You are on page 1of 27

International Journal of Biological Macromolecules 101 (2017) 931–957

Contents lists available at ScienceDirect

International Journal of Biological Macromolecules


journal homepage: www.elsevier.com/locate/ijbiomac

Review

Recent advances in enzyme extraction strategies: A comprehensive


review
Shamraja S. Nadar, Rohini G. Pawar, Virendra K. Rathod ∗
Department of Chemical Engineering, Institute of Chemical Technology, Matunga (E), Mumbai 400019, India

a r t i c l e i n f o a b s t r a c t

Article history: The increasing interest of industrial enzymes demands for development of new downstream strate-
Received 6 December 2016 gies for maximizing enzyme recovery. The significant efforts have been focused on the development of
Received in revised form 7 March 2017 newly adapted technologies to purify enzymes in catalytically active form. Recently, an aqueous two
Accepted 10 March 2017
phase system (ATPS) is emerged as powerful tools for efficient extraction and purification of enzymes
Available online 14 March 2017
due to their versatility, lower cost, process integration capability and easy scale-up. The present review
gives an overview of effect of parameters such as tie line length, pH, neutral salts, properties of polymer
Keywords:
and salt involved in traditional polymer/polymer and polymer/salt ATPS for enzyme recovery. Further,
Enzymes
Extraction
advanced ATPS have been developed based on alcohols, surfactants, micellar compounds to avoid tedious
Purification recovery steps for getting desired enzyme. In order to improve the selectivity and efficiency of ATPS,
Aqueous two phase system recent approaches of conventional ATPS combined with different techniques like affinity ligands, ionic
Three phase partitioning liquids, thermoseparating polymers and microfluidic device based ATPS have been reviewed. Moreover,
CLEAs three phase partitioning is also highlighted for enzymes enrichment as a blooming technology for effi-
Nanoflowers ciently integrated bioseparation techniques. At the end, it includes an overview of CLEAs technology and
organic–inorganic nanoflowers preparation as novel strategies for simultaneous extraction, purification
and immobilization of enzymes.
© 2017 Elsevier B.V. All rights reserved.

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 932
2. Aqueous two phase system . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 932
2.1. Factors affecting partitioning of enzymes in aqueous two phase partitioning . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 933
2.1.1. Nature and concentration of polymer . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 933
2.1.2. Polymer/salt aqueous two phase partitioning system . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 935
2.1.3. Polymer/polymer aqueous two phase partitioning system . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 937
2.1.4. Effect of pH on partitioning of enzymes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 937
2.1.5. Effect of volume ratio and tie-lines length on partitioning of enzymes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 938
2.1.6. Effect of neutral salts on enzyme partitioning . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 938
3. Alcohol/salt based aqueous two phase partitioning system . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 938
4. Surfactant/alcohol based aqueous two phase partitioning system . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 939
5. Advanced aqueous two phase partitioning system . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 939
5.1. Aqueous two-phase affinity partitioning system . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 940
5.2. Ionic liquid based aqueous two phase partitioning system . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 942
5.3. Thermoseparating polymer-based aqueous two-phase system . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 942
5.4. Aqueous micellar two-phase system . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 944
5.5. Microfluidic platform for ATPS . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 945

∗ Corresponding author.
E-mail address: vk.rathod@ictmumbai.edu.in (V.K. Rathod).

http://dx.doi.org/10.1016/j.ijbiomac.2017.03.055
0141-8130/© 2017 Elsevier B.V. All rights reserved.
932 S.S. Nadar et al. / International Journal of Biological Macromolecules 101 (2017) 931–957

6. Three phase partitioning . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 945


7. Simultaneous purification and immobilization strategies . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 948
8. Future perspectives and challenges . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 949
9. Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 950
Acknowledgement . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 950
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 950

1. Introduction well as they should be sustainable, efficient and biocompatible with


the enzyme conformational structure [14,15]. So there is a need
Many chemical transformation processes are carried out in to develop easy, fast, scalable and economically feasible methods
extreme conditions, high temperature and pressure involving harsh to extract enzymes either from fermentation or natural sources
and hazardous processing which led to higher consumption of (Fig. 1). In this review, we have explained various types of extrac-
chemicals and energy [1]. Also, harmful by-products are pro- tion methods with their modifications and parameters affecting the
duced due to non-specificity which have huge negative impact on extraction of enzymes for the optimization and design of efficient
the environment [2]. Hence, there is a need for development of and economic downstream processes. Also, we have mentioned
clean, bio-compatible, reliable and benign processes for industrial certain methods of simultaneous purification and immobilization
manufacturing of chemicals. Biocatalysis has emerged as a pos- of enzyme which make its application economically feasible.
sible eco-friendly and greener catalysts, meeting the demands of
green chemistry and sustainable development which is directing 2. Aqueous two phase system
more and more researchers to exploit it [3]. Mainly biocatalysts
are divided into two types: whole cells and isolated enzymes. Aqueous two phase partitioning system (ATPS), a liquid–liquid
Compared to whole cells, isolated enzymes offer several bene- fractionation method has been recognized as a superior, versa-
fits, including simpler reaction apparatus, higher productivity due tile and well-established process for separation [16]. It seems
to higher catalyst concentration and simpler product purifica- to be a promising alternative method in which clarification,
tion owing to elimination of undesirable side reaction during cell partial purification and concentration are integrated into one
growth [4,5]. Also, enzymes possess some excellent properties; step. It is very simple and easy method which is considered
high reaction rates, highly selective and specific working under as an efficient approach for purification and recovery of wide
mild conditions. Additionally, enzymatic based processes reduce range of biomolecules [17]. Generally, ATPS are composed of
adverse impact of manufacturing on our eco-system by reducing aqueous solution of two incompatible polymer/polymer and
the consumption of large amount of raw materials, energy and the polymer/salts components. When the concentration of these com-
subsequent generation of waste (as compared to chemical based ponents increases above a certain critical value, it results into
process) which is beneficial for both industries and environment formation of two phases. The extraction of enzymes with ATPS is
[6,7]. an interesting alternative to traditional separation methods mainly
Industrial enzymes originate from microorganisms which are due to retention of catalytic activity, maintenance of stability due to
fermented primarily by using renewable resources (Fig. 1) [8]. With non-denaturing conditions, high biocompatibility, lower environ-
the advancement in molecular biology and media formulation, the mental burdens, low energy consumption and relative reliability
production yield of enzyme (i.e. enzyme per mass of whole cell) in scale-up [18]. Also, these single step isolation processes allow
has increased significantly. Though there is an increasing demand simultaneous purification of target molecules and removal of con-
for enzyme production, an improvement in downstream processes taminants, therefore bringing about shorter processing time.
has been neglected. The recovery and purification of enzyme typ- Success of aqueous two-phase systems depends on the ability to
ically involves numerous process steps including; precipitation manipulate system parameters (phase composition) so as to obtain
with ammonium sulphate and/or organic solvents, filtration or cen- appropriate partition coefficients (K, the ratio of biomolecule con-
trifugation, dialysis, often followed by several chromatographic centration in top phase to that of bottom phase) and selectivity for
separation techniques such as ion exchange chromatography, size proper separation of target protein. However, the partition mech-
exclusion chromatography, hydrophobic interaction chromatogra- anisms are still not completely understood. Indeed, the extent of
phy or affinity chromatography [9,10]. At the end of the purification partitioning depends not only on biomolecule properties but also
process steps, the enzymes must be concentrated by lyophilization on system properties. The biomolecule properties include many
or ultra-filtration. Although these multistep purification processes variables such as: hydrophobicity, molecular conformation, molec-
seem to be effective, they suffer from many disadvantages such ular mass, concentration of the protein, flexible chain polymers,
as high cost, time consuming and difficulty in scale up. Also, dur- protein net electrical charge and isoelectric point (pI) [19–21]. Also,
ing such a long procedure, energy expenditure in these processes it involves the different chemical and physical interactions such as
is high especially for viscous fermentation broth. Apart from that, Van der Waals and ionic interactions of the biomolecules with the
solvent based extraction can cause considerable denaturation of surrounding phase along with hydrogen bonding and hydropho-
enzyme molecules [11]. Hence, these tedious procedures for the bic interaction too play an important role in the partitioning. In
recovery of biological products have a major impact on the final addition to it, system properties depend on molecular weight and
product cost as separation and purification accounts for a major concentration of phase forming polymer, type of phase forming salt,
fraction (70–80%) of the total bioprocess cost [12]. So, there is a concentration of salt and phase volume ratio [22–24].
demand for the development of cost-effective, mild downstream, Generally, the development of useful strategies for purifica-
more integrated separation and purification techniques with the tion of enzyme by ATPS is divided into four main steps (Fig. 2).
minimum number of steps [13]. The first step is physicochemical characterisation of the feed-
The retention of catalytic activity for enzymes is important for stock (properties of enzyme as well as contaminants), followed
the success of the extraction processes. In this context, significant by second and third steps where the type of ATPS (polymer/salt,
efforts have been focused on the development of new or adapted polymer/polymer or advanced ATPS) and system parameters (poly-
technologies for the purification of enzymes, with lower costs, as mer, tie-line length, volume ratio and pH) are selected, respectively.
S.S. Nadar et al. / International Journal of Biological Macromolecules 101 (2017) 931–957 933

Fig. 1. Simplified representation of the fractionation of enzymes from fermentation by aqueous two-phase processes and three phase partitioning.

Fig. 2. Practical strategy to ATPS process development and scale-up for the downstream processing of enzymes.

The final step is to evaluate the influence of process parameters interactions such as van der Waals, hydrophobic, hydrogen bond
(such as sample loading, concentration, addition of neutral salts, and ionic interactions. The appropriate partitioning of enzymes
system geometry and recycling of chemicals) on the target product (partitioning efficiency) is generally estimated in terms of purifica-
recovery and purity [19,25]. However, the different combinations tion factor (PF), partition coefficient of enzyme (Ke ), yield, specific
of phase forming component have been applied for partitioning activity and activity recovery.
and recovering various enzyme. Once the product of interest and
major contaminant are characterized, the bottom and top phase 2.1.1. Nature and concentration of polymer
forming components of ATPS must be selected. There are different Polyethylene glycol (PEG) is one of the most employed compo-
combinations of ATPS available (such as polymer/polymer ATPS, nent in ATPS as it is produced worldwide in large quantities and
polymer/salt ATPS and ATPS forming alternative components) in wide range of molecular weights. Molecular weight of PEG has
which have been reported and factors involved in the separation a strong influence on the partitioning behaviour of biomolecules
of enzymes are discussed in this review. [32]. Hence, it is necessary to screen a number of different molecu-
lar weights of PEG for partitioning biomolecules in one phase. PEG
2.1. Factors affecting partitioning of enzymes in aqueous two with 400–8000 Da is widely used to partition enzyme molecules
phase partitioning from crude fermentation broth or from natural sources. The parti-
tioning behaviour depends on the relative hydrophobicity of the
Although the mechanism of biomolecules partition in ATPS is polymeric compound (i.e. the number of carbons present in an
very complex, it is believed that the properties of polymer (polymer aliphatic chain, surface hydrophobicity) [33]. As the molecular
type, concentration, molecular weight) and the other components weight of PEG increases, hydrophobic characteristic of the parti-
(salt, polymer) constituting the phase are probably the most vital tioned enzyme and its partitioning coefficient linearly increase [34].
factors affecting the separation of biomolecules in different phases The PEG with low molecular weight (below 1000 Da) is highly sol-
[26–30]. Lee et al. conducted studies by varying a broad array of the uble due to shorter polymer chains with hydrophilic end group
factors involved in ATPS. The influence of phase forming polymers [35]. Hence, higher concentration of polymer is required in order
should be considered firstly, including concentration of polymer to form two phases while better partitioning can be achieved due
and its molecular weight [31]. These parameters directly influence to low interfacial tension [21,36]. It has also been reported that
protein partition by changing the number of chemical and physical higher molecular weight of PEG results in lower partition coeffi-
934 S.S. Nadar et al. / International Journal of Biological Macromolecules 101 (2017) 931–957

cient factor on accounting of increased chain length. This further recovery of SOD was reportes as 78.9%. During partitioning of
increase the excluded volume, i.e. reduced available free volume SOD, it was found that PEG below 1000 Da is not suitable due
for the protein in the PEG rich phase which leads to partitioning of to its high miscibility, and hence, it required higher concentra-
the biomolecules to the bottom phase [37]. Therefore, for efficient tions for inducing a two-phase system. Also, lower molecular
partitioning of enzyme, appropriate selection of polymer is needed weight PEG may draw all proteins to the top phase that will
on the basis of its nature and properties along with favourable cause poor separation with low purification of the target pro-
interactions with enzyme. tein. On the other hand, PEG above 8000 strongly increased
From these reports and as summarized in Tables 1 and 2, it is the viscosity which resulted into denaturation effect thereby
seen that wide range of PEG has been employed successfully for effi- not suitable for ATPS [47]. Kirincic and Klofutar reported that
cient separation of a wide variety of enzymes. In one of the studies, the increase in PEG (400–20000) molecular weight increases the
the researchers performed extractive purification of recombinant viscosity (0.959 × 103 –1.601 × 103 kg m−1 s−1 ) of an aqueous solu-
peroxidase isozyme c from insect larvae. They observed decrease tion of PEG, ranging among 0.959 × 103 kg m−1 s−1 (PEG 400),
in partition coefficient from 128 to 0.5 with increase in PEG molec- 1.119 × 103 kg m−1 s−1 (PEG 4000) and 1.601 × 103 kg m−1 s−1 (PEG
ular weight. The shift of peroxidase from the top to the bottom 20000) [48]. However, the polymer molecular weights influence
phase was observed with the increase in PEG molecular weight partitioning of enzyme by changing the polymer–enzyme interac-
[38]. Another study on the downstreaming of lipase from Rhizopus tion and by altering the phase diagram. Thus, the choice of the most
microsporus was done by ATPS containing PEG 2000 and ammo- suitable intermediate molecular weight of PEG is very important for
nium sulphate. The lipase was partitioned into top phase with yield increasing the extraction efficiency of the ATPS.
and purification factor of 92.3% and 19.8-fold respectively [39]. Besides the molecular weight, choice of PEG concentration has
From above examples, it can be seen that selection of appropri- the most significant effect and provide favourable partitioning of
ate molecular weight of PEG help to propel the enzymes either top enzyme in ATPS. Generally, PEG concentrations are varied in the
or bottom phase in ATPS. In the study by Kianmehr et al. inves- range of 7–21% w/w to study enzyme partition. In the study carried
tigated the influence of PEG molecular weight (1000–8000) on out by Ratanapongleka, the researchers extracted laccase (from L.
partitioning of recombinant P. fluorescens d-galactose dehydroge- polychrous) with significantly high purification factor and yield in
nase (GalDH) in a system composed of PEG/ammonium sulphate. top phase by ATPS, composed of low concentration of PEG 4000
The PEG 4000 was found to be favourable for GalDH partitioning and phosphate salt [49]. Similarly, serine protease was extracted
and showed high affinity toward the top polymer phase. Further from Streblus asper leaves, the partition coefficient decreased sig-
increase in molecular weight of PEG (from 4000 to 8000) resulted nificantly at a low concentration of PEG 4000. This might be due
in less available space in the upper phase, which ledin the decrease to low PEG concentration, hydrophobic interaction between PEG
of partition coefficient (from 45 to 14) due to exclusion effect and the surface of protein enhances and it leads to efficient sep-
[40]. In one of the study, simultaneous separation and purification aration of enzyme molecules [50]. On the other hand, the high
of ␤-galactosidase and ␤-glucosidase from barley (Hordeum vul- PEG concentration shows negative effect on the partition coeffi-
gare) was carried out by employing a wide range of PEG molecular cient of the enzyme. In the extraction of a yeast invertase with
weights (400, 1000, 1500, 4000, 6000 and 8000 Da). For low molec- PEG 3000 and magnesium sulphate system, partitioning of inver-
ular weight PEG (below 1500 Da), both the enzymes partitioned to tase was significantly affected by the high PEG concentration. The
top phase whereas for high molecular weight PEG (above 6000 Da), PEG concentration (10%) resulted in the highest purification factors
both the enzymes partitioned to bottom phase. PEG 1500 gave bet- (3.2-fold) with the yield of 134%, whereas at higher concentration of
ter differential partitioning of ␤-galactosidase and ␤-glucosidase PEG (from 10 to 20%), purification factors drastically decreased with
into opposite phases in terms of activity recovery (60 and 65%) and retention of lower enzyme activity [42]. The reason for this reduc-
activity partition coefficient (2.45 and 0.47) which indicates selec- tion in enzyme activity recovery was attributed to denaturation
tive partitioning of ␤-glucosidase and ␤-galactosidase between two due to break-down of enzyme conformation with the hydropho-
phase [41]. So the manipulation in PEG molecular weight can selec- bic interaction between PEG and enzyme [39]. Additionally, high
tively portioned enzyme from crude fermentation broth. PEG concentration in the system results in the increase of viscos-
The source of enzyme also play an important role in selecting ity and interfacial tension which resulted in more difficulties in
suitable PEG molecular weight which revealed by following studies. partition of the enzyme molecules [26]. However, in some cases,
Yuzugullu et al. partitioned invertase from potato tuber (Solanum higher concentration of PEG found to be favourable which showed
tuberosum) with PEG 4000 in PEG/ammonium sulphate ATPS to improved yield and partitioning of enzyme. In the recovery of lipase
get increased the purity of 5.11-fold with the recovered activity from Penicillium cyclopium, the highest yield was obtained at higher
of 197%, whereas Yücekan et al. partitioned the same enzyme from concentration of PEG 4000 (20%) as compared to lower concentra-
tomato (Lycopersicon esculentum) with PEG 3000 in PEG/sodium tion of PEG (with same molecular weight) in the top phase. Authors
sulphate system to obtain 5.5-fold purity with the recovered activ- have claimed that this was probably due to higher concentration of
ity of 90%. In both cases, invertase was partitioned into the bottom PEG which led to favourable hydrophobic interactions with lipase
salt rich phase [30,42]. From above studies, it is clear that the prop- [51]. Similarly, relatively smaller molecular weight enzyme such as
erties of enzyme (i.e. molecular weight, conformation of enzyme) SOD was obtained at the top phase with increasing concentration
play a vital role while extraction. Similarly, lipase produced by of PEG due to the hydrophobic interactions [52]. Hence, selection of
Bacillus sp. and R. porus was purified by PEG 8000/potassium phos- appropriate PEG concentration is needed for good separation and
phate and PEG 2000/ammonium sulphate system at bottom phase purification which is dependent on the type of ATPS and surface
with best purification factor and yield [43,44]. This shows that enzyme properties.
the distribution of targeted molecules is dependent on biochem- In most of the cases, the effect of polymer concentration on
ical and structural properties of proteins and molecular weight of enzyme partitioning is straightforward. For effective partitioning
PEG. However, there is no clear correlation between partition coef- of any enzymes, both the parameters (i.e. PEG concentration and
ficients and molecular weight of enzyme for different molecular its molecular weight) need to be selected appropriately. However,
weight of PEG [45,46]. the molecular weights of polymer which influences the partition by
The partitioning and purification of superoxide dismutase (SOD) altering the phase diagram and by changing the polymer–protein
(small molecular weight enzyme) from P. chrysosporium was done interaction; thereby, section of both concentration and molecular
by using PEG 3350/potassium phosphate at pH 7.0.The total weight of polymer becomes complicated. Moreover, researchers
S.S. Nadar et al. / International Journal of Biological Macromolecules 101 (2017) 931–957 935

Table 1
Extraction of enzymes by using polymer/salt system.

Enzyme Source Polymer Salt Kp/PF Yield Ref.

Luciferase Photinuspyralis PEG 1500 (14%) Ammonium sulphate (16%) 13.69-fold – [60]
D-Galactose Pseudomonas fluorescens PEG 4000 (15%) Ammonium sulphate (11.8%) 58.9% 92.8% [40]
dehydrogenase
Invertase Baker’s yeast PEG 3350 (10%) Magnesium sulphate (15%) 8.81-fold 77% [61]
Hexokinase Baker’s yeast PEG 1500 (13.5%) Sodium Citrate (17.5%) 1.87-fold – [68]
Lipase Penicillium cyclopium PEG 4000 (15%) Potassium phosphate (70%) 9-fold 95.7% [51]
Endo-mannanase Bacillus sp. CFR1601 PEG 3350 (12.0%) Sodium sulphate (12.0%) – 95.4% [57]
Lipase Rhizopusniveus PEG 4000 (32.5%) Sodium carbonate (18.4%) 19.3-fold 92.8% [44]
Lipase Rhizopusmicrosporus PEG 2000 (20%) Ammonium sulphate (12%) – 92.3% [39]
Invertase Lycopersiconesculentum PEG 3000 (15%) Ammonium sulphate (12%) 5.5-fold 88% [30]
Glutaminase Zygosaccharomyces rouxii PEG 2000 (14%) Sodium sulphate (10%) 1.95-fold 69.1% [64]
Acid Invertase Solanum Tuberosum PEG 4000 (12.5%) Sodium sulphate 5.11-fold – [42]
(15%)
Superoxide Phanerochaetechrysosporium PEG 3350 (24%) Potassium phosphate (5%) 0.17 – [47]
dismutase
Laccase Lentinuspolychrous PEG 4000 phosphate salt 88.3 99.1% [253]
(12%) (16%)
Lipase Bacillus sp. PEG 8000 (20%) Potassium phosphate (18%) 123.89-fold – [43]
Alcohol Baker’s yeast PEG 2000 (15%) Potassium phosphate (10%) 6.6-fold – [254]
dehydrogenase
Polyphenol oxidase Ananas comosus Merr. PEG 1500 (18%) Phosphate (14%) 2.7-fold – [66]
Amylase Bacillus subtilis PEG 1000 (16.7%) Potassium phosphate (14.8%) 5.4-fold – [80]
Phytase Schizophyllum commune PEG 1500 (22%) Sodium citrate (14%) 2.63 367% [255]
␤-Xylosidase Trichoderma koningii G-39 PEG 6200 (10%) Sodium phosphate (5%) 8.8-fold 100% [256]
Invertase Baker’s yeast PEG 3000 (15%) Magnesium sulphate (23%) 6.2-fold – [37]
Cyclodextrin Bacillus cereus PEG 8000 (19.0%) Sodium Citrate (11.5%) 16.3-fold 70% [257]
glycosyltransferase
Lipoxygenase Glycine Max PEG 6000 Ammonium sulphate 4.38-fold – [258]
Serine Protease Streblus asper PEG 6000 (16%) Magnesium sulphate (15%) 14.4-fold 96.7% [50]
Laccase Hericium erinaceus (Bull.:Fr) PEG 8000 (17%) Potassium phosphate (12.2%) 8.03 99% [259]
Pers.
proteases C. perfringens. PEG 10000 (22%) Sodium Citrate (8%) 4.2-fold 131% [28]
␤-Galactosidase Aspergilus niger PEG 2000 (15.24%) Ammonium sulphate (17.14%) 19.42-fold – [260]
Polyphenol Potato Peel PEG 1500 (17.62%) potassium phosphate buffer (15.11%) 3.7 77.8% [261]
Oxidase
Polyphenol oxidase Solanum tuberosum PEG 8000 (5%) Phosphate (28.5%) 15.7-fold 97.0% [262]
Proteinase Tuna spleen PEG 1000 (15%) Magnesium sulphate (20%) 6.61-fold 69.0% [27]
␣-Amylase hog pancreas PEG 2000 (11.7%) Fructose-1,6-bisphosphate trisodium 3.2-fold 79% [263]
salt (19%)
␣-Galactosidase Aspergillus oryzae PEG 4000 (12%) Phosphate (11.9%) 3.6-fold 87.7% [264]

Table 2
Extraction of enzymes by using polymer/polymer system.

Enzyme Source Polymer Polymer PF Yield Ref.

Serine protease Mango peel PEG 4000 Dextran 14.37-fold 97.3% [69]
Amyloglucosidase Aspergillus niger PEG 4000 Polyacrylate 15000 2.0-fold 86% [76]
Pectinases – PEG 4000 Crude dextran 1.66-fold – [74]
Alkaline phosphatase Bacillus licheniformis PEG 4000 Dextran T500 5.23-fold – [71]
L-glutaminase Bacillus cereus MTCC 1305 PEG 4000 (8.5%) Dextran T500 1.31-fold – [70]
Lipase Burkholderia pseudomallei PEG 8000 Dextran T500 2.44-fold 92.1% [73]

cannot isolate the effect of polymer molecular weight from the factorial approaches could make this technique a very useful and
effect of polymer concentration [14,53,54]. The design of experi- helpful tool for purification processes [55–57].
ment is very useful methodology in finding the optimum partition
condition. Thereby, these two factors should be considered simul- 2.1.2. Polymer/salt aqueous two phase partitioning system
taneously for modelling of protein partitioning in ATPS. An ATPS Since chemical cost is always a dominant factor in any sepa-
composed of PEG and phosphate ATPS is used for purification of ration and purification process, an inexpensive polymer/salt ATPS
a highly thermostable and alkaline active recombinant xylanase has been widely favoured for commercial application [58]. In
from B. halodurans [29]. Response surface methodology (RSM) PEG/salt ATPS, the choice of a phase-forming salt has directly
and artificial neural network (ANN) have been used to develop facilitated the separation and extraction of the target molecules
predictive models for optimization of purification process param- between the phases (Fig. 3) [39,59]. To ensure the efficiency of
eters (PEG molecular weight and its concentrations) and both the extraction, the partitioning is carried out by employing vari-
models have been able to predict appropriate conditions for extrac- ous phase-forming salts. The salts have ability to manipulate the
tion. The results demonstrating partitioning behaviour with higher hydrophobic interactions between biomolecules in ATPS. The use
prediction accuracy was obtained by ANN with high correlation of different salts, change the environment around the enzymes
coefficient (R2 ) compared to RSM. Also, Desai et al. found that the which lead to change in the partition behaviour [25]. The example
average percentage error for ANN and RSM models were 6.5 and of enzyme extraction by polymer/salt system is shown in Table 1.
20% indicating the superiority of ANN in capturing the nonlinear Priyanka et al. employed various phase forming salts; magnesium
behaviour of the system. This superiority of ANN over other multi sulphate, potassium phosphate, sodium sulphate and ammonium
936 S.S. Nadar et al. / International Journal of Biological Macromolecules 101 (2017) 931–957

Fig. 3. Schematic diagram illustrating the partitioning of enzyme and contaminant proteins (impurities) in a polymer/polymer and polymer/salt ATPS.

sulphate together with PEG 1500 to arrive at the most suitable ATPS with multivalent anions of high-charge density limits such inter-
to purify luciferase from fireflies (Photinus pyralis). It was observed actions with the polymer chain, leading to salt-depleted zones and
that luciferase preferentially partitioning to the top phase with consequently formation of two phases. Hence use of anionic salts
PEG 1500/ammonium sulphate system exhibited highest enzyme for the partitioning of enzymes is more beneficial [64].
activity recovery (97.30%) with 3.61-fold of purification factor [60]. Apart from the salt type, another important factor which
In another study, the PEG/ammonium sulphate was found to influences the partitioning of enzyme molecule in ATPSs is the
be the best in terms of activity recovery and differential partition- concentration of utilized salt [61]. As evident from previous lit-
ing of both the enzymes ␤-galactosidase and ␤-glucosidase from erature, the recovery of enzyme was increased with an increase
barley [41]. Aqueous two-phase extraction of ␤-d-fructofuronoside in concentration of salt to a maximum value and further increase
fructohydrolase (␤-d-FF) was performed by using PEG 3350/salt in concentration drastically affected purification factor as well
(ammonium sulphate, sodium sulphate, sodium phosphate, potas- as activity recovery. In the separation of proteins (cytochrome c,
sium phosphate, magnesium sulphate and trisodium citrate) trypsin, bovine serum albumin and myoglobin) by the ATPS system,
system. Amongst these salts, the highest ␤-d-FF activity of 84.7% Zhao et al. used the different concentration of ammonium sulphate
with 3.78-fold purification was observed in case of magnesium sul- in the range of 10–18%. It was observed that as concentration of
phate [61]. In polymer/salt ATPS, sometimes salts show activity ammonium sulphate increased, the top phase volume decreased
inhibition during the partitioning of enzymes. In the separa- and all proteins retained in the bottom phase [65]. In many cases,
tion of endo-mannanase from Bacillus sp., magnesium sulphate enzyme recovery is not affected significantly while purification
and ammonium sulphate displayed complete inhibition of endo- fold is more sensitive with respect to salt concentrations. Also, the
mannanase as no activity was detected in either top or bottom distribution of enzyme is controlled by concentration of the salt.
phases [57]. The probable reason for these results was the stronger The partition coefficient of acid invertase increased rapidly with
ionic strength and protein aggregation (due to salting out effect) an increase in salt concentration from 13% to 20%. The maximum
leads to protein denaturation [62]. enzyme recovery (128%) with a purification factor of 3.89-fold was
Generally, two phases in ATPS have to be isoosmotic and elec- obtained at 15% of salt concentration combined with PEG 4000 sys-
trically neutral, when salt is added to polymer solution, cations tem [42]. Moreover, PEG/potassium phosphate system is employed
and anions distribute themselves differentially among the phases for separation and purification of enzymes mixture polyphenol
depending on their affinity with aqueous phase [26]. Thereby, oxidase (PPO) and bromelain from pineapple (Ananas comosus L.
an interfacial potential is produced differently which may help Merr.. The results showed about 228% activity recovery and 4.0-fold
in the partition of biomolecules (proteins). The performance of increase in purification factor in case of bromelain whereas, 90%
salts in phase separation is reflected in the lyotropic series (a activity recovery and 2.7-fold increase in PPO purity. The increase
classification of ions based upon salting-out or salting-in ability) in salt concentration resulted in “salting out effect” which increased
[63]. The effectiveness of salts with multi-charged anions is as per partitioning of the both the enzymes to the top phase. However,
SO4 −2 > HPO4 −2 > C2 H3 O2 −1 > Cl−1 , whereas, cations are commonly purity of bromelain in the top phase was low whereas purity of
ranked in the following order: NH4 + > K+ > Na+ > Mg2+ > Ca2+ . In gen- polyphenol oxidase increased at higher potassium phosphate con-
eral, proteins with the negative charge tend to partition to the top centration due to precipitation of enzyme at the interface [66].
phase in PEG/salt systems while those with the positive charge usu- The polymer/phosphate salts and polymer/sulphate salt sys-
ally go to the bottom phase [37]. The feasibility of utilizing ATPS tems have been most commonly used for separation of enzymes
composed of PEG 2000 and ammonium sulphate for the purifi- [22]. However, use of these salts in large scale led to a strong
cation of lipase from Rhizopus microsporus fermentation culture negative impact on the environment. In order to reduce environ-
was investigated by Anvari. The lipase partitioning was much more mental problems. citrate (biodegradable and nontoxic in nature)
strongly affected by anion species than by cations [39]. In the case and ammonium carbonate (volatile in nature) are favoured because
of cationic salts, the interaction of cations with water molecules is of their high selectivity, less pollution and biocompatibility [67].
readily replaced by PEG-cation interactions. On the other hand, salts In one of the studies, Oliveira et al. employed citrate in PEG/salt
S.S. Nadar et al. / International Journal of Biological Macromolecules 101 (2017) 931–957 937

ATPS to extract hexokinase from baker’s yeast (Saccharomyces cere- from Aspergillus niger using ATPS, PEG 4000 and sodium poly-
visiae), to obtain high enzymatic recovery in the top phase with acrylate 15,000. The purification factor and the recovery yield of
high partition coefficient [68]. The choice of a phase-forming salt amyloglucosidase were determined as 1.9-fold and 86% respec-
and its concentration have a direct influence on the separation and tively. The poly(acrylic acid) is harmless, relatively inexpensive,
extraction of enzymes in ATPS due to its major effect on the system easily handled and strong negatively charged polymer (pH above
environment. 7.0) which shows strong electrostatic interaction with charged
biomolecules. In addition to that, the phase separation in this
2.1.3. Polymer/polymer aqueous two phase partitioning system system is relatively fast and has low viscosity as compared to tra-
The another type of ATPS is polymer/polymer system in which ditional PEG/dextran systems [76]. Another study involved use of
polymers were employed as another component which not only thermoseparating polymer as a one of the polymer component in
provides good stability to proteins but also making them compati- polymer/polymer system. Lysozyme and trypsin partitioned into a
ble due to high water content (>90%). Two mutually incompatible two-phase formed by maltodextrin (a low-cost starch derivative)
different polymers beyond a critical concentration have ten- and a copolymer of ethylene oxides and propylene oxides (ther-
dency to form two-phase systems in water solutions (Fig. 3) [16]. moreactive). This system was more cost efficient as polymers were
PEG–dextran system is practised widely in the partitioning of readily recycled by temperature-induced phase partitioning with-
enzymes [69]. Selected examples of the use of polymer/polymer out costly ultrafiltration or chromatography steps [77]. In addition,
ATPSs for recovery and purification of enzymes are presented in the polymer/polymer system can be used in affinity partition by
Table 2. Singh et al. carried out partitioning studies of L-glutaminase modifying one of the polymer in the two-phase system for selective
produced from Bacillus cereus MTCC 1305 in different PEG–salt and and enhance separation which is explained in later part of review.
PEG–dextran ATPS. The PEG–dextran system was found to be most
suitable for partitioning of L-glutaminase as compared to PEG–salt 2.1.4. Effect of pH on partitioning of enzymes
system. The maximum partition coefficient (1.31) with 2.09 U activ- Principally, the partitioning of enzymes into two phases in the
ity was obtained for PEG–dextran system. Authors have claimed ATPS depends on their isoelectric points (pI, the pH at which net
that PEG-salt based ATPS causes denaturation of the biological charge on enzyme is neutral). The addition of salts and the change
structureand dissociation (protein unfolding) of the ligand-protein of pH or ionic strength have often been used to alter the partitioning
complex [70]. To understand the effect of different molecular of the enzyme and improve the selectivity of the two-phase extrac-
weight of dextran on enzyme extraction, Pandey et al. performed tion. According to Albertsson’s equation, the partition coefficient of
partitioning of alkaline phosphatase from B. licheniformis by using charged biomolecules is influenced by short-range (van der Waals)
ATPS which composed of PEG and two different dextran (dex- and long-range (electrostatic) molecular interactions [16]. The pH
tran T40 and dextran T500). PEG 4000/dextran T 500 system was of the system affects the net charge of protein and ion composi-
selected as most appropriate ATPS for alkaline phosphatase pro- tion, surface character of contaminating materials which causes
duction based on greater partition coefficient [71]. Furthermore, variation in their partitioning into the top and bottom phases [37].
dextran demonstrates stabilizing effect on enzymes (due to enzyme The influence of pH on partitioning of recombinant Bacillus
and polysaccharide interaction), which apparently maintains the sphaericus phenylalanine dehydrogenase (PheDH) was observed in
enzyme activity in the ATPS system. The dextran T500 is com- PEG/ammonium sulphate ATPS. The pH of the system varied from
monly employed for partitioning of enzymes in polymer/polymer 3 to 8 and the maximum activity recovery (120%) and yield (90%)
system which has less influence on the partition coefficient [72]. was obtained at pH 7.8. With increasing pH, net negative charge of
So, concentration of dextran is considered to attain higher purifi- the protein increased simultaneously. So, the interaction between
cation factor. This is investigated in the study carried out by Ooi protein and PEG-rich phase became stronger and partition coef-
et al. They performed extractive fermentation using ATPS which ficient elevated [78]. As a general rule in polymer/salt systems,
incorporated PEG 8000 and dextran T 500 (5–20%) to extract most proteins with acidic pI and consequently negative surface
lipase derived from Burkholderia pseudomallei. At lower concen- charges prefer to move to the top phase, while positively charged
tration of dextran, higher yield of 92% with highly active lipase proteins move to the bottom phase. Most of the biomolecules,
was obtained [73]. However, the ATPS based on PEG and dex- especially proteins and enzymes, are stable at neutral pH which is
tran is very expensive because of the high cost of phase forming favourable condition to conduct the ATPS partitioning. The pH value
polymers. So, expensive dextran fraction with narrow molecular above 7 is suitable for the PEG/phosphate system and pH below 6.5
weight distribution can be replaced by crude dextran. The use of is compatible with the PEG/sulphate system. Partition behaviour
crude dextran with PEG 4000 in ATPS has been evaluated in parti- of glutaminase (pI is around 5) from Zygosaccharomyces rouxii at
tioning of pectinases. Almost complete one-sided partition of endo different pH (3–9) was studied in an ATPS of PEG 2000/sodium
and exo-pectinase was achieved with highest total endo- and exo- sulphate. It was found that as pH increased towards 7, partition
pectinase yields (72.41% and 69.46%, activity recovery respectively) coefficient decreased upto 9.44 with the corresponding yield of
with purification fold of 1.73 and 2.14, respectively. However, crude 90.42%. Further increment in pH led to decrease in yield upto
dextran showed rather high viscosity in solution which results into 40% [79].
problem for phase separation [74]. This, in fact, limits the imple- Bezerra et al. investigated the effect of pH on the purification fac-
mentation of dextran based system at the large scale for separation tor of alpha amylase fermented from Bacillus subtilis. They found
of biomolecules [15]. that purification factor was increased from 0.3 to 1.8-fold in the
The above problem may be solved by using an economic alterna- PEG 1000/potassium phosphate ATPS, when the pH rose from 5.0
tive polymer that substitutes for dextran with equivalent partition to 8.0 [80]. In another study, extraction of lipase from Rhizopus
properties. So, there has been an interest in developing new poly- microsporus fermentation culture was carried out by Anvari. An
mer systems which can enhance partitioning of enzymes between increase in pH of the ATPS (PEG 4000/K2 HPO4 ) from 7.0 to 9.0
the two phases based on the electrostatic interactions [24]. Pereira reduced the partition coefficient of lipase from 7.94 to 4.45 and
et al. used polyacrylate as a polymer for partitioning and recov- activity recovered from 81.1 to 70.6% [39]. Enzymes are slightly
ery of lipase was derived from Leucosporidium scottii L117 which instable in the acidic area, but it was dramatically lost their activ-
showed 40.5 activity balance (ratio of total enzyme activity in top ity at higher pH (above 9.0). The effect of pH (in the range of
and bottom phase to enzyme activity in crude sample) [75]. In 4–11) on partition behaviour of a plant peroxidase extracted from
another study, Lizzy et al. extracted and purified amyloglucosidase the leaves of Ipomoea palmetta was studied in ATPS composed of
938 S.S. Nadar et al. / International Journal of Biological Macromolecules 101 (2017) 931–957

PEG 1550/ammonium sulphate. The effect of pH was found to be and considered in conventional ATPS methods. Also, they are usu-
different on partitioning behaviour of enzyme in different phase ally require long processing time and also expensive. Therefore,
systems. At extreme pH conditions (below pH 4 and above 11), in such a case, the addition of neutral salt can improve the par-
enzymes deactivate due to conformational changes. Their partition tition of target biomolecule which significantly increase process
coefficients in case of sodium sulphate, ammonium sulphate and yield as well as selectivity [63]. It is known that the presence of
potassium phosphate phase systems were 8.5, 7.0 and 10.3, respec- ions has influence on the partition behaviour of proteins through
tively with the corresponding yield of 90, 85 and 73% [81]. Hence, the “salting out” effect. The effect of adding salts to an ATPS is
the partition of enzymes can be manipulated either by changing salt dependent on the type of system and salt that is used [53]. Gen-
component of the system or by altering overall pH of the system. erally, the addition of neutral salts affects the electrical potential
difference between top and bottom phases and protein hydropho-
2.1.5. Effect of volume ratio and tie-lines length on partitioning of bicity. An increase in the hydrophobicity will decrease the amount
enzymes of bound water, which keeps the final composition of the system
For an efficient separation in ATPS, it is essential to find high constant. This may result in the exposure of hydrophobic patches of
recovery, purification of enzyme and favourable partition coeffi- protein surface, which will promote hydrophobic interactions with
cient which is influenced by phase volume ratio (ratio of volume the polymer phase [29,87]. In general, water structure making ions
of top phase to that of bottom phase). All two-phase systems with (Li+ , Na+ , Ca2+ , Mg2+ , NH4+ , F− , SO4 2− , CO3 2− , PO4 2− , CH3 COO− )
overall composition represented by one of the tie-lines give phases favour more hydrophilic phase, while, water structure breaking
with same composition but different volumes [15]. Tie line length ions (K+ , Rb+ , Cs+ , I− , Cl− , Br− , SCN− , NO3− , ClO4− ) favour more
(TLL) can affect biomolecule partitioning by interfacial tension and hydrophobic phase. Moreover, neutral salt addition affects enzyme
hydrophobicity between two phases of ATPS. The ATPS becomes partitioning by altering the partition coefficient of proteins in rela-
more hydrophobic with increasing TLL due to reduction of water tion to their charge [38]. Furthermore, it increases the ionic strength
availability [82]. An increase in the TLL causes an increase in the and enhances the migration of low molecular weight compounds
protein partition coefficient that, in turn, increases the yield of pro- towards the polymer phase, especially in PEG <4000 [15,88]. How-
teins in the top phase due to reduction of the bottom phase [62]. ever, the addition of high concentration of neutral salts may cause
Increasing TLL in the PEG/salt system makes salting-out more effec- denaturation of proteins in the existing system, thus lower con-
tive, leading to shift of proteins to the top PEG-rich phase [83]. If centration range from 0.1 to 1.0 M or 1 to 8% w/v is preferred to
protein solubility in the PEG phase is insufficient, protein will pre- enhance the yield of the enzyme in ATPS [14].
cipitate at the interface. Solubility and salting-out depend on the The different neutral salts give different purification, activity
properties of individual proteins. Therefore, a different response recovery and yields. In order to determine the effect of neutral
will be expected when a mixture of protein is handled [84]. salts on partitioning of invertase, various salts such as; KCl, NaCl,
In the study involving purification of luciferase using MnCl2 , Na2 CO3 , MgCl2 , MgSO4 in PEG/sodium sulphate ATPS were
PEG/ammonium sulphate ATPS, the phase volume ratio was varied evaluated. The researchers obtained maximum purification of 5.5-
in the range of 0.12–2.50 with tie line of 16% (w/w). It was found that fold and 154% activity recovery for KCl. A selective increase in
the specific activity and degree of purification of luciferase in the the partition coefficient for invertase is caused mostly by surface
top phase increased with the decrease in volume ratio [60]. On the hydrophobicity differences between the proteins [30]. Another key
other hand, in the recovery of lipase from Penicillium cyclopium by factor affecting the enzyme partition in two phase systems is the
ATPS, the phase volume ratio increased by approximately 5.8 times concentration of neutral salt used. In the purification of lipases pro-
to augment enzyme yield by 24% in the top phase while concen- duced by Bacillus sp. ITP-001 using PEG and potassium phosphate,
tration of phosphate was reduced by 4.4%. However, purification partition coefficient of enzyme was almost constant with NaCl addi-
factor of enzyme was reduced from 3.5 to 2.3-fold in the top phase. tion till 6%, although the purification factor increased significantly
The increase in yield of enzyme by varying phase volume ratio from 59.93 to 141.65-fold. Further addition of NaCl decreased the
along the lower tie-line, results in reduction of salt concentration in partition coefficient of lipase [43]. Similar results were observed
ATPS which would be beneficial from the both environmental and with Aspergillus terreus lipase purification in the presence of NaCl
economic reasons [51]. ATPS has also been used to further concen- in ATPS system of PEG and potassium phosphate. The purification
trate and recover fibrinolytic crude extract by using six different factor with NaCl was 12.5% higher than that of ATPS system without
volume ratios above and below the equilibrium composition and NaCl [89].
42.6% of TLL. The highest purification factor was 7.91-fold at 1.5
volume ratio. Above and below this volume ratio, purification was
not significant. In defined ATPS, the composition of the top phase 3. Alcohol/salt based aqueous two phase partitioning
proportionally increases with the volume ratio along the same TLL. system
Hence, target enzyme had been partitioned into top phase (polymer
rich phase) due to existence of more free volumes [85]. In contrast In both polymer/salt and polymer/polymer systems, the target
to that, when the volume ratio decreases over the limit of solu- biomolecules are accumulated in either the polymer-rich phase
bility of fibrinolytic enzyme in the PEG-rich phase, the fibrinolytic or salt-rich phase. The recovery of biomolecules from phase-
enzyme precipitates and gets accumulated at the interphase. There- forming chemicals/polymers is very convoluted [90]. Also, it has
fore, it leads to loss of fibrinolytic enzyme by precipitation. It could been widely reported that it involves tedious purification oper-
be minimised by higher volume ratios of respective ATPS. However, ations, such as ultrafiltration and crystallization often needed to
higher volume ratio also has a drawback as it leads to the dilution get desired proteins from these conventional ATPS [91]. Mostly,
of enzyme concentrations [86]. these systems require back-extraction of target biomolecule into
salt-rich bottom phase, or additional complicated operations (such
2.1.6. Effect of neutral salts on enzyme partitioning as ion exchange chromatography, diafiltration, crystallization or
The desired biomolecule has to be preferentially partitioned ultrafiltration) to remove the phase-forming components from tar-
in one phase, whereas the interfering substances (other proteins, get product [92]. Also,in many cases, protein (enzyme) which has
polysaccharides and contaminants etc.) should be partitioned in low solubility in aqueous solutions tends to aggregate in poly-
the other phase to achieve a significant extraction. However, the mer/salt system which ultimately led to loss of enzyme [93].
partition of target enzyme is not as one sided as might be expected Furthermore, recycling of bottom phase forming salt is very compli-
S.S. Nadar et al. / International Journal of Biological Macromolecules 101 (2017) 931–957 939

cated. This require large amounts of polymers/chemicals, leading to and the bottom alcohol phase which can be recycled [15]. Non-
environmental pollution and high operating costs [94]. Therefore, ionic surfactants have been employed for solubilizing membrane-
advanced and improved ATPS have been developed using alco- associated proteins without loss of their biological activity. During
hols, surfactants, micellar compounds [95]. Also, ionic liquid, and solubilisation, the non-ionic surfactant swaps most lipid molecules
thermo-separating polymers (TSPs) have been employed to extract in contact with the hydrophobic domain of the protein and leads to
enzymes from fermentation broth and natural sources [96,97]. the formation of a soluble protein-surfactant mixed micelle [106].
The alcohol/salt ATPS is formed by mixing two immiscible com- The physicochemical environments micelle-rich and in the micelle-
ponents which composed of alcohol and salt solutions. In this poor phases form the basis for an effective separation and makes
system, the biomolecules were partitioned by the hydrophobic surfactant and alcohol based system, a convenient and potentially
interaction of organic solvent combined with the salting-out effect useful method for the separation and purification of biomolecules
of the salt-rich phase [98]. The salt-rich bottom phase also influ- like enzymes and proteins [107].
ences the hydrophobic interactions between proteins as salt ions It is important to make a note that the effect of surfactants
interact with the oppositely charged residues of the protein, result- on partition of enzyme depends on specific and selective chemi-
ing in the dehydration. Thus, the hydrophobic interaction between cal interactions between the enzyme molecules. It is manifested in
the enzyme and organic solvent led to the desirable partition and the work carried out by Zhong et al. The researchers used different
transfer of the enzyme to the top phase of the system. Addition- surfactant (Triton X-100, Tween 80, SDS)/mannitol to purify poly-
ally, the target protein can be easily extracted and recovered by galacturonase enzyme from Durio zibethinus. Tween 80 exhibited
removing the alcohol using evaporation [99]. However, an increase effective enzymatic partitioning with 11.2-fold purification factor
inalcohol concentration in two phase system results in the par- and yield of 93.2%. While, an anionic surfactant SDS showed the
tial dehydration of the bottom phase [100]. Alcohol-salt ATPS have minimal effect on the enzyme separation. It has been suspected
also been used for the recovery of low and high molecular weight that ionic surfactant attached to protein might interrupt the ter-
biomolecules [101]. Additionally, the alcohol/salt ATPS has less tox- tiary conformational which affect the enzyme activity [108,109].
icity impact to the environment and is economic as compared to the It has been verified that the interaction between ionic surfactants
conventional ATPS [14]. Also, it presents some additional advan- and proteins is a combination of electrostatic as well as hydropho-
tagessuch as high polarity, low viscosity, low cost, and ease of bic forces [110]. Thus, the surfactant head (hydrophilic) group plays
recovery from the alcohol-rich phase [102]. a determining role in protein-surfactant interactions that preferen-
Various examples of extraction of enzyme by using alcohol/salt tially begins with strong ionic bonds formation between surfactant
ATPS system have been summarized in Table 3. Pectinase from polar groups. The protein-surfactant mechanism would greatly
mango (Mangifera indica) waste was purified using an aqueous inhibit the enzyme separation in two phase system and directly
organic phase system of ethanol and potassium phosphate. The reduce the purification factor [111,112].
salting-out effect resulted in the molecular exclusion of pectinase In surfactant and alcohol (Tween 80/xylitol) based ATPS sys-
to the upper phase and contaminants moving from organic solvent tem, different concentrations of surfactant were employed to purify
top phase to salt-rich bottom phase. Based on the system, pecti- lipase from Pumpkin (Cucurbita moschata) Seeds. The lipase activ-
nase was extracted with the purification factor of 11.7-fold, with ity was augmented by increasing Tween 80 concentrations upto
97.1% yield [62]. The selection of alcohol and salts play an important 10%, while the activity decreased beyond this concentration due to
role in the partitioning of enzyme. Ooi et al. employed differ- partially inactivation of enzyme. The inactivation of enzyme was
ent alcohol-based top phase (ethanol, 1-propanol and 2-propanol) attributed to the binding of Tween 80 surfactant to the proteins,
and a salt-based bottom phase (ammonium sulphate, potassium which disrupted the majority of the globular structures resulting
phosphate and sodium citrate) to evaluate for their effectiveness into reduction in purification factor [113,114]. The high recovery
in lipase recovery from Burkholderia pseudomallei. The maximum of enzymes along with recycling of phase forming components is
lipase recovery was achieved in 2-propanol/potassium phosphate advantage of surfactant/alcohol based system. The purification of
ATPSwith purification factor of 13.5-fold and a yield of 99%. The polygalacturonase from Durio zibethinus murry was successfully
authors have claimed that the negatively charged lipase is repelled done by using Triton X-100 and mannitol ATPS to get 16.1-fold
from salt-rich bottom phase and longer hydrocarbon chain alco- purification factor and 97.3% yield. Further, surfactant and alcohol
hol facilitated the interaction with lipase, and thus enhanced the was successively recycled for 5 times with minor loss of phase form-
partitioning [103]. ing component. This could be explained by the selective interaction
The adverse effect of alcohol based systems during purification of the surfactant toward desirable protein into the top phase after
of enzyme have also been reported. In the recovery of superoxide being reused several times [115]. Therefore, this surfactant/alcohol
dismutase from Kluyveromyces marxianus by ethanol and potas- based system presents a promising alternative to the conventional
sium phosphate ATPS, the SOD activity in each phase of the systems ATPS which includes beneficial properties like inexpensive phase
decreased dramatically upto 50%. This loss in catalytic activity was formation compounds, mildness of the recovery conditions (high
attributed to structural and conformational changes and subse- water content) and simplicity of the procedure. However, these
quent denaturalization of the protein due to the high concentration system cannot be suitable for the enzymes which are sensitive to
of ethanol in the system [104,105]. Therefore, it limits the usage of surfactant as well as alcohol. Vicente et al. observed that during the
ethanol/salt ATPS for the recovery of certain enzymes. recovery of bromelain (cysteine endopeptidases) from pineapple
stem by surfactant based ATPS, enzymatic activity was reduced by
40% due to unfavourable interaction with surfactant Triton X-114
4. Surfactant/alcohol based aqueous two phase partitioning [116].
system

Recently, a novel approach for ATPS based on surfactant and 5. Advanced aqueous two phase partitioning system
alcohol (mostly sugar alcohol) have been developed. It has ability
to retain the biological activity of enzymes over other conventional The growing demand for biotechnological fine chemicals and
ATPS has been reported (Table 4). This system makes it possible to biological molecules, in particular proteins has led to the develop-
create two phases; the top surfactant rich phase (containing non- ment of specific separation and purification methods. In general,
ionic and ionic surfactants such as Triton X-100, Tween 80, SDS) ATPS has proven to be effective for the recovery of biological prod-
940 S.S. Nadar et al. / International Journal of Biological Macromolecules 101 (2017) 931–957

Table 3
Extraction of enzymes by using alcohol/salt-based aqueous two-phase system.

Enzyme Source Alcohol Salt PF Yield Ref.

Lipase Burkholderia pseudomallei 2-Propanol (16%) Potassium phosphate (16%) 13.5-fold 99% [103]
Pectinase Mangifera indica Ethanol (19%) Potassium phosphate (22%) 11.7-fold 97.1% [265]
SuperoxideDismutase (SOD) Kluyveromyces marxianus Ethanol (17%) Potassium phosphate (26%) 9.8-fold 80% [105]
Serine protease Mangifera Indica 2-Propanol (16%) Potassium phosphate (20%) 11.6-fold 96.7% [62]

Table 4
Extraction of enzymes by using surfactant/alcohol based aqueous two-phase system.

Enzyme Source Surfactant Alcohol Kp Yield Ref.

Lipase Leucosporidium scottii L117 Triton X-114 McIlvainebuffer 4.77 88.94% [75]
Polygalacturonase Durio zibethinus Murry Tween- 80 (25%) Mannitol (22%) 16.1 97.3% [266]
Lipase Cucurbita moschata Triton X-100 (24%) Xylitol (20%) 16.4 97.4% [114]

ucts from different origins [19,117]. However, one major drawback modified polymer concentration could also enhance the recovery
concerning the use of ATPS is their lack of specificity. Also, tra- and purification yield of specific enzyme in affinity ATPS. Neverthe-
ditional methods for purifying and separating proteins are highly less, appropriate levels of ligands should be added to the system
complex, time consuming and expensive. Although the manipula- as their higher ligand concentration could introduce shielding
tion of the system parameters may favour the selective partitioning effect towards target biomolecules and also higher concentration
of the desired compound towards a particular phase in traditional of modified polymer can generate steric hindrance between active
ATPS, specificity is not high enough to accomplish complete separa- receptor sites and target molecules [98,117]. These possible alter-
tion between product and contaminants [98]. Hence, the modified natives emphasize the importance of the different parameters in
ATPS was employed as down streaming strategies to enhance the affinity ATPS systems, in order to design efficient and novel purifi-
purification of biomolecules which is briefly summarized as below. cation strategies.
Affinity ligand provides high resolution which can be coupled
5.1. Aqueous two-phase affinity partitioning system with traditional ATPS. The polymer component (PEG or dex-
tran) has to be chemically active to purify enzymes.The several
Affinity aqueous two phase partitioning provides a highly com- example of affinity aqueous two phase system for enzyme extrac-
patible system, based on the specific interaction between the tion is listed in Table 5. These strategies could be developed
biomolecule and affinity ligands which can be manipulated in order using derivatives of PEG as affinity ligands such as PEG-benzoate,
to develop more predictable purification strategies. Therefore, it PEG palmitate, PEG phenylacetamide or PEG trimethylamine
can bepossible to get specific partition coefficients for a defined [122]. In one of the studies, modified PEG and sulphate ATPS
ATPS [104]. Generally, it consists of chemical modification of at was utilized to purify ␤-Galactosidase and glucose oxidase from
least one phase forming component of the ATPS by coupling affinity Kluyveromyces lactis and Aspergillus niger fermentation. In this par-
ligand (Fig. 4 Left). Therefore, selective interaction between spe- ticular case PEGs were modified as: PEG-trimethylamine (TMA
cific ligands in one of the polymeric phases of the system and ionic interaction ligand), PEG-benzoate (pseudo specific ligand),
biomolecule of interest followed by their partition towards the PEG-palmitate (hydrophobic ligand) and PEG-Coomassie brilliant
modified polymer rich phase is achieved [118]. Another way of blue G-250 (pseudo-specific ligand). The glucose oxidase was
affinity partitioning ATPS can also be attained by the addition of free partitioned toward the PEG-Coomassie at bottom phase with
ligands (chemical or biological molecules) in conventional ATPS in 19-fold purification and 80% recovery, while the contaminants pro-
order to encourage a partition shift of the desired target molecule pelled to the upper phase. ␤-Galactosidase was best extracted in
instead of the chemical modification of the phase forming com- PEG–TMA systems (7-fold purification increase with 80% recov-
ponent (Fig. 4 Right) [53]. At preparative scale, these systems also ery). Authors have claimed that it was because of repletion between
present important advantages such as: high ligand concentration positively charged ligand TMA and negatively induced-charged ␤-
per unit volume of polymer solution, which renders high concen- galactosidase favouring its partition towards the bottom phase. The
tration of bound target biomolecule in one of the phases of system affinity attractions between active groups in polymer and contam-
(process intensification) and fast equilibrium achieving [119]. As inant proteins, which in turn provides a new strategy and open a
it combines the selective affinity technique with the robust tradi- new research area for purifying target enzyme molecules [123]. The
tional liquid–liquid ATPS, it can be implemented for the specific interaction between affinity ligand and specific biomolecule results
recovery of diverse biological molecules with the significant purifi- in an efficient separation process. However, the extended protocol
cation with improved yield. time and relative complexity within activation process may limit
Most of the reported investigations about affinity partitioning their use.
belong to polymer/polymer ATPS. Very few reports are available on An affinity ATPS purification strategies using free ligands has
affinity partitioning in polymer/salt ATPS mainly due to the inter- been implemented to recover various enzymes. In this methodol-
ference of high salt concentration with the biospecific interactions ogy, neither polymer activation nor ligand coupling are required,
[120]. Based on our literature, we have identified four additional which ultimately reduce the time and chemical consumption [117].
parameters usually studied for affinity ATPS; pH level for product Sharma et al. investigated an affinity ATPS consisting of PEG/salt for
partitioning, ionic strength of the system, affinity ligand nature and purification of phospholipase D in the presence of alginate (a known
its concentration (or modified polymers concentration). These fac- macroaffinity ligand) from peanuts and carrots. Incorporation of
tors must be taken into consideration in order to optimise ATPS alginate in the PEG phase was resulted in 91 and 93% of the enzyme
as affinity interactions between enzymes and affinity ligands are activity from peanuts and carrots respectively [124]. Another study
highly reliant on the ionic state of the active groups (according mentions the application of affinity ATPS using free ligand addi-
to pH) and the ionic strength of the system (depending on con- tion in the partition of chitinase. The systems were formed by PEG
centration of salt) [121]. In addition, ligand concentration and 6000/potassium phosphate ATPS in the presence of free chitosan
S.S. Nadar et al. / International Journal of Biological Macromolecules 101 (2017) 931–957 941

Fig. 4. Schematic diagram illustrating the partitioning of enzyme and contaminant proteins (impurities) in aqueous two phase affinity partitioning system: addition of a free
ligand (Right) or modification of PEG with ligands (Left).

Table 5
Extraction of enzymes by using aqueous two-phase affinity partitioning system.

Enzyme System Modification Remark Ref.

␤-Galactosidase Dextran T500/PEG 8000 Dextran-benzoyl Kp decrease: 0.036–0.018 [267]


Glucoamylase PEG 300/Phosphate Starch as free ligand Kp decrease from 6.6 to 0.73 [268]
Chitinase PEG 6000/Phosphate Chitosan as free ligand 90% recovery in UP after final elution [125]
Penicillin acylase PEG 4000/Phosphate PEG-benzoate 14-fold increase of Kp (0.1–1.4) and 60% [119]
recovery in a single
Cutinase PEG 1500/Phosphate Butyrate as free ligand 98% recovery in first extraction and Kp of 135 [269]
Penicillin acylase PEG 4000/phosphate PEG-trimethylamine Kp of 2.1 with 70% recovery in single step [119]
extraction
Phospholipase D PEG 6000/phosphate Alginate as free ligand 85% recovery in UP and 78-fold purification [270]
factor
Penicillin acylase PEG 4000/Phosphate PEG-palmitate Kp of 3.22 and 0.56 for contaminant proteins; [119]
75% recovery
␤-Mannanase Guar Gum/PES 200 Galactomannan as FL Kp increase from 1.37 to 7.06 [271]
Glucose-6-phosphate dehydrogenase PEG 6000/PES 100 Cibacron Blue F3GA (FL) ATPS with dye induced an increase in [117]
Kp from 0.73 to 1.59
Cellulase PEO/Dextran T500 Maleic acid-PEO 2-Fold increase in Kp for cellulose with [117]
40% modification rate at PEO phase

in solution as affinity ligand. Chitinase was extracted from differ- resulting polymer-bound metal can be used to augment the par-
ent sources such as puffballs (Lycoperdon umbrinum), Neurospora titioning of metal binding proteins into the polymer-rich phase in
crassa and cabbage (Brassica oleracea). At lower concentration of PEG/salt or PEG/polymer ATPS [126]. In one of the studies, affin-
chitosan (0.1%), chitinase recovered from N. crassa exhibited higher ity of papain towards metal ions (Cd, Co, Zn and Cu ions) was
activity recovery (86%) with 34-fold purification increase in top investigated. The papain enzyme was extracted from papaya fruit
phase, while, extraction from puffballs and cabbage exhibited a (Carica papaya) by traditional ATPS composed of PEG/ammonium
38-fold and 20-fold of purification with total activity recovery of sulphate and metal affinity ATPS. The yield was found to be 74.5,
88 and 80% respectively in the PEG rich phase. On the other hand, 72.3, 53.3 and 53.5% for Cu2+ , Zn2+ , Co2+ and Cd2+ ion respectively
as the concentration of ligands increased, purification factor and whereas for conventional ATPS, the yield was found to be 33.3%.
yield of enzyme reduced drastically. The authors suggested that This showed that ATPS coupled with metal ion enhanced the extrac-
high concentration of free ligands caused the non-specific interac- tion of enzyme due to affinity of enzyme towards metal ions [127].
tions between the chitinase and the affinity ligand, which lowering Another comparative experiments determined that, lactate dehy-
the overall binding capacity probably due to “crowding effect”. drogenase was extracted efficiently by using PEG/hydroxypropyl
Based on the results, authors demonstrated the potential of affin- ATPS in the presence of copper ions (Cu2+ ) and authors confirm that
ity partitioning of enzymes, emphasizing the effect of polymer a strong interaction between the copper ion and enzyme allows
characteristics (molecular weight and concentration), on ligand to concentrate in a specific phase [122]. Freire et al. evaluated
distribution through the system, hence those critical parameters the affinity dye based ATPS for papain partitioning from crude
should be considered in order to develop efficient recovery and latex using tri-azine dyes (CIBACRON Blue F3GA and PROCION Red
purification strategies [125]. HE3G). These dyes have highly specific functional groups and act
Metal affinity partitioning ATPS exploits the affinity of tran- as affinity factors in most of chromatographic applications. It was
sition metal ions for electron-rich amino acid residues (cysteine found that initial increase in PEG molecular mass and dye concen-
and histidine) present in enzymes. The metal ions are partially tration resulted in high partition coefficient value while further
chelated and coupled to a linear polymer (PEGor dextran), the increase in dye concentration reduced the partition coefficient of
942 S.S. Nadar et al. / International Journal of Biological Macromolecules 101 (2017) 931–957

papain. In addition to that when polymer concentration increased This has been demonstrated in the study carried out by Ventura
to maximum, partition coefficient of papain enzyme reduced along et al. The researchers purified lipase B from Candida Antarctica using
with the purity factor. The dyes CIBACRON Blue F3GA and PROCION IL/potassium salts [142]. The results suggested that the increase in
Red HE3G combined with PEG/ammonium salt resulted into 11.6 the alkyl chain length induces drastic increase in partition coef-
and 13.4-fold purification factor [128]. Affinity partitioning ATPS ficient of other protein than partition coefficient of enzyme which
has a wide scope for enzyme purification, however the application remains consistently very low. The increase in the alkyl chain length
of aqueous two-phase affinity partitioning system for recovery of makes the ionic liquid more hydrophobic, decreasing the Coulom-
high commercial value biologicals products is expected. bic interactions and increasing the dispersive forces that occur
between the enzyme molecules and the ionic liquids at the IL-rich
5.2. Ionic liquid based aqueous two phase partitioning system phase. These factors seem to partition the contaminant proteins
into upper IL-rich phase. Due to its low isoelectric point (pI 6.0),
In last decade, ionic liquids (ILs) have emerged as an alternative the lipase is negatively charged at these pH conditions (pH 7.0)
phase forming component for ATPS particularly when proteins are which resulted in the increase of its hydrophilic character, creating
easily denatured and lose their biological activity in contact with stronger affinity of enzyme for the bottom salt-rich phase [145].
organic solvents [129]. They have unique properties such as neg- The difference observed in the partitioning behaviour between the
ligible vapour pressure, non-flammability and high thermal and contaminant proteins and enzyme, must result from differences
chemical stability with wide electrochemical window [129,130]. in their hydrophilicity/hydrophobicity that will allow the parti-
Most water-soluble ILs are salting-in inducing electrolytes (known tion of contaminant proteins and enzyme into different phases. The
as “chaotropic” salts) in the presence of aqueous solutions and enzyme recovery obtained were consistently above 96% with purifi-
salting-out inorganic salts (known as “kosmotropic” salts) result- cation factor of 2.6-fold for 1-methyl-3-octylimidazolium chloride
ing in the formation of two phases in ATPS [131–133]. The driving ([C8 mim]Cl). The presence of IL in the medium is reported to have a
force for phase separation in IL ATPS is the competition between harmful effect on the enzyme activity, though not very significant at
the salt and IL for water molecules. The higher affinity of the inor- low concentration level [146]. The increase in the purification fac-
ganic salt for water induces a migration of water away from the IL tor must be the result of elimination of the contaminant proteins,
ions which decrease their hydration and reducing the solubility of which are acting as enzymatic inhibitors.
ILs in aqueous solution [134]. The commonly employed inorganic Aforementioned studies involving ILs in extraction, purification
salts are ammonium, sodium or potassium based salts of multiple and recovery of enzymes depict that, in many cases, the substitution
charged anions, such as carbonate, citrate phosphate or sulphate of toxic and flammable organic solvents is not only beneficial for the
(strong salting-out inducing anions) [135,136]. Recently, the possi- environment but also results in higher yields in catalytically active
bility of extracting some macromolecules (proteins and enzymes) form compared to the classic ATPS. Even though ILs can perform
using ILs has been explored as ILs have the potential to retain high better than organic solvents in biotechnological applications, much
enzyme activity and stability with significant extraction efficiency research still needs to be done, especially concerning synthesis and
[137]. The ATPS formed by ILs and salts are reported to give higher the recovery of the IL due to its high cost. In addition, up-scaling
recoveries of the proteins as compared to conventional ATPS com- and integration of the newly advanced processes to larger scale
posing PEG and inorganic salt [138]. In addition, IL-based ATPS also needs further exploring. Nevertheless, the research in the field of
exhibit better process properties, such as little emulsion formation, ILs for extraction and biocatalysis (in synthesis) is very interesting
lower viscosity and fast phase separation [139]. On the whole, it and found to be very promising.
seems that water-soluble ILs are likely to be more appropriate for
the isolation of enzymes which can be considered as an economi- 5.3. Thermoseparating polymer-based aqueous two-phase system
cal and efficient extraction approach for those separation systems
[129]. Therefore, IL based ATPS offer the opportunity to combine the An advanced and improved ATPS have been developed by using
extraction and concentration of biocatalysts in active form [140]. thermoseparating polymers (TSPs) consisting of a high water con-
The enzyme extraction by using IL is summarized in Table 6. tent (70–90% w/w) that provides a mild environment for separation
Deive et al. have compared IL based system with conventional of sensitive biomolecules (mostly enzymes). It requires a lower
PEG based system for extraction of three different lipases; CalB, amount of phase forming components for two phases formation
CalA and Thermomyces lanuginosus lipase. The partition coefficients with thermoseparating ATPS, thereby, minimizing the risks of both
in the ILs system was two to three folds higher than those obtained salting out and precipitation of proteins and preventing enzyme
with PEG based systems. The author attributed this favourable par- denaturation [113,147]. Moreover, phase separation can be done
tition ratio to the large difference in polarity between the two more rapidly and selectively at room temperature due to efficient
phases in the presence of IL [141–143]. In another study, the sepa- mass transfer facilitated by high interfacial contact area. Addition-
ration and purification of lipolytic enzyme produced by Bacillus sp. ally, the phase-forming components of thermoseparating ATPS are
was done from the fermentation broth by using IL based ATPS. At generally more safe, non-flammable, non-toxic and relatively envi-
the end of the production phase, improved purification factors and ronmentally friendly in nature [148,149]. Along with ease of phase
enzyme recovery efficiencies was observed[140]. In many cases, separation and biomolecule partitioning, it has the ability to han-
imidazolium-based ionic liquids have proven to be the most effi- dle high capacity and reduce the volume of subsequent purification
cient cations, and [C4 SO4 ], [C2 SO4 ] and Cl− were chosen as the steps, large-scale purification using thermoseparating ATPS can
optimal anions [129]. Other works have focused on the extraction of be easily and reliably predicted from laboratory data [150,151].
a peroxidase and two different alcohol dehydrogenases, using imi- Thus, thermoseparating ATPS is a potential solution for industrial
dazolium and ammonium based ionic liquids. In the latter work, demand of highly-efficient large-scale and cost-effective separa-
apart from the increased specific activity for two different alco- tion technology with short processing time [152]. Various examples
hol dehydrogenases, it also manifested that the presence of an IL of extraction of enzymes by using ATPS based on TSP are mentioned
provides the opportunity to combine an extraction process with in Table 7.
efficiencies of 90% and the increased performance in the biocat- The temperature-induced ATPSs give rise to development of
alytic reaction [144]. TSPs with distinctive thermoseparating properties and easy recy-
The extraction of enzyme also depends on the chemical nature clability (Fig. 5) [153]. The common employed TSPs are random,
of ILi.e. cation alkyl chain length and the anion and cation core. diblock and triblock copolymers of hydrophilic ethylene oxide (EO)
S.S. Nadar et al. / International Journal of Biological Macromolecules 101 (2017) 931–957 943

Table 6
Extraction of enzymes by using ionic liquid based aqueous two-phase system.

Enzyme Source Ionic liquid Salt Remark Ref.

Lipase B Candida antarctica ([C2 C1 im]Cl, Dipotassium Maximum [142]


[C7 H7 C1 im]Cl, phosphate and purification and recovery were
[C4 C1 im][CF3 SO3 ], Potassium dihydrogen obtained for cations with a C8
[C4 C1 im][N(CN)2 ], phosphate side alkyl chain, the [N(CN)2]
[C4 C1 im][C1 SO3 ], anion which belonging to the
[C4 C1 pyr]Cl, pyridinium family
[C4 C1 py]Cl,
[C4 C1 pip]Cl,
[C8 C1 py][N(CN)2 ])
Lipase Bacillus [C4 C1 im]Cl Dipotassium IL-based ATPS was superior to [272]
sp. ITP-001 phosphate and PEG-based ATPS for the
Potassium dihydrogen purification
phosphate
Alcohol Lactobacillus brevis Ammoeng100TM , Dipotassium The presence of IL exhibited [273]
dehydrogenases and thermophilic Ammoeng101TM , phosphate and positive stability-enhancement
bacterium Ammoeng110TM Potassium dihydrogen effect on enzymes during
phosphate purification.
Horseradish [C4 C1 im]Cl Dipotassium The lyotropy is beneficial to [274]
Peroxidase phosphate enzyme activity maintaining
which should be consider
while choosing proper IL-ABS
as extraction system
Lipase A Candida antarctica [C2 C1 im][C4 SO4 ] Ammonium sulphate High charge–density salts and [143]
amino acids have been studied
in order to evaluate their
potential as phase forming
agents and as stabilizers of
enzyme.

Table 7
Extraction of enzymes by using thermoseparating aqueous two-phase system.

Enzyme Source Thermo- Salt/ Recovery Polymer recycling Ref.


separating alcohol efficiency
Polymer

␣-Amylase Leucosporidium EOPO 2500 Sulphate 91% yield and – [147]


scottii L117 3.31-fold PF
CGTase Bacillus cereus EOPO 3900 Phosphate 87% yield 2 times [156]
and 13.1-fold PF
Endo- Kluyveromyces UCON Sulphate Up to 99% yield and – [275]
Polygalacturonase marxianus 10-fold PF
Laccase Peniophora cinerea UCON Potassium 134% yield and – [276]
dihydrogen 1.31-fold PF
phosphate
Lipase Burkholderia EOPO 3900 Phosphate 99% yield and 3 times [90]
cenocepacia strain 14-fold PF
ST8
Laccase T. versicolor UCON Dipotassium Kp = 0.272 – [97]
phosphate
Lipase Burkholderia EOPO 3900 – 99% yield and – [157]
cepacia 14-fold PF
Lysozyme Hen egg white EO50 PO50 Dipotassium 85% yield and 3 times [159]
phosphate 16.9-fold PF
Thermostable Methanococcus PEO–PPO Ammonium Up to 90% and – [277]
␣-amylase jannaschii sulphate 3.31-fold PF
L-asparaginase E. coli 11303 EO40 PO60 -870 Potassium 73.3% yield 2 times [158]
dihydrogen
phosphate and
Dipotassium
phosphate
Thermo-acidic Hylocereus EOPO 2500 2-propanol 96.6% yield and 5 times [278]
amylase polyrhizus 14.3-fold PF

and hydrophobic propylene oxide (PO). The EOPO copolymers have employing random copolymer of EOPO and compared it with tra-
been usedas replacement of PEG with different type of salts for ditional PEG/ammonium sulphate ATPS. The 3.33-fold purification
partitioning. These polymers have a wide range of application as factor was observed for copolymer/salt system, whereas it was
their EO/PO ratio and molecular weight can be greatly varied which 2.90-fold for PEG/salt system [150]. In another extraction studies,
shows significant influence on the separation of biomolecules Ng et al. purified cyclodextrin glycosyltransferase from B. cereus
[154]. Other examples of TSPs include poly ethyl hydroxyethyl upto 13.1-fold with a yield of 87% and the recovery upto 80% by
cellulose, poly (vinylcaprolactam), poly (N-isopropylacrylamide) ATPS composed of EOPO 3900 and potassium phosphate salt [156].
and non-ionic surfactants [155]. Peeples et al. purified a recom- The concentration and molecular weight of EOPO also shows
binant, thermostable ␣-amylase from Methanococcus jannaschii by a significant influence on the partitioning of protein due to
944 S.S. Nadar et al. / International Journal of Biological Macromolecules 101 (2017) 931–957

Fig. 5. Schematic diagram illustrating the principles of a temperature-separating polymer based ATPS.

change in hydrophilic to hydrophobic ratio. The hydrophobicity EO/PO ratio, as well as block size and molecular mass [93,148].
of the polymer increase with molecular weight due to increase Also, the LCST of TSPs can be lowered by making the polymer
in hydrophobic area to hydrophilic group ratio. The extraction of more hydrophobic (i.e. increase of PO content) or by addition of
lipase derived from Burkholderia cepaci was carried out withdiffer- hydrophilic salt (e.g. sodium sulphate), acids/bases or ionic sur-
ent molecular weights (970, 2500, 3900 and 12000 Da) by varying factants (e.g. SDS) which promote partitioning of target products
concentrations (2–20%) of EOPO. The purification factor increased [147,162,164].
with increase in molecular weight till EOPO 3900 and further In one of the studies conducted by Ng et al., employed ATPS com-
increase in molecular weight resulted into lowering of purification posed of TSP and phosphate salt for the recovery of cyclodextrin
factor [157]. In partitioning of L-asparaginase by PEO–PPO–PEO glycosyltransferase from Bacillus cereus and recycled phase com-
(triblock copolymers)/phosphate system, the purification factor ponents over four successive extractions. The purification factor
showed an increasing trend for increasing triblock copolymer and the yield of lipase were obtained 14 and 99%, respectively even
molar mass. Further, the yield of L-asparaginase obtained by after four successive purification cycles[156]. In most of the recy-
triblock copolymer/phosphate system was 73.3%, while for con- cling studies, the yield and purity fall following the recycling cycles
ventional (PEG/salt) ATPS, it was 52.1% [158]. due to increasing contaminants in bottom phase after each cycle,
System pH plays a crucial role in TSPs based ATPS as it modifies thus, recommending that the EOPO copolymer can only be recy-
the net charge on protein molecules and thus, influence partitioning cled and reused for few times. However, there are exceptions in
of the target product. The influence of pH (7–10) on lysozyme par- both lysozyme and lipase extraction studies which showed that
titioning in EO50 PO50 /potassium phosphate ATPS was studied by the parameters such as purity, yield and volume ratio are quite
Dembczyński et al. The pH 9 was found to be effective for extraction stable, regardless of high accumulation of contaminant protein in
of lysozyme (pI 10.9) [159]. Some enzymes such as fungal laccase the bottom phase throughout four consecutive recoveries [90,165].
(pI 6) and l-asparaginase (pI 4.9) have been extracted within range Thermo-acidic amylase from red pitaya (Hylocereus polyrhizus) peel
of acidic pH. The hydrophobic interaction dominates at pH around was satisfactorily partitioned into the polymer-rich top phase in
pI of biomolecules due to zero net charge of proteins, while electro- the system composed of 30% (w/w) EOPO 2500 and 15% (w/w) 2-
static forces show greater effect at other pH. Besides that, system propanol at pH 5.0. The amylase was successfully recovered with a
pH generally has no effect on the cloud point (CP) of TSPs, with high purification factor of 14.3-fold along with the yield of 96.6%
the exception of cationic hydrophobically modified ethylene oxide and copolymer was also recovered and recycled above 97%, making
polymer for which CP increased rapidly at pH <9 [160]. the method more economical than the traditional ATPS method.
Apart from outstanding advantages of temperature-induced In thermoseparating based ATPS, simplicity of product recovery
separation, TSPs can be recycled and reutilized for multiple times. and economic chemical cost provide a huge scope in extrac-
After heating beyond a critical temperature known as lower criti- tion of enzymes. However, novel TSPs should be developed with
cal solution temperature (LCST) or cloud point (CP), the solubility of unique characteristics and also, different combinations of two
TSPs decreases and solution turns into turbid and hazy to form two phase system needs to be explored. It is necessary to find corre-
phases eventually [161]. In these systems, the top phase has almost lations between different parameters and predict the partitioning
100% water content, while the bottom phase contains copolymer behaviour by using mathematical models. Additionally, kinetics of
[162]. This feature helps in easy extraction of target product and bioseparation and underlying mechanisms need be further studied
helps to recycle and reutilization of polymers possible without the and simplified into simpler concepts.
need for additional third component or extra separation processes.
This makes the separation process not only a greener and economic
downstreaming technique, but also makes the recovery more effi- 5.4. Aqueous micellar two-phase system
cient. The commonly used PEG polymer in two phase partitioning is
also a TSP. However, LCST above 100 ◦ C (low-molecular-mass PEG The aqueous micellar two-phase system (AMTPS) is another
has a LCST as high as 180 ◦ C) is unsuitable in most case of thermo variation in ATPS, which is also known as surfactant-mediated
separation of biomaterials [154]. On the other hand, hydrophobi- phase separation. It is a more efficient method for purification of
cally modified EOPO has low LCST around 12 ◦ C at 3% w/w, which biomolecules. AMTPS utilizes a binary phase micellar system with
means that thermo separation can be done near room tempera- the help of surfactants at low concentrations in aqueous solutions to
ture [163]. LCST of thermoseparating polymers depends on the induce micelles [166]. Once the temperature of the surfactant solu-
tion reaches a CP temperature, two phases are formed in AMTPS due
S.S. Nadar et al. / International Journal of Biological Macromolecules 101 (2017) 931–957 945

to difference between the physicochemical environments [167]. establish a stable interface without surfactants or special surface
When the temperature is above the CP, the surfactant molecules treatment. In recent years, the parallel laminar flow of ATPS in
self-aggregate to maintain an aqueous (surfactant-depleted) phase microfluidic devices is widely studied, serving as an ideal plat-
and results into two phase formation i.e. in a bottom, micelle- form for rapidly extracting and separating biomolecules [174,175].
rich phase and a top, micelle-poor phase [168]. The most common Compared with conventional batch processes, microfluidic proto-
phase-forming nonionic surfactants used are Triton X-114 and cols offer a number of advantages. Microfluidic systems can easily
other alkyl polyoxyethylenes (Cm EOn ) such as n-decyltetraethylene manage microliters to picoliters of fluid volumes, which reduce
oxide. The partitioning of proteins (enzymes) are governed primar- sample volumes and reagent costs. Also, short diffusion distances
ily by steric, repulsive and excluded-volume interactions between of microfluidic systems translate into faster and more efficient sep-
the globular hydrophilic proteins and the non-charged cylindrical aration without further need of mixing [176,177]. Moreover, two
micelles, which drive desired biomolecules preferential into the phases are easily recovered from different exit branches by using
bottom micelle-rich phase while simultaneously eliminating most designed channel architectures, whereas in the standard batch
of the contaminants (proteins) to the micellar poor top phase, due processes, the phases are separated in a settler which usually con-
to excluded-volume interactions between the contaminates and sumes a significant amount of time. Further, separation processes
micelles. The formation of micelles represent a delicate balance of by microfluidic device can allow real time monitoring of the status
several intermolecular forces including those of the hydrophobic, of a separation process by using optical detection techniques and
van der Waals, steric and electrostatic types. The resulting micelles performing multidimensional separations on the basis of affinity
are labile microstructures formed by the non-covalent binding of partitioning in ATPS [178].
individual surfactant molecules, contrary to polymers, which are In the past few years, a number of groups around the globe have
formed by the covalent binding of individual monomers [169]. Also, discovered the opportunities related to ATPS in microfluidic devices
micellar size and shape, as well as the associated micellar solution for enzyme separation. Novak et al. used a three-stream ATPS with
properties can be tuned in situ by varying solution conditions, such PEG in the outer stream and potassium phosphate in the middle
as temperature, surfactant concentration, salt type and concentra- stream to extract the enzyme ␣-amylase. They demonstrated a sig-
tion. nificant improvement in separation time. The microfluidic system
Jaramillo et al. carried out extraction of the pectinase (from performed extraction within 10 s, with extraction efficiency (i.e.
Aspergillus oryzae) by using a micellar Triton X-114/sodium phos- the difference in concentration between the inlet and outlet of the
phate AMTPS system. This system gave highest yield of 375.5% in microchannel of enzyme) of 52%. On the other hand, conventional
the top phase [170]. In another studies, Duarte et al. extracted lipase macro-scale ATPS (composed of PEG 4000 and potassium phos-
produced by psychrotrophic yeast Leucosporidium scottii using phate) extracted ␣-amylase with an extraction efficiency of 74% in
AMTPS composed of Triton X-114 (TX-114)/McIlvaine buffer pH 2.5 h. Further, the researchers have claimed that during continuous
7.0 and compared it with PEG/phosphate salts and PEG/polyacrylic parallel flow extraction processing, simultaneous phase separation
acid. Lipase preferentially partitioned into the micelle-rich phase occurred at the exit of the microchannel system, which resulted
with the purification factor 1.32-fold which is much higher than in shortening of the process time as compared to the conventional
traditional ATPS (compose of PEG/phosphate) systems [75]. Amid batch extraction [179]. In another study, Hahn et al. separated car-
et al. investigated the influence of different non-ionic surfactants bonic anhydrase and beta-galactosidase using a microfluidic ATPS.
(Pluronic L31, Pluronic L81, Pluronic L61, Pluronic L121, TX-114) The dextran phase containing enzyme was sandwiched between
and different types of salts (K2 SO4 , KNO3 KH2 PO4 , and KCl) on the two PEG phases and electric field was applied perpendicular to
purification of serine protease from kesinai leaves. The protease was the interface in a channel. They demonstrated that separation of
partitioned into bottom micellar phase with high purification fac- protein can be achieved through differences in electrophoretic
tor (10.3-fold) and yield of 92% in the AMTPS composed of Pluronic mobility and/or partition coefficients in microfluidic ATPS [180].
L61/KNO3 . [112]. In above all cases, the enhanced purification was Undoubtedly, ATPS combined with microfluidic system can
observed due to improve protein solubility in bottom phase by the provide a platform for continuous separation/purification of
salt addition and increase the hydrophobic interaction between macromolecules like proteins/enzymes. However, in order to
the surfactant phases. In addition to that, the protease from the bot- achieve higher extraction efficiency by using microfluidic sys-
tom phase was re-extracted to a new aqueous phaseand recycle the tem, it is necessary to screen different combinations of number
surfactant. In another studies for easy recycling, thermoseparating of inlet/outlet of the device to generate stable vertical interface
polymer triblock copolymer (PEOn –PPOm –PEOn ) was incorporated between the two phases, enabling better partitioning of desired
with Triton X-114 to extract lipase (from Burkholderia sp. ST8). biomolecules. Also, integration of multi-dimensional separations
Lipase recovery was achieve with purification factor of 7.2-fold and (i.e. electrochemical potentials) with ATPS along or across the
0.032 partition coefficient [171]. microchannel will improve or shift the selectivity in microfluidic
ATPS [172].
5.5. Microfluidic platform for ATPS

In conventional ATPS, separation of two phases usually takes 6. Three phase partitioning
few hours, and in some cases agitation is required to mix the two
phases. The partitioning of two phase system typically depends In recent years, three phase partitioning (TPP) has emerged
on the diffusion coefficient. Recently, the microfluidic technology as a simple, rapid, efficient, modest, inexpensive and integrated
has emerged as a promising tool which offers a new approach bioseparation technique for recovery, purification and enrichment
of integration and miniaturization to address issues associated of biomolecules such as enzymes from complex mixture [181]. It is
with traditional APTS. The microfluidic devices contain a num- easily scalable and can be employed directly with crude slurry and
ber of co-flowing streams of immiscible phases guided through a is performed at room temperature. It does not involve any addi-
microchannel [172,173]. The length of the diffusion path is adjusted tion of polymer in the system to be removed later [96,182,183].
by geometric constraints or hydrodynamic means which speeds It consists of the sequential addition of a sufficient amount of
up the mass transfer. Two-phase flow of ATPS in microfluidic salt (ammonium sulphate, potassium phosphate, and sodium cit-
chips allows the low interfacial tension resulting in high capil- rate etc.) and an organic solvent (t-butanol, 2-butanol, 1-propanol,
lary number even at low flow rates, making it straightforward to and 2-propanol etc.) in the crude extract. The reaction mixture
946 S.S. Nadar et al. / International Journal of Biological Macromolecules 101 (2017) 931–957

Table 8
a Extraction of enzymes from fermentation broth by using three phase partitioning (TPP).

Enzyme Source Salt Organic solvent pH Purification factor Ref.

a-Galactosidase and invertase Aspergillus oryzae Ammonium tert-butanol 4.3 15 and 12-fold respectively [206]
sulphate
Serratiopeptidase Serratia marcescens Ammonium tert-butanol 7 9.4-fold [191]
sulphate
Laccase Coriolopsis trogii Ammonium tert-butanol 5 15.6-fold [213]
sulphate
Fibrinolytic enzyme Bacillus sphaericus MTCC 3672. Ammonium tert-butanol 9 16.15-fold [220]
sulphate
Aryl Alcohol Oxidase Pleurotus ostreatus Ammonium tert-butanol 6 10.19-fold [185]
sulphate
Laccase Pleurotus ostreatus Ammonium tert-butanol – 7.22-fold [279]
sulphate
Exo-inulinase Aspergillus niger Ammonium tert-butanol 4 10.2-fold [212]
sulphate
L-asparaginase Capsicum annuum L. Ammonium tert-butanol 8.5 6.83-fold [280]
sulphate
Naringinase Aspergillus brasiliensis Ammonium tert-butanol 5.5 4.4-fold [209]
sulphate
Pecinase Aspergillus niger Ammonium tert-butanol – 10-fold [199]
sulphate
␤-galactosidase Lactobacillus acidophilus Ammonium tert-butanol 7 7.5-fold [281]
sulphate
Laccase Ganoderma sp. WR-1 Ammonium tert-butanol 7 13.2-fold [282]
sulphate
Invertase Saccharomyces cerevisiae Ammonium tert-butanol 4 15-fold [204]
sulphate

Table 8b Extraction of enzyme from natural sources by using three phase partitioning (TPP).
Enzyme Source Salt Organic solvent pH Purification factor Ref.

␣-Galactosidase Citrullus vulgaris (watermelon) Ammonium tert-butanol 5.5 2.7-fold [211]


sulphate
Ipomoea peroxidase Ipomoea palmate (leaves) Ammonium tert-butanol – 18-fold [200]
sulphate
Proteases Papaya peels Ammonium tert-butanol 7 10.1-fold [208]
sulphate
Invertase Solanum tuberosum (Potato tubers) Ammonium tert-butanol – 5.67-fold [283]
sulphate
Polyphenol oxidase Malus domestica (Fuji apple) Ammonium tert-butanol – 54.41-fold [284]
sulphate
Pectinase Lycopersicon esculentum (Tomato) Ammonium tert-butanol 6.5 9-fold [199]
sulphate
␣-Galactosidase Solanum muricatum (Pepino) Ammonium tert-butanol 5.5 6.2-fold [192]
sulphate
␣-Galactosidase Lycopersicon esculentum (Tomato) Ammonium tert-butanol 4 4.3-fold [205]
sulphate
␤-Galactosidase Cicer arietinum (Chick pea) Ammonium tert-butanol 6.8 10.1-fold [195]
sulphate
Peroxidase Citrus sinenses (orange peels) Ammonium tert-butanol 6 18.20-fold [190]
sulphate
Catalase Solanum tuberosum (Sweet Potato) Ammonium tert-butanol 7 14.1-fold [203]
sulphate
Invertase Lycopersicon esculentum (tomato) Ammonium tert-butanol 4.5 8.6-fold [197]
sulphate

was mixed well and decantation was carried out. The mixture tulated as key contributors for the partitioning of the target enzyme
separates into three distinct phases: a t-butanol rich upper layer [185,186]. The TPP process is affected by physiological parameters
and an aqueous lower layer which are formed [184]. The third like temperature, pH salt type, co-solvent and their concentration.
phase is formed by a protein-enriched in between the polar and The excellent results can be obtained by streaming these parame-
non-polar solvent. During the separation, some small molecular ters. Various enzymes were extracted from fermentation broth as
weight and non-polar compound impurities (such as lipids, phe- well as from natural sources which are summarized in Table 8a and
nolics and some detergents lipids and pigments) are accumulated 8b. Gu et al. showed the effectiveness of TPP for the extraction of
in upper t-butanol phase while polar compound impurities (like ␣-galactosidase from Aspergillus niger by comparing it with ATPS
protein contaminant, saccharides and cell debris) get concentrated (which are considered as efficient and economical strategies). For
in the lower aqueous phase (Fig. 6) [96,185]. The probable reason ATPS comprising PEG/phosphate, highest purification factor upto
behind this protein isolation is that organic solvent binds to the 3.25-fold with 84.41% activity recovery were obtained. On the other
precipitated enzymes, thereby increasing their buoyancy and caus- hand, in TPP system made up of ammonium sulphate and t-butanol
ing them to float above the denser aqueous salt layer and forms was exhbited excellent purification (6.27-fold) with 97.21% activity
the third phase (interphase). Additionally, combination of salting recovery [187].
out, isotonic precipitation, co-solvent precipitation, osmolytic and The solubility of proteins/enzymes is mainly dependent on the
kosmotropic cause enzyme hydration shifts which have been pos- effect of ionic strength of the solution [188]. Hence, it is essential to
S.S. Nadar et al. / International Journal of Biological Macromolecules 101 (2017) 931–957 947

not only the overall conformation changes but also alter the sec-
ondary structure content of the enzyme resulted into increased
flexibility in activated enzyme molecule [197,198]. A remarkable
increase in the catalytic activity and yield of pectinase, peroxidase,
proteinase K and trypsin inhibitor obtained from TPP have been
reported [199–202]. Hence, it is necessary to select appropriate salt
and its concentration during enzyme extraction by TPP system.
Another important parameter is pH value of partitioning
medium and isoelectric point (pI) of target protein which directly
influences the recovery and purification factor of proteins. Distri-
bution and partitioning of biomolecules in TPP systems alternated
with pH due to electrostatic interactions between charged pro-
tein (enzyme) and phases [203]. Electrostatic forces and binding of
anionic salts (i.e. SO4 2− , HPO4 2− , and C3 H5 O(COO)3 3− ) to cationic
protein molecules, which promote conformational shrinkage and
macromolecular contraction, are the main causes of the strong salt
anion-pH dependency in salting out [189,204]. Proteins tend to pre-
cipitate most readily at their pI. When system pH is above the p I of
Fig. 6. Schematic diagram illustrating the partitioning of enzyme and (polar and protein, it will attain net negative charge and would be propelled
non-polar) contaminant impurities in three phase partitioning. to the bottom aqueous phase. On the other hand, if system pH for
the TPP process is lower than the pI of the protein, enzymes get
precipitated or accumulated between the two phases in TPP. This
observe the effect of various salts and salt concentration on parti- might be due to the electrostatic component of the reactions when
tioning of enzyme in TPP system. Salts such as ammonium sulphate, anion/cation of the salt binding to oppositely charged proteins
magnesium sulphate, and sodium sulphate have been mostly used leading to the partition of most contaminant proteins into aque-
in TPP process [189]. At lower salt concentration it may not able ous bottom phase above isoelectric point. Akardere et al. purified
to alter the hydrophobic surface of enzyme leading to insufficient invertase from Saccharomyces cerevisiae upto 15-fold purity with
precipitation of desired enzyme. On the other hand, at higher salt 363% activity recovery at pH 4.5 while at pH 5.0 provided 8.5-fold
concentration, water molecules are attracted by salt ions result- purification with 300% activity recovery [204]. Similarly, Çalci et al.
ing in enhanced protein–protein interactions and the coagulation reported that at pH 4.5, ␣-galactosidase from tomato was extracted
of protein molecules through hydrophobic interactions [190]. Sul- upto 4.3-fold purification with 80% yield in the system composed
phate ions being early member of the hofmeister series interact of ammonium sulphate and t-butanol which was increased by two
well with water, forming hydrogen bonds and dehydrating pro- folds at pH 6.0 [205]. Dhananjay et al. reported the extraction of
teins [96]. Ammonium sulphate showed the maximum yield and ␣-galactosidase and invertase from Aspergillus oryzae which were
recovery in most of the cases of TPP. This could be due to ammo- accumulated in a coherent middle phase in a TPP system under dif-
nium sulphate, as it has a high hydration capacity and affinity for ferent pH conditions. The pH of the TPP system was adjusted at 4.3
water compared to other salts. Large proportion of water molecules for ␣-galactosidase to get 50% activity recovery with 15-fold purity
are involved in hydration of sulphate ions thereby increasing their whereas invertase were extracted at 5.0 pH with 12-fold purity and
effective radius and acting as pushing kosmotrope through its sig- 54% activity recovery [206].
nificant hydration. It also promotes protein stability by favouring In TPP system, the solvent plays a vital role in the formation of
hydrophobic interactions [184]. In addition to that, various other phases. Mostly tertiary butanol (t-butanol) is employed as organic
researchers have reported that, the salting out effect in TPP is asso- solvent as it acts as a kosmotrope in synergism with salt kos-
ciated with kosmotropy, ionic strength effects and the binding of motropy [200]. The phase formation so far dependent on t-butanol
sulphate to cationic sites of a protein [191,192]. Additionally, Rao and salt, a C4 alcohol which is a differentiating solvent and ammo-
et al. claimed that large sulphate ions crowd together in aqueous nium sulphate as effective kosmotropic agent. t-Butanol binds to
phase and alter the solubility of protein in bottom phase, finally the precipitated proteins, thereby increasing their buoyancy and
pushing it into the organic phase due to increase in the interfa- causing the precipitates to float above the denser aqueous salt
cial tension. Higher interfacial tension generates a greater polarity layer. t-Butanol appears to be kosmotropic and crowding agent
difference and a higher density difference between both phases at room temperature, resulting in “enzyme–t-butanol coprecipi-
in the salt system [193]. However, with an increase in concentra- tate” which float above denser aqueous salt layer [203,207]. It is
tion of ammonium sulphate, the degree of purification factor and believed that because of its size and branched structure, t-butanol
activity recovery decreased significantly [194,195]. The ammonium does not easily penetrate interior to the enzyme molecules and
sulphate saturation (40–60%) resulted in the 16.2% yield of nattok- hence does not cause denaturation [184]. t-Butanol is not univer-
inase from Bacillus natto NRRL-3666 with a 1.46-fold purification. sal solvent to be always used for TPP, other C3 , C4 and co-solvents
Further increase in ammonium sulphate saturation (80–95%) dras- may also serve because they too act as a differentiating co-solvents
tically reduced the yield (0.9%) and purity (0.23-fold) [196]. Apart [208,209]. Ketnawa et al. used t-butanol, 1-butanol, 1-propanol and
from ammonium sulphate salt, various researcher used different 2-propanol to purify alkaline proteases derived from fish viscera.
type of salts in TPP. The salts such as sodium citrate, potassium 2-propanol provided higher purification as compared to other sol-
phosphate were used to get the maximum recovery of alkaline pro- vents, whereas, t-butanol provided higher activity recovery for the
teases from farmed giant catfish viscera. Sodium citrate provided same proteases [189]. Similarly, in the extraction of ␣-galactosidase
the highest recovery (142%) with purification (4.65-fold) followed from fermented media of A. oryzae, t-butanol gave the 12-fold
by ammonium sulphate and potassium phosphate. In some cases, purification factor and 92% activity recovery while 1-butanol, 2-
It has been often observed that three-phase partitioning leads to propanol and 1-propanol gave 69, 35 and 40% recovery, respectively
simultaneous activation of the enzyme molecules which (if the [188]. In this TPP based protocol for enzyme partitioning, t-butanol
enzyme recovery is high) resultsinto an apparently observed higher has been found to consistently perform better than all other organic
yield (more than 250%) [189]. During TPP, enzymes were undergo solvents. The concentration of t-butanol need to be optimum to get
948 S.S. Nadar et al. / International Journal of Biological Macromolecules 101 (2017) 931–957

synergic kosmotrope effect with salt. This is because higher solvent ing their industrial applications economically feasible [224]. One of
quantities attract more water from aqueous phase which leads to the effective means to decrease the cost of an enzyme is the reduc-
increased salt concentration in aqueous phase and precipitation of tion of separation and purification cost, which can be achieved by
enzyme at interface [210]. Besides, t-butanol can be recycled by either reducing the number of purification steps and/or increasing
easy evaporation at 82.4 ◦ C (boiling point) [96]. recovery rates or developing simultaneous purification and immo-
Apart from pH, temperature is an important operation param- bilization [225]. There are few strategies in which the enzymes are
eter affecting the enzyme configuration and overall stability [124]. purified and immobilized parallelly. It not only reduces the steps
The use of low temperatures in solvent or salt precipitation of biocatalyst preparation but also makes application of biocatalyst
prevents the heat generation, thus ensures minimal protein denat- economically feasible [226–228].
uration [211,212]. At lower temperature (4 ◦ C) the recovery was From past decades, cross linked enzyme aggregates (CLEAs)
same as at the ambient temperature which might be due to the have emerged as a versatile and novel carrier free immobilization
low movement of particle solvent at lower temperature. However technique [229]. The enzyme immobilization by CLEAs involves
at 25 ◦ C, the conformation of enzyme changed into highly active two steps: precipitation of an enzyme from aqueous solution fol-
form.While, enzyme activity recovery decreased at higher temper- lowed by cross linking with a bi-functional reagent [230]. In the first
ature (131% activity recovery at 45 ◦ C) in comparison to ambient step the soluble enzyme is aggregated by the addition of precipi-
temperature (154%, 31 ◦ C) but considerably increase in the purifi- tating agents such as salts (ammonium sulphate), water-miscible
cation fold of the enzyme in the precipitate. This may be due to the organic solvents (methanol, ethanol, propanol, DMSO, etc.) and
synergistic effects of the phase components along with tempera- non-ionic polymers (PEG) to an aqueous solution of enzyme. In
ture, which decreases the selectivity of extraction, thus reducing the aggregate, the enzyme molecules are held together by non-
the yield as well as the degree of purification [213]. covalent bonding without perturbation of their tertiary structure,
The partitioning of enzyme is always restricted due to mass i.e. without denaturation. In the second step, the formed aggregates
transfer driven phenomenon and any improvement in mass trans- are chemically cross linked to each other by a bifunctional reagent
fer leads to enhanced partitioning and subsequent purity of the (mostly glutaraldehyde) via the reaction of amino groups of Lys
target enzyme [214]. Hence, ultrasound is employed as the pro- residues on the external surface of the enzyme (Fig. 7) [231,232].
cess intensification tool in the extraction of enzymes in recent After chemical cross linking, the aggregates become permanently
times [215]. An ultrasonic wave leads to creation of small rapidly locked into insoluble pre-organized superstructure with the reten-
growing bubbles; collapse of such bubbles is accompanied by cre- tion of their catalytic activity [233,234]. Immobilization using the
ating shock waves that cause strong shear formation of gas filled CLEA technique combines purification and immobilization into sin-
bubbles [216]. The subsequent collapse of these bubbles imparts gle unit operation. Therefore it is possible to isolate an enzyme in
shock wave and mechanical shear to the surrounding environ- its immobilized form directly from a fermentation broth or any nat-
ment. The change in local energy densities and mechanical shears ural sources [229,230]. During the CLEAs preparation, the effects of
incurred due to cavitational phenomenon accelerates the mass precipitating agent, concentration, cross-linkers concentration and
transfer across the different phases [217]. In ultrasound assisted cross-linking time are need to be studied to get highly active, robust
TPP (UATPP), ultrasound power, duty cycle and irradiation time and stable biocatalyst [235]. Yusof et al. prepared CLEAs of seven
are critical operating parameters to be considered while extrac- hydrolase enzymes (amylase, mannanase, glucosidase, glucanase,
tion of biomolecules [218,219]. Avhad et al. extracted fibrinolytic protease, lipase and fructosyltransferase) obtained from cocoa pod
enzyme from Bacillus sphaericus by using ultrasound coupled TPP husk [236]. Fazary et al. prepared cross-linked feruloyl esterase
system. UATPP showed increase in purity and activity recovery of aggregate (CLEA) from Aspergillus awamori which were thermally
fibrinolytic enzyme by 7-fold and 4-fold respectively, as compared stable in the range of 25–85 ◦ C as compared to native form [237].
to conventional TPP [220]. Pakhale et al. purified serratiopeptidase Most of the cases, glutaraldehyde has been used as a cross-linker
from Serratia marcescens NRRL B 23112 with UATPP. The process in the preparation of CLEAs. However, it can’t be employed for all
time for UATPP was significantly reduced to 5 min from 60 min enzymes, especially sensitive to it. The small size of glutaraldehyde
as compared to conventional TPP. The overall results showed that cross-linker gets easily penetrated into the enzyme active site and
the UATPP is an effective technique for the purification of serra- reacting with catalytically active amino acids residues leading to
tiopeptidase with maximum purity, activity recovery and reduced enzyme inactivation. Moreover, cross linking of enzyme aggregates
processing time [191]. These stories of success highlight the advan- by glutaraldehyde forms compact and supramolecular structures
tage of ultrasound technique with TPP for the establishment of easy hindering the active sites of enzyme [238,239]. To overcome these,
and simple processes to implement which is attractive from an Nadar et al. prepared CLEAs by precipitating crude enzyme and
economic viewpoint. subsequently cross-linked them by biocompatible, biodegradable,
non-toxic, renewable and macromolecular polysaccharide based
cross-linker viz. agar, chitosan, dextran, pectin and gum arabic to
7. Simultaneous purification and immobilization strategies get highly active CLEAs [240,241]. However, the CLEAs preparation
is inadequate for all enzymes since crosslinking largely requires
Apart from the inherent advantages of enzyme, the industrial amino groups in the form of Lys residues on the external surface of
applicability of enzyme is always obstructed due to insufficient the enzyme. Hence, it may lead to low mechanical stability or even
stability in operational conditions, difficulties in recovery from releasing enzyme into the reaction media during reaction [242].
reaction mixture and reusability for multiple cycles [6,221,222]. Recently, another approach was used to prepare CLEAs in pres-
Also, the use of commercial enzymes in soluble form not only ence of magnetic nanoparticle called as a magnetic CLEAs. It
leads to the waste but also it contaminates the product, ultimately was simply prepared by addition of amino functionalized mag-
increasing the purification cost of desired product. These hur- netite nanoparticles as an additive into enzyme (with low Lys
dles can be prevailed by the enzyme immobilization tools [223]. residue content) solution, which further precipitate enzyme to
The immobilization greatly enhances the properties of enzymes form aggregates and then cross-linking of enzyme aggregates and
in terms of thermal stability, tolerance to extremely high pH and nanoparticles [243]. The prepared magnetic CLEAs showed high
organic solvents, selectivity and activity to meet the demands of thermal, mechanical stable and non-leachable CLEAs due to suf-
practical uses. It also facilitates the efficient recovery and re-use of ficient cross linking of enzyme aggregates [244]. These magnetic
the enzyme, thus enabling its cost-effective use and thereby mak- nanoparticles not only provide the large surface area for enzyme
S.S. Nadar et al. / International Journal of Biological Macromolecules 101 (2017) 931–957 949

Fig. 7. Schematic diagram illustrating preparation of cross-linked enzyme aggregates.

anchoring but also provide higher operational stability. Addition- techniques applied for the selective recovery and purification of
ally, magnetic nanoparticles help to ease recovery from reaction biomolecules could possibly hinder ATPS application at industrial
mixture by using the external magnetic field, instead of using tradi- levels. The development and application of ATPS is limited due
tional filtration and centrifugation [245,246]. Talekar et al. prepared to two major restrictions. Firstly, the limited predictive process
the magnetic CLEAs of crude alpha amylase which retained almost design due to the poor understanding and lack of the knowledge of
100% residual activity even after six cycles of reusability in batch the responsible mechanisms for the behaviour of enzymes in ATPS
mode [242]. In addition to that, CLEAs have also been cross-linked and interactions solute–solvent between target product and con-
to magnetic mesoporous silica particles and it maintained its initial taminants. Secondly, monitoring the characteristics of proteins is
activity even after 35 days of storage [247]. the basic requirement for assessment of bioprocesses, which are
Recently, construction of organic–inorganic hybrid material is affected frequently by the presence of different polymers or salts.
a rapidly expanding field of material chemistry for its advanced Considerable time is needed to build first recovery process of the
designs with a specific structure and functionality [248,249]. When experiment, while big budget is needed for installation and limited
crude enzyme is mixed with metal ion solution, it binds specifically output of the purification units. Compared with a novel alcohol/salt
with enzyme which initiate synthesis of hybrid nanoflowers of that ATPS, polymer/polymer, polymer/salt and surfactant/alcohol ATPS
enzyme. This would result in simultaneous purification and immo- systems have some disadvantages such as the high cost, viscos-
bilization of enzyme into hybrid organic–inorganic nanoflowers. ity, ionic strength as well as slow segregation (Table 9). There are
The binding of a ligand to one site (which we designate as the difficulties in isolating the extracted enzymes from the polymer
allosteric site) on a protein molecule indirectly manipulates the phase as well as from micellar phase by re-extraction and envi-
properties of another specific site (the functional site and in ronmental pollution resulting from the recycling of phase-forming
enzymes called the active site) on the same enzyme as a con- polymers. Hence, the ATPS is expected to overcome the drawbacks
sequence of conformational changes. Organic–inorganic hybrid and challenges by rapid evolution theoretically and experimentally.
nanoflowers were prepared by using copper ions as the inor- The modification and improvement in ATPSs such as free
ganic component and enzyme as the organic component (Fig. 8). affinity ligands or using modified or activated copolymers are
Recently, in the work of Zare et al. a simple approach was devel- more effective due to the high efficiency and selective separa-
oped for fabrication of enzyme–inorganic hybrid nanoflowers by tion and purification which ultimately increasing recovery yields
using HRP as the organic component and Cu3 (PO4 )2 ·3H2 O as the of biomolecules. The novel type of ATPS combined with other tech-
inorganic component [250]. In a typical experiment, the aqueous niques such as chromatography which might help intensifying the
CuSO4 solution was added to phosphate buffered saline (PBS) con- capability of existing ATPS for recovering biocatalyst at commercial
taining enzyme. The copper phosphate nano-crystals and enzymes scale. Also, the use of 3-D mapping for analysing protein pro-
were self-assembled to form the high surface area hierarchical files allows the identification of the molecular properties from the
nanoflowers embedded with enzymes [251]. Nadar et al. synthe- main protein contaminants. This information will facilitate estab-
sized hybrid glucoamylase nanoflowers which exhibited excellent lishment of concentration and specific purification conditions for
reusability and reproducibility in cycle analysis [252]. In another enzymes. Further, experimental design can be used as a strategy
study, lactoperoxidase was purified from bovine milk and simul- to screen and optimise the purification process conditions, allow-
taneously immobilized by copper metal ions (Cu2+ ) via a greener ing a fast assessment of the different experimental parameters
approach to form nanoflowers. This resulted in enhancement in involved as well as their possible interactions in the optimal ATPS
catalytic activity and stability [226]. system for partitioning of desired biocatalyst. In the last few years,
In a nutshell, the advantages provide by CLEAs technology and some studies focused on the development of excellent polymers
hybrid nanoflowers, the simultaneous purification and immobiliza- forming ATPS for higher recovering. The development of smart
tion strategies exhibit a great potential in industrial application, polymers which by responding to a change in temperature (PNB ) or
following greener approach. pH (PADB ) or light (PNBC ) are able to change conformation, thereby
allowing the recovery of enzyme of interest along with the poly-
mer in opposite phases. Another breakthrough could arise from
8. Future perspectives and challenges the integration of functional magnetic particles into ATPS, which
could speed up phase separation and enhance process throughput.
To fulfil the demand of a growing market of industrial enzymes, Extractive fermentation and purification is one of the emerging
the development of new and innovative downstream methods for continues extraction strategies containing in situ product recov-
purification of catalytically active enzymes is of great importance. ery and simultaneously performed fermentation, separation and
The main evaluation between these techniques determined their purification of enzymes instantaneously. Moreover, the simulta-
efficiency in terms of purity and yield for a specific product, and the
economic aspects of each. The well-established chromatographic
950 S.S. Nadar et al. / International Journal of Biological Macromolecules 101 (2017) 931–957

Fig. 8. Schematic diagram illustrating preparation of organic-inorganic hybrid nanoflowers.

Table 9
comparison of different types of ATPS.

Type Advantages Disadvantages

Polymer/polymer ATPS Easily amenable and modifiable High viscosity


Polymer/salt ATPS Lower viscosity and less time consuming High ionic strength
Alcohol/salt ATPS Low cost, lower viscosity, less time consuming Denaturation of labile enzyme, high volatility
and recycling of alcohol of alcohol
Surfactant/alcohol ATPS Recycling of alcohol, low viscosity Denaturation of labile enzyme
Aqueous affinity two phase system High resolution technique, high selectivity Time consuming, relative complexity within
activation process
Ionic liquid based ATPS Low viscosity, Non-flammability, strong High cost
solubilizing power, high thermal stability
Thermo separating polymer based ATPS Simplicity of product recovery, cheap chemical Denaturation of heat labile enzyme
cost and environmental friendliness, easy
recycling of polymers
Aqueous micellar two phase system High selectivity and recycling of surfactant High cost and not compatible to thermo
sensitive enzyme
Aqueous two-phase flotation High concentration coefficient, soft separation, Denaturation of heat labile and sheer sensitive
low dosage of organic solvent, simple enzyme
operation and low environmental impact

neous immobilization and purification of enzymes make it a great CLEAs and organic–inorganic nanoflowers preparation are of high
impact on down streaming and various industrial applications. potential.

Acknowledgement
9. Conclusion
The authors would like to acknowledge the University Grants
Commission (UGC) of India for financial assistance to the research
An efficient extraction/purification technology needs to offer
work.
several benefits includes less time-consuming operations, lower
consumption of energy and resources and lower labour costs. Over
the last decades, ATPS has been reported as an attractive alterna- References
tive or platform in downstream processing that can be adopted by
industries due to its high capacity and easy scalability. This review [1] J. Hagen, Industrial Catalysis: A Practical Approach, Wiley-VCH, 2006.
[2] V. Buchholz, Biocatalysts and Enzyme Technology, Wiley, 2012.
presented parameters that contribute to the establishment of effi- [3] A.A. Homaei, R. Sariri, F. Vianello, R. Stevanato, Enzyme immobilization: an
cient ATPS processes to get highly purified enzyme in active forms. update, J. Chem. Biol. 6 (2013) 185–205, http://dx.doi.org/10.1007/s12154-
Also, the modifications of ATPS and process integration for aug- 013-0102-9.
[4] U. Hanefeld, L. Cao, E. Magner, Enzyme immobilisation: fundamentals and
menting recovery and purification of enzymes were highlighted. application, Chem. Soc. Rev. 42 (2013) 6211, http://dx.doi.org/10.1039/
Although, the potential of ATPS for the recovery of catalytically c3cs90042h.
active enzyme has been proven, the technique has not been widely [5] A.S. Hiltrud Lenke, BIOTECHNOLOGY—Vol. IV Biotransformations,
Encyclopedia of Life Support Systems, Eolss Publ, 2009.
exploited at a commercial scale. Another attempt has been made
[6] R.A. Sheldon, S. van Pelt, Enzyme immobilisation in biocatalysis: why, what
to develop a simple, more efficient and economical method for and how, Chem. Soc. Rev. 42 (2013) 6223–6235, http://dx.doi.org/10.1039/
separation and purification of enzyme molecules by three-phase C3CS60075K.
[7] K.R. Jegannathan, P.H. Nielsen, Environmental assessment of enzyme use in
partitioning. The rapid, economic and single pot purification meth-
industrial production—a literature review, J. Clean. Prod. 42 (2013) 228–240,
ods have good potential to purify enzymes with high catalytic http://dx.doi.org/10.1016/j.jclepro.2012.11.005.
activity. An effective means to decrease the enzyme cost is reduc- [8] O. Kirk, T.V. Borchert, C.C. Fuglsang, Industrial enzyme applications, Curr.
tion of its separation and purification cost, which can be achieved Opin. Biotechnol. 13 (2002) 345–351, http://dx.doi.org/10.1016/S0958-
1669(02)00328-2.
by integrated purification and immobilization techniques. So as [9] K.S.M.S. Raghavarao, N.K. Rastogi, M.K. Gowthaman, N.G. Karanth, Aqueous
discussed in this review article, there are few strategies such as two-phase extraction for downstream processing of enzymes/proteins, Adv.
S.S. Nadar et al. / International Journal of Biological Macromolecules 101 (2017) 931–957 951

Appl. Microbiol. 41 (1995) 97–171, http://dx.doi.org/10.1016/S0065- [32] B. Mattiasson, R. Kaul, Use of aqueous two-phase systems for recovery and
2164(08)70309-5. purification in biotechnology, ACS Symp. Ser. (1986) 78–92, http://dx.doi.
[10] T.M. Przybycien, N.S. Pujar, L.M. Steele, Alternative bioseparation org/10.1021/bk-1986-0314.ch007.
operations: life beyond packed-bed chromatography, Curr. Opin. Biotechnol. [33] B.A. Andrews, J.A. Asenjo, Theoretical and experimental evaluation of
15 (2004) 469–478, http://dx.doi.org/10.1016/j.copbio.2004.08.008. hydrophobicity of proteins to predict their partitioning behavior in aqueous
[11] R.M. Banik, A. Santhiagu, B. Kanari, Technological aspects of extractive two phase systems: a review, Sep. Sci. Technol. 45 (2010) 2165–2170,
fermentation using aqueous two-phase systems, World. J. Microbiol. http://dx.doi.org/10.1080/01496395.2010.507436.
Biotechnol. 19 (2003) 337–348, http://dx.doi.org/10.1023/ [34] G. Tubio, B. Nerli, G. Picó, Relationship between the protein surface
A:1023940809095. hydrophobicity and its partitioning behaviour in aqueous two-phase
[12] O. Aguilar, M. Rito-Palomares, Aqueous two-phase systems strategies for the systems of polyethyleneglycol-dextran, J. Chromatogr. B Anal. Technol.
recovery and characterization of biological products from plants, J. Sci. Food Biomed. Life Sci. 799 (2004) 293–301, http://dx.doi.org/10.1016/j.jchromb.
Agric. 90 (2010) 1385–1392, http://dx.doi.org/10.1002/jsfa.3956. 2003.10.060.
[13] H.E. Schoemaker, D. Mink, M.G. Wubbolts, Dispelling the [35] J. Raghava Rao, B.U. Nair, Novel approach towards recovery of
myths—biocatalysis in industrial synthesis, Science (2003) 299 (80-.). glycosaminoglycans from tannery wastewater, Bioresour. Technol. 102
[14] A.M. Azevedo, P.A.J. Rosa, I.F. Ferreira, M.R. Aires-Barros, (2011) 872–878, http://dx.doi.org/10.1016/j.biortech.2010.09.013.
Chromatography-free recovery of biopharmaceuticals through aqueous [36] D. Forciniti, C.K. Hall, M.-R. Kula, Influence of polymer molecular weight and
two-phase processing, Trends Biotechnol. 27 (2009) 240–247, http://dx.doi. temperature on phase composition in aqueous two-phase systems, Fluid
org/10.1016/j.tibtech.2009.01.004. Phase Equilib. 61 (1991) 243–262, http://dx.doi.org/10.1016/0378-
[15] H. Yang, A.M. Goja, M. Cui, C. Li, Aqueous two-phase extraction advances for 3812(91)80002-D.
bioseparation, J. Bioprocess. Biotech. 44172 (2013) 2155–9821, http://dx. [37] T. Karkas, Characteristics of invertase partitioned in poly(ethylene
doi.org/10.4172/2155-9821.1000140. glycol)/magnesium sulfate aqueous two-phase system, Biochem. Eng. J. 60
[16] P.A. Albertsson, Partition of Cell Particles and Macromolecules: Separation (2012) 142–150, http://dx.doi.org/10.1016/j.bej.2011.11.005.
and Purification of Biomolecules, Cell Organelles, Membranes, and Cells in [38] A.M. Targovnik, O. Cascone, M.V. Miranda, Extractive purification of
Aqueous Polymer Two-phase Systems and Their Use in Biochemical Analysis recombinant peroxidase isozyme c from insect larvae in aqueous two-phase
and Biotechnology, Wiley, 1986. systems, Sep. Purif. Technol. 98 (2012) 199–205, http://dx.doi.org/10.1016/j.
[17] H.S. Mohammadi, E. Omidinia, Process integration for the recovery and seppur.2012.08.004.
purification of recombinant Pseudomonas fluorescens proline [39] P. Taylor, M. Anvari, Extraction of lipase from Rhizopus microsporus
dehydrogenase using aqueous two-phase systems, J. Chromatogr. B Anal. fermentation culture by aqueous two-phase partitioning, Biotechnol.
Technol. Biomed. Life Sci. 929 (2013) 11–17, http://dx.doi.org/10.1016/j. Biotechnol. Equip. (2015), http://dx.doi.org/10.1080/13102818.2015.
jchromb.2013.03.024. 1042406.
[18] B. Neelwarne, Red beet biotechnology: food and pharmaceutical [40] A. Kianmehr, M. Pooraskari, B. Mousavikoodehi, S.S. Mostafavi, Recombinant
applications, red beet biotechnol, Food Pharm. Appl. (2012) 1–435, http:// d-galactose dehydrogenase partitioning in aqueous two-phase systems:
dx.doi.org/10.1007/978-1-4614-3458-0. effect of pH and concentration of PEG and ammonium sulfate, Bioresour.
[19] J. Benavides, M. Rito-Palomares, Practical experiences from the Bioprocess. (2014) 1–8.
development of aqueous two-phase processes for the recovery of high value [41] A.B. Hemavathi, K.S.M.S. Raghavarao, Differential partitioning of
biological products, J. Chem. Technol. Biotechnol. 83 (2008) 133–142, http:// ␤-galactosidase and ␤-glucosidase using aqueous two phase extraction,
dx.doi.org/10.1002/jctb.1844. Process Biochem. 46 (2011) 649–655, http://dx.doi.org/10.1016/j.procbio.
[20] B.A. Andrews, A.S. Schmidt, J.A. Asenjo, Correlation for the partition 2010.11.008.
behavior of proteins in aqueous two-phase systems: effect of surface [42] Y. Yuzugullu, Y.A. Duman, Aqueous two-phase (PEG4000/Na2SO4)
hydrophobicity and charge, Biotechnol. Bioeng. 90 (2005) 380–390, http:// extraction and characterization of an acid invertase from potato tuber
dx.doi.org/10.1002/bit.20495. (Solanum tuberosum), Prep. Biochem. Biotechnol. 45 (2015) 696–711,
[21] J.C. Salgado, B.A. Andrews, M.F. Ortuzar, J.A. Asenjo, Prediction of the http://dx.doi.org/10.1080/10826068.2014.943373.
partitioning behaviour of proteins in aqueous two-phase systems using only [43] J.M.P. Barbosa, R.L. Souza, A.T. Fricks, G.M. Zanin, C.M.F. Soares, Á.S. Lima,
their amino acid composition, J. Chromatogr. A 1178 (2008) 134–144, http:// Purification of lipase produced by a new source of Bacillus in submerged
dx.doi.org/10.1016/j.chroma.2007.11.064. fermentation using an aqueous two-phase system, J. Chromatogr. B Anal.
[22] A. Glyk, T. Scheper, S. Beutel, PEG?salt aqueous two-phase systems: an Technol. Biomed. Life Sci. 879 (2011) 3853–3858, http://dx.doi.org/10.1016/
attractive and versatile liquid–liquid extraction technology for the j.jchromb.2011.10.035.
downstream processing of proteins and enzymes, Appl. Microbiol. [44] D. Aradhana, H.P. Sreeja, G. Sharmila, C. Muthukumaran, Optimization of
Biotechnol. 99 (2015) 6599–6616, http://dx.doi.org/10.1007/s00253-015- Rhizopus niveus lipase partitioning by an aqueous biphasic system, Chem.
6779-7. Eng. Technol. 37 (2014) 1191–1197, http://dx.doi.org/10.1002/ceat.
[23] J. Benavides, O. Aguilar, B. Lapizco-Encinas, M. Rito-Palomares, Extraction 201300652.
and purification of bioproducts and nanoparticles using aqueous two-phase [45] J.A. Asenjo, B.A. Andrews, Aqueous two-phase systems for protein
systems strategies, Chem. Eng. Technol. 31 (2008) 838–845, http://dx.doi. separation: a perspective, J. Chromatogr. A 1218 (2011) 8826–8835, http://
org/10.1002/ceat.200800068. dx.doi.org/10.1016/j.chroma.2011.06.051.
[24] S. Saravanan, J.R. Rao, B.U. Nair, T. Ramasami, Aqueous two-phase [46] G. Picó, D. Romanini, B. Nerli, B. Farruggia, Polyethyleneglycol molecular
poly(ethylene glycol)-poly(acrylic acid) system for protein partitioning: mass and polydispersivity effect on protein partitioning in aqueous
influence of molecular weight, pH and temperature, Process Biochem. 43 two-phase systems, J. Chromatogr. B Anal. Technol. Biomed. Life Sci. 830
(2008) 905–911, http://dx.doi.org/10.1016/j.procbio.2008.04.011. (2006) 286–292, http://dx.doi.org/10.1016/j.jchromb.2005.11.008.
[25] P.A.J. Rosa, I.F. Ferreira, A.M. Azevedo, M.R. Aires-Barros, Aqueous two-phase [47] B. Kavakcıoğlu, B. Tongul, L. Tarhan, Aqueous two-phase system purification
systems: a viable platform in the manufacturing of biopharmaceuticals, J. for superoxide dismutase induced by menadione from Phanerochaete
Chromatogr. A 1217 (2010) 2296–2305, http://dx.doi.org/10.1016/j.chroma. chrysosporium, Artif. Cells Nanomed. Biotechnol. 2016 (1401) 1–9, http://dx.
2009.11.034. doi.org/10.3109/21691401.2016.1160404.
[26] R. Hatti-kaul, Aqueous two-phase systems, Mol. Biotechnol. 19 (2001) [48] S. Kirinčič, C. Klofutar, Viscosity of aqueous solutions of poly(ethylene
697–713, http://dx.doi.org/10.1385/MB:19:3:269. glycol)s at 298.15 K, Fluid Phase Equilib. 155 (1999) 311–325, http://dx.doi.
[27] S. Klomklao, S. Benjakul, W. Visessanguan, B.K. Simpson, H. Kishimura, org/10.1016/S0378-3812(99)00005-9.
Partitioning and recovery of proteinase from tuna spleen by aqueous [49] K. Ratanapongleka, Partitioning behavior of laccase from Lentinus
two-phase systems, Process Biochem. 40 (2005) 3061–3067, http://dx.doi. polychrous lev in aqueous two phase systems, Songklanakarin J. Sci.
org/10.1016/j.procbio.2005.03.009. Technol. 34 (2012) 69–76.
[28] T.S. Porto, G.M. Medeiros e Silva, C.S. Porto, M.T.H. Cavalcanti, B.B. Neto, J.L. [50] A. Mehrnoush, S. Mustafa, A.M.M. Yazid, Heat-treatment aqueous two phase
Lima-Filho, A. Converti, A.L.F. Porto, A. Pessoa, Liquid–liquid extraction of system for purification of serine protease from kesinai (Streblus asper)
proteases from fermented broth by PEG/citrate aqueous two-phase system, leaves, Molecules 16 (2011) 10202–10213, http://dx.doi.org/10.3390/
Chem. Eng. Process. Process Intensif. 47 (2008) 716–721, http://dx.doi.org/ molecules161210202.
10.1016/j.cep.2006.12.004. [51] M.G. Antov, D.Ž. Ivetić, Z.D. Knežević Jugović, Single step recovery of lipase
[29] F. Rahimpour, R. Hatti-Kaul, G. Mamo, Response surface methodology and from Penicillium cyclopium by aqueous two-phase extraction, Sep. Sci.
artificial neural network modelling of an aqueous two-phase system for Technol. 51 (2016) 622–628, http://dx.doi.org/10.1080/01496395.2015.
purification of a recombinant alkaline active xylanase, Process Biochem. 51 1117106.
(2016) 452–462, http://dx.doi.org/10.1016/j.procbio.2015.12.018. [52] B. Kavakcıoğlu, B. Tongul, L. Tarhan, Aqueous two-phase system purification
[30] I. Yücekan, S. Önal, Partitioning of invertase from tomato in poly(ethylene for superoxide dismutase induced by menadione from Phanerochaete
glycol)/sodium sulfate aqueous two-phase systems, Process Biochem. 46 chrysosporium, Artif. Cells Nanomed. Biotechnol. (2016) 1–9, http://dx.doi.
(2011) 226–232, http://dx.doi.org/10.1016/j.procbio.2010.08.015. org/10.3109/21691401.2016.1160404.
[31] K. Berggren, H.-O. Johansson, F. Yjerneld, Effects of salts and the surface [53] P.A.J. Rosa, A.M. Azevedo, M.R. Aires-Barros, Application of central
hydrophobicity of proteins on partitioning in aqueous two-phase systems composite design to the optimisation of aqueous two-phase extraction of
containing thermoseparating ethylene oxide-propylene oxide copolymers, J. human antibodies, J. Chromatogr. A 1217 (2007) 50–60, http://dx.doi.org/
Chromatogr. A 718 (1995) 67–79, http://dx.doi.org/10.1016/0021- 10.1016/j.chroma.2006.11.075.
9673(95)00657-5. [54] A.M. Azevedo, P.A.J. Rosa, I.F. Ferreira, J. de Vries, T.J. Visser, M.R.
Aires-Barros, Downstream processing of human antibodies integrating an
952 S.S. Nadar et al. / International Journal of Biological Macromolecules 101 (2017) 931–957

extraction capture step and cation exchange chromatography, J. system for amyloglucosidase purification: equilibrium diagrams and
Chromatogr. B 877 (2009) 50–58, http://dx.doi.org/10.1016/j.jchromb.2008. partitioning studies, Sep. Purif. Technol. 118 (2013) 888–894, http://dx.doi.
11.014. org/10.1016/j.seppur.2013.08.039.
[55] G.R. Pazuki, V. Taghikhani, M. Vossoughi, Prediction of the partition [77] B. Bolognese, B. Nerli, G. Picó, Application of the aqueous two-phase systems
coefficients of biomolecules in polymer–polymer aqueous two-phase of ethylene and propylene oxide copolymer-maltodextrin for protein
systems using the artificial neural network model, Part. Sci. Technol. 28 purification, J. Chromatogr. B Anal. Technol. Biomed. Life Sci. 814 (2005)
(2010) 67–73, http://dx.doi.org/10.1080/02726350903408175. 347–353, http://dx.doi.org/10.1016/j.jchromb.2004.10.057.
[56] K.M. Desai, S.A. Survase, P.S. Saudagar, S.S. Lele, R.S. Singhal, Comparison of [78] H.S. Mohamadi, E. Omidinia, R. Dinarvand, Evaluation of recombinant
artificial neural network (ANN) and response surface methodology (RSM) in phenylalanine dehydrogenase behavior in aqueous two-phase partitioning,
fermentation media optimization: case study of fermentative production of Process Biochem. 42 (2007) 1296–1301, http://dx.doi.org/10.1016/j.procbio.
scleroglucan, Biochem. Eng. J. 41 (2008) 266–273, http://dx.doi.org/10.1016/ 2007.06.005.
j.bej.2008.05.009. [79] S. Bolar, R. Iyyaswami, P.D. Belur, Purification of glutaminase from
[57] P.K. Srivastava, M. Kapoor, Extracellular endo-mannanase from Bacillus sp. Zygosaccharomyces rouxii in polyethylene glycol-Sodium sulphate aqueous
CFR1601: economical production using response surface methodology and two-phase system, Sep. Sci. Technol. 50 (2015) 1360–1368, http://dx.doi.
downstream processing using aqueous two phase system, Food Bioprod. org/10.1080/01496395.2014.967410.
Process. 91 (2013) 672–681, http://dx.doi.org/10.1016/j.fbp.2013.06.006. [80] R.P. Bezerra, F.K.S.L. Borba, K.A. Moreira, J.L. Lima-Filho, A.L.F. Porto, A.C.
[58] K.S. Vijayaragavan, A. Zahid, J.W. Young, C.L. Heldt, Separation of porcine Chaves, Extraction of amylase from fermentation broth in poly (ethylene
parvovirus from bovine serum albumin using PEG-salt aqueous two-phase glycol) salt aqueous two-phase system, Braz. Arch. Biol. Technol. 49 (2006)
system, J. Chromatogr. B Anal. Technol Biomed. Life Sci. 967 (2014) 547–555.
118–126, http://dx.doi.org/10.1016/j.jchromb.2014.07.025. [81] N.D. Srinivas, R.S. Barhate, K.S.M.S. Raghavarao, Aqueous two-phase
[59] K. Nandini, N. Rastogi, Liquid–liquid extraction of lipase using aqueous extraction in combination with ultrafiltration for downstream processing of
two-phase system, Food, Bioprocess Technol. (2011), http://dx.doi.org/10. Ipomoea peroxidase, J. Food Eng. 54 (2002) 1–6, http://dx.doi.org/10.1016/
1007/s11947-008-0160-0 (Accessed 24 September 2016). S0260-8774(01)00170-4.
[60] B.S. Priyanka, N.K. Rastogi, K.S.M.S. Raghavarao, M.S. Thakur, Downstream [82] Y. Liu, Z. Wu, Y. Zhang, H. Yuan, Partitioning of biomolecules in aqueous
processing of luciferase from fireflies (Photinus pyralis) using aqueous two-phase systems of polyethylene glycol and nonionic surfactant, Biochem.
two-phase extraction, Process Biochem. 47 (2012) 1358–1363, http://dx.doi. Eng. J. 69 (2012) 93–99, http://dx.doi.org/10.1016/j.bej.2012.08.018.
org/10.1016/j.procbio.2012.04.022. [83] M. Estela da Silva, T. Teixeira Franco, Purification of soybean peroxidase
[61] M.C. Madhusudhan, K.S.M.S. Raghavarao, Aqueous two phase extraction of (Glycine max) by metal affinity partitioning in aqueous two-phase systems,
invertase from baker’s yeast: effect of process parameters on partitioning, J. Chromatogr. B Biomed. Sci. Appl. 743 (2000) 287–294, http://dx.doi.org/
Process Biochem. 46 (2011) 2014–2020, http://dx.doi.org/10.1016/j.procbio. 10.1016/S0378-4347(00)00172-9.
2011.07.014. [84] G.B. Ferreira, A.F. Evangelista, J.B. Severo Junior, R.R. de Souza, J.C.C. Santana,
[62] M. Amid, M. Shuhaimi, M.Z. Islam Sarker, M.Y. Abdul Manap, Purification of E.B. Tambourgi, E. Jordão, Partitioning optimization of proteins from Zea
serine protease from mango (Mangifera Indica Cv. Chokanan) peel using an mays malt in ATPS PEG 6000/CaCl2, Braz. Arch. Biol. Technol. 50 (2007)
alcohol/salt aqueous two phase system, Food Chem. 132 (2012) 1382–1386, 557–564, http://dx.doi.org/10.1590/S1516-89132007000300020.
http://dx.doi.org/10.1016/j.foodchem.2011.11.125. [85] S. Chethana, C.A. Nayak, K.S.M.S. Raghavarao, Aqueous two phase extraction
[63] J. Huddleston, A. Veide, K. Köhler, J. Flanagan, S.-O. Enfors, A. Lyddiatt, The for purification and concentration of betalains, J. Food Eng. 81 (2007)
molecular basis of partitioning in aqueous two-phase systems, Trends 679–687, http://dx.doi.org/10.1016/j.jfoodeng.2006.12.021.
Biotechnol. 9 (1991) 381–388, http://dx.doi.org/10.1016/0167- [86] S. Mohamed Ali, T.C. Ling, S. Muniandy, Y.S. Tan, J. Raman, V. Sabaratnam,
7799(91)90130-A. Recovery and partial purification of fibrinolytic enzymes of Auricularia
[64] S. Bolar, P.D. Belur, R. Iyyaswami, Partitioning studies of glutaminase in polytricha (Mont.) Sacc by an aqueous two-phase system, Sep. Purif.
polyethylene glycol and salt-based aqueous two-phase systems, Chem. Eng. Technol. 122 (2014) 359–366, http://dx.doi.org/10.1016/j.seppur.2013.11.
Technol. 36 (2013) 1378–1386, http://dx.doi.org/10.1002/ceat.201200677. 016.
[65] X.-Y. Zhao, F. Qu, M. Dong, F. Chen, A.-Q. Luo, J.-H. Zhang, Separation of [87] F. Hachem, B.A. Andrews, J.A. Asenjo, Hydrophobic partitioning of proteins
proteins by aqueous two-phase extraction system combined with liquid in aqueous two-phase systems, Enzyme Microb. Technol. 19 (1996)
chromatography, Chin. J. Anal. Chem. 40 (2012) 38–42, http://dx.doi.org/10. 507–517, http://dx.doi.org/10.1016/S0141-0229(96)80002-D.
1016/S1872-2040(11)60520-3. [88] A.M. Azevedo, P.A.J. Rosa, I.F. Ferreira, M.R. Aires-Barros, Optimisation of
[66] B.R. Babu, N.K. Rastogi, K.S.M.S. Raghavarao, Liquid-liquid extraction of aqueous two-phase extraction of human antibodies, J. Biotechnol. 132
bromelain and polyphenol oxidase using aqueous two-phase system, Chem. (2007) 209–217, http://dx.doi.org/10.1016/j.jbiotec.2007.04.002.
Eng. Process. Process Intensif. 47 (2008) 83–89, http://dx.doi.org/10.1016/j. [89] R. Gulati, R. Saxena, R. Gupta, Fermentation and downstream processing of
cep.2007.08.006. lipase from Aspergillus terreus, Process Biochem. 36 (2000) 149–155, http://
[67] Y.M. Lu, Y.Z. Yang, X.D. Zhao, C.B. Xia, Bovine serum albumin partitioning in dx.doi.org/10.1016/S0032-9592(00)00201-6.
polyethylene glycol (PEG)/potassium citrate aqueous two-phase systems, [90] P.L. Show, C.P. Tan, M.S. Anuar, A. Ariff, Y.A. Yusof, S.K. Chen, T.C. Ling,
Food Bioprod. Process. 88 (2010) 40–46, http://dx.doi.org/10.1016/j.fbp. Primary recovery of lipase derived from Burkholderia cenocepacia strain
2009.12.002. ST8 and recycling of phase components in an aqueous two-phase system,
[68] G.G.G. Oliveira, D.P. Silva, I.C. Roberto, M. Vitolo, A. Pessoa, Partition Biochem. Eng. J. 60 (2012) 74–80, http://dx.doi.org/10.1016/j.bej.2011.10.
behavior and partial purification of hexokinase in aqueous two-phase 005.
polyethylene glycol/citrate systems, Appl. Biochem. Biotechnol. 105–108 [91] C. Kepka, J. Rhodin, R. Lemmens, F. Tjerneld, P.-E. Gustavsson, Extraction of
(2003) 787–798, http://dx.doi.org/10.1385/abab:108:1-3:787. plasmid DNA from Escherichia coli cell lysate in a thermoseparating
[69] A. Mehrnoush, S. Mustafa, M.Z.I. Sarker, A.M.M. Yazid, Optimization of aqueous two-phase system, J. Chromatogr. A 1217 (2004) 95–104, http://dx.
serine protease purification from mango (mangifera indica cv. chokanan) doi.org/10.1016/j.chroma.2003.10.028.
peel in polyethylene glycol/dextran aqueous two phase system, Int. J. Mol. [92] R. Dembczyński, W. Białas, K. Regulski, T. Jankowski, Lysozyme extraction
Sci. 13 (2012) 3636–3649, http://dx.doi.org/10.3390/ijms13033636. from hen egg white in an aqueous two-phase system composed of ethylene
[70] P. Singh, R.M. Banik, Partitioning studies of l-glutaminase production by oxide-propylene oxide thermoseparating copolymer and potassium
Bacillus cereus MTCC 1305 in different PEG-salt/dextran, Bioresour. Technol. phosphate, Process Biochem. 45 (2010) 369–374, http://dx.doi.org/10.1016/
114 (2012) 730–734, http://dx.doi.org/10.1016/j.biortech.2012.03.046. j.procbio.2009.10.011.
[71] S.K. Pandey, R.M. Banik, Extractive fermentation for enhanced production of [93] J. Persson, L. Nyström, H. Ageland, F. Tjerneld, Purification of recombinant
alkaline phosphatase from Bacillus licheniformis MTCC 1483 using aqueous apolipoprotein A-1Milano expressed in Escherichia coli using aqueous
two-phase systems, Bioresour. Technol. 102 (2011) 4226–4231, http://dx. two-phase extraction followed by temperature-induced phase separation, J.
doi.org/10.1016/j.biortech.2010.12.066. Chromatogr. B Biomed. Sci. Appl. 711 (1998) 97–109, http://dx.doi.org/10.
[72] S. Raja, V.R. Murty, V. Thivaharan, V. Rajasekar, V. Ramesh, Aqueous two 1016/S0378-4347(98)00029-2.
phase systems for the recovery of biomolecules—a review, Sci. Technol. 1 [94] Z.-G. Li, Y.-Q. Sun, W.-L. Zheng, H. Teng, Z.-L. Xiu, A novel and
(2012) 7–16, http://dx.doi.org/10.5923/j.scit.20110101.02. environment-friendly bioprocess of 1,3-propanediol fermentation
[73] C.W. Ooi, S.L. Hii, S.M.M. Kamal, A. Ariff, T.C. Ling, Extractive fermentation integrated with aqueous two-phase extraction by ethanol/sodium
using aqueous two-phase systems for integrated production and purification carbonate system, Biochem. Eng. J. 80 (2013) 68–75, http://dx.doi.org/10.
of extracellular lipase derived from Burkholderia pseudomallei, Process 1016/j.bej.2013.09.014.
Biochem. 46 (2011) 68–73, http://dx.doi.org/10.1016/j.procbio.2010.07.014. [95] M. Moradi, Y. Yamini, Surfactant roles in modern sample preparation
[74] M.G. Antov, D.M. Peri, S.N. Pejin, Pectinases partitioning in aqueous techniques: a review, J. Sep. Sci. 35 (2012) 2319–2340, http://dx.doi.org/10.
two-phase systems: an integration of the systems poly (ethylene 1002/jssc.201200368.
glycol)/crude dextran and poly (ethylene glycol)/ammonium sulphate, J. [96] Z.J. Tan, C.Y. Wang, Y.J. Yi, H.Y. Wang, W.L. Zhou, S.Y. Tan, F.F. Li, Three phase
Serb. Chem. Soc. 69 (2004) 299–307. partitioning for simultaneous purification of aloe polysaccharide and
[75] A.W.F. Duarte, A.M. Lopes, J.V.D. Molino, A. Pessoa, L.D. Sette, Liquid–liquid protein using a single-step extraction, Process Biochem. 50 (2015) 482–486,
extraction of lipase produced by psychrotrophic yeast Leucosporidium http://dx.doi.org/10.1016/j.procbio.2015.01.004.
scottii L117 using aqueous two-phase systems, Sep. Purif. Technol. (2015) [97] S.C. Silvério, O. Rodríguez, A.P.M. Tavares, J.A. Teixeira, E.A. Macedo, Laccase
1–11, http://dx.doi.org/10.1016/j.seppur.2015.10.001. recovery with aqueous two-phase systems: enzyme partitioning and
[76] L.A. Pereira Alcântara, K.S. Do Nascimento, C.A. Mourão, V.P.R. Minim, L.A. stability, J. Mol. Catal. B Enzym. 87 (2013) 37–43, http://dx.doi.org/10.1016/
Minim, Aqueous two-phase poly(ethylene glycol)-sodium polyacrylate j.molcatb.2012.10.010.
S.S. Nadar et al. / International Journal of Biological Macromolecules 101 (2017) 931–957 953

[98] Y.K. Yau, C.W. Ooi, E.-P. Ng, J.C.-W. Lan, T.C. Ling, P.L. Show, Current [119] M.R. Gavasane, V.G. Gaikar, Aqueous two-phase affinity partitioning of
applications of different type of aqueous two-phase systems, Bioresour. penicillin acylase from E. coli in presence of PEG-derivatives, Enzyme
Bioprocess. 2 (2015) 49, http://dx.doi.org/10.1186/s40643-015-0078-0. Microb. Technol. 32 (2003) 665–675, http://dx.doi.org/10.1016/S0141-
[99] Y. Guan, T.H. Lilley, T.E. Treffry, C.-L. Zhou, P.B. Wilkinson, Use of aqueous 0229(03)00032-2.
two-phase systems in the purification of human interferon-␣1 from [120] S.D. Flanagan, S.H. Barondes, Affinity partitioning. A method for purification
recombinant Escherichia coli, Enzyme Microb. Technol. 19 (1996) 446–455, of proteins using specific polymer-ligands in aqueous polymer two-phase
http://dx.doi.org/10.1016/S0141-0229(96)00051-8. systems, J. Biol. Chem. 250 (1975) 1484–1489, http://www.ncbi.nlm.nih.
[100] R.C.F. Bonomo, L.A. Minim, J.S.R. Coimbra, R.C.I. Fontan, L.H. Mendes da Silva, gov/pubmed/1112812 (Accessed 29 September 2016).
V.P.R. Minim, Hydrophobic interaction adsorption of whey proteins: effect [121] P.A.J. Rosa, A.M. Azevedo, I.F. Ferreira, J. de Vries, R. Korporaal, H.J. Verhoef,
of temperature and salt concentration and thermodynamic analysis, J. T.J. Visser, M.R. Aires-Barros, Affinity partitioning of human antibodies in
Chromatogr. B 844 (2006) 6–14, http://dx.doi.org/10.1016/j.jchromb.2006. aqueous two-phase systems, J. Chromatogr. A 1162 (2007) 103–113, http://
06.021. dx.doi.org/10.1016/j.chroma.2007.03.067.
[101] W. Zhi, Q. Deng, Purification of salvianolic acid B from the crude extract of [122] Z.Q. Lin, D.Q. Yao, S.J. Mei, L.H. Zhu, Preparation of Iminodiacetic
Salvia miltiorrhiza with hydrophilic organic/salt-containing aqueous Acid-Polyethylene Glycol for Immobilized Metal Ion Affinity Partitioning,
two-phase system by counter-current chromatography, J. Chromatogr. A
1116 (2006) 149–152, http://dx.doi.org/10.1016/j.chroma.2006.03.036. . 8 (2000) 310–314.
[102] T. Tianwei, H. Qing, L. Qiang, Purification of glycyrrhizin from Glycyrrhiza [123] S.B. Dhoot, J.M. Dalal, V.G. Gaikar, Purification of glucose oxidase and
uralensis Fisch with ethanol/phosphate aqueous two phase system, ␤-galactosidase by partitioning in a PEG-salt aqueous two-phase system in
Biotechnol. Lett. (2002), the presence of PEG-derivatives, Sep. Sci. Technol. 42 (2007) 1859–1881,
http://link.springer.com/article/10.1023/A:1019850531640 (accessed http://dx.doi.org/10.1080/01496390701310355.
September 27, 2016). [124] S. Sharma, M.N. Gupta, Purification of phospholipase D from Dacus carota by
[103] C.W. Ooi, B.T. Tey, S.L. Hii, S.M.M. Kamal, J.C.W. Lan, A. Ariff, T.C. Ling, three-phase partitioning and its characterization, Protein Expression Purif.
Purification of lipase derived from Burkholderia pseudomallei with 21 (2001) 10–6316, http://dx.doi.org/10.1006/prep.2000.1357.
alcohol/salt-based aqueous two-phase systems, Process Biochem. 44 (2009) [125] S. Teotia, R. Lata, M.N. Gupta, Chitosan as a macroaffinity ligand: purification
1083–1087, http://dx.doi.org/10.1016/j.procbio.2009.05.008. of chitinases by affinity precipitation and aqueous two-phase extractions, J.
[104] J. Simental-Martínez, B. Montalvo-Hernández, M. Rito-Palomares, J. Chromatogr. A 1052 (2004) 85–91, http://dx.doi.org/10.1016/j.chroma.2004.
Benavides, Application of aqueous two-phase systems for the recovery of 08.096.
bioactive low-molecular weight compounds, Sep. Sci. Technol. 49 (2014) [126] Y. Xu, M.A. Souza, M.Z.R. Pontes, M. Vitolo, A. Pessoa, Liquid–liquid
1872–1882, http://dx.doi.org/10.1080/01496395.2014.904878. extraction of enzymes by affinity aqueous two-phase systems, Braz. Arch.
[105] J. Simental-Martínez, M. Rito-Palomares, J. Benavides, Potential application Biol. Technol. 46 (2003) 741–750, http://dx.doi.org/10.1590/S1516-
of aqueous two-phase systems and three-phase partitioning for the 89132003000400030.
recovery of superoxide dismutase from a clarified homogenate of K [127] S. Rathnasamy, R. Kumaresan, Integration and investigation of metal ligands
luyveromyces marxianus, Biotechnol. Prog. 30 (2014) 1326–1334, http://dx. in aqueous two phase extraction and its partition coefficient studies in
doi.org/10.1002/btpr.1979. papain extraction, Indian J. Biotechnol. 12 (2013) 225–230.
[106] R.B. Haga, V.C. Santos-Ebinuma, M. de Siqueira Cardoso Silva, A. Pessoa, C.O. [128] S. Rathnasamy, R. Kumaresan, Partition coefficient studies in integrated
Rangel-Yagui, Clavulanic acid partitioning in charged aqueous two-phase aqueous two phase (ATPi) extraction with free tri-azine dye ligands for
micellar systems, Sep. Purif. Technol. 103 (2013) 273–278, http://dx.doi.org/ papain partitioning from its crude latex, Res. J. Biotechnol. 9 (2014) 66–77.
10.1016/j.seppur.2012.10.034. [129] M.G. Freire, A.F.M. Cláudio, J.M.M. Araújo, J.A.P. Coutinho, I.M. Marrucho,
[107] A.M. Lopes, V.C. Santos-Ebinuma, A. Pessoa Júnior, C.O. Rangel-Yagui, J.N.C. Lopes, L.P.N. Rebelo, Aqueous biphasic systems: a boost brought about
Influence of salts on the coexistence curve and protein partitioning in by using ionic liquids, Chem. Soc. Rev. 41 (2012) 4966, http://dx.doi.org/10.
nonionic aqueous two-phase micellar systems, Braz. J. Chem. Eng. 31 (2014) 1039/c2cs35151j.
1057–1064, http://dx.doi.org/10.1590/0104-6632.20140314s00002677. [130] Z. Du, Y.-L. Yu, J.-H. Wang, Extraction of proteins from biological fluids by
[108] E.A. Ermakova, N.L. Zakhartchenko, Y.F. Zuev, Effect of surface potential of use of an ionic liquid/aqueous two-phase system, Chemistry 13 (2007)
reverse micelle on enzyme-substrate complex formation, Colloids Surf. A 2130–2137, http://dx.doi.org/10.1002/chem.200601234.
317 (2008) 297–302, http://dx.doi.org/10.1016/j.colsurfa.2007.10.027. [131] Z. Li, Y. Pei, L. Liu, J. Wang, (Liquid + liquid) equilibria for (acetate-based ionic
[109] H. Zhong, G. ming Zeng, X.Z. Yuan, H. yan Fu, G.H. Huang, F.Y. Ren, liquids + inorganic salts) aqueous two-phase systems, J. Chem. Thermodyn.
Adsorption of dirhamnolipid on four microorganisms and the effect on cell 42 (2010) 932–937, http://dx.doi.org/10.1016/j.jct.2010.03.010.
surface hydrophobicity, Appl. Microbiol. Biotechnol. 77 (2007) 447–455, [132] Y. Lu, W. Lu, W. Wang, Q. Guo, Y. Yang, Thermodynamic studies of
http://dx.doi.org/10.1007/s00253-007-1154-y. partitioning behavior of cytochrome c in ionic liquid-based aqueous
[110] U. Sivars, F. Tjerneld, Mechanisms of phase behaviour and protein two-phase system, Talanta 85 (2011) 1621–1626, http://dx.doi.org/10.1016/
partitioning in detergent/polymer aqueous two-phase systems for j.talanta.2011.06.058.
purification of integral membrane proteins, Biochim. Biophys. Acta 2000 [133] C.L.S. Louros, A.F.M. Cláudio, Neves C.M.S.S, M.G. Freire, I.M. Marrucho, J.
(1474) 133–146, http://dx.doi.org/10.1016/S0304-4165(99)00208-1. Pauly, J.A.P. Coutinho, Extraction of biomolecules using phosphonium-based
[111] N. Chen, J.-B. Fan, J. Xiang, J. Chen, Y. Liang, Enzymatic hydrolysis of ionic liquids + K3PO4 aqueous biphasic systems, Int. J. Mol. Sci. 11 (2010)
microcrystalline cellulose in reverse micelles, Biochim. Biophys. Acta 2006 1777–1791, http://dx.doi.org/10.3390/ijms11041777.
(1764) 1029–1035, http://dx.doi.org/10.1016/j.bbapap.2006.03.015. [134] C.M.S.S. Neves, S.P.M. Ventura, M.G. Freire, I.M. Marrucho, J.A.P. Coutinho,
[112] M. Amid, M.Y. Abd Manap, M. Shuhaimi, Purification of a novel protease Evaluation of cation influence on the formation and extraction capability of
enzyme from kesinai plant (Streblus asper) leaves using a surfactant-salt ionic-liquid-based aqueous biphasic systems, J. Phys. Chem. B 113 (2009)
aqueous micellar two-phase system: a potential low cost source of enzyme 5194–5199, http://dx.doi.org/10.1021/jp900293v.
and purification method, Eur. Food Res. Technol. 237 (2013) 601–608, [135] Y. Pei, Z. Li, L. Liu, J. Wang, H. Wang, Selective separation of protein and
http://dx.doi.org/10.1007/s00217-013-2037-3. saccharides by ionic liquids aqueous two-phase systems, Sci. China Chem.
[113] C.H. Tan, P.L. Show, C.W. Ooi, E.P. Ng, J.C.W. Lan, T.C. Ling, Novel lipase 53 (2010) 1554–1560, http://dx.doi.org/10.1007/s11426-010-4025-9.
purification methods—a review of the latest developments, Biotechnol. J. 10 [136] Y. Pei, J. Wang, K. Wu, X. Xuan, X. Lu, Ionic liquid-based aqueous two-phase
(2015) 31–44, http://dx.doi.org/10.1002/biot.201400301. extraction of selected proteins, Sep. Purif. Technol. 64 (2009) 288–295,
[114] M. Amid, M.Y. Manap, M. Hussin, S. Mustafa, A novel aqueous two phase http://dx.doi.org/10.1016/j.seppur.2008.10.010.
system composed of surfactant and xylitol for the purification of lipase from [137] Joana R. Trindade, Zoran P. Visak, Marijana Blesi, Isabel M. Marrucho, João
pumpkin (Cucurbita moschata) seeds and recycling of phase components, A.P. Coutinho, José N. Canongia Lopes, Luis P.N. Rebelo, Salting-out effects in
Molecules 20 (2015) 11184–11201, http://dx.doi.org/10.3390/ aqueous ionic liquid solutions: cloud-point temperature shifts, J. Phys.
molecules200611184. Chem. B 11 (2007) 4737–4741, http://dx.doi.org/10.1021/JP067022D.
[115] M. Amid, F.A. Asmadi, M. Hussin, M.Y. Manap, M.Z. Islam Sarker, C.G. Hean, A [138] S. Oppermann, F. Stein, U. Kragl, Ionic liquids for two-phase systems and
novel aqueous micellar two-phase system composed of surfactant and their application for purification, extraction and biocatalysis, Appl.
mannitol for purification of polygalacturonase enzyme from Durio Microbiol. Biotechnol. 89 (2011) 493–499, http://dx.doi.org/10.1007/
zibethinus Murray and recycling phase components, Sep. Sci. Technol. 51 s00253-010-2933-4.
(2016) 968–975, http://dx.doi.org/10.1080/01496395.2016.1142562. [139] M. Moniruzzaman, K. Nakashima, N. Kamiya, M. Goto, Recent advances of
[116] F.A. Vicente, L.D. Lario, A. Pessoa, S.P.M. Ventura, Recovery of bromelain enzymatic reactions in ionic liquids, Biochem. Eng. J. 48 (2010) 295–314,
from pineapple stem residues using aqueous micellar two-phase systems http://dx.doi.org/10.1016/j.bej.2009.10.002.
with ionic liquids as co-surfactants, Process Biochem. (2015), http://dx.doi. [140] S.P.M. Ventura, R.L.F. de Barros, J.M. de Pinho Barbosa, C.M.F. Soares, S. Á.
org/10.1016/j.procbio.2016.01.004. Lima, et al., Coutinho, Production and purification of an extracellular
[117] F. Ruiz-Ruiz, J. Benavides, O. Aguilar, M. Rito-Palomares, Aqueous two-phase lipolytic enzyme using ionic liquid-based aqueous two-phase systems,
affinity partitioning systems: current applications and trends, J. Chromatogr. Green Chem. 14 (2012) 734, http://dx.doi.org/10.1039/c2gc16428k.
A 1244 (2012) 1–13, http://dx.doi.org/10.1016/j.chroma.2012.04.077. [141] F.J. Deive, A. Rodríguez, A.B. Pereiro, J.M.M. Araújo, M.A. Longo, M.A.Z.
[118] A. Cordes, J. Flossdorf, M.-R. Kula, Affinity partitioning: development of Coelho, J.N.C. Lopes, J.M.S.S. Esperança, et al., Rebelo, ionic liquid-based
mathematical model describing behavior of biomolecules in aqueous aqueous biphasic system for lipase extraction, Green Chem. 13 (2011)
two-phase systems, Biotechnol. Bioeng. 30 (1987) 514–520, http://dx.doi. 390–396, http://dx.doi.org/10.1039/C0GC00075B.
org/10.1002/bit.260300408. [142] S.P.M. Ventura, S.G. Sousa, M.G. Freire, L.S. Serafim, S. Á. Lima, J.A.P.
Coutinho, Design of ionic liquids for lipase purification, J. Chromatogr. B
954 S.S. Nadar et al. / International Journal of Biological Macromolecules 101 (2017) 931–957

Anal. Technol. Biomed. Life Sci. 879 (2011) 2679–2687, http://dx.doi.org/10. purification, Biotechnol. Bioeng. 66 (1999) 247–257, http://dx.doi.org/10.
1016/j.jchromb.2011.07.022. 1002/(SICI)1097-0290(1999)66:4<247:AID-BIT6>3.0.CO;2-5.
[143] F.J. Deive, A. Rodríguez, L.P.N. Rebelo, I.M. Marrucho, Extraction of Candida [164] M.T. Cunha, F. Tjerneld, J.M. Cabral, Maria Raquel Aires-Barros, Effect of
antarctica lipase A from aqueous solutions using imidazolium-based ionic electrolytes and surfactants on the thermoseparation of an ethylene
liquids, Sep. Purif. Technol. 97 (2012) 205–210, http://dx.doi.org/10.1016/j. oxide–propylene oxide random copolymer in aqueous solution, J.
seppur.2011.12.013. Chromatogr. B Biomed. Sci. Appl. 711 (1998) 53–60, http://dx.doi.org/10.
[144] S. Dreyer, U. Kragl, Ionic liquids for aqueous two-phase extraction and 1016/S0378-4347(98)00021-8.
stabilization of enzymes, Biotechnol. Bioeng. 99 (2008) 1416–1424, http:// [165] R. Dembczynski, W. Bialas, T. Jankowski, Recycling of phase components
dx.doi.org/10.1002/bit.21720. during lysozyme extraction from hen egg white in the EO50PO50/K2HPO4
[145] Q. Shang, W. Li, Q. Jia, D. Li, Partitioning behavior of amino acids in aqueous aqueous two-phase system, Biochem. Eng. J. 51 (2010) 24–31, http://dx.doi.
two-phase systems containing polyethylene glycol and phosphate buffer, org/10.1016/j.bej.2010.04.011.
Fluid Phase Equilib. 219 (2004) 195–203, http://dx.doi.org/10.1016/j.fluid. [166] C. Liu, D.T. Kamei, J.A. King, D.I. Wang, D. Blankschtein, Separation of
2004.01.032. proteins and viruses using two-phase aqueous micellar systems, J.
[146] E.A. Snellman, E.R. Sullivan, R.R. Colwell, Purification and properties of the Chromatogr. B Biomed. Sci. Appl. 711 (1998) 127–138, http://dx.doi.org/10.
extracellular lipase, LipA, of Acinetobacter sp. RAG-1, Eur. J. Biochem. 269 1016/S0378-4347(98)00013-9.
(2002) 5771–5779, http://dx.doi.org/10.1046/j.1432-1033.2002.03235.x. [167] F.H. Quina, W.L. Hinze, Surfactant-mediated cloud point extractions: an
[147] Y.K. Leong, J.C.W. Lan, H.S. Loh, T.C. Ling, C.W. Ooi, P.L. Show, environmentally benign alternative separation approach, Ind. Eng. Chem.
Thermoseparating aqueous two-phase systems: recent trends and Res. 38 (1999) 4150–4168, http://dx.doi.org/10.1021/IE980389N.
mechanisms, J. Sep. Sci. 39 (2016) 640–647, http://dx.doi.org/10.1002/jssc. [168] H. Tani, T. Kamidate, H. Watanabe, Aqueous micellar two-phase systems for
201500667. protein separation, Anal. Sci. 14 (1998) 875–888, http://dx.doi.org/10.2116/
[148] A.P.B. Rabelo, E.B. Tambourgi, A. Pessoa, Bromelain partitioning in two-phase analsci.14.875.
aqueous systems containing PEO-PPO-PEO block copolymers, J. Chromatogr. [169] L.P. Malpiedi, B.B. Nerli, D.S.P. Abdala, P. De Alcântara Pessôa-Filho, A.
B 807 (2004) 61–68, http://dx.doi.org/10.1016/j.jchromb.2004.03.029. Pessoa, Aqueous micellar systems containing Triton X-114 and Pichia
[149] A. Nilsson, H.-O. Johansson, M. Mannesse, M.R. Egmond, F. Tjerneld, pastoris fermentation supernatant: a novel alternative for single
Partitioning of peptide-tagged proteins in aqueous two-phase systems using chain-antibody fragment purification, Sep. Purif. Technol. 132 (2014)
hydrophobically modified micelle-forming thermoseparating polymer, 295–301, http://dx.doi.org/10.1016/j.seppur.2014.05.045.
Biochim. Biophys. Acta 2002 (1601) 138–148, http://dx.doi.org/10.1016/ [170] P.M. Duque Jaramillo, H.A. Rocha Gomes, F.G. De Siqueira, M.
S1570-9639(02)00462-4. Homem-De-Mello, E.X.F. Filho, P.O. Magalhães, Liquid–liquid extraction of
[150] M. Li, T.L. Peeples, Purification of hyperthermophilic archaeal amylolytic pectinase produced by Aspergillus oryzae using aqueous two-phase micellar
enzyme (MJA1) using thermoseparating aqueous two-phase systems, J. system, Sep. Purif. Technol. 120 (2013) 452–457, http://dx.doi.org/10.1016/
Chromatogr. B Anal. Technol. Biomed. Life Sci. 807 (2004) 69–74, http://dx. j.seppur.2013.09.020.
doi.org/10.1016/j.jchromb.2004.03.030. [171] C.W. Ooi, C.P. Tan, S.L. Hii, A. Ariff, S. Ibrahim, T.C. Ling, Primary recovery of
[151] S. Moreira, S.C. Silvério, E.A. Macedo, A.M.F. Milagres, J.A. Teixeira, S.I. lipase derived from Burkholderia sp. ST8 with aqueous micellar two-phase
Mussatto, Recovery of Peniophora cinerea laccase using aqueous two-phase system, Process Biochem. 46 (2011) 1847–1852, http://dx.doi.org/10.1016/j.
systems composed by ethylene oxide/propylene oxide copolymer and procbio.2011.06.014.
potassium phosphate salts, J. Chromatogr. A 1321 (2013) 14–20, http://dx. [172] W. Lee, P. Tseng, D. Di, Microtechnology for Cell Manipulation and Sorting,
doi.org/10.1016/j.chroma.2013.10.056. Microsystems and Nanosystems, Springer International Publishing, 2017,
[152] I.F. Ferreira, A.M. Azevedo, P.A.J. Rosa, M.R. Aires-Barros, Purification of http://dx.doi.org/10.1007/978-3-319-44139-9.
human immunoglobulin G by thermoseparating aqueous two-phase [173] R. Hu, X. Feng, P. Chen, M. Fu, H. Chen, L. Guo, B.F. Liu, Rapid, highly efficient
systems, J. Chromatogr. A 1195 (2008) 94–100, http://dx.doi.org/10.1016/j. extraction and purification of membrane proteins using a microfluidic
chroma.2008.04.077. continuous-flow based aqueous two-phase system, J. Chromatogr. A 1218
[153] S.C. Silvério, O. Rodríguez, A.P.M. Tavares, J.A. Teixeira, E.A. MacEdo, Laccase (2011) 171–177, http://dx.doi.org/10.1016/j.chroma.2010.10.090.
recovery with aqueous two-phase systems: enzyme partitioning and [174] S. Hardt, T. Hahn, Microfluidics with aqueous two-phase systems, Lab Chip
stability, J. Mol. Catal. B Enzym. 87 (2013) 37–43, http://dx.doi.org/10.1016/ (2011), http://dx.doi.org/10.1039/c1lc20569b.
j.molcatb.2012.10.010. [175] Y. Huang, T. Meng, T. Guo, W. Li, W. Yan, X. Li, S. Wang, Z. Tong, Aqueous
[154] F.A. Hasmann, V.C. Santos, D.B. Gurpilhares, A. Pessoa-Junior, I.C. Roberto, two-phase extraction for bovine serum albumin (BSA) with co-laminar flow
Aqueous two-phase extraction using thermoseparating copolymer: a new in a simple coaxial capillary microfluidic device, Microfluid. Nanofluid. 16
system for phenolic compounds removal from hemicelullosic hydrolysate, J. (2014) 483–491, http://dx.doi.org/10.1007/s10404-013-1245-2.
Chem. Technol. Biotechnol. 83 (2008) 167–173, http://dx.doi.org/10.1002/ [176] E. Espitia-Saloma, P. Vázquez-Villegas, O. Aguilar, M. Rito-Palomares,
jctb.1779. Continuous aqueous two-phase systems devices for the recovery of
[155] J. Persson, A. Kaul, F. Tjerneld, Polymer recycling in aqueous two-phase biological products, Food Bioprod. Process. 92 (2014) 101–112, http://dx.
extractions using thermoseparating ethylene oxide-propylene oxide doi.org/10.1016/j.fbp.2013.05.006.
copolymers, J. Chromatogr. B Biomed. Sci. Appl. 743 (2000) 115–126, http:// [177] R.R.G. Soares, D.F.C. Silva, P. Fernandes, A.M. Azevedo, V. Chu, J.P. Conde,
dx.doi.org/10.1016/s0378-4347(00)00213-9. M.R. Aires-Barros, Miniaturization of aqueous two-phase extraction for
[156] H.S. Ng, C.P. Tan, M.N. Mokhtar, S. Ibrahim, A. Ariff, C.W. Ooi, T.C. Ling, biological applications: from micro-tubes to microchannels, Biotechnol. J.
Recovery of Bacillus cereus cyclodextrin glycosyltransferase and recycling of (2016) 1–15, http://dx.doi.org/10.1002/biot.201600356.
phase components in an aqueous two-phase system using [178] M.P.C. Marques, P. Fernandes, Microfluidic devices: useful tools for
thermo-separating polymer, Sep. Purif. Technol. 89 (2012) 9–15, http://dx. bioprocess intensification, Molecules 16 (2011) 8368–8401, http://dx.doi.
doi.org/10.1016/j.seppur.2011.12.028. org/10.3390/molecules16108368.
[157] P.L. Show, C.P. Tan, M. Shamsul Anuar, A. Ariff, Y.A. Yusof, S.K. Chen, T.C. [179] U. Novak, M. Lakner, I. Plazl, P. Žnidaršič-Plazl, Experimental studies and
Ling, Extractive fermentation for improved production and recovery of modeling of ␣-amylase aqueous two-phase extraction within a microfluidic
lipase derived from Burkholderia cepacia using a thermoseparating polymer device, Microfluid. Nanofluid. 19 (2015) 75–83, http://dx.doi.org/10.1007/
in aqueous two-phase systems, Bioresour. Technol. 116 (2012) 226–233, s10404-015-1550-z.
http://dx.doi.org/10.1016/j.biortech.2011.09.131. [180] T. Hahn, G. Münchow, S. Hardt, Electrophoretic transport of biomolecules
[158] J.H. Zhu, X.L. Yan, H.J. Chen, Z.H. Wang, In situ extraction of intracellular across liquid–liquid interfaces, J. Phys. Condens. Matter 23 (2011) 184107,
l-asparaginase using thermoseparating aqueous two-phase systems, J. http://dx.doi.org/10.1088/0953-8984/23/27/279502.
Chromatogr. A 1147 (2007) 127–134, http://dx.doi.org/10.1016/j.chroma. [181] I. Roy, M.N. Gupta, Three-phase affinity partitioning of proteins, Anal.
2007.02.035. Biochem. 300 (2002) 11–14, http://dx.doi.org/10.1006/abio.2001.5367.
[159] R. Dembczyński, W. Białas, T. Jankowski, Partitioning of lysozyme in [182] N.N. Kurmudle, S.B. Bankar, I.B. Bajaj, M.V. Bule, R.S. Singhal,
aqueous two-phase systems containing ethylene oxide-propylene oxide Enzyme-assisted three phase partitioning: a novel approach for extraction
copolymer and potassium phosphates, Food Bioprod. Process. 91 (2013) of turmeric oleoresin, Process Biochem. 46 (2011) 423–426, http://dx.doi.
292–302, http://dx.doi.org/10.1016/j.fbp.2012.11.001. org/10.1016/j.procbio.2010.09.010.
[160] M. Jönsson, H.-O. Johansson, Protein partitioning in thermoseparating [183] H.H. Wang, C.L. Chen, T.L. Jeng, J.M. Sung, Comparisons of ␣-amylase
systems of a charged hydrophobically modified ethylene oxide polymer, J. inhibitors from seeds of common bean mutants extracted through three
Chromatogr. A 983 (2003) 133–144, http://dx.doi.org/10.1016/S0021- phase partitioning, Food Chem. 128 (2011) 1066–1071, http://dx.doi.org/10.
9673(02)01695-3. 1016/j.foodchem.2011.04.015.
[161] M. Li, J. Kim, T.L. Peeples, Amylase partitioning and extractive bioconversion [184] C. Dennison, R. Lovrien, Three phase partitioning: concentration and
of starch using thermoseparating aqueous two-phase systems, J. Biotechnol. purification of proteins, Protein Expr. Purif. 161 (1997) 149–161.
93 (2002) 15–26. [185] V.V. Kumar, V.S. Rapheal, Induction and purification by three-phase
[162] J. Persson, L. Nyström, H. Ageland, F. Tjerneld, Purification of recombinant partitioning of Aryl Alcohol Oxidase (AAO) from Pleurotus ostreatus, Appl.
proteins using thermoseparating aqueous two-phase system and polymer Biochem. Biotechnol. 163 (2011) 423–432, http://dx.doi.org/10.1007/
recycling, J. Chem. Technol. Biotechnol. 74 (1999) 238–243, http://dx.doi. s12010-010-9050-9.
org/10.1002/(SICI)1097-4660(199903)74:3<238:AID-JCTB29>3.0.CO;2-Z. [186] G.M. Rather, J. Mukherjee, P.J. Halling, M.N. Gupta, Activation of alpha
[163] H.-O. Johansson, J. Persson, F. Tjerneld, Thermoseparating water/polymer chymotrypsin by three phase partitioning is accompanied by aggregation,
system: a novel one-polymer aqueous two-phase system for protein PLoS One 7 (2012), http://dx.doi.org/10.1371/journal.pone.0049241.
S.S. Nadar et al. / International Journal of Biological Macromolecules 101 (2017) 931–957 955

[187] F. Gu, J. Gao, J. Xiao, Q. Chen, H. Ruan, G. He, Efficient methods of purification Technol. 118 (2013) 835–841, http://dx.doi.org/10.1016/j.seppur.2013.08.
of ␣-galactosidase from Aspergillus niger: aqueous two-phase system versus 040.
three-phase partitioning, Rom. Biotechnol. Lett. 17 (2012) 7853–7862. [212] V. Vinoth Kumar, M.P. Premkumar, V. Kumar Sathyaselvabala, S.
[188] A. Sharma, S. Sharma, M.N. Gupta, Purification of alkaline phosphatase from Dineshkirupha, J. Nandagopal, S. Sivanesan, Aspergillus niger exo-inulinase
chicken intestine by three-phase partitioning and use of phenyl-Sepharose purification by three phase partitioning, Eng. Life Sci. 11 (2011) 607–614,
6 B in the batch mode, Bioseparation (2000) 155–161. http://dx.doi.org/10.1002/elsc.201000180.
[189] S. Ketnawa, S. Benjakul, O. Martínez-Alvarez, S. Rawdkuen, Three-phase [213] Y. Liu, M. Yan, J. Huang, Three-phase partitioning for purification of laccase
partitioning and proteins hydrolysis patterns of alkaline proteases derived produced by coriolopsis trogii under solid fermentation, Am. J. Food
from fish viscera, Sep. Purif. Technol. 132 (2014) 174–181, http://dx.doi.org/ Technol. 10 (2015) 127–134, http://dx.doi.org/10.3923/ajft.2015.127.134.
10.1016/j.seppur.2014.05.006. [214] P.R. Gogate, A.M. Kabadi, A review of applications of cavitation in
[190] M.D. Vetal, V.K. Rathod, Three phase partitioning a novel technique for biochemical engineering/biotechnology, Biochem. Eng. J. 44 (2009) 60–72,
purification of peroxidase from orange peels (Citrus sinenses), Food Bioprod. http://dx.doi.org/10.1016/j.bej.2008.10.006.
Process. 94 (2015) 284–289, http://dx.doi.org/10.1016/j.fbp.2014.03.007. [215] S.S. Nadar, V.K. Rathod, Sonochemical effect on activity and conformation of
[191] S.V. Pakhale, S.S. Bhagwat, Purification of serratiopeptidase from Serratia commercial lipases, Appl. Biochem. Biotechnol. (2016) 1–19, http://dx.doi.
marcescens NRRL B 23112 using ultrasound assisted three phase org/10.1007/s12010-016-2294-2.
partitioning, Ultrason. Sonochem. 31 (2016) 532–538, http://dx.doi.org/10. [216] T.W. Charpe, V.K. Rathod, Extraction of glycyrrhizic acid from licorice root
1016/j.ultsonch.2016.01.037. using ultrasound: process intensification studies, Chem. Eng. Process.
[192] A. Şen, M. Eryilmaz, H. Bayraktar, S. Önal, Purification of ␣-galactosidase Process Intensif. 54 (2012) 37–41, http://dx.doi.org/10.1016/j.cep.2012.01.
from pepino (Solanum muricatum) by three-phase partitioning, Sep. Purif. 002.
Technol. 83 (2011) 130–136, http://dx.doi.org/10.1016/j.seppur.2011.09. [217] Y.X. Guo, J. Han, D.Y. Zhang, L.H. Wang, L.L. Zhou, An ammonium
026. sulfate/ethanol aqueous two-phase system combined with ultrasonication
[193] P.R. Rao, V.K. Rathod, Rapid extraction of andrographolide from for the separation and purification of lithospermic acid B from Salvia
Andrographis paniculata Nees by three phase partitioning and miltiorrhiza Bunge, Ultrason. Sonochem. 19 (2012) 719–724, http://dx.doi.
determination of its antioxidant activity, Biocatal. Agric. Biotechnol. 4 org/10.1016/j.ultsonch.2011.12.017.
(2015) 586–593, http://dx.doi.org/10.1016/j.bcab.2015.08.016. [218] S.M. Harde, R.S. Singhal, Extraction of forskolin from Coleus forskohlii roots
[194] M. Gagaoua, N. Hoggas, K. Hafid, Three phase partitioning of zingibain, a using three phase partitioning, Sep. Purif. Technol. 96 (2012) 20–25, http://
milk-clotting enzyme from Zingiber officinale Roscoe rhizomes, Int. J. Biol. dx.doi.org/10.1016/j.seppur.2012.05.017.
Macromol. (2014) 1–8, http://dx.doi.org/10.1016/j.ijbiomac.2014.10.069. [219] J.A. Chougle, R.S. Singhal, O.-D. Baik, Recovery of astaxanthin from
[195] Y. Duman, E. Kaya, Purification, recovery, and characterization of chick pea paracoccus NBRC 101723 using ultrasound-assisted three phase
(Cicer arietinum) ␤-galactosidase in single step by three phase partitioning partitioning (UA-TPP), Sep. Sci. Technol. 49 (2014) 811–818, http://dx.doi.
as a rapid and easy technique, Protein Expression Purif. 91 (2013) 155–160, org/10.1080/01496395.2013.872146.
http://dx.doi.org/10.1016/j.pep.2013.08.003. [220] D.N. Avhad, S.S. Niphadkar, V.K. Rathod, Ultrasound assisted three phase
[196] R. Garg, B.N. Thorat, Nattokinase purification by three phase partitioning partitioning of a fibrinolytic enzyme, Ultrason. Sonochem. 21 (2014)
and impact of t-butanol on freeze drying, Sep. Purif. Technol. 131 (2014) 628–633, http://dx.doi.org/10.1016/j.ultsonch.2013.10.002.
19–26, http://dx.doi.org/10.1016/j.seppur.2014.04.011. [221] R.A. Sheldon, Enzyme immobilization: the quest for optimum performance,
[197] B. Özer, E. Akardere, E.B. Çelem, S. Önal, Three-phase partitioning as a rapid Adv. Synth. Catal. 349 (2007) 1289–1307, http://dx.doi.org/10.1002/adsc.
and efficient method for purification of invertase from tomato, Biochem. 200700082.
Eng. J. 50 (2010) 110–115, http://dx.doi.org/10.1016/j.bej.2010.04.002. [222] E.T. Hwang, M.B. Gu, Enzyme stabilization by nano/microsized hybrid
[198] G.M. Rather, M.N. Gupta, Three phase partitioning leads to subtle structural materials, Eng. Life Sci. 13 (2013) 49–61, http://dx.doi.org/10.1002/elsc.
changes in proteins, Int. J. Biol. Macromol. 60 (2013) 134–140, http://dx.doi. 201100225.
org/10.1016/j.ijbiomac.2013.05.018. [223] D.N. Tran, K.J. Balkus, Perspective of recent progress in immobilization of
[199] A. Sharma, M.N. Gupta, Purification of pectinases by three-phase enzymes, ACS Catal. 1 (2011) 956–968, http://dx.doi.org/10.1021/
partitioning, Biotechnol. Lett. 23 (2001) 1625–1627, http://dx.doi.org/10. cs200124a.
1023/A:1011984517432. [224] C. Garcia-Galan, Á. Berenguer-Murcia, R. Fernandez-Lafuente, R.C.
[200] A.V. Narayan, M.C. Madhusudhan, K.S.M.S. Raghavarao, Extraction and Rodrigues, Potential of different enzyme immobilization strategies to
purification of ipomoea peroxidase employing three-phase partitioning, improve enzyme performance, Adv. Synth. Catal. 353 (2011) 2885–2904,
Appl. Biochem. Biotechnol. 151 (2008) 263–272, http://dx.doi.org/10.1007/ http://dx.doi.org/10.1002/adsc.201100534.
s12010-008-8185-4. [225] O. Barbosa, C. Ortiz, Á. Berenguer-Murcia, R. Torres, R.C. Rodrigues, R.
[201] R.K. Wati, T. Theppakorn, S. Benjakul, S. Rawdkuen, Three-phase Fernandez-Lafuente, Strategies for the one-step immobilization-purification
partitioning of trypsin inhibitor from legume seeds, Process Biochem. 44 of enzymes as industrial biocatalysts, Biotechnol. Adv. 33 (2015) 435–456,
(2009) 1307–1314, http://dx.doi.org/10.1016/j.procbio.2009.07.002. http://dx.doi.org/10.1016/j.biotechadv.2015.03.006.
[202] R.K. Singh, S. Gourinath, S. Sharma, I. Roy, M.N. Gupta, C. Betzel, A. [226] C. Altinkaynak, I. Yilmaz, Z. Koksal, H. Özdemir, I. Ocsoy, N. Özdemir,
Srinivasan, T.P. Singh, Enhancement of enzyme activity through three-phase Preparation of lactoperoxidase incorporated hybrid nanoflower and its
partitioning: crystal structure of a modified serine proteinase at 1.5 A excellent activity and stability, Int. J. Biol. Macromol. 84 (2016) 402–409,
resolution, Protein Eng. 14 (2001) 307–313, http://dx.doi.org/10.1093/ http://dx.doi.org/10.1016/j.ijbiomac.2015.12.018.
PROTEIN/14.5.307. [227] D.S. Raja, W.-L. Liu, H.-Y. Huang, C.-H. Lin, Immobilization of protein on
[203] Y.A. Duman, E. Kaya, Three-phase partitioning as a rapid and easy method nanoporous metal-organic framework materials, Comments Inorg. Chem. 35
for the purification and recovery of catalase from sweet potato tubers (2015) 332–350, http://dx.doi.org/10.1080/02603594.2015.1059827.
(Solanum tuberosum), Appl. Biochem. Biotechnol. 170 (2013) 1119–1126, [228] X. Wu, M. Hou, J. Ge, Metal–organic frameworks and inorganic nanoflowers:
http://dx.doi.org/10.1007/s12010-013-0260-9. a type of emerging inorganic crystal nanocarrier for enzyme immobilization,
[204] E. Akardere, B. Özer, E.B. Çelem, S. Önal, Three-phase partitioning of Catal. Sci. Technol. 5 (2015) 5077–5085, http://dx.doi.org/10.1039/
invertase from Baker’s yeast, Sep. Purif. Technol. 72 (2010) 335–339, http:// C5CY01181G.
dx.doi.org/10.1016/j.seppur.2010.02.025. [229] L. Cao, L. van Langen, R.A. Sheldon, Immobilised enzymes: carrier-bound or
[205] E. Çalci, T. Demir, E. Biçak Çelem, S. Önal, Purification of tomato carrier-free? Curr. Opin. Biotechnol. 14 (2003) 387–394, http://dx.doi.org/
(Lycopersicon esculentum) ␣-galactosidase by three-phase partitioning and 10.1016/S0958-1669(03)00096-X.
its characterization, Sep. Purif. Technol. 70 (2009) 123–127, http://dx.doi. [230] S. Talekar, A. Pandharbale, M. Ladole, S. Nadar, M. Mulla, K. Japhalekar, K.
org/10.1016/j.seppur.2009.09.004. Pattankude, D. Arage, Carrier free co-immobilization of alpha amylase,
[206] S.K. Dhananjay, V.H. Mulimani, Purification of ␣-galactosidase and invertase glucoamylase and pullulanase as combined cross-linked enzyme aggregates
by three-phase partitioning from crude extract of Aspergillus oryzae, (combi-cleas): a tri-enzyme biocatalyst with one pot starch hydrolytic
Biotechnol. Lett. 30 (2008) 1565–1569, http://dx.doi.org/10.1007/s10529- activity, Bioresour. Technol. 147 (2013) 269–275, http://dx.doi.org/10.1016/
008-9722-5. j.biortech.2013.08.035.
[207] S. Rawdkuen, P. Chaiwut, P. Pintathong, S. Benjakul, Three-phase [231] S. Talekar, A. Joshi, G. Joshi, P. Kamat, R. Haripurkar, S. Kambale, Parameters
partitioning of protease from Calotropis procera latex, Biochem. Eng. J. 50 in preparation and characterization of cross linked enzyme aggregates
(2010) 145–149, http://dx.doi.org/10.1016/j.bej.2010.04.007. (CLEAs), RSC Adv. 3 (2013) 12485–12511, http://dx.doi.org/10.1039/
[208] P. Chaiwut, P. Pintathong, S. Rawdkuen, Extraction and three-phase c3ra40818c.
partitioning behavior of proteases from papaya peels, Process Biochem. 45 [232] R.A. Sheldon, Cross-linked enzyme aggregates as industrial biocatalysts, Org.
(2010) 1172–1175, http://dx.doi.org/10.1016/j.procbio.2010.03.019. Process Res. Dev. 15 (2011) 213–223, http://dx.doi.org/10.1021/op100289f.
[209] M. Shanmugaprakash, V. Vinothkumar, J. Ragupathy, D.A. Reddy, [233] R. a Sheldon, Cross-linked enzyme aggregates (CLEAs): stable and recyclable
Biochemical characterization of three phase partitioned naringinase from biocatalysts, Biochem. Soc. Trans. 35 (2007) 1583–1587, http://dx.doi.org/
Aspergillus brasiliensis MTCC 1344, Int. J. Biol. Macromol. 80 (2015) 10.1042/BST0351583.
418–423, http://dx.doi.org/10.1016/j.ijbiomac.2015.06.057. [234] S. Talekar, G. Joshi, R. Chougle, B. Nainegali, S. Desai, A. Joshi, S. Kambale, P.
[210] A. Sharma, M.N. Gupta, Three phase partitioning as a large-scale separation Kamat, R. Haripurkar, S. Jadhav, S. Nadar, Preparation of stable cross-linked
method for purification of a wheat germ bifunctional protease/amylase enzyme aggregates (CLEAs) of NADH-dependent nitrate reductase and its
inhibitor, Process Biochem. 37 (2001) 193–196. use for silver nanoparticle synthesis from silver nitrate, Catal. Commun. 53
[211] H. Bayraktar, S. Önal, Concentration and purification of ␣-galactosidase from (2014) 62–66, http://dx.doi.org/10.1016/j.catcom.2014.05.003.
watermelon (Citrullus vulgaris) by three phase partitioning, Sep. Purif.
956 S.S. Nadar et al. / International Journal of Biological Macromolecules 101 (2017) 931–957

[235] S. Talekar, S. Desai, M. Pillai, N. Nagavekar, S. Ambarkar, S. Surnis, M. Ladole, fruiting bodies in aqueous two-phase system, J. Biosci. Bioeng. xx (2015)
S. Nadar, M. Mulla, Carrier free co-immobilization of glucoamylase and 6–11, http://dx.doi.org/10.1016/j.jbiosc.2016.01.016.
pullulanase as combi-cross linked enzyme aggregates (combi-CLEAs), RSC [260] G. Khayati, M. Anvari, N. Shahidi, Partitioning of b-galactosidase in aqueous
Adv. 3 (2013) 2265–2271, http://dx.doi.org/10.1039/C2RA22657J. two-phase systems containing polyethyleneglycol and phosphate salts,
[236] F. Yusof, Cocoa pod husk, a new source of hydrolase enzymes for Fluid Phase Equilib. 385 (2015) 147–152, http://dx.doi.org/10.1016/j.fluid.
preparation of cross - linked enzyme aggregate, SpringerPlus 5 (2016) 1–20. 2014.11.003.
[237] A.E. Fazary, S. Ismadji, Y.H. Ju, Biochemical studies on native and cross-linked [261] S.S. Niphadkar, M.D. Vetal, V.K. Rathod, Purification and characterization of
aggregates of Aspergillus awamori feruloyl esterase, Int. J. Biol. Macromol. polyphenol oxidase from waste potato peel by aqueous two phase
44 (2009) 240–248, http://dx.doi.org/10.1016/j.ijbiomac.2008.12.012. extraction, Prep. Biochem. Biotechnol. 6068 (2014) 37–41, http://dx.doi.org/
[238] C. Mateo, J.M. Palomo, L.M. Van Langen, F. Van Rantwijk, R.A. Sheldon, A. 10.1080/10826068.2014.940970.
New, Mild cross-linking methodology to prepare cross-linked enzyme [262] B.K. Vaidya, H.K. Suthar, S. Kasture, S. Nene, Purification of potato polyphenol
aggregates, Biotechnol. Bioeng. 86 (2004) 273–276, http://dx.doi.org/10. oxidase (PPO) by partitioning in aqueous two-phase system, Biochem. Eng. J.
1002/bit.20033. 28 (2006) 161–166, http://dx.doi.org/10.1016/j.bej.2005.10.006.
[239] Q. Zhen, M. Wang, W. Qi, R. Su, Z. He, Preparation of ␤-mannanase CLEAs [263] X. Chen, G. Xu, X. Li, Z. Li, H. Ying, Purification of an ␣-amylase inhibitor in a
using macromolecular cross-linkers, Catal. Sci. Technol. 3 (2013) 1937, polyethylene glycol/fructose-1,6-bisphosphate trisodium salt aqueous
http://dx.doi.org/10.1039/c3cy20886a. two-phase system, Process Biochem. 43 (2008) 765–768, http://dx.doi.org/
[240] S. Talekar, S. Nadar, A. Joshi, G. Joshi, Pectin cross-linked enzyme aggregates 10.1016/j.procbio.2008.03.003.
(pectin-CLEAs) of glucoamylase, RSC Adv. 4 (2014) 59444–59453, http://dx. [264] K. Naganagouda, V.H. Mulimani, Aqueous two-phase extraction (ATPE): An
doi.org/10.1039/C4RA09552A. attractive and economically viable technology for downstream processing of
[241] S.S. Nadar, A.B. Muley, M.R. Ladole, P.U. Joshi, Macromolecular cross-linked Aspergillus oryzae ␣-galactosidase, Process Biochem. 43 (2008) 1293–1299,
enzyme aggregates (M-CLEAs) of ␣-amylase, Int. J. Biol. Macromol. 84 http://dx.doi.org/10.1016/j.procbio.2008.07.016.
(2016) 69–78, http://dx.doi.org/10.1016/j.ijbiomac.2015.11.082. [265] M. Amid, M.Y. Abdul Manap, S. Mustafa, Purification of pectinase from
[242] S. Talekar, V. Ghodake, T. Ghotage, P. Rathod, P. Deshmukh, S. Nadar, M. mango (Mangifera indica L. cv. Chokanan) waste using an aqueous organic
Mulla, M. Ladole, Novel magnetic cross-linked enzyme aggregates (magnetic phase system: a potential low cost source of the enzyme, J. Chromatogr. B
CLEAs) of alpha amylase, Bioresour. Technol. 123 (2012) 542–547. Anal. Technol. Biomed. Life Sci. 931 (2013) 17–22, http://dx.doi.org/10.1016/
[243] A. Bhattacharya, B.I. Pletschke, Magnetic cross-linked enzyme aggregates j.jchromb.2013.05.009.
(CLEAs): a novel concept towards carrier free immobilization of [266] F.A. Vicente, L.P. Malpiedi, F.A.E. Silva, A. Pessoa, J.A.P. Coutinho, S.P.M.
lignocellulolytic enzymes, Enzyme Microb. Technol. 61–62 (2014) 17–27, Ventura, Design of novel aqueous micellar two-phase systems using ionic
http://dx.doi.org/10.1016/j.enzmictec.2014.04.009. liquids as co-surfactants for the selective extraction of (bio)molecules, Sep.
[244] S.S. Nadar, V.K. Rathod, Magnetic macromolecular cross linked enzyme Purif. Technol. 135 (2014) 259–267, http://dx.doi.org/10.1016/j.seppur.
aggregates (CLEAs) of glucoamylase, Enzyme Microb. Technol. 83 (2016) 2014.06.045.
78–87, http://dx.doi.org/10.1016/j.enzmictec.2015.10.009. [267] M. Lu, F. Tjerneld, Interaction between tryptophan residues and
[245] Á. Cruz-Izquierdo, E.A. Picó, C. López, J.L. Serra, M.J. Llama, Magnetic hydrophobically modified dextran: effect on partitioning of peptides and
Cross-Linked Enzyme Aggregates (mCLEAs) of Candida antarctica lipase: an proteins in aqueous two-phase systems, J. Chromatogr. A 766 (1997)
efficient and stable biocatalyst for biodiesel synthesis, PLoS One 9 (2014) 99–108, http://dx.doi.org/10.1016/S0021-9673(96)00994-6.
e115202, http://dx.doi.org/10.1371/journal.pone.0115202. [268] T. de Gouveia, B.V. Kilikian, Bioaffinity extraction of glucoamylase in
[246] U.V. Sojitra, S.S. Nadar, V.K. Rathod, A magnetic tri-enzyme nanobiocatalyst aqueous two-phase systems using starch as free bioligand, J. Chromatogr. B
for fruit juice clarification, Food Chem. 213 (2016) 296–305, http://dx.doi. Biomed. Sci. Appl. 743 (2000) 241–246, http://dx.doi.org/10.1016/S0378-
org/10.1016/j.foodchem.2016.06.074. 4347(00)00031-1.
[247] U.V. Sojitra, S.S. Nadar, V.K. Rathod, Immobilization of pectinase onto [269] S. Fernandes, G. Johansson, R. Hatti-Kaul, Purification of recombinant
chitosan magnetic nanoparticles by macromolecular cross-linker, cutinase by extraction in an aqueous two-phase system facilitated by a fatty
Carbohydr. Polym. 157 (2016) 677–685, http://dx.doi.org/10.1016/j.carbpol. acid substrate, Biotechnol. Bioeng. 73 (2001) 465–475, http://dx.doi.org/10.
2016.10.018. 1002/bit.1081.
[248] S.W. Lee, S.A. Cheon, M. Il Kim, T.J. Park, Organic–inorganic hybrid [270] S. Teotia, M. Gupta, Purification of phospholipase D by two-phase affinity
nanoflowers: types, characteristics, and future prospects, J. Nanobiotechnol. extraction, J. Chromatogr. A 1025 (2004) 297–301, http://dx.doi.org/10.
13 (2015) 54, http://dx.doi.org/10.1186/s12951-015-0118-0. 1016/j.chroma.2003.10.104.
[249] S.S. Nadar, V.K. Rathod, Facile synthesis of glucoamylase embedded [271] M. Antov, L. Anderson, A. Andersson, F. Tjerneld, H. Stålbrand, Affinity
metal-organic frameworks (glucoamylase-MOF) with enhanced stability, partitioning of a Cellulomonas fimi ␤-mannanase with a mannan-binding
Int. J. Biol. Macromol. 95 (2017) 511–519, http://dx.doi.org/10.1016/j. module in galactomannan/starch aqueous two-phase system, J. Chromatogr.
ijbiomac.2016.11.084. A 1123 (2006) 53–59, http://dx.doi.org/10.1016/j.chroma.2006.05.021.
[250] J. Ge, J. Lei, R.N. Zare, Protein–inorganic hybrid nanoflowers, Nat. [272] S.P.M. Ventura, R.L.F. de Barros, J.M. de Pinho Barbosa, C.M.F. Soares, S. Á.
Nanotechnol. 7 (2012) 428–432, http://dx.doi.org/10.1038/nnano.2012.80. Lima, J.A.P. Coutinho, Production and purification of an extracellular
[251] B. Somturk, M. Hancer, I. Ocsoy, N. Özdemir, Synthesis of copper ion lipolytic enzyme using ionic liquid-based aqueous two-phase systems,
incorporated horseradish peroxidase-based hybrid nanoflowers for Green Chem. 14 (2012) 734, http://dx.doi.org/10.1039/c2gc16428k.
enhanced catalytic activity and stability, Dalton Trans. 44 (2015) [273] S. Dreyer, U. Kragl, Ionic liquids for aqueous two-phase extraction and
13845–13852, http://dx.doi.org/10.1039/C5DT01250C. stabilization of enzymes, Biotechnol. Bioeng. 99 (2008) 1416–1424, http://
[252] S.S. Nadar, S.D. Gawas, V.K. Rathod, Self-assembled organic–inorganic dx.doi.org/10.1002/bit.21720.
hybrid glucoamylase nanoflowers with enhanced activity and stability, Int. J. [274] Q. Cao, L. Quan, C. He, N. Li, K. Li, F. Liu, Partition of horseradish peroxidase
Biol. Macromol. 92 (2016) 660–669, http://dx.doi.org/10.1016/j.ijbiomac. with maintained activity in aqueous biphasic system based on ionic liquid,
2016.06.071. Talanta 77 (2008) 160–165, http://dx.doi.org/10.1016/j.talanta.2008.05.055.
[253] K. Ratanapongleka, J. Phetsom, Extraction in two-phase systems and some [275] M. Pereira, Y.-T. Wu, A. Venâncio, J. Teixeira, Aqueous two-phase extraction
properties of laccase from lentinus polychrous, Int. J. Chem. Mol. Nucl. using thermoseparating polymer: a new system for the separation of
Mater. Metall. Eng. 5 (2011) 733–736. endo-polygalacturonase, Biochem. Eng. J. 15 (2003) 131–138, http://dx.doi.
[254] M.C. Madhusudhan, K.S.M.S. Raghavarao, S. Nene, Integrated process for org/10.1016/S1369-703X(02)00190-0.
extraction and purification of alcohol dehydrogenase from Baker’s yeast [276] S. Moreira, S.C. Silvério, E.A. Macedo, A.M.F. Milagres, J.A. Teixeira, S.I.
involving precipitation and aqueous two phase extraction, Biochem. Eng. J. Mussatto, Recovery of Peniophora cinerea laccase using aqueous two-phase
38 (2008) 414–420, http://dx.doi.org/10.1016/j.bej.2007.08.007. systems composed by ethylene oxide/propylene oxide copolymer and
[255] D.N.X. Salmon, A. Walter, T.S. Porto, K.A. Moreira, L.P. de S. Vandenberghe, potassium phosphate salts, J. Chromatogr. A 1321 (2013) 14–20, http://dx.
C.R. Soccol, A.L.F. Porto, M.R. Spier, Aqueous two-phase extraction for partial doi.org/10.1016/j.chroma.2013.10.056.
purification of Schizophyllum commune phytase produced under [277] M. Li, T.L. Peeples, Purification of hyperthermophilic archaeal amylolytic
solid-state fermentation, Biocatal. Biotransform. 32 (2014) 45–52, http://dx. enzyme (MJA1) using thermoseparating aqueous two-phase systems, J.
doi.org/10.3109/10242422.2013.872633. Chromatogr. B 807 (2004) 69–74, http://dx.doi.org/10.1016/j.jchromb.2004.
[256] I.H. Pan, Y.K. Li, Rapid process for purification of an extracellular 03.030.
␤-xylosidase by aqueous two-phase extraction, J. Chromatogr. B Biomed. [278] M. Amid, Y. Manap, N.K. Zohdi, A novel aqueous two phase system
Sci. Appl. 754 (2001) 179–184, http://dx.doi.org/10.1016/S0378- composed of a thermo-separating polymer and an organic solvent for
4347(00)00604-6. purification of thermo-Acidic amylase enzyme from red pitaya (Hylocereus
[257] H.S. Ng, C.P. Tan, S.K. Chen, M.N. Mokhtar, A. Ariff, T.C. Ling, Primary capture polyrhizus) peel, Molecules 19 (2014) 6635–6650, http://dx.doi.org/10.
of cyclodextrin glycosyltransferase derived from Bacillus cereus by aqueous 3390/molecules19056635.
two phase system, Sep. Purif. Technol. 81 (2011) 318–324, http://dx.doi.org/ [279] V.V. Kumar, V. Sathyaselvabala, S.D. Kirupha, A. Murugesan, T. Vidyadevi, S.
10.1016/j.seppur.2011.07.039. Sivanesan, Application of response surface methodology to optimize three
[258] M.C. Lakshmi, M.C. Madhusudhan, K.S.M.S. Raghavarao, Extraction and phase partitioning for purification of laccase from pleurotus ostreatus, Sep.
purification of lipoxygenase from soybean using aqueous two-phase system, Sci. Technol. 46 (2011) 1922–1930, http://dx.doi.org/10.1080/01496395.
Food Bioprocess Technol. 5 (2012) 193–199, http://dx.doi.org/10.1007/ 2011.583306.
s11947-009-0278-8. [280] M. Shanmugaprakash, C. Jayashree, V. Vinothkumar, S.N.S. Senthilkumar, S.
[259] D. Rajagopalu, P.L. Show, Y.S. Tan, S. Muniandy, V. Sabaratnam, T.C. Ling, Siddiqui, V. Rawat, M. Arshad, Biochemical characterization and antitumor
Recovery of laccase from processed Hericium erinaceus (Bull.: Fr) Pers. activity of three phase partitioned l-asparaginase from Capsicum annuum L,
S.S. Nadar et al. / International Journal of Biological Macromolecules 101 (2017) 931–957 957

Sep. Purif. Technol. 142 (2015) 258–267, http://dx.doi.org/10.1016/j.seppur. [283] Y. Duman, E. Kaya, Purification and recovery of invertase from potato tubers
2014.12.036. (Solanum tuberosum) by three phase partitioning and determination of
[281] H.S. Choonia, S.S. Lele, Three phase partitioning of ␤-galactosidase produced kinetic properties of purified enzyme, Turk. J. Biochem. 39 (2014) 443–448,
by an indigenous Lactobacillus acidophilus isolate, Sep. Purif. Technol. 110 http://dx.doi.org/10.5505/tjb.2014.78949, Research Article [Araştırma
(2013) 44–50, http://dx.doi.org/10.1016/j.seppur.2013.02.033. Makalesi].
[282] S. Rajeeva, S.S. Lele, Three-phase partitioning for concentration and [284] F. Liu, J.H. Zhao, X. Wen, Y.Y. Ni, Purification and structural analysis of
purification of laccase produced by submerged cultures of Ganoderma sp. membrane-bound polyphenol oxidase from Fuji apple, Food Chem. 183
WR-1, Biochem. Eng. J. 54 (2011) 103–110, http://dx.doi.org/10.1016/j.bej. (2015) 72–77, http://dx.doi.org/10.1016/j.foodchem.2015.03.027.
2011.02.006.

You might also like