Skip to main content
Protein Science : A Publication of the Protein Society logoLink to Protein Science : A Publication of the Protein Society
. 2009 Jan;18(1):24–36. doi: 10.1002/pro.3

Effect of trehalose on protein structure

Nishant Kumar Jain 1, Ipsita Roy 1,*
PMCID: PMC2708026  PMID: 19177348

Abstract

Trehalose is a ubiquitous molecule that occurs in lower and higher life forms but not in mammals. Till about 40 years ago, trehalose was visualized as a storage molecule, aiding the release of glucose for carrying out cellular functions. This perception has now changed dramatically. The role of trehalose has expanded, and this molecule has now been implicated in a variety of situations. Trehalose is synthesized as a stress-responsive factor when cells are exposed to environmental stresses like heat, cold, oxidation, desiccation, and so forth. When unicellular organisms are exposed to stress, they adapt by synthesizing huge amounts of trehalose, which helps them in retaining cellular integrity. This is thought to occur by prevention of denaturation of proteins by trehalose, which would otherwise degrade under stress. This explanation may be rational, since recently, trehalose has been shown to slow down the rate of polyglutamine-mediated protein aggregation and the resultant pathogenesis by stabilizing an aggregation-prone model protein. In recent years, trehalose has also proved useful in the cryopreservation of sperm and stem cells and in the development of a highly reliable organ preservation solution. This review aims to highlight the changing perception of the role of trehalose over the last 10 years and to propose common mechanisms that may be involved in all the myriad ways in which trehalose stabilizes protein structures. These will take into account the structure of trehalose molecule and its interactions with its environment, and the explanations will focus on the role of trehalose in preventing protein denaturation.

Keywords: anhydrobiosis, carbohydrates, protein structure, trehalose

Introduction

The use of osmolytes in the stabilization of biomolecules is an old trick of nature. These small molecules counteract the various stress conditions that an organism encounters. The mechanism by which this stabilization occurs is not fully understood; in fact, there may not be a universal rule that works for all compatible solutes. These solutes range from sugars to polyols, amino acids and their derivatives, and so forth.1,2 Among these, the interest in trehalose has shown periodic crests and troughs with various theories being advanced to explain the astonishing properties that have, at times, been attributed to it. Although a number of articles reporting the stabilization of various biomolecules by trehalose (and in some cases, sucrose) have appeared in the recent literature, none of them has attempted to unravel the common thread that may link the various studies together. For example, studies that concentrate on the physical or chemical properties of trehalose and its solution pay little attention to finding out a correlation with biological activity.311 Similarly, articles reporting gene expression profiling relevant to trehalose as a bioprotector do not make a serious attempt to relate the protective action to physical and chemical properties of the molecule.1217 This review attempts to bridge this gap and to find out how data from physicists, chemists, and life scientists can be collected and collated so that an understanding of general interest to protein scientists may emerge.

Trehalose is a white, odorless powder with relative sweetness 45% that of sucrose. It is a bisacetal, nonreducing homodisaccharide in which two glucose units are linked together in a α-1,1-glycosidic linkage (α-d-glucopyranosyl-α-d-glucopyranoside; mycose, mushroom sugar). Some of the properties of trehalose are listed in Table I. Because of the inherent properties of trehalose, namely prevention of starch retrogradation and stabilization of proteins and lipids, it has proved quite useful in a number of industries including food processing, cosmetics, pharmaceutics, and so forth.18,19 The restraining factor in the industrial use of trehalose has been the high cost of manufacturing the sugar. With the development of an enzyme-based method (from Arthrobacter sp. Q36) for trehalose synthesis,20 the use is expected to increase. US Food and Drug Administration granted generally recognized as safe status to the disaccharide in 2000, which will certainly increase the scope of the areas where this disaccharide is being used.

Table I.

Physical and Chemical Properties of Trehalose

Parameter Value
Chemical formula C12H22O11 (anhydrous)
C12H22O11·2H2O (dihydrate)
Molecular weight 342.3 g mol−1 (anhydrous)
378.3 g mol−1 (dihydrate)
Appearance White, odourless powder
Sweetness (relative) 45% that of sucrose
Polymorphs Two crystalline forms: anhydrous (Tα, Tβ) and dihydrate (Th), one well-defined amorphous form
Chemical nature Nonreducing disaccharide (α-d-glucopyranosyl-α-d- glucopyranoside)
Relative density 1.22 g cm−3 (at 25°C and weight fraction = 0.5)
Glass transition temperature 117°C
Thermostability >99% (120°C/90 min)
Melting point 203°C (anhydrous)
97°C (dihydrate)
Heat of fusion 53.4 kJ mol−1 (anhydrous)
57.8 kJ mol−1 (dihydrate)
Hygroscopicity None at <90% RH
Solubility in water 68.9 g/100 g (at 20°C)
Freezing temperature (100 mg mL−1 water) −197°C
Optical rotation [α]D = +178°C
Viscosity 8.2 cp (at 40°C and weight fraction = 0.5)
Chemical reactivity Does not usually react with proteins
Regulatory status GRAS (generally regarded as safe) by US FDA

RH, relative humidity.

Trehalose is present quite widely in the biological world14,19 and performs different functions (Table II). For example, in prokaryotes, trehalose is thought to serve as a carbon source, a structural component, or as a compatible solute in halophiles and cyanobacteria.47 In Escherichia coli, trehalose is synthesized in response to high osmolarity.22 On the other hand, Bacillus subtilis utilizes trehalose only as a carbon source and not for osmoregulation.21 The disaccharide is also present in many other species of bacteria, including Streptomyces, where trehalose is stored in the aerial hyphae and spores.24 It forms a part of the cell wall in different species of mycobacteria, including Mycobacterium tuberculosis and the phylogenetically related corynaebacteria (including Corynebacterium glutamicum).12,23 Free trehalose is also found in the latter during hyperosmotic stress.

Table II.

Applications of Trehalose in Various Areas of Stress Protection

Applications Reference(s)
In nature
 Carbon/energy source 21
 Osmoregulation 22
 Structural component of bacterial cell wall 12,23
 Dessication protectant 24,25
 Cryoprotection 13
 Immunogenicity (cord factor in mycobacteria) 26
 Growth regulator in plants 27,28
In laboratory
 Treatment of xerophthalmia and xeroderma 29,30
 Inhibition of osteoclast differentiation as a model for treatment of osteoporesis 31
 Suppression of “senior” body odor 20
 Stabilization of drug molecules against stress 32,33
 Stabilization of vaccines and liposomes 34
 Drought resistance in crop plants 15
 Preservation of mammalian cells against dessication 35
 Protection against thermal stress 36
 Cryoprotection of human fibroblasts and oocytes 37,38
 Stabilization of model phospholipids against leakage 39
 Protection against hypoxia in mammalian cells 40
 Protection against anoxia-induced protein aggregation 41
 Huntington's disease mouse model 42
 Cell line model of OPMD 43
 Osteoporesis in mouse models 44
 Aggregation of β-amyloid 45
 Aggregation of prion protein 46

Since nature is the source of clues, the table lists some of the areas where trehalose occurs naturally and exhibits its protective action against exposure of the organism to abiotic stress. This is followed by some examples where trehalose has been used as a “designer” protectant in different fields.

In the animal kingdom, trehalose has been found in very high amounts in the adult roundworm (about 6% of the dry weight) and in the eggs of roundworm (about 9% of the dry weight).47 In fact, tardigrades and other anhydrobiotic organisms have been proposed as model organisms for space research because of their inherent resistance to various harmful stimuli, an effect that is attributed mainly to a high level of trehalose in their systems.48 Trehalose has not been found in higher vertebrates to date even though the enzyme required to hydrolyze trehalose (trehalase) has been reported. In humans, the enzyme is found in the brush border cells of the epithelial membrane of the small intestine and in the proximal tubules of the kidneys.49,50 Their role is probably limited to the metabolism of any ingested trehalose and was elucidated when a patient with inherited trehalase deficiency suffered from diarrhoea following intake of mushrooms.51 The presence of trehalase is surprising, since humans lack another hydrolase, namely cellulase, even though the corresponding substrate, cellulose, is much more widespread than trehalose.

Physical Properties of Trehalose

The solid state and solution properties of trehalose impart to it the characteristics that are held to be responsible for the bioprotective role of trehalose. In this section, we describe some of the more widely accepted theories that may explain its extraordinary properties. Many more conjectures abound, all of them with more criticism than support, and will not be discussed here.

Solid state polymorphs

One of the most important reasons why trehalose is such an important bioprotectant is due to the existence of a number of polymorphs, both in the crystalline as well as amorphous states. At least two fully crystalline forms and several more transitory intermediates between crystalline and amorphous forms are known5255 (see Fig. 1). The most commonly occurring crystalline form, trehalose dihydrate (depicted as Th), is stable at room temperature.55 Careful dehydration of the dihydrate under defined conditions leads to the formation of the anhydrous crystal (depicted as Tβ).56 Dehydration of the dihydrate by heating at temperatures below 85°C, at low scanning rates, results in another anhydrous form (depicted as Tα).57 The low rate of scanning results in a low rate of water loss, ensuring that the crystalline lattice does not degenerate into a more relaxed form. This transformation is reversible, and the anhydrous form can be hydrated back to the dihydrate form without any loss of integrity of the crystalline structure. The anhydrous crystalline form Tα is thought to absorb moisture, undergoing a reversible transition to the crystalline dihydrate form.54 This reversibility, without alteration of the three-dimensional structure of the disaccharide, is important in its protective action.

Figure 1.

Figure 1

Transformation path of trehalose forms. Dihydrate trehalose Th is formed at equilibrium from the solution state and can be transformed reversibly to Tα. In addition, depending on the temperature and rate of water loss, Th can give either an amorphous form or the Tγ form. Reprinted from Ref. 54, with permission from Elsevier.

Glass transition temperature

Glass transition temperature (Tg), the temperature above which transition from viscous to fluid state occurs and the components acquire greater mobility, is the property that is invoked the most while explaining the “unusual effects” exhibited by trehalose.25,58,59 Melts of carbohydrates can be cooled to room temperature without undergoing crystallization. This transition to the glassy state or the vitrification theory has been put forward as the most widely accepted hypothesis to explain the bioprotective action of trehalose.58,60,61 Glasses typically have very high viscosities, with the result that any molecule that is encapsulated by a glassy matrix becomes, for all practical purposes, immobile and hence stable when exposed to stress conditions. Though controversy still reigns over the exact mechanism by which trehalose acts as a desiccation-protectant, two points of view have emerged; these need not be mutually exclusive. In the case of “immobilization theory,” trehalose is viewed as a cocoon that traps the biomolecule inside a glassy matrix, like amber-encasing insects.58,62 The movement of proteins is restricted by the sugar matrix. As discussed earlier, trehalose can transit between one crystalline form and another, without relaxing its structural integrity.54 Trehalose has the highest glass transition temperature of all the disaccharides.63,60 In general, addition of water to an amorphous substance increases its mobility leading to a decrease in Tg. Though this anticipated decrease does occur in the case of trehalose, Tg is still much higher than sucrose or maltose, neither of which exhibit so many polymorphic forms.63 This is probably the reason why trehalose is preferred over sucrose even though the latter has been postulated to play a similar role in some plants. In amorphous trehalose, local pockets of crystalline dihydrate exist, which trap residual water molecules, immobilizing them when water is scarce. This, along with the reversible transition between the dihydrate and anhydrous crystalline forms, is proposed to be the mechanism by which trehalose traps a biomolecule inside it and protects it during desiccation.6 Partial evidence for this hypothesis has recently been provided using positron annihilation lifetime spectroscopy to study the effect of dehydration on trehalose.6 In the glassy state, the average size of the free volume hole between trehalose molecules increases with increasing water activity, pointing to the role of water as a plasticizer by breaking hydrogen bonds between trehalose molecules. On the other hand, when water is removed from Tα anhydrous crystals, the channels become increasingly elongated. Under limiting water conditions, the channels are visualized as infinitely long cylindrical holes. Thus, the “anomalous” properties of trehalose are ascribed to the high stability of the glassy state and a specific interaction with water molecules in the crystalline state.6 Data for glass transition and melting of trehalose/water systems are highly disparate and make comparison of results from different groups difficult.63 It needs to be emphasized that the Tg of trehalose is not unusual but is at the highest end of all disaccharides and in the same league as tetrasaccharides.54Tg of a trehalose solution is higher than that of a sucrose solution, at all water activities. Thus, a sample incubated along with trehalose and stored at 37°C is still around 80°C below its Tg (assuming the Tg of trehalose to be ∼117°C, a widely accepted value),63 whereas the same sample, in the presence of sucrose, will be only 28°C below its Tg (which is 65°C). Because of the higher Tg of trehalose as compared to sucrose, a sample incubated with sucrose is more likely to absorb moisture, resulting in a decrease in its Tg to the storage temperature (37°C in the above case), which will cause the sample to degrade. Since the Tg of trehalose is much higher than sucrose, even if a sample containing trehalose were to absorb moisture and its Tg were to decrease, it will still not reach a stage where the Tg goes below the storage temperature of 37°C. That is how trehalose proves to be a better antidote to stress exposure. A trehalose containing sample, on absorbing water, produces more of the dihydrate crystalline form. Other disaccharides like α-melibiose monohydrate, α-lactose monohydrate, and so forth are also capable of transition to the crystalline hydrate but do not show similar bioprotective effects during desiccation and/or other stress conditions.61 Trehalose has also been implicated in the protection of organisms against cold-induced stress. Restoration of otsA/otsB genes from an E. coli deletion mutant for these two genes leads to an expected increase in trehalose production and also to an increase in the viability of the mutant at 4°C.64 It is hypothesized that trehalose prevents the inactivation and aggregation of proteins at lower temperature and stabilizes the cell membrane by delaying the onset of phase shift from liquid crystal to gel state.64 Trehalose is also thought to increase the cytoplasmic viscosity, thus decreasing the possibility of formation of intracellular ice crystals which are often fatal.65

Solution properties

Trehalose has been classified as a kosmotrope or water-structure maker, that is the interaction between trehalose/water is much stronger than water/water interaction and may be involved in its bioprotective action.66 Raman scattering experiments reveal the disruption of the tetrahedral network of water molecules on addition of trehalose.9 This “destructuring” of the water network by trehalose and ordering the water molecules around itself (as a kosmotrope) does not allow the formation of ice and makes trehalose one of the best cryoprotectants known. Maltose and sucrose follow trehalose in this behavior. Above a specific concentration (30%), sugar molecules force their hydrogen-bonding imprint on the water network, and trehalose turns out to be the most successful in this as is clear from the decrease in the “open” band intensity [ν(OH)].9 The decrease in the band intensity for the open conformation (O—H vibrational mode in tetrabonded water molecule) with increasing concentration of disaccharides reiterates the destructuring effect of the disaccharides on the tetrahedral hydrogen bonded network of water. Thirty percent appears to be the threshold value of concentration beyond which the disaccharides, especially trehalose, form their own hydrogen-bonded network with water and the disruption caused by the disaccharide becomes more apparent. Thus, trehalose is capable of disturbing the bonding arrangement of water more than maltose or sucrose, decreasing the freezing temperature of biological solutions to a lower value than the other two disaccharides. This is consistent with the earlier observation that disaccharides hamper crystallization of proteins by decreasing the amount of freezable water.67 Molecular dynamic (MD) simulations have confirmed that the addition of disaccharides to water leads to a drastic reorganization of the hydrogen bonded network of the latter and a significant increase in the fraction of population forming hydrogen bonds.5,9 This effect is again much more pronounced for trehalose than sucrose or maltose and is attributed to the higher hydration number for trehalose. The pattern of water distribution around the glycosidic oxygen is found to be different for all the disaccharides and is also different from other oxygens in disaccharides.5 The characteristic first solvation peak at 2.8 Å, which is well defined in all the other cases, is found to be missing for the glycosidic oxygens of the three (1,1)-linked disaccharides (including trehalose) under study.5 Instead, this peak shifts to 3.5 Å for trehalose, with lowered peak density. Thus, the glycosidic oxygen is not easily hydrated as compared to the rest of the oxygens, and this effect is most pronounced in the case of trehalose. Also, the radial distribution function around the glycosidic oxygen in trehalose shows restricted distribution of water molecules when compared with other oxygens and other disaccharides. The hydration number of the glycosidic oxygen is lower than all other oxygens and is particularly low in the case of trehalose (0.028 when compared with an average value of 0.153 for the glycosidic oxygen in other disaccharides).5 Thus, water molecules are nonuniformly distributed around oxygens of disaccharides, and this distribution plays an important role in the behavior of trehalose. MD simulations reveal that the dynamic motion about the glycosidic linkage is much slower in trehalose than in all other disaccharides in aqueous solutions. (The lifetimes of internal motions about the glycosidic linkage in case of trehalose, isotrehalose, and neotrehalose are 4.74, 3.13, and 3.16 ns, respectively. This value is 2.89 ns in case of maltose).5 Since the lability of the glycosidic linkage is probably the most important parameter in determining the conformation, these results, coupled with results indicating ordered distribution of water around the glycosidic oxygen of trehalose, explain how trehalose is able to maintain its conformation even in the aqueous milieu. The preferential exclusion theory has been cited in situations where water is in excess.6870 According to this, trehalose, or any other sugar molecule, does not interact directly with the biological macromolecule. Water is said to be excluded from the solvation layer of the biomolecule and is ordered around trehalose. With increase in the concentration of trehalose in bulk water, there is a competition between trehalose and the biomolecule for the available water. This competition causes water molecules to be destructured around biomolecules and “structured” around trehalose, making it a kosmotrope or water structure-maker. It is possible that depending on the structure of the protein/biomolecule in question, trehalose will be able to manipulate the water structure around itself, such that the protein/biomolecule is stabilized. Though the distribution of water molecules around trehalose is not uniform, they are oriented around trehalose in such a way that an ordered structure, with hydrogen bonds in all directions, is formed.

Indirect evidence for the manipulation of surrounding water layer by trehalose has also come from NMR and ultrasonic studies which reveal that trehalose has a very high affinity for water.71,72 NMR relaxation time measurements show that trehalose has the highest hydration ability among all the saccharides studied. Although trehalose lacks internal hydrogen bonds, at very high concentrations, the two glucose rings fold over, forming intermolecular hydrogen bonds.71,72,73 Ultrasonic studies support this hypothesis since the compressibility of trehalose solutions strongly depends on concentration.72 As water is added, unfolding occurs, making hydrogen bonding sites available for water molecules. Thus trehalose offers its protective action by reducing the activity of water. Trehalose orders the water network, at least up to the third hydration shell, reducing the amount of freezable water. According to the water replacement theory, which is an alternate hypothesis presented to explain the unique properties of trehalose, the disaccharide is thought to substitute water around the biomolecule, maintaining its three-dimensional structure by providing sites with hydrogen-bonding species.7477 This substitution also restricts the mobility of the biological macromolecule. Direct experimental evidence is available for this. When freeze-dried porcine pancreatic lipase is redissolved in water, the enzyme becomes flexible and active (and also susceptible to thermal denaturation). When suspended in any organic solvent, on the other hand, the enzyme shows very low activity. If freeze-drying is carried out in the presence of trehalose or a two-component system such as polyethylene glycol–trehalose, the same enzyme shows very high activity in organic medium as well.78 This is because the water molecules are replaced with trehalose molecules that provide hydrogen-bonding network, maintaining the three-dimensional structure of the active enzyme. How much of the interaction is a direct one between trehalose and the biomolecule remains doubtful. The mechanism of stabilization by trehalose or any other cosolute during freeze-drying is infinitely more complex than simple cryoprotection.34,79 Many cosolutes which act as efficient cryoprotectants fail to protect the protein from drying/desiccation-induced stress. The only exceptions are disaccharides.16,80,81 FT-IR studies carried out with trehalose and proteins have confirmed that hydrogen bonding does occur between the sugar and the protein and that this is a requirement for the preservation of labile proteins during desiccation. In the case of lysozyme, the position of the amide I band shifts from 1652 cm−1 for the fully hydrated enzyme to 1659 cm−1 when it is lyophilized in the absence of any excipients. In the presence of trehalose, the band regains the original position it occupied for the enzyme in solution.80 Similarly, the shift in the amide II band from 1543 cm−1 (for the fully hydrated enzyme) to 1530 cm−1 (for the enzyme lyophilized in the absence of any excipients) is reversed when lyophilization is carried out in the presence of trehalose. At higher concentrations of trehalose, sublimation results in crystallization of the disaccharide, and hydrogen bonding is no longer possible. Under these conditions, the spectrum of lysozyme resembles that of the enzyme freeze-dried in the absence of trehalose.80 Thus, hydrogen bonding is a prerequisite for stabilization of proteins by trehalose during freeze-drying. The stabilization to stress experienced by a protein during lyophilization is complicated by the fact that the stabilizer excipient should have both cryoprotective as well as lyoprotective properties. It is here that trehalose emerges the winner. A monosaccharide like glucose might be an efficient lyoprotectant but it provides no protection against freezing.82 Trehalose, on the contrary, offers both cryo- as well as lyoprotection during freeze-drying. It stabilizes the partially unfolded state of the protein (rather than stabilizing the native structure).83 Trehalose is thought to replace the water shell around proteins/membranes and to preserve the three-dimensional structure of the biomolecule.75 There is a huge body of evidence in support of this viewpoint as well, especially when proteins are freeze-dried for preservation. FT-IR and other studies have pointed to the gradual replacement of water by sugars, mainly trehalose, but also polymers like polyethylene glycol, and so forth.8487 In many cases where comparisons have been carried out, trehalose has come out at the top, not only among disaccharides but in comparison with other excipients as well.86,87 Simultaneous use of trehalose and ionic salts has been shown to have a synergistic effect on the freeze-drying stability of hen egg white lysozyme.88 This stabilization results from the interaction of trehalose with the protein, as confirmed by changes in the CO stretching vibration, resulting from the destructuring of the surrounding water molecules. This study also serves as a reminder that, during freeze-drying, the mixture does not remain homogeneous and a concentration gradient of components of the lyophilization mixture is set up.88 Proposing any hypothesis needs to take into account this often-ignored fact. Recent studies on structural relaxation times with human growth hormone have shown only a weak correlation between the secondary structure and aggregation propensity of the protein.89 This case is one of the few exceptions where addition of sucrose results in a stable, though more mobile, formulation than trehalose. There may be other reasons for this observation. Since trehalose is a nonreducing sugar, there is no free aldehyde group available to undergo a Maillard-type reaction with amino groups of proteins. Additionally, the α-1,1-glycosidic bond in trehalose is much less susceptible to hydrolysis than the α,β-1,2 glycosidic bond of the other nonreducing sugar, sucrose. In fact, under suboptimal conditions, the behavior of sucrose resembles that of glucose, and the rate of condensation reaction is ∼2000 times faster than with trehalose.90 This could be a major reason why trehalose is preferred over sucrose when a protein is lyophilized or when an organism faces desiccation, both scenarios where water availability is low.

The data presented earlier make it quite clear that trehalose, by virtue of its structure, has acquired some extraordinary properties. However, one question that begs an answer is as follows: are solution properties enough? It is difficult to see how the characteristics of trehalose in aqueous solution will help explain the stabilization effect observed when it is used as an excipient during freeze-drying. In this case, the vitrification properties of trehalose assume importance. Though the changes occurring during the initial replacement of water by trehalose are relevant, it is perhaps judicious to consider the changes that trehalose itself may face in low water conditions before looking at it as a stabilizer for biomolecules under low water conditions. In other words, the mechanism by which trehalose offers stabilization to a biomolecule in water-excess medium is probably different from the mechanism by which it does so in a low-water situation.

Biopreservative Action of Trehalose

A few areas where trehalose is active have been listed in Table II. This section deals specifically with cases where interaction of trehalose with proteins is involved. A number of polyhydroxyl compounds, including sugars, are used by organisms to overcome stress conditions like heat, cold, desiccation, and so forth. Trehalose, and to some extent sucrose, constitute a major category of these molecules. Their importance lies in the manner in which they can stabilize proteins as well as lipid bilayer. In the case of proteins, the most commonly invoked hypothesis to explain the transition from native to denatured state, in terms of thermodynamic parameters, is Tanford's transfer model.91 The basis of this model is that energetics of osmolytes on protein stability is an additive term of the free-energy contributions of polar and nonpolar groups exposed on the native and denatured states of a protein. As shown,92,93 when the solubility of a protein in 1M osmolyte solution is greater than in water, the solubility ratio (of the protein in water and 1M osmolyte) is less than 1, the apparent transfer free energy is negative, and the interaction between the protein and the osmolyte is energetically favorable. If this ratio is more than 1, the interaction between the protein and the osmolyte becomes unfavorable. As pointed out by the authors, unfavorable interaction of trehalose with the peptide backbone of the partially formed intermediate causes preferential exclusion of the osmolyte from the protein–water interface, thus increasing its free energy.92 This is complemented by a favorable interaction of trehalose with the peptide backbone of the native protein, which increases the thermodynamic barrier between the native and the partially folded forms. In fact, it has been shown recently that the hydration of the peptide backbone in the presence of an osmolyte depends on the type of osmolyte and the size of the side chains.94 The effect of this exclusion on the equilibrium constant of unfolding is mathematically expressed by the Wyman linkage relationship.95 According to this, the change in the equilibrium constant due to a change in the osmolyte concentration is expressed as the difference in the number of osmolyte molecules bound by the folded and unfolded protein molecules. As stated earlier, the unfavorable interaction of the peptide backbone with the osmolyte has been identified as the thermodynamic force responsible for the increase in chemical potential of the system.92 The unfolded state has more solvent-exposed surface area than the folded state, resulting in preferential exclusion and increase in chemical potential. The unfolded state is thus destabilized with respect to the folded state. This relationship, though widely accepted, does not take into account the effect of the osmolyte on the protein structure. Studies on the stabilization of the ribosomal protein S6 by trehalose have shown that trehalose does not alter the two-state character of the equilibrium unfolding of the protein.96 The stabilization induced by trehalose is nonspecific and is not localized to any particular secondary structure motif. This also agrees well with the change in transfer of free energy on unfolding and the stabilization inferred from thermal transitions.96 In addition, trehalose favors the collapse of the unfolded state coupled with an increase in the rate of folding and a decrease in the rate of unfolding of the protein.96 In case of unfolding of cutinase by heat, trehalose increases the melting temperature of the enzyme by delaying thermal unfolding of an intermediate.97 Trehalose compensates for the altered entropy of the process by increasing the contribution of enthalpy to the process, making unfolding a thermodynamically unfavorable process.

On exposure to heat, yeast produces trehalose, along with heat shock proteins, on a large scale.98 It has now been shown in yeast that trehalose stabilizes partially folded proteins during heat shock and is rapidly hydrolyzed on activation of the partially folded form by molecular chaperones.99 Trehalose offers protection to yeast glucose-6-phosphate dehydrogenase and phosphoglucoisomerase against thermoinactivation.100 Induction of thermotolerance by trehalose is also inferred from the fact that the level of trehalose is correlated with thermotolerance.100 Surprisingly, trehalose has also been found to inhibit aggregation of denatured proteins (e.g., bovine glutamic dehydrogenase) following heat shock.100 Whether trehalose aids the function of the chaperone or vice versa is not clear. In some cases, trehalose is thought to influence chaperone activity of heat shock proteins. There is evidence that heat shock leads to increased synthesis of components of the trehalose-6-phosphate synthase complex in Saccharomyces cerevisiae.101 Yeast mutants defective in genes for trehalose anabolism display much lower survival rates than the native species.36 Adding confusion to this controversy are reports which indicate that the synthesis of heat shock proteins is neither necessary nor sufficient for the induction of thermotolerance in yeast.102,103 On the other hand, E. coli mutants with no otsA or otsB do display reduced thermotolerance.104 More conclusive proof can be derived from the study of the effect of trehalose on the thermoinactivation of enzymes. In one study, trehalose has been found to protect yeast pyrophosphatase against exposure at 50°C much better than other sugars like sucrose, fructose, or glucose.105 This is attributed to the lower concentration of water in trehalose solutions. A 1.5M trehalose solution contains 62.5% water by volume; at a similar molar concentration, sucrose and maltose solutions contain 87% and 86% water by volume, respectively. This enhanced hydrated volume of trehalose results in increased viscosity. If the volumetric concentrations of water in sucrose and maltose solutions are equalized to that in a 1.5M trehalose solution, the respective concentrations required turn out to be 4.3M and 4M. Since these disaccharides are insoluble at such high molar concentrations, a 4.4M glycerol solution is prepared, which contains 62.5% water. At this concentration, the levels of thermoprotection afforded to pyrophosphatase by both trehalose and glycerol are identical,105 proving that it is the hydrated volume and not the molar concentration per se that determines the amount of stabilization offered. Fluorescence emission studies have confirmed that both trehalose and glycerol prevent unfolding of the enzyme when used at the same volumetric concentration. When used at the same molar concentration as trehalose, both sucrose and maltose fail to prevent the unfolding of the enzyme at 50°C to any significant extent.105 It is possible that trehalose and other sugars solubilize in the bulk water and are excluded from the solvation layer of the protein (preferential exclusion theory). This leads to a decrease in the solvation layer around the protein, restricting its mobility, and stabilizing it against stress conditions. As the hydrated radius of a trehalose molecule is ∼2.5 times higher than that of sucrose or maltose, the size-exclusion effects become more pronounced and the stabilization effect is high.105

One of the most exciting areas where trehalose has shown some promise is in therapeutics. Trehalose has already proved its usefulness as a protectant against oxidative stress. As stated earlier, trehalose accumulation can be induced in S. cerevisiae following exposure to gentle stress conditions such as mild heat shock98 or the presence of a proteasome inhibitor.106 These cells show increased tolerance when exposed to a free radical generating system. Mutants unable to synthesize trehalose show exceedingly low viability when exposed to oxidative stress. This effect can be reversed if trehalose is made available to the mutants.106 The reaction of oxygen radicals with proteins gives rise to free carbonyl groups in the latter. It is hypothesized that exposure to H2O2 causes aggregation of proteins and that the presence of trehalose overcomes this damage, presumably by acting as a free radical scavenger. Trehalose-deficient mutants of Candida albicans are extremely sensitive to exposure to H2O2 while the same condition induces trehalose accumulation in wild-type cells, with improved life spans.107 In mammalian cells, O2 deprivation or hypoxia for 5–10 min results in irreversible tissue damage.40 Drosophila however can survive exposure to complete N2 atmosphere for up to 4 h without any damage. In this case, overexpression of TPS1 gene leads to accumulation of trehalose with concomitant increase in tolerance to O2 deprivation.41 Trehalose is also believed to reduce anoxia-induced protein aggregation in vitro.41 Transfection of Drosophila TPS1 gene into HEK-293 cells has given rise to populations that show decreased trehalose metabolism when grown in glucose-deficient media. These transfected cells show increased trehalose accumulation when exposed to hypoxic stress and show much higher survival rates as compared to control cells. The amount of aggregated as well as ubiquitinated proteins in the transfected cells is much less when exposed to O2-depleted atmosphere.41 Thus stabilization of proteins by trehalose is probably responsible for survival of lower organisms when exposed to hypoxic stress.

Expansion of glutamine units in many proteins results in their aggregation leading to neurodegenerative diseases. One of these is the Huntington's disease where expansion of CAG units occurs in the first exon of the gene coding for huntingtin protein.108 Although the premise that protein aggregation is a precursor of the disease is debatable, cell culture studies have shown that amelioration of aggregation does lead to relieving the complications in the cellular machinery which ultimately cause cell death. A mutant myoglobin has been engineered, which has an expanded polyglutamine stretch (35 residues) and is a good model for polyglutamine diseases.42 A library of small molecules has been screened in vitro for their ability to inhibit protein aggregation caused by the presence of expanded polyglutamine units. The compounds chosen are such that they are nontoxic and suitable for oral administration. Disaccharides emerge as successful candidates with, perhaps not surprisingly, trehalose surfacing as the one which causes the most significant reduction in aggregation in vitro.42 Other disaccharides such as sucrose, maltose, and melibiose do cause a reduction in aggregation but the effect is marginal as compared to trehalose which exhibits a dose-dependent behavior. The same results can be reproduced in mouse neuroblastoma cell lines expressing a truncated N-terminal huntingtin containing 150 glutamine residues, fused to an enhanced green fluorescent protein (EGFP).42 Interestingly, the trehalose metabolite glucose does not show any significant effect, confirming that it is trehalose itself and not its ability to act as a source of glucose which is responsible for the alleviation of symptoms. Increasing the concentration of trehalose does not affect the expression of polyglutamine-containing truncated huntingtin. Overexpression of E. coli otsA and otsB results in the accumulation of trehalose in the above cell line and inhibition of intracellular aggregation. Oral administration of trehalose (at 0.05M) to R6/2 transgenic mice (a mouse model of Huntington's disease) reduces dilatation of lateral ventricles.42 The average brain weight of the transgenic mice also increases. More importantly, immunohistochemical staining (for ubiquitin-positive protein aggregates) confirms the decrease in intranuclear aggregation in motor cortex, striatum, and liver upon administration of trehalose.42 As mentioned earlier, trehalose is mostly impermeable to plasma membrane. However, it can easily be loaded into mammalian cells using fluid-phase endocytosis and pinocytosis109 by simply being in contact with the cells, as in this case, via oral administration of this molecule. The stabilization of an engineered myoglobin containing 35 glutamine residues upon administration of trehalose is of the same order as that of an engineered myoglobin containing a nonpathogenic stretch of glutamine residues. Thus, the stabilization may have resulted from the interaction of the polyglutamine stretch with trehalose rather than the truncated N-terminal huntingtin, which does not fold easily even in the presence of trehalose. This is so since trehalose has no effect on the stability of wild-type, nonengineered myoglobin. It is also important to recollect that trehalose is rapidly hydrolyzed once the stabilized partially folded protein is taken over by chaperones for correct and complete folding.99 Trehalose, in fact, has been shown to inhibit protein folding. Thus, in all probability, trehalose interacts with the expanded polyglutamine stretch and stabilizes it early before aggregation can set in.

Another codon reiteration disease is oculopharyngeal muscular dystrophy (OPMD), which is caused by the abnormal expansion of the polyalanine stretch in the poly(A) binding nuclear protein 1 (PABPN1).110 The expanded GCG repeat units in the PABPN1 gene express a protein that aggregates as tubular filaments within the nuclei of skeletal muscle fibres. These aggregates are presumed to be toxic and are crucial in OPMD pathology. Trehalose has been shown to decrease aggregation and toxicity of mutants in a COS7 cell line expressing EGFP tagged-PABPN1.43 Transgenic A17-1 mice (mouse models of OPMD) show a decrease in aggregation (as seen by immunolabeling for PABPN1-positive aggregates in the nuclei of bicep section of treated mice) accompanied by attenuation of muscle weakness upon oral administration of trehalose. Addition of cyclohexamide (an inhibitor of protein synthesis) and lactacystin (an inhibitor of trehalose-induced clearance of mutant PABPN1) confirms that trehalose promotes the clearance of mutant PABPN1 via proteasomes.43 No comparison with other disaccharides has been made in this study. Perhaps not surprisingly, trehalose has also been found to inhibit aggregation of β-amyloid (Aβ) to form amyloid plaques, the pathological hallmark of Alzheimer's disease, in a dose-dependent manner.45 Interestingly, however, this inhibition follows different patterns in case of Aβ-40 and Aβ-42. While in the former, inhibition of aggregation is seen at all concentrations of trehalose studied, the effect is seen only at the highest concentration in case of Aβ-42. Also, trehalose is able to reverse the cytotoxicity caused by Aβ-40, but not by Aβ-42. This can be correlated with the retention of toxic oligomeric structures by Aβ-42 even in the presence of trehalose. Because of the presence of two more relatively nonpolar amino acid residues, Aβ-42 is thought to be more hydrophobic and hence more prone to aggregation than Aβ-40. Interaction between hydrophobic patches results in the loss of ordering of surrounding water molecules, decreasing the entropy and thus free energy of the process. Addition of trehalose cannot overcome this thermodynamic barrier and hence trehalose is unable to disassemble the toxic, oligomeric assemblies of Aβ-42.45 There has been a recent report of trehalose reducing the size of PrPSc (a protease-resistant form of the prion protein PrPC) aggregates in prion-infected ScN2A cells and protecting these cells against oxidative stress.46 More research is necessary however before trehalose can be prescribed as a therapeutic molecule in this case, since breaking up of infectious prion aggregates may result in formation of smaller particles that can act as nuclei for further aggregation.

Trehalose has also been found useful in relieving the symptoms of osteoporesis.44 Symptoms of estrogen deficiency following ovariectomy in mice, namely decrease in bone weight and loss of calcium and phosphorus levels in the femur, are reversed upon oral administration of trehalose, which has no effect on the weight of uterus.44 Thus, trehalose does not act in the same manner as estrogen but rather by the inhibition of osteoclast differentiation leading to reduced femoral bone loss.

Trehalose has also been found to act as a stabilizer to improve the shelf-life of therapeutic proteins. Work with model proteins has shown that trehalose is able to abrogate the moisture-induced aggregation of bovine serum albumin, by interfering with the formation of intermolecular disulphide bonds.33 Electrophoresis studies with recombinant human serum albumin in the solid state have shown the formation of a completely amorphous state in the presence of trehalose and sucrose and stabilization of the protein during storage over a 4-month period at 35°C.111 Similar kind of stabilization effects have been shown by another sugar, sorbitol, in the stabilization of tetanus and diphtheria toxoids against moisture-induced aggregation.32 Trehalose has also demonstrated favorable properties in the stabilization of recombinant botulinum serotype A vaccine during lyophilization112 and IgG1 in spray-dried formulations.113

Conclusions

The various explanations used to elucidate the bioprotective role of trehalose have been covered in sufficient detail in the previous sections. Three main rationales have emerged (see Fig. 2). None of these, on its own, is sufficient to explain the bioprotection offered by trehalose. The situation “on the ground” is probably an amalgamation of these theories, which need not be mutually exclusive. It needs to be reiterated that none of the physical or chemical properties of trehalose discussed earlier is sufficient in itself to explain the bioprotective action that trehalose offers across the spectrum of stress conditions that an organism faces. As summarized later, these properties are neither “unusual” nor “exceptional.” They lie at one end of the range (either highest or lowest) and in some cases, even in between, for disaccharides.

  • Nonreducing sugar

  • Relatively inert glycosidic linkage

  • Absence of internal hydrogen bonds

  • Existence of a number of polymorphs

  • Transition between polymorphs without affecting crystallinity

  • High glass transition temperature

  • High fragility

  • High hydrophilicity and hydration number

  • Large hydrated volume

  • Kosmotropy or water structure “maker”

  • Restricted distribution of water molecules

  • Flexibility of glucose rings to expand and contract

  • Ability to increase cytoplasmic viscosity: decrease in intracellular ice formation

  • Depression of melting temperature of lipids

  • Stabilization of partially folded proteins

Figure 2.

Figure 2

Various theories to explain the “exceptional” properties of trehalose. (A) Vitrification theory assumes that trehalose forms a glassy matrix that acts as a cocoon and presumably physically shields the protein or indeed cells from abiotic stresses. (B) Preferential exclusion theory, on the other hand, proposes that there is no direct interaction between trehalose and protein (or biomolecule). Instead, as can be seen, addition of trehalose to bulk water sequesters water molecules away from the protein, decreasing its hydrated radius and increasing its compactness and consequently stability. (C) Water replacement theory talks of substitution of water molecules by trehalose-forming hydrogen bonds, maintaining the three-dimensional structure and stabilizing biomolecules.

There may be individual molecules that have exceptional properties, but taken together, trehalose, with the synergistic action of all these factors, seems to offer the maximum bioprotection. It seems unlikely that there is a hitherto undiscovered parameter which will provide a solution to the “trehalose question.” So, is trehalose the elixir of life? As far as countering stress conditions by organism is concerned, probably yes. Does it work because it has magical properties? No. Trehalose works simply because it possesses the best parameters that nature can provide, none of which, on its own, will at best, be capable of lending more than a helping hand. As the critical analysis mentioned earlier shows, what works is probably a cumulative effect of all the theories that have been put forward; the relative contribution of each parameter is adjusted according to the stress factor that is encountered.

References

  • 1.Ignatova Z, Gierasch LM. Inhibition of protein aggregation in vitro and in vivo by a natural osmoprotectant. Proc Natl Acad Sci USA. 2006;103:13357–13361. doi: 10.1073/pnas.0603772103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Ignatova Z, Gierasch LM. Effects of osmolytes on protein folding and aggregation in cells. Methods Enzymol. 2007;428:355–372. doi: 10.1016/S0076-6879(07)28021-8. [DOI] [PubMed] [Google Scholar]
  • 3.Bordat P, Lerbret A, Demaret J-P, Affouard F, Descamps M. Comparative study of trehalose, sucrose and maltose by molecular modelling. Europhys Lett. 2005;65:41–47. [Google Scholar]
  • 4.Chen B, Fowler A, Bhowmick S. Forced and natural convective drying of trehalose/water thin films: implication in the desiccation preservation of mammalian cells. J Biomech Eng. 2006;128:335–346. doi: 10.1115/1.2187051. [DOI] [PubMed] [Google Scholar]
  • 5.Choi Y, Cho KW, Jeong K, Jung S. Molecular dynamic simulations of trehalose as a ‘dynamic reducer’ for solvent water molecules in the hydration shell. Carbohydr Res. 2006;341:1020–1028. doi: 10.1016/j.carres.2006.02.032. [DOI] [PubMed] [Google Scholar]
  • 6.Kilburn D, Townrow S, Muenier V, Richardson R, Alam A, Ubbink J. Organization and mobility of water in amorphous and crystalline trehalose. Nat Mater. 2006;5:632–635. doi: 10.1038/nmat1681. [DOI] [PubMed] [Google Scholar]
  • 7.Kuttel MM, Naidoo KJ. Ramachandran free-energy surfaces for disaccharides: trehalose, a case study. Carbohydr Res. 2005;340:875–879. doi: 10.1016/j.carres.2005.01.031. [DOI] [PubMed] [Google Scholar]
  • 8.Lerbret A, Bordat P, Affouard F, Descamps M, Migliardo F. How homogeneous are the trehalose, maltose, and sucrose water solutions? An insight from molecular dynamics simulations. J Phys Chem B. 2005;109:11046–11057. doi: 10.1021/jp0468657. [DOI] [PubMed] [Google Scholar]
  • 9.Lerbret A, Bordat P, Affouard F, Guinet Y, Hedoux A, Paccou L, Prevost D, Descamps M. Influence of homologous disaccharides on the hydrogen-bond network of water: complementary Raman scattering experiments and molecular dynamics simulations. Carbohydr Res. 2005;340:881–887. doi: 10.1016/j.carres.2005.01.036. [DOI] [PubMed] [Google Scholar]
  • 10.Magazú S, Migliardo F, Mondelli C, Vadala M. Correlation between bioprotective effectiveness and dynamic properties of trehalose–water, maltose–water and sucrose–water mixtures. Carbohydr Res. 2005;340:2796–2801. doi: 10.1016/j.carres.2005.09.026. [DOI] [PubMed] [Google Scholar]
  • 11.Singh KJ, Roos YJ. State transitions and freeze concentration in trehalose-protein-cornstarch mixtures. LWT. 2006;39:930–938. [Google Scholar]
  • 12.Carpinelli J, Kraemer R, Agosin E. Metabolic engineering of Corynebacterium glutamicum for trehalose over production: role of the TreYZ trehalose biosynthetic pathway. Appl Environ Microbiol. 2006;72:1949–1955. doi: 10.1128/AEM.72.3.1949-1955.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Duong T, Barrangou R, Russell WM, Klaenhammer TR. Characterisation of the tre locus and analysis of trehalose cryoprotection in Lactobacillus acidophilus NCFM. Appl Environ Microbiol. 2006;72:1218–1225. doi: 10.1128/AEM.72.2.1218-1225.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Elbein AD, Pan YT, Pastuszak I, Carroll D. New insights on trehalose: a multifunctional molecule. Glycobiology. 2003;13:17R–27R. doi: 10.1093/glycob/cwg047. [DOI] [PubMed] [Google Scholar]
  • 15.Garg AK, Kim J-K, Owens TG, Ranwala AP, Choi YD, Kochian LV, Wu RJ. Trehalose accumulation in rice plants confers high tolerance levels to different abiotic stresses. Proc Natl Acad Sci USA. 2002;99:15898–15903. doi: 10.1073/pnas.252637799. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Haines AH. Non-equivalence ofd- andl-trehalose in stabilizing alkaline phosphatase against freeze-drying and thermal stress. Is chiral recognition involved? Org Biomol Chem. 2006;4:702–706. doi: 10.1039/b514473f. [DOI] [PubMed] [Google Scholar]
  • 17.Higo A, Katoh H, Ohmori K, Ikeuchi M, Ohmori M. The role of a gene cluster for trehalose metabolism in dehydration tolerance of the filamentous cyanobacterium Anabaena sp. PCC 7120. Microbiology. 2006;152:979–987. doi: 10.1099/mic.0.28583-0. [DOI] [PubMed] [Google Scholar]
  • 18.Komes D, Lovrić T, Ganić KK, Gracin L. Study oftrehalose addition on aroma retention in dehydrated strawberry puree. Food Technol Biotechnol. 2003;41:111–120. [Google Scholar]
  • 19.Richards AB, Krakowa S, Dexter LB, Schmid H, Wolterbeek APM, Waalkens-Berendsen DH, Shigoyuki A, Kurimoto M. Trehalose: a review of properties, history of use and human tolerance, and results of multiple safety studies. Food Chem Toxicol. 2003;40:871–898. doi: 10.1016/s0278-6915(02)00011-x. [DOI] [PubMed] [Google Scholar]
  • 20.Higashiyama T. Novel functions and applications of trehalose. Pure Appl Chem. 2002;74:1263–1269. [Google Scholar]
  • 21.Whatmore AM, Reed RH. Determination of turgor pressure in Bacillus subtilis: a possible role for K+ in turgor regulation. J Gen Microbiol. 1990;136:2521–2526. doi: 10.1099/00221287-136-12-2521. [DOI] [PubMed] [Google Scholar]
  • 22.Kempf B, Bremer E. Uptake and synthesis of compatible solutes as microbial stress responses to high-osmolality environments. Arch Microbiol. 1998;170:319–330. doi: 10.1007/s002030050649. [DOI] [PubMed] [Google Scholar]
  • 23.Murphy HN, Stewart GR, Mischenko VV, Apt AS, Harris R, McAlister MS, Driscoll PC, Young DB, Robertson BD. The OtsAB pathway is essential for trehalose biosynthesis in Mycobacterium tuberculosis. J Biol Chem. 2005;280:14524–14529. doi: 10.1074/jbc.M414232200. [DOI] [PubMed] [Google Scholar]
  • 24.Rueda B, Miguelez EM, Hardisson C, Manzanal MB. Changes in glycogen and trehalose content of Streptomyces brasiliensis during growth in liquid cultures under sporulating and non-sporulating conditions. FEMS Microbiol Lett. 2001;194:181–185. doi: 10.1111/j.1574-6968.2001.tb09466.x. [DOI] [PubMed] [Google Scholar]
  • 25.Crowe LM. Lessons from nature: the role of sugar in anhydrobiosis. Comp Biochem Phys A. 2002;131:505–513. doi: 10.1016/s1095-6433(01)00503-7. [DOI] [PubMed] [Google Scholar]
  • 26.Rao V, Gao F, Chen B, Jacobs WR, Jr, Glickman MS. Trans-cyclopropanation of mycolic acids on trehalose dimycolate suppresses Mycobacterium tuberculosis-induced inflammation and virulence. J Clin Invest. 2006;116:1660–1667. doi: 10.1172/JCI27335. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Rolland F, Moore B, Sheen J. Sugar sensing and signaling in plants. Plant Cell. 2002;14:s185–s205. doi: 10.1105/tpc.010455. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Eastmond PJ, Graham IA. Is trehalose-6-phosphate a regulator of sugar metabolism in plants? J Exp Bot. 2003;54:533–537. doi: 10.1093/jxb/erg039. [DOI] [PubMed] [Google Scholar]
  • 29.Matsuo T. Trehalose protects corneal epithelial cells from death by drying. Br J Ophthalmol. 2001;85:610–612. doi: 10.1136/bjo.85.5.610. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Norcia MA. Compositions and methods for wound management. Official Gazette US Pat Trademark Office. 2000;1232:424–448. [Google Scholar]
  • 31.Arai C, Kohguchi M, Akamatsu S, Arai N, Yoshizane C, Hasegawa N, Hanaya T, Arai S, Ikeda M, Kurimoto M. Trehalose suppresses lipopolysaccharide-induced osteoclastogenesis bone marrow in mice. Nutr Res. 2001;21:993–999. doi: 10.1016/s0271-5317(01)00315-3. [DOI] [PubMed] [Google Scholar]
  • 32.Schwendeman SP, Constantino HR, Gupta RK, Siber GR, Klibanov AM, Langer R. Stabilization of tetanus and diphtheria toxoids against moisture-induced aggregation. Proc Natl Acad Sci USA. 1995;92:11234–11238. doi: 10.1073/pnas.92.24.11234. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Jain NK, Roy I. Role of trehalose in moisture-induced aggregation of bovine serum albumin. Eur J Pharm Biopharm. 2008;69:824–834. doi: 10.1016/j.ejpb.2008.01.032. [DOI] [PubMed] [Google Scholar]
  • 34.Crowe JH, Leslie SM, Crowe LM. Is vitrification sufficient to preserve liposomes during freeze-drying? Cryobiology. 1994;31:355–366. doi: 10.1006/cryo.1994.1043. [DOI] [PubMed] [Google Scholar]
  • 35.Guo N, Puhlev I, Brown DR, Mansbridge J, Levine F. Trehalose expression confers desiccation tolerance on human cells. Nat Biotechnol. 2000;18:168–171. doi: 10.1038/72616. [DOI] [PubMed] [Google Scholar]
  • 36.Sun WQ, Leopold AC. Cytoplasmic vitrification and survival of anhydrobiotic organisms. Comp Biochem Physiol. 1997;117A:327–333. [Google Scholar]
  • 37.Eroglu A, Russo MJ, Bieganski R, Fowler A, Cheley S, Bayley H, Toner M. Intracellular trehalose improves the survival of cryopreserved mammalian cells. Nat Biotechnol. 2000;18:163–167. doi: 10.1038/72608. [DOI] [PubMed] [Google Scholar]
  • 38.Eroglu A, Toner M, Toth TL. Beneficial effect of microinjected trehalose on the cryosurvival of human oocytes. Fertil Steril. 2002;77:152–158. doi: 10.1016/s0015-0282(01)02959-4. [DOI] [PubMed] [Google Scholar]
  • 39.Crowe JH, Hoekstra FA, Nguyen KHN, Crowe LM. Is vitrification involved in depression of the phase transition temperature in dry phospholipids? Biochim Biophys Acta. 1996;1280:187–196. doi: 10.1016/0005-2736(95)00287-1. [DOI] [PubMed] [Google Scholar]
  • 40.Haddad GG, Ma E. Neuronal tolerance to O2 deprivation in drosophila: novel approaches using genetic models. Neuroscientist. 2001;7:538–550. doi: 10.1177/107385840100700610. [DOI] [PubMed] [Google Scholar]
  • 41.Chen Q, Haddad GG. Role of trehalose phosphate synthase and trehalose during hypoxia: from flies to mammals. J Exp Biol. 2004;207:3125–3129. doi: 10.1242/jeb.01133. [DOI] [PubMed] [Google Scholar]
  • 42.Tanaka M, Michida Y, Niu S, Ikeda T, Jana NR, Doi H, Kurosawa M, Nekooki M, Nukina N. Trehalose alleviates polyglutamine-mediated pathology in a mouse model of Huntington's disease. Nat Med. 2004;10:148–154. doi: 10.1038/nm985. [DOI] [PubMed] [Google Scholar]
  • 43.Davies JE, Sarkar S, Rubinsztein DC. Wild-type PABPN1 is anti-apoptotic and reduces toxicity of the oculopharyngeal muscular dystrophy mutation. Hum Mol Genet. 2006;15:23–31. doi: 10.1093/hmg/ddm382. [DOI] [PubMed] [Google Scholar]
  • 44.Nishizaki Y, Yoshizane C, Toshimori Y, Arai N, Akamatsu S, Hanaya T, Arai S, Ikeda M, Kurimot M. Disaccharide-trehalose inhibits bone resorption in ovariectomized mice. Nutr Res. 2000;20:653–664. [Google Scholar]
  • 45.Liu R, Barkhordarian H, Emadi S, Park CB, Sierks MR. Trehalose differentially inhibits aggregation and neurotoxicity of beta amyloid 40 and 42. Neurobiol Dis. 2005;20:74–81. doi: 10.1016/j.nbd.2005.02.003. [DOI] [PubMed] [Google Scholar]
  • 46.Béranger F, Crozet C, Goldsborough A, Lehmann S. Trehalose impairs aggregation of PrPSc molecules and protects prion-infected cells against oxidative damage. Biochem Biophys Res Commun. 2008;334:44–48. doi: 10.1016/j.bbrc.2008.06.094. [DOI] [PubMed] [Google Scholar]
  • 47.Arguelles JC. Physiological roles of trehalose in bacteria and yeasts: a comparative analysis. Arch Microbiol. 2000;174:217–224. doi: 10.1007/s002030000192. [DOI] [PubMed] [Google Scholar]
  • 48.Jönsson KI. Tardigrades as a potential model organism in space research. Astrobiology. 2007;7:757–766. doi: 10.1089/ast.2006.0088. [DOI] [PubMed] [Google Scholar]
  • 49.Mercer J, Eagles ME, Talbot IC. Brush border enzymes in coeliac disease: histochemical evaluation. J Clin Pathol. 1990;43:307–312. doi: 10.1136/jcp.43.4.307. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Sasai-Takedatsu M, Taketani S, Nagata N, Furukawa T, Tokunaga R, Kojima T, Kobayashi Y. Human trehalase: characterization, localization, and its increase in urine by renal proximal tubular damage. Nephron. 1996;73:179–185. doi: 10.1159/000189037. [DOI] [PubMed] [Google Scholar]
  • 51.Bergoz R. Trehalose malabsorption causing intolerance to mushrooms. Report of a probable case. Gastroenterology. 1971;60:909–912. [PubMed] [Google Scholar]
  • 52.McGarvey OS, Kett VL, Craig DQM. An investigation into the crystallisation of α,α-trehalose from the amorphous state. J Phys Chem B. 2003;107:6614–6620. [Google Scholar]
  • 53.Nagase H, Endo T, Ueda H, Nakagaki M. An anhydrous polymorphic form of trehalose. Carbohydr Res. 2002;337:167–173. doi: 10.1016/s0008-6215(01)00294-4. [DOI] [PubMed] [Google Scholar]
  • 54.Sussich F, Skopec C, Brady J, Cesaro A. Reversible dehydration of trehalose and anhydrobiosis: from solution state to an exotic crystal? Carbohydr Res. 2001;334:165–176. doi: 10.1016/s0008-6215(01)00189-6. [DOI] [PubMed] [Google Scholar]
  • 55.Taga T, Senma M, Osaki K. The crystal and molecular structure of trehalose dihydrate. Acta Crystallogr B. 1972;28:3258–3263. [Google Scholar]
  • 56.Jeffrey GA, Nanni R. The crystal structure of anhydrous alpha,alpha-trehalose at −150 degrees. Carbohydr Res. 1985;137:21–30. doi: 10.1016/0008-6215(85)85146-6. [DOI] [PubMed] [Google Scholar]
  • 57.Nagase H, Endo T, Ueda H, Nakai T. Influence of dry conditions on dehydration of α,α-trehalose dihydrate. STP Pharm Sci. 2003;13:269–275. [Google Scholar]
  • 58.Crowe JH, Carpenter JF, Crowe LM. The role of vitrification in anhydrobiosis. Annu Rev Physiol. 1998;60:73–103. doi: 10.1146/annurev.physiol.60.1.73. [DOI] [PubMed] [Google Scholar]
  • 59.Crowe JH, Crowe LM, Oliver AE, Tsvetkova N, Wolkers W, Tablin F. The trehalose myth revisited: introduction to a symposium on stabilization of cells in the dry state. Cryobiology. 2001;43:89–105. doi: 10.1006/cryo.2001.2353. [DOI] [PubMed] [Google Scholar]
  • 60.Green JL, Angell CA. Phase relations and vitrification in saccharide-water solutions and the trehalose anomaly. J Phys Chem. 1989;93:2880–2882. [Google Scholar]
  • 61.Roos Y. Melting and glass transitions of low molecular weight carbohydrates. Carbohydr Res. 1993;238:39–48. [Google Scholar]
  • 62.Liu Q, Schmidt RK, Teo B, Karplus PA, Bray JW. Molecular dynamics studies of the hydration of α,α-trehalose. J Am Chem Soc. 1997;119:7851–7862. [Google Scholar]
  • 63.Chen T, Fowler A, Toner M. Literature review: supplemented phase diagram of the trehalose–water binary mixture. Cryobiology. 2000;40:277–282. doi: 10.1006/cryo.2000.2244. [DOI] [PubMed] [Google Scholar]
  • 64.Kandror O, DeLeon A, Goldberg AL. Trehalose synthesis is induced upon exposure of Escherichia coli to cold and is essential for viability at low temperatures. Proc Natl Acad Sci USA. 2002;99:9727–9732. doi: 10.1073/pnas.142314099. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Rudolph AS, Crowe JH, Crowe LM. Effects of three stabilizing agents—proline, betaine, and trehalose-on membrane phospholipids. Arch Biochem Biophys. 1986;245:134–143. doi: 10.1016/0003-9861(86)90197-9. [DOI] [PubMed] [Google Scholar]
  • 66.Branca C, Maccarrone S, Magazú S, Maisano G, Bennington SM, Taylor J. Tetrahedral order in homologous disaccharide-water mixtures. J Chem Phys. 2005;22:174513. doi: 10.1063/1.1887167. [DOI] [PubMed] [Google Scholar]
  • 67.Forbes RT, Davis KG, Hindle M, Clarke JG, Maas J. Water vapor sorption studies on the physical stability of a series of spray-dried protein/sugar powders for inhalation. J Pharm Sci. 1998;87:1316–1321. doi: 10.1021/js980081l. [DOI] [PubMed] [Google Scholar]
  • 68.Sola-Penna M, Meyer-Fernandes JR. Trehalose protects yeast pyrophosphatase against structural and functional damage induced by guanidinium chloride. Z Naturforsch Sect C Biosci. 1996;51:160–164. doi: 10.1515/znc-1996-3-405. [DOI] [PubMed] [Google Scholar]
  • 69.Timasheff SN. A physicochemical basis for the selection of osmolytes by nature. In: Osmond CB, Bolis CL, Somero GN, editors. Water and life: comparative analysis of water relationships at organic, cellular and molecular levels. Berlin: Springer Verlag; 1992. pp. 70–86. [Google Scholar]
  • 70.Timasheff SN. The control of protein stability and association by weak interactions with water: how do solvents affect these processes? Annu Rev Biophys Biomol Struct. 1993;22:67–97. doi: 10.1146/annurev.bb.22.060193.000435. [DOI] [PubMed] [Google Scholar]
  • 71.Kawai H, Sakurai M, Inoue Y, Chujo R, Kobayashi S. Hydration of oligosaccharides: anomalous hydration ability of trehalose. Cryobiology. 1992;29:599–606. doi: 10.1016/0011-2240(92)90064-9. [DOI] [PubMed] [Google Scholar]
  • 72.Magazú S, Migliardo P, Musolino AM, Sciortino MT. α,α-Trehalose-water solutions. 1. Hydration phenomena and anomalies in the acoustic properties. J Phys Chem B. 1997;101:2348–2351. [Google Scholar]
  • 73.Elias ME, Elias AM. Trehalose + water fragile system: properties and glass transition. J Mol Liq. 1999;83:303–310. [Google Scholar]
  • 74.Cho CH, Singh S, Robinson GW. Understanding all of water's anomalies with a nonlocal potential. J Chem Phys. 1997;107:7979–7988. [Google Scholar]
  • 75.Crowe JH, Crowe LM, Chapman D. Preservation of membranes in anhydrobiotic organisms: the role of trehalose. Science. 1984;223:701–703. doi: 10.1126/science.223.4637.701. [DOI] [PubMed] [Google Scholar]
  • 76.Franks F. Solvation interactions of proteins in solution. Philos Trans R Soc Lond B Life Sci. 1977;278:33–57. doi: 10.1098/rstb.1977.0032. [DOI] [PubMed] [Google Scholar]
  • 77.Liu Q, Brady JW. Anisotropic solvent structuring in aqueous sugar solutions. J Am Chem Soc. 1996;118:12276–12286. [Google Scholar]
  • 78.Roy I, Sharma A, Gupta MN. Obtaining higher transesterification rates with subtilisin Carlsberg in nonaqueous media. Bioorg Med Chem Lett. 2004;14:887–889. doi: 10.1016/j.bmcl.2003.12.021. [DOI] [PubMed] [Google Scholar]
  • 79.Roy I, Gupta MN. Freeze-drying of proteins: some emerging concerns. Biotechnol Appl Biochem. 2004;39:165–177. doi: 10.1042/BA20030133. [DOI] [PubMed] [Google Scholar]
  • 80.Carpenter JF, Crowe JH. An infrared spectroscopic study of the interactions of carbohydrates with dried proteins. Biochemistry. 1989;28:3916–3922. doi: 10.1021/bi00435a044. [DOI] [PubMed] [Google Scholar]
  • 81.Jovanovic N, Bouchard A, Hofland GW, Witkamp GJ, Crommelin DJ, Jiskoot W. Distinct effects of sucrose and trehalose on protein stability during supercritical fluid drying and freeze-drying. Eur J Pharm Sci. 2006;27:336–345. doi: 10.1016/j.ejps.2005.11.003. [DOI] [PubMed] [Google Scholar]
  • 82.Carpenter JF, Hand SC, Crowe LM, Crowe JH. Cryoprotection of phosphofructokinase with organic solutes: characterization of enhanced protection in the presence of divalent cations. Arch Biochem Biophys. 1986;250:505–512. doi: 10.1016/0003-9861(86)90755-1. [DOI] [PubMed] [Google Scholar]
  • 83.Sola-Penna M, Ferreira-Pereira A, Lemos AP, Meyer-Fernandes JR. Carbohydrate protection of enzyme structure and function against guanidinium chloride treatment depends on the nature of carbohydrate and enzyme. Eur J Biochem. 1997;248:24–29. doi: 10.1111/j.1432-1033.1997.00024.x. [DOI] [PubMed] [Google Scholar]
  • 84.Clegg JS, Seitz P, Seitz W, Hazlewood CF. Cellular responses to extreme water loss: the water-replacement hypothesis. Cryobiology. 1982;19:306–316. doi: 10.1016/0011-2240(82)90159-6. [DOI] [PubMed] [Google Scholar]
  • 85.Crowe JH, Oliver AE, Hoekstra FA, Crowe LM. Stabilization of dry membranes by mixtures of hydroxyethyl starch and glucose: the role of vitrification. Cryobiology. 1997;35:20–30. doi: 10.1006/cryo.1997.2020. [DOI] [PubMed] [Google Scholar]
  • 86.Maury M, Murphy K, Kumar S, Mauerer A, Lee G. Spray-drying of proteins: effects of sorbitol and trehalose on aggregation and FT-IR amide I spectrum of an immunoglobulin G. Eur J Pharm Biopharm. 2005;59:251–261. doi: 10.1016/j.ejpb.2004.07.010. [DOI] [PubMed] [Google Scholar]
  • 87.Sastry GH, Agmon N. Trehalose prevents myoglobin collapse and preserves its internal mobility. Biochemistry. 1997;36:7097–7108. doi: 10.1021/bi9626057. [DOI] [PubMed] [Google Scholar]
  • 88.Ragoonanan V, Aksan A. Heterogeneity in desiccated solutions: implications for biostabilization. Biophys J. 2008;94:2212–2227. doi: 10.1529/biophysj.107.110684. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Pikal MJ, Rigsbee D, Roy ML, Galreath D, Kovach KJ, Wang BS, Carpenter JF, Cicerone MT. Solid state chemistry of proteins: II. The correlation of storage stability of freeze-dried human growth hormone (hGH) with structure and dynamics in the glassy solid. J Pharm Sci. 97:5106–5121. doi: 10.1002/jps.21374. [DOI] [PubMed] [Google Scholar]
  • 90.O'Brien J. Stability of trehalose, sucrose and glucose to nonenzymatic browning in model systems. J Food Sci. 1996;61:679–682. [Google Scholar]
  • 91.Tanford C. Isothermal unfolding of globular proteins in aqueous urea solutions. J Am Chem Soc. 1964;86:2050–2059. [Google Scholar]
  • 92.Auton M, Bolen DW. Predicting the energetics of osmolyte-induced protein folding/unfolding. Proc Natl Acad Sci USA. 2005;102:15065–15068. doi: 10.1073/pnas.0507053102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Auton M, Bolen DW. Application of the transfer model to understand how naturally occurring osmolytes affect protein stability. Methods Enzymol. 2007;428:397–418. doi: 10.1016/S0076-6879(07)28023-1. [DOI] [PubMed] [Google Scholar]
  • 94.Auton M, Bolen DW, Rösgen J. Structural thermodynamics of protein preferential solvation: osmolyte salvation of proteins, aminoacids, and peptides. Proteins. 73:802–813. doi: 10.1002/prot.22103. [DOI] [PubMed] [Google Scholar]
  • 95.Wyman J. Linkage graphs: a study in the thermodynamics of macromolecules. Q Rev Biophys. 1984;17:453–488. doi: 10.1017/s0033583500004881. [DOI] [PubMed] [Google Scholar]
  • 96.Chen L-Y, Cabrita GJM, Otzen DE, Melo EP. Stabilization of the ribosomal protein S6 by trehalose is counterbalanced by the formation of a putative off-pathway species. J Mol Biol. 2005;351:402–416. doi: 10.1016/j.jmb.2005.05.056. [DOI] [PubMed] [Google Scholar]
  • 97.Baptista RP, Pedersen S, Cabrita GJ, Otzen DE, Cabral JM, Melo EP. Thermodynamics and mechanism of cutinase stabilization by trehalose. Biopolymers. 2008;89:538–547. doi: 10.1002/bip.20926. [DOI] [PubMed] [Google Scholar]
  • 98.Ellis T. Proteins as molecular chaperones. Nature. 1987;328:378–379. doi: 10.1038/328378a0. [DOI] [PubMed] [Google Scholar]
  • 99.Singer MA, Lindquist S. Multiple effects of trehalose on protein folding in vitro and in vivo. Mol Cell. 1998;1:639–648. doi: 10.1016/s1097-2765(00)80064-7. [DOI] [PubMed] [Google Scholar]
  • 100.Hottiger T, de Virgilio C, Hall MN, Boller T, Wiemken A. The role of trehalose synthesis for the acquisition of thermotolerance in yeast. II. Physiological concentrations of trehalose increase the thermal stability of proteins in vitro. Eur J Biochem. 1994;219:187–193. doi: 10.1111/j.1432-1033.1994.tb19929.x. [DOI] [PubMed] [Google Scholar]
  • 101.de Virgilio C, Buerckert N, Bell W, Jenoe P, Boller T, Wiemken A. Disruption of TPS2, the gene encoding the 100-kDa subunit of the trehalose-6-phosphate synthase/phosphatase complex in Saccharomyces cerevisiae, causes accumulation of trehalose-6-phosphate and loss of trehalose-6-phosphate phosphatase activity. Eur J Biochem. 1993;212:315–323. doi: 10.1111/j.1432-1033.1993.tb17664.x. [DOI] [PubMed] [Google Scholar]
  • 102.Praekelt UM, Meacock MP. HSP12, a new small heat shock gene of Saccharomyces cerevisiae: analysis of structure, regulation and function. Mol Gen Genet. 1990;223:97–106. doi: 10.1007/BF00315801. [DOI] [PubMed] [Google Scholar]
  • 103.Smith BJ, Yaffe MP. Uncoupling thermotolerance from the induction of heat shock proteins. Proc Natl Acad Sci USA. 1991;88:11091–11094. doi: 10.1073/pnas.88.24.11091. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Hengge-Aronis R, Klein W, Lange R, Rimmele M, Boos W. Trehalose synthesis genes are controlled by the putative sigma factor encoded by rpoS and are involved in stationary-phase thermotolerance in Escherichia coli. J Bacteriol. 1991;173:7918–7924. doi: 10.1128/jb.173.24.7918-7924.1991. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Sola-Penn M, Meyer-Fernandes JR. Stabilization against thermal inactivation promoted by sugars on enzyme structure and function: why is trehalose more effective than other sugars? Arch Biochem Biophys. 1998;360:10–14. doi: 10.1006/abbi.1998.0906. [DOI] [PubMed] [Google Scholar]
  • 106.Benaroudj N, Lee DH, Goldberg AL. Trehalose accumulation during cellular stress protects cells and cellular proteins from damage by oxygen radicals. J Biol Chem. 2001;276:24261–24267. doi: 10.1074/jbc.M101487200. [DOI] [PubMed] [Google Scholar]
  • 107.González-Párraga P, Hernández JA, Argüelles JC. Role of antioxidant enzymatic defences against oxidative stress H2O2 and the acquisition of oxidative tolerance in Candida albicans. Yeast. 2003;20:1161–1169. doi: 10.1002/yea.1029. [DOI] [PubMed] [Google Scholar]
  • 108.Huntington Collaborative Research Group. A novel gene containing a trinucleotide repeat that is expanded and unstable on Huntington's disease chromosomes. Cell. 1993;72:971–983. doi: 10.1016/0092-8674(93)90585-e. [DOI] [PubMed] [Google Scholar]
  • 109.Ma X, Jamil K, MacRae T, Clegg J, Russell JM, Villeneuve TS, Eulothe M, Sun Y, Crowe JH, Tablin F, Oliver AE. A small stress protein acts synergistically with trehalose to confer desiccation tolerance on mammalian cells. Cryobiology. 2005;51:15–28. doi: 10.1016/j.cryobiol.2005.04.007. [DOI] [PubMed] [Google Scholar]
  • 110.Brias B, Bouchard JP, Xie YG, Rochefort DL, Chretien N, Tome FM, Lafreniere RG, Rommens JM, Uyama E, Nohira O, Blumen S, Korczyn AD, Heutink P, Mathieu J, Duranceau A, Codère F, Fardeau M, Rouleau GA. Short GCG expansions in the PABP2 gene cause oculopharyngeal muscular dystrophy. Nat Genet. 1998;18:164–167. doi: 10.1038/ng0298-164. [DOI] [PubMed] [Google Scholar]
  • 111.Han Y, Jin BS, Lee SB, Sohn Y, Joung JW, Lee JH. Effects of sugar additives on protein stability of recombinant human serum albumin during lyophilization and storage. Arch Pharm Res. 2007;30:1124–1131. doi: 10.1007/BF02980247. [DOI] [PubMed] [Google Scholar]
  • 112.Roy S, Henderson I, Nayar R, Randolph TW, Carpenter JF. Effect of pH on stability of recombinant botulinum serotype A vaccine in aqueous solution and during storage of freeze-dried formulations. J Pharm Sci. 2008;97:5132–5146. doi: 10.1002/jps.21409. [DOI] [PubMed] [Google Scholar]
  • 113.Schüle S, Schulz-Fademrecht T, Garidel P, Bechtold-Peters K, Frieß W. Stabilization of IgG1 in spray-dried powders for inhalation. Eur J Pharm Biopharm. 69:793–807. doi: 10.1016/j.ejpb.2008.02.010. [DOI] [PubMed] [Google Scholar]

Articles from Protein Science : A Publication of the Protein Society are provided here courtesy of The Protein Society

RESOURCES