You are on page 1of 5

Chitosan as healing material: P

a g e | 19

An International Journal
J. Med. & Allied Health Sciences 2(1),19-23, 2013 HATAM Publishers

Journal of Medical & Allied Health Sciences


Journal homepage: http://www.hgpub.com/index.php/jmahs/jmahs.html

Chitosan: A novel material for healing


Anu Gopal , Vinay Kant , Ajai Lawrence Dsilva , Radhika Syam ,Sreela L ,Vijayta Gupta , Dhirendra 1 1 Kumar , Dinesh Kumar
1 1, * 1 2 3 4 5

Division of Pharmacology and Toxicology; Division of Veterinary Biotechnology; Division of Bacteriology IVRI, Bareilly, UP, India, 243122 4 Dept. of Livestock Production Management, NDRI, Karnal, Hariyana, India, 132001 5 Department of Chemistry, University of Jammu *Corresponding author: anugopal04vet@gmail.com ARTICLE INFORMATION Article history
Received 22 February 2013 Revised 30 March 2013 Accepted 11 April 2013 Available online 15 April 2013

ABSTRACT Chitosan is a natural biopolymer derived from chitin and is known in the wound management area for its wide range of properties. It has the ability to affect the functions of various cells involved in wound healing by stimulating the release of various cytokines and growth factors. Also its biodegradability and bio-compatibility facilitates its use as an excellent biomaterial. Various wound healing agents have been recently introduced into market utilizing these properties of chitosan.

Keywords
Chitosan wound healing polymer collagen

2013 HATAM: Publishers. All rights Reserved. 1. Introduction Chitosan, one of the most abundant, renewable, nontoxic and biodegradable carbohydrate polymers, has recently attracted the interest of pharmaceutical, biomedical and food arenas, due to its favourable physico-chemical and biological properties. Chitosan is actually a copolymer of glucosamine and N-acetyl glucosamine units linked by 1-4 glucosidic bonds, obtained by alkaline deacetylation of chitin. The in vitro cytotoxic activity of chitosan was found to be minimal, and also it possess favourable characteristics for promoting physiologically ordered dermal regeneration during wound healing. Wound healing progresses through a series of interdependent and overlapping stages involving a variety of cellular and matrix components, acting together to re-establish the integrity of damaged tissue (Boateng et al., 2008). The role of chitin and

chitosan in wound healing is amazing as evidenced by the recently published scientific papers and patents. In addition to lack of toxicity and allergenicity, its biocompatible, biodegradable and bioactive nature facilitates its use for diverse applications in pharmaceutical and medical fields (Illum, 1998, Singla et al., 2001). Chitosan in addition to all these possess excellent antifungal and antibacterial properties also (Allan et al., 1979, Nelson et al., 1994). Thus these properties could be an added advantage for chitosan based materials to be used in hostile environments of wounds. 2. Desirable properties of chitosan as a wound healing agent 1. Minimal foreign body reaction 2. Availability of chemical side groups for attachment to other molecules

Journal of Medical & Allied Health Sciences, Volume 2, Issue 1, April 2013

Chitosan as healing material: P

a g e | 20

3. 4. 5. 6. 7. 8. 9. 10.

Analgesic Promotion of granulation tissue formation Facilitation of re-epithelization Limitation of scar formation and retraction Hemostatic Antibacterial Mild processing conditions Minimum scar formation

Semi permeable biological dressing using chitosan and its derivatives have shown to prevent dehydration and contamination of the wound, by maintaining a sterile wound exudate under the dry scab, optimizing the conditions for healing. The application of chitosan hydrogels has also been demonstrated to effectively interact with and protect the wound ensuring good moist healing environment (Senel & Mc Clure, 2004). In addition, chitosan modulates the functions of inflammatory cells and subsequently promotes granulation and organization . There is also good evidence that chitosan can beneficially influence every separate stage of wound healing. Peculiarity of chitosan to foster adequate granulation tissue formation accompanied by angiogenesis and regular deposition of thin collagen fibers, further enhances correct repair of dermoepidermal lesions. The main biochemical effects of chitins and chitosans are macrophage and neutrophil activation, cytokine production, giant cell migration and stimulation of type IV collagen synthesis (Shi et al., 2006). 3. Effect of chitosan on various cells involved in wound healing The healing process involves four phases that overlap in time and space: hemostasis, inflammation, proliferation and tissue remodeling, occurring through a complex interplay between various cytokines and growth factors which are produced by the variety of cells involved in healing (Diegelmann & Evans, 2004). So study of the influence of chitosan on these cells has proved to be beneficial in explaining its role in wound healing. In skin, wound healing begins within minutes with hemostasis marked by the appearance of platelets. Chitosan is a hemostat, which helps in natural blood clotting and heamostasis, blocks nerve endings and hence reduces pain (Paul & Sharma 2004). The effects of chitosan on various cells which are impotant in healing are as follows 3.1 Neutrophils Neutrophil influx is an early inflammatory response that is essential for the clearance of bacteria and cellular debris during cutaneous wounding. Accumulation and migration of immune cells like macrophages and neutrophils in the wound bed is thus necessary for bodys defence against such external infections. Studies have shown that wounds applied with chitosan show accumulation of

polymorphonuclear cells, angiogenesis and healing with minimum scarring (Usami et al., 1997). It has been also suggested that the mechanism by which chitin and chitosan act at a wound site involves activation of neutrophils and macrophages, favouring the release of various cytokines and growth factors involved in healing. Histological findings of various studies also suggest that chitosan stimulate the migration of polymorphonuclear and mononuclear cells, and accelerate the reformation of connective tissue and angiogenesis (Mori et al., 1997). Chitosan has been also reported to elicit neutrophil migration that results in the prolonged presence of neutrophils within chitosan matrices (Hidaka et al., 1999, Ueno et al., 1999, VandeVord et al., 2002). Although neutrophil inltration is indicative of inammation, other inammatory symptoms such as erythema, fever, or abscess formation have not been observed when chitosan is implanted in vivo (Minami et al., 1998). Thus chitosan helps to maintain a healthy inflammatory stage of the healing. 3.2 Macrophages Macrophage is one of the most important immunocompetent cell types and has a pivotal role in host defence and immuno-adjusting. Activated macrophages can release cytokines, such as interleukin-1 (IL-1), tumor necrosis factor-alpha (TNF-), various other growth factors and reactive oxygen intermediates, to defend against microbial infections and aid in healing. Macrophage activation is probably the main effect of administration of chitin based dressings causing the production of IL-1 and TNF . Studies have showed that chitin/chitosan derivatives have the ability to up-regulate the production of IL-1 and TNF and colony-stimulating factor by macrophages and also it had an in vitro stimulatory effect on both macrophage nitric oxide (NO) production and chemotaxis (Peluso et al., 1994). The macrophage NO secretion was attributed to the N-acetylglucosamine unit of the chitosan molecule rather than to the glucosamine residue. Thus by having a stimulatory effect on macrophages and neutrophils chitosan effectively act on the inflammatory phase of the healing . 3.3 Fibroblasts The fibroblast is a cell type, which synthesizes various growth factors like TGF-1 and deposits a collagen-rich extracellular matrix helping in the regeneration of a healthy skin. There are several reports about effect of chitosan on fibroblast growth. Even though TGF 1 is one of most important growth factor involved in wound healing studies have demonstrated that excess TGF 1 from fibroblasts in later stages of healing can cause excessive scarring of the wounds (Gauglitz et al., 2011). Various studies have suggested the inability of fibroblast to attach and proliferate in the presence

Journal of Medical & Allied Health Sciences, Volume 2, Issue 1, April 2013

Chitosan as healing material: P

a g e | 21

of chitosan in the later stages of healing which could be beneficial for scarless healing (Malette et al., 1986, Schmidt et al., 1993). But again several studies showed its proliferative effect also, suggesting the degree of deacetylation is a factor determining its effect on fibroblasts. Chitosans with relatively high degrees of deacetylation strongly stimulated fibroblast proliferation while samples with lower levels of deacetylation showed less activity (Howling et al., 2001). The stimulatory effect on fibroblast proliferation required the presence of serum in the culture medium suggesting that the chitosan may be interacting with growth factors present in the serum and potentiating their effect. These studies suggests that chitosan could accelerate wound healing by enhancing the functions of inflammatory cells, such as polymorphonuclear leukocytes (PMN) (phagocytosis, and production of osteopontin and leukotriene B4), macrophages (phagocytosis, and production of IL-1, TGF-1 and Platelet derived growth factor (PDGF)), and fibroblasts (production of IL-8) (Senel & Mc Clure, 2004). As a result, chitosan promotes granulation and organization, and therefore, it is beneficial for open wounds (Ueno et al., 2001 ). Effect of chitosan on various other important cells like keratinocytes, platelets etc also have been studied but the results were found to be variable. 3.4 Effect of chitosan on collagen Collagen is the major constituent of the extracellular matrix. The fibroblasts are responsible for the synthesis of collagen, primarily type I and type III which forms the initial components of granulation tissue of the wound (Desmouliere & Gabbiani, 1994). It plays a pivotal role in maintaining skin structure and accounts for as much as 70% of the weight of the skin. Chitosan will gradually depolymerize to N acetyl D-glucosamine which stimulates the fibroblasts and help in collagen synthesis. It mainly stimulates the type I and type IV collagen synthesis causing a healing with minimum scar formation (Minami et al., 1993). 3.5 Scarless healing An attractive and superior possibility of chitosan compared to other wound healing agents is that it could be used in wound management products to check excessive fibrosis. Fetal wounds heal without fibrosis or scar formation and this peculiarity is attributed to its high hyaluronic acid content in the foetal tissues (Lorenz et al., 1993). Prolonged presence of hyaluronic acid in the matrix of foetal wounds creates an environment that promotes foetal fibroblast movements (Longaker et al., 1991). The chemical structure of chitosan which is similar to that of hyaluronic acid suggesting that it would also create a condition similar to foetal wound healing . Chitosan, is composed of D-glucosamine,

which is also present in disaccharide subunits of hyaluronic acid, thus structurally mimicking hyaluronic acid and could exert similar effects. Early migration of fibroblasts and their proliferation in the wound area is implicated in wound scarring (Jobson et al., 1998). Studies using chitosan-polyvinyl pyrrolidone hydrogels have found to be produce minimum scarring of the wounds by modulating the fibroblast growth and selectively promoting the epithelial growth (Risbud et al., 2000). 3.6 Chitosan combined products for healing To improve the healing process, chitosan has been combined with a variety of modified materials such as growth factors, extra- cellular matrix components and antibacterial agents. Studies have shown that the incorporation of basic fibroblast growth factor (bFGF) into chitosan accelerated the rate of healing (Obara et al., 2003). Various composites based on chitosan also have reported recently like the silver nano composites, with good antibacterial and wound healing potency (Chambers et al., 2007, Fredriksson et al., 2009). These composite materials found to be beneficial in wound healing due to its biocompatibility and ability to provide excellent stability for a sustained release of nanoparticles (Vimala et al., 2010). Also studies have showed that metals with antibacterial + + properties like Cu2 and Ag are having synergistic effects with chitosan causing the formation of efficient composite materials (Leonida et al., 2011). 4. Conclusion So chitosan has attracted much attention not only due to its broad spectrum antibacterial activity but also due to its most abundant natural, renewable, biocompatible, and biodegradable polymeric nature with enormous metal complexation capacity. Wet spun alginate composite containing 0.15 2.0% chitin nanofibrils was characterized in view of its use as a wound dressing. These alginate dressings with chitin nanofibrils were less sucessptible to lysozymes and proved to be efficient for healing (Watthanaphanit et al., 2008). Curcumin coated chitosan-alginate blend also showed superior wound healing abilities when used on in vivo models (Yuvarani et al., 2012). Some wound-covering materials have been developed from chitin nonwoven fabric and polyelectrolyte complexes of chitosan with its own anionic derivatives (sulfated chitosan or N-carbo- xybutyl chitosan) and these materials were also found to be effective in regenerating the wounded skin tissue (Dang & Leong 2006). Chitosan has also proved to have good characteristics for the attachment, proliferation and viability of mesenchymal stem cells, with which it can be considered for use in stem cell therapies cell transplantation and tissue regeneration (Muzzarelli & Muzzarelli, 2008). Various bioactive chitosan scaffold co cultured with keratinocyte and

Journal of Medical & Allied Health Sciences, Volume 2, Issue 1, April 2013

Chitosan as healing material: P

a g e | 22

fibroblast cells have also been introduced recently with accelerated wound closure properties (Paul et al., 2012). Also chitosan appears to have no adverse effects after implantation in tissue and for this reason it has been used for a wide range of biomedical applications such as artificial skin substitute (Lucarotti et al., 2013). So it is clear that chitosan with its unique combination of biological, physical, and chemical properties, present a novel, wound healing biopolymer. References Allan, C. R., & Hadwiger, L. A. (1979). The fungicidal effect of chitosan on fungi of varying cell wall composition. Exp. Mycol., 3, 285-289. Boateng, J. S., Matthews, K. H., Stevens, H. N., & Eccleston, G. M. (2008). Wound healing dressings and drug delivery systems: a review. J. Pharm. Sci., 97(8), 2892-2923. Chambers, H., Dumville, J. C., & Cullum, N. (2007). Silver treatments for leg ulcers: a systematic review. Wou,nd Repair Regen., 15, 165-173. Dang, J. M., & Leong, K. W. (2006). Natural polymers for gene delivery and tissue engineering. Adv. Drug Deliv. Rev., 58(4), 487499. Desmouliere, A., & Gabbiani, G. (1994). Modulation of fibroblastic cytoskeletal features during pathological situations: the role of extracellular matrix and cytokines. Cell Motil. Cytoskelet., 29, 195-203. Diegelmann, R.F., & Evans, M. C. (2004). Wound healing: an overview of acute,fibrotic and delayed healing. Front. Bioci., 9, 263-289. Fredriksson, C., Kratz, G., & Huss, F. (2009). Accumulation of silver and delayed reepithelialization in normal human skin: an exvivo study of different silver dressings. Wounds, 21, 116-123. Gauglitz, G. G., Korting, H. C., Pavicic, T., Ruzicka, T., & Jeschke, M. G. (2011). Hypertrophic Scarring and Keloids: Pathomechanisms, Current and Emerging Treatment Strategies. Mol Med., 17(1-2), 113-125. Hidaka, Y., Ito, M., Mori, K., Yagasaki, H., & Kafrawy, A. H. (1999). Histopathological and immuno histochemical studies of membranes of deacetylated chitin derivatives implanted over rat calvaria. J. Biomed. Mater. Res., 46(3), 418-23. Howling, G. I., Dettmar, P. W., Goddard, P. A., Hampson, F. C., Dornish, M., & Wood, E. J. (2001). The effect of chitin and chitosan on the proliferation of human skin fibroblasts and keratinocytes in vitro. Biomaterials, 22(22), 2959-2966. Illum, L. (1998). Chitosan and its use as a pharmaceutical excipient. Pharm. Res., 15, 1326-1231.

Jobson, T. M., Billington, C. K., & Hall, I. P. (1998). Regulation of proliferation of human colonic subepithelial myofibroblasts by mediators important in intestinal inflammation. J. Clin. Invest., 101, 2650- 2657 Leonida, M. D., Banjade, S., Vo, T., Anderle, G., Haas, G. J., & Philips, N. (2011). Nanocomposite materials with antimicrobial activity based on chitosan. Int. J. Nano Biomaterials., 3(4), 316-334. Longaker, M. T., Chiu, E. S., Adzick, N. S., Stern, M., Harrison, M. R., & Stern, R. (1991). Studies in fetal wound healing. V. A prolonged presence of hyaluronic acid characterizes fetal wound fluid. Ann. Surg., 213, 292-296 Lorenz, H. P., & Adzick, N. S. (1993). Scarless skin wound repair in the fetus. West. J. Med., 159, 350-355. Lucarotti, C., Oddo, C. M., Vitiello, N., & Carrozza, M. C. (2013). Synthetic and bio-artificial tactile sensing: a review. Sensors, 13(2), 1435-66. Malette, W. G., Quigley, H. J., & Adicks, E. D. (1986). From chitosan effect in vascular surgery, Tissue culture and tissue regenration. In Chitin in nature and technology. Edited by Muzzarelli R, Jeuniaux C, Gooday G. New York: Plenum Press, 435442. Minami, S., Okamoto, Y., Tanioka, S., Sashiwa, H., Saimoto, H., Matsuhashi, A., & Shigemasa, Y. (1993). From effects of chitosan on wound healing. In Carbohydrates and carbohydrate st polymers. Vol 1. 1 edition. Edited by Yalpani M. Illinois: ATL Press, 141-152. Minami, S., Suzuki, H., Okamoto, Y., Fujinaga, T., & Shigemasa, Y.(1998). Chitin and chitosan activate complement via the alternative pathway. Carbohydr. Polym., 36(3), 151-155. Mori, T., Okumura, M., Matsuura, M., Ueno, K., Tokura, S., Okamoto, Y,, Minami, S., & Fujinaga, T. (1997). Effects of Chitin and Its Derivatives on the Proliferation and Cytokine Production of Fibroblasts in vitro. Biomaterials, 18, 947- 951. Muzzarelli, R. A. A., & Muzzarelli, C. (2008). From chitin and chitosan hydrogels. In Handbook of hydrocolloids. 2nd edition. Edited by Philips GO, Williams A. Cambridge: Woodhead Publishing Ltd, 368- 370. Nelson, J. L., Alexander, J. W., Gianotti, L., Chalk, C. L., & Pyles, T. (1994). Influence of dietary fiber on microbial growth in vitro and bacterial translocation after burn injury in mice. Nutrition, 10, 32-36. Obara, K., Ishihara, M., Ishizuka, T., Fujita, M., Ozeki, Y., Maehara, T., Saito, Y., Yura, H., Matsui, T., Hattori, H., Kikuchi, M., & Kurita, A. (2003). Photo crosslinkable chitosan hydrogel containing fibroblast growth factor-2 stimulates wound healing in healing-impaired mice. Biomaterials, 24, 34373444.

Journal of Medical & Allied Health Sciences, Volume 2, Issue 1, April 2013

Chitosan as healing material: P

a g e | 23

Paul, W., & Sharma, C. P. (2004). Chitin and alginates wound dressings: A short review. Trends Biomater. Artif. Organs., 18, 18-23. Paul, W., Rekha, M. R., Mohanan, P. V., & Chandra, P. S. (2012). Bioactive chitosan scaffold co cultured with keratinocyte and fibroblast cells. Trends Biomater. Artif. Organs, 26(1), 89-92. Peluso, G., Petillo, O., Ranieri, M., Santin, M., Ambrosio, L., Calabro, D., Avallone, B., & Balsamo, G. (1994). Chitosan-mediated stimulation of macrophage function. Biomaterials, 15(15), 1215-1220. Risbud, M., Hardikar, A., & Bhonde, R. (2000). Growth modulation of fibroblasts by chitosanpolyvinyl pyrrolidone hydrogel: Implications for wound management? J. Biosci., 25(25), 31- 34. Schmidt, R. J., Chung, L. Y., Andews, A. M., Spyratou, O., & Turner, T. D. (1993). Biocompatibility of wound management products: a study of the effects of various polysaccharides on murine L929 fibroblast proliferation and macrophage respiratory burst. J. Pharm. Pharmacol., 45, 508- 513. Senel, S., & Mc Clure, S. J. (2004). Potential applications of chitosan in veterinary medicine. Adv. Drug Deliver. Rev., 56, 1467-1480. Shi, C. M., Zhu, Y., Ran, X. Z., Wang, M., Su, Y., & Cheng, T. M. (2006). Therapeutic potential of chitosan and its derivatives in regenerative medicine. J. Surg. Res., 133, 185192. Singla, A. K., & Chawla, M. (2001). Chitosan: some pharmaceutical and biological aspects-an update. J. Pharm. Pharmacol., 53, 1047-1067. Ueno, H., Mori, T., & Fujinaga, T. (2001).Topical formulations and wound healing applications of chitosan. Adv Drug Deliv Rev., 52(2), 105-115.

Ueno, H., Yamada, H., Tanaka, I., Kaba, N., Matsuura, M., Okumura, M., Kadosawa, T., & Fujinaga, T. (1999). Accelerating effects of chitosan for healing at early phase of experimental open wound in dogs. Biomaterials, 20(15), 14071414. Usami, Y., Okamoto, Y., Minami, S., Matsuhashi, A., & Shigemasa, Y. (1997). Influence of chain length of N-acetyl-D glucosamine and Dglucosamine residues on direct and complement mediated chemotactic activities of canine polymorphonuclear cells. Carbohydr. Polym., 32, 115122 VandeVord, P. J., Matthew, H. W., DeSilva, S. P., Mayton, L., Wu, B., & Wooley, P. H. (2002). Evaluation of the biocompatibility of a chitosan scaffold in mice. J Biomed Mater Res., 59(3), 585- 590. Vimala, K., Mohan, Y. M., Sivudu, K. S., Varaprasad, K., Ravindra, S., Reddy, N. N., Padma, Y., Sreedhar, B., & Mohanaraju, K. (2010). Fabrication of porous chitosan films impregnated with silver nanoparticles: a facile approach for superior antibacterial application. Colloid Surface B., 76(1), 248-258. Watthanaphanit, A., Supaphol, P., Tamura, H., Tokura, S., & Rujiravanit, R. (2008). Fabrication, structure, and properties of chitin whisker-reinforced alginate nanocomposite fibers. J. App. Polym. Sci., 110, 890-899. Yuvarani, I., Senthil, K. S., Jayachandran, V., SeKwon, K., & Sudha P. N. (2012). Preparation and Characterization of Curcumin Coated Chitosan-Alginate Blend for Wound Dressing Application. J. Biomat. Tissue Eng., 2(1), 5460.

Journal of Medical & Allied Health Sciences, Volume 2, Issue 1, April 2013

You might also like