Potential Applications of Enzymes Immobi

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 12

Biotechnology Advances 30 (2012) 512–523

Contents lists available at SciVerse ScienceDirect

Biotechnology Advances
journal homepage: www.elsevier.com/locate/biotechadv

Research review paper

Potential applications of enzymes immobilized on/in nano materials: A review


Shakeel Ahmed Ansari a, Qayyum Husain b,⁎
a
Department of Biochemistry, Faculty of Life Sciences, Aligarh Muslim University, Aligarh-202002, India
b
Faculty of Applied Medical Sciences, Jazan University, Jazan, Post Box 2092, Saudi Arabia

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

Article history: Several new types of carriers and technologies have been implemented in the recent past to improve traditional
Received 23 May 2011 enzyme immobilization which aimed to enhance enzyme loading, activity and stability to decrease the enzyme
Received in revised form 26 August 2011 biocatalyst cost in industrial biotechnology. These include cross-linked enzyme aggregates, microwave-assisted
Accepted 12 September 2011
immobilization, click chemistry technology, mesoporous supports and most recently nanoparticle-based immo-
Available online 17 September 2011
bilization of enzymes. The union of the specific physical, chemical, optical and electrical properties of nanoparti-
Keywords:
cles with the specific recognition or catalytic properties of biomolecules has led to their appearance in myriad
Nanoparticles novel biotechnological applications. They have been applied time and again for immobilization of industrially
Immobilization important enzymes with improved characteristics. The high surface-to-volume ratio offered by nanoparticles
Biosensors resulted in the concentration of the immobilized entity being considerably higher than that afforded by experi-
Biomedical applications mental protocols based on immobilization on planar 2-D surfaces. Enzymes immobilized on nanoparticles
showed a broader working pH and temperature range and higher thermal stability than the native enzymes.
Compared with the conventional immobilization methods, nanoparticle based immobilization served three im-
portant features; (i) nano-enzyme particles are easy to synthesize in high solid content without using surfactants
and toxic reagents, (ii) homogeneous and well defined core–shell nanoparticles with a thick enzyme shell can be
obtained, and (iii) particle size can be conveniently tailored within utility limits. In addition, with the growing
attention paid to cascade enzymatic reaction and in vitro synthetic biology, it is possible that co-immobilization
of multi-enzymes could be achieved on these nanoparticles.
© 2011 Elsevier Inc. All rights reserved.

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 512
2. Magnetic nanoparticles in enzyme immobilization . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 513
3. Enzyme immobilization on novel nanoparticles . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 515
4. Enzyme immobilization on nonmagnetic nanoparticles . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 516
5. Nanoparticles-based enzymes in biosensors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 519
6. Nanobiocatalysis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 520
7. Concluding remarks. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 521
Acknowledgments . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 521
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 521

1. Introduction
Abbreviations: GOX, glucose oxidase; NSP, nickel-impregnated silica paramagnetic
particles; EI-NSP, enzyme immobilized on NSP; HRP, horseradish peroxidase; ITO, indi- Recent interest in nanotechnology has provided a wealth of diverse
um tin oxide; GOS, galacto-oligosaccharides; CGTase, cyclodextrin glycosyl transferase; nanoscaffolds that could potentially support enzyme immobilization
SiMNs, silica-coated magnetic nanoparticles; PAN-co-MMA, poly-acrylonitrile-co- due to their potential applications in biotechnology, immunosensing
methyl-methacrylate; PEG, polyethylene glycol; PEI, polyethyleneimine; PAZO, poly and biomedical areas. Immobilization of enzymes is advantageous for
[1-[4-(3-carboxy-4-hydroxyphenylazo) benzenesulfonamido]-1,2-ethanediyl, sodium
commercial application due to convenience in handling, ease of separa-
salt].
⁎ Corresponding author. Tel.: + 966 595021577(Mobile). tion of enzymes from the reaction mixture and reuse, low product cost
E-mail address: qayyumbiochem@gmail.com (Q. Husain). and a possible increase in thermal and pH stability (Husain, 2010; Lei et

0734-9750/$ – see front matter © 2011 Elsevier Inc. All rights reserved.
doi:10.1016/j.biotechadv.2011.09.005
S.A. Ansari, Q. Husain / Biotechnology Advances 30 (2012) 512–523 513

al., 2002; Tischer and Wedekind, 1999; Wang, 2006). An important re- The premise of using nanoscale structures for immobilization is to
quirement for protein immobilization is that the matrix should provide reduce diffusion limitations and maximize the functional surface area
a biocompatible and inert environment, i.e. it should not interfere with to increase enzyme loading (Xie et al., 2009). In addition, the physical
the native structure of the protein, which thereby could compromise its characteristics of nanoparticles such as enhanced diffusion and parti-
biological activity (Mitchell et al., 2002). cle mobility can impact inherent catalytic activity of attached en-
Enzymes immobilized to nanosized scaffolds such as spheres, fibers zymes (Jia et al., 2003). Thermal stability, increased surface area and
and tubes have recently been reported (Kim et al., 2006a, b; Martin irradiation resistance are the other advantages that expedite their po-
and Kohli, 2003; Yim et al., 2003). Consequently, a number of processes tential applications in photodetectors, solar cells, biosensors, nano-
for enzyme immobilization in silica nanotubes (Mitchell et al., 2002), generators and ceramics (Moghaddam et al., 2009). In the last few
within phospholipid bilayers (Hamachi et al., 1994), on self-assembled years, several types of nanoparticles have been extensively employed
monolayers (Hung et al., 2006), in Langmuir–Blodgett films (Troitsky in the production of various nanostructures like nanorods, nanotubes,
et al., 2003), within a polymer matrix (Kim et al., 2005a; Kim et al., nanowires, nanorings etc. (Ali and Winterer, 2010; Ni et al., 2007).
2005b), galleries of R-zirconium phosphate (Kumar and McLendon, Amongst all, biosystems and its components have a remarkable im-
1997), mesoporous materials (Lei et al., 2006), on polystyrene latex par- pact as biotechnology deals with nanometer size molecules such as
ticles (Caruso and Mohwald, 1999), gold nanoparticles assembled on protein and nucleic acids. Several bio-nano processes have been de-
polymers and zeolite (Bhat et al., 2003; Phadtare et al., 2003) and in ther- veloped using fabricated nanoscale structures with biomolecules as
mally evaporated fatty lipid films (Sastry et al., 2002) have been devel- nanoblocks (Yamashita, 2001).
oped, each with its characteristic pros and cons. Moreover, polymer
microspheres and various metal nanoparticles have also been successful- 2. Magnetic nanoparticles in enzyme immobilization
ly conjugated with proteins and enzymes (Schuler and Caruso, 2000;
Willner et al., 2006). The surface modification of nanoparticles via en- Magnetic fields have been utilized in support systems to study
zyme molecules and biomembrane probes provided unique biofunction- enzyme immobilization (Bayramoglu et al., 2008; Gupta and
ality to them. Qhobosheane et al. (2001) have immobilized lactate Gupta, 2005; Kuroiwa et al., 2008; Pimentel et al., 2007; Selim et
dehydrogenase (LDH) and glutamate dehydrogenase onto the silica al., 2007). Several magnetic particles and magnetic supports such
nanoparticles in order to modify them biochemically. The immobilized as microspheres of various biomaterials encapsulating the magnetic
preparations showed excellent enzymatic activities and detection capa- particles and copolymers with magnetic particles have been used
bility. They used dye-doped silica nanoparticles as biomarkers for cell with good results (Dyal et al., 2003; Koneracka et al., 1999; Kouassi
staining to demonstrate their bioapplicability in several biochemical et al., 2005; Saiyed et al., 2003). The high surface to volume ratio pro-
and biotechnological applications. Thus, these approaches demonstrated vided by the magnetic nanoparticles favors high binding capacity and
the feasibility of utilizing such nanoparticles in biosensing and biomark- high catalytic specificity of the conjugated enzyme (Johnson et al.,
ing applications. 2008; Konwarh et al., 2009). In addition, magnetic field susceptibility

Table 1
Enzymes immobilized on magnetic nanoparticles and their biotechnological applications.

Enzyme Nanoparticle utilized Applications Kinetic parameters References

Cholesterol oxidase Fe3O4 Analysis of total Free enzyme: Ea = Kouassi et al., 2005
nanoparticles cholesterol 13.6 kJ/mol;
in serum immobilized
enzyme: Ea = 9.3
kJ/mol
Haloalkane dehalogenase Silica coated Production of – Johnson et al., 2008
iron oxide fusion proteins
nanoparticle containing
dehalogenase
sequences
Laccase Chitosan-magnetic nanoparticle Bioremediation Free enzyme: Km Kalkan et al., 2009
of environmental (10−2 mM) = 5.69,
pollutants Vmax (10−2 mM min−1) = 7.70;
immobilized enzyme:
Km (10−2 mM) = 10.69, Vmax
(10−2 mM min−1) = 14.00
Keratinase Fe3O4 Synthesis of keratin Soluble enzyme: Konwarh et al., 2009
nanoparticle specific activity = 37 U mg− 1;
immobilized enzyme:
specific activity = 128.97 U mg− 1
α-Amylase Cellulose-coated magnetite nanoparticles Starch degradation Immobilized enzyme: Namdeo and Bajpai, 2009
Km = 7.5 × 10− 7 μmol ml− 1, rmax
(maximum degradation rate)
= 0.04 × 10− 6 μmol− 1 g− 1 min− 1
β Galactosidase Con A layered ZnO nanoparticles Lactose hydrolysis Free enzyme: Ansari and Husain, 2011a
Km = 2.38 mM,
Vmax = 0.520 mM min− 1;
immobilized enzyme:
Km = 5.88 mM,
Vmax = 0.460 mM min−1
Lipase Fe3O4 nanoparticles Hydrolysis of pNPP Immobilized enzyme: Huang et al., 2003
Ea = 6.4 kJ mol−1,
specific activity = 1.07 μmol min−1
mg, Km = 0.4 mM
514 S.A. Ansari, Q. Husain / Biotechnology Advances 30 (2012) 512–523

revealed a mechanism for efficient recovery of the enzyme complex oxide superparamagnetic nanoparticles using affinity peptides. The
thereby preventing the enzyme contamination of the final product. En- enzyme was cloned from Xanthobacter autotrophicus strain GJ10
zyme stability is maximized with nanoscaled support with the addition- into Escherichia coli to produce fusion proteins containing dehalogen-
al advantages of possible modulation of the catalytic specificity, lower ase sequences with C-terminal polypeptide repeats that have a specif-
transfer resistance to solve the diffusion problem and lower operational ic affinity for either silica or iron oxide. Researchers are employing
cost (Bruno et al., 2005). Table 1 lists various magnetic nanoparticles current technologies and developing new applications that utilize en-
used for the immobilization of enzymes and their biotechnological zymes immobilized on nanoparticles. Many enzymes currently used
applications. in biotechnology, including glucose oxidase and peroxidase, have
Entrapment of enzymes has also been performed by using iron–silica been covalently immobilized to magnetic nanoparticles (MNPs)
composites because of its better biocompatibility (Besanger et al., 2003). using several different ligands. Glucose oxidase that was used to mea-
Prakasham et al. (2010) utilized nickel-impregnated silica paramagnetic sure blood glucose was covalently immobilized on MNPs. It was
particles (NSP) as biocatalyst immobilization matrices. A greater catalytic found to be stable over a wide range of pH and temperature range
behavior of enzyme immobilized on NSP (EI-NSP) compared to its solu- and retained significant activity for 3 months (Rossi et al., 2004). Li-
ble counterpart revealed that immobilization of an enzyme on these NSP pase, which is important for processing lipids in food and pharmaceu-
protects its deactivation at higher and lower pH. Moreover, the improved tical industries, has been covalently immobilized on nanoparticles by
enzyme activity might be attributed to the fact that enzyme configura- a carbodiimide linkage and exhibited significant activity after
tion was altered under an immobilized environment. Such a variation 1 month of storage (Dyal et al., 2003). Trypsin and chymotrypsin
of improved activity under immobilized conditions has been observed have also been stabilized against denaturation and self-digestion at
with other microbial systems and enzymes (Ramkrishna and Prakasham, the air–water interface by immobilization on iron and gold nanopar-
1999; Srinivasulu et al., 2002). In addition, immobilization of diastase on ticles (Hansen et al., 2006; Jordan et al., 2006). Streptokinase, which
NSP was evaluated for starch hydrolysis at different temperatures rang- has been used as a therapeutic agent, was covalently immobilized to
ing from 30 to 60 °C at pH 5.0. EI-NSP showed improved starch hydroly- MNPs by carbodiimide linkage for use in localized lysis of blood
sis activity compared to a free enzyme at all investigated temperatures clots in vivo. The magnetic properties of the particle-streptokinase
and it showed enhanced thermostability as a result of immobilization congener would allow focusing on treatment at the exact location
(Prakasham et al., 2007). where the clot is present, thus reducing the amount of enzyme re-
Kim et al. (2009) coated magnetic particles (10 nm) with silica (di- quired and in turn decreasing the risk of eliciting an immune re-
ameter of 40 nm) and charged them with Cu 2+ ions via a multidentate sponse (Fernandes et al., 2006; Koneracka et al., 2002).
ligand, imino-diacetic acid for the immobilization of His-tagged Bacillus Kouassi et al. (2005) successfully bound cholesterol oxidase to
stearothermopilus L1 lipase. Microporous silica gel with a mean particle Fe3O4 nanoparticles via carbodiimide activation and confirmed its
diameter of 115 μm was used as a comparative support material. The binding by FT-IR spectroscopy. The binding efficiency was between
molar ratio of Cu2+ to imino-diacetic acid was found to be 1:1.14 and 98% and 100% irrespective of the amount of particles used. Kinetic
1:1.99 in the silica gel and the silica-coated magnetic nanoparticles studies revealed that the stability and activity of the enzyme was sig-
(SiMNs), respectively. The specific activity of the immobilized enzyme nificantly improved against pH, temperature and substrate concen-
exhibited the following order: Cu2+-charged SiMN N SiMNN Cu2+- tration upon its binding to nanoparticles in comparison to the free
charged silica gel N silica gel. Lipase immobilized on Cu2+charged enzyme. The activation energy for free and bound cholesterol oxidase
SiMNs retained 70% activity after its 5th repeated use while other was 13.6 and 9.3 kJ mol −1, respectively. The study demonstrated that
immobilized preparations lost considerably very high activity under the stability and activity of this enzyme could be enhanced via attach-
similar experimental conditions. Similarly, keratinase from Bacillus ment to the magnetic nanoparticles, which could henceforth contrib-
subtilis immobilized onto polyethylene glycol-supported Fe3O4 super- ute to the better use of enzymes in various biological and clinical
paramagnetic nanoparticles showed a marked increase in stability applications.
against various types of physical and chemical denaturants as compared Pseudomonas cepacia lipase immobilized onto magnetic nanopar-
to its soluble counterpart (Konwarh et al., 2009). ticles via carbodiimide activation prepared by co-precipitating ferric
Levison et al. (1998) prepared Fe2O3 nanoparticles by sonication and ferrous chlorides in alkaline solution under hydrothermal condi-
of Fe (CO) in decalin and then annealed amorphous Fe2O3 nanoparti- tions showed a binding efficiency of 90% and the corresponding activ-
cles for their subsequent use in immobilizing industrial enzymes. ity recovery of 70% when the weight ratio of lipase bound to magnetic
Several investigators have earlier reported an improvement in the nanoparticles was 0.09. The thermal stability was significantly im-
stability of Candida rugosa lipase when immobilized on Fe2O3 mag- proved after immobilization thereby attracting its use in the produc-
netic nanoparticles. This system offered a relatively simple technique tion of biodiesel (Mak et al., 2009). Nevertheless, organic–inorganic
for separating and reusing enzymes over a longer period than that for complex technology was exploited to prepare zinc tetra-amino-
free enzymes alone and for enzymes immobilized by physisorption. phthalocyanine-Fe3O4 nanoparticle composites. Laccase bound to
The separation of an immobilized enzyme was facilitated by using a such support via glutaraldehyde by the active amino groups of mag-
magnet where either the substrate solution was removed while the netic carriers showed immobilization yields and Km value of 25%
immobilized enzyme was held in place with a magnetic field or vice and 20.1 μM, respectively. Immobilized laccase had good thermal,
versa (Dyal et al., 2003; Gardimalla et al., 2005). In another study, C. storage and operational stability, which could be used as the sensing
rugosa lipase (CRL) was immobilized on novel nanosupport, phos- biocomponent for the fiber optic biosensor based on enzyme catalysis
phorous-containing polyurethanes. Pre-treatment of the support (Huang et al., 2007).
with polar or non-polar solvents had a negative impact on lipase A novel and efficient immobilization of β galactosidase from
loading. Non-ionic surfactants increased the immobilization yield of Aspergillus oryzae was recently developed by using magnetic Fe3O4-
CRL by 30–40%. Immobilization has improved the thermal stability chitosan nanoparticles as support. The magnetic Fe3O4-chitosan
of enzyme and exhibited a half-life of 70 h at 55 °C. The highest syn- nanoparticles were prepared by electrostatic adsorption of chitosan
thetic activity of CRL-polyurethane preparations were observed at onto the surface of Fe3O4 nanoparticles made through co-precipita-
aw = 0.33. The biocatalyst retained 80% activity after fifteen subse- tion of Fe 2+ and Fe 3+. β Galactosidase was covalently immobilized
quent five-hour cycles when utilized in esterification of palmitic onto the nanocomposites using glutaraldehyde as activating agent.
acid with cetyl alcohol (Guncheva et al., 2011). The immobilization process was optimized by examining immobi-
Johnson et al. (2008) demonstrated the feasibility of specifically lized time, crosslinking time, enzyme concentration, glutaraldehyde
attaching haloalkane dehalogenase to silica-coated or uncoated iron concentration, and initial pH values of glutaraldehyde and enzyme
S.A. Ansari, Q. Husain / Biotechnology Advances 30 (2012) 512–523 515

solution. As a result, the immobilized enzyme presented a higher (SDS-PAGE). An immobilized two-enzyme system showed optimal activ-
storage, pH and thermal stability than the soluble enzyme. Galacto- ity at pH 7.0 and 8.0 for (S)-1,2-propanediol and L-tagatose production,
oligosaccharide (GOS) was formed with lactose as substrate by respectively. It retained 70% activity after 1 week of repeated use. The
using the immobilized enzyme as biocatalyst with a maximum yield use of affinity magnetic nanoparticles offers the advantage of a one-pot
of 15.5% (w/v) obtained when 50% lactose was hydrolyzed (Pan et purification of His(6)-tagged GatDH and FDH followed by the production
al., 2009). of rare sugar and chiral diol (Demira et al., 2011).
Recently, cellulosic ethanol has received a lot of attention as an al-
ternative transportation fuel to reduce global dependence on oil. 3. Enzyme immobilization on novel nanoparticles
However, due to high cost of enzymes, the process is not currently
economically feasible. Cellulase multi-enzyme mixtures along with Gold nanoparticles serve as excellent biocompatible surfaces for
extra β glucosidase are the key enzymes for biomass degradation. the immobilization of enzymes and proteins since the interaction be-
Glucose oxidase (GOX) was selected as a model enzyme to demon- tween amino and cysteine groups of proteins with gold nanoparticles
strate the immobilization strategies. Three different size magnetic is as strong as that of the commonly used thiols. Thus, amino acids
nanoparticles (5 nm, 25 nm and 50 nm) were fabricated to explore and proteins may be directly immobilized on gold nanoparticles
the effect of various particle sizes on diffusion efficiency. Two differ- without any modification (Crespilho et al., 2006; Lia et al., 2010; Xu
ent methods, co-precipitation and oxidation of Fe (OH)2 were used et al., 2007). Gold nanoparticles can be easily and efficiently functio-
to fabricate different sizes of magnetic nanoparticles functionalized nalized with thiolated molecules and carboxylic groups, which in
with amine groups by 3-(amino propyl) triethoxysilane. Glutaralde- turn, are conjugated with amino groups of the proteins (Jun et al.,
hyde was used as a crosslinking agent between functionalized mag- 2001; Vertegel et al., 2004). Enzyme immobilized on polyurethane
netic nanoparticles and GOX. It was found that activity of GOX microsphere–gold nanoparticles has shown enhanced storage and
immobilized on different sized magnetic nanoparticles retained thermal stability (Kumar et al., 2003). Phadtare et al. (2004) have in-
marked activity after 40 day storage at various temperatures. Trans- vestigated the assembly of gold nanoparticles on the surface of the
mission electron micrograph (TEM) showed that the morphology of amine-functionalized zeolite microspheres for the formation of zeo-
magnetic nanoparticles was spherical and the sizes agreed with re- lite-gold nanoparticle “core–shell” structures and its use in immobi-
sults obtained from those of a Brunauer, Emmett, Teller method. lizing fungal protease. The assembly of gold nanoparticles on the
The magnetic strength of nanoparticles was analyzed via a physical zeolite surface occurs through the amino groups present in 3-amino-
property measurement system. X-ray photoelectron spectroscopy con- propyltrimethoxysilane. The fungal proteases bound to the massive
firmed each step of the magnetic nanoparticle surface modification and “core–shell” structures were easily separated from the reaction medium
successful enzyme immobilization. Recycling stability studies showed by mild centrifugation and exhibited excellent reuse characteristics. The
20% loss in activity during 10 consecutive recycles for large (50 nm) biocatalytic activity of fungal protease in bioconjugate was marginally
and medium (25 nm) size enzyme magnetic nanoparticles. enhanced relative to the free enzyme in solution. It has also shown an
Laccase is a potentially attractive catalyst for bioremediation of envi- enhancement in pH and temperature stability and a shift in the temper-
ronmental pollutants especially organic substances such as chlorophe- ature-optima.
nols and aromatic hydrocarbons of petrochemical industry, toxins of Yang et al. (2006) have studied multilayer films of GOX on Au-
olive oil, mill wastewater, de-lignification in paper industry and dye de- nanoparticle electrode surface using cysteamine as a covalent attach-
colorization in textile industry. Laccase from Trametes versicolor immo- ment cross-linker. However, the main disadvantage of this method
bilized onto chitosan coated magnetite nanoparticles by using reversed- was the addition of a redox mediator to the reaction medium which
phase suspension adsorption retained about 71% of its initial activity at was less efficient than using an immobilized redox mediator. Moreover,
the end of its 30th repeated use (Kalkan et al., 2009). submicron-sized poly (N-isopropyl acrylamide)/polyethyleneimine
α Amylase immobilized on Fe2O3 nanoparticle had significantly core–shell microgels were prepared in aqueous media by using tert-
greater thermal, storage and operational stability as compared to its butyl hydroperoxide as an initiator and then the gold nanoparticles
free counterpart. Both the soluble and immobilized enzymes were (8 nm) were formed on the surface of microgels. The amino groups on
characterized by Fourier transform infrared spectroscopy (FT-IR), the polyethyleneimine (PEI) chains act as the binder for the assembly
atomic force microscopy (AFM), thermogravimetric analysis (TGA) of gold nanoparticles/microgel complex. The microgels were highly sta-
and differential thermal analysis (DTA). The results for the continu- ble with gold nanoparticles on their extended PEI chains in aqueous
ous hydrolysis of starch in a batch process suggested that α amylase media, and this multi-scale nanoparticle complex could be recovered
immobilized on Fe2O3 nanoparticle might be a good candidate for from water and redispersed in water. The nanogold/microgel particles
the hydrolysis of starch and other polysaccharides used in food indus- were further conjugated with the enzymes; horseradish peroxidase
tries (Khan et al., Communicated). Recently, catalytic removal of (HRP) and urease. It was found that under identical assay conditions,
bisphenol A was investigated with hemoglobin immobilized on the enzyme/nanogold/microgel systems exhibited enhanced biocata-
amino-modified magnetic nanoparticles (Tang et al., 2011). The lytic activity over free enzymes in solution, especially at lower enzyme
amino-modified magnetite nanoparticles were firstly prepared by concentrations. Moreover, HRP/nanogold/microgel systems showed
the co-precipitation of Fe 2+ and Fe 3+ with NH3·H2O and then modi- higher activity at various pH, temperatures and storage stability as com-
fied by 3-aminopropyltriethoxysilane. It was observed that immobi- pared to free enzyme (Xu et al., 2007). The interaction of colloidal gold
lized hemoglobin could remove bisphenol A up to 80.3%. The particles with proteins/enzymes have been employed for the immobili-
immobilization had a beneficial effect on the stability of hemoglobin zation of pepsin and xanthine oxidase (Gole et al., 2001; Zhao et al.,
and conversions of bisphenol A. 1996). The high surface area of host gold nanoparticles renders the
Rapid immobilization with one-pot purification of galactitol dehy- immobilized enzyme “quasi free” and at the same time retained the ad-
drogenase (GatDH) and formate dehydrogenase (FDH) was achieved vantages of immobilization such as ease in reusing, an enhanced tempo-
by using iminodiacetic acid with chelated Co2+ modified magnetic ral and thermal stability, etc.
nanoparticles as a carrier. Lactate dehydrogenase (LDH) from recombi- Crespilho et al. (2009) demonstrated an extremely promising ap-
nant E. coli and FDH commencing Candida methylica were used as an proach for constructing enzyme biosensors. They immobilized urease
auxiliary enzyme for the regeneration of NADH/NAD +. The affinity on electroactive nanostructured membranes made up of poly-aniline
magnetic nanoparticles were characterized by scanning electron mi- and silver nanoparticles (AgNP) stabilized in polyvinyl alcohol
croscopy (SEM) and FTIR while the purity of GatDH and FDH was (PAni/PVA-AgNP). Fabrication of this modified electrode comprised
assayed by sodium dodecyl sulfate-polyacrylamide gel electrophoresis chemical deposition of poly-aniline followed by drop-coating of
516 S.A. Ansari, Q. Husain / Biotechnology Advances 30 (2012) 512–523

PVA-AgNP and urease, resulting in a final ITO/PAni/PVA-AgNP/urease action of electrocatalytic activity of platinum nanoparticles and CNT.
electrode configuration. Investigation on the performance of this The combined electrocatalytic activity permitted low-potential detec-
electrode toward urea hydrolysis via amperometric measurements tion of H2O2 with remarkably improved sensitivity. Platinum-CNT
revealed a fast increase in cathodic current with a well-defined peak based biosensor responded more sensitively to glucose than CNT-
upon addition of urea to the electrolytic solution. A modified elec- based biosensor. The sensitivity of platinum-CNT based biosensor
trode with an immobilized enzyme promoted an efficient conversion was four times greater than that of CNT-based biosensor. Similarly,
of urea into ammonium and bicarbonate ions. Km app
of 2.7 mM L − 1 in- Zhao et al. (2007) have demonstrated a sensitive amperometric glu-
dicated that the electrode architecture employed might be advanta- cose biosensor based on platinum nanoparticles and CNTs electrode.
geous for fabrication of enzymatic devices with improved biocatalytic Platinum-nanoparticles enhanced the electrocatalytic activity of elec-
properties. This applied potential ensures minimization of interference trode for electro-oxidating H2O2. GOX electrode exhibited good re-
effects when the biosensor was used in real and complex matrices such producibility and excellent response performance to glucose with
as biological media, food and beverages. The measurements carried out linear range from 1 × 10 −5 to 7 × 10 −3 mol L −1 and fast response
with H2O2 and glucose demonstrated that this new approach was ex- time within 5 s.
tremely promising for the construction of enzyme biosensors.
GOX was covalently immobilized on thiolated gold nanoparticles
via N-ethyl-N′-(3-dimethyl-aminopropyl) carbodiimide and N- 4. Enzyme immobilization on nonmagnetic nanoparticles
hydroxysuccinimide. Its binding was confirmed by UV–visible, FT-
IR, CD and TEM studies. The immobilized enzyme exhibited a re- Nanoscaled materials provide upper limits in balancing the key
sponse time of 30 s, a shelf life of more than 6 months and improved factors that determine the efficiency of biocatalysts including surface
tolerance to both pH and temperature. Lower Km (3.74 mg dL −1) of area, mass transfer resistance and effective enzyme loading. The ex-
immobilized GOX showed enhanced activity which indicated a fa- ploitation of various nanomaterials such as nanoparticles, nanofibers,
vorable conformational change in the GOX structure upon attach- nanotubes and nanoporous matrices for biotechnological applications
ment to thiolated gold nanoparticles. These GOX thiolated gold has been excellently reviewed by Wang (2006). Beyond their high
nanoparticles were used for estimating glucose up to 300 mg dL -1. surface area to volume ratios, nanoscale biocatalyst systems exhibit
The result showed that these biocompatible thiol-modified gold unique behaviors that distinguish them from traditional immobilized
nanoparticles might serve as a promising matrix for the immobiliza- systems and extend their remarkable applications in engineering of
tion of other enzymes and proteins with enhanced stability and ac- biomolecular catalysis (Jia et al., 2011; Kim et al., 2011).
tivity for biosensor applications (Pandey et al., 2007). Recently, Several workers have developed a one-step method for preparing
gold nanoparticles were used to immobilize acetylcholinesterase. cellulase-immobilized on nanoparticles that consisted of well-defined
AFM demonstrated that gold nanoparticles enhanced interaction of poly-methyl methacrylate (PMMA) cores and cellulase shells (Ho et
the enzyme with the gold electrode. The modified electrode was al., 2008; Kin et al., 2008). Immobilized cellulase showed improved
exploited for electrochemical detection of thiocholine at the gold thermostability and retained significantly very high activity at
surface after hydrolyzing acetylthiocholine by the immobilized enzyme. broader pH range as compared to soluble counterpart. These nano-
The sensor response to acetylthiocholine was significantly reduced and particles have thick and evenly distributed enzyme shells and this
the utility of the electrode was limited in the absence of nanoparticle preparation can be produced in high concentrations of up to 18%
layer. The ability of the nanoparticle-based sensor to reliably measure (w/w) solid content. Thus, this method might provide a new com-
concentrations of the organophosphate pesticide carbofuran at lesser mercially viable route to the immobilization of thermally stable en-
concentrations was demonstrated by monitoring the inhibition of hy- zyme to form nanoparticle enzyme complexes (Ho et al., 2008).
drolysis of acetylthiocholine. Table 2 illustrates some common nanoparticles that have been
Mahmoud et al. (2009) have prepared a novel nanocomposite employed in the recent past for immobilizing enzymes of major in-
consisting of cellulose nanocrystals functionalized with gold nanopar- dustrial applications.
ticles. Cyclodextrin glycosyl transferase (CGTase) and alcohol oxidase Eldin et al. (2010) prepared copolymers of poly-acrylonitrile-
conjugated on an activated cellulose nanocrystal/gold nanoparticle co-methyl methacrylate (PAN-co-MMA) nanospheres using a
matrix exhibited a significant biocatalytic activity with excellent en- precipitation–polymerization technique and ethylenediamine by
zyme stability and without apparent original activity loss. The recov- covalent attachment with carbonyl groups of PAN-co-MMA nano-
ered specific activities were 70% and 95% for CGTase and alcohol particles. This preparation was used for the immobilization of
oxidase, respectively. Similarly, bioconjugates of an enzyme and β galactosidase using glutaraldehyde as a coupling agent. The catalytic
gold nanorods by fabrication via electrostatic interactions between activity of the immobilized β galactosidase was 22.35 μM min−1 g−1
anionic GOX and positively charged gold nanorods were constructed. of nanospheres. Operational, thermal and storage stabilities were
The assembled processes were monitored by UV–vis spectra, zeta po- found to increase upon immobilization. Different factors affecting the
tential measurements and transmission electron microscopy (TEM). modification and activation processes were studied and the resultant
The enzyme activity of the obtained bioconjugates displayed relative- activated nanospheres were characterized by FT-IR, TGA SEM. Polyure-
ly enhanced thermostability behavior in contrast with that of free en- thane powder from hexamethylene diisocyanate and butanediol has
zyme. Free GOX in solution had lost 78% of its relative activity at 90 °C also been exploited to immobilize Penicillium canescens β galactosidase
while enzyme immobilized on gold nanorods retained about 39.3% by covalent binding between the amino groups of enzyme and the iso-
activity under similar incubation conditions (Ma and Ding, 2009). cyanate groups in polyurethane powder (Budriene et al., 2005). Candida
Nanosized dispersions of platinum have gained considerable at- antarctica lipase B is one of the most recognized biocatalysts used in a
tention because of their size-dependent electrical, chemical and opti- broad range of synthetic applications of industrial importance including
cal properties. The catalytic and electrochemical activity of highly kinetic resolutions, aminolysis, esterification and trans-esterification
dispersed platinum nanoparticles has been the focus of keen interest (Anderson et al., 1998; Ghanem, 2008; Schmid and Verger, 1998).
in the fabrication of amperometric biosensors (Genies et al., 1998; Li Several workers have reported increased utilization of immobilized
et al., 2007; Park and Cheon, 2001; You et al., 2003). Platinum nano- enzyme in chemical transformations and polymerization reactions
particle-doped sol–gel solution was used as a binder for multi-walled (Kumar et al., 2000; Mahapatro et al., 2004). Miletic et al. (2010) have
carbon nanotubes (CNT) for the fabrication of electrochemical sen- immobilized lipase on polystyrene nanoparticles and found that the
sors (Yang et al., 2006). The resulting CNT-silicate material brought activity of immobilized enzyme was remarkably improved as compared
new capabilities for electrochemical devices by using synergistic to the free enzyme.
S.A. Ansari, Q. Husain / Biotechnology Advances 30 (2012) 512–523 517

Table 2
Enzymes immobilized on non magnetic nanoparticles and their applications.

Enzyme Nanoparticle used Applications Kinetic parameters References

Glucose oxidase Thiolated gold nanoparticle Estimation of glucose level Immobilized enzyme: Pandey et al., 2007
up to 300 mg mL−1 Km = 3.74 mM, soluble
enzyme = 5.85 mM
Lipase Polystyrene nanoparticle Aminolysis, esterification, – Miletic et al., 2010
trans-esterification
α-Chymotrypsin Polystyrene nanoparticles Proteolysis (cleaves peptide Immobilized enzyme: Jia et al., 2003
amide bonds) Km = 31.7 μM, kcat = 20.0 s−1;
soluble enzyme:
Km = 47.8 μM, kcat = 17.8 s−1
β galactosidase POS-PVA GOS synthesis Immobilized enzyme: Neri et al., 2009
k1 = 1.41 h−1;
soluble enzyme: k1 = 1.16 h−1
CGTase + alcohol oxidase Activated cellulose nanocrystal/gold Enhanced enzyme _ Mahmoud et al., 2009
nanoparticle matrix loading and stability
Diastase Silica coated nickel nanoparticle Starch hydrolysis Immobilized enzyme: Prakasham et al.,
Km = 8,414 mM, 2007
Vmax = 4.92 μM min−1 mg−1;
soluble enzyme:
Km = 10,176 mM,
Vmax = 2.71 μM min−1 mg−1

α-Chymotrypsin and lipase were immobilized in hierarchically- NADH were incorporated into two porous silica glass supports
ordered mesocellular mesoporous silica (HMMS) in a simple and ef- (30 nm and 100 nm pore size) to obtain effective shuttling of cova-
fective way by enzyme adsorption followed by glutaraldehyde cross- lently bound NADH between LDH and GDH. It was observed that
linking. It resulted in the formation of nanometer scale crosslinked the glass of 30 nm pore size afforded enzyme activities which were
enzyme aggregates (CLEAs) entrapped in the mesocellular pores of two-folds as observed for the glass of 100 nm pore size thereby indi-
HMMS (37 nm) which did not leach out of HMMS through narrow cating that the former provided better enzyme−cofactor integration.
mesoporous channels (13 nm). CLEA of α-chymotrypsin (CLEA-CT) A similar multienzyme system immobilized on nonporous polysty-
in HMMS showed high enzyme loading capacity and significantly in- rene particles of 500 nm diameter was only 2% active as the glass-
creased enzyme stability even after 2 weeks under a rigorously shak- supported system. It is believed that nanoporous structure of the
ing condition, while the free enzyme demonstrated a rapid glass supports enhanced molecular interactions among the immobi-
inactivation due to the enzyme leaching. Moreover, CLEA of lipase lized enzymes and cofactor, thus improving the catalytic efficiency
in HMMS retained 30% specific activity with greatly enhanced stabil- of the system. Applications of such catalysts in industrial chemical
ity (Kim et al., 2007). processing may provide efficient in situ cofactor regeneration, easy
Immobilization of enzymes and proteins on activated supports catalyst/cofactor reuse and simple product purification.
permitted simplification of the reactor design and may be used to im- Synthesis of GOS by A. oryzae β galactosidase immobilized on POS-
prove some enzyme properties (Gaberc-Porekar and Menart, 2001; PVA was reported by Neri et al. (2008). A maximum GOS concentration
Iyer and Ananthnarayan, 2008; Mateo et al., 2007). Supports contain- of 26% (w/v) of total sugars was achieved at 55% lactose conversion
ing epoxy groups proved useful to generate intense multipoint cova- from 50% (w/v) lactose solution at pH 4.5 and 40 °C. Trisaccharides
lent attachment with different nucleophiles placed on the surface of accounted for more than 81% of the total GOS produced. GOS formation
enzyme molecules (e.g., amino, thiol, hydroxyl groups). However, was not considerably affected by pH and temperature. Its formation was
the intermolecular reaction between epoxy groups and soluble en- unaffected by enzyme immobilization in the POS-PVA matrix, thus indi-
zymes was extremely slow. Here, enzyme immobilization was per- cating the absence of diffusional limitations in the enzyme carrier. Fur-
formed via a two-step process: (i) an initial physical or chemical thermore, this immobilized derivative could be reused 10 times with
intermolecular interaction of the enzyme surface with the new func- the retainment of nearly 84% of the initial activity. Wan et al. (2007)
tional groups introduced on the support surface, and (ii) a subsequent have exploited electrospun nanofibers bearing porphyrin pendants to
intense intramolecular multipoint covalent reaction between the nu- covalently immobilize catalase thereby improving the activity and sta-
cleophiles of the immobilized enzyme and the epoxy groups of the bility of the immobilized enzyme. The facilitation of electron transfer
supports. The first immobilization might involve different enzyme re- between the immobilized enzyme and the ability of porphyrin pen-
gions which were further rigidified by multipoint covalent attach- dants to retain the active conformation of the catalase might be respon-
ment (Mateo et al., 2007). sible for the improvement in its activity and stability.
Liu et al. (2009) reported covalent attachment of NAD(H) to silica Hamlin et al. (2007) have reported a successful immobilization and
nanoparticles and found successful coordination of particle-immobi- improved stabilization of β galactosidase on polyelectrolyte surfaces. β
lized enzymes which enabled multistep biotransformation. Moreover, Galactosidase was covalently immobilized in a channel of a borosilicate
silica nanoparticle-attached glutamate dehydrogenase, LDH and glass microchip by using channel specific coating technology (Xiong
NAD(H) were prepared and applied to catalyze the coupled reactions and Regneir, 2001). Immobilization of β galactosidase onto polyelectro-
for production of α-ketoglutarate and lactate with the cofactor regener- lyte multilayer assemblies of the polyanion, poly [1-[4-(3-carboxy-4-
ated within the reaction cycle. The use of particle-attached cofactor hydroxyphenylazo) benzenesulfonamido]-1,2-ethanediyl, sodium salt]
promises a new biochemical processing strategy for cofactor-depen- (PAZO) and the polycation, PEI constructed by electrostatic self-assem-
dent biotransformation. bly has been monitored. A single layer of β galactosidase was deposited
In another study, multistep reactions catalyzed by a covalently over a precursor film comprising up to five bilayers of the PEI/PAZO
immobilized enzyme−cofactor−enzyme system were achieved (El- polyelectrolyte pair. The enzyme was deposited on both PEI and PAZO
Zahab et al., 2004). LDH, glucose dehydrogenase (GDH) and cofactor surfaces.
518 S.A. Ansari, Q. Husain / Biotechnology Advances 30 (2012) 512–523

Quartz crystal microbalance with dissipation monitoring, single- in biocatalytic and biosensing applications. Similarly, chitosan nano-
wavelength ellipsometry and UV–visible absorption spectroscopy particles were prepared and used as carriers for immobilizing enzyme
revealed differences in both the amount of β galactosidase incorpo- (Tang et al., 2007). They choose various methods for preparing chitosan
rated in each of the multilayer assemblies and the resulting enzyme nanoparticles and the effect of several factors such as the molecular
packing density in the films. The enzymatic films were immersed in weight of chitosan, concentration of chitosan, TPP concentration and so-
a reaction solution containing o-nitrophenyl β-D-galactopyranoside lution pH on the size of chitosan nanoparticles were studied. The results
and absorbance measurements were used to monitor the concentra- showed that solution pH, TPP concentration and chitosan concentration
tion of o-nitrophenol. Although the data indicated that a comparable affected the size of chitosan nanoparticles significantly. The adequacy of
amount of β galactosidase was incorporated onto both surfaces, enzy- the model equation for predicting the orders of the size of chitosan
matic activity was substantially inhibited when β galactosidase was nanoparticles was verified effectively by the validation data. The mini-
immobilized on the polyanionic surface compared to the enzyme on mum particle size was about 42± 5 nm. The optimal conditions of im-
the polycationic surface. The difference in catalytic activities reflects mobilization were as follows: 1 mg of neutral proteinase was
the different abilities of two polyelectrolytes to screen the protein ac- immobilized on chitosan nanoparticles for about 15 min at 40 °C and
tive site from the substrate environment. In both assemblies, the pro- the obtained enzyme activity yield was 84.3%.
tein interpenetrated the PEI/PAZO multilayer, disrupting the J- A successful immobilization of GOX on the surface of silica nano-
aggregated state of the PAZO chromophores. This work demonstrated particles (SNPs) was done via physical entrapment within photo-po-
that the charge, conformation and composition of underlying poly- lymerized hydrogels prepared from two different molecular weights
electrolyte cushions have a significant effect on the structure and (575 and 8000 Da) of polyethylene glycol (PEG) (Jang et al., 2010).
function of an immobilized protein within functional nanoassemblies The hydrogel entrapment resulted in a decrease in reaction rate and
(Hamlin et al., 2007). Chitosan, modified by linolenic acid and 1- an increase in apparent Km of silica nanoparticle-immobilized GOX,
ethyl-3-(3-dimethylaminopropyyl)-carbodiimide coupling displayed but these negative effects were minimized by using hydrogel with a
improved enzyme loading capacity (0.04 to 37.57%) with increasing higher MW PEG, which provided higher water content and larger
BSA concentration (Liu et al., 2005). mesh size. The catalytic rate of the PEG 8000 hydrogel was about
Daubresse et al. (1996) reported immobilization of alkaline phos- ten times faster than that of the PEG 575 hydrogel because of en-
phatase by physical entrapment within colloidal particles produced hanced mass transfer. A long-term stability test demonstrated that
by inverse micro-emulsion polymerization. Functionality was inde- SNP-immobilized GOX entrapped within the hydrogel maintained
pendently imparted to the nanoparticle surface by copolymerization more than 60% of its initial activity after a week, whereas non-entrapped
of acrylamide, N, N′ methylene-bis-acrylamide with N-acryloyl-1,6- SNP-immobilized GOX and entrapped GOX without SNP immobilization
diaminohexane and acrylic acid. They studied the effect of functional retained less than 20% of their initial activity. Incorporation of SNPs into
co-monomers on the size and zeta potential of the reactive latexes. hydrogel enhanced the mechanical strength of the hydrogel six-fold rel-
Integrity of the immobilized enzyme was ascertained from its catalyt- ative to bare hydrogels. Moreover, a hydrogel microarray entrapping
ic activity towards hydrolysis of p-nitrophenylphosphate. This meth- SNP-immobilized GOX was fabricated using photolithography and suc-
od also conferred the immobilized enzyme with greater stability cessfully used for quantitative glucose detection. Nattokinase purified
against environmental changes such as pH and temperature, as well from Bacillus subtilis using ionic exchange chromatography was success-
as higher selectivity toward the substrates. Kim et al. (2006a, b) fully immobilized upon polyhydroxybutyrate nanoparticles (Deepak et
have demonstrated successful immobilization of M. javanicus lipase al., 2009). Immobilization resulted in the enhanced stability with a 20%
on silica nanoparticles. The attachment of glutaraldehyde and 1,4 increase in enzyme activity. Moreover, the activity was completely
phenylene diisothiocyanate onto the ethylene diamine-activated sili- retained for 25 days upon storage at 4 °C. β Galactosidase from Kluyvero-
ca nanoparticles resulted in a remarkable increase in the enzyme myces lactis was covalently immobilized onto a mPOS-PVA using glutar-
loading capacity and activity. Immobilized lipase retained a high aldehyde as activating agent. A soluble and immobilized enzyme
level of activity over a wider range of pH than the free form. Thermal displayed the same pH-optimum (6.5) and temperature-optimum
stability was considerably enhanced by the immobilization. Since this (50 °C). However, the immobilized enzyme showed a marked increase
nanoparticle-based enzyme immobilization preserves high enzyme in stability against various physical and chemical denaturants. The appar-
app
loading and activity with enhanced stability, it can be applied to var- ent Km and activation energy for both soluble and immobilized β galac-
ious enzymatic applications, such as biosensors and bioremediations. tosidases were evaluated. Immobilized β galactosidase presented higher
In the past, Li et al. (2003a, b) have succeeded in coupling pepsin to operational, greater lactose hydrolyzing activity and thermal stability
alumina in an orientation-specific manner. A comparative analysis than the soluble enzyme.
revealed that conjugated pepsin retained higher enzymatic activity In the author's laboratory, ZnO nanoparticles (ZnO-NP) were
upon immobilization mainly because of the lack of diffusion limita- exploited for the immobilization of A. oryzae β galactosidase by sim-
tions of the substrate. Additionally, upon attachment to the alumina ple bioaffinity based adsorption. Ansari and Husain (2011a, b) dem-
nanoparticles, the thermal stability of pepsin was enhanced. Mao et onstrated a novel and an efficient method for the immobilization of
al. (2006) developed novel core–shell nanoparticles consisting of A. oryzae β galactosidase by using concanavalin A (Con A) layered
poly(methyl-methacrylate) cores coated with synthetic polymer zinc oxide nanoparticles as bioaffinity support. AFM analysis showed
and other biopolymers shells via direct graft copolymerization of that the prepared matrix has an advantageous microenvironment and
methyl methacrylate. Much greater enzymatic yield (90%) was large surface area available for binding a significant amount of en-
achieved by this novel procedure of enzyme immobilizations. Naik zyme. Con A layered ZnO-NP retained 84% of the enzyme activity
et al. (2004) demonstrated the entrapment of cobalt–platinum and upon immobilization. Soluble and immobilized β galactosidase exhib-
cadmium–selenium and zinc sulfide nanoparticles within the silica ited the same pH-optimum at pH 4.5 while the temperature-opti-
matrix. Both free and entrapped cadmium–selenium and zinc sulfide mum was increased from 50 °C to 60 °C for the immobilized
nanoparticles exhibited similar emission profiles. It was observed that enzyme. The free enzyme lost 81% activity when it was exposed at
addition of magnetic nanoparticles to the entrapped enzyme–silica 60 °C for 2 h whereas an immobilized enzyme retained 60% activity
matrix facilitated easy separation of the entrapped enzyme from the under similar experimental conditions. Michaelis constant, Km was
product. Iron oxide nanoparticles were entrapped along with the en- 2.38 mM and 5.88 mM for free and immobilized β galactosidase, re-
zyme in a silica matrix using the bio-silicification reaction which spectively. Vmax for the soluble and the immobilized enzyme was
resulted in the reusability of entrapped enzyme after magnetic separa- 0.520 mM/min and 0.500 mM/min, respectively. The functional
tion. Biomolecules entrapped using this method is widely applicable groups present in native and parent compounds were monitored by
S.A. Ansari, Q. Husain / Biotechnology Advances 30 (2012) 512–523 519

FTIR. Immobilized enzyme retained more than 80% activity even after nanoparticles. The biosensor showed excellent electro-catalytical re-
its 6th repeated use. In another study, comparative stability studies of sponse to the oxidation of glucose when ferrocene-methanol was
β galactosidase immobilized on native ZnO and ZnO-NP were investi- used as an artificial redox mediator. It exhibited good stability, repro-
gated. The activity yield of immobilization was 85% for the enzyme ducibility and long life time. Furthermore, Kiapp of 2.7 mM for GOX on
adsorbed on ZnO-NP as compared to the enzyme adsorbed on native the nickel oxide nanoparticles exhibited excellent bio-electrocatalytic
ZnO, which retained only 60% β galactosidase upon immobilization. activity of immobilized enzyme toward glucose oxidation. In addition,
ZnO-NP adsorbed β galactosidase exhibited greater thermal, storage this glucose biosensor showed fast amperometric response (3 s) with
and operational stability as compared to ZnO adsorbed β galactosi- a sensitivity of 446.2 nA mM−1, detection limit of 24 μM and wide con-
dase. In addition, ZnO-NP adsorbed β galactosidase hydrolyzed a centration range of 30 μM to 5 mM.
greater amount of lactose from milk and whey as compared to simply A novel disposable biosensor for rapid determination of H2O2 was
ZnO adsorbed β galactosidase (Husain et al., 2011). prepared by entrapping HRP in a colloidal gold nanoparticle modified
In the recent past, Al2O3-NPs has been exploited as a potential candi- chitosan membrane (Au-chitosan) to modify the ITO electrode. This
date in targeted drug delivery and biosensor application (Frey et al., 1997; membrane was very efficient for retaining the enzyme activity and
Heilmann et al., 2003) but no study has been carried out which could preventing its leakage out of the membrane. The biosensor was char-
strengthen its importance in immobilizing industrially important en- acterized by SEM, AFM and electrochemical methods. Parameters af-
zymes. Thus, a high yield immobilized preparation was obtained by spe- fecting the performance of the biosensor, including concentrations
cific adsorption of K. lactis β galactosidase on a highly efficient and of o-phenylenediamine and pH of substrate solution were optimized
selective bioaffinity support, Con A layered Al2O3-NPs. TGA of bioaffinity and the linear calibration range was obtained from 0.01 to 0.5 mM
support revealed 6% loss in weight at 600 °C whereas its thermal decom- with a correlation coefficient of 0.997 (n = 8). The amperometric re-
position was observed at 350 °C by DTA. No significant change was no- sponse of the biosensor did not show an obvious decrease after the
ticed in the band intensity of pUC19 plasmid upon its treatment with substrates were injected continuously 34 times into the flow cell.
Con A layered Al2O3-NPs. Comet assay further exhibited negligible change The prepared biosensor was economic due to the low-cost of ITO
in tail length of comet after treating the lymphocytes by bioaffinity ma- film electrode obtained from industrial mass production. It enabled
trix. AFM revealed a large surface area of Con A layered Al2O3-NPs for a simple, sensitive and rapid determination for H2O2 and thus, pro-
binding higher amounts of the enzyme. Moreover, FTIR has shown bind- vided a useful platform for the development of biosensors. The bio-
ing of β galactosidase on bioaffinity support by exhibiting a broadening in sensor possesses good detection precision, acceptable accuracy and
peaks at 3220.61 cm−1 and 3447.27 cm−1. Soluble and immobilized β ga- storage stability for fabrication in batch (Lin et al., 2007). The devel-
lactosidase exhibited the same pH-optima at pH 7.0. However, the immo- oped sensor showed rapid electrocatalytic response for monitoring
bilized enzyme exhibited enhanced pH stability and a broad spectrum H2O2 and provided a promising platform for the development of bio-
temperature optimum. Immobilized β galactosidase was found to be sensors, affinity supports and immobilized enzyme reactors.
highly stable against product inhibition by galactose and retained 85% ac- Glucose oxidase was immobilized on tin oxide using a membrane
tivity after its sixth repeated use. Due to easy production, excellent tem- entrapment method which provided a simple process for fabricating
perature stability, non-toxic nature and greater specificity of the enzyme electrode. The immobilized glucose oxidase showed a rela-
bioaffinity support, it might serve as a powerful biorecognition probe in tively large current response as compared to other glucose biosen-
biosensor applications (Ansari and Husain, 2011a, b). sors. It exhibited good reproducibility and stability. The sensitivity
of the biosensor was 19.55 μA mM −1 and a linear response was ob-
5. Nanoparticles-based enzymes in biosensors served between 0 and 3 mM glucose. Such an amperometric glucose
biosensor can be used for detecting glucose in blood, urine, beverages
Special attention has been given to bioassay applications using and fermentation systems (Park et al., 2008).
nanomaterials to develop biosensors, biomedical devices, biofuel β Galactosidase was covalently immobilized onto a gold-coated
cells and bioreactors (Akella and Mitra, 2007; Cui and Gao, 2003; magneto-elastic film via a self assembled monolayer (SAM) of ω-car-
Penn et al., 2003; Ramanavicius et al., 2005). DNA, antibodies and en- boxylic acid alkylthiol containing terminal carboxylic group. The en-
zymes have been immobilized to nanostructure materials by cova- zyme was then attached to the monolayer via carbodiimide/N-
lent binding, adsorption and entrapment to develop sensitive hydroxysuccinimide-mediated coupling (Ball et al., 2003). The immo-
biosensors and bioassay methods (Li et al., 2003a, b; Liu and Ju, bilized enzyme exhibited a Km of 1.2 mM by using indolyl galactopyr-
2002). Recently, a large number of enzyme biosensors based on anoside as substrate. Kinetic parameters for non-precipitating and
nanoparticles (Besteman et al., 2003; Xiao et al., 2003) or nanotubes precipitating enzymatic substrates were examined by competitive in-
have been developed. The ability to tailor the properties of nanoma- hibition. Moreover, this method could also be employed for screening
terials offers excellent prospects for enhancing the performance of enzyme inhibitors or determining concentrations of enzyme sub-
enzyme-based biocatalytic sensors. strates or inhibitors based on wireless, magnetoelastic transduction.
Biosensors has many advantages such as high sensitivity, fast re- Li et al. (2003a, b) prepared a novel method for immobilizing an-
sponse, low cost, immunity from electrical and magnetic disturbances, tibodies (antigens) based on magnetic nanoparticles for piezoelectric
geometrical versatility, remote sensing capability, small size and light- immunoassay. The goat-anti-IgG antibody as the model analyte was
weight, and can provide an alternative to laboratory-based measure- first covalently immobilized to magnetic nanoparticles which were
ment methods (D'souza, 2001; Kishen et al., 2003). Salimi et al. surface modified with amino-groups. The magnetic bionanoparticles
(2007) have successfully co-deposited GOX on nickel-oxide (NiO) thus formed were attached to the surfaces of quartz crystal with the
nanoparticles at a glassy carbon electrode. Cyclic voltametry was used help of a permanent magnet. The detection of IgG was performed
for electro-deposition of NiO nanoparticle and GOX immobilization. with the prepared sensor. The piezoelectric immunosensor can deter-
The direct electron transfer of immobilized GOX displayed a pair of mine IgG in the range of 0.6–4.9 μg ml –1 with a detection limit of
well defined and nearly reversible redox peaks with a formal potential 0.36 μg ml –1. The magnetic bionanoparticles and immunocomplex
(E0′) of −0.420 V in pH 7.0 phosphate buffer and the response showed layer could be removed easily by taking away the magnetic field,
a surface controlled electrode process. The surface coverage and hetero- making the regeneration of a piezoelectric sensor easier and more
geneous electron transfer rate constant of GOX immobilized on NiO film efficient.
glassy carbon electrode were 9.45 × 10−13 mol cm− 2 and 25.2±0.5 s −1 A mediator less biosensor was fabricated with a double-sided micro-
which indicated a high enzyme loading ability of the NiO nanoparticles porous gold electrode by successively immobilizing a mixed self-assem-
and great facilitation of the electron transfer between GOX and NiO bled monolayer comprising of carboxylic acid and thiol-terminated
520 S.A. Ansari, Q. Husain / Biotechnology Advances 30 (2012) 512–523

thiolate (DL-thiorphan and 1,8-octanedithiol, GOX) and finally a gold With an increasing H2O2 concentration the amperometric response of
nanoparticle (Au-NP) on one working side. The double-sided micropo- the sensor increased. The linear response range of the sensor to H2O2
rous gold electrodes were formed by plasma sputtering of gold on a po- concentration was from 1.7 × 10−6 to 5.3 × 10 −4 mol L −1 with a corre-
rous nylon substrate, yielding a face-to-face type two-electrode lation coefficient of 0.998 (n = 12) and a detection limit of
electrochemical cell. While the straight chain molecule 1,8-octane- 9.2 × 10−7 mol L −1 at a signal-to-noise ratio of 3. The apparent Michae-
dithiol forms a dense insulating monolayer, the side armed DL-thior- lis–Menten constant was determined to be 0.31 mM L −1. The storage
phan forms a low density layer for the diffusion of redox couples to stability of the sensor was examined by amperometric response at
the electrode surface. The mixed self-assembled monolayer not only H2O2 concentration of 106 μM L −1. After 60 days of storage and inter-
provided the linking functional groups for both enzyme and gold nano- mittently measuring every 5 days, the results showed that the response
particles but also resulted in an appropriately spaced monolayer for di- of the sensor still maintain about 80% of the original values (Zhang et al.,
rect electron transfer process from the center of the redox enzyme to 2004a, b).
the electrode surface. GOX was covalently immobilized onto carboxyl- Recently, single enzyme nanoparticles have been used for various
ic-acid terminated monolayer gold nanoparticles by simply dispensing applications in bioscience research from biosensors to new drug de-
it in colloidal gold solution. It was observed that the resulting ampero- velopment, food, environmental monitoring, proteomics, biomarker
metric biosensor exhibited quantitatively the same response to glucose analysis and in virus detection. Biological signal transducers are wide-
in the presence and of dissolved oxygen, which provides evidence that ly used for the manufacture of nanobiosensors (Bhinder and Dadra,
the gold nanoparticles immobilized on and around the GOX promote di- 2009). Kishen et al. (2003) have developed a fiber optic biosensor in
rect electron transfer from the enzyme to the electrode, assuming the the recent past to monitor Streptococci activity in human saliva. Wu
role of a common redox mediator (Zhang et al., 2006). The steady- et al. (2009a, b) presented a novel bionanocomposite film consisting
state response of H2O2 biosensor was measured by using selenium of glucose oxidase/platinum/functional grapheme sheets/chitosan
nanoparticles. It achieved 95% of steady-state currents within 10 s. for glucose sensing. The biosensor exhibited good sensitivity with a
detection limit of 0.6 μM glucose and better responses due to its
large surface area and fast electron transfer of graphene and Pt nano-
Table 3
Nanoparticles exploited to synthesize enzyme based biosensors.
particles. It had good reproducibility and long-term stability. The in-
terfering signals from ascorbic and uric acids were negligible. This
Enzyme Type of nanoparticle Application References work showed that the hybrid GOD/Pt/FGS/chitosan nanocomposite-
immobilized
based biosensor has great potential for clinical utility and home care
Glucose Selenium H2O2 biosensor, Zhang et al., for rapid monitoring of glucose. The graphene-based biosensor fabri-
oxidase nanoparticle determination of free 2004a, b
cation method demonstrated in this work was readily applicable to
glucose in body fluids,
food and agricultural the fabrication of other biosensors based on oxidases such as biosen-
products sors for cholesterol, alcohol, lactate, acetylcholine, hypoxanthine and
Uricase ZnO nanoparticle Uric acid biosensor, Zhang et al., xanthine (Wang and Lin, 2008). Several other nanoparticle based en-
evaluation of serum 2004a, b
zymes that have been utilized to develop biosensors of industrial im-
uric acid
Glucose Gold nanoparticle Biosensors, catalytic Yang et al., 2006;
portance and clinical applications have been summed up in Table 3.
oxidase nanodevice to Li et al., 2007 A reagentless uric acid biosensor based on uricase immobilized on
construct nanoreactors ZnO nanorods was developed by Zhang et al. (2004a, b). ZnO nanorods
Laccase Zinc-tetra-amino- Fiber optic biosensor, Huang et al., derived electrode retained enzyme activity and could enhance the elec-
phthalocyanine- catechol biosensor and 2007; Zhang et
tron transfer between the enzyme and the electrode. This sensor showed
Fe3O4, laccase- in degrading bisphenol al., 2007;
modified silica A Galliker et al., a high thermal stability up to 85 °C and an electrocatalytic activity to the
nanoparticles, 2010 oxidation of uric acid without the presence of an electron mediator. The
magnetic core–shell electrocatalytic response showed a linear dependence on the uric acid
(Fe3O4–SiO2) concentration ranging from 5.0×10−6 to 1.0×10−3 mol L−1 at 3σ. The
nanoparticles
Glucose Nickel oxide Amperometric Salimi et al.,
biosensor developed for phenol detection on ZnO nanorods exhibited
oxidase nanoparticle biosensor and 2007 good performance showing response time less than 5 s upon adding phe-
electrochemical nol to the buffer solution. The sensitivity and detection limit were im-
investigation of glucose proved to 103.08 μA mM−1 and 0.623 μM due to the pretreatment of
oxidase
the gold electrode (Gu et al., 2009).
Glucose Platinum Biosensors, naked eye Zhao et al., 2007;
oxidase nanoparticle, gold detection of glucose in Radhakumary
nanoparticles urine and Sreenivasan, 6. Nanobiocatalysis
2011
Urease Silver nanoparticle, Biosensors, analysis of Crespilho et al., Driven by the recent advances in molecular biotechnology and
phosphonate grafted urea content in blood, 2009, Sahoo et
iron oxide urine, alcoholic al., 2011
nanoscale science and engineering, development of nanobiocatalysts
nanoparticle beverages, natural has shown a multiscale trend that includes the optimization of protein
water and molecular structures, manipulation of nanoscale environments of the
environmental enzymes, microscale properties of materials and fluids and operational
wastewaters
factors of macroscale reactors (Wang, 2009). Compared with enzymes
Peroxidase Gold-chitosan Rapid deterioration of Lin et al., 2007
nanoparticle H2O2, water treatment, immobilized on micrometric supports, a nanobiocatalyst could achieve
pharmaceutical and a much higher enzyme loading capacity and significantly enhanced
biomedical applications mass transfer efficiency. Recent advances of nanobiocatalytic ap-
Glucose Pt/FGS/chitosan, Glucose biosensor, Wu et al., 2009a, proaches have improved the performance of protein digestion by
oxidase silica nanoparticle quantitative estimation b, Jang et al.,
of glucose 2010
using various nanomaterials such as nanoporous materials, magnetic
Cyclodextrin Cellulose-silver Biosensors, Ma and Ding, nanoparticles and polymer nanofibers. Trypsin stabilization was
glycosyl nanoparticle determination of 2009; Mahmoud achieved in the form of trypsin-coated nanofibers which showed negli-
transferase, methanol and methyl et al., 2009; Ling gible decrease in activity after repeated uses for 1 year and exhibited
alcohol ester content of pectin and Heng, 2010
greater resistance to proteolysis, therefore it can be employed in the de-
oxidase
velopment of automated, high-throughput and on-line digestion
S.A. Ansari, Q. Husain / Biotechnology Advances 30 (2012) 512–523 521

systems. This novel method implicates improved performance of pro- full occupation of the surface of the support by the enzyme. More-
tein digestion in speed, detection sensitivity, recyclability and trypsin over, the use of pure enzymes avoids undesired side reactions cata-
stability (Kim et al., 2010). A simple and unique method was developed lyzed by contaminant enzymes of crude preparations. To overcome
to prepare bioactive materials through a spin coating process (Tong et the problems associated with enzyme purification like time-consum-
al., 2008). These workers have investigated the preparation of protease, ing, high cost, enzyme inactivation, etc., one suitable solution would
subtilisin carlsberg-coated plastic films and examined their activity for be to exploit nanoparticles for immobilization by highly selective im-
hydrolysis of chicken egg albumin (CEA). This novel method produced mobilization procedures. The utility of such highly efficient matrices
enzymatic coatings with a typical loading of 13 μg cm−2 and retained can be further extended in the development of therapeutic agents
46% chicken egg albumin activity in aqueous solutions and exhibited re- as novel targeted drug delivery for treating cancer patients across
markably improved thermal stability. the globe.
Wang et al. (2009) have developed an attractive approach for fab-
ricating a nanobiocatalyst by immobilizing chloroperoxidase on iron
oxide MNPs which were cheap and biocompatible with a core con- Acknowledgments
taining multiple iron oxide MNPs and a thick polymer shell. Several
enzymes such as hydrolase, glucose oxidase and alcohol dehydroge- The authors are thankful to the Council of Science and Technology,
nase have been immobilized on such MNPs in the recent past with Lucknow, Uttar Pradesh for funding the project entitled “Immobiliza-
great success (Blakemore, 1975; Kim et al., 2008; Petros and DeSi- tion of plant and fungal β galactosidase by using bioaffinity supports
mone, 2010). —its application in the hydrolysis of lactose in whey and milk.” The
Ginet et al. (2011) have functionalizing lipid-coated nano-magnets authors extend their thanks to Dr. Humaira Ashraf and Dr. Rukhsana
with phosphohydrolase and successfully engineered an easy-to-purify, Satar for their critical reading and providing valuable suggestion in
robust and recyclable biocatalyst to degrade ethyl-paraoxon, a commonly preparing this manuscript.
used pesticide. They genetically fused the opd gene from Flavobacterium
sp. ATCC 27551 encoding a paraoxonase tomamC, an abundant protein References
of the magnetosome membrane in Magnetospirillum magneticum AMB-
1. The MamC protein acts as an anchor for the paraoxonase to the magne- Akella S, Mitra CK. Electrochemical studies of glucose oxidase immobilized on glutathi-
one coated gold nanoparticles. Ind J Biochem Biophys 2007;44:82–7.
tosome surface, thus producing magnetic nanoparticles displaying phos-
Ali M, Winterer M. ZnO nanocrystals: surprisingly alive. Chem Mater 2010;22:85–91.
phohydrolase activity. Magnetosomes functionalized with Opd were Anderson EM, Karin M, Kirk O. One biocatalyst—many applications: the use of Candida
easily recovered from genetically modified AMB-1 cells. The catalytic antarctica B lipase in organic synthesis. Biocatal Biotransform 1998;37:1608–37.
Ansari SA, Husain Q. Designing and surface modification of ZnO nanoparticles for bio-
properties of the immobilized Opd which were measured by the hydroly-
medical applications. Food Chem Toxicol 2011a;49:2107–15.
sis of ethyl-paraoxon exhibited Km (and kcat) values of 58 μM (and Ansari SA, Husain Q. Immobilization of Kluyveromyces lactis β galactosidase on conca-
178 s− 1) and 43 μM (and 314 s− 1) for the immobilized and purified en- navalin A layered aluminium oxide nanoparticles — its future aspects in biosensor
zyme, respectively. The immobilized enzyme was found to be quite stable applications. J Molec Catal B: Enzym 2011b;70:119–26.
Ball JC, Puckett LG, Bachas LG. Covalent immobilization of β galactosidase onto a gold-
over repeated uses for pesticide degradation and thus it makes it applica- coated magnetoelastic transducer via a self-assembled monolayer: toward a mag-
ble for pesticide bioremediation in contaminated effluents. netoelastic biosensor. Anal Chem 2003;75:6932–7.
Magnetosomes are nano-sized particles of magnetite (Fe3O4) or Bayramoglu G, Yilmaz M, Senel AU, Arica MY. Preparation of nanofibrous polymer
grafted magnetic poly(GMA-MMA)-g-MAA beads for immobilization of trypsin
greigite (Fe3S4) that are biomineralized by a phylogenetically diverse via adsorption. Biochem Eng J 2008;40:262–74.
group of prokaryotes, the magnetotactic bacteria. They cover a broad Besanger TR, Chen Y, Deisingh AK, Hodgson R, Jin W, Mayer S, et al. Screening of inhib-
range of applications such as magnetic resonance imaging, hyperther- itors using enzymes entrapped in sol–gel-derived materials. Anal Chem 2003;75:
2382–91.
mia cancer treatment or molecules and cell sorting. Functional soluble Besteman K, Lee J, Wiertz FGM, Heering HA, Dekker C. Enzyme-coated carbon nano-
proteins (luciferase) have been grafted on magnetosome surfaces by tubes as single-molecule biosensors. Nano Lett 2003;3:727–30.
chemical coupling of specific ligands to membrane lipids or proteins, Bhat RR, Genzer J, Chaney BN, Sugg HW, Liebmann-Vinson A. Controlling the assembly
of nanoparticles using surface grafted molecular and macromolecular gradients.
or displayed by gene fusion using as an anchor proteins specifically tar-
Nanotechnology 2003;14:1145–52.
geted to the magnetosome membrane (Lang et al., 2007). The latter ap- Bhinder SS, Dadra P. Application of nanostructures and new nanoparticles as advanced
proach circumvents the purification steps, as the enzyme is genetically biomaterials. Asian J Chem 2009;21:167–71.
Blakemore R. Magnetotactic bacteria. Science 1975;190:377–9.
targeted to the membrane surrounding the crystal: subsequently, the
Bruno LM, Coelho JS, Melo EHM, Lima-Filho JL. Characterization of Mucor miehei lipase
functionalized magnetite particles can be magnetically recovered after immobilized on polysiloxane-polyvinyl alcohol magnetic particles. World J Microb
a simple disruption of the cells. The production of magnetotactic bacte- Biotechnol 2005;21:189–92.
ria can be achieved with a reasonable yield under controlled conditions Budriene S, Gorochovceva N, Romaskevic T, Yugova LV, Miezeliene A, Dienys G, et al. β
Glactosidase from Penicillium canescens: properties and immobilization. Centr Eur J
in bioreactors. In another study, single-step production of a reusable Chem 2005;3:95-105.
magnetic biocatalyst was carried out for bioremediation of pollutants Caruso F, Mohwald H. Protein multilayer formation on colloids through a stepwise self-
from soil and water by utilizing in vivo immobilization of a bacterial assembly technique. J Am Chem Soc 1999;121:6039–46.
Crespilho FN, Ghica ME, Florescu M, Nart FC, Oliveira ON, Brett CMA. A strategy for enzyme
phosphotriesterase. This dimeric zinc metalloenzyme was able to hy- immobilization on layer-by-layer dendrimer-gold nanoparticle electrocatalytic mem-
drolyze organophosphorous neurotoxins such as pesticides (i.e. para- brane incorporating redox mediator. Electrochem Commun 2006;8:1665–70.
oxon) or some nerve agents (Van Dyk and Pletschke, 2011). Crespilho FN, Iost RM, Travain SA, Oliveira JrON, Zucolotto V. Enzyme immobilization
on Ag nanoparticles/polyaniline nanocomposites. Biosens Bioelec 2009;24:3073–7.
Cui D, Gao H. Advances and prospects of bionanomaterials. Biotechnol Prog 2003;19:683–92.
7. Concluding remarks D'souza SF. Microbial biosensors. Biosens Bioelect 2001;16:337–53.
Daubresse C, Grandfils C, Jérome R, Teyssie P. Enzyme immobilization in reactive nano-
particles produced by inverse microemulsion polymerization. Coll Polym Sci
Nanocrystalline and nanoporous metal oxide surfaces are serving 1996;274:482–9.
as novel matrices for enzyme immobilization. Recently, the use of Deepak V, Pandian SBRK, Kalishwaralal K, Gurunathan S. Purification, immobilization,
nanoparticles has emerged as a versatile tool for generating excellent and characterization of nattokinase on PHB nanoparticles. Biores Technol
2009;100:6644–6.
supports for enzyme stabilization due to their small size and large
Demira AS, Talpura FN, Sopacia SB, Kohring G-W, Celik A. Selective oxidation and re-
surface area. Nanoparticles strongly influence the mechanical proper- duction reactions with cofactor regeneration mediated by galactitol-, lactate- and
ties of the material like stiffness and elasticity and provide biocom- formate dehydrogenases immobilized on magnetic nanoparticles. J Biotechnol
patible environments for enzyme immobilization. The availability of 2011;152:176–83.
Dyal A, Loos K, Noto M, Chang SW, Spagnoli C, Shafi KVPM, et al. Activity of Candida
pure enzymes is another key factor in preparing very active immobi- rugosa lipase immobilized on Fe2O3 magnetic nanoparticles. J Am Chem Soc
lized biocatalysts for the development of biosensors since it facilitated 2003;125:1685–6.
522 S.A. Ansari, Q. Husain / Biotechnology Advances 30 (2012) 512–523

Eldin MMS, Zatahry AAE, Al-Sabah MB, Elaassar MR. Immobilization of β galactosidase Kim BC, Nair S, Kim J, Kwak JH, Grate JW, Kim SH, et al. Preparation of biocatalytic
onto copolymers nanoparticles of poly (acrylonitrile-co-methylmethacrylate): char- nanofibres with high activity and stability via enzyme aggregate coating on poly-
acterization and application to whey hydrolysis. Nanotechnol Conf 2010. U.S.A. mer nanofibres. Nanotechnology 2005a;16:S382–8.
El-Zahab B, Jia H, Wang P. Enabling multienzyme biocatalysis using nanoporous mate- Kim BK, Kim YH, Won K, Chang H, Choi Y, Kong K, et al. Electrical properties of polyani-
rials. Biotechnol Bioeng 2004;87:178–83. line nanofibre synthesized with biocatalyst. Nanotechnology 2005b;16:1177–81.
Fernandes EGR, Queiroz AAAD, Abraham GA, Roman JS. Anti-thrombogenic properties Kim J, Grate JW, Wang P. Nanostructures for enzyme stabilization. Chem Eng Sci
of bioconjugate streptokinase-polyglycerol dendrimers. J Mater Sci Mater Med 2006a;61:1017–26.
2006;17:105–11. Kim MI, Ham HO, Oh SD, Park HG, Chang HN, Choi SH. Immobilization of Mucor javani-
Frey A, Neutra MR, Robey FA. Peptomer aluminum oxide nanoparticle conjugates as sys- cus lipase on effectively functionalized silica nanoparticles. J Mol Catal B: Enzym
temic and mucosal vaccine candidates: synthesis and characterization of a conjugate 2006b;39:62–8.
derived from the C4 domain of HIV-1MN Gp120. Bioconj Chem 1997;8:424–33. Kim M, Kim J, Lee J, Jia H, Na HB, Youn JK, et al. Crosslinked enzyme aggregates in hier-
Gaberc-Porekar V, Menart V. Perspectives of immobilized-metal affinity chromatogra- archically ordered mesoporous silica: a simple and effective method for enzyme
phy: a review. J Biochem Biophys Methods 2001;49:335–60. stabilization. Biotechnol Bioeng 2007;96:210–8.
Galliker P, Hommes G, Schlosser D, Philippe F-X, Shahgaldian P. Laccase-modified silica Kim J, Grate JW, Wang P. Nanobiocatalysis and its potential applications. Trends Bio-
nanoparticles efficiently catalyze the transformation of phenolic compounds. J Coll technol 2008;26:639–46.
Interf Sci 2010;349:98-105. Kim H, Kwon HS, Ahn J, Lee CH, Ahn IS. Evaluation of a silica-coated magnetic nanoparticle
Gardimalla HMR, Mandal D, Stevens PD, Yen M, Gao Y. Superparamagnetic nanoparti- for the immobilization of a His-tagged lipase. Biocatal Biotrans 2009;27:246–53.
cle-supported enzymatic resolution of racemic carboxylates. Chem Commun Kim J, Kim BC, Lopez-Ferrer D, Petritis K, Smith RD. Nanobiocatalysis for protein diges-
2005;37:4432–4. tion in proteomic analysis. Proteomics 2010;10:687–99.
Genies L, Faure R, Durand R. Electrochemical reduction of oxygen on platinum nano- Kin MH, Xuepu M, Lianquan Gu, Pei Li. Facile route to enzyme immobilization: core–
particles in alkaline media. Electrochim Acta 1998;44:1317–27. shell nanoenzyme particles consisting of well-defined poly(methyl methacrylate)
Ghanem A. Lipase-catalyzed kinetic resolution of racemates: a versatile method in chi- cores and cellulase shells. Langmuir 2008;24:11036–42.
ral separation. Tetrahedron 2008;63:1721–32. Kishen A, John MS, Lim CS, Asundi A. A fiber optic biosensor (FOBS) to monitor mutant
Ginet N, Pardoux R, Adryanczyk G, Garcia D, Brutesco C. Single-step production of a recyclable Streptococci in human saliva. Biosens Bioelectron 2003;18:1371–8.
nanobiocatalyst for organophosphate pesticides biodegradation using functionalized Koneracka M, Kopcansky P, Antalmk M, Timko M, Ramchand CN, Lobo D, et al. Immo-
bacterial magnetosomes. PLoS One 2011;6:e21442. doi:10.1371/journal.pone.0021442. bilization of proteins and enzymes to the magnetic particles. J Magnet Magn Mater
Gole A, Dash C, Ramakrishnan V, Sainkar SR, Mandale AB, Rao M, et al. Pepsin–gold col- 1999;201:427–30.
loid conjugates: preparation, characterization and enzymatic activity. Langmuir Koneracka M, Kopcansky P, Timko M, Ramchand CN. Direct binding procedure of pro-
2001;17:1674–9. teins and enzymes to fine magnetic particles. J Magn Mater 2002;252:409–11.
Gu BX, Xu CX, Zhu GP, Liu SQ, Chen LY, Li XS. Tyrosinase immobilization on ZnO nanor- Konwarh R, Karak N, Rai SK, Mukherjee AK. Polymer-assisted iron oxide magnetic
ods for phenol detection. J Phys Chem B 2009;113:377–81. nanoparticle immobilized keratinase. Nanotechnology 2009;20:225–35.
Guncheva M, Tashev E, Zhiryakova D, Tosheva T, Tzokova N. Immobilization of lipase Kouassi GK, Irudayaraj J, McCarty G. Examination of cholesterol oxidase attachment to
from Candida rugosa on novel phosphorous-containing polyurethanes: Applica- magnetic nanoparticles. J Nanobiotechnol 2005;3:1–9.
tion in wax ester synthesis. Process Biochem 2011;46:923–30. Kumar CV, McLendon GL. Nanoencapsulation of cytochrome c and horseradish perox-
Gupta AK, Gupta M. Synthesis and surface engineering of iron oxide nanoparticles for idase at the galleries of a-zirconium phosphate. Chem Mater 1997;9:863–70.
biomedical applications. Biomaterials 2005;26:3995–4021. Kumar A, Gross RA, Jendrossek D. Poly (3-hydroxybutyrate)-depolymerase from Pseu-
Hamachi I, Fujita A, Kunitake T. Enhanced N-demethylase activity of cytochrome C domonas lemoignei: catalysis of esterifications in organic media. J Org Chem
bound to a phosphate-bearing synthetic bilayer membrane. J Am Chem Soc 2000;65:7800–6.
1994;116:8811–2. Kumar A, Mandal S, Selvakannan PR, Pasricha R, Mandale AB, Sastry M. Fractal gold
Hamlin RE, Dayton TL, Johnson LE, Johal MSA. QCM study of the immobilization of β ga- nanostructures produced by the spontaneous reduction of chloroaurate ions in
lactosidase on polyelectrolyte surfaces: effect of the terminal polyion on enzymatic thermally evaporated hexadecylaniline thin films. Langmuir 2003;9:6277–82.
surface activity. Langmuir 2007;23:4432–7. Kuroiwa T, Noguchi Y, Nakajima M, Sato S, Mukataka S, Ichikawa S. Production of chit-
Hansen MJ, Natale N, Kornacki P, Paszczynski AJ. Conjugating magnetic nanoparticles osan oligosaccharides using chitosanase immobilized on amylase coated magnetic
for application in health and environmental research. Paper presented at the nanoparticles. Process Biochem 2008;43:62–9.
48th Annual Idaho Academy of Science Meeting and Symposium, Moscow, Lang C, Schuler D, Faivre D. Synthesis of magnetite nanoparticles for bio- and nano-
Idaho; 2006. 4 June 2006. technology: genetic engineering and biomimetics of bacterial magnetosomes.
Heilmann A, Teuscher N, Kiesow A, Janasek D. Nanoporous aluminum oxide as a novel Macromol Biosci 2007;7:144–51.
support material for enzyme biosensors. J Nanosci Nanotechnol 2003;3:375–9. Lei C, Shin Y, Liu J, Ackerman EJ. Entrapping enzyme in a functionalized nanoporous
Ho KM, Mao X, Gu L, Li P. Facile route to enzyme immobilization: core–shell nanoen- support. J Am Chem Soc 2002;124:11242–3.
zyme particles consisting of well-defined poly (methyl methacrylate) cores and Lei CH, Shin YS, Magnuson JK, Fryxell G, Lasure LL, Elliott DC, et al. Characterization of
cellulase shells. Langmuir 2008;24:11036–42. functionalized nanoporous supports for protein confinement. Nanotechnology
Huang SH, Liao MH, Chen DH. Direct binding and characterization of lipase onto mag- 2006;17:5531–8.
netic nanoparticles. Biotechnol Prog 2003;19:1095–100. Levison PR, Dennis J, Badger SE, Hathi P, Davis MJ, Brue IJ, et al. Recent developments of
Huang J, Liu C, Xiao H, Wang J, Jiang D, Erdan GU. Zinc tetraaminophthalocyanine–Fe3O4 magnetic beads for use in nucleic acid purification. J Chromatogr A 1998;816:107–11.
nanoparticle composite for laccase immobilization. Int J Nanomed 2007;2:775–84. Li J, He X, Wu Z, Wang K, Shen G, Yu R. Piezoelectric immunosensor based on magnetic
Hung SW, Hwang JK, Tseng F, Chang JM, Chen CC, Chieng CC. Molecular dynamics sim- nanoparticles with simple immobilization procedures. Analy Chim Acta
ulation of the enhancement of cobra cardiotoxin and E6 protein binding on mixed 2003a;481:191–8.
self-assembled monolayer molecules. Nanotechnology 2006;17:S8-S13. Li J, Wang J, Gavalas VG, Atwood DA, Bachas LG. Alumina-pepsin hybrid nanoparticles
Husain Q. β Galactosidase and their potential applications. Crit Rev Biotechnol with orientation-specific enzyme coupling. Nanoletters 2003b;3:55–8.
2010;30:41–62. Li D, He Q, Cui Y, Duan L, Li J. Immobilization of glucose oxidase onto gold nanoparticles
Husain Q, Ansari SA, Fahad M, Azam A. Immobilization of Aspergillus oryzae β galacto- with enhanced thermostability. Biochem Biophys Res Commun 2007;355:488–93.
sidase on zinc oxide nanoparticles via simple adsorption mechanism. Int J Biol Lia Y, Schluesenerb HJ, Xu S. Gold nanoparticle-based biosensors. Gold Bull 2010;43:
Macromol 2011;49:37–43. 29–41.
Iyer PV, Ananthanarayan L. Enzyme stability and stabilization: Aqueous and non-aque- Lin J, Qu W, Zhang S. Disposable biosensor based on enzyme immobilized on Au-
ous environment. Process Biochem 2008;43:1019–32. chitosan-modified indium tin oxide electrode with flow injection amperometric
Jang E, Park S, Park S, Lee Y, Kim D, Kim B, et al. Fabrication of polyethylene glycol- analysis. Analyt Biochem 2007;360:288–93.
based hydrogels entrapping enzyme-immobilized silica nanoparticles. Polym Adv Ling YP, Heng LY. A potentiometric formaldehyde biosensor based on immobilization
Technol 2010;21:476–82. of alcohol oxidase on acryloxysuccinimide-modified acrylic microspheres. Sensors
Jia H, Zhu G, Wang P. Catalytic behaviors of enzymes attached to nanoparticles: the ef- 2010;10:9963–81.
fect of particle mobility. Biotechnol Bioeng 2003;84:406–14. Liu S, Ju H. Renewable reagentless hydrogen peroxide sensor based on direct electron
Jia H, Zhao X, Kim J. Wang P. Carbon nanotube composite electrodes for biofuel cells. transfer of horseradish peroxidase immobilized on colloidal gold-modified elec-
Nanoscale Science and Engineering in Biomolecular Catalysis. ACS Symposium Se- trode. Anal Biochem 2002;307:110–6.
ries 2011;986:273–88. Liu CG, Desai KSH, Chen G, Park HJ. Linolenic acid-modified chitosan for formation of
Johnson AK, Zawadzka AM, Deobald LA, Crawford RL, Paszczynski AJ. Novel method for self-assembled nanoparticles. J Agric Food Chem 2005;53:437–41.
immobilization of enzymes to magnetic nanoparticles. J Nanopart Res 2008;10: Liu W, Zhang S, Wang P. Nanoparticle-supported multi-enzyme biocatalysis with in
1009–25. situ cofactor regeneration. J Biotechnol 2009;139:102–7.
Jordan BJ, Hong R, Gider B, Hill J, Emrick T, Rotello VM. Stabilization of α-chymotrypsin Ma Z, Ding T. Bioconjugates of glucose oxidase and gold nanorods based on electrostat-
at air–water interface through surface binding to gold nanoparticle scaffolds. Soft ic interaction with enhanced thermostability. Nanosc Res Lett 2009;4:1236–40.
Matter 2006;2:558–60. Mahapatro A, Kumar A, Kalra B, Gross RA. Solvent free adipic acid/1,8 octanediol con-
Jun L, Zhou W, Kumbhar J, Wiemann J, Fang J, Carpentier EE, et al. Gold-coated iron densation polymerization characterized by Candida antarctica B lipase. Macromol-
(Fe@Au) nanoparticles: synthesis, characterization and magnetic field induced ecule 2004;37:35–40.
self-assembly. Solid State Chem 2001;159:26–31. Mahmoud KA, Male KB, Hrapovic S, Luong JHT. Cellulose nanocrystal/gold nanoparticle
Kalkan NA, Aksoy EA, Aksoy S, Hasirci N. Chitosan coated magnetic nanoparticles for composite as a matrix for enzyme immobilization. Appl Mater Inter 2009;1:
adsorption of laccase. Abstract no. 605, published in Nanotr6 Conf; 2009. 1383–6.
Khan MJ, Aazam A, Husain Q. α Amylase immobilized on Fe2O3 nanoparticle and its ap- Mak KH, Yu CY, Kuan IC, Lee SL. Immobilization of Pseudomonas cepacia lipase onto
plication in starch hydrolysis. Communicated. magnetic nanoparticles for biodiesel production. Sci Technol Vision 2009;5:19–23.
S.A. Ansari, Q. Husain / Biotechnology Advances 30 (2012) 512–523 523

Mao X, Huang J, Fai LM, Du Z, Ma L, Huang Z, et al. Novel core–shell nanoparticles and Schmid RD, Verger R. Lipases: interfacial enzymes with attractive applications. Ang
their application in high-capacity immobilization of enzymes. Appl Biochem Bio- Chem (Int Ed) 1998;37:1609–16.
technol 2006;35:229–39. Schuler C, Caruso F. Preparation of enzyme multilayers on colloids for biocatalysis.
Martin CR, Kohli P. The emerging field of nanotube biotechnology. Nat Rev Drug Discov Macromol Rapid Commun 2000;21:750–3.
2003;2:29–37. Selim KMK, Ha YS, Kim SJ, Chang Y, Kim TJ, Lee GH, et al. Surface modification of mag-
Mateo C, Grazu V, Palomo JM, Lopez-Gallego F, Fernandez-Lafuente R, Guisan JM. Im- netite nanoparticles using lactobionic acid and their interaction with hepatocytes.
mobilization of enzymes on heterofunctional epoxy supports. Nature Protocol Biomaterials 2007;28:710–6.
2007;2:1022–33. Srinivasulu B, Prakasham RS, Annapurna J, Srinivas S, Ellaiah P, Ramakrishna SV. Neo-
Miletic N, Abetz V, Ebert K, Loos K. Immobilization of Candida antarctica lipase B on mycin production with free and immobilized cells of Streptomyces marinensis in
polystyrene nanoparticles. Macromol Rapid Commun 2010;31:71–4. an airlift reactor. Process Biochem 2002;38:593–8.
Mitchell DT, Lee SB, Trofin L, Li N, Nevanen TK, Soderlund H, et al. Smart nanotubes for Tang Z, Qian J, Shi L. Preparation of chitosan nanoparticles as carrier for immobilized
bioseparations and biocatalysis. J Am Chem Soc 2002;124:11864–5. enzyme. Appl Biochem Biotechnol 2007;136:77–96.
Moghaddam AB, Tayebe Nazari T, Badraghi J, Kazemzad M. Synthesis of ZnO nanopar- Tang T, Fan H, Ai S, Han R, Qiu Y. Hemoglobin (Hb) immobilized on amino-modified
ticles and electrodeposition of polypyrrole/ZnO nanocomposite film. Int J Electro- magnetic nanoparticles for the catalytic removal of bisphenol A. Chemosphere
chem Sci 2009;4:247–57. 2011;83:255–64.
Naik RA, Tomczak MM, Luckarift HR, Spain JC, Stonea MO. Entrapment of enzymes and nano- Tischer W, Wedekind F. Immobilized enzymes: methods and applications. Top Curr
particles using biomimetically synthesized silica. Chem Commun 2004;66:1684–5. Chem 1999;200:95-126.
Namdeo M, Bajpai SK. Immobilization of α-amylase onto cellulose-coated magnetite Tong X, Trivedi A, Jia H, Zhang M, Wang P. Enzymic thin film coatings for bioactive ma-
(CCM) nanoparticles and preliminary starch degradation study. J Mol Catal B: terials. Biotechnol Prog 2008;24:714–9.
Enzym 2009;59:134–9. Troitsky VI, Berzina TS, Pastorino L, Bernasconi E, Nicolini C. A new approach to the de-
Neri DFM, Balcao VM, Carneiro-da-Cunha MG, Carvalino jr LB, Teixeira JA. Immobiliza- position of nanostructured biocatalytic films. Nanotechnology 2003;14:597–602.
tion of β galactosidase from Kluyveromyces lactis onto a polysiloxane-polyvinyl al- Van Dyk JS, Pletschke B. Review on the use of enzymes for the detection of organochlo-
cohol magnetic (mPOS-PVA) composite for lactose hydrolysis. Catal Commun rine, organophosphate and carbamate pesticides in the environment. Chemo-
2008;9:2334–9. sphere 2011;82:291–307.
Neri DFM, Balcao VM, Costa RS, Rocha ICAP, Ferreira EMFC, Torres DPM, et al. Galacto- Vertegel AA, Siegel RW, Dordick JS. Silica nanoparticle size influences the structure and
oligosaccharides production during lactose hydrolysis by free Aspergillus oryzae β enzymatic activity of adsorded lysozyme. Langmuir 2004;20:6800–7.
galactosidase and immobilized on magnetic polysiloxane-polyvinyl alcohol. Food Wan LS, Ke BB, Wu J, Xu ZK. Catalase immobilization on electrospun nanofibers: effects
Chem 2009;115:92–9. of porphyrin pendants and carbon nanotubes. J Phys Chem C 2007;111:14091–7.
Ni Y, Cao X, Wu G, Hu G, Yang Z, Wei X. Preparation, characterization and property Wang P. Nanoscale biocatalyst systems. Curr Opin Biotechnol 2006;17:574–9.
study of zinc oxide nanoparticles via a simple solution-combusting method. Nano- Wang P. Multi-scale features in recent development of enzymic biocatalyst systems.
technology 2007;18:155–61. Appl Biochem Biotechnol 2009;152:343–52.
Pan C, Hu B, Li W, Sun Y, Ye H, Zeng X. Novel and efficient method for immobilization Wang J, Lin Y. Functionalized carbon nanotubes and nanofibers for biosensing applica-
and stabilization of β galactosidase by covalent attachment onto magnetic Fe3O4- tions. Trends Anal Chem 2008;27:619–26.
chitosan nanoparticles. J Mol Catal B: Enzym 2009;61:208–15. Wang W, Xu Y, Wang DIC, Li Z. Recyclable nanobiocatalyst for enantioselective sulfox-
Pandey P, Singh SP, Arya SK, Gupta V, Datta M, Singh S, et al. Application of thiolated idation: facile fabrication and high performance of chloroperoxidase-coated mag-
gold nanoparticles for enhancement of glucose oxidase activity. Langmuir netic nanoparticles with iron oxide core and polymer shell. J Am Chem Soc
2007;23:3333–7. 2009;131:12892–3.
Park JI, Cheon J. Synthesis of “solid solution” and “core–shell” type cobalt–platinum magnetic Willner I, Baron R, Willer B. Growing metal nanoparticles by enzymes. Adv Mater
nanoparticles via trans-metalation reactions. J Am Chem Soc 2001;123:5743–6. 2006;18:1109–20.
Park JY, Kim YH, Seong A, Yoo YJ. Amperometric determination of glucose based on the Wu H, Wang J, Kang X, Wang C, Wang D, Liu J, et al. Glucose biosensor based on immo-
direct electron transfer between glucose oxidase and tin oxide. Biotechnol Bioproc bilization of glucose oxidase in platinum nanoparticles/graphene/chitosan nano-
Eng 2008;13:431–5. composite film. Talanta 2009a;80:403–6.
Penn S, He L, Natan M. Nanoparticles for bioanalysis. Curr Opin Chem Biol 2003;7:609–15. Wu Z, Zhang B, Yan B. Regulation of enzyme activity through interactions with nano-
Petros RA, DeSimone JM. Strategies in the design of nanoparticles for therapeutic appli- particles. Int J Mol Sci 2009b;10:4198–209.
cations. Nat Rev Drug Discov 2010;9:615–27. Xiao Y, Patolsky F, Katz JF, Hainfeld E, Willner I. Plugging into enzymes: nanowiring of
Phadtare S, Kumar A, Vinod VP, Dash C, Palaskar DV, Rao M, et al. Direct assembly of redox-enzymes by a gold nanoparticle. Science 2003;299:1877–81.
gold nanoparticle “shells” on polyurethane microsphere “cores” and their applica- Xie T, Wang A, Huang L, Li H, Chen Z, Wang Q, et al. Recent advances in the support and
tion as enzyme immobilization templates. Chem Mater 2003;15:1944–9. technology used in enzyme immobilization. Afr J Biotechnol 2009;8:4724–33.
Phadtare S, Vinod VP, Mukhopadhyay K, Kumar A, Rao M, Chaudhari RV, et al. Immobi- Xiong L, Regneir FE. Channel specific coatings on microfabricated chips. J Chromat A
lization and biocatalytic activity of fungal protease on gold nanoparticle-loaded ze- 2001;924:165–76.
olite microspheres. Biotechnol Bioeng 2004;85:629–37. Xu J, Zeng F, Wu S, Liu X, Hou C, Tong Z. Gold nanoparticles bound on microgel particles
Pimentel MCB, Leao ABF, Melo EHM, Ledingham WM, Lima-Filho JL, Sivewright M. Im- and their application as an enzyme support. Nanotechnology 2007;18:265–73.
mobilization of Candida rugosa Iipase on magnetized dacron: kinetic study. Art. Yamashita I. Fabrication of a two-dimensional array of nanoparticles using ferritin mol-
Cell. Blood Substit. Biotechnol 2007;35:221–35. ecule. Thin Solid Films 2001;393:12–8.
Prakasham RS, Sarala DG, Rajya LK, Subba R. Novel synthesis of ferric impregnated sil- Yang M, Yang Y, Liu Y, Shen G, Yu R. Platinum nanoparticles-doped sol–gel/carbon
ica nanoparticles and their evaluation as a matrix for enzyme immobilization. J nanotubes composite electrochemical sensors and biosensors. Biosens Bioelectron
Phys Chem C 2007;111:3842–7. 2006;21:1125–31.
Prakasham RS, Devi GS, Rao CS, Sivakumar VSS, Satish T, Sharma PN. Nickel-impregnat- Yim TJ, Kim DY, Karajanagi SS, Lu TM, Kane R, Dordick JS. Silicon nanocolumns as novel
ed silica nanoparticle synthesis and their evaluation for biocatalyst immobilization. nanostructured supports for enzyme immobilization. J Nanosci Nanotechnol
Appl Biochem Biotechnol 2010;160:1888–95. 2003;3:479–82.
Qhobosheane M, Santra S, Zhang P, Tan W. Biochemically functionalized silica nano- You T, Niwa O, Tomita M, Hirono S. Characterization of platinum nanoparticle-
particles. Analyst 2001;126:1274–8. embedded carbon film electrode and its detection of hydrogen peroxide. Anal
Radhakumary C, Sreenivasan K. Naked eye detection of glucose in urine using glucose Chem 2003;75:2080–5.
oxidase immobilized gold nanoparticles. Anal Chem 2011;83:2829–33. Zhang F, Wang X, Ai S, Suna Z, Wan Q, Zhub Z, et al. Immobilization of uricase on ZnO
Ramanavicius A, Kausaite A, Ramanaviciene A. Polypyrrole-coated glucose oxidase nanorods for a reagentless uric acid biosensor. Anal Chim Acta 2004a;519:155–60.
nanoparticles for biosensor design. Sens Actuat B 2005;111:532–9. Zhang J, Zhang SY, Xu JJ, Chen HY. A new method for the synthesis of selenium nano-
Ramkrishna SV, Prakasham RS. Microbial fermentation with immobilized cells. Curr Sci particles and the application to construction of H2O2 biosensor. Chin Chem Lett
1999;77:87-100. 2004b;15:1345–8.
Rossi LM, Quach AD, Rosenzweig Z. Glucose oxidase magnetite nanoparticle bioconju- Zhang F, Cho SS, Yang SH, Seo SS, Cha GS, Nam H. Gold nanoparticle-based mediatorless
gate for glucose sensing. Anal Bioanal Chem 2004;380:606–13. biosensor prepared on microporous electrode. Electroanalysis 2006;18:217–22.
Sahoo B, Sahu SK, Pramanik P. A novel method for the immobilization of urease on Zhang Y, Zeng GM, Tang L, Yu HY, Li JB. Catechol biosensor based on immobilizing lac-
phosphonate grafted iron oxide nanoparticle. J Mol Catal B: Enzym 2011;69: case to modified core–shell magnetic nanoparticles supported on carbon paste
95-102. electrode. Huan Jing Ke Xue 2007;28:2320–5.
Saiyed ZM, Telang SD, Ramchand CN. Application of magnetic techniques in the field of Zhao J, O'Daly JP, Henkens RW, Stonehuerner J, Crumbliss AL. A xanthine oxidase/col-
drug discovery and biomedicine. Biomag Res Technol 2003;1:2–8. loidal gold enzyme electrode for amperometric biosensor applications. Biosens
Salimi A, Sharifi E, Noorbakhsh A, Soltanian S. Immobilization of glucose oxidase on Bioelectron 1996;11:493–502.
electrodeposited nickel oxide nanoparticles: direct electron transfer and electroca- Zhao K, Zhuang S, Chang Z, Songm H, Dai L, He P, et al. Amperometric glucose biosensor
talytic activity. Biosens Bioelect 2007;22:3146–53. based on platinum nanoparticles combined aligned carbon nanotubes electrode.
Sastry M, Rao M, Ganesh KN. Electrostatic assembly of nanoparticles and biomacromo- Electroanalysis 2007;19:1069–74.
lecules. Acc Chem Res 2002;35:847–55.

You might also like