Download as pdf or txt
Download as pdf or txt
You are on page 1of 16

CRITICAL REVIEWS IN BIOTECHNOLOGY, 2018

VOL. 38, NO. 3, 335–350


https://doi.org/10.1080/07388551.2017.1355294

REVIEW ARTICLE

Practical insights on enzyme stabilization


Carla Silvaa, Madalena Martinsa, Su Jingb, Jiajia Fuc and Artur Cavaco-Pauloa,b
a
Centre of Biological Engineering (CEB), University of Minho, Braga, Portugal; bInternational Joint Research Laboratory for Textile and
Fiber Bioprocesses, Jiangnan University, Wuxi, China; cKey Laboratory of Science and Technology of Eco-Textiles, Ministry of Education,
Jiangnan University, Wuxi, Jiangsu, China

ABSTRACT ARTICLE HISTORY


Enzymes are efficient catalysts designed by nature to work in physiological environments of living Received 9 November 2016
systems. The best operational conditions to access and convert substrates at the industrial level Revised 4 January 2017
are different from nature and normally extreme. Strategies to isolate enzymes from extremophiles Accepted 17 May 2017
can redefine new operational conditions, however not always solving all industrial requirements.
The stability of enzymes is therefore a key issue on the implementation of the catalysts in indus- KEYWORDS
trial processes which require the use of extreme environments that can undergo enzyme instabil- Stabilization; enzymes;
ity. Strategies for enzyme stabilization have been exhaustively reviewed, however they lack a formulation; molecular
practical approach. This review intends to compile and describe the most used approaches for interactions; industrial
enzyme stabilization highlighting case studies in a practical point of view. catalysis

Industrial demands of enzymes Enzyme properties and the need of


stabilization strategies
Major applications of enzymes are in industries of deter-
gents, food processing, animal nutrition, juice and fla- Enzymes are protein molecules consisting of folded
vorings, cosmetics, medication, pharmaceuticals, polypeptide chains of amino acids that are essential to
leather, silk, chemical, and for research and develop- perform an array of biological functions. The order of
ment [1,2]. The industrial applications are only feasible these amino acids in a protein determines its tertiary
if catalysts are stabilized against temperature, extreme structure through molecular geometry and intramolecu-
pH, and in the presence of alkalis, acids, salts, and sur- lar chemical interactions [7,8]. Depending on the amino
factants [3,4]. Major applications of enzymes are at high acid composition, these proteins can incorporate both
temperature (e.g. washing at 60–70  C, starch gelatiniza- acidic and basic functional groups, which play an
tion at 100  C, textile desizing at 80–90  C), or may vary important role in their structure. Therefore, their expres-
depending on the substrate and product solubility and sion can be related to resistance of unfolding forces
stability. They can occur under high salt concentration because of protein’s conformation results in a less sol-
(food industry) and alkaline conditions and/or in the uble state due to the occurrence of structural changes,
presence of surfactants (detergents). Reactions can be aggregation, and/or precipitation [9]. Enzyme’s solubil-
realized at gas–liquid interface (for reactions consuming ity can change as their structure undergoes modifica-
(e.g. O2) or producing (e.g. CO2) gas), liquid–liquid inter- tions resulting from the exposure of different residues
face for aqueous-organic two-liquid phase reactions to the surrounding environment. A few of the remark-
where organic phase is used as a carrier for the sub- able features of enzymes are their functional diversity
strate and/or product and in the presence of organic and versatility derived from each constituent amino
solvents [3,5]. Hence, the need to stabilize enzymes acid having a different side chain with a specific chemis-
against thermodeactivation, deactivation in the pres- try and polarity, and from flexible and numerous ways
ence of surfactants and alkaline pH is imperative [6]. in which polypeptide chains can fold.
Enzyme stability can be enhanced following different Enzymes undergo a variety of denaturation reactions
routes which include strategies for stabilization in aque- during production, storage, and their application in
ous and non-aqueous environments. industry. Denaturation is the unfolding of the tertiary

CONTACT Artur Cavaco-Paulo artur@deb.uminho.pt Centre of Biological Engineering, University of Minho, 4710-057 Braga, Portugal
These authors contributed equally to this work.
ß 2017 Informa UK Limited, trading as Taylor & Francis Group
336 C. SILVA ET AL.

Figure 1. Illustration of (a) electron density profiles perpendicular to the air/water interface for lysozyme injected into buffer solu-
tions with pH 3 and pH 11.5 (pI) (adapted from Yano et al. [19]); (b) net charge enzyme as a function of pH (adapted from
Pihlasalo et al. [142]); topdown image shows the effect of several physical and biological agents responsible for denaturation with
the respective target sites and the corresponding effects on enzymes.

structure of the enzyme to a disordered polypeptide in contrast, the adsorption efficiency decreases as pH
which key residues are no longer aligned closely increases, since the particles and protein repel each
enough to continue the participation in functional or other due to repulsive charges, both having a negative
structure stabilizing interactions [3]. If the denaturing charge [20,21].
influence is removed this tendency can be reversed. The achievement of stable and active enzymes is
Enzymes must be formulated in order to maintain often a challenging effort because they have not
proper folding to perform their biological functions evolved naturally to be used in industrial environments.
[10–12]. Their integrity under extreme conditions might Their biological activity depends on the three-dimen-
involve high excipient concentrations [11] whose inter- sional native structure, hence catalytically active, and
actions are aided by several forces, such as hydrogen any significant conformational change can lead to their
bonds, solvation forces, electrostatic, Van der Waal inactivation [12]. Manifestations of enzyme instability
forces, amongst others [13–15]. These interactions can arise from aggregation, loss of biological functionality,
be controlled through screening of the charge by and exposure to extreme conditions or even slight var-
increasing the salt concentration of the solution or by iations of temperature or pH, that can induce abrupt
changing the charge density on the surface [16,17]. conformational changes and subsequent loss of their
Generally, at low ionic strength, the observed adsorp- biological activity [22]. A number of enzyme-based
tion is to be maximumal at the isoelectric point [18], processes have been commercialized for producing
whereas the unfolding of the tertiary structure of pro- valuable products, however despite their great potential
tein reaches a maximum because of the large intramo- they have been hampered by undesirable stability, cata-
lecular electrostatic repulsion, as observed in Figure 1(a) lytic efficiency, and low specificity. For this reason, the
[19,20]. The unfolding of the tertiary structure is greater exploitation of methodologies for enzyme stabilization
at pH 11.5 which is its isoelectric point (pI). Proteins is imperative for the progress in biotechnology and for
have low solubility at pI, because the effective charge potential protein-templates discovery (Figure 2).
of the molecule is zero (Figure 1(b)) and with decreased Along this review, practical insights related with
repulsion and electrostatic interaction between the mol- enzyme stabilization, highlighting experimental aspects
ecules, they form clumps that tend to precipitate. In for each methodology developed will be provide. This
CRITICAL REVIEWS IN BIOTECHNOLOGY 337

Figure 2. Schematic representation of different enzyme stabilization methodologies.

will be described along the text and practical examples interesting enzyme classes that are active and stable
will be specified in Table 1. under extreme conditions have been used to maximize
reactions in the food and paper industry, detergents,
drugs, toxic waste removal, and drilling for oil. These
Enzyme-stabilization strategies
enzymes can be produced from the thermophiles
Screening and isolation of enzymes from through either optimized fermentation of the microor-
extremophiles ganisms or cloning of fast-growing mesophiles by
recombinant DNA technology. It has been found that
The interest in extremophiles stems from their surpris-
psychrophilic enzymes can assist enhance yield of heat-
ing properties being superior to traditional catalysts
sensitive products, halophilic enzymes that are stable in
and allowing the performance of industrial processes
high salt concentrations serve as models for biocatalysis
under harsh conditions in which conventional proteins
in low-water media and thermophilic enzymes that are
are denaturated. There has been extensive research on
highly resistant to proteases, detergents, and chaotropic
the structural proteins and key metabolic enzymes that
agents, which may also afford resistance to the effects
are responsible for the organisms’ unusual properties.
of organic solvents. Within these enzyme classes are
Recent research has focused on the identification of
included: esterases/lipases, glycosidases, aldolases, nitri-
extremozymes relevant for industrial biocatalysis lases/amidases, phosphatases, and racemases [24,25].
[23,24]. The most evident examples of the success of extremo-
Extremophiles are organisms that have evolved to philes, used in industrial large scale, are the starch
exist in a variety of extreme environments and fall into degrading enzymes, a-amylases [26]. Lipases and pro-
a number of different classes that include thermophiles, teases, which are resistant to high temperatures, have
acidophiles, alkalophiles, psychrophiles, and barophiles also been the subject of study. In the textile industry,
(piezophiles) and others. They can survive at high and extremophilic enzymes find applications in several proc-
low temperatures (5 to 130  C), extreme pH (0–12), esses ranging from fiber preparation to finishing and
high salt concentrations (3–35%), and high pressure subsequent laundering of textiles (see Table 1:
(1000 bar). They have adapted to thrive in ecological Examples 1, 2, 3).
niches such as deep-sea hydrothermal vents, hot
springs, and sulfataric fields [25]. As a result, these
microorganisms produce unique biocatalysts that func-
Engineering of enzymes
tion under conditions in which their mesophilic coun- In the past, an enzyme-based process was designed
terparts could not survive, permitting the development around the limitations of the enzyme. Currently, the
of additional industrial processes [24,25]. Many enzyme is engineered to fit process specifications [27].
338 C. SILVA ET AL.

Protein engineering is the design and construction of role in distorting the glucose ring into a more reactive
novel proteins, usually by manipulation of their genes. conformation [40]. Some other relevant examples of
The rational protein design or direct evolution allows enzyme engineering are given in this review in Table 1.
the alteration of enzyme properties to meet the limita-
tions in their applications [28]. Rational design uses
Functionalization of enzymes
structural and mechanistic information together with
molecular modeling for the prediction of changes in Covalent chemical modification has emerged as a
the protein structure in order to alter or induce the powerful alternative to site-directed mutagenesis and
desired properties. Advances in computer technology direct evolution for tailoring enzymes. Chemical modifi-
have helped to create better protein models to improve cation of amino acid sidechains allows a greater, almost
predictions for rational design, but structure–activity unlimited, variety of groups to be introduced, but the
relationships are still not trivial. In directed evolution, reactions used for their introduction are typically non-
mutant libraries are created by random changes, specific in nature. Thus, despite many potential advan-
screened for the desired property and the variants tages, several classical methods used for protein
showing promising results are subjected to further modification create mixtures of proteins as a result
rounds of evolution. More researchers today employ of poor discrimination or insufficiently efficient
combined methods of these two strategies, called chemistry [41].
focused-directed evolution and semi-rational design A vast number of nonspecific modifications has been
[29,30]. The advances in genomics, proteomics, and reviewed during the last decades. As examples, we
informatics have been creating new opportunities to highlight the introduction of hydrophobic and hydro-
exploit a large amount of biological data [31–33]. philic groups into chymotrypsin which stabilize the
The increased industrial requirements relate to the enzyme against thermal denaturation [42]. The nonspe-
use of biocatalysts in aggressive non-natural media like cific modification, as judged by 2,4,6-trinitrobenzenesul-
organic solvents, ionic liquids, and physiologic fluids. fonic acid (TNBS) titration, of amines in Candida rugosa
This has led to the development of novel strategies to lipase (CRL) with benzyloxycarbonyl (Z), Z-NO2, lauroyl,
develop enzymes with improved performances [34]. and acetyl-enhanced enantioselectivity in lipase-
Improving the activity of an industrial enzyme is often a catalyzed esterification of 2-(4-substituted phenoxy)pro-
primary goal. This is partly because naturally available panoic acids with n-BuOH in isopropyl ether [43].
enzymes are usually not optimally suited for many proc- Phosphorylation, glycosylation, and farnesylation are
esses in industrial applications. Many enzymes used in also examples of enzyme modifications modulating
the textile industry, such cellulases, amylases, lipases, their catalytic activity [44]. The chemical modification of
and even proteases, act on insoluble substrates. cellulose by maleic anhydride and N-bromosuccinimide
Therefore, the rate of substrate turnover may be limited was attempted to improve detergent stability [45].
by diffusion, and controlled by enzyme mobility at the Glutaraldehyde crosslinking and pegylation of
surface or by on/off enzyme desorption rates [35]. enzymes surface have been other strategies to enhance
These, in turn, are often related to the surface proper- biocatalyst stability [46]. The use of glutaraldehyde was
ties of the enzyme and the conditions at the interface firstly reported by Quiocho and Richards [47] and has
between the enzyme and substrate [36]. The experi- triggered a series of improvements on highly stable
mental results from several site-directed variants with CLECs (insoluble crosslinked enzyme crystals). This tech-
structural modeling of fungal lipase from Rhizopus ory- nology involves the crosslinking of enzyme microcrys-
zae have provided much insights into the molecular tals with bifunctional reagents, such as glutaraldehyde
mechanism(s) of catalysis [36]. Substitutions at Glu87 [41,46,48]. The behavior of several enzymes, normally
and Trp89 in the lid region have been suggested to used for textile finishing, modified by glutaraldehyde,
alter the activity of the lipase from Humicola lanuginosa such as the formation of dimers and higher oligomers
(lipolase) [37]. Cellulases and xylanases have become a and the production of enzymatic aggregates with pre-
major focus in recent years due to their ability to pro- served activity, was studied by Silva et al. [49] (see
vide the soft feel of stone-washed jeans in textile proc- Table 1 for other examples).
essing, fabric care benefits (such as color crispness) Pegylation is a broadly used strategy characterized
when used in laundry detergents [38], and reduction of by the modification of a protein or peptide by the link-
the quantity of chemicals required for bleaching in the ing of one or more methoxy polyethylene glycol chains,
pulp and paper industry, thereby minimizing environ- a nontoxic and nonimmunogenic polymer. Pegylation
mental impact [39]. Tyr169 in the Trichoderma reesei cel- changes the physical and chemical properties of the
lobiohydrolase II catalytic domain plays an important biomolecule, such as its conformation, electrostatic
CRITICAL REVIEWS IN BIOTECHNOLOGY 339

Table 1. Enzyme stabilization strategies (practical applications).


Stabilization strategies Enzyme How to … Effect Application References
Screening and isola- Amylase Acidophile, thermophile and Increase oxidative stability Desizing of cotton [116,117]
tion of enzymes psychrophile/
from extremophiles
Protease Thermophile, psychrophile, Shrinkage reduction; Degumming of silk and [118–121,123]
halophile, alkaliphile improved dyeability production of sand wash
effect
Esterase Thermophile Depilling and improvement Polyester hydrolysis and [112–125]
of dyeing of polyester depolymerization; ring-
fibers open polymerization
Engineering of Cutinase Fusion of cutinase with two Modify acetate cellulose Surface modification of [126]
enzymes CBM (cellulose-binding fibers and increase acetate cellulose fibers
models) hydrophilicity
Cutinase Genetic modification by site- Enhance activity towards Synthetic fibers modification [127]
directed mutagenesis synthetic fibers (polyester
and polyamide)
Protease Construction of a recombin- Increase the molecular Alternative method to chlor- [121]
ant protein with subtilisin weight of subtilisin E to ine treatment of wool
E and elastin-like polymer use as alternative to
chlorine treatment of
wool
Laccase Site-directed mutagenesis in Increase oxidation of Phenolics oxidation for poly- [128]
four phenylalanine near phenolics merization; textile waste
the active site water treatment
Xylanase XT6 Directed evolution and site- 52 increase in thermal sta- Biobleaching [129]
directed mutagenesis bility; Kopt increase by 10  C;
catalytic efficiency increase
by 90%
Functionalization of Laccase PEGylation of laccase with Improve stability and cata- Polymerization of phenolics; [50]
enzymes mPEG in presence of lytic activity bioremediation
cianoborohydride
Glucose-oxidase PEGylation of GOx followed Using this stabilization strat- Enzymatic bleaching of cot- [54]
by controlled chemical egy, size increases and ton; hydrogen peroxide
modification with aggregation due to inter- generation
glutaraldehyde molecular crosslinking are
avoided; PEGylated
enzyme retains 73% ori-
ginal activity after 4
weeks at 37  C (vs. 8.2%
retention for control).
Immobilization of a-amylase Chemical cross-linking of Enhance thermal and stor- Desizing of cotton [61]
enzymes enzyme aggregates with age stability; Free
amino functionalized enzyme:
magnetite nanoparticles Km ¼ 0.93 ± 0.014
(CLEAs) Vmax ¼ 85 ± 0.11; mag-
netic CLEAs:
Km ¼ 0.21 ± 0.019
Vmax ¼ 81 ± 0.27
b-glucosidase Covalent immobilization of Immobilized enzyme: Lignocellulose hydrolysis [130]
b-1,4-glucosidase from Kl ¼ 3.8 lM, Vmax ¼ 3347
Agaricus arvensis onto (lmol min1);
functionalized silicon soluble enzyme:
oxide nanoparticles Km ¼ 2.5 mM,
Vmax ¼ 3028 lmol min1
mg1
Laccase Laccase formulation was Increase affinity and activity Textile waste water [131]
immobilized on modified to the substrate treatment
green coconut fiber silan-
ized with 3-glycidoxypro-
pyltrimethoxysilane
Protease Esperase, was covalently Increase thermal stability Wool shrink-resist finish [132]
linked to Eudragit S-100, of wool
a reversible soluble–insol-
uble polymer by carbodii-
mide coupling
Laccase Laccase was immobilized Increase stability on textile Waste water treatment [133]
onto polyamide 6.6 fibers surfaces
Candida antarctica Immobilization on macropo- Catalyze organic chemical Polyester synthesis by ring- [134]
lipase B (CALB) rous resin of poly(methyl reactions opening polymerization
methacrylate of various lactones and
polycondensation reac-
tions of diacid/diol
substrates
(continued)
340 C. SILVA ET AL.

Table 1. Continued
Stabilization strategies Enzyme How to … Effect Application References
Cutinase from Immobilization of cutinase Catalyze organic chemical Synthesis of polyamides [135]
Fusarium solani on ewatit reactions
Beads or in the form of
cross-linked enzyme
aggregates (CLEA)
Medium engineering Ribonuclease Addition of sucrose, trehal- Increase stabilization – [66]
ose, glucose, maltose and
ribose to enzyme
formulations
Lysozyme Addition of PEG mixed Increase solubility and – [92]
solvents stability
Protease Addition of pyridinium and Stabilization by kosmotropic Food industry; wool [136]
imidazolium-based ionic anions and destabilization finishing
liquids by chaotropic anions;
Hofmeister series in
general
Enzyme encapsulation Laccase Encapsulation into in Increase stability Textiles and cosmetics [137]
liposomes
Amylase Encapsulation into magnetic Increase stability Food industry [138]
chitosan beads
Laccase Encapsulation in oil-in-water Increase enzyme stability at Textile and cosmetics [139]
proteinaceous micro- high temperatures and
emulsions high shear stress
Enzyme stabilization Lipase Study the stability of lipase Increase enzyme stability at Lipid hydrolysis [140]
by silk on both water-soluble 37  C without need to
and insoluble silk fibroin use cryoprotectantes,
films emulsifiers or covalent
immobilization
Glucose-oxidase GOx immobilized in air dried Improved enzyme activity Ampometric sensors for glu- [141]
silk films cose detection

binding, size, and hydrophobicity, improving the cata- crystals [53]. Support binding can be physical or chem-
lytic behavior of enzymes and their stability in aqueous ical, involving weak or covalent bonds. Physical bonding
media. The greatly expanded hydrodynamic volume of is weak and hardly able to keep the enzyme fixed to
the PEG–enzyme conjugate resulting from mPEG ability the carrier under industrial conditions. The support can
to coordinate water molecules can be responsible for be a synthetic resin, an inorganic polymer such as zeo-
these changes [50,51]. Despite being explored mainly in lite or silica, or a biopolymer. Entrapment involves inclu-
the pharmaceutical area, this technique has been sion of an enzyme in a polymer network such as an
extended to other areas for the modification and stabil- organic polymer or silica-gel, or a membrane device
ization of a broad range of enzymes. Examples of suc- such as a hollow fiber or a microcapsule. It requires the
ceeded enzyme pegylation are presented in Table 1. synthesis of the polymeric network in the presence of
the enzyme. The covalent attachment involves the
cross-linking of enzyme aggregates or crystals, using a
Immobilization of enzymes
bifunctional reagent, to prepare carrier-free macropar-
Enzymes are generally expensive and unlike conven- ticles [52,53]. The ideal immobilization procedure for a
tional heterogeneous chemical catalysts, most of them given enzyme is the one that allows a high turnover
operate dissolved in water in homogeneous catalysis rate of the enzyme while retaining high catalytic activity
systems, leading to product contamination and ruling over time. Recently, the major focus of enzyme immo-
out their recovery and reuse [52]. One of the most bilization is the development of robust enzymes that
used strategies to overcome these drawbacks is immo- are not only active but also stable and selective in
bilization. It is a technical process in which enzymes are organic solvents. Moreover, several new types of carriers
fixed to or within solid supports, creating a heteroge- and technologies have been implemented which aimed
neous immobilized enzyme system [52]. The solid sup- to enhance enzyme loading, activity, and stability
port stabilizes the enzyme structure maintaining its decreasing though the biocatalyst cost in industrial
activity. applications [52,54–56]. These include: cross-linked
Approaches used for the design of immobilized enzyme aggregates, microwave-assisted immobilization,
enzymes include a variety of methods: adsorption, cova- click chemistry technology, mesoporous supports, and
lent binding, entrapment with nanofibrous polymers, nanoparticle-based immobilization. The high surface-to-
nanoparticles, cross-linked enzyme aggregates, or volume ratio offered by nanoparticles resulted in the
CRITICAL REVIEWS IN BIOTECHNOLOGY 341

concentration of the immobilized entity being higher solvation to polar residues of the biocatalyst and other
than that afforded by 2-D surface protocols [55]. intervening molecules in order to facilitate protein con-
Enzyme immobilization envisage between other formational changes during the biocatalytic process and
goals, to increase the activity of the catalyst. The to speed up the reaction. There are many different
enhancement of enzyme activity upon immobilization approaches to control water activity in biotransforma-
depends on: the microenvironment, partition effect, dif- tions, but the control of media composition is the sim-
fusion effect, conformational change, molecular orienta- plest one.
tion, conformational flexibility, conformation induction,
and binding mode. It has been observed that many The role of solvents
immobilized enzymes exhibit higher activity than the
Solvents can be categorized as: water-miscible (mono-
corresponding native enzyme [57–59] and only few
phasic aqueous–organic systems, including some ionic
reports about the decrease of Km and increase Vmax
liquid systems), non-aqueous (monophasic organic sys-
upon formation of crosslinked enzyme aggregates
tem), water-immiscible (multiphasic aqueous–organic
(CLEAs) [60,61] (see Table 1 for other examples related
systems, and most of ionic liquid systems described to
with enzyme immobilization on textile, bioremediation
date), anhydrous systems (including solvent free sys-
industries, and polymer synthesis).
tems), supercritical fluids and gas phase, and reversed
micelles. The effects of polar and non-polar solvents on
Medium engineering enzyme activity are quite dissimilar; both reduce the
enzyme activity for different reasons. In polar solvents,
Enzymes for industrial purposes are sold on the basis of
water stripping is the major, but not the only reason for
the overall activity. The manufacturer will usually rec-
reduction of enzyme activity. Water stripping is referred
ommend storage conditions and quote the expected
to the ability of polar organic solvents to displace water
rate of activity loss under those conditions. It is of pri-
molecules from the protein surface, to be replaced by
mary importance to the enzyme producer and customer
solvent molecules which rigidify the molecular structure
that the enzymes retain their activity during storage
of the enzyme and concomitantly affects the enzyme
and use. Some enzymes retain their activity under oper-
turn-over [64]. Additionally, polar solvents can interfere
ational conditions for weeks or even months but others
with the ionic interactions of the protein and/or break-
do not. Excipients are required to increase the long-
ing polar interactions which induce at least a partial
term stability of enzymes following processing and stor-
unfolding of the molecular structure, especially in
age [62]. Moreover, despite enzymes are recognized by
enzymes with polar/dipolar transitions states and inter-
their several advantages has biocatalysts, like bio-
mediates. High conversion efficiency in non-aqueous
degradability, high specificity, and activity under mild
homogeneous biocatalysis can be achieved only with
conditions, they fail for most reactions when water is
high enzyme solubility and stability in the water–organic
not the preferable solvent. For this, enzymes must be
mixture.
tolerant to the presence of organics and the medium
engineering by addition of inorganic salts, polyols, and
Polyols, sugars, and salt addition (/ionic liquids)
sugars to the enzyme aqueous solution seems to be an
efficient approach [62,63]. Addition of polyols and sugars to aqueous solutions of
The structural stability of enzymes is controlled by proteins promotes strengthening of the hydrophobic
the interactions between protein molecules and the sur- interactions among non-polar amino acid residues, lead-
rounding solvent molecules [63]. Changes in the micro- ing to protein rigidification and enhancing thermostabil-
environment of the biocatalyst not only are able to ity [63]. Moreover, it has been postulated that the effect
modulate the enzyme activity and stability but also to of polyols on the activity of water seems to govern their
shape the enzyme selectivity. One of the main variables stabilization effect. Some theories state that the protect-
in biotransformations is the water content in the ive effect of sugars against destabilization when added
enzyme microenvironment. The water activity is relevant to the medium replace water molecules that are
since it involves the effect of water mass action on the removed from the hydration shells of the proteins
chemical equilibrium [64]. Besides, the role of water is [63,65–68] (Figure 3). Other hypothesis postulate that
complex and diverse, since water is able to participate the protective moieties are excluded from the surfaces
directly as a substrate, and/or during the transition of the protein entities and thus the available water mol-
states and/or as reaction product. Moreover, water can ecules in solution can interact with the protein entity,
take part in the reaction not only directly, but in an stabilizing its native configuration. Due to the preferen-
equally relevant role as a “‘lubricant”’, providing tial exclusion of the protective sugar moieties from the
342 C. SILVA ET AL.

Figure 3. Mechanisms for protein interaction with small and macromolecules and their effect on protein stabilization.

(immediate domain) hydration shell of protein entities, binding to them and disrupting the localized structure
sugar moieties shape a protective and stabilizing shield of water (the chaotropic effect). From this, it can be
around those biomolecules. The basis of this phenom- seen why ammonium sulfate and potassium hydrogen
enon is the difference in size between molecules of phosphate are a powerful enzyme stabilizers whereas
water and those of the sugar moieties. Essentially, a shell sodium thiosulfate and calcium chloride destabilize
is formed around the protein at the radius of closest enzymes. Many enzymes are specifically stabilized by
approach between the protein and the sugar moiety, a low concentrations of cations which may or may not
shell that is impenetrable to the sugar moieties but is form part of the active site, for example Ca2þ stabilizes
penetrable to water, resulting in an excess of water in a-amylases and Co2þ stabilizes glucose isomerases. At
the vicinity of the protein: preferential hydration. high concentrations (e.g. 20% NaCl), salt discourages
Preferential hydration of proteins is favored due to microbial growth due to its osmotic effect. In addition,
stronger interactions between sugar and water mole- ions can offer some protection against oxidation to
cules compared to those between sugar and protein groups such as thiols by salting-out the dissolved oxy-
molecules [69–71]. Such preferential exclusion increases gen from solution. The stabilization property of these
the chemical potential of the protein molecule efficient salts is governed by a competition phenom-
[69,72–75], proportionally to the solvent exposed sur- enon between salt exclusion and salt binding effects.
face area. According to the Le Chatelier's principle, sugar These salts increase the surface tension at water–
osmolytes favor the more compact state (viz, the native, enzyme interface and strengthen hydrophobic interac-
folded state, F) over the structurally expanded state (viz. tions by keeping hydrophobic moieties away from
the unfolded, denatured state, U), which leads to an water molecules [80–83]. Dissolved salts in aqueous sol-
increase of the Gibbs free energy change associated utions have a strong influence on protein-protein inter-
with the denaturation process (F () U) in the pres- actions and on the aggregates formed. The effect of
ence of osmolytes (one should remember at this point salts on protein solubility and stability have been
that DGD ¼ RTln([U]/[F])) [72,76–79]. known for more than a century and were first reported
In general, enzymes are stabilized by increasing their by Franz Hofmeister [84]. A variety of inorganic ions has
concentration and the ionic strength of the environ- been tested for their ability to precipitate various pro-
ment. Neutral salts compete with enzymes for water teins from solution. The overall order of a particular
and bind to charged groups or dipoles. This may result anion’s and cation’s effectiveness as a protein precipi-
in the interactions between enzyme's hydrophobic tant is generally as follows:
areas being strengthened and causing the enzyme mol- CO3 2 > SO4 2 > S2 O3 2 > H2 PO4 > F > Cl
ecules to compress and making them more resistant to > Br > NO3 > I > ClO4  > SCN
thermal unfolding reactions. Not all salts are equally
NðCH3 Þ4 þ > NH4 þ > Csþ > Rbþ > Kþ > Naþ > Liþ
effective in stabilizing hydrophobic interactions, some > Ca2þ > Mg2þ
are much more effective at their destabilization by
CRITICAL REVIEWS IN BIOTECHNOLOGY 343

Anions to the left of chloride are well hydrated and micelles; due to their hydrophobic nature, the alkyl
help salt proteins out of solution. Anions to the right of chains of organic cations in aqueous solutions are sur-
chloride generally salt proteins into solution and are rounded by water molecules forming “cage-like” struc-
more weakly hydrated. The salting-out anions have a tures so-called “hydrophobic hydration”; the inclusion
higher charge-to-volume ratio and generally have fairly of other molecules and macromolecules into the poly-
weak polarizabilities [84]. Work conducted on lysozyme meric IL network results in the formation of polar and
provided evidence that above protein isoelectric point, non-polar regions; the aqueous solution of free enzyme
the macromolecule bears a negative charge, and a dir- can be surrounded by the IL network retaining its activ-
ect Hofmeister series is observed. Chaotropes like I, ity [91]. A considerable number of enzymes are not sol-
ClO4, and SCN help to unfold proteins and salt them uble in most common ILs, being suspended in the
into solution. Kosmotropes, like SO42 and F, lead to reaction media with low water content. Thus, the net-
the stabilization of the folded state and cause a salting- work theory is not sufficient to explain the enzyme sta-
out effect. If the pH of the solution is below pl of the bilization by the ILs [91]. The impact of individual ions
protein, the macromolecules are net positively charged must also be considered. The effect of ILs aqueous solu-
and an inverse Hofmeister series is observed. tions on enzyme activity follows the ion kosmotropicity
Chaotropic anions become more effective at salting-out (Hofmeister series): kosmotropic anions and chaotropic
cations stabilize the enzyme, while chaotropic anions
proteins from solution than kosmotropic anions [85,86]
and kosmotropic cations destabilize (see examples in
(see Table 1 for other examples).
Table 1).
Ionic liquids are eco-friendly solvent media which
play an important role in several enzymatic reactions
[87]. ILs are composed of an organic cation involving Polymers and surfactants
imidazolium, pyrrolidinium, pyridinium, ammonium,
The stabilizing effect of polymers addition to enzyme
quanidinium, and other cations with a variety of sub-
formulations may promote the exclusion of protein
stituents and an organic anion (halides, tetrafluorobo-
molecules from part of the solvent and prevent detri-
rate, hexa fluorophosphates, and larger anions
mental effects of the environment upon the enzyme
containing sulfonynol or fluoroalkyl groups) [87]. ILs
molecules. By the inclusion of PEG, one of the most
have many favorable properties such as low vapor pres-
used stabilizing polymers, protein molecules became
sure, a wide liquid range, low flammability, high ionic hydrated, explained by a steric (hindrance) exclusion
conductivity, high thermal conductivity, high dissol- mechanism due to large difference in sizes between
ution capability toward many substrates, high thermal water and PEG molecules, with PEG being excluded
and chemical stability, and a wide electrochemical from enzyme vicinity [63,92].
potential window [88,89]. Because of these unique Enzyme-contained in a reverse micellar system is also
properties, ILs have been widely recognized as solvents a feasible strategy for enzyme stabilization [93]. The
or (co-)catalysts in a variety of applications including reversed micelle is formed by surfactant amphiphiles
organic catalysis, inorganic synthesis, biocatalysis, poly- self-aggregated in the bulk apolar organic solvents. The
merization, and engineering fluids. Typical IL cations are surfactant molecules assemble themselves with the
nitrogen-containing (such as alkylammonium, N,N0 -dia- polar head to the inner side and the apolar tail in con-
lkylimidazolium, N-alkylpyridinium, and pyrrolidinium), tact with the organic solvent. This self-aggregation only
or phosphorous-containing (such as alkylphosphonium). occurs when the surfactant concentration is above the
The common choices of anions include halides, BF 4, critical micelle concentration (CMC). Because of the
     
PF6 , CH3CO2 , CF3CO2 , NO3 , Tf2N , [(CF3SO2) 2N ], formed polar cores, reversed micellar system allows
[RSO4], and [RPO4] [88]. nanometer-sized aqueous droplets stabilized in it.
It has been postulated that some ILs properties, such The process of enzyme solubilization in reversed
as polarity, hydrogen-bond basicity, and nucleophilicity micelles results in the formation of enzyme-containing
of anions, Hofmeister series, hydrophobicity, and viscos- reversed micellar system, and the entrapped enzymes
ity, influence greatly the enzyme’s catalytic behavior have enhanced activities under those conditions as
[90]. Imidazolium ILs can form H-bonded polymeric suited in the lipid bilayers of biological membranes [93].
supramolecules, so-called organized nanostructures The surfactant concentration influences greatly the
with polar and non-polar regions responsible for enzyme solubilization and its catalytic efficiency. Some
enzyme stabilization; many ILs contain hydrophilic and works reported the decrease of catalytic activity with
lipophilic segments which turn them amphiphilic the increase of surfactant concentration in the organic
and behaving as surfactants forming aggregates and solvent [94].
344 C. SILVA ET AL.

Storage stabilization of enzymes gel, in a membrane device or in a microcapsule. The


entrapment requires the synthesis of a polymeric net-
Enzymes are generally best stored at 4  C in clean,
work in the presence of the enzyme to be encapsulated
autoclaved glassware or polypropylene tubes. Storage
[96]. Nanoparticles have been used as enzyme immobil-
at room temperature often leads to protein degradation
ization carriers [97]. Enzymes immobilized onto nano-
and/or inactivity, commonly as a result of microbial
sized scaffolds like spheres, fibers, and tubes have been
growth. For short-term storage (1 day to a few weeks),
reported [98]. Effective enzyme loading on nanopar-
many proteins may be stored in simple buffers at 4  C.
ticles has been achieved for up to 10 wt% due to a large
For long-term storage for 1 month to 1 year, some
surface area per unit mass of nanoparticles. Overall,
researchers choose to bead single-use aliquots of the
nanoparticles are considered to be an ideal support for
protein in liquid nitrogen for storage in clean plastic enzyme immobilization due to their minimized diffu-
containers under liquid nitrogen. This method involves sional limitations, maximum surface area per unit mass,
adding the protein solution dropwise (about 100 ll and high enzyme loading capability [98]. Theoretical
each) into a pool of liquid nitrogen, then collecting the and experimental studies have demonstrated that par-
drop-sized frozen beads and storing them in cryovials ticle mobility, which is governed by particle size and
under liquid nitrogen. solution viscosity, can impact the intrinsic activity of the
Frozen at 20  C or 80  C is the more common form particle-attached enzymes [97].
of cold protein storage. Because freeze-thaw cycles One of the most studied templates for enzyme immo-
decrease protein stability, samples for frozen storage are bilization is the encapsulation in silica matrices. Studies
best dispensed and prepared in single-use aliquots so on the immobilization of enzymes within biosilica nano-
that, once thawed, the protein solution will not have to particles formed by reaction of a silicate precursor with a
be refrozen. Alternatively, addition of 50% glycerol, ethyl- silica-precipitating peptide, demonstrated that the reac-
ene glycol, or DMSO will prevent solutions from freezing tion rapidly formed a network of fused silica nanospheres
at 20  C, enabling repeated use from a single stock that entrapped the peptide [99]. These nanodevices have
without warming (i.e. thawing). These inert substances been explored and demonstrated several advantages
modify the physicochemical properties of aqueous solu- compared with other encapsulation methods. They are
tions. They are preferentially excluded from contact with inexpensive, no need for chemical reagents for synthesis,
the protein surface, being the protein molecule hydrated, the immobilization occurs in seconds, the particles forma-
thus accounting their protective effect [63]. tion is done at mild conditions, the matrix can be dis-
Dilute protein solutions (<1 mg/ml) are more prone solved to release the entrapped enzyme and have a high
to inactivation and activity loss as a result of low-level level of stabilization uptake [98].
binding to the storage vessel. Therefore, it is common Most techniques for obtaining nanoparticles that
practice to add “carrier” or “filler” protein, such as puri- contain enzymes have been based on the so-called
fied bovine serum albumin (BSA) to 1–5 mg/ml “nanoentrapment” approach using the water-in-oil
(0.1–0.5%), to dilute protein solutions to protect them microemulsion system (also called reverse micelles),
against such degradation and loss. Many compounds which leads to discrete nanoparticles through polymer-
may be added to enzyme solutions to lengthen shelf ization in the water phase or water–oil interface [100].
life, namely cryoprotectants such as glycerol or ethylene Within these nanodevices the size and the number of
glycol to stabilize proteins by preventing the formation enzyme molecules within a reverse micelle is difficult to
of ice crystals at 20  C that destroy the protein struc- control. To overcome this, a new synthetic approach
ture; protease inhibitors to prevent proteolytic cleavage was reported under the name of “single enzyme nano-
of proteins; anti-microbial agents such as sodium azide particles (SENs)”, in which an organic-inorganic hybrid
(NaN3) to inhibit microbial growth; metal chelators such polymer network of a thickness of less than a few nano-
as EDTA to avoid metal-induced oxidation of –SH meters was built up from the surface of the enzyme.
groups and help to maintain the protein in a reduced This approach is therefore in stark contrast to the con-
state; reducing agents such a dithiothreitol (DTT) and 2- ventional nanoentrapment described above. SEN syn-
mercaptoethanol (2-ME) to maintain the protein in the thesis consists of three steps: (i) the enzyme surface is
reduced state by preventing oxidation of cysteines [95]. modified in water by acryloylation; (ii) the vinyl polymer
is grown from the enzyme surface in hexane; and (iii)
the attached polymer chains undergo orthogonal poly-
Enzyme encapsulation
merization via silanol condensation crosslinking [100].
The encapsulation of enzymes contemplates the entrap- SENs constitute a new approach in industrial enzyme
ment in a polymer network (gel lattice), in a silica sol– research [101,102]. The SEN form means that every
CRITICAL REVIEWS IN BIOTECHNOLOGY 345

single enzyme molecule is surrounded with a porous potential for the immobilization and preservation of
polymer matrix layer on a nanometer scale, resulting in enzymes [108]. The mechanisms underlying enzyme sta-
higher stability of the enzyme without any significant bilization by silk fibers can be divided in two groups:
limitation to mass transfer of the substrate from solu- mechanisms of stabilization in the liquid state and solid
tion to the active site. Individual magnetic enzyme states. In the liquid state, preferential exclusion, where
nanoparticles were produced by Yang et al. (2008) via silk fibroin is characterized by its steric exclusion ability
surface modification and aqueous polymerization of on proteins solution, and hydrophobic and electrostatic
each separate enzyme molecule. Their results show that binding. The later effect is explained by the silk’s amphi-
the stability of the enzyme and organic/inorganic net- philic nature, allowing for shielding of exposed hydro-
work composites is retained over a wide pH range phobic patches, by the high degree of hydrophobicity
(pH ¼ 5.5–9.0) and at high temperatures (60–70  C) in and by its high charge which by interaction with pro-
organic solvents [103]. tein surroundings can alter the exposed surface charge
One-dimensional nanostructured materials such as distribution, changing the protein–protein interaction
fibers, wires, rods, belts, tubes, spirals, and rings have [108]. In the solid state, the mechanisms of stabilization
attracted the interest for enzyme encapsulation. are related with the water replacement hypothesis and
Electrospun nanofibers are simple and most used for glass dynamics or vitrification hypothesis [108].
this purpose. They are long, uniform in diameter, and
can be diversified in composition [96].
Major remarks and future perspectives
The enzyme encapsulation into liposomes is a prom-
ising strategy to stabilize and prevent denaturation. By Enzyme stabilization does not end on the methodolo-
controlling lipid membrane permeability it is possible to gies described during discussion, it is important to note
control the permeability of liposomes increasing the that there are procedures wherein diverse stabilization
permeation of the substrate. These devices allow the methods have been applied simultaneously or consecu-
stabilization of and succeed to maintain the full enzyme tively to afford the best stabilization conditions, higher
function [104]. yield, and recovery of the support material after enzyme
Enzyme immobilization is an important strategy to inactivation [3].
enhance the stability and recoverability of enzymes and Examples have been discussed: (i) the case of site-
to facilitate the separation of enzymes from reaction directed mutagenesis of penicillin G acylase from
products. However, enzyme purification followed by Escherichia coli for better immobilization [109], (ii)
separate chemical steps to allow immobilization on a immobilization followed by modification of penicillin
solid support reduces the efficiency and yield of the acylase with aldehyde-dextran [110], (iii) multipoint
active enzyme covalent attachment of penicillin acylase and later
Strategies that use enzymes as a buildingblock for entrapping the enzyme into a hyperhydrophilic nanoen-
biomaterials have been developed to overcome some vironment [111], (iv) PEG-ylation and subsequent immo-
drawbacks related with enzyme purification and chem- bilization of creatinine amidohydrolase for biosensor
ical separation. Recently, Zhou and coworkers described development [112], (v) site-directed mutagenesis of
polypeptide constructs that self-assemble spontan- thermolysin-like neutral protease from Bacillus stearo-
eously into nanofibrils with fused active enzyme subu- thermophilus followed by immobilization [113], (vi) use
nits displayed on the amyloid fibril surface. They of stabilizing additives in combination with covalently
showed that the fibrils can be recycled and reused in immobilized a-chymotrypsin [114], and (vii) chemical
functional assays both in conventional batch processes modification of surface groups of penicillin G acylase fol-
and in a continuous-flow microreactor [105]. They also lowed by immobilization of the modified enzyme [115].
developed enzymatically active microgels that are stabi- The selection of the best stabilization approach from
lized by amyloid nanofibrils [106]. The protein nanofi- the plethora of methods available is highly dependent
brils were also applied to generate colloidosome-like on the enzyme–stabilizer interactions and possible
two-dimensional crosslinked networks of nanostruc- changes in the catalytic efficiency of the enzymes.
tures templated by all-aqueous emulsions, which were Optimized stabilization parameters will lead to more
nominated as fibrillosomes [107]. efficient enzymes as well as to an increase of the eco-
nomic potential of the existing enzymatic processes in
novel areas where the catalysts have not been applied
Enzyme stabilization by silk
due to their instability.
Silk fibroin is a biologically derived protein from domes- The growing replacement of conventional chemical
ticated silkworm cocoons and is a material with great methods in laboratories and industries by enzyme-
346 C. SILVA ET AL.

based strategies demands continuous research and protein structure, folding, and stability. CBE Life Sci
improvement of the stabilization strategies. New meth- Educ. 2010;9:189–195.
[11] Burioni R, Cassi D, Cecconi F, et al. Topological ther-
ods and/or combination of different techniques would
mal instability and length of proteins. Proteins.
allow to overcome the current limitations related with 2004;55:529–535.
the enzyme stabilization decreasing costs and storage [12] Bommarius AS, Paye MF. Stabilizing biocatalysts.
problems and improving commercialization. Chem Soc Rev. 2013;42:6534–6565.
[13] Saptarshi SR, Duschl A, Lopata AL. Interaction of
nanoparticles with proteins: relation to bio-reactivity
Disclosure statement of the nanoparticle. J Nanobiotechnology. 2013;11:
26–21.
No potential conflict of interest was reported by the authors. [14] Salloum DS, Schlenoff JB. Protein adsorption modal-
ities on polyelectrolyte multilayers. Biomacromolecules.
2004;5:1089–1096.
Funding [15] Jeong S. Analytical methods and formulation factors
The authors would like to thank Portuguese Science and to enhance protein stability in solution. Arch Pharm
Technology Foundation (FCT) under the scope of the stra- Res. 2012;35:1871–1886.
tegic funding of UID/BIO/04469/2013 unit and COMPETE [16] Sinha N, Mohan S, Lipschultz CA, et al. Differences in
2020 (POCI-01-0145-FEDER-006684) and BioTecNorte oper- electrostatic properties at antibody–antigen binding
ation (NORTE-01-0145-FEDER-000004) funded by European sites: implications for specificity and cross-reactivity.
Regional Development Fund under the scope of Norte2020 – Biophys J. 2002;83:2946–2968.
Programa Operacional Regional do Norte. They would like [17] Andrade JD, Hlady V. Protein adsorption materials
also to thank the Program for Changjiang Scholars and biocompatibility: a tutorial review. Berlin Heidelberg:
Innovative Research Team in University (IRT1135). This study Springer, 1986. p. 1–63.
was also supported by Chinese Government Scholarship [18] van der Veen M, Norde W, Stuart MC. Electrostatic
interactions in protein adsorption probed by compar-
under China Scholarship Council (No. 201606790036) and
ing lysozyme and succinylated lysozyme. Colloids
Chinese Foundation Key projects of governmental cooper-
Surf B. 2004;35:33–40.
ation in International Scientific and Technological Innovation
[19] Yano YF, Uruga T, Tanida H, et al. Protein salting out
(No. 2016 YFE0115700).
observed at an air  water interface. J Phys Chem
Lett. 2011;2:995–999.
References [20] Dill KA. Dominant forces in protein folding.
Biochemistry. 1990;29:7133–7155.
[1] Kumar VSD, Sangwan P. Industrial enzymes trends: [21] Shen DZ, Xue YH, Kang Q, et al. Monitoring of
scope and relevance. 1st ed. Chapter: 10. In: Beniwal adsorption of lysozyme onto quartz using an elec-
V, Sharma AK, editors. Global market scenario of trode-separated piezoelectric sensor. Microchem J.
industrial enzymes. India: Nova Science Publishers; 1998;60:1–7.
2014. p.173–196. [22] Dobson CM. Protein folding and misfolding. Nature.
[2] Beilen JB, Li Z. Enzyme technology: an overview. Curr 2003;426:884–890.
Opin Biotechnol. 2002;13:338–344. [23] Demirjian DC, Mor{s-Varas F, Cassidy CS. Enzymes
[3] Iyer PV, Ananthanarayan L. Enzyme stability and sta- from extremophiles. Curr Opin Chem Biol. 2001;5:
bilization—aqueous and non- aqueous environment. 144–151.
Process Biochem. 2008;43:1019–1032. [24] Bergquist PL, Morgan HW, Saul D. Selected enzymes
[4]  again C. Enzyme stabilization—recent experimen-
O’F from extreme thermophiles with applications in bio-
tal progress. Enzyme Microb Technol. 2003;33: technology. Curr Biotechnol. 2014;3:45–59.
137–49. [25] Haki GD, Rakshit SK. Developments in industrially
[5] Kirk O, Borchert TV, Fuglsang CC. Industrial enzyme important thermostable enzymes: a review. Bioresour
applications. Curr Opin Biotechnol. 2002;13:345–351. Technol. 2003;89:17–34.
[6] Gianfreda L, Scarfi MR. Enzyme stabilization: state of [26] Andrade CMMC Pereira Jr, N, Antranikian G.
the art. Mol Cell Biochem. 1991;100:97–128. Extremely thermophilic microorganisms and their
[7] Horwitz J, Bova MP, Ding L, et al. crystallin: function polymer-hidrolytic enzymes. Rev Microbiol. 1999;30:
and structure. Eye. 1999;13:403–408. 287–298.
[8] Katyal N, Deep S. Revisiting the conundrum of trehal- [27] Bornscheuer UT, Huisman GW, Kazlauskas RJ, et al.
ose stabilization. Phys Chem Chem Phys. 2014;16: Engineering the third wave of biocatalysis. Nature.
26746–26761. 2012;485:185–194.
[9] Chang BS, Yeung B. Physical stability of protein phar- [28] Davids T, Schmidt M, Bo €ttcher D, et al. Strategies for
maceuticals. formulation and process development the discovery and engineering of enzymes for bioca-
strategies for manufacturing biopharmaceuticals. talysis. Curr Opin Chem Biol. 2013;17:215–220.
California: John Wiley & Sons, Inc. 2010. p. 69–104. [29] Behrens GA, Hummel A, Padhi SK, et al. Discovery
[10] Robic S. Mathematics, thermodynamics, and model- and protein engineering of biocatalysts for organic
ing to address ten common misconceptions about synthesis. Adv Synth Catal. 2011;353:2191–2215.
CRITICAL REVIEWS IN BIOTECHNOLOGY 347

[30] Lutz S. Beyond directed evolution—semi-rational pro- [49] Silva CJ, Sousa F, Guebitz G, Cavaco-Paulo A.
tein engineering and design. Curr Opin Biotechnol. Chemical modifications on proteins using glutaralde-
2010;21:734–743. hyde. Food Technol Biotechnol. 2004;42:51–16.
[31] Bommarius AS, Blum JK, Abrahamson MJ. Status of [50] Mayolo-Deloisa K, Gonzalez-Gonzalez M, Simental-
protein engineering for biocatalysts: how to design Martınez J, et al. Aldehyde PEGylation of laccase
an industrially useful biocatalyst. Curr Opin Chem from Trametes versicolor in route to increase its sta-
Biol. 2011;15:194–200. bility: effect on enzymatic activity. J Mol Recognit.
[32] Bo€ ttcher D, Bornscheuer UT. Protein engineering of 2015;28:173–179.
microbial enzymes. Curr Opin Microbiol. 2010;13: [51] Lopez-Cruz JI, Viniegra-Gonzalez G, Hernandez-
274–282. Arana A. Thermostability of native and pegylated
[33] Woodley JM. Protein engineering of enzymes for pro- myceliophthora thermophila laccase in aqueous and
cess applications. Curr Opin Chem Biol. 2013;17: mixed solvents. Bioconjugate Chem. 2006;17:
310–316. 1093–1098.
[34] Otten LG, Hollmann F, Arends IWCE. Enzyme engin- [52] Homaei AA, Sariri R, Vianello F, et al. Enzyme immo-
eering for enantioselectivity: from trial-and-error to bilization: an update. J Chem Biol. 2013;6:185–205.
rational design? Trends Biotechnol. 2010;28:46–54. [53] Asgher M, Shahid M, Kamal S, et al. Recent trends
[35] Brode PF, Erwin CR, Rauch DS, et al. Subtilisin and valorization of immobilization strategies and lig-
BPN‘Variants: increased hydrolytic activity on surface- ninolytic enzymes by industrial biotechnology. J Mol
bound substrates via decreased surface activity. Catal B Enzym. 2014;101:56–66.
Biochemistry. 1996;35:3162–3169. [54] Ritter DW, Newton JM, McShane MJ. Modification of
[36] Beer HD, Wohlfahrt G, McCarthy JEG, et al. Analysis PEGylated enzyme with glutaraldehyde can enhance
of the catalyic mechanism of a fungal lipase using stability while avoiding intermolecular crosslinking.
computer-aided design and structural mutants. RSC Adv. 2014;4:28036–28040.
Protein Eng Des Sel. 1996;9:507–517. [55] Ansari SA, Husain Q. Potential applications of
[37] Martinelle M, Holmquist M, Clausen IG, et al. The role enzymes immobilized on/in nano materials: a review.
of Glu87 and Trp89 in the lid of Humicola lanuginosa Biotechnol Adv. 2012;30:512–523.
lipase. Protein Eng Des Sel. 1996;9:519–524. [56] Cowan DA, Fernandez-Lafuente R. Enhancing the
[38] Pedersen S, Lange NK, Nissen AM. Novel industrial functional properties of thermophilic enzymes by
enzyme applications. Ann Ny Acad Sci. 1995;750: chemical modification and immobilization. Enzyme
376–390. Microb Technol. 2011;49:326–346.
[39] Jegannathan KR, Nielsen PH. Environmental assess- [57] Taylor RF. A comparison of various commercially-
ment of enzyme use in industrial production – a lit- available liquid chromatographic supports for immo-
erature review. J Clean Prod. 2013;42:228–240. bilization of enzymes and immunoglobulins. Anal
[40] Koivula A, Reinikainen T, Ruohonen L, et al. The Chim Acta. 1985;172:241–248.
active site of Trichoderma reesei cellobiohydrolase II: [58] Palomo JM. Lipases enantioselectivity alteration by
the role of tyrosine 169. Protein Eng. 1996;9:691–699. immobilization techniques. Curr Bioact Comp. 2008;4:
[41] Davis BG. Chemical modification of biocatalysts. Curr 126–138.
Opin Biotechnol. 2003;14:379–386. [59] Kamori M, Hori T, Yamashita Y, et al. Immobilization
[42] Vinogradov AA, Kudryashova EV, Grinberg VY, et al. of lipase on a new inorganic ceramics support,
The chemical modification of a-chymotrypsin with toyonite, and the reactivity and enantioselectivity of
both hydrophobic and hydrophilic compounds stabil- the immobilized lipase. J Mol Catal B Enzym.
izes the enzyme against denaturation in water–or- 2000;9:269–274.
ganic media. Protein Eng. 2001;14:683–689. [60] Sangeetha K, Emilia Abraham T. Preparation and
[43] Ueji S-i, Tanaka H, Hanaoka T, et al. Effects of chem- characterization of cross-linked enzyme aggregates
ical modification of lipase on its enantioselectivity in (CLEA) of Subtilisin for controlled release applica-
organic solvents. Chem Lett. 2001;30:1066–1067. tions. Int J Biol Macromol. 2008;43:314–319.
[44] Dıaz-Rodrıguez A, Davis BG. Chemical modification in [61] Talekar S, Ghodake V, Ghotage T, et al. Novel mag-
the creation of novel biocatalysts. Curr Opin Chem netic cross-linked enzyme aggregates (magnetic
Biol. 2011;15:211–219. CLEAs) of alpha amylase. Bioresour Technol.
[45] Bund RK, Singhal RS. Chemical modification of cellu- 2012;123:542–547.
lase by maleic anhydride and N-bromosuccinimide [62] Stepankova V, Bidmanova S, Koudelakova T, et al.
for improved detergent stability. J Surfact Deterg. Strategies for stabilization of enzymes in organic
2002;5:1–4. solvents. ACS Catal. 2013;3:2823–2836.
[46] DeSantis G, Jones JB. Chemical modification of [63] Balc~ao VM, Vila MMDC. Structural and functional sta-
enzymes for enhanced functionality. Curr Opin bilization of protein entities: state-of-the-art. Adv
Biotechnol. 1999;10:324–330. Drug Deliv Rev. 2015;93:25–41.
[47] Quiocho FA, Richards FM. Intermolecular cross linking [64] Hudson EP, Eppler RK, Clark DS. Biocatalysis in semi-
of a protein in the crystalline state: carboxypepti- aqueous and nearly anhydrous conditions. Curr Opin
dase-A. Proc Natl Acad Sci USA. 1964;52:833–839. Biotechnol. 2005;16:637–643.
[48] Govardhan CP. Crosslinking of enzymes for improved [65] Scharnagl C, Reif M, Friedrich J. Stability of proteins:
stability and performance. Curr Opin Biotechnol. temperature, pressure and the role of the solvent.
1999;10:331–335. Biochim Biophys Acta. 2005;1749:187–213.
348 C. SILVA ET AL.

[66] Miyawaki O. Hydration state change of proteins upon [84] Kherb J, Flores SC, Cremer PS. Role of carboxylate
unfolding in sugar solutions. Biochim Biophys Acta. side chains in the cation hofmeister series. J Phys
2007;1774:928–935. Chem B. 2012;116:7389–7397.
[67] Kumar V, Chari R, Sharma VK, et al. Modulation of [85] Zhang Y, Cremer PS. The inverse and direct
the thermodynamic stability of proteins by polyols: Hofmeister series for lysozyme. Proc Natl Acad Sci
significance of polyol hydrophobicity and impact USA. 2009;106:15249–15253.
on the chemical potential of water. Int J Plast. [86] Sedlak E, Stagg L, Wittung-Stafshede P. Effect of
2011;413:19–28. Hofmeister ions on protein thermal stability: Roles of
[68] Saenger W. Structure and dynamics of water sur- ion hydration and peptide groups? Arch Biochem
rounding biomolecules. Annu Rev Biophys Biophys Biophys. 2008;479:69–73.
Chem. 1987;16:93–114. [87] Gao W-W, Zhang F-X, Zhang G-X, et al. Key factors
[69] Arakawa T, Prestrelski SJ, Kenney WC, et al. Factors affecting the activity and stability of enzymes in ionic
affecting short-term and long-term stabilities of pro- liquids and novel applications in biocatalysis.
teins. Adv Drug Deliv Rev. 1993;10:1–28. Biochem Eng J. 2015;99:67–84.
[70] Wong YH, Tayyab S. Protein stabilizing potential of [88] Zhao H. Ionic liquids as (Co-)solvents for hydrolytic
simulated honey sugar cocktail under various enzymes. Ionic liquids in biotransformations and
denaturation conditions. Process Biochem. 2012;47: organocatalysis. Hoboken (NJ): John Wiley & Sons,
1933–1943. Inc., 2012. p. 151–227.
[71] Carpenter JF, Crowe JH. The mechanism of cryopro- [89] Weingartner H, Cabrele C, Herrmann C. How ionic
tection of proteins by solutes. Cryobiology. 1988;25: liquids can help to stabilize native proteins. Phys
244–255. Chem Chem Phys. 2012;14:415–426.
[72] Haque I, Singh R, Moosavi-Movahedi AA, et al. Effect [90] Yang Z. Hofmeister effects: an explanation for the
of polyol osmolytes on DgD, the Gibbs energy of sta- impact of ionic liquids on biocatalysis. J Biotechnol.
bilisation of proteins at different pH values. Biophys
2009;144:12–22.
Chem. 2005;117:1–12. [91] Zhao H. Protein stabilization and enzyme activation
[73] Haque I, Singh R, Ahmad F, et al. Testing polyols’ com-
in ionic liquids: specific ion effects. J Chem Technol
patibility with Gibbs energy of stabilization of proteins
Biotechnol. 2016;91:25–50.
under conditions in which they behave as compatible
[92] Shulgin IL, Ruckenstein E. Preferential hydration and
osmolytes. FEBS Lett. 2005;579:3891–3898.
solubility of proteins in aqueous solutions of poly-
[74] Ebel C, Eisenberg H, Ghirlando R. Probing protein-
ethylene glycol. Biophys Chem. 2006;120:188–198.
sugar interactions. Biophys J. 2000;78:385–393.
[93] Liang Y, Yuan X, Zeng G, et al. Effects of surfactants
[75] Timasheff SN, Arakawa T. Mechanism of protein pre-
on enzyme-containing reversed micellar system. Sci
cipitation and stabilization by co-solvents. J Cryst
China Chem. 2011;54:715–723.
Growth. 1988;90:39–46.
[94] Brown ED, Yada RY, Marangoni AG. The dependence
[76] Sanchez-Ruiz JM. Protein kinetic stability. Biophys
of the lipolytic activity of Rhizopus arrhizus lipase on
Chem. 2010;148:1–15.
[77] Becktel WJ, Schellman JA. Protein stability curves. surfactant concentration in Aerosol-OT/isooctane
Biopolymers. 1987;26:1859–1877. Peptide Science reverse micelles and its relationship to enzyme struc-
Section. ture. Biochim Biophys Acta. 1993;1161:66–72.
[78] Miyawaki O, Tatsuno M. Thermodynamic analysis of [95] Simpson RJ. Stabilization of Proteins for Storage.
alcohol effect on thermal stability of proteins. Cold Spring Harb Protoc. 2010;2010:pdb.top79.
J Biosci Bioeng. 2011;111:198–203. [96] Hwang ET, Gu MB. Enzyme stabilization by nano/
[79] Linhananta A, Hadizadeh S, Plotkin SS. An effective microsized hybrid materials. Eng Life Sci. 2013;13:
solvent theory connecting the underlying mecha- 49–61.
nisms of osmolytes and denaturants for protein sta- [97] Jia H, Zhu G, Wang P. Catalytic behaviors of enzymes
bility. Biophys J. 2011;100:459–468. attached to nanoparticles: the effect of particle
[80] Kristjansson MM, Kinsella JE. Protein and enzyme sta- mobility. Biotechnol Bioeng. 2003;84:406–414.
bility: structural, thermodynamic, and experimental [98] Betancor L, Luckarift HR. Bioinspired enzyme encap-
aspects. In: John EK, editor. Advances in food and sulation for biocatalysis. Trends Biotechnol. 2008;26:
nutrition research. Netherlands: Academic Press; 566–572.
1991. p. 237–316. [99] Poulsen N, Sumper M, Kro €ger N. Biosilica formation
[81] Lavelle L, Fresco JR. Stabilization of nucleic acid in diatoms: characterization of native silaffin-2 and its
triplexes by high concentrations of sodium and role in silica morphogenesis. Proc Natl Acad Sci USA.
ammonium salts follows the Hofmeister series1. 2003;100:12075–12080.
Biophys Chem. 2003;105:681–699. [100] Kim J, Grate JW, Wang P. Nanobiocatalysis and its
[82] Kamiyama T, Sadahide Y, Nogusa Y, et al. Polyol- potential applications. Trends Biotechnol. 2008;26:
induced molten globule of cytochrome c: an evi- 639–646.
dence for stabilization by hydrophobic interaction. [101] Kim J, Grate JW. Single-enzyme nanoparticles arm-
Biochim Biophys Acta. 1999;1434:44–57. ored by a nanometer-scale organic/inorganic net-
[83] Timasheff SN, Arakawa T. Mechanism of protein pre- work. Nano Lett. 2003;3:1219–1222.
cipitation and stabilization by co-solvents. J Cryst [102] Kim J, Jia H, Lee C-w, et al. Single enzyme nanopar-
Growth. 1988;90:39–46. ticles in nanoporous silica: a hierarchical approach to
CRITICAL REVIEWS IN BIOTECHNOLOGY 349

enzyme stabilization and immobilization. Enzyme [118] Levene R, Cohen Y, Barkai D. Applying proteases to
Microb Technol. 2006;39:474–480. confer improved shrink resistance to wool. J Soc
[103] Yang Z, Si S, Zhang C. Magnetic single-enzyme nano- Dyers Colour. 1996;112:6–10.
particles with high activity and stability. Biochem [119] Freddi G, Mossotti R, Innocenti R. Degumming of silk
Biophys Res Commun. 2008;29:169–175. fabric with several proteases. J Biotechnol. 2003;106:
[104] Colletier J-P, Chaize B, Winterhalter M, et al. Protein 101–112.
encapsulation in liposomes: efficiency depends on [120] Shen J, Rushforth M, Cavaco-Paulo A, et al.
interactions between protein and phospholipid Development and industrialisation of enzymatic
bilayer. BMC Biotechnol. 2002;2:9. shrink-resist process based on modified proteases for
[105] Zhou X-M, Entwistle A, Zhang H, et al. Self-assembly wool machine washability. Enzyme Microb Technol.
of amyloid fibrils that display active enzymes. 2007;40:1656–1661.
ChemCatChem. 2014;6:1961–1968. [121] Araujo R, Silva C, Machado R, et al. Proteolytic
[106] Zhou X-M, Shimanovich U, Herling TW, et al. enzyme engineering: a tool for wool.
Enzymatically active microgels from self-assembling Biomacromolecules. 2009;10:1655–1661.
protein nanofibrils for microflow chemistry. ACS [122] Hajighasemi M, Nocek BP, Tchigvintsev A, et al.
Nano. 2015;9:5772–5781. Biochemical and structural insights into enzymatic
[107] Song Y, Shimanovich U, Michaels TCT, et al. depolymerization of polylactic acid and other
Fabrication of fibrillosomes from droplets stabilized polyesters by microbial carboxylesterases.
by protein nanofibrils at all-aqueous interfaces. Nat Biomacromolecules. 2016;17:2027–2039.
Commun. 2016;7:12934. [123] Yang Y, Yu Y, Zhang Y, et al. Lipase/esterase-
[108] Li AB, Kluge JA, Guziewicz NA, et al. Silk-based stabil- catalyzed ring-opening polymerization: a green poly-
ization of biomacromolecules. J Control Release. ester synthesis technique. Process Biochem. 2011;46:
2015;219:416–430. 1900–1908.
[109] Montes T, Graz u V, Lo pez-Gallego F, et al. Genetic [124] Silva CM, Carneiro F, O’Neill A, et al. Cutinase—a
modification of the penicillin G acylase surface to new tool for biomodification of synthetic fibers.
improve its reversible immobilization on ionic J Polym Sci A Polym Chem. 2005;43:2448–2450.
exchangers. Appl Environ Microbiol. 2007;73: [125] Ronkvist ÅM, Xie W, Lu W, et al. Cutinase-catalyzed
312–319. hydrolysis of poly(ethylene terephthalate).
[110] Fernandez-Lafuente R, Rosell CM, Caanan-Haden L, Macromolecules. 2009;42:5128–5138.
et al. Facile synthesis of artificial enzyme nano-envi- [126] Matama T, Ara ujo R, G€ ubitz GM, et al.
ronments via solid-phase chemistry of immobilized Functionalization of cellulose acetate fibers with
derivatives: dramatic stabilization of penicillin acylase engineered cutinases. Biotechnol Progress. 2010;26:
versus organic solvents. Enzyme Microb Technol. 636–643.
1999;24:96–103. [127] Araujo R, Silva C, O’Neill A, et al. Tailoring cutinase
[111] Abian O, Mateo C, Fernandez-Lorente G, et al. activity towards polyethylene terephthalate and poly-
Stabilization of immobilized enzymes against water- amide 6,6 fibers. J Biotechnol. 2007;128:849–857.
soluble organic cosolvents and generation of hyper- [128] Galli C, Gentili P, Jolivalt C, et al. How is the reactivity
hydrophilic micro-environments surrounding enzyme of laccase affected by single-point mutations?
molecules. Biocatal Biotransformation. 2001;19: Engineering laccase for improved activity towards
489–503. sterically demanding substrates. Appl Microbiol
[112] Hassani L, Ranjbar B, Khajeh K, et al. Horseradish per- Biotechnol. 2011;91:123–131.
oxidase thermostabilization: the combinatorial effects [129] Zhang Z-G, Yi Z-L, Pei X-Q, et al. Improving the ther-
of the surface modification and the polyols. Enzyme mostability of Geobacillus stearothermophilus xyla-
Microb Technol. 2006;38:118–125. nase XT6 by directed evolution and site-directed
[113] Chan L, Cross HF, She JK, et al. Covalent attachment mutagenesis. Bioresour Technol. 2010;101:9272–9278.
of proteins to solid supports and surfaces via sor- [130] Singh RK, Zhang Y-W, Nguyen N-P-T, et al. Covalent
tase-mediated ligation. PLoS One. 2007;2:e1164. immobilization of b-1,4-glucosidase from Agaricus
[114]  K, Szava A, Maria Simon L. Stabilization of vari-
Laszlo arvensis onto functionalized silicon oxide nanopar-
ous a-chymotrypsin forms in aqueous-organic media ticles. Appl Microbiol Biotechnol. 2011;89:337–344.
by additives. J Mol Catal B Enzym. 2001;16:141–146. [131] v~ao RO, Silverio SC, Tavares APM, et al. Green
Cristo
[115] Montes T, Grazu V, Lo pez-Gallego F, et al. Chemical coconut fiber: a novel carrier for the immobilization
modification of protein surfaces to improve their of commercial laccase by covalent attachment for
reversible enzyme immobilization on ionic exchang- textile dyes decolourization. World J Microbiol
ers. Biomacromolecules. 2006;7:3052–3058. Biotechnol. 2012;28:2827–2838.
[116] Araujo R, Casal M, Cavaco-Paulo A. Application of [132] Silva CJSM Zhang Q, Shen J, et al. Immobilization of
enzymes for textile fibres processing. Biocatal proteases with a water soluble–insoluble reversible
Biotransformation. 2008;26:332–349. polymer for treatment of wool. Enzyme Microb
[117] Chand N, Nateri AS, Sajedi RH, et al. Enzymatic desiz- Technol. 2006;39:634–640.
ing of cotton fabric using a Ca2þ-independent [133] Silva C, Silva CJ, Zille A, et al. Laccase immobilization
a-amylase with acidic pH profile. J Mol Catal B on enzymatically functionalized polyamide 6,6 fibres.
Enzym. 2012;83:46–50. Enzyme Microb Technol. 2007;41:867–875.
350 C. SILVA ET AL.

[134] Miletic N, Nastasovic A, Loos K. Immobilization of macromolecular substrates under a time-varying


biocatalysts for enzymatic polymerizations: possibil- magnetic field. J Biotechnol. 2010;148:119–127.
ities, advantages, applications. Bioresour Technol. [139] Martins M, Azoia N, Silva C, et al. Stabilization of
2012;115:126–135. enzymes in micro-emulsions for ultrasound proc-
[135] Stavila E, Arsyi RZ, Petrovic DM, et al. Fusarium solani esses. Biochem Eng J. 2015;93:115–118.
pisi cutinase-catalyzed synthesis of polyamides. Eur [140] Lu S, Wang X, Lu Q, et al. Stabilization of enzymes
Polymer J. 2013;49:834–842. in silk films. Biomacromolecules. 2009;10:1032–1042.
[136] Zhao H, Olubajo O, Song Z, et al. Effect of kosmotro- [141] Demura M, Asakura T, Kuroo T. Immobilization
picity of ionic liquids on the enzyme stability in of biocatalysts with bombyx mori silk fibroin by
aqueous solutions. Bioorg Chem. 2006;34:15–25. several kinds of physical treatment and its applica-
[137] Yoshimoto M. Stabilization of enzymes through encap- tion to glucose sensors. Biosensors. 1989;4:
sulation in liposomes. In: Minteer DS, editor. Enzyme 361–372.
stabilization and immobilization: methods and proto- [142] Shaw KL, Grimsley GR, Yakovlev GI, et al. The effect
cols. Totowa, NJ: Humana Press; 2011. p. 9–18. of net charge on the solubility, activity, and stability
[138] Yang K, Xu N-S, Su WW. Co-immobilized enzymes in of ribonuclease Sa. Protein Sci Soc. 2001;10:
magnetic chitosan beads for improved hydrolysis of 1206–1215.

You might also like