Generic placeholder image

Current Stem Cell Research & Therapy

Editor-in-Chief

ISSN (Print): 1574-888X
ISSN (Online): 2212-3946

Review Article

Chitosan in Biomedical Engineering: A Critical Review

Author(s): Shabnam Mohebbi, Mojtaba Nasiri Nezhad*, Payam Zarrintaj, Seyed Hassan Jafari, Saman Seyed Gholizadeh, Mohammad Reza Saeb* and Masoud Mozafari*

Volume 14, Issue 2, 2019

Page: [93 - 116] Pages: 24

DOI: 10.2174/1574888X13666180912142028

Price: $65

Abstract

Biomedical engineering seeks to enhance the quality of life by developing advanced materials and technologies. Chitosan-based biomaterials have attracted significant attention because of having unique chemical structures with desired biocompatibility and biodegradability, which play different roles in membranes, sponges and scaffolds, along with promising biological properties such as biocompatibility, biodegradability and non-toxicity. Therefore, chitosan derivatives have been widely used in a vast variety of uses, chiefly pharmaceuticals and biomedical engineering. It is attempted here to draw a comprehensive overview of chitosan emerging applications in medicine, tissue engineering, drug delivery, gene therapy, cancer therapy, ophthalmology, dentistry, bio-imaging, bio-sensing and diagnosis. The use of Stem Cells (SCs) has given an interesting feature to the use of chitosan so that regenerative medicine and therapeutic methods have benefited from chitosan-based platforms. Plenty of the most recent discussions with stimulating ideas in this field are covered that could hopefully serve as hints for more developed works in biomedical engineering.

Keywords: Chitosan, biomaterial, biomedical engineering, stem cell, tissue engineering, gene therapy.

[1]
Hafshejani TM, Zamanian A, Venugopal JR, et al. Antibacterial glass-ionomer cement restorative materials: A critical review on the current status of extended release formulations. J Control Release 2017; 262: 317-28.
[2]
Zarrintaj P, Saeb MR, Ramakrishna S, Mozafari M. Biomaterials selection for neuroprosthetics. Curr Opin in Biomed Eng 2018; 6: 99-109.
[3]
Zarrintaj P, Manouchehri S, Ahmadi Z, et al. Agarose-based biomaterials for tissue engineering. Carbohydr Polym 2018; 187: 66-84.
[4]
Atoufi Z, Zarrintaj P, Motlagh GH, Amiri A, Bagher Z, Kamrava SK. A novel bio electro active alginate-aniline tetramer/agarose scaffold for tissue engineering: Synthesis, characterization, drug release and cell culture study. J Biomater Sci Polym Ed 2017; 28: 1617-38.
[5]
Sari MG, Saeb MR, Shabanian M, et al. Epoxy/starch-modified nano-zinc oxide transparent nanocomposite coatings: A showcase of superior curing behavior. Prog Org Coat 2018; 115: 143-50.
[6]
Zarrintaj P, Bakhshandeh B, Saeb MR, et al. Oligoaniline-based conductive biomaterials for tissue engineering. Acta Biomater 2018; 72: 16-34.
[7]
Zarrintaj P, Bakhshandeh B, Rezaeian I, Heshmatian B, Ganjali MR. A novel electroactive agarose-aniline pentamer platform as a potential candidate for neural tissue engineering. Sci Rep 2017; 7: 17187.
[8]
Derakhshandeh MR, Eshraghi MJ, Hadavi MM, et al. Diamond-like carbon thin films prepared by pulsed-DC PE-CVD for biomedical applications. Surf Innov 2018; 6: 167-75.
[9]
Derakhshandeh SMR, Eshraghi MJ, Javaheri M, et al. Diamond-like carbon-deposited films: A new class of bio-corrosion protective coatings. Surf Innov 2018; 1-42.
[10]
Sadeghi-Kiakhani M, Khamseh S, Rafie A, Tekieh SMF, Zarrintaj P, Saeb MR. Thermally stable antibacterial wool fabrics surface-decorated by TiON and TiON/Cu thin films. Surf Innov 2018; 1-30.
[11]
Rahmati M, Milan PB, Samadikuchaksaraei A, et al. Ionically crosslinked thermoresponsive chitosan hydrogels formed in situ: A conceptual basis for deeper understanding. Macromol Mater Eng 2017; 302.
[12]
Elieh-Ali-Komi D, Hamblin MR. Chitin and chitosan: Production and application of versatile biomedical nanomaterials. Int J Adv Res 2016; 4: 411.
[13]
Islam S, Bhuiyan MR, Islam M. Chitin and chitosan: Structure, properties and applications in biomedical engineering. J Polym Environ 2017; 25: 854-66.
[14]
Kazami N, Sakaguchi M, Mizutani D, et al. A simple procedure for preparing chitin oligomers through acetone precipitation after hydrolysis in concentrated hydrochloric acid. Carbohydr Polym 2015; 132: 304-10.
[15]
Wang R, Song X, Xiang T, et al. Mussel-inspired chitosan-polyurethane coatings for improving the antifouling and antibacterial properties of polyethersulfone membranes. Carbohydr Polym 2017; 168: 310-9.
[16]
Younes I, Rinaudo M. Chitin and chitosan preparation from marine sources. Structure, properties and applications. Mar Drugs 2015; 13: 1133-74.
[17]
Tian M, Tan H, Li H, You C. Molecular weight dependence of structure and properties of chitosan oligomers. RSC Advances 2015; 5: 69445-52.
[18]
Kumari S, Rath P, Kumar ASH, Tiwari T. Extraction and characterization of chitin and chitosan from fishery waste by chemical method. Environ Technol Innov 2015; 3: 77-85.
[19]
Garcia-Valdez O, Champagne P, Cunningham MF. Graft modification of natural polysaccharides via reversible deactivation radical polymerization. Prog Polym Sci 2017; 76: 151-73.
[20]
Sudha P, Gomathi T, Aisverya S. Recent research in the applications of chitin, chitosan and oligosaccharides. Green Polym Environ Pollut Control 2016; p. 303.
[21]
Choi C, Nam J-P, Nah J-W. Application of chitosan and chitosan derivatives as biomaterials. J Ind Eng Chem 2016; 33: 1-10.
[22]
Halim AS, Keong LC, Zainol I, Rashid AHA. Biocompatibility and biodegradation of chitosan and derivativesChitosan-based systems for biopharmaceuticals: Delivery, targeting and polymer therapeutics¶ 2012: 57-73
[23]
Ghasemi HH, Rezvani Z, Nazm BM, et al. Chitosan-intercalated montmorillonite/poly (vinyl alcohol) nanofibers as a platform to guide neuronlike differentiation of human dental pulp stem cells. ACS Appl Mater Interfaces 2017; 9: 11392-404.
[24]
Aliramaji S, Zamanian A, Mozafari M. Super-paramagnetic responsive silk fibroin/chitosan/magnetite scaffolds with tunable pore structures for bone tissue engineering applications. Mater Sci Eng C 2017; 70: 736-44.
[25]
Motevalian M, Karimi M, Chauhan N, Habibi Y, Mozafari M. Chitosan-functionalized poly (lactide-co-glycolide) nanoparticles: ultimate breaking through the brain’s tight security gateway. Bioinspired Biomimetic and Nanobiomater 2016; 5(2): 74-84.
[26]
Hur S-J, Kim Y-C, Choi I, Lee S-K. The effects of biopolymer encapsulation on total lipids and cholesterol in egg yolk during in vitro human digestion. Int J Mol Sci 2013; 14: 16333-47.
[27]
Saeedi M, Vahidi O, Goodarzi V, Saeb MR, Izadi L, Mozafari M. A new prospect in magnetic nanoparticle-based cancer therapy: Taking credit from mathematical tissue-mimicking phantom brain models. Nanomed Nanotechnol Biol Med 2017; 13: 2405-14.
[28]
Bakhshandeh B, Zarrintaj P, Oftadeh MO, et al. Tissue engineering; strategies, tissues, and biomaterials. Biotechnol Genet Eng Rev 2017; 33: 144-72.
[29]
Zou P, Yang X, Wang J, et al. Advances in characterisation and biological activities of chitosan and chitosan oligosaccharides. Food Chem 2016; 190: 1174-81.
[30]
Chen K, Guo B, Luo J. Quaternized carboxymethyl chitosan/organic montmorillonite nanocomposite as a novel cosmetic ingredient against skin aging. Carbohydr Polym 2017; 173: 100-6.
[31]
Jimtaisong A, Saewan N. Utilization of carboxymethyl chitosan in cosmetics. Int J Cosmet Sci 2014; 36: 12-21.
[32]
Morsy R, Ali SS, El-Shetehy M. Development of hydroxyapatite-chitosan gel sunscreen combating clinical multidrug-resistant bacteria. J Mol Struct 2017; 1143: 251-8.
[33]
Libio IC, Demori R, Ferrão MF, Lionzo MI, da Silveira NP. Films based on neutralized chitosan citrate as innovative composition for cosmetic application. Mater Sci Eng C 2016; 67: 115-24.
[34]
Nilforoushzadeh MA, Amirkhani MA, Zarrintaj P, et al. Skin care and rejuvenation by cosmeceutical facial mask. J Cosmet Dermatol 2018; 17(5): 693-702.
[35]
Luo X, Zeng J, Liu S, Zhang L. An effective and recyclable adsorbent for the removal of heavy metal ions from aqueous system: magnetic chitosan/cellulose microspheres. Bioresour Technol 2015; 194: 403-6.
[36]
Madill EA, Garcia-Valdez O, Champagne P, Cunningham MF. CO2-responsive graft modified chitosan for heavy metal (nickel) recovery. Polym 2017; 9: 394.
[37]
Tsai B, Garcia-Valdez O, Champagne P, Cunningham MF. Poly (Poly (Ethylene Glycol) Methyl Ether Methacrylate) Grafted Chitosan for Dye Removal from Water. Processes 2017; 5: 12.
[38]
Quinlan PJ, Grishkewich N, Tam KC. Removal of 2‐naphthoxyacetic acid from aqueous solution using quaternized chitosan beads. Can J Chem Eng 2017; 95: 21-32.
[39]
Aider M. Chitosan application for active bio-based films production and potential in the food industry. LWT-Food Sci Techno 2010; 43: 837-42.
[40]
Kashyap PL, Xiang X, Heiden P. Chitosan nanoparticle based delivery systems for sustainable agriculture. Int J Biol Macromol 2015; 77: 36-51.
[41]
Santo Pereira AE, Silva PM, Oliveira JL, Oliveira HC, Fraceto LF. Chitosan nanoparticles as carrier systems for the plant growth hormone gibberellic acid. Colloids Surf B 2017; 150: 141-52.
[42]
El Hadrami A, Adam LR, El Hadrami I, Daayf F. Chitosan in plant protection. Mar Drugs 2010; 8: 968-87.
[43]
Vikele L, Laka M, Sable I. et al. Effect of chitosan on properties of paper for packaging. Cellul Chem Technol 2017; 51: 67-73.
[44]
Zemljič Lf, Valh Jv, Kreže T. Preparation of antimicrobial paper sheets using chitosan. Cellul Chem Technol 2017; 51(1-2): 75-81.
[45]
Rahmati M, Samadikuchaksaraei A, Mozafari M. Insight into the interactive effects of β-glycerophosphate molecules on thermosensitive chitosan-based hydrogels. Bioinspir Biomim Nan 2016; 5: 67-73.
[46]
Nazemi K, Azadpour P, Moztarzadeh F, Urbanska A, Mozafari M. Tissue-engineered chitosan/bioactive glass bone scaffolds integrated with PLGA nanoparticles: A therapeutic design for on-demand drug delivery. Mater Lett 2015; 138: 16-20.
[47]
Samadikuchaksaraei A, Gholipourmalekabadi M, Farhadihosseinabadi B, Rezvani Z, Mozafari M. Carboxymethyl chitosan/forsterite bone tissue engineering scaffolds: Correlations between composition and physico-chemical characteristics. Biointerface Res Appl Chem 2016; 6: 1222-8.
[48]
Zarrintaj P, Urbanska A, Gholizadeh SS, Goodarzi V, Saeb MR, Mozafari M. A facile route to the synthesis of anilinic electroactive colloidal hydrogels for neural tissue engineering applications. J Colloid Interface Sci 2018; 516: 57-66.
[49]
Farokhi M, Mottaghitalab F, Samani S, et al. Silk fibroin/hydroxyapatite composites for bone tissue engineering. Biotechnol Adv 2017; 36: 68-91.
[50]
Kargozar S, Baino F, Hamzehlou S, Hill RG, Mozafari M. Bioactive glasses: Sprouting angiogenesis in tissue engineering. Trends Biotechnol 2018; 36: 430-44.
[51]
Farokhi M, Mottaghitalab F, Shokrgozar MA, Kaplan DL, Kim H-W, Kundu SC. Prospects of peripheral nerve tissue engineering using nerve guide conduits based on silk fibroin protein and other biopolymers. Int Mater Rev 2017; 62: 367-91.
[52]
Muzzarelli RA, El Mehtedi M, Bottegoni C, Gigante A. Physical properties imparted by genipin to chitosan for tissue regeneration with human stem cells: A review. Int J Biol Macromol 2016; 93: 1366-81.
[53]
Fouladiha H, Marashi S-A, Shokrgozar MA, Farokhi M, Atashi A. Applications of a metabolic network model of mesenchymal stem cells for controlling cell proliferation and differentiation. Cytotechnology 2018; 70: 331-8.
[54]
Shojaei S, Tafazzoli-Shadpour M, Shokrgozar MA, Haghighipour N, Jahromi FH. Stress phase angle regulates differentiation of human adipose-derived stem cells toward endothelial phenotype. Prog Biomater 2018; 1-11.
[55]
Bakhshandeh B, Zarrintaj P, Oftadeh MO, et al. Tissue engineering; strategies, tissues, and biomaterials. Biotechnol Genet Eng Rev 2018; 1-29.
[56]
Zarrintaj P, Rezaeian I, Bakhshandeh B, Heshmatian B, Ganjali MR. Bio-Conductive Scaffold Based on Agarose-Polyaniline for Tissue Engineering. J Skin Stem Cell 2017; 4: e67394.
[57]
Gholipourmalekabadi M, Zhao S, Harrison BS, Mozafari M, Seifalian AM. Oxygen-generating biomaterials: A new, viable paradigm for tissue engineering? Trends Biotechnol 2016; 34: 1010-21.
[58]
Cheung RCF, Ng TB, Wong JH, Chan WY. Chitosan: An update on potential biomedical and pharmaceutical applications. Mar Drugs 2015; 13: 5156-86.
[59]
Liu X, Ma L, Mao Z, Gao C. Chitosan-based biomaterials for tissue repair and regeneration Chitosan for Biomaterials II. Springer 2011; pp. 81-127.
[60]
Han TTY, Shridhar A, Robb K, Kornmuller A, Brown CF, Flynn LE. Natural materials as smart scaffolds for tissue engineering. Smart Mater Tissue Eng 2016; pp. 124-62.
[61]
Yu LM, Kazazian K, Shoichet MS. Peptide surface modification of methacrylamide chitosan for neural tissue engineering applications. J Biomed Mater ResPart A 2007; 82: 243-55.
[62]
Shirosaki Y, Hayakawa S, Osaka A, et al. Challenges for nerve repair using chitosan-siloxane hybrid porous scaffolds. BioMed Res Int 2014; 153808.
[63]
Cooper A, Bhattarai N, Zhang M. Fabrication and cellular compatibility of aligned chitosan–PCL fibers for nerve tissue regeneration. Carbohydr Polym 2011; 85: 149-56.
[64]
Crompton K, Goud J, Bellamkonda R, et al. Polylysine-functionalised thermoresponsive chitosan hydrogel for neural tissue engineering. Biomater 2007; 28: 441-9.
[65]
Wang G, Wang X, Huang L. Feasibility of chitosan-alginate (Chi-Alg) hydrogel used as scaffold for neural tissue engineering: A pilot study in vitro. Biotechnol Biotechnol Equip 2017; 31: 766-73.
[66]
Lu G, Kong L, Sheng B, Wang G, Gong Y, Zhang X. Degradation of covalently cross-linked carboxymethyl chitosan and its potential application for peripheral nerve regeneration. Eur Polym J 2007; 43: 3807-18.
[67]
Zhang L, Chen Y, Xu H, et al. Preparation and evaluation of an injectable chitosan-hyaluronic acid hydrogel for peripheral nerve regeneration. J Wuhan Uni Technol-Mater Sci Ed 2016; 31: 1401-7.
[68]
Azizi S, Heshmatian B, Amini K, Raisi A, Azimzadeh M. Alpha-lipoic acid loaded in chitosan conduit enhances sciatic nerve regeneration in rat. Iran J Basic Med Sci 2015; 18: 228.
[69]
Thien DVH, Hsiao SW, Ho MH, Li CH, Shih JL. Electrospun chitosan/hydroxyapatite nanofibers for bone tissue engineering. J Mater Sci 2013; 48: 1640-5.
[70]
Zhang Y, Venugopal JR, El-Turki A, Ramakrishna S, Su B, Lim CT. Electrospun biomimetic nanocomposite nanofibers of hydroxyapatite/chitosan for bone tissue engineering. Biomater 2008; 29: 4314-22.
[71]
Frohbergh ME, Katsman A, Botta GP, et al. Electrospun hydroxyapatite-containing chitosan nanofibers crosslinked with genipin for bone tissue engineering. Biomater 2012; 33: 9167-78.
[72]
Jiang T, Nukavarapu SP, Deng M, et al. Chitosan–poly (lactide-co-glycolide) microsphere-based scaffolds for bone tissue engineering: In vitro degradation and in vivo bone regeneration studies. Acta Biomater 2010; 6: 3457-70.
[73]
Xu T, Yang H, Yang D, Yu Z-Z. Polylactic acid nanofiber scaffold decorated with chitosan islandlike topography for bone tissue engineering. ACS Appl Mater Interfaces 2017; 9: 21094-104.
[74]
Hsu Sh, Whu SW, Hsieh SC, Tsai CL, Chen DC, Tan TS. Evaluation of chitosan‐alginate‐hyaluronate complexes modified by an RGD‐containing protein as tissue‐engineering scaffolds for cartilage regeneration. Artif Organs 2004; 28: 693-703.
[75]
Correia CR, Moreira-Teixeira LS, Moroni L, et al. Chitosan scaffolds containing hyaluronic acid for cartilage tissue engineering. Tissue Eng Part C Methods 2011; 17: 717-30.
[76]
Jin R, Teixeira LM, Dijkstra PJ, et al. Injectable chitosan-based hydrogels for cartilage tissue engineering. Biomater 2009; 30: 2544-51.
[77]
Silva JM, Georgi N, Costa R, et al. Nanostructured 3D constructs based on chitosan and chondroitin sulphate multilayers for cartilage tissue engineering. PLoS One 2013; 8: e55451.
[78]
Park H, Choi B, Hu J, Lee M. Injectable chitosan hyaluronic acid hydrogels for cartilage tissue engineering. Acta Biomater 2013; 9: 4779-86.
[79]
Badhe RV, Bijukumar D, Chejara DR, et al. A composite chitosan-gelatin bi-layered, biomimetic macroporous scaffold for blood vessel tissue engineering. Carbohydr Polym 2017; 157: 1215-25.
[80]
Du F, Wang H, Zhao W, et al. Gradient nanofibrous chitosan/poly ɛ-caprolactone scaffolds as extracellular microenvironments for vascular tissue engineering. Biomater 2012; 33: 762-70.
[81]
Zhang L, Ao Q, Wang A, et al. A sandwich tubular scaffold derived from chitosan for blood vessel tissue engineering. J Biomed Mater. Res Part A 2006; 77: 277-84.
[82]
Zhang W, Zhao L, Ma J, et al. Electrospinning of fucoidan/chitosan/poly (vinyl alcohol) scaffolds for vascular tissue engineering. Fibers Polym 2017; 18: 922-32.
[83]
Zhu C, Fan D, Duan Z, et al. Initial investigation of novel human‐like collagen/chitosan scaffold for vascular tissue engineering. J Biomed Mater ResPart A 2009; 89: 829-40.
[84]
Tangsadthakun C, Kanokpanont S, Sanchavanakit N, Banaprasert T, Damrongsakkul S. Properties of collagen/chitosan scaffolds for skin tissue engineering. J Met Mater Miner 2017; 16: 37-44.
[85]
Luangbudnark W, Viyoch J, Laupattarakasem W, Surakunprapha P, Laupattarakasem P. Properties and biocompatibility of chitosan and silk fibroin blend films for application in skin tissue engineering. Sci World J 2012; 2012: 697201.
[86]
Han C-m, Zhang L-p, Sun J-z, Shi H-f, Zhou J, Gao C-Y. Application of collagen-chitosan/fibrin glue asymmetric scaffolds in skin tissue engineering. J Zhejiang Univ Sci B 2010; 11: 524-30.
[87]
Ma L, Gao C, Mao Z, et al. Collagen/chitosan porous scaffolds with improved biostability for skin tissue engineering. Biomater 2003; 24: 4833-41.
[88]
Sarkar SD, Farrugia BL, Dargaville TR, Dhara S. Chitosan–collagen scaffolds with nano/microfibrous architecture for skin tissue engineering. J Biomed Mater Res Part A 2013; 101: 3482-92.
[89]
Fan J, Yang J. Preparation and characterization of a chitosan/galactosylated hyaluronic acid/heparin scaffold for hepatic tissue engineering. J Biomater Sci Polym Ed 2017; 28: 569-81.
[90]
Yang J, Chung TW, Nagaoka M, Goto M, Cho C-S, Akaike T. Hepatocyte-specific porous polymer-scaffolds of alginate/galactosylated chitosan sponge for liver-tissue engineering. Biotechnol Lett 2001; 23: 1385-9.
[91]
Chen F, Tian M, Zhang D, et al. Preparation and characterization of oxidized alginate covalently cross-linked galactosylated chitosan scaffold for liver tissue engineering. Mater Sci Eng C 2012; 32: 310-20.
[92]
Tripathi A, Melo JS. Preparation of a sponge-like biocomposite agarose–chitosan scaffold with primary hepatocytes for establishing an in vitro 3D liver tissue model. RSC Advances 2015; 5: 30701-10.
[93]
Semnani D, Naghashzargar E, Hadjianfar M, et al. Evaluation of PCL/chitosan electrospun nanofibers for liver tissue engineering. Int J Polym Mater Polym Biometer 2017; 66: 149-57.
[94]
Milan PB, Lotfibakhshaiesh N, Joghataie M, Ai J, Pazouki A, Kaplan D, et al. Accelerated wound healing in a diabetic rat model using decellularized dermal matrix and human umbilical cord perivascular cells. Acta Biomater 2016; 45: 234-46.
[95]
Farokhi M, Mottaghitalab F, Fatahi Y, Khademhosseini A, Kaplan DL. Overview of silk fibroin use in wound dressings. Trends Biotechnol 2018; 36(9): 907-22.
[96]
Boateng J, Catanzano O. Advanced therapeutic dressings for effective wound healing-a review. J Pharm Sci 2015; 104: 3653-80.
[97]
Ahmed S, Ikram S. Chitosan based scaffolds and their applications in wound healing. Achv Life Sci 2016; 10: 27-37.
[98]
Dragostin OM, Samal SK, Dash M, et al. New antimicrobial chitosan derivatives for wound dressing applications. Carbohydr Polym 2016; 141: 28-40.
[99]
Sezer AD, Cevher E. Biopolymers as wound healing materials: Challenges and new strategies. Biomater App Nanomed: InTech 2011.
[100]
Patrulea V, Ostafe V, Borchard G, Jordan O. Chitosan as a starting material for wound healing applications. Eur J Pharm Biopharm 2015; 97: 417-26.
[101]
Takeo M, Lee W, Ito M. Wound healing and skin regeneration. Cold Spring Harb Perspect Med 2015; 5: a023267.
[102]
Zhang J, Xia W, Liu P, et al. Chitosan modification and pharmaceutical/biomedical applications. Mar Drugs 2010; 8: 1962-87.
[103]
Liu H, Wang C, Li C, et al. A functional chitosan-based hydrogel as a wound dressing and drug delivery system in the treatment of wound healing. RSC Advances 2018; 8: 7533-49.
[104]
Rahmati M, Pennisi CP, Mobasheri A, Mozafari M. Bioengineered scaffolds for stem cell applications in tissue engineering and regenerative medicine. Adv Exp Med Biol 2018; 1-17.
[105]
Wu Y-Y, Jiao Y-P, Xiao L-L, et al. Experimental study on effects of adipose-derived stem cell–seeded silk fibroin chitosan film on wound healing of a diabetic rat model. Ann Plast Surg 2018; 80: 572.
[106]
Gomathysankar S, Halim AS, Yaacob NS. Proliferation of keratinocytes induced by adipose-derived stem cells on a chitosan scaffold and its role in wound healing, a review. Arch Plast Surg 2014; 41: 452.
[107]
Young HE, Hyer L, Black Jr A.C., Robinson Jr J.S.. Chapter 1: Adult stem Cells: From bench-top to bedside. Tissue regeneration: where nano-structure meets biology: World Sci 2014; p. 1-59
[108]
Shou K, Huang Y, Qi B, et al. Induction of mesenchymal stem cell differentiation in the absence of soluble inducer for cutaneous wound regeneration by a chitin nanofiber‐based hydrogel. J Tissue Eng Regen Med 2018; 12: e867-0.
[109]
Jang H, Kim TW, Yoon S, et al. O-GlcNAc regulates pluripotency and reprogramming by directly acting on core components of the pluripotency network. Cell Stem Cell 2012; 11: 62-74.
[110]
Jayakumar R, Prabaharan M, Kumar PS, Nair S, Tamura H. Biomaterials based on chitin and chitosan in wound dressing applications. Biotechnol Adv 2011; 29: 322-37.
[111]
Tamer TM, Valachová K, Hassan MA, et al. Chitosan/hyaluronan/edaravone membranes for anti-inflammatory wound dressing: In vitro and in vivo evaluation studies. Mater Sci Eng C 2018; 90: 227-35.
[112]
Noori S, Kokabi M, Hassan Z. Poly (vinyl alcohol)/chitosan/honey/clay responsive nanocomposite hydrogel wound dressing. J Appl Polym Sci 2018; 135: 46311.
[113]
Li Q, Lu F, Zhou G, et al. Silver inlaid with gold nanoparticle/chitosan wound dressing enhances antibacterial activity and porosity, and promotes wound healing. Biomacromole 2017; 18: 3766-75.
[114]
Khorasani MT, Joorabloo A, Moghaddam A, Shamsi H. MansooriMoghadam Z. Incorporation of ZnO nanoparticles into heparinised polyvinyl alcohol/chitosan hydrogels for wound dressing application. Int J Biol Macromol 2018; 114: 1203-15.
[115]
Cao Z, Shen Z, Luo X, et al. Citrate-modified maghemite enhanced binding of chitosan coating on cellulose porous membranes for potential application as wound dressing. Carbohydr Polym 2017; 166: 320-8.
[116]
Alavarse AC, de Oliveira Silva FW, Colque JT, et al. Tetracycline hydrochloride-loaded electrospun nanofibers mats based on PVA and chitosan for wound dressing. Mater Sci Eng C 2017; 77: 271-81.
[117]
Baghaie S, Khorasani MT, Zarrabi A, Moshtaghian J. Wound healing properties of PVA/starch/chitosan hydrogel membranes with nano Zinc oxide as antibacterial wound dressing material. J Biomater Sci Polym Ed 2017; 28: 2220-41.
[118]
Bajpai S, Ahuja S, Chand N, Bajpai M. Nano cellulose dispersed chitosan film with Ag NPs/Curcumin: An in vivo study on Albino Rats for wound dressing. Int J Biol Macromol 2017; 104: 1012-9.
[119]
Behera SS, Das U, Kumar A, Bissoyi A, Singh AK. Chitosan/TiO2 composite membrane improves proliferation and survival of L929 fibroblast cells: Application in wound dressing and skin regeneration. Int JF Biol Macromol 2017; 98: 329-40.
[120]
Kaygusuz H, Torlak E, Akın-Evingür G, Özen İ, von Klitzing R, Erim FB. Antimicrobial cerium ion-chitosan crosslinked alginate biopolymer films: a novel and potential wound dressing. Int J Biol Macromol 2017; 105: 1161-5.
[121]
Vedakumari WS, Ayaz N, Karthick AS, Senthil R, Sastry TP. Quercetin impregnated chitosan–fibrin composite scaffolds as potential wound dressing materials—Fabrication, characterization and in vivo analysis. Eur J Pharm Sci 2017; 97: 106-12.
[122]
Zhou X, Wang H, Zhang J, et al. Functional poly (ε-caprolactone)/chitosan dressings with nitric oxide-releasing property improve wound healing. Acta Biomater 2017; 54: 128-37.
[123]
Lu Z, Gao J, He Q, et al. Enhanced antibacterial and wound healing activities of microporous chitosan-Ag/ZnO composite dressing. Carbohydr Polym 2017; 156: 460-9.
[124]
Yoon S-J, Hyun H, Lee D-W, Yang DH. Visible light-cured glycol chitosan hydrogel containing a beta-cyclodextrin-curcumin inclusion complex improves wound healing in vivo. Molecules 2017; 22: 1513.
[125]
Koosehgol S, Ebrahimian-Hosseinabadi M, Alizadeh M, Zamanian A. Preparation and characterization of in situ chitosan/polyethylene glycol fumarate/thymol hydrogel as an effective wound dressing. Mater Sci Eng C 2017; 79: 66-75.
[126]
Karahaliloglu Z, Kilicay E, Denkbas EB. Antibacterial chitosan/silk sericin 3D porous scaffolds as a wound dressing material. Artif Cells Nanomed Biotechnol 2017; 45: 1172-85.
[127]
Liang D, Lu Z, Yang H, Gao J, Chen R. Novel asymmetric wettable AgNPs/chitosan wound dressing: in vitro and in vivo evaluation. ACS Appl Mater Interfaces 2016; 8: 3958-68.
[128]
Farandos NM, Yetisen AK, Monteiro MJ, Lowe CR, Yun SH. Contact lens sensors in ocular diagnostics. Adv Healthc Mater 2015; 4: 792-810.
[129]
Sheardown H. Critical role for drug delivery in the development of new ophthalmic treatments. Future Med Chem 2012; 4: 2123-5.
[130]
Lorentz H, Sheardown H. Ocular delivery of biopharmaceuticals Mucosal delivery of biopharmaceuticals. Springer 2014; pp. 221-59.
[131]
Wadhwa S, Paliwal R, Paliwal SR, Vyas SP. Chitosan and its role in ocular therapeutics. Mini Rev Med Chem 2009; 9: 1639-47.
[132]
Başaran E, Yazan Y. Ocular application of chitosan. Expert Opin Drug Deliv 2012; 9: 701-12.
[133]
Xin-Yuan S, Tian-Wei T. New contact lens based on chitosan/gelatin composites. J Bioact Compat Polym 2004; 19: 467-79.
[134]
Behl G, Iqbal J, O’Reilly NJ, McLoughlin P, Fitzhenry L. Synthesis and characterization of poly (2-hydroxyethylmethacrylate) contact lenses containing chitosan nanoparticles as an ocular delivery system for dexamethasone sodium phosphate. Pharm Res 2016; 33: 1638-48.
[135]
Kim H-J, Zhang K, Moore L, Ho D. Diamond nanogel-embedded contact lenses mediate lysozyme-dependent therapeutic release. ACS Nano 2014; 8: 2998-3005.
[136]
Vil’danova R, Sigaeva N, Kukovinets O, et al. Modification of hyaluronic acid and chitosan, aimed at developing hydrogels for ophthalmology. Russ J Appl Chem 2014; 87: 1547-57.
[137]
Fischak C, Klaus R, Werkmeister RM, et al. Effect of topically administered chitosan-N-acetylcysteine on corneal wound healing in a rabbit model. J Ophthalmol 2017; 2017: 5192924.
[138]
Srivastava GK, Rodriguez-Crespo D, Singh AK, et al. Chitosan feasibility to retain retinal stem cell phenotype and slow proliferation for retinal transplantation. BioMed Res Int 2014; 2014: 287896.
[139]
Kmiec M, Pighinelli L, Tedesco M, Silva M, Reis V. Chitosan-properties and applications in dentistryAdv Tissue Eng Regen Med Open Access 2017; 2: 00035
[140]
Sano H, Shibasaki K-I, Matsukubo T, Takaesu Y. Effect of chitosan rinsing on reduction of dental plaque formation. Bull Tokyo Dent Coll 2003; 44: 9-16.
[141]
Chen C-Y, Chung Y-C. Antibacterial effect of water-soluble chitosan on representative dental pathogens Streptococcus mutans and Lactobacilli brevis. J Appl Oral Sci 2012; 20: 620-7.
[142]
Costa E, Silva S, Madureira A, Cardelle-Cobas A, Tavaria F, Pintado M. A comprehensive study into the impact of a chitosan mouthwash upon oral microorganism’s biofilm formation in vitro. Carbohydr Polym 2014; 101: 1081-6.
[143]
Costa E, Silva S, Costa M, et al. Chitosan mouthwash: Toxicity and in vivo validation. Carbohydr Polym 2014; 111: 385-92.
[144]
Mohire NC, Yadav AV. Chitosan-based polyherbal toothpaste: As novel oral hygiene product. Indian J Dent Res 2010; 21: 380.
[145]
Matsunaga T, Yanagiguchi K, Yamada S, Ohara N, Ikeda T, Hayashi Y. Chitosan monomer promotes tissue regeneration on dental pulp wounds. J Biomed L Mater Res Part A 2006; 76: 711-20.
[146]
Soares DG, Rosseto HL, Basso FG, Scheffel DS, Hebling J. COSTA CAdS. Chitosan-collagen biomembrane embedded with calcium-aluminate enhances dentinogenic potential of pulp cells. Braz Oral Res 2016; 30(1): e54.
[147]
Li F, Liu X, Zhao S, Wu H, Xu HH. Porous chitosan bilayer membrane containing TGF-β1 loaded microspheres for pulp capping and reparative dentin formation in a dog model. Dent Mater 2014; 30: 172-81.
[148]
Perchyonok VT, Felitti R, Zhang S. Chitosan bio-active designer materials and orthodontics: Development and evaluation of novel materials as enamel protective agents. Biointerface Res Appl Chem 2016; 6: 1303-7.
[149]
Li X, Wang X, Zhao T, et al. Guided bone regeneration using chitosan-collagen membranes in dog dehiscence-type defect model. J Oral Maxillofac Surg 2014; 72(304): 1-14.
[150]
Kumar KA, Kumar J, Sarvagna J, Gadde P, Chikkaboriah S. Hemostasis and post-operative care of oral surgical wounds by hemcon dental dressing in patients on oral anticoagulant therapy: A split mouth randomized controlled clinical trial. JCDR 2016; 10: ZC37.
[151]
Ezoddini-Ardakani F, Azam AN, Yassaei S, Fatehi F, Rouhi G. Effects of chitosan on dental bone repair. Health 2011; 3: 200.
[152]
Vaca-Cornejo F, Reyes HM, Jiménez SHD, Velázquez RAL, Jiménez JMD. Pilot Study Using a Chitosan-Hydroxyapatite Implant for Guided Alveolar Bone Growth in Patients with Chronic Periodontitis. J Funct Biomater 2017; 8: 29.
[153]
Suzuki S, Masuda Y, Morisaki H, Yamada Y, Kuwata H, Miyazaki T. The study of chitosan-citrate solution as a root canal irrigant: A preliminary reportOral Hyg Health 2014 ;2: 2332-07021000142
[154]
Sana FA, Yurtsever MÇ, Bayrak GK, Tunçay EÖ, Kiremitçi AS, Gümüşderelioğlu M. Spreading, proliferation and differentiation of human dental pulp stem cells on chitosan scaffolds immobilized with RGD or fibronectin. Cytotechnol 2017; 69: 617-30.
[155]
Hashemi-Beni B, Khoroushi M, Foroughi MR, Karbasi S, Khademi AA. Cytotoxicity assessment of polyhydroxybutyrate/chitosan/ nano-bioglass nanofiber scaffolds by stem cells from human exfoliated deciduous teeth stem cells from dental pulp of exfoliated deciduous tooth. Dent Res J 2018; 15: 136.
[156]
Rysz J, Gluba-Brzózka A, Franczyk B, Jabłonowski Z, Ciałkowska-Rysz A. Novel biomarkers in the diagnosis of chronic kidney disease and the prediction of its outcome. Int J Mol Sci 2017; 18: 1702.
[157]
Jing SB, Li L, Ji D, Takiguchi Y, Yamaguchi T. Effect of chitosan on renal function in patients with chronic renal failure. J Pharm Pharmacol 1997; 49: 721-3.
[158]
Yoon HJ, Moon ME, Park HS, et al. Effects of chitosan oligosaccharide (COS) on the glycerol-induced acute renal failure in vitro and in vivo. Food Chem Toxicol 2008; 46: 710-6.
[159]
Xing R, Liu S, Yu H, et al. Protective effect of sulfated chitosan of C3 sulfation on glycerol-induced acute renal failure in rat kidney. Int J Biol Macromol 2014; 65: 383-8.
[160]
Boure T, Vanholder R. Which dialyser membrane to choose? Nephrol Dial Transplant 2004; 19: 293-6.
[161]
Amiji MM. Permeability and blood compatibility properties of chitosan-poly (ethylene oxide) blend membranes for haemodialysis. Biomater 1995; 16: 593-9.
[162]
Amiji MM. Surface modification of chitosan membranes by complexation-interpenetration of anionic polysaccharides for improved blood compatibility in hemodialysis. J Biomater Sci Polym Ed 1997; 8: 281-98.
[163]
Yu D-G, Jou C-H, Lin W-C, Yang M-C. Surface modification of poly (tetramethylene adipate-co-terephthalate) membrane via layer-by-layer assembly of chitosan and dextran sulfate polyelectrolyte multiplayer. Colloids Surf B 2007; 54: 222-9.
[164]
Zhang Q, Lu X, Yang S, Zhang Q, Zhao L. Preparation of anticoagulant polyvinylidene fluoride hollow fiber hemodialysis membranesBiomed Eng/Biomed Technol 2017; 62: 57-65
[165]
Gao J, Liu R, Wu J, et al. The use of chitosan based hydrogel for enhancing the therapeutic benefits of adipose-derived MSCs for acute kidney injury. Biomate 2012; 33: 3673-81.
[166]
Lesher-Perez SC, Segura T, Moraes C. Getting there is half the battle: Recent advances in delivering therapeutics. Integr Biol 2016; 8: 8-11.
[167]
Liechty WB, Kryscio DR, Slaughter BV, Peppas NA. Polymers for drug delivery systems. Annu Rev Chem Biomol Eng 2010; 1: 149-73.
[168]
Elgadir MA, Uddin MS, Ferdosh S, Adam A, Chowdhury AJK, Sarker MZI. Impact of chitosan composites and chitosan nanoparticle composites on various drug delivery systems: A review. J Food Drug Anal 2015; 23: 619-29.
[169]
Hudson D, Margaritis A. Biopolymer nanoparticle production for controlled release of biopharmaceuticals. Crit Rev Biotechnol 2014; 34: 161-79.
[170]
Amsden B. Novel biodegradable polymers for local growth factor delivery. Eur J Pharm Biopharm 2015; 97: 318-28.
[171]
Li S, Xiong Y, Zhang X. Poloxamer surface modified trimethyl chitosan nanoparticles for the effective delivery of methotrexate in osteosarcoma. Biomed Pharmacother 2017; 90: 872-9.
[172]
Liu S, Yang S, Ho PC. Intranasal administration of carbamazepine-loaded carboxymethyl chitosan nanoparticles for drug delivery to the brain. Asian J Pharm Sci 2018; 13: 72-81.
[173]
García J, Ruiz-Durántez E, Valderruten N. Interpenetrating polymer networks hydrogels of chitosan and poly (2-hydroxyethyl methacrylate) for controlled release of quetiapine. React Funct Polym 2017; 117: 52-9.
[174]
Song M, Li L, Zhang Y, Chen K, Wang H, Gong R. Carboxymethyl-β-cyclodextrin grafted chitosan nanoparticles as oral delivery carrier of protein drugs. React Funct Polym 2017; 117: 10-5.
[175]
Efiana NA, Mahmood A, Lam HT, Zupančič O, Leonaviciute G, Bernkop-Schnürch A. Improved mucoadhesive properties of Self-Nanoemulsifying Drug Delivery Systems (SNEDDS) by introducing acyl chitosan. Int J Pharm 2017; 519: 206-12.
[176]
Mohammed L, Ragab D, Lin S, Said S, Gomaa H, Mequanint K. Preparation and characterization of glycol chitosan-Fe3O4 Core–shell magnetic nanoparticles for controlled delivery of progesterone. J Biomater Tissue Eng 2017; 7: 561-70.
[177]
Zhao Z, He M, Yin L, et al. Biodegradable nanoparticles based on linoleic acid and poly (β-malic acid) double grafted chitosan derivatives as carriers of anticancer drugs. Biomacromol 2009; 10: 565-72.
[178]
Xu J, Strandman S, Zhu JX, Barralet J, Cerruti M. Genipin-crosslinked catechol-chitosan mucoadhesive hydrogels for buccal drug delivery. Biomater 2015; 37: 395-404.
[179]
Akhlaghi SP, Tiong D, Berry RM, Tam KC. Comparative release studies of two cationic model drugs from different cellulose nanocrystal derivatives. EEur J Pharm Biopharm 2014; 88: 207-15.
[180]
Cui F, Qian F, Zhao Z, Yin L, Tang C, Yin C. Preparation, characterization, and oral delivery of insulin loaded carboxylated chitosan grafted poly (methyl methacrylate) nanoparticles. Biomacromol 2009; 10: 1253-8.
[181]
Bhumkar DR, Joshi HM, Sastry M, Pokharkar VB. Chitosan reduced gold nanoparticles as novel carriers for transmucosal delivery of insulin. Pharm Res 2007; 24: 1415-26.
[182]
Ganguly S, Dash AK. A novel in situ gel for sustained drug delivery and targeting. Int J Pharm 2004; 276: 83-92.
[183]
Fischer A. Gene therapy: Myth or reality? C R Biol 2016; 339: 314-8.
[184]
Ramamoorth M, Narvekar A. Non-viral vectors in gene therapy-an overview. JCDR 2015; 9: GE01.
[185]
Sharma D, Singh J. Synthesis and characterization of fatty acid grafted chitosan polymer and their nanomicelles for nonviral gene delivery applications. Bioconjug Chem 2017; 28: 2772-83.
[186]
Kim Y-M, Park S-C, Jang M-K. Targeted gene delivery of polyethyleneimine-grafted chitosan with RGD dendrimer peptide in αvβ3 integrin-overexpressing tumor cells. Carbohydr Polym 2017; 174: 1059-68.
[187]
Sato T, Nakata M, Yang Z, Torizuka Y, Kishimoto S, Ishihara M. In vitro and in vivo gene delivery using chitosan/hyaluronic acid nanoparticles: Influences of molecular mass of hyaluronic acid and lyophilization on the transfection efficiency. J Gene Med 2017; 19(8): e2968.
[188]
Darvishi MH, Nomani A, Hashemzadeh H, Amini M, Shokrgozar MA, Dinarvand R. Targeted DNA delivery using a biotinylated chitosan carrier to cancer cells. Biotechnol Appl Biochem 2017; 64(3): 423-32.
[189]
Meng T, Wu J, Yi H, et al. A spermine conjugated stearic acid-g-chitosan oligosaccharide polymer with different types of amino groups for efficient p53 gene therapy. Colloids Surf B 2016; 145: 695-705.
[190]
Eslaminejad T, Nematollahi-Mahani SN, Ansari M. Cationic β-cyclodextrin–chitosan conjugates as potential carrier for pmCherry-C1 gene delivery. Mol Biotechnol 2016; 58: 287-98.
[191]
Liu X, Mo Y, Liu X, et al. Synthesis, characterisation and preliminary investigation of the haemocompatibility of polyethyleneimine-grafted carboxymethyl chitosan for gene delivery. Mater Sci Eng C 2016; 62: 173-82.
[192]
Picola IPD, Shi Q, Fernandes JC, et al. Chitosan derivatives for gene transfer: effect of phosphorylcholine and diethylaminoethyl grafts on the in vitro transfection efficiency. J Biomater Sci Polym Ed 2016; 27: 1611-30.
[193]
Soofiyani SR, Hallaj-Nezhadi S, Lotfipour F, Hosseini AM, Baradaran B. Gene therapy based on interleukin-12 loaded chitosan nanoparticles in a mouse model of fibrosarcoma. Iran J Basic Med Sci 2016; 19: 1238.
[194]
Kim Y-K, Zhang M, Lu J-J, et al. PK11195-chitosan-graft-polyethylenimine-modified SPION as a mitochondria-targeting gene carrier. J Drug Target 2016; 24: 457-67.
[195]
Siegel RL, Miller KD, Jemal A. Cancer statistics, 2018. CA A Cancer J Clin 2018; 68: 7-30.
[196]
Babu A, Ramesh R. Multifaceted applications of chitosan in cancer drug delivery and therapy. Mar Drugs 2017; 15: 96.
[197]
Barua S, Ramos J, Potta T, et al. Discovery of cationic polymers for non-viral gene delivery using combinatorial approaches. Comb Chem High Throughput Screen 2011; 14: 908-24.
[198]
Yang G, Wang X, Fu S, Tang R, Wang J. pH-triggered chitosan nanogels via an ortho ester-based linkage for efficient chemotherapy. Acta Biomater 2017; 60: 232-43.
[199]
Rastegari B, Karbalaei-Heidari HR, Zeinali S, Sheardown H. The enzyme-sensitive release of prodigiosin grafted β-cyclodextrin and chitosan magnetic nanoparticles as an anticancer drug delivery system: Synthesis, characterization and cytotoxicity studies. Colloids Surf B 2017; 158: 589-601.
[200]
Irvine DJ, Hanson MC, Rakhra K, Tokatlian T. Synthetic nanoparticles for vaccines and immunotherapy. Chem Rev 2015; 115: 11109-46.
[201]
Khalil DN, Smith EL, Brentjens RJ, Wolchok JD. The future of cancer treatment: immunomodulation, CARs and combination immunotherapy. Nat Rev Clin Oncol 2016; 13: 273.
[202]
Meiliana A, Dewi NM, Wijaya A. Cancer Immunotherapy: A Review. The Indonesian Biomed J 2016; 8: 1-20.
[203]
Chen P-G, Huang Z-H, Sun Z-Y, et al. Chitosan nanoparticles based nanovaccines for cancer immunotherapy. Pure Appl Chem 2017; 89: 931-9.
[204]
Temizoz B, Kuroda E, Ishii KJ. Vaccine adjuvants as potential cancer immunotherapeutics. Int Immunol 2016; 28: 329-38.
[205]
Monette A, Ceccaldi C, Assaad E, Lerouge S, Lapointe R. Chitosan thermogels for local expansion and delivery of tumor-specific T lymphocytes towards enhanced cancer immunotherapies. Biomater 2016; 75: 237-49.
[206]
Singh PK, Srivastava AK, Dev A, Kaundal B, Choudhury SR, Karmakar S. 1, 3β-Glucan anchored, paclitaxel loaded chitosan nanocarrier endows enhanced hemocompatibility with efficient anti-glioblastoma stem cells therapy. Carbohydr Polym 2018; 180: 365-75.
[207]
Amini N, Vousooghi N, Alizade A, et al. Transplantation of adipose tissue-derived stem cells into brain through cerebrospinal fluid in rat models: Protocol development and initial outcome data. Curr Stem Cell Res Ther 2018. [Epub ahead of print].
[208]
Kargozar S, Mozafari M, Hashemian SJ, et al. Osteogenic potential of stem cells‐seeded bioactive nanocomposite scaffolds: A comparative study between human mesenchymal stem cells derived from bone, umbilical cord Wharton’s jelly, and adipose tissue. J Biomed Mater Res Part B: B Appl Biomater 2018; 106: 61-72.
[209]
Xu B, Li Y, Deng B, Liu X, Wang L, Zhu QL. Chitosan hydrogel improves mesenchymal stem cell transplant survival and cardiac function following myocardial infarction in rats. Exp Ther Med 2017; 13: 588-94.
[210]
Zhang J, Lu X, Feng G, et al. Chitosan scaffolds induce human dental pulp stem cells to neural differentiation: Potential roles for spinal cord injury therapy. Cell Tissue Res 2016; 366: 129-42.
[211]
Chien Y, Chang Y-L, Li H-Y, et al. Synergistic effects of carboxymethyl-hexanoyl chitosan, cationic polyurethane-short branch PEI in miR122 gene delivery: Accelerated differentiation of iPSCs into mature hepatocyte-like cells and improved stem cell therapy in a hepatic failure model. Acta Biomater 2015; 13: 228-44.
[212]
Hsu S-H, Kuo W-C, Chen Y-T, et al. New nerve regeneration strategy combining laminin-coated chitosan conduits and stem cell therapy. Acta Biomater 2013; 9: 6606-15.
[213]
Alhadrami HA. Biosensors: Classifications, medical applications and future prospective. Biotechnol Appl Biochem 2017; 65(3): 497-508.
[214]
Al-Mokaram A, Amir MA, Yahya R, Abdi MM, Mahmud HNME. The development of non-enzymatic glucose biosensors based on electrochemically prepared polypyrrole–chitosan–titanium dioxide nanocomposite films. Nanomater 2017; 7: 129.
[215]
Fartas FM, Abdullah J, Yusof NA, Sulaiman Y, Saiman MI. Biosensor based on tyrosinase immobilized on graphene-decorated gold nanoparticle/chitosan for phenolic detection in aqueous. Sensors 2017; 17: 1132.
[216]
Teepoo S, Dawan P, Barnthip N. Electrospun chitosan-gelatin biopolymer composite nanofibers for horseradish peroxidase immobilization in a hydrogen peroxide biosensor. Biosens 2017; 7: 47.
[217]
Sorouri R, Bagheri H, Afkhami A, Salimian J. Fabrication of a novel highly sensitive and selective immunosensor for botulinum neurotoxin serotype A based on an effective platform of electrosynthesized gold nanodendrites/chitosan nanoparticles. Sensors 2017; 17: 1074.
[218]
Dong X-X, Yang J-Y, Luo L, et al. Portable amperometric immunosensor for histamine detection using Prussian blue-chitosan-gold nanoparticle nanocomposite films. Biosens Bioelectron 2017; 98: 305-9.
[219]
Hassanein A, Salahuddin N, Matsuda A, Kawamura G, Elfiky M. Fabrication of biosensor based on Chitosan-ZnO/Polypyrrole nanocomposite modified carbon paste electrode for electroanalytical application. Mater Sci Eng C 2017; 80: 494-501.
[220]
Tammam SN, Khalil MA, Gawad EA, Althani A, Zaghloul H, Azzazy HM. Chitosan gold nanoparticles for detection of amplified nucleic acids isolated from sputum. Carbohydr Polym 2017; 164: 57-63.
[221]
Vadivambal R, Jayas DS. Bio-imaging: Principles, techniques, and applications: CRC Press 2015; 1: 381
[222]
Restani RB, Morgado PI, Ribeiro MP, Correia IJ, Aguiar‐Ricardo A, Bonifácio VD. Biocompatible polyurea dendrimers with pH‐dependent fluorescence. Angew Chem Int Ed 2012; 51: 5162-5.
[223]
Pan X, Ren W, Gu L, Wang G, Liu Y. Photoluminescence from chitosan for bio-imaging. Aust J Chem 2014; 67: 1422-6.
[224]
Terai T, Nagano T. Fluorescent probes for bioimaging applications. Curr Opin Chem Biol 2008; 12: 515-21.
[225]
Qian C-G, Chen Y-L, Feng P-J, et al. Conjugated polymer nanomaterials for theranostics. Acta Pharmacol Sin 2017; 38: 764.
[226]
Knight DK, Shapka SN, Amsden BG. Characterization of glycol chitosan: a potential material for use in biomedical and pharmaceutical applications: a comparison of fractionation techniques. ACS Pub 2006; 11: 227-42.
[227]
Rhee J-K, Park OK, Lee A, Yang DH, Park K. Glycol chitosan-based fluorescent theranostic nanoagents for cancer therapy. Mar Drugs 2014; 12: 6038-57.
[228]
Abhari RE, Martins JA, Morris HL, Mouthuy P-A, Carr A. Synthetic sutures: Clinical evaluation and future developments. J Biomater Appl 2017; 32: 410-21.
[229]
Viju S, Thilagavathi G. Effect of chitosan coating on the characteristics of silk-braided sutures. J Ind Text 2013; 42: 256-68.
[230]
Maslova M, Uspenskii S, Gal’Braikh L, Kil’Deeva N. Surgical sutures modified with polysaccharide composites. Fibre Chem 2016; 48: 253-7.
[231]
Hu W, Huang Z-M. Development of Functional Sutures through Electrospinning 2007 16(7): 991-4
[232]
Diggery RC, Grint DT. Catheters: Types, Applications, and Potential Complications. Nova Sci 2012.
[233]
Akaraborworn O. A review in emergency central venous catheterization. Chin J Traumatol 2017; 20: 137-40.
[234]
Anjum S, Singh S, Benedicte L, Roger P, Panigrahi M, Gupta B. Biomodification strategies for the development of antimicrobial urinary catheters: overview and advances. Global Challenges 2017; 2(1): 201700068.
[235]
Martinez LR, Mihu MR, Tar M, et al. Demonstration of antibiofilm and antifungal efficacy of chitosan against candidal biofilms, using an in vivo central venous catheter model. J Infect Dis 2010; 201: 1436-40.
[236]
Cobrado L, Silva-Dias A, Azevedo M, Pina-Vaz C, Rodrigues A. In vivo antibiofilm effect of cerium, chitosan and hamamelitannin against usual agents of catheter-related bloodstream infections. J Antimicrob Chemother 2012; 68: 126-30.
[237]
Mendoza G, Regiel-Futyra A, Tamayo A, et al. Chitosan-based coatings in the prevention of intravascular catheter-associated infections. J Biomater Appl 2018; 32: 725-37.
[238]
Wang DA, Ji J, Sun YH, Yu GH, Feng LX. Blends of stearyl poly (ethylene oxide) coupling‐polymer in chitosan as coating materials for polyurethane intravascular catheters. J Biomed Mater ResPart A 2001; 58: 372-83.
[239]
Yang SH, Lee YSJ, Lin FH, Yang JM, Chen KS. Chitosan/poly (vinyl alcohol) blending hydrogel coating improves the surface characteristics of segmented polyurethane urethral catheters. J Biomed Mater Res B Appl Biomater 2007; 83: 304-13.
[240]
Niemczyk A, El Fray M, Franklin SE. Friction behaviour of hydrophilic lubricious coatings for medical device applications. Tribol Int 2015; 89: 54-61.

Rights & Permissions Print Cite
© 2024 Bentham Science Publishers | Privacy Policy