Next Article in Journal
Varying Synthesis Conditions and Comprehensive Characterization of Fluorine-Doped Hydroxyapatite Nanocrystals in a Simulated Body Fluid
Next Article in Special Issue
Pink-Beam Serial Synchrotron Crystallography at Pohang Light Source II
Previous Article in Journal
Continuous Isolation of Particles with Varying Aspect Ratios up to Thin Needles Achieving Free-Flowing Products
Previous Article in Special Issue
Processing of Multicrystal Diffraction Patterns in Macromolecular Crystallography Using Serial Crystallography Programs
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Perspective

A Short Review on Cryoprotectants for 3D Protein Structure Analysis

1
Department of Lifestyle Medicine, College of Environmental and Bioresource Sciences, Jeonbuk National University, Gobong-ro 79, Iksan 54596, Korea
2
Department of Botany, Dagon University, Min Ye Kyawswar Street 52, East Dagon Township, Yangon 11422, Myanmar
3
Advanced Materials Division, Korea Research Institute of Chemical Technology (KRICT), Gajeong-ro 141, Daejeon 34114, Korea
4
Department of Chemistry, College of Natural Science, Jeonbuk National University, Baekje-daero 567, Jeonju 54896, Korea
5
Department of Oriental Medicine Resources, College of Environmental and Bioresource Sciences, Jeonbuk National University, Gobong-ro 79, Iksan 54596, Korea
6
Advanced Institute of Environment and Bioscience, College of Environmental & Bioresources Sciences, Jeonbuk National University, Gobong-ro 79, Iksan 54596, Korea
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Crystals 2022, 12(2), 138; https://doi.org/10.3390/cryst12020138
Submission received: 8 December 2021 / Revised: 14 January 2022 / Accepted: 15 January 2022 / Published: 19 January 2022
(This article belongs to the Special Issue Serial X-ray Crystallography)

Abstract

:
The three-dimensional structure of protein is determined by analyzing diffraction data collected using X-ray beams. However, X-ray beam can damage protein crystals during data collection, lowering the quality of the crystal data. A way to prevent such damage is by treating protein crystals with cryoprotectants. The cryoprotectant stabilizes the protein crystal and prevents lowering the quality of the diffraction data. Many kinds of cryoprotectants are commercially available, and various treatment methods have also been reported. However, incorrect selection or treatment of such cryoprotectants may lead to deterioration of crystal diffraction data when using X-ray beams.

1. Introduction

The first protein crystal was observed about 180 years ago [1,2]. However, crystallizing protein remained questionable until the 1880s, when plant seed proteins were crystallized for purification [3,4]. Since then, the PDB (Protein Data Bank) has been established in 1971 [5], and more than 170,000 biological macromolecular structures have been registered in the PDB. About 85% of the biological macromolecule structures registered in the PDB were determined by X-rays [6].
X-ray crystallography is the most advantageous technique for determining protein structure [7]. It typically consists of nine steps: target protein selection, genetic engineering for increase the quantity of target proteins, purification, crystallization, treatment with cryoprotectants before freezing via liquid nitrogen, data collection, calculating the electron density map, refinement & validation, and finally the model building of target proteins [8] (Figure 1). The refined 3D structure of target protein gives new ideas to creating mutants of the target protein, i.e., the better functioning target protein in the cell.
Protein crystals kept at room temperature dramatically deteriorate when they are exposed to high-intensity X-rays [10]. Therefore, protecting and/or lowering radiation damage of protein crystals is essential for collecting better data sets. Sometimes it is possible for protein crystals exposed at 100 K to tolerate a much longer total X-ray exposure without seriously affecting the diffraction quality, so it can generally collect more than one complete dataset from a single crystal [11]. However, protein crystals also contain water, which transforms into hexagonal ice during cooling, expanding its volume [12]. Therefore, a cryoprotectant, such as sugars, glycerol, ethylene glycol, and polyethylene glycols (PEG) [13,14], that induces protein dehydration is essential (Figure 2) [15].
Caution should be exercised because high concentration cryoprotectants may also cause various side effects to protein crystals, such as volume reduction and morphology change of protein crystals, reductions of scattering contrasts, and an increase in solvent viscosity. However, the studies of cryoprotectants are few. This short review introduces widely used cryoprotectants, the cause and/or effect of cryoprotectants during cryoprotection, and their effectiveness on the diffraction quality of protein crystals.

2. What Is Cryoprotection, and When Do We Use Cryoprotectants?

Cryoprotectants inhibit and preserve protein crystal damage during freezing. Cryoprotection is the immersion of crystals in cryosolution for a short period, typically less than 10 s or longer than 1 day [16,17,18]. Some cryoprotectants may dissolve protein crystals; therefore, soaking times (damage) should be stringently monitored [19,20]. Cryoprotectant use also depends on crystallization conditions. Since the crystallization precipitant is almost identical to the cryoprotectant, the mother liquor is used for cryoprotection. If the precipitant concentration of the mother liquor is insufficient to protect the crystal, an additional cryoprotectant can be added.
Various types of cryoprotectants, such as salts, organic solvents, sugars, polyols, and polymers, are listed in Table 1. In many cases, the low molecular weight PEGs, such as PEG 200, 400, and 600, are appropriate cryoprotectants. Low molecular weight polyols (PEGs), sugars, and organic solvents penetrate the crystal lattice and cause dehydration [19,21,22]. Polymers, including high molecular weight PEGs, such as PEG 3350, 4000, and 6000, do not penetrate crystals but coat the crystal lattice, and are referred to as non-penetrating cryoprotectants [23]. Similarly, oils do not penetrate crystals but provide barriers between the crystal surface and air. Alcohols and salts also act as dehydration molecules via vapor and osmotic effects.
In PDB structures reported in approximately 1000 publications (2015–2021), polymers have been used as precipitating agents in 63% of structures (Figure S1a,b). Crystallization using organic compounds and salts as precipitants were used in 22% (Figure S1a,c) and 15% (Figure S1a,d) of structures, respectively. Since polymer- and organic compound-mediated crystallization has been performed at sufficient concentrations for cryo-cooling, such cryoprotectants are used less frequently when compared with salt crystallization approaches (63%, Figure S2a). The most frequently applied added cryoprotectant was penetrating cryoprotectants such as glycerol and/or EG (Figure S2b). This statistic result was similar to previous research result by Farley, C & Juers, DH [24]. However, cryoprotection using vapor and osmotic effects is rarely used (Figure S2b).
Thermal contractions are classified with contraction coefficients according to the solvent temperature. The thermal contraction value of cryoprotectants at cryogenic temperatures is important for determining the concentration of penetrating cryoprotectant for successful cryoprotection. Indeed, glycerol, as a popular cryoprotectant, is used at 25–30%, which is the concentration of glycerol to the equilibrium between extension and contraction values at 77 K [25]. Further, 20% DMSO is close to the equilibrium between extension and contraction values at 77 K; however, DMSO is limited due to its biochemical toxicity.

3. X-ray Damage and Diffraction Quality

The damage to protein crystals by X-rays occurs in stages. Nave [26] reported that radiation induces damage to crystals either directly or indirectly. In crystal damage, direct molecular destruction by radiation is called primary damage. Secondary damage refers to the damage caused by free radicals produced by direct and/or indirect causes after radiation exposure. In addition, the crystal lattice may become unstable or destroyed due to primary and secondary damage. This phenomenon is called the domino phenomenon [27].
Various studies have reported the damage to protein crystals caused by X-rays [28,29,30,31]. Alkire et al. reported that various commercial cryoloops determine the differ diffraction data quality according to the stiffness and aperture diameter of the cryoloop [32]. Michael et al. developed crystals in cryoloops and collected diffraction data at 100 K in cryoprotection-free conditions. These authors suggested this method was advantageous as it could lead to technical advances, such as automated systems for protein structural analyses using X-rays, crystallization, cryoprotection, and data collection [33].
Although the cryoprotection condition is more limited than the crystallization condition (commercial crystallization screening kits: about 2500 and cryoprotection screening kits: about 100) (Table S1), it is evident that one of the important factors in preventing the damage and affecting the diffraction pattern is crystal cooling. If this is processed incorrectly, i.e., it causes ice rings within the diffraction pattern and damages the crystal itself. Crystals were grown using ammonium sulfate (2.8–3.2 M) causes ice rings and low data quality, despite being able to protect the crystals by the osmotic effect (Figure 3a,b). However, mis-cryoprotection can be improved using glycerol and various cryoprotectants (Figure 3c,d). Glycerol, which is popularly used for cryoprotection, affects carbonic anhydrase II activity. However, conformational changes in the carbonic anhydrase II structure are not identified in active and substrate binding sites, despite glycerol being used as a cryoprotectant [34].

4. How to Improve the Diffraction Quality of Protein Crystal?

Sometimes, high intensity of X-ray beam induces the poor quality of X-ray data and unsuitable for diffraction studies. These can be overcome by internal factors of synchrotron, such as attenuation time, oscillation range, and exposure time, but they cannot be a fundamental solution [35]. The post crystallization method can help to solve this problem. Post crystallization methods can convert poor diffracting crystals into data quality crystals through the various cryoprotection method [7].
Crystal annealing reduces the disorder of the flash cooling method and can increase the diffraction quality of the protein crystals [36]. There are three different types of crystal annealing: macromolecular crystal annealing (MCA) and flash annealing (FA). MCA removes a cryocooled crystal from the cold gas stream. It places in 300 mL cryo-solution, and after 3 min of equilibration, the crystal is re-cooled in cryo-stream [36]. The FA method blocks the cold-stream for 1.5–2 s three times and 6 s for the interval of each thawing step [37]. MCA treatment is better and more reproducible than others, the FA method is defective for most of the crystals [38].
Crystal dehydration is a treatment that produces the most remarkable improvements in the diffraction resolution of protein crystals [39]. Dehydration removes the excess solvent, tightens the protein molecules, and reduces the size of solvent channels. By this way, it improves the crystal order and diffraction resolution. Many studies have been reported that crystal dehydration method can convert poor quality crystals (>10 Å) to high-resolution data quality crystals (<2 Å) [40,41,42,43,44,45]. Removing excess solvent can make successful flash cooling easy, especially for large initial solvent contents crystals. After dehydration, protein crystals change the structural transformation and yield the alternative crystal packing; it is not possible or difficult to achieve directly during the crystal growth stage. Therefore, crystal dehydration is the most effective treatment for improving the crystal diffraction properties than the other post crystallization treatments [7].
Crystal soaking is similar to dehydration treatment. Dehydration can cause the shrinking of the crystal lattice and lower the solvent content of the crystal. Crystal soaking treatment without changing the unit cell or solvent content and notably improves the crystal diffraction quality. A higher concentration of precipitating agents can increase the quality of the diffraction resolution without evaporation or decrease the unit cell parameters. Glycerol is the most widely used cryoprotecting agent in crystal soaking solutions to improve the quality of protein crystals [46].
In the multistep soaking method, which is one of the alternative methods, crystals are soaked 2–3 times using different cryoprotectant solutions [47]. This method is considered useful when the diffraction pattern is not optimized by a commercially available cryoprotectant kit. When either of the two cryoprotectants has only a marginal effect on the protein crystal, the combining of the cryoprotectants (two or sometimes more) show a significant improvement in crystal quality. This method, however, may require as much time as that required by the previous method because it searches for the second and subsequent cryoprotectants.
The other approach is reported to increase the diffraction data quality in cryoprotection process. High-pressure cryocooling has been devised as an alternative method and has been successfully applied in various technical and scientific studies for cryoprotection of protein crystals [48,49,50]. This method requires the conservation of crystal hydration because pressure is applied to the crystal using dry helium gas. This method involves mounting the protein crystals from the native mother liquor onto a cryoloop using a droplet of oil, applying a pressure of 200 MPa in He gas to the crystals, cooling the crystal under pressure and then releasing the pressure. The crystal is then removed from the apparatus under liquid nitrogen and handled thereafter in a manner similar to a normal cryocooled crystal. These methods are not sufficiently adequate for low resolution diffracting crystals since the relatively high background scattering due to the hydrating materials.

5. The Problem in Structural Analysis

The chemical properties of cryoprotectants can absorb radicals generated by radiation. It has also been reported to reduce damage to crystals, primarily due to changes in protein-protein interactions [51]. However, added molecules, such as cryoprotectants, should not affect the protein structure except for the ligand. It rarely occurs using the penetrate cryoprotectant.
Kim showed that trehalose induces morphological changes in the structure of GAPDH through binding to S-loop [52]. Trehalose is one of the cryoprotectants that induces inhibiting protein hydration [53]. The apo- and holo-structures of GAPDH rotate 2.4° and 3.1°, respectively, compared to the trehalose bonded structure. There are two hypotheses. One is that the change in shape is a result of the penetration of the cryoprotectant. Some penetrating cryoprotectants, such as low-weight polymers, polyols, and saccharide groups, bind to the protein structure. The GAPDH structure in the PDB has been reported to bind to GAPDH as a cryoprotectant. However, the cryoprotectant does not affect key domains such as the active site and protein interaction domain. Interestingly, trehalose binds to the S-loop of GAPDH, a key residue in the protein-protein interaction [54,55]. In contrast, other cryoprotectants such as maltose and sucrose do not bind GAPDH in Kim’s work. It is supposed that the protein structure might undergo a conformational change in some cases through cryoprotection because of hydration and dehydration in internal cavities as well as protein surface [56]. In these reasons, the collection of X-ray diffraction has been conducted at room temperature (RT), recently.
X-ray diffraction data of dihydrofolate reductase (DHFR) were collected at both RT and cryogenic temperatures (CT). As expected, CT data set resolution was higher than the RT data set. However, while more ensemble water molecules were generated at RT, interestingly, the number of water molecules interacting with proteins was greater at CT. In addition, Arg 52 residues interacting with folate were more flexible at RT [57]. There were also reported that the hidden ligands binding site neither active nor catalytic site binding site of some protein could be revealed in RT conditions, but not in CT conditions [58,59]. Previous studies also reported that crystallography conducted using serial femtoseconds and microfluidic platforms were useful during RT conditions, but required a large amount of protein crystals for measurement [60]. In addition, the crystal damage of X-ray beam is still unsolved. Therefore, it is necessary to find suitable cryoprotectants and methods when proceeding with crystal cooling [61,62,63,64].

6. Conclusions

Cryocrystallographic methods are essential in protein crystallography. In X-ray crystallography, cryoprotection plays an important role in improving data quality. Recently, cryoprotection has been tried under various conditions, such as the multistep soaking method and high pressure, to improve data quality. However, cryoprotectants can also affect the structure of the protein. Due to various factors, the choice of cryoprotectant depends on practical experience. Therefore, this short review could have a positive effect as an empirical basis for the future in X-ray crystallography.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/cryst12020138/s1, Figure S1. The statistic analysis of precipitants for crystallization in the structures reported to the PDB in the period of 2015 to 2021, Figure S2. The use of additional cryoproteiction, Table S1. Lists of commercially available cryoprotectants from two representative companies

Author Contributions

Conceptualization, K.J., H.G.K., H.-W.C. and Y.J.K.; investigation, K.J., H.G.K. and S.H.S.H.; visualization, K.J., H.G.K., and M.K.; writing—original draft resources, K.J., H.G.K., H.-W.C. and Y.J.K. writing—review and editing, H.-W.C. and Y.J.K.; funding acquisition, Y.J.K.; All authors have read and agreed to the published version of the manuscript.

Funding

This research was supported by the Bio & Medical Technology Development Program and ICT(NRF-2017M3A9F6029733) and Basic Science Research Program of the National Research Foundation (NRF) funded by the Ministry of Science (NRF-2021R1I1A3060013).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

Y.J. Kim is grateful to Pohang Accelerator Laboratory [21] where the data were collected on beamline 7A. H.-W. Choe thanks JBNU and DGIST for his chair professorship in the period of 2014–2021 at JBNU and in the period of 2017–2019 at DGIST. He is currently working as an emeritus professor at the Department of Chemistry, College of Natural Science, JBNU, Republic of Korea. S.H.S. Hlaing thanks GKS (Global Korea Scholarship, GKS.G.2019.429) which is currently sponsored by the Korean Government.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Funke, O. Über das Milzvenenblut. Z. Rat. Med. 1851, 1, 172–179. [Google Scholar]
  2. Hunefeld, F. Die Chemismus in der Thierischen Organization; Brockhaus: Leipzig, Germany, 1840; pp. 158–163. [Google Scholar]
  3. Ritthausen, H. Über die Eiweisskorper verschiedenen Oelsamen. Pfluegers Arch. 1880, 21, 81–104. [Google Scholar] [CrossRef]
  4. Osborne, T. The proteids of the Brazil nut. Am. Chem. J. 1891, 13, 212–218. [Google Scholar]
  5. Protein Data Bank. Protein data bank. Nat. New Biol. 1971, 233, 233. [Google Scholar]
  6. wwPDB Consortium. Protein Data Bank: The single global archive for 3D macromolecular structure data. Nucleic Acids Res. 2018, 47, D520–D528. [Google Scholar]
  7. Kim, Y.J.; Hofmann, K.P.; Ernst, O.P.; Scheerer, P.; Choe, H.-W.; Sommer, M.E. Crystal structure of pre-activated arrestin p44. Nature 2013, 497, 142–146. [Google Scholar] [CrossRef]
  8. Heras, B.; Martin, J.L. Post-crystallization treatments for improving diffraction quality of protein crystals. Acta Crystallogr. Sect. D Biol. Crystallogr. 2005, 61, 1173–1180. [Google Scholar] [CrossRef] [Green Version]
  9. Chayen, N.E.; Saridakis, E. Protein crystallization: From purified protein to diffraction-quality crystal. Nat. Methods 2008, 5, 147–153. [Google Scholar] [CrossRef]
  10. Garman, E.F.; Owen, R.L. Cryocooling and radiation damage in macromolecular crystallography. Acta Crystallogr. Sect. D Biol. Crystallogr. 2006, 62, 32–47. [Google Scholar] [CrossRef] [Green Version]
  11. Pflugrath, J.W. Practical macromolecular cryocrystallography. Acta Cryst. F Struct. Biol. Commun. 2015, 71, 622–642. [Google Scholar] [CrossRef]
  12. Kriminski, S.; Caylor, C.L.; Nonato, M.C.; Finkelstein, K.D.; Thorne, R.E. Flash-cooling and annealing of protein crystals. Acta Crystallogr. Sect. D Biol. Crystallogr. 2002, 58, 459–471. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Maleknia, S.D.; Ralston, C.Y.; Brenowitz, M.D.; Downard, K.M.; Chance, M.R. Determination of macromolecular folding and structure by synchrotron x-ray radiolysis techniques. Anal. Biochem. 2001, 289, 103–115. [Google Scholar] [CrossRef]
  14. O’Neill, P.; Stevens, D.L.; Garman, E. Physical and chemical considerations of damage induced in protein crystals by synchrotron radiation: A radiation chemical perspective. J. Synchrotron Radiat. 2002, 9, 329–332. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Xiao, B.; Gamblin, S.J. The effects of cryoprotectant on crystal stability. J. Cryst. Growth 1996, 168, 244–247. [Google Scholar] [CrossRef]
  16. Garman, E. Cool data: Quantity AND quality. Acta Crystallogr. Sect. D Biol. Crystallogr. 1999, 55, 1641–1653. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Petock, J.M.; Wang, Y.-F.; DuBois, G.C.; Harrison, R.W.; Weber, I.T. Effects of different post-crystallization soaking conditions on the diffraction of Mtcp1 crystals. Acta Crystallogr. Sect. D Biol. Crystallogr. 2001, 57, 763–765. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  18. Walker, L.J.; Moreno, P.O.; Hope, H. Cryocrystallography: Effect of cooling medium on sample cooling rate. J. Appl. Crystallogr. 1998, 31, 954–956. [Google Scholar] [CrossRef] [Green Version]
  19. Vera, L.; Stura, E.A. Strategies for protein cryocrystallography. Cryst. Growth Des. 2013, 14, 427–435. [Google Scholar] [CrossRef]
  20. López-Jaramillo, F.; Moraleda, A.; González-Ramírez, L.; Carazo, A.; García-Ruiz, J. Soaking: The effect of osmotic shock on tetragonal lysozyme crystals. Acta Crystallogr. Sect. D Biol. Crystallogr. 2002, 58, 209–214. [Google Scholar] [CrossRef] [Green Version]
  21. Anand, K.; Pal, D.; Hilgenfeld, R. An overview on 2-methyl-2, 4-pentanediol in crystallization and in crystals of biological macromolecules. Acta Crystallogr. Sect. D Biol. Crystallogr. 2002, 58, 1722–1728. [Google Scholar] [CrossRef]
  22. Vera, L.; Czarny, B.; Georgiadis, D.; Dive, V.; Stura, E.A. Practical use of glycerol in protein crystallization. Cryst. Growth Des. 2011, 11, 2755–2762. [Google Scholar] [CrossRef]
  23. Farley, C.; Juers, D.H. Efficient cryoprotection of macromolecular crystals using vapor diffusion of volatile alcohols. J. Struct. Biol. 2014, 188, 102–106. [Google Scholar] [CrossRef] [Green Version]
  24. Garman, E.F.; Doublié, S. Cryocooling of macromolecular crystals: Optimization methods. Methods Enzymol. 2003, 368, 188–216. [Google Scholar]
  25. Alcorn, T.; Juers, D.H. Progress in rational methods of cryoprotection in macromolecular crystallography. Acta Crystallogr. Sect. D Biol. Crystallogr. 2010, 66, 366–373. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Nave, C. Radiation damage in protein crystallography. Radiat. Phys. Chem. 1995, 45, 483–490. [Google Scholar] [CrossRef]
  27. Henderson, R. Cryo-protection of protein crystals against radiation damage in electron and X-ray diffraction. Proc. R. Soc. Lond. Ser. B Biol. Sci. 1990, 241, 6–8. [Google Scholar]
  28. Moffat, K.; Bilderback, D.; Schildkamp, W.; Volz, K. Laue diffraction from biological samples. Nucl. Instrum. Methods Phys. Res. Sect. A Accel. Spectrometers Detect. Assoc. Equip. 1986, 246, 627–635. [Google Scholar] [CrossRef]
  29. Moffat, K. Time-resolved macromolecular crystallography. Annu. Rev. Biophys. Biophys. Chem. 1989, 18, 309–332. [Google Scholar] [CrossRef] [PubMed]
  30. Getzoff, E.; Jones, K.; McRee, D.; Moffat, K.; Ng, K.; Rivers, M.; Schildkamp, W.; Singer, P.; Spanne, P.; Sweet, R. Laue diffraction protein crystallography at the National Synchrotron Light Source. Nucl. Instrum. Methods Phys. Res. Sect. B Beam Interact. Mater. At. 1993, 79, 249–255. [Google Scholar] [CrossRef] [Green Version]
  31. Gonzalez, A.; Nave, C. Radiation damage in protein crystals at low temperature. Acta Crystallogr. Sect. D Biol. Crystallogr. 1994, 50, 874–877. [Google Scholar] [CrossRef] [PubMed]
  32. Alkire, R.; Rotella, F.; Duke, N. Testing commercial protein crystallography sample mounting loops for movement in a cold-stream. J. Appl. Crystallogr. 2013, 46, 525–536. [Google Scholar] [CrossRef]
  33. Berger, M.A.; Decker, J.H.; Mathews, I.I. Diffraction study of protein crystals grown in cryoloops and micromounts. J. Appl. Crystallogr. 2010, 43, 1513–1518. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Aggarwal, M.; Boone, C.D.; Kondeti, B.; Tu, C.; Silverman, D.N.; McKenna, R. Effects of cryoprotectants on the structure and thermostability of the human carbonic anhydrase II–acetazolamide complex. Acta Crystallogr. Sect. D Biol. Crystallogr. 2013, 69, 860–865. [Google Scholar] [CrossRef] [Green Version]
  35. Teng, T.Y.; Moffat, K. Primary radiation damage of protein crystals by an intense synchrotron X-ray beam. J. Synchrotron Radiat. 2000, 7, 313–317. [Google Scholar] [CrossRef]
  36. Harp, J.M.; Timm, D.E.; Bunick, G.J. Macromolecular crystal annealing: Overcoming increased mosaicity associated with cryocrystallography. Acta Crystallogr. Sect. D Biol. Crystallogr. 1998, 54, 622–628. [Google Scholar] [CrossRef] [PubMed]
  37. Yeh, J.I.; Hol, W.G. A flash-annealing technique to improve diffraction limits and lower mosaicity in crystals of glycerol kinase. Acta Crystallogr. Sect. D Biol. Crystallogr. 1998, 54, 479–480. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  38. Harp, J.M.; Hanson, B.L.; Timm, D.E.; Bunick, G.J. Macromolecular crystal annealing: Evaluation of techniques and variables. Acta Crystallogr. Sect. D Biol. Crystallogr. 1999, 55, 1329–1334. [Google Scholar] [CrossRef] [PubMed]
  39. Heras, B.; Edeling, M.A.; Byriel, K.A.; Jones, A.; Raina, S.; Martin, J.L. Dehydration converts DsbG crystal diffraction from low to high resolution. Structure 2003, 11, 139–145. [Google Scholar] [CrossRef]
  40. Cramer, P.; Müller, C.W. Engineering of diffraction-quality crystals of the NF-κB P52 homodimer: DNA complex. FEBS Lett. 1997, 405, 373–377. [Google Scholar] [CrossRef] [Green Version]
  41. Pang, S.S.; Guddat, L.W.; Duggleby, R.G. Crystallization of the FAD-independent acetolactate synthase of Klebsiella pneumoniae. Acta Crystallogr. Sect. D Biol. Crystallogr. 2002, 58, 1237–1239. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Kodandapani, R.; Vijayan, M. Protein hydration and water structure: X-ray analysis of a closely packed protein crystal with very low solvent content. Acta Crystallogr. Sect. D Biol. Crystallogr. 1993, 49, 234–245. [Google Scholar]
  43. Dobrianov, I.; Kriminski, S.; Caylor, C.; Lemay, S.; Kimmer, C.; Kisselev, A.; Finkelstein, K.; Thorne, R. Dynamic response of tetragonal lysozyme crystals to changes in relative humidity: Implications for post-growth crystal treatments. Acta Crystallogr. Sect. D Biol. Crystallogr. 2001, 57, 61–68. [Google Scholar] [CrossRef] [Green Version]
  44. Fu, Z.-Q.; Du Bois, G.C.; Song, S.P.; Harrison, R.W.; Weber, I.T. Improving the diffraction quality of MTCP-1 crystals by post-crystallization soaking. Acta Crystallogr. Sect. D Biol. Crystallogr. 1999, 55, 5–7. [Google Scholar] [CrossRef]
  45. Izard, T.; Sarfaty, S.; Westphal, A.; De Kok, A.; Hol, W.G. Improvement of diffraction quality upon rehydration of dehydrated icosahedral Enterococcus faecalis pyruvate dehydrogenase core crystals. Protein Sci. 1997, 6, 913–915. [Google Scholar] [CrossRef] [Green Version]
  46. Sousa, R. Use of glycerol, polyols and other protein structure stabilizing agents in protein crystallization. Acta Crystallogr. Sect. D Biol. Crystallogr. 1995, 51, 271–277. [Google Scholar] [CrossRef] [PubMed]
  47. Senda, M.; Hayashi, T.; Hatakeyama, M.; Takeuchi, K.; Sasaki, A.T.; Senda, T. Use of multiple cryoprotectants to improve diffraction quality from protein crystals. Cryst. Growth Des. 2016, 16, 1565–1571. [Google Scholar] [CrossRef]
  48. Kim, C.U.; Kapfer, R.; Gruner, S.M. High-pressure cooling of protein crystals without cryoprotectants. Acta Crystallogr. Sect. D Biol. Crystallogr. 2005, 61, 881–890. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  49. Kim, C.U.; Hao, Q.; Gruner, S.M. High-pressure cryocooling for capillary sample cryoprotection and diffraction phasing at long wavelengths. Acta Crystallogr. Sect. D Biol. Crystallogr. 2007, 63, 653–659. [Google Scholar] [CrossRef]
  50. Kim, C.U.; Wierman, J.L.; Gillilan, R.; Lima, E.; Gruner, S.M. A high-pressure cryocooling method for protein crystals and biological samples with reduced background X-ray scatter. J. Appl. Crystallogr. 2013, 46, 234–241. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  51. Kuwamoto, S.; Akiyama, S.; Fujisawa, T. Radiation damage to a protein solution, detected by synchrotron X-ray small-angle scattering: Dose-related considerations and suppression by cryoprotectants. J. Synchrotron Radiat. 2004, 11, 462–468. [Google Scholar] [CrossRef]
  52. Kim, Y.J. A cryoprotectant induces conformational change in glyceraldehyde-3-phosphate dehydrogenase. Acta Cryst. F Struct. Biol. Commun. 2018, 74, 277–282. [Google Scholar] [CrossRef] [PubMed]
  53. Corradini, D.; Strekalova, E.G.; Stanley, H.E.; Gallo, P. Microscopic mechanism of protein cryopreservation in an aqueous solution with trehalose. Sci. Rep. 2013, 3, 1218. [Google Scholar] [CrossRef] [PubMed]
  54. Fermani, S.; Trivelli, X.; Sparla, F.; Thumiger, A.; Calvaresi, M.; Marri, L.; Falini, G.; Zerbetto, F.; Trost, P. Conformational selection and folding-upon-binding of intrinsically disordered protein CP12 regulate photosynthetic enzymes assembly. J. Biol. Chem. 2012, 287, 21372–21383. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Omar Abuzaid, A.; Elena López-Calcagno, P.; Lawson, T.; Anne Raines, C. Arabidopsis CP12 mutants have reduced levels of phosphoribulokinase and impaired function of the Calvin–Benson cycle. J. Exp. Bot. 2017, 68, 2285–2298. [Google Scholar] [CrossRef] [Green Version]
  56. Skrzypczak-Jankun, E.; Borbulevych, O.; Zavodszky, M.; Baranski, M.; Padmanabhan, K.; Petricek, V.; Jankun, J. Effect of crystal freezing and small-molecule binding on internal cavity size in a large protein: X-ray and docking studies of lipoxygenase at ambient and low temperature at 2.0 Å resolution. Acta Crystallogr. Sect. D Biol. Crystallogr. 2006, 62, 766–775. [Google Scholar] [CrossRef]
  57. Keedy, D.A.; Van Den Bedem, H.; Sivak, D.A.; Petsko, G.A.; Ringe, D.; Wilson, M.A.; Fraser, J.S. Crystal cryocooling distorts conformational heterogeneity in a model Michaelis complex of DHFR. Structure 2014, 22, 899–910. [Google Scholar] [CrossRef] [Green Version]
  58. Fischer, M.; Shoichet, B.K.; Fraser, J.S. One crystal, two temperatures: Cryocooling penalties alter ligand binding to transient protein sites. ChemBioChem 2015, 16, 1560. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  59. Fraser, J.S.; Clarkson, M.W.; Degnan, S.C.; Erion, R.; Kern, D.; Alber, T. Hidden alternative structures of proline isomerase essential for catalysis. Nature 2009, 462, 669–673. [Google Scholar] [CrossRef]
  60. Maeki, M.; Ito, S.; Takeda, R.; Ueno, G.; Ishida, A.; Tani, H.; Yamamoto, M.; Tokeshi, M. Room-temperature crystallography using a microfluidic protein crystal array device and its application to protein–ligand complex structure analysis. Chem. Sci. 2020, 11, 9072–9087. [Google Scholar] [CrossRef]
  61. Samygina, V.R.; Antonyuk, S.V.; Lamzin, V.S.; Popov, A.N. Improving the X-ray resolution by reversible flash-cooling combined with concentration screening, as exemplified with PPase. Acta Crystallogr. Sect. D Biol. Crystallogr. 2000, 56, 595–603. [Google Scholar] [CrossRef] [Green Version]
  62. Rodgers, D.W. Cryocrystallography. Structure 1994, 2, 1135–1140. [Google Scholar] [CrossRef] [Green Version]
  63. Garman, E.F.; Schneider, T.R. Macromolecular cryocrystallography. J. Appl. Crystallogr. 1997, 30, 211–237. [Google Scholar] [CrossRef] [Green Version]
  64. Juers, D.H.; Matthews, B.W. Cryo-cooling in macromolecular crystallography: Advantages, disadvantages and optimization. Q. Rev. Biophys. 2004, 37, 105. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Overall processes of protein structure analysis using X-ray. p44 (PDB entry: 4J2Q [9]) also known as a splicing variant of arrestin that is actively working on in our laboratory, has been used for this figure, i.e.,SDS-PAGE, crystal, diffraction pattern, electron density map and final model).
Figure 1. Overall processes of protein structure analysis using X-ray. p44 (PDB entry: 4J2Q [9]) also known as a splicing variant of arrestin that is actively working on in our laboratory, has been used for this figure, i.e.,SDS-PAGE, crystal, diffraction pattern, electron density map and final model).
Crystals 12 00138 g001
Figure 2. The basic principle of cryoprotection in protein crystals. Directly frozen and/or mis-selected cryoprotectant used protein crystals lead to ice rings on the diffraction caused by cooling environment. The reason is that water molecules contained in protein crystals undergo phase transition from water to hexagonal ice through rapid cooling with liquid nitrogen. This causes the volume of protein crystals to expand, destroys the crystal lattice, and leads to lowering the quality of data. Therefore, proper treatment with cryoprotectants in protein crystals is indispensable for better quality X-ray crystal data.
Figure 2. The basic principle of cryoprotection in protein crystals. Directly frozen and/or mis-selected cryoprotectant used protein crystals lead to ice rings on the diffraction caused by cooling environment. The reason is that water molecules contained in protein crystals undergo phase transition from water to hexagonal ice through rapid cooling with liquid nitrogen. This causes the volume of protein crystals to expand, destroys the crystal lattice, and leads to lowering the quality of data. Therefore, proper treatment with cryoprotectants in protein crystals is indispensable for better quality X-ray crystal data.
Crystals 12 00138 g002
Figure 3. The importance of cryoprotection in diffraction quality. Incorrect cryoprotection of GAPDH protein forms an ice ring and lowering the quality of X-ray data (a,b). However, correct selection of cryoprotection for GAPDH crystal leads to much better quality of X-ray data (c,d).
Figure 3. The importance of cryoprotection in diffraction quality. Incorrect cryoprotection of GAPDH protein forms an ice ring and lowering the quality of X-ray data (a,b). However, correct selection of cryoprotection for GAPDH crystal leads to much better quality of X-ray data (c,d).
Crystals 12 00138 g003
Table 1. The types of cryoprotectants.
Table 1. The types of cryoprotectants.
Penetrate
 Sugars (25~50%)
 Glucose Sucrose Maltose Xylitol Inositol
 Trehalose Raffinose Erythriol
 Polyols (25~50%)
 Glycerol Ethylene glycol Diethylene glycol Propylene glycerol 1,2-propanediol
 2,3-butanediol 1,6-hexanediol 2-methyl-2,4-pentanediol
 Low weight PEGs (25~50%)
 PEG 200 PEG 400 PEG 600 etc
Non-penetrate
 High weigh PEGs (25~50%)
 PEG 3350 PEG 6000 PEG 8000 etc.
 Oils
 Paraffin oil Olive oil Canola oil Perfluoropolyether oil,
 Parabar 10312 Turbomolecular pump oil
Others
 Vapor phase (alcohols)
 Methanol Ethanol Isopropanol
 Osmotics (salts, 50~90%)
 Lithium acetate Lithium chloride Lithium formate Sodium malonate Magnesium acetate
 Sodium chloride Sodium formate Sodium nitrate Ammonium sulfate
(concentration)
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Jang, K.; Kim, H.G.; Hlaing, S.H.S.; Kang, M.; Choe, H.-W.; Kim, Y.J. A Short Review on Cryoprotectants for 3D Protein Structure Analysis. Crystals 2022, 12, 138. https://doi.org/10.3390/cryst12020138

AMA Style

Jang K, Kim HG, Hlaing SHS, Kang M, Choe H-W, Kim YJ. A Short Review on Cryoprotectants for 3D Protein Structure Analysis. Crystals. 2022; 12(2):138. https://doi.org/10.3390/cryst12020138

Chicago/Turabian Style

Jang, Kiyoung, Hyun Gi Kim, Sandi Hnit San Hlaing, MinSoung Kang, Hui-Woog Choe, and Yong Ju Kim. 2022. "A Short Review on Cryoprotectants for 3D Protein Structure Analysis" Crystals 12, no. 2: 138. https://doi.org/10.3390/cryst12020138

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop