2.213. Chitosan

17
2.213. Chitosan M A Barbosa, A P Pe ˆgo, and I F Amaral, Universidade do Porto, Porto, Portugal ã 2011 Elsevier Ltd. All rights reserved. 2.213.1. Sources, Analysis, and Properties 222 2.213.1.1. Chemical Structure 222 2.213.1.2. Solution Properties 222 2.213.1.3. Chitosan Preparation: Chitin Isolation and N-deacetylation 223 2.213.1.4. Chitosan Characterization 223 2.213.1.4.1. Degree of acetylation 223 2.213.1.4.2. Molecular weight 224 2.213.1.5. General Aspects of Biological Behavior 225 2.213.1.5.1. Biocompatibility 225 2.213.1.5.2. Cytocompatibility 225 2.213.1.5.3. Bacteriostatic and fungostatic properties 225 2.213.1.5.4. Enzymatic degradation 226 2.213.1.5.5. Immunoadjuvancy 226 2.213.1.5.6. Hemostatic and blood clotting properties 226 2.213.1.5.7. Cell-binding properties 226 2.213.1.5.8. Wound-healing properties 226 2.213.1.5.9. Bone-healing properties 226 2.213.1.6. Chitosan Functionalization 227 2.213.2. Processing 227 2.213.2.1. Films and Porous Scaffolds (Freeze-Drying and Freeze-Gelling) 227 2.213.2.2. Nanofibers 227 2.213.2.3. Polyelectrolyte Complexes 228 2.213.2.4. Micro- and Nanoparticles 229 2.213.2.5. Cross-linking 229 2.213.3. Biomedical Applications 229 2.213.3.1. Wound Management 229 2.213.3.2. Tissue Repair and Regeneration 230 2.213.3.3. Delivery of Therapeutic Agents 232 2.213.3.4. Other Applications 234 2.213.4. Future Prospects 235 References 235 Glossary Coacervation The process that results in the aggregation of molecules or colloidal particles under the action of electrostatic attractive forces. Degree of acetylation (DA) Molar fraction of N-acetylated units in chitin/chitosan. Electrospinning Technique used to produce nanofibers, based on the application of a sufficiently high voltage between a needle and a metallic collector, resulting in a very thin jet of fluid which is projected against a collector. Endotoxin A toxin of internal origin. Endotoxins should be absent from chitosan used for biomedical applications. Freeze-drying (of chitosan) Polymer solutions are frozen to temperatures that cause the formation of ice crystals, which are removed by sublimation under vacuum, producing a porous structure. Freeze-gelling (of chitosan) A method alternative to freeze-drying to produce 3D-scaffolds. The method is based on freezing and subsequent extraction of the solution-rich phase by a nonsolvent for the polymer, while the polymer-rich phase is gelled under the action of a neutralizing agent. Glycosaminoglycans A gel-forming repeating disaccharide units of the extracellular matrix. Neuroma A growth or tumor of nerve tissue. Polycation A macromolecule with many positively charged groups. Polyelectrolyte complexes Self-assembled structures formed by reacting two oppositely charged polyelectrolytes in an aqueous solution. Proteoglycans A constituent of the extracellular matrix resulting from the association of a protein and glycosaminoglycans. 221

description

chitosan

Transcript of 2.213. Chitosan

  • 2.213. Chitosanto, Porto, Portugal

    2. 22. 22. 22. 32. 32.2 32.2 42. 52.2 52.2 52.2 52.2 62.2 62.2 62.2 62.213.1.5.8. Wound-healing properties 2262.213.1.5.9. Bone-healing properties 226

    to temperatures that cause the formation of ice crystals, resulting from the association of a protein andwhich are removed by sublimation under vacuum,

    producing a porous structure.

    glycosaminoglycans.2.213.1.6. Chitosan Functionalization 2272.213.2. Processing 2272.213.2.1. Films and Porous Scaffolds (Freeze-Drying and Freeze-Gelling) 2272.213.2.2. Nanofibers 2272.213.2.3. Polyelectrolyte Complexes 2282.213.2.4. Micro- and Nanoparticles 2292.213.2.5. Cross-linking 2292.213.3. Biomedical Applications 2292.213.3.1. Wound Management 2292.213.3.2. Tissue Repair and Regeneration 2302.213.3.3. Delivery of Therapeutic Agents 2322.213.3.4. Other Applications 2342.213.4. Future Prospects 235References 235

    GlossaryCoacervation The process that results in the aggregation of

    molecules or colloidal particles under the action of

    electrostatic attractive forces.

    Degree of acetylation (DA) Molar fraction of N-acetylated

    units in chitin/chitosan.

    Electrospinning Technique used to produce

    nanofibers, based on the application of a sufficiently high

    voltage between a needle and a metallic collector, resulting

    in a very thin jet of fluid which is projected against a

    collector.

    Endotoxin A toxin of internal origin. Endotoxins

    should be absent from chitosan used for biomedical

    applications.

    Freeze-drying (of chitosan) Polymer solutions are frozen

    Freeze-gelling (of chitosan) A method alternative to

    freeze-drying to produce 3D-scaffolds. The method

    is based on freezing and subsequent extraction of the

    solution-rich phase by a nonsolvent for the polymer, while

    the polymer-rich phase is gelled under the action of a

    neutralizing agent.

    Glycosaminoglycans A gel-forming repeating disaccharide

    units of the extracellular matrix.

    Neuroma A growth or tumor of nerve tissue.

    Polycation A macromolecule with many positively charged

    groups.

    Polyelectrolyte complexes Self-assembled structures

    formed by reacting two oppositely charged polyelectrolytes

    in an aqueous solution.

    Proteoglycans A constituent of the extracellular matrix 2011 Elsevier Ltd. All rights reserved.

    213.1. Sources, Analysis, and Properties 22213.1.1. Chemical Structure 22213.1.2. Solution Properties 22213.1.3. Chitosan Preparation: Chitin Isolation and N-deacetylation 22213.1.4. Chitosan Characterization 2213.1.4.1. Degree of acetylation 2213.1.4.2. Molecular weight 22213.1.5. General Aspects of Biological Behavior 2213.1.5.1. Biocompatibility 2213.1.5.2. Cytocompatibility 2213.1.5.3. Bacteriostatic and fungostatic properties 2213.1.5.4. Enzymatic degradation 2213.1.5.5. Immunoadjuvancy 2213.1.5.6. Hemostatic and blood clotting properties 22M A Barbosa, A P Pego, and I F Amaral, Universidade do Por

    13.1.5.7. Cell-binding properties 22221

  • AbbreviationsA Absorbance

    DA Degree of acetylation

    EC Endothelial cells

    ECM Extracellular matrix

    FN Fibronectin

    FT-IR Fourier transform infrared spectroscopy

    GAG Glycosaminoglycan

    H&E Hematoxylin and Eosin

    lobster, and shrimp shells, adjacent sheets have opposite direc-

    tions, and thus it has an antiparallel chain arrangement. In

    b- h he squidge

    it

    sh

    ad

    cosamine units by acid, the electrostatic repulsions between

    NH3 groups lead to the destruction of interchain attractive

    interactions, such as hydrogen bonds and hydrophobic inter-

    t pH lower

    a

    -

    -

    h

    -

    s

    e

    e

    f

    e

    c

    s

    f

    FiD-b-

    222 Materials of Biological Originnus Loligo, adjacent sheets have the same direction, and thus

    has a parallel chain arrangement. In g-chitin, every thirdeet has the opposite direction to the previous two sheets. In

    dition to intrasheet interchain hydrogen bonds, a-chitin also

    OO

    NHRHO

    OO

    HO

    O

    NHRCH2OH

    CH2OH

    gure 1 Chemical structure of chitosan, a linear copolymer ofglucosamine (RH) and N-acetyl D-glucosamine (R COCH3) in a(14) linkage. Glucosamine is the predominant repeating unit.chitin, w ich is the form occurring in the pen of tthan its pKa, which may range from 6.5 to 7, chitosan ispolycation and at pH 4.0 and below, it is completely proto

    nated.6 Chitosan solubility depends on chitosan charge den

    sity, which is tightly connected with structural parameters suc

    as DA, chain length, and distribution of N-acetylated glucos

    amine units, as well as on environmental parameters, such a

    pH, ionic strength, and dielectric constant of the media.7 Th

    solubility range increases on increasing the DA, due to th

    increase of the steric hindrance related to the increase o

    the number of the acetyl groups, together with the increas

    of the intrinsic pKa. According to Sorlier et al.,6 the intrinsi

    pKa of chitosan increases from 6.46 to 6.8 as the DA increase

    from 5% to 35%, respectively, revealing an increase oactions, and consequently to chitosan solubility. AHA Hyaluronic acid

    HLC Human-like collagen

    IVD Intervertebral disc

    LbL Layer-by-layer

    Mn Number average molecular weight

    2.213.1. Sources, Analysis, and Properties

    2.213.1.1. Chemical Structure

    Chitosan is a linear copolymer of D-glucosamine and N-acetyl-

    D-glucosamine in a b-(14) linkage, in which glucosamine isthe predominant repeating unit (Figure 1). Chitosan itself may

    be found in the mycelia of certain fungi in association with

    other polysaccharides, but is mostly obtained by deacetylation

    of chitin. Chitin is the second most abundant polysaccharide

    in nature after cellulose, occurring in the cell walls of certain

    fungi1 and yeasts, in plants as the equivalent to cellulose, and

    in many invertebrate groups such as mollusks and arthropods

    as the fibrous support of the inorganic mineral phase of their

    exoskeleton, as an alternative to collagen.1 Chitin is a high

    molecular weight crystalline polysaccharide, which is theoreti-

    cally comprised entirely of N-acetylated D-glucosamine units.

    Naturally occurring chitin, however, is mostly present as a

    copolymer, containing different proportions ofN-glucosamine

    units, dependent on the source.2 In chitin, the chains are

    arranged in sheets or stacks, the chains of each sheet having

    the same direction and being bonded through intrasheet

    hydrogen bonds between two adjacent chains. Naturally occur-

    ring chitins are found in three polymorphic forms, a-, b-, andg-chitin, which differ in the arrangement of chains within thecrystalline regions. In a-chitin, which is the one found in crab,MSC Mesenchymal stem cells

    Mw Weight average molecular weight

    NMR Nuclear magnetic resonance

    PDGF Platelet-derived growth factor

    PECs Polyelectrolyte complexes

    PEO Poly(ethylene oxide)

    PLGA Poly(lactic-co-glycolic acid)

    PLLA Poly(L-lactic acid)

    SEC Size exclusion chromatography

    SEM Scanning electron microscopy

    TCP Tricalcium phosphate

    TGF-b1 Transforming growth factor beta 1

    g-PGA Gamma-poly(glutamic acid)

    presents hydrogen bonds between adjacent chains. These inter-

    sheet bondings are responsible for the lack of swelling in water

    of a-chitin, whereas b-chitin swells readily in water and formshydrates.2 Chitosan is also crystalline, but as compared to

    chitin, presents a longer distance between adjacent chains

    belonging to the same sheet, due to the removal of theN-acetyl

    groups during the conversion from chitin to chitosan, which

    hold together adjacent chains through C(2)NHOC(7)hydrogen bonds.2 Instant differentiation between chitin and

    chitosan can be made based on their solubility. While chitin is

    soluble in N,N-dimethylacetamide (DMAc) in the presence of

    510% (w/v) lithium chloride and insoluble in dilute acid

    solutions, the reverse is true for chitosan.2,3 In chitin/chitosan

    terminology, the molar fraction of N-acetylated units is termed

    the degree of acetylation (DA), expressed in percentage,

    or fraction of N-acetylated units (FA).4,5 Since a DA around or

    lower than 50% is usually required for chitosan solubility in

    dilute acidic solutions, the term chitosan is applied both to

    fully-deacetylated chitin and partially deacetylated chitin with

    DAs 50%.

    2.213.1.2. Solution Properties

    Chitosan is neither soluble in water nor in organic solvents.

    However, after protonation of amine functionalities from glu-

  • Chitosan 223ried out at as low temperature as possible, under inert atmo-

    sphere, such as nitrogen or argon, or in the presence of oxygen

    scavengers or reducing agents, such as NaBH4. As chitin is not

    soluble in such systems, deacetylation occurs under heteroge-

    neous conditions. During deacetylation of a-chitin, NaOH actsinitially on the amorphous regions of the polymer, and only

    afterwards on the crystalline regions. Heterogeneous deacetyla-

    tion leads therefore to a block distribution of acetylated units,

    rather than a random distribution of the same. As a result, the

    characteristic infrared (IR) bands attributed to crystallinizationcationicity of amine functionalities on increasing the DA. As a

    result, chitosans with DAs in the range of 4555% are water-

    soluble, providing that the N-acetylated units are randomly

    distributed. In the presence of high ionic strengths, solubility

    is reduced. The high concentration of protons leads to the

    screening of the electrostatic interactions occurring between

    polymeric chains, with subsequent establishment of chain

    interactions and polymer precipitation. As a result, chitosan

    is not soluble in strong acids such as hydrochloric acid solu-

    tions with molarities higher than 0.1M.7

    2.213.1.3. Chitosan Preparation: Chitin Isolation andN-deacetylation

    Commercially available chitin is most commonly prepared

    from the exoskeletons of crab, shrimp, and prawn, obtained

    as waste from the seafood processing industry. In these, chitin

    is tightly associated with proteins, inorganic material (mainly

    CaCO3), pigments, and lipids. Deproteinization and deminer-

    alization are generally carried out by treatment with 12M

    NaOH at 70 C or higher temperature, and 1.25M HCl atroom temperature, respectively, deproteinization being usually

    done prior to demineralization. Both treatments may lead to

    the cleavage of chitin polymeric chains. In this sense, a number

    of alternative methods have been proposed in order to mini-

    mize the hydrolysis of glycosidic linkages during chitin extrac-

    tion, including the use of proteolytic enzymes to remove

    protein and EDTA to remove mineral. Finally, the pigments

    present in the exoskeletons of crustaceans can be extracted with

    ethanol, acetone, or oxidizing agents such as KMnO4.2 The

    preparation of squid chitin, although similar, occurs under

    milder conditions, as b-chitin is composed exclusively of chitinand proteins, with only traces of metal salts.8

    Deacetylation may be carried out under acid or basic

    conditions, but basic conditions are preferred, due to the sus-

    ceptibility of chitin glycosidic linkages to acid hydrolysis. The

    deacetylation of a-chitin is usually carried out using strongaqueous bases at 90150 C for a few hours, to produce chit-osan with a DA between 5% and 30%.2,4,5,9 High reaction

    temperatures reduce the time required for deacetylation, but

    result in increased hydrolysis of polymeric chains. Deacetyla-

    tion of chitin proceeds rapidly in 50% (w/v) aqueous NaOH at

    100 C during the first hour of alkali treatment, but extensionof the reaction time results rather in chain hydrolysis than in

    significant deacetylation.2 To obtain chitosans with low DAs

    (

  • including elemental analysis, colloid titration with a polya-

    nion, dye adsorption, and spectroscopies such as ultraviolet

    (UV), IR, and liquid/solid state nuclear magnetic resonance

    (NMR). The advantages and drawbacks of each technique

    have been discussed.2,4,5 Among these, high-resolution proton

    NMR (1H NMR) is considered the most accurate technique for

    the determination of the DA of chitosan. 1H NMR is usually

    performed in D2O containing DCl, the DA value being deter-

    mined from the relative integrals of acetyl (N-acetyl and

    AcOH) and combined H2H6 protons.16,17 For chitosans

    with high acetyl contents, the use of solid state 13C CP/MAS

    NMR is preferred to 1H NMR, since complete dissolution of

    the sample prior to analysis is required for 1H NMR. Because

    of its simplicity associated with accuracy, IR spectroscopy

    is the most frequently used technique. The use of FT-IR spec-

    troscopy for the determination of the DA is based on the

    variation of an absorbance band characteristic of N-acetyl

    groups, as a function of an internal reference band. The DA

    value can be extrapolated from a calibration curve estab-

    lished using an absolute technique, such as NMR. A number

    of different methods have been proposed, differing in terms

    of the analytical and reference IR bands used, as well as in

    terms of the baselines used for the determination of the corre-

    spondent absorbance values.18 One of the most frequently

    employed method is the one described by Baxter et al.19

    It uses the amide I band at 1655 cm1 as the analytical band

    This relation was validated by dye adsorption, for DAs

    comprised between 0 and 55%. Later on, Brugnerotto et al.20

    proposed the use of the amide III band (CN stretching cou-

    pled with NH in plane deformation) at 1320 cm1 as theanalytical band and the band at 1420 cm1 as the internalreference band. The authors analyzed chitin/chitosan samples

    covering the entire range of DAs, and used samples from

    different sources. A very good linear correlation between the

    ratio of absorbance bands (A1320/A1420) and the experimental

    values obtained from NMR was found in all the range of DA

    values, which could be expressed by the following relation:

    A1320=A1420 0:3822 0:03133DA % Brugnerotto et al. found superior agreement between the

    experimental and estimated DA values using this ratio of absor-

    bance bands than those involving the band at 3450 cm1

    (Figure 2).

    2.213.1.4.2. Molecular weightDepending on its source and preparation procedure, chitosan

    molecular weight may range from 300 to over than 1000 kD,

    squid pen chitosans presenting usually higher molecular

    weight, as compared to chitosans obtained from the exoskele-

    tons of crustaceans.21 Several techniques can be used to esti-

    mate the average molecular weight of chitosan. Among them

    are capillary viscometry, ultracentrifugation, and size exclusion

    chromatography (SEC) coupled with light scattering. Whatever

    l.

    um

    pos

    224 Materials of Biological Origin(the baseline is the one originally proposed for the A1655/A2867ratio) and the hydroxyl band at 3450 cm1 as the internalreference band. The sample has, therefore, to be perfectly dry

    and the IR spectra immediately recorded. The DA is deter-

    mined as follows:

    DA % A1655=A3450 115

    20003000400020

    25

    30

    35

    40

    45

    50

    55

    60

    65

    70

    75

    %T

    3431

    Baxter et al. Brugnerotto et a

    Waven

    Figure 2 Squid pen chitosan infrared spectra, showing the baselines proA1320/A1420 and A1655/A3450 ratio, respectively.the technique used, the tendency of the polymer chains to

    aggregate in solution constitutes a problem.2,22 SEC is the

    most direct one, providing, in a single measurement, the

    weight-average molecular weight and the number-average

    molecular weight. From these two values, the polydispersity

    40010001500

    1657

    14211317

    ber (cm1)

    ed by Brugnerotto et al.20 and Baxter et al.19 for the determination of the

  • Chitosan 225tion of neutrophils, which are cells usually associated with

    acute inflammation, no evidence of other signs associated

    with an inflammatory response, such as erytema and edema,

    were found. A very low incidence of chitosan-specific

    immune reactions was observed and, with time, collagen

    deposition within and surrounding the implant, with capsule

    formation, was found. However, the capsule was always

    highly cellular and its thickness decreased over time. Angio-

    genic activity associated with the external implant surface was

    observed. Overall, chitosan with DA 8% was found to have a

    high degree of biocompatibility. Recently, we assessed the

    inflammatory response to chitosan porous scaffolds with

    two different DAs (DA 4% and DA 15%) using a subcutane-

    ous air-pouch model of inflammation.28 Implantation of

    chitosan scaffolds with DA 15% induced a higher recruit-

    ment of neutrophils and increased adhesion of inflammatory

    cells during the early phase of implantation, while DA 4%

    merely caused a slight increase in the number of leukocytes

    present in the inflammatory exudates. With time, chitosanindex Mw/Mn can be easily determined. Still, this technique

    requires previous calibration of the SEC system with narrow

    polydispersity standards of knownmolecular weight. The asso-

    ciation of a light-scattering detector with SEC provides infor-

    mation on the absolute molecular weight as well as on

    molecular size parameters, such as the radius of gyration Rg.

    The analysis is usually performed using CH3COOH/

    CH3COONa buffers at pH near 4.5 as mobile phase, salt

    being added to screen electrostatic repulsion between proto-

    nated amine groups in chitosan.23 For the analysis, the refrac-

    tive index increment value (dn/dC) of chitosan in the

    CH3COONa/CH3COOH system used is required. The dn/dC

    values for chitosan may be found in literature, including some

    that are dependent on the DA.24 In this case, previous charac-

    terization of the DA may be required. Alternatively and if

    possible, the dn/dC value can be measured using a differential

    refractometer. SEC coupled with light scattering is also the

    technique described in ASTM guidelines for the determination

    of chitosan molecular weight.

    2.213.1.5. General Aspects of Biological Behavior

    2.213.1.5.1. BiocompatibilityOne of the major issues that have to be addressed while

    envisaging biomedical applications of a biomaterial is bio-

    compatibility. The nontoxicity of chitosan films was initially

    showed by Rao and Sharma25 using standard in vivo toxico-

    logical tests to evaluate chitosan safety and haemostatic

    potential. The biocompatibility of films with different DAs

    was examined by Tomihata and Ikada,26 using a rat subcuta-

    neous implant model. While chitosan films with DA 31%

    induced a relatively severe inflammatory reaction, with

    almost complete resorption after 4weeks of implantation,

    films with lower DAs led to a lower inflammatory reaction

    and degraded at a slower rate. Films with DAs 16% showeda very mild tissue reaction. The biocompatibility of chitosan

    porous scaffolds (DA 8%) was addressed by VandeVord

    et al.,27 using a mouse intraperitoneal and subcutaneous

    implant model. Histological assessment revealed an early

    migration of neutrophils into the implantation area, which

    resolved over implantation time. Besides this early accumula-2.213.1.5.3. Bacteriostatic and fungostatic propertiesChitosan exhibits an intrinsic antibacterial activity, inhibiting

    bacteria and fungi growth. As an example, in Staphylococcus

    aureus cultures, chitosan treatment promotes structural

    changes in the so-called membranewall complex leading to

    the impairment of surface cell structures and to bacterial2.213.1.5.2. CytocompatibilityA wide number of cells have been successfully cultured on 2D

    and 3D chitosan matrices envisaging cell-based regenerative

    therapies, among them keratinocytes,29 chondrocytes,30,31

    osteoblasts,3234 hepatocytes,35 and Schwann cells.36 The DA

    was found to be an important parameter affecting cell adhe-

    sion, lower DAs favoring cell adhesion. This effect was reported

    for a number of anchorage-dependent cells, such as keratino-

    cytes,29 fibroblasts,29,37 dorsal root ganglion neurons,38,39 and

    Schwann cells.40 In our lab, we investigated the DA effect on

    the behavior of osteogenic cells on chitosan films and porous

    matrices, using DAs in the range of 449%.32,33,41,42 These

    studies revealed a similar tendency to increased cell adhesion

    on decreasing the DA, and showed that differences in the DA as

    small as 9% can be critical in terms of osteoblastic response to

    chitosan.41,42 For instance, in the case of rat bone marrow

    stromal cells, cell adhesion, cytoskeleton organization, prolif-

    eration, and osteogenic differentiation were only observed on

    chitosan with DA 4%, while the same were hampered on

    chitosans with higher DAs (13%). We hypothesize that theDA could influence cell adhesion and osteoblast differentia-

    tion by influencing the adsorbed layer of adhesion proteins.

    For that, we performed protein adsorption studies using125I-fibronectin. In line with the higher cell adhesion levels

    found, chitosan with DA 4% showed the highest fibronectin

    (FN) adsorption both from a single FN protein solution and

    from diluted serum.32,33 From these results we may speculate

    that protonated amine groups from glucosamine units in chit-

    osan may modulate cell adhesion to chitosan by promoting

    the adsorption of cell adhesive proteins such as FN.

    To improve cell behavior on chitosan matrices, several

    attempts have been made, including physiadsorption of adhe-

    sive proteins43 and covalent binding of cell adhesion pep-

    tides.44,45 For instance, endothelial cell (EC) adhesion to

    porous chitosan matrices is significantly enhanced upon previ-

    ous incubation of chitosan matrices in an FN solution.46 Inter-

    estingly, we found that EC adhesion to FN-coated chitosan

    matrices is also dependent on the DA. While cell adhesion

    was impaired on DA 15%, ECs were able to adhere, spread,

    and colonize chitosan matrices with DA 4%. Later on, protein

    adsorption studies on scaffolds with DA 4% revealed a higher

    number of exposed FN cell-binding domains, as well as greater

    ability to adsorb FN and to retain and exchange adsorbed FN in

    the presence of competitive proteins, in agreement with the

    higher cell numbers found.scaffolds with DA 15% induced the formation of a thicker

    collagenous capsule and a high infiltration of inflammatory

    cells within the scaffold. Since inflammation and healing are

    interrelated, these results showed the importance of the care-

    ful selection of the DA while developing chitosan porous

    implants for tissue repair and regeneration.

  • 47

    226 Materials of Biological Origindeath. The biological mechanisms underlying this property

    remain poorly understood. Bacterial growth inhibition is

    believed to be related to chitosan ability to establish electro-

    static interactions between chitosan cationic amino groups and

    anions, such as N-acetylmuramic acid, sialic acid, and neura-

    minic acid, present on the bacterial cell wall. In addition to

    electrostatic interactions, hydrophobic interactions resultant

    from the presence of N-acetylated residues in chitosan are

    also thought to contribute to chitosan bacteriostatic properties,

    highly acetylated chitosans being reported to be excellent floc-

    culants of Escherichia coli suspensions.48

    2.213.1.5.4. Enzymatic degradationIn nature, chitosan can be hydrolysed by chitinases, chitosanases,

    and lysozymes, as well as by nonspecific hydrolases, such as

    a-amilases and lipases. In addition, like all polysaccharides, chit-osan is vulnerable to acid hydrolysis and to oxidativereductive

    depolymerization reactions.49,50 In vivo, the human enzymes

    involved in chitosan hydrolysis are only partially known.

    In human serum partially, N-acetylated chitosans are

    mainly depolymerized by lysozyme.50 Lysozyme is normally

    present in the human serum, saliva, and other fluids, hydro-

    lyzing preferentially the b-(14) glicosidic linkages betweenN-acetylglucosamine (NAGA) and N-acetylmuramic acid

    residues that occur in the cell walls of bacteria. Therefore,

    in addition to its natural substrate, lysozyme can hydrolyse

    partially N-acetylated chitosans. The active site of lysozyme

    binds six sugar rings, and three consecutiveN-acetyl-D-glucos-

    amine residues are required for lysozyme catalytic activity.51

    As a consequence, the susceptibility of chitosan to lysozyme

    depolymerization in vitro depends not only on the DA but

    also on the distribution of N-acetylated units along chitosan

    chains. The initial degradation rate increases with the DA,

    and this increase is more pronounced for N-acetylated chit-

    osans prepared under homogeneous conditions than for

    chitosans obtained by heterogeneous deacetylation.50,52 The

    same trend was observed in vivo upon subcutaneous implan-

    tation of chitosan films in a rat animal model.26 Chitosans

    with low DAs may last several months in vivo.27 Crystallinity

    is another parameter significantly influencing chitosan sus-

    ceptibility to lysozyme hydrolysis, by reducing lysozyme

    accessibility to the substrate. For instance, the low degrada-

    tion rates reported for chitosans with very low DAs have been

    frequently associated with the high levels of crystallinity and

    inter-molecular bindings present in these chitosans.26,27

    Finally, chitosan solubility may eventually shade the effect

    of lysozyme on chitosan degradation. This is the case of the

    accelerated mass loss observed for chitosans with DAs close

    to 50%, attributed to the enhanced solubility of chitosan mole-

    cules at physiologic pH.38,39

    The enzymatic hydrolysis of chitosan in wound-healing

    process was addressed by Muzarelli.49 Upon hydrolysis of

    chitosan by lysozyme, the oligomers released activate macro-

    phages, inducing the production of diffusible molecules,

    such as interferon, tumor necrosis factor-a, and interleukin-1.Activated macrophages secrete more lysozyme as well as

    N-acetyl-b-D-glucosaminidase and chitinase, which furthercatalyze the depolymerization of chitosan into monomers.

    These become available for further incorporation into hyalur-

    onate, keratan sulfate, and chondroitin sulfate.2.213.1.5.5. ImmunoadjuvancyChitosan is chemotactic for neutrophils, which has been

    attributed to specific interactions of chitosan or its oligosac-

    charides with neutrophils receptors, such as selectins.27 In

    addition to being chemotactic to neutrophils, chitosan shows

    a biological aptitude for activating macrophages for tumorici-

    dal activity and for production of interleukin-1,53 as well as

    nitric oxide.54 The immunoadjuvant properties of chitosan

    have been attributed to the NAGA units, rather than to

    the glucosamine units. Macrophage activation appears to be

    dependent on binding of NAGA to specific cell membrane

    receptors, namely involved in the binding of mannose- and

    NAGA-glycoproteins.54

    2.213.1.5.6. Hemostatic and blood clotting propertiesChitosan is a powerful hemostatic agent25 that induces blood

    clotting, even in the presence of extensive anticoagulation ther-

    apy.55 Blood clotting was suggested to be related to the possible

    formation of polyelectrolyte complexes (PECs), involving chit-

    osan amino functionalities and negatively charged acidic groups

    present at the surface of erytrocytes. Benesch and Tengvall56

    suggested that the procoagulant activity of chitosan could be

    related to the ability of chitosan to bind fibrinogen. In contrast,

    chitin displays anticoagulant properties, increasing upon

    O-sulfation,57 which is attributed to its similarity to heparin, a

    naturally occurring sulfated glycosaminoglycan (GAG) used as

    anticoagulant agent in clinic.

    2.213.1.5.7. Cell-binding propertiesChitosan is able to agglutinate a variety of mammalian cells in

    suspension. Cell adhesion to chitosan is attributed to nonspe-

    cific electrostatic interactions occurring directly between pro-

    tonated amine groups from glucosamine units and negatively

    charged carboxylate and sulfate groups found in cell surface

    proteoglycans.29,58 Close to the physiologic pH, the majority

    of chitosan ammonium groups are dissociated and subse-

    quently uncharged. Still, the presence of a small amount of

    nondissociated ammonium groups in chitosan chains is suffi-

    cient to provide enough cationic sites and allow the establish-

    ment of electrostatic interactions.30

    2.213.1.5.8. Wound-healing propertiesChitosan has been found to accelerate dermal wound healing

    and inhibit fibroplasia, showing a biological aptitude to stim-

    ulate cell proliferation and the deposition of an orderly

    organized connective tissue.59 This behavior has been related

    to chitosan ability to activate macrophages for cytokine

    production (transforming growth factor beta 1 (TGF-b1)and platelet-derived growth factor (PDGF)), upon hydrolysis

    by lysozyme.60,61 Moreover, Howling et al.62 suggested

    that chitosan may interact with growth factors present in

    serum, potentiating their effect, as chitosan has a stimulatory

    effect on dermal fibroblast proliferation when added to serum-

    containing culture medium, highly deacetylated chitosans

    producing a stronger mitogenic response, as compared to sam-

    ples with lower levels of deacetylation.

    2.213.1.5.9. Bone-healing propertiesThe osteogenic potential of chitosan was first reported by

    Borah et al.63 in 1992. In this study, chitosan was used to

  • Chitosan 227In the control group, no sign of ostegenesis or reparative

    process was observed and bone marrow was rich in adipo-

    cytes. The modified chitosan was reported to have a stimula-

    tory effect on bone formation. Although several works have

    demonstrated the osteoconductive properties of chitosan, it

    was often combined with therapeutic molecules, growth fac-

    tors, or calcium phosphates. The bone regenerative properties

    of unmodified chitosan are reported in a study of Park et al.66

    In this study, unmodified and PDGF-releasing chitosan

    sponges were applied to rat calvarial defects. Both matrices

    led to a significant increase in new bone formation, as com-

    pared to untreated defects, which became completely filled

    with fibrous connective tissue. In addition, a marked increase

    of bone formation and mineralization was observed in the

    presence of PDGF, as expected. The subsequent studies of

    Lee et al.67 supported these results. The effect of the DA

    on osteogenesis was addressed by Hidaka et al.,68 who

    implanted subperiosteally over the calvaria of rats mem-

    branes prepared from hydroxyapatite and chitosan. DAs of

    0, 6, 20, 30, and 35% were used. The authors reported for

    DAs 20% a marked inflammatory reaction, followedby accumulation of osteocalcin positive cells at the site of

    implantation, while for lower DAs a mild inflammation

    with minimal osteogenesis was observed.

    2.213.1.6. Chitosan Functionalization

    Chitosan has both reactive amino or amido groups at C(2)

    positions, as well as primary and secondary hydroxyl groups

    at C(6) and C(3) positions which can be used to prepare

    derivatives with well-defined structures and biological pro-

    perties under mild reaction conditions. Two modification

    reactions were already addressed in this chapter, namely dea-

    cetylation and N-acetylation. Other chemical modifications

    often explored to prepare versatile precursors and chitosan

    derivatives for biomedical applications include other acyla-

    tion reactions, N-phthaloylation, Schiffs base formation,

    N,O-carboxymethylation, N-carboxyalkylation, and graft

    copolymerization.2,9 Graft copolymerization, in particular,

    has been extensively used to introduce side chains onto chit-

    osan, namely to obtain tailored hybrid materials composed

    of natural polysaccharides and synthetic polymers.treat bone defects, made in the endochondral long bones of

    the rabbit. Chitosan reportedly stimulated osteogenesis with

    closure of the critical size bone defects, as compared to

    controls, in 812 weeks. As time progressed, the possible oste-

    ogenic, osteoconducting, and osteoinducting properties

    became the subject of investigation. The most significant

    works demonstrating the osteoconductive properties of chito-

    san are possibly those carried out by Muzarelli et al.64,65 In the

    first work, Muzzarelli et al.65 treated bone defects made in

    the tibiae of rabbits with freeze-dried methyl pyrrolidinone

    chitosan. The experimental sites showed signs of neoformed

    bone tissue, as opposed to controls, originating from the pre-

    existing bone, as well as from the periosteum. Subsequently,

    Muzzarelli et al. prepared a modified chitosan (DA 8%)

    carrying imidazole groups, to treat bone defects made in the

    femoral condyle of sheep. Within 40 days after surgery,

    the neoformed tissue occluded the surgical hole and assumed

    a trabecular structure in the peripheral area of the lesion.2.213.2. Processing

    Chitosan is extremely versatile in terms of processing. Films,

    micro- and nanoparticles, porous scaffolds, micro- and nano-

    fibers, and meshes can be produced with chitosan for a wide

    range of applications.

    2.213.2.1. Films and Porous Scaffolds (Freeze-Drying andFreeze-Gelling)

    Taking advantage of the solubility of chitosan in mildly acidic

    conditions, films can be produced by casting the resulting gel

    on a flat surface and then allowing the solvent to evaporate.

    Neutralization with an alkaline solution, washing, and drying

    are often employed to stabilize the films. These can then be

    used as substrates for cell culture experiments.

    Porous scaffolds can be prepared by freeze-drying, also

    known as lyophilization. Acidic chitosan solutions are frozen

    to temperatures of 20 to 80 C, which causes formation ofice crystals. The rate and temperature of freezing influence the

    size of the ice crystals and consequently the size of the pores

    formed during the sublimation phase. The latter is carried out

    in a vacuum at low temperature, in order to avoid melting of

    the chitosan-rich phase. After elimination of the ice crystals,

    drying can be continued to eliminate unfrozen water mole-

    cules. Pores with dimensions in the range 40250mm and

    porosities of 80% can be obtained. By applying a temperature

    gradient along a certain direction, preferential orientation of

    the pores can be obtained to produce structures with aligned

    porosity. Figure 3 presents laser scanning confocal and scan-

    ning electron microscopy (SEM) images of chitosan sponges

    with two DAs (4% and 30%), which have been colonized

    by MG-63 cells.42 It shows that a better spreading is reached

    with the lower DA.

    In some cases, the freeze-drying method can damage the

    pore walls during evaporation of the solvent. Also, formation

    of skin can also occur. To avoid these problems, the method of

    freeze-gelling has been proposed for producing porous struc-

    tures of poly(L-lactic acid) (PLLA), poly(lactic-co-glycolic acid)

    (PLGA), chitosan, and alginate.69 The method is based on the

    extraction of the solution-rich phase by a nonsolvent for the

    polymer. Exemplifying for the case of chitosan, the procedure

    is the following. An acetic acid solution of chitosan is frozen

    to 20 C, which causes the formation of ice crystals. To formthe pores, the ice crystals are removed by immersing the frozen

    chitosan solution in an NaOH/ethanol solution at 20 C.Neutralization of the acid by the sodium hydroxide causes

    the gelation of chitosan, whereas the ethanol dissolves the ice

    crystals. Subsequent evaporation of the ethanol at room tem-

    perature produces the pores. The method has also been applied

    to produce composite scaffolds of chitosan and gamma-poly

    (glutamic acid) (g-PGA) for the delivery of rhBMP-2.70 Con-centrations of chitosan and g-PGA were 4% and 1%, respec-tively, in a 0.2M acetic acid solution. Freezing was done

    at 80 C and gelation was achieved by immersing the frozenchitosan/g-PGA in a 3-M NaOH/ethanol solution at 20 C.

    2.213.2.2. Nanofibers

    Nanofibers of chitosan can be produced by electrospinning.

    This technique has become extremely useful for the

  • Dimagidiud. M

    228 Materials of Biological Originpreparation of nanofibers for tissue engineering, due to the

    wide range of polymers that can be processed to obtain nano-

    fibers with diameters in the range 20400nm. Briefly, the

    application of a sufficiently high voltage between a needle

    and a metallic collector overcomes the surface tension holding

    a drop of liquid at the tip of the needle, resulting in a very thin

    jet of fluid being projected against the collector. The solvent is

    evaporated during the trajectory between needle and collector,

    resulting in a nonwoven structure. One of problems with

    electrospun meshes is the slow cell infiltration due to the

    high packing density of the nanofibers. To circumvent this

    problem fast degrading nanofibers can be combined with

    slow degrading fibers. This strategy has been developed by,71

    who have co-electrospun poly(epsilon-caprolactone) with

    poly(ethylene oxide) (PEO) from two separate spinnerets.

    The former polymer is slowly degradable, whereas the second

    one is water-soluble, acting as a sacrificial component of the

    scaffold. Seeding these scaffolds with mesenchymal stem cells

    (MSCs) has show that cell infiltration improved with the frac-

    tion of sacrificial polymer. High porosities (of the order of 95%

    to 97%) can be obtained also by coating microfibers with

    Figure 3 Fluorescence microscopy and scanning electron microscopicDAs. Red lines: chitosan walls of the pores; nuclei were stained with prop(green). Adapted from Amaral, I. F; Sampaio, P.; Barbosa, M. A. J. BiomeDA 30%nanofibers, as demonstrated by.72 Scaffolds made from poly-

    caprolactone nanofibers electrospun onto polylactic acid

    microfibers supported infiltration of human chondrocytes,

    with higher porosities favoring cell infiltration.

    2.213.2.3. Polyelectrolyte Complexes

    The self-assembly of polymer chains due to electrostatic inter-

    actions is the basis for the formation of PECs. PECs are formed

    by reacting two oppositely charged polyelectrolytes in an aque-

    ous solution. PECs are generally biocompatible networks and

    can be easily produced in the lab. PECs between chitosan

    and many polyanions have been described. Polysaccharides

    such as alginate, chondroitin sulfate, dextran sulfate and gel-

    lum gum form PECs with chitosan. Similarly, polyamino

    acids (e.g., poly-L-Lysine and poly(aspartic acid)), proteins

    (e.g., collagen), and glycosoaminoglycans (e.g., hyaluronic

    acid, HA) also bind electrostatically to chitosan, via anionic

    carboxylic or sulfate groups. The cationic amino groups ofchitosan are responsible for the formation of electrostatic

    bonds with the anionic groups of the other polyelectrolyte.

    The formation of PEC hydrogels requires that the two

    polymers are oppositely charged. Since the pKa of chitosan

    is in the vicinity of 6.5, the other polymer must have a

    lower pKa for electrostatic aggregation to occur. If attraction

    is too strong, a precipitate, and not a hydrogel, will form.

    By modifying the ionic strength of the aqueous solution,

    for example, by addition of salts such as NaCl, the electro-

    static interactions can be modulated so that a homogeneous

    hydrogel is formed. Functionalization of chitosan with posi-

    tively charged (e.g., glycolchitosan73), or negatively charged

    (e.g., sulfate32,33) groups expands the range of PECs

    that can be produced. Since no cross-linkers are required to

    form the hydrogel, these materials usually have excellent

    cytocompatibilty.

    Using the principles that govern PEC formation, a film can

    be produced using the layer-by-layer (LbL) method, by alter-

    nate immersion of a substrate in solutions of positively and

    negatively charged polyelectrolytes. LbL films are usually very

    thin (typically

  • form the chitosan/DNA complex nanoparticles. Chitosan

    anteed during washing, which has limited the use of this

    has been reported to have no such toxic effects. Cross-linking

    2.213.3. Biomedical Applications

    Because of its well-known biocompability, chitosan is being

    widely explored for biomedical and pharmaceutical applica-

    tions. In the following sections the main areas of application

    and research will be discussed.

    2.213.3.1. Wound Management

    Chitosan is referenced in the wound management field for its

    hemostatic properties85 Furthermore, the biological properties

    including bacteriostatic and fungistatic86 properties are partic-

    ularly useful for wound treatment. It also affects inflammatory

    cell function that helps in faster wound healing, and has an

    aptitude to stimulate cell proliferation and histoarchitectural

    tissue organization.87

    Chitosan has been explored as a topical hemostatic agent in

    a variety of forms,88 reaching the market in recent years. Agents

    being commercialized include: (1) a dressing, which works by

    becoming extremely adherent when in contact with blood,

    sealing the wound and controlling bleeding (HemConMedical

    Strain (%)

    Chitosan 229cross-linker. Genipin, which is a natural product used in tradi-

    tional Chinese medicine and extracted from gardenia fruit,nanoparticles with various ratios of chitosan to plasmid OP-1

    were used to transfect chondrocytes.81 Nanoparticles with a

    chitosan/plasmid weight ratio of 10:1 entered the cells and

    resulted in the expression of the plasmid, which maintained

    its structural integrity. The size of the plasmid did, however,

    affect the efficiency of transfection to the cells.

    2.213.2.5. Cross-linking

    Like in other polymers, cross-linking of chitosan molecules is

    often used to increase the mechanical and chemical stability of

    chitosan. Cross-linking may involve covalent binding of chit-

    osan molecules to chitosan molecules, or to other polymer

    chains. A hybrid polymer network is formed in this case.

    The covalent bonds are formed between the two different

    molecules, but they may also be established between mole-

    cules of the same polymer. Another possibility is to add to the

    chitosan solution a nonreacting polymer before cross-linking.

    The chains of this polymer become entangled in the structure

    of the cross-linked chitosan, contributing to its physical rein-

    forcement. These are called semi-interpenetrating networks

    (semi-IPN). The reinforcing polymer can be subsequently

    cross-linked to form a full-IPN. The most common cross-

    linkers that have been used are dialdehydes, and particularly

    glutaraldhyde.82 The aldehyde group forms imine bonds with

    the amino groups of chitosan and the reaction can be carried

    out in aqueous medium, without the need for other molecules

    to initiate the reaction. One of the disadvantages of the

    method is the toxicity of glutaraldhyde. Removal of residual

    glutaraldehyde molecules from the hydrogel cannot be guar-bilayers and using higher concentrations of cross-linker, the

    effect on adhesion was more pronounced.

    2.213.2.4. Micro- and Nanoparticles

    Micro- and nanoparticles of chitosan can be prepared by vari-

    ous methods. The most common method consists in ionotro-

    pic gelation of chitosan molecules by anions, such as the

    polyanion tripolyphosphate (TPP)77 and sulfate.78 The experi-

    mental procedure involves drop-wise addition of the anion to

    a chitosan solution under agitation. Agitation can be per-

    formed mechanically, ultrasonically, or by a combination of

    both. The rate of agitation controls the size of the particles.

    Chitosan nanoparticles prepared by gelation with TPP were

    uptaken by A549 cells in percentages that depended on the

    molecular weight (Mw) and DA.79 Uptake decreased by 26%

    when the Mw decreased from 213 000 to 10 000 and by 41%

    when the DA increased from 12% to 54%. The nanoparticles

    cytotoxicity was lower for the higher DA.

    Nanoparticles have been produced for incorporating

    DNA, proteins and therapeutic agents. A complex coacervation

    method has been often used for incorporating DNA. In com-

    plex coacervation, separation of two oppositely charged col-

    loids occurs. Normally, a chitosan solution (e.g., in sodium

    acetate buffer) and a DNA solution (e.g., in sodium sulfate) are

    preheated to 50 C and then quickly mixed and vortexed to80of chitosan/PEO blends with genipin resulted in more stable

    and elastic films.83 Figure 4 shows the stressstrain curves

    of chitosan/PEO obtained with different concentrations of

    genipin. 0.1% genipin resulted in films that exhibited a strain

    to fracture of 90%. Genipin has also been used to stabilize

    polylectrolyte multilayers of chitosan/hyaluronan and chito-

    san/alginate.76 Cell adhesion was markedly influenced by

    cross-linking. Water-soluble chitosan chlorides (low and high

    molecular weights) and chitosan glutamates (low and

    high molecular weights) were cross-linked using various con-

    centrations (5% to 20%) of genipin, to encapsulate cells

    removed from bovine intervertebral discs.84 A cell viability of

    95% was obtained with the high molecular weight chitosan

    glutamate cross-linked with 5% genipin.Figure 4 Stressstrain curves of chitosan/poly(ethylene oxide)(PEO) films cross-linked with genipin. The PEO used (LPEO) had aMw 600. Reproduced from Jin, J.; Song, M.; Hourston, D. J.Biomacromolecules 2004, 5(1), 162168, with permission from Elsevier.0

    7

    14

    21

    28

    35

    0.01% 0.1%

    0.8%

    5% water

    0%

    CSR/LPEO50

    Str

    ess

    (MP

    a)

    0 20 40 60 80 100

    0.5%

  • well as bone fillers, such as cements, adhesives for tissue repair,

    230 Materials of Biological Originand scaffolds for tissue engineering. Among the latter, skin,

    bone, cartilage, and nervous tissues are those where more

    investigation is taking place, involving the use of chitosan as

    a scaffolding material or as an extracellular matrix (ECM)

    analog.9496 The popularity of chitosan for tissue repair and

    regeneration is owing to the fact that it can be readily processed

    into a variety of forms that include fibers, films, sponges, or

    hydrogels. This provides the possibility to mimic the shape of

    target tissue or interfaces. Moreover, the similarity of its chem-

    ical structure to some polysaccharide constituents of the ECM

    and the possibility to chemically modify it to impart desired

    functionalities add to the great potential of chitosan as a bio-

    material for tissue repair and regeneration.

    In orthopedics, chitosan has been often used in combina-

    tion with ceramics, such as hydroxiapatite and other calcium-

    containing ceramics to produce composites. These have been

    investigated as bone fillers, and many were found suitable as

    bone-filling materials.97,98 The advantage of this approach is to

    develop bone substitute materials, which combine the biode-

    gradability, strength, and flexibility of chitosan with the osteo-

    genic potential and hardness of the mineral filler. In addition,

    the chitosan matrix acts as a binder, preventing postoperative

    migration of the mineral phase.99 Chitosan has also been

    explored as an adjuvant additive, to render calcium phosphate

    cements injectable and to enable their use in minimally inva-

    sive surgical procedures.100 Other bone cements combining

    chitosan, hydroxiapatite, and poly(methyl methacrylate) wereTechnologies, Inc.); (2) high surface area flakes which, when

    in contact with blood, swell, gel, and stick together to make a

    gel-like clot (being commercialized by MedTrade Products

    Ltd.); and (3) a chitosan-coated nonwoven pad used in the

    management of bleeding wounds (Abbot Vascular, USA). The

    mechanisms underlying the action of chitosan are not com-

    pletely understood, but it has been suggested to involve

    vasoconstriction, and the rapid mobilization of red blood

    cells, clotting factors and platelets to the site of the injury as a

    result of the positive charge on the chitosan molecule.89 This

    action is reported to occur even in patients on anticoagulants.

    Because of its high absorption capacity of fluids, wound-

    healing properties, adhesiveness, antibacterial activity, and

    film forming properties, chitosan has been explored for burn

    and wound dressings.90 The modification of chitosan proper-

    ties, either at the structural level or by association with other

    materials, has been explored60,61 to potentially improve its

    biological performance. Despite these efforts, the commercial

    exploration of a wound dressing material based on chitosan is

    yet in its infancy.

    Although the apparent potential of chitin and chitosan deri-

    vatives in the preparation of sutures have long been recognized,

    there is still no commercial production of chitin-based absorb-

    able suture materials because of insufficient elasticity of chitin

    threads and certain limitations of their processability into the

    fiber form.91,92 Strategies to overcome some of these hurdles

    have been thoroughly reviewed by Pillai and coworkers.93

    2.213.3.2. Tissue Repair and Regeneration

    Internal medical applications for tissue repair and regeneration

    include orthopedic implants, such as bone pins and plates, asalso developed, and reported to provide porous spaces for

    osteoconduction, after chitosan degradation.101 Finally, the

    association of growth factors with chitosan-based bone fillers

    has also been investigated, as a combinative strategy of con-

    trolled local drug delivery and bone regenerative therapy.49

    Besides being used as a filler material, chitosan has also been

    employed as a coating material, in order to improve titanium

    and hydroxyapatite implants osteointegration.102104 Envisa-

    ging to improve the biocompatibility of electrolytically depos-

    ited apatite coatings on Ti alloys, Wang et al.105 prepared a

    hybrid calcium phosphate/chitosan coating. When compared

    to apatite coatings, this hybrid coating revealed to be more

    cytocompatible towards bone marrow stromal cells.

    There are a wide variety of adhesives available for use in

    surgery, ranging from cyanoacrylates to fibrin-based mixtures.

    Chitosan use in the design of new tissue adhesives was moti-

    vated by the fact that it can bind to collagen due to hydrogen

    bonding and polyanionicpolycationic interactions. It is said

    to overcome some of the limitations of currently available

    materials. It is not blood-derived, and in light activated appli-

    cations can be used without significant temperature raise.

    Lauto and coworkers106,107 have developed flexible and insol-

    uble strips of chitosan-based adhesives that incorporate indo-

    cyanin green dye, for use as a bandage to fix rectangular

    sections of sheep intestine using a diode laser. The chitosan

    bandage bonded effectively to tissue without sutures and pre-

    served the ECM structure avoiding irreversible thermal dena-

    turation of imbedded bioactive proteins. A photo-cross-linked

    chitosan hydrogel showed strong sealing ability when tested in

    a rabbit animal model of a punctured carotid artery and lung,

    stopping the bleeding and air leakage, respectively.108 The

    sealing ability of the chitosan hydrogel was found to be similar

    or even stronger than that of fibrin.

    Table 1 provides examples of works being developed in the

    field of tissue engineering using chitosan as a scaffold/matrix

    material.

    Many studies have been reported on the use of chitosan as a

    scaffold in skin tissue engineering due to its many advantages

    for wound healing, such as hemostasis, accelerating tissue

    regeneration, and stimulating the fibroblast synthesis of colla-

    gen.121 The use of chitosan for the preparation of skin substi-

    tutes was successfully explored in a blend with bovine collagen

    types I and III and GAGs, where fibroblast and keratinocytes

    have been cocultured. The blend-based porous substrate acts as

    a scaffold for fibroblasts, thereby producing a living dermal

    equivalent, which once epithelialized results in reconstructed

    skin.122 The use of this skin substitute has been proven to

    lead not only to the reconstruction of surface epithelia for

    the treatment of pathological conditions of skin, but also as

    a testing platform for pharmatoxicologic studies, serving as a

    validated alternative to animal tests.

    In articular cartilage tissue engineering, the ideal scaffold

    should mimic the natural environment in the articular cartilage

    matrix, which is highly hydrated and rich in ECM components,

    such as type II collagen and GAGs. These are known to play a

    key role in the expression of the chondrocytic phenotype

    and in supporting chondrogenesis in vitro as well as in vivo.

    Chitosan has been found particularly interesting for hyaline

    cartilage tissue engineering, due to its structural similarity with

    HA, a GAG abundant in the ECM of cartilage.123 The

  • Table 1 Current applications of chitosan scaffolds in tissue engineering

    Application Scaffold DA (%) Scaffold properties Performed biological studies References

    Skin Asymmetric genepin-cross-linked chitosan membranecontaining collagen Inanospheres

    15 Asymmetricmembrane(porosity 4258%)

    In vitro static primary culture of rat skinfibrobasts

    In vivo evaluation of skin regeneration in arat animal model

    109

    Cartilage Chitosan/b-chitin scaffolds 2 Porous (pore size50100mm)

    Rabbit chondrocytes (isolated fromarticular cartilage) culture and ECMproduction assessment

    110

    Cartilage Temperature-responsivechitosan hydrogel (withb-sodiumglycerophosphate andhydroxyethyl cellulose)

    14 Hydrogel In vitro cultures with adult sheep articularcondrocytes

    In vivo implantation in articular cartilagedefects in the sheep model

    111

    Nucleuspulposus ofintervertebraldiscs (IVD)

    Chitosan/glycerophosphategels Chitosan/glycerophosphate/hydroxyethyl cellulose gelsChitosan/genipin gels

    Chitosan:5% and21%Glutamatesalt ofchitosan:15%

    Hydrogel In vitro primary cultures of peripheralannulus fibrosus and the centralnucleus pulposus cells isolated fromIVDs from the tails of bovine steers

    112

    Bone Chitosan/TCP scaffolds n.m. Porous (pore size100mm)

    Fetal rat calvarial osteoblastic cellsproliferation, viability, anddifferentiation

    113

    Bone Chitosan/nonsinteredhydroxylapatite particles

    12 BilayeredPorous layer (poresize 100400mm;porosity 30%)

    Cytotoxicity of extracts (fibroblasts)Osteogenic and chondrogenicdifferentiation studies withmesenchymal stem cell (from humanadipose tissue)

    114

    Bone Chitosan and PDGF-BBloaded chitosan scaffolds

    n.m. Porous (pore size100mm)

    Cytotoxicity tests: rat calvarialosteoblastic cells adhesion andproliferationBone regeneration ability in a ratcalvarial defect

    66

    Bone Chitosan and chitosan/chondroitin sulfatescaffoldsChitosan/PLLA scaffoldsChitosan coated PLLAscaffolds(all loaded or not withPDGF-BB)

    30 Porous (pore size100150mm)Porous (pore size150200mm)Porous (pore size100150mm)

    Rat calvarial osteoblastic cells adhesionand proliferationBone regeneration ability in a ratcalvarial defect

    67

    Bone Chitosan/gelatin andchitosan/gelatin/b-TCPmacroporous scaffolds

    10 Porous (pore size322mm)Porous (pore size185420mm)

    Biocompatibility evaluation(subcutaneous implantation in a rabbitmodel)

    115

    Bone Chitosan scaffolds andchitosan scaffoldsmodified with RGDS orRGES peptides

    15 n. m. Rat osteoblast-like cells (ROS 17/2.8)adhesion, proliferation, anddifferentiation

    45

    Nerve Chitosan conduits containingoriented filaments ofpolyglycolic acid

    7.7 Nonporous chitosanconduit containingoriented filamentsof polyglycolic acid

    Bridging sciatic nerve across a 30-mmdefect in Beagle dogs

    116

    Nerve Chitosan 8 Nonporous conduit Implantation of extramedullary conduitsseeded with neural stem and progenitorcells derived from transgenic greenfluorescent protein rats after spinalcord transection

    117

    Ligament Chitosan-based hyaluronanhybrid polymer fibers

    19 Fibers Rabbit fibroblast adhesion andproliferation; ECM productionassessment

    118

    (Continued)

    Chitosan 231

  • fold

    s wrag160 nm and chitosan films

    /chitffol 9rosi

    232 Materials of Biological Originpossibility of using chitosan in the hydrogel form is an added

    value in such application. In this form it may closely match the

    natural mechanical properties of hyaline cartilage. Further-

    more, its application can be performed by aminimally invasive

    method. A number of in situ gelling chitosan-based hydrogels

    are currently under investigation for this purpose.124,125

    In recent years, considerable attention has been given to the

    application of chitosan-based materials in the field of orthope-

    dic tissue engineering. Interesting characteristics that render

    chitosan suitable for this purpose are its biocompatibility/bio-

    degradability, structural similarity to ECMGAGs, intrinsic anti-

    bacterial nature, ability to bind anionic molecules such as

    growth factors, GAGs, and DNA, as well as its ability to be

    molded into porous structures suitable for cell ingrowth and

    osteoconduction.126 In this sense, a wide number of support

    matrices in the form of injectable gels or porous scaffolds have

    been developed for bone tissue-engineering applications. Most

    often, chitosan is used in combination with a variety of materi-

    als, such as ceramics, PLLA, gelatin, GAGs, as well as growth

    factors, in an attempt to improve its mechanical properties,

    osteoconduction, and ability to induce bone regeneration (see

    Table 1). Binding with cell adhesive motifs has also been

    explored, in order to promote cell adhesion.

    Although chitosan has been used as a scaffold for articular

    cartilage and bone formation by direct differentiation of mesen-

    chymal cells into chondrocytes and osteoblast, respectively, only

    recently it has been used as a template for endochondral ossifi-

    cation.31,127 The endochondral ossification pathway involves an

    intermediate cartilage stage and is responsible for the formation

    of long bones, vertebra, and the cranial base during develop-

    ment. Oliveira et al. have subcutaneously implanted a transient

    cartilage scaffold based on chitosan and a permanent cartilage

    scaffold in nude mice (see Figure 5). Only in the former, the

    Table 1 (Continued)

    Application Scaffold DA (%) Scaf

    Liver Galactosylated chitosanelectrospun nanofibers

    15 Fiberaveof

    Vascular Human-like collagen (HLC,produced by recombinantE. coli)/chitosan tubularscaffolds

    2025 HLCsca46po

    n. m. not mentioned.depositionof ectopic bonewas detected, as early as 1month after

    implantation. After 3months, bone trabeculae and bone mar-

    row cavities were formed inside the scaffolds. The bone depos-

    itedwas similar to the boneof themice vertebra. Interestingly, no

    bone formation was observed in control implants, for the time

    span of the study.

    When the extent of nerve damage prohibits the direct

    approximation of the two nerve stumps in peripheral nerve

    repair, autologous nerve grafting is considered the surgical

    treatment of choice. Artificial nerve guides, where a tube

    bridges the nerve gap, offer a promising alternative, preventing

    the sacrifice of a donor nerve and possible neuroma formation

    at the donor site. Furthermore, the use of synthetic nerveguides reduces operation time and prevents the mismatch

    between the damaged nerve and the graft. Peripheral nerve

    correction performed with chitosan-based nerve conduits has

    been explored with success.116 Chitosan-based conduits pres-

    ent a number of advantages over other proposed guides,

    namely, the readiness of tuning their physical properties38,39

    and surface chemistry by changing the DA of chitosan, which

    in turn influences cell adhesion behavior of the key player cells

    in the nerve regeneration process.40 The surface modification

    of chitosan scaffolds with cell adhesion proteins or motifs,

    such as the pentapeptides YIGSR and IKVAV, which bind to

    neural cell membrane receptors, has also been investigated

    either by physiadsorption128 or chemical grafting.129131 The

    use of chitosan-based conduits for spinal cord injury treatment

    is also under study with the first in vivo studies showing

    promising results.117,132,133 The incorporation into the scaf-

    folds of proteins that can enhance nerve regeneration is being

    assessed as well.134 Moreover, the possibility of culturing neu-

    ral stem cells in these constructs also opens new avenues for

    addressing nerve lesion treatments.135

    A transversal problem in the field of tissue engineering has

    been the vascularization of the tissue-engineered constructs

    postimplantation. The insufficient supply of oxygen and nutri-

    ents to the inner part of the cellmatrix constructs in the initial

    phases after implantation136 was found to be contributing to

    the limited success of many of the proposed tissue regeneration

    strategies. To overcome this unsolved issue, a number of stra-

    tegies are being developed to create a vascular network able to

    deliver oxygen and nutrients throughout an implanted cell

    matrix construct. One of the promising approaches has been

    the precolonization of the scaffolds with vascular cell types,

    such as ECs or endothelial progenitor cells.137,138 We have

    shown that the endothelization of chitosan scaffolds was

    Hepatocytes function assessmentosan tubulard (pore sizemm;ty 85%)

    In vitro human venous fibroblast cultureIn vivo biocompatability assessment inrabbits livers

    120properties Performed biological studies References

    ith ane diameter

    Attachment and culture of primary rathepatocytes on the fibrous scaffolds

    119found to be possible when these were precoated with the

    ECM protein FN.46 However, as observed for other cell types,

    the DA was found to be a key parameter modulating EC adhe-

    sion to FN-coated chitosan scaffolds (see Figure 6) by influen-

    cing the adsorbed protein layer. The selection of suitable

    DAs will therefore be highly important for the design of new

    vascularization strategies.

    2.213.3.3. Delivery of Therapeutic Agents

    The use of chitosan in the pharmaceutical industry spans from

    its application as an excipient139 to its application as a vehicle

    for the controlled delivery of therapeutic agents.

  • (q)(p)

    1st month 2nd month 3rd month 4th month

    (a)

    (e)

    (i)

    (j)

    (m) (n)

    (f) (g) (h)

    5th month

    ExperimentalVertebraControl

    (b) (c) (d)

    50mm

    200mm200mm200mm200mm

    50mm

    2 mm

    50mm50mm

    200mm

    50mm

    50mm 50mm 50mm

    50mm50mm

    200mm 200mm(l)

    (o)

    (r)

    (k)

    Figure 5 Histological analysis of chondrocyte/chitosan scaffolds implanted in the subcutaneous region of nude mice. Axial paraffin sections were cutthrough the body of the mouse (I, 5month time point) to include the experimental and control scaffolds. Sections were stained with H&E andphotographed. Details of bone forming in experimental scaffolds during the first 4months of implantation are shown in images (a) through (d). Images(e), (f), (g), and (h) correspond to high magnifications of (a), (b), (c), and (d), respectively, showing the beginning of vascular invasion (arrow heads).The control scaffolds do not undergo visible changes during the first 5months of implantation, and therefore only the 5th month is included in thisfigure. A complete section is shown after 5months of implantation in image (i). Control scaffold is on the left and experimental is seen on the right side ofthe image. Details of control scaffold (j), vertebral body (k), and experimental scaffold (l) can be observed. Higher magnifications of control (m, p),mouse vertebra (n, q), and experimental scaffold (o, r) show tissue details. Deep in the control sample (m), only cartilage tissue can be observed.Vascular invasion of cartilage (arrow head in o) and bone deposition (arrow in o) can be observed in the experimental sample. Detail of vertebra (n, q)shows the presence of bone (arrow) and blood vessels (arrow head). Note bone formation deep in the experimental scaffold as well as the presence ofblood vessels (r). Adapted from Oliveira, S. M.; Mijares, D. Q.; Turner, G.; Amaral, I. F.; Barbosa, M. A.; Teixeira, C. C. Tissue Eng. 2009, 15(3): 635643,with permission from Liebert.

    Chitosan 233

  • 234 Materials of Biological Origin72h

    DA 4%

    (a) (d)

    4h

    300mmTaking advantage of the fact that positively charged chitosan

    can bind to negatively charged lipids and bile salt components,

    interfering with the lipid absorption process in the gut, chito-

    san has been introduced in the market as a weight and choles-

    terol control agent. However, the efficacy of its use has been

    questioned in the field.140,141 Furthermore, chitosan has

    been used for the production of controlled release implant

    systems for delivery of hormones, proteins, and other peptides

    over extended periods of time.142144 The mucoadhesive prop-

    erties and transmucosal absorption-promoting characteristics

    of chitosan have been exploited especially for nasal145,146 and

    oral delivery147151 of polar drugs to include peptides, proteins,

    and nucleic acids and for vaccine development.132,133,152

    The immune adjuvant behavior of chitosan153 has also been

    drawing the attention of the field to its application in vaccine

    development154156 and cancer treatment.157159

    Additionally, its cationic nature and high charge density in

    slightly acidic conditions allow chitosan to form PECs nucleic

    (b)

    (c)

    (e)

    (f)

    144

    300mm

    300mm

    Figure 6 Confocal laser scanning microscopy of human pulmonary microvascaffolds previously incubated in a 40mgml1 fibronectin solution. Cells werImages were collected after 4, 72, and 144 h after cell seeding. HPMEC-ST1.6the scaffolds (a) to (f). Cell spreading as well as the cellular layer covering themagnification images (d) to (f). In these, the 3D scaffold is shown in blue duchitosan scaffolds with DA 15%, very few cells were found after 72 and 144 hfor which the same bar corresponds to 75 mm. Adapted from Amaral, I. F.; Upermission from Elseiver.(g) 300mm75mmDA 15%acids both DNA and RNA.94,95,160,161 Although chitosan is

    able to condense nucleic acids and protect them from nuclease

    degradation, its main advantage over other nonviral vectors

    relies on its low cytotoxicty and biodegradability. However,

    chitosans poor solubility under physiological conditions and

    low transfection efficacy has impeded its use as a nucleic acid

    carrier. To overcome such limitations, a number of strategies for

    chitosan modification have been proposed by our group and

    others. These include the increase of chitosan overall positive

    charge by quaternization,162 modification of its buffering capac-

    ity163 and functionalization of chitosan with cell-binding mole-

    cules for receptor-mediated cell internalization.164

    2.213.3.4. Other Applications

    Other potential applications of chitosan include cosmetics,

    contact lenses, dialysis membranes, and coatings of the inner

    lumen of blood-contacting polymeric tubes, onto which an

    (h)

    (i) 300mm

    300mm

    75mm

    74.14mm

    scular endothelial cells (HPMEC-ST1.6R cell line) grown on chitosane labeled with calcein AM resulting in green fluorescence of viable cells.R cells seeded on scaffolds with DA 4% were able to spread and colonizepore walls after 144 h of cell culture are shown in more detail in the highere to chitosan autofluorescence upon excitation by the 405 nm laser. Onof cell culture (h) and (i). Scale bar is 300 mm except for images (d) to (f),nger, R. E.; Fuchs, S. et al. Biomaterials 2009, 30(29), 54655475, with

  • tested in depth.

    16. Fernandez-Megia, E.; Novoa-Carballal, R.; Quinoa, E.; Riguera, R. Carbohydr.Polym. 2005, 61(2), 155161.

    Chitosan 23517. Hirai, A.; Odani, H.; Nakajima, A. Polym. Bull. 1991, 26(1), 8794.18. Shigemasa, Y.; Matsuura, H.; Sashiwa, H.; Saimoto, H. Int. J. Biol. Macromol.

    1996, 18(3), 237242.References

    1. Rudall, K. M. J. Polym. Sci. C Polym. Symp. 1969, 28PC, 83.2. Roberts, G. A. F. Chitin Chemistry. Macmillan: London, 1992.3. Khor, E. Chitin: Fulfilling a Biomaterials Promise. Elsevier Science: Oxford, UK,

    2001.4. Roberts, G. A. F. In Advances in Chitin Science; Domard, A., Roberts, G. A. F.,

    Varum, K. M., Eds.; Jacques Andre Publisher: Lyon, 1997; Vol. II, pp 2231.5. Roberts, G. A. F. In Chitin Handbook; Muzzarelli, R. A. A., Peter, M. G., Eds.; Atec:

    Grottammare, 1997; pp 127132.6. Sorlier, P.; Denuziere, A.; Viton, C.; Domard, A. Biomacromolecules 2001, 2(3),

    765772.7. Cauchie, H.-M. In Advances in Chitin Science; Domard, A., Roberts, G. A. F.,

    Varum, K. M., Eds.; Jacques Andre: Lyon, 1997; Vol. II, pp 3239.8. Kurita, K. In Chitin Handbook; Muzzarelli, R. A. A., Peter, M. G., Eds.; Atec:

    Grottammare, 1997; pp 491493.9. Kurita, K. Prog. Polym. Sci. 2001, 26(9), 19211971.

    10. Mima, S.; Miya, M.; Iwamoto, R.; Yoshikawa, S. J. Appl. Polym. Sci. 1983, 28(6),19091917.

    11. Lamarque, G.; Viton, C.; Domard, A. Biomacromolecules 2004, 5(3),9921001.

    12. Tchemtchoua, V. T.; Atanasova, G.; et al. Anal. Biochem. 2009, 393(1), 145147.13. Struszczyk, H. J. Appl. Polym. Sci. 1987, 33(1), 177189.14. Vachoud, L.; Zydowicz, N.; Domard, A. Carbohydr. Res. 1997, 302(34),

    169177.15. ASTM International Standard Guide for Characterization and Testing of Chitosan

    Salts as Starting Materials Intended for Use in Biomedical and Tissue-EngineeredMedical Product Applications. ASTM F 2103 01(2007)e2. ASTM International:West Conshohocken, PA, 2007.antithrombotic agent is deposited. Comprehensive reading on

    these and other applications of chitosan in the biomedical field

    can be found in Dumitriu.165

    2.213.4. Future Prospects

    Chitosan cationicity and enzymatic degradability associated

    with structural analogy with ECM glycosaminoglicans (GAGs)

    contribute together to the increasing interest in its application as

    a biomaterial. A limitation to its use in clinics is related to the

    low reproductivity of chitosan physicochemical characteristics

    among batches as well as the presence of nonexpected contami-

    nants. In this sense, chitosan obtained by biotechnology pro-

    cesses may constitute a better alternative to those presently

    obtained from marine animal sources. Furthermore, for the

    development of advanced chitosan-based biomaterials, inter-

    national standards concerning the range of requirements for

    chitosan use in medical devices are still missing.

    The possibility to covalently and ionically bound proteins

    and peptides to chitosan, combined with morphologically and

    mechanically tuned scaffolds for tissue repair/regeneration

    and targeted delivery of biologically-active entities for cell

    therapies are fields that remain vastly unexplored. The possi-

    bility of producing matrices with various degradation kinetics

    and inflammatory responses offers ample opportunities to

    expand the use of chitosan-based materials. In spite of

    the extensive research that has been carried out with these

    materials, a lot of clinically useful possibilities have not been19. Baxter, A.; Dillon, M.; Taylor, K. D. A.; Roberts, G. A. F. Int. J. Biol. Macromol.1992, 14(3), 166169.

    20. Brugnerotto, J.; Lizardi, J.; Goycoolea, F. M.; Arguelles-Monal, W.; Desbrieres, J.;Rinaudo, M. Polymer 2001, 42(8), 35693580.

    21. Hudson, S. M.; Smith, C. In Biopolymers from Renewable Resources;Kaplan, D. L., Ed.; Springer-Verlag: Berlin, 1998; pp 96118.

    22. Anthonsen, M. W.; Varum, K. M.; Hermansson, A. M.; Smidsrod, O.; Brant, D. A.Carbohydr. Polym. 1994, 25(1), 1323.

    23. Terbojevich, M.; Cosani, A. In Chitin Handbook; Muzzarelli, R. A. A., Peter, M. G.,Eds.; Atec: Grottammare, 1997; pp 87101.

    24. Schatz, C.; Viton, C.; Delair, T.; Pichot, C.; Domard, A. Biomacromolecules 2003,4(3), 641648.

    25. Rao, S. B.; Sharma, C. P. J. Biomed. Mater. Res. 1997, 34(1), 2128.26. Tomihata, K.; Ikada, Y. Biomaterials 1997, 18(7), 567575.27. VandeVord, P. J.; Matthew, H. W. T.; DeSilva, S. P.; Mayton, L.; Wu, B.;

    Wooley, P. H. J. Biomed. Mater. Res. 2002, 59(3), 585590.28. Barbosa, J. N.; Amaral, I. F.; Aguas, A. P.; Barbosa, M. A. J. Biomed. Mater. Res. A

    2010, 93(1), 2028.29. Chatelet, C.; Damour, O.; Domard, A. Biomaterials 2001, 22(3), 261268.30. Denuziere, A.; Ferrier, D.; Damour, O.; Domard, A. Biomaterials 1998, 19(14),

    12751285.31. Oliveira, S. M.; Amaral, I. F.; Barbosa, M. A.; Teixeira, C. C. Tissue Eng. 2009,

    15(3), 625634.32. Amaral, I. F.; Granja, P. L.; Barbosa, M. A. J. Biomater. Sci. Polym. Ed. 2005,

    16(12), 15751593.33. Amaral, I. F.; Lamghari, M.; Sousa, S. R.; Sampaio, P.; Barbosa, M. A. J. Biomed.

    Mater. Res. 2005, 75A(2), 387397.34. Lahiji, A.; Sohrabi, A.; Hungerford, D. S.; Frondoza, C. G. J. Biomed. Mater. Res.

    2000, 51(4), 586595.35. Elcin, Y. M.; Dixit, V.; Gitnick, G. Artif. Organs 1998, 22(10), 837846.36. Yuan, Y.; Zhang, P. Y.; Yang, Y. M.; Wang, X.; Gu, X. S. Biomaterials 2004,

    25(18), 42734278.37. Prasitsilp, M.; Jenwithisuk, R.; Kongsuwan, K.; Damrongchai, N.; Watts, P.

    J. Mater. Sci. Mater. Med. 2000, 11(12), 773778.38. Freier, T.; Koh, H. S.; Kazazian, K.; Shoichet, M. S. Biomaterials 2005, 26(29),

    58725878.39. Freier, T.; Montenegro, R.; Koh, H. S.; Shoichet, M. S. Biomaterials 2005, 26(22),

    46244632.40. Cao, W. L.; Jing, D. H.; Li, J. M.; Gong, Y. D.; Zhao, N. M.; Zhang, X. F.

    J. Biomater. Applic. 2005, 20(2), 157177.41. Amaral, I. F.; Cordeiro, A. L.; Sampaio, P.; Barbosa, M. A. J. Biomater. Sci. Polym.

    Ed. 2007, 18(4), 469485.42. Amaral, I. F.; Sampaio, P.; Barbosa, M. A. J. Biomed. Mater. Res. 2006, 76A(2),

    335346.43. Cuy, J. L.; Beckstead, B. L.; Brown, C. D.; Hoffman, A. S.; Giachelli, C. M.

    J. Biomed. Mater. Res. 2003, 67A(2), 538547.44. Chung, T. W.; Lu, Y. F.; Wang, S. S.; Lin, Y. S.; Chu, S. H. Biomaterials 2002,

    23(24), 48034809.45. Ho, M. H.; Wang, D. M.; et al. Biomaterials 2005, 26(16), 31973206.46. Amaral, I. F.; Unger, R. E.; et al. Biomaterials 2009, 30(29), 54655475.47. Didenko, L. V.; Gerasimenko, D. V.; et al. Bull. Exp. Biol. Med. 2005, 140(3),

    356360.48. Strand, S. P.; Vandvik, M. S.; Varum, K. M.; Ostgaard, K. Biomacromolecules

    2001, 2(1), 126133.49. Muzzarelli, R. A. A. Cell. Mol. Life Sci. 1997, 53, 131140.50. Varum, K. M.; Myhr, M. M.; Hjerde, R. J.; Smidsrod, O. Carbohydr. Res. 1997,

    299(12), 99101.51. Varum, K. M.; Holme, H. K.; Izume, M.; Stokke, B. T.; Smidsrod, O. Biochim.

    Biophys. Acta Gen. Subj. 1996, 1291(1), 515.52. Nordtveit, R. J.; Varum, K. M.; Smidsrod, O. Carbohydr. Polym. 1994, 23(4),

    253260.53. Nishimura, K.; Nishimura, S.; Seo, H.; Nishi, N.; Tokura, S.; Azuma, I. J. Biomed.

    Mater. Res. 1986, 20(9), 13591372.54. Peluso, G.; Petillo, O.; et al. Biomaterials 1994, 15(15), 12151220.55. Klokkevold, P. R.; Fukayama, H.; Sung, E. C.; Bertolami, C. N. J. Oral Maxillofac.

    Surg. 1999, 57(1), 4952.56. Benesch, J.; Tengvall, P. Biomaterials 2002, 23(12), 25612568.57. Hagiwara, K.; Kuribayashi, Y.; et al. Carbohydr. Polym. 1999, 39(3),

    245248.58. Henriksen, I.; Green, K. L.; Smart, J. D.; Smistad, G.; Karlsen, J. Int. J. Pharm.

    1996, 145(12), 231240.59. Muzzarelli, R.; Baldassarre, V.; et al. Biomaterials 1988, 9(3), 247252.60. Ueno, H.; Mori, T.; Fujinaga, T. Adv. Drug Deliv. Rev. 2001, 52(2), 105115.

  • 236 Materials of Biological Origin61. Ueno, H.; Nakamura, F.; Murakami, M.; Okumura, M.; Kadosawa, T.; Fujinaga, T.Biomaterials 2001, 22(15), 21252130.

    62. Howling, G. I.; Dettmar, P. W.; Goddard, P. A.; Hampson, F. C.; Dornish, M.;Wood, E. J. Biomaterials 2001, 22(22), 29592966.

    63. Borah, G.; Scott, B.; Wortham, K. In Advances in Chitin and Chitosan; Brine, C. J.,Sandford, P. A., Zikakis, J. P., Eds.; Elsevier: Amsterdam, 1992; pp 206215.

    64. Muzzarelli, R. A. A.; Mattiolibelmonte, M.; et al. Biomaterials 1994, 15(13),10751081.

    65. Muzzarelli, R. A. A.; Zucchini, C.; et al. Biomaterials 1993, 14(12), 925929.66. Park, Y. J.; Lee, Y. M.; Park, S. N.; Sheen, S. Y.; Chung, C. P.; Lee, S. J.

    Biomaterials 2000, 21(2), 153159.67. Lee, J. Y.; Nam, S. H.; et al. J. Control. Release 2002, 78(13), 187197.68. Hidaka, Y.; Ito, M.; Mori, K.; Yagasaki, H.; Kafrawy, A. H. J. Biomed. Mater. Res.

    1999, 46(3), 418423.69. Ho, M. H.; Kuo, P. Y.; et al. Biomaterials 2004, 25(1), 129138.70. Hsieh, C. Y.; Hsieh, H. J.; Liu, H. C.; Wang, D. M.; Hou, L. T. Dent. Mater. 2006,

    22(7), 622629.71. Baker, B. M.; Gee, A. O.; et al. Biomaterials 2008, 29(15), 23482358.72. Thorvaldsson, A.; Stenhamre, H.; Gatenholm, P.; Walkenstrom, P.

    Biomacromolecules 2008, 9(3), 10441049.73. Sakai, S.; Ono, T.; Ijima, H.; Kawakami, K. J. Microencapsul. 2000, 17(6),

    691699.74. Picart, C.; Schneider, A.; et al. Adv. Funct. Mater. 2005, 15(11), 17711780.75. Manna, U.; Bharani, S.; Patil, S. Biomacromolecules 2009, 10(9), 26322639.76. Hillberg, A. L.; Holmes, C. A.; Tabrizian, M. Biomaterials 2009, 30(27),

    44634470.77. Calvo, P.; RemunanLopez, C.; VilaJato, J. L.; Alonso, M. J. J. Appl. Polym. Sci.

    1997, 63(1), 125132.78. Berthold, A.; Cremer, K.; Kreuter, J. J. Control. Release 1996, 39(1), 1725.79. Huang, M.; Khor, E.; Lim, L. Y. Pharm. Res. 2004, 21(2), 344353.80. Mao, H. Q.; Roy, K.; et al. J. Control. Release 2001, 70(3), 399421.81. Xu, X.; Capito, R. M.; Spector, M. J. Biomed. Mater. Res. A 2008, 84(4),

    10381048.82. Schiffman, J. D.; Schauer, C. L. Biomacromolecules 2007, 8(2), 594601.83. Jin, J.; Song, M.; Hourston, D. J. Biomacromolecules 2004, 5(1), 162168.84. Mwale, F.; Iordanova, M.; Demers, C. N.; Steffen, T.; Roughley, P.; Antoniou, J.

    Tissue Eng. 2005, 11(12), 130140.85. Paul, W.; Sharma, C. P. Trends Biomater. Artif. Organs 2004, 18(1), 1823.86. Rinaudo, M. Polym. Int. 2008, 57(3), 397430.87. Park, C. J.; Gabrielson, N. P.; Pack, D. W.; Jamison, R. D.; Johnson, A. J. W.

    Biomaterials 2009, 30(4), 436444.88. Seyednejad, H.; Imani, M.; Jamieson, T.; Seifalian, A. M. Br. J. Surg. 2008, 95

    (10), 11971225.89. Neuffer, M. C.; McDivitt, J.; Rose, D.; King, K.; Cloonan, C. C.; Vayer, J. S. Mil.

    Med. 2004, 169(9), 716720.90. Mi, F. L.; Shyu, S. S.; Wu, Y. B.; Lee, S. T.; Shyong, J. Y.; Huang, R. N.

    Biomaterials 2001, 22(2), 165173.91. Mikhailov, G. M.; Lebedeva, M. F. Russ. J. Appl. Chem. 2007, 80(5), 685694.92. Nakajima, M.; Atsumi, K.; Kifune, K.; Miura, K.; Kanamaru, H. Jpn. J. Surg. 1986,

    16(6), 418424.93. Pillai, C. K. S.; Paul, W.; Sharma, C. P. Prog. Polym. Sci. 2009, 34(7),

    641678.94. Dang, J. M.; Leong, K. W. Adv. Drug Deliv. Rev. 2006, 58(4), 487499.95. Kim, I. Y.; Seo, S. J.; et al. Biotechnol. Adv. 2008, 26(1), 121.96. Muzzarelli, R. A. A. Carbohydr. Polym. 2009, 76(2), 167182.97. Cho, B. C.; Park, J. W.; Baik, B. S.; Kwon, I. C.; Kim, I. S. J. Craniofac. Surg.

    2002, 13(6), 783793.98. Kawakami, T.; Antoh, M.; Hasegawa, H.; Yamagishi, T.; Ito, M.; Eda, S.

    Biomaterials 1992, 13(11), 759763.99. Khor, E.; Lim, L. Y. Biomaterials 2003, 24(13), 23392349.

    100. Leroux, L.; Hatim, Z.; Freche, M.; Lacout, J. L. Bone 1999, 25(2), 31S34S.101. Kim, S. B.; Kim, Y. J.; et al. Biomaterials 2004, 25(26), 57155723.102. Bumgardner, J. D.; Wiser, R.; Elder, S. H.; Jouett, R.; Yang, Y.; Ong, J. L.

    J. Biomater. Sci. Polym. Ed. 2003, 14(12), 14011409.103. Bumgardner, J. D.; Wiser, R.; et al. J. Biomater. Sci. Polym. Ed. 2003, 14(5),

    423438.104. Mattiolibelmonte, M.; Biagini, G.; et al. J. Bioact. Compat. Polym. 1995, 10(3),

    249257.105. Wang, J.; de Boer, J.; de Groot, K. J. Dent. Res. 2004, 83(4), 296301.106. Lauto, A. Lasers Surg. Med. 2009, 41(5), 366371.107. Lauto, A.; Stoodley, M.; et al. Lasers Surg. Med. 2007, 39(1), 1927.108. Ishihara, M.; Ono, K.; et al. New Dev. Glycomed. 2001, 1223, 251257.109. Chen, K. Y.; Liao, W. J.; et al. Biomacromolecules 2009, 10(6), 16421649.110. Suzuki, D.; Takahashi, M.; et al. J. Mater. Sci. Mater. Med. 2008, 19(3),13071315.

    111. Hao, T.; Wen, N.; Cao, J. K.; et al. Osteoarthr. Cartil. 2010, 18(2), 257265.112. Roughley, P.; Hoemann, C.; DesRosiers, E.; Mwale, F.; Antoniou, J.; Alini, M.

    Biomaterials 2006, 27(3), 388396.113. Lee, Y. M.; Park, Y. J. J. Periodontol. 2000, 71(3), 410417.114. Malafaya, P. B.; Pedro, A. J.; Peterbauer, A.; Gabriel, C.; Redl, H.; Reis, R. L.

    J. Mater. Sci. Mater. Med. 2005, 16(12), 10771085.115. Yin, Y. J.; Ye, F.; Cui, J. F.; Zhang, F. J.; Li, X. L.; Yao, K. D. J. Biomed. Mater.

    Res. A 2003, 67A(3), 844855.116. Wang, X.; Hu, W.; Cao, Y.; Yao, J.; Wu, J.; Gu, X. Brain 2005, 128(Pt 8),

    18971910.117. Nomura, H.; Zahir, T.; et al. Tissue Eng. A 2008, 14(5), 649665.118. Funakoshi, T.; Majima, T.; et al. J. Biomed. Mater. Res. 2005, 74A(3), 338346.119. Feng, Z. Q.; Chu, X. H.; et al. Biomaterials 2009, 30(14), 27532763.120. Zhu, C.; Fan, D.; et al. J. Biomed. Mater. Res. A 2009, 89(3), 829840.121. Muzzarelli, R. A.; Mattioli-Belmonte, M.; Pugnaloni, A.; Biagini, G. Exs 1999, 87,

    251264.122. Auxenfans, C.; Builles, N.; et al. Pathol. Biol. 2009, 57(4), 290298.123. Suh, J. K. F.; Matthew, H. W. T. Biomaterials 2000, 21(24), 25892598.124. Jin, R.; Moreira Teixeira, L. S.; et al. Biomaterials 2009, 30(13), 25442551.125. Tan, H.; Chu, C. R.; Payne, K. A.; Marra, K. G. Biomaterials 2009, 30(13),

    24992506.126. Di Martino, A.; Sittinger, M.; Risbud, M. V. Biomaterials 2005, 26(30),

    59835990.127. Oliveira, S. M.; Mijares, D. Q.; Turner, G.; Amaral, I. F.; Barbosa, M. A.;

    Teixeira, C. C. Tissue Eng. 2009, 15(3), 635643.128. Itoh, S.; Yamaguchi, I.; et al. Brain Res. 2003, 993(12), 111123.129. Kato, K.; Utani, A.; et al. Biochemistry 2002, 41(35), 1074710753.130. Matsuda, A.; Kobayashi, H.; Itoh, S.; Kataoka, K.; Tanaka, J. Biomaterials 2005,

    26(15), 22732279.131. Wang, W.; Itoh, S.; et al. J. Biomed. Mater. Res. A 2008, 85A(4), 919928.132. Li, G. P.; Liu, Z. G.; Liao, B.; Zhong, N. S. Cell Mol. Immunol. 2009, 6(1), 4550.133. Li, X.; Yang, Z.; Zhang, A.; Wang, T.; Chen, W. Biomaterials 2009, 30(6),

    11211132.134. Chavez-Delgado, M. E.; Mora-Galindo, J.; et al. J. Biomed. Mater. Res. B Appl.

    Biomater. 2003, 67B(2), 702711.135. Wang, A.; Ao, Q.; et al. Tsinghua Sci. Technol. 2006, 11(4), 415420.136. Moon, J. J.; West, J. L. Curr. Top. Med. Chem. 2008, 8(4), 300310.137. Fuchs, S.; Motta, A.; Migliaresi, C.; Kirkpatrick, C. J. Biomaterials 2006, 27(31),

    53995408.138. Unger, R. E.; Peters, K.; Wolf, M.; Motta, A.; Migliaresi, C.; Kirkpatrick, C. J.

    Biomaterials 2004, 25(21), 51375146.139. Illum, L. Pharm. Res. 1998, 15(9), 13261331.140. Mhurchu, C. N.; Dunshea-Mooij, C.; Bennett, D.; Rodgers, A. Obes. Rev. 2005, 6

    (1), 3542.141. Pittler, M. H.; Ernst, E. Am. J. Clin. Nutr. 2004, 79(4), 529536.142. Janes, K. A.; Calvo, P.; Alonso, M. J. Adv. Drug Deliv. Rev. 2001, 47(1), 8397.143. Liu, Z. H.; Jiao, Y. P.; Wang, Y. F.; Zhou, C. R.; Zhang, Z. Y. Adv. Drug Deliv. Rev.

    2008, 60(15), 16501662.144. Prabaharan, M.; Mano, J. F. Drug Deliv. 2005, 12(1), 4157.145. Goycoolea, F. M.; Lollo, G.; Remunan-Lopez, C.; Quaglia, F.; Alonso, M. J.

    Biomacromolecules 2009, 10(7), 17361743.146. Teijeiro-Osorio, D.; Remunan-Lopez, C.; Alonso, M. J. Biomacromolecules 2009,

    10(2), 243249.147. Hejazi, R.; Amiji, M. Int. J. Pharm. 2002, 235(12), 8794.148. Hejazi, R.; Amiji, M. J. Control. Release 2003, 89(2), 151165.149. Hejazi, R.; Amiji, M. Pharm. Dev. Technol. 2003, 8(3), 253262.150. Hejazi, R.; Amiji, M. Int. J. Pharm. 2004, 272(12), 99108.151. Thanou, M.; Verhoef, J. C.; Junginger, H. E. Adv. Drug Deliv. Rev. 2001, 50,

    S91S101.152. Vila, A.; Sanchez, A.; et al. Eur. J. Pharm. Biopharm. 2004, 57(1), 123131.153. Nishimura, K.; Nishimura, S.; et al. Vaccine 1985, 3(5), 379384.154. Westerink, M. A. J.; Smithson, S. L.; et al. Vaccine 2001, 20(56),

    711723.155. Yang, Y.; Chen, J.; et al. Comp. Immunol. Microbiol. Infect. Dis. 2007, 30(1),

    1932.156. Zaharoff, D. A.; Rogers, C. J.; Hance, K. W.; Schlom, J.; Greiner, J. W. Vaccine

    2007, 25(11), 20852094.157. Chen, W. R.; Adams, R. L.; Carubelli, R.; Nordquist, R. E. Cancer Lett. 1997, 115

    (1), 2530.158. Chen, W. R.; Korbelik, M.; Bartels, K. E.; Liu, H.; Sun, J.; Nordquist, R. E.

    Photochem. Photobiol. 2005, 81(1), 190195.

  • 159. Seo, S. H.; Han, H. D.; Noh, K. H.; Kim, T. W.; Son, S. W. Clin. Exp. Metastasis2009, 26(3), 179187.

    160. Borchard, G. Adv. Drug Deliv. Rev. 2001, 52(2), 145150.161. Lai, W. F.; Lin, M. C. J. Control. Release 2009, 134(3), 158168.162. Kean, T.; Roth, S.; Thanou, M. J. Control. Release 2005, 103(3), 643653.163. Moreira, C.; Oliveira, H.; Pires, L. R.; Simoes, S.; Barbosa, M. A.; Pego, A. P.

    Acta Biomater. 2009, 5(8), 29953006.164. Murata, J.-i.; Ohya, Y.; Ouchi, T. Carbohydr. Polym. 1996, 29(1), 6974.

    165. Dumitriu, S. Polysaccharides in Medical Applications. Marcel Dekker:New York, 1996.

    Relevant Web