Anda di halaman 1dari 17

Carbohydrate Polymers 151 (2016) 172188

Contents lists available at ScienceDirect

Carbohydrate Polymers
journal homepage: www.elsevier.com/locate/carbpol

Review

A review of chitosan and its derivatives in bone tissue engineering


R. LogithKumar 1 , A. KeshavNarayan 1 , S. Dhivya 1 , A. Chawla 1 , S. Saravanan,
N. Selvamurugan
Department of Biotechnology, School of Bioengineering, SRM University, Kattankulathur, Tamil Nadu, India

a r t i c l e

i n f o

Article history:
Received 4 March 2016
Received in revised form 24 April 2016
Accepted 15 May 2016
Available online 18 May 2016
Keywords:
Biomaterials
Chitosan
Stem cells
Osteoblasts
Bone tissue engineering

a b s t r a c t
Critical-sized bone defects treated with biomaterials offer an efcient alternative to traditional methods involving surgical reconstruction, allografts, and metal implants. Chitosan, a natural biopolymer is
widely studied for bone regeneration applications owing to its tunable chemical and biological properties.
However, the potential of chitosan to repair bone defects is limited due to its water insolubility, faster
in vivo depolymerization, hemo-incompatibility, and weak antimicrobial property. Functionalization of
chitosan structure through various chemical modications provides a solution to these limitations. In
this review, current trends of using chitosan as a composite with other polymers and ceramics, and its
modications such as quaternization, carboxyalkylation, hydroxylation, phosphorylation, sulfation and
copolymerization in bone tissue engineering are elaborated.
2016 Elsevier Ltd. All rights reserved.

Contents
1.
2.

3.

Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 173
Quaternized chitosan . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 174
2.1.
N,N,N-trimethyl chitosan (TMC) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 174
2.2.
N-(2-hydroxyl) propyl-3-trimethylammonium chitosan chloride . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 178
2.3.
Other quaternized chitosans . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 178
Carboxyalkyl chitosan . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 179
3.1.
Carboxymethyl chitosan . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 179
3.2.
Carboxymethyl chitosan derivatives . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 179

Abbreviations: ACP, amorphous calcium phosphate; ALP, alkaline phosphatase; BBC, bioactive bone cement; bFGF, basic broblast growth factor; BTE, bone tissue engineering; BHA, butylated hydroxy anisole; BMP-2, bone morphogenetic protein-2; CDH, calcium-decit hydroxyapatite; CHPTAC, (N-(3-chloro-2-hydroxypropyl) trimethyl
ammonium chloride); CS, chitosan; CMC, carboxymethyl chitosan; COL-I, collagen-I; CPC, calcium phosphate cement; CT, computed tomography; DAH, 1,6-diaminohexane;
DCPA, dicalcium phosphate anhydrous; DCPD, dicalcium phosphate dihydrate; DD, degree of dimethylation; DDD, degenerative disc disease; DMC, dimethyl chitosan;
DQ, degree of quaternization; ECM, extracellular matrix; EDAX, energy dispersive X-ray analysis; GAG, glycosaminoglycans; GEN, genipin; GLU, glutaraldehyde; GP, glycerophosphate; GTMAC, glycidyl trimethyl ammonium chloride; HACC, hydroxypropyltrimethylammonium chloride chitosan; HBC, hydroxybutyl chitosan; HDPs, human
dermal progenitor cells; HEC, hydroxyethylchitosan; HPLCs, human periodontal ligament cells; HTCC, N-(2-hydroxyl) propyl-3-trimethylammonium chitosan chloride;
HPC, hydroxypropyl chitosan; HPLCs, human periodontal ligament cells; HTEC, N-(2-hydroxyl) propyl-3-triethyl ammonium chitosan chloride; HyA, hyaluronic acid;
LBL, layer by layer; MMA, methyl methacrylate; MMT, montmorillonite; MMA, methyl methacrylate; MPCM, monocalcium phosphate monohydrate; MRSA, methicillinresistant Staphylococcus aureus; MSCs, mesenchymal stem cells; MTX, methotrexate; nHAp, nano hydroxyapatite; NBHPDCS, N-(5-bromic-2-hydroxyl-phenyl)-N, N-dimethyl
chitosan; NHNPDCS, N-(2-hydroxyl-5-nitro-phenyl)-N, N-dimethyl chitosan; NHPDCS, N-(2-hydroxyl-phenyl)-N, N-dimethyl chitosan; NMPC, N-methylene phosphonic chitosan; NHS-QPS, (4-(2,5-Dioxo-pyrrolidin-1-yloxycarbonyl)-benzyl)-triphenyl-phosphonium bromide; N, N-DCMCS, N, N-dicarboxymethyl chitosan; NP, nanoparticle; OBs,
osteoblasts; OREC, organic rectorite; PAA, polyacrylic acid; PAMPS, poly(2-acrylamido-2-methylpropanesulfonic acid); P-COS, phosphorylated chito-oligosaccharides; PCPC-1,
monocalcium phosphate monohydrate; PCTS, sodium-phosphorylated chitosan; PEC, polyelectrolyte complex; PEG, polyethylene glycol; PHEMA, polymethylmethacrylateco-polyhydroxyethylmethacrylate; PIA, polysaccharide intracellular adhesin; PMMA, poly methyl methacrylate; PVA, poly vinyl alcohol; PVP, poly vinyl pyrrolidone; TBDMS,
di-tert-butyl dimethylsilyl; TCP, tricalcium phosphate; TMC, N, N, N trimethyl chitosan; TMCMC, tri methyl carboxymethyl chitosan; TNF, tumor necrosis factor; 26SCS, 2-N,
6-O-sulfated chitosan; CT, micro computed tomography.
Corresponding author at: Department of Biotechnology School of Bioengineering SRM University, Kattankulathur 603 203, Tamil Nadu, India.
E-mail addresses: selvamn2@yahoo.com, selvamurugan.n@ktr.srmuniv.ac.in (N. Selvamurugan).
1
These authors equally contributed.
http://dx.doi.org/10.1016/j.carbpol.2016.05.049
0144-8617/ 2016 Elsevier Ltd. All rights reserved.

R. LogithKumar et al. / Carbohydrate Polymers 151 (2016) 172188

4.
5.
6.
7.
8.

173

Hydroxyalkyl chitosan . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 181


Phosphorylated chitosan . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 181
Sulfated chitosan . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 182
Copolymer of chitosan . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 183
Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 184
Acknowledgements . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 185
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 185

1. Introduction
A worldwide increase in the occurrence of pathological fractures
coupled with risk factors during surgical interventions underlines the importance of bone tissue engineering (BTE). Cell-free
constructs and cell-loaded constructs with various chemical and
biological cues act as a template to support bone regeneration
process. They can serve as an alternative treatment method to
conventional grafting procedures. Polymers that can recapitulate natural bone extracellular matrix (ECM) architecture with
the necessary biochemical and load-bearing properties have been
analyzed for in vivo bone regeneration applications (Swetha
et al., 2010; Saranya, Moorthi, Saravanan, Devi, & Selvamurugan,
2011).
Chitosan (CS) is a deacetylated form of chitin procured
mainly from the exoskeleton of crustaceans. It is a linear
polymer composed of randomly distributed units namely: (1 4)2-acetamido-2-deoxy--d-glucan (N-acetyl-d-glucosamine; NAG)
and (1 4)-2-amino-2-deoxy--d-glucan (d-glucosamine) linked
by (1 4) linkages (Jayakumar, Nagahama, Furuike, & Tamura,
2008; Jayakumar, Prabaharan, Nair, Tamura, & 2010b) as shown
in Fig. 1. The degree of deacetylation represents the molar ratio
of the d-glucosamine units to the sum of both NAG and dglucosamine units (Croisier and Jerome, 2013). Depolymerization
of CS can occur by enzymes like glucosaminidases, lipases, and
lysozyme. Chitosan possesses structural resemblance with glycosaminoglycans (GAG), one of the components of ECM that
interacts with collagen bers playing an important role in cellcell adhesion. Chitosan, upon depolymerisation yields bioactive
chito-oligosaccharides with superior anti-microbial properties, and
its monomeric products (glucosamine) metabolized or excreted
from the body. Therefore, CS is biodegradable and has excellent
biocompatibility with almost all the tissues of the body. Chitosan has displayed signicant osteoconductivity, but minimal
osteoinductive property. It induces proliferation of osteoblast cells,
mesenchymal cells and induces in vivo neovascularization (CostaPinto, Reis, & Neves, 2011; Kim et al., 2008; Saravanan, Sameera,
Moorthi, & Selvamurugan, 2013). For orthopedic applications, CS
in various geometries like sponges, bers, lms and other complex
structures are prepared. Thus, CS satises most of the properties supporting its candidature for tissue engineering applications

(Jiang, James, Kumbar, & Laurencin, 2014; Niranjan et al., 2013;


Saranya, Saravanan, Moorthi, Ramyakrishna, & Selvamurugan,
2011).
To enhance the properties of CS for BTE applications, other
polymers and inorganic materials added to CS, and used as 3D
lyophilized scaffolds, hydrogels/lm, and electrospun mats. Chitosan hydrogel membranes with nano-hydroxyapatite (nHAp)
signicantly increased the crystallinity of the composite and
showed excellent biocompatibility with MG-63 cells (Madhumathi
et al., 2009). The addition of nHAp and nano-bioactive glass ceramics (nBGC) to CS/gelatin signicantly improved cell attachment and
proliferation of MG-63 osteoblast-like cells (Peter et al., 2010a,
2010b). The inclusion of nanoscale silicon dioxide and zirconia particles into CS matrix signicantly reduced the degradation rate of
pristine CS scaffolds and enhanced protein adsorption property
(Pattnaik et al., 2011). Protein adsorption and biomineralization are
essential factors that determine the bioactivity of material used in
BTE. Incorporation of carbohydrate anionic moiety like chondroitin
4-sulfate into CS matric increased apatite deposition which facilitated the spreading of bone marrow stromal cells and signicantly
enhanced the compressive modulus (Park et al., 2013). The addition of diopside particles (CaMgSi2 O6 ) to CS matrix up regulated
the expression of osteoblast differentiation marker genes namely
alkaline phosphatase (ALP) and type I collagen (COL-I) and exhibited biocompatibility in vivo in a rat model (Kumar et al., 2014).
The inclusion of bioceramic materials to polymers may enhance
their mechanical property. There was an increase in compressive strength by 33.07% and enhancement in the proliferation of
mouse preosteoblastic cells (MC3T3-E1) upon addition of nHAp to
CS (Zhang, Myers, Wallace, Brandt, & Choong, 2014).
Chitosan as one of the biopolymers in bio-composite scaffolds
has been widely used for BTE applications. The blending of CS with
an anionic polysaccharide alginate stabilized the system by electrostatic interaction of them (Venkatesan, Bhatnagar, Manivasagan,
Kang, & Kim, 2015). Incorporation of nano-silicon dioxide particles to CS-alginate composite displayed a signicant increase in
protein adsorption and apatite deposition, and they were nontoxic to osteoblasts (Sowjanya et al., 2013). Also, incorporation
of nHAp to CS/alginate matrix enhanced the mechanical property of the scaffold as well as stimulated the differentiation of
mouse pre-osteoblastic cells (MC3T3-E1) cells to osteocytes to

Fig. 1. Chemical Structures of chitin and chitosan. Reprinted with permission from Younes and Rinaudo (2015). Chitin and chitosan preparation from marine sources.
Structure, properties and applications. Marine drugs, 13(3), 11331174. 2015, MDPI.

174

R. LogithKumar et al. / Carbohydrate Polymers 151 (2016) 172188

Cs/alginate scaffold (Kim et al., 2015). Cold atmospheric plasma


treated CS/HAp scaffolds displayed surface roughness and wettability, which selectively enhanced the protein adsorption of
bronectin and vitronectin. This modication resulted in greater
mesenchymal stem cells (MSCs) inltration into the scaffolds with
increased collagen deposition and mineralization by the end of 3
weeks (Wang, Cheng et al., 2013; Wang, Xie et al., 2013). The inclusion of chicken feather keratin nanoparticles with CS signicantly
improved the protein adsorption, and they were biocompatible
with human osteoblastic cells (Saravanan et al., 2013). Doping and
addition of antibacterial metal ions such as nanophase copper,
zinc and silver to CS polymer signicantly improved the antimicrobial property (Niranjan et al., 2013; Saravanan et al., 2011;
Tripathi et al., 2012). Chitosan/hyaluronic acid scaffolds upon addition of calcium phosphate cement exhibited a signicant increase
in ALP activity with no signicant change in the rate of osteoblastic cell proliferation (Hesaraki and Nezafati, 2014). The presence
of fucoidan in CS/alginate scaffold stimulated the proliferation of
MG-63 osteoblast-like cells with signicant enhancement in ALP
activity and apatite deposition over the scaffolds (Venkatesan,
Bhatnagar, & Kim, 2014). The addition of nHAp to CS/gelatin matrix
not only increased the mechanical property of the scaffolds but also
stimulated the proliferation and differentiation of induced pluripotent stem cells of gingival broblasts to osteocytes (Isikli, Hasirci, &
Hasirci, 2012; Ji et al., 2015, 2016). Fucoidan in TCP-CS scaffolds
increased the compressibility, apatite deposition, and osteocalcin
release, which favor osteogenic differentiation of human mesenchymal stromal cells in vitro (Puvaneswary et al., 2015).
Chitosan and its bio-composite scaffolds have been widely
tested under in vivo conditions. Chitosan/gelatin scaffolds promoted osteoblast proliferation in vivo, their degradation occurred
completely in 12 weeks, and thus, these scaffolds have the potential for bone healing process (Ma et al., 2014). The encapsulation
of MC3T3-E1 osteoblast-like cells in -tricalcium phosphateCS-alginate microcapsules and their injection in the dorsal
subcutaneous area of mice showed collagen deposition and neovascularization at the site of injection. Also, the characteristic
osteoid-like structures were seen at the end of 8 weeks (He, Dong,
Chen, & Lin, 2013; Qiao et al., 2013). An osteoinductive bone graft
prepared by combining calcium phosphate with CS and its implantation in the rats calvarial defects resulted in a signicantly higher
matrix deposition rate as well as osteon formation. Surprisingly,
implantation with the scaffolds replaced the entire calvarial defect
with host bone tissue. An injectable thermosensitive hydrogel containing CS along with Zn, nHAp and -glycerolphosphate (-GP)
showed its potential towards bone formation in vivo as evidenced
by increased apatite and collagen deposition in the defective bone
area (Dhivya, Saravanan, Sastry & Selvamurugan, 2015).
Despite the numerous applications of CS as a stand-alone polymer or as a composite with other polymers and ceramic particles,
its use is limited. Firstly, CS solubility in aqueous solutions is pH
dependent. Since CS has a pKa of 6.5 and its semi-crystalline nature
favors strong intra/inter molecular hydrogen bonding, the solubility of CS at neutral pH is limited. Secondly, CS is quickly and
easily degraded in vivo due to the presence of (1 4) glycosidic linkages, which are hydrolytically cleaved by the abundance
of lysozyme (approximately, 1315 mg/L) present in the body (Lim
et al., 2008). Hence, the study focused on CS with its side chain
modications to address their solubility and controlled biodegradation (Feng and Xia, 2011; Lafeur et al., 2013). Additionally, the
polycationic nature of CS can induce thrombosis, red blood cell
aggregation and hemolysis making it unsuitable for tissue engineering applications. Therefore, many studies focused on altering
the surface charge on the CS chain to enhance its hemocompatibility (Balan and Verestiuc, 2014; Yalinca, Yilmaz, Taneri, Bullici,
& Tuzmen, 2013). Scaffolds with a suitable antimicrobial prop-

erty were required to prevent the implant-associated infections


(Hedrick, Adams, & Sawyer, 2006; Qiu, Zhang, An, & Wen, 2007).
However, CS has little antimicrobial property at neutral pH due to
the protonation of amino groups that occurs only in acidic medium.
Finally, CS has low bioactivity due to the lack of charged and reactive
groups in its structure, which otherwise are necessary to facilitate protein adsorption. Thus, immobilizing scaffolds with proteins
like bronectin was essential for increasing the bioactivity of CS
based scaffolds (Custodio, Alves, Reis, & Mano, 2010). Table 1 summarizes several modications at C2 or C5 positions of CS by an
introduction of various reactive groups or by copolymerization
with other polymers of interest or by grafting with biological and
synthetic macromolecules, which render appropriate bone regenerative properties.
2. Quaternized chitosan
The modication of CS side chain yields to a variety of derivatives
for biomedical applications. Chitosan was modied into N,N,Ntrimethyl chitosan (TMC) by reaction with methyl iodide catalyzed
by sodium hydroxide at 60 C into N-methyl-2-pyrrolidinone. In a
second step, the substitution of iodide ions by chloride ion by an ion
exchange process yielded TMC (Fig. 2). In another example, N-(2hydroxyl) propyl-3-trimethylammonium chitosan chloride (HTCC)
was prepared by dissolving CS in NaOH/CHPTAC aqueous solution by a pressure-equalizing dropping funnel (Fan et al., 2015).
Other modied CSs were such as N-(2-hydroxyl) propyl-3-triethyl
ammonium chitosan chloride (HTEC) and N-(2-hydroxyl-phenyl)N,N-dimethyl chitosan (NHPDCS) (Benediktsdottir, Gudjonsson,
Baldursson, & Masson, 20114; de Britto, Celi Goy, Campana Filho,
& Assis, 2011; Guo et al., 2007; Yang et al., 2015). Chitosan exerts
antimicrobial activity by disrupting the negatively charged outer
membrane of the microbes, and the modications of CS structure
would further enhance its antimicrobial activity that is one of the
features required in BTE.
2.1. N,N,N-trimethyl chitosan (TMC)
Positive charge rich trimethyl chitosan (TMC) displayed
enhanced antibacterial activity compared to CS against both E. coli
and S. aureus (Jia and Xu, 2001). The ratio of DD (degree of
dimethylation) to DQ (degree of quaternization) seems to control
mucoadhesive, cytotoxic and other physicochemical properties of
TMC (Haas, Ravi Kumar, Borchard, Bakowsk, & Lehr, 2005). The
anionic groups of lipopolysaccharide in Gram-negative and lipoteichoic acids in Gram-positive bacteria act as a molecular linkage for
quaternized and trimethylated amino groups in TMC (Raafat, Von
Bargen, Haas, & Sahl, 2008). The electrostatic interaction between
these groups can disrupt the function and integrity of the bacterial membrane. The antibacterial property of TMC increased with
increase in pH of the medium (Xu, Xin, Li, Huang, & Zhou, 2010). It is
important to note that chitosan chemistry inuences the shielding
effect of surface ion pairs resulting in either partial O-methylation
or free of O-methylation. Removal of these surface ion pairs by
dialysis showed an increase in the mobility of alkyl chains, conferring an active interaction at the surface of the bacterial envelope
resulting in accelerated the antibacterial activity. In addition, TMC
without O-methylation reduced the cytotoxicity and increased its
solubility at physiological pH. Di-tert-butyl dimethylsilyl (TBDMS)
is a commonly used protectant of OH group during methylation,
which is highly stable under the reaction conditions while the
removal is also a facile process (Benediktsdottir et al., 2011; Martins
et al., 2015).
In addition to the antibacterial property of TMC, TMC could also
used as a polycation in developing polyelectrolyte complexes by

R. LogithKumar et al. / Carbohydrate Polymers 151 (2016) 172188

175

Table 1
Chemically modied derivatives of Chitosan and the improved properties offered by their reactive groups.
Derivatives

Subtypes

Quaternized Chitosan

N,N,N-Trimethyl chitosan (TMC)

High absorption efciency (Britto & Assis, 2007)


Mucoadhesion (Pardeshi and Belgamwar, 2016; Raafat,
Von Bargen, Haas, & Sahl, 2008)

N-(2-hydroxyl)
propyl-3-trimethylammonium
chitosan chloride (HTCC or HACC)

Better moisture retention and absorption compared to


chitosan (Huang et al., 2014)
Inhibited the expression of intracellular adhesion genes
which can mediate biolm formation on implants (Peng
et al., 2011)
High transfection efciency when compared to Chitosan
(Xiao et al., 2012)

N-(2-hydroxyl-substituted
phenyl)-N,N-dimethyl chitosan
(NXRPDCS)

Strong antifungal activities against many resistant


strains (Guo et al., 2007)

N-Carboxy-methyl Chitosan (N-CMC)

High metal chelation ability-bound strongly to Ca2+ ions


and promoted apatite formation (Mourya and Inamdar,
2008; Mourya et al., 2010)

O-Carboxymethyl Chitosan (O-CMC)

Promoted adhesion and proliferation in human


broblast cells (Wongpanit et al., 2005)

N,O-Carboxymethyl chitosan
(N,O-CMC)

High water retentivity and excellent gel forming


capacity (Jayakumar, Prabaharan, Nair, & Tamura, 2010)
Exhibited negligible immunogenicity (Shalumon et al.,
2009).

Hydroxyethyl chitosan

High moisture absorption rate and highly hydrophilic

Hydroxypropyl chitosan

99% inhibition against most of the bacteria (Peng, Han,


Liu, & Xu, 2005)

Hydroxybutyl chitosan

Rapid gelation kinetics and displayed good in vivo


stability (Dang et al., 2006)

N-Acetyl chitosan

Due to hydrophobic pockets it caused lesser lysozyme


absorption and higher bronectin adsorption
(Tangpasuthadol, Pongchaisirikul, & Hoven, 2003)
Lower swelling index maintains its mechanical strength
(Le Tien, Lacroix, Ispas-Szabo, & Mateescu, 2003)

Carboxyalkyl Chitosan

Hydroxyalkyl
Chitosan

N-Acyl Chitosan

Reactive group

Important properties

176

R. LogithKumar et al. / Carbohydrate Polymers 151 (2016) 172188

Table 1 (Continued)
Derivatives

Thiolated Chitosan

Subtypes

Reactive group

Important properties

N-Carboxyacyl chitosan

Formed stable hydrogels owing to its amphoteric nature


(Hirano & Moriyasu, 2004)

Chitosan cysteine conjugate

Rapid formation of hydrogel promotes cell attachment


and inltration (Bernkop-Schnurch, Hornof, & Zoidl,
2004; Riva et al., 2011)
Gel interacted with BMP-2 causing no change in its
swelling behavior (Kurniawan Fudholi, & Susidarti,
2012)

Chitosan2-iminothiolane conjugate
(or Chitosan-4-Thio-butylamidine)

Highly porous structure for cell inltration (Vijapur


Sreenivas, Patil, Vijapur, & Patwari, 2012)
High solubility over wide range of pH (Masuko et al.,
2005)

Chitosan-Thioglycolic acid

Excellent in situ gelling capacity: Formed gels rapidly at


body temperature (Sakloetsakun, Hombach, &
Bernkop-Schnurch, 2009)

Attributing to its negative charge, low interfacial energy


and high binding capacity to Ca2 +, and it acted as a
strong nucleator for high biomineralization (Xu, Neoh,
Lin, & Kishen, 2011)
Enhanced adipose derived stem cell growth in vitro (Yeh
and Lin, 2012)

Phosphorylated Chitosan

Sulfated Chitosan

2-N, 6-O-sulfated chitosan (26SCS)

Structural analogy to heparin, made it highly


hemo-compatible (Shen et al., 2011)

Semi-synthetic resins
of Chitosan

Copolymer of chitosan with methyl


methacrylate

Good mechanical and compressive strength (>10 MPa)


(Endogan et al., 2014)
Uniformly arranged nanoporous structure which caused
sustained time-release of drugs and proteins (Shen et al.,
2011)

Copolymer of Chitosan with


Acrylamide

Thermo-responsive gels with 3-D porous architecture,


good interconnectivity, elasticity and displayed
appreciable osteoconductivity (Liao, Chen, & Chen, 2011)

Glycol Chitosan

High water solubility at wide range of pH (Chung et al.,


2003)
Faster biodegradability for early bone healing
application (Park, Cho, Chung, Kwon, & Jeong, 2003)

Glucosamine Chitosan

Better water solubility with enhanced antimicrobial


properties (Chen & Zhao, 2012)

Sugar derivatives of
Chitosan

R. LogithKumar et al. / Carbohydrate Polymers 151 (2016) 172188

177

Table 1 (Continued)
Derivatives

Subtypes

Specic Ligand conjugated Chitosan

Arginylglycylaspartic acid
(RGD),TyrIleGlySer- Arg(YIGSR),
le-Lys-Val-Ala-Val(IKVAV),
A99a(ALRGDN) and others

Reactive group

Important properties
Acceleration of integrin mediated osteoblast
attachment, proliferation and differentiation (Tsai, Chen,
& Liu, 2013; Wang et al., 2014)

Formed viscoelastic injectable hydrogels with


biostability and high metal adsorption capacity (Wang,
2006)

Glycol Chitosan

Glutaraldehyde Chitosan

found biocompatible and biostable complex which


induced vascularization and tissue formation (Ma et al.,
2003; Wu, Black, Santacana Laftte, & Patrick, 2007)

Chitosan-ascorbate conjugate

Excellent antioxidant property with high scavenging


and chelation abilities (Tian Tian, Wang, & Mo, 2009)

Cyanoethyl Chitosan

Enhanced bactericidal nature

Imidazole Chitosan

Inhibited Ca2 + release from bone (Di Martino, Sittinger,


& Risbud, 2005)
Slightly osteoinductive (Muzzarelli et al., 1994)

Fig. 2. Quaternization of chitosan. Chitosan was modied into N,N,N-trimethyl chitosan. Reprinted with permission from, Mourya and Inamdar (2008). Chitosan-modications
and applications: opportunities galore. Reactive and Functional polymers, 68(6), 10131051. Elsevier, 2008.

layer by layer (LBL) assembley, a surface modication strategy benecial over crosslinking, in preventing the effects of altered charge
density, reactivity, and degradability of the material. Almodovar
and Kipper (2011) developed an LBL assembley using TMC as a
polycation, heparin as a polyanion. This PEC-LBL assembley showed
to promote FGF-2 adsorption serving as suitable matrices for the
delivery of growth factors for cell and tissue engineering. A yet
another study on the coating of cortical bone with TMC-heparin
polyelectrolyte multilayers showed that they can serve as periosteum mimics, deliver osteoprogenitor cells, and improve bone
allograft healing (Romero et al., 2015). Thus, while the application
of TMC much explored regarding its antibacterial properties, efforts
are needed in considering the polycationic nature of TMC to extend
further its applications in tissue engineering as vehicles for growth
factor delivery.
O-carboxymethyl-N,N,N-trimethyl-chitosan (CMTMC) scaffolds
had no negative inuence when treated with human dermal

progenitor (HDP) cells (Patrulea et al., 2015). Further, by immobilization of 10 nmol cm2 and ve nmol cm2 RGDC peptide on the
surface of 1,6-diaminohexane (DAH)-CMTMC, there was the successful attachment of HDP cells as well as the promotion of cell proliferation compared to DAH-CMTMC scaffolds without the RGDC
peptide (Patrulea et al., 2016). Novel Electrospun silver nanoparticles (Ag Nps) loaded nanobrous mats containing TMC, polyacrylic
acid (PAA) or poly(2-acrylamido-2-methylpropanesulfonic acid)
(PAMPS) showed superior antibacterial activity against E. coli and
S. aureus than the membranes without TMC and Ag NPs (Kalinov
et al., 2015). Polyalanine treatment to quaternized CS improved
its biocompatibility and antibacterial activities (Zhao, Li, Guo, &
Ma, 2015). The antibacterial nature and the wound healing properties of TMC bers, prepared by the methylation of CS bers
using iodomethane under alkaline conditions has been recently
assessed by Zhou et al. (2016). The resulting TMC bers with varying degrees of quaternization indicated their higher antibacterial

178

R. LogithKumar et al. / Carbohydrate Polymers 151 (2016) 172188

Fig. 3. Effects of CS and TMC bers in rat would model in vivo over 12-day treatment period. Micrographs displaying the wounds at day 0, 3, 6, 9, 12. The modied CS bers
(TMC) had enhanced wound healing property. Reprinted with permission from, Zhou et al. (2016). Biomaterials based on N, N, N-Trimethyl chitosan bers in wound dressing
applications. International journal of biological macromolecules. Elsevier, 2016.

activity against E. coli and S. aureus compared to CS bers. The TMC


bers also exhibited enhanced swelling properties attributed to the
positive surplus charges in its chain. These modied CS bers tested
in vivo in rat full-thickness excision models displayed reduced
wound maceration, wound re-epithelialization, eschar development over 12 days compared to the unmodied CS bers (Fig. 3).

2.2. N-(2-hydroxyl) propyl-3-trimethylammonium chitosan


chloride
N-(2-hydroxyl) propyl-3-trimethylammonium chitosan chloride (HTCC) showed stronger antibacterial activity under alkaline
condition (Qin et al., 2004). It had enhanced antibacterial potency
compared to CS against Methicillin-resistant Staphylococcus aureus
and Staphylococcus epidermidis Porphyromonas gingivalis (P. gingivalis ATCC33277), Prevotella intermedia (P. intermedia ATCC 25611),
Actinobacillus actinomycetemcomitans (A. actinomycetemcomitans
Y4) and Streptococcus mutans (S. mutans Ingbritt C) (Martinez
et al., 2010; Peng et al., 2010). In addition to antibacterial activity,
the role of HTCC in tissue regeneration was studied. Hydroxypropyltrimethylammonium chloride chitosan accelerated the
proliferation of human periodontal ligament cells (HPDLCs) even at
the lower concentration as compared to pristine CS (Qiu Xia et al.,
2010). The CS-HTCC/GP aqueous solution injected intramuscularly
into the rumps of spraguedawley rats formed a gel-like plug at
the injection site and showed a good amount of collagen deposition at the end of 9th week. Chitosan-HTCC/glycerophosphate (GP)
thermosensitive hydrogel loaded with basic broblast growth factor (CS-HTCC/GPbFGF) effectively enhanced the new periodontal
support tissues in dogs. The regenerative tissues seemed complete to ll the fraction areas, and a signicant amount of new
bone formation observed (Croisier and Jerome, 2013). Quaternized
CS derivatives like O-(2-hydroxyl) propyl-3-trimethyl ammonium
chitosan chloride (O-HTCC) and HTEC have also displayed superior
antioxidant properties exhibiting dose-dependent reducing power
and lipid peroxidation inhibition effect (Cui, Tang, & Yin, 2012;
Wan, A., Xu, Sun, & Li, 2013). The addition of CMC to HTCC improved
its mechanical property (Hu, Wang, & Wang 2016). Their results
indicated the formation of strong intermolecular hydrogen bond
between OH group of HTCC and CMC, which lead to the increased
rigidity and enhanced the tensile strength and elongation at break
values.

2.3. Other quaternized chitosans


The quaternized CS (QCS) displayed a signicant role in inhibition of the expression of icaA gene (that is involved in secretion
of polysaccharide intracellular adhesin) in bacteria on titanium
implants in a biolm prevention assay (Peng et al., 2011). Use
of antibiotics such as gentamicin loaded in PMMA (Poly (methyl
methacrylate)) bone cement was standard practice for preventing
infections in joint arthroplasty and osteomyelitis. This practice had
not only given rise to antibiotic resistant bacteria like methicillinresistant Staphylococcus aureus (MRSA) but also shown to reduce
proliferation of hMSCs and the viability of osteoblast cells (Parker,
Clegg, & Taylor, 2012). However, hydroxypropyl trimethylammonium chloride chitosan (HACC) with 26% degree of substitution
loaded in PMMA inhibited biolm formation (Tan, Guo, Yang, Xu,
& Tang, 2012). It also resulted in better apatite deposition, cell
attachment and differentiation of hMSCs towards osteoblast lineage. Expression levels of COL-I and ALP were signicantly higher
compared to both gentamicin loaded PPMA and CSPMMA (Tan
et al., 2012b). The quaternized CS derivatives with increasing
degree of substitution (DS) showed antifungal potency in vitro
on the mycelial growth of Aspergillus avus. Increasing the DS of
CS exhibited an almost six-fold increase in the antifungal activity at only one-fourth concentration of the deacetylated chitosan
polymer. Quaternized CSsincluding N-(2-hydroxyl-phenyl)-N,
N-dimethyl CS (NHPDCS), N-(5-chloro-2-hydroxyl-phenyl)-N, Ndimethyl CS (NCHPDCS), N-(2-hydroxyl-5-nitro-phenyl)-N, Ndimethyl CS (NHNPDCS) and N-(5-bromic-2-hydroxyl-phenyl)N,N-dimethyl CS (NBHPDCS), N,N,N-(diethylcinnamyl) CS (QC1)
and N,N,N-(diethyl-p-dimethylaminobenzyl) CS (QC3) showed better antifungal activities compared to the unmodied CS (Badawy
and Rabea, 2014; Guo et al., 2007).
An injectable in situ forming an electroactive hydrogel, containing QCS grafted polyaniline with oxidized dextran (o-dex) as crosslinker showed excellent antimicrobial properties in vitro and in vivo
(Zhao, Li, Guo, & Ma, 2015). The design and synthesis of a series of
novel quaternized derivatives of CS, dihydroxy quaternary ammonium salts (QAS) containing long chain alkyl bromides reported.
The results indicated that the increase in the alkyl chain length
increased the antibacterial activity (Wang et al., 2016). Additionally, a reactive antibacterial compound (4-(2,5-dioxo-pyrrolidin1-yloxycarbonyl)-benzyl)-triphenyl-phosphonium bromide (NHSQPS) showed effective antimicrobial activity towards E. coli and S.

R. LogithKumar et al. / Carbohydrate Polymers 151 (2016) 172188

aureus compared to CS and also the compound was solubilized in


water between a wide range of pH 312 (Zhu et al., 2016).
Layer by layer self-assembling polymers into composite lms
is a technique to improve the antibacterial efciency of implant
materials. Organic rectorite (OREC) is a layered silicate material
well known for their superior mechanical, anti-infective properties owing to their larger interlayer distance. Self-assembled
QCMC/OREC/alginate nanobrous mats layered onto cellulose
nanobers displayed enhanced antibacterial activity against E. coli
and S. aureus compared to cellulose lms (Jiang et al., 2015). As cellulose based composites are mechanically strong matrices, the LBL
assembly of QCMC/OREC/Alg onto cellulose mats could act as cytocompatible, mechanically strong antiinfective materials for tissue
engineering applications. Recently, a novel method of modication
to QCS was adopted by reacting CS with (3-chloro-2-hydroxypropyl
trimethyl ammonium chloride) in the presence of alkali or urea
resulting in the highest degree of substitution (You et al., 2016). The
prepared QCS was used to develop a hydrogel by in situ polymerization with PAA. There existed a strong electrostatic interaction
between the quaternary ammonium group of CS and the carboxyl
group of PAA resulting in enhanced toughness, self-recovery/shape
memory properties with tunable mechanical properties. An electrostatic LBL lm assembly comprising of heparin (HP), CS and
its derivatives namely DMC, TMC was aimed to develop an efcient anti-inammatory coating to medical devices and implants
(Follmann et al., 2016). Because inammatory responses are universal host defense mechanisms against foreign agents, which on
the other hand hinders the success of many medical implants.
3. Carboxyalkyl chitosan
Amongst other derivatives of CS, carboxyalkyl CS was widely
studied due to its ease of synthesis, a high degree of hydrophilicity and numerous potential applications (Riva et al., 2011).
Carboxyalkylation of CS yields carboxyethyl, carboxybutyl, carboxymethyl CS and other derivatives. The carboxy ( COOH) groups
impart anionic charges on the CS chain, making it amphoteric with
a wide range of biomedical applications. Carboxyalkyl CSs were
water soluble at a wide range of pH, biodegradable, biocompatible,
and were non-toxic to human tissues. Grafting succinic acid to carboxyalkyl CS improved water solubility and transfection efciency
(Ahmed and Ikram, 2015).
3.1. Carboxymethyl chitosan
The most commonly used CS derivative was carboxymethyl
chitosan (CMC). CMC is amphiprotic ether derived from CS. The
alkylation of CS with monohalocarboxylic acid at different reaction
conditions conferred the selectivity form of N- or O-carboxymethyl
derivatives (Fei Liu, Lin Guan, Zhi Yang, Li, & De Yao, 2001).
Carboxymethylation introduced carboxymethyl ( COOCH3 ) group
in the primary or secondary hydroxyl groups and the amino
groups bonded to the glucopyranose unit (Riva et al., 2011).
Amphoteric nature, enhanced aqueous solubility, biocompatibility,
biodegradability and minimal immunogenicity of CMC supported
its candidature in BTE applications (Ibrahim, El-Zairy, & Mosaad,
2015; Shalumon et al., 2009). By reductive alkylation and direct
alkylation, carboxymethylation of CS prepared. However, the former one was not preferred as it utilized expensive chemicals,
produces toxic by-products like NaCN and HCN, and would be difcult to scale-up to industrial level (Mourya, Inamdar, & Tiwari,
2010; Shalumon et al., 2009). The degree of substitution of CS was
directly proportional to the concentration of NaOH used in the
direct alkylation process, but it was independent of the degree of
deacetylation of CS. The optimum concentration of NaOH was found

179

to be%; higher concentrations could lead to the depolymerization


of the CS chains (Chen, Tian, & Du, 2004).
There are reports available showing the potential role played
by CMC along with other polymers and biomaterials in BTE. Carboxymethyl chitosan/HAp scaffolds showed better bone formation
than HAp alone with negligible cytotoxicity on MC3T3-E1 cells
and high blood adsorbing capacity (Tokura and Tamura, 2001).
The layering of CMC on titanium implants prevented implantassociated infection and subsequent implant failure. This strategy
highly reduced S. aureus and S. epidermidis adhesion on the implants
(Shi, Neoh, Kang, Poh, & Wang, 2009). Carboxymethyl chitosan/PVA
(poly vinyl alcohol) electrospun scaffold supported hMSC attachment and proliferation with no apparent cytotoxicity (Shalumon
et al., 2009). Injectable gels of CMCgelatinnHAp stimulated proliferation of osteoblasts and showed no evidence of inammation
at the site of implantation (Mishra et al., 2011). Carboxymethyl
chitosan/gelatin/-TCP composites fabricated via ultrasonic radiation mediated crosslinking were porous in structure with optimum
mechanical strength. These scaffolds showed biocompatible and
promoted bone regeneration when implanted in the mandibular
region of beagle dogs. Furthermore, the micro-CT analysis clearly
indicated new bone formation with a volumetric density (BV/TV%)
of 13.3% (Zhou et al., 2012). MTT assay showed that CMC (800 mg/L)
with a combination of platelet-derived growth factor-BB (10 g/L)
could promote proliferation as well as differentiation of HPDLCs (Ji
et al., 2010).
3.2. Carboxymethyl chitosan derivatives
Derivatives of CMC can be amino substituted (N-CMC), hydroxyl
substituted (O-CMC) or both amino and hydroxyl substituted (N, OCMC). Fig. 4 represents a schematic representation of the chemical
synthesis of these derivatives. These derivatives were cytocompatible, biodegradable and exerted superior antibacterial properties.
Further, N-CMC supported chondrocyte adhesion and possesses
metal ion chelating ability (Mourya et al., 2010). O-CMC stimulated
broblast proliferation (Fei Liu et al., 2001) while N, O-CMC had
unique gel forming capacity with excellent water retentive properties (Jayakumar, Nagahama et al., 2010; Jayakumar, Selvamurugan,
2010; Jayakumar, Prabaharan et al., 2010). N-CMC possessed selective metal adsorption capacity and it readily bound to Ca2+ ions and
promoted biomineralization at the site of implantation (Kyzas and
Bikiaris, 2015). O-CMC-BMP-2 modied titanium surfaces proved
the great extent of biomineralization of both osteoblast cells and
hMSCs along with enhancement of osseointegration and longevity
of the titanium implants (Shi, Neoh, Kang, Poh, & Wang, 2009).
N, O-CMC/n--TCP (tricalcium phosphate) composites in a mass
proportion of 1:1 highly promoted apatite formation on its surface when subjected to varied chemical conditions of simulated
body uids (Liu et al., 2013). Lee and Kamarul (2014) prepared a
copolymer scaffold comprising of PVA/N-O-CMC/PEG, physically
cross-linked by 50 KGy e-beam radiation. The strong crosslinking
mediated by the e-beam radiation enhanced the physical properties
of the scaffold such as porosity, pore size, and conferred superior
swelling properties. The scaffolds also showed increased chondrocyte proliferation on its surface with high expression of GAG and
collagen type II (Lee and Kamarul, 2014).
O-Carboxymethyl chitosan-based injectable hydrogel containing alginate (Alg), nano brin particles displayed higher resistance
to mechanical compression. The Alg/O-CMC/nano brin (AOF) composite promoted the differentiation of adipose derived stem cells
to adipocytes for adipose tissue engineering (Jaikumar et al., 2015).
PVA/CMC/montmorillonite (MMT) based hydrogel nanocomposite system displayed enhanced antimicrobial activity better than
the standard drug penicillin G (Sabaa, Abdallah, Mohamed, &
Mohamed, 2015). Li et al. (2015) developed a CMC/modied starch

180

R. LogithKumar et al. / Carbohydrate Polymers 151 (2016) 172188

Fig. 4. Carboxyalkylation of Chitosan. Chitosan was modied into N-carboxymethyl chitosan, O-carboxymethyl chitosan and N, O-carboxymethyl chitosan. Reprinted with
permission from, Mourya and Inamdar (2008). Chitosan-modications and applications: opportunities galore. Reactive and Functional polymers, 68(6), 10131051. Elsevier,
2008.

based in situ injectable hydrogel with a superior mechanical property. Oxidized cholesterol starch as a non-cytotoxic crosslinker
used in the hydrogel synthesis mediated stable covalent bond formation, by the condensation of aldehyde groups of oxidized starch
with the primary amine group of CMC. Besides being an antibacterial material, upon optimizing the crosslinker concentration, the
structural and mechanical properties of the hydrogel could be
tailor made to specic applications. Hao, Chen, Yu, Liu, and Sun
(2016) recently developed the CMC-multiwalled carbon nanotube
based strong antibacterial composite upon coordination of metal
ions Cu and Zn. They displayed enhanced antimicrobial activity
against S. aureus, E. coli and V. anguillarum. In another application,
CMC grafted polyacrylic acid served as superabsorbent polymers

in the case of serious hemorrhages, and these polymers promoted


the concentration of coagulation factors, RBCs and platelets. The
enhanced roughness, porous surface structure, high water retention ability and availability of positively charged NH3 + cations of
the polymers promoted the adhesion of negatively charged RBCs
onto the surface of the wound thereby facilitated hemostasis (Chen,
Huang et al., 2016; Chen, Zhang et al., 2016).
N, O-CMC nanoparticles displayed better antibacterial property
against S. aureus than both O-CMC and CS nanoparticles, and the
inhibition of bacterial growth seemed to increase with an increase
in the concentration of these nanoparticles (Anitha et al., 2009).
N, O-CMCzinc compound exerted a better antimicrobial activity than CSzinc complex against S. aureus (Patale and Patravale,

R. LogithKumar et al. / Carbohydrate Polymers 151 (2016) 172188

2011). Carboxymethyl chitosan displayed an inhibition efciency


of >90% against bacterial adhesion. It prevented biolm formation
at the efciency of 63.1% of Gram-positive bacteria and 70.6% of
Gram-negative bacteria after 1 of biolm initiation. The proposed
mechanism behind this phenomenon is the neutralization of the
bacterial surface charge leading to occulation of the bacterial cell
population (Tan, Han, Ma, & Yu, 2011). Kaya et al. (2014) demonstrated the antimicrobial and antioxidant activities possessed by
O-CMC. Water soluble N, O-CMC developed by Patrulea, Applegate,
Ostafe, Jordan, and Borchard (2015) showed its non-cytotoxic and
non-immunogenic nature, which are the features also required in
BTE.
4. Hydroxyalkyl chitosan
Hydroxyalkyl chitosans can be obtained by the substitution
reaction of CS with epoxides at either amino or hydroxyl groups
giving N-hydroxyalkyl or O-hydroxyalkyl CS derivatives, respectively (Fig. 5). The ratio of O/N-substitution was determined by
the choice of catalyst (NaOH or HCl) used in the reaction and the
reaction temperature (Donges, Reichel, & Kessler, 2000; Mourya
and Inamdar, 2008). For tissue engineering applications, only a few
derivatives of hydroxyalkyl CS such as hydroxybutyl, hydroxyethyl
and hydroxypropyl chitosan have been explored. Hydroxypropyl
chitosan (HPC) at a concentration of, grafted with maleic acid,
owing to its capability to form coordination bonds displayed better
inhibitory effects up to 99.9% against both S. aureus and E.coli within
30 min of contact time (Peng, Han, Liu, & Xu, 2005). Hydroxybutyl
chitosan (HBC) provided an extra advantage of a rapid gelation
kinetics and better in vivo stability over that of collagen and therefore, established itself as a suitable polymer (Dang et al., 2006).
Hydroxybutyl chitosan was shown to be biocompatible and was
minimally toxic to ve-wt% when exposed to hMSCs and intervertebral disk cells as a potential candidate to treat degenerative disc
disease (DDD). Its thermosensitive properties underlined its probable use to form gels at body temperature in as quick as 30. The
cell viability in the presence of HBC was similar to that in the presence of collagen in a 2-weeks treatment period. It promoted the
proliferation of L929 mouse broblast cells with minimal toxicity suggesting its application in wound healing (Wei et al., 2009).
Cellulose/O-hydroxyethyl chitosan bers with an HEC concentration of 6.2% displayed reduction of E. coli growth and had moisture
absorption rate of 13.55% (Xu, Xin et al., 2010; Xu, Zhuang et al.,
2010).
Clay nanocomposites emerged to be superior materials in
improving the mechanical properties of polymeric materials
with strength, modulus, and dimensional stability. A modied
HTCC-nanoclay (montmorillonite) composite showed antibacterial
activity, cytocompatibility, and cell growth (Aliabadi, Dastjerdi, &
Kabiri, 2013). The inclusion of nanoclay to HTCC resulted in strongly
intercalated network structure, which allowed the bacteria to effectively entrapped, and damaged them. However, further studies are
warranted to investigate and support the applications of hydroxyalkyl CS in BTE.
5. Phosphorylated chitosan
The synthesis of phosphorylated CS (P-CS) involves the simultaneous reaction of the phosphorous acid and formaldehyde to CS
in the acidic aqueous acidic resulting in the formation of watersoluble N-mono- and di-phosphonicmethylene chitosan (Fig. 6).
Phosphorylated CS displays better ionic conductivity and swelling
index. Although it decreased crystallinity, its tensile strength
remained similar to CS. Phosphorylated CS exhibited considerably rough surface morphology unlike CS (Jayakumar, Nagahama,

181

Furuike, & Tamura, 2008; Jayakumar, Selvamurugan, Nair, Tokura,


& Tamura, 2008). Phosphorous pentoxide (P2 O5 ) reaction with CS
yielded water-soluble phosphorylated CS with a high degree of substitution (Tachaboonyakiat, Netswasdi, Srakaew, & Opaprakasit,
2010). Phosphorylated CS blend alginate lms showed induction
of apatite layer after immersing it in simulated body uid (SBF)
for 21 days (Jayakumar et al., 2009). The solubility of CS based
hydrogels above pH 6 reduced its potential for tissue engineering applications. To overcome this limitation, the phosphorylated
derivative of CS, N-methylene phosphonic chitosan (NMPC) was
synthesized by cross-linking of NMPC with either glutaraldehyde
(NMPC-GLU) or genipin (NMPC-GEN). While NMPC-GEN hydrogel was better among the two, both the hydrogels showed higher
elastic modulus, better compressive strengths and increased the
proliferation of cells.
A biodegradable P-CS/monetite (dicalcium phosphate
anhydrous-DCPA) composite showed enhanced mechanical
performance compared to DCPA or unphosphorylated CS-DCPA
(Boroujeni, Zhou, Luchini, & Bhaduri, 2014). The addition of 5 w%
P-CS powders to DCPA decreased the setting time as well as
increased the youngs modulus, compressive strength by two-fold
(Fig. 7a,b). However, it was observed that further increase in
w% of P-CS from 5% to 10% reversed the increasing mechanical
properties. The composite showed cytocompatibility, supported
the proliferation of MC3T3-E1 cells, and hence suggested as ideal
bone cement for load bearing applications.
Calcium phosphate cement has been used as llers at the site
of injury for its remarkable osteoconductive and osseointegrative
properties (Winge, Reikeras, & Rokkum, 2011), yet they lack the
mechanical strength required for bone tissue implants. The binding afnity of calcium to reactive functional groups followed the
order: phosphorylate > carboxylate > amino > hydroxyl groups. The
addition of P-CS up to 2 wt% to monocalcium phosphate monohydrate (PCPC-1) and calcium oxide cement, and up to 10 wt%
to dicalcium phosphate dihydrate and calcium hydroxide cement
(PCPC-2) increased their compressive strength and youngs modulus. The leaching experiment showed that P-CS did not leach out
for up to two months when implanted in vivo. Hence, P-CS could
bind tightly to inorganic phase during the setting process of CPC
(calcium phosphate cement), thus, can be effective as bone ller in
clinical applications.
Enzymatic degradation of CS formed chito-oligosaccharides
(Aam et al., 2010) and phosphorylated chito-oligosaccharides (PCOS) up to 100 g/mL concentration showed no cytotoxicity to
MG-63 osteoblast-like cells. Also, low molecular weight P-COS
seemed to promote cell proliferation. ALP activity was signicantly
increased in the presence of P-COS as compared to CS (Venkatesan,
Pangestuti, Qian, Ryu, & Kim, 2010). To achieve the mechanical
strength, a P-CS/CS/n-HAp composite was prepared by the coprecipitation method and it displayed a compressive strength of
70.25 Mpa. The compressive strength of the composite remained
64% of the original (45.07 Mpa) after soaking in simulated body
uid (SBF) for . Hence, it displayed controlled biodegradation and
excellent bioactivity for BTE purposes (Li, Huang, Wang, Ma, & Xie,
2011). The incorporation of P-CS powders at ve-wt% to the widely
used bone cement DCPA reduced its setting time and doubled its
compressive strength. Both DCPA and DCPAP-CS increased cell
proliferation (MC3T3-E1). The results showed that both the materials were as cytocompatible as HAp. The cell morphology of attached
osteoblast cells in 7 days culture testied the biocompatibility of
DCPAP-CS with up to 5-wt% of P-CS (Boroujeni et al., 2014).
The lack of negatively charged functionalities on the surface of
CS limits the osteoblast cell attachment. Hence, its modications
with plasma induced phosphonic groups signicantly improved
osteoblast attachment, viability, and proliferation (Lopez-Perez, Da
Silva, Sousa, Pashkuleva, & Reis, 2010). It has been reported that P-

182

R. LogithKumar et al. / Carbohydrate Polymers 151 (2016) 172188

Fig. 5. Hydroxyalkylation of chitosan. Reprinted with permission from, Mourya and Inamdar (2008). Chitosan-modications and applications: opportunities galore. Reactive
and Functional polymers, 68(6), 10131051. Elsevier, 2008.

CS was completely blood compatible, had lesser platelet reactivity


unlike CS, and enhanced adipose-derived stem cell growth in vitro
(Yeh and Lin, 2012). Metals like Zn have been widely used for their
antimicrobial properties in complex with biomaterials (Thian et al.,
2013). However, metals can cause signicant cytotoxicity if used
above a threshold concentration. Sodium-phosphorylated chitosan
(PCTS) and ZnO showed their application in periodontal dressings.
The PCTS did not change the crystalline nature of ZnO and reduced
its cytotoxicity by forming a complex with its protonated hydroxyl
groups. The composite was biocompatible with primary human
gingival broblast cells (Srakaew, Ruangsri, Suthin, Thunyakitpisal,
& Tachaboonyakiat, 2011). Phosphorylated CS signicantly promoted proliferation of both primary human osteoblasts (OBs) and
the OB like stromal cell (the component of the giant cell tumor
of bone cells; up to 1000-g ml1 concentration) only after seven
days of treatment. A study on the osteogenic effect of the P-CS suggested that it regulates the level of osteoclastogenic factors, and
receptor activator of nuclear factor kappa B ligand (NFKBL) and
osteoprotegerin (OPG) expression (Tang et al., 2011). Partially demineralized dentine sections modied by covalent immobilization of
P-CS showed better deposition of calcium phosphate on the dentine
surface (Xu, Neoh, Lin, & Kishen, 2011). All these properties ensure
P-CSs candidature as a promising biomaterial for BTE applications.

6. Sulfated chitosan
Chitosan chain can be sulfated using sulphuric acid or sulphonic
acid salts (Rajasree and Rahate, 2013; Zhang, Zhang, Ma, Yang, &
Nie, 2015). The sulfated CS played a major in bone metabolism
and enhanced bone morphogenetic protein-2 (BMP-2) activity in
bone (Takada et al., 2003). The half-life of BMP-2 in culture was
prolonged with heparin, a sulfated polysaccharide (Hosseinkhani,
Khademhosseini, & Kobayashi, 2007; Zhao et al., 2006). The addition of sulfated CS to calcium-decit hydroxyapatite (CDH) loaded

with BMP-2 increased in release prole of BMP-2 as compared


to the composite without sulfated CS. The initial burst following gradual release was essential for the growth of new bone
tissue (Zhao et al., 2011). Sulfated CS stimulated proliferation of
both primary human osteoblasts and the OB like stromal cell
component of the giant cell tumor (GCT) of bone cells at a concentration of 100 g ml1 . However, it inhibited cell proliferation
above 1000 g ml1 ; this effect was more pronounced in primary
human GCT stromal cells as compared to osteoblast cells (Tang et al.,
2011).
Neovascularization at the site of a bone defect is necessary for
the development of a new brous tissue. Modied heparin displayed vascular tissue formation as reported previously (Janairo
et al., 2012; Wang, Cheng et al., 2013; Wang, Xie et al., 2013). Sulfated N-carboxymethyl CS inhibited thrombin and factor Xa similar
to antithrombin. It also caused no hemolysis when human blood
samples treated with it (Zhou et al., 2013). In another study, 2N, 6-O-sulfated chitosan (26SCS) based nanoparticle (S-NP) was
successfully developed and loaded with BMP-2 (BMP-2/S-NP) and
then fabricated with gelatin BMP-2/S-NP (BMP-2/S-NP/G) to repair
critical-sized bone defect of rabbit radius. The gelatin BMP-2/S-NP
treatment signicantly increased both peripheral and new vessel
formation. Therefore, vascularisation contributed by BMP-2/S-NP
in the critical defect site and controlled release of BMP-2 led to
increased bone augmentation (Cao et al., 2014). The photopolymerizable hydrogel incorporated with rhBMP-2 and 2-N, 6-O-sulfated
CS nanoparticles displayed excellent cytocompatibility, cell attachment and cell in the growth of human MSCs. In vitro results
indicated higher ALP activity and mineralization in the presence of
2-N, 6-O-sulfated CS nanoparticles. Bone formation was conrmed
in rat thigh defect and rabbit radius defect models (Cao et al., 2014;
Peschel, Zhang, Fischer, & Groth, 2012). These results indicated that
sulfated CS can have potential towards BTE purposes.

R. LogithKumar et al. / Carbohydrate Polymers 151 (2016) 172188

183

Fig. 6. Synthesis of Phosphorylated Chitosans. Reprinted with permission from, Mourya and Inamdar (2008). Chitosan-modications and applications: opportunities galore.
Reactive and Functional polymers, 68(6), 10131051. Elsevier, 2008.

Fig. 7. Effect of p-CS loaded DCPA cements on the mechanical properties. a) Compressive strength of DCPA, DCPAp-chitosan (5 wt%), DCPAp-chitosan (10 wt%) and
DCPAuntreated chitosan (5 wt%). b) Youngs modulus of DCPA, DCPAp-chitosan (5 wt%), DCPAp-chitosan (10 wt%), and DCPAuntreated chitosan (5 wt%). Reprinted with
permission from Boroujeni et al. (2014). Development of monetite/phosphorylated chitosan composite bone cement.Journal of Biomedical Materials Research Part B: Applied
Biomaterials,102(2), 260266. 2013 Wiley Periodicals, Inc.

7. Copolymer of chitosan
The grafted copolymers improve the physicochemical properties of synthetic or natural polymers. Poly(methyl methacrylate)

(PMMA) is one of the most commonly used bone cement owing


to its ability to form a strong mechanical bond with the implant.
This acrylic resin is currently the widely used material for the construction of removable denture and is the basis of dental composite

184

R. LogithKumar et al. / Carbohydrate Polymers 151 (2016) 172188

Table 2
Superior properties of chitosan derivatives/chemically modied chitosan based composites favouring bone tissue engineering.
Derivatives/Modications
of Chitosan

Bioactive
Cells treated
molecules/Crosslinkers/proteins

Properties

References

Carboxymethyl
chitosan

Ulvan

Immortalized mouse lung


broblasts cell line (L929)

Barros et al., 2013

Carboxymethyl
Chitosan

genipin carbodiimide local


recombinant human
BMP-2
Zein Silica

Human osteoblastic cell


line (SaOS-2)

Increased Ca and P absorption in


the bulk, enchanced mechanical
performance.
Cytocompatible, promoted cell
attachment and proliferation.

Reves, Bumgardne, &


Haggard (2013)

hMSCs

Enhanced antibacterial activity.

Zhou et al. (2014)

BMP-2 photopolymerisable
hydrogels

hMSCs

Sustained growth factor delivery


and enhanced bioactivity.

Cao et al. (2014)

Chitosan Hydroxyapatite

Good compressive strength.

alkaline phosphatase (ALP)


polydopamine Ti
polyethylene glycol
dimethacrylate (PEGDA)
N,N-dimethylacrylamide
(DMMA)
Hydroxyapatite

hMSCs

Enhanced cellular ALP activity and


calcium deposition by osteoblasts.
Improved mechanical behaviour,
thermal stability. Promoted cell
attachment and proliferation.

Li, Huang, Wang, Ma & Xie


(2011)
Zheng, Neoh & Kang (2016)

hydroxypropyltrimethyl
ammonium chloride
chitosan(HACC)
2-N, 6-O-sulfated
chitosan
nanoparticles
4-phosphorylated
chitosan
carboxymethyl
chitosan
Methacryloyloxy ethyl
carboxyethyl
chitosan
chitosanpolylactic
acid
chitosan-graftpolycaprolactone
(CHS-g-PCL)
N-methylene
phosphonic chitosan
(NMPC)

SW1353

Greatly inuenced nucleation and


growth of crystalline HA.
Enhanced cell mineralization and
ALP activity

Silicatein

SaOS-2

Genipin

Human osteoblastic cell


line (MG-63)

Thiolated chitosan

beta-glycerophosphate
hydroxyapatite

hMSCs

Glycol chitosan (G-CS)

Hyaluronic acid/nHAp

MC-3T3-E1

HACC

Alginate oyster shell


powder

MC-3T3-E1, MG-63 cells

lling materials (Nayak, 2010). It is better than other materials


regarding aesthetics, easy manipulation, low cost, and its ability to
distribute the implant load at the site of implantation. However,
PMMA does not adhere so well to the surrounding bone tissue,
and it is not suitable for the high load bearing sites (Dorozhkin,
2011). The addition of hydroxypropyltrimethylammonium chloride chitosan (HACC) to PMMA showed signicantly higher apatite
deposition, ALP activity and promoted attachment and proliferation of hMSCs. These cells on PMMA-HACC displayed signicantly
higher expression of COL-1, osteopontin (OPN) and osteocalcin
(OC) by the end of 21 days as compared to PMMA-CS, PMMAgentamicin, and PMMA (Tan, Guo et al., 2012). In another study,
the addition of PMMA-co-PHEMA (polymethylmethacrylateco-polyhydroxyethylmethacrylate) to CS/HAp blend showed a
signicant increase in Youngs modulus and stiffness, and enhanced
water absorption capacity of the scaffolds (Bhowmick, Banerjee,
Kumar, & Kundu, 2013). PMMA-HACC promoted a stronger bond
with the surrounding bone, as compared to others in rabbit femoral
condyle (Tan, Ao, Ma, & Tang, 2013). The CS-PMMA composite
increased its compressive strength and promoted new osteoid formation compared to PMMA alone (Endogan et al., 2014). The effects
of using CMC as an additive to methotrexate (MTX) loaded PMMA
bone cement signicantly increased the bending modulus, bending strength and compressive strength of the PMMA composite
(Liu et al., 2015). In comparison to the MTX-PMMA group, CMCMTX-PMMA improved the osseointegration with the host tissue
proving to be an ideal composite over MTX-PMMA as an antitumour bone cement. CS-g-PVCL is a grafted copolymer where CS
grafted with PVCL polymers of varying chain lengths through ami-

Better cell adhesion, cell-cell


interaction, proliferation. Increased
ALP activity and calcium
deposition.
Porous structure with a uniform
distribution of nHAp with
appropriate degradation rate and
low cytotoxicity.
Faster enzymatic degradation of
the scaffold within 4 weeks.
Increased compressive strength,
protein absorption &
biomineralization.

Ma et al. (2010)

Cai et al. (2009)


Wiens, Elkhooly,
Schroder,Mohamed and
Muller (2014)
Datta, Dhara and
Chatterjee (2012)

Liu et al. (2014)

Huang et al., 2016 Huang,


Zhang, Wu and Xu (2016)
Chen, Huang et al. (2016);
Chen, Zhang et al. (2016)

dation reaction by activation of the terminal carboxylic group. The


copolymers were water-soluble at low temperature and were thermosensitive. The thermal behavior was dependent on the grafted
chain length. For future drug delivery purposes, BTE and regenerative medicine, these thermosensitive characteristics can be useful
(Fernandez-Quiroz et al., 2015)
The nanocomposites of CS-graft-(methyl methacrylate) (CS-gMMA) containing Ag nanoparticles used to avoid the implantassociated infections, and the result showed excellent antimicrobial properties (up to 9398%) with increased mechanical strength.
Even though PMMA loaded with antibiotics were used in dermal
llers, and joint arthroplasties, the release of antibiotics was quick
and hence the possibility of altering the mechanical and fatigue
strength of PMMA (Zhang, Myers et al., 2014). The quaternized
CS (HACC) loaded with PMMA in the cavity displayed signicantly
better antibacterial property against the resistant bacteria as compared to PMMA, PMMA-gentamicin or PMMA-CS (Tan, Ao, Ma, Lin,
& Tang, 2014). The treatment of CS with carbon disulde under
alkaline condition formed dithiocarbamate CS. Dithiocarbamate CS
with a metal ion complex showed a prolonged antibacterial activity (Kim et al., 2014). To understand better about CS derivatives
and their properties, we listed the various modications of chitosan
along with bioactive molecules employed in bone tissue engineering (Table 2).
8. Conclusion
This review summarized CS composites and chemically
modied CS for their application in BTE. Modied-CS based scaf-

R. LogithKumar et al. / Carbohydrate Polymers 151 (2016) 172188

folds/hydrogels displayed superior physical, chemical, mechanical


and biological properties unlike its counterpart serving to be excellent vehicles for accelerating bone regeneration. Also, this review
consolidated that the empirical renement of CS has candidly
thrown open new avenues for the treatment of bone defects. Thus,
an effort took here provided insights into the past and current
trends of using modied CS polymer. Integration and processing of
the differential properties offered by various modied CS composites would be further benecial for treating bone and bone related
defects.
Acknowledgements
We thank Anbuselvan Thambidurai and Pallavi Chatterjee for
their technical help in the manuscript preparation. We also thank
the Council of Scientic and Industrial Research (CSIR), India (Grant
No. 60(0110)/13/EMR-II to N.S.) and the SRM University for nancial support.
References
Aam, B. B., Heggset, E. B., Norberg, A. L., Sorlie, M., Varum, K. M., & Eijsink, V. G.
(2010). Production of chitooligosaccharides and their potential applications in
medicine. Marine Drugs, 8(5), 14821517.
Ahmed, S., & Ikram, S. (2015). Chitosan & its derivatives: a review in recent
innovations. International Journal of Pharmaceutical Sciences and Research, 6(1),
1427.
Aliabadi, M., Dastjerdi, R., & Kabiri, K. (2013). HTCC-Modied nanoclay for tissue
engineering applications: a synergistic cell growth and antibacterial efciency.
BioMed Research International, 2013, 17.
Almodovar, J., & Kipper, M. J. (2011). Coating electrospun chitosan nanobers with
polyelectrolyte multilayers using the polysaccharides heparin and N, N,
N-trimethyl chitosan. Macromolecular Bioscience, 11(1), 7276.
Anitha, A., Rani, V. D., Krishna, R., Sreeja, V., Selvamurugan, N., Nair, S. V., et al.
(2009). Synthesis, characterization, cytotoxicity and antibacterial studies of
chitosan, O-carboxymethyl and N, O-carboxymethyl chitosan nanoparticles.
Carbohydrate Polymers, 78(4), 672677.
Badawy, M. E., & Rabea, E. I. (2014). Synthesis and antifungal property of N-(aryl)
and quaternary N-(aryl) chitosan derivatives against Botrytis cinerea. Cellulose,
21(4), 31213137.
Balan, V., & Verestiuc, L. (2014). Strategies to improve chitosan hemocompatibility:
a review. European Polymer Journal, 53, 171188.
Barros, A. A. A., Alves, A., Nunes, C., Coimbra, M. A., Pires, R. A., & Reis, R. L. (2013).
Carboxymethylation of ulvan and chitosan and their use as polymeric
components of bone cements. Acta Biomaterialia, 9(11), 90869097.
Benediktsdottir, B. E., Gaware, V. S., Runarsson, O. V., Jonsdottir, S., Jensen, K. J., &
Masson, M. (2011). Synthesis of N, N, N-trimethyl chitosan homopolymer and
highly substituted N-alkyl-N, N-dimethyl chitosan derivatives with the aid of
di-tert-butyldimethylsilyl chitosan. Carbohydrate Polymers, 86(4), 14511460.
Benediktsdottir, B. E., Gudjonsson, T., Baldursson, O., & Masson, M. (2014).
N-alkylation of highly quaternized chitosan derivatives affects the paracellular
permeation enhancement in bronchial epithelia in vitro. European Journal of
Pharmaceutics and Biopharmaceutics, 86(1), 5563.
Bhowmick, A., Banerjee, S., Kumar, R., & Kundu, P. P. (2013).
Hydroxyapatite-packed chitosan-PMMA nanocomposite: a promising material
for construction of synthetic bone. In multifaceted development and
application of biopolymers for biology. Biomedicine and Nanotechnology, 254,
135167.
Boroujeni, N. M., Zhou, H., Luchini, T. J., & Bhaduri, S. B. (2014). Development of
monetite/phosphorylated chitosan composite bone cement. Journal of
Biomedical Materials Research Part B: Applied Biomaterials, 102(2), 260266.
Cai, X., Tong, H., Shen, X., Chen, W., Yan, J., & Hu, J. (2009). Preparation and
characterization of homogeneous chitosanpolylactic acid/hydroxyapatite
nanocomposite for bone tissue engineering and evaluation of its mechanical
properties. Acta Biomaterialia, 5(7), 26932703.
Cao, L., Werkmeister, J. A., Wang, J., Glattauer, V., McLean, K. M., & Liu, C. (2014).
Bone regeneration using photocrosslinked hydrogel incorporating rhBMP-2
loaded 2-N, 6-O-sulfated chitosan nanoparticles. Biomaterials, 35(9),
27302742.
Chen, L., Tian, Z., & Du, Y. (2004). Synthesis and pH sensitivity of carboxymethyl
chitosan-based polyampholyte hydrogels for protein carrier matrices.
Biomaterials, 25(17), 37253732.
Chen, T. Y., Huang, H. C., Cao, J. L., Xin, Y. J., Luo, W. F., & Ao, N. J. (2016). Preparation
and characterization of alginate/HACC/oyster shell powder biocomposite
scaffolds for potential bone tissue engineering applications. RSC Advances,
6(42), 3557735588.
Chen, Y., Zhang, Y., Wang, F., Meng, W., Yang, X., Li, P., et al. (2016). Preparation of
porous carboxymethyl chitosan grafted poly (acrylic acid) superabsorbent by
solvent precipitation and its application as a hemostatic wound dressing.
Materials Science and Engineering: C, 63, 1829.

185

Chung, T. W., Lu, Y. F., Wang, H. Y., Chen, W. P., Wang, S. S., Lin, Y. S., et al. (2003).
Growth of human endothelial cells on different concentrations of
Gly-Arg-Gly-Asp grafted chitosan surface. Articial Organs, 27(2), 155161.
Costa-Pinto, A. R., Reis, R. L., & Neves, N. M. (2011). Scaffolds based bone tissue
engineering: the role of chitosan. Tissue Engineering Part B: Reviews, 17(5),
331347.
Croisier, F., & Jerome, C. (2013). Chitosan-based biomaterials for tissue
engineering. European Polymer Journal, 49(4), 780792.
Cui, L., Tang, C., & Yin, C. (2012). Effects of quaternization and PEGylation on the
biocompatibility, enzymatic degradability and antioxidant activity of chitosan
derivatives. Carbohydrate Polymers, 87(4), 25052511.
Custodio, C. A., Alves, C. M., Reis, R. L., & Mano, J. F. (2010). Immobilization of
bronectin in chitosan substrates improves cell adhesion and proliferation.
Journal of Tissue Engineering and Regenerative Medicine, 4(4), 316323.
Dang, J. M., Sun, D. D., Shin-Ya, Y., Sieber, A. N., Kostuik, J. P., & Leong, K. W. (2006).
Temperature-responsive hydroxybutyl chitosan for the culture of
mesenchymal stem cells and intervertebral disk cells. Biomaterials, 27(3),
406418.
Datta, P., Dhara, S., & Chatterjee, J. (2012). Hydrogels and electrospun nanobrous
scaffolds of N-methylene phosphonic chitosan as bioinspired osteoconductive
materials for bone grafting. Carbohydrate Polymers, 87(2), 13541362.
De Britto, D., Celi Goy, R., Campana Filho, S. P., & Assis, O. B. (2011). Quaternary
salts of chitosan: history, antimicrobial features: and prospects. International
Journal of Carbohydrate Chemistry, 2011, 112.
Dhivya, S., Saravanan, S., Sastry, T. P., & Selvamurugan, N. (2015).
Nanohydroxyapatite-reinforced chitosan composite hydrogel for bone tissue
repair in vitro and in vivo. Journal of Nanobiotechnology, 13, 40.
Di Martino, A., Sittinger, M., & Risbud, M. V. (2005). Chitosan: a versatile
biopolymer for orthopaedic tissue-engineering. Biomaterials, 26(30),
59835990.
Donges R., Reichel D., & Kessler B. (2000) U.S. Patent No. 6, 090, 928. Washington,
DC: U.S. Patent and Trademark Ofce.
Dorozhkin, S. V. (2011). Biocomposites and hybrid biomaterials based on calcium
orthophosphates. Biomatter, 1(1), 356.
Endogan, T., Kiziltay, A., Kose, G. T., Comunoglu, N., Beyzadeoglu, T., & Hasirci, N.
(2014). Acrylic bone cements: effects of the poly (methyl methacrylate)
powder size and chitosan addition on their properties. Journal of Applied
Polymer Science, 131(3).
Fan, L., Yang, J., Wu, H., Hu, Z., Yi, J., Tong, J., et al. (2015). Preparation and
characterization of quaternary ammonium chitosan hydrogel with signicant
antibacterial activity. International Journal of Biological Macromolecules, 79,
830836.
Fei Liu, X., Lin Guan, Y., Zhi Yang, D., Li, Z., & De Yao, K. (2001). Antibacterial action
of chitosan and carboxymethylated chitosan. Journal of Applied Polymer Science,
79(7), 13241335.
Feng, Y., & Xia, W. (2011). Preparation, characterization and antibacterial activity
of water-soluble O-fumaryl-chitosan. Carbohydrate Polymers, 83(3),
11691173.
Fernandez-Quiroz, D., Gonzalez-Gomez, A., Lizardi-Mendoza, J., Vazquez-Lasa, B.,
Goycoolea, F. M., San Roman, J., et al. (2015). Effect of the molecular
architecture on the thermosensitive properties of chitosan-g-poly
(N-vinylcaprolactam). Carbohydrate Polymers, 134, 92101.
Follmann, H. D., Naves, A. F., Martins, A. F., Felix, O., Decher, G., Muniz, E. C., et al.
(2016). Advanced broblast proliferation inhibition for biocompatible coating
by electrostatic layer-by-layer assemblies of heparin and chitosan derivatives.
Journal of Colloid and Interface Science, 474, 917.
Guo, Z., Xing, R., Liu, S., Zhong, Z., Ji, X., Wang, L., et al. (2007). The inuence of the
cationic of quaternized chitosan on antifungal activity. International Journal of
Food Microbiology, 118(2), 214217.
Haas, J., Ravi Kumar, M. N., Borchard, G., Bakowsky, U., & Lehr, C. M. (2005).
Preparation and characterization of chitosan and trimethyl-chitosan-modied
poly-(epsilon-caprolactone) nanoparticles as DNA carriers. A.A.P.S.
PharmSciTech, 6(1), 2230.
Hao, X., Chen, S., Yu, H., Liu, D., & Sun, W. (2016). Metal ion-coordinated
carboxymethylated chitosan grafted carbon nanotubes with enhanced
antibacterial properties. RSC Advances, 6(1), 3943.
He, Y., Dong, Y., Chen, X., & Lin, R. (2013). Ectopic osteogenesis and scaffold
biodegradation of tissue engineering bone composed of chitosan and
osteo-induced bone marrow mesenchymal stem cells in vivo. Chinese Medical
Journal, 127(2), 322328.
Hedrick, T. L., Adams, J. D., & Sawyer, R. G. (2006). Implant-associated infections:
an overview. Journal of Long-term Effects of Medical Implants, 16(1), 8399.
Hesaraki, S., & Nezafati, N. (2014). In vitro biocompatibility of chitosan/hyaluronic
acid-containing calcium phosphate bone cements. Bioprocess and Biosystems
Engineering, 37(8), 15071516.
Hosseinkhani, H., Hosseinkhani, M., Khademhosseini, A., & Kobayashi, H. (2007).
Bone regeneration through controlled release of bone morphogenetic
protein-2 from 3-D tissue engineered nano-scaffold. Journal of Controlled
Release, 117(3), 380386.
Hu, D., Wang, H., & Wang, L. (2016). Physical properties and antibacterial activity
of quaternized chitosan/carboxymethyl cellulose blend lms. LWT-Food Science
and Technology, 65, 398405.
Huang, J., Cheng, Z. H., Xie, H. H., Gong, J. Y., Lou, J., Ge, Q., et al. (2014). Effect of
quaternization degree on physiochemical and biological activities of chitosan
from squid pens. International Journal of Biological Macromolecules, 70,
545550.

186

R. LogithKumar et al. / Carbohydrate Polymers 151 (2016) 172188

Huang, Y., Zhang, X., Wu, A., & Xu, H. V. (2016). An injectable nano-hydroxyapatite
(n-HA)/glycol chitosan (G-CS)/hyaluronic acid (HyA) composite hydrogel for
bone tissue engineering. RSC Advances, 6(40), 3352933536.
Ibrahim, H. M., El-Zairy, E. M. R., & Mosaad, R. M. V. (2015). Preparation,
characterization and median lethal dose (LD50) of carboxymethyl chitosan as
target Drug Delivery. International Journal of Advanced Research, 3(1), 865873.
Isikli, C., Hasirci, V., & Hasirci, N. (2012). Development of porous
chitosangelatin/hydroxyapatite composite scaffolds for hard
tissue-engineering applications. Journal of Tissue Engineering and Regenerative
Medicine, 6(2), 135143.
Jaikumar, D., Sajesh, K. M., Soumya, S., Nimal, T. R., Chennazhi, K. P., Nair, S. V., et al.
(2015). Injectable alginate-O-carboxymethyl chitosan/nano brin composite
hydrogels for adipose tissue engineering. International Journal of Biological
Macromolecules, 74, 318326.
Janairo, R. R. R., Henry, J. J., Lee, B. L. P., Hashi, C. K., Derugin, N., Lee, R., et al. (2012).
Heparin-modied small-diameter nanobrous vascular grafts NanoBioscience.
IEEE Transactions on, 11(1), 2227.
Jayakumar, R., Nagahama, H., Furuike, T., & Tamura, H. (2008). Synthesis of
phosphorylated chitosan by novel method and its characterization.
International Journal of Biological Macromolecules, 42(4), 335339.
Jayakumar, R., Rajkumar, M., Freitas, H., Selvamurugan, N., Nair, S. V., Furuike, T.,
et al. (2009). Preparation, characterization, bioactive and metal uptake studies
of alginate/phosphorylated chitin blend lms. International Journal of Biological
Macromolecules, 44(1), 107111.
Jayakumar, R., Chennazhi, K. P., Muzzarelli, R. A. A., Tamura, H., Nair, S. V., &
Selvamurugan, N. V. (2010). Chitosan conjugated DNA nanoparticles in gene
therapy. Carbohydrate Polymers, 79(1), 18.
Jayakumar, R., Prabaharan, M., Nair, S. V., & Tamura, H. (2010). Novel chitin and
chitosan nanobers in biomedical applications. Biotechnology Advances, 28(1),
142150.
Jayakumar, R., Prabaharan, M., Nair, S. V., Tokura, S., Tamura, H., & Selvamurugan,
N. (2010). Novel carboxymethyl derivatives of chitin and chitosan materials
and their biomedical applications. Progress in Material Science, 55(7), 675709.
Ji, Q. X., Deng, J., Xing, X. M., Yuan, C. Q., Yu, X. B., Xu, Q. C., et al. (2010).
Biocompatibility of a chitosan-based injectable thermosensitive hydrogel and
its effects on dog periodontal tissue regeneration. Carbohydrate Polymers,
82(4), 11531160.
Ji, J., Tong, X., Huang, X., Wang, T., Lin, Z., Cao, Y., et al. (2015). Sphere-shaped
nano-hydroxyapatite/chitosan/gelatin 3D porous scaffolds increase
proliferation and osteogenic differentiation of human induced pluripotent
stem cells from gingival broblasts. Biomedical Material, 10(4), 045005.
Ji, J., Tong, X., Huang, X., Zhang, J., Qin, H., & Hu, Q. (2016). Patient-derived human
induced pluripotent stem cells from gingival broblasts composited with
dened nanohydroxyapatite/chitosan/gelatin porous scaffolds as potential
bone graft substitutes. Stem Cells Translational Medecine, 5(1), 95105.
Jia, Z., & Xu, W. (2001). Synthesis and antibacterial activities of quaternary
ammonium salt of chitosan. Carbohydrate Research, 333(1), 16.
Jiang, T., James, R., Kumbar, S. G., & Laurencin, C. T. (2014). Chitosan as a
biomaterial: structure, properties: and applications in tissue engineering and
drug delivery. Natural and Synthetic Biomedical Polymers, 5, 91113.
Jiang, L., Lu, Y., Liu, X., Tu, H., Zhang, J., Shi, X., et al. (2015). Layer-by-layer
immobilization of quaternized carboxymethyl chitosan/organic rectorite and
alginate onto nanobrous mats and their antibacterial application.
Carbohydrate Polymers, 121, 428435.
Kalinov, K. N., Ignatova, M. G., Manolova, N. E., Markova, N. D., Karashanova, D. B., &
Rashkov, I. B. (2015). Novel antibacterial electrospun materials based on
polyelectrolyte complexes of a quaternized chitosan derivative. RSC Advances,
5(67), 5451754526.
Kaya, M., Cakmak, Y. S., Baran, T., Asan-Ozusaglam, M., Mentes, A., & Tozak, K. O.
(2014). New chitin, chitosan, and O-carboxymethyl chitosan sources from
resting eggs of Daphnia longispina (Crustacea); with physicochemical
characterization, and antimicrobial and antioxidant activities. Biotechnology
and Bioprocess Engineering, 19(1), 5869.
Kim, I. Y., Seo, S. J., Moon, H. S., Yoo, M. K., Park, I. Y., Kim, B. C., et al. (2008).
Chitosan and its derivatives for tissue engineering applications. Biotechnology
Advances, 26(1), 121.
Kim, Y. T., Yum, S., Heo, J. S., Kim, W., Jung, Y., & Kim, Y. M. (2014). Dithiocarbamate
chitosan as a potential polymeric matrix for controlled drug release. Drug
Development and Industrial Pharmacy, 40(2), 192200.
Kim, H. L., Jung, G. Y., Yoon, J. H., Han, J. S., Park, Y. J., Kim, D. G., et al. (2015).
Preparation and characterization of nano-sized
hydroxyapatite/alginate/chitosan composite scaffolds for bone tissue
engineering. Materials Science and Engineering: C, 54, 2025.
Kumar, J. P., Lakshmi, L., Jyothsna, V., Balaji, D. R., Saravanan, S., Moorthi, A., et al.
(2014). Synthesis and characterization of diopside particles and their
suitability along with chitosan matrix for bone tissue engineering in vitro and
in vivo. Journal of Biomedical Nanotechnology, 10(6), 970981.
Kurniawan, D. W., Fudholi, A., & Susidarti, R. A. (2012). Synthesis of thiolated
chitosan as matrix for the preparation of metformin hydrochloride
microparticles. Research in Pharmacy, 2, 2635.
Kyzas, G. Z., & Bikiaris, D. N. (2015). Recent modications of chitosan for adsorption
applications: a critical and systematic review. Marine Drugs, 13(1), 312337.
Lafeur, F., Hintzen, F., Rahmat, D., Shahnaz, G., Millotti, G., & Bernkop-Schnurch, A.
(2013). Enzymatic degradation of thiolated chitosan. Drug Development and
Industrial Pharmacy, 39(10), 15311539.

Le Tien, C., Lacroix, M., Ispas-Szabo, P., & Mateescu, M. A. (2003). N-acylated
chitosan: hydrophobic matrices for controlled drug release. Journal of
Controlled Release, 93(1), 113.
Lee, S. Y., & Kamarul, T. (2014). N O: carboxymethyl chitosan enhanced scaffold
porosity and biocompatibility under e-beam irradiation at 50 kGy.
International Journal of Biological Macromolecules, 64, 115122.
Li, B., Huang, L., Wang, X., Ma, J., & Xie, F. (2011). Biodegradation and compressive
strength of phosphorylated chitosan/chitosan/hydroxyapatite bio-composites.
Materials & Design, 32(8), 45434547.
Li, Y., Tan, Y., Xu, K., Lu, C., Liang, X., & Wang, P. (2015). In situ crosslinkable
hydrogels formed from modied starch and O-carboxymethyl chitosan. RSC
Advances, 5(38), 3030330309.
Liao, H. T., Chen, C. T., & Chen, J. P. (2011). Osteogenic differentiation and ectopic
bone formation of canine bone marrow-derived mesenchymal stem cells in
injectable thermo-responsive polymer hydrogel. Tissue Engineering Part C:
Methods, 17(11), 11391149.
Lim, S. M., Song, D. K., Oh, S. H., Lee-Yoon, D. S., Bae, E. H., & Lee, J. H. (2008). In vitro
and in vivo degradation behavior of acetylated chitosan porous beads Journal
of Biomaterials Science. Polymer Edition, 19(4), 453466.
Liu, Y. Y., Chu, S. L., Li, N. N., Bao, X. F., Gao, S., Sha, L., et al. (2013). Degradation
property of N, O-CMC/n--TCP composites with different mass proportion in
simulated body uid. Journal of Jilin University Science Edition, 43(1), 552556.
Liu, X., Chen, Y., Huang, Q., He, W., Feng, Q., & Yu, B. (2014). A novel
thermo-sensitive hydrogel based on thiolated
chitosan/hydroxyapatite/beta-glycerophosphate. Carbohydrate Polymers, 110,
6269.
Liu, B. M., Li, M., Yin, B. S., Zou, J. Y., Zhang, W. G., & Wang, S. Y. (2015). Effects of
incorporating carboxymethyl chitosan into PMMA bone cement containing
methotrexate. PUBLIC LIBRARY OF SCIENCE, 10(12).
Lopez-Perez, P. M., Da Silva, R. M., Sousa, R. A., Pashkuleva, I., & Reis, R. L. (2010).
Plasma-induced polymerization as a tool for surface functionalization of
polymer scaffolds for bone tissue engineering: an in vitro study. Acta
Biomaterialia, 6(9), 37043712.
Ma, L., Gao, C., Mao, Z., Zhou, J., Shen, J., Hu, X., et al. (2003). Collagen/chitosan
porous scaffolds with improved biostability for skin tissue engineering.
Biomaterials, 24(26), 48334841.
Ma, G., Yang, D., Li, Q., Wang, K., Chen, B., Kennedy, J. F., et al. (2010). Injectable
hydrogels based on chitosan derivative/polyethylene glycol dimethacrylate/N,
N-dimethylacrylamide as bone tissue engineering matrix. Carbohydrate
Polymers, 79(3), 620627.
Ma, K., Cai, X., Zhou, Y., Zhang, Z., Jiang, T., & Wang, Y. (2014). Osteogenetic
property of a biodegradable three-dimensional macroporous hydrogel coating
on titanium implants fabricated via EPD. Biomedical Materials, 9(1), 015008.
Madhumathi, K., Shalumon, K. T., Rani, V. D., Tamura, H., Furuike, T., Selvamurugan,
N., et al. (2009). Wet chemical synthesis of chitosan hydrogelhydroxyapatite
composite membranes for tissue engineering applications. International
Journal of Biological Macromolecules, 45(1), 1215.
Martinez, L. R., Mihu, M. R., Tar, M., Cordero, R. J., Han, G., Friedman, A. J., et al.
(2010). Demonstration of antibiolm and antifungal efcacy of chitosan
against candidal biolms, using an in vivo central venous catheter model.
Journal of Infectious Diseases, 201(9), 14361440.
Martins, A. F., Facchi, S. P., Follmann, H. D., Gerola, A. P., Rubira, A. F., & Muniz, E. C.
(2015). Shielding effect of surface ion pairs on physicochemical and
bactericidal properties of N, N, N-trimethyl chitosan salts. Carbohydrate
Research, 402, 252260.
Masuko, T., Minami, A., Iwasaki, N., Majima, T., Nishimura, S. I., & Lee, Y. C. (2005).
Thiolation of chitosan. Attachment of proteins via thioether formation.
Biomacromoelecules, 6(2), 880884.
Mishra, D., Bhunia, B., Banerjee, I., Datta, P., Dhara, S., & Maiti, T. K. (2011).
Enzymatically crosslinked
carboxymethylchitosan/gelatin/nano-hydroxyapatite injectable gels for
in situ bone tissue engineering application. Materials Science and Engineering:
C, 31(7), 12951304.
Mourya, V. K., & Inamdar, N. N. (2008). Chitosan-modications and applications:
opportunities galore. Reactive and Functional Polymers, 68(6), 10131051.
Mourya, V. K., Inamdar, N. N., & Tiwari, A. (2010). Carboxymethyl chitosan and its
applications. Advanced Materials Letters, 1(1), 1133.
Muzzarelli, R. A. A., Mattioli-Belmonte, M., Tietz, C., Biagini, R., Ferioli, G., Brunelli,
M. A., et al. (1994). Stimulatory effect on bone formation exerted by a modied
chitosan. Biomaterials, 15(13), 10751081.
Nayak, Y. (2010). HydroxyapatiteTZP Composites: Processing, Mechanical
Properties, Microstructure and in Vitro Bioactivity (Doctoral dissertation).
Doctoral Dissertation.
Niranjan, R., Koushik, C., Saravanan, S., Moorthi, A., Vairamani, M., &
Selvamurugan, N. (2013). A novel injectable temperature-sensitive zinc doped
chitosan/-glycerophosphate hydrogel for bone tissue engineering.
International Journal of Biological Macromolecules, 54, 2429.
Pardeshi, C. V., & Belgamwar, V. S. (2016). Controlled synthesis of N, N: N-trimethyl
chitosan for modulated bioadhesion and nasal membrane permeability.
International Journal of Biological Macromolecules, 82, 933944.
Park, J. H., Cho, Y. W., Chung, H., Kwon, I. C., & Jeong, S. Y. (2003). Synthesis and
characterization of sugar-bearing chitosan derivatives: aqueous solubility and
biodegradability. Biomacromolecules, 4(4), 10871091.
Park, H., Choi, B., Nguyen, J., Fan, J., Sha, S., Klokkevold, P., et al. (2013). Anionic
carbohydrate-containing chitosan scaffolds for bone regeneration.
Carbohydrate Polymers, 97(2), 587596.

R. LogithKumar et al. / Carbohydrate Polymers 151 (2016) 172188


Parker, R. A., Clegg, P. D., & Taylor, S. E. (2012). The in vitro effects of antibiotics on
cell viability and gene expression of equine bone marrow-derived
mesenchymal stromal cells. Equine Veterinary Journal, 44(3), 355360.
Patale, R. L., & Patravale, V. B. (2011). O, N-carboxymethyl chitosanzinc complex:
a novel chitosan complex with enhanced antimicrobial activity. Carbohydrate
Polymers, 85(1), 105110.
Patrulea, V., Applegate, L. A., Ostafe, V., Jordan, O., & Borchard, G. (2015). Optimized
synthesis of O-carboxymethyl-N, N, N-trimethyl chitosan. Carbohydrate
Polymers, 122, 4652.
Patrulea, V., Hirt-Burri, N., Jeannerat, A., Applegate, L. A., Ostafe, V., Jordan, O., et al.
(2016). Peptide-decorated chitosan derivatives enhance broblast adhesion
and proliferation in wound healing. Carbohydrate Polymers, 142, 114123.
Pattnaik, S., Nethala, S., Tripathi, A., Saravanan, S., Moorthi, A., & Selvamurugan, N.
(2011). Chitosan scaffolds containing silicon dioxide and zirconia nano
particles for bone tissue engineering. International Journal of Biological
Macromolecules, 49(5), 11671172.
Peng, Y., Han, B., Liu, W., & Xu, X. (2005). Preparation and antimicrobial activity of
hydroxypropyl chitosan. Carbohydrate Research, 340(11), 18461851.
Peng, Z. X., Wang, L., Du, L., Guo, S. R., Wang, X. Q., & Tang, T. T. (2010). Adjustment
of the antibacterial activity and biocompatibility of hydroxypropyltrimethyl
ammonium chloride chitosan by varying the degree of substitution of
quaternary ammonium. Carbohydrate Polymers, 81(2), 275283.
Peng, Z. X., Tu, B., Shen, Y., Du, L., Wang, L., Guo, S. R., et al. (2011). Quaternized
chitosan inhibits icaA transcription and biolm formation by Staphylococcus
on a titanium surface. Antimicrobial Agents and Chemotherapy, 55(2), 860866.
Peschel, D., Zhang, K., Fischer, S., & Groth, T. (2012). Modulation of osteogenic
activity of BMP-2 by cellulose and chitosan derivatives. Acta Biomaterialia, 8(1),
183193.
Peter, M., Binulal, N. S., Nair, S. V., Selvamurugan, N., Tamura, H., & Jayakumar, R.
(2010). Novel biodegradable chitosangelatin/nano-bioactive glass ceramic
composite scaffolds for alveolar bone tissue engineering. Chemical Engineering
Journal, 158(2), 353361.
Peter, M., Ganesh, N., Selvamurugan, N., Nair, S. V., Furuike, T., Tamura, H., et al.
(2010). Preparation and characterization of
chitosangelatin/nanohydroxyapatite composite scaffolds for tissue
engineering applications. Carbohydrate Polymers, 80(3), 687694.
Puvaneswary, S., Talebian, S., Raghavendran, H. B., Murali, M. R., Mehrali, M., A,
A. M., et al. (2015). Fabrication and in vitro biological activity of
TCP-Chitosan-Fucoidan composite for bone tissue engineering. Carbohydrate
Polymers, 134, 799807.
Qiao, P., Wang, J., Xie, Q., Li, F., Dong, L., & Xu, T. (2013). Injectable calcium
phosphatealginatechitosan microencapsulated MC3T3-E1 cell paste for
bone tissue engineering in vivo. Materials Science and Engineering: C, 33(8),
46334639.
Qin, C., Xiao, Q., Li, H., Fang, M., Liu, Y., Chen, X., et al. (2004). Calorimetric studies
of the action of chitosan-N-2-hydroxypropyl trimethyl ammonium chloride on
the growth of microorganisms. International Journal of Biological
Macromolecules, 34(1), 121126.
Qiu, Y., Zhang, N., An, Y. H., & Wen, X. (2007). Biomaterial strategies to reduce
implant-associated infections. The International Journal of Articial Organs,
30(9), 828841.
Raafat, D., Von Bargen, K., Haas, A., & Sahl, H. G. (2008). Insights into the mode of
action of chitosan as an antibacterial compound. Applied and Environmental
Microbiology, 74(12), 37643773.
Rajasree, R., & Rahate, K. P. (2013). An overview on various modications of
chitosan and its applications. International Journal of Pharmaceutical Sciences
and Research, 4(11), 4175.
Reves, B. T., Bumgardner, J. D., & Haggard, W. O. (2013). Fabrication of crosslinked
carboxymethylchitosan microspheres and their incorporation into composite
scaffolds for enhanced bone regeneration. Journal of Biomedical Materials
Research Part B:Applied Biomaterials, 101(4), 630639.
Riva, R., Ragelle, H., Des Rieux, A., Duhem, N., Jerome, C., & Preat, V. (2011).
Chitosan and chitosan derivatives in drug delivery and tissue engineering. In
Chitosan for Biomaterials II, 244, 1944.
Romero, R., Chubb, L., Travers, J. K., Gonzales, T. R., Ehrhart, N. P., & Kipper, M. J.
(2015). Coating cortical bone allografts with periosteum-mimetic scaffolds
made of chitosan, trimethyl chitosan and heparin. Carbohydrate Polymers, 122,
144151.
Sabaa, M. W., Abdallah, H. M., Mohamed, N. A., & Mohamed, R. R. (2015). Synthesis:
characterization and application of biodegradable crosslinked carboxymethyl
chitosan/poly (vinyl alcohol) clay nanocomposites. Materials Science and
Engineering: C, 56, 363373.
Sakloetsakun, D., Hombach, J. M., & Bernkop-Schnurch, A. (2009). In situ gelling
properties of chitosan-thioglycolic acid conjugate in the presence of oxidizing
agents. Biomaterials, 30(31), 61516157.
Saranya, N., Moorthi, A., Saravanan, S., Devi, M. P., & Selvamurugan, N. (2011).
Chitosan and its derivatives for gene delivery. International Journal of Biological
Macromolecules, 48(2), 234238.
Saranya, N., Saravanan, S., Moorthi, A., Ramyakrishna, B., & Selvamurugan, N.
(2011). Enhanced osteoblast adhesion on polymeric nano-scaffolds for bone
tissue engineering. Journal of Biomedical Nanotechnology, 7(2), 238244.
Saravanan, S., Nethala, S., Pattnaik, S., Tripathi, A., Moorthi, A., & Selvamurugan, N.
(2011). Preparation, characterization and antimicrobial activity of a
bio-composite scaffold containing chitosan/nano-hydroxyapatite/nano-silver
for bone tissue engineering. International Journal of Biological Macromolecules,
49(2), 188193.

187

Saravanan, S., Sameera, D. K., Moorthi, A., & Selvamurugan, N. (2013). Chitosan
scaffolds containing chicken feather keratin nanoparticles for bone tissue
engineering. International Journal of Biological Macromolecules, 62, 481486.
Shalumon, K. T., Binulal, N. S., Selvamurugan, N., Nair, S. V., Menon, D., Furuike, T.,
et al. (2009). Electrospinning of carboxymethyl chitin/poly (vinyl alcohol)
nanobrous scaffolds for tissue engineering applications. Carbohydrate
Polymers, 77(4), 863869.
Shen, S. C., Ng, W. K., Shi, Z., Chia, L., Neoh, K. G., & Tan, R. B. H. (2011). Mesoporous
silica nanoparticle-functionalized poly (methyl methacrylate)-based bone
cement for effective antibiotics delivery. Journal of Materials Science: Materials
in Medicine, 22(10), 22832292.
Shi, Z., Neoh, K. G., Kang, E. T., Poh, C. K., & Wang, W. (2009). Surface
functionalization of titanium with carboxymethyl chitosan and immobilized
bone morphogenetic protein-2 for enhanced osseointegration.
Biomacromolecules, 10(6), 16031611.
Sowjanya, J. A., Singh, J., Mohita, T., Sarvanan, S., Moorthi, A., Srinivasan, N., et al.
(2013). Biocomposite scaffolds containing chitosan/alginate/nano-silica for
bone tissue engineering. Colloids and Surfaces B: Biointerfaces, 109, 294300.
Srakaew, V., Ruangsri, P., Suthin, K., Thunyakitpisal, P., & Tachaboonyakiat, W.
(2011). Sodium-phosphorylated chitosan/zinc oxide complexes and evaluation
of their cytocompatibility: an approach for periodontal dressing. Journal of
Biomaterials Applications, 27, 403412.
Swetha, M., Sahithi, K., Moorthi, A., Srinivasan, N., Ramasamy, K., & Selvamurugan,
N. (2010). Biocomposites containing natural polymers and hydroxyapatite for
bone tissue engineering. International Journal of Biological Macromolecules,
47(1), 14.
Tachaboonyakiat, W., Netswasdi, N., Srakaew, V., & Opaprakasit, M. (2010).
Elimination of inter-and intramolecular crosslinks of phosphorylated chitosan
by sodium salt formation. Polymer Journal, 42(2), 148156.
Takada, T., Katagiri, T., Ifuku, M., Morimura, N., Kobayashi, M., Hasegawa, K., et al.
(2003). Sulfated polysaccharides enhance the biological activities of bone
morphogenetic proteins. Journal of Biological Chemistry, 278(44), 4322943235.
Tan, Y., Han, F., Ma, S., & Yu, W. (2011). Carboxymethyl chitosan prevents formation
of broad-spectrum biolm. Carbohydrate Polymers, 84(4), 13651370.
Tan, H., Guo, S., Yang, S., Xu, X., & Tang, T. (2012). Physical characterization and
osteogenic activity of the quaternized chitosan-loaded PMMA bone cement.
Acta Biomaterialia, 8(6), 21662174.
Tan, H., Peng, Z., Li, Q., Xu, X., Guo, S., & Tang, T. (2012). The use of quaternised
chitosan-loaded PMMA to inhibit biolm formation and downregulate the
virulence-associated gene expression of antibiotic-resistant staphylococcus.
Biomaterials, 33(2), 365377.
Tan, H., Ao, H., Ma, R., & Tang, T. (2013). Quaternised chitosan-loaded
polymethylmethacrylate bone cement: biomechanical and histological
evaluations. Journal of Orthopaedic Translation, 1(1), 5766.
Tan, H. L., Ao, H. Y., Ma, R., Lin, W. T., & Tang, T. T. (2014). In vivo effect of
quaternized chitosan-loaded polymethylmethacrylate bone cement on
methicillin-resistant staphylococcus epidermidis infection of the tibial
metaphysis in a rabbit model. Antimicrobial Agents and Chemotherapy, 58(10),
60166023.
Tang, T., Zhang, G., Lau, C. P., Zheng, L. Z., Xie, X. H., Wang, X. L., et al. (2011). Effect
of water-soluble P-chitosan and S-chitosan on human primary osteoblasts and
giant cell tumor of bone stromal cells. Biomedical Materials, 6(1), 015004.
Tangpasuthadol, V., Pongchaisirikul, N., & Hoven, V. P. (2003). Surface modication
of chitosan lms: effects of hydrophobicity on protein adsorption.
Carbohydrate Research, 338(9), 937942.
Thian, E. S., Konishi, T., Kawanobe, Y., Lim, P. N., Choong, C., Ho, B., et al. (2013).
Zinc-substituted hydroxyapatite: a biomaterial with enhanced bioactivity and
antibacterial properties. Journal of Materials Science: Materials in Medicine,
24(2), 437445.
Tian, X. L., Tian, D. F., Wang, Z. Y., & Mo, F. K. (2009). Synthesis and evaluation of
Chitosan-Vitamin C complex. Indian Journal of Pharmaceutical Sciences, 71(4),
371376.
Tokura, S., & Tamura, H. (2001). O-Carboxymethyl-chitin concentration in
granulocytes during bone repair. Biomacromolecules, 2(2), 417421.
Tripathi, A., Saravanan, S., Pattnaik, S., Moorthi, A., Partridge, N. C., &
Selvamurugan, N. (2012). Bio-composite scaffolds containing
chitosan/nano-hydroxyapatite/nano-copperzinc for bone tissue engineering.
International Journal of Biological Macromolecules, 50(1), 294299.
Tsai, W. B., Chen, Y. R., & Liu, H. L. (2013). RGD-conjugated crosslinked chitosan
scaffolds for culture and osteogenic differentiation of mesenchymal stem cells.
Journal of the Taiwan Institute of Chemical Engineers, 44(1), 17.
Venkatesan, J., Pangestuti, R., Qian, Z. J., Ryu, B., & Kim, S. K. (2010). Biocompatibility
and alkaline phosphatase activity of phosphorylated chitooligosaccharides on
the osteosarcoma MG63 cell line. Journal of Functional Biomaterials, 1(1), 313.
Venkatesan, J., Bhatnagar, I., & Kim, S. K. (2014). Chitosan-alginate biocomposite
containing fucoidan for bone tissue engineering. Marine Drugs, 12(1),
300316.
Venkatesan, J., Bhatnagar, I., Manivasagan, P., Kang, K. H., & Kim, S. K. (2015).
Alginate composites for bone tissue engineering: a review. International
Journal of Biological Macromolecules, 72, 269281.
Vijapur, L. S., Sreenivas, S. A., Patil, S. H., Vijapur, P. V., & Patwari, P. K. (2012).
Thiolated chitosans: a novel mucoadhesive polymers: a review. International
Research Journal of Pharmacy, 3(4), 5157.
Wan, A., Xu, Q., Sun, Y., & Li, H. (2013). Antioxidant activity of high molecular
weight chitosan and N, O-quaternized chitosans. Journal of Agricultural and
Food Chemistry, 61(28), 69216928.

188

R. LogithKumar et al. / Carbohydrate Polymers 151 (2016) 172188

Wang, M., Cheng, X., Zhu, W., Holmes, B., Keidar, M., & Zhang, L. G. (2013). Design
of biomimetic and bioactive cold plasma-modied nanostructured scaffolds
for enhanced osteogenic differentiation of bone marrow-derived
mesenchymal stem cells. Tissue Engineering Part A, 20(56), 10601071.
Wang, X. L., Xie, X. H., Zhang, G., Chen, S. H., Yao, D., He, K., et al. (2013). Exogenous
phytoestrogenic molecule icaritin incorporated into a porous scaffold for
enhancing bone defect repair. Journal of Orthopaedic Research, 31(1), 164172.
Wang, Y., Peng, W., Liu, X., Zhu, M., Sun, T., Peng, Q., et al. (2014). Study of bilineage
differentiation of human-bone-marrow-derived mesenchymal stem cells in
oxidized sodium alginate/N-succinyl chitosan hydrogels and synergistic effects
of RGD modication and low-intensity pulsed ultrasound. Acta Biomaterialia,
10(6), 25182528.
Wang, C. H., Liu, W. S., Sun, J. F., Hou, G. G., Chen, Q., Cong, W., et al. (2016).
Non-toxic O-quaternized chitosan materials with better water solubility and
antimicrobial function. International Journal of Biological Macromolecules, 84,
418427.
Wang, W. (2006). A novel hydrogel crosslinked hyaluronan with glycol chitosan.
Journal of Materials Science: Materials in Medicine, 17(12), 12591265.
Wei, C. Z., Hou, C. L., Gu, Q. S., Jiang, L. X., Zhu, B., & Sheng, A. L. (2009). A
thermosensitive chitosan-based hydrogel barrier for post-operative adhesions
prevention. Biomaterials, 30(29), 55345540.
Wiens, M., Elkhooly, T. A., Schroder, H. C., Mohamed, T. H., & Muller, W. E. (2014).
Characterization and osteogenic activity of a silicatein/biosilica-coated
chitosan-graft-polycaprolactone. Acta Biomaterialia, 10(10), 44564464.
Winge, M. I., Reikeras, O., & Rokkum, M. (2011). Calcium phosphate bone cement: a
possible alternative to autologous bone graft. A radiological and biomechanical
comparison in rat tibial bone. Archives of Orthopaedic and Trauma Surgery,
131(8), 10351041.
Wongpanit, P., Sanchavanakit, N., Pavasant, P., Supaphol, P., Tokura, S., &
Rujiravanit, R. (2005). Preparation and characterization of microwave-treated
carboxymethyl chitin and carboxymethyl chitosan lms for potential use in
wound care application. Macromolecular Bioscience, 5(10), 10011012.
Wu, X., Black, L., Santacana-Laftte, G., & Patrick, C. W. (2007). Preparation and
assessment of glutaraldehyde-crosslinked collagenchitosan hydrogels for
adipose tissue engineering. Journal of Biomedical Materials Research Part A,
81(1), 5965.
Xiao, B., Wan, Y., Wang, X., Zha, Q., Liu, H., Qiu, Z., et al. (2012). Synthesis and
characterization of N-(2-hydroxy) propyl-3-trimethyl ammonium chitosan
chloride for potential application in gene delivery. Colloids and Surfaces B:
Biointerfaces, 91, 168174.
Xu, T., Xin, M., Li, M., Huang, H., & Zhou, S. (2010). Synthesis, characteristic and
antibacterial activity of N, N, N-trimethyl chitosan and its carboxymethyl
derivatives. Carbohydrate Polymers, 81(4), 931936.
Xu, X., Zhuang, X., Cheng, B., Xu, J., Long, G., & Zhang, H. (2010). Manufacture and
properties of cellulose/O-hydroxyethyl chitosan blend bers. Carbohydrate
Polymers, 81(3), 541544.
Xu, Z., Neoh, K. G., Lin, C. C., & Kishen, A. (2011). Biomimetic deposition of calcium
phosphate minerals on the surface of partially demineralized dentine modied
with phosphorylated chitosan. Journal of Biomedical Materials Research Part B:
Applied Biomaterials, 98(1), 150159.
Yalinca, Z., Yilmaz, E., Taneri, B., Bullici, F., & Tuzmen, S. (2013). Blood contact
properties of ascorbyl chitosan Journal of Biomaterials Science. Polymer
Edition, 24(17), 19691987.

Yang, X., Zhang, C., Qiao, C., Mu, X., Li, T., Xu, J., et al. (2015). A simple and
convenient method to synthesize
N-[(2-hydroxyl)-propyl-3-trimethylammonium] chitosan chloride in an ionic
liquid. Carbohydrate Polymers, 130, 325332.
Yeh, H. Y., & Lin, J. C. (2012). Surface phosphorylation for polyelectrolyte complex
of chitosan and its sulfonated derivative: surface analysis, blood compatibility
and adipose derived stem cell contact properties Journal of Biomaterials
Science. Polymer Edition, 23(1-4), 233250.
You, J., Xie, S., Cao, J., Ge, H., Xu, M., Zhang, L., et al. (2016). Quaternized
chitosan/poly (acrylic acid) polyelectrolyte complex hydrogels with tough,
self-recovery, and tunable mechanical properties. Macromolecules, 49(3),
10491059.
Younes, I., & Rinaudo, M. (2015). Chitin and chitosan preparation from marine
sources. Structure, properties and applications. Marine Drugs, 13(3),
11331174.
Zhang, B. G., Myers, D. E., Wallace, G. G., Brandt, M., & Choong, P. F. (2014). Bioactive
coatings for orthopaedic implantsrecent trends in development of implant
coatings. International Journal of Molecular Sciences, 15(7), 1187811921.
Zhang, X., Zhang, Y., Ma, G., Yang, D., & Nie, J. (2015). The effect of the prefrozen
process on properties of a chitosan/hydroxyapatite/poly (methyl
methacrylate) composite prepared by freeze drying method used for bone
tissue engineering. RSC Advances, 5(97), 7967979686.
Zhao, B., Katagiri, T., Toyoda, H., Takada, T., Yanai, T., Fukuda, T., et al. (2006).
Heparin potentiates the in vivo ectopic bone formation induced by bone
morphogenetic protein-2. Journal of Biological Chemistry, 281(32),
2324623253.
Zhao, J., Shen, G., Liu, C., Wang, S., Zhang, W., Zhang, X., et al. (2011). Enhanced
healing of rat calvarial defects with sulfated chitosan-coated calcium-decient
hydroxyapatite/bone morphogenetic protein 2 scaffolds. Tissue Engineering
Part A, 18(12), 185197.
Zhao, X., Li, P., Guo, B., & Ma, P. X. (2015). Antibacterial and conductive injectable
hydrogels based on quaternized chitosan-graft-polyaniline/oxidized dextran
for tissue engineering. Acta Biomaterialia, 26, 236248.
Zheng, D., Neoh, K. G., & Kang, E. T. (2016). Bifunctional coating based on
carboxymethyl chitosan with stable conjugated alkaline phosphatase for
inhibiting bacterial adhesion and promoting osteogenic differentiation on
titanium. Applied Surface Science, 360, 8697.
Zhou, Y., Xu, L., Zhang, X., Zhao, Y., Wei, S., & Zhai, M. (2012). Radiation synthesis of
gelatin/CM-chitosan/-tricalcium phosphate composite scaffold for bone
tissue engineering. Materials Science and Engineering: C, 32(4), 9941000.
Zhou, Y., Yang, H., Liu, X., Mao, J., Gu, S., & Xu, W. (2013). Potential of
quaternization-functionalized chitosan ber for wound dressing. International
Journal of Biological Macromolecules, 52, 327332.
Zhou, P., Xia, Y., Cheng, X., Wang, P., Xie, Y., & Xu, S. (2014). Enhanced bone tissue
regeneration by antibacterial and osteoinductive silica-HACC-zein composite
scaffolds loaded with rhBMP-2. Biomaterials, 35(38), 1003310045.
Zhou, Z., Yan, D., Cheng, X., Kong, M., Liu, Y., Feng, C., et al. (2016). Biomaterials
based on N, N, N-Trimethyl chitosan bers in wound dressing applications.
International Journal of Biological Macromolecules, 89, 471476.
Zhu, D., Cheng, H., Li, J., Zhang, W., Shen, Y., Chen, S., et al. (2016). Enhanced
water-solubility and antibacterial activity of novel chitosan derivatives
modied with quaternary phosphonium salt. Materials Science and Engineering:
C, 61, 7984.

Anda mungkin juga menyukai