Download as pdf or txt
Download as pdf or txt
You are on page 1of 10

Bioresource Technology 337 (2021) 125399

Contents lists available at ScienceDirect

Bioresource Technology
journal homepage: www.elsevier.com/locate/biortech

Chemoenzymatic production of chitooligosaccharides employing ionic


liquids and Thermomyces lanuginosus chitinase
Manish Kumar a, Jogi Madhuprakash b, Venkatesh Balan c, Amit Kumar Singh d, V. Vivekanand e,
Nidhi Pareek a, *
a
Department of Microbiology, School of Life Sciences, Central University of Rajasthan, Ajmer 305817, Rajasthan, India
b
Department of Plant Sciences, School of Life Sciences, University of Hyderabad, Prof. CR Rao Road, Gachibowli, Hyderabad 500046, India
c
Department of Engineering Technology, College of Technology, University of Houston, Sugar Land, TX 77479, USA
d
Department of Mechanical Engineering, Malaviya National Institute of Technology, Jaipur 302017, Rajasthan, India
e
Centre for Energy and Environment, Malaviya National Institute of Technology, Jaipur 302017, Rajasthan, India

H I G H L I G H T S G R A P H I C A L A B S T R A C T

• Chemoenzymatic COS generation


employing ionic liquids and
T. lanuginosus chitinase.
• IL pretreated chitin appeared amor­
phous and could facilitate enzyme
accessibility.
• Optimization led to 1.5 and 1.3 fold
enhanced (GlcNAc)2 and (GlcNAc)3
production.
• 0.1 g of both (GlcNAc)2 and (GlcNAc)3
was purified from 1 g TBP pretreated
chitin.
• Proficient bio-catalytic valorization of
coastal residuals to chito-bioactives.

A R T I C L E I N F O A B S T R A C T

Keywords: The aim of this work was to study a two-step chemoenzymatic method for production of short chain chitooli­
Chitin gosaccharides. Chitin was chemically pretreated using sulphuric acid, sodium hydroxide and two different ionic
Chitooligosaccharides liquids, 1-Ethyl-3-methylimidazolium bromide and Trihexyltetradecylphosphonium bis(2,4,4-trimethylpentyl)
Chitinase
phosphinate under mild processing conditions. Pretreated chitin was further hydrolyzed employing purified
Pretreatment
Ionic liquid
chitinase from Thermomyces lanuginosus ITCC 8895. Trihexyltetradecylphosphonium bis(2,4,4-trimethylpentyl)
phosphinate treated chitin appeared amorphous and resulted in generation of 1.10 ± 0.89 mg ml− 1 of
(GlcNAc)2 and 1.07 ± 0.92 mg ml− 1 of (GlcNAc)3. Further derivation of optimum conditions through two-factor-
9 run experiments resulted in to 1.5 and 1.3 fold increments in (GlcNAc)2 and (GlcNAc)3 production, respec­
tively. 0.1 g of both (GlcNAc)2 and (GlcNAc)3 has been purified from the Trihexyltetradecylphosphonium bis
(2,4,4-trimethylpentyl)phosphinate pretreated chitin (1 g) employing cation exchange chromatography. The
present study will lay the foundation for development of a green sustainable solution for cost effective upcycling
of coastal residual resources to chito-bioactives.

* Corresponding author.
E-mail address: nidhipareek@curaj.ac.in (N. Pareek).

https://doi.org/10.1016/j.biortech.2021.125399
Received 22 March 2021; Received in revised form 5 June 2021; Accepted 8 June 2021
Available online 11 June 2021
0960-8524/© 2021 Elsevier Ltd. All rights reserved.
M. Kumar et al. Bioresource Technology 337 (2021) 125399

1. Introduction acetate pretreated chitin hydrolysed using Streptomyces albolongus chi­


tinase. Notable COS production has been reported using chemical pre­
Chitin-derived products are of immense significance in the current treatment of chitin followed by enzymatic hydrolysis (Husson et al.,
scenario due to their vast biological applications to produce nano­ 2017; Xu et al., 2020). However, limited literature is available on the
particles and nanofibers, and other applications in drug and gene de­ employment of ionic liquids for pretreatment of chitin for further
livery, nutrition fortification and wound dressing (Yang et al., 2017; Das enzymatic hydrolysis. Moreover, scarce reports are available on the
et al., 2015; Lodhi et al., 2014). The chitooligosaccharides (COS) are the utilization of thermostable chitinase for COS production. Although
homo- or hetro-oligomers of N-acetyl-D-glucosamine (GlcNAc) and D- conventional chemicals viz. acids, alkali have considered as effective
glucosamine (GlcN) which are linked by β-1,4-glycosidic bonds. The pretreatment agents for chitin disintegration; but low product quality
COS are derived from chitin or chitosan (CHS) with the degree of and environmental concerns boost the search for contemporary agents
polymerization (DP) ranging from 2 to 20 and can be prepared by viz. ILs. The reusablility of ILs could be crutial in economic prospective
chemical or enzyme processing methods (Kaczmarek et al., 2019). The for the development of an efficient process for COS production.Thus, the
value for COS is defined by its length and the degree of acetylation (DA) present study explores the impact of traditional harsh chemicals (H2SO4
(Singh et al., 2020; Liu et al., 2019; Zhao et al., 2019; Şenel, 2019). Amid and NaOH) and ILs pretreatment of chitin followed by hydrolysis using
chitin and its derivatives, COS have attracted a wide range of application purified chitinase from thermophilic fungus, T. lanuginosus (Kumar
including antimicrobial, antioxidant, gene therapy, immune cell prolif­ et al., 2018) for COS production. The two ILs 1-Ethyl-3-methylimidazo­
eration, and tumor growth inhibition due to its superior solubility, low lium bromide (EMB) and Trihexyltetradecylphosphonium bis(2,4,4-
viscosity, and increased absorption properties in the intestine for anti- trimethylpentyl)phosphinate (TBP) used in this study were selected on
obesity applications (Phil et al., 2018; Liaqat and Eltem, 2018). the basis of their low cost and to the best of our knowledge this is the first
The chemical production approaches are rapid and produce high report of employing these ILs for chitin pretreatment. IL pretreated
yield; however the hydrolysis reactions are difficult to control, resulting chitin is found to be highly porous and amorphous when compared to
in GlcNAc as a major product (Hamed et al., 2016). The enzymatic untreated chitin, thereby facilitating chitinase accessibility during hy­
methods of producing COS appeared favorable as they do not produce drolysis producing high COS yield when compared to untreated and
any toxic byproducts benefiting the environment, high specificity so that acid/based pretreated chitin. COS purification protocol has also been
reaction can be controlled and hydrolysis reaction are carried at low developed using preparative cation exchange chromatography, and
temperatures (Pan et al., 2019; Hamer et al., 2015). However, the quantified them using high performance liquid chromatography (HPLC).
enzymatic approach still face some limitations such as low COS yield
and high cost of production (Kaczmarek et al., 2019; Jung and Park, 2. Materials and methods
2014). The recent knowledge in the field of chitinolytic enzymes espe­
cially chitinase from different mesophilic and thermophilic bacteria and Two ILs EMB and TBP, N-Acetylglucosamine (GlcNAc), and dimeric
fungi have given several options that will help to overcome some of the N-Acetylglucosamine (GlcNAc)2, and trimeric N-Acetylglucosamine
limitations of enzymatic approaches and expected to play a major role in (GlcNAc)3 were acquired from Sigma-Aldrich, Germany. The α-chitin
the development of robust bioprocess (Kidibule et al., 2020; Hamid processed from shrimp shells was obtained from HiMedia, India.
et al., 2013). The production of COS is dependent on both the type of Colloidal chitin was prepared from the shrimp shell chitin flakes using
chitinolytic enzymes used and recalcitrant nature of chitin (Kidibule the reported protocol (Kumar et al., 2017). Briefly, 5 g of shrimp’s shells
et al., 2020; 2018). Several biopolymers including proteins, cellulose, chitin was mildly agitated with 100 ml of ice-cold concentrated hydro­
hemicellulose and chitin have been successfully pretreated using chloric acid in a glass bottle and maintained at 4℃ for 24 h. Then the
different chemical, physical and biological methods (Mukherjee et al., reaction mixture added to ice-cold ethanol (96%, 500 ml) and kept
2020; Villa-Lerma et al., 2013; Guoxiang et al., 2010). Among these under rapid stirring and maintained at 4℃ for 24 h. The precipitate was
methods, chemical pretreatment has been found to be cost effective and separated by centrifugation (12,000g, 25 min, and 4℃) and washed
are reported to increase the porosity and increase enzyme accessibility with distilled water until the pH reached neutral. The colloidal chitin
so that chitinolytic enzymes could act more efficiently (Suryawanshi was further dried overnight, ground to fine powder using mortar and
et al., 2019; Li et al., 2019). Since harsh chemicals results in the for­ pestle and stored at room temperature before further use. Thin layer
mation of undesirable toxic side products, several researchers are chromatography (TLC) silica gel sheets, chromatography cation ex­
exploring mild pretreatment chemical such as ionic liquids (ILs) change media such as SP-Sepharose were purchased from Sigma-
(Sánchez et al., 2018; Li et al., 2016). Recently, a combination of both Aldrich, Germany. All other chemicals used in the study were analyt­
chemical and enzymatic processing called chemoenzymatic approaches ical grade.
are getting popular for the development of cost effect method of pro­
ducing COS (Ogata, 2020; Harmsen et al., 2020; Queda et al., 2019). 2.1. Fungal strain and chitinase production
ILs are organic salts that are liquid at a temperature below 100 ◦ C
(Lei et al., 2017). ILs contains cations such as imidazolium, pyridinium, The chitinase producing thermophilic (45 ◦ C) fungal strain used in
pyrrolidinium, or ammonium; organic anions like CH3COO‾ or inor­ the present study, T. lanuginous ITCC 8895 (earlier known as Humicola
ganic anions like Cl‾, Br‾, and BF4‾ (Chiappe and Pieraccini, 2005). ILs grisea) was isolated from the sandy loam soil of Jaisalmer, Rajasthan,
have shown superior dissolution and processability behavior with bio­ India. Chitinase was produced and purified at Microbial Catalysis and
macromolecules because of their high chemical and thermal stability, Process Engineering Laboratory, Department of Microbiology, Central
high ionic conductivity, non-inflammability, and trivial vapor pressure University of Rajasthan, India using reported procedures (Kumar et al.,
(Chiappe and Pieraccini, 2005). ILs can be recycled due to their low 2018; 2017). The chitinase production was carried out under submerged
volatility, which will reduce chemical pollution to the environment. fermentation employing the optimum levels of nutritional variables
Though they do not contribute to air pollution due to negligible vapor (chitin, 7.49; colloidal chitin, 4.91; KH2PO4, 0.87; K2HPO4, 0.68; KCl,
pressure ILs, their impact on aquatic and terrestrial eco-system is yet to 0.19; NH4Cl, 1.0; MgSO4⋅7H2O, 0.20; yeast extract, 5.50 gl− 1) (Kumar
be assessed (Steudte, 2014). ILs have been well extensively explored for et al., 2017) and process conditions (pH, 6.50; Temperature, 47 ◦ C;
the dissolution of lignocellulose to produce biofuels and biochemicals Inoculum size, 5 discs) (Kumar et al., 2018). Chitinase purification was
(Usmani et al., 2020; Brandt et al., 2013). Same knowledge can be carried out by using cation and anion exchange chromatography (Kumar
applied to pretreat chitin (Shamshina and Berton, 2020; Husson et al., et al., 2018).
2017). Li et al. (2019) reported the generation of GlcNAc (175.62 mg)
and (GlcNAc)2 (341.70 mg) from 1 g of 1-ethyl-3lmethylimidazolium

2
M. Kumar et al. Bioresource Technology 337 (2021) 125399

2.2. Chitin pretreatment Chitinase assay was performed by incubating enzyme and 1% colloidal
chitin with citrate buffer (100 mM, pH 3.0) in 1:1 ratio (v/v) 70℃ for 60
The COS production was carried out by the employing a two-step min. The reaction mixture was centrifuged (10,000g, 5 min.) and the
chemoenzymatic process. First, chitin was subjected to different pre­ supernatant was analysed for reducing sugar through 3,5-Dinitrosalicy­
treatment using chemicals such as H2SO4, NaOH and ILs (EMB and TBP) clic acid method. Here, chitinase was used in the term of specific activity
followed by enzymatic hydrolysis. Chitin (10% w/v) was first combined and one unit (U) of chitinase activity was calculated as the amount of
with different pretreatment chemicals at 10:1 ratio, followed by incu­ enzyme required to release 1 µmol of GlcNAc per min under the standard
bation in a water bath (Tempo, India) at 100 ◦ C for 90 min using pre­ assay conditions (Kumar et al., 2018). Small aliquot of samples were
viously reported protocol (Usmani et al., 2020; Hou et al., 2017). withdrawn using pipette at various time intervals (0, 0.25, 0.5, 1, 2, 4, 6,
Further, the pretreated chitin was filtered, washed until pH reached 6.0, 8, 10, 12, 24 and 48 h) and the enzymatic reaction was stopped by
and dried in an oven (50 ◦ C, overnight). The obtained pretreated dry boiling (100 ◦ C, 10 min) using a heat block followed by cooling down on
chitin was stored at room temperature for further use. ice and centrifugation (10,000g, 4 ◦ C, 5 min).

2.3. Physico-chemical analysis of chitin 2.5. Thin-layer chromatography

The untreated as well as the pretreated chitin were subjected to The hydrolyzed products were analysed using TLC Silica gel 60
scanning electron microscopic (SEM) analysis to study the effect of (Merck, Germany). Samples (1 µl) from untreated and pretreated enzy­
pretreatment on the structure and morphological properties. Briefly, the matic hydrolysate were spotted on the TLC. The spots were developed in
dried chitin samples were taken in small amounts on double-sided car­ the solvent system containing n-butanol: methanol: ammonia solution
bon tape adhered to aluminum stubs. The stubs were then sputter coated (25%): water (5:4:2:1, v/v/v/v) in closed jar. The developed spots were
with gold and examined using FEI Nova NanoSEM 450 electron micro­ analysed by spraying with aniline-diphenylamine reagent followed by
scope (Nova, Netherlands). The sample micrographs at different mag­ heating in a hot air oven set at 150 ◦ C (Tempo, India) for 10 min
nifications were captured using a secondary electron detector at an (Purushotham et al., 2012). The quantitative analysis of spots present in
accelerating voltage of 15 kV. TLC plates were analysed using JustTLC software (Sweday, Sweden).
Both untreated and pretreated chitin samples were subjected to
Fourier Transform Infrared (FTIR) spectroscopy (PerkinElmer, USA). 2.6. Design of experiments
The spectra were generated by employing Attenuated Total Reflectance
(ATR) mode. All the chitin samples were scanned from 4000 to 400 cm− 1 The derivation of the optimum levels of T. lanuginous chitinase and
to study the alternation in the functional groups, spectra were compared chitin (untreated and pretreated) could be achieved through statistical
and analysed through the stack graph prepared using OriginPro (Ori­ optimization by two-factor design matrix. The range of TBP pretreated
ginLab, USA). The DA (%) was determined by using the following chitin was kept between 1 and 10% (w/v), while purified T. lanuginosus
equation (Gbenebor et al., 2017): chitinase was taken in a range of 1–5 U mg− 1 (Table 1). A two-level 9 run
experiment was conducted and the response in terms of (GlcNAc)2 and
DA(%) = A1655 / A3450 × 100 / 1.33 (1) (GlcNAc)3 production was taken from the mean value of hydrolysis
-1
Where, A1655 is the absorbance of amide at 1655 cm and A3450 is experiment run in triplicates.
the absorbance of the hydroxyl group at 3450 cm− 1.
The X-ray powder diffraction of untreated and pretreated chitin was 2.7. COS purification
recorded using X-Ray Diffractometer, PANalytical X’Pert Pro (Malvern
Panalytical, Netherlands). The diffraction pattern was recorded at a scan The hydrolysis product obtained under optimized conditions con­
rate of 0.2◦ /s between 2θ angles of 5◦ and 30◦ under Cu radiation (40 tains different DP’s. The hydrolysate was subjected to the extraction step
mA, 40 kV). The X-ray diffractograms were analysed and plotted as a to remove GlcNAc and other impurities using cation exchange chro­
stacked graph through OriginPro (OriginLab, USA). The crystallinity matography using SP-Sepharose media (Sigma-Aldrich, USA). The col­
index (CI %) was calculated using the following equation (Focher et al., umn was equilibrated with citrate buffer (100 mM, pH 3.0) and the
1990): hydrolysis product was then loaded on the top of the column. Elution
buffer contains a varying concentration of sodium chloride (50–500
CI (%) = [(I110 − − Iam )/I110 ] × 100 (2) mM) and the collected fractions were subjected to TLC to determine the
relative concentration of GlcNAc and COS. The fraction containing
Where, I110 (arbitrary units) is the maximum intensity of the (1 1 0) peak (GlcNAc)2 and (GlcNAc)3 were pooled and desalted using Sephadex G-
at 2θ = 19◦ , and Iam (arbitrary units) is the amorphous diffraction at 2θ 10 (Sigma-Aldrich, USA). Further, the active fractions were concen­
= 12◦ . trated, forzen using deep freezer at − 80 ◦ C and lyophilized using a
The thermogravimetric analysis (TGA) and differential thermal FreeZone freeze dryer (Labconco, USA). The COS fractions were ana­
analysis (DTA) of untreated and pretreated chitin were analysed with lysed using HPLC (Waters 1500 series, Ireland) using Asahipak NH2P-
the help of EXSTAR TG/DTA SII 6300 (Hitachi, Japan). The thermo­ 50E column (4.6 × 250 mm) (Shodex, China) connected to a RI detector.
grams were obtained by using chitin samples (5 mg) in an alumina pan The HPLC separation was carried out at room temperature (25 ◦ C) using
by purging nitrogen at 200 ml/min. Alumina powder was used as the acetonitrile water mixture (70:30, v/v) as the mobile phase at a flow rate
reference for TGA analysis. The DTA measurement was obtained at of 1.0 ml min− 1. The concentration of (GlcNAc)2 and (GlcNAc)3 was
35–700 ◦ C at a heating rate of 10 ◦ C/min. The graphical representation quantified using a standard curve generated using commercial COS.
of data was done employing OriginPro (OriginLab, USA).
Table 1
2.4. Enzymatic hydrolysis of untreated and pretreated chitin Experimental variables at different levels in the two-factor design for COS
production.
Chitin to COS conversion efficiency of untreated and pretreated Variable Component Levels
chitin samples was evaluated by using purified T. lanuginous chitinase.
− 1 0 +1
The untreated and pretreated chitin samples (1%, w/v) were mixed with
citrate buffer (100 mM, pH 3.0). Enzyme hydrolysis was carried out for X1 TBP pretreated chitin (%, w/v) 1.0 5.5 10.0
X2 T. lanuginosus chitinase (U mg− 1) 1.0 3.0 5.0
48 h using chitinase at an enzyme loading of 2.5 U mg− 1 at 70 ◦ C.

3
M. Kumar et al. Bioresource Technology 337 (2021) 125399

3. Results and discussion chitin. The characteristic peaks of stretching vibrations of amide I, II,
and III were detected at 1626, 1552, and 1310 cm− 1. The bands at 3258
3.1. Chitinase production, purification and characterization cm− 1 and 3100 cm− 1 in untreated and pretreated chitin indicated the
stretching and bending vibration of aliphatic –OH. The absorption peak
Purified chitinase from T. lanuginosus ITCC 8895 was employed for at 2878 cm− 1 represented the C–H stretching of the –CH3 group. The
enzymatic hydrolysis of chitin following pretreatment. The chitinase absorption band at 1626 cm− 1 demonstrates the stretching vibration of
production, derivation of optimum nutritional and process conditions, the carbonyl group. The bending vibration of amide II (N–H bending)
purification and characterization were performed in the earlier studies from acetamide groups was observed at 1552 cm− 1 and the stretching
(Kumar et al., 2017; 2018). Briefly, T. lanuginosus was isolated and pu­ vibration of amide III (C–N stretching) from acetamide group appeared
rified from the soil sample of Jaisalmer, Rajasthan, India. The strain at 1310 cm− 1. Moreover, the peak at 1014 cm− 1 depicted the stretching
showed the maximum level of chitinase activity (120 ± 0.90 U l− 1) vibration for C–O–C of the glucosamine ring in the untreated as well as
following 72 hr of submerged fermentation. Under derived nutritional pretreated chitin. The characteristic ring stretching band for β-1,4
and process conditions the chitinase production was enhanced by 1.8 glycosidic bonds was noticed in the range of 886 cm− 1 and 896 cm− 1
folds (213.57 ± 6.4 U l− 1). and in the case of pretreated chitin, a reduction in the peak intensity
Further, chitinase (50 kDa) was purified from the culture superna­ could be observed. The decreased intensity of peak indicated the
tants of T. lanuginosus to homogeneity using ion exchange chromatog­ disruption of chitin during pretreatment. (Sivaramakrishna et al., 2020;
raphy. The enzyme was purified to 3.8 fold with 17.1% recovery. The Kumari et al., 2015; Dahmane et al., 2014). The DA of untreated and
optimal temperature and pH of the purified chitinase were 70℃ and 3.0 pretreated chitin are presented in supplementary materials.
respectively. Chitinase showed stability over a wide range of pH (2.0 –
11.0) and temperature (30-90℃). Half life of the purified chitinase was 3.2.3. XRD
169.06 min at its optimal temperature. Moreover, detremination of ki­ X-ray diffraction (XRD) analysis of untreated and pretreated chitin
netic parameters viz. Km, Vmax, kcat/Km and substrate specificity illus­ was carried out to study the crystallinity and recalcitrance properties.
trated that the purified chitinase exhibited highest activity towards The crystallinity of chitin is a key attribute for their efficient enzymatic
colloidal chitin as depicted from its lower Km value (0.11 mg ml− 1). The hydrolysis (Mohan et al., 2018). A comparative x-ray diffractogram of
enzyme activity was notably enhanced by Mn2+ and Co2+ (173 and untreated and pretreated chitin samples are shown in supplementary
144%). materials. The characteristic strong peaks at 2θ values of 9.5◦ , 19.2◦ ,
23.4◦ , and 26.4◦ can be observed in the untreated and the pretreated
3.2. Impact of pretreatment on the physicochemical properties of chitin chitin samples. The peak at 2θ value = 19.5◦ corresponds to the char­
acteristic plane (1 1 0) of crystalline chitin and a decrease in the in­
The pretreatment of chitin was performed by employing various tensities of the peak was observed for pretreated chitin, indicating that
chemicals viz. H2SO4, NaOH and ILs (EMB and TBP) to reduce the the crystallinity of the chitin was decreased following pretreatment,
recalcitrance towards enzymatic action. Structural and physico- which is further supported by the CI data obtained by using the equation
chemical characterization (SEM, FTIR, XRD, TGA and DTA) was per­ (1) (Ibitoye et al., 2018). The CI of untreated chitin was highest (81.0%),
formed following pretreatment to study the alterations occurred due to followed by H2SO4, NaOH, TBP, and TBP pretreated chitin, i.e., 80.5%,
the pretreatment to enhnace the substrate accessibility. The pretreated 79.2%, 78.1%, and 76.6%, respectively (see supplementary materials).
chitin appeared more clear and amorphous as compared to the untreated The notable decrease in CI of EMB and TBP pretreated chitin suggested
chitin. The effectiveness of the pretreatment process was illustrated by that the IL pretreatment has made chitin amorphous and facilitate chi­
decreased crystallinity, an opened structure and functional group tinase accessibility and increase the production of COS.
analysis.
3.2.4. TGA & DTA
3.2.1. SEM measurements The TGA and DTA analysis of untreated and pretreated chitin sam­
The alterations in the surface morphology of chitin following ples was performed to study thermal stability following pretreatment.
chemical pretreatment could be observed by comparing the scanning The degradation of untreated and pretreated chitin started in the range
electron micrographs of untreated and pretreated chitin samples. The of 50–100 ◦ C and is recognized by the evaporation of water molecules.
micrographs were taken at 20,000x magnification and 5 µm resolution After the removal of water molecules, the decomposition occurred be­
illustrated significant variance in the morphology. The micrograph of tween 250 and 360 ◦ C that led to the degradation of chitin (see sup­
untreated chitin (see supplementary materials) showed an intact and plementary materials). The temperature at which the sample began to
uniformly pitted crystalline structure with very random pores. The tight lose weight (Tonset) for untreated chitin was 285 ◦ C and the completion
and orderly fashion of the microfibril stacks could be seen on the surface of the degradation process occurred at 360 ◦ C. For H2SO4 and NaOH
of the untreated chitin. However, in the case of H2SO4 and NaOH pre­ pretreated chitin, the Tonset and the completion temperature were found
treated chitin (see supplementary materials), an improved surface to be 275 and 355 ◦ C, respectively. While, the Tonset and the completion
disintegration of the crystalline structure could be observed as compared temperature of EMB and TBP pretreated chitin was observed to be 265
to the untreated chitin. Furthermore, the micrographs of EMB and TBP and 350 ◦ C, respectively. The pretreated chitin experienced a slightly
pretreated chitin (see supplementary materials) revealed significant earlier onset temperature (Tonset) when compared to the untreated chitin
disruption of the structure when compared to untreated chitin. The due to the decrease in crystallinity after chemical pretreatment. Most of
microfibrils appeared to be arranged in bundles even after pretreatment. the weight loss of chitin samples occurred before 360 ◦ C due to pyrolysis
Moreover, enhanced porosity of EMB and TBP pretreated chitin has and resulted in char residue of carbonaceous material. Similar pattern of
resulted into increased surface area to facilitate enzyme accessibility thermogram for chitin could be observed in the previous studies (Feás
during hydrolysis. et al., 2020).
Moreover, the DTA thermogram of untreated and pretreated chitin
3.2.2. FTIR spectroscopy show less difference (see supplementary materials) with one major sharp
The FTIR spectroscopy of untreated and pretreated chitin are shown peak for the core thermal decomposition at 360 ◦ C. Comparatively less
in supplementary materials with characteristic peaks for chitin as re­ intense peaks were observed ~ 550 ◦ C due to the presence of recalcitrant
ported before (Sivaramakrishna et al., 2020). A similar pattern of ab­ materials and traces amount of chemicals. A slight shift in the peak of
sorption bands was exhibited in both untreated and pretreated chitin untreated chitin ~ 360 ◦ C indicated the requirement of high tempera­
which confirm that chemical pretreatment process did not decompose ture for the decomposition of untreated chitin as compared to the

4
M. Kumar et al. Bioresource Technology 337 (2021) 125399

pretreated chitin. Hence, the pretreated chitin appeared to be more (GlcNAc)2 and (GlcNAc)3 were achieved after 2 h of the enzymatic hy­
amorphous, less thermostable and amenable for enzymatic hydrolysis. drolysis, i.e., 0.88 and 0.78 mg ml− 1, respectively which is noticeably
The appearance of positive peaks in the TGA thermogram depicted the higher when compared to the H2SO4 and NaOH pretreated chitin
evolution of heat, i.e., the reaction was exothermic suggesting the (Fig. 2d). Similarly, TBP pretreated chitin resulted in 1.10 and 1.07 mg
change in the crystalline phase. ml− 1 of (GlcNAc)2 and (GlcNAc)3 (Fig. 2e). As the hydrolysis time pro­
gressed, prolonged COS hydrolysis has been resulted into accumulation
of GlcNAc. The GlcNAc concentration after 48 h of hydrolysis for un­
3.3. Enzymatic hydrolysis
treated chitin, H2SO4, NaOH, EMB, and TBP pretreated chitin were 0.84,
0.57, 0.90, 0.51, and 1.65 mg ml− 1, respectively.
Different pretreated chitin was subjected to purified T. lanuginosus
chitinase followed by TLC analysis to evaluate the COS conversion ef­
ficiency. TLC analysis revealed different hydrolyzed products GlcNAc, 3.4. Derivation of optimum reaction conditions for enhanced COS
chitobiose (GlcNAc)2, and chitotriose (GlcNAc)3 in the hydrolysate production
(Fig. 1). Relatively high titers of COS (GlcNAc)2 and (GlcNAc)3 were
detected during the initial period of hydrolysis. However, a further in­ COS production could be augmented by deriving the optimum
crease in hydrolysis time resulted in decrease of COS and increase in enzyme and substrate concentration during hydrolysis employing two-
GlcNAc. The production level of GlcNAc, (GlcNAc)2, and (GlcNAc)3 are factor statistical optimization. The two-factor design and the corre­
presented in Fig. 2. sponding (GlcNAc)2 and (GlcNAc)3 yield are shown in Table 2. The two-
The highest level of (GlcNAc)2 and (GlcNAc)3 produced after 1 hr of factor-9 run experiment showed significant variation in (GlcNAc)2 and
enzymatic hydrolysis using untreated chitin was 0.35 and 0.33 mg ml− 1 (GlcNAc)3 production levels i.e. 0.24–1.68 mg ml− 1 and 0.23–1.39 mg
respectively (Fig. 2). The H2SO4 pretreated chitin resulted in a ml− 1, respectively (Table 2). The TLC analysis of the hydrolysate from
maximum of 0.76 and 0.43 mg ml− 1 of (GlcNAc)2 and (GlcNAc)3, the 9 run experiments has been shown in Fig. 3. The highest concen­
respectively (Fig. 2b). Further, NaOH pretreated chitin showed 0.72 mg tration of (GlcNAc)2 and (GlcNAc)3 was obtained after 2 h of hydrolysis.
ml− 1 of (GlcNAc)2 after 0.5 h enzyme hydrolysis; while the highest level 1.5 and 1.3 fold increase has been observed in (GlcNAc)2 and (GlcNAc)3
of (GlcNAc)3, i.e., 0.69 mg ml− 1 was detected at the beginning of production following two-factor optimization. The observed variations
enzymatic hydrolysis (Fig. 2c). The presence of (GlcNAc)2 and in the production levels of (GlcNAc)2 and (GlcNAc)3 depicted the sig­
(GlcNAc)3 at the beginning of hydrolysis for H2SO4 and NaOH pre­ nificance of optimizing enzyme and substrate ratio for the hydrolysis.
treated chitin show the rapid disintegration of chitin. Moreover, EMB The highest level of (GlcNAc)2 and (GlcNAc)3 production (1.7 and 1.4
and TBP pretreated chitin illustrated controlled conversion reaction mg ml− 1) was achieved when the ratio of TBP pretreated chitin (%, w/v)
patterns (Fig. 2d & 2e). With EMB pretreated chitin, the highest level of and chitinase (U mg− 1) from T. lanuginosus was 1:5 (Fig. 4 RUN1). The

Fig. 1. Time-course TLC analysis of GlcNAc, (GlcNAc)2 and (GlcNAc)3 generation after hydrolysis. Here, substrates such as (a) untreated chitin; (b) H2SO4; (c) NaOH;
(d) EMB and (e) TBP pretreated chitin were combined with purified T. lanuginosus chitinase during hydrolysis. The COS formation were monitored at different
interval of time (given in hours).

5
M. Kumar et al. Bioresource Technology 337 (2021) 125399

Fig. 2. Time-course analysis of GlcNAc, (GlcNAc)2, and (GlcNAc)3 generation during hydrolysis of chitin. Here, different chitinase hydrolysis product produced using
(a) untreated chitin; (b) H2SO4; (c) NaOH; (d) EMB; and (e) TBP using chitinase from T. lanuginosus are shown.

COS production. However, the significant increment in (GlcNAc)2 and


Table 2
(GlcNAc)3 production could be observed with a comparatively low
Two-factor design matrix and (GlcNAc)2 and (GlcNAc)3 yield after chitinase
solids loading using TBP pretreated chitin. The decrease in levels of
hydrolysis when using TBP pretreated chitin. Here, X1 is TBP pretreated chitin
(GlcNAc)2 and (GlcNAc)3 production at high solids loading of pretreated
(%, w/v) and X2 is T. lanuginosus chitinase (U mg− 1). The (GlcNAc)2 and
(GlcNAc)3 concentrations in mg ml− 1 following hydrolysis (2 h) were taken as chitin suggested that substrate inhibition that could lower the rate of
response. hydrolysis.
Run X1 X2 Response
(GlcNAc)2 (mg ml− 1) (GlcNAc)3 (mg ml− 1) 3.5. COS purification
1 1 5 1.68 1.39
2 10 1 0.24 0.23 The removal of impurities from the hydrolysate was done by
3 1 1 0.38 0.24 empolying cation exchange chromatography. TLC analysis of the
4 10 5 0.40 0.31
different fractions showed that the major portion of impurities and
5 5.5 3 0.45 0.38
6 1 3 0.50 0.40 GlcNAc elute in the NaCl gradient of 50 to 200 mM. The elution of
7 5.5 5 0.41 0.30 (GlcNAc)2 and (GlcNAc)3 were mainly observed in the NaCl gradient of
8 5.5 1 0.31 0.30 250 and 300 mM respectively. Wu et al. (2017) also showed the appli­
9 10 3 0.35 0.29 cability of cation exchange chromatography fro the seperation of COS
from the mixture of various COS. The active fractions for (GlcNAc)2 and
effect of (GlcNAc)2 and (GlcNAc)3 production against the two variables, (GlcNAc)3 production were desalted and subjected to HPLC analysis.
i.e., TBP pretreated chitin (%, w/v) and chitinase (U mg− 1) from The quantification of (GlcNAc)2 and (GlcNAc)3 produced via coupled
T. lanuginosus could be studied through response surface plots (Fig. 5). chemoenzymatic disintegration was performed using HPLC. COS were
The increasing level of chitinase showed significant enhancement in separated using NH2P-50E (4.6 × 250 mm) cation exchange prepartive
column using 70% acetonitrile at a flow rate of 1 ml min− 1. The

6
M. Kumar et al. Bioresource Technology 337 (2021) 125399

Fig. 3. Time-course TLC analysis of GlcNAc, (GlcNAc)2 and (GlcNAc)3 generation from TBP pretreated chitin under different RUN conditions.

retention depends on the hydrophilic interaction between the stationary chip2 from Paenibacilus pasadenesis) hydrolysis system. In another study
phase and its hydroxyl amino group content in COS. The presence of chitinase from S. albolongus ATCC 27,414 was employed on 1-ethyl-3-
more hydroxyl and amino groups in COS enhances the hydrogen- methylimidazolium acetate pretreated chitin and resulted in the gen­
bonding interactions with the stationary phase, which influences the eration of (GlcNAc)2 following 48 hr of hydrolysis (Li et al., 2019).
retention time. The retention time for (GlcNAc)2 and (GlcNAc)3 in the Additionally, the present study could be considered significant in terms
standard samples and TBP pretreated chitin hydrolysate after 2 h was of additional generation of (GlcNAc)3 besides (GlcNAc)2.
found 5.2 and 8.2 min. respectively (see supplementary materials). The
final concentration of (GlcNAc)2 and (GlcNAc)3 achieved was 1.05 and 4. Conclusions
1.02 mg ml− 1. About 0.1 g of both (GlcNAc)2 and (GlcNAc)3 could be
purified when 1 g of TBP pretreated chitin was hydrolyzed using chiti­ A new chemoenzymatic method for production of short chain COS
nase with a total COS yield of 20%. In another study, when chitin was has been developed using ILs pretreated chitin followed by hydrolysis
pretreated using 1-ethyl-3-methylimidazolium acetate IL followed by using purified chitinase from T. lanuginosus. The highest levels of COS
enzymatic hydrolysis through chitinase from Streptomyces griseues that ((GlcNAc)2, 1.10; (GlcNAc)3, 1.07 mg ml− 1)) could be achieved from
resulted in the generation of 0.06 g of (GlcNAc)2 from 1 g of chitin TBP pretreated chitin. Further, two-factor optimization and chromato­
(Husson et al., 2017). Similarly, Significant production of (GlcNAc)2 has graphic seperation resulted in generation of 0.1 g of both (GlcNAc)2 and
been reported by Xu et al. (2018) from 1-butyl-3methylimidazolium (GlcNAc)3 from TBP pretreated chitin (1 g). The study will open up the
acetate pretreated crab shell chitin and double-chitinase (chip1 and way for further studies to enhance the COS yield with simultaneous cost

7
M. Kumar et al. Bioresource Technology 337 (2021) 125399

Fig. 4. Time-course analysis of GlcNAc, (GlcNAc)2, and (GlcNAc)3 production from TBP pretreated chitin.

reduction to benefit industries that are dependent on these substrates for CRediT authorship contribution statement
development of several agricultural and health products along with
valorization of marine waste. Manish Kumar: Data curation, Methodology, Formal analysis,
Writing - original draft. Jogi Madhuprakash: Resources, Writing - re­
view & editing. Venkatesh Balan: Resources, Formal analysis, Writing -

8
M. Kumar et al. Bioresource Technology 337 (2021) 125399

facility. The authors also highly acknowledged the Central University of


Rajasthan, Rajasthan, India for providing the necessary facilities to
complete this work. Dr. Balan thank DOD (Grant No. W911NF2010281)
for partially funding this project and State of Texas for his startup funds.

Appendix A. Supplementary data

Supplementary data to this article can be found online at https://doi.


org/10.1016/j.biortech.2021.125399.

References

Brandt, A., Gräsvik, J., Hallett, J.P., Welton, T., 2013. Deconstruction of lignocellulosic
biomass with ionic liquids. Green Chem. 15, 550–583.
Chiappe, C., Pieraccini, D., 2005. Ionic liquids: Solvent properties and organic reactivity.
J. Phys. Org. Chem. 18, 275–297.
Dahmane, E.M., Taourirte, M., Eladlani, N., Rhazi, M., 2014. Extraction and
characterization of chitin and chitosan from Parapenaeus longirostris from Moroccan
local sources. Int. J. Polym. Anal. Ch. 19, 342–351.
Das, S.N., Madhuprakash, J., Sarma, P., Purushotham, P., Suma, K., Manjeet, K.,
Rambabu, S., Gueddari, N.E.E., Moerschbacher, B.M., Podile, A.R., 2015.
Biotechnological approaches for field applications of chitooligosaccharides (COS) to
induce innate immunity in plants. Crit. Rev. Biotechnol. 35, 29–43.
Feás, X., Vázquez-Tato, M.P., Seijas, J.A., Nikalje, P.G., Fraga-López, F., 2020. Extraction
and physicochemical characterization of chitin derived from the Asian hornet, Vespa
velutina Lepeletier 1836 (Hym.: Vespidae). Molecules 25. https://doi.org/10.3390/
molecules25020384.
Focher, B., Beltrame, P., Naggi, A., Torri, G., 1990. Alkaline N-deacetylation of chitin
enhanced by flash treatments. Reaction kinetics and structure modifications.
Carbohydr. Polym. 12, 405–418.
Gbenebor, O.P., Adeosun, S.O., Lawal, G.I., Jun, S., Olaleye, S.A., 2017. Acetylation,
crystalline and morphological properties of structural polysaccharide from shrimp
exoskeleton. Eng. Sci. Technol. Int. J. 20, 1155–1165.
Guoxiang, L., Yumin, D., Yongzhen, T., Yating, L., Sheng, L., Xianwen, H., Jianhong, Y.,
2010. Dilute solution properties of four natural chitin in NaOH/urea aqueous
system. Carbohydr. Polym. 80, 970–976.
Hamed, I., Özogul, F., Regenstein, J.M., 2016. Industrial applications of crustacean by-
products (chitin, chitosan, and chitooligosaccharides): A review. Trends Food Sci.
Technol. 48, 40–50.
Hamer, S.N., Cord-Landwehr, S., Biarnés, X., Planas, A., Waegeman, H.,
Moerschbacher, B.M., Kolkenbrock, S., 2015. Enzymatic production of defined
chitosan oligomers with a specific pattern of acetylation using a combination of
chitin oligosaccharide deacetylases. Sci. Rep. 5 https://doi.org/10.1038/srep08716.
Hamid, R., Khan, M.A., Ahmad, M., Ahmad, M.M., Abdin, M.Z., Musarrat, J., Javed, S.,
2013. Chitinases: An update. J. Pharm. Bioallied Sci. 5, 21. https://doi.org/10.4103/
0975-7406.106559.
Harmsen, R.A., Aam, B.B., Madhuprakash, J., Hamre, A.G., Goddard-Borger, E.D.,
Withers, S.G., Eijsink, V.G., Sørlie, M., 2020. Chemoenzymatic synthesis of chito-
oligosaccharides with alternating N-D-acetylglucosamine and D-glucosamine. ACS
Biochem. 2020 https://doi.org/10.1021/acs.biochem.0c00839.
Hou, Q., Ju, M., Li, W., Liu, L., Chen, Y., Yang, Q., 2017. Pretreatment of lignocellulosic
Fig. 5. Response surface plots of (a) (GlcNAc)2 and (b) (GlcNAc)3 production as biomass with ionic liquids and ionic liquid-based solvent systems. Molecules 22.
a function of chitinase and TBP pretreated chitin. All the value of response are https://doi.org/10.3390/molecules22030490.
expressed as the mean values of three independent assays ± stan­ Husson, E., Hadad, C., Huet, G., Laclef, S., Lesur, D., Lambertyn, V., Jamali, A., Gottis, S.,
dard deviations. Sarazin, C., Van Nhien, A.N., 2017. The effect of room temperature ionic liquids on
the selective biocatalytic hydrolysis of chitin via sequential or simultaneous
strategies. Green Chem. 19, 4122–4131.
review & editing. Amit Kumar Singh: Formal analysis, Writing - review Ibitoye, E., Lokman, I., Hezmee, M., Goh, Y., Zuki, A., Jimoh, A., 2018. Extraction and
physicochemical characterization of chitin and chitosan isolated from house cricket.
& editing. V. Vivekanand: Formal analysis, Writing - review & editing.
Biomed. Mater. 13 https://doi.org/10.1088/1748-605X/aa9dde.
Nidhi Pareek: Conceptulization, Funding acquisition, Supervision, Jung, W.-J., Park, R.-D., 2014. Bioproduction of chitooligosaccharides: Present and
Project administration, Writing - review & editing. perspectives. Mar. Drugs 12, 5328–5356.
Kaczmarek, M.B., Struszczyk-Swita, K., Li, X., Szczesna-Antczak, M., Daroch, M., 2019.
Enzymatic modifications of chitin, chitosan and chitooligosaccharides. Front.
Bioeng. Biotechnol. 7, 243. https://doi.org/10.3389/fbioe.2019.00243.
Declaration of Competing Interest Kidibule, P.E., Santos-Moriano, P., Jiménez-Ortega, E., Ramírez-Escudero, M., Limón, M.
C., Remacha, M., Plou, F.J., Sanz-Aparicio, J., Fernández-Lobato, M., 2018. Use of
The authors declare that they have no known competing financial chitin and chitosan to produce new chitooligosaccharides by chitinase Chit42:
Enzymatic activity and structural basis of protein specificity. Microb. Cell Fact. 17
interests or personal relationships that could have appeared to influence https://doi.org/10.1186/s12934-018-0895-x.
the work reported in this paper. Kidibule, P.E., Santos-Moriano, P., Plou, F.J., Fernández-Lobato, M., 2020. Endo-
chitinase Chit33 specificity on different chitinolytic materials allows the production
of unexplored chitooligosaccharides with antioxidant activity. Biotechnol. Rep. 27
Acknowledgments https://doi.org/10.1016/j.btre.2020.e00500.
Kumar, M., Brar, A., Vivekanand, V., Pareek, N., 2017. Production of chitinase from
The authors would like to acknowledge the Department of Science thermophilic Humicola grisea and its application in production of bioactive
chitooligosaccharides. Int. J. Biol. Macromol. 104, 1641–1647.
and Technology (Grant No. DST/INSPIRE/04/2014/002644) and Sci­ Kumar, M., Brar, A., Vivekanand, V., Pareek, N., 2018. Process optimization, purification
ence and Engineering Research Board (Grant No. ECR/2018/002633), and characterization of a novel acidic, thermostable chitinase from Humicola grisea.
Government of India for financial support. Authors also acknowledged Int. J. Biol. Macromol. 116, 931–938.

Material Research Centre, MNIT, Jaipur, India and Institute Instru­


mentation Centre, IIT Roorkee, Roorkee, India for instrumentation

9
M. Kumar et al. Bioresource Technology 337 (2021) 125399

Kumari, S., Rath, P., Kumar, A.S.H., Tiwari, T.N., 2015. Extraction and characterization Sánchez, Á., Mengíbar, M., Fernández, M., Alemany, S., Heras, A., Acosta, N., 2018.
of chitin and chitosan from fishery waste by chemical method. Environ. Technol. 3, Influence of preparation methods of chitooligosaccharides on their physicochemical
77–85. properties and their anti-inflammatory effects in mice and in RAW264. 7
Lei, Z., Chen, B., Koo, Y.-M., MacFarlane, D.R., 2017. Introduction: Ionic liquids. Chem. Macrophages. Mar. Drugs. 16 https://doi.org/10.3390/md16110430.
Rev. 117 https://doi.org/10.1021/acs.chemrev.7b00246. Şenel, B., 2019. In vitro preliminary studies of chitooligosaccharide coated
Li, J., Huang, W.-C., Gao, L., Sun, J., Liu, Z., Mao, X., 2019. Efficient enzymatic nanostructured lipidic nanoparticles for efficient gene delivery. J. Res. Pharma. 23,
hydrolysis of ionic liquid pretreated chitin and its dissolution mechanism. 671–681.
Carbohydr. Polym. 211, 329–335. Shamshina, J.L., Berton, P., 2020. Use of ionic liquids in chitin biorefinery: A systematic
Li, K., Xing, R., Liu, S., Li, P., 2016. Advances in preparation, analysis and biological review. Front. Bioeng. Biotechnol. 8 https://doi.org/10.3389/fbioe.2020.00011.
activities of single chitooligosaccharides. Carbohydr. Polym. 139, 178–190. Singh, A., Benjakul, S., Huda, N., Xu, C., Wu, P., 2020. Preparation and characterization
Liaqat, F., Eltem, R., 2018. Chitooligosaccharides and their biological activities: A of squid pen chitooligosaccharide–epigallocatechin gallate conjugates and their
comprehensive review. Carbohydr. Polym. 184, 243–259. antioxidant and antimicrobial activities. RSC Adv. 10, 33196–33204.
Liu, Y., Xing, R., Liu, S., Qin, Y., Li, K., Yu, H., Li, P., 2019. Effects of Sivaramakrishna, D., Bhuvanachandra, B., Mallakuntla, M.K., Das, S.N., Ramakrishna, B.,
chitooligosaccharides supplementation with different dosages, molecular weights Podile, A.R., 2020. Pretreatment with KOH and KOH-urea enhanced hydrolysis of
and degrees of deacetylation on growth performance, innate immunity and α-chitin by an endo-chitinase from Enterobacter cloacae subsp. cloacae. Carbohydr.
hepatopancreas morphology in Pacific white shrimp (Litopenaeus vannamei). Polym. 235 https://doi.org/10.1016/j.carbpol.2020.115952.
Carbohydr. Polym. 226 https://doi.org/10.1016/j.carbpol.2019.115254. Steudte, S., 2014. Investigations on the stability and ecotoxicity of selected ionic liquid
Lodhi, G., Kim, Y.-S., Hwang, J.-W., Kim, S.-K., Jeon, Y.-J., Je, J.-Y., Ahn, C.-B., Moon, S.- cations and anions. Ph.D. thesis. University of Bremen.
H., Jeon, B.-T., Park, P.-J., 2014. Chitooligosaccharide and its derivatives: Suryawanshi, N., Jujjavarapu, S.E., Ayothiraman, S., 2019. Marine shell industrial
Preparation and biological applications. BioMed Res. Int. 2014 https://doi.org/ wastes–an abundant source of chitin and its derivatives: Constituents, pretreatment,
10.1155/2014/654913. fermentation, and pleiotropic applications-A revisit. Int. J. Environ. Sci. Technol. 16,
Mohan, M., Deshavath, N.N., Banerjee, T., Goud, V.V., Dasu, V.V., 2018. Ionic liquid and 1–22.
sulfuric acid-based pretreatment of bamboo: Biomass delignification and enzymatic Usmani, Z., Sharma, M., Gupta, P., Karpichev, Y., Gathergood, N., Bhat, R., Gupta, V.K.,
hydrolysis for the production of reducing sugars. Ind. Eng. Chem. Res. 57, 2020. Ionic liquid based pretreatment of lignocellulosic biomass for enhanced
10105–10117. bioconversion. Bioresour. Technol. 304 https://doi.org/10.1016/j.
Mukherjee, S., Behera, P.K., Madhuprakash, J., 2020. Efficient conversion of crystalline biortech.2020.123003.
chitin to N, N’-diacetylchitobiose by the enzyme cocktail produced by Paenibacillus Villa-Lerma, G., Gonzalez-Marquez, H., Gimeno, M\., Lopez-Luna, A., Barzana, E., Shirai,
sp. LS1. Carbohydr. Polym. 250 https://doi.org/10.1016/j.carbpol.2020.116889. K., 2013. Ultrasonication and steam-explosion as chitin pretreatments for chitin
Ogata, M., 2020. Chemoenzymatic synthesis and function of chitin derivatives. Curr. oligosaccharides production by chitinases of Lecabicillium lecanii. Bioresorce.
Pharm. Des. 26, 3522–3529. Technol. 146, 794-798.
Pan, M., Li, J., Lv, X., Du, G., Liu, L., 2019. Molecular engineering of chitinase from Wu, Y., Lu, W.P., Wang, J., Gao, Y., Guo, Y., 2017. Rapid and convenient separation of
Bacillus sp. DAU101 for enzymatic production of chitooligosaccharides. Enzyme chitooligosaccharides by ion-exchange chromatography. IOP Conference Series:
Microb. Technol. 124, 54–62. Materials Science and Engineering 275, 012008.
Phil, L., Naveed, M., Mohammad, I.S., Bo, L., Bin, D., 2018. Chitooligosaccharide: An Xu, T., Qu, M., Liu, H., Cao, D., Xu, C., Wang, L., Qi, B., 2020. Chitin degradation
evaluation of physicochemical and biological properties with the proposition for potential and whole-genome sequence of Streptomyces diastaticus strain CS 1801.
determination of thermal degradation products. Biomed. Pharmacother. 102, AMB Expr. 10, 29.
438–451. Xu, P., Wu, X.L., Guo, X.X., Tang, J., Zong, M.H., Lou, W.Y., 2018. Double-chitinase
Purushotham, P., Sarma, P., Podile, A.R., 2012. Multiple chitinases of an endophytic hydrolysis of crab shell chitin pretreated by ionic liquid to generate chito-
Serratia proteamaculans 568 generate chitin oligomers. Bioresour. Technol. 112, oligosaccharide. ACS Sustain. Chem. Eng. 7, 1683–1691.
261–269. Yang, F., Luan, B., Sun, Z., Yang, C., Yu, Z., Li, X., 2017. Application of
Queda, F., Covas, G., Silva, T., Santos, C.A., Bronze, M.R., Cañada, F.J., Corvo, M.C., chitooligosaccharides as antioxidants in beer to improve the flavour stability by
Filipe, S.R., Marques, M.M.B., 2019. A top-down chemo-enzymatic approach protecting against beer staling during storage. Biotechnol. Lett. 39, 305–310.
towards N-acetylglucosamine-N-acetylmuramic oligosaccharides: Chitosan as a Zhao, M., Gu, L., Li, Y., Chen, S., You, J., Fan, L., Wang, Y., Zhao, L., 2019.
reliable template. Carbohydr. Polym. 224 https://doi.org/10.1016/j. Chitooligosaccharides display anti-tumor effects against human cervical cancer cells
carbpol.2019.115133. via the apoptotic and autophagic pathways. Carbohydr. Polym. 224 https://doi.org/
10.1016/j.carbpol.2019.115171.

10

You might also like