, M. S. (2013). Optimization of protein buffer cocktails using Thermofluor. Acta Cryst. F69, 209-214.]). Melting-temperature methods are also applicable for the assessment of the thermo­dynamic stability of membrane proteins in detergents and have been successfully applied to the analysis of the stability of the acetylcholine receptor in Brij-35 (Yeh et al., 2006[Yeh, A. P., McMillan, A. & Stowell, M. H. B. (2006). Rapid and simple protein-stability screens: application to membrane proteins. Acta Cryst. D62, 451-457.]).

Potential caveats of the Thermofluor technique include the possibility of protein–dye interactions that many adversely affect the protein stability, a phenomenon that was observed in the study of GroEL when using the ANS dye (Smoot et al., 2001[Smoot, A. L., Panda, M., Brazil, B. T., Buckle, A. M., Fersht, A. R. & Horowitz, P. M. (2001). The binding of bis-ANS to the isolated GroEL apical domain fragment induces the formation of a folding intermediate with increased hydrophobic surface not observed in tetradecameric GroEL. Biochemistry, 40, 4484-4492.]). Additionally, aberrant or false-positive thermal shifts are fairly common, and careful analysis of the melt-curve data must be carried out; tools such as Meltdown are available to assist in this effort (Rosa et al., 2015[Rosa, N., Ristic, M., Seabrook, S. A., Lovell, D., Lucent, D. & Newman, J. (2015). Meltdown: a tool to help in the interpretation of thermal melt curves acquired by differential scanning fluorimetry. J. Biomol. Screen. 20, 898-905.]). It is also important to note that not all proteins unfold in a well-defined sigmoid melting curve, including proteins with high disulfide-bond content (e.g. albumin) and proteins from thermophilic organisms such as T. maritima. Finally, a negative Thermofluor result (i.e. no change in Tm) does not necessarily indicate a lack of binding for small molecules, but could simply mean that the small molecule does not stabilize the protein further. This is of particular relevance for proteins which have high melting temperatures in the absence of ligands.

6. Improving protein stability and the concept of crystallizability

Many techniques are available for increasing the crystallizability of a protein, and the central theme of these techniques is the improvement of both the compositional and the conformational stability of the protein sample (Table 4[link]). These methods include truncations, buffers, ligands, purification tags, reductive methylation, surface-entropy reduction (SER), in situ proteolysis, Thermofluor, deuterium-exchange mass spectrometry (DXMS) and disulfide engineering. A complementary review of protein-engineering approaches for improving the properties of proteins for crystallization studies is provided by Ruggiero et al. (2012[Ruggiero, A., Smaldone, G., Squeglia, F. & Berisio, R. (2012). Enhanced crystallizability by protein engineering approaches: a general overview. Protein Pept. Lett. 19, 732-742.]).

Table 4
Methods for modifying protein stability and improving protein crystallizability

Many of these techniques, in some way, affect both the compositional and conformational stability of the protein. Many of these methods have been successfully deployed as so-called `salvage' methods in high-throughput structural genomics consortia.

  Protein crystallizability- and stability-modifying methods
  Truncations and domain selection Buffer screening Ligands and additive screening Fused affinity tags and crystallization chaperones Reductive alkylation
Compositional stability Yes Yes Yes Yes Yes
Conformational stability Yes Yes Yes Yes Yes
Example Removal of disordered regions Stabilize buffers via ionic changes Binding to stabilize protein Stabilize protein with rigid fusion of tag Provides entropic benefit upon crystallization
Key references Yumerefendi et al. (2010[Yumerefendi, H., Tarendeau, F., Mas, P. J. & Hart, D. J. (2010). ESPRIT: an automated, library-based method for mapping and soluble expression of protein domains from challenging targets. J. Struct. Biol. 172, 66-74.]), Reich et al. (2006[Reich, S., Puckey, L. H., Cheetham, C. L., Harris, R., Ali, A. A. E., Bhattacharyya, U., Maclagan, K., Powell, K. A., Prodromou, C., Pearl, L. H., Driscoll, P. C. & Savva, R. (2006). Combinatorial domain hunting: an effective approach for the identification of soluble protein domains adaptable to high-throughput applications. Protein Sci. 15, 2356-2365.]), Klock et al. (2008[Klock, H. E., Koesema, E. J., Knuth, M. W. & Lesley, S. A. (2008). Combining the polymerase incomplete primer extension method for cloning and mutagenesis with microscreening to accelerate structural genomics efforts. Proteins, 71, 982-994.]), Gräslund et al. (2008[Gräslund, S., Sagemark, J., Berglund, H., Dahlgren, L. G., Flores, A., Hammarström, M., Johansson, I., Kotenyova, T., Nilsson, M., Nordlund, P. & Weigelt, J. (2008). The use of systematic N- and C-terminal deletions to promote production and structural studies of recombinant proteins. Protein Expr. Purif. 58, 210-221.]) Reinhard et al. (2013[Reinhard, L., Mayerhofer, H., Geerlof, A., Mueller-Dieckmann, J. & Weiss, M. S. (2013). Optimization of protein buffer cocktails using Thermofluor. Acta Cryst. F69, 209-214.]) Chung (2007[Chung, C. (2007). The use of biophysical methods increases success in obtaining liganded crystal structures. Acta Cryst. D63, 62-71.]), Hassell et al. (2007[Hassell, A. M. et al. (2007). Crystallization of protein-ligand complexes. Acta Cryst. D63, 72-79.]) Smyth et al. (2003[Smyth, D. R., Mrozkiewicz, M. K., McGrath, W. J., Listwan, P. & Kobe, B. (2003). Crystal structures of fusion proteins with large affinity tags. Protein Sci. 12, 1313-1322.]) Rice et al. (1977[Rice, R. H., Means, G. E. & Brown, W. D. (1977). Stabilization of bovine trypsin by reductive methylation. Biochim. Biophys. Acta, 492, 316-321.]), Tan et al. (2014[Tan, K. et al. (2014). Salvage of failed protein targets by reductive alkylation. Methods Mol. Biol. 1140, 189-200.]), Walter et al. (2006[Walter, T. S., Meier, C., Assenberg, R., Au, K. F., Ren, J., Verma, A., Nettleship, J. E., Owens, R. J., Stuart, D. I. & Grimes, J. M. (2006). Lysine methylation as a routine rescue strategy for protein crystallization. Structure, 14, 1617-1622.])
  Protein crystallizability- and stability-modifying methods
  Surface mutagenesis, surface-entropy reduction (SER) and deglycosylation In situ proteolysis Thermofluor Deuterium-exchange mass spectrometry (DXMS) Disulfide engineering
Compositional stability Yes Yes No No Yes
Conformational stability Yes Yes Yes Yes Yes
Example Entropic stabilization of large, flexible, surface-exposed residues Removal of flexible loops Analysis of thermal stability (Tm) Identification of flexible regions Stabilization of quaternary structure of crystal packing
Key references Cooper et al. (2007[Cooper, D. R., Boczek, T., Grelewska, K., Pinkowska, M., Sikorska, M., Zawadzki, M. & Derewenda, Z. (2007). Protein crystallization by surface entropy reduction: optimization of the SER strategy. Acta Cryst. D63, 636-645.]), Goldschmidt et al. (2014[Goldschmidt, L., Eisenberg, D. & Derewenda, Z. S. (2014). Salvage or recovery of failed targets by mutagenesis to reduce surface entropy. Methods Mol. Biol. 1140, 201-209.]) Dong et al. (2007[Dong, A. et al. (2007). In situ proteolysis for protein crystallization and structure determination. Nature Methods, 4, 1019-1021.]), Wernimont & Edwards (2009[Wernimont, A. & Edwards, A. (2009). In situ proteolysis to generate crystals for structure determination: an update. PLoS One, 4, e5094.]) Reinhard et al. (2013[Reinhard, L., Mayerhofer, H., Geerlof, A., Mueller-Dieckmann, J. & Weiss, M. S. (2013). Optimization of protein buffer cocktails using Thermofluor. Acta Cryst. F69, 209-214.]), Ristic et al. (2015[Ristic, M., Rosa, N., Seabrook, S. A. & Newman, J. (2015). Formulation screening by differential scanning fluorimetry: how often does it work? Acta Cryst. F71, 1359-1364.]) Englander (2006[Englander, S. W. (2006). Hydrogen exchange and mass spectrometry: a historical perspective. J. Am. Soc. Mass Spectrom. 17, 1481-1489.]), Englander & Kallenbach (1983[Englander, S. W. & Kallenbach, N. R. (1983). Hydrogen exchange and structural dynamics of proteins and nucleic acids. Q. Rev. Biophys. 16, 521-655.]) Forse et al. (2011[Forse, G. J., Ram, N., Banatao, D. R., Cascio, D., Sawaya, M. R., Klock, H. E., Lesley, S. A. & Yeates, T. O. (2011). Synthetic symmetrization in the crystallization and structure determination of CelA from Thermotoga maritima. Protein Sci. 20, 168-178.]), Quistgaard (2014[Quistgaard, E. M. (2014). A disulfide polymerized protein crystal. Chem. Commun. 50, 14995-14997.])

Several software packages and algorithms have been developed to assess the so-called crystallizability of a protein (Smialowski & Frishman, 2010[Smialowski, P. & Frishman, D. (2010). Protein crystallizability. Methods Mol. Biol. 609, 385-400.]; Derewenda, 2010[Derewenda, Z. S. (2010). Application of protein engineering to enhance crystallizability and improve crystal properties. Acta Cryst. D66, 604-615.]; Ruggiero et al., 2012[Ruggiero, A., Smaldone, G., Squeglia, F. & Berisio, R. (2012). Enhanced crystallizability by protein engineering approaches: a general overview. Protein Pept. Lett. 19, 732-742.]). These include DisMeta (Huang et al., 2014[Huang, Y. J., Acton, T. B. & Montelione, G. T. (2014). DisMeta: a meta server for construct design and optimization. Methods Mol. Biol. 1091, 3-16.]), XtalPred (Jahandideh et al., 2014[Jahandideh, S., Jaroszewski, L. & Godzik, A. (2014). Improving the chances of successful protein structure determination with a random forest classifier. Acta Cryst. D70, 627-635.]; Slabinski et al., 2007[Slabinski, L., Jaroszewski, L., Rychlewski, L., Wilson, I. A., Lesley, S. A. & Godzik, A. (2007). XtalPred: a web server for prediction of protein crystallizability. Bioinformatics, 23, 3403-3405.]), POODLE (Shimizu, 2014[Shimizu, K. (2014). POODLE: tools predicting intrinsically disordered regions of amino acid sequence. Methods Mol. Biol. 1137, 131-145.]), MFDp2 (Mizianty et al., 2014[Mizianty, M. J., Uversky, V. & Kurgan, L. (2014). Prediction of intrinsic disorder in proteins using MFDp2. Methods Mol. Biol. 1137, 147-162.]), MoRFpred (Disfani et al., 2012[Disfani, F. M., Hsu, W.-L., Mizianty, M. J., Oldfield, C. J., Xue, B., Dunker, A. K., Uversky, V. N. & Kurgan, L. (2012). MoRFpred, a computational tool for sequence-based prediction and characterization of short disorder-to-order transitioning binding regions in proteins. Bioinformatics, 28, i75-i83.]), RFCRYS (Jahandideh & Mahdavi, 2012[Jahandideh, S. & Mahdavi, A. (2012). RFCRYS: sequence-based protein crystallization propensity prediction by means of random forest. J. Theor. Biol. 306, 115-119.]), XANNpred (Overton et al., 2011[Overton, I. M., van Niekerk, C. A. & Barton, G. J. (2011). XANNpred: neural nets that predict the propensity of a protein to yield diffraction-quality crystals. Proteins, 79, 1027-1033.]), SVMCRYS (Kandaswamy et al., 2010[Kandaswamy, K. K., Pugalenthi, G., Suganthan, P. N. & Gangal, R. (2010). SVMCRYS: an SVM approach for the prediction of protein crystallization propensity from protein sequence. Protein Pept. Lett. 17, 423-430.]), SCMCRYS (Charoenkwan et al., 2013[Charoenkwan, P., Shoombuatong, W., Lee, H.-C., Chaijaruwanich, J., Huang, H.-L. & Ho, S.-Y. (2013). SCMCRYS: predicting protein crystallization using an ensemble scoring card method with estimating propensity scores of P-collocated amino acid pairs. PLoS One, 8, e72368.]), CRYSTALP2 (Kurgan et al., 2009[Kurgan, L., Razib, A. A., Aghakhani, S., Dick, S., Mizianty, M. & Jahandideh, S. (2009). CRYSTALP2: sequence-based protein crystallization propensity prediction. BMC Struct. Biol. 9, 50.]), MetaPPCP (Mizianty & Kurgan, 2009[Mizianty, M. J. & Kurgan, L. (2009). Meta prediction of protein crystallization propensity. Biochem. Biophys. Res. Commun. 390, 10-15.]) and ParCrys (Overton et al., 2008[Overton, I. M., Padovani, G., Girolami, M. A. & Barton, G. J. (2008). ParCrys: a Parzen window density estimation approach to protein crystallization propensity prediction. Bioinformatics, 24, 901-907.]). Many of these packages use a template-based approach to analyze the propensity of a protein to crystallize by comparison with known crystal structures. However, some of these packages, including XtalPred, POODLE and DisMeta, utilize sequence-based predictions to identify regions of low complexity, disorder, transmembrane and signal peptides.

It is important to note that the propensity for disorder calculated by most of these methods, and in turn the propensity for crystallization, is largely predicted on the basis of a single polypeptide chain in isolation. Clearly, protein–protein complexes can often result in stabilization of the constituent proteins, as is commonly observed for IDPs (see §[link]2.2). Therefore, particularly for crystallographic studies, it is often essential to study the protein–protein complex as a whole; the individual proteins are often too disordered or unstable when uncomplexed.

6.1. Truncations and domain selection

Selection of the shortest possible domain is often preferable and computational tools such as Expression of Soluble Proteins by Random Incremental Truncation (ESPRIT) and combinatorial domain hunting (CDH) are available to assist in this effort (Reich et al., 2006[Reich, S., Puckey, L. H., Cheetham, C. L., Harris, R., Ali, A. A. E., Bhattacharyya, U., Maclagan, K., Powell, K. A., Prodromou, C., Pearl, L. H., Driscoll, P. C. & Savva, R. (2006). Combinatorial domain hunting: an effective approach for the identification of soluble protein domains adaptable to high-throughput applications. Protein Sci. 15, 2356-2365.]; Yumerefendi et al., 2010[Yumerefendi, H., Tarendeau, F., Mas, P. J. & Hart, D. J. (2010). ESPRIT: an automated, library-based method for mapping and soluble expression of protein domains from challenging targets. J. Struct. Biol. 172, 66-74.]). Truncation of a protein to the shortest possible fragment can often be a key factor in successful structure solution. For example, amyloid fibers are of tremendous medical importance in diseases such Alzheimer's and prion diseases and their partially disordered structure has traditionally hindered structural analysis using crystallographic techniques. However, shorter fragments of only 6–7 amino acids in length, which also form fibrils, were used to produce microcrystals and to determine the structure (Moshe et al., 2016[Moshe, A., Landau, M. & Eisenberg, D. (2016). Preparation of crystalline samples of amyloid fibrils and oligomers. Methods Mol. Biol. 1345, 201-210.]; Sawaya et al., 2007[Sawaya, M. R., Sambashivan, S., Nelson, R., Ivanova, M. I., Sievers, S. A., Apostol, M. I., Thompson, M. J., Balbirnie, M., Wiltzius, J. J., McFarlane, H. T., Madsen, A. O., Riekel, C. & Eisenberg, D. (2007). Atomic structures of amyloid cross-β spines reveal varied steric zippers. Nature (London), 447, 453-457.]). Another example is the production of structured truncation arrays of a target protein generated using polymerase incomplete primer extension cloning methods (Klock & Lesley, 2009[Klock, H. E. & Lesley, S. A. (2009). The polymerase incomplete primer extension (PIPE) method applied to high-throughput cloning and site-directed mutagenesis. Methods Mol. Biol. 498, 91-103.]). Using this technique, structural genomics consortia such as the Joint Center for Structural Genomics (JCSG) and the Structural Genomics Institute, Karolinska Institutet have been able to generate several thousands of truncations for targets recalcitrant to crystallization (Klock et al., 2008[Klock, H. E., Koesema, E. J., Knuth, M. W. & Lesley, S. A. (2008). Combining the polymerase incomplete primer extension method for cloning and mutagenesis with microscreening to accelerate structural genomics efforts. Proteins, 71, 982-994.]; Gräslund et al., 2008[Gräslund, S., Sagemark, J., Berglund, H., Dahlgren, L. G., Flores, A., Hammarström, M., Johansson, I., Kotenyova, T., Nilsson, M., Nordlund, P. & Weigelt, J. (2008). The use of systematic N- and C-terminal deletions to promote production and structural studies of recombinant proteins. Protein Expr. Purif. 58, 210-221.]).

In addition to truncations of the protein, it is also important to consider other mutations of the protein. Stability-enhancing mutations in membrane proteins are surprisingly common and some estimates suggest that ∼10% of random mutations will confer some level of stability on the protein (Bowie, 2001[Bowie, J. U. (2001). Stabilizing membrane proteins. Curr. Opin. Struct. Biol. 11, 397-402.]). For example, two valine-to-alanine substitutions in the transmembrane portion of the M13 coat protein were found to enhance thermal stability (Deber et al., 1993[Deber, C. M., Khan, A. R., Li, Z., Joensson, C., Glibowicka, M. & Wang, J. (1993). Val→Ala mutations selectively alter helix-helix packing in the transmembrane segment of phage M13 coat protein. Proc. Natl Acad. Sci. USA, 90, 11648-11652.]). The reasons for the stability-enhancing effects of mutations are not always immediately obvious from analysis of the structure. It has been suggested that membrane proteins are required to be inherently flexible, and therefore conformationally unstable, in order to maintain function in the restricted environment of the membrane (Bowie, 2001[Bowie, J. U. (2001). Stabilizing membrane proteins. Curr. Opin. Struct. Biol. 11, 397-402.]).

6.2. Buffer screening

The buffer in which a protein is solubilized exerts an influence on its stability (Davis-Searles et al., 2001[Davis-Searles, P. R., Saunders, A. J., Erie, D. A., Winzor, D. J. & Pielak, G. J. (2001). Interpreting the effects of small uncharged solutes on protein-folding equilibria. Annu. Rev. Biophys. Biomol. Struct. 30, 271-306.]). Therefore, buffer screening is a powerful method for stabilizing proteins for crystallographic applications and also for the formulation of biologics. One of the primary methods used for high-throughput buffer screening is Thermofluor (see §[link]5.4). Using such approaches it is possible to screen libraries of hundreds of different buffers and pH combinations, and the stabilizing effect can be easily inferred from the change in Tm (ΔTm; Reinhard et al., 2013[Reinhard, L., Mayerhofer, H., Geerlof, A., Mueller-Dieckmann, J. & Weiss, M. S. (2013). Optimization of protein buffer cocktails using Thermofluor. Acta Cryst. F69, 209-214.]; Ristic et al., 2015[Ristic, M., Rosa, N., Seabrook, S. A. & Newman, J. (2015). Formulation screening by differential scanning fluorimetry: how often does it work? Acta Cryst. F71, 1359-1364.]). Using these approaches, interesting protein-stabilizing buffers have been identified. For example, citrate, bis-tris and N-(2-acetamido)iminodiacetic acid (ADA) have all been identified as having statistically significant stabilizing effects on the proteins tested (Ristic et al., 2015[Ristic, M., Rosa, N., Seabrook, S. A. & Newman, J. (2015). Formulation screening by differential scanning fluorimetry: how often does it work? Acta Cryst. F71, 1359-1364.]). As a more extreme example of buffer screening, IIPs (see §[link]2.2) such as V22-SH3 are insoluble in traditional buffer systems and can only be solubilized in pure water (Liu & Song, 2009[Liu, J. & Song, J. (2009). Insights into protein aggregation by NMR characterization of insoluble SH3 mutants solubilized in salt-free water. PLoS One, 4, e7805.]).

6.3. Ligands and additive screening

Ligand binding can also significantly help to stabilize the protein, particularly from the perspective of conformational stability. Co-crystal structures of proteins bound to cofactors, prosthetic groups, substrates, drugs and inhibitors are often the holy grail of structural biology; somewhat fortunately for the structural biologist, ligands often have a stabilizing effect on the protein and can increase the chances of successful crystallization. It is important to remember that although soaking of compounds through the crystal lattice to the active site is often possible, it may also bring about conformational changes in the protein on binding of the ligand. Therefore, ab initio crystal screening in the presence of the ligand may be required in order to obtain crystals (Hassell et al., 2007[Hassell, A. M. et al. (2007). Crystallization of protein-ligand complexes. Acta Cryst. D63, 72-79.]). The judicial use of bioanalytical techniques, such as Thermofluor and NMR, is key for guiding the successful production of a ligand-bound crystal structure (see §[link]5).

In addition to the stabilizing effects of small-molecule ligands, it is also possible to identify ions and other organic additives that stabilize the protein or even the crystal. For example, Thermofluor was used to identify magnesium ions as a stabilizing influence on the enzyme DapD from Mycobacterium tuberculosis, and the subsequent addition of MgCl2 to the crystallization solution resulted in larger crystals (Reinhard et al., 2013[Reinhard, L., Mayerhofer, H., Geerlof, A., Mueller-Dieckmann, J. & Weiss, M. S. (2013). Optimization of protein buffer cocktails using Thermofluor. Acta Cryst. F69, 209-214.]; Schuldt et al., 2009[Schuldt, L., Weyand, S., Kefala, G. & Weiss, M. S. (2009). The three-dimensional structure of a mycobacterial DapD provides insights into DapD diversity and reveals unexpected particulars about the enzymatic mechanism. J. Mol. Biol. 389, 863-879.]). In this case, stabilization of the quaternary structure results from the addition of magnesium ions, and two tightly coordinated Mg2+ ions were identified in the homotrimer interface of the crystal structure (Schuldt et al., 2009[Schuldt, L., Weyand, S., Kefala, G. & Weiss, M. S. (2009). The three-dimensional structure of a mycobacterial DapD provides insights into DapD diversity and reveals unexpected particulars about the enzymatic mechanism. J. Mol. Biol. 389, 863-879.]). Other examples of stabilizing additives include the commonly used precipitants polyethylene glycol (PEG) and 2-methyl-2,4-pentanediol (MPD), which are both often observed bound to crystal structures. In the case of MPD it has been proposed that it stabilizes the protein by promoting the hydration of the protein surface by binding to exposed hydrophobic surface residues such as leucine (Anand et al., 2002[Anand, K., Pal, D. & Hilgenfeld, R. (2002). An overview on 2-methyl-2,4-pentanediol in crystallization and in crystals of biological macromolecules. Acta Cryst. D58, 1722-1728.]). Building on this theme, additive screens such as `Silver Bullets' (Hampton Research) have been assembled that can stabilize or initiate cross-linking between proteins further promoting crystal lattice formation (McPherson & Cudney, 2006[McPherson, A. & Cudney, B. (2006). Searching for silver bullets: an alternative strategy for crystallizing macromolecules. J. Struct. Biol. 156, 387-406.]).

6.4. Fused affinity tags and crystallization chaperones

As discussed in §[link]4.1, affinity purification tags such as MBP are often used to aid in both protein stability and solubility during the course of protein expression and purification. Larger tags such as MBP are usually removed prior to crystallization trials, as the flexibility of the tag can interfere with crystal packing. However, in some cases smaller tags such as His can often be left on without unduly affecting crystal packing or protein function (Bucher et al., 2002[Bucher, M. H., Evdokimov, A. G. & Waugh, D. S. (2002). Differential effects of short affinity tags on the crystallization of Pyrococcus furiosus maltodextrin-binding protein. Acta Cryst. D58, 392-397.]). The primary reason for removing large tags is the reduced conformational stability resulting from the flexible linker between the target protein and its affinity tag. Several groups have successfully `engineered out' this linker flexibility by inserting a string of alanine residues in place of the usual protease cleavage site in the linker (Smyth et al., 2003[Smyth, D. R., Mrozkiewicz, M. K., McGrath, W. J., Listwan, P. & Kobe, B. (2003). Crystal structures of fusion proteins with large affinity tags. Protein Sci. 12, 1313-1322.]). This concept has resulting in several crystal structures of proteins fused to MBP, including gp21 (Kobe et al., 1999[Kobe, B., Center, R. J., Kemp, B. E. & Poumbourios, P. (1999). Crystal structure of human T cell leukemia virus type 1 gp21 ectodomain crystallized as a maltose-binding protein chimera reveals structural evolution of retroviral transmembrane proteins. Proc. Natl Acad. Sci. USA, 96, 4319-4324.]), SarR (Liu et al., 2001[Liu, Y., Manna, A., Li, R., Martin, W. E., Murphy, R. C., Cheung, A. L. & Zhang, G. (2001). Crystal structure of the SarR protein from Staphylococcus aureus. Proc. Natl Acad. Sci. USA, 98, 6877-6882.]), MATa1 (Ke & Wolberger, 2003[Ke, A. & Wolberger, C. (2003). Insights into binding cooperativity of MATa1/MATα2 from the crystal structure of a MATa1 homeodomain-maltose binding protein chimera. Protein Sci. 12, 306-312.]) and MCL1 (Clifton et al., 2015[Clifton, M. C. et al. (2015). A maltose-binding protein fusion construct yields a robust crystallography platform for MCL1. PLoS One, 10, e0125010.]). This concept has led to the idea of crystallization chaperones (Bukowska & Grütter, 2013[Bukowska, M. A. & Grütter, M. G. (2013). New concepts and aids to facilitate crystallization. Curr. Opin. Struct. Biol. 23, 409-416.]). Example uses of crystallization chaperones include the application of Fab antibody fragments to study the neurotransmitter sodium symporter LeuT in various conformational states (Krishnamurthy & Gouaux, 2012[Krishnamurthy, H. & Gouaux, E. (2012). X-ray structures of LeuT in substrate-free outward-open and apo inward-open states. Nature (London), 481, 469-474.]) and the fusion of T4 lysozyme to G-protein-coupled receptor (Zou et al., 2012[Zou, Y., Weis, W. I. & Kobilka, B. K. (2012). N-Terminal T4 lysozyme fusion facilitates crystallization of a G protein coupled receptor. PLoS One, 7, e46039.]). These approaches are proving to be useful for the stabilization of membrane proteins and as aids in their structure determination (Lieberman et al., 2011[Lieberman, R. L., Culver, J. A., Entzminger, K. C., Pai, J. C. & Maynard, J. A. (2011). Crystallization chaperone strategies for membrane proteins. Methods, 55, 293-302.]).

6.5. Reductive alkylation

Modification of the protein surface is a well established strategy for enhancing protein crystallization and can be achieved using site-directed mutagenesis (see §[link]6.6) or chemical modification (Derewenda, 2004[Derewenda, Z. S. (2004). Rational protein crystallization by mutational surface engineering. Structure, 12, 529-535.]). A common method of chemical modification is the reductive methylation of the -amino groups of solvent-exposed lysine residues. This is performed using the reducing agents dimethylamine–borane and formaldehyde (Means, 1977[Means, G. E. (1977). Reductive alkylation of amino groups. Methods Enzymol. 47, 469-478.]). In recent years, this technique has become one of the workhorse `salvage' techniques of structural genomics consortia (Tan et al., 2014[Tan, K. et al. (2014). Salvage of failed protein targets by reductive alkylation. Methods Mol. Biol. 1140, 189-200.]; Walter et al., 2006[Walter, T. S., Meier, C., Assenberg, R., Au, K. F., Ren, J., Verma, A., Nettleship, J. E., Owens, R. J., Stuart, D. I. & Grimes, J. M. (2006). Lysine methylation as a routine rescue strategy for protein crystallization. Structure, 14, 1617-1622.]; Sledz et al., 2010[Sledz, P., Zheng, H., Murzyn, K., Chruszcz, M., Zimmerman, M. D., Chordia, M. D., Joachimiak, A. & Minor, W. (2010). New surface contacts formed upon reductive lysine methylation: improving the probability of protein crystallization. Protein Sci. 19, 1395-1404.]). Reductive methylation is believed to function via the introduction of new surface contacts, therefore promoting crystal lattice formation (Sledz et al., 2010[Sledz, P., Zheng, H., Murzyn, K., Chruszcz, M., Zimmerman, M. D., Chordia, M. D., Joachimiak, A. & Minor, W. (2010). New surface contacts formed upon reductive lysine methylation: improving the probability of protein crystallization. Protein Sci. 19, 1395-1404.]).

Recent developments of the technique include the use of ethylation and isopropylation, although fewer targets are available to assess the performance of such techniques (Tan et al., 2014[Tan, K. et al. (2014). Salvage of failed protein targets by reductive alkylation. Methods Mol. Biol. 1140, 189-200.]). Another important development of the method is the use of cysteine alkylation for the structure determination of membrane proteins. This method was first used for the determination of the β1-adrenergic G-protein-coupled receptor structure (Warne et al., 2008[Warne, T., Serrano-Vega, M. J., Baker, J. G., Moukhametzianov, R., Edwards, P. C., Henderson, R., Leslie, A. G. W., Tate, C. G. & Schertler, G. F. X. (2008). Structure of a β1-adrenergic G-protein-coupled receptor. Nature (London), 454, 486-491.]) and is in common use for many GPCR studies (Columbus, 2015[Columbus, L. (2015). Post-expression strategies for structural investigations of membrane proteins. Curr. Opin. Struct. Biol. 32, 131-138.]). Cysteine alkylation stabilizes the protein by preventing the formation of disulfide bonds, and in the case of the β1-adrenergic receptor functions by stabilizing the monomers and preventing oligomers forming (Mathiasen et al., 2014[Mathiasen, S., Christensen, S. M., Fung, J. J., Rasmussen, S. G. F., Fay, J. F., Jorgensen, S. K., Veshaguri, S., Farrens, D. L., Kiskowski, M., Kobilka, B. & Stamou, D. (2014). Nanoscale high-content analysis using compositional heterogeneities of single proteoliposomes. Nature Methods, 11, 931-934.]).

In addition to alkylation, other chemical modifications such as fluorination can be carried out. Fluorine is all but absent from biological systems, but stabilizes proteins as a result of the `fluorous effect' (Buer & Marsh, 2014[Buer, B. C. & Marsh, E. N. (2014). Design, synthesis, and study of fluorinated proteins. Methods Mol. Biol. 1216, 89-116.]; Marsh, 2014[Marsh, E. N. (2014). Fluorinated proteins: from design and synthesis to structure and stability. Acc. Chem. Res. 47, 2878-2886.]). This effect results in an unusual propensity to undergo phase separation and causes an increase in the buried surface area in the hydrophobic core of fluorinated proteins. Fluorinated proteins can be generated using the highly fluorinated amino acid hexafluoroleucine. The crystal structure of a designed four-helical bundle protein, α4H, reveals that the fluorinated residues pack well into the hydrophobic core of the protein with little perturbation of the structure (Buer et al., 2012[Buer, B. C., Meagher, J. L., Stuckey, J. A. & Marsh, E. N. (2012). Structural basis for the enhanced stability of highly fluorinated proteins. Proc. Natl Acad. Sci. USA, 109, 4810-4815.]). This method would clearly perturb the structure and the function of some proteins, but may be a method worthy of further investigation for enabling structural studies of very unstable proteins.

6.6. Surface mutagenesis, surface-entropy reduction (SER) and deglycosylation

Mutagenesis of surface-exposed amino acids is a proven method for engineering proteins with improved stability and chance of crystallization (Derewenda, 2004[Derewenda, Z. S. (2004). Rational protein crystallization by mutational surface engineering. Structure, 12, 529-535.], 2010[Derewenda, Z. S. (2010). Application of protein engineering to enhance crystallizability and improve crystal properties. Acta Cryst. D66, 604-615.]; Derewenda & Vekilov, 2006[Derewenda, Z. S. & Vekilov, P. G. (2006). Entropy and surface engineering in protein crystallization. Acta Cryst. D62, 116-124.]). Amongst the first uses of mutagenesis to enhance crystallizability was the transplant of key crystal contacts from the rat ferritin structure onto the human ortholog, which was previously unsolved (Lawson et al., 1991[Lawson, D. M., Artymiuk, P. J., Yewdall, S. J., Smith, J. M., Livingstone, J. C., Treffry, A., Luzzago, A., Levi, S., Arosio, P., Cesareni, G., Thomas, C. D., Shaw, W. V. & Harrison, P. M. (1991). Solving the structure of human H ferritin by genetically engineering intermolecular crystal contacts. Nature (London), 349, 541-544.]). In this example, Lys86 of the human protein was mutated to Glu to mimic the Ca2+-binding site observed in the crystal contacts of the rat ortholog. Clearly, this technique is useful if an orthologous structure is available. However, this is often not the case and one is shooting in the dark when choosing surface residues to mutate.

To address this issue, Derewenda and coworkers made a series of mutations to RhoGDI targeting glutamate and lysine residues on the surface of the protein (Longenecker et al., 2001[Longenecker, K. L., Garrard, S. M., Sheffield, P. J. & Derewenda, Z. S. (2001). Protein crystallization by rational mutagenesis of surface residues: Lys to Ala mutations promote crystallization of RhoGDI. Acta Cryst. D57, 679-688.]; Mateja et al., 2002[Mateja, A., Devedjiev, Y., Krowarsch, D., Longenecker, K., Dauter, Z., Otlewski, J. & Derewenda, Z. S. (2002). The impact of Glu→Ala and Glu→Asp mutations on the crystallization properties of RhoGDI: the structure of RhoGDI at 1.3 Å resolution. Acta Cryst. D58, 1983-1991.]; Czepas et al., 2004[Czepas, J., Devedjiev, Y., Krowarsch, D., Derewenda, U., Otlewski, J. & Derewenda, Z. S. (2004). The impact of Lys→Arg surface mutations on the crystallization of the globular domain of RhoGDI. Acta Cryst. D60, 275-280.]). These residues have high conformational entropy and rarely participate in protein–protein interfaces within the crystal (Lo Conte et al., 1999[Conte, L. L., Chothia, C. & Janin, J. (1999). The atomic structure of protein-protein recognition sites. J. Mol. Biol. 285, 2177-2198.]; Baud & Karlin, 1999[Baud, F. & Karlin, S. (1999). Measures of residue density in protein structures. Proc. Natl Acad. Sci. USA, 96, 12494-12499.]). Therefore, they represent attractive targets for modification of crystal contacts. In an attempt to reduce the conformational entropy on the protein surface, these residues were mutated to either alanine, arginine or aspartate. The original set of mutable residues has more recently been expanded to include glutamine. The SERp server is an excellent resource for identifying such residues (Goldschmidt et al., 2007[Goldschmidt, L., Cooper, D. R., Derewenda, Z. S. & Eisenberg, D. (2007). Toward rational protein crystallization: a web server for the design of crystallizable protein variants. Protein Sci. 16, 1569-1576.], 2014[Goldschmidt, L., Eisenberg, D. & Derewenda, Z. S. (2014). Salvage or recovery of failed targets by mutagenesis to reduce surface entropy. Methods Mol. Biol. 1140, 201-209.]; https://bit.ly/1LFjdyk ). This site evaluates the solvent exposure, secondary structure, surface entropy and evolutionary conservation for sets of glutamate, glutamine or lysine residues. Analysis of the evolutionary conservation is used as a guide to avoid the mutation of residues that may be structurally or functionally significant. Finally, the site suggests appropriate clusters of residues matching these criteria as suitable for mutagenesis. These techniques have been used in conjunction with the fused affinity-tag and molecular-chaperone approach discussed above (see §[link]6.4) to determine the structure of a RACK1A-MBP fusion protein (Moon et al., 2010[Moon, A. F., Mueller, G. A., Zhong, X. & Pedersen, L. C. (2010). A synergistic approach to protein crystallization: combination of a fixed-arm carrier with surface entropy reduction. Protein Sci. 19, 901-913.]).

Finally, the presence of PTMs (see §[link]3.1) on the surface of the protein, in particular glycosylation, must be carefully considered. N- and O-linked sugars occur frequently on the surface of eukaryotic proteins and their chemical heterogeneity and conformational freedom can result in significant conformational variability. Strategies to deal with glycosylation on the protein surface include expression using a deglycosylation-deficient CHO cell strains such as lec1 (Puthalakath et al., 1996[Puthalakath, H., Burke, J. & Gleeson, P. A. (1996). Glycosylation defect in Lec1 Chinese hamster ovary mutant is due to a point mutation in N-acetylglucosaminyltransferase I gene. J. Biol. Chem. 271, 27818-27822.]) or an HEK293S strain such as GnTI (Reeves et al., 2002[Reeves, P. J., Callewaert, N., Contreras, R. & Khorana, H. G. (2002). Structure and function in rhodopsin: high-level expression of rhodopsin with restricted and homogeneous N-glycosylation by a tetracycline-inducible N-acetylglucosaminyltransferase I-negative HEK293S stable mammalian cell line. Proc. Natl Acad. Sci. USA, 99, 13419-13424.]). Another strategy is the mutagenesis of consensus glycosylation sites, such as the Asn-X-Ser/Thr sequon, which is targeted for N-linked glycosylation (Mellquist et al., 1998[Mellquist, J. L., Kasturi, L., Spitalnik, S. L. & Shakin-Eshleman, S. H. (1998). The amino acid following an Asn-X-Ser/Thr sequon is an important determinant of N-linked core glycosylation efficiency. Biochemistry, 37, 6833-6837.]), and Ser or Thr residues, which are targeted for O-linked glycosyl­ation. Alternatively, endoglycosylases such as endoglycosidase H (Endo H) and PNGase F can be used. In the case of PNGase F the glycosylated asparagine residue is converted to an aspartic acid, thus removing the sugar completely. Another approach is the use of N-glycosylation inhibitors such as swainsonine and kifunensine (Elbein, 1987[Elbein, A. D. (1987). Inhibitors of the biosynthesis and processing of N-linked oligosaccharide chains. Annu. Rev. Biochem. 56, 497-534.]). An effective combination approach uses glycosylation inhibitors during expression, followed by treatment with Endo H. Such an approach has been used to generate diffraction-quality crystals of sRPTPμ and s19A (Chang et al., 2007[Chang, V. T., Crispin, M., Aricescu, A. R., Harvey, D. J., Nettleship, J. E., Fennelly, J. A., Yu, C., Boles, K. S., Evans, E. J., Stuart, D. I., Dwek, R. A., Jones, E. Y., Owens, R. J. & Davis, S. J. (2007). Glycoprotein structural genomics: solving the glycosylation problem. Structure, 15, 267-273.]). Also, prudent selection of the expression system can be deployed to vary the glycosylation patterns. For example, Sf insect cells generally produce simpler glycosylation patterns of the GlucNAcMan5 type and often glycosylate at fewer sites, whereas yeast cells can often hyperglycosylate proteins.

It must be noted that the presence of glycans on the protein surface can sometimes aid in crystallization by mediating important crystal contacts. For example, a complex of the densely glycosylated Hepatitis C virus E2 (HCV E2) glycoprotein bound to a broadly neutralizing antibody failed to crystallize using numerous deglycosylation strategies, including Endo H treatment or N-linked glycan-sequon mutagenesis. In this example, only the fully glycosylated HCV E2 protein crystallized, and the resulting structure revealed a key crystal contact mediated by an N-linked glycan interacting non­specifically with a neighboring symmetry mate within the crystal (Kong et al., 2013[Kong, L., Giang, E., Nieusma, T., Kadam, R. U., Cogburn, K. E., Hua, Y., Dai, X., Stanfield, R. L., Burton, D. R., Ward, A. B., Wilson, I. A. & Law, M. (2013). Hepatitis C virus E2 envelope glycoprotein core structure. Science, 342, 1090-1094.]). Full or partial glycosylation may also be necessary to crystallize complexes involving glycan-dependent protein interactions (Kong et al., 2014[Kong, L., Stanfield, R. & Wilson, I. (2014). HIV Glycans in Infection and Immunity, edited by R. Pantophlet, pp. 117-141. New York: Springer.]). These examples illustrate that glycosylation may also have a stabilizing effect on proteins, especially for crystallization, and the presence of glycans is not always deleterious.

6.7. In situ proteolysis

The presence of proteases in a protein sample submitted for crystallization trials is clearly of significance and can lead to significant composition instability over the time course of the experiment. For example, it was serendipitously discovered that a penicillium fungus growing in a crystallization drop was responsible for cleaving ∼200 residues off the yeast CPSF-100 protein and was essential for successful crystallization (Mandel et al., 2006[Mandel, C. R., Gebauer, D., Zhang, H. & Tong, L. (2006). A serendipitous discovery that in situ proteolysis is essential for the crystallization of yeast CPSF-100 (Ydh1p). Acta Cryst. F62, 1041-1045.]; Bai et al., 2007[Bai, Y., Auperin, T. C. & Tong, L. (2007). The use of in situ proteolysis in the crystallization of murine CstF-77. Acta Cryst. F63, 135-138.]). This finding initiated so-called in situ proteolysis, in which protein samples are crystallized in the presence of a panel of proteases such as trypsin and chymotrypsin (Dong et al., 2007[Dong, A. et al. (2007). In situ proteolysis for protein crystallization and structure determination. Nature Methods, 4, 1019-1021.]; Wernimont & Edwards, 2009[Wernimont, A. & Edwards, A. (2009). In situ proteolysis to generate crystals for structure determination: an update. PLoS One, 4, e5094.]). More recently, these techniques have been combined with mass-spectrometric analysis to help identify peptide fragments that are stable over the time frame of a crystallization experiment (Gheyi et al., 2010[Gheyi, T., Rodgers, L., Romero, R., Sauder, J. M. & Burley, S. K. (2010). Mass spectrometry guided in situ proteolysis to obtain crystals for X-ray structure determination. J. Am. Soc. Mass Spectrom. 21, 1795-1801.]). In a similar fashion, limited proteolysis can be used to identify stable domains of membrane and globular proteins. The membrane protects the transmembrane portions of the protein from protease cleavage and more compact forms of the membrane protein can be produced. Such an approach was used to generate a stable 55 kDa construct of the P pilus PapC that was subsequently used for crystal structure determination (Remaut et al., 2008[Remaut, H., Tang, C., Henderson, N. S., Pinkner, J. S., Wang, T., Hultgren, S. J., Thanassi, D. G., Waksman, G. & Li, H. (2008). Fiber formation across the bacterial outer membrane by the chaperone/usher pathway. Cell, 133, 640-652.]).

6.8. Thermal stability screening

The thermal stability of a protein displays a good level of correlation with the crystallizability of a protein. For example, a large-scale Thermofluor study of 657 protein samples showed that only 23% of the proteins with a Tm of <43°C yielded crystals. In contrast, 49% of the proteins with a Tm of >45°C yielded crystals (Dupeux et al., 2011[Dupeux, F., Röwer, M., Seroul, G., Blot, D. & Márquez, J. A. (2011). A thermal stability assay can help to estimate the crystallization likelihood of biological samples. Acta Cryst. D67, 915-919.]; see §[link]5.4 and Fig. 4[link]). However, beyond 45°C the Tm does not appear to be particularly predictive of crystallizability; there is no significant increase in crystallization frequency for proteins with a Tm of between 45 and 96°C (Dupeux et al., 2011[Dupeux, F., Röwer, M., Seroul, G., Blot, D. & Márquez, J. A. (2011). A thermal stability assay can help to estimate the crystallization likelihood of biological samples. Acta Cryst. D67, 915-919.]). Furthermore, there are no known structural features that correlate well with Tm (Kumar et al., 2000[Kumar, S., Tsai, C.-J. & Nussinov, R. (2000). Factors enhancing protein thermostability. Protein Eng. Des. Sel. 13, 179-191.]), and the Tm appears to be highly solvent-dependent (Faria et al., 2004[Faria, T. Q., Lima, J. C., Bastos, M., Maçanita, A. L. & Santos, H. (2004). Protein stabilization by osmolytes from hyperthermophiles: effect of mannosylglycerate on the thermal unfolding of recombinant nuclease A from Staphylococcus aureus studied by picosecond time-resolved fluorescence and calorimetry. J. Biol. Chem. 279, 48680-48691.]). It is important to note that other features of the melting curve, aside from the Tm, may be useful to the crystallographer. For example, a high initial signal generated by a Thermofluor assay may indicate that an exposed hydrophobic surface of the native protein may be interacting with the dye used in the assay. Proteins displaying such profiles appear to have a lower rate of crystallization (36.6%; Dupeux et al., 2011[Dupeux, F., Röwer, M., Seroul, G., Blot, D. & Márquez, J. A. (2011). A thermal stability assay can help to estimate the crystallization likelihood of biological samples. Acta Cryst. D67, 915-919.]). Similarly, a wide thermal denaturation peak can be indicative of noncooperative unfolding behavior, which might be a useful consideration for construct modification (Morar-Mitrica et al., 2013[Morar-Mitrica, S., Nesta, D. & Crotts, G. (2013). Differential scanning calorimetry (DSC) for biopharmaceutical development: old concepts, new applications. BioPharma Asia, 2(4), 44-55.]). Ultimately, measures of thermal stability may be useful as a broad, qualitative assessment of the suitability of a protein for crystallization.

[Figure 4]
Figure 4
Thermofluor assay of protein melting temperature in the presence of stabilizing ligands. (a) Example melting curves for a protein of unknown function from Eubacterium siraeum (ZP_02421384.1) in the presence of various adenosine- and ribose-containing ligands (grey), adenosine diphosphate (ADP, green), adenosine triphosphate (ATP, red) and control sample with no ligand (blue dashed line). ADP and ATP result in a shift in melting temperature (ΔTm) of 3 and 8°C, respectively. (b) Matrix of 72 proteins of unknown function screened in a Thermofluor assay against a panel of 327 ligands. The ΔTm is indicated by the size and color of the data points ranging from 10 to 70°C. Data kindly provided by Anna Grezchnik of the Joint Center for Structural Genomics (JCSG).

6.9. Deuterium-exchange mass spectrometry (DXMS)

Prior to crystallization, it is often desirable to remove protein termini and low-complexity regions owing to their inherent flexibility (Derewenda, 2010[Derewenda, Z. S. (2010). Application of protein engineering to enhance crystallizability and improve crystal properties. Acta Cryst. D66, 604-615.]). Disordered regions of the protein often hamper crystallization and the design of protein constructs trimmed of such regions can be guided using deuterium-exchange mass spectrometry (DXMS; Englander, 2006[Englander, S. W. (2006). Hydrogen exchange and mass spectrometry: a historical perspective. J. Am. Soc. Mass Spectrom. 17, 1481-1489.]; Konermann et al., 2011[Konermann, L., Pan, J. & Liu, Y.-H. (2011). Hydrogen exchange mass spectrometry for studying protein structure and dynamics. Chem. Soc. Rev. 40, 1224-1234.]; Spraggon et al., 2004[Spraggon, G., Pantazatos, D., Klock, H. E., Wilson, I. A., Woods, V. L. Jr & Lesley, S. A. (2004). On the use of DXMS to produce more crystallizable proteins: structures of the T. maritima proteins TM0160 and TM1171. Protein Sci. 13, 3187-3199.]; Figs. 5[link]a and 5[link]b).

[Figure 5]
Figure 5
Deuterium-exchange mass spectrometry (DXMS) analysis. DXMS was used to guide the construct design used for determining the crystal structure of a putative ethanolamine-utilization protein from Salmonella typhimurium. (a) The left side of the figure shows that the N-terminal portion of the protein is more disordered, or unstructured in solution, as the backbone amide protons are more susceptible to exchange. Deuterium-labelled proteolytic fragments are highlighted in red. (b) The right side of the figure shows that the C-terminal portion of the protein is more ordered, or structured in solution, as the backbone-amide protons are less susceptible to exchange. The region selected for truncation is denoted by a blue arrow. (c) Ribbon diagram of the final crystal structure determined for residues 98–229 showing the compact and ordered structure (loops are shown in green, α-helices in red and β-­strands in yellow; PDB entry 2pyt ; Joint Center for Structural Genomics, unpublished work). (d) The same structure colored according to the B-factor value, highlighting the stable core of the protein in blue and the more flexible outer regions in green through red. Data kindly provided by Scott Lesley of the Joint Center for Structural Genomics (JCSG).

Using this technique, proteins are exposed to deuterated solvent and the deuterium is allowed to exchange with the backbone amide protons of the protein. The exchange rates are dependent on the degree of solvent exposure and the integrity of the local secondary structure. Therefore, this method gives a measure of the conformational flexibility of the protein in solution (Englander & Kallenbach, 1983[Englander, S. W. & Kallenbach, N. R. (1983). Hydrogen exchange and structural dynamics of proteins and nucleic acids. Q. Rev. Biophys. 16, 521-655.]). These rates can be obtained by measuring the change in the mass of the deuterated peptides using proteolytic mass spectrometry. DXMS has been successfully applied to structural genomics targets, guiding the deletion of flexible regions that previously hindered the growth of diffraction-quality crystals (Spraggon et al., 2004[Spraggon, G., Pantazatos, D., Klock, H. E., Wilson, I. A., Woods, V. L. Jr & Lesley, S. A. (2004). On the use of DXMS to produce more crystallizable proteins: structures of the T. maritima proteins TM0160 and TM1171. Protein Sci. 13, 3187-3199.]; Pantazatos et al., 2004[Pantazatos, D., Kim, J. S., Klock, H. E., Stevens, R. C., Wilson, I. A., Lesley, S. A. & Woods, V. L. Jr (2004). Rapid refinement of crystallographic protein construct definition employing enhanced hydrogen/deuterium exchange MS. Proc. Natl Acad. Sci. USA, 101, 751-756.]; Fig. 5). Although the utility of DXMS is often limited by the lengths of the proteolytic fragments that can be produced, sometimes restricting the area of investigation to less than 50% of the protein sequence, it can often provide a key insight for the design of crystallizable constructs. Furthermore, when coupled with structure, DXMS can be mapped onto the protein structure to provide a three-dimensional map of flexibility that can further guide protein-engineering efforts and mechanistic studies (Guttman et al., 2012[Guttman, M., Kahn, M., Garcia, N. K., Hu, S.-L. & Lee, K. K. (2012). Solution structure, conformational dynamics, and CD4-induced activation in full-length, glycosylated, monomeric HIV gp120. J. Virol. 86, 8750-8764.]; Kong et al., 2010[Kong, L., Huang, C.-C., Coales, S. J., Molnar, K. S., Skinner, J., Hamuro, Y. & Kwong, P. D. (2010). Local conformational stability of HIV-1 gp120 in unliganded and CD4-bound states as defined by amide hydrogen/deuterium exchange. J. Virol. 84, 10311-10321.]; Zhang et al., 2012[Zhang, A. P., Bornholdt, Z. A., Liu, T., Abelson, D. M., Lee, D. E., Li, S., Woods, V. L. Jr & Saphire, E. O. (2012). The ebola virus interferon antagonist VP24 directly binds STAT1 and has a novel, pyramidal fold. PLoS Pathog. 8, e1002550.]; Chung et al., 2011[Chung, K. Y., Rasmussen, S. G. F., Liu, T., Li, S., DeVree, B. T., Chae, P. S., Calinski, D., Kobilka, B. K., Woods, V. L. Jr & Sunahara, R. K. (2011). Conformational changes in the G protein Gs induced by the β2 adrenergic receptor. Nature (London), 477, 611-615.]).

6.10. Disulfide engineering

Disulfide engineering is a well established method for stabilizing proteins and for studying and modifying protein function and dynamics (Dombkowski et al., 2014[Dombkowski, A. A., Sultana, K. Z. & Craig, D. B. (2014). Protein disulfide engineering. FEBS Lett. 588, 206-212.]). Intermolecular disulfide bonds between proteins in the crystal lattice have been observed and have led to the coining of the phrase `spontaneously polymerizing protein crystals' (Quistgaard, 2014[Quistgaard, E. M. (2014). A disulfide polymerized protein crystal. Chem. Commun. 50, 14995-14997.]). Disulfide engineering can be used to introduce these intermolecular disulfides into the protein in an attempt to stabilize the lattice and promote crystallization. Furthermore, studies have shown that symmetrical proteins, such as homodimers, tend to crystallize more readily (Wukovitz & Yeates, 1995[Wukovitz, S. W. & Yeates, T. O. (1995). Why protein crystals favour some space groups over others. Nature Struct. Mol. Biol. 2, 1062-1067.]). Using T4 lysozyme as a model system, it has been demonstrated that the introduction of disulfide bonds can be used to make monomeric proteins dimerize and increasing the chance of lattice formation (Banatao et al., 2006[Banatao, D. R., Cascio, D., Crowley, C. S., Fleissner, M. R., Tienson, H. L. & Yeates, T. O. (2006). An approach to crystallizing proteins by synthetic symmetrization. Proc. Natl Acad. Sci. USA, 103, 16230-16235.]; Heinz & Matthews, 1994[Heinz, D. W. & Matthews, B. W. (1994). Rapid crystallization of T4 lysozyme by intermolecular disulfide cross-linking. Protein Eng. Des. Sel. 7, 301-307.]). This has been termed `synthetic symmetrization' and can be a useful tool for assisting in the crystallization of monomeric proteins and protein–protein complexes which display asymmetry. Proteins that have been successfully crystallized using this approach include CelA from Thermotoga maritima (Forse et al., 2011[Forse, G. J., Ram, N., Banatao, D. R., Cascio, D., Sawaya, M. R., Klock, H. E., Lesley, S. A. & Yeates, T. O. (2011). Synthetic symmetrization in the crystallization and structure determination of CelA from Thermotoga maritima. Protein Sci. 20, 168-178.]). Tools for identifying residues suitable for disulfide engineering are available, including Disulfide by Design 2.0 (Craig & Dombkowski, 2013[Craig, D. B. & Dombkowski, A. A. (2013). Disulfide by Design 2.0: a web-based tool for disulfide engineering in proteins. BMC Bioinformatics, 14, 346.]; https://bit.ly/1NbV2tQ ). One caveat of this approach is the potential of disulfide bonds to adopt different conformations that may promote conformational flexibility. For example, the disulfide bond connecting the I-EGF1 and I-EGF2 domains of β2 integrin is able to accommodate a >20 Å hinge motion between the domains (Shi et al., 2007[Shi, M., Foo, S. Y., Tan, S.-M., Mitchell, E. P., Law, S. K. A. & Lescar, J. (2007). A structural hypothesis for the transition between bent and extended conformations of the leukocyte β2 integrins. J. Biol. Chem. 282, 30198-30206.]; Smagghe et al., 2010[Smagghe, B. J., Huang, P.-S., Ban, Y.-E. A., Baker, D. & Springer, T. A. (2010). Modulation of integrin activation by an entropic spring in the β-knee. J. Biol. Chem. 285, 32954-32966.]).

7. Future outlook

Protein stability does mean many different things to many different scientists. However, on a global level, it can be considered as the ability of a protein to maintain structure and function in a particular environment. If the environment of interest is normal physiological conditions then the net summation of all contributing forces must add up to provide a small negative ΔG, therefore favoring a stable folded protein. However, not all proteins operate in standard physiological environments and many other factors must be considered. For example, many proteins, such as FeFe hydrogenase, are sensitive to oxygen and changes to the structure must be considered under an oxygen-free environment (Mulder et al., 2011[Mulder, D. W., Shepard, E. M., Meuser, J. E., Joshi, N., King, P. W., Posewitz, M. C., Broderick, J. B. & Peters, J. W. (2011). Insights into [FeFe]-hydrogenase structure, mechanism, and maturation. Structure, 19, 1038-1052.]). However, the growth of such enzymes under anaerobic conditions is both costly and difficult to achieve on a large scale, and considerable effort is being made to generate oxygen-tolerant hydrogenases for use on an industrial scale (Fritsch et al., 2013[Fritsch, J., Lenz, O. & Friedrich, B. (2013). Structure, function and biosynthesis of O2-tolerant hydrogenases. Nature Rev. Microbiol. 11, 106-114.]). The stability of such enzymes is of great interest to the biofuels industry, with the potential for biological hydrogen-gas production (Kim & Kim, 2011[Kim, D.-H. & Kim, M.-S. (2011). Hydrogenases for biological hydrogen production. Bioresour. Technol. 102, 8423-8431.]). Similarly, light-sensitive proteins such as opsins, photolyase and photosystems I and II all have unique structural features that enable them to utilize the energy of photons to carry out biological functions. Other interesting examples include the light-, oxygen- and voltage-sensitive domains (LOVs) found in plants and algae that undergo conformational changes and covalent binding of an FMN moiety under illumination with blue light (Briggs, 2007[Briggs, W. R. (2007). The LOV domain: a chromophore module servicing multiple photoreceptors. J. Biomed. Sci. 14, 499-504.]; Kottke et al., 2006[Kottke, T., Hegemann, P., Dick, B. & Heberle, J. (2006). The photochemistry of the light-, oxygen-, and voltage-sensitive domains in the algal blue light receptor phot. Biopolymers, 82, 373-378.]). These examples illustrate how nonstandard environments must be considered in any discussion of protein stability.

Traditionally, discussions of protein stability have focused on soluble, folded and more classical globular proteins. However, large percentages of the proteome are predicted to contain unstructured, insoluble and aggregated proteins in the form of IDPs, IIPs and APRs (see §[link]2.2). Such proteins, and their unwieldy conformational stability, represent a challenge for protein crystallographers, who usually simply remove such regions during the cloning stage. However, these families of proteins are of tremendous medical importance and deletion of these regions at the cloning stage is no longer meaningful. Crystallographic techniques are evolving to help to deal with such proteins, particularly in the areas of microcrystallo­graphy, next-generation synchrotron sources and X-ray free-electron lasers (XFELs) (Neutze & Moffat, 2012[Neutze, R. & Moffat, K. (2012). Time-resolved structural studies at synchrotrons and X-ray free electron lasers: opportunities and challenges. Curr. Opin. Struct. Biol. 22, 651-659.]; Spence et al., 2012[Spence, J. C., Weierstall, U. & Chapman, H. N. (2012). X-ray lasers for structural and dynamic biology. Rep. Prog. Phys. 75, 102601.]; Gruner & Lattman, 2015[Gruner, S. M. & Lattman, E. E. (2015). Biostructural science inspired by next-generation X-ray sources. Annu. Rev. Biophys. 44, 33-51.]; Weckert, 2015[Weckert, E. (2015). The potential of future light sources to explore the structure and function of matter. IUCrJ, 2, 230-245.]; Moukhametzianov et al., 2008[Moukhametzianov, R., Burghammer, M., Edwards, P. C., Petitdemange, S., Popov, D., Fransen, M., McMullan, G., Schertler, G. F. X. & Riekel, C. (2008). Protein crystallography with a micrometre-sized synchrotron-radiation beam. Acta Cryst. D64, 158-166.]; Igarashi et al., 2008[Igarashi, N., Ikuta, K., Miyoshi, T., Matsugaki, N., Yamada, Y., Yousef, M. S. & Wakatsuki, S. (2008). X-ray beam stabilization at BL-17A, the protein microcrystallography beamline of the Photon Factory. J. Synchrotron Rad. 15, 292-295.]; Fischetti et al., 2009[Fischetti, R. F., Xu, S., Yoder, D. W., Becker, M., Nagarajan, V., Sanishvili, R., Hilgart, M. C., Stepanov, S., Makarov, O. & Smith, J. L. (2009). Mini-beam collimator enables microcrystallography experiments on standard beamlines. J. Synchrotron Rad. 16, 217-225.]).

Recent developments in XFEL light sources are enabling higher resolution studies of protein dynamics on a femto­second timescale (Uervirojnangkoorn et al., 2015[Uervirojnangkoorn, M., Zeldin, O. B., Lyubimov, A. Y., Hattne, J., Brewster, A. S., Sauter, N. K., Brunger, A. T. & Weis, W. I. (2015). Enabling X-ray free electron laser crystallography for challenging biological systems from a limited number of crystals. Elife, 4, e05421.]). From a crystallization perspective, such light sources present a unique set of challenges to the crystallographer that are somewhat orthogonal to the traditional problems faced when using more traditional diffraction methods. One such requirement is the need for microcrystalline sample material that can be injected into the laser path. Additionally, such techniques also allow larger and more dynamic protein structures to be determined, particularly those which possess conformational disorder and thus hinder the formation of larger, more ordered crystals. Recent examples using XFEL techniques include structures of the complex between synaptotagmin 1 and the neuronal SNARE (Zhou et al., 2015[Zhou, Q. et al. (2015). Architecture of the synaptotagmin-SNARE machinery for neuronal exocytosis. Nature (London), 525, 62-67.]). Clearly, XFEL technology, especially when coupled with associated hybrid methods, such as cryo-EM, ultrafast electron diffraction (UED) and double electron–electron resonance (DEER), will help mitigate many of the problems associated with protein conformation stability and its effect on protein crystallization (Wakatsuki, 2016[Wakatsuki, S. (2016). In Synchrotron Light Sources and Free-Electron Lasers, edited by E. Jaeschke, S. Khan, J. R. Schneider & J. B. Hastings. New York: Springer. In the press.]). Next-generation synchrotron sources will be capable of providing smaller and brighter X-ray beams, and most importantly with a higher coherence fraction (Weckert, 2015[Weckert, E. (2015). The potential of future light sources to explore the structure and function of matter. IUCrJ, 2, 230-245.]). Such coherent beams will provide exciting opportunities for the study of protein dynamics on ever smaller timescales and will be essential for the study of unstable proteins that are currently inaccessible using traditional crystallographic techniques.

In summary, we will end this review with the title of an excellent paper by the late Fred Richards: Protein stability: still an unsolved problem (Richards, 1997[Richards, F. M. (1997). Protein stability: still an unsolved problem. Cell. Mol. Life Sci. 53, 790-802.]). Although the problem is still largely unsolved, considerable progress has been made towards the study of protein stability, disorder and dynamics. Structural methods such as crystallography, NMR and cryo-EM are central to this endeavor and the exploration of innovative hybrid methods will be vital.

Acknowledgements

This work was sponsored in part by contributions from k.-k. Hofkristallamt, Vista, California, USA and Austrian Science Fund (FWF) project P28395-B26. We would like to thank members of the Joint Center for Structural Genomics for fruitful discussions and contribution of the Thermofluor and DXMS data.

References

First citationAltmann, F., Staudacher, E., Wilson, I. B. & März, L. (1999). Insect cells as hosts for the expression of recombinant glycoproteins. Glycoconj. J. 16, 109–123.  CrossRef PubMed CAS Google Scholar
First citationAlushin, G. M., Lander, G. C., Kellogg, E. H., Zhang, R., Baker, D. & Nogales, E. (2014). High-resolution microtubule structures reveal the structural transitions in αβ-tubulin upon GTP hydrolysis. Cell, 157, 1117–1129.  CrossRef CAS PubMed Google Scholar
First citationAmarasinghe, C. & Jin, J.-P. (2015). The use of affinity tags to overcome obstacles in recombinant protein expression and purification. Protein Pept. Lett. 22, 885–892.  CrossRef CAS PubMed Google Scholar
First citationAnand, K., Pal, D. & Hilgenfeld, R. (2002). An overview on 2-methyl-2,4-pentanediol in crystallization and in crystals of biological macromolecules. Acta Cryst. D58, 1722–1728.  Web of Science CrossRef CAS IUCr Journals Google Scholar
First citationAnfinsen, C. B. (1973). Principles that govern the folding of protein chains. Science, 181, 223–230.  CrossRef CAS PubMed Web of Science Google Scholar
First citationAranko, A. S., Wlodawer, A. & Iwai, H. (2014). Nature's recipe for splitting inteins. Protein Eng. Des. Sel. 27, 263–271.  CrossRef CAS PubMed Google Scholar
First citationBachmair, A., Finley, D. & Varshavsky, A. (1986). In vivo half-life of a protein is a function of its amino-terminal residue. Science, 234, 179–186.  CrossRef CAS PubMed Web of Science Google Scholar
First citationBai, Y., Auperin, T. C. & Tong, L. (2007). The use of in situ proteolysis in the crystallization of murine CstF-77. Acta Cryst. F63, 135–138.  Web of Science CrossRef CAS IUCr Journals Google Scholar
First citationBanatao, D. R., Cascio, D., Crowley, C. S., Fleissner, M. R., Tienson, H. L. & Yeates, T. O. (2006). An approach to crystallizing proteins by synthetic symmetrization. Proc. Natl Acad. Sci. USA, 103, 16230–16235.  Web of Science CrossRef PubMed CAS Google Scholar
First citationBarth, A. (2007). Infrared spectroscopy of proteins. Biochim. Biophys. Acta, 1767, 1073–1101.  CrossRef PubMed CAS Google Scholar
First citationBaud, F. & Karlin, S. (1999). Measures of residue density in protein structures. Proc. Natl Acad. Sci. USA, 96, 12494–12499.  Web of Science CrossRef PubMed CAS Google Scholar
First citationBieri, M., Kwan, A. H., Mobli, M., King, G. F., Mackay, J. P. & Gooley, P. R. (2011). Macromolecular NMR spectroscopy for the non-spectroscopist: beyond macromolecular solution structure determination. FEBS J. 278, 704–715.  CrossRef CAS PubMed Google Scholar
First citationBoivin, S., Kozak, S. & Meijers, R. (2013). Optimization of protein purification and characterization using Thermofluor screens. Protein Expr. Purif. 91, 192–206.  Web of Science CrossRef CAS PubMed Google Scholar
First citationBowie, J. U. (2001). Stabilizing membrane proteins. Curr. Opin. Struct. Biol. 11, 397–402.  Web of Science CrossRef PubMed CAS Google Scholar
First citationBriggs, W. R. (2007). The LOV domain: a chromophore module servicing multiple photoreceptors. J. Biomed. Sci. 14, 499–504.  CrossRef PubMed CAS Google Scholar
First citationBrondyk, W. H. (2009). Selecting an appropriate method for expressing a recombinant protein. Methods Enzymol. 463, 131–147.  CrossRef PubMed CAS Google Scholar
First citationBrunori, M. (2014). Variations on the theme: allosteric control in hemoglobin. FEBS J. 281, 633–643.  CrossRef CAS PubMed Google Scholar
First citationBrutscher, B., Felli, I. C., Gil-Caballero, S., Hošek, T., Kümmerle, R., Piai, A., Pierattelli, R. & Sólyom, Z. (2015). NMR methods for the study of instrinsically disordered proteins structure, dynamics, and interactions: general overview and practical guidelines. Adv. Exp. Med. Biol. 870, 49–122.  CrossRef PubMed Google Scholar
First citationBruylants, G., Wouters, J. & Michaux, C. (2005). Differential scanning calorimetry in life science: thermodynamics, stability, molecular recognition and application in drug design. Curr. Med. Chem. 12, 2011–2020.  Web of Science CrossRef PubMed CAS Google Scholar
First citationBryan, P. N. & Orban, J. (2010). Proteins that switch folds. Curr. Opin. Struct. Biol. 20, 482–488.  CrossRef CAS PubMed Google Scholar
First citationBucher, M. H., Evdokimov, A. G. & Waugh, D. S. (2002). Differential effects of short affinity tags on the crystallization of Pyrococcus furiosus maltodextrin-binding protein. Acta Cryst. D58, 392–397.  Web of Science CrossRef CAS IUCr Journals Google Scholar
First citationBuer, B. C. & Marsh, E. N. (2014). Design, synthesis, and study of fluorinated proteins. Methods Mol. Biol. 1216, 89–116.  CrossRef CAS PubMed Google Scholar
First citationBuer, B. C., Meagher, J. L., Stuckey, J. A. & Marsh, E. N. (2012). Structural basis for the enhanced stability of highly fluorinated proteins. Proc. Natl Acad. Sci. USA, 109, 4810–4815.  CrossRef CAS PubMed Google Scholar
First citationBukowska, M. A. & Grütter, M. G. (2013). New concepts and aids to facilitate crystallization. Curr. Opin. Struct. Biol. 23, 409–416.  Web of Science CrossRef CAS PubMed Google Scholar
First citationBulawa, C. E., Connelly, S., Devit, M., Wang, L., Weigel, C., Fleming, J. A., Packman, J., Powers, E. T., Wiseman, R. L., Foss, T. R., Wilson, I. A., Kelly, J. W. & Labaudinière, R. (2012). Tafamidis, a potent and selective transthyretin kinetic stabilizer that inhibits the amyloid cascade. Proc. Natl Acad. Sci. 109, 9629–9634.  CrossRef CAS PubMed Google Scholar
First citationCalderone, T. L., Stevens, R. D. & Oas, T. G. (1996). High-level misincorporation of lysine for arginine at AGA codons in a fusion protein expressed in Escherichia coli. J. Mol. Biol. 262, 407–412.  CrossRef CAS PubMed Google Scholar
First citationCarver, T. E. et al. (2005). Decrypting the biochemical function of an essential gene from Streptococcus pneumoniae using Thermofluor technology. J. Biol. Chem. 280, 11704–11712.  Web of Science CrossRef PubMed CAS Google Scholar
First citationChang, V. T., Crispin, M., Aricescu, A. R., Harvey, D. J., Nettleship, J. E., Fennelly, J. A., Yu, C., Boles, K. S., Evans, E. J., Stuart, D. I., Dwek, R. A., Jones, E. Y., Owens, R. J. & Davis, S. J. (2007). Glycoprotein structural genomics: solving the glycosylation problem. Structure, 15, 267–273.  Web of Science CrossRef PubMed CAS Google Scholar
First citationCharoenkwan, P., Shoombuatong, W., Lee, H.-C., Chaijaruwanich, J., Huang, H.-L. & Ho, S.-Y. (2013). SCMCRYS: predicting protein crystallization using an ensemble scoring card method with estimating propensity scores of P-collocated amino acid pairs. PLoS One, 8, e72368.  Web of Science CrossRef PubMed Google Scholar
First citationChatterjee, S. & Mayor, S. (2001). The GPI-anchor and protein sorting. Cell. Mol. Life Sci. 58, 1969–1987.  CrossRef PubMed CAS Google Scholar
First citationChong, S., Mersha, F. B., Comb, D. G., Scott, M. E., Landry, D., Vence, L. M., Perler, F. B., Benner, J., Kucera, R. B., Hirvonen, C. A., Pelletier, J. J., Paulus, H. & Xu, M.-Q. (1997). Single-column purification of free recombinant proteins using a self-cleavable affinity tag derived from a protein splicing element. Gene, 192, 271–281.  CrossRef CAS PubMed Web of Science Google Scholar
First citationChou, P. Y. & Fasman, G. D. (1974). Conformational parameters for amino acids in helical, β-sheet, and random coil regions calculated from proteins. Biochemistry, 13, 211–222.  CrossRef CAS PubMed Web of Science Google Scholar
First citationChung, C. (2007). The use of biophysical methods increases success in obtaining liganded crystal structures. Acta Cryst. D63, 62–71.  CrossRef IUCr Journals Google Scholar
First citationChung, K. Y., Rasmussen, S. G. F., Liu, T., Li, S., DeVree, B. T., Chae, P. S., Calinski, D., Kobilka, B. K., Woods, V. L. Jr & Sunahara, R. K. (2011). Conformational changes in the G protein Gs induced by the β2 adrenergic receptor. Nature (London), 477, 611–615.  CrossRef CAS PubMed Google Scholar
First citationClifton, M. C. et al. (2015). A maltose-binding protein fusion construct yields a robust crystallography platform for MCL1. PLoS One, 10, e0125010.  CrossRef PubMed Google Scholar
First citationColumbus, L. (2015). Post-expression strategies for structural investigations of membrane proteins. Curr. Opin. Struct. Biol. 32, 131–138.  CrossRef CAS PubMed Google Scholar
First citationCompiani, M. & Capriotti, E. (2013). Computational and theoretical methods for protein folding. Biochemistry, 52, 8601–8624.  CrossRef CAS PubMed Google Scholar
First citationConte, L. L., Chothia, C. & Janin, J. (1999). The atomic structure of protein–protein recognition sites. J. Mol. Biol. 285, 2177–2198.  CrossRef CAS PubMed Google Scholar
First citationCooper, D. R., Boczek, T., Grelewska, K., Pinkowska, M., Sikorska, M., Zawadzki, M. & Derewenda, Z. (2007). Protein crystallization by surface entropy reduction: optimization of the SER strategy. Acta Cryst. D63, 636–645.  Web of Science CrossRef CAS IUCr Journals Google Scholar
First citationCraig, D. B. & Dombkowski, A. A. (2013). Disulfide by Design 2.0: a web-based tool for disulfide engineering in proteins. BMC Bioinformatics, 14, 346.  Google Scholar
First citationCzepas, J., Devedjiev, Y., Krowarsch, D., Derewenda, U., Otlewski, J. & Derewenda, Z. S. (2004). The impact of Lys→Arg surface mutations on the crystallization of the globular domain of RhoGDI. Acta Cryst. D60, 275–280.  Web of Science CrossRef CAS IUCr Journals Google Scholar
First citationDaniel, E., Onwukwe, G. U., Wierenga, R. K., Quaggin, S. E., Vainio, S. J. & Krause, M. (2015). ATGme: open-source web application for rare codon identification and custom DNA sequence optimization. BMC Bioinformatics, 16, 303.  Google Scholar
First citationDavies, D. R. (1964). A correlation between amino acid composition and protein structure. J. Mol. Biol. 9, 605–609.  CrossRef PubMed CAS Web of Science Google Scholar
First citationDavis-Searles, P. R., Saunders, A. J., Erie, D. A., Winzor, D. J. & Pielak, G. J. (2001). Interpreting the effects of small uncharged solutes on protein-folding equilibria. Annu. Rev. Biophys. Biomol. Struct. 30, 271–306.  PubMed CAS Google Scholar
First citationDe Baets, G., Schymkowitz, J. & Rousseau, F. (2014). Predicting aggregation-prone sequences in proteins. Essays Biochem. 56, 41–52.  CrossRef PubMed Google Scholar
First citationDeber, C. M., Khan, A. R., Li, Z., Joensson, C., Glibowicka, M. & Wang, J. (1993). Val→Ala mutations selectively alter helix–helix packing in the transmembrane segment of phage M13 coat protein. Proc. Natl Acad. Sci. USA, 90, 11648–11652.  CrossRef CAS PubMed Google Scholar
First citationDekker, K., Yamagata, H., Sakaguchi, K. & Udaka, S. (1991). Xylose (glucose) isomerase gene from the thermophile Thermus thermophilus: cloning, sequencing, and comparison with other thermostable xylose isomerases. J. Bacteriol. 173, 3078–3083.  CAS PubMed Google Scholar
First citationDemirdöven, N., Cheatum, C. M., Chung, H. S., Khalil, M., Knoester, J. & Tokmakoff, A. (2004). Two-dimensional infrared spectroscopy of antiparallel β-sheet secondary structure. J. Am. Chem. Soc. 126, 7981–7990.  PubMed Google Scholar
First citationDerewenda, Z. S. (2004). Rational protein crystallization by mutational surface engineering. Structure, 12, 529–535.  Web of Science CrossRef PubMed CAS Google Scholar
First citationDerewenda, Z. S. (2010). Application of protein engineering to enhance crystallizability and improve crystal properties. Acta Cryst. D66, 604–615.  Web of Science CrossRef CAS IUCr Journals Google Scholar
First citationDerewenda, Z. S. & Vekilov, P. G. (2006). Entropy and surface engineering in protein crystallization. Acta Cryst. D62, 116–124.  Web of Science CrossRef CAS IUCr Journals Google Scholar
First citationDieci, G., Bottarelli, L., Ballabeni, A. & Ottonello, S. (2000). tRNA-assisted overproduction of eukaryotic ribosomal proteins. Protein Expr. Purif. 18, 346–354.  CrossRef PubMed CAS Google Scholar
First citationDill, K. A. (1990). Dominant forces in protein folding. Biochemistry, 29, 7133–7155.  CrossRef CAS PubMed Web of Science Google Scholar
First citationDisfani, F. M., Hsu, W.-L., Mizianty, M. J., Oldfield, C. J., Xue, B., Dunker, A. K., Uversky, V. N. & Kurgan, L. (2012). MoRFpred, a computational tool for sequence-based prediction and characterization of short disorder-to-order transitioning binding regions in proteins. Bioinformatics, 28, i75–i83.  CrossRef CAS PubMed Google Scholar
First citationDombkowski, A. A., Sultana, K. Z. & Craig, D. B. (2014). Protein disulfide engineering. FEBS Lett. 588, 206–212.  CrossRef CAS PubMed Google Scholar
First citationDong, A. et al. (2007). In situ proteolysis for protein crystallization and structure determination. Nature Methods, 4, 1019–1021.  Web of Science CrossRef PubMed CAS Google Scholar
First citationDunker, A. K. & Oldfield, C. J. (2015). Back to the future: nuclear magnetic resonance and bioinformatics studies on intrinsically disordered proteins. Adv. Exp. Med. Biol. 870, 1–34.  CrossRef PubMed Google Scholar
First citationDupeux, F., Röwer, M., Seroul, G., Blot, D. & Márquez, J. A. (2011). A thermal stability assay can help to estimate the crystallization likelihood of biological samples. Acta Cryst. D67, 915–919.  Web of Science CrossRef CAS IUCr Journals Google Scholar
First citationElbein, A. D. (1987). Inhibitors of the biosynthesis and processing of N-linked oligosaccharide chains. Annu. Rev. Biochem. 56, 497–534.  CrossRef CAS PubMed Google Scholar
First citationEnglander, S. W. (2006). Hydrogen exchange and mass spectrometry: a historical perspective. J. Am. Soc. Mass Spectrom. 17, 1481–1489.  CrossRef PubMed CAS Google Scholar
First citationEnglander, S. W. & Kallenbach, N. R. (1983). Hydrogen exchange and structural dynamics of proteins and nucleic acids. Q. Rev. Biophys. 16, 521–655.  CrossRef CAS PubMed Google Scholar
First citationEricsson, U. B., Hallberg, B. M., DeTitta, G. T., Dekker, N. & Nordlund, P. (2006). Thermofluor-based high-throughput stability optimization of proteins for structural studies. Anal. Biochem. 357, 289–298.  Web of Science CrossRef PubMed CAS Google Scholar
First citationEvdokimov, A. G. et al. (2007). Serendipitous discovery of novel bacterial methionine aminopeptidase inhibitors. Proteins, 66, 538–546.  CrossRef PubMed CAS Google Scholar
First citationFaria, T. Q., Lima, J. C., Bastos, M., Maçanita, A. L. & Santos, H. (2004). Protein stabilization by osmolytes from hyperthermophiles: effect of mannosylglycerate on the thermal unfolding of recombinant nuclease A from Staphylococcus aureus studied by picosecond time-resolved fluorescence and calorimetry. J. Biol. Chem. 279, 48680–48691.  CrossRef PubMed CAS Google Scholar
First citationFields, P. A., Dong, Y., Meng, X. & Somero, G. N. (2015). Adaptations of protein structure and function to temperature: there is more than one way to `skin a cat'. J. Exp. Biol. 218, 1801–1811.  CrossRef PubMed Google Scholar
First citationFischer, S., Handrick, R. & Otte, K. (2015). The art of CHO cell engineering: a comprehensive retrospect and future perspectives. Biotechnol. Adv. 33, 1878–1896.  CrossRef CAS PubMed Google Scholar
First citationFischetti, R. F., Xu, S., Yoder, D. W., Becker, M., Nagarajan, V., Sanishvili, R., Hilgart, M. C., Stepanov, S., Makarov, O. & Smith, J. L. (2009). Mini-beam collimator enables microcrystallography experiments on standard beamlines. J. Synchrotron Rad. 16, 217–225.  Web of Science CrossRef CAS IUCr Journals Google Scholar
First citationForse, G. J., Ram, N., Banatao, D. R., Cascio, D., Sawaya, M. R., Klock, H. E., Lesley, S. A. & Yeates, T. O. (2011). Synthetic symmetrization in the crystallization and structure determination of CelA from Thermotoga maritima. Protein Sci. 20, 168–178.  Web of Science CrossRef CAS PubMed Google Scholar
First citationFritsch, J., Lenz, O. & Friedrich, B. (2013). Structure, function and biosynthesis of O2-tolerant hydrogenases. Nature Rev. Microbiol. 11, 106–114.  CrossRef CAS Google Scholar
First citationFu, W., Lin, J. & Cen, P. (2007). 5-Aminolevulinate production with recombinant Escherichia coli using a rare codon optimizer host strain. Appl. Microbiol. Biotechnol. 75, 777–782.  CrossRef PubMed CAS Google Scholar
First citationGheyi, T., Rodgers, L., Romero, R., Sauder, J. M. & Burley, S. K. (2010). Mass spectrometry guided in situ proteolysis to obtain crystals for X-ray structure determination. J. Am. Soc. Mass Spectrom. 21, 1795–1801.  Web of Science CrossRef CAS PubMed Google Scholar
First citationGiordanetto, F., Schäfer, A. & Ottmann, C. (2014). Stabilization of protein–protein interactions by small molecules. Drug Discov. Today, 19, 1812–1821.  CrossRef CAS PubMed Google Scholar
First citationGoldschmidt, L., Cooper, D. R., Derewenda, Z. S. & Eisenberg, D. (2007). Toward rational protein crystallization: a web server for the design of crystallizable protein variants. Protein Sci. 16, 1569–1576.  Web of Science CrossRef PubMed CAS Google Scholar
First citationGoldschmidt, L., Eisenberg, D. & Derewenda, Z. S. (2014). Salvage or recovery of failed targets by mutagenesis to reduce surface entropy. Methods Mol. Biol. 1140, 201–209.  CrossRef CAS PubMed Google Scholar
First citationGoyal, S., Qin, H., Lim, L. & Song, J. (2015). Insoluble protein characterization by circular dichroism (CD) spectroscopy and nuclear magnetic resonance (NMR). Methods Mol. Biol. 1258, 371–385.  CrossRef PubMed Google Scholar
First citationGräslund, S., Sagemark, J., Berglund, H., Dahlgren, L. G., Flores, A., Hammarström, M., Johansson, I., Kotenyova, T., Nilsson, M., Nordlund, P. & Weigelt, J. (2008). The use of systematic N- and C-terminal deletions to promote production and structural studies of recombinant proteins. Protein Expr. Purif. 58, 210–221.  Web of Science PubMed Google Scholar
First citationGruner, S. M. & Lattman, E. E. (2015). Biostructural science inspired by next-generation X-ray sources. Annu. Rev. Biophys. 44, 33–51.  Web of Science CrossRef CAS PubMed Google Scholar
First citationGuttman, M., Kahn, M., Garcia, N. K., Hu, S.-L. & Lee, K. K. (2012). Solution structure, conformational dynamics, and CD4-induced activation in full-length, glycosylated, monomeric HIV gp120. J. Virol. 86, 8750–8764.  CrossRef CAS PubMed Google Scholar
First citationGuzman, L. M., Belin, D., Carson, M. J. & Beckwith, J. (1995). Tight regulation, modulation, and high-level expression by vectors containing the arabinose PBAD promoter. J. Bacteriol. 177, 4121–4130.  CAS PubMed Web of Science Google Scholar
First citationGuzzo, A. V. (1965). The influence of amino acid sequence on protein structure. Biophys. J. 5, 809–822.  CrossRef CAS PubMed Google Scholar
First citationHall, M. P. (2014). Biotransformation and in vivo stability of protein biotherapeutics: impact on candidate selection and pharmacokinetic profiling. Drug Metab. Dispos. 42, 1873–1880.  CrossRef PubMed Google Scholar
First citationHassell, A. M. et al. (2007). Crystallization of protein–ligand complexes. Acta Cryst. D63, 72–79.  Web of Science CrossRef CAS IUCr Journals Google Scholar
First citationHay, R. T. (2005). SUMO. Mol. Cell, 18, 1–12.  CrossRef PubMed CAS Google Scholar
First citationHeinz, D. W. & Matthews, B. W. (1994). Rapid crystallization of T4 lysozyme by intermolecular disulfide cross-linking. Protein Eng. Des. Sel. 7, 301–307.  CrossRef CAS Web of Science Google Scholar
First citationHermans, J., Anderson, A. G. & Yun, R. H. (1992). Differential helix propensity of small apolar side chains studied by molecular dynamics simulations. Biochemistry, 31, 5646–5653.  CrossRef PubMed CAS Google Scholar
First citationHirata, R., Ohsumk, Y., Nakano, A., Kawasaki, H., Suzuki, K. & Anraku, Y. (1990). Molecular structure of a gene, VMA1, encoding the catalytic subunit of H+-translocating adenosine triphosphatase from vacuolar membranes of Saccharomyces cerevisiae. J. Biol. Chem. 265, 6726–6733.  CAS PubMed Google Scholar
First citationHuai, Q., Kim, H.-Y., Liu, Y., Zhao, Y., Mondragon, A., Liu, J. O. & Ke, H. (2002). Crystal structure of calcineurin–cyclophilin–cyclo­sporin shows common but distinct recognition of immunophilin–drug complexes. Proc. Natl Acad. Sci. USA, 99, 12037–12042.  Web of Science CrossRef PubMed CAS Google Scholar
First citationHuang, Y. J., Acton, T. B. & Montelione, G. T. (2014). DisMeta: a meta server for construct design and optimization. Methods Mol. Biol. 1091, 3–16.  CrossRef CAS PubMed Google Scholar
First citationIgarashi, N., Ikuta, K., Miyoshi, T., Matsugaki, N., Yamada, Y., Yousef, M. S. & Wakatsuki, S. (2008). X-ray beam stabilization at BL-17A, the protein microcrystallography beamline of the Photon Factory. J. Synchrotron Rad. 15, 292–295.  Web of Science CrossRef CAS IUCr Journals Google Scholar
First citationJahandideh, S., Jaroszewski, L. & Godzik, A. (2014). Improving the chances of successful protein structure determination with a random forest classifier. Acta Cryst. D70, 627–635.  Web of Science CrossRef IUCr Journals Google Scholar
First citationJahandideh, S. & Mahdavi, A. (2012). RFCRYS: sequence-based protein crystallization propensity prediction by means of random forest. J. Theor. Biol. 306, 115–119.  Web of Science CrossRef CAS PubMed Google Scholar
First citationJarvis, D. L. (2009). Baculovirus–insect cell expression systems. Methods Enzymol. 463, 191–222.  CrossRef PubMed CAS Google Scholar
First citationJiménez, M. A. (2014). Design of monomeric water-soluble β-hairpin and β-sheet peptides. Methods Mol. Biol. 1216, 15–52.  PubMed Google Scholar
First citationJulien, J.-P. et al. (2015). Design and structure of two HIV-1 clade C SOSIP.664 trimers that increase the arsenal of native-like Env immunogens. Proc. Natl Acad. Sci. USA, 112, 11947–11952.  CrossRef CAS PubMed Google Scholar
First citationKandaswamy, K. K., Pugalenthi, G., Suganthan, P. N. & Gangal, R. (2010). SVMCRYS: an SVM approach for the prediction of protein crystallization propensity from protein sequence. Protein Pept. Lett. 17, 423–430.  CrossRef CAS PubMed Google Scholar
First citationKane, J. F. (1995). Effects of rare codon clusters on high-level expression of heterologous proteins in Escherichia coli. Curr. Opin. Biotechnol. 6, 494–500.  CrossRef CAS PubMed Web of Science Google Scholar
First citationKapust, R. B. & Waugh, D. S. (1999). Escherichia coli maltose-binding protein is uncommonly effective at promoting the solubility of polypeptides to which it is fused. Protein Sci. 8, 1668–1674.  Web of Science CrossRef PubMed CAS Google Scholar
First citationKe, A. & Wolberger, C. (2003). Insights into binding cooperativity of MATa1/MATα2 from the crystal structure of a MATa1 homeodomain-maltose binding protein chimera. Protein Sci. 12, 306–312.  Web of Science CrossRef PubMed CAS Google Scholar
First citationKery, V., Elleder, D. & Kraus, J. P. (1995). δ-Aminolevulinate increases heme saturation and yield of human cystathionine β-synthase expressed in Escherichia coli. Arch. Biochem. Biophys. 316, 24–29.  CrossRef CAS PubMed Google Scholar
First citationKhoury, G. A., Baliban, R. C. & Floudas, C. A. (2011). Proteome-wide post-translational modification statistics: frequency analysis and curation of the Swiss-Prot database. Sci. Rep. 1, 90.  CrossRef Google Scholar
First citationKim, D.-H. & Kim, M.-S. (2011). Hydrogenases for biological hydrogen production. Bioresour. Technol. 102, 8423–8431.  CrossRef CAS PubMed Google Scholar
First citationKlock, H. E., Koesema, E. J., Knuth, M. W. & Lesley, S. A. (2008). Combining the polymerase incomplete primer extension method for cloning and mutagenesis with microscreening to accelerate structural genomics efforts. Proteins, 71, 982–994.  Web of Science CrossRef PubMed CAS Google Scholar
First citationKlock, H. E. & Lesley, S. A. (2009). The polymerase incomplete primer extension (PIPE) method applied to high-throughput cloning and site-directed mutagenesis. Methods Mol. Biol. 498, 91–103.  CrossRef PubMed CAS Google Scholar
First citationKobe, B., Center, R. J., Kemp, B. E. & Poumbourios, P. (1999). Crystal structure of human T cell leukemia virus type 1 gp21 ectodomain crystallized as a maltose-binding protein chimera reveals structural evolution of retroviral transmembrane proteins. Proc. Natl Acad. Sci. USA, 96, 4319–4324.  Web of Science CrossRef PubMed CAS Google Scholar
First citationKomander, D. & Rape, M. (2012). The ubiquitin code. Annu. Rev. Biochem. 81, 203–229.  Web of Science CrossRef CAS PubMed Google Scholar
First citationKonermann, L., Pan, J. & Liu, Y.-H. (2011). Hydrogen exchange mass spectrometry for studying protein structure and dynamics. Chem. Soc. Rev. 40, 1224–1234.  CrossRef CAS PubMed Google Scholar
First citationKong, L., Giang, E., Nieusma, T., Kadam, R. U., Cogburn, K. E., Hua, Y., Dai, X., Stanfield, R. L., Burton, D. R., Ward, A. B., Wilson, I. A. & Law, M. (2013). Hepatitis C virus E2 envelope glycoprotein core structure. Science, 342, 1090–1094.  CrossRef CAS PubMed Google Scholar
First citationKong, L., Huang, C.-C., Coales, S. J., Molnar, K. S., Skinner, J., Hamuro, Y. & Kwong, P. D. (2010). Local conformational stability of HIV-1 gp120 in unliganded and CD4-bound states as defined by amide hydrogen/deuterium exchange. J. Virol. 84, 10311–10321.  CrossRef CAS PubMed Google Scholar
First citationKong, L., Stanfield, R. & Wilson, I. (2014). HIV Glycans in Infection and Immunity, edited by R. Pantophlet, pp. 117–141. New York: Springer.  Google Scholar
First citationKopera, E., Bal, W., Lenarčič Živkovič, M., Dvornyk, A., Kludkiewicz, B., Grzelak, K., Zhukov, I., Zagórski-Ostoja, W., Jaskolski, M. & Krzywda, S. (2014). Atomic resolution structure of a protein prepared by non-enzymatic His-tag removal. Crystallographic and NMR study of GmSPI-2 inhibitor. PLoS One, 9, e106936.  CrossRef PubMed Google Scholar
First citationKottke, T., Hegemann, P., Dick, B. & Heberle, J. (2006). The photochemistry of the light-, oxygen-, and voltage-sensitive domains in the algal blue light receptor phot. Biopolymers, 82, 373–378.  CrossRef PubMed CAS Google Scholar
First citationKrężel, A., Kopera, E., Protas, A. M., Poznański, J., Wysłouch-Cieszyńska, A. & Bal, W. (2010). Sequence-specific NiII-dependent peptide bond hydrolysis for protein engineering. Combinatorial library determination of optimal sequences. J. Am. Chem. Soc. 132, 3355–3366.  PubMed Google Scholar
First citationKrishnamurthy, H. & Gouaux, E. (2012). X-ray structures of LeuT in substrate-free outward-open and apo inward-open states. Nature (London), 481, 469–474.  CrossRef CAS PubMed Google Scholar
First citationKrishnan, V. V. & Rupp, B. (2012). Macromolecular structure determination: comparison of X-ray crystallography and NMR spectroscopy. eLS, doi:10.1002/9780470015902.a0002716.pub2.  Google Scholar
First citationKumar, S., Tsai, C.-J. & Nussinov, R. (2000). Factors enhancing protein thermostability. Protein Eng. Des. Sel. 13, 179–191.  CrossRef CAS Google Scholar
First citationKurgan, L., Razib, A. A., Aghakhani, S., Dick, S., Mizianty, M. & Jahandideh, S. (2009). CRYSTALP2: sequence-based protein crystallization propensity prediction. BMC Struct. Biol. 9, 50.  Google Scholar
First citationKwan, A. H., Mobli, M., Gooley, P. R., King, G. F. & Mackay, J. P. (2011). Macromolecular NMR spectroscopy for the non-spectroscopist. FEBS J. 278, 687–703.  CrossRef CAS PubMed Google Scholar
First citationKyratsous, C. A. & Panagiotidis, C. A. (2012). Heat-shock protein fusion vectors for improved expression of soluble recombinant proteins in Escherichia coli. Methods Mol. Biol. 824, 109–129.  CrossRef CAS PubMed Google Scholar
First citationKyratsous, C. A., Silverstein, S. J., DeLong, C. R. & Panagiotidis, C. A. (2009). Chaperone-fusion expression plasmid vectors for improved solubility of recombinant proteins in Escherichia coli. Gene, 440, 9–15.  CrossRef PubMed CAS Google Scholar
First citationLawson, D. M., Artymiuk, P. J., Yewdall, S. J., Smith, J. M., Livingstone, J. C., Treffry, A., Luzzago, A., Levi, S., Arosio, P., Cesareni, G., Thomas, C. D., Shaw, W. V. & Harrison, P. M. (1991). Solving the structure of human H ferritin by genetically engineering intermolecular crystal contacts. Nature (London), 349, 541–544.  CrossRef PubMed CAS Web of Science Google Scholar
First citationLayton, C. J. & Hellinga, H. W. (2011). Quantitation of protein–protein interactions by thermal stability shift analysis. Protein Sci. 20, 1439–1450.  CrossRef CAS PubMed Google Scholar
First citationLazaridis, T. & Karplus, M. (2002). Thermodynamics of protein folding: a microscopic view. Biophys. Chem. 100, 367–395.  CrossRef Google Scholar
First citationLeibly, D. J., Nguyen, T. N., Kao, L. T., Hewitt, S. N., Barrett, L. K. & Van Voorhis, W. C. (2012). Stabilizing additives added during cell lysis aid in the solubilization of recombinant proteins. PLoS One, 7, e52482.  Web of Science CrossRef PubMed Google Scholar
First citationLi, S.-C., Goto, N. K., Williams, K. A. & Deber, C. M. (1996). α-Helical, but not β-sheet, propensity of proline is determined by peptide environment. Proc. Natl Acad. Sci. USA, 93, 6676–6681.  CrossRef CAS PubMed Google Scholar
First citationLieberman, R. L., Culver, J. A., Entzminger, K. C., Pai, J. C. & Maynard, J. A. (2011). Crystallization chaperone strategies for membrane proteins. Methods, 55, 293–302.  Web of Science CrossRef CAS PubMed Google Scholar
First citationLin, Z., Zhao, Q., Zhou, B., Xing, L. & Xu, W. (2015). Cleavable self-aggregating tags (cSAT) for protein expression and purification. Methods Mol. Biol. 1258, 65–78.  CrossRef PubMed Google Scholar
First citationLiu, J. & Song, J. (2009). Insights into protein aggregation by NMR characterization of insoluble SH3 mutants solubilized in salt-free water. PLoS One, 4, e7805.  CrossRef PubMed Google Scholar
First citationLiu, Y., Manna, A., Li, R., Martin, W. E., Murphy, R. C., Cheung, A. L. & Zhang, G. (2001). Crystal structure of the SarR protein from Staphylococcus aureus. Proc. Natl Acad. Sci. USA, 98, 6877–6882.  Web of Science CrossRef PubMed CAS Google Scholar
First citationLiu, Z. & Huang, Y. (2014). Advantages of proteins being disordered. Protein Sci. 23, 539–550.  CrossRef CAS PubMed Google Scholar
First citationLiu, Z.-Q., Mahmood, T. & Yang, P.-C. (2014). Western blot: technique, theory and trouble shooting. N. Am. J. Med. Sci. 6, 160.  PubMed Google Scholar
First citationLongenecker, K. L., Garrard, S. M., Sheffield, P. J. & Derewenda, Z. S. (2001). Protein crystallization by rational mutagenesis of surface residues: Lys to Ala mutations promote crystallization of RhoGDI. Acta Cryst. D57, 679–688.  Web of Science CrossRef CAS IUCr Journals Google Scholar
First citationLonghi, S., Lieutaud, P. & Canard, B. (2010). Conformational disorder. Methods Mol. Biol. 609, 307–325.  CrossRef CAS PubMed Google Scholar
First citationMalakhova, O. A., Yan, M., Malakhov, M. P., Yuan, Y., Ritchie, K. J., Kim, K. I., Peterson, L. F., Shuai, K. & Zhang, D.-E. (2003). Protein ISGylation modulates the JAK-Stat signaling pathway. Genes Dev. 17, 455–460.  CrossRef PubMed CAS Google Scholar
First citationMandel, C. R., Gebauer, D., Zhang, H. & Tong, L. (2006). A serendipitous discovery that in situ proteolysis is essential for the crystallization of yeast CPSF-100 (Ydh1p). Acta Cryst. F62, 1041–1045.  Web of Science CrossRef CAS IUCr Journals Google Scholar
First citationMarsh, E. N. (2014). Fluorinated proteins: from design and synthesis to structure and stability. Acc. Chem. Res. 47, 2878–2886.  CrossRef CAS PubMed Google Scholar
First citationMateja, A., Devedjiev, Y., Krowarsch, D., Longenecker, K., Dauter, Z., Otlewski, J. & Derewenda, Z. S. (2002). The impact of Glu→Ala and Glu→Asp mutations on the crystallization properties of RhoGDI: the structure of RhoGDI at 1.3 Å resolution. Acta Cryst. D58, 1983–1991.  Web of Science CrossRef CAS IUCr Journals Google Scholar
First citationMathiasen, S., Christensen, S. M., Fung, J. J., Rasmussen, S. G. F., Fay, J. F., Jorgensen, S. K., Veshaguri, S., Farrens, D. L., Kiskowski, M., Kobilka, B. & Stamou, D. (2014). Nanoscale high-content analysis using compositional heterogeneities of single proteoliposomes. Nature Methods, 11, 931–934.  CrossRef CAS PubMed Google Scholar
First citationMcPherson, A. & Cudney, B. (2006). Searching for silver bullets: an alternative strategy for crystallizing macromolecules. J. Struct. Biol. 156, 387–406.  Web of Science CrossRef PubMed CAS Google Scholar
First citationMeans, G. E. (1977). Reductive alkylation of amino groups. Methods Enzymol. 47, 469–478.  CrossRef CAS PubMed Google Scholar
First citationMellquist, J. L., Kasturi, L., Spitalnik, S. L. & Shakin-Eshleman, S. H. (1998). The amino acid following an Asn-X-Ser/Thr sequon is an important determinant of N-linked core glycosylation efficiency. Biochemistry, 37, 6833–6837.  CrossRef CAS PubMed Google Scholar
First citationMills, K. V., Johnson, M. A. & Perler, F. B. (2014). Protein splicing: how inteins escape from precursor proteins. J. Biol. Chem. 289, 14498–14505.  CrossRef CAS PubMed Google Scholar
First citationMizianty, M. J. & Kurgan, L. (2009). Meta prediction of protein crystallization propensity. Biochem. Biophys. Res. Commun. 390, 10–15.  Web of Science CrossRef PubMed CAS Google Scholar
First citationMizianty, M. J., Uversky, V. & Kurgan, L. (2014). Prediction of intrinsic disorder in proteins using MFDp2. Methods Mol. Biol. 1137, 147–162.  CrossRef CAS PubMed Google Scholar
First citationMonod, J., Wyman, J. & Changeux, J.-P. (1965). On the nature of allosteric transitions: a plausible model. J. Mol. Biol. 12, 88–118.  CrossRef PubMed CAS Web of Science Google Scholar
First citationMoon, A. F., Mueller, G. A., Zhong, X. & Pedersen, L. C. (2010). A synergistic approach to protein crystallization: combination of a fixed-arm carrier with surface entropy reduction. Protein Sci. 19, 901–913.  Web of Science CAS PubMed Google Scholar
First citationMorar-Mitrica, S., Nesta, D. & Crotts, G. (2013). Differential scanning calorimetry (DSC) for biopharmaceutical development: old concepts, new applications. BioPharma Asia, 2(4), 44–55.  Google Scholar
First citationMoshe, A., Landau, M. & Eisenberg, D. (2016). Preparation of crystalline samples of amyloid fibrils and oligomers. Methods Mol. Biol. 1345, 201–210.  CrossRef PubMed Google Scholar
First citationMottonen, J., Strand, A., Symersky, J., Sweet, R. M., Danley, D. E., Geoghegan, K. F., Gerard, R. D. & Goldsmith, E. J. (1992). Structural basis of latency in plasminogen activator inhibitor-1. Nature (London), 355, 270–273.  CrossRef PubMed CAS Web of Science Google Scholar
First citationMoukhametzianov, R., Burghammer, M., Edwards, P. C., Petitdemange, S., Popov, D., Fransen, M., McMullan, G., Schertler, G. F. X. & Riekel, C. (2008). Protein crystallography with a micrometre-sized synchrotron-radiation beam. Acta Cryst. D64, 158–166.  Web of Science CrossRef CAS IUCr Journals Google Scholar
First citationMujtaba, S., He, Y., Zeng, L., Yan, S., Plotnikova, O., Sachchidanand, Sanchez, R., Zeleznik-Le, N. J., Ronai, Z. & Zhou, M.-M. (2004). Structural mechanism of the bromodomain of the coactivator CBP in p53 transcriptional activation. Mol. Cell, 13, 251–263.  CrossRef PubMed CAS Google Scholar
First citationMulder, D. W., Shepard, E. M., Meuser, J. E., Joshi, N., King, P. W., Posewitz, M. C., Broderick, J. B. & Peters, J. W. (2011). Insights into [FeFe]-hydrogenase structure, mechanism, and maturation. Structure, 19, 1038–1052.  CrossRef CAS PubMed Google Scholar
First citationNettleship, J. E., Watson, P. J., Rahman-Huq, N., Fairall, L., Posner, M. G., Upadhyay, A., Reddivari, Y., Chamberlain, J. M., Kolstoe, S. E., Bagby, S., Schwabe, J. W. & Owens, R. J. (2015). Transient expression in HEK 293 cells: an alternative to E. coli for the production of secreted and intracellular mammalian proteins. Methods Mol. Biol. 1258, 209–222.  CrossRef PubMed Google Scholar
First citationNeutze, R. & Moffat, K. (2012). Time-resolved structural studies at synchrotrons and X-ray free electron lasers: opportunities and challenges. Curr. Opin. Struct. Biol. 22, 651–659.  Web of Science CrossRef CAS PubMed Google Scholar
First citationNovikova, O., Topilina, N. & Belfort, M. (2014). Enigmatic distribution, evolution, and function of inteins. J. Biol. Chem. 289, 14490–14497.  CrossRef CAS PubMed Google Scholar
First citationOldfield, C. J., Meng, J., Yang, J. Y., Yang, M. Q., Uversky, V. N. & Dunker, A. K. (2008). Flexible nets: disorder and induced fit in the associations of p53 and 14-3-3 with their partners. BMC Genomics, 9, S1.  Google Scholar
First citationOlson, M. A., Zabetakis, D., Legler, P. M., Turner, K. B., Anderson, G. P. & Goldman, E. R. (2015). Fusion to a highly stable consensus albumin binding domain allows for tunable pharmacokinetics. Protein Eng. Des. Sel. 28, 395–402.  CrossRef CAS PubMed Google Scholar
First citationOltersdorf, T. et al. (2005). An inhibitor of Bcl-2 family proteins induces regression of solid tumours. Nature (London), 435, 677–681.  CrossRef PubMed CAS Google Scholar
First citationOverton, I. M., Padovani, G., Girolami, M. A. & Barton, G. J. (2008). ParCrys: a Parzen window density estimation approach to protein crystallization propensity prediction. Bioinformatics, 24, 901–907.  Web of Science CrossRef PubMed CAS Google Scholar
First citationOverton, I. M., van Niekerk, C. A. & Barton, G. J. (2011). XANNpred: neural nets that predict the propensity of a protein to yield diffraction-quality crystals. Proteins, 79, 1027–1033.  CrossRef CAS PubMed Google Scholar
First citationPace, C. N., Fu, H., Fryar, K. L., Landua, J., Trevino, S. R., Shirley, B. A., Hendricks, M. M., Iimura, S., Gajiwala, K., Scholtz, J. M. & Grimsley, G. R. (2011). Contribution of hydrophobic interactions to protein stability. J. Mol. Biol. 408, 514–528.  CrossRef CAS PubMed Google Scholar
First citationPace, C. N., Fu, H. et al. (2014). Contribution of hydrogen bonds to protein stability. Protein Sci. 23, 652–661.  CrossRef CAS PubMed Google Scholar
First citationPace, C. N. & Scholtz, J. M. (1998). Biophys. J. 75, 422–427.  Web of Science CAS PubMed Google Scholar
First citationPace, C. N., Scholtz, J. M. & Grimsley, G. R. (2014). Forces stabilizing proteins. FEBS Lett. 588, 2177–2184.  PubMed Google Scholar
First citationPantazatos, D., Kim, J. S., Klock, H. E., Stevens, R. C., Wilson, I. A., Lesley, S. A. & Woods, V. L. Jr (2004). Rapid refinement of crystallographic protein construct definition employing enhanced hydrogen/deuterium exchange MS. Proc. Natl Acad. Sci. USA, 101, 751–756.  Web of Science CrossRef PubMed CAS Google Scholar
First citationPapaneophytou, C. P. & Kontopidis, G. (2014). Statistical approaches to maximize recombinant protein expression in Escherichia coli: a general review. Protein Expr. Purif. 94, 22–32.  CrossRef CAS PubMed Google Scholar
First citationPark, M. H., Nishimura, K., Zanelli, C. F. & Valentini, S. R. (2010). Functional significance of eIF5A and its hypusine modification in eukaryotes. Amino Acids, 38, 491–500.  CrossRef PubMed CAS Google Scholar
First citationPauling, L., Corey, R. B. & Branson, H. R. (1951). The structure of proteins: two hydrogen-bonded helical configurations of the polypeptide chain. Proc. Natl Acad. Sci. USA, 37, 205–211.  CrossRef PubMed CAS Web of Science Google Scholar
First citationPaulus, H. (2000). Protein splicing and related forms of protein autoprocessing. Annu. Rev. Biochem. 69, 447–496.  Web of Science CrossRef PubMed CAS Google Scholar
First citationPelton, J. T. & McLean, L. R. (2000). Spectroscopic methods for analysis of protein secondary structure. Anal. Biochem. 277, 167–176.  CrossRef PubMed CAS Google Scholar
First citationPrivalov, P. L. & Dragan, A. I. (2007). Microcalorimetry of biological macromolecules. Biophys. Chem. 126, 16–24.  CrossRef PubMed CAS Google Scholar
First citationProthero, J. W. (1966). Correlation between the distribution of amino acids and alpha helices. Biophys. J. 6, 367–370.  CrossRef CAS PubMed Google Scholar
First citationPugach, P. et al. (2015). A native-like SOSIP.664 trimer based on an HIV-1 subtype B env gene. J. Virol. 89, 3380–3395.  CrossRef CAS PubMed Google Scholar
First citationPuthalakath, H., Burke, J. & Gleeson, P. A. (1996). Glycosylation defect in Lec1 Chinese hamster ovary mutant is due to a point mutation in N-acetylglucosaminyltransferase I gene. J. Biol. Chem. 271, 27818–27822.  CrossRef CAS PubMed Google Scholar
First citationQing, G., Ma, L.-C., Khorchid, A., Swapna, G. V. T., Mal, T. K., Takayama, M. M., Xia, B., Phadtare, S., Ke, H., Acton, T., Montelione, G. T., Ikura, M. & Inouye, M. (2004). Cold-shock induced high-yield protein production in Escherichia coli. Nature Biotechnol. 22, 877–882.  CrossRef CAS Google Scholar
First citationQuistgaard, E. M. (2014). A disulfide polymerized protein crystal. Chem. Commun. 50, 14995–14997.  CrossRef CAS Google Scholar
First citationRabut, G. & Peter, M. (2008). Function and regulation of protein neddylation. EMBO Rep. 9, 969–976.  Web of Science CrossRef PubMed CAS Google Scholar
First citationRamachandran, G. N., Ramakrishnan, C. & Sasisekharan, V. (1963). Stereochemistry of polypeptide chain configurations. J. Mol. Biol. 7, 95–99.  CrossRef PubMed CAS Web of Science Google Scholar
First citationReeves, P. J., Callewaert, N., Contreras, R. & Khorana, H. G. (2002). Structure and function in rhodopsin: high-level expression of rhodopsin with restricted and homogeneous N-glycosylation by a tetracycline-inducible N-acetylglucosaminyltransferase I-negative HEK293S stable mammalian cell line. Proc. Natl Acad. Sci. USA, 99, 13419–13424.  Web of Science CrossRef PubMed CAS Google Scholar
First citationRehm, T., Huber, R. & Holak, T. A. (2002). Application of NMR in structural proteomics: screening for proteins amenable to structural analysis. Structure, 10, 1613–1618.  CrossRef PubMed CAS Google Scholar
First citationReich, S., Puckey, L. H., Cheetham, C. L., Harris, R., Ali, A. A. E., Bhattacharyya, U., Maclagan, K., Powell, K. A., Prodromou, C., Pearl, L. H., Driscoll, P. C. & Savva, R. (2006). Combinatorial domain hunting: an effective approach for the identification of soluble protein domains adaptable to high-throughput applications. Protein Sci. 15, 2356–2365.  CrossRef PubMed CAS Google Scholar
First citationReinhard, L., Mayerhofer, H., Geerlof, A., Mueller-Dieckmann, J. & Weiss, M. S. (2013). Optimization of protein buffer cocktails using Thermofluor. Acta Cryst. F69, 209–214.  Web of Science CrossRef IUCr Journals Google Scholar
First citationRemaut, H., Tang, C., Henderson, N. S., Pinkner, J. S., Wang, T., Hultgren, S. J., Thanassi, D. G., Waksman, G. & Li, H. (2008). Fiber formation across the bacterial outer membrane by the chaperone/usher pathway. Cell, 133, 640–652.  CrossRef PubMed CAS Google Scholar
First citationRice, R. H., Means, G. E. & Brown, W. D. (1977). Stabilization of bovine trypsin by reductive methylation. Biochim. Biophys. Acta, 492, 316–321.  CrossRef CAS PubMed Google Scholar
First citationRichards, F. M. (1997). Protein stability: still an unsolved problem. Cell. Mol. Life Sci. 53, 790–802.  CrossRef CAS PubMed Google Scholar
First citationRichardson, J. S. (1981). The anatomy and taxonomy of protein structure. Adv. Protein Chem. 34, 167–339.  CrossRef CAS PubMed Google Scholar
First citationRistic, M., Rosa, N., Seabrook, S. A. & Newman, J. (2015). Formulation screening by differential scanning fluorimetry: how often does it work? Acta Cryst. F71, 1359–1364.  Web of Science CrossRef IUCr Journals Google Scholar
First citationRogers, S., Wells, R. & Rechsteiner, M. (1986). Amino acid sequences common to rapidly degraded proteins: the PEST hypothesis. Science, 234, 364–368.  CrossRef CAS PubMed Google Scholar
First citationRonda, L., Bruno, S. & Bettati, S. (2013). Tertiary and quaternary effects in the allosteric regulation of animal hemoglobins. Biochim. Biophys. Acta, 1834, 1860–1872.  CrossRef CAS PubMed Google Scholar
First citationRosa, N., Ristic, M., Seabrook, S. A., Lovell, D., Lucent, D. & Newman, J. (2015). Meltdown: a tool to help in the interpretation of thermal melt curves acquired by differential scanning fluorimetry. J. Biomol. Screen. 20, 898–905.  CrossRef CAS PubMed Google Scholar
First citationRuggiero, A., Smaldone, G., Squeglia, F. & Berisio, R. (2012). Enhanced crystallizability by protein engineering approaches: a general overview. Protein Pept. Lett. 19, 732–742.  CrossRef CAS PubMed Google Scholar
First citationRupp, B. (2015). Origin and use of crystallization phase diagrams. Acta Cryst. F71, 247–260.  Web of Science CrossRef IUCr Journals Google Scholar
First citationSanchez-Ruiz, J. M. (1995). Differential scanning calorimetry of proteins. Subcell. Biochem. 24, 133–176.  CAS PubMed Google Scholar
First citationSawaya, M. R., Sambashivan, S., Nelson, R., Ivanova, M. I., Sievers, S. A., Apostol, M. I., Thompson, M. J., Balbirnie, M., Wiltzius, J. J., McFarlane, H. T., Madsen, A. O., Riekel, C. & Eisenberg, D. (2007). Atomic structures of amyloid cross-β spines reveal varied steric zippers. Nature (London), 447, 453–457.  Web of Science CrossRef PubMed CAS Google Scholar
First citationSchrödinger, E. (1945). What is Life? The Physical Aspect of the Living Cell. New York: McMillan.  Google Scholar
First citationSchuldt, L., Weyand, S., Kefala, G. & Weiss, M. S. (2009). The three-dimensional structure of a mycobacterial DapD provides insights into DapD diversity and reveals unexpected particulars about the enzymatic mechanism. J. Mol. Biol. 389, 863–879.  Web of Science CrossRef PubMed CAS Google Scholar
First citationSeabrook, S. A. & Newman, J. (2013). High-throughput thermal scanning for protein stability: making a good technique more robust. ACS Comb. Sci. 15, 387–392.  Web of Science CrossRef CAS PubMed Google Scholar
First citationSecchiero, P., Bosco, R., Celeghini, C. & Zauli, G. (2011). Recent advances in the therapeutic perspectives of nutlin-3. Curr. Pharm. Des. 17, 569–577.  CrossRef CAS PubMed Google Scholar
First citationSelzer, L., Kant, R., Wang, J. C., Bothner, B. & Zlotnick, A. (2015). Hepatitis B virus core protein phosphorylation sites affect capsid stability and transient exposure of the C-terminal domain. J. Biol. Chem. 290, 28584–28593  CrossRef CAS PubMed Google Scholar
First citationSemisotnov, G. V., Rodionova, N. A., Razgulyaev, O. I., Uversky, V. N., Gripas', A. F. & Gilmanshin, R. I. (1991). Study of the `molten globule' intermediate state in protein folding by a hydrophobic fluorescent probe. Biopolymers, 31, 119–128.  CrossRef CAS PubMed Web of Science Google Scholar
First citationShi, M., Foo, S. Y., Tan, S.-M., Mitchell, E. P., Law, S. K. A. & Lescar, J. (2007). A structural hypothesis for the transition between bent and extended conformations of the leukocyte β2 integrins. J. Biol. Chem. 282, 30198–30206.  CrossRef PubMed CAS Google Scholar
First citationShimizu, K. (2014). POODLE: tools predicting intrinsically disordered regions of amino acid sequence. Methods Mol. Biol. 1137, 131–145.  CrossRef CAS PubMed Google Scholar
First citationShumway, S. D., Maki, M. & Miyamoto, S. (1999). The PEST domain of IκBα is necessary and sufficient for in vitro degradation by μ-calpain. J. Biol. Chem. 274, 30874–30881.  CrossRef PubMed CAS Google Scholar
First citationSikic, K. & Carugo, O. (2009). CARON – average RMSD of NMR structure ensembles. Bioinformation, 4, 132–133.  CrossRef PubMed Google Scholar
First citationSlabinski, L., Jaroszewski, L., Rychlewski, L., Wilson, I. A., Lesley, S. A. & Godzik, A. (2007). XtalPred: a web server for prediction of protein crystallizability. Bioinformatics, 23, 3403–3405.  Web of Science CrossRef PubMed CAS Google Scholar
First citationSledz, P., Zheng, H., Murzyn, K., Chruszcz, M., Zimmerman, M. D., Chordia, M. D., Joachimiak, A. & Minor, W. (2010). New surface contacts formed upon reductive lysine methylation: improving the probability of protein crystallization. Protein Sci. 19, 1395–1404.  Web of Science CrossRef CAS PubMed Google Scholar
First citationSmagghe, B. J., Huang, P.-S., Ban, Y.-E. A., Baker, D. & Springer, T. A. (2010). Modulation of integrin activation by an entropic spring in the β-knee. J. Biol. Chem. 285, 32954–32966.  CrossRef CAS PubMed Google Scholar
First citationSmialowski, P. & Frishman, D. (2010). Protein crystallizability. Methods Mol. Biol. 609, 385–400.  CrossRef CAS PubMed Google Scholar
First citationSmith, C. K., Withka, J. M. & Regan, L. (1994). A thermodynamic scale for the β-sheet forming tendencies of the amino acids. Biochemistry, 33, 5510–5517.  CrossRef CAS PubMed Web of Science Google Scholar
First citationSmoot, A. L., Panda, M., Brazil, B. T., Buckle, A. M., Fersht, A. R. & Horowitz, P. M. (2001). The binding of bis-ANS to the isolated GroEL apical domain fragment induces the formation of a folding intermediate with increased hydrophobic surface not observed in tetradecameric GroEL. Biochemistry, 40, 4484–4492.  Web of Science CrossRef PubMed CAS Google Scholar
First citationSmyth, D. R., Mrozkiewicz, M. K., McGrath, W. J., Listwan, P. & Kobe, B. (2003). Crystal structures of fusion proteins with large affinity tags. Protein Sci. 12, 1313–1322.  Web of Science CrossRef PubMed CAS Google Scholar
First citationSomero, G. N. (2004). Adaptation of enzymes to temperature: searching for basic `strategies'. Comp. Biochem. Physiol. B Biochem. Mol. Biol. 139, 321–333.  CrossRef PubMed Google Scholar
First citationSørensen, H. P. & Mortensen, K. K. (2005). Advanced genetic strategies for recombinant protein expression in Escherichia coli. J. Biotechnol. 115, 113–128.  PubMed Google Scholar
First citationSpence, J. C., Weierstall, U. & Chapman, H. N. (2012). X-ray lasers for structural and dynamic biology. Rep. Prog. Phys. 75, 102601.  Web of Science CrossRef PubMed Google Scholar
First citationSpencer, M. L., Theodosiou, M. & Noonan, D. J. (2004). NPDC-1, a novel regulator of neuronal proliferation, is degraded by the ubiquitin/proteasome system through a PEST degradation motif. J. Biol. Chem. 279, 37069–37078.  CrossRef PubMed CAS Google Scholar
First citationSpiegel, H., Schinkel, H., Kastilan, R., Dahm, P., Boes, A., Scheuermayer, M., Chudobová, I., Maskus, D., Fendel, R., Schillberg, S., Reimann, A. & Fischer, R. (2015). Optimization of a multi-stage, multi-subunit malaria vaccine candidate for the production in Pichia pastoris by the identification and removal of protease cleavage sites. Biotechnol. Bioeng. 112, 659–667.  CrossRef CAS PubMed Google Scholar
First citationSpraggon, G., Pantazatos, D., Klock, H. E., Wilson, I. A., Woods, V. L. Jr & Lesley, S. A. (2004). On the use of DXMS to produce more crystallizable proteins: structures of the T. maritima proteins TM0160 and TM1171. Protein Sci. 13, 3187–3199.  CrossRef PubMed CAS Google Scholar
First citationStickle, D. F., Presta, L. G., Dill, K. A. & Rose, G. D. (1992). Hydrogen bonding in globular proteins. J. Mol. Biol. 226, 1143–1159.  PubMed CAS Web of Science Google Scholar
First citationStriebel, F., Imkamp, F., Özcelik, D. & Weber-Ban, E. (2014). Pupylation as a signal for proteasomal degradation in bacteria. Biochim. Biophys. Acta, 1843, 103–113.  CrossRef CAS PubMed Google Scholar
First citationStudier, F. W. (1991). Use of bacteriophage T7 lysozyme to improve an inducible T7 expression system. J. Mol. Biol. 219, 37–44.  CrossRef CAS PubMed Web of Science Google Scholar
First citationSugase, K., Dyson, H. J. & Wright, P. E. (2007). Mechanism of coupled folding and binding of an intrinsically disordered protein. Nature (London), 447, 1021–1025.  CrossRef PubMed CAS Google Scholar
First citationSurma, M. A., Szczepaniak, A. & Króliczewski, J. (2014). Comparative studies on detergent-assisted apocytochrome b6 reconstitution into liposomal bilayers monitored by zetasizer instruments. PLoS One, 9, e111341.  CrossRef PubMed Google Scholar
First citationTan, K. et al. (2014). Salvage of failed protein targets by reductive alkylation. Methods Mol. Biol. 1140, 189–200.  CrossRef CAS PubMed Google Scholar
First citationTanner, J. J., Hecht, R. M. & Krause, K. L. (1996). Determinants of enzyme thermostability observed in the molecular structure of Thermus aquaticus D-glyceraldehyde-3-phosphate dehydrogenase at 2.5 Å resolution. Biochemistry, 35, 2597–2609.  CrossRef CAS PubMed Web of Science Google Scholar
First citationTegel, H., Tourle, S., Ottosson, J. & Persson, A. (2010). Increased levels of recombinant human proteins with the Escherichia coli strain Rosetta(DE3). Protein Expr. Purif. 69, 159–167.  CrossRef PubMed CAS Google Scholar
First citationTowbin, H., Staehelin, T. & Gordon, J. (1979). Electrophoretic transfer of proteins from polyacrylamide gels to nitrocellulose sheets: procedure and some applications. Proc. Natl Acad. Sci. USA, 76, 4350–4354.  CrossRef CAS PubMed Web of Science Google Scholar
First citationUervirojnangkoorn, M., Zeldin, O. B., Lyubimov, A. Y., Hattne, J., Brewster, A. S., Sauter, N. K., Brunger, A. T. & Weis, W. I. (2015). Enabling X-ray free electron laser crystallography for challenging biological systems from a limited number of crystals. Elife, 4, e05421.  Web of Science CrossRef Google Scholar
First citationUversky, V. N. & Dunker, A. K. (2010). Understanding protein non-folding. Biochim. Biophys. Acta, 1804, 1231–1264.  Web of Science CrossRef CAS PubMed Google Scholar
First citationVasina, J. A. & Baneyx, F. (1997). Expression of aggregation-prone recombinant proteins at low temperatures: a comparative study of the Escherichia coli cspA and tac promoter systems. Protein Expr. Purif. 9, 211–218.  CrossRef CAS PubMed Google Scholar
First citationVolkmann, G. & Iwaï, H. (2010). Protein trans-splicing and its use in structural biology: opportunities and limitations. Mol. Biosyst. 6, 2110–2121.  Web of Science CrossRef CAS PubMed Google Scholar
First citationWakatsuki, S. (2016). In Synchrotron Light Sources and Free-Electron Lasers, edited by E. Jaeschke, S. Khan, J. R. Schneider & J. B. Hastings. New York: Springer. In the press.  Google Scholar
First citationWalker, C. S., Shetty, R. P., Clark, K., Kazuko, S. G., Letsou, A., Olivera, B. M. & Bandyopadhyay, P. K. (2001). On a potential global role for vitamin K-dependent γ-carboxylation in animal systems: evidence for a γ-glutamyl carboxylase in Drosophila. J. Biol. Chem. 276, 7769–7774.  CrossRef PubMed CAS Google Scholar
First citationWallace, B. A. & Janes, R. W. (2010). Synchrotron radiation circular dichroism (SRCD) spectroscopy: an enhanced method for examining protein conformations and protein interactions. Biochem. Soc. Trans. 38, 861–873.  Web of Science CrossRef CAS PubMed Google Scholar
First citationWalls, D. & Loughran, S. T. (2011). Tagging recombinant proteins to enhance solubility and aid purification. Methods Mol. Biol. 681, 151–175.  CrossRef CAS PubMed Google Scholar
First citationWalter, T. S., Meier, C., Assenberg, R., Au, K. F., Ren, J., Verma, A., Nettleship, J. E., Owens, R. J., Stuart, D. I. & Grimes, J. M. (2006). Lysine methylation as a routine rescue strategy for protein crystallization. Structure, 14, 1617–1622.  Web of Science CrossRef PubMed CAS Google Scholar
First citationWang, J., Cao, Z., Zhao, L. & Li, S. (2011). Novel strategies for drug discovery based on intrinsically disordered proteins (IDPs). Int. J. Mol. Sci. 12, 3205–3219.  CrossRef CAS PubMed Google Scholar
First citationWarne, T., Serrano-Vega, M. J., Baker, J. G., Moukhametzianov, R., Edwards, P. C., Henderson, R., Leslie, A. G. W., Tate, C. G. & Schertler, G. F. X. (2008). Structure of a β1-adrenergic G-protein-coupled receptor. Nature (London), 454, 486–491.  Web of Science CrossRef PubMed CAS Google Scholar
First citationWeckert, E. (2015). The potential of future light sources to explore the structure and function of matter. IUCrJ, 2, 230–245.  Web of Science CrossRef CAS PubMed IUCr Journals Google Scholar
First citationWells, J. A. & McClendon, C. L. (2007). Reaching for high-hanging fruit in drug discovery at protein–protein interfaces. Nature (London), 450, 1001–1009.  CrossRef PubMed CAS Google Scholar
First citationWernimont, A. & Edwards, A. (2009). In situ proteolysis to generate crystals for structure determination: an update. PLoS One, 4, e5094.  Web of Science CrossRef PubMed Google Scholar
First citationWhisstock, J. C. & Bottomley, S. P. (2006). Molecular gymnastics: serpin structure, folding and misfolding. Curr. Opin. Struct. Biol. 16, 761–768.  CrossRef PubMed CAS Google Scholar
First citationWhisstock, J. C., Skinner, R., Carrell, R. W. & Lesk, A. M. (2000). Conformational changes in serpins. I. The native and cleaved conformations of α1-antitrypsin. J. Mol. Biol. 296, 685–699.  CrossRef PubMed CAS Google Scholar
First citationWhiteheart, S. W., Shenbagamurthi, P., Chen, L., Cotter, R. J. & Hart, G. W. (1989). Murine elongation factor 1 alpha (EF-1 alpha) is posttranslationally modified by novel amide-linked ethanolamine-phosphoglycerol moieties. Addition of ethanolamine-phosphoglycerol to specific glutamic acid residues on EF-1 alpha. J. Biol. Chem. 264, 14334–14341.  CAS PubMed Google Scholar
First citationWhitmore, L. & Wallace, B. A. (2008). Protein secondary structure analyses from circular dichroism spectroscopy: methods and reference databases. Biopolymers, 89, 392–400.  Web of Science CrossRef PubMed CAS Google Scholar
First citationWhitmore, L., Woollett, B., Miles, A. J., Klose, D. P., Janes, R. W. & Wallace, B. A. (2011). PCDDB: the Protein Circular Dichroism Data Bank, a repository for circular dichroism spectral and metadata. Nucleic Acids Res. 39, D480–D486.  CrossRef CAS PubMed Google Scholar
First citationWillis, B. T. M. & Pryor, A. W. (1975). Thermal Vibrations in Crystallography. Cambridge University Press.  Google Scholar
First citationWood, D. W. (2014). New trends and affinity tag designs for recombinant protein purification. Curr. Opin. Struct. Biol. 26, 54–61.  CrossRef CAS PubMed Google Scholar
First citationWright, P. E. & Dyson, H. J. (1999). Intrinsically unstructured proteins: re-assessing the protein structure–function paradigm. J. Mol. Biol. 293, 321–331.  Web of Science CrossRef PubMed CAS Google Scholar
First citationWukovitz, S. W. & Yeates, T. O. (1995). Why protein crystals favour some space groups over others. Nature Struct. Mol. Biol. 2, 1062–1067.  CrossRef CAS Web of Science Google Scholar
First citationYakimov, A., Rychkov, G. & Petukhov, M. (2014). De novo design of stable α-helices. Methods Mol. Biol. 1216, 1–14.  CrossRef CAS PubMed Google Scholar
First citationYamasaki, M., Li, W., Johnson, D. J. & Huntington, J. A. (2008). Crystal structure of a stable dimer reveals the molecular basis of serpin polymerization. Nature (London), 455, 1255–1258.  Web of Science CrossRef PubMed CAS Google Scholar
First citationYeh, A. P., McMillan, A. & Stowell, M. H. B. (2006). Rapid and simple protein-stability screens: application to membrane proteins. Acta Cryst. D62, 451–457.  Web of Science CrossRef CAS IUCr Journals Google Scholar
First citationYumerefendi, H., Tarendeau, F., Mas, P. J. & Hart, D. J. (2010). ESPRIT: an automated, library-based method for mapping and soluble expression of protein domains from challenging targets. J. Struct. Biol. 172, 66–74.  Web of Science CrossRef CAS PubMed Google Scholar
First citationYun, R. H., Anderson, A. & Hermans, J. (1991). Proline in α-helix: stability and conformation studied by dynamics simulation. Proteins, 10, 219–228.  CrossRef PubMed CAS Google Scholar
First citationZhang, A. P., Bornholdt, Z. A., Liu, T., Abelson, D. M., Lee, D. E., Li, S., Woods, V. L. Jr & Saphire, E. O. (2012). The ebola virus interferon antagonist VP24 directly binds STAT1 and has a novel, pyramidal fold. PLoS Pathog. 8, e1002550.  CrossRef PubMed Google Scholar
First citationZhao, Q., Frederick, R., Seder, K., Thao, S., Sreenath, H., Peterson, F., Volkman, B. F., Markley, J. L. & Fox, B. G. (2004). Production in two-liter beverage bottles of proteins for NMR structure determination labeled with either 15N- or 13C–15N. J. Struct. Funct. Genomics, 5, 87–93.  CrossRef PubMed CAS Google Scholar
First citationZhou, Q. et al. (2015). Architecture of the synaptotagmin–SNARE machinery for neuronal exocytosis. Nature (London), 525, 62–67.  CrossRef CAS PubMed Google Scholar
First citationZhou, X.-X., Wang, Y.-B., Pan, Y.-J. & Li, W.-F. (2008). Differences in amino acids composition and coupling patterns between mesophilic and thermophilic proteins. Amino Acids, 34, 25–33.  Web of Science CrossRef PubMed CAS Google Scholar
First citationZou, Y., Weis, W. I. & Kobilka, B. K. (2012). N-Terminal T4 lysozyme fusion facilitates crystallization of a G protein coupled receptor. PLoS One, 7, e46039.  Web of Science CrossRef PubMed Google Scholar

This is an open-access article distributed under the terms of the Creative Commons Attribution (CC-BY) Licence, which permits unrestricted use, distribution, and reproduction in any medium, provided the original authors and source are cited.

Journal logoSTRUCTURAL BIOLOGY
COMMUNICATIONS
ISSN: 2053-230X
Follow Acta Cryst. F
Sign up for e-alerts
Follow Acta Cryst. on Twitter
Follow us on facebook
Sign up for RSS feeds