Generic placeholder image

Current Pharmaceutical Design

Editor-in-Chief

ISSN (Print): 1381-6128
ISSN (Online): 1873-4286

General Review Article

Lock Stock and Barrel of Wound Healing

Author(s): Gitika A. Dhingra, Malkiet Kaur, Manjinder Singh, Geeta Aggarwal and Manju Nagpal*

Volume 25, Issue 38, 2019

Page: [4090 - 4107] Pages: 18

DOI: 10.2174/1381612825666190926163431

Price: $65

Abstract

Any kind of injury may lead to wound formation. As per World Health Organization Report, “more than 5 million people die each year due to injuries. This accounts for 9% of the world’s population death, nearly 1.7 times the number of fatalities that result from HIV/AIDS, tuberculosis and malaria combined. In addition, ten million people suffer from non-fatal injuries which require treatment”. This scenario leads to increased health and economic burden worldwide. Rapid wound healing is exigent subject-field in the health care system. It is imperative to be updated on wound care strategies as impaired wound healing may lead to chronic, non-healing wounds and thus further contributes to the national burden. This article is a comprehensive review of wound care strategies. The first and second part of this review article focuses on the understanding of wound, its types and human body’s healing mechanism. Wound healing is natural, highly coordinated process that starts on its own, immediately after the injury. However, individual health condition influences the healing process. Discussion of factors affecting wound healing has also been included. Next part includes the detailed review of diverse wound healing strategies that have already been developed for different types of wound. A detailed description of various polymers that may be used has been discussed. Amongst drug delivery systems, oligomers, dendrimers, films, gels, different nano-formulations, like nanocomposites, nanofibers, nanoemulsions and nanoparticles are discussed. Emphasis on bandages has been made in this article.

Keywords: Chronic wound, wound healing strategies, nanomaterials, smart bandages, pathophysiology, nanoemulsions.

[1]
WHO online [Home page on internet] injury and violence- the facts 2014. [Accessed on 15 March 2019]. Available at https://www.who.int/
[2]
Walshe C. Living with a venous leg ulcer: a descriptive study of patients’ experiences. J Adv Nurs 1995; 22(6): 1092-100.
[http://dx.doi.org/10.1111/j.1365-2648.1995.tb03110.x] [PMID: 8675863]
[3]
Reiber GE, Boyko EJ, Smith DG. Lower extremity foot ulcers and amputations in diabetes. Diabetes in America 1995; 2: 409-27.
[4]
Diabetes in north america: millions of feet at risk of amputations 65th annual sessions of the american diabetes association. International Diabetes Federation. San Diego 2005.
[5]
Tripathy JP, Jagnoor J, Prasad BM, Ivers R. Cost of injury care in India: cross-sectional analysis of national sample survey 2014. Inj Prev 2018; 24(2): 116-22.
[http://dx.doi.org/10.1136/injuryprev-2017-042318] [PMID: 28724552]
[6]
Frykberg RG, Banks J. Challenges in the treatment of chronic wounds. Adv Wound Care (New Rochelle) 2015; 4(9): 560-82.
[http://dx.doi.org/10.1089/wound.2015.0635] [PMID: 26339534]
[7]
Posnett J, Franks PJ. The burden of chronic wounds in the UK. Nurs Times 2008; 104(3): 44-5.
[PMID: 18293879]
[8]
Augustin M, Brocatti LK, Rustenbach SJ, Schäfer I, Herberger K. Cost-of-illness of leg ulcers in the community. Int Wound J 2014; 11(3): 283-92.
[http://dx.doi.org/10.1111/j.1742-481X.2012.01089.x] [PMID: 23020710]
[9]
Andreu V, Mendoza G, Arruebo M, Irusta S. Smart dressings based on nanostructured fibers containing natural origin antimicrobial, anti-inflammatory, and regenerative compounds. Materials (Basel) 2015; 8(8): 5154-93.
[http://dx.doi.org/10.3390/ma8085154] [PMID: 28793497]
[10]
Merriam Webster online [homepage on internet] dictionary-wound [accessed on 15th march 2019]. Available at https://www.merrian.webster.com
[11]
Qin Y, Ed. Medical Textile Materials. USA: Woodhead Publishing 2015.
[12]
Rajendran S, Ed. Advanced textiles for wound care. USA: Woodhead Publishing 2018.
[13]
Wound management and prevention [homepage on internet] Case closed: A dressing for chronic and acute wounds [accessed on 16 march 2019] available at https://www.o-wn.com
[14]
Simmons BP. Guideline for prevention of surgical wound infections. Infect Control Hosp Epidemiol 1982; 3(S2): S188-96.
[http://dx.doi.org/10.1017/S0195941700056733]
[15]
Cho H, Blatchley MR, Duh EJ, Gerecht S. Acellular and cellular approaches to improve diabetic wound healing. Adv Drug Deliv Rev 2019; 146: 267-88.
[http://dx.doi.org/10.1016/j.addr.2018.07.019] [PMID: 30075168]
[16]
Rajkumar S. Nadar, Selvakumar. Nanotechnology in wound healing- a review. Glob J Nanomed 2017; 3(1): 1-4.
[17]
Shrivastav A, Mishra AK, Ali SS, Ahmad A, Abuzinadah MF, Khan NA. In vivo models for assesment of wound healing potential: a systematic review. Wound medicine 2018; 20:43-53. [http://dx.doi.org/10.1016/j.wndm.2018.01.003] [18] Wound source online [home page on internet] the four stages of wound healing. [Accessed on 20 march 2019]. Available at
[19]
Beck E, Duckert F, Ernst M. The influence of fibrin stabilizing factor on the growth of fibroblats in vitro and wound healing. Thromb Haemost 1961; 8(02): 485-91.
[http://dx.doi.org/10.1055/s-0038-1654579]
[20]
Kalashnikova I, Das S, Seal S. Nanomaterials for wound healing: Scope and advancement. Nanomedicine (Lond) 2015; 10(16): 2593-612.
[http://dx.doi.org/10.2217/nnm.15.82] [PMID: 26295361]
[21]
Guo S, Dipietro LA. Factors affecting wound healing. J Dent Res 2010; 89(3): 219-29.
[http://dx.doi.org/10.1177/0022034509359125] [PMID: 20139336]
[22]
Wound source online [home page on internet] Factors effecting wound healing in chronic wounds [Accessed on 20 march 2019] available at https://www.woundsource.com
[23]
Wound source online [home page on internet] wound characteristics that affect wound healing [Accessed on 21 march 2019] available at https://woundsource.com
[24]
Elite learning online [home page on internet] 10 factors that affect the wound healing process all nurses should know [Accessed on 21st march 2019] available at https://www.elitecme.com
[25]
Wilson JA, Clark JJ. Obesity: impediment to postsurgical wound healing. Adv Skin Wound Care 2004; 17(8): 426-35.
[http://dx.doi.org/10.1097/00129334-200410000-00013] [PMID: 15492679]
[26]
Centers for disease control and prevention [home page on internet] Division of nutrition, physical activity and obesity [Accessed on 30th march 2019] available at https://www.cdc.gov
[27]
Pharmaceutical Press; 1990 [home page on internet] stephen thomas. Wound management and dressings [Accessed on 30th march 2019] available at. www.researchgate.net
[28]
Bhat S, Kumar A. Biomaterials and bioengineering tomorrow’s healthcare. Biomatter 2013; 3(3)e24717
[http://dx.doi.org/10.4161/biom.24717] [PMID: 23628868]
[29]
Ngo YH, Li D, Simon GP, Garnier G. Paper surfaces functionalized by nanoparticles. Adv Colloid Interface Sci 2011; 163(1): 23-38.
[http://dx.doi.org/10.1016/j.cis.2011.01.004] [PMID: 21324427]
[30]
Ramasamy M, Lee J. Recent nanotechnology approaches for prevention and treatment of biofilm-associated infections on medical devices. BioMed Res Int 2016; 20161851242
[http://dx.doi.org/10.1155/2016/1851242]
[31]
Younan GJ, Heit YI, Dastouri P, et al. Mast cells are required in the proliferation and remodeling phases of microdeformational wound therapy Plast Reconstr Surg 2011; 128(6): e 649-58.
[http://dx.doi.org/10.1097/PRS.0b013e318230c55d] [PMID: 22094766]
[32]
Heit YI, Dastouri P, Helm DL, et al. Foam pore size is a critical interface parameter of suction-based wound healing devices. Plast Reconstr Surg 2012; 129(3): 589-97.
[http://dx.doi.org/10.1097/PRS.0b013e3182402c89] [PMID: 22090246]
[33]
Rajendran NK, Kumar SS, Houreld NN, Abrahamse H. A review on nanoparticle based treatment for wound healing. J Drug Deliv Sci Technol 2018; 44: 421-30.
[http://dx.doi.org/10.1016/j.jddst.2018.01.009]
[34]
Dai T, Tanaka M, Huang YY, Hamblin MR. Chitosan preparations for wounds and burns: antimicrobial and wound-healing effects. Expert Rev Anti Infect Ther 2011; 9(7): 857-79.
[http://dx.doi.org/10.1586/eri.11.59] [PMID: 21810057]
[35]
Ahmed S, Ikram S. Chitosan based scaffolds and their applications in wound healing Achievements in the life sciences 2016; 10(1): 27-37.
[http://dx.doi.org/10.1016/j.als.2016.04.001]
[36]
Ghica MV, Albu MG, Leca M, Popa L, Moisescu ST. Design and optimization of some collagen-minocycline based hydrogels potentially applicable for the treatment of cutaneous wound infections. Pharmazie 2011; 66(11): 853-61.
[PMID: 22204131]
[37]
Kempf M, Miyamura Y, Liu PY, et al. A denatured collagen microfiber scaffold seeded with human fibroblasts and keratinocytes for skin grafting. Biomaterials 2011; 32(21): 4782-92.
[http://dx.doi.org/10.1016/j.biomaterials.2011.03.023] [PMID: 21477857]
[38]
Mitra T, Sailakshmi G, Gnanamani A, et al. Preparation and characterization of a thermostable and biodegradable biopolymers using natural cross-linker. Int J Biol Macromol 2011; 48(2): 276-85.
[http://dx.doi.org/10.1016/j.ijbiomac.2010.11.011] [PMID: 21126533]
[39]
Kohane DS, Langer R. Polymeric biomaterials in tissue engineering. Pediatr Res 2008; 63(5): 487-91.
[http://dx.doi.org/10.1203/01.pdr.0000305937.26105.e7] [PMID: 18427292]
[40]
[41]
Burkatovskaya M, Tegos GP, Swietlik E, Demidova TNP, Castano A, Hamblin MR. Use of chitosan bandage to prevent fatal infections developing from highly contaminated wounds in mice. Biomaterials 2006; 27(22): 4157-64.
[http://dx.doi.org/10.1016/j.biomaterials.2006.03.028] [PMID: 16616364]
[42]
Mohanty C, Das M, Sahoo SK. Sustained wound healing activity of curcumin loaded oleic acid based polymeric bandage in a rat model. Mol Pharm 2012; 9(10): 2801-11.
[http://dx.doi.org/10.1021/mp300075u] [PMID: 22946786]
[43]
Lowe A, Bills J, Verma R, Lavery L, Davis K, Balkus KJ Jr. Electrospun nitric oxide releasing bandage with enhanced wound healing. Acta Biomater 2015; 13: 121-30.
[http://dx.doi.org/10.1016/j.actbio.2014.11.032] [PMID: 25463501]
[44]
Mu X, Yu H, Zhang C, et al. Nano-porous nitrocellulose liquid bandage modulates cell and cytokine response and accelerates cutaneous wound healing in a mouse model. Carbohydr Polym 2016; 136: 618-29.
[http://dx.doi.org/10.1016/j.carbpol.2015.08.070] [PMID: 26572394]
[45]
Huebner KL, Kunkel AK, McConnel CS, Callan RJ, Dinsmore RP, Caixeta LS. Evaluation of horn bud wound healing following cautery disbudding of preweaned dairy calves treated with aluminum-based aerosol bandage. J Dairy Sci 2017; 100(5): 3922-9.
[http://dx.doi.org/10.3168/jds.2016-12192] [PMID: 28259415]
[46]
Khatami M, Varma RS, Zafarnia N, Yaghoobi H, Sarani M, Kumar VG. Applications of green synthesized Ag, ZnO and Ag/ZnO nanoparticles for making clinical antimicrobial wound-healing bandages. Sustainable Chemistry and Pharmacy 2018; 10: 9-15.
[http://dx.doi.org/10.1016/j.scp.2018.08.001]
[47]
Pal A, Goswami D, Cuellar HE, Castro B, Kuang S, Martinez RV. Early detection and monitoring of chronic wounds using low-cost, omniphobic paper-based smart bandages. Biosens Bioelectron 2018; 117: 696-705.
[http://dx.doi.org/10.1016/j.bios.2018.06.060] [PMID: 30014943]
[48]
BASF global online [home page on internet] topical gels [Accessed on 25th march 2019] available at https://pharmaceutical.basf.com
[49]
Avinash S, Gowda DV, Suresh J, Ram AS, Srivastava A, Osmani RM. Formulation and evaluation of topical gel using eupatorium glandulosum michx for wound healing activity. Schol Res Lib 2016; 8(8): 255-66.
[50]
Schoukens G. Bioactive dressings to promote wound healing Advanced textiles for wound care. Woodhead Publishing 2009; pp. 114-52.
[http://dx.doi.org/10.1533/9781845696306.1.114]
[51]
Dhivya S, Padma VV, Santhini E. Wound dressings - a review. Biomedicine (Taipei) 2015; 5(4): 22.
[http://dx.doi.org/10.7603/s40681-015-0022-9] [PMID: 26615539]
[52]
Patel NA, Patel M, Patel RP. Formulation and evaluation of polyherbal gel for wound healing. Int Res J Pharm 2011; 1(1): 15-20.
[53]
Aly UF. Preparation and evaluation of novel topical gel preparations for wound healing in diabetics. Int J Pharm Pharm Sci 2012; 4(4): 76.
[54]
Khan AW, Kotta S, Ansari SH, Sharma RK, Kumar A, Ali J. Formulation development, optimization and evaluation of aloe vera gel for wound healing. Pharmacogn Mag 2013; 9(Suppl. 1): S6-S10.
[http://dx.doi.org/10.4103/0973-1296.117849] [PMID: 24143047]
[55]
Patil S, George T, Mahadik K. Green synthesized nanosilver loaded silk fibroin gel for enhanced wound healing. J Drug Deliv Sci Technol 2015; 30: 30-6.
[http://dx.doi.org/10.1016/j.jddst.2015.09.001]
[56]
Rao BN, Fathima SR, Viswanath V, Prakash KG, Padmini DS, Reddy PS. Novel citric acid dendritic hydrogels for the delivery of econazole nitrate and its antifungal activity. J Applied Pharm Sci 2016; 6(12): 094-102.
[http://dx.doi.org/10.7324/JAPS.2016.601213]
[57]
Ramane S, Syed V, Biyani K. Evaluation of wound healing activity of polyherbal gel a novel herbal formulation. Int J Res Pharm Biomed Sci 2011; 3: 39-42.
[58]
Jain S, Jain DK. Formulation and evaluation of herbal gel for wound healing. J Drug Deliv Ther 2017; 7(7): 32-4.
[59]
Devi DR, Lakshna SS, Parvathi SV, Hari BV. Investigation of wound healing effect of topical gel of Albizia amara leaves extract. S Afr J Bot 2018; 119: 400-9.
[http://dx.doi.org/10.1016/j.sajb.2018.10.005]
[60]
Ferreira MO, de Lima IS, Morais AÍ, et al. Chitosan associated with chlorhexidine in gel form: synthesis, characterization and healing wounds applications. J Drug Deliv Sci Technol 2019; 49: 375-82.
[http://dx.doi.org/10.1016/j.jddst.2018.12.003]
[61]
Shao F, Yang A, Yu DM, Wang J, Gong X, Tian HX. Bio-synthesis of Barleria gibsoni leaf extract mediated zinc oxide nanoparticles and their formulation gel for wound therapy in nursing care of infants and children. J Photochem Photobiol B 2018; 189: 267-73.
[http://dx.doi.org/10.1016/j.jphotobiol.2018.10.014] [PMID: 30419522]
[62]
Jangde R, Srivastava S, Singh MR, Singh D. In vitro and in vivo characterization of quercetin loaded multiphase hydrogel for wound healing application. Int J Biol Macromol 2018; 115: 1211-7.
[http://dx.doi.org/10.1016/j.ijbiomac.2018.05.010] [PMID: 29730004]
[63]
Alvarado-Gomez E, Martínez-Castañon G, Sanchez-Sanchez R, Ganem-Rondero A, Yacaman MJ, Martinez-Gutierrez F. Evaluation of anti-biofilm and cytotoxic effect of a gel formulation with Pluronic F-127 and silver nanoparticles as a potential treatment for skin wounds. Mater Sci Eng C 2018; 92: 621-30.
[http://dx.doi.org/10.1016/j.msec.2018.07.023] [PMID: 30184789]
[64]
Dev SK, Choudhury PK, Srivastava R, Sharma M. Antimicrobial, anti-inflammatory and wound healing activity of polyherbal formulation. Biomed Pharmacother 2019; 111: 555-67.
[http://dx.doi.org/10.1016/j.biopha.2018.12.075] [PMID: 30597309]
[65]
Li W, Gao F, Kan J, Deng J, Wang B, Hao S. Synthesis and fabrication of a keratin-conjugated insulin hydrogel for the enhancement of wound healing. Colloids Surf B Biointerfaces 2019; 175: 436-44.
[http://dx.doi.org/10.1016/j.colsurfb.2018.12.020] [PMID: 30562718]
[66]
Kulshrestha S, Chawla R, Alam MT, Adhikari JS, Basu M. Efficacy and dermal toxicity analysis of Sildenafil citrate based topical hydrogel formulation against traumatic wounds. Biomed Pharmacother 2019; 112108571
[http://dx.doi.org/10.1016/j.biopha.2019.01.032] [PMID: 30798130]
[67]
Cardoso AM, de Oliveira EG, Coradini K, et al. Chitosan hydrogels containing nanoencapsulated phenytoin for cutaneous use: skin permeation/penetration and efficacy in wound healing. Mater Sci Eng C 2019; 96: 205-17.
[http://dx.doi.org/10.1016/j.msec.2018.11.013] [PMID: 30606527]
[68]
de Sousa FD, Vasconselos PD, da Silva AFB, et al. Hydrogel and membrane scaffold formulations of frutalin (breadfruit lectin) within a polysaccharide galactomannan matrix have potential for wound healing. Int J Biol Macromol 2019; 121: 429-42.
[http://dx.doi.org/10.1016/j.ijbiomac.2018.10.050] [PMID: 30326222]
[69]
Yu JR, Navarro J, Coburn JC, et al. Current and future perspectives on skin tissue engineering: key feature of biomedical research translation assessment and clinical application. Adv Healthc Mater 2019; 8(5)e1801471
[http://dx.doi.org/10.1002/adhm.201801471] [PMID: 30707508]
[70]
TVL HB. Vidyavathi M, Kavitha K, Sastry TP, Suresh-Kumar RV. Preparation and evaluation of chitosangelatin composite films for wound healing activity. Trends Biomater Artif Organs 2010; 24(3): 123-30.
[71]
Renuka M, Nishadh P, Jigar S, Tejal M. Mucoadhesive wound healing film of doxycycline hydrochloride. Int J Drug Dev and Res 2012; 4: 128-40.
[72]
Qureshi MA, Khatoon F. Development of citric acid cross linked poly (vinyl alcohol) hydrogel film, its degradability and effect of temperature, pH. Adv Sci Lett 2014; 20(7-8): 1414-9.
[http://dx.doi.org/10.1166/asl.2014.5546]
[73]
Ojeda-Martínez ML, Yáñez-Sánchez I, Velásquez-Ordoñez C, et al. Skin wound healing with chitosan thin films containing supported silver nanospheres. J Bioact Compat Polym 2015; 30(6): 617-32.
[http://dx.doi.org/10.1177/0883911515590495]
[74]
Devi N, Dutta J. Preparation and characterization of chitosan-bentonite nanocomposite films for wound healing application Int J Biol Macromol 2017; 104(Pt B): 1897-904.
[http://dx.doi.org/10.1016/j.ijbiomac.2017.02.080] [PMID: 28242331]
[75]
Campos MG, Rawls HR, Mei LH, Satsangi N. In vitro wound healing model: effects of chitosan films loaded with gentamicin and silver sulfadiazine on the wound filling rate Adv Tissue Eng Regen Med Open Access 2017; 2(3): 00031
[http://dx.doi.org/10.15406/atroa.2017.02.00031]
[76]
Zou Q, Cai B, Li J, Li J, Li Y. In vitro and in vivo evaluation of the chitosan/Tur composite film for wound healing applications. J Biomater Sci Polym Ed 2017; 28(7): 601-15.
[http://dx.doi.org/10.1080/09205063.2017.1289036] [PMID: 28277010]
[77]
Patel S, Srivastava S, Singh MR, Singh D. Preparation and optimization of chitosan-gelatin films for sustained delivery of lupeol for wound healing Int J Biol Macromol 2018; 107(Pt B): 1888-97.
[http://dx.doi.org/10.1016/j.ijbiomac.2017.10.056] [PMID: 29037874]
[78]
Rostami H, Mohammadi R, Asri-Rezaei S, Tehrani AA. Evaluation of application of chitosan/nano sodium selenite biodegradable film on full thickness excisional wound healing in rats. Iran J Vet Surg 2018; 13(1): 14-22.
[79]
Yasuda K, Ogushi M, Nakashima A, Nakano Y, Suzuki K. Accelerated wound healing on the skin using a film dressing with β-glucan paramylon. In Vivo 2018; 32(4): 799-805.
[80]
Liu X, Hao W, Lok CN, Wang YC, Zhang R, Wong KK. Dendrimer encapsulation enhances anti-inflammatory efficacy of silver nanoparticles. J Pediatr Surg 2014; 49(12): 1846-51.
[http://dx.doi.org/10.1016/j.jpedsurg.2014.09.033] [PMID: 25487498]
[81]
Deng X, Li X, Chen W, Zhao T, Huang W, Qian H. Design, synthesis and biological evaluation of peptide dendrimers with wound healing promoting activity. Med Chem Res 2017; 26(3): 580-6.
[http://dx.doi.org/10.1007/s00044-016-1777-6]
[82]
Zhou T, Chen K, Kong LM, et al. Synthesis, iron binding and antimicrobial properties of hexadentate 3-hydroxypyridinones-terminated dendrimers. Bioorg Med Chem Lett 2018; 28(14): 2504-12.
[http://dx.doi.org/10.1016/j.bmcl.2018.05.058] [PMID: 29886020]
[83]
Kang YO, Jung JY, Cho D, Kwon OH, Cheon JY, Park WH. Antimicrobial silver chloride nanoparticles stabilized with chitosan oligomer for the healing of burns. Materials (Basel) 2016; 9(4): 215.
[http://dx.doi.org/10.3390/ma9040215] [PMID: 28773340]
[84]
Nguyen MH, Lee SE, Tran TT, et al. A simple strategy to enhance the in vivo wound-healing activity of curcumin in the form of self-assembled nanoparticle complex of curcumin and oligochitosan. Mater Sci Eng C 2019; 98: 54-64.
[http://dx.doi.org/10.1016/j.msec.2018.12.091] [PMID: 30813056]
[85]
Nikita G, Vivek P, Chhaya G. Wound-healing activity of an oligomer of alkannin/shikonin, isolated from root bark of onosma echioides. Nat Prod Res 2015; 29(16): 1584-8.
[http://dx.doi.org/10.1080/14786419.2014.986126] [PMID: 25572039]
[86]
Ma H, Liu Y, Zhu C, et al. Multivalent urea bond assembly of polyacrylate oligomers with improved mechanical strength and high self-healing efficiency. React Funct Polym 2019; 137: 79-87.
[http://dx.doi.org/10.1016/j.reactfunctpolym.2018.12.023]
[87]
Aghamohamadi N, Sanjani NS, Majidi RF, Nasrollahi SA. Preparation and characterization of aloe vera acetate and electrospinning fibers as promising antibacterial properties materials. Mater Sci Eng C 2019; 94: 445-52.
[http://dx.doi.org/10.1016/j.msec.2018.09.058] [PMID: 30423728]
[88]
Moradkhannejhad L, Abdouss M, Nikfarjam N, Mazinani S, Heydari V. Electrospinning of zein/propolis nanofibers; antimicrobial properties and morphology investigation. J Mater Sci Mater Med 2018; 29(11): 165.
[http://dx.doi.org/10.1007/s10856-018-6174-x] [PMID: 30392146]
[89]
Chen JP, Chiang Y. Bioactive electrospun silver nanoparticles-containing polyurethane nanofibers as wound dressings. J Nanosci Nanotechnol 2010; 10(11): 7560-4.
[http://dx.doi.org/10.1166/jnn.2010.2829] [PMID: 21137982]
[90]
Xu C, Xu F, Wang B, Lu T. Electrospinning of poly (ethylene-co-vinyl alcohol) nanofibres encapsulated with Ag nanoparticles for skin wound healing. J Nanomater 2011; 2011: 3.
[http://dx.doi.org/10.1155/2011/201834]
[91]
Shalumon KT, Anulekha KH, Nair SV, Nair SV, Chennazhi KP, Jayakumar R. Sodium alginate/poly(vinyl alcohol)/nano ZnO composite nanofibers for antibacterial wound dressings. Int J Biol Macromol 2011; 49(3): 247-54.
[http://dx.doi.org/10.1016/j.ijbiomac.2011.04.005] [PMID: 21635916]
[92]
Charernsriwilaiwat N, Opanasopit P, Rojanarata T, Ngawhirunpat T. Lysozyme-loaded, electrospun chitosan-based nanofiber mats for wound healing. Int J Pharm 2012; 427(2): 379-84.
[http://dx.doi.org/10.1016/j.ijpharm.2012.02.010] [PMID: 22353400]
[93]
Augustine R, Dominic EA, Reju I, Kaimal B, Kalarikkal N, Thomas S. Electrospun polycaprolactone membranes incorporated with ZnO nanoparticles as skin substitutes with enhanced fibroblast proliferation and wound healing. RSC Advances 2014; 4(47): 24777-85.
[http://dx.doi.org/10.1039/c4ra02450h]
[94]
Monteiro N, Martins M, Martins A, et al. Antibacterial activity of chitosan nanofiber meshes with liposomes immobilized releasing gentamicin. Acta Biomater 2015; 18: 196-205.
[http://dx.doi.org/10.1016/j.actbio.2015.02.018] [PMID: 25749293]
[95]
Safaee-Ardakani MR, Hatamian-Zarmi A, Sadat SM, et al. Electrospun schizophyllan/polyvinyl alcohol blend nanofibrous scaffold as potential wound healing. Int J Biol Macromol 2019; 127: 27-38.
[http://dx.doi.org/10.1016/j.ijbiomac.2018.12.256] [PMID: 30597239]
[96]
Es-haghi A, Mashreghi M, Bazaz MR, Homayouni-Tabrizi M, Darroudi M. Fabrication of biopolymer based nanocomposite wound dressing: evaluation of wound healing properties and wound microbial load IET nanobiotechnology 2016; 11(5): 517-22.
[97]
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(6): 3958-68.
[http://dx.doi.org/10.1021/acsami.5b11160] [PMID: 26800283]
[98]
Akturk O, Kismet K, Yasti AC, et al. Collagen/gold nanoparticle nanocomposites: a potential skin wound healing biomaterial. J Biomater Appl 2016; 31(2): 283-301.
[http://dx.doi.org/10.1177/0885328216644536] [PMID: 27095659]
[99]
Sathiyaseelan A, Shajahan A, Kalaichelvan PT, Kaviyarasan V. Fungal chitosan based nanocomposites sponges-An alternative medicine for wound dressing Int J Biol Macromol 2017; 104(Pt B): 1905-15.
[http://dx.doi.org/10.1016/j.ijbiomac.2017.03.188] [PMID: 28373049]
[100]
Raja IS, Fathima NN. Gelatin-cerium oxide nanocomposite for enhanced excisional wound healing. ACS Applied Bio Materials 2018; 1(2): 487-95.
[http://dx.doi.org/10.1021/acsabm.8b00208]
[101]
Hernández Martínez SP, Rivera González TI, Franco Molina MA, et al. A novel gold calreticulin nanocomposite based on chitosan for wound healing in a diabetic mice model. Nanomaterials (Basel) 2019; 9(1): 75.
[http://dx.doi.org/10.3390/nano9010075] [PMID: 30625974]
[102]
Science direct online [Home page on internet] Nanoemulsions [Accessed on 2nd April 2019] available at https://www.sciencedirect.com
[103]
Song Z, Sun H, Yang Y, et al. Enhanced efficacy and anti-biofilm activity of novel nanoemulsions against skin burn wound multi-drug resistant MRSA infections. Nanomedicine (Lond) 2016; 12(6): 1543-55.
[http://dx.doi.org/10.1016/j.nano.2016.01.015] [PMID: 26961464]
[104]
Alam P, Ansari MJ, Anwer MK, Raish M, Kamal YK, Shakeel F. Wound healing effects of nanoemulsion containing clove essential oil. Artif Cells Nanomed Biotechnol 2017; 45(3): 591-7.
[http://dx.doi.org/10.3109/21691401.2016.1163716] [PMID: 28211300]
[105]
Thomas L, Zakir F, Mirza MA, Anwer MK, Ahmad FJ, Iqbal Z. Development of curcumin loaded chitosan polymer based nanoemulsion gel: in vitro, ex vivo evaluation and in vivo wound healing studies. Int J Biol Macromol 2017; 101: 569-79.
[http://dx.doi.org/10.1016/j.ijbiomac.2017.03.066] [PMID: 28322948]
[106]
Shanmugapriya K, Kim H, Saravana PS, Chun BS, Kang HW. Astaxanthin-alpha tocopherol nanoemulsion formulation by emulsification methods: investigation on anticancer, wound healing, and antibacterial effects. Colloids Surf B Biointerfaces 2018; 172: 170-9.
[http://dx.doi.org/10.1016/j.colsurfb.2018.08.042] [PMID: 30172200]
[107]
Bonferoni MC, Riva F, Invernizzi A, et al. Alpha tocopherol loaded chitosan oleate nanoemulsions for wound healing. Evaluation on cell lines and ex vivo human biopsies, and stabilization in spray dried trojan microparticles. Eur J Pharm Biopharm 2018; 123: 31-41.
[http://dx.doi.org/10.1016/j.ejpb.2017.11.008] [PMID: 29155053]
[108]
Konop M, Damps T, Misicka A, Rudnicka L. Certain aspects of silver and silver nanoparticles in wound care: a minireview. J Nanomater 2016; 2016: 47.
[http://dx.doi.org/10.1155/2016/7614753]
[109]
Ahmadi M, Adibhesami M. The effect of silver nanoparticles on wounds contaminated with pseudomonas aeruginosa in mice: An experimental study. Iran J Pharm Res 2017; 16(2): 661-9.
[PMID: 28979320]
[110]
Mordorski B, Rosen J, Friedman A. Nanotechnology as an innovative approach for accelerating wound healing in diabetes. Diabetes Management 2015; 5(5): 329-32.
[http://dx.doi.org/10.2217/dmt.15.28]
[111]
Rigo C, Ferroni L, Tocco I, et al. Active silver nanoparticles for wound healing. Int J Mol Sci 2013; 14(3): 4817-40.
[http://dx.doi.org/10.3390/ijms14034817] [PMID: 23455461]
[112]
Singh R, Singh D. Chitin membranes containing silver nanoparticles for wound dressing application. Int Wound J 2014; 11(3): 264-8.
[http://dx.doi.org/10.1111/j.1742-481X.2012.01084.x] [PMID: 22958740]
[113]
Hebeish A, El-Rafie MH, El-Sheikh MA, Seleem AA, El-Naggar ME. Antimicrobial wound dressing and anti-inflammatory efficacy of silver nanoparticles. Int J Biol Macromol 2014; 65: 509-15.
[http://dx.doi.org/10.1016/j.ijbiomac.2014.01.071] [PMID: 24530328]
[114]
Leu JG, Chen SA, Chen HM, et al. The effects of gold nanoparticles in wound healing with antioxidant epigallocatechin gallate and α-lipoic acid. Nanomedicine (Lond) 2012; 8(5): 767-75.
[http://dx.doi.org/10.1016/j.nano.2011.08.013] [PMID: 21906577]
[115]
Gobin AM, O’Neal DP, Watkins DM, Halas NJ, Drezek RA, West JL. Near infrared laser-tissue welding using nanoshells as an exogenous absorber. Lasers Surg Med 2005; 37(2): 123-9.
[http://dx.doi.org/10.1002/lsm.20206] [PMID: 16047329]
[116]
Khan MI, Behera SK, Paul P, et al. Biogenic Au@ ZnO core-shell nanocomposites kill staphylococcus aureus without provoking nuclear damage and cytotoxicity in mouse fibroblasts cells under hyperglycemic condition with enhanced wound healing proficiency. Med Microbiol Immunol (Berl) 2018; 5: 1-21.
[PMID: 30291475]
[117]
Mariselvam R, Ranjitsingh AJ, Padmalatha C, Selvakumar PM. Green synthesis of copper quantum dots using rubia cardifolia plant root extracts and its antibacterial properties. Mater Sci 2014; 3(4): 191.
[118]
Boateng JS, Matthews KH, Stevens HN, Eccleston GM. Wound healing dressings and drug delivery systems: a review. J Pharm Sci 2008; 97(8): 2892-923.
[http://dx.doi.org/10.1002/jps.21210] [PMID: 17963217]
[119]
Bowler PG, Duerden BI, Armstrong DG. Wound microbiology and associated approaches to wound management. Clin Microbiol Rev 2001; 14(2): 244-69.
[http://dx.doi.org/10.1128/CMR.14.2.244-269.2001] [PMID: 11292638]
[120]
Newman MD, Stotland M, Ellis JI. The safety of nanosized particles in titanium dioxide- and zinc oxide-based sunscreens. J Am Acad Dermatol 2009; 61(4): 685-92.
[http://dx.doi.org/10.1016/j.jaad.2009.02.051] [PMID: 19646780]
[121]
Sankar R, Dhivya R, Shivashangari KS, Ravikumar V. Wound healing activity of origanum vulgare engineered titanium dioxide nanoparticles in wistar albino rats. J Mater Sci Mater Med 2014; 25(7): 1701-8.
[http://dx.doi.org/10.1007/s10856-014-5193-5] [PMID: 24682905]
[122]
Javanmardi S, Ghojoghi A, Divband B, Ashrafi J. Titanium dioxide nanoparticle/gelatin: a potential burn wound healing biomaterial. Wounds 2018; 30(12): 372-9.
[PMID: 30507548]
[123]
Nanowerk online [home page on internet] the promise of nitric oxide-releasing nanoparticles as wound healing agent [Accessed on 2nd April 2019] available at https://www.nanowerk.com
[124]
Miller C, McMullin B, Ghaffari A, et al. Gaseous nitric oxide bactericidal activity retained during intermittent high-dose short duration exposure. Nitric Oxide 2009; 20(1): 16-23.
[http://dx.doi.org/10.1016/j.niox.2008.08.002] [PMID: 18789393]
[125]
Tocco I, Zavan B, Bassetto F, Vindigni V. Nanotechnology-based therapies for skin wound regeneration. J Nanomater 2012; 2012: 4.
[http://dx.doi.org/10.1155/2012/714134]
[126]
Barraud N, Schleheck D, Klebensberger J, et al. Nitric oxide signaling in pseudomonas aeruginosa biofilms mediates phosphodiesterase activity, decreased cyclic di-GMP levels, and enhanced dispersal. J Bacteriol 2009; 191(23): 7333-42.
[http://dx.doi.org/10.1128/JB.00975-09] [PMID: 19801410]
[127]
Han G, Martinez LR, Mihu MR, Friedman AJ, Friedman JM, Nosanchuk JD. Nitric oxide releasing nanoparticles are therapeutic for staphylococcus aureus abscesses in a murine model of infection. PLoS One 2009; 4(11)e7804
[http://dx.doi.org/10.1371/journal.pone.0007804] [PMID: 19915659]
[128]
Matson JB, Zha RH, Stupp SI. Peptide self-assembly for crafting functional biological materials. Curr Opin Solid State Mater Sci 2011; 15(6): 225-35.
[http://dx.doi.org/10.1016/j.cossms.2011.08.001] [PMID: 22125413]
[129]
Zhu J, Marchant RE. Design properties of hydrogel tissue-engineering scaffolds. Expert Rev Med Devices 2011; 8(5): 607-26.
[http://dx.doi.org/10.1586/erd.11.27] [PMID: 22026626]
[130]
Haines-Butterick L, Rajagopal K, Branco M, et al. Controlling hydrogelation kinetics by peptide design for three-dimensional encapsulation and injectable delivery of cells. Proc Natl Acad Sci USA 2007; 104(19): 7791-6.
[http://dx.doi.org/10.1073/pnas.0701980104] [PMID: 17470802]
[131]
Wang S, Nagrath D, Chen PC, Berthiaume F, Yarmush ML. Three-dimensional primary hepatocyte culture in synthetic self-assembling peptide hydrogel. Tissue Eng Part A 2008; 14(2): 227-36.
[http://dx.doi.org/10.1089/tea.2007.0143] [PMID: 18333775]
[132]
Mohamed A, Xing MM. Nanomaterials and nanotechnology for skin tissue engineering. Int J Burns Trauma 2012; 2(1): 29-41.
[PMID: 22928165]
[133]
Cherubino M, Rubin JP, Miljkovic N, Kelmendi-Doko A, Marra KG. Adipose-derived stem cells for wound healing applications. Ann Plast Surg 2011; 66(2): 210-5.
[http://dx.doi.org/10.1097/SAP.0b013e3181e6d06c] [PMID: 21200308]
[134]
Kucharzewski M, Rojczyk E, Wilemska-Kucharzewska K, Wilk R, Hudecki J, Los MJ. Novel trends in application of stem cells in skin wound healing. Eur J Pharmacol 2019; 843: 307-15.
[http://dx.doi.org/10.1016/j.ejphar.2018.12.012] [PMID: 30537490]
[135]
Pastar I, Stojadinovic O, Tomic-Canic M. Role of keratinocytes in healing of chronic wounds. Surg Technol Int 2008; 17: 105-12.
[PMID: 18802889]
[136]
Lee KB, Choi J, Cho SB, et al. Topical embryonic stem cells enhance wound healing in diabetic rats. J Orthop Res 2011; 29(10): 1554-62.
[http://dx.doi.org/10.1002/jor.21385] [PMID: 21469178]
[137]
Hu MS, Leavitt T, Malhotra S, et al. Stem cell-based therapeutics to improve wound healing. Plast Surg Int 2015; 2015383581
[http://dx.doi.org/10.1155/2015/383581] [PMID: 26649195]
[138]
Clayton ZE, Tan RP, Miravet MM, et al. Induced pluripotent stem cell-derived endothelial cells promote angiogenesis and accelerate wound closure in a murine excisional wound healing model. Biosci Rep 2018; 38(4)pii BSR20180563
[http://dx.doi.org/10.1042/BSR20180563] [PMID: 29976773]
[139]
Ding DC, Shyu WC, Lin SZ. Mesenchymal stem cells. Cell Transplant 2011; 20(1): 5-14.
[http://dx.doi.org/10.3727/096368910X] [PMID: 21396235]
[140]
Schlosser S, Dennler C, Schweizer R, et al. Paracrine effects of mesenchymal stem cells enhance vascular regeneration in ischemic murine skin. Microvasc Res 2012; 83(3): 267-75.
[http://dx.doi.org/10.1016/j.mvr.2012.02.011] [PMID: 22391452]
[141]
Zahorec P, Koller J, Danisovic L, Bohac M. Mesenchymal stem cells for chronic wounds therapy. Cell Tissue Bank 2015; 16(1): 19-26.
[http://dx.doi.org/10.1007/s10561-014-9440-2] [PMID: 24651970]
[142]
Bourin P, Bunnell BA, Casteilla L, et al. Stromal cells from the adipose tissue-derived stromal vascular fraction and culture expanded adipose tissue-derived stromal/stem cells: a joint statement of the international federation for adipose therapeutics and science (IFATS) and the international society for cellular therapy (ISCT). Cytotherapy 2013; 15(6): 641-8.
[http://dx.doi.org/10.1016/j.jcyt.2013.02.006] [PMID: 23570660]
[143]
Merceron C, Portron S, Masson M, et al. The effect of two- and three-dimensional cell culture on the chondrogenic potential of human adipose-derived mesenchymal stem cells after subcutaneous transplantation with an injectable hydrogel. Cell Transplant 2011; 20(10): 1575-88.
[http://dx.doi.org/10.3727/096368910X557191] [PMID: 21294960]
[144]
Frese L, Dijkman PE, Hoerstrup SP. Adipose tissue-derived stem cells in regenerative medicine. Transfus Med Hemother 2016; 43(4): 268-74.
[http://dx.doi.org/10.1159/000448180] [PMID: 27721702]
[145]
Li Y, Zhang J, Yue J, Gou X, Wu X. Epidermal stem cells in skin wound healing. Adv Wound Care (New Rochelle) 2017; 6(9): 297-307.
[http://dx.doi.org/10.1089/wound.2017.0728] [PMID: 28894637]
[146]
Zhang CP, Fu XB. Therapeutic potential of stem cells in skin repair and regeneration. Chin J Traumatol 2008; 11(4): 209-21.
[http://dx.doi.org/10.1016/S1008-1275(08)60045-0] [PMID: 18667118]
[147]
Zare S, Anjomshoa M, Kurd S, et al. Regenerative medicine: novel approach in burn wound healing. J Skin Stem Cell 2015; 2(2)e30351
[http://dx.doi.org/10.17795/jssc30351]
[148]
Chen Z, Wang Y, Shi C. Therapeutic implications of newly identified stem cell populations from the skin dermis. Cell Transplant 2015; 24(8): 1405-22.
[http://dx.doi.org/10.3727/096368914X682431] [PMID: 24972091]
[149]
Yan J, Tie G, Wang S, et al. Diabetes impairs wound healing by Dnmt1-dependent dysregulation of hematopoietic stem cells differentiation towards macrophages. Nat Commun 2018; 9(1): 33.
[http://dx.doi.org/10.1038/s41467-017-02425-z] [PMID: 29295997]
[150]
Chatterjee P, Chakraborty B, Nandy S. Aloe vera plant: review with significant pharmacological activities. Mintage J Pharm Med Sci 2013; 2(3): 21-4.
[151]
Saini S, Dhiman A, Nanda S. Traditional indian medicinal plants with potential wound healing activity: a review. Int J Pharm Sci Res 2016; 7(5): 1809.
[152]
Purohit SK, Solanki R, Soni MK, Mathur V. Experimental evaluation of aloe vera leaves pulp as topical medicament on wound healing. Int J Pharmacol Res 2012; 2(3): 110-2.
[http://dx.doi.org/10.7439/ijpr.v2i3.702]
[153]
Sabale P, Bhimani B, Prajapati C, Sabale V. An overview of medicinal plants as wound healers. J Appl Pharm Sci 2012; 2(11): 143-50.
[154]
Boudreau MD, Beland FA. An evaluation of the biological and toxicological properties of Aloe barbadensis (miller), aloe vera. J Environ Sci Health C Environ Carcinog Ecotoxicol Rev 2006; 24(1): 103-54.
[http://dx.doi.org/10.1080/10590500600614303] [PMID: 16690538]
[155]
Hashemi SA, Madani SA, Abediankenari S. The review on properties of Aloe vera in healing of cutaneous wounds. BioMed Res Int 2015; 2015714216
[http://dx.doi.org/10.1155/2015/714216]
[156]
Dan MM, Sarmah P, Vana DR, Dattatreya A. Wound healing: concepts and updates in herbal medicine. Inte J Med Res Health Sci 2018; 7(1): 170-81.
[157]
Majumdar A, Sangole P. Alternative approaches to wound healingwound healing-new insights into ancient challenges. London, UK: IntechOpen Ltd 2016.
[http://dx.doi.org/10.5772/63636]
[158]
Yadav KH, Kumar JR, Basha SI, Deshmukh GR, Gujjula R, Santhamma B. Wound healing activity of topical application of aloe vera gel in experimental animal models. Int J Pharma Bio Sci 2012; 3(2): 63-72.
[159]
Mediratta PK, Sharma KK, Singh S. Evaluation of immunomodulatory potential of ocimum sanctum seed oil and its possible mechanism of action. J Ethnopharmacol 2002; 80(1): 15-20.
[http://dx.doi.org/10.1016/S0378-8741(01)00373-7] [PMID: 11891082]
[160]
Prakash P, Gupta N. Therapeutic uses of ocimum sanctum linn (tulsi) with a note on eugenol and its pharmacological actions: a short review. Indian J Physiol Pharmacol 2005; 49(2): 125-31.
[PMID: 16170979]
[161]
Pattanayak P, Behera P, Das D, Panda SK. Ocimum sanctum Linn. A reservoir plant for therapeutic applications: an overview. Pharmacogn Rev 2010; 4(7): 95-105.
[http://dx.doi.org/10.4103/0973-7847.65323] [PMID: 22228948]
[162]
Goel A, Kumar S, Singh DK, Bhatia AK. Wound healing potential of ocimum sanctum linn. With induction of tumor necrosis factor-alpha. Indian J Exp Biol 2010; 48(4): 402-6.
[PMID: 20726339]
[163]
Maier CM, Chan PH. Role of superoxide dismutases in oxidative damage and neurodegenerative disorders. Neuroscientist 2002; 8(4): 323-34.
[http://dx.doi.org/10.1177/107385840200800408] [PMID: 12194501]
[164]
Udupa SL, Shetty S, Udupa AL, Somayaji SN. Effect of ocimum sanctum linn. On normal and dexamethasone suppressed wound healing. Indian J Exp Biol 2006; 44(1): 49-54.
[PMID: 16430091]
[165]
Raina R, Parwez S, Verma PK, Pankaj NK. Medicinal plants and their role in wound healing. Online Veterinary J 2008; 3(1): 21.
[166]
Chundran NK, Husen IR, Rubianti I. Effect of neem leaves extract (azadirachta indica) on wound healing. Althea Med J 2015; 2(2): 199-203.
[http://dx.doi.org/10.15850/amj.v2n2.535]
[167]
Maan P, Yadav KS, Yadav NP. Wound healing activity of Azadirachta indica A. Juss stem bark in mice. Pharmacogn Mag 2017; 13(Suppl. 2): S316-20.
[http://dx.doi.org/10.4103/0973-1296.210163] [PMID: 28808399]
[168]
Wagle BR, Chetri DK. Evaluation of wound healing properties of neem (azadirachta indica) in dogs. Int J Herb Med 2017; 5(4): 5-7.
[169]
Chainani-Wu N. Safety and anti-inflammatory activity of curcumin: a component of tumeric (curcuma longa). J Altern Complement Med 2003; 9(1): 161-8.
[http://dx.doi.org/10.1089/107555303321223035] [PMID: 12676044]
[170]
Akbik D, Ghadiri M, Chrzanowski W, Rohanizadeh R. Curcumin as a wound healing agent. Life Sci 2014; 116(1): 1-7.
[http://dx.doi.org/10.1016/j.lfs.2014.08.016] [PMID: 25200875]
[171]
Tejada S, Manayi A, Daglia M, et al. Wound healing effects of curcumin: a short review. Curr Pharm Biotechnol 2016; 17(11): 1002-7.
[http://dx.doi.org/10.2174/1389201017666160721123109] [PMID: 27640646]
[172]
Derakhshandeh H, Kashaf SS, Aghabaglou F, Ghanavati IO, Tamayol A. Smart bandages: the future of wound care. Trends Biotechnol 2018; 36(12): 1259-74.
[http://dx.doi.org/10.1016/j.tibtech.2018.07.007] [PMID: 30197225]
[173]
Panchal C, Patel R, Jadav K, Mashru R. smart bandage for specific wound healing. Eur J Pharm Med Res 2018; 5(8): 267-75.
[174]
Innovation and economic development [home page on internet] smart bandage for real-time wireless monitoring of chronic wounds [Accessed on 5th April 2019] available at: https://innovation.kaust.edu.sa
[175]
Puffett N, Martin L, Chow MK. Cohesive short-stretch vs four-layer bandages for venous leg ulcers Br J Community Nurs 2006; 11: (Sup3): S6-11.
[http://dx.doi.org/10.12968/bjcn.2006.11.Sup3.21211]
[176]
Kassal P, Kim J, Kumar R, et al. Smart bandage with wireless connectivity for uric acid biosensing as an indicator of wound status. Electrochem Commun 2015; 56: 6-10.
[http://dx.doi.org/10.1016/j.elecom.2015.03.018]
[177]
C|net online [home page on internet] paint-on bandage changes color as your wound heals [Accessed on 5th April 2019]. Available at: www.cnet.com
[178]
ProDigitalWeb online [home page on internet] smart bandage signals infection by turning fluorescent [Accessed on 5th April 2019] available at: http://www.prodigitalweb.com
[179]
MailOnline [home page on internet] the smart bandage that turns yellow to provide an early warning of infections [Accessed on 10th April 2019] available at: https://www.dailymail.co.uk
[180]
Guinovart T, Valdés-Ramírez G, Windmiller JR, Andrade FJ, Wang J. Bandage based wearable potentiometric sensor for monitoring wound pH. Electroanalysis 2014; 26(6): 1345-53.
[http://dx.doi.org/10.1002/elan.201300558]
[181]
Mostafalu P, Tamayol A, Rahimi R, et al. Smart bandage for monitoring and treatment of chronic wounds. Small 2018; 14(33)e1703509
[http://dx.doi.org/10.1002/smll.201703509] [PMID: 29978547]

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