Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 + 2801 word(s) 2801 2021-11-26 04:12:35 |
2 format correct Meta information modification 2801 2022-01-18 09:25:42 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Mihai, M. Nanomaterials for Wound Healing and Infection Control. Encyclopedia. Available online: https://encyclopedia.pub/entry/18391 (accessed on 05 July 2024).
Mihai M. Nanomaterials for Wound Healing and Infection Control. Encyclopedia. Available at: https://encyclopedia.pub/entry/18391. Accessed July 05, 2024.
Mihai, Mara. "Nanomaterials for Wound Healing and Infection Control" Encyclopedia, https://encyclopedia.pub/entry/18391 (accessed July 05, 2024).
Mihai, M. (2022, January 18). Nanomaterials for Wound Healing and Infection Control. In Encyclopedia. https://encyclopedia.pub/entry/18391
Mihai, Mara. "Nanomaterials for Wound Healing and Infection Control." Encyclopedia. Web. 18 January, 2022.
Nanomaterials for Wound Healing and Infection Control
Edit

Wound healing has been intensely studied in order to develop an "ideal" technique that achieves expeditious recovery and reduces scarring to the minimum, thus ensuring function preservation. The classic approach to wound management is represented by topical treatments, such as antibacterial or colloidal agents, in order to prevent infection and promote a proper wound-healing process. Nanotechnology studies submicroscopic particles (maximum diameter of 100 nm), as well as correlated phenomena. Metal nanoparticles (e.g., silver, gold, zinc) are increasingly being used in dermatology, due to their beneficial effect on accelerating wound healing, as well as treating and preventing bacterial infections. Other benefits include: ease of use, less frequent dressing changes and a constantly moist wound environment. This entry highlights recent findings regarding nanoparticle application in wound management.

wound healing infections control nano-dressings nanoparticles chronic infection

1. Definition and Classification

Nanotechnology studies the synthesis, structure and dynamic of atomic and molecular nanometric particles (maximum diameter of 100 nm), termed nanoparticles, upon which nanoproducts are built [1]. By downscaling to nanometric range, a particle’s surface increases exponentially, whereas its volume decreases, which leads to particular physico-chemical characteristics that account for numerous medical applications. Dynamic light scattering (DLS), fluorescence correlation spectroscopy (FCS) and Raman scattering (RS) offer information regarding the hydrodynamic size distribution of nanoparticles, whereas circular dichroism (CD), Infrared spectroscopy (IR) and Mass spectroscopy (MS) help in investigating their structure, conformation and surface characteristics. In addition, electron microscopy in general and transmission electron microscopy (TEM) and scanning electron microscopy (SEM) in particular, can achieve resolutions of less than 1 nm, thus making them valuable investigational tools employed in studying the properties and dynamics (i.e., diffusion, adherence) of nanomaterials. Through TEM, the morphology of nanomaterials can be investigated, whereas SEM recreates a superficial image of the nanomaterial, thus being solely employed in the study of dimensions and overall shape [2].
The size and shape of nanoparticles are key properties that determine their biological efficiency, by influencing active substance delivery (carrier circulation), penetrability (either directly through cell membranes or through phagocytosis) and cellular responses (receptor recognition) [2][3][4].
Due to their antibacterial properties and low toxicity profile, metal nanoparticles such as silver, gold and zinc represent ideal candidates for integration in wound dressings [5]. The main types of nanomaterials used for wound treatment are represented by nanoparticles, nanocomposites, coatings and scaffolds (Figure 1).
Figure 1. Main types of nanomaterials which can be used for wound treatment.
Nanomaterials for tissue regeneration can be developed under different structures: nanoparticles, nanospheres, nanocapsules, nanoemulsions, nanocarriers and nanocolloids [1].
Two principal types of nanoparticles (NPs) are commonly employed in wound therapy: (1) NPs that possess intrinsic characteristics that aid wound closure; (2) NPs used as delivery vectors for therapeutic agents. The former can be divided into metallic/metal oxide nanomaterials and nonmetallic nanomaterials (Figure 1) [6].
Regarding metallic and metal oxide nanoparticles, the most studied are silver, gold and zinc compounds due to their unique properties: antibacterial activity and reduced penetration into the skin. Their effectiveness, as well as toxicity, relies on key features, for example: dimension and architecture (smaller particles are more biologically active), surface functionalization, zeta potential and polydispersity index [7]. Zeta potential estimates the particle surface charge which influences receptor binding capacity as well as the penetrability of cellular barriers [3]. Other particle surface characteristics, such as porosity, chemical structure and heterogeneity, hydrolytic stability, determine the biological behavior of nanomaterials, including their interactions with other biomolecules, which may influence their biodistribution [4][8].
Nanospheres are small polymeric matrix systems composed of a fixed porous polymer onto which active substances (e.g., aminoacids, minerals or organic substances) can bind. As a result, active compounds become more stable and exhibit increased biocompatibility and improved pharmaceutical properties [1].
Nanocapsules are capable of enclosing an active agent within their structure, releasing it at a specific time, ensuring a more controlled delivery. They can also improve the penetration of active ingredients into deeper layers of the dermis. On the other hand, the effectiveness of the substances may be reduced [1].
Nanoemulsions represent homogenous, thermally stable oil-in-water emulsions with a droplet size of maximum 100 nm that also contain surfactant and can easily incorporate biologically active substances, preventing their sedimentation. They are also characterized by high solubilization capacity and fluidity and decreased viscosity. Their disadvantage would be the uncontrolled accumulation of active substances in reticular dermis or subcutaneous fat [1].
Nanocarriers structurally resemble liposomes, but contain synthetic, non-ionic surfactants. In addition, they express surface receptors capable of binding to specific loci, thus increasing the efficacy of the active agent, while also minimizing any adverse reactions [1].
Nanocolloids are composed of 1.5 to 5 nm non-ionic metal nanoparticles highly scattered in demineralized water due to their perpetual Brownian motion which allows them to penetrate eukaryotic and prokaryotic cells, thus exhibiting antimicrobial activity. Moreover, they are characterized by good electrical conductivity, being positively charged particles [1].

2. Nanoparticles

2.1. Silver Nanoparticles

Silver nanoparticles (AgNPs) might overcome the limitations of standard silver compounds. Due to their increased surface-to-volume ratio, AgNPs are more potent at reduced concentrations, thus lowering their toxicity. Pure silver nanoparticles can modulate anti-inflammatory cytokine release, in order to promote rapid wound closure without increasing scarring [6]. By inducing the differentiation of myofibroblasts from normal fibroblasts, AgNPs promote wound contractility, thus accelerating the healing process [8]. Moreover, AgNPs stimulate epidermal re-epithelialization through the proliferation and relocation of keratinocytes [8]. However, Szmyd et al. reported that higher concentrations of AgNPs decrease keratinocyte viability, metabolism, as well as migration and differentiation of these cells, through the activation of caspase 3 and 7 (proteases involved in programmed cell death) and dose-dependent DNA damage [9]. In order to reduce side effects, silver nanoparticles can be used in low doses along with antimicrobial drugs to achieve increased efficiency. This nanomaterial proved a high bacteria-killing performance against Gram negative pathogens and promoted wound healing [10]. Similar results have been reported by Holban et al. who coated polyester-nylon dressings with AgNPs which interfered with bacterial colonization as well as biofilm synthesis, while maintaining a low toxicity profile [11].
The main anti-bacterial mechanism of AgNPs is creating sulfuric bonds with either bacterial cell membrane proteins or thiol groups of various enzymes, especially those involved in the respiratory chain, thus leading to apoptosis [8]. Moreover, since DNA contains sulfurous and phosphorous bonds, AgNPs can interfere with DNA synthesis during cell division, thus inhibiting bacterial multiplication [8].
Moreover, silver nanoparticles were used for numerous clinical trials in the therapy of wounds, especially burns and chronic wounds (i.e., diabetic ulcers) [12]. Currently, there are some commercially available dressings containing AgNPs. Such example is Acticoat©-wound dressing containing AgNPs with average size of 15 nm. The proved properties of Anticoat© are: wound healing, infection reduction on the site of the wound, and pain reduction, aspects observed in most tested patients. However, this coating is still under investigation for the therapy of burns. Recent clinical trials concluded that Anticoat© may be efficient in avoiding infections in burns when applied together with silver sulphadiazine and chlorhexidine digluconate cream [13].

2.2. Gold Nanoparticles

Gold nanoparticles (AuNPs) represent a solid option when it comes to wound therapy due to their chemical stability and capacity of absorbing near infrared light, while, at the same time, being relatively easy to synthetize [1]. Moreover, by tuning the surface plasmon resonance, AuNP gels gain thermoresponsiveness, as described by Arafa et al., who demonstrated their antibacterial and healing properties both in vitro and in vivo, backed by histopathologic examinations [14]. AuNPs can either directly target the bacterial cell wall, or they can bind to bacterial DNA, blocking the double-helix from uncoiling during replication or transcription, thus exerting bactericidal and bacteriostatic properties. 

2.3. Zinc Oxide Nanoparticles

Zinc oxide nanoparticles (ZnONPs) represent a reliable antibacterial agent, by inducing bacterial cell membrane perforations. Moreover, when incorporated in hydrogel-based wound dressings [6], the overall contact time is increased, promoting keratinocyte migration, thus improving re-epithelialization [8]. In addition, in a recent study, a microporous chitosan hydrogel/ZnONPs dressing presented a high capacity of absorbing wound exudates and enabled the formation of hemostatic blood clots, while also displaying antibacterial properties with little cytotoxicity [15].
However, the intrinsic high toxicity of these material limit their usage in wound healing therapies [6]. For example, high concentrations of ZnONPs are associated with mitochondrial dysfunction in keratinocytes, causing the release of lactate dehydrogenase. Moreover, ZnONPs produce reactive oxygen species, while also inhibiting the expression of superoxide dismutase and glutathione peroxidase genes in human keratinocytes, inducing cell membrane oxidative stress and apoptosis. In addition, it has been described that ZnONPs generate carcinogenic transformations [16].
In a skin wound mouse model, ZnONPs achieve successful wound closure and aesthetic wound healing, suggesting their capability as an effective antimicrobial tissue adhesive [17].

3. Nanocomposites or Composite Nanoparticles

Artificial synthesis of NPs is limited due to the fact that it involves high costs, energy consumption and additional resources to dispose of toxic by-products. Therefore, non-polluting, cheap and renewable sources such as plant extracts have gained popularity as they contain alkaloids, phenols, amino acids and proteins which are employed in the reduction and stabilization of Ag ions in AgNPs. 
Gelatin is a natural, biodegradable, highly biocompatible, non-immunogenic polymer, rich in hydroxyproline that can reduce AgNO3 in situ through the hydroxyl groups. Moreover, its non-polar aminoacids help stabilize AgNPs. As a result, Ye et al. developed gelatin-chitosan-Ag, a new composite with a high density of pores with diameters of 100–250 µm, in which AgNPs were initially mixed with chitosan, cross-linked with tannic acid and cryodesiccated. The compound exhibited wound healing and anti-bacterial properties, while maintaining low cytotoxicity [18]. In addition, Shao et al. used Barleria gibsoni aqueous leaves to synthetize a ZnONPs gel efficient in treating both Gram-positive and Gram-negative infected burns [19].
Polymeric nanomaterial therapy employs polymeric nanoparticles (e.g., chitosan) as wound dressings or as delivery vectors due to the fact that they exhibit antibacterial and re-epithelialization properties [6]. Biopolymers are biocompatible polymeric networks that can absorb significant quantities of liquid, while also ensuring a moist wound environment [20].
Chitosan is a hydrophilic biopolymer commonly extracted from the exoskeleton of crustacean arthropods, characterized by high bioavailability and low toxicity, in addition to being one of the few polymers with antibacterial properties [20]. Chemically, this biopolymer is a linear polysaccharide containing D-glucosamine and N-acetyl glucosamine units. Apart from its affinity to metals, proteins and dyes, chitosan can also form complexes with anions, such as sulfate and phosphate, due to its cationic nature [21]. Moreover, its degradation products can stimulate synthesis of extracellular matrix components [20].
Various forms of chitosan have been investigated for wound-healing therapies, such as hydrogels, membranes, films, sponges and scaffolds [21]. Chitosan nanoparticles are characterized by superior permeability, antibacterial properties and immune modulation, while also being non-toxic to human cells. Polydopamine nanoparticles were loaded with ciprofloxacin which was released on demand through near-infrared light stimulation, even though the hydrogel complex exhibited minimal leakage under physiological conditions [22].
Hernandez Martinez et al. described a nanocomposite of AuNPs, chitosan and calreticulin for the therapy of diabetic lesions. Calreticulin is a 46 kDa calcium-binding protein found in the endoplasmic reticulum which regulates cytosolic and endoplasmic reticulum calcium levels. In addition, calreticulin acts as a molecular chaperone that controls the proper folding of proteins. The gold nanoparticles-chitosan-calreticulin composite promoted the growth, migration and differentiation of keratinocytes, fibroblasts and endothelial cells, without affecting cell viability, in vitro as well as in vivo. Moreover, the histological examination described granulation, re-epithelization and increased collagen synthesis [23].
Bacterial cellulose is a biopolymer synthetized by various microorganisms, such as AcetobacterPseudomonasSalmonella, through carbohydrate fermentations. Bacterial cellulose represents an excellent skin substitute due to its physico-chemical traits, such as very high surface area per unit, increased biocompatibility, hydrophilicity and nontoxicity. Bacterial nanocellulose is characterized by three-dimensional porous networks capable of retaining a high quantity of water, thus ensuring a damp environment, proper for wound healing [24]. Khalid et al. analyzed the healing properties of bacterial cellulose combined with ZnONPs and discovered that the added ZnONPs granted antibacterial properties [25]. Moniri et al. combined bacterial nanocellulose obtained from the fermentation of Gram-negative Gluconacetobacter xylinus, with silver nanoparticles (BNC-Ag). The nanocomposite improved wound healing and reduced Staphylococcus aureus colonization in vitro [26].

Nano-Carriers for Wound Healing

Nanomaterials can also act as carriers for therapeutic agents, controlling their release. Nitric oxide exhibits a crucial role in inflammatory pathways, cellular growth, angiogenesis and extracellular matrix deposition and remodeling. In addition, nitric oxide exhibits wide spectrum antibacterial properties, while also interfering with biofilm synthesis. As a result, many studies have tried to produce a delivery system with high loading capacity, controlled release and reduced cytotoxicity [6]. Nitric oxide-releasing poly(lactic-co-glycolic acid)-polyethylenimine nanoparticles were designed to provide prolonged nitric oxide release, antibacterial efficacy against methicillin-resistant Staphylococcus aureus and Pseudomonas aeruginosa, as well as accelerated wound closure in vivo [27].
Curcumin, the dynamic component of the nutritional spice turmeric, also presented interest due to its antibiotic and antioxidant properties, as well as stimulating the production of granulation tissue. A recent study attempted to increase curcumin’s poor solubility by encapsulating it into a saline-hydrogel nanoparticle vector, and the results demonstrated in vitro efficacy against methicillin-resistant Staphylococcus aureus and enhanced wound closure activity [28]. Ghaffari et al. stated that both gel and ointment preparations containing curcumin-Ampicillin solid lipid nanoparticles exhibited antibacterial effects, while also increasing wound closure rate compared to control and placebo groups, both in vitro and in vivo, without additional toxicity [29]. In addition, in a mouse model experiment, Moradi et al. studied the associated effect of pulse photo-bio-modulation with curcumin loaded super-paramagnetic iron oxide nanoparticles and reported accelerated wound healing and an important reduction in Staphylococcus aureus colonies [30].

4. Coatings and Scaffolds

Another application of nanomaterials is the production of scaffolds that mimic the properties of extracellular matrix. Several techniques are used to produce them, such as electro-spinning, self-assembly and phase separation. The most employed is electro-spinning which creates porous polymeric nanofibers that can be used as hybrid scaffolds for the adherence and development of fibroblasts in wounds [6]. Other nano-polymers, such as dendrimers, can exhibit anti-inflammatory properties, while also forming networks that can incorporate antibacterial agents (e.g., silver) [31].
Using a porcine model of superficial partial thickness wounds, Haik et al. studied the healing properties of a polymer nanofiber dressing applied by a mobile handheld electrospinning device. Regarding safety (risk of infection, delayed healing) the results were similar to the traditional paraffin tulle gras dressing. The non-touch technique and the ease of application represent advantages of this therapeutic option [32].
Electrospun chitosan-poly-vinyl alcohol nanofibrous blend scaffolds were applied to treat models of diabetic wounds in rats, showing increased healing rates compared with controls [33].
The in vivo study in Wistar rats of a electrospun nanofiber membrane enhanced with silver nanoparticles showed multiple favorable effects: low cytotoxicity, long-term antibacterial activity, reduced inflammation and enhanced wound healing [34].
An electrospun composite scaffold with curcumin showed low cytotoxicity and anti-oxidant effect in vitro, as well as enhanced wound healing in vivo, in a full-thickness dermal model for Wistar rats [35].
Other nanoscale system compounds attempted to deliver growth factors, such as recombinant human epidermal growth factor, in order to stimulate the healing of full-thickness diabetic wounds. However, their application was limited by the highly proteolytic environment, as well as the down-regulation of the associated growth factor receptors and signaling molecules in chronic lesions [36]. In contrast, other authors described better results when using basic fibroblast growth factor (bFGF). For example, Zhang et al. described a hydrogel that employed Ca2+ as a cross-linker between carboxyl groups (–COO) and that was capable of releasing preloaded bFGF. They observed that both calcium and bFGF stimulated fibroblast growth and differentiation, and that the effect was more potent in the early phases of re-epithelialization, inducing wound contraction, both in vitro and in vivo. In addition the hydrogel presented two profiles of bFGF release: an initial burst which accounted for approximately 40% in the first 12 h and a secondary, prolonged phase, which ensured a constant release of bFGF over 8 days. These dynamics suggest that the compound achieves increased levels and stability of local bFGF over a longer period of time [37].
Furthermore, electro-spun nano-fibrous meshes have been used as networks for gene encapsulation in wound-dressing materials. Gene-activated matrix therapy, which associates gene therapy and tissue engineering, has the advantage of increasing or decreasing the expression of a target gene involved in regeneration. DNA is also more stable compared to growth factors. However, more studies need to be performed in order to obtain additional data about gene therapy effectiveness, bioavailability and safety in the presence of wound bacterial colonization [6].
The multiple properties of nanomaterials regarding antimicrobial effects, the stimulation of wound healing by inducing angiogenesis, re-epithelialization, granulation and/or collagen synthesis, as well as their role in the prevention of abnormal scarring represent advantages compared to standard treatment and arguments towards their use in clinical practice (Table 1).
Table 1. Properties of nanomaterials developed for wound management. bFGF—basic fibroblast growth factor.
Material Properties References
Antibacterial Stimulation of Wound Healing 1 Prevention of Abnormal Scarring
Nanoparticles Silver nanoparticles + + + [6][8][10][11]
Gold nanoparticles + + Unknown [14][8][38]
Zinc oxide nanoparticles + + Unknown [6][15][8][39]
Composite nanoparticles Eg. chitosan nanoparticles, nanocellulose + + + [6][40][18][20][41][42][43][22][23][44][45][25][26]
Nanocarriers Eg. Nitric oxide-releasing poly(lactic-co-glycolic acid)-polyethylenimine nanoparticles, curcumin loaded super-paramagnetic iron oxide + + Unknown [27][46][28][29][30]
Coatings and Scaffolds Eg. hydrogel preloaded with bFGF, Hydrogels reinforced with AgNPs ++ ++ Unknown [47][48][49]
1 (angiogenesis, re-epithelialization, granulation and/or collagen synthesis).

References

  1. Niska, K.; Zielinska, E.; Radomski, M.W.; Inkielewicz-Stepniak, I. Metal nanoparticles in dermatology and cosmetology: Interactions with human skin cells. Chem. Biol. Interact. 2018, 295, 38–51.
  2. Lin, P.C.; Lin, S.; Wang, P.C.; Sridhar, R. Techniques for physicochemical characterization of nanomaterials. Biotechnol. Adv. 2014, 32, 711–726.
  3. Ferrari, M. Nanogeometry: Beyond drug delivery. Nat. Nanotechnol. 2008, 3, 131–132.
  4. George, S.; Lin, S.; Ji, Z.; Thomas, C.R.; Li, L.; Mecklenburg, M.; Meng, H.; Wang, X.; Zhang, H.; Xia, T.; et al. Surface defects on plate-shaped silver nanoparticles contribute to its hazard potential in a fish gill cell line and zebrafish embryos. ACS Nano 2012, 6, 3745–3759.
  5. Negut, I.; Grumezescu, V.; Grumezescu, A.M. Treatment Strategies for Infected Wounds. Molecules 2018, 23.
  6. Hamdan, S.; Pastar, I.; Drakulich, S.; Dikici, E.; Tomic-Canic, M.; Deo, S.; Daunert, S. Nanotechnology-Driven Therapeutic Interventions in Wound Healing: Potential Uses and Applications. ACS Cent. Sci. 2017, 3, 163–175.
  7. Jamil, B.; Abbasi, R.; Abbasi, S.; Khan, S.U.; Ihsan, A.; Javed, S.; Bokhari, H.; Imran, M. Encapsulation of Cardamom Essential Oil in Chitosan Nano-composites: In-vitro Efficacy on Antibiotic-Resistant Bacterial Pathogens and Cytotoxicity Studies. Front. Microbiol. 2016, 7, 1580.
  8. Vijayakumar, V.; Samal, S.K.; Mohanty, S.; Nayak, S.K. Recent advancements in biopolymer and metal nanoparticle-based materials in diabetic wound healing management. Int. J. Biol. Macromol. 2019, 122, 137–148.
  9. Szmyd, R.; Goralczyk, A.G.; Skalniak, L.; Cierniak, A.; Lipert, B.; Filon, F.L.; Crosera, M.; Borowczyk, J.; Laczna, E.; Drukala, J.; et al. Effect of silver nanoparticles on human primary keratinocytes. Biol. Chem. 2013, 394, 113–123.
  10. Pal, S.; Nisi, R.; Stoppa, M.; Licciulli, A. Silver-Functionalized Bacterial Cellulose as Antibacterial Membrane for Wound-Healing Applications. ACS Omega 2017, 2, 3632–3639.
  11. Radulescu, M.; Andronescu, E.; Dolete, G.; Popescu, R.C.; Fufă, O.; Chifiriuc, M.C.; Mogoantă, L.; Bălşeanu, T.-A.; Mogoșanu, G.D.; Grumezescu, A.M.; et al. Silver Nanocoatings for Reducing the Exogenous Microbial Colonization of Wound Dressings. Materials 2016, 9, 345.
  12. Boroumand, Z.; Golmakani, N.; Boroumand, S. Clinical trials on silver nanoparticles for wound healing. Nanomed. J. 2018, 5, 186–191.
  13. Fong, J.; Wood, F.; Fowler, B. A silver coated dressing reduces the incidence of early burn wound cellulitis and associated costs of inpatient treatment: Comparative patient care audits. Burns 2005, 31, 562–567.
  14. Arafa, M.G.; El-Kased, R.F. Thermoresponsive gels containing gold nanoparticles as smart antibacterial and wound healing agents. Sci. Rep. 2018, 8, 13674.
  15. Kumar, P.T.; Lakshmanan, V.-K.; Anilkumar, T.; Ramya, C.; Reshmi, P.; Unnikrishnan, A.; Nair, S.V.; Jayakumar, R. Flexible and microporous chitosan hydrogel/nano ZnO composite bandages for wound dressing: In vitro and in vivo evaluation. ACS Appl. Mater. Interfaces 2012, 4, 2618–2629.
  16. Yang, H.; Liu, C.; Yang, D.; Zhang, H.; Xi, Z. Comparative study of cytotoxicity, oxidative stress and genotoxicity induced by four typical nanomaterials: The role of particle size, shape and composition. J. Appl. Toxicol. 2009, 29, 69–78.
  17. Gao, Y.; Han, Y.; Cui, M.; Tey, H.L.; Wang, L.; Xu, C. ZnO nanoparticles as an antimicrobial tissue adhesive for skin wound closure. J. Mater. Chem. B 2017, 5, 4535–4541.
  18. Ye, H.; Cheng, J.; Yu, K. In situ reduction of silver nanoparticles by gelatin to obtain porous silver nanoparticle/chitosan composites with enhanced antimicrobial and wound-healing activity. Int. J. Biol. Macromol. 2019, 121, 633–642.
  19. Shao, F.; Yang, A.; Yu, D.M.; Wang, J.; Gong, X.; Tian, H.X. 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, 267–273.
  20. Shariatinia, Z. Pharmaceutical applications of chitosan. Adv. Colloid Interface Sci. 2019, 263, 131–194.
  21. Biranje, S.S.; Madiwale, P.V.; Patankar, K.C.; Chhabra, R.; Dandekar-Jain, P.; Adivarekar, R.V. Hemostasis and anti-necrotic activity of wound-healing dressing containing chitosan nanoparticles. Int. J. Biol. Macromol. 2019, 121, 936–946.
  22. Gao, G.; Jiang, Y.-W.; Jia, H.-R.; Wu, F.-G. Near-infrared light-controllable on-demand antibiotics release using thermo-sensitive hydrogel-based drug reservoir for combating bacterial infection. Biomaterials 2019, 188, 83–95.
  23. Hernandez Martinez, S.P.; Rivera González, T.; Franco Molina, M.; Bollain y Goytia, J.; Martínez Sanmiguel, J.; Zárate Triviño, D.; Rodríguez Padilla, C. A Novel Gold Calreticulin Nanocomposite Based on Chitosan for Wound Healing in a Diabetic Mice Model. Nanomaterials 2019, 9, 75.
  24. Fu, L.; Zhang, J.; Yang, G. Present status and applications of bacterial cellulose-based materials for skin tissue repair. Carbohydr. Polym. 2013, 92, 1432–1442.
  25. Khalid, A.; Khan, R.; Ul-Islam, M.; Khan, T.; Wahid, F. Bacterial cellulose-zinc oxide nanocomposites as a novel dressing system for burn wounds. Carbohydr. Polym. 2017, 164, 214–221.
  26. Moniri, M.; Moghaddam, A.B.; Azizi, S.; Rahim, R.A.; Zuhainis, S.W.; Navaderi, M.; Mohamad, R. In vitro molecular study of wound healing using biosynthesized bacteria nanocellulose/silver nanocomposite assisted by bioinformatics databases. Int. J. Nanomed. 2018, 13, 5097–5112.
  27. Nurhasni, H.; Cao, J.; Choi, M.; Kim, I.; Lee, B.L.; Jung, Y.; Yoo, J.-W. Nitric oxide-releasing poly(lactic-co-glycolic acid)-polyethylenimine nanoparticles for prolonged nitric oxide release, antibacterial efficacy, and in vivo wound healing activity. Int. J. Nanomed. 2015, 10, 3065–3080.
  28. Krausz, A.E.; Adler, B.L.; Cabral, V.; Navati, M.; Doerner, J.; Charafeddine, R.A.; Chandra, D.; Liang, H.; Gunther, L.; Clendaniel, A.; et al. Curcumin-encapsulated nanoparticles as innovative antimicrobial and wound healing agent. Nanomed. Nanotechnol. Biol. Med. 2015, 11, 195–206.
  29. Ghaffari, S.; Alihosseini, F.; Sorkhabadi, S.M.R.; Bidgoli, S.A.; Mousavi, S.E.; Haghighat, S.; Nasab, A.A.; Kianvash, N. Nanotechnology in Wound Healing; Semisolid Dosage Forms Containing Curcumin-Ampicillin Solid Lipid Nanoparticles, in-Vitro, Ex-Vivo and in-Vivo Characteristics. Adv. Pharm. Bull. 2018, 8, 395–400.
  30. Moradi, A.; Kheirollahkhani, Y.; Fatahi, P.; Abdollahifar, M.-A.; Amini, A.; Naserzadeh, P.; Ashtari, K.; Ghoreishi, S.K.; Chien, S.; Rezaei, F.; et al. An improvement in acute wound healing in mice by the combined application of photobiomodulation and curcumin-loaded iron particles. Lasers Med. Sci. 2018, 34, 779–791.
  31. Abdel-Sayed, P.; Kaeppeli, A.; Siriwardena, T.; Darbre, T.; Perron, K.; Jafari, P.; Reymond, J.-L.; Pioletti, D.P.; Applegate, L.A.; Kaeppli, A. Anti-Microbial Dendrimers against Multidrug-Resistant P. aeruginosa Enhance the Angiogenic Effect of Biological Burn-wound Bandages. Sci. Rep. 2016, 6, 22020.
  32. Haik, J.; Kornhaber, R.; Blal, B.; Harats, M. The Feasibility of a Handheld Electrospinning Device for the Application of Nanofibrous Wound Dressings. Adv. Wound Care 2017, 6, 166–174.
  33. Gholipour-Kanani, A.; Bahrami, S.H.; Rabbani, S. Effect of novel blend nanofibrous scaffolds on diabetic wounds healing. IET Nanobiotechnol. 2016, 10, 1–7.
  34. Dong, R.H.; Jia, Y.-X.; Qin, C.-C.; Zhan, L.; Yan, X.; Cui, L.; Zhou, Y.; Jiang, X.; Long, Y.-Z. In situ deposition of a personalized nanofibrous dressing via a handy electrospinning device for skin wound care. Nanoscale 2016, 8, 3482–3488.
  35. Fu, S.Z.; Meng, X.H.; Fan, J.; Yang, L.L.; Wen, Q.L.; Ye, S.J.; Lin, S.; Wang, B.Q.; Chen, L.L.; Wu, J.B.; et al. Acceleration of dermal wound healing by using electrospun curcumin-loaded poly(epsilon-caprolactone)-poly(ethylene glycol)-poly(epsilon-caprolactone) fibrous mats. J. Biomed. Mater. Res. B Appl. Biomater. 2014, 102, 533–542.
  36. Chu, Y.; Yu, D.; Wang, P.; Xu, J.; Li, D.; Ding, M. Nanotechnology promotes the full-thickness diabetic wound healing effect of recombinant human epidermal growth factor in diabetic rats. Wound Repair Regen. 2010, 18, 499–505.
  37. Zhang, X.; Kang, X.; Jin, L.; Bai, J.; Liu, W.; Wang, Z.; Ji, L. Stimulation of wound healing using bioinspired hydrogels with basic fibroblast growth factor (bFGF). Int. J. Nanomed. 2018, 13, 3897–3906.
  38. Marza, S.; Magyari, K. Skin wound regeneration with bioactive glass-gold nanoparticles ointment. Biomed. Mater. 2019, 14, 025011.
  39. Khan, M.I.; Behera, S.K.; Paul, P.; Das, B.; Suar, M.; Jayabalan, R.; Fawcett, D.; Poinern, G.E.J.; Tripathy, S.K.; Mishra, A. Biogenic 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. 2018.
  40. Sood, R.; Chopra, D.S. Optimization of reaction conditions to fabricate Ocimum sanctum synthesized silver nanoparticles and its application to nano-gel systems for burn wounds. Mater. Sci. Eng. C Mater. Biol. Appl. 2018, 92, 575–589.
  41. Chen, Y.; Dan, N.; Dan, W.; Liu, X.; Cong, L. A novel antibacterial acellular porcine dermal matrix cross-linked with oxidized chitosan oligosaccharide and modified by in situ synthesis of silver nanoparticles for wound healing applications. Mater. Sci. Eng. C Mater. Biol. Appl. 2019, 94, 1020–1036.
  42. Hajji, S.; Ben Khedir, S.; Hamza-Mnif, I.; Hamdi, M.; Jedidi, I.; Kallel, R.; Boufi, S.; Nasri, M. Biomedical potential of chitosan-silver nanoparticles with special reference to antioxidant, antibacterial, hemolytic and in vivo cutaneous wound healing effects. Biochim. Biophys. Acta Gen. Subj. 2019, 1863, 241–254.
  43. Holban, A.M.; Grumezescu, V.; Grumezescu, A.M.; Vasile, B.S.; Trusca, R.; Cristescu, R.; Socol, G.; Iordache, F. Antimicrobial nanospheres thin coatings prepared by advanced pulsed laser technique. Beilstein J. Nanotechnol. 2014, 5, 872–880.
  44. Sun, T.; Zhan, B.; Zhang, W.; Qin, D.; Xia, G.; Zhang, H.; Peng, M.; Li, S.-A.; Zhang, Y.; Gao, Y.; et al. Carboxymethyl chitosan nanoparticles loaded with bioactive peptide OH-CATH30 benefit nonscar wound healing. Int. J. Nanomed. 2018, 13, 5771–5786.
  45. Tayeb, A.H.; Amini, E.; Ghasemi, S.; Tajvidi, M. Cellulose Nanomaterials-Binding Properties and Applications: A Review. Molecules 2018, 23, 2684.
  46. Wang, T.; Zheng, Y.; Shi, Y.; Zhao, L. pH-responsive calcium alginate hydrogel laden with protamine nanoparticles and hyaluronan oligosaccharide promotes diabetic wound healing by enhancing angiogenesis and antibacterial activity. Drug Deliv. Transl. Res. 2019, 9, 227–239.
  47. Wang, S.; Yan, C.; Zhang, X.; Shi, D.; Chi, L.; Luo, G.; Deng, J. Antimicrobial peptide modification enhances the gene delivery and bactericidal efficiency of gold nanoparticles for accelerating diabetic wound healing. Biomater. Sci. 2018, 6, 2757–2772.
  48. Moura, J.; Borsheim, E.; Carvalho, E. The Role of MicroRNAs in Diabetic Complications-Special Emphasis on Wound Healing. Genes 2014, 5, 926–956.
  49. Tartarini, D.; Mele, E. Adult Stem Cell Therapies for Wound Healing: Biomaterials and Computational Models. Front. Bioeng. Biotechnol. 2015, 3, 206.
More
Information
Contributor MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register :
View Times: 619
Entry Collection: Biopharmaceuticals Technology
Revisions: 2 times (View History)
Update Date: 18 Jan 2022
1000/1000
Video Production Service