You are on page 1of 13

European Polymer Journal 49 (2013) 780–792

Contents lists available at SciVerse ScienceDirect

European Polymer Journal


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

Feature Article

Chitosan-based biomaterials for tissue engineering


Florence Croisier 1, Christine Jérôme ⇑
Center for Education and Research on Macromolecules (CERM), Department of Chemistry, University of Liège, Allée de la Chimie 3, B6a, 4000 Liège, Belgium

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

Article history: Derived from chitin, chitosan is a unique biopolymer that exhibits outstanding properties,
Received 10 October 2012 beside biocompatibility and biodegradability. Most of these peculiar properties arise from
Received in revised form 12 December 2012 the presence of primary amines along the chitosan backbone. As a consequence, this poly-
Accepted 15 December 2012
saccharide is a relevant candidate in the field of biomaterials, especially for tissue engi-
Available online 23 January 2013
neering. The current article highlights the preparation and properties of innovative
chitosan-based biomaterials, with respect to their future applications. The use of chitosan
Keywords:
in 3D-scaffolds – as gels and sponges – and in 2D-scaffolds – as films and fibers – is dis-
Chitosan scaffolds
Biomaterials
cussed, with a special focus on wound healing application.
Tissue engineering Ó 2013 Elsevier Ltd. Open access under CC BY-NC-ND license.
Wound healing

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 781
2. Chitosan: structure and extraction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 781
3. Structure–properties relationship . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 781
4. Remarkable properties of chitosan as biomaterial . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 782
5. Chitosan 3D-scaffolds for tissue engineering . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 783
5.1. Chitosan hydrogels . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 783
5.1.1. Physically associated chitosan hydrogels . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 783
5.1.2. Chitosan cross-linked by coordination complex . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 785
5.1.3. Chemically cross-linked chitosan hydrogels . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 785
5.2. Chitosan sponges. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 785
6. Chitosan 2D-scaffolds for wound dressing . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 785
6.1. Chitosan films . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 786
6.1.1. Improving properties of chitosan free-standing films . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 786
6.1.2. Chitosan thin films . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 786
6.2. Chitosan porous nanofiber membranes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 788
7. Conclusions. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 789
Acknowledgements . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 789
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 789

⇑ Corresponding author. Tel.: +32 4 366 34 91; fax: +32 4 366 34 97.
E-mail addresses: fcroisier@ulg.ac.be (F. Croisier), C.Jerome@ulg.ac.be (C. Jérôme).
URL: http://www.cerm.ulg.ac.be (C. Jérôme).
1
Tel.: +32 4 366 34 69; fax: +32 4 366 34 97.

0014-3057 Ó 2013 Elsevier Ltd. Open access under CC BY-NC-ND license.


http://dx.doi.org/10.1016/j.eurpolymj.2012.12.009
F. Croisier, C. Jérôme / European Polymer Journal 49 (2013) 780–792 781

1. Introduction under severe alkaline conditions or by enzymatic hydroly-


sis in the presence of particular enzymes, among of chitin
Over the last years, many attempts have been made to deacetylase [5,6].
replace petrochemical products by renewable, biosourced After cellulose, chitin is the second most abundant bio-
components. Abundant naturally occurring polymers – as polymer [2] and is commonly found in invertebrates – as
starch, collagen, gelatin, alginate, cellulose and chitin [1] crustacean shells or insect cuticles – but also in some
– represent attractive candidates as they could reduce mushrooms envelopes, green algae cell walls, and yeasts
the actual dependence on fossil fuels, and consequently [7–9]. At industrial scale, the two main sources of chitosan
have a positive environmental impact. The most challeng- are crustaceans and fungal mycelia; the animal source
ing part of this approach is to obtain bio-based materials shows however some drawbacks as seasonal, of limited
with properties equivalent to those of fully synthetic prod- supplies and with product variability which can lead to
ucts, from a functional point of view. In this respect, chito- inconsistent physicochemical characteristics [10]. The
san is a quite unique bio-based polymer: its intrinsic mushroom source offers the advantage of a controlled pro-
properties are so singular and valuable that chitosan pos- duction environment all year round that insures a better
sesses no actual petrochemical equivalent. Consequently, reproducibility of the resulting chitosan [11], the physical
the inherent characteristics of chitosan make it exploitable properties of extracted chitosan being notably related to
directly for itself. the growth substrate composition. Moreover, the vegetable
source is generally preferred to the crustacean one from an
2. Chitosan: structure and extraction allergenic point of view – the produced chitosan being
safer for biomedical and healthcare applications [12]. The
Chitosan is a linear, semi-crystalline polysaccharide mushroom-extracted chitosan typically presents a nar-
composed of (1 ? 4)-2-acetamido-2-deoxy-b-D-glucan rower molecular mass distribution than the chitosan pro-
(N-acetyl D-glucosamine) and (1 ? 4)-2-amino-2-deoxy- duced from seafood [12], and may also differ in terms of
b-D-glucan (D-glucosamine) units [2]. The structure of this molecular mass, DD and distribution of deacetylated
polymer is depicted in Fig. 1. As such, chitosan is not exten- groups [11,13]. Chitosan DD greatly varies between 60
sively present in the environment – however, it can be eas- and 100% while its molecular weight typically ranges from
ily derived from the partial deacetylation of a natural 300 to 1000 kDA [1], depending on the source and prepara-
polymer: the chitin (Fig. 2). The deacetylation degree tion. Chitosan oligomers can be prepared by degradation of
(DD) of chitosan, giving indication of the number of amino chitosan using specific enzyme [15] or reagent as hydrogen
groups along the chains, is calculated as the ratio of D-glu- peroxide [16].
cosamine to the sum of D-glucosamine and N-acetyl D-glu- At the time of its production, it is difficult to predict
cosamine. To be named ‘‘chitosan’’, the deacetylated chitin chitosan characteristics. Therefore, the current approach
should contain at least 60% of D-glucosamine residues [3,4] consists in analyzing the resulting product composition
(which corresponds to a deacetylation degree of 60). The and properties rather than in targeting predefined charac-
deacetylation of chitin is conducted by chemical hydrolysis teristics by controlling the process of production. The char-
acterization of chitosan is thus quite of importance but it is
not easygoing nor straightforward. Being a natural-based
O compound, chitosan may be contaminated by organic
and inorganic impurities, and presents broad polydisper-
sity, this is why producers mainly refer to samples viscos-
OH HN ity rather than molecular mass. Chitosan is also poorly
O HO soluble, except in acidic medium (as it will be discussed
O O in the following paragraph) which makes analyzes difficult
HO O to perform. Different methods including pH-potentiomet-
NH2 OH ric titration, IR-spectroscopy, 1H NMR spectroscopy, but
also UV-spectroscopy, colloidal titration and enzymatic
Fig. 1. Chemical structure of chitosan. degradation are reported in the literature for the determi-
nation of chitosan deacetylation degree [17], while its
molecular mass is typically deduced from viscosimetry or
O determined by size exclusion chromatography [2].

OH HN
3. Structure–properties relationship
O HO
O O The presence of amino groups in the chitosan structure
HO O
(Fig. 1) differentiates chitosan from chitin, and gives to this
HN OH
polymer many peculiar properties. Indeed, the amino
groups of the D-glucosamine residues might be protonated
providing solubility in diluted acidic aqueous solutions
O
(pH < 6) [14]. In contrast, practical applications of chitin
Fig. 2. Chemical structure of chitin. are extremely limited due to its poor solubility, if any
782 F. Croisier, C. Jérôme / European Polymer Journal 49 (2013) 780–792

[18]. Interestingly, the aqueous solubility of chitosan is pH- The haemostatic activity of chitosan can also be related
dependent allowing processability under mild conditions to the presence of positive charges on chitosan backbone.
[4], which opens prospects to a wide range of applications, Indeed, red blood cells membranes are negatively charged,
particularly in the field of cosmetics [2]. and can thus interact with the positively charged chitosan.
Due to these amino groups, chitosan also efficiently Besides, chitin shows less effective haemostatic activity
complexes various species, such as metal ions, and there- than chitosan, which tends to confirm this explanation
fore is often used for the treatment of waste waters, puri- [32–34].
fying them by recovering heavy metals [2]. The complexing Due to its positive charges, chitosan can also interact
ability of chitosan is further exploited for beverages (wine, with the negative part of cells membrane, which can lead
juices. . .) clarification [19]. to reorganization and an opening of the tight junction pro-
Chitosan with protonated amino groups becomes a teins, explaining the permeation enhancing property of
polycation that can subsequently form ionic complexes this polysaccharide. As for mucoadhesion, if chitosan DD
with a wide variety of natural or synthetic anionic increases, the permeation ability also increases [35].
species [4], such as lipids, proteins, DNA and some nega- The case of chitosan antimicrobial activity is slightly
tively charged synthetic polymers as poly(acrylic acid) more complex; two main mechanisms have been reported
[4,18,20,21]. As a matter of fact, chitosan is the only posi- in the literature to explain chitosan antibacterial and
tively charged, naturally occurring polysaccharide [18]. antifungal activities. In the first proposed mechanism,
As a polyelectrolyte, chitosan can notably be employed positively charged chitosan can interact with negatively
for the preparation of multilayered films, using layer-by- charged groups at the surface of cells, and as a conse-
layer deposition technique [22]. This particular case will quence, alter its permeability. This would prevent essential
be detailed later on. materials to enter the cells or/and lead to the leaking of
The amino and alcohol functions along chitosan chains fundamental solutes out of the cell. The second mechanism
enable this polysaccharide to form stable covalent bon- involves the binding of chitosan with the cell DNA (still via
dings with other species. Non-specific reactions can be protonated amino groups), which would lead to the inhibi-
performed on the alcohol groups, as etherification and tion of the microbial RNA synthesis. Chitosan antimicrobial
esterification reactions [2] but remarkably, the amino property might in fact result from a combination of both
group of D-glucosamine residues may be specifically quat- mechanisms [23,36].
ernized or reacted with aldehyde functions under mild The polycationic nature of chitosan also allows explain-
conditions through reductive amination [2]. Various func- ing chitosan analgesic effects. Indeed, the amino groups of
tionalizations can be introduced along chitosan backbone the D-glucosamine residues can protonate in the presence
by this technique to further extend chitosan field of of proton ions that are released in the inflammatory area,
applications. resulting in an analgesic effect [37].
Besides, chitosan exhibits other remarkable intrinsic Now, to explain chitosan biodegradability, it is impor-
properties: this polysaccharide exhibits antibacterial activ- tant to remember that chitosan is not only a polymer bear-
ity [23,24], along with antifungal [9], mucoadhesive [25], ing amino groups, but also a polysaccharide, which
analgesic [9] and haemostatic properties [26]. It can be bio- consequently contains breakable glycosidic bonds. Chito-
degraded into non-toxic residues [27,28] – the rate of its san is actually degraded in vivo by several proteases, and
degradation being highly related to the molecular mass mainly lysozyme [9,38,39]. Till now, eight human chitinas-
of the polymer and its deacetylation degree – and has es have been identified, three of them possessing enzy-
proved to some extent biocompatibility with physiological matic activity on chitosan [40]. The biodegradation of
medium [29,30]. All these singular features make chitosan chitosan leads to the formation of non-toxic oligosaccha-
an outstanding candidate for biomedical applications. rides of variable length. These oligosaccharides can be
incorporated in metabolic pathways or be further excreted
4. Remarkable properties of chitosan as biomaterial [41]. The degradation rate of chitosan is mainly related to
its degree of deacetylation, but also to the distribution of
As already mentioned, several remarkable properties of N-acetyl D-glucosamine residues and the molecular mass
chitosan offered unique opportunities to the development of chitosan [42–44].
of biomedical applications. The elucidation of their mecha- To explain the relationship between chitosan biodegra-
nism will lead to a better understanding of chitosan med- dation and DD, it is important to remember that chitosan is
ical and pharmaceutical interest. a semi-crystalline polymer; crystallinity is indeed maximum
The presence of the protonable amino group along for a DD equal to 0 or 100% (chitin or fully deacetylated
D-glucosamine residues allows to elucidate most of chito- chitosan, respectively), and decreases for intermediate
san properties. Indeed, the mucoadhesion of chitosan for DD. Yet, as polymer crystallinity is inversely related to
example, can be explained by the presence of negatively the biodegradation kinetic, when chitosan DD decreases
charged residues (sialic acid) in the mucin – the glycopro- (close to 60%), its crystallinity also decreases, which results
tein that composes the mucus. In acidic medium, chitosan in an increase of the biodegradation rate. Besides, the dis-
amino groups are positively charged and can thus interact tribution of acetyl residues along chitosan will also affect
with the mucin. This mucoadhesion is directly related to its crystallinity, and consequently the biodegradation rate.
the DD of chitosan: actually, if chitosan DD increases, the Finally, it can be reasonably assumed that smaller chitosan
number of positive charges also increases, which leads to chains will be more rapidly degraded into oligosaccharides
improved mucoadhesive properties [31]. than chitosan with higher molecular mass.
F. Croisier, C. Jérôme / European Polymer Journal 49 (2013) 780–792 783

Considering all the aforementioned properties, it is not of the gel, making it able to soak up/absorb large quantities
surprising that chitosan was, is and will be tested in many of water while remaining insoluble in the liquid phase [12].
biomedical and pharmaceutical applications, notably for Hydrogels are interesting biomaterials as their high water
sutures, dental and bone implants and as artificial skin content makes them compatible with a majority of living
[2]. Within the framework of these tests, chitosan has tissues. Moreover, they are soft and bendable, which min-
shown biocompatibility [9,45] and was notably approved imizes the damage to the surrounding tissue during and
by the Food and Drug Administration (FDA) for use in after implantation in the patient [38]. The mechanical
wound dressings [46]. properties of hydrogels tend to mimic those of the soft
However, the compatibility of chitosan with physiolog- body-tissues, which allows the gels to insure both func-
ical medium depends on the preparation method (residual tional and morphological characteristics of the tissue to
proteins could indeed cause allergic reactions) and on the be repaired [38]. This is why hydrogels are often used as
DD – biocompatibility increases with DD increase. Chito- biomedical scaffolds for tissue replacements, but also for
san actually proved to be more cytocompatible in vitro other biomedical applications such as drug and growth fac-
than chitin. Indeed, while the number of positive charges tor delivery [52–54].
increases, the interaction between cells and chitosan in- Three main types of chitosan hydrogels have been
creases as well, which tends to improve biocompatibility developed, presenting either reversible or irreversible gela-
[47]. tion. Chitosan can indeed be either physically associated,
Besides, some chemical modifications of chitosan struc- coordinated with metal ions or irreversibly/chemically
ture could induce toxicity [38]. cross-linked into hydrogels [38].
After describing chitosan main properties in the previ-
ous paragraphs, the following will focus on the potential 5.1.1. Physically associated chitosan hydrogels
applications of chitosan as a biomaterial, and more pre- The formation of ‘‘physical’’ hydrogels is based on the
cisely on its processing methods for uses as biomedical reversible interactions that can occur between polymer
3D-scaffolds for tissue engineering, and 2D-scaffolds espe- chains. Those interactions have a non-covalent nature,
cially for wound healing purposes. such as electrostatic interactions, hydrophobic interactions
or hydrogen bondings [55,56].
5. Chitosan 3D-scaffolds for tissue engineering Those interactions can be dependent of various param-
eters as pH, concentration, temperature. . ., which make
During the fabrication of implantable scaffolds particu- them not very stable, exhibiting reversible gelation. The
lar attention should be paid to body compatibility, swelling of those hydrogels can be tuned by adjusting the
mechanical properties, scaffold morphology and porosity, nature and the quantities of each component, in order to
as well as healing and tissue replacement capacity [48]. increase or decrease the number of interactions. As a rule,
According to literature, the main requirements for the fewer interactions will lead to a softer gel while a higher
elaboration of tissue engineering scaffolds also include that number of interactions will give a tighter and stiffer gel.
the scaffold should not induce acute or chronic response, It is remarkable to observe that chitosan is able to form
should be biodegradable so that the cured tissue will be a gel, all by itself, without the need of any additive. The
able to replace the biomaterial, possess surface properties process is based on the neutralization of chitosan amino
that will promote cell attachment, differentiation and pro- groups and thus the inhibition of the repulsion between
liferation, have suitable mechanical properties for handling chitosan chains. The formation of the hydrogel then occurs
and to mimic the damaged tissue, and finally, be manufac- via hydrogen bonds, hydrophobic interactions and chito-
tured into variety of shapes [49,50]. san crystallites [38,70], as illustrated in Fig. 3.
Due to its aforementioned remarkable properties, chito- Further on, chitosan hydrogels can be formed by blend-
san appears thus as a relevant candidate for the prepara- ing chitosan with other water-soluble non-ionic polymers,
tion of such biomaterials, which could substitute for as poly(vinyl alcohol) (PVA) [55,71]. Thermo-sensitive
missing or damaged tissue and organ [48], and allow cell chitosan hydrogels can be obtained by blending chitosan
attachment and proliferation [51] provided that 3D-scaf- with polyol salts, as glycerol phosphate disodium salt
folds might be produced. Such development relies thus [72]. Chitosan structure can also be modified to form phys-
on robust processing methods for chitosan making able ical hydrogels: chitosan-g-poly(ethylene glycol) (PEG)
to adjust the scaffold properties at best to the diseased tis- graft copolymer is able of self-organization depending on
sue requirements. Therefore, methodologies have been the temperature to form stable hydrogels [73].
developed to finely shape chitosan hydrogels and foams Thanks to the polycationic nature of chitosan in acidic
(or sponges) as pertinent 3D-scaffolds applicable for tissue conditions, formation of chitosan hydrogels through elec-
engineering. These will be reviewed in the following trostatic interactions involving small-size polyanions but
sections. also polyelectrolytes appears straightforward (Fig. 4a).
Positively charged chitosan will interact with nega-
5.1. Chitosan hydrogels tively charged molecules, such as phosphates, sulfates
and citrates ions [57,58], to form hydrogels. Varying con-
Gels are composed of a solid phase, typically represent- centration and size of the anionic species as well as the
ing less than 10% of the total volume of the gel, and a liquid numbers of D-glucosamine units vs. N-acetyl-D-glucosa-
phase. In hydrogels, the liquid phase is water (and some- mine units allows fine tuning of the swelling of the ob-
times adjuvants). The solid phase ensures the consistency tained hydrogel.
784 F. Croisier, C. Jérôme / European Polymer Journal 49 (2013) 780–792

H bonds
Hydrophobic interactions
Crystallites

Fig. 3. Schematic representation of physical chitosan hydrogel, without any additives.

Non-covalent cross-linking (a) Coordination complex cross-linking (b) Covalent cross-linking (c)

Via electrostatic interactions with anions


(as phosphates, sulfates and citrates) or
polyanions (as proteins (gelatin and Via amide or ester bonds formation
Via coordination bonds with metal ions
collagen…) and anionic polysaccharides (with glyoxal, glutaraldehyde and
(as Pd(II), Pt (II) and Mo(VI)).
(hyaluronic acid…)). Genipin…).
Also via H bonds, hydrophobic
interactions and crystallization

Fig. 4. Schematic representation of three types of chitosan hydrogels, prepared in presence of additives. Gels can be formed through non-covalent cross-
linking (a), coordination complex cross-linking (b) and covalent cross-linking (c).

Larger negatively charged molecules, natural and syn- It should be noted that electrostatic interactions can oc-
thetic polyanions, can also give gels with chitosan. In the cur with other secondary interactions, as for example
natural polyanion category, proteins (as gelatin, collagen, hydrogen bonding [68,69]. However, the interactions be-
keratin, albumin and fibroin) [59–61], anionic polysaccha- tween charged chitosan and anions or polyanions are
rides (as hyaluronic acid, alginate, pectin, heparin, xanthan, stronger than these secondary bonding. The preparation
dextran sulfate, chodroitin sulfate, fucoidan) [62–65], of ionic chitosan hydrogels avoids the use of catalyst or
carboxymethyl cellulose and glycosaminoglycans [62,66] toxic reacting agent, which is a prerequisite for biomedical
have been reported for such purpose. Synthetic polyanions applications.
– as poly(acrylic acid) (PAA) – were also used to form The easiness of gelation without the need of toxic addi-
hydrogels [67]. The charge density of chitosan and of the tives, in conditions compatible with the human body led to
polyanion will play a major role in the swelling and stabil- the development of injectable solutions that exhibit a
ity of the formed hydrogels. Proper adjustment of the pH of sol/gel transition upon injection in the body, and offer
the medium is thus important. the unique opportunity to shape the gel within the diseased
F. Croisier, C. Jérôme / European Polymer Journal 49 (2013) 780–792 785

tissue used as mold or template, perfectly adjusting the


scaffold to the defect. However, the mechanical resis-
tance/strength of the aforementioned hydrogels is quite
limited and uncontrolled dissolution of the gels can occur
[74]. Besides, it is difficult to accurately control the size
of the hydrogel pores. Therefore, irreversible chemical
cross-linking of the hydrogels have been investigated.

5.1.2. Chitosan cross-linked by coordination complex


Coordinate-covalent bonds can also occur with chitosan
via metal ions as Pt (II), Pd (II), and Mo (VI) [68,69] leading
to the formation of another type of hydrogels, but less sui- 100 µm
ted for biomedical use. This gelation is depicted in Fig. 4b.
Fig. 5. SEM image of a chitosan sponge prepared by freeze-drying.
5.1.3. Chemically cross-linked chitosan hydrogels
The formation of those hydrogels occurs via covalent than the hydroxyl groups, they are mostly used for this
bonding between polymer chains (Fig. 4c). The obtained purpose since the cross-linking degree might consume
hydrogels are much more stable than the previous physi- only few of the amino groups initially present on chitosan.
cally associated hydrogels since the gelation is irreversible. Chitosan with high DD is then of interest as starting
However, this approach requires the chemical modification material.
of the primary structure of chitosan, which could alter its
initial properties, particularly if amino groups are involved 5.2. Chitosan sponges
in the reaction. In addition, the involved reaction might be
a source of contamination by toxic residual reactants or Sponges are nothing else than foams with an open
catalyst traces. porosity. These solid structures are able to absorb high
Both the amino and the hydroxyl groups of chitosan can amount of fluids (more than 20 times their dry weight),
form a variety of linkages, as amide and ester bonding, but due to their micro-porosity. They typically offer good cell
also Schiff base formation [55,75], which can lead to chito- interaction, still being soft and flexible [80].
san hydrogels formation. Small multifunctional molecules Chitosan sponges are mainly used as wound healing
or polymeric cross-linker can both be used to react with materials, as they can soak up the wound exudates, while
chitosan and induce cross-linking. Chitosan can also be helping the tissue regeneration. Chitosan sponges also find
firstly modified by activated functional groups before application in bone tissue engineering, as a filling material
inducing cross-linking by photo-induced reaction or enzy- [81].
matic catalyzed reactions [38]. These sponges are mainly obtained by freeze drying
A straightforward way to obtain irreversible chitosan (lyophilization), a simple efficient process consisting in
hydrogels is the use of dialdehyde cross-linkers, such as freezing a solution of chitosan followed by sublimation of
glyoxal and glutaraldehyde, which fastly react with the the solvent under reduced pressure (a SEM image of a
amino groups of the chitosan D-glucosamine units and, to chitosan sponge prepared by this technique is presented
a lesser extent, with the hydroxyl groups of chitosan in Fig. 5). For example, chitosan [54], chitosan/tricalcium
[62]. Tripolyphosphate, ethylene glycol, diglycidyl ether phosphate (TCP) [81,82], and chitosan/collagen sponges
and diisocyanate can also play the role of cross-linker to [83] were prepared by this technique and used as scaffolds
obtain chitosan hydrogels. However, all the aforemen- for bone regeneration. Chitosan–ZnO composite sponges
tioned cross-linkers may impart toxicity to the formed also prepared by freeze-drying [80] showed good swelling,
hydrogels, which is a major concern given the future appli- antibacterial and haemostatic activities, confirming their
cation of the hydrogels as biomaterials [76]. potential healing in wound dressing application.
Lately, a natural derived cross-linker, the genipin Cross-linked chitosan sponges loaded with a model of
(C11H14O5, isolated from Genipa Americana in 1960 and antibiotic drug – the norfloxacin – were prepared by sol-
then from Gardenia jasminoides Ellis) has been used as vent evaporation technique [84]. Fibrillar structure was
an alternative cross-linking agent [62]. This cross-linker obtained and those sponges showed promising use for
degrades more slowly than other cross-linkers (such as wound dressing application.
glutaraldehyde) and its toxicity has not yet been estab- Recently, efforts were dedicated to the use of supercrit-
lished [62,77]. Genipin can react with primary amine and ical carbon dioxide (scCO2) as a green medium to induce
thus cross-link polymers containing such amino groups porosity to chitosan scaffolds [85].
[77,78], like proteins and chitosan. The scCO2 method allows the preparation of porous
The properties of the resulting hydrogels will rely on chitosan scaffolds, suitable for cell cultures directly upon
the cross-linking density and the ratio of cross-linker mol- depressurization.
ecules to the moles of polymer repeating units [79].
It is important to note that chemical cross-linking of 6. Chitosan 2D-scaffolds for wound dressing
chitosan that involves chemical modifications of chitosan
primary amine might induce modifications of chitosan In the particular case of wound dressing, specific
properties. However, these groups being more reactive requirements have to be met to encounter efficiency. The
786 F. Croisier, C. Jérôme / European Polymer Journal 49 (2013) 780–792

wound dressing should be non-toxic and non-allergenic, chitosan film containing 87.31 mg/m2 of phosphorus –
allow gas exchanges, keep moist environment, protect NB: conductivity measurements were realized after
the wound against microbial organisms and absorb wound hydratation of the films) [94], (ii) chitosan blends with
exudates [80]. As mentioned above, chitosan sponges have minocycline hydrochloride, to prepare films that accelerate
been found to fit quite well these requirements. Neverthe- the healing of burns (reduction of the wound size) [95], (iii)
less, as far as skin tissues are concerned, more 2D-shaped chitosan/poly(vinyl alcohol)/alginate films by casting/
scaffolds, such as films and porous membranes, are supple- solvent evaporation method, in order to create a moist
mentary candidates, spreading the panel of structures environment within the framework of wound healing
available to tackle the challenges addressed in skin repair. [96], and (iv) non-adherent film of b-glucan and chitosan
[97,98].
6.1. Chitosan films Heterogeneous chemical modification of the chitosan
film can tune the surface properties of the film; for in-
Chitosan films can be easily prepared by wet casting stance, when a stearoyl group was attached to the chitosan
from chitosan salt solutions, followed by drying (typically films, they became more hydrophobic (contact angle of
using oven or infrared (IR) drying [86]). For example, Hem- pure chitosan film being 89 ± 6°, while 101 ± 4° was found
ConÒ bandage is an engineered chitosan acetate prepara- for N-stearoylchitosan film) and promoted proteins
tion designed as a haemostatic dressing [80]. adsorption. When chitosan films were reacted with succi-
nic anhydride or phthalic anhydride, it resulted in more
6.1.1. Improving properties of chitosan free-standing films hydrophilic films (contact angles of 56 ± 2° and 51 ± 7°
To improve the properties of those films, some physico- were found for N-succinylchitosan and N-phthalylchitosan
chemical processes have been tested. For examples, (i) films, respectively), which promoted lysozyme adsorption
nitrogen or argon plasma [87] treatment of chitosan mem- [99]. Interestingly, a thermo-sensitive film was prepared
branes increases the film surface roughness and improves by combining thiolated chitosan, poly(N-isopropyl acryl-
the cell adhesion and proliferation (especially for films amide) and ciprofloxacin, so that by decreasing the tem-
treated with nitrogen plasma at 20 W during 20 min), (ii) perature, the film can easily be removed from wound
ozone or UV irradiation promotes surface modification of [100].
chitosan films leading to depolymerization of chitosan Finally, incorporation of inorganic particles in these
[88], (iii) by introducing silica particles or poly(ethylene films allows synergistic effects of both materials leading
glycol) [89] into the chitosan films, their porosity can be to high performance healing. Ag nanoparticles possess
artificially modified with macro and micro-pores (pores identified antimicrobial activity, notably used in silver-
size ranging 0.5–100 lm). based wound dressings [101]. Those Ag particles can be
As far as mechanical properties are concerned, blending added to chitosan film, to further avoid microorganism
and chemical modifications of chitosan have proven effi- proliferation [102–105]. Polyphosphate procoagulant can
ciency. To improve the films ductility, chitosan can be also be incorporated to the chitosan/Ag films [24]. Beside
blended (or copolymerized) with poly(ethylene glycol) Ag, zinc oxide (ZnO) nanoparticles are non-toxic and pres-
(PEG), which results in a decrease of the modulus with ent antibacterial and good photocatalytic activities [106].
an increase of the strain at break (for 50/50 chitosan/PEO Like the Ag nanoparticles, they can be incorporated in
blended film, the decrease of the modulus was 56%, while chitosan films. Complex films containing chitosan, Ag
the increase of the strain at break was 125%, in comparison nanoparticles and ZnO particles were prepared [107]. They
to pure chitosan film) [90]. Amidation of the chitosan films possess pronounced antibacterial activity (especially the
(up to 50%) can occur with thermal treatment; it leads to film containing 0.1 wt.% Ag and 10 wt.% ZnO), thus promis-
significant strengthening of the films and to a decrease of ing application in wound dressing field.
the film solubility in aqueous media [91]. Chitosan films
prepared in presence of dialdehyde starch, that plays the 6.1.2. Chitosan thin films
role of cross-linking agent, show improved mechanical The properties of the wound dressing surface in contact
properties and better water-swelling (best values were ob- to the diseased body are particularly of importance to pro-
tained with a dialdehyde starch content of 5%, with a ten- vide efficient healing. Multilayer coatings and nanostruc-
sile strength of 113.1 MPa and an elongation at break of tured thin films have thus also been investigated in order
27%) [92]. via Michael addition reaction, chitosan/polyeth- to tune the surface properties of various medical devices.
ylene glycol diacrylate (PEGDA) blended films were devel- Two methods to design chitosan thin films in order to
oped [93], showing enhanced swelling (the maximum modify the surface of a performed substrate will be shortly
swelling was observed at a ratio of CS/PEGDA equal to described: Langmuir–Blodgett (LB), and the layer-by-layer
40/60) and good mechanical properties (the maximum val- (LBL) deposition techniques. These two processes are of
ues of tensile strength and modulus were obtained at a ra- importance since both of them allow to control parameters
tio of CS/PEGDA equal to 70/30). such as film thickness, composition, morphology, and even
Other noteworthy developments are also the prepara- roughness [18].
tion of: (i) phosphorylated chitosan films (prepared from To form a Langmuir [108] monolayer at the air–water
the reaction of orthophosphoric acid and urea in DMF), interface, a solution in an organic volatile solvent of an
which present higher ionic conductivity than normal chito- amphiphilic water-insoluble (macro)molecule is spread
san films (conductivity of pure chitosan film being equal to on the surface of water. When the organic solvent is evap-
8.3  105 S cm1, and reaching 1.2  103 S cm1 for a orated, the hydrophilic part of the amphiphile is anchored
F. Croisier, C. Jérôme / European Polymer Journal 49 (2013) 780–792 787

Fig. 6. Schematic representation of Langmuir film (a) (left) and Langmuir–Blodgett process (b) (right).

Fig. 7. Schematic representation of LBL process.

to the aqueous subphase, while the hydrophobic part is di- with a cinnamoyl chromophore were synthesized [114].
rected toward air [18] (as depicted in Fig. 6a). The Lang- Mixed films of cholesterol and chloroform-soluble O,O-di-
muir–Blodgett (LB) process [109] consists in transferring palmitoyl chitosan are an example of two-component LB
this Langmuir monolayer onto a solid support. This can films [115].
be done by immersing (or emerging) a solid support in On the other hand, the layer-by-layer (LBL) deposition
(or from) the aqueous subphase to recover the monolayer technique is based on the alternated adsorption of materi-
(Fig. 6b). By repeating the process, multilayered films can als bearing complementary charged or functional groups
be obtained. [22,112], in aqueous medium. A schematic representation
Chitosan being poorly soluble in organic solvents, of LBL process is presented in Fig. 7. A large number of mol-
chemical modification is required for this process to be ecules can be used, including synthetic and natural poly-
performed. Chitosan modified with long alkyl chains at- electrolytes [111], nanotubes [116] and biomolecules
tached to the primary hydroxyl and amino groups, was [117]. Such polyelectrolyte multilayered films find many
the first example of LB chitosan films formation [18,110], applications in the field of sensors [118], filters, but also
this derivatization making the resulting chitosan soluble as biomaterial for tissue engineering [119].
in chloroform. Derivatives of chitosan pentamers were The polycationic nature of chitosan makes this polymer
used to produce LB films [111,112], while other amphi- well-suited for LBL processes. As a consequence, LBL chito-
philic chitosans, namely, N,N-dialkyl-chitosans, were also san-based films are involved in many applications as sen-
reported [113]. All these films find application mainly in sors, drug delivery systems, haemostatic devices, bone
drug release and drug delivery systems, since the amphi- implants and wound dressings [18].
philic films are able to entrap various drugs with efficacy. Chitosan can be LBL co-deposited with several species
To facilitate the optical characterization of the LB films, on metallic devices [18]. For sensing applications, en-
amphiphilic chitosans bearing an alkyl group combined zymes, antigens, nucleic acids, carbon nanotubes, phthalo-
788 F. Croisier, C. Jérôme / European Polymer Journal 49 (2013) 780–792

cyanines, and porphyrins can be trapped by this process in during ESP, repulsive forces between charged species
a chitosan matrix that helps maintaining the biomolecule within polymer backbone arise as a consequence of the
activity. For coronary stents, chitosan/heparin LBL films application of an important electric field. This phenomenon
are particularly interesting for their anticoagulation prop- restricts the formation of continuous fibers and often leads
erties [18]. to the creation of particles [133,134].
To improve mechanical properties of chitosan LBL films, A few examples of pure electrospun chitosan fibers are
which might be especially important when deposited in reported; chitosan (nano)fibers were successfully pro-
soft substrates, chitosan can be combined in the film with duced from trifluoroacetic acid (TFA) and dichloromethane
clays such as Na-montmorillonite [120], with carbon nano- (DCM) mixtures [135,136]. It was suggested [135] that the
tubes [121], or with cellulose nanowhiskers [122]. amino groups of the chitosan form salts with TFA [137],
LBL films entirely made of polysaccharides, such as which destroy the interactions between chitosan mole-
chitosan and hyaluronic acid [123], and of chitosan and cules and facilitate the ESP process. In addition, the use
alginate [119] offer great prospect for wound dressing pur- of DCM allows improving the homogeneity of the produced
pose, as they have shown abilities to accelerate the healing. fibers. Another group used TFA in conjugation with glutar-
aldehyde, to produce cross-linked chitosan fibers [138]. Fi-
6.2. Chitosan porous nanofiber membranes nally, the use of concentrated aqueous acetic acid was
considered [139,140].
There are several ways to produce chitosan fibers; the To facilitate the electrospinning of chitosan, some
first example was reported as early as 1926 [124,125]. authors report on the use of modified chitosan such as
Fibers were notably produced using dry [126] and wet hexanoyl chitosan [141] and PEGylated chitosan [142].
spinning [127] from acetic acid solution. Lithium chloride/ Nevertheless, the easiest way to produce chitosan (nano)-
N,N-dimethylacetamide was also used as a solvent [128]. fibers is the electrospinning of chitosan as a blend with an-
In order to decrease production cost and to improve fiber other polymer. Many examples of such chitosan blend
properties, blends of chitosan with other polymers were fibers produced by ESP are reported in the literature.
also considered: sodium alginate [129], tropocollagen The electrospinning of chitosan in presence of poly(eth-
[130], cellulose, sodium hyaluronate, sodium heparin, ylene oxide) (PEO) [143–147] and ultrahigh-molecular-
sodium chondroitin sulfate [124], poly(acrylic acid) [131] weight poly(ethylene oxide) (UHMWPEO) [148] are
were thereby employed. commonly reported. A SEM image of chitosan/PEO nanofibers
In recent years, electrospinning (ESP) [132] has distin- is presented in Fig. 9. Poly(vinyl alcohol) (PVA) [149–151]
guished as a versatile technique for the preparation of can also be used to form chitosan (nano)fibers. The uses
polymer fibers from a few nanometers to microns in diam- of poly(ethylene terephtalate (PET) [152], poly(vinyl
eter, depending on the processing conditions. ESP uses a pyrrolidone) (PVP) [153] and poly(lactic acid) (PLA) [154]
high voltage to create an electrically charged jet of polymer were also reported.
solution or melt which can lead to fibers formation. Fig. 8 Chitosan/PEO nanofibers prepared by ESP exhibit cellu-
shows a schematic representation of this technique. The lar biocompatibility [133] [147]. The structure of the fiber
mats of fibers produced by this method possess high mats was found to promote the attachment of human cells,
porosity, high specific surface area, and are able to mimic while preserving their morphology and viability [147]. The
the natural extracellular matrix, which makes these mats addition of UHMWPEO to chitosan ESP solution allows
great candidates for biomaterial applications. Chitosan forming fibers ranging from less than an hundred nanome-
(nano)fibers find many applications for the preparation of ters to a few tens micrometers [148], while adding PVP al-
wound dressing and for tissue engineering [133]. lows decreasing the diameter of chitosan-based fibers
However, as chitosan is a polyelectrolyte in acidic [153]. These blends thus offer great prospects for designing
medium, its electrospinning turns out to be tricky. Indeed, tissue engineering scaffolds [133]. The joint use of PVA and
chitosan allows the preparation of fibers without beaded

10 µm

Fig. 8. Schematic representation of the electrospinning set-up. Fig. 9. SEM image of electrospun chitosan/PEO blended fibers.
F. Croisier, C. Jérôme / European Polymer Journal 49 (2013) 780–792 789

Table 1
Main advantages, disadvantages and applications of the chitosan biomaterials presented.

Biomaterial type Specifications Main advantage(s) Main disadvantage(s) Main biomedical application(s)
Hydrogels (3D)  Physically associ- Soft flexible non-toxic Not stable (uncontrolled Tissue replacements/engineering
ated (reversible) dissolution may occur) drug/growth factor delivery
Low mechanical resistance
Pore size difficult to control
 Chemically cross- Soft flexible stable May be toxic
linked controlled pore size
(irreversible)
Crosslinking may affect
chitosan intrinsic properties
Sponges (3D) High porosity May shrivel Tissue engineering (filling material)
 Free-standing Soft Low porosity Wound dressings skin substitutes
Films (2D)  Thin (LB) Material coating Laborious for the Coatings for a variety of scaffolds
construction of multilayers wound dressings skin substitutes
 Thin (LBL) Material coating Many steps
Multilayer
construction
Porous Membranes (2D) Nanofibers High porosity Coatings for a variety of scaffolds
Mimic skin ESP of pure chitosan difficult wound dressings skin substitutes
extracellular matrix

defects; chitosan/PVA nanofibers are notably used for a Acknowledgements


variety of biomedical applications, as they also proved bio-
compatible [149–151]. Chitosan/PET nanofibers present, F.C. is grateful to the Belgian ‘‘Fonds pour la Formation à
besides biocompatibility, enhanced antibacterial proper- la Recherche dans l’Industrie et dans l’Agriculture’’ (FRIA)
ties, in comparison to pure PET fibers or chitin/PET fibers for financial support. The present work has been supported
[148]. by the Science Policy Office of the Belgian Federal Govern-
The addition of another polymer to facilitate the ESP ment (IAP 7/05). CERM thanks the Walloon Region for sup-
process is actually a way to improve the properties of the porting researches on chitosan in the frame of CHITOPOL,
resulting fibers such as the mechanical properties, the bio- TARGETUM and GOCELL projects.
compatibility, and the antibacterial behavior [133]. Elec-
trospun chitosan-based nanofiber mats appear thus today
References
as an emerging biomaterial in wound dressing applications
sustained by the apparition of products such as ChitoflexÒ [1] Liu X, Ma L, Mao Z, Gao C. Chitosan-based biomaterials for tissue
on the market. repair and regeneration. Chitosan for biomaterials II. In: Jayakumar
R, Prabaharan M, Muzzarelli RAA, editors. Advances in polymer
science. Heidelberg: Springer Berlin; 2011. p. 81–127.
7. Conclusions [2] Rinaudo M. Chitin and chitosan: properties and applications. Prog
Polym Sci 2006;31:603–32.
[3] Acosta N, Jimenez C, Borau V, Heras A. Extraction and
Chitosan is a quite unique biosourced polymer charac- characterization of chitin from crustaceans. Biomass Bioenergy
terized by primary amines along the backbone. Such struc- 1993;5:145–53.
ture imparts to this polysaccharide not only highly [4] Madihally SV, Matthew HWT. Porous chitosan scaffolds for tissue
engineering. Biomaterials 1999;20:1133–42.
valuable physico-chemical properties but also particular [5] Jayakumar R, Menon D, Manzoor K, Nair SV, Tamura H. Biomedical
interactions with proteins, cells and living organisms. In applications of chitin and chitosan based nanomaterials. A short
addition, this polycation (upon protonation in acidic condi- review. Carbohydr Polym 2010;82:227–32.
[6] Venkatesan J, Kim S-K. Chitosan composites for bone tissue
tions) shows improved solubility in aqueous media which engineering – an overview. Mar Drugs 2010;8:2252–66.
allows the processing of this material in very mild condi- [7] Rane KD, Hoover DG. Production of chitosan by fungi. Food
tions in a wide variety of shapes. Soft gels and thin films, Biotechnol 1993;7:11–33.
[8] Aranaz I, Harris R, Heras A. Chitosan amphiphilic derivatives.
so as stronger sponges and nanofiber mats are thus easily Chemistry and applications. Curr Org Chem 2010;14:308–30.
made available with good control on their composition, [9] Aranaz I, Mengibar M, Harris R, Panos I, Miralles B, Acosta N, et al.
function and morphology. The main advantages and disad- Functional characterization of chitin and chitosan. Curr Chem Biol
2009;3:203–30.
vantages of the chitosan biomaterials presented in this
[10] Kannan M, Nesakumari M, Rajarathinam K, Ranjit SAJA. Production
article are pointed out in Table 1, with respect to their bio- and characterization of mushroom chitosan under solid-state
medical applications. Therefore, this polymer is very fermentation conditions. Adv Biol Res 2010;4:10–3.
[11] Wu T, Zivanovic S, Draughon FA, Sams CE. Chitin and chitosan
attractive for biomedical uses, notably in tissue engineer-
value-added products from mushroom waste. J Agricult Food Chem
ing. The wide panel of chitosan properties and processed 2004;52:7905–10.
materials gives to this biosourced polymer a quite promis- [12] Riva R, Ragelle H, des Rieux A, Duhem N, Jérôme C, Préat V. Chitosan
ing future as biomaterial as demonstrated by emerging and chitosan derivatives in drug delivery and tissue engineering.
Chitosan for biomaterials II. In: Jayakumar R, Prabaharan M,
products on the market notably in the wound dressing Muzzarelli RAA, editors. Advances in polymer science. Heidelberg:
field. Springer Berlin; 2011. p. 19–44.
790 F. Croisier, C. Jérôme / European Polymer Journal 49 (2013) 780–792

[13] Teng WL, Khor E, Tan TK, Lim LY, Tan SC. Concurrent production of [40] Funkhouser JD, Aronson Jr NN. Chitinase family GH18: evolutionary
chitin from shrimp shells and fungi. Carbohydr Res insights from the genomic history of a diverse protein family. BMC
2001;332:305–16. Evol Biol. 2007; 7:96:16 pp.
[14] Leedy M, Martin H, Norowski P, Jennings J, Haggard W, Bumgardner [41] Pangburn SH, Trescony PV, Heller J. Lysozyme degradation of
J. Use of chitosan as a bioactive implant coating for bone-implant partially deacetylated chitin, its films and hydrogels. Biomaterials
applications. Chitosan for biomaterials II. In: Jayakumar R, 1982;3:105–8.
Prabaharan M, Muzzarelli RAA, editors. Advances in polymer [42] Aiba S. Studies on chitosan: 4. Lysozymic hydrolysis of partially N-
science. Heidelberg: Springer Berlin; 2011. p. 129–65. acetylated chitosans. Int J Biol Macromol 1992;14:225–8.
[15] Nanba T, Izume M. Cosmetics containing chitosan oligosaccharides. [43] Tomihata K, Ikada Y. In vitro and in vivo degradation of films in
1987; JP 62273905 A: 12p. chitin and its deacetylated derivatives. Biomaterials
[16] Zhang Y, Zhang X-W, Li Y-M. Study on the preparation of chitosan 1997;18:567–75.
oligomer through the chitosan degraded by hydrogen peroxide. [44] Zhang H, Neau SH. In vitro degradation of chitosan by a commercial
Anhui Nongye Kexue 2011;39:13060–2. enzyme preparation: effect of molecular weight and degree of
[17] Balazs N, Sipos P. Limitations of pH-potentiometric titration for the deacetylation. Biomaterials 2001;22:1653–8.
determination of the degree of deacetylation of chitosan. Carbohydr [45] Thanou M, Verhoef JC, Junginger HE. Oral drug absorption
Res 2006;342:124–30. enhancement by chitosan and its derivatives. Adv Drug Delivery
[18] Pavinatto FJ, Caseli L, Oliveira Jr ON. Chitosan in nanostructured Rev 2001;52:117–26.
thin films. Biomacromolecules 2010;11:1897–908. [46] Wedmore I, McManus JG, Pusateri AE, Holcomb JB. A special report
[19] Craveiro AA, Craveiro AC, Pontes DF, Brasil IM, Cesar LT, Cabral MdF on the chitosan-based hemostatic dressing: experience in current
et al. Use of chitosan in clarification of beverages. 2012; BR combat operations. J Trauma 2006;60:655–8.
2010001795 A2: 11p. [47] Chatelet C, Damour O, Domard A. Influence of the degree of
[20] Takahashi T, Takayama K, Machida Y, Nagai T. Characteristics of acetylation on some biological properties of chitosan films.
polyion complexes of chitosan with sodium alginate and sodium Biomaterials 2000;22:261–8.
polyacrylate. Int J Pharm 1990;61:35–41. [48] Dutta P, Rinki K, Dutta J. Chitosan: a promising biomaterial for
[21] Kim T-H, Jiang H-L, Jere D, Park I-K, Cho M-H, Nah J-W, et al. tissue engineering scaffolds. Chitosan for biomaterials II. In:
Chemical modification of chitosan as a gene carrier in vitro and Jayakumar R, Prabaharan M, Muzzarelli RAA, editors. Advances in
in vivo. Prog Polym Sci 2007;32:726–53. polymer science. Heidelberg: Springer Berlin; 2011. p. 45–79.
[22] Decher G. Fuzzy nanoassemblies: toward layered polymeric [49] Olteanu CE, Enescu D. Chitosan involved tissue engineering and
multicomposites. Science 1997;277:1232–7. regenerative medicine. Roum Biotechnol Lett 2007;12:3217–33.
[23] Sudarshan NR, Hoover DG, Knorr D. Antibacterial action of chitosan. [50] Zhu X, Chian KS, Chan-Park MBE, Lee ST. Effect of argon-plasma
Food Biotechnol 1992;6:257–72. treatment on proliferation of human-skin-derived fibroblast on
[24] Ong S-Y, Wu J, Moochhala SM, Tan M-H, Lu J. Development of a chitosan membrane in vitro. J Biomed Mater Res, Part A
chitosan-based wound dressing with improved hemostatic and 2005;73A:264–74.
antimicrobial properties. Biomaterials 2008;29:4323–32. [51] Jagur-Grodzinski J. Biomedical applications of polymers 2001–
[25] Lehr CM, Bouwstra JA, Schacht EH, Junginger HE. In vitro evaluation 2002. e-Polym. 2003; Paper No. 12.
of mucoadhesive properties of chitosan and some other natural [52] Muzzarelli RAA, Muzzarelli C. Chitosan chemistry: relevance to the
polymers. Int J Pharm 1992;78:43–8. biomedical sciences. Adv Polym Sci 2005;186:151–209.
[26] Yang J, Tian F, Wang Z, Wang Q, Zeng Y-J, Chen S-Q. Effect of [53] Drury JL, Mooney DJ. Hydrogels for tissue engineering: scaffold
chitosan molecular weight and deacetylation degree on hemostasis. design variables and applications. Biomaterials 2003;24:4337–51.
J Biomed Mater Res, Part B 2007;84B:131–7. [54] Seol Y-J, Lee J-Y, Park Y-J, Lee Y-M, Ku Y, Rhyu I-C, et al. Chitosan
[27] Bagheri-Khoulenjani S, Taghizadeh SM, Mirzadeh H. An sponges as tissue engineering scaffolds for bone formation.
investigation on the short-term biodegradability of chitosan with Biotechnol Lett 2004;26:1037–41.
various molecular weights and degrees of deacetylation. Carbohydr [55] Berger J, Reist M, Mayer JM, Felt O, Gurny R. Structure and
Polym 2009;78:773–8. interactions in chitosan hydrogels formed by complexation or
[28] Vaarum KM, Myhr MM, Hjerde RJN, Smidsroed O. In vitro aggregation for biomedical applications. Eur J Pharm Biopharm
degradation rates of partially N-acetylated chitosans in human 2004;57:35–52.
serum. Carbohydr Res 1997;299:99–101. [56] Boucard N, Viton C, Domard A. New aspects of the formation of
[29] VandeVord PJ, Matthew HWT, DeSilva SP, Mayton L, Wu B, Wooley physical hydrogels of chitosan in a hydroalcoholic medium.
PH. Evaluation of the biocompatibility of a chitosan scaffold in Biomacromolecules 2005;6:3227–37.
mice. J Biomed Mater Res 2002;59:585–90. [57] Shu XZ, Zhu KJ. Controlled drug release properties of ionically
[30] Sashiwa H, Aiba S. Chemically modified chitin and chitosan as cross-linked chitosan beads: the influence of anion structure. Int J
biomaterials. Prog Polym Sci 2004;29:887–908. Pharm 2002;233:217–25.
[31] He P, Davis SS, Illum L. In vitro evaluation of the mucoadhesive [58] Shen EC, Wang C, Fu E, Chiang CY, Chen TT, Nieh S. Tetracycline
properties of chitosan microspheres. Int J Pharm 1998;166: release from tripolyphosphate–chitosan cross-linked sponge: a
75–88. preliminary in vitro study. J Periodontal Res 2008;43:642–8.
[32] Park P-J, Je J-Y, Jung W-K, Ahn C-B, Kim S-K. Anticoagulant activity [59] Yin YJ, Yao KD, Cheng GX, Ma JB. Properties of polyelectrolyte
of heterochitosans and their oligosaccharide sulfates. Eur Food Res complex films of chitosan and gelatin. Polym Int 1999;48:429–32.
Technol 2004;219:529–33. [60] Mao JS, Cui YL, Wang XH, Sun Y, Yin YJ, Zhao HM, et al. A
[33] Rao SB, Sharma CP. Use of chitosan as a biomaterial: studies on preliminary study on chitosan and gelatin polyelectrolyte complex
its safety and hemostatic potential. J Biomed Mater Res 1997;34: cytocompatibility by cell cycle and apoptosis analysis. Biomaterials
21–8. 2004;25:3973–81.
[34] Klokkevold PR, Fukayama H, Sung EC, Bertolami CN. The effect of [61] Jiang T, Zhang Z, Zhou Y, Liu Y, Wang Z, Tong H, et al. Surface
chitosan (poly-N-acetyl glucosamine) on lingual hemostasis in functionalization of titanium with chitosan/gelatin via
heparinized rabbits. J Oral Maxillofac Surg 1999;57:49–52. electrophoretic deposition: characterization and cell behavior.
[35] Smith J, Wood E, Dornish M. Effect of chitosan on Epithelial Cell Biomacromolecules 2010;11:1254–60.
Tight Junctions. Pharm Res 2004;21:43–9. [62] Djerassi C, Gray JD, Kinci FA. Naturally occurring oxygen
[36] Chung Y-C, Chen C-Y. Antibacterial characteristics and activity of heterocycles. IX. Isolation and characterization of genipin. J Org
acid-soluble chitosan. Bioresour Technol 2008;99:2806–14. Chem 1960;25:2174–7 [Francesko A, Tzanov T. Chitin, chitosan and
[37] Okamoto Y, Kawakami K, Miyatake K, Morimoto M, Shigemasa Y, derivatives for wound healing and tissue engineering. Adv Biochem
Minami S. Analgesic effects of chitin and chitosan. Carbohydr Eng/Biotechnol 2011; 125: 1–27].
Polym 2002;49:249–52. [63] Sezer AD, Cevher E, Hatipoglu F, Ogurtan Z, Bas AL, Akbuga J.
[38] Dash M, Chiellini F, Ottenbrite RM, Chiellini E. Chitosan – a versatile Preparation of fucoidan–chitosan hydrogel and its application as
semi-synthetic polymer in biomedical applications. Prog Polym Sci burn healing accelerator on rabbits. Biol Pharm Bull
2011;36:981–1014. 2008;31:2326–33.
[39] Kean T, Thanou M. Chitin and chitosan: sources, production and [64] Boddohi S, Moore N, Johnson PA, Kipper MJ. Polysaccharide-based
medical applications. In: Williams PA, editor. Renewable resources polyelectrolyte complex nanoparticles from chitosan, heparin, and
for functional polymers and biomaterials. Polysaccharides, proteins hyaluronan. Biomacromolecules 2009;10:1402–9.
and polyesters. 1st ed. London: Royal Society of, Chemistry; 2011. [65] Il’ina AV, Varlamov VP. Chitosan-based polyelectrolyte complexes:
p. 292–318. a review. Appl Biochem Microbiol 2005;41:5–11.
F. Croisier, C. Jérôme / European Polymer Journal 49 (2013) 780–792 791

[66] Denuziere A, Ferrier D, Damour O, Domard A. Chitosan– [92] Tang R, Du Y, Fan L. Dialdehyde starch-crosslinked chitosan films
chondroitin sulfate and chitosan–hyaluronate polyelectrolyte and their antimicrobial effects. J Polym Sci Part B: Polym Phys
complexes: biological properties. Biomaterials 1998;19: 2003;41:993–7.
1275–85. [93] Zhang X, Yang D, Nie J. Chitosan/polyethylene glycol diacrylate
[67] de Oliveira HCL, Fonseca JLC, Pereira MR. Chitosan-poly(acrylic films as potential wound dressing material. Int J Biol Macromol
acid) polyelectrolyte complex membranes: preparation, 2008;43:456–62.
characterization and permeability studies. J Biomater Sci Polym [94] Wan Y, Creber KAM, Peppley B, Bui VT. Synthesis, characterization
Ed 2008;19:143–60. and ionic conductive properties of phosphorylated chitosan
[68] Brack HP, Tirmizi SA, Risen Jr WM. A spectroscopic and viscometric membranes. Macromol Chem Phys 2003;204:850–8.
study of the metal ion-induced gelation of the biopolymer chitosan. [95] Aoyagi S, Onishi H, Machida Y. Novel chitosan wound dressing
Polymer 1997;38:2351–62. loaded with minocycline for the treatment of severe burn wounds.
[69] Dambies L, Vincent T, Domard A, Guibal E. Preparation of chitosan Int J Pharm 2007;330:138–45.
gel beads by ionotropic molybdate gelation. Biomacromolecules [96] Pei HN, Chen XG, Li Y, Zhou HY. Characterization and ornidazole
2001;2:1198–205. release in vitro of a novel composite film prepared with chitosan/
[70] Ladet S, David L, Domard A. Multi-membrane hydrogels. Nature poly(vinyl alcohol)/alginate. J Biomed Mater Res Part A
2008;452:76–9. 2008;85A:566–72.
[71] Cascone MG, Maltinti S, Barbani N, Laus M. Effect of chitosan and [97] Kofuji K, Huang Y, Tsubaki K, Kokido F, Nishikawa K, Isobe T, et al.
dextran on the properties of poly(vinyl alcohol) hydrogels. J Mater Preparation and evaluation of a novel wound dressing sheet
Sci: Mater Med 1999;10:431–5. comprised of b-glucan–chitosan complex. React Funct Polym
[72] Chenite A, Chaput C, Wang D, Combes C, Buschmann MD, 2010;70:784–9.
Hoemann CD, et al. Novel injectable neutral solutions of [98] Xu H, Ma L, Shi H, Gao C, Han C. Chitosan-hyaluronic acid hybrid
chitosan form biodegradable gels in situ. Biomaterials film as a novel wound dressing: in vitro and in vivo studies. Polym
2000;21:2155–61. Adv Technol 2007;18:869–75.
[73] Bhattarai N, Ramay HR, Gunn J, Matsen FA, Zhang M. PEG-grafted [99] Tangpasuthadol V, Pongchaisirikul N, Hoven VP. Surface
chitosan as an injectable thermosensitive hydrogel for sustained modification of chitosan films. Effects of hydrophobicity on
protein release. J Controlled Release 2005;103:609–24. protein adsorption. Carbohydr Res 2003;338:937–42.
[74] Peppas N, editor. Hydrogels in medicine and pharmacy, vol. 1. [100] Radhakumary C, Antonty M, Sreenivasan K. Drug loaded
Fundamentals: CRC Press; 1986. thermoresponsive and cytocompatible chitosan based hydrogel as
[75] Hennink WE, van NCF. Novel crosslinking methods to design a potential wound dressing. Carbohydr Polym 2011;83:705–13.
hydrogels. Adv Drug Delivery Rev 2002;54:13–36. [101] Rai M, Yadav A, Gade A. Silver nanoparticles as a new generation of
[76] Leung H-W. Ecotoxicology of glutaraldehyde: review of antimicrobials. Biotechnol Adv 2009;27:76–83.
environmental fate and effects studies. Ecotoxicol Environ Saf [102] Lu S, Gao W, Gu HY. Construction, application and biosafety of
2001;49:26–39. silver nanocrystalline chitosan wound dressing. Burns
[77] Sung HW, Huang RN, Huang LL, Tsai CC. In vitro evaluation of 2008;34:623–8.
cytotoxicity of a naturally occurring cross-linking reagent for [103] Mi F-L, Wu Y-B, Shyu S-S, Chao A-C, Lai J-Y, Su C-C. Asymmetric
biological tissue fixation. J Biomater Sci Polym Ed 1999;10:63–78. chitosan membranes prepared by dry/wet phase separation: a new
[78] Sung H-W, Liang IL, Chen C-N, Huang R-N, Liang H-F. Stability of a type of wound dressing for controlled antibacterial release. J
biological tissue fixed with a naturally occurring crosslinking agent Membr Sci 2003;212:237–54.
(genipin). J Biomed Mater Res 2001;55:538–46. [104] Thomas V, Yallapu MM, Sreedhar B, Bajpai SK. Fabrication,
[79] Park K, Shalaby WSE, Park H. Biodegradable hydrogels for drug characterization of chitosan/nanosilver film and its potential
delivery. Technomic; 1993. antibacterial application. J Biomater Sci Polym Ed 2009;20:
[80] Jayakumar R, Prabaharan M, Sudheesh KPT, Nair SV, Tamura H. 2129–44.
Biomaterials based on chitin and chitosan in wound dressing [105] Vimala K, Mohan YM, Sivudu KS, Varaprasad K, Ravindra S, Reddy
applications. Biotechnol Adv 2011;29:322–37. NN, et al. Fabrication of porous chitosan films impregnated with
[81] Costa-Pinto AR, Reis RL, Neves NM. Scaffolds based bone tissue silver nanoparticles: a facile approach for superior antibacterial
engineering: the role of chitosan. Tissue Eng Part B application. Colloids Surf B 2010;76:248–58.
2011;17:331–47. [106] Cohen ML. The theory of real materials. Ann Rev Mater Sci
[82] Lee Y-M, Park Y-J, Lee S-J, Ku Y, Han S-B, Choi S-M, et al. Tissue 2000;30:1–26.
engineered bone formation using chitosan/tricalcium phosphate [107] Li L-H, Deng J-C, Deng H-R, Liu Z-L, Li X-L. Preparation,
sponges. J Periodontol 2000;71:410–7. characterization and antimicrobial activities of chitosan/Ag/ZnO
[83] Arpornmaeklong P, Pripatnanont P, Suwatwirote N. Properties of blend films. Chem Eng J 2010;160:378–82.
chitosan–collagen sponges and osteogenic differentiation of rat- [108] Langmuir I. Constitution and fundamental properties of solids and
bone-marrow stromal cells. Int J Oral Maxillofac Surg liquids. II. Liquids. J Am Chem Soc 1917;39:1848–906.
2008;37:357–66. [109] Blodgett KB. Films built by depositing successive unimolecular
[84] Denkbas EB, Ozturk E, Ozdemir N, Kececi K, Agalar C. Norfloxacin- layers on a solid surface. J Am Chem Soc 1935;57:1007–22.
loaded chitosan sponges as wound dressing material. J Biomater [110] Nishimura S, Miura Y, Ren L, Sato M, Yamagishi A, Nishi N, et al. An
Appl 2004;18:291–303. efficient method for the syntheses of novel amphiphilic
[85] Rinki K, Tripathi S, Dutta PK, Dutta J, Hunt AJ, Macquarrie DJ, et al. polysaccharides by regio- and thermoselective modifications of
Direct chitosan scaffold formation via chitin whiskers by a chitosan. Chem Lett 1993:1623–6.
supercritical carbon dioxide method: a green approach. J Mater [111] Fendler JH. Self-assembled nanostructured materials. Chem Mater
Chem 2009;19:8651–5. 1996;8:1616–24.
[86] Khan TA, Peh KK, Ch’ng HS. Mechanical, bioadhesive strength and [112] Hammond PT. Form and function in multilayer assembly: new
biological evaluations of chitosan films for wound dressing. J Pharm applications at the nanoscale. Adv Mater 2004;16:1271–93.
Pharm Sci 2000;3:303–11. [113] Li M, Su S, Xin M, Liao Y. Relationship between N,N-dialkyl chitosan
[87] Silva SS, Luna SM, Gomes ME, Benesch J, Pashkuleva I, Mano JF, et al. monolayer and corresponding vesicle. J Colloid Interface Sci
Plasma surface modification of chitosan membranes: 2007;311:285–8.
characterization and preliminary cell response studies. Macromol [114] Wu Y, Hisada K, Maeda S, Sasaki T, Sakurai K. Fabrication and
Biosci 2008;8:568–76. structural characterization of the Langmuir–Blodgett films from a
[88] Matienzo LJ, Winnacker SK. Dry processes for surface modification new chitosan derivative containing cinnamate chromophores.
of a biopolymer: chitosan. Macromol Mater Eng 2002;287: Carbohydr Polym 2007;68:766–72.
871–80. [115] Tong Y, Wang S, Xu J, Chua B, He C. Synthesis of O,O0 -dipalmitoyl
[89] Clasen C, Wilhelms T, Kulicke WM. Formation and characterization chitosan and its amphiphilic properties and capability of
of chitosan membranes. Biomacromolecules 2006;7:3210–22. cholesterol absorption. Carbohydr Polym 2005;60:229–33.
[90] Kolhe P, Kannan RM. Improvement in ductility of chitosan through [116] Kim SN, Rusling JF, Papadimitrakopoulos F. Carbon nanotubes for
blending and copolymerization with PEG: FTIR investigation of electronic and electrochemical detection of biomolecules. Adv
molecular interactions. Biomacromolecules 2003;4:173–80. Mater 2007;19:3214–28.
[91] Zotkin MA, Vikhoreva GA, Kechek’yan AS. Thermal modification of [117] He J-A, Samuelson L, Li L, Kumar J, Tripathy SK. Oriented
chitosan films in the form of salts with various acids. Vysokomol bacteriorhodopsin/polycation multilayers by electrostatic layer-
Soedin Ser A Ser B 2004;46:359–63. by-layer assembly. Langmuir 1998;14:1674–9.
792 F. Croisier, C. Jérôme / European Polymer Journal 49 (2013) 780–792

[118] Lee S-H, Kumar J, Tripathy SK. Thin film optical sensors employing [145] Matsumoto H, Yako H, Minagawa M, Tanioka A. Characterization
polyelectrolyte assembly. Langmuir 2000;16:10482–9. of chitosan nanofiber fabric by electrospray deposition:
[119] Wittmer CR, Phelps JA, Lepus CM, Saltzman WM, Harding MJ, Van electrokinetic and adsorption behavior. J Colloid Interface Sci
TPR. Multilayer nanofilms as substrates for hepatocellular 2007;310:678–81.
applications. Biomaterials 2008;29:4082–90. [146] Ojha SS, Stevens DR, Hoffman TJ, Stano K, Klossner R, Scott MC,
[120] Darder M, Colilla M, Ruiz-Hitzky E. Biopolymer-clay et al. Fabrication and characterization of electrospun chitosan
nanocomposites based on chitosan intercalated in nanofibers formed via templating with polyethylene oxide.
montmorillonite. Chem Mater 2003;15:3774–80. Biomacromolecules 2008;9:2523–9.
[121] Wang S-F, Shen L, Zhang W-D, Tong Y-J. Preparation and [147] Bhattarai N, Edmondson D, Veiseh O, Matsen FA, Zhang M.
mechanical properties of chitosan/carbon nanotubes composites. Electrospun chitosan-based nanofibers and their cellular
Biomacromolecules 2005;6:3067–72. compatibility. Biomaterials 2005;26:6176–84.
[122] Liu Y, Hu N. Enhanced DNA oxidative damage by acrylonitrile and [148] Zhang YZ, Su B, Ramakrishna S, Lim CT. Chitosan nanofibers from
protecting DNA from damage with an outer catalase layer: an an easily electrospinnable UHMWPEO-doped chitosan solution
electrochemical study. Electroanalysis 2008;20:2671–6. system. Biomacromolecules 2008;9:136–41.
[123] Salomaki M, Kankare J. Influence of synthetic polyelectrolytes on [149] Huang X-J, Ge D, Xu Z-K. Preparation and characterization of stable
the growth and properties of Hyaluronan–Chitosan multilayers. chitosan nanofibrous membrane for lipase immobilization. Eur
Biomacromolecules 2009;10:294–301. Polym J 2007;43:3710–8.
[124] Pillai CKS, Paul W, Sharma CP. Chitin and chitosan polymers: [150] Duan B, Yuan X, Zhu Y, Zhang Y, Li X, Zhang Y, et al. A nanofibrous
chemistry, solubility and fiber formation. Prog Polym Sci composite membrane of PLGA-chitosan/PVA prepared by
2009;34:641–78. electrospinning. Eur Polym J 2006;42:2013–22.
[125] Kunike G. Chitin and chitosan. J Soc Dyers Colorists 1926;42:24. [151] Li L, Hsieh Y-L. Chitosan bicomponent nanofibers and nanoporous
[126] Rigby GW. Process for the preparation of films and filaments and fibers. Carbohydr Res 2006;341:374–81.
products thereof. 1936; US 2040880: p. 4. [152] Jung K-H, Huh M-W, Meng W, Yuan J, Hyun SH, Bae J-S, et al.
[127] East GC, Qin Y. Wet spinning of chitosan and the acetylation of Preparation and antibacterial activity of PET/chitosan nanofibrous
chitosan fibers. J Appl Polym Sci 1993;50:1773–9. mats using an electrospinning technique. J Appl Polym Sci
[128] Agboh OC, Qin Y. Chitin and chitosan fibers. Polym Adv Technol 2007;105:2816–23.
1997;8:355–65. [153] Ignatova M, Manolova N, Rashkov I. Novel antibacterial fibers of
[129] Tamura H, Tsuruta Y, Tokura S. Preparation of chitosan-coated quaternized chitosan and poly(vinyl pyrrolidone) prepared by
alginate filament. Mater Sci Eng C 2002;C20:143–7. electrospinning. Eur Polym J 2007;43:1112–22.
[130] Hirano S, Zhang M, Nakagawa M, Miyata T. Wet spun chitosan– [154] Peesan M, Rujiravanit R, Supaphol P. Electrospinning of hexanoyl
collagen fibers, their chemical N-modifications, and blood chitosan/polylactide blends. J Biomater Sci Polym Ed
compatibility. Biomaterials 2000;21:997–1003. 2006;17:547–65.
[131] Ohkawa K, Ando M, Shirakabe Y, Takahashi Y, Yamada M, Shirai H,
et al. Preparing chitosan-poly(acrylic acid) composite fibers by self-
assembly at an aqueous solution interface. Text Res J
2002;72:120–4. Florence Croisier received her master’s degree
[132] Formhals A. Artificial threads and method of producing same. in chemical sciences from the University of
1940; US 2187306. p. 4. Liège (ULg, Belgium) in 2007. She is currently
[133] Jayakumar R, Prabaharan M, Nair SV, Tamura H. Novel chitin and finishing her Ph.D. under the supervision of
chitosan nanofibers in biomedical applications. Biotechnol Adv Professor Christine Jérôme in the Center for
2010;28:142–50. Education and Research on Macromolecules
[134] Min B-M, Lee SW, Lim JN, You Y, Lee TS, Kang PH, et al. Chitin and (ULg, Belgium). Her research focuses on the
chitosan nanofibers: electrospinning of chitin and deacetylation of preparation of chitosan-based nanofibers
chitin nanofibers. Polymer 2004;45:7137–42. with multilayered structure, using a combi-
[135] Ohkawa K, Cha D, Kim H, Nishida A, Yamamoto H. Electrospinning nation of electrospinning and layer-by-layer
of chitosan. Macromol Rapid Commun 2004;25:1600–5. deposition techniques.
[136] Sangsanoh P, Supaphol P. Stability improvement of electrospun
chitosan nanofibrous membranes in neutral or weak basic aqueous
solutions. Biomacromolecules 2006;7:2710–4.
[137] Hasegawa M, Isogai A, Onabe F, Usuda M, Atalla RH.
Characterization of cellulose-Chitosan blend films. J Appl Polym
Sci 1992;45:1873–9. Christine Jérôme completed her PhD on con-
[138] Schiffman JD, Schauer CL. Cross-linking chitosan nanofibers. jugated polymers for aqueous waste treat-
Biomacromolecules 2007;8:594–601. ments in 1998 at the University of Liege,
[139] Geng X, Kwon O-H, Jang J. Electrospinning of chitosan dissolved in Belgium and then worked as a post-dosctoral
concentrated acetic acid solution. Biomaterials 2005;26:5427–32. researcher on developing the electrografting
[140] Vrieze S, Westbroek P, Camp T, Langenhove L. Electrospinning of process of acrylates. In 2000, she joined the
chitosan nanofibrous structures: feasibility study. J Mater Sci University of Ulm in Germany as a recipient of
2007;42:8029–34. the Humboldt scholarship and studied the
[141] Neamnark A, Rujiravanit R, Supaphol P. Electrospinning of synthesis of functional magnetic nanohybrids.
hexanoyl chitosan. Carbohydr Polym 2006;66:298–305.
She returned to the University of Liege in
[142] Du J, Hsieh Y-L. PEGylation of chitosan for improved solubility and
2001 as Research Associate of the National
fiber formation via electrospinning. Cellulose 2007;14:543–52.
Foundation of the Scientific Research in the
[143] Spasova M, Manolova N, Paneva D, Rashkov I. Preparation of
chitosan-containing nanofibers by electrospinning of chitosan/ group of Professor R. Jérôme, where she became Professor in 2006 and
poly(ethylene oxide) blend solutions. e-Polym; 2004 [No pp. director of the Center for Education and Research on Macromolecules in
given]. 2007. Today full Professor, her research interests include biosourced
[144] Duan B, Dong C, Yuan X, Yao K. Electrospinning of chitosan polymers, functional macromolecular systems and advanced biomateri-
solutions in acetic acid with poly(ethylene oxide). J Biomater Sci als.
Polym Ed 2004;15:797–811.

You might also like