You are on page 1of 8

Food Chemistry 256 (2018) 419–426

Contents lists available at ScienceDirect

Food Chemistry
journal homepage: www.elsevier.com/locate/foodchem

Review

Modifications of konjac glucomannan for diverse applications T

Fan Zhu
School of Chemical Sciences, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand

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

Keywords: Konjac glucomannan (KGM) is a major polysaccharide from the corm of Amorphophallus konjac. Native KGM has
Film limited uses and has been chemically/physically/enzymatically modified to expand the range of functional
Encapsulation properties. This mini-review summarises the recent advances of modifying KGM for diverse food and nonfood
Biodegradable applications, focusing on the chemical and physical modifications. The chemical methods include substitution,
Chemical and physical modifications
grafting, cross-linking, oxidation, and deacetylation, whereas the physical modifications are electrospinning,
Super absorbent
microfluidic spinning, γ-irradiation, extrusion, and electric field processing. The modified KGM has been used in
Konjac glucomannan
Underutilized species a range of different applications, including biodegradable film, emulsion, medical and pharmaceutical material,
encapsulation and controlled release, fish feed and functional food ingredient, separation medium, aerogel,
liquid crystal, absorbent for removal of pollutants in waste water, and so on. These KGM-based products tend to
be biodegradable, biocompatible, and non-toxic with improved functional performance.

1. Introduction There are various health effects associated with the consumption of
KGM and related products. The health effects include weight loss and
Konjac (Amorphophallus konjac) of the Araceae (aroid) family is satiety, oral health, enhancing the growth and viability of beneficial
native to warm subtropical and tropical Asia. It is widely grown as a organisms in colon, binding of nutrients (e.g., cholesterol), and so on
cash crop with high yields in countries such as China, India, and Japan (Tester & Al-Ghazzewi, 2016). There has been increasing interest in
(Behera & Ray, 2017). Apart from starch, glucomannan is the major using KGM for the formulation of functional foods.
component of the konjac corm which may accounts for ∼70% of the Apart from food applications, KGM has been used in other areas
dry weight of refined flour (Behera & Ray, 2017; Huang, Jin, Ye, Hu, & such as for biomedical and environmental uses. Compared with syn-
Wang, 2016; Zhu, 2016). Konjac glucomannan (KGM) is consisted of D- thetic and fossil oil based polymers, natural polysaccharides including
mannose and D-glucose residues (linked by β-1,4 bonds) with a ratio of KGM tend to be renewable, environmentally friendly (biodegradable),
1.6:1 or 1.4:1, depending on the genotypes (Behera & Ray, 2017; and biocompatible (Zia, Zia, Zuber, Ahmad, & Muneer, 2016). KGM has
Zhang, Chen, & Yang, 2014). On average, there is an acetyl group on been a research focus in the last few years. This is partially reflected by
the C-6 position of the backbone per every 9–19 sugar residues. Side some recent reviews on various aspects of KGM (Behera & Ray, 2017;
chains may exist with a degree of branching of ∼8%. The molecular Tester & Al-Ghazzewi, 2016; Zhang et al., 2014; Zia et al., 2016). For
weight of KGM ranges from 500 k to 2000 k, depending on the plant example, Zhang et al. (2014) reviewed the modifications of KGM for the
source and processing methods. KGM tends to have a homogeneous size uses in oral colon targeting drug delivery systems. Zia et al. (2016)
distribution with a polydispersity (MW/Mn) of 1.21 (Behera & Ray, reviewed the properties and applications of KGM based polyurethanes.
2017; Zhang et al., 2014). The nutritional properties and health effects of KGM and the hydro-
KGM has good water binding and gelation capacity as well as film lyzed products have also been reviewed lately (Behera & Ray, 2017;
forming property (Zhang et al., 2014). These functional properties Tester & Al-Ghazzewi, 2016). These reviews pointed out that native
make konjac corm powder and KGM excellent food ingredients. Konjac KGM has a very limited range of functionalities. It is commonly mod-
powder has been a food item in China and Japan since ancient times. ified to expand the functional variation for diverse applications (Zhang,
There are a few KGM based food products from the markets such as Xie, & Gan, 2005). During the last few years, many different types of
noodles and cakes which are locally popular. KGM is a GRAS (generally chemical and physical modification methods such as grafting and
regarded as safe) food additive as approved by Health Canada in Ca- electrospinning have been used to create new properties of KGM for
nada and FDA (Food and Drug Administration) in USA. It is also ap- desired applications (e.g., as wound dressing hydrogel and for the re-
proved by European Union as E425 (Tester & Al-Ghazzewi, 2016). moval of heavy metals from waste water) (Chen, Zhang, Ding, Lin, &

Abbreviations: CKGM, carboxymethyl KGM; DS, degree of substitution; DP, degree of polymerization; KGM, konjac glucomannan; TEMPO, 2,2,6,6-tetramethyl-1-piperidiny-1-oxyl
E-mail address: fzhu5@yahoo.com.

https://doi.org/10.1016/j.foodchem.2018.02.151
Received 6 September 2017; Received in revised form 21 November 2017; Accepted 27 February 2018
Available online 06 March 2018
0308-8146/ © 2018 Elsevier Ltd. All rights reserved.
F. Zhu Food Chemistry 256 (2018) 419–426

Lu, 2017; Shahbuddin, Bullock, MacNeil, & Rimmer, 2014). A review produce mixed esters varying in the DS (Danjo, Enomoto-Rogers,
on the recent progress in this field would greatly support the current Takemura, & Iwata, 2014). The mixed esters had number based mole-
exploitation of KGM as versatile biomaterial. cular weights of 3–6 × 105 and DP of > 1 × 103. The mixed esters had
The present mini-review summarises the chemical and physical a much higher decomposition temperature (∼360 °C) than KGM
modifications of KGM and the various applications, focusing on the (274 °C). The acylation of hydroxyl groups of KGM much decreased the
literatures from the past 5 years. The basics of KGM such as extraction, intramolecular dehydration, improving the thermostability. The mixed
structure, and physicochemical properties have been reviewed pre- esters of KGM were made into transparent films (Danjo et al., 2014).
viously (Behera & Ray, 2017; Zhang et al., 2014), and thus, are not Increasing DS of acetylation increased the tensile strength of the films,
covered in this review. Readers are encouraged to refer to the previous whereas increasing DS of butyrylation enhanced the elongation at
publications to gain the basic background information on KGM. There break. This is possibly due to the plasticizing effect of butyryl group.
has been a rather large number of publications on the chemical and The resulting films had comparable mechanical properties to those of
physical modifications of KGM from the last 5 years or so. Therefore, commonly used polymers such as PMMA, suggesting the potential
only representative papers have been intentionally selected in the commercial applications of KGM based films (Danjo et al., 2014).
current review in order to comprehensively cover the various mod-
ifications and applications of KGM, while not exceeding the word limit 2.1.2. Acylation with acyl carbon chains for film formation
of the Journal. The applications of the modified KGM, instead of the Apart from KGM acetate and butyrate as described in the previous
modification/synthesis process (e.g., reaction conditions), are empha- section, KGM was acylated with acyl carbon chains (n from 2 to 12)
sized to give the paper a focus. In some of the reported cases, combined (Enomoto-Rogers, Ohmomo, Takemura, & Iwata, 2014), which has
modifications were employed (e.g., both physical and chemical), which, been the only systematic study on KGM acrylated with different carbon
in this review, is categorised according to the major and emphasized chain lengths so far. DS of these KGM esters reached 3 with DP varying
type of modification. In general, this review is categorised into sub- from 0.9 × 103 (KGM laurate) to 2.6 × 103 (KGM propionate). In-
sections primarily based on the types of modifications. For example, creasing carbon number of acyl chains decreased the glass transition
within the chemical modification section, subsections on substitution-, temperature (Tg) of the samples. KGM acetate (described in the above
chain elongation-, and depolymerisation-type modifications are pro- section) was also acylated with acyl carbon chains (n from 3 to 18).
duced. Readers are also encouraged to refer to the Supplementary These mixed esters had DS ranging from 0.7 (KGM acetate stearate) to
material (figures and tables) for much increased readability and clarity. 2.2 (KGM acetate propionate) and DP varying from 100 (KGM acetate
The enzymatically modified KGM, which has been recently reviewed palmitate) to 330 (KGM acetate butyrate). Therefore, KGM was par-
(Tester & Al-Ghazzewi, 2016), is not covered here. This review may tially degraded during the synthesis of the mixed esters. The mixed
provide a basis to further develop konjac as a sustainable crop. esters of KGM had higher Tg than those of respective homoesters. The
acyl groups introduced onto KGM may disrupt the molecular interac-
2. Chemical modifications tions and the hydrogen bond formation. The esters were insoluble in
ethanol and water but soluble in tetrahydrofuran, ethyl acetate, and
Many types of chemical modification methods and the combinations chloroform. The esters were dissolved in chloroform and casted into
have been conducted on KGM (Supplementary Table 1 and Fig. 1). The transparent and colorless films (Enomoto-Rogers et al., 2014). In-
major modifications include substitution, chain elongation, and de- creasing chain length of acryl groups increased the elongation at break,
gradation with some examples of the themes illustrated (Supplementary while decreasing the Young’s moduli and tensile strengths of the films.
Fig. 1). These modified KGM have been used in diverse applications as Overall, it becomes obvious that the functional properties of the films of
described below. KGM esters could be mostly controlled by the type and DS of acryl
groups (Danjo, Enomoto-Rogers, Takemura, & Iwata, 2014; Enomoto-
2.1. Substitution Rogers et al., 2013, 2014).

2.1.1. Acetylation for film formation 2.1.3. Carboxymethylation for film formation, paper strengthening, and
Environmental concerns require the production of biodegradable controlled release
films to replace the fossil oil based products. KGM triacetate and acetate Carboxymethyl KGM (CKGM) is one of the most commonly used
with different degrees of substitution (DS) were prepared for thermo- modified KGM. The introduced carboxymethyl group decreased the
plastic film production (Enomoto-Rogers, Ohmomo, & Iwata, 2013). At interactions (intermolecular hydrogen bonding) among KGM, rendering
a DS of 3, the number based molecular weights of the KGM triacetate the KGM more liquid like (Kobayashi, Tsujihata, Hibi, & Tsukamoto,
was 3 × 105 and the degree of polymerisation (DP) was 1 × 103. In- 2002). The effect of carboxymethylation on the water binding proper-
creasing DS of both acetates increased the decomposition temperatures ties of KGM was further studied (Xiao, Dai, Wang, Ni, & Yan, 2015). The
of KGM. Increasing DS also decreased the glass transition temperature modification decreased the equilibrium moisture content (25 °C), water
of the acetates (e.g., from 219 °C at DS of 1.3 to 178 °C at DS of 3), absorption, and solubility of KGM. Atomic force microscopy (AFM)
probably due to the effect on hydrogen bonding. The KGM acetate was analysis showed that the modification increased the folding of the KGM
made into films by casting the solution. The films were transparent. molecules, possibly making them less interactive with water molecules
Increasing DS decreased the elongation at break and the tensile strength (Xiao et al., 2015).
of the films as illustrated in Supplementary Fig. 2A, which may be due CKGM was mixed with soy protein isolate (SPI) to produce biode-
to the effect of acetylation on hydrogen bond formation and inter- gradable films (Wang et al., 2014). Both hydrogen bonding and Mail-
molecular interactions of KGM molecules (Enomoto-Rogers et al., lard reactions occurred between CKGM and SPI, making the two com-
2013). Huang, Takahashi, Kobayashi, Kawase, and Nishinari (2002) ponents compatible. The interactions increased the tensile strength and
studied the effect of acetylation on the gelation properties of KGM and elongation at break, while decreasing the oxygen permeability of the
found that increasing DS increased the elasticity of KGM gel. This ap- composite films. Increasing CKGM proportion decreased the surface
peared to be similar to the result that increasing DS decreased the roughness and water adsorption, while increasing the surface wett-
tensile strength of KGM film (Enomoto-Rogers et al., 2013), though it ability of the films (Wang et al., 2014). The decreased water adsorption
should be noted that film and gel are different structural systems. Only could be attributed to the increased hydrophobicity of KGM by car-
one study so far reported the production of KGM acetate based film boxymethylation as described by Xiao et al. (2015).
(Enomoto-Rogers et al., 2013). CKGM has been used as a paper strengthening agent (Wang, He,
An additional group (butyryl) was attached to the KGM acetate to Wang, & Song, 2015). Addition of CKGM (0.9%) increased the density,

420
F. Zhu Food Chemistry 256 (2018) 419–426

burst and tensile indexes, and folding endurance of the paper. CKGM DNA and drug delivery where emulsion systems are needed (Long et al.,
increased the bonding force between the paper fibers due to electro- 2016). KGM octenyl succinate was produced by a microwave based
static interactions. An excessive amount of CKGM caused charge re- method under alkaline conditions (Meng et al., 2014). Emulsifying
pulsion, weakening the paper structure (Wang et al., 2015). In addition capacity and emulsion stability (up to 90 days) of the KGM octenyl
to CKGM, polyamide-epichlorohydrin (PAE) was used to synergistically succinate were excellent, which indicates the potential use of this KGM
increase the wet strength of the paper. Increasing degree of carbox- product in emulsion based food/controlled release systems.
ymethylation of KGM increased both the dry and wet tensile indexes of Gurusmatika, Nishi, Harmayani, Pranoto, and Sugahara (2017) showed
the paper, which could be attributed to the altered charges of CKGM that octenyl succinate of glucomannan from A. oncophyllus possessed
(Wang et al., 2015). immunomodulatory activity by stimulating macrophages in vitro.
CKGM (negatively charged), together with 2-hydroxypropyl tri- Therefore, it may be expected that KGM octenyl succinate, apart from
methyl ammonium chloride chitosan (HACC) (positively charged), have being an emulsifier, also has this biological activity.
been used to produce nanospheres/microspheres through electrostatic
interactions for controlled release applications (Shi et al., 2016). The 2.1.7. Phthalation for waste water treatment
size and zeta potential of the composite spheres ranged from 600 to One study so far reported the phthalation of KGM for waste water
1460 nm and 39 to 50 mV, respectively. A maximum of encapsulation treatment (Zhang, Han, Yao, Pang, & Luo, 2013). KGM was reacted with
efficiency of ovalbumin (as a model ingredient) at 72% was achieved phthalic anhydride using pyridine as the catalyst to form KGM phtha-
through process optimization. In vitro controlled release test (at pH 7.4) lates with different DS up to 0.55. The synthetic process is illustrated in
showed that ovalbumin was released over a period of 24 h (Shi et al., Supplementary Fig. 1A. The KGM phthalate as a solid biosorbent was
2016). Such a composite sphere system can be used for controlled re- used to remove the copper ions in waste water (Zhang et al., 2013). Cu
lease of macromolecules such as proteins and vaccines, though in vivo (II) adsorption capacity of the KGM phthalate depended on the DS, pH,
and clinical studies remain to be conducted. and the amount of phthalate present. The Cu(II) adsorption was mostly
of chemisorption and the equilibrium was established within 2 h. At an
2.1.4. Cationization for waste water treatment and paper strengthening acidic pH, a maximum adsorption of 17 mg/g was achieved for KGM
Cationic KGM was produced by reacting with (2-methacryloylox- phthalate, which was higher than that of common biosorbents (e.g.,
yethyl)trimethylammonium chloride and 3-chloro-2-hydroxypropyl that of grape stalks was 12 mg/g). When the pH was higher than 10, a
trimethylammonium chloride as a flocculant for waste water treatment KGM phthalate (100 mg/L) effectively removed 99% of Cu(II) from the
and as a paper strengthening agent, respectively (Ren, Zhang, Qin, & Li, solution. The KGM phthalate appeared to be an efficient biodegradable
2016; Wang, He, Yan, & Song, 2017). The reaction partially decom- absorbent to remove Cu(II) in waste water (Zhang et al., 2013).
posed KGM and the grafting was initiated by free radicals on the KGM
skeleton. The modified KGM had DS ranging from 0.09 to 0.3. The 2.1.8. Sulfation for drug
cationic KGM showed good flocculating capacity at pH of 1–9 and in the KGM was hydrolyzed by sulfuric acid before sulfation reaction with
presence of salt. In a model system, the cationic KGM with a DS of 0.2 piperidine-N-sulfonic acid in DMSO or with SO3-pyridine complex in
decreased the turbidity of a kaolin suspension (0.1%) by up to 90% at pyridine at high temperature (60 °C) (Bo, Muschin, Kanamoto,
pH 7. The flocculating capacity of cationic KGM could be due to the Nakashima, & Yoshida, 2013). KGM sulfates with DS of 1.3–1.4 and
electrostatic interactions with the negatively charged contaminants in molecular weights of 0.4 × 104–1.0 × 104 were prepared (Bo et al.,
water (Ren et al., 2016). Similarly, cationic KGM also interacted with 2013). The in vitro anti-HIV and anti-coagulant activities of KGM sul-
negatively charged surface of pulp fibers, improving the paper making fates were tested. The anti-HIV capacity of the sulfate was comparable
performance (Wang et al., 2017). Quantitively, addition of cationic to that of an anti-HIV drug (ddC), whereas the blood anti-coagulant
KGM (1%) enhanced the tensile (by 11%) and burst (by 13%) indexes as activity was similar to that of curdlan and dextran sulfates. Mechanistic
well as the folding endurance (by 72%) of the paper. Scanning electron studies suggested that these bioactivities were much due to electrostatic
microscopy (SEM) analysis showed that the paper fibers became more interactions between the amino groups of targets and the KGM sulfate
compact with the addition of the cationic KGM (Wang et al., 2017). (Bo et al., 2013). A previous study showed that curdlan (a naturally
occurring polysaccharide with a linear 1,3-β pyranoside structure)
2.1.5. Cyanoethylation for liquid crystal formation sulfate also possessed anti-AIDS virus activity (Yoshida et al., 1990).
Cyanoethyl KGM with various DS (0.1–2.1) were synthesized by Curdlan sulfate had an EC50 (50% effective concentration of sulfated
adding acrylonitrile in N, N-dimethylformamide solution (Shen, Li, glucomannan on HIV) of 0.1 μg/mL, which was lower than KGM sulfate
Zhang, Wan, & Gao, 2012). The successful introduction of cyanoethyl (0.7 μg/mL). Therefore, the advantage of using KGM sulfate as an anti-
groups was confirmed by FTIR (Fourier-transform infrared spectro- AIDS drug remains to be explored in a comprehensive way.
scopy) and NMR (nuclear magnetic resonance) spectroscopy analysis
which are the routine type of analysis for verifying the modification. 2.2. Chain elongation
Cyanoethyl KGM with a DS of 2.1 dissolved in 1-allyl-3-methylimida-
zolium chloride (AMIMCl) exhibited liquid crystalline behaviors when 2.2.1. Cross-linking
the concentration reached 16%. Rheological analysis revealed the for- 2.2.1.1. Modifying rheology. KGM was cross-linked by borate for
mation of crystalline regions in isotropic phase, mesophase, and ani- rheological modifications (Ratcliffe, Williams, English, & Meadows,
sotropic phase (Shen et al., 2012). The advantages of this kind of liquid 2013). Rheological analysis of small strain deformation showed that the
crystal polymers over the synthetic ones can be non-toxicity, biode- cross-linking greatly slowed the network dynamics while increasing the
gradability, and the clearly defined thermotropic and lyotropic meso- network connectivity (Ratcliffe et al., 2013). Such a modification in
phase behaviors. KGM rheology may only provide a fundamental basis for the
formulation of food and health products, which remains to be linked
2.1.6. Esterification for emulsification to any practical application.
KGM has been esterified with different functional groups to produce
emulsifiers (Long, Pan, & Yao, 2016; Meng, Zheng, Wang, Liang, & 2.2.1.2. Waste water treatment. KGM was cross-linked with calcium
Zhong, 2014). KGM-oleic acid ester with a DS of 0.37 was produced oxide in the presence of graphene oxide (GO) to form hydrogel (Gan,
through a lipase-catalyzed reaction in isooctane (Long et al., 2016). The Shang, Hu, Yuen, & Jiang, 2015). SEM analysis showed that the
KGM ester is amphiphilic and may have potential applications as food hydrogel of KGM and GO mixtures had a much more compact
flavoring agents, surface coating materials, and medical material for structure with a rougher surface than that of KGM. The hydrogel of

421
F. Zhu Food Chemistry 256 (2018) 419–426

cross-linked KGM was used to remove dyes (methyl blue and methyl through self-aggregation with the aid of sonication (Ha et al., 2011).
orange) in waste water. The incorporation of GO greatly enhanced the The size of the formed particles was dependent on pH and ionic
dye adsorbing capacity of hydrogel. The adsorption behaviors of the strength. In vitro release test showed a sustainable releasing behavior of
hydrogel for both dyes followed a pseudo-second-order and were well etoposide from the encapsulation system (Ha et al., 2011), though in
described by the Freundlich isotherm model (Gan et al., 2015). The vivo test remains to be conducted for any practical application.
resulting hydrogel may be used for water purifications.
2.2.2.3. Enhancing oil recovery. KGM-g-AM (acrylamide)-g-AA (acrylic
2.2.1.3. Protein desalting. Another study reported the cross-linking of acid)-g-APEG (allyl polyoxyethylene ether) was produced through free-
KGM using epoxy chloropropane as the cross-linker (Xu, Yao, Yu, Zhou, radical copolymerization (Gou et al., 2017) (Supplementary Fig. 1E).
& Zhao, 2014) (Supplementary Fig. 1B). Nanospheres with the Structural analysis showed that the resulting graft-copolymer had a
diameters of 40–50 nm were produced from the cross-linked KGM much reduced crystallinity than the KGM and had a porous
through emusification. The nanospheres can be easily dispersed with microstructure. The graft-copolymer showed good biodegradability
good flowing property. They also have high moisture adsorption and with over 60% of the mass degraded in 4 weeks by a bio-degradation
desorption capacities. These properties suggested that they may be used test. Compared to partially hydrolyzed polyacrylamide, the copolymer
as adsorbents which can be easily recycled (Xu et al., 2014). Acid- showed better salt resistance, anti-shearing capacity, temperature
hydrolysed KGM was also cross-linked to produce microspheres tolerance, and desired viscoelasticity. The resistance factor
(Supplementary Fig. 2B) (Xiong, Zhou, Sun, Li, & Zhao, 2014). The (RF = 14.1) and residual resistance factor (RRF = 4.95) of the
pre-treatment by acid hydrolysis greatly reduced the viscosity of KGM copolymer were obtained from a core flood test, which are indicative
solution. The microspheres showed great stability in a range of pH of the ability for mobility control and flooding effect with improved oil
(1–14) and in commonly used buffer solutions as well as to autoclaving. recovery (by 14%). The results suggested that the KGM-g-AM-g-AA-g-
Protein-desalting experiments showed that the cross-linked KGM APEG copolymer could replace the acrylamide based conventional
microspheres showed similar performance to a commerical gel copolymers for enhanced oil recovery in oil field mining (Gou et al.,
(Sephadex G25). The modified KGM can be an economic alternative 2017).
in the protein desalting application (Xiong et al., 2014).
2.2.2.4. Molecular-imprinting for extraction of bioactive compound. KGM
2.2.2. Graft copolymers based molecular-imprinted copolymer (MIP) was produced using
2.2.2.1. Biomedical application. Three KGM based hydrogels were acrylic acid as a functional monomer and N,N′-
produced (Shahbuddin et al., 2014). KGM was cross-linked by methylenebisacrylamide as a cross-linker (Tian, Zhou, Xiong, & Lu,
termination of radicals from oxidation with Ce(IV). Semi-IPN (semi 2017) (Supplementary Fig. 1F). Caffeine was used as the template for
interpenetrating network) was formed from copolymerization of formation of the molecular-imprinted copolymer. SEM analysis showed
PEGDA [poly(ethylene glycol)-diacrylate] and NVP (N- that the molecular imprinting process produced much more
vinylpyrrolidone) with KGM. Graft-conetworks were also formed by aggregation for the copolymers to form larger particles. The
cross-linking KGM with Ce(IV). SEM analysis showed that the samples molecular imprinting technique greatly improved the adsorption
had smooth surfaces with differences in the cross-section morphology. (from 22 to 63 mg/g) and selectivity (selective factor increased from
The semi-IPNs and the graft-conetwork had porous structure possibly 0.9 to 2.5) of the copolymer for caffeine extraction. Langmuir equation
due to phase separation during the reactions. The hydrogels were well described the adsorption isotherms of the MIP. The selective
cytocompatible, while the graft-conetworks can stimulate fibroblast adsorption of caffeine by the MIP was successfully applied in a range of
metabolic activity and the migration of keratinocytes and fibroblasts. beverage samples with high recovery rates from 91 to 99%. The MIP
Therefore, the KGM based hydrogels can be used for wound healing obtained is cost-effective for rapid and sensitive extraction of caffeine
(Shahbuddin et al., 2014). from beverage samples (Tian et al., 2017). It may be expected that other
types of bioactive compounds can be extracted by KGM based MIPs,
2.2.2.2. Encapsulation. CKGM was grafted with poly(ethylene glycol) which remains to be tested. Molecularly imprinted biopolymers based
(PEG) to form graft copolymers (CKGM-g-PEG) (Li, Xia, Branham, Ha, & on other types of polysaccharides such as chitosan have been developed
Wu, 2011). The CKGM-g-PEG (0.2%) was mixed with α-cyclodextrin for extraction of targeted chemical components (Wang et al., 2014). It is
(6%) for self-assembly to encapsulate glucose oxidase (GOX) in solution not clear if the KGM based MIP has any advantage over MIPs based on
(Supplementary Fig. 1C). The self-assembly appeared in the form of other types of natural polysaccharides. Comparative studies are needed
hollow nanospheres/microspheres with the size ranging from 340 nm to to explore this aspect.
1.2 μm, depending on the DS (PEG substitution) and the concentration.
Transmission electron microscopy (TEM) analysis revealed the 2.2.2.5. Waste water treatment. CKGM was conjugated with
distinguishable outer shell and central core of the particles. A polydopamine as an absorbent to remove Pb(II) in waste water (Chen
maximum of encapsulation efficiency (67%) was recorded. The et al., 2017). This modified KGM is placed in this subsection rather than
leakage rate of GOX from the spheres was as low as 2%, while the in the carboxymethylation section (Section 2.1.3) because the grafting
substrate (glucose) easily penetrated into the spheres to get oxidized by is the major reaction resulting in the function described here (for waste
the encapsulated GOX. The encapsulation greatly increased the water treatment). The KGM based copolymer showed a high adsorption
thermostability, the pH range for optimal enzymatic activity, and the capacity (a maximum of 95 mg/g at 25 °C) and can be easily recovered
storability of GOX. The spheres showed great in vitro biocompatibility by using HCl (0.001 M) as the desorbing agent. The Pb(II) adsorption
with the L929 cells (MTT viability assay), suggesting potential process was well described by Langmuir linear model. The process was
biomedical uses (Li et al., 2011). endothermic and spontaneous in nature. The kinetics was well
CKGM (hydrophilic) was conjugated with cholesterol (hydrophobic) described by the intra-particle diffusion model and a pseudo-second
to become amphiphilic (Ha, Wu, Wang, Peng, & Ding, 2011) order equation. Possible interactions were electrostatic attraction and
(Supplementary Fig. 1D). The amphiphilic conjugates formed self-ag- chelation between the functional groups (eOH/eNH2) of the
gregates with very low critical aggregation concentrations of copolymer and Pb(II) in solution (Chen et al., 2017).
2.59 × 10−3–5.89 × 10−3 mg/mL. The size of the self-aggregates
ranged from 391 nm to 1 μm as revealed by dynamic laser light-scat- 2.2.2.6. Water adsorption. KGM was conjugated with acrylic acid and
tering technique. The amphiphilic conjugate was used to encapsulate acrylamide (KGM-AA-AM) or acrylic acid, acrylamide, and kaolin
etoposide (a hydrophobic drug as a model bioactive compound) (KGM-AA-AM-K) through a frontal polymerization technique (Li, Ji,

422
F. Zhu Food Chemistry 256 (2018) 419–426

Xia, & Li, 2012). SEM analysis revealed very porous microstructure of through hydrogen bonding. The composite aerogel showed great ca-
both co-polymers. The copolymer KGM-AA-AM-K was used as a water pacity to remove arsenite [As(III)] (up to 30 mg/g) and arsenate [As
absorbent, and showed great water adsorption capacity (1.9 kg/g in (V)] (up to 12 mg/g) in solution (pH 7, 50 °C) (Ye et al., 2017). KGM
distilled water and 93 g/g in saline). The porous structure, numerous based aerogels, which are biodegradable, may be used as filter tip of
OH groups of KGM, and the charge on kaolin contributed to the high cigarettes to reduce the impact on the environment and to filter away
water adsorption of the KGM based copolymer (Li et al., 2012). Li et al. some harmful substance of the smoke. Another study also explored the
(2009) synthesized KGM-g-acrylic acid polymers using a frontal potential of deacetylated KGM-soy protein isolate (SPI) conjugate for
polymerization technique, and found that the maximum water the removal of Cu(II) which is one of the most common pollutants in
absorbency reached 1.3 kg/g, which is not as high as the result of Li waste water (Liu et al., 2016). Comparative analysis showed that the
et al. (2012). Chen, Zhang, and Li (2015) prepared a superabsorbent of incorporation of SPI into the deacetylated KGM greatly increased the Cu
KGM graft-acrylic acid polymers using trimethylolpropane (II) adsorption capacity in solution by 5 times (up to 63 mg/g at 25 °C).
trimethacrylate as a cross-linker. The swelling of this co-polymers This increase could be attributed to the strong chelating property of SPI
reached a maximum at pH 7.4, though the water absorbency was not for the ion. The adsorption process also followed a pseudo-second-order
studied. Yang, Zhang, Yang, Yan, and Jiang (2017) found that the equation. Luo, Liu, Deng, and Lin (2011) explored the application of
aggregate size of KGM based super-absorbents greatly affected the carboxylic acid functionalized deacetylated KGM for the removal of
water adsorption capacity, though the structure of the studied copper(II) from aqueous solution in a fixed-bed column system. The
absorbents was not specified. Small particles with a lower adsorption cylinder shaped KGM based material was prepared by a single screw
capacity reached the maximum of adsorption faster than large particles extruder. SEM analysis showed that the material was porous. The ad-
which tend to have a higher adsorption capacity (Yang et al., 2017). sorption–desorption analysis of copper(II) removal showed that a
Overall, it appeared that KGM based graft copolymers can possess great maximum adsorption of 35 mg/g was achieved. The adsorbed copper
water adsorption capacity which may be maximized by altering their ions could be easily desorbed by using HCl solution (0.01 mol/L) with
structural properties. There are super-absorbents based on other types an efficiency of 98% (Luo et al., 2011). The above mentioned studies all
of natural polysaccharides such as starch (Zhong et al., 2013). showed that different deacetylated KGM based material have great
Comparative studies are needed to reveal if KGM based super- potential for the removal of heavy metals from waste water. However,
absorbents may have any advantage over those of other natural in order to be commercially successful, cost estimation and technolo-
polysaccharides. gical feasibility for large scale production should be analyzed to reveal
which one of these reported KGM polymers may be practically em-
2.3. Depolymerization ployed.

2.3.1. Deacetylation for wound dressing, moisture-resistant film, and waste 2.3.2. Oxidation for encapsulation, and fish feed and hard capsule
water treatment formulation
KGM is deacetylated in an alkali solution [e.g., Ca(OH)2] and the Oxidized KGM has been used as shell material for hard capsule
deacetylated KGM has been used to formulate wound dressing and preparation, encapsulation of bioactives for controlled release, and as
moisture-resistant films (Huang, Chu, Huang, Wu, & Tsai, 2015; Wei an ingredient for fish feed formulation (Chen, Zhao, Liu, Li, & Liu, 2016;
et al., 2015), aerogel for arsenic removal in solution (Ye, Jin, Huang, Shi et al., 2017; Zhang et al., 2013). KGM was TEMPO (2,2,6,6-tetra-
Hu, & Li, 2017), and biosorbent for the removal of copper(II) from methyl-1-piperidiny-1-oxyl)-oxidized on the C-6 position of sugar units
waste water (Liu, Zou, Peng, Hu, & Liu, 2016). The deacetylated KGM to various degrees (up to 80%) (Chen et al., 2016). Degree of oxidation
can more easily form junction zones and network due to the reduced at 80% gave increased transparency and solubility of KGM, while de-
steric hindrance (Huang et al., 2015). The Ca(OH)2-modified KGM creasing the viscosity. The increasing number of carboxyl groups may
showed great in vitro biocompatibility with L929 fibroblast and HaCaT enhance the charge repulsion among the KGM molecules and disrupt
keratinocyte cells. In vivo tests employing male Sprague-Dawley rats their interactions for decreased viscosity (Chen et al., 2016). The oxi-
were also conducted. The results showed that the films of deacetylated dized KGM was made into films for hard capsule formulation. SEM
KGM inhibited the activation and absorption of platelets, while pro- analysis showed that the oxidation process increased the smoothness of
moting wound contractility at an early stage of healing. Collagen se- KGM films. The oxidation increased the hydrophobicity, tensile
cretion and development of epithelial coverage and granulation tissues strength, and elongation at break of the films. The introduced COO−
in the wounds of rats treated with the films were recorded for 2 weeks groups may efficiently bind more water which may be a plasticizer for
after the surgery. Overall, the deacetylated KGM has great potential for the films to increase the elongation. In vitro drug (erythromycin stearate
wound dressing applications (Huang et al., 2015). as a model compound) dissolution test (0.1 M HCl and 37 °C) showed
Deacetylated KGM solution was casted into film which was further that the shell rupture time for the KGM of 80% oxidation was 5–10 min
coated with shellac and stearic acid mixtures by brushing (Wei et al., with 80% of the drug being released within 30–45 min. Overall, the
2015). The moisture resistant properties of the resulting composite oxidized KGM appeared to be suitable for making the gastric soluble
films were studied. The shellac based coating, where the stearic acid hard capsules (Chen et al., 2016).
was dispersed, intimately adhered onto surface of the films of deace- The COO− groups of TEMPO-oxidized KGM were cross-linked by
tylated KGM through physical interactions. The coating, which is hy- Fe3+ to form microspheres for the encapsulation of anthocyanins (hy-
drophobic, greatly improved the moisture barrier properties and tensile drophilic) and β-carotene (hydrophobic) through ultrasonic emulsifi-
strength of the films. Increasing concentration of stearic acid greatly cation (Shi et al., 2017). The microspheres were further coated with
decreased the water vapor permeability of the composite film, while chitosan (positively charged) for targeted delivery. The co-encapsula-
increasing the water contact angle. The high moisture-resistant prop- tion of both bioactives was confirmed by confocal laser scanning mi-
erties of the coated KGM based films may find applications in food croscopy. The controlled release of both bioactives were studied in vitro
products which need strict moisture management (Wei et al., 2015). using a simulated gastrointestinal tract (Shi et al., 2017). The co-en-
Deacetylated KGM was mixed with graphene (GO), iron, and man- capsulation system effectively retained both anthocyanins and β-car-
ganese oxide (FMO) in solution before freeze-drying to form composite otene in the gastric environment, and released them under the in-
aerogel (Supplementary Fig. 2C) (Ye et al., 2017). SEM analysis showed testinal conditions. The co-encapsulation also increased the thermal
that the aerogels are highly porous with very rough surface. The stability of the antioxidants (Shi et al., 2017).
aerogels with the addition of GO had great mechanical strength, which KGM was oxidized by H2O2 to be used as a feed ingredient for a type
was attributed to the molecular interactions between KGM and GO of fish (Schizothorax prenanti) (Zhang, Wu, Wang, & Wang, 2013). Up to

423
F. Zhu Food Chemistry 256 (2018) 419–426

8 g/kg of oxidized KGM was added into the fish diet. Various physio- tungstic acid ions got cross-linked with the OH groups of the C-6 po-
logical and biochemical markers of the fish, such as specific growth sition of KGM sugar units (Wang, Jiang, Lin, Pang, and Liu, 2016). A
rate, erythrocyte phagocytic index, and content of IgM, were monitored maximum of springiness of 1.21 mm for the composite gel was obtained
and compared with those of the control group. The addition of the under optimized conditions (sodium tungstate concentration of 0.3%,
modified KGM into the feed significantly improved the growth and aslo KGM concentration of 0.5%, voltage of 25 V, and treatment time of
the immune functions of the fish (Zhang et al., 2013). These effects may 12 min). SEM analysis showed that the gel was made of thin strands and
be related to the improvement of oxidized KGM on the gut health of the was highly porous with non-uniformed pore size. The gels showed
fish. Two more similar studies confirmed that oxidized KGM could pseudo-plastic behaviors and had low flow behavior indexes. The
improve intestinal morphology and enhance the growth performance, freeze-dried gels showed a high porosity with a homogeneous dis-
while modulating intestinal microflora of S. prenanti (Zhang, Wu, Xu, & tribution of pores. The KGM-tungsten gels were produced on positive
Yao, 2017; Zheng, Wu, & Xu, 2015). However, the structure of the electrode and may be used to carry bioactives/drugs with negative
oxidized KGM should be studied to explore the structure–function re- charges for targeted delivery (Wang, Zhuang, et al., 2016). Preparation
lationship of the product. of gels using electric fields may become a facile technique in creating
novel properties of KGM based products.
3. Physical modifications
3.4. Extrusion for modified rheology and water binding properties
There has been increasing demand for non-chemically and physi-
cally modified ingredients from the market due to consumers’ pre- KGM was extruded under 4 different conditions (solid content of 24
ference for “natural and healthy” products. A range of physical mod- or 33%, temperature at 90 or 110 °C, with or without die restriction)
ifications, including electrospinning, microfluidic spinning, extrusion, before grinding (Tatirat, Charoenrein, & Kerr, 2012). The extrusion
electric field processing, and γ-irradiation, have been conducted on somewhat increased the size (e.g., from 112 to 144 μm) and roughness
KGM to create a range of novel functionalities (Supplementary Table 2). of the KGM particles, while decreasing the water adsorption up to one
Some of the physical modifications such as extrusion and γ-irradiation third of the original value. The degree of crystallinity increased when
also induce chemical changes on KGM, altering the functional proper- the die restriction and high temperature (110 °C) were used. The visc-
ties. osity of KGM solution (0.5%) decreased, which could be readily at-
tributed to the decreased molecular weight induced by extrusion (from
3.1. Electrospinning for biomedical scaffold 1.2 × 106 to 2.7 × 105). The shearing and higher temperature of ex-
trusion conditions mostly affected the properties of KGM. The extruded
KGM and chitosan (ratio of 60/15–50/25 with a total solid con- KGM with reduced water absorption and viscosity may reduce the
centration of 1.5%) were mixed in a diluted acetic acid solution (10%, choking risk of ingesting KGM-containing products (Tatirat et al.,
v/v) for electrospinning to form nanofibers (Nie, Shen, Zhou, Jiang, & 2012). The extruded KGM may be used in the formulation of food,
Chen, 2011). Increasing chitosan concentration decreased the diameter health and nutraceutical products with much reduced choking risk.
of the nanofibers from 350 to 180 nm (Supplementary Fig. 2D). KGM in
aqueous solution was also electro-spun to form nanofibers, and in- 3.5. γ-Irradiation
creasing KGM concentration from 0.6 to 1.2% increased the diameters
of the nanofibers from 150 to 350 nm. The in vitro biocompatibility of 3.5.1. Degradation
the resulting nanofibers was studied to reveal their potential biomedical Prawitwong, Takigami, and Phillips (2007) studied the impact of γ-
uses. The resulting nanofibers showed great adherence to bone me- irradiation (0.5–100 kGy) on molar mass and properties of KGM. In-
senchymal stem cells than the bulk films (prepared from solution creasing irradiation dosage increased the free radical yield which
casting) due to the nanofibrous and highly porous nature (Nie et al., caused chain scission. The molecular weight and intrinsic viscosity of
2011). Zhu, Lin, Zhang, and Luo (2015) prepared KGM grafted with KGM were decreased greatly by the treatment. No new chemical groups
styrene (St)/butyl acrylate (BA) (KGM-g-St/BA) and used the graft co- were introduced except for a small amount of carbonyl groups. KGM
polymer for electrospinning. The grafting greatly increased the hydro- was degraded by γ-ray at 50 kGy with or without H2O2 (Pan et al.,
phobicity of the electrospun fiber films of KGM-g-St/BA, which remains 2013). γ-Ray and H2O2 synergistically degraded the KGM as revealed
to be tested for any specific application (Zhu et al., 2015). Overall, the by molecular weight analysis. This may be due to the free radical re-
electrospun nanofibers based on KGM have potential for biomedical actions, resulting in molecular scissions. The treatment also reduced the
applications, which remains to be tested in clinical situation. degree of crystallinity and viscosity of KGM. The polydispersity index of
the resulting KGM reached 1, suggesting a much uniformed molecular
3.2. Microfluidic spinning for wound dressing and micro-reactor for size distribution (Pan et al., 2013). The degraded KGM with much re-
fluorescence reaction duced viscosity may give reduced choking risk as well as altered texture
for formulation healthy food and nutraceutical products.
A KGM and sodium polyacrylate (PAAS) mixture (1:1, w/w) was
made into microfibers through a microfluidic spinning technology 3.5.2. Antioxidant material
(0.1 mL/h and 800 rad/min) (MST) (Mu, Ni, Wang, Yuan, & Yan, 2017) KGM was subjected to γ-irradiation at 100 kGy to obtain the de-
(Supplementary Fig. 2E). The microfibers are consistent in size with a graded products with a good dispersibility (Jian et al., 2017). The ir-
width of 100 μm. They can be easily arranged into microgrids and radiation greatly reduced the number average molecular size of KGM
microarrays for the recognition of amines and targeted reactions. The from 8.3 × 105 (native KGM) to 4.2 × 104 (at 100 kGy), while some-
KGM based microfibers were uniformly loaded with ofloxacin (an an- what increasing the polydispersity from 1.1 to 1.3. The antioxidant
tibiotics) for potential wound dressing applications (Mu et al., 2017). capacity of the degraded KGM against H2O2 induced damage on LO2
cells was studied. The cells pretreated with the degraded KGM product
3.3. Electric field processing for composite gel formation had increased survival rates and activities of GSH-Px (glutathione
peroxidase) and CAT (catalase). The pretreatment also reduced the
KGM (up to 0.5%) and tungsten composite gels were prepared under accumulation of reactive oxygen species in the cells. The in vitro anti-
direct current electric fields (treatments up to 30 V and 15 min) with oxidant capacity of the degraded KGM could be due to the radical
sodium tungstate (up to 0.5%) (Wang, Zhuang, Li, Pang, & Liu, 2016). scavenging effect (Jian et al., 2017). There are diverse health benefits of
Mechanistic studies showed that under acid condition the isopoly- hydrolysed KGM such as enhancing the immune response in human

424
F. Zhu Food Chemistry 256 (2018) 419–426

body and binding nutrients like cholesterol (Tester & Al-Ghazzewi, activity. KGM can be abundantly and sustainably obtained from konjac
2016). Therefore, it may be expected that γ-irradiated KGM also pos- cultivation with high yields. The modified KGM tends to be biode-
sesses these biological activities, which remain to be better studied. The gradable and biocompatible with little impact on the environment. It
degraded KGM product may be used as an ingredient in functional food becomes evident that modified KGM has great potential to be used in
formulations for health promoting. various food and non-food applications.
It is obvious that there are many research opportunities to better
4. Comparative studies are needed develop KGM for diverse applications. KGM from other konjac species
such as A. guripingensis tends to have different functional properties
In some cases, different modifications of KGM are related to the from that of A. konjac. The KGM of other species may also be modified
same type of applications as described in the above 2 sections (Sections to create a wider range of functionalities which may expand the ap-
2 and 3) (Supplementary Tables 1 and 2). A few examples are given plications. Furthermore, the correct konjac species from which KGM is
here. For waste water treatment, modified KGM based on cationization derived should be specified in the report, requiring input from plant
(e.g., as flocculant) (Wang et al., 2017), phthalation (e.g., for Cu(II) biologist. Compared with other well-known and better studied plant
removal) (Zhang et al., 2013), cross-linking [e.g., for removal of dyes polysaccharides such as starch, cellulose, and pectin, KGM is still to be
(methyl blue and methyl orange)] (Gan et al., 2015), graft copolymers modified by a range of different methods. Especially, physical mod-
[e.g., for Pb (II) removal] (Chen et al., 2017), and deacetylated KGM ifications such as ultrasound and pulsed electric field, which appear to
based aerogel (e.g., for arsenite removal) (Ye et al., 2017) were sug- be green and appealing to the public, remain to be more conducted on
gested. Both KGM-oleic acid ester and KGM octenyl succinate showed KGM. A combination of different types of modifications (physical,
good emulsifying capacity (Long et al., 2016; Meng et al., 2014). KGM chemical, and enzymatic) may be used to create a wide range of KGM
graft copolymers (Shahbuddin et al., 2014), deacetylated KGM (Huang functionalities. In some cases, different modifications of KGM led to
et al., 2015), and microfluidic spun KGM (Mu et al., 2017) were sug- similar uses. For example, both cationic and carboxymethyl KGMs can
gested to have great potential for wound dressing and biomedical ap- be ingredients for paper strengthening. Comparative studies should be
plications. KGM acylated with different functional groups (Enomoto- conducted to verify which modified KGM can better achieve the mission
Rogers et al., 2013), acetylated KGM (Enomoto-Rogers et al., 2014), in a holistic view. There is a need to compare KGM based products with
and CKGM-protein composite (Wang et al., 2014) were shown to have those based on other natural polysaccharides such as chitosan and
good film forming capacity. All the studies claimed that the modified starch for the same categories of applications. So that any advantages of
KGM was suitable for some specific applications mentioned above. using KGM based products may be revealed to support commercial
Therefore, comparative studies/analysis are needed to confirm which exploitation. Some of the studies reported the synthesis of the modified
type of modified KGM is best suitable for specific applications. KGM and only suggested some potential applications (e.g., oleic acid
Another consideration is that if the modified-KGM based products ester). The actual application of these modified KGMs should be further
described in this review have any advantage over products based on tested. In some applications of modified KGM such as encapsulation and
other types of natural polysaccharides (e.g., starch). For example, both controlled release, in vivo and clinical studies should be conducted to
KGM sulfate and curdlan sulfate showed anti-HIV activity (Bo et al., verify the usefulness of the reported results. The majority of the reports
2013). Both KGM and chitosan based MIP could be used for the ex- reviewed here are laboratory-based. For commercial applications, the
traction of chemical components (Tian et al., 2017; Wang et al., 2014). performance of the modified KGM should be compared with that of
Therefore, comparative studies are needed to reveal if there is any samples which are commonly used from the current market. For food
advantage of using KGM based products over those of other natural and pharmaceutical/biomedical applications, the safety and toxicity of
polysaccharides and also over the commerical products (e.g., anti-HIV the modified KGM products (e.g., KGM based emulsifiers) should be
drugs). vigorously tested for potential clinical applications and to meet the
Overall, comparison between the resultant modified-KGM (de- regulations and approval from government agencies such as FDA (Food
scribed in this review) and the current/commercial products should be and Drug Administration). Finally, at a more fundamental level, the
conducted to reveal any technological advantage as well as possible structure-functionality/application relationships of modified KGM
disadvantage of KGM based products. Cost estimation, technological should be systematically explored to provide a strong scientific basis to
feasibility, and the impact of the synthetic process as well as the final better develop KGM as a versatile natural polysaccharide.
KGM products on the environment should be taken into consideration.
Clinical studies and pilot-scale trials should be conducted to distinguish Declaration
if the reported studies are mainly due to the pure academic curiosity or
if they have any potential industrial applications. The author declares no conflict of interest.

5. Conclusions and future research directions Appendix A. Supplementary data

KGM has been modified by various chemical and physical methods Supplementary data associated with this article can be found, in the
or the combinations. The chemical methods include substitution-type online version, at http://dx.doi.org/10.1016/j.foodchem.2018.02.151.
modifications (acetylation, carboxymethylation, cationization, cya-
noethylation, esterification with oleic acid, octenyl succination, References
phthalation, and sulfation), chain elongation-type modifications (cross-
link and grafting), and degradation-type modifications (deacetylation Behera, S. S., & Ray, R. C. (2017). Nutritional and potential health benefits of konjac
and oxidation). The physical methods include electrospinning, micro- glucomannan, a promising polysaccharide of elephant foot yam, Amorphophallus
konjac K. Koch: A review. Food Reviews International, 33, 22–43.
fluidic spinning, extrusion, electric field processing, and γ-irradiation. Bo, S., Muschin, T., Kanamoto, T., Nakashima, H., & Yoshida, T. (2013). Sulfation and
The resulting modified KGM has been used in many different applica- biological activities of konjac glucomannan. Carbohydrate Polymers, 94, 899–903.
tions. They include thermoplastic film formation, paper strengthening Chen, P. P., Zhang, H. P., Ding, J., Lin, X. Y., Lu, X., et al. (2017). Carboxylmethyl konjac
glucomannan conjugated polydopamine composites for Pb(II) removal. Carbohydrate
agent, emulsifier, biosorbent for water and pollutants in waste water, Polymers, 162, 62–70.
drug formulation, protein desalting, wound dressing, encapsulation of Chen, J., Zhang, W., & Li, X. (2015). Preparation and characterization of a novel super-
functional ingredient for controlled release, enhanced recovery of fossil absorbent of konjac glucomannanpoly(acrylic acid) with trimethylolpropane tri-
methacrylate cross-linker. RSC Advances, 5, 38417–38423.
oil, molecular imprinting polymer for extraction, fish feed ingredient, Chen, Y., Zhao, H., Liu, X., Li, Z., Liu, B., et al. (2016). TEMPO-oxidized Konjac
micro-reactor for fluorescence reaction, and enhanced antioxidant

425
F. Zhu Food Chemistry 256 (2018) 419–426

glucomannan as appliance for thepreparation of hard capsules. Carbohydrate Shen, C., Li, W., Zhang, L., Wan, C., & Gao, S. (2012). Synthesis of cyanoethyl konjac
Polymers, 143, 262–269. glucomannan and its liquid crystalline behavior in an ionic liquid. Journal of Polymer
Danjo, T., Enomoto-Rogers, Y., Takemura, A., & Iwata, T. (2014). Syntheses and prop- Research, 19, 9758.
erties of glucomannan acetate butyrate mixed esters. Polymer Degradation and Shi, M., Bai, J., Zhao, L., Yu, X., Liang, J., Liu, Y., et al. (2017). Co-loading and intestine-
Stability, 109, 373–378. specific delivery of multiple antioxidants in pH-responsive microspheres based on
Enomoto-Rogers, Y., Ohmomo, Y., & Iwata, T. (2013). Syntheses and characterization of TEMPO-oxidized polysaccharides. Carbohydrate Polymers, 157, 858–865.
konjac glucomannan acetate and their thermal and mechanical properties. Shi, C., Zhu, P., Chen, N., Ye, X., Wang, Y., & Xiao, S. (2016). Preparation and sustainable
Carbohydrate Polymers, 92, 1827–1834. release of modified konjacglucomannan/chitosan nanospheres. International Journal
Enomoto-Rogers, Y., Ohmomo, Y., Takemura, A., & Iwata, T. (2014). Syntheses of glu- of Biological Macromolecules, 91, 609–614.
comannan esters and their thermal andmechanical properties. Carbohydrate Polymers, Tatirat, O., Charoenrein, S., & Kerr, W. L. (2012). Physicochemical properties of extru-
101, 592–599. sion-modified konjac glucomannan. Carbohydrate Polymers, 87, 1545–1551.
Gan, L., Shang, S., Hu, E., Yuen, C. W. M., & Jiang, S. X. (2015). Konjac glucomannan/ Tester, R. F., & Al-Ghazzewi, F. H. (2016). Beneficial health characteristics of native and
graphene oxide hydrogel with enhanced dyesadsorption capability for methyl blue hydrolysed konjac (Amorphophallus konjac) glucomannan. Journal of the Science of
and methyl orange. Applied Surface Science, 357, 866–872. Food and Agriculture, 96, 3283–3291.
Gou, S., Li, S., Feng, M., Zhang, Q., Pan, Q., Wen, J., et al. (2017). Novel biodegradable Tian, D. T., Zhou, Y. C., Xiong, L., & Lu, F. T. (2017). Synthesis and properties of caffeine
graft-modified water-soluble copolymer using acrylamide and konjac glucomannan molecularly imprinted polymers based on konjac glucomannan. Advances in Polymer
for enhanced oil recovery. Industrial & Engineering Chemistry Research, 56, 942–951. Technology, 201, 21575.
Gurusmatika, S., Nishi, K., Harmayani, E., Pranoto, Y., & Sugahara, T. (2017). Wang, M., He, W., Wang, S., & Song, X. (2015). Carboxymethylated glucomannan as
Immunomodulatory activity of octenyl succinic anhydride modified porang paper strengthening agent. Carbohydrate Polymers, 125, 334–339.
(Amorphophallus oncophyllus) glucomannan on mouse macrophage-like J774.1 cells Wang, M., He, W., Yan, R., & Song, X. (2017). A study of cationic glucomannan as a paper
and mouse primary peritoneal macrophages. Molecules, 22, 1187. strength agent. Journal of Macromolecular Science, Part A: Pure and Applied Chemistry,
Ha, W., Wu, H., Wang, X. L., Peng, S. L., Ding, L. S., et al. (2011). Self-aggregates of 54, 216–220.
cholesterol-modified carboxymethyl konjac glucomannan conjugate: Preparation, Wang, L., Jiang, Y., Lin, Y., Pang, J., & Liu, X. Y. (2016). Rheological properties and
characterization, and preliminary assessment as a carrier of etoposide. Carbohydrate formation mechanism of DC electric fields induced konjac glucomannan-tungsten
Polymers, 86, 513–519. gels. Carbohydrate Polymers, 142, 293–299.
Huang, Y. C., Chu, H. W., Huang, C. C., Wu, W. C., & Tsai, J. S. (2015). Alkali-treated Wang, Y., Wang, E., Wu, Z., Li, H., Zhu, Z., Zhu, X., et al. (2014). Synthesis of chitosan
konjac glucomannan film as a novel wound dressing. Carbohydrate Polymers, 117, molecularly imprinted polymers for solid-phase extraction of methandrostenolone.
778–787. Carbohydrate Polymers, 101, 517–523.
Huang, Q., Jin, W., Ye, S., Hu, Y., Wang, Y., et al. (2016). Comparative studies of konjac Wang, L., Xiao, M., Dai, S., Song, J., Ni, X., et al. (2014). Interactions between carbox-
flours extracted from Amorphophallus guripingensis and Amorphophallus rivirei: Based ymethyl konjac glucomannan and soyprotein isolate in blended films. Carbohydrate
on chemical analysis and rheology. Food Hydrocolloids, 57, 209–216. Polymers, 101, 136–145.
Huang, L., Takahashi, R., Kobayashi, S., Kawase, T., & Nishinari, K. (2002). Gelation Wang, L., Zhuang, Y., Li, J., Pang, J., & Liu, X. (2016). The textural properties and mi-
behavior of native and acetylated konjac glucomannan. Biomacromolecules, 3, crostructure of konjac glucomannan–tungsten gels induced by DC electric fields. Food
1296–1303. Chemistry, 212, 256–263.
Jian, W., Tu, L., Wu, L., Xiong, H., Pang, J., & Sun, Y. M. (2017). Physicochemical Wei, X., Pang, J., Zhang, C., Yu, C., Chen, H., & Xie, B. (2015). Structure and properties of
properties and cellular protection against oxidation of degraded Konjac glucomannan moisture-resistant konjac glucomannanfilms coated with shellac/stearic acid coating.
prepared by γ-irradiation. Food Chemistry, 231, 42–50. Carbohydrate Polymers, 118, 119–125.
Kobayashi, S., Tsujihata, S., Hibi, N., & Tsukamoto, Y. (2002). Preparation and rheolo- Xiao, M., Dai, S., Wang, L., Ni, X., Yan, W., et al. (2015). Carboxymethyl modification of
gical characterization of carboxymethyl konjac glucomannan. Food Hydrocolloids, 16, konjac glucomannan affects waterbinding properties. Carbohydrate Polymers,
289–294. 130, 1–8.
Li, J., Ji, J., Xia, J., & Li, B. (2012). Preparation of konjac glucomannan-based super- Xiong, Z., Zhou, W., Sun, L., Li, X., Zhao, D., et al. (2014). Konjac glucomannan micro-
absorbent polymers by frontal polymerization. Carbohydrate Polymers, 87, 757–763. spheres for low-cost desalting of protein solution. Carbohydrate Polymers, 111, 56–62.
Li, Q., Xia, B., Branham, M., Ha, W., Wu, H., et al. (2011). Self-assembly of carboxymethyl Xu, P., Yao, Q., Yu, N., Zhou, Y., Zhao, F., et al. (2014). Narrow-dispersed Konjac glu-
konjac glucomannan-g-poly(ethylene glycol) and (α-cyclodextrin) to biocompatible comannan nanospheres with high moisture adsorption and desorption ability by in-
hollow nanospheres for glucose oxidase encapsulation. Carbohydrate Polymers, 86, verse emulsion crosslinking. Materials Letters, 137, 59–61.
120–126. Yang, F., Zhang, M., Yang, H., Yan, W., & Jiang, F. (2017). Effect of aggregate size on
Li, J., Zhang, X., Chen, J., Xia, J., Ma, M. H., & Li, B. (2009). Frontal polymerization liquid absorption characteristics of konjac glucomannan superabsorbent. Journal of
synthesis and characterization of konjac glucomannan-graft-acrylic acid polymers. Applied Polymer Science, 134, 45416.
Journal of Polymer Science Part A: Polymer Chemistry, 47, 3391–3398. Ye, S., Jin, W., Huang, Q., Hu, Y., Li, Y., et al. (2017). Da-KGM based GO-reinforced
Liu, F., Zou, H., Peng, J., Hu, J., Liu, H., et al. (2016). Removal of copper(II) using FMBO-loaded aerogels for efficient arsenic removal in aqueous solution. International
deacetylated konjac glucomannan conjugated soy protein isolate. International Journal of Biological Macromolecules, 94, 527–534.
Journal of Biological Macromolecules, 86, 338–344. Yoshida, T., Hatanaka, K., Uryu, T., Kaneko, Y., Yasuda, N., Mimura, T., et al. (1990).
Long, X. Y., Pan, J. X., & Yao, L. Q. (2016). Lipase-catalyzed esterification of konjac Synthesis and structural analysis of curdlan sulfate with potent anti-AIDS virus ac-
glucomannan in isooctane. Environmental Progress & Sustainable Energy, 35, tivity. Macromolecules, 23, 3717–3722.
1149–1155. Zhang, C., Chen, J. D., & Yang, F. Q. (2014). Konjac glucomannan, a promising poly-
Luo, X., Liu, F., Deng, Z., & Lin, X. (2011). Removal of copper(II) from aqueous solution in saccharide for OCDDS. Carbohydrate Polymers, 104, 175–181.
fixed-bed column by carboxylic acid functionalized deacetylated konjac gluco- Zhang, C., Han, B., Yao, X., Pang, L., & Luo, X. (2013). Synthesis of konjac glucomannan
mannan. Carbohydrate Polymers, 86, 753–759. phthalate as a new biosorbent for copper ion removal. Journal of Polymer Research,
Meng, F., Zheng, L., Wang, Y., Liang, Y., & Zhong, G. (2014). Preparation and properties 20, 34.
of konjac glucomannan octenyl succinate modified by microwave method. Food Zhang, L., Wu, Y., Wang, L., & Wang, H. (2013). Effects of oxidized konjac glucomannan
Hydrocolloids, 38, 205–210. (OKGM) on growth and immune function of Schizothorax prenanti. Fish & Shellfish
Mu, R. J., Ni, Y., Wang, L., Yuan, Y., Yan, Z., et al. (2017). Fabrication of ordered konjac Immunology, 35, 1105–1110.
glucomannan microfiber arrays via facile microfluidic spinning method. Materials Zhang, L., Wu, Y., Xu, H., & Yao, Y. (2017). Effects of oxidized konjac glucomannan on
Letters, 196, 410–413. the intestinal microbial flora and intestinal morphology of Schizothorax prenanti.
Nie, H., Shen, X., Zhou, Z., Jiang, Q., Chen, Y., et al. (2011). Electrospinning and char- Aquaculture International, 25, 233–250.
acterization of konjac glucomannan/chitosan nanofibrous scaffolds favoring the Zhang, Y. Q., Xie, B. J., & Gan, X. (2005). Advance in the applications of konjac gluco-
growth of bone mesenchymal stem cells. Carbohydrate Polymers, 85, 681–686. mannan and its derivatives. Carbohydrate Polymers, 60, 27–31.
Pan, T., Peng, S., Xu, Z., Xiong, B., Wen, C., Yao, M., et al. (2013). Synergetic degradation Zheng, Q., Wu, Y., & Xu, H. (2015). Effect of dietary oxidized konjac glucomannan on
of konjac glucomannan by γ-ray irradiation and hydrogen peroxide. Carbohydrate Schizothorax prenanti growth performance, body composition, intestinal morphology
Polymers, 93, 761–767. and intestinal microflora. Fish Physiology and Biochemistry, 41, 733–743.
Prawitwong, P., Takigami, S., & Phillips, G. O. (2007). Effects of γ-irradiation on molar Zhong, K., Lin, Z. T., Zheng, X. L., Jiang, G. B., Fang, Y. S., Mao, X. Y., & Liao, Z. W.
mass and properties of Konjac mannan. Food Hydrocolloids, 21, 1362–1367. (2013). Starch derivative-based superabsorbent with integration of water-retaining
Ratcliffe, I., Williams, P. A., English, R. J., & Meadows, J. (2013). Small strain de- and controlled-release fertilizers. Carbohydrate Polymers, 92, 1367–1376.
formation measurements of konjac glucomannan solutions and the influence of bo- Zhu, F. (2016). Structure, properties, and applications of aroid starch. Food Hydrocolloids,
rate cross-linking. Carbohydrate Polymers, 95, 272–281. 52, 378–392.
Ren, W. J., Zhang, A. Q., Qin, S. Y., & Li, Z. K. (2016). Synthesis and evaluation of a novel Zhu, J., Lin, X., Zhang, Z., & Luo, X. (2015). Preparation and characterization of KGM-g-
cationic konjac glucomannan-based flocculant. Carbohydrate Polymers, 144, 238–244. St/BA fibers and core/shell PCL/KGM-g-St/BA fibers. RSC Advances, 5, 24975–24983.
Shahbuddin, M., Bullock, A. J., MacNeil, S., & Rimmer, S. (2014). Glucomannan-poly(N- Zia, F., Zia, K. M., Zuber, M., Ahmad, H. B., & Muneer, M. (2016). Glucomannan based
vinyl pyrrolidinone) bicomponent hydrogels for wound healing. Journal of Materials polyurethanes: A critical short review of recent advances and future perspectives.
Chemistry B, 2, 727–738. International Journal of Biological Macromolecules, 87, 229–236.

426

You might also like