Journal Home > Volume 15 , Issue 4

Poor wound treatment impacts millions of humans worldwide, increasing deaths and costs. Wounds have three key complications: (a) a lack of an adequate environment for cell migration, proliferation, and angiogenesis; (b) microbial infection; and (c) unstable and prolonged inflammation.Regrettably, contemporary therapeutic treatments have not entirely tackled these basic difficulties and thus have insufficient medical accomplishment. The incorporation of the extraordinary capabilities of nanomaterials in wound healing has achieved major successes over the years. Nanomaterials can promote a variety of cellular and molecular processes that assist in the wound microenvironment through antibacterial, anti-inflammatory, and angiogenic activities, potentially shifting the surroundings from nonhealing to healing. The current review focuses on novel techniques, with a particular focus on recent revolutionary wound healing and infection control tactics based on nanomaterials, such as nanoparticles, nanocomposites, and scaffolds, which are discussed in depth. Furthermore, the effectiveness of nanoparticles as carriers for therapeutic compounds in wound-healing applications has been investigated which provide researchers an up-to-date sources on the use of nanomaterials and their creative ways that can improve wound-healing uses.


menu
Abstract
Full text
Outline
About this article

Nanoparticles-assisted Wound Healing: A Review

Show Author's information Sarah Habeeb Hamed1Ebaa Adnan Azooz2( )Emad Abbas Jaffar Al-Mulla3( )
The Gifted Students` School in Najaf, the General Directorate of Education Al-Najaf, Ministry of Education, Iraq
Medical Laboratory Technology Department, College of Medical Technology, the Islamic University, Najaf, Iraq
College of Health and Medical Techniques, Al-Furat Al-Awsat Technical University, An-Najaf, Iraq

Abstract

Poor wound treatment impacts millions of humans worldwide, increasing deaths and costs. Wounds have three key complications: (a) a lack of an adequate environment for cell migration, proliferation, and angiogenesis; (b) microbial infection; and (c) unstable and prolonged inflammation.Regrettably, contemporary therapeutic treatments have not entirely tackled these basic difficulties and thus have insufficient medical accomplishment. The incorporation of the extraordinary capabilities of nanomaterials in wound healing has achieved major successes over the years. Nanomaterials can promote a variety of cellular and molecular processes that assist in the wound microenvironment through antibacterial, anti-inflammatory, and angiogenic activities, potentially shifting the surroundings from nonhealing to healing. The current review focuses on novel techniques, with a particular focus on recent revolutionary wound healing and infection control tactics based on nanomaterials, such as nanoparticles, nanocomposites, and scaffolds, which are discussed in depth. Furthermore, the effectiveness of nanoparticles as carriers for therapeutic compounds in wound-healing applications has been investigated which provide researchers an up-to-date sources on the use of nanomaterials and their creative ways that can improve wound-healing uses.

Keywords: gold nanoparticles (Au NPs), zinc oxide nanoparticles (ZnO NPs), wounds caring, sliver nanoparticle (Ag NPs)

References(76)

[1]

M.H. Kim. Nanoparticle-based therapies for wound biofilm infection: Opportunities and challenges. IEEE Transactions on NanoBioscience, 2016, 15(3): 294−304. https://doi.org/10.1109/TNB.2016.2527600

[2]

Y. Peng, Y. Liu, X.L. Lu, et al. Ag-Hybridized plasmonic Au-triangular nanoplates: Highly sensitive photoacoustic/Raman evaluation and improved antibacterial/photothermal combination therapy. Journal of Materials Chemistry B, 2018, 6(18): 2813−2820. https://doi.org/10.1039/c8tb00617b

[3]

T. Brunella, T. Giovanni, B. Barbara, et al. Use of polylactide-Co-glycolide-nanoparticles for lysosomal delivery of a therapeutic enzyme in glycogenosis type II fibroblasts. Journal of Nanoscience and Nanotechnology, 2015, 15(4): 2657−2666. https://doi.org/10.1166/jnn.2015.9251

[4]

A. Kushwaha, L. Goswami, B.S. Kim. Nanomaterial-based therapy for wound healing. Nanomaterials, 2022, 12(4): 618. https://doi.org/10.3390/nano12040618

[5]

D.M. dos Santos, D.S. Correa, E.S. Medeiros, et al. Advances in functional polymer nanofibers: From spinning fabrication techniques to recent biomedical applications. ACS Applied Materials &Interfaces, 2020, 12(41): 45673−45701. https://doi.org/10.1021/acsami.0c12410

[6]

X.L. Lu, R.H. Chen, J. Lv, et al. High-resolution bimodal imaging and potent antibiotic/photodynamic synergistic therapy for osteomyelitis with a bacterial inflammation-specific versatile agent. Acta Biomaterialia, 2019, 99: 363−372. https://doi.org/10.1016/j.actbio.2019.08.043

[7]

E.A. Azooz, E.A. Azooz, R.K. Kadhum. Rapid palladium preconcentration and spectrophotometric determination in water and soil samples. Analytical and Bioanalytical Chemical Research, 2022, 9(3): 251−258.

[8]

N. Siebert, W.G. Xu, E. Grambow, et al. Erythropoietin improves skin wound healing and activates the TGF-β signaling pathway. Laboratory Investigation, 2011, 91(12): 1753−1765. https://doi.org/10.1038/labinvest.2011.125

[9]

A.K.M. Al-Toriahi, E.A. Azooz, E.A.J. Al-Mulla. Metal nanoparticles and nano-filters for the disposal of hospital waste: A review. Nano Biomedicine and Engineering, 2023, 15(2): 179−190. https://doi.org/10.26599/NBE.2023.9290017

[10]

C.A. Cobbold, J.A. Sherratt. Mathematical modelling of nitric oxide activity in wound healing can explain keloid and hypertrophic scarring. Journal of Theoretical Biology, 2000, 204(2): 257−288. https://doi.org/10.1006/jtbi.2000.2012

[11]

M.K. Swamy, U.R. Sinniah. Patchouli (Pogostemon cablin Benth.): Botany, agrotechnology and biotechnological aspects. Industrial Crops and Products, 2016, 87: 161−176. https://doi.org/10.1016/j.indcrop.2016.04.032

[12]
S.K. Mohanty, M.K. Swamy, U.R. Sinniah, et al. Leptadenia reticulata (Retz.) Wight & Arn. (Jivanti): Botanical, agronomical, phytochemical, pharmacological, and biotechnological aspects. Molecules, 2017, 22(6): 1019.
DOI
[13]

A.R. Hussein, M.S. Gburi, N.M. Muslim, et al. A greenness evaluation and environmental aspects of solidified floating organic drop microextraction for metals: A review. Trends in Environmental Analytical Chemistry, 2023, 37: e00194. https://doi.org/10.1016/j.teac.2022.e00194

[14]

V. Vijayakumar, S.K. Samal, S. Mohanty, et al. Recent advancements in biopolymer and metal nanoparticle-based materials in diabetic wound healing management. International Journal of Biological Macromolecules, 2019, 122: 137−148. https://doi.org/10.1016/j.ijbiomac.2018.10.120

[15]

S. Hamdan, I. Pastar, S. Drakulich, et al. Nanotechnology-driven therapeutic interventions in wound healing: Potential uses and applications. ACS Central Science, 2017, 3(3): 163−175. https://doi.org/10.1021/acscentsci.6b00371

[16]
M.A.M. Hassan, A.H. Mohammed, , Hameed, E. M. Application of Aloe vera gel blended polymer-collagen scaffolds for bone tissue engineering. Nano Biomedicine and Engineering, 2023, 15(2): 118–125.
DOI
[17]

T.J. Shaw, P. Martin. Wound repair at a glance. Journal of Cell Science, 2009, 122(18): 3209−3213. https://doi.org/10.1242/jcs.031187

[18]

E.A. Azooz, H.S.A. Al-Wani, M.S. Gburi, et al. Recent modified air-assisted liquid–liquid microextraction applications for medicines and organic compounds in various samples: A review. Open Chemistry, 2022, 20(1): 525−540. https://doi.org/10.1515/chem-2022-0174

[19]

K.P. Hoversten, L.J. Kiemele, A.M. Stolp, et al. Prevention, diagnosis, and management of chronic wounds in older adults. Mayo Clinic Proceedings, 2020, 95(9): 2021−2034. https://doi.org/10.1016/j.mayocp.2019.10.014

[20]

M.L. Wang, X.W. Huang, H.X. Zheng, et al. Nanomaterials applied in wound healing: Mechanisms, limitations and perspectives. Journal of Controlled Release, 2021, 337: 236−247. https://doi.org/10.1016/j.jconrel.2021.07.017

[21]

G. Han, R. Ceilley. Chronic wound healing: A review of current management and treatments. Advances in Therapy, 2017, 34: 599−610. https://doi.org/10.1007/s12325-017-0478-y

[22]

M.M. Mihai, M.B. Dima, B. Dima, et al. Nanomaterials for wound healing and infection control. Materials, 2019, 12(13): 2176. https://doi.org/10.3390/ma12132176

[23]

C.Y. Mao, Y.M. Xiang, X.M. Liu, et al. Repeatable photodynamic therapy with triggered signaling pathways of fibroblast cell proliferation and differentiation to promote bacteria-accompanied wound healing. ACS Nano, 2018, 12(2): 1747−1759. https://doi.org/10.1021/acsnano.7b08500

[24]

R. Ahmadi, E.A. Azooz, Y. Yamini, et al. Liquid-liquid microextraction techniques based on in situ formation/decomposition of deep eutectic solvents. TrAC Trends in Analytical Chemistry, 2023, 161: 117019. https://doi.org/10.1016/j.trac.2023.117019

[25]

S.S.D. Kumar, N.K. Rajendran, N.N. Houreld, et al. Recent advances on silver nanoparticle and biopolymer-based biomaterials for wound healing applications. International Journal of Biological Macromolecules, 2018, 115: 165−175. https://doi.org/10.1016/j.ijbiomac.2018.04.003

[26]

T.G. MPharm, T. Nigusse, M.D.D. MPharm. Silver nanoparticles as real topical bullets for wound healing. Journal of the American College of Clinical Wound Specialists, 2011, 3(4): 82−96. https://doi.org/10.1016/j.jcws.2012.05.001

[27]

W. Sim, R. Barnard, M.A.T. Blaskovich, et al. Antimicrobial silver in medicinal and consumer applications: A patent review of the past decade (2007–2017). Antibiotics, 2018, 7(4): 93. https://doi.org/10.3390/antibiotics7040093

[28]

J.S. Möhler, W. Sim, M.A.T. Blaskovich, et al. Silver bullets: A new lustre on an old antimicrobial agent. Biotechnology Advances, 2018, 36(5): 1391−1411. https://doi.org/10.1016/j.biotechadv.2018.05.004

[29]

R. Szmyd, A.G. Goralczyk, L. Skalniak, et al. Effect of silver nanoparticles on human primary keratinocytes. Biological Chemistry, 2013, 394(1): 113−123. https://doi.org/10.1515/hsz-2012-0202

[30]

M. Ahmadi, M. Adibhesami. The effect of silver nanoparticles on wounds contaminated with Pseudomonas aeruginosa in mice: An experimental study. Iranian Journal of Pharmaceutical Research, 2017, 16(2): 661−669.

[31]

S. Pal, R. Nisi, M. Stoppa, et al. Silver-functionalized bacterial cellulose as antibacterial membrane for wound-healing applications. ACS Omega, 2017, 2(7): 3632−3639. https://doi.org/10.1021/acsomega.7b00442

[32]
J. Wu, Y.D. Zheng, W.H. Song, et al. In situ synthesis of silver-nanoparticles/bacterial cellulose composites for slow-released antimicrobial wound dressing. Carbohydrate Polymers, 2014, 102: 762–771.
DOI
[33]

E.A. Azooz, G.J. Shabaa, E.A.J. Al-Mulla. Methodology for preconcentration and determination of silver in aqueous samples using cloud point extraction. Brazilian Journal of Analytical Chemistry, 2021, 9(35): 39−48. https://doi.org/10.30744/brjac.2179-3425.ar-61-2021

[34]

R. Singh, D. Singh. Chitin membranes containing silver nanoparticles for wound dressing application. International Wound Journal, 2014, 11(3): 264−268. https://doi.org/10.1111/j.1742-481x.2012.01084.x

[35]
W. Hu, S. Chen, X. Li, et al.. In situ synthesis of silver chloride nanoparticles into bacterial cellulose membranes. Materials Science and Engineering: C, 2009, 29(4): 1216–1219.
DOI
[36]

K. Niska, E. Zielinska, M.K. Radomski, et al. Metal nanoparticles in dermatology and cosmetology: Interactions with human skin cells. Chemico-Biological Interactions, 2018, 295: 38−51. https://doi.org/10.1016/j.cbi.2017.06.018

[37]

M.G. Arafa, R.F. El-Kased, M.M. Elmazar. Thermoresponsive gels containing gold nanoparticles as smart antibacterial and wound healing agents. Scientific Reports, 2018, 8: 13674. https://doi.org/10.1038/s41598-018-31895-4

[38]

M.A. Sherwani, S. Tufail, A.A. Khan, et al. Gold nanoparticle-photosensitizer conjugate based photodynamic inactivation of biofilm producing cells: Potential for treatment of C. albicans infection in BALB/c mice. PLoS One, 2015, 10(7): e0131684. https://doi.org/10.1371/journal.pone.0131684

[39]

O. Akturk, K. Kismet, A.C. Yasti, et al. Collagen/gold nanoparticle nanocomposites: A potential skin wound healing biomaterial. Journal of Biomaterials Applications, 2016, 31(2): 283−301. https://doi.org/10.1177/0885328216644536

[40]

L. Shahzadi, A.A. Chaudhry, A.R. Aleem, et al. Development of K-doped ZnO nanoparticles encapsulated crosslinked chitosan based new membranes to stimulate angiogenesis in tissue engineered skin grafts. International Journal of Biological Macromolecules, 2018, 120: 721−728. https://doi.org/10.1016/j.ijbiomac.2018.08.103

[41]
P.C. Balaure, A.M. Holban, A.M. Grumezescu, et al. In vitro and in vivo studies of novel fabricated bioactive dressings based on collagen and zinc oxide 3D scaffolds. International Journal of Pharmaceutics, 2019, 557: 199–207.
DOI
[42]

F.A. Wannas, E.A. Azooz, R.K. Ridha, et al. Separation and micro determination of zinc(II) and cadmium(II) in food samples using cloud point extraction method. Iraqi Journal of Science, 2023, 64(3): 1046−1061. https://doi.org/10.24996/ijs.2023.64.3.2

[43]

R. Rakhshaei, H. Namazi. A potential bioactive wound dressing based on carboxymethyl cellulose/ZnO impregnated MCM-41 nanocomposite hydrogel. Materials Science and Engineering:C, 2017, 73: 456−464. https://doi.org/10.1016/j.msec.2016.12.097

[44]

E.A.J. Al-Mulla, K. Al-Janabi. Extraction of cobalt(II) from aqueous solution by N, N’-carbonyl difatty amides. Chinese Chemical Letters, 2011, 22(4): 469−472. https://doi.org/10.1016/j.cclet.2010.10.037

[45]

J.U. Choi, S.W. Lee, R. Pangeni, et al. Preparation and in vivo evaluation of cationic elastic liposomes comprising highly skin-permeable growth factors combined with hyaluronic acid for enhanced diabetic wound-healing therapy. Acta Biomaterialia, 2017, 57: 197−215. https://doi.org/10.1016/j.actbio.2017.04.034

[46]

E.A.J. Al-Mulla. Preparation of polylactic acid/epoxidized palm oil/fatty nitrogen compounds modified clay nanocomposites by melt blending. Polymer Science Series A, 2011, 53(2): 149−157. https://doi.org/10.1134/s0965545x11020015

[47]

A. Naskar, H. Khan, R. Sarkar et al. Anti-biofilm activity and food packaging application of room temperature solution process based polyethylene glycol capped Ag-ZnO-graphene nanocomposite. Materials Science and Engineering:C, 2018, 91: 743−753. https://doi.org/10.1016/j.msec.2018.06.009

[48]

Y.Z. Zhou, R. Chen, T.T. He, et al. Biomedical potential of ultrafine Ag/AgCl nanoparticles coated on graphene with special reference to antimicrobial performances and burn wound healing. ACS Applied Materials &Interfaces, 2016, 8(24): 15067−15075. https://doi.org/10.1021/acsami.6b03021

[49]

E.A.J. Al-Mulla, W.M.Z.W. Yunus, N.A.B. Ibrahim, et al. Epoxidized palm oil plasticized polylactic acid/fatty nitrogen compound modified clay nanocomposites: Preparation and characterization. Polymers and Polymer Composites, 2010, 18(8): 451−460. https://doi.org/10.1177/096739111001800806

[50]
R.H. Dong, Y.X. Jia, C.C. Qin, et al. In situ deposition of a personalized nanofibrous dressing via a handy electrospinning device for skin wound care. Nanoscale, 2016, 8(6): 3482–3488.
DOI
[51]

G.J. Shabaa, F.A. Semysim, R.K. Ridha, et al. Air-assisted dual-cloud point extraction coupled with flame atomic absorption spectroscopy for the separation and quantification of zinc in pregnant women’s serum. Journal of the Iranian Chemical Society, 2023, 20(9): 2277−2284. https://doi.org/10.1007/s13738-023-02834-6

[52]

G. Han, L.R. Martinez, M.R. Mihu, et al. Nitric oxide releasing nanoparticles are therapeutic for staphylococcus aureus abscesses in a murine model of infection. PLoS One, 2009, 4(11): e7804. https://doi.org/10.1371/journal.pone.0007804

[53]

L.R. Martinez, G. Han, M. Chacko, et al. Antimicrobial and healing efficacy of sustained release nitric oxide nanoparticles against Staphylococcus aureus skin infection. Journal of Investigative Dermatology, 2009, 129(10): 2463−2469. https://doi.org/10.1038/jid.2009.95

[54]

M.R. Mihu, U. Sandkovsky, G. Han, et al. The use of nitric oxide releasing nanoparticles as a treatment againstAcinetobacter baumanniiin wound infections. Virulence, 2010, 1(2): 62−67. https://doi.org/10.4161/viru.1.2.10038

[55]
E.A. Azooz, J.R. Moslim, S.M. Hameed, et al. Aspirin in Food Samples for Separation and Micro Determination of Copper(II) using Cloud Point Extraction/Solvation Method. Nano Biomedcine and Engineering, 2021,13(1): 62–71.
DOI
[56]

B.D. Ulery, L.S. Nair, C.T. Laurencin. Biomedical applications of biodegradable polymers. Journal of Polymer Science Part B:Polymer Physics, 2011, 49(12): 832−864. https://doi.org/10.1002/polb.22259

[57]

H.K. Makadia, S.J. Siegel. Poly lactic-co-glycolic acid (PLGA) as biodegradable controlled drug delivery carrier. Polymers, 2011, 3(3): 1377−1397. https://doi.org/10.3390/polym3031377

[58]

V.P. Torchilin. Drug targeting. European Journal of Pharmaceutical Sciences, 2000, 11: S81−S91. https://doi.org/10.1016/S0928-0987(00)00166-4

[59]
E.A. Azooz, R.K. Ridha, H.A. Abdulridha. The Fundamentals and Recent Applications of Micellar System Extraction for Nanoparticles and Bioactive Molecules: A Review. Nano Biomedicine and Engineering, 2021, 13(3): 264–278.
DOI
[60]

F. Norouzinezhad, F. Ghaffari, A. Norouzinejad, et al. Cutaneous leishmaniasis in Iran: Results from an epidemiological study in urban and rural provinces. Asian Pacific Journal of Tropical Biomedicine, 2016, 6(7): 614−619. https://doi.org/10.1016/j.apjtb.2016.05.005

[61]

A. Stejskalová, B.D. Almquist. Using biomaterials to rewire the process of wound repair. Biomaterials Science, 2017, 5(8): 1421−1434. https://doi.org/10.1039/c7bm00295e

[62]

M.I.N. Ahamed, S. Sankar, P.M. Kashif, et al. Evaluation of biomaterial containing regenerated cellulose and chitosan incorporated with silver nanoparticles. International Journal of Biological Macromolecules, 2015, 72: 680−686. https://doi.org/10.1016/j.ijbiomac.2014.08.055

[63]

N. Levi-Polyachenko, R. Jacob, C. Day, et al. Chitosan wound dressing with hexagonal silver nanoparticles for hyperthermia and enhanced delivery of small molecules. Colloids and Surfaces B:Biointerfaces, 2016, 142: 315−324. https://doi.org/10.1016/j.colsurfb.2016.02.038

[64]

R. Jayakumar, M. Prabaharan, P.T. Sudheesh Kumar, et al. Biomaterials based on chitin and chitosan in wound dressing applications. Biotechnology Advances, 2011, 29(3): 322−337. https://doi.org/10.1016/j.biotechadv.2011.01.005

[65]

S.H. Hsu, Y.B. Chang, C.L. Tsai, et al. Characterization and biocompatibility of chitosan nanocomposites. Colloids and Surfaces B:Biointerfaces, 2011, 85(2): 198−206. https://doi.org/10.1016/j.colsurfb.2011.02.029

[66]

R.A.B. Sanad, H.M. Abdel-Bar. Chitosan-hyaluronic acid composite sponge scaffold enriched with Andrographolide-loaded lipid nanoparticles for enhanced wound healing. Carbohydrate Polymers, 2017, 173: 441−450. https://doi.org/10.1016/j.carbpol.2017.05.098

[67]

P.S. Rabbani, A. Zhou, Z.M. Borab, et al. Novel lipoproteoplex delivers Keap1 siRNA based gene therapy to accelerate diabetic wound healing. Biomaterials, 2017, 132: 1−15. https://doi.org/10.1016/j.biomaterials.2017.04.001

[68]

F.H.J. Al-Shemmari, E.A.J. Al-Mulla, A.A. Rabah. A comparative study of different surfactants for natural rubber clay nanocomposite preparation. Rendiconti Lincei, 2014, 25(3): 409−413. https://doi.org/10.1007/s12210-014-0307-z

[69]

W.H. Hoidy, M.B. Ahmad, E.A.J. Al-Mulla. Chemical synthesis and characterization of palm oil-based difatty Acyl thiourea. Journal of Oleo Science, 2010, 9(5): 229−233. https://doi.org/10.5650/jos.59.229

[70]

K.H. Gathwan, I.H.T. Al-Karkhi, E.A.J. Al-Mulla. Hepatic toxicity of nickel chloride in mice. Research on Chemical Intermediates, 2013, 39: 2537−2542. https://doi.org/10.1007/s11164-012-0780-x

[71]

N. Volkova, M. Yukhta, O. Pavlovich, et al. Application of cryopreserved fibroblast culture with Au nanoparticles to treat burns. Nanoscale Research Letters, 2016, 11: 22. https://doi.org/10.1186/s11671-016-1242-y

[72]
M. Mirzahosseinipour, K. Khorsandi, R. Hosseinzadeh, et al. Antimicrobial photodynamic and wound healing activity of curcumin encapsulated in silica nanoparticles. Photodiagnosis and Photodynamic Therapy, 2020, 29: 101639.
DOI
[73]

I.A. Mohammed, E.A.J. Al-Mulla, N.K.A. Kadar, et al. Structure-property studies of thermoplastic and thermosetting polyurethanes using palm and soya oils-based polyols. Journal of Oleo Science, 2013, 62(12): 1059−1072. https://doi.org/10.5650/jos.62.1059

[74]

M.M. Radhi, E.A. Jaffar Al-Mulla, W.T. Tan. Electrochemical characterization of the redox couple of Fe(III)/Fe(II) mediated by grafted polymer electrode. Research on Chemical Intermediates, 2014, 40(1): 179−192. https://doi.org/10.1007/s11164-012-0954-6

[75]

N.M. Muslim, B.K. Hussain, N.M. Abdulhussein, et al. Determination of selenium in black tea leaves using the air-assisted cloud point extraction method: evaluation the method's environmental performance. Analytical Bioanalytical Chemistry Research, 2024, 11(1): 10−21.

[76]

C.Y. Tong, W. Zou, W.M. Ning, et al. Synthesis of DNA-guided silver nanoparticles on a graphene oxide surface: Enhancing the antibacterial effect and the wound healing activity. RSC Advances, 2018, 8(49): 28238−28248. https://doi.org/10.1039/c8ra04933e

Publication history
Copyright
Rights and permissions

Publication history

Received: 23 June 2023
Revised: 15 August 2023
Accepted: 13 September 2023
Published: 07 November 2023
Issue date: December 2023

Copyright

© The Author(s) 2023.

Rights and permissions

This is an open-access article distributed under  the  terms  of  the  Creative  Commons  Attribution  4.0 International  License (CC BY) (http://creativecommons.org/licenses/by/4.0/), which  permits  unrestricted  use,  distribution,  and reproduction in any medium, provided the original author and source are credited.

Return