Recent updates in nanotechnological advances for wound healing: A narrative review
-
Hitesh Chopra
, Saurov Mahanta
, Satya Kumar Avula
Abstract
The repair and remodeling of injured tissues, known as wound healing, is a multifaceted process. The use of nanotechnology to speed up the healing process of wounds by promoting the right kind of mobility through different mechanisms has shown a lot of promise. Several nanotechnologies with novel properties have emerged in recent years, each of which targets a different issue associated with wound-healing processes. Through their antibacterial, anti-inflammatory, and angiogenic actions, nanomaterials may alter the wound microenvironment from a non-healing to a healing state. Wound dressings including hydrogels, gelatin sponges, films, and bandages may all benefit from the use of nanoparticles (NPs) to keep harmful microbes out. The addition of bioactive substances like antibiotics, NPs, and growth factors to certain dressings may further boost their efficacy. In conclusion, this review sheds light on wound healing that may be aided by the special features of materials based on nanotechnology. Although nanomaterials for wound healing show great promise, further study is needed before this promising area can convert its findings into consumer-friendly solutions.
1 Introduction
Skin is a highly flexible and diverse organ that protects us from external environments (for example, chemicals, physical entities, and UV light). This strident outer environment frequently causes skin damage, but due to the presence of intricate healing systems, our skin allows it to heal swiftly and efficiently [1,2,3]. Regardless of the individual’s high inherent reparative ability, several cellular components poorly respond at the damage site, thereby delaying wound healing. However, the pathogenic systemic modifications associated with aging or unrestrained diabetes typically produce such attenuation. In fact, the primary risk factors for the development of chronic wounds (wounds that take more than 12 weeks to heal) are age and severe diabetes. Pessimistically, these chronic lesions (i.e., mostly venous ulcers, pressure sores, and diabetic foot ulcers [DFUs]) constitute an immediate need for noble and modern treatment methods worldwide [4]. Clinically, wound healing has always been a significant challenge, thus novel approaches and tools are desperately required. As nanotechnology has advanced rapidly in this century, nanomaterials developed via the integration of many domains have found use in important areas including healthcare, chemistry, and even the military [5,6,7,8,9,10]. Nanotechnology is the broad term for creating and making things whose function depends on their structure at the nanoscale, which is usually defined as 100 nm or less [11]. With their high adsorption capacity, drug loading, and antibacterial capabilities, nanoparticles (NPs) have found widespread usage in the wound-healing industry in recent years [12,13,14,15,16]. Temporary skin replacements, or wound dressings, serve a crucial role in hemostasis, infection management, and facilitating wound closure. This article examines the present state of knowledge on skin restoration and describes the abnormal cellular activities behind chronic wound-healing disorders. Emerging research tools will be crucial for decoding the cellular and molecular mechanisms behind the intensity of the diseases and the healing method. The studies were searched using search engines such as Scopus, Google scholar, PubMed, and Google patents. The manuscripts were searched using terms such as wound, nanoformulations, nanofiber, hydrogels, NPs, artificial intelligence (AI), and wound healing. However, the results shown in languages other than English were excluded. This review provides a detailed understanding of how nanoformulations for wound healing are acting at the molecular level and what kinds of recent advances have taken place. This shall enlighten the readers to think beyond the traditional wound-healing formulations.
2 Wound-healing stages
Wound healing in the skin seems to be primarily motivated by the spontaneous repair of the skin’s barrier function for countering further injury or contamination. To foster this process, a huge number of cells and mediators are required to be involved from the outset [17,18]. Research on the transition phase of wound healing starting from an inflammatory to a proliferative stage is ongoing and supposed to involve many intrinsic hidden factors [19], such as molecular patterns, associated with damage as well as pathogens detected by toll-like receptors and further involve to induce and maintain the inflammation in skin cells [20,21]. To reach the site of injury, the leukocytes, especially neutrophil granulocytes, move along an increasing gradient of chemokine [22,23] and the inflammatory response is further boosted by neutrophils, which secrete many pro-inflammatory cytokines [24]. Many other studies have also revealed the role of cytokines and chemokines in wound healing [25]. In addition, wound healing is believed to be enhanced by the epidermal growth factor (EGF) receptor pathway, which has been implicated in this process [26]. In contrast, macrophages are key players in the transition from inflammation to proliferation [27]. For example, research has revealed that in the absence of macrophages during the inflammatory and proliferative phases of wound healing, a decrease in tissue formation or bleeding occurs [28]. Therefore, the process has failed to proceed into the next phase. On the other hand, pathogen-specific molecular patterns and damage-associated molecular patterns activate skin-resident macrophages as well as monocytes that are separate from invading ones [27,28,29]. During the recovery phase, there is an increase in macrophages with a reparative function in the M2 subgroup, which produces anti-inflammatory mediators and extracellular matrix (ECM), which leads to fibroblast proliferation, angiogenesis, and phagocytosis of neutrophils (i.e., efferocytosis), bacteria, and cell debris to minimize extra injury to the wound site [30]. The present M1–M2 transition paradigm is supported by this phenomenon [31]. Non-healing or chronic wounds, such as venous ulcers and diabetic wounds, emerge if the M1–M2 transition is not efficiently regulated [32,33,34]. The earlier description proved the importance of macrophages in the efficient healing process of skin wounds. Null mice could heal skin wounds in the same length of time as wild mice, and Martin et al. [35] revealed that other cells may take over several tasks of macrophages. The absence of scarring in PU.1-null mice casts doubt on the role of the inflammatory response in skin wound healing [35].
The cutaneous wounds are healed by resurfacing the epithelium and contracting the wound site. The efficient wound-healing process is influenced by one of two mechanisms depending on the species. Even while rats recover largely by contracting, in humans this process accounts for up to 80% of the healing process [36]. Granulation tissue is formed before the formation of epithelial covering, which begins tissue regeneration, which refers to the secondary intention of healing for this kind of wound healing.
Besides the above conditions, there are also some reports on the most severe cases of third-intention healing including septic circumstances, in which patients deliberately, but temporarily leave their infected wounds unattended until the sickness subsides, at which point the wounds are closed. Stapling or reconstructive plastic surgery may be used to seal wounds that have been stabilized and put in excellent condition [37]. Using this easy-to-understand wound-healing taxonomy, the physician may predict whether a patient would have complete skin regeneration or just partial tissue restoration due to scarring. For wounds that are small, superficial, and clean, the blood clot production is limited to closing the wound site and eliminating the smallest cell debris. In fact, more time is required in the first phases of wound healing, which include hemostasis and cell debris clearance, necrotic tissue removal, and the formation of granulation tissue in deep, large wounds that are infected. It takes a few hours for keratinocytes to change from cobblestone-shaped to flat-migratory, and this process starts during the first few hours after injury [38]. In the early days of wound resurfacing, leap-frogging cells were described as keratinocytes moving over one another and onto the wound bed without displaying any unique migration characteristics [39]. There are many studies that reveal that the cells or even whole cell rows drag others along as they crawl over the incision [40,41,42]. Additionally, a large part of this process may be attributed to the development of the extension membrane or epidermal tongue and lamellipodial crawling and shuffling [43,44]. Furthermore, the keratinocytes around the wound site produce the epidermal tongue and also rearrange the cytoskeleton, while a leapfrog-like extension of the incision across the tongue follows the process [45]. The activated keratinocytes in the wound matrix’s front row drive them forward, away from fibrin, fibronectin, and vitronectin, which are produced from blood clots (lamellipodial crawling). However, instead of moving toward the wound center, front-row cells modify their shape, free their cell–cell connections, restructure themselves, and leave the front edge [45]; but when contact inhibition reaches the center of the wound, keratinocyte migration ends and the wound covering gets completed [45]. Even in thin layers, adipose tissue impairs the ability of the dermis, fascia, and muscle to cover wounds. For epithelial mobility, all tissues other than the dermis are needed in the development of granulation tissue. Moreover, along with diverse entities of cells related to the wound, the skin microflora such as Gram-positive and Gram-negative bacteria are present in the same tissue which have also a certain role in the wound healing processes. In the skin, the matrix is composed of type I collagen, glycoproteins (including hyaluronic acid [HA]), fibronectin, and hyaluronan and when a skin injury causes cellular damage, the invading adult stem cells are generally responsible for healing it, whereas the hair follicle bulge and interfollicular epidermis niche stem cells replace lost cells in epidermal regeneration [46,47,48]. In non-healing ulcers, in which the cell pool is limited due to prolonged inflammation produced by infection, hypoxia, ischemia, and/or profuse exudates display disruption of the epidermal stem cell niche [49]. Therefore, stem cell research is promoting stem cell usage as a remedy for wounds that are usually unable to heal. When it comes to healing a wound, the stem cells play an important role throughout the process, which enables the inflammation to be resolved as well as cell migration, proliferation, and differentiation [50,51,52]. However, their complex strategy of work, though, is still a mystery. The various stages of wound healing are illustrated in Figure 1.

Various phases of wound healing such as bleeding and hemostasis, inflammation, proliferation, and remodeling.
Epidermal barrier repair is very successful in healthy individuals, where biochemical mechanisms are activated when the skin is wounded to repair the damage. But when certain circumstances come opposite to the normal cell regulations, the incidence of delayed wound healing increases. The proliferation of pro-inflammatory cytokines and proteases (such as elastase), low levels of growth factors, and aged fibroblasts are common findings in chronic wound-healing studies [53]. Wound fibrosis may be predicted by looking at the inflammatory response. Consequently, the lack of inflammation in the uterus is touted as evidence of the extraordinary ability of the baby to heal scars without scarring [24]. For example, persistent wound inflammation is marked by an increase in reactive oxygen species (ROS) levels due to a reduction in macrophage phagocytic capacity in the neutrophil population [54]. In the battle against pathogenic microbes, ROS are critical; however, too much amount may cause tissue damage by boosting oxidative stress and, therefore, the level of matrix metalloproteinase (MMP). A prolonged open wound with an infection risk is not known to induce chronic inflammation but neither is known to cause chronic inflammation (Figure 2). This is shown by the impaired cell migration seen in chronic wounds, which have elevated levels of MMPs. Defective granulation tissue is caused by the breakdown of the ECM and the inability of fibroblasts to respond to growth cues. The infusion of innate immune cells into chronic wounds may negatively affect the healing process; however, other immune cell lineages, such as Langerhans cells, have been related to better healing outcomes [55]. Additionally, keratinocyte migration and re-epithelialization might be hampered by biofilms (mono- or polymicrobial) producing their own proteases [46]. Microbes often attach themselves to surfaces in the beginning. Despite this, their first attachment may be reversed, and they still are susceptible to drugs. As they develop, they form a biofilm that is resistant to antibiotics and immunological control because of the mucopolysaccharide matrix they produce from microbial and host components [56]. Keratinocytes are unable to migrate despite their high proliferation near the chronic wound margins because of their inability to activate and differentiate. To elicit a fibroblast response, high amounts of proinflammatory cytokines are expected, which will lead to an increase in MMP synthesis and a reduction in tissue inhibitor of matrix metalloproteinase secretion. Tissue injury and hypoxia are caused by the formation of fibrin and plasma protein-rich “cuffs” around microvessels in the form of fibrin and plasma [55].

The schematic representation of open wound risk and its immunological response. The image depicts the role of various immunological mediators involved in wound management.
The chemicals that microbes create and secrete, known as “virulence factors,” contribute to the pathogenicity of the microbe. Adhesins, capsules, endotoxins, exotoxins, flagella, lipases, pilli, and proteases are only a few of the various virulence factors that exist. They may play a variety of tasks, including aiding in microbial adhesion, invasion, or both, and promoting microbial growth in a host by evading host detection, blocking phagocytosis, and controlling intracellular survival. Microorganisms such as Gram-negative and Gram-positive bacteria, fungi, and viruses all generate proteases [57,58,59,60]. Numerous pathogenic bacteria, such as Staphylococcus, Streptococcus spp., Enterococcus spp., and Pseudomonas aeruginosa, generate a wide variety of proteases [61,62]. Regulatory factors, which regulate the transcription of protease genes in response to the local environment of the bacterium, may have a role in mediating the synthesis and release of bacterial proteases. Several environmental conditions, such as nutrients, development stage, osmolarity, pH, and temperature, might affect production. Quorum sensing is another possible impact [63,64,65,66,67,68]. Such elements could show up in the context of a soft-tissue illness. Higher levels of the P. aeruginosa elastase have been linked to increased inflammation and tissue damage [69,70], while protease-deficient P. aeruginosa strains have been found to be less virulent than their protease-producing counterparts in burn wound mouse models. Proteases produced by bacteria may sometimes provoke an immune response in the host. Several host elastolysis proteases, such as human leukocyte elastase and human neutrophil elastase [71], are stimulated by P. aeruginosa elastase A (LasA) protease. Although this seems paradoxical for survival since it helps remove bacterial organisms from the site, a prolonged inflammatory phase may result in a protracted activation of the host’s immune response, including host proteases, leading to wound chronicity [62,72]. Without curing the infection, the host’s immune system may actively deteriorate the tissue around it. This may allow the infection to spread to neighboring and deeper tissues.
Chronic wounds have been shown to have a pH environment ranging from 7.15 to 8.9 [73,74]. Both healing and non-healing wounds exhibit this degree of variation. Lower healing rates have been shown in both acute and chronic wounds with an alkaline pH [75]. An acidic pH is critical for wound healing, and it is important to note that the wound’s pH shifts from neutral to acidic as healing advances [76]. The scientists found that the tissue type, not wound severity, was associated with pH. Indeed, an increase in metabolic burden on the wound due to the presence of necrotic tissue and devitalized tissue results in tissue hypoxia [77]. Alkaline environments promote the disintegration of ECM and, by extension, the wound itself [78].
3 Nanotechnologies for wound healing
Currently, chronic wounds are treated by covering the wound, preventing bacteria by destroying and removing dead tissue, moisturizing it, and absorbing increased quantities of fluid [79]. Nanotechnology involves the study of structure as well as dynamics along with the synthesis of atomic as well as molecular particles (nanometric) upon which the building of nanoproducts is done [80]. Due to their qualities [81], nanotechnology platforms have shown new potential and advantages in the area. Recent advances in nanotechnology have enabled the transfer of biomolecules like DNA/RNA or GFs, which may be used in chronic wound healing, to new application areas in the field of medication delivery. Their compact size and physicochemical qualities enable intracellular delivery of these biomolecules or medications, preserve them from degradation, and improve drug penetration into the wound [82,83,84,85,86,87]. Together, these factors allow topical administration and prolong the half-life of therapeutic medicines, hence reducing the number of administrations and associated expenses. In addition, the encapsulation of medications and biomolecules inside nanocarriers permits various drug release patterns that may be tailored to the needs of wound healing. In the following sections, we will brief on how the NPs, nanofibers, and self-assembled nanocarriers are used in the treatment of chronic wounds focusing on those that have shown good wound-healing outcomes in diabetic animal models.
3.1 Electrospun nanofibers
Electrospinning technology, as a process for manufacturing ultrafine fibers, has experienced decades of development [88]. The electrostatic spinning equipment consists largely of four components: a high-voltage generator, a fluid driver, a spinneret, and a collection mechanism [89]. After the voltage is applied during the electrospinning technique, the original electrospinning fluid undergoes a gradual change in morphology until it reaches the critical voltage form of a Taylor cone. There are two stages of whiplash when the liquid jet reaches its maximum length. For example, the jet may be prolonged to micrometers or even a few hundred nanometers in length by using solvent volatilization [90,91]. For nanofibers to change their shape and size, it is necessary to regulate many factors, such as the molecular weight and viscosity of the polymer as well as the flow rate, the receiving distance, and the ambient conditions (humidity or temperature) [92,93]. Electrospinning is a simple, top-down, one-step preparation approach that provides nanofibers with microscopic pore size, high porosity, and a structure akin to ECM (as shown in Figure 3). As a consequence, it has gained considerable attention from researchers and is used to generate functionalized nanofibers for biomedical and other purposes (as shown in Figure 3) [94,95,96]. Moreover, electrospinning technology is consistently refined and increased. Figure 3 depicts the process of formation of nanofibers using the electrospinning technique, and their use as wound dressings for wound-healing activity, while the last part of Figure 3 defines the healing action of nanofibers including various inflammatory mediators.

Various processes involved in the preparation of nanofibers and their applications in the wound-healing process.
Nanofibers filled with berberine have been developed as a therapy for DFUs and were fabricated using electrospun cellulose acetate/gels (CA/gel) and filled with berberine (Beri) [97]. The hemocompatibility test showed that the produced dressings induced less hemolysis than the positive control (RBC lysed in distilled water). Moreover, the berberine-infused dressing displayed less hemolysis than the pure CA/gel dressing. Positive control and CA/gel dressings were shown to have lower antibacterial activity than the Beri dressing. Berberine inclusion in the CA/gel nanofiber matrix is responsible for the improved bactericidal action of CA/gel/Beri dressing described in research, and this value is advantageous for wound-dressing applications [97].
To improve wound healing by absorbing exudate at the wound site, speeding hemostasis, and preventing bacteria development and inflammation, Chen et al. created a three-dimensional (3D) layered nanofiber sponge (3D-AgMOF-CUR) with an in situ formed silver-metal organic framework (Ag-MOF) and curcumin (CUR) [98]. Reduced inflammation, higher collagen deposition, increased granulation and angiogenesis, and quicker skin regeneration are all examples of 3D-AgMOF-improved CUR’s wound dressing effectiveness. In part, this is due to its unique 3D layered structure, which allows for increased water absorption and air permeability during wound healing, as well as superior hemostatic capabilities. A possible antibacterial property of Ag-MOF formed on the sponge is enhanced if wound infection is assumed. This property might help to avoid infection and unhealing by rapidly destroying bacteria in the wound. Aside from reducing inflammation, increasing collagen deposition, and stimulating skin tissue regrowth, CUR loading may also speed wound healing.
Another study describes the development of a polycaprolactone-polyethylene glycol-egg yolk oil (PCL-PEG-EYO) scaffold for burn healing, using egg yolk oil, polycaprolactone, and polyethylene glycol (PEG) [99]. Results showed that the scaffold had a uniform morphology, hydrophilicity, and improved cell viability and attachment compared to the PCL-PEG scaffold. It also demonstrated antibacterial activity and enhanced wound closure, re-epithelialization, angiogenesis, and collagen synthesis in vivo. Overall, the PCL-PEG-EYO nanofibrous scaffold shows potential for the management of full-thickness burn wounds.
The study reported by Bouhajeb et al. described the fabrication of an implantable dressing material for reducing anti-pressure ulcer disease using electrospun poly(lactic acid) (PLA) nanofibers containing the hydroethanolic extract of T. ramosissimum [100]. The optimized synthesis conditions resulted in a homogeneous and nonwoven mat structure with identified chemical compounds including flavonoids, monoterpenoids, hydroxycinnamic derivatives, and phenolic acids. In vivo wound-healing investigations on induced pressure ulcers in mice showed that the implantable 17-PLA/HE NF material improved wound-healing capabilities, reduced the injury area, and provided a high wound closure percentage from the first days of application. Histological assays showed complete re-epithelialization with 17-PLA/HE NFs. This study offers the possibility to fabricate implantable materials containing only the determined released compounds for further research.
Anaya Mancipe et al. reported on a coaxial electrospinning process to fabricate a polycaprolactone-collagen/poly(vinyl alcohol) (PCL-Col_1/PVA) core–shell nanofiber mat for potential use as a skin wound dressing [101]. The PCL core improves mechanical properties, while PVA improves collagen processability. The triple helix structure of collagen is preserved, and common post-processing can be avoided. The resulting nanofiber mat exhibits high liquid absorption, structural stability, hydrophilicity, and collagen release capacity. The optimized nanofiber mat showed no cytotoxicity, making it a potential novel dressing for skin damage regeneration, especially for chronic wound treatment.
The research reported by Koohzad and Asoodeh discussed the development of a bioresponsive nanodrug delivery system for tissue engineering applications [102]. The system comprises HA–chitosan (CS)–polyvinyl alcohol complex nanofibers that are pH-sensitive, exhibit excellent properties for wound dressing, and are cross-linked using citric acid. The nanofiber scaffold is designed to release peptide molecules that accelerate wound healing, which was confirmed by in vitro and in vivo studies. The peptide-loaded nanofiber samples showed better adhesion, proliferation, migration, and fibroblast cell growth than other groups. In vivo studies in a mouse model demonstrated that the designed nanofiber was gradually absorbed without causing dryness or infection, and it showed excellent tissue repair ability. Gene expression studies indicate that the antimicrobial peptide promotes the inflammatory phase of wound healing in a shorter time frame by accelerating the tumor necrosis factor-α cytokine response. Various other studies reported on the development of electrospinned nanofibers for wound-healing purpose are given in Table 1.
Data of various polymers and their electrospinned nanofibers for wound healing
Polymer | Drug | Ref. |
---|---|---|
Honey, CS, and PVA | Cleome droserifolia (CE) and Allium sativum aqueous extract | [103] |
PCL/gelatin o/w (oil/water) nanofiber mat | Ketoprofen | [104] |
PVA/CS nanofiber mats | Tetracycline HCl | [105] |
Silk fibroin (SF)/PCL asymmetric membrane | Thymol | [106] |
PU/dextran nanofiber mats | Ciprofloxacin HCl | [107] |
Zein/Ag nanocomposite mats | Silver | [108] |
PCL nanofiber | CUR | [109] |
Collagen nanofiber mats | AgNPs | [110] |
Polygalacturonic acid and HA nanofibrous mat | AgNPs | [111] |
CS oligosaccharide/poly(vinyl alcohol) nanofiber | AgNPs | [112] |
SF asymmetric membrane | KR12 (antimicrobial motif derived from human cathelicidin peptide) | [113] |
Gelatin/PVA | Pine honey | [114] |
Gelatin/CS | Cinnamon | [115] |
Gelatin/PCL | Cinnamon | [116] |
Gelatin | Centella asiatica extract | [117] |
Gelatin/starch | Lawsonia inermis (henna) | [118] |
Gelatin/PCL | Lawsone (2-hydroxy-1,4-naphthoquinone) | [119] |
Gelatin/CA | Zataria multiflora | [120] |
Gelatin | Cinnamaldehyde, or Limonene, or Eugenol | [121] |
Gelatin/poly(lactic-co-glycolic acid) (PLGA) | Hypericum capitatum var. capitatum (HCC) extract | [122] |
Gelatin/PCL | Clove essential oil | [123] |
Gelatin/PVA | Carica papaya | [124] |
Gelatin/PCL | Oregano oil | [125] |
Gelatin | Chondroitin sulfate | [126] |
Gelatin/PCL | Trimethoxysilylpropyloctadecyldimethyl ammonium chloride (QAS) | [127] |
Gelatin/SF | Astragaloside IV | [128] |
Gelatin/SF | Astragaloside IV | [129] |
Gelatin/PCL | (+)-Catechin | [130] |
Gelatin/PCL | Ketoprofen | [131] |
Gelatin/PCL | Taurine (2-aminoethane sulfonic acid) | [132] |
Gelatin/PLA | Ciprofloxacin | [133] |
Gelatin/PVA | Gentamicin | [134] |
Gelatin/PCL/ZIF-8 | Gentamicin | [135] |
Gelatin methacryloyl (GelMA)/PCL | Cephalexin | [136] |
Gelatin/PVA | Cephradine | [137] |
Gelatin/SF | Ceftazidime | [138] |
Gelatin/PLGA | Liraglutide | [139] |
Gelatin/PU | Silver-sulfadiazine | [140] |
Gelatin/polyhydroxy butyric acid | Silver-sulfadiazine | [141] |
Gelatin | Vitamins A and E | [142] |
Gelatin/PLA | EGF | [143] |
Heparin/gelatin/PCL | Basic fibroblast growth factor (bFGF) | [144] |
Collagen nanofiber scaffold | Ostholamide | [145] |
CS/polyethylene oxide nanofibers | Silver and zinc NPs | [146] |
CA and soy protein hydrolysate | — | [147] |
Polycaprolactone | Spirulina extract | [148] |
Polycaprolactone nanofibrous mat | α-Lactalbumin | [149] |
3.2 NPs for wound healing
The difficulties involved with the care of refractory wounds are often linked to microbial contamination and infection. Elimination of these is essential for prompt wound healing [150]. The growth of multidrug-resistant infections has led to an increase in the usage of antimicrobial treatments based on NPs. As potent antimicrobials, metallic NPs are currently thoroughly explored and investigated. It is known that the NPs’ antibacterial activity is proportional to the surface area in contact with microbes. While the healing properties of silver have been known for millennia, its therapeutic potential has recently been greatly enhanced by the use of silver NPs in wound dressings and topical treatments. Silver ion release from wound dressings, hydrogels, lotions, and sprays may be prolonged with the help of these NPs. Silver nanoparticles’ (AgNPs’) sustained antibacterial action and less cytotoxicity toward healthy cells result from their regulated release of silver ions. AgNPs aid in wound healing in ways other than their antibacterial capabilities. They have been found to stimulate the growth of new blood vessels, a process essential for delivering oxygen and nutrients to injured tissue throughout the healing process. In addition, the healing process may be aided by AgNPs because of their ability to control inflammatory reactions. They have been shown to have antioxidant capabilities, making for an optimal wound-healing environment by shielding cells from oxidative stress.
The use of AgNPs has become evident in several clinical trials in wound therapy, particularly chronic wounds and burns [151]. It has been shown that Ag-alginates and collagen preparations are being used in clinical settings and that the efficacy of these products has been well studied. In most cases, the kind of silver used in the preparation of these condiments is a mystery. Nanocrystalline Ag is utilized in Acticoat instead of ionic Ag, which is used in other materials. Acticoat has been shown to reduce wound healing time for severe partial-thickness burns when compared to standard paraffin gauze dressings in a retrospective study of burn wounds in humans [152]. Overall, AgNP-based dressings are more effective than Ag sulfadiazine cream in terms of effectiveness [153]. AgNPs accelerate the process of healing by promoting contractility of wounds by induction of differentiation of myofibroblasts from normal fibroblasts. Further re-epithelialization of the epidermis is stimulated by AgNPs through keratinocyte proliferation as well as relocation [154]. Antibacterial wound dressings made of silver nanocomposites, which combine AgNPs with hydrogels for improved antibacterial activity and cytocompatibility, have been widely studied. Again, in animal trials, it has been observed that inorganic particles like silica adhere tightly to open wounds. This leads to the integration of AgNPs into silica NPs (mesoporous) with the help of disulfide bonds. The resultant compound has demonstrated excellent antimicrobial activities with minor toxicity at the cellular level [155].
Preclinical investigations have considered electrospinned scaffolds as well as those made using different techniques and AgNPs. Silver and CS nanocomposite dressings were synthesized and evaluated in rats with severe partial-thickness wounds using nanoscale and self-assembly technologies. Compared to Ag sulfadiazine, the nanocomposite dressings considerably accelerated wound healing and reduced Ag levels in the blood and tissues [156]. When exposed to a wound dressing made of cellobiose, CS, and AgNPs, laboratory rats with experimental wounds recovered more quickly than their untreated counterparts [157]. The wound contraction in rats treated with silver-based dressings was larger than that in rats treated with distilled water or ionic or nanocrystalline silver dressings [157]. In comparison to a commercially available silver alginate cream, guar gum alkylamine impregnated with AgNPs showed faster wound healing in rats [158]. The proliferation and migration of keratinocytes at the site of wound healing were aided by the nano-biomaterial. AgNPs derived from Naringi crenulata leaf extracts increased wound healing in rats when applied topically [159]. For wound healing and collagen and granulation tissue deposition, activated carbon fibers containing AgNPs showed great biocompatibility in vitro [160]. An alginate fiber-integrated AgNP increased fibroblast movement, decreased inflammation, and improved wound healing in preclinical and in vitro experiments, respectively [161]. In vivo studies of AgNPs for wound healing showed that in addition to decreasing inflammation and regulating fibrogenic cytokines, these NPs have antimicrobial properties [162]. Biosynthesis of AgNPs using Phytophthora infestans microbes revealed stability and better wound contraction ability when compared to Ag sulfadiazine in an in vivo excision wound model [153]. Synthesis employing the two glycosaminoglycans (chondroitin and acharan) as reducing agents improved the endurance of these composites without any observed aggregates.
To promote wound closure, these nanocomposites were applied to the wound surface and accelerated the formation of new tissue, including granulation tissue and collagen, in a mouse wound healing model [163]. Sulfobetaine was treated with a silver–clay nanohybrid to produce an antibacterial polymer with a long-lasting and protein-resistance antibacterial activity that was regulated by diffusion [164].
These biopolymers and NPs have considerable potential to improve wound healing, especially in the treatment of DFUs [154], which are still a huge issue and are connected to high amputation rates and clinical expenses. Testing of antimicrobial peptide-AgNPs composite on diabetic rats by Dai et al. has shown that the compound has wide-spectrum activity without causing resistance, as well as improved wound healing without affecting the dermal tissues [165]. Silver, which has a lot of antibacterial properties on several levels [166], makes it less likely that multidrug-resistant bacteria would acquire resistance. The wound-healing effects of AgNP were studied in albino rats by Kumar et al. AgNP-containing cream formulations reduced wound area, increased collagen deposition, fewer macrophages, necrosis, and increased levels of fibrous fibroblasts in mice [167]. When applied to bandages after thermal damage in BALB/C mice, AgNPs (14 nm) decreased inflammation and scarring while also preventing bacterial development [168]. Albino rats have also been proven to benefit from biosynthesized AgNP ointments, which demonstrate the usefulness of AgNPs [169]. Adibhesami et al. reported that the common pathogen Staphylococcus aureus, which causes wound infections, was suppressed by AgNPs in the wounds of mice implanted with the NP. Wounds infected with bacteria recovered more rapidly and efficiently in this study, with no side effects [170]. It was shown that nano-biocomposites containing AgNPs expedited wound healing in diabetic mice by dramatically increasing the expression of collagen and growth factors, improving re-epithelialization, vasculogenesis, and collagen deposition in comparison to control groups [171].
According to recent research [166], AgNPs may speed up myofibroblast differentiation, which in turn increases wound contraction and healing time. Silver has also been proved to promote keratinocyte proliferation and relocation throughout the healing process. An excisional wound model in mice was used to study the impact of AgNPs on skin cells. According to histology and wound model experiments, AgNPs increase keratinocyte proliferation and migration as well as keratinocyte growth and maturation, which results in wound contraction. AgNP-treated wounds healed faster than those in the control group [172,173]. While wound healing, Frankova et al. studied the effects of AgNPs on primary human keratinocyte and fibroblast cells to observe how they compared to the effects of AgNPs on the most exposed cells. Proinflammatory cytokines and bacterial growth inhibition are benefits of AgNPs in wound healing. A study by Tian et al. found that AgNPs may cause an inflammatory response in a mouse model when applied topically. To better understand the function of cytokines in wound healing, scientists employed quantitative real-time RT-PCR (reverse transcription polymerase chain reaction) to examine the levels of TGF-β1 (transforming growth factor-beta-1), IL-10 (interleukin 10), VEGF (vascular endothelial growth factor), and IFN-α (interferon alpha). Changes in the mRNA levels of many cytokines were shown by AgNPs in the cytokine profile. According to Tian et al.’s findings [162], scars would be less obvious if they were coated with AgNPs. In another study, researchers formulated AgNP-based biocompatible film. In vitro experiments on HDFa cell lines confirmed the biocomposite films’ biocompatibility and revealed their antibacterial effectiveness against S. aureus. In vivo testing on rabbits revealed that the C2P4.10.Ag1-IBF film sample showed reduced fibrosis through the expression of TNFAIP8 (TNF alpha-induced protein 8) factors, MHCII (major histocompatibility complex class II) as an anti-apoptotic marker that promotes immune cooperation among local cells, SMA as a marker of the presence of myofibroblasts moving toward the interepithelial spaces in preparation for epithelialization, and Cox2 as an indicator of inflammation [174].
There is a promotion of healing and inhibition of colonization of microbes by gold nanoparticles (AuNPs). Growth as well as differentiation of keratinocytes is enhanced by AuNPs at low concentrations [175]. When AuNPs are combined with polymers or stem cells, wound-healing activity increases. The CS-AuNP enhances the free radical scavenging activity of AuNPs several-fold. As a result, polycationic CS is an important intermediary molecule in the production of AuNPs. Combining the Tegaderm dressing with chitin-AuNPs boosted hemostasis and accelerated wound healing in a rat surgical wound model, according to Volkova et al. [176]. In a second rat in vitro study, AuNPs were delivered to burn sites together with cryopreserved human fibroblasts (CrHFC-AuNP). Enhanced collagen deposition was seen in CrHFC-AuNP-treated wounds compared to non-treated wounds [177]. Gold nanoshells have shown considerable absorption cross-links without photobleaching, allowing laser-tissue repair. Using molecular chemistry and self-assembly techniques, peptide NPs may be created. Peptide-based NPs with medicinal uses, such as drug delivery, are a hot topic in synthetic biology at present. Peptide nanostructures may also be employed in the study of cell signaling and the development of biological therapies. As a result, these self-assembled peptide scaffolds may be functionally changed so that they can better interact with a variety of cell and tissue types. Peptide hydrogels, which are often used as examples of biocompatibility and cytocompatibility, are compatible with a wide range of mammalian cells and biological systems. Because of their ability to transport therapeutic substances to the site of damage and stimulate tissue regeneration, lipid NPs play a crucial role in the healing process. These lipid-based NPs, including phospholipids and solid lipids, offer desirable qualities for use in wound healing. The capacity of lipid NPs to encapsulate and distribute therapeutic substances such as drugs, growth hormones, and nucleic acids directly to the wound site is one of their key roles in wound healing. Lipidomic NPs preserve and improve the stability of these therapeutic substances, allowing for their continuous release at the wound site. By delivering the medicine directly to the site of the injury, this technique speeds up the healing process and lowers the negative effects of systemic dosing. Wound healing is aided by the innate capabilities of lipid NPs. Because of their compact size and high surface area to volume ratio, for instance, they may efficiently absorb cells and penetrate the wound bed. As a result, more therapeutic chemicals are able to reach the surrounding cells and tissues, boosting their potential to heal. The lipid-based structure of these NPs makes them biocompatible and less likely to induce unfavorable immune responses since they match the makeup of cell membranes. Further improving their targeting effectiveness and therapeutic efficacy is the addition of ligands to the surface of lipid NPs that preferentially bind to receptors expressed on wound-associated cells [178,179,180,181,182,183,184].
Among the lipid NPs, the capability to augment the accumulation of drugs in the skin is exhibited by liposomes thereby contributing to the healing of wounds. For the development of liposomes and lipid NPs, phospholipids are commonly used. Lipids exhibit biocompatibility and biodegradability as well as controlled release and greater loading of drugs. They also facilitate the transport of drugs because of their capability to convert lipids of skin into fluid [185].
When manufactured peptides are employed, peptides may be folded into fibrils and activated with the right cell culture media. When evenly given in cultured cells, these peptide hydrogels had no negative impact on cell survival. Peptide hydrogels, recently produced, seem to promote cell adhesion and hepatocyte development in vitro, according to preliminary investigations [186]. As a result, liver tissue regeneration is significantly affected. Native endothelial cells (ECs) may benefit from an improved microenvironment when given using peptide hydrogels [187].
Peptide amphiphile (PA) systems with bioactive epitopes have been used to improve scaffolds for certain cell types of interest. Cell adhesion antigens, for example, have been proven to be effective when combined with a specific ligand. RGDS and PA collaborated to prove an alternative support system for bone marrow mononuclear cells and enamel epithelial cells. The experimental group missing the RGDS epitope had fewer bone marrow mononuclear cells than the control group [188]. The hydrogels made by including PA into an adhesion epitope had a progressive cell-responsive matrix, and this offered a favorable environment for the proliferation of dental stem cells, according to a separate study. The combination of several HAs has also recently been identified to generate a self-sealing pouch [189]. For tissue regeneration, this bag, which is filled with the proper liquids, may surely include human mesenchymal stem cells (MSCs). Conjugated polypeptides have been found to promote bone regeneration and functional recovery of chondrocytes in bone injury [190]. In tissue regeneration, peptide hydrogel biomaterial outperformed more traditional natural biomaterial in terms of efficacy and safety. Cell proliferation and differentiation during tissue repair were first aided by self-assembled nanostructures. You et al. formulated AgNPs (Nag) loaded with CS and collagen scaffolds [191]. The use of dermal scaffolds made of collagen and covered with AgNPs might serve as a novel non-toxic antibacterial dressing. The membrane is readily destroyed by NAg, which then enters the microbial body and converts into silver ions (Ag+) in the cytoplasm, harming the intracellular structure as a secondary effect. To interact with the Gram-negative bacteria E. coli, as a result, the possibility exists that NAg with the same surface area, but different shapes, might have distinct biological effects (as shown in Figure 4).

Antibacterial effect of NAg on wound-healing mediators. Various inflammatory mediators involved in the wound-healing action of Nag are defined.
Various other types of NP-based systems for wound healing are defined in Table 2.
Data for various NP-based systems for wound-healing purposes
Nanoparticle system | Reported activity | Ref. |
---|---|---|
Polydopamine/puerarin (PDA/PUE) NP-incorporated polyethylene glycol diacrylate hybrid hydrogel (PEG-DA/PDA/PUE) | Hydrogel showed excellent cell proliferation and antioxidant activity, including reduced ROS and increased glutathione peroxidase and superoxide dismutase activity under oxidative stress conditions | [192] |
Cerium oxide NP incorporated electrospinned poly(3-hydroxybutyrate-co-3-hydroxyvalerate) | NPs incorporating(poly(3-hydroxybutyric acid-co-3-hydroxyvaleric acid) membranes have high potential as wound dressings to boost cell proliferation and vascularization and increase diabetic wound healing | [193] |
Collagen/lipid NP-CUR cryostructurates | These hydrogels offer a framework for cell contact with NIH 3T3 fibroblasts as early as 4 h after seeding, without cytotoxicity | [194] |
AgNP-impregnated CS-PEG hydrogel | Chronic diabetic wound healing with improved antimicrobial, antioxidant, and wound-healing properties | [195] |
Cerium oxide NP-loaded polyvinyl alcohol nanogels | Nanogels that can be used for surgical wounds | [196] |
Synthetic CS-based composite hydrogel, named SNPECHG, incorporating silver ions (Ag+) and NP-encapsulated EGF | Histological study proved wound re-epithelization, collagen deposition, and faster collagen maturation | [197] |
Guar gum/CUR-stabilized AgNP hydrogels | Histopathology shows that the hydrogel composite accelerates tissue remodeling through normal routes, boosting the creation of fibroblasts and new blood vessels | [198] |
Thiolated hyaluronic acid (HA-SH) and bioactive silver-lignin nanoparticles (Ag@Lig NPs) based hydrogels | In situ self-assembly of HA-SH and Ag@Lig NPs supply nanocomposite hydrogels that prevent wound chronicity factors such as overexpressed proteolytic and oxidative enzymes and high bacterial load | [199] |
Erbium borate NPs | Promoted scarless wound healing | [200] |
Insufficient delivery of the desired concentration of antibiotics to the target bacterial cells is a major limitation of conventional antibiotic or antimicrobial treatment against biofilm infections in non-healing chronic wounds [201]. This is associated with the presence of a complex matrix network of biofilms and the avascular nature of chronic wounds [202]. If this occurs, not only may the therapeutic efficacy of orally or topically delivered medications be significantly reduced, but drug resistance could also be induced. Since extracellular polymeric substance (EPS)-forming biofilms may bind soluble antimicrobials to matrix components, preventing them from reaching target cells, they can reduce the diffusion of antimicrobial drugs to individual bacterial cells [203].
NP-encapsulated medicines have a greater chance of penetrating EPS and reaching their intended targets than do free drug molecules allowing for more effective therapeutic antibiotic administration to bacteria. NPs that are both biocompatible and biodegradable have been used as carriers for antibiotics, increasing their efficacy and allowing them to remain in the body for longer. The use of liposomes and polymeric NPs as carriers for antibiotic delivery against biofilms [204] is one example. Because their lipid bilayer structure is similar to that of cell membranes, liposome NPs have found widespread usage as drug delivery carriers in numerous biological applications [205]. The encapsulated medicine may be delivered to the cell membranes or the inside of the bacterium once the liposome NP fuses with the bacterial cell wall [206]. Liposomal antibiotic delivery to biofilms has been shown to be more therapeutically effective than free antibiotic administration, according to recent investigations. For instance, the β-lactam antibiotic piperacillin can be protected from hydrolysis by staphylococcal β-lactamase if it is encapsulated within liposomes [207]. This increased activity for piperacillin-encapsulated liposomes against S. aureus was correlated with the drug’s resistance to hydrolysis by β-lactamase. Liposomal encapsulation of gentamycin has been shown to dramatically increase its antibacterial effectiveness against P. aeruginosa biofilms, as shown in a separate investigation by Mugabe et al. [208].
NPs constructed from PLGA are well-studied polymeric NPs. The benefits of PLGA NPs include their biocompatibility and biodegradability, as well as the ability to tune the degradation profile of PLGA to regulate the release kinetics of loaded medicines [209]. Antibiotics delivered by PLGA NPs have been proven to be effective against numerous bacterial species, including P. aeruginosa [210], S. aureus [210], and Escherichia coli [211]. Lately, a novel kind of drug delivery platform called lipid–polymer hybrid NPs has evolved [212], which combines the benefits of liposomes and polymeric NPs. The core of the hybrid NP is formed of polymeric NPs, and the lipid layers around it provide structural stability and regulated biodegradability [213]. Lipids are very biocompatible; therefore, the hybrid NP has the benefits of lipids as well. Few studies have been conducted on the use of lipid–polymeric hybrid NPs for the controlled release of antibiotics [213], but it has been shown that hybrid NPs exhibit a higher cellular delivery efficacy than either liposomes or polymeric NPs when targeting cancer cells [214]. Because of this, the hybrid NPs may serve as an alternate antibiotic delivery platform to liposomes or polymeric NPs for the treatment of wound biofilm infections.
NPs’ potential to boost healing is impressive, and there are many opportunities for further development and use. Nevertheless, it should be noted that the wound site is not protected by unbroken skin. Since NPs utilized for wound healing are in touch with living tissue, ensuring their biological safety is essential before application [215]. Skin irritation and allergy are examples of the transdermal noxiousness of NPs that are often cited. Carbon nanotubes (CNTs) and nickel NPs, for example, have been linked to skin hypersensitivity due to their ions and surface coatings [216]. Transdermal skin exposure to NPs has been shown to aggravate preexisting skin inflammation, irritation, and psoriasis [217,218]. Exposure to NPs has been shown in certain investigations to cause oxidative stress, autophagy, and programmed cell death in fibroblasts and keratinocytes [219]. An NP’s toxicity is decided by its size, shape, surface charge, consistency, and concentration. Thus, it is essential to modify the physicochemical features of new NPs when developing them for wound therapy, in order to lessen their toxicity to skin cells [220].
The study by Vijayakumar et al. focused on using probiotic bacteria to produce NP-conjugated probiotics for improved antibacterial and wound-healing activity [221]. The isolated Lactiplanti bacillus plantarum strain was found to synthesize AgNPs, which were confirmed using ultraviolet-Vis spectroscopy, Fourier transform infrared spectroscopy, and high-resolution transmission electron microscopy. The biosynthesized AgNPs showed increased antioxidant and antibacterial activities and demonstrated significant wound-healing capabilities with 96% wound closure observed through an in vitro scratch-wound assay. The study provides a new green synthesis platform for probiotic bacteria to produce AgNPs, showing potential for wound healing against infectious pathogens.
Another study developed a method for synthesizing stable Nzo NP gels using zinc acetate as a precursor and optimized the synthesis parameters to achieve the most stable gels at room temperature, with a molar concentration of zinc C(Zn2+) ranging from 0.05 to 0.2 M at pH 8. Hydroxyethyl cellulose was found to be the optimal polysaccharide for stabilizing the ZnO NP gels, which showed irregularly shaped particles assembled into aggregates, with sizes ranging from 150 to 1,400 nm [222]. Dressings have been prepared by incorporating ZnO NPs in a matrix (bioresorbable), which is constituted of collagen and essential oil (1%). Such wound dressing has facilitated the closure of wounds and has helped in the prevention of the growth of bacteria both in vivo and in vitro [223]. The gel of ZnO NPs modified with hydroxyethyl cellulose was found to have a regenerative effect on burn wounds, significantly higher than that of the control group and the group treated with a gel of ZnO microparticles and hydroxyethyl cellulose. The healing rate of burn wounds in animals treated with gel of ZnO NPs with hydroxyethyl cellulose was 16.23% higher than in animals treated with gel of ZnO microparticles with hydroxyethylcellulose and 24.33% higher than in the control group treated with hydroxyethylcellulose. Successful impregnation of ZnO NPs in the cellulose membranes of bacteria was carried out. It has been shown to exhibit antibacterial activity against E. coli, S. aureus, and P. aeruginosa. In a burn model of mice ZnO containing bacterial cellulose, nanocomposites have shown the activity of healing significantly with fine regeneration of tissue proven by analysis histologically [224]. AuZnO core–shell composite NPs have been found to exhibit anti-biofilm as well as antibacterial activities against Staphylococcus haemolyticus (methicillin resistant) and S. aureus by releasing ROS. Thus, the proficiency of wound healing is accelerated [225].
Delayed wound healing is a common problem in many cases due to disruptions in the physiological healing process using the current wound-healing procedures. Hydrogel wound dressings have shown promise in enhancing granulation tissue and epithelium formation in the wound area by providing a moist environment. However, challenges such as exudate accumulation, bacterial proliferation, and reduced levels of growth factors have limited their effectiveness. To address these challenges, researchers developed a hydrogel wound dressing by loading platelet-rich fibrin–CS NPs into gelatin–CS hydrogel (Gel-CH/CH-PRF) using a solvent mixing method [226]. The goal was to evaluate the characteristics of the hydrogel dressings, sustained release of proteins, and reduction in the risk of infection in the wound area. The Gel-CH/CH-PRF hydrogel demonstrated excellent swelling behavior, porosity, absorption of wound exudates, and proper vapor permeability rate, providing the requisite moisture without dehydration around the wound area. The hydrogel dressing also showed a sustained release of proteins with excellent control over the release. Additionally, the hydrogel dressing showed high antimicrobial activity against both Gram-positive and Gram-negative bacteria. The presence of CS in the hydrogels resulted in the lowest scavenging capacity-50 value and the highest 2,2-diphenylpicrylhydrazyl (DPPH) radical scavenging activity at a concentration of 25 μg mL−1 for the Gel-CH/CH-PRF hydrogel. The hydrogels also demonstrated excellent cell viability and proliferation. In vivo studies on a full-thickness wound model showed that the Gel-CH/CH-PRF hydrogel dressing significantly increased wound closure and epidermis thickness. The findings demonstrate the potential of the Gel-CH/CH-PRF hydrogel as an ideal wound dressing for accelerated wound healing.
Researchers developed a multifunctional wound material by modifying camelina oil bodies with AgNPs and covalently bonding them with human fibroblast growth factor 2 (hFGF2) for the treatment of bacterial infection wounds [227]. The AgNPs-hFGF2-OB demonstrated broad-spectrum antibacterial activity against S. aureus and E. coli, and effectively promoted the migration of NIH/3T3 cells, showing good biocompatibility. In a mouse bacterial infection wound model, the AgNPs-hFGF2-OB group showed a significantly higher wound healing rate compared to other treatment groups, especially on the seventh day after treatment. Therefore, AgNPs-hFGF2-OB has the potential to inhibit bacterial growth, promote collagen deposition, granulation tissue regeneration, and angiogenesis, without any significant toxicity, making it a promising wound dressing for the repair of bacterial infection wounds.
A new approach using a mussel-inspired strategy was developed to create a multifunctional hydrogel named CAC/PDA/Cu(H2O2), which is composed of carboxymethyl CS/sodium alginate (Alg)-based hydrogel with a PDA coating induced by H2O2/CuSO4 [228]. The CAC/PDA/Cu(H2O2) hydrogel has excellent biocompatibility, mechanical properties, and degradability, and it is capable of antibacterial, anti-inflammatory, and angiogenesis-promoting effects in vitro. In a rat model of methicillin-resistant S. aureus (MRSA)-induced infection, CAC/PDA/Cu(H2O2) hydrogel efficiently eliminates MRSA, reduces inflammatory expression, promotes angiogenesis, and shortens wound-healing time. On days 7, 11, and 14, the CAC/PDA/Cu(H2O2) hydrogel exhibited the best wound-healing rates at 80.63 ± 2.44, 92.45 ± 2.26, and 97.86 ± 0.66%, respectively. The multifunctional hydrogel provides a promising potential for wound management and therapy for wound healing.
3.3 Hydrogels
Hydrogels have been studied as a viable wound dressing material in recent years [229]. Hydrogel wound dressings are used for both incisional and excisional wounds [230,231]. Hydrogels fit the definition of a 3D cross-linked polymeric network structure because they can hold a large amount of water while preserving their structural integrity after expanding [1,2,232,233,234]. The hydrogels consist of several hydrophilic groups that crossinteract thereby causing the formation of a matrix (3D), which can trap liquids, viz. exudates from the wound [235]. Materials for tissue regeneration that mimic natural 3D structures have elasticity like native tissue, can supply a moist environment, and absorb wound exudates including biomimetic hydrogels. These hydrogels allow for gas exchange to prevent the growth of anaerobic bacteria, act as a barrier to bacteria growth, and improve epithelization and cell migration into wound hydrogels (Figure 5) [236,237]. Another possibility is injectable hydrogels. Using injectable hydrogels for skin regeneration has several advantages, including the ability to fill wounds with uneven space, adherence to wounds, and the ability to contain bioactive substances and cells in situ [238] Traditional hydrogels may not be effective for wound healing because of their poor mechanical properties and inability to mimic the native tissue architecture [239]. In contrast, conventional hydrogels can only supply a moist environment and are restricted in their capabilities. Hydrogels having diverse functionalities that enhance wound healing have been the subject of several investigations. Added wound healing capabilities may be provided by multifunctional hydrogels in addition to creating a milieu replicating the natural tissue ECM. To reduce microbial invasion and hydrogel dehydration, these hydrogels may also be constructed in a bilayer form.

Representation of advantages of hydrogel cell therapies for wound healing. The hydrogels can prevent the entry of pathogens on the wound site, as they act as a protective barrier.
Due to the advancement in the field of synthetic chemistry and materials science, in situ formation of hydrogel is now possible upon delivery through standard needles. This ensures minimum invasion while delivering therapeutic payloads, and can help in induction of the regeneration process of the part of the body that is damaged [240].
Hydrogels with antimicrobial NPs have been widely investigated. In the study of wound healing, zinc oxide nanoparticles (ZnO NPs) [241], AgNPs [242], and AuNPs [243] have been used as nanocomposites in hydrogels. Antibacterial effects of most metallic NPs are decided by adherence to the membrane, destruction of the cell wall, and leakage through the outer layer of bacterial components, such as nucleic acids. Suppression of protein synthesis is the last step. In the case of metallic oxide NPs, photocatalysis serves as the principal antibacterial mechanism. There are two types of free radicals that are formed when UV irradiation strikes metallic oxide NPs: oxygen and hydroxyl radicals [244,245]. The antibacterial properties of metal NPs may be altered by a wide range of factors. AuNP’s form and surface modification were examined in one study, for example, to see how they affect wound healing. Based on the results of this study, a thermosensitive hydrogel incorporating AuNPs was developed for wound healing (rods of AuNR or spheres of AuNS). As a result, of this research, the charge of material was altered using PEG, PAH (polycyclic aromatic hydrocarbon), and PAA (peroxyacetic acid). After 21 days of therapy with poloxamer hydrogels having PEG-modified and PAH-modified AuNRs, significant wound healing without scarring was seen. In addition, compared to the other groups, the hydrogel having PAH-AuNRs exhibited considerable collagen deposition, suggesting that the lesion had healed. Despite the nanocomposite hydrogels’ antibacterial characteristics, the PEG-AuNR group had a low number of bacterial colonies. There was no bacterial growth when PAH-AuNR wounds were swapped in vivo for wounds from other groups. PAA-AuNRs, PEG-AuNS, and poloxamer hydrogels all showed mixed bacterial growth [246]. There may be a correlation between wound healing and the formation of gold aggregates in the form of gold–protein aggregation and a better organization of collagen in wounds treated with the PAH-AuNRs hydrogel. Using PEG-AuNRs hydrogel may help heal wounds because of its hydrophilicity, high absorption, and adhesiveness. The higher antibacterial activity of AuNRs in comparison to AuNS may be explained by the fact that wound healing is influenced by NP shape [247].
Anthocyanins, which are flavonoids present in a variety of fruits and vegetables, have been shown to have a wide range of beneficial effects on human health, including anti-inflammation, antibacterial, antioxidant, and angiogenesis. Hydrogels were created by adding hesperidin to the alginate/CS solution of Bagher et al. [248]. Researchers found that combining hesperidin with hydrogel improved wound healing positively. Polyphenols in grapefruit seed extract (GFSE) are classified as tannins because of the presence of these compounds. Hydroxypropyl cellulose and sodium carboxymethyl cellulose hydrogel films have improved their ability to suppress bacteria growth after being treated with GFSE [249]. A toxicologically diverse, marine natural product is found in a variety of Chinese herbal treatments such as Sophora flavescens, Radix sophorae, and many more. Although it helped maintain a good wound-healing environment, the marine-loaded konjac/fish gelatin hydrogel also inhibited the spread of germs from developing on the wound surface [250]. These hydrogels may be used in wrist-healing applications.
Huang et al. produced CS and polyethylene glycol diacrylate (PEGDA) hydrogels [251] by directly combining PEGDA/CS-PSI hydrogels with trp-rich PSI and CS. Although PSI has a broad variety of antibacterial characteristics, its cytotoxicity is low, and it does not build bacterial resistance when overused. PSI-loaded hydrogels outperform PSI-free hydrogels in terms of antibacterial efficacy against E. coli and S. aureus.
Chronic wounds were treated with GelMA and recombinant tropoelastinmethacryloyl-substituted (MeTro) hydrogels [252]. Later, AMP Tet213 was put right into the hydrogel solution, which was then used to photo-cross-link the hydrogel precursor solution. The bactericidal activities of AMP Tet213 and ZnO NPs were also studied using MeTro/GelMAZnO hydrogels. In terms of both qualitative and quantitative antibacterial activity, AMP Tet213 and ZnO NPs were compared. To address the limitations of ZnO NPs, AMP-based antibacterial techniques may be able to supply an alternative. Because of their long-term activity, noble metal ions have long-term suppressive effects on bacterial growth and photothermal properties. When combined with heat treatment, inherent antibacterial activity may have a quick, highly effective, and long-lasting antibacterial impact. Gallic acid-modified Ag (GA-Ag) NPs were implanted into the carrageenan network structure to form an antibacterial hydrogel [253]. GA-AgNPs may be employed as a photothermal agent to kill bacteria when exposed to NIR irradiation.
CNTs supply a long-term antibacterial activity that is persistent in the presence of light. NIR radiation may be converted to heat energy with their help. CNTs may be added to hydrogels to improve cell adhesion, proliferation, and differentiation. CNT-based hydrogels for PTA treatment of bacterial wounds have been attracting increasing attention during the last several years. Antibacterial sticky and conductive gelatin-grafted-dopamine (GT-DA)/CS/CNT composite hydrogels were generated by the oxidative coupling of catechol groups between GT-DA and PDA-coated CNTs [254]. Guo et al. have developed a CEC-PF CNT hydrogel that has excellent NIR stimuli responsiveness and remarkable photothermal antibacterial activity. Antibacterial properties and therapeutic efficacy were equivalents between the PTA group and the antibiotic-loaded group [255]. These photothermal hydrogels are a promising way for the treatment of infected skin defect wounds.
Researchers have developed an all-natural hydrogel wound dressing with intrinsic immunomodulatory ability that can regulate macrophage heterogeneity by increasing the conversion of pro-inflammatory macrophages to anti-inflammatory phenotypes [256]. The hydrogel is bioadhesive, scavenges ROS, and has antibacterial properties, providing effective protection to the trauma surface from secondary damage. The prepared FGMA/FG/PA hydrogel has been shown to have good anti-infective and immunomodulatory abilities in vitro and in vivo experiments. The hydrogel can regulate the immune microenvironment at the trauma site, shorten the inflammation period, promote vascular regeneration, and have good therapeutic effects on diabetic trauma. The hydrogel has been found to be able to activate the immune regulation of macrophages, and it shows promising applications for diabetic wound repair and other immune-related disease treatments. The hydrogel can potentially improve wound healing by regulating macrophage heterogeneity, and it is expected to have significant implications for the development of advanced wound dressings. Interestingly, it has been found that compared to hydrogels without bFGF and wound-healing products commercially available, bioinspired hydrogels with bFGF can cause significant enhancement of proliferation of cells. Moreover, they are also more efficient in inducing re-epithelialization of the wound, deposition of collagen, and contraction of the wound without any significant inflammation and toxicity [257].
Titanium-oxo-clusters (TOCs) are widely used in various fields but their application in biomedical areas is limited due to their unstable structure and lack of antibacterial and anti-inflammatory properties. Luo et al. developed a stable, antibacterial, and anti-inflammatory Ag-titanium-oxo-cluster (Ag-TOC) by introducing silver and salicylic acid using a solvothermal method [258]. The Ag9Ti4 cluster was then incorporated into a dopamine-containing hydrogel system, which demonstrated good antibacterial and photothermal properties in vitro. The Ag9Ti4 hydrogel system also showed better anti-inflammatory and wound-healing ability in vivo under NIR conditions. The study suggests that the Ag-TOC hydrogel system could provide a new strategy for the expanding biomedical applications of TOCs.
Adipose-derived stem cells (ADSC)-originating exosomes (ADSC-Exos) have been identified as a potential strategy for diabetic wound repair. However, the fast decrease in biological activity and unknown biological mechanisms limit their clinical application. To address these issues, hypoxia-pretreated ADSC-Exos (ADSC-HExos)-embedded GelMA hydrogels (GelMA-HExos) were developed via non-covalent force and physical embedding, creating a new approach for clinical treatment of diabetic wound repair [259]. In vitro, GelMA-HExos hydrogels had a loose porous structure, stable degradation, and expansion rate. In vivo, GelMA-HExos hydrogels promoted wound healing in diabetic mice. Furthermore, ADSC-HExos had an enhanced therapeutic effect, in which circ-Snhg11 expression was increased. circ-Snhg11-modified ADSC-Exos increased the migratory, proliferative, and blood vessel regeneration potential of vascular ECs. Overexpression (OE) of NFE2L2-HIF1α or inhibition of miR-144-3p – both of which are members of the miR-144-3p/NFE2L2/HIF1α pathway downstream of circ-Snhg11 – reversed the therapeutic effects of circ-Snhg11 [259]. This study explored the effects and downstream targets of hypoxic engineered exosome hydrogels in managing diabetic wound repair and these hydrogels are expected to have application possibilities in other disease areas. The regulation mechanism study found that circ-Snhg11 delivery from GelMA-HExo incremented survival and maintained EC function, possibly via the activation of miR-144-3p/NFE2L2/HIF1α signaling. These findings suggest a new therapeutic strategy for patients with diabetic ulcer. However, further investigation is needed to reverse the problems associated with poor mechanical properties, low biological activity, short duration of effect, and high risk of sudden release of exosomes [259].
A new physical dual-network multifunctional hydrogel adhesive has been designed based on polysaccharide material for the treatment of full-thickness skin defects infected with multidrug-resistant bacteria [260]. The hydrogel uses ureido-pyrimidinone (UPy)-modified Bletilla striata polysaccharide and dopamine-conjugated di-aldehyde-HA as matrix materials for strong biocompatibility and wound-healing ability. The catechol-Fe3+ and quadrupole hydrogen-bonding cross-linking of UPy-dimer provide a highly dynamic physical dual-network structure that imparts good rapid self-healing, injectability, shape adaptation, NIR/pH responsiveness, high tissue adhesion, and mechanical properties of this hydrogel. Bioactivity experiments showed that the hydrogel also possesses antioxidant, hemostatic, photothermal-antibacterial, and wound-healing effects. This functionalized hydrogel shows promise as a candidate for clinical treatment of full-thickness bacteria-stained wound dressing materials.
The development of a thermosensitive hydrogel that undergoes a sol–gel transition at body temperature has been presented as an effective wound dressing for promoting healing and preventing infections. A study introduces a ROS-scavenging hydrogel based on polydopamine-modified poly(ε-caprolactone-co-glycolide)-b-poly(ethyleneglycol)-b-poly(ε-caprolactone-co-glycolide) (PDA/P2) triblock copolymer [261]. The PDA/P2 solution at 30 wt% concentration forms a gel at 34–38°C and was found to have ROS-scavenging properties through DPPH and ABTS assays and intracellular ROS downregulation in RAW264.7 cells. Additionally, AgNPs were encapsulated in the hydrogel to provide antibacterial activity against E. coli and S. aureus. An in vivo study on an S. aureus-infected rat model demonstrated that the PDA/P2–4@Ag hydrogel dressing could promote wound healing by inhibiting bacterial growth, alleviating the inflammatory response, and inducing angiogenesis and collagen deposition. This study highlights a new approach to preparing temperature-sensitive hydrogel-based multifunctional wound dressings.
The development of novel hemostatic materials with wound-healing properties has gained significant attention in recent years [262]. A series of ultrafast cross-linked adhesive hydrogels have been developed using biomass-derived materials such as Schiff base and ionic coordinate bonds among catechol-conjugated gelatin, dialdehyde cellulose nanocrystals, calcium ions, and ferric iron. The hydrogels exhibit adjustable gelation time and mechanical properties by altering the contents of dialdehyde cellulose nanocrystals and ferric iron. The hydrogels are also endowed with self-healing and injectable performance, enhanced adhesiveness, near-infrared responsiveness, and antibacterial activity due to the introduction of catechol groups and the formation of catechol–Fe complexes. The hydrogels demonstrate rapid hemostasis and biodegradability in vitro and in vivo experiments. Furthermore, the hydrogels accelerate the regeneration of wound tissues in a rat full-thickness skin defect model, suggesting their potential as hemostatic materials for wound-healing applications in the biomedical field.
3.4 Smart dressings
In most instances, dressings based on nanotechnology are used for delivering actives to the wound bead. When the size of the materials is reduced to nanoscales, there is a change in the physicochemical features. This can also help in influencing and accelerating the process of healing [263,264]. The pH of healthy skin is between 4.0 and 6.0. Further evidence that the pH of the wound environment changes from alkaline to neutral and then acidic as a wound heals was reported by Gethin, who also discussed the significance of pH in acute and chronic wound healing [265]. But this is a widely accepted concept [266]. The wound site has a lower pH than the surrounding skin, regardless of the pH. pH-responsive dressings have been utilized to treat infected and chronic wounds [267,268,269] based on this pH difference. Externally controlled change and pH change may be categorized as stimulus-sensitive materials using the following: the work of Kiaee et al. [268] demonstrates an example of an externally controlled wound dressing that reacts to pH variations. The pH of the wound dressing was adjusted using electrical voltage throughout the study. Wounds were treated with CS NPs enclosed in a PEG-diacrylate/laponite dressing.
A whole new hydrogel material constituted of genipin and polysaccharide of Bletilla striata crosslinked with CS has been used for wound dressing. This combination presented comparatively better features than CS crosslinked solely with genipin. However, this material exhibited poor activity against bacteria. Thus, to overcome this issue, the proposal of a nanohybrid has been given. The nanohybrid in its final formulation is incorporated with AgNPs. The dressing with nanohybrid provides permeation of gas and has the ability to retain water. Moreover, it suppresses the proliferation of bacteria and augments the proliferation of fibroblasts thereby showing its great potential for using in order to facilitate the healing of wounds further [270].
Zinc and copper wires were used as anodes and cathodes, respectively, to apply a direct current voltage to the hydrogel. As the positive charge moved away from the anode in this experiment, a redox reaction at the electrode increased the pH (the cathode was not immersed in the hydrogel). The deprotonation and shrinking of CS NPs in a basic environment, and the weakening of the electrostatic link between laponite and these NPs as pH rises, might promote drug release from these particles. With this externally managed pH change, wound dressing drug release may be consistently and swiftly increased. The external control is cumbersome in practice for this pH-sensitive wound dressing, which requires a very high pH value for best medicine release.
The acidic environment of the stomach, on the other hand, is a given. They developed an acidic pH-responsive hydrogel to prevent stomach bleeding, and this enhanced gastric wound healing considerably [271]. When acryloyl-6-aminocaproic acid and acryloyl-NHS (AA) were polymerized, hydrogels were created. This hydrogel shows exceptional adhesion properties due to its reactivity with amino groups found in biological tissues. The hydrogels expanded less in acidic solution because of an intramolecular hydrogen link between carboxyl groups at low pH (pH 2.0). Because of its low swell ratio, this hydrogel could endure an acidic environment. In a pig gastrointestinal hemorrhage model, these hydrogels have been found to successfully minimize delayed bleeding. The hydrogel’s initiator and catalyst, if used, might be cytotoxic. Gel-preparation research in the future will use safer techniques.
NIR-responsive wound dressings may be used in a variety of ways, including releasing gaseous molecules and creating photothermal effects. NIR activation of a hydrogel-NP composite wound dressing from Liu et al. effectively inhibits bacterial infection and releases NO to stimulate angiogenesis [272]. ZIF-8 (zeolitic imidazolate framework-8) changed with PDA (BNN6) and was used as a loading agent. Hydrogels composed of gelatin methacrylate and oxide dextran were used to enclose them (GelMA). This increased the photothermal activity of NPs, which resulted in the death of bacteria when exposed to NIR light. BNN6, on the other hand, may continually create NO when subjected to an 808 nm NIR laser. With the help of our NIR-responsive multifunctional wound dressing, wound healing and angiogenesis were significantly improved. The photothermal effect combined with other NIR-triggered releases may be a solution for adaptive wound dressings that can employ NIR light to its maximum potential.
Chronic wounds, such as diabetic ulcers, supply a significant challenge to wound healing. Diabetics often have abnormalities in wound-healing angiogenesis, which may cause healing to be delayed or cause other issues. Wounds become more prone to infection in hyperglycemia. In the treatment of diabetic wounds, a wound dressing that reacts to changes in blood glucose may be beneficial. An easy-to-implement method for making wound dressings that respond to changes in blood glucose is the glucose-responsive drug release method. When Zhao et al. developed a glucose-triggered drug release mechanism using the Schiff-base and phenylboronate ester reaction, it was the method they employed [273]. For the crosslinking of CS with PVA, we used phenylboronate (CSPBA) and PEG with a benzaldehyde cap, while for the amino group modification we used CSPBA. This two-network configuration proved the hydrogel’s pH and glucose reactivity. If glucose competes with PVA for the synthesis of phenylboronate esters with CSPBA, the crosslinking of this hydrogel may weaken, allowing insulin release from CSPBA hydrogels. This glucose-responsive wound dressing may help control blood glucose levels in addition to speeding up wound healing. A whole new approach to the treatment of wounds and sickness is offered by this study.
Wang et al. developed an injectable polysaccharide-based bandage including exosomes (FEP@exosome) for the treatment of diabetic wounds [274]. FEP@exosome dressing’s pH-responsive release of exosomes, as well as its antibacterial, rapid hemostasis, tissue-adhesive, and UV shielding characteristics, may help angiogenesis. These results imply that the long-term release of bioactive chemicals from an exosome-based dressing and the pace with which the wound heals might work together to speed up healing and minimize scar formation. “Exosome-based wound dressing” treatments based on the release of therapeutic chemicals have also been reported in the literature [275,276,277]. Patients no longer have to travel far to a local clinic for traditional hospital-centered wound management, which is shifting its focus toward the development of multifunctional and self-sustaining wound dressings, which emphasize long-term, continuous monitoring, early diagnosis, and prompt treatment for the benefit of both patients and clinicians.
3.5 Microneedles (MNs)
The use of MNs in medicine is on the rise. They can help with medication delivery [278,279] and are minimally invasive devices that can go through the skin [280]. Due to their capability to deliver transdermally and greater surface area specifically, there is recognition of MNs as encouraging biomaterials for the healing of wounds [281]. There is evidence that their use aids in the repair of scar tissue in dermatology as well [282]. They will not cause any discomfort since they are designed to go just 50–100 µm deep into the skin without touching any nerves. MNs manufactured from carbohydrates offer a number of benefits [283], in contrast to those from more traditional materials like silicon, metal, ceramic, or silica glass. Before shaping the MNs, the medications may be mixed into the mixture, and then the MNs can dissolve into the skin. Yao et al. developed a moldable, biodegradable, and wound-friendly Zn-MOF encapsulated methacrylate hyaluronic acid (MeHA) MN array (MNA) to speed up the healing process [284]. Such an MNA displays good antibacterial activity and great biocompatibility due to the damaging ability against the bacterium capsule and oxidative stress of the zinc ion emitted by the Zn-MOF. Furthermore, the photo crosslinked MeHA in the degradable MNA has better capabilities for releasing the loaded active ingredients slowly and gradually to prevent further wound injury. Also helpful for tissue regeneration is the low molecular weight HA that is produced when MeHA is hydrolyzed. The Zn-MOF-encased degradable MNA has been shown to significantly speed up epithelial regeneration and neovascularization due to these properties [284].
Zeng et al. introduced an MN dressing with synergistic flexibility to promote wound healing in diabetic patients [285]. The anti-diabetic medicine metformin may be placed onto MNs, which can then puncture the skin of diabetic rats to elicit a reaction for managing blood glucose levels. CaO2@polydopamine (CaO2@PDA) is a unique multifunctional nanosystem incorporated into polycaprolactone and gelatin (PCL/Gel) Electrospinned nanofiber films, as MN back patches to reduce inflammation, increase oxygen delivery, and soak up any exudate that could otherwise be left behind. As a bonus, the CaO2@PDA applied to the back patches showed promising antibacterial activity against S. aureus and E. coli. In addition, a high CD31 and low TNF- level was shown by the as-fabricated flexible MN dressings loaded with metformin and CaO2@PDA NPs, resulting in rapid diabetic skin-wound closure.
Mechanical strength and toughness are important for MNs to avoid deformation and breakage during penetration of the stratum corneum [286]. MNs should have their size perfected to maximize medication load with minimal needle size. Furthermore, various MN sizes and uses need distinct production methods. Based on their research, Du et al. concluded that the mechanical characteristics of MNs were similarly affected by the molecular weight of the polymer and the kinds of drug loadings [287]. The MNs were made using HA of two distinct molecular weights (10 and 300 kDa) and two HA MNs (10 and 300 kDa) were loaded with bupivacaine and lidocaine, respectively. Therefore, HA (10 kDa) MN and HA (300 kDa) MN have comparable mechanical strength, exceeding those of their drug-loaded MNs.
A double-layer MN patch for diabetic wound healing was developed, comprising an HA-loaded antibiotic drug and an angiogenic drug-containing CS and SF substrate [288]. The system showed excellent mechanical properties, biocompatibility, antibacterial, and angiogenesis-promoting capabilities. The tip of the MN was loaded with tetracycline hydrochloride for early antibacterial activity, while the substrate containing deferoxamine promoted wound contraction and angiogenesis. In vivo experiments in diabetic rats demonstrated reduced inflammation and accelerated wound healing. DMN@TCH/DFO has the potential for clinical applications in diabetic wound healing.
A core–shell HA MN patch has been developed which contains artificial nanovesicles derived from ferrum-MSCs and the needle tips are encapsulated with PDA NPs. MSC-derived artificial nanovesicles augment the mRNA and protein expression in a more heightened manner in comparison to the exosomes secreted naturally and the yield production is also increased [289,290]. The development of a methacrylate gelatin (GelMA) MNs patch has been reported. It is used for achieving the release of exosomes and tazarotene transdermally and in a controlled manner. In this context, it is to be noted that tazarotene is a drug that has got similarity to retinoic acid and it causes effective promotion of angiogenesis: regeneration of hair follicle as well as collagen in repair of wound. Clinically, the MN patch mentioned above can be used to repair diabetic wounds [291,292].
Researchers have developed a multifunctional MN patch for rapid wound healing through chemo-photodynamic antibacterial effects and sustained release of growth factors [293]. The MN tips contain antibiotics and bioactive small molecule-encapsulated metal–organic frameworks (MOFs) that dissolve and deliver the payloads to the wound upon piercing the skin. Upon light irradiation, the MOF-based NPs convert O2 into 1O2, which synergistically removes pathogenic bacteria from the wound, reducing the required antibiotic amount. The NPs also achieve continuous growth factor release in the wound tissue, promoting epithelial tissue and neovascularization, and accelerating chronic wound healing. The MOF-based MN patches offer a simple, safe, and effective alternative for chronic wound management. Regeneration of epithelium and neovascularization can be accelerated dramatically by a degradable MNA encapsulated with Zn-MOF. Such results are indicative of the fact that when a degradable MNA is used combinedly with MOFs, it is highly effective in the promotion of healing of wounds [281].
Researchers have developed triple-helical recombinant collagen (THRC) dressings that accelerate the healing of acute skin wounds caused by MNs and photodamage [294]. The dressings were found to be biocompatible and non-irritating in cell and animal studies. Histological analysis of animal models showed that the THRC dressings effectively healed damaged dermis by accelerating re-epithelialization and enhancing collagen deposition. The dressings were also found to exhibit rapid epithelialization rates similar to commercial bovine collagen dressings in MN-injured rat defects. The triple-helical structure of collagen was confirmed by circular dichroism measurements. In vitro tests using L929 fibroblasts revealed that THRC dressings were highly biocompatible and promoted the proliferation and adhesion of fibroblasts. The research suggests that THRC dressings could provide an advanced treatment for acute skin wounds and have attractive applications in postoperative care for facial rejuvenation.
Wang et al. developed patch for diabetic wounds to treat them non-invasively [295]. A PLGA-based MN patch was loaded with magnesium hydride (MgH2) macroparticles (MN-MgH2) for transdermal administration and extended release of H2 and Mg ions (Mg2+). MgH2 is a commonplace kind of H2 storage for portable uses. MgH2 has more storage and generation capacity for H2 than Mg powder and is substantially more stable at room temperature. However, MgH2 treatment is a relatively unexplored area. The resulting Mg2+ has the potential to convert macrophages into repairing M2 macrophages and boost angiogenesis to reduce hyperglycemic-induced microvascular damage. Hence, MgH2 may be a useful therapeutic agent for diabetic wound healing. However, using a PLGA-based MN patch allows for transdermal delivery of MgH2 with minimal invasiveness, protects MgH2 from contact with water to prolong its lifetime, and permits sustainable release of MgH2 into the physiological microenvironment, all of which have previously been demonstrated for subcutaneous drug delivery. Moreover, the local acidity was increased by the dissolved PLGA and its breakdown products (lactic acid and glycolic acid), which promoted the release of Mg2+ from Mg(OH)2. As a consequence, MN-MgH2 has been shown to improve wound healing in vitro and in vivo by decreasing ROS production, promoting M2 polarization, increasing cell proliferation and migration, and enhancing angiogenesis and tissue regeneration [295].
One of the leading causes of mortality is excessive bleeding, especially after trauma. Controlling fatalities requires immediate action to stop bleeding after a trauma, such as a gunshot. Hydrogels, which change phase from liquid to a crosslinked solid, stick to tissue and provide hemostatic effects; they have largely replaced sutures and staples in surgical applications. These materials are not ready for immediate use since they need extensive preparation and sophisticated processing, such as photo-/thermoactivation. By nanoengineering of GelMA, a popular cell-friendly biopolymer, researchers have generated hemostatic MNAs with silicate nanoplatelets (SNs) that can greatly enhance the contact area with blood and stick to the wound through an interlocking mechanism [295]. They demonstrated that the clotting time may be reduced by 89% in vitro by doping GelMA MNAs with only 2% w/v of SNs, which is not achievable with hydrogels of the same composition. After applying designed GelMA-SN MNAs to the perforated liver wound, they noticed a 92% decrease in bleeding compared to the untreated damage group. Moreover, the hemostatic MNAs might degrade in vivo in a timeframe (4 weeks) that is suitable for the wound-healing process without eliciting substantial inflammatory reactions. Because of their good bio/hemocompatibility and biodegradability, hemostatic MNAs may be placed on the site post-surgery to prevent internal bleeding without causing difficulties or requiring removal. It is also possible to enhance the hemostatic and wound healing effects of the MNAs by loading them with medicines, growth factors, or other biologics.
Thermal and mineral ingredients present in underwater hot springs offer enough energy for life to arise in these conditions [296,297]. Certain disorders may be treated by soaking in a spa bath heated to between 30 and 42°C and infused with a variety of trace elements and minerals (including iron, metasilicates, and Ag+). Mice suffering from hind limb ischemia may benefit from EC tube formation and angiogenesis when subjected to thermal stimulation [298,299]. Spa baths have been found to speed up wound healing in a nude rat model by increasing the vascular density of granulation tissue [298]. Using photothermal effects and the release of bioactive ions (Fe2+ and
![Figure 6
Illustration representing preparation and application of PCL/AgNPs/BP nanofibers. (a) Fabrication of nanofibers using electrospinning technique. (b) NIR-assisted action of nanofibers to promote wound healing. Reproduced with permission from Zhao et al. [301].](/document/doi/10.1515/ntrev-2023-0129/asset/graphic/j_ntrev-2023-0129_fig_006.jpg)
Illustration representing preparation and application of PCL/AgNPs/BP nanofibers. (a) Fabrication of nanofibers using electrospinning technique. (b) NIR-assisted action of nanofibers to promote wound healing. Reproduced with permission from Zhao et al. [301].
3.6 Role of AI in wound healing
The printing settings in 3D (bio)printing must be optimized for the production of scaffolds with desired architectures and acceptable homogeneities [234]. Conventional methods for improving the settings often need the operators’ earlier knowledge and time-consuming optimization tests. Furthermore, such a conventional approach may be less successful due to the fast expansion in the variety of biomaterial inks and the geometrical complexity of the scaffolds to be constructed. To overcome this difficulty, researchers suggested the use of an AI-assisted image-analysis algorithm in conjunction with a programmable pneumatic extrusion (bio)printer to create a high-throughput printing-condition-screening system (AI-HTPCSS) [287]. In a high-throughput way, the AI-HTPCSS decides the best printing parameters for producing hydrogel designs with consistent morphology and arrangement. Results reveal that scaffolds printed under the greatest circumstances have desirable mechanical qualities, in vitro biological performances, and effectiveness in speeding the healing of diabetic wounds in vivo.
Interest in applying AI to simulate the healing process of wounds has been increasing. One example of such an AI model is described in an article published in “Advances In Wound Care” and used to predict the healing of chronic wounds within 12 weeks [302]. The research concluded that the AI model could accurately predict wound healing. Wound features, rather than patient demographics, were shown to be better indicators of success in the model. AI has shown promise for predicting pressure ulcers in other research as well, and has shown promise as a tool for predicting when wounds will heal, and it has the potential to do the same for evaluating wounds. A report from 2022 claims that AI software that analyzes digital images has great potential to provide reliable data on wound evaluation [303] in contrast to the subjectivity of human observers while evaluating wound size and tissue changes manually. By eliminating observer bias, computer-based image analysis may greatly enhance wound evaluation. The results of wound treatment will improve, and so will the satisfaction of patients.
The utilization of AI-based software and telemedicine has the potential to be a crucial factor in the provision of uninterrupted healthcare services to the populace by means of prognostication and evaluation. The implementation of this technology would facilitate enhanced accessibility for a broader range of patients to avail the services of proficient wound care specialists. The utilization of camera applications, AI, and telemedicine in conjunction with smartphones enables patients and their careers to capture images of their persistent wounds, which are subsequently employed in the evaluation and treatment of the wounds. The images can be expeditiously shared via an application, enabling healthcare professionals to promptly assess the visuals in real-time. The proposed solution would facilitate remote monitoring of chronic wounds by wound care specialists, thereby enabling them to assess the wound status even in the absence of in-person clinic visits. The relevance of telemedicine’s accessibility is evident as certain patients with chronic wounds may be confined to their beds and unable to physically attend appointments with their healthcare provider. In the absence of an application, these individuals would encounter difficulties in obtaining essential healthcare services. Considering these factors, it is evident that the implementation of AI and telemedicine holds immense potential to transform the delivery of wound care to patients. The implementation of this approach has the potential to reduce expenses, enhance efficacy, and promote equitable access to healthcare.
4 Patents of nanoformulations for wound healing
The patents of nanoformulations for wound healing are given in Table 3.
Various important patents employing nanoformulations for wound healing applications
Patent No. | Description | Ref. |
---|---|---|
UUS10058455B2 | The invention relates to wound dressings made up of numerous patterned protuberances that contain medicinal NPs, which are placed in direct contact with the wound to facilitate the healing process. Additionally, the patterned protuberances and galvanic action can provide targeted and enhanced healing capabilities. The invention provides a promising avenue for the development of advanced wound dressings that can improve patient outcomes and reduce healing time | [304] |
AAU2019101531A4 | The invention is a hemostatic and antibacterial material for medical use, consisting of a composite carrier made of iron oxide/kaolin and zinc oxide on the surface of the carrier. Research has found that the composite carrier and zinc oxide have a synergistic effect that enhances their hemostatic and antibacterial properties. The use of this material is expected to lead to faster wound healing | [305] |
CCN102698313B | The invention is a nano-silver antibacterial hydrogel and a method of preparation. It is made up of 3–20% natural polymer or its derivative, 0–20% synthetic polymer, and 0.005–0.2% nano-silver. The hydrogel is prepared by blending the natural polymer or its derivative, the synthetic polymer, a compound containing silver ions, and water, and using a radiation cross-linking method. The hydrogel has good biocompatibility, uniform distribution of nano-silver, slow-release capabilities, and effectively inhibits bacteria such as E. coli | [306] |
CCN102657893B | The invention is a medical nano-fiber sponge material with a porosity of 90–98% and a diameter of 50–1,000 nm. It comprises 0.5–8.5% bioactive glass fine particles, 65–88% gelatin, 0.2–5.0% HA, 0.2–5.0% CS, and water. The resulting sponge has excellent bioactivity, bacteriostatic properties, and mechanical strength, making it suitable for regenerative repair of various skin wounds and skin tissue engineering | [307] |
CCN113274539B | The invention is a self-powered wound patch comprising a PVDF/CNTs fiber patch with a drug-loaded PDA-CS hydrogel coating. The hydrogel consists of dopamine hydrochloride, CS, and either CUR, EGF, or asiaticoside, as well as other components. The patch stops bleeding and absorbs exudates while also providing anti-inflammatory, antibacterial, and moisturizing effects to improve the microenvironment for wound healing | [308] |
CCN114533947B | The invention describes a carbon-based bimetallic nano-cluster with antibacterial activity and the capability of promoting wound healing. The nano-cluster consists of gold and silver embedded in a carbon skeleton using a microwave-assisted method. The nano-cluster has good biocompatibility with the biological system and is low in toxicity | [309] |
WWO2022184149A1 | The method involves preparing a nanoemulsion of perfluorodecalin by adding an emulsifier to a part of a PBS buffer solution and then subjecting it to ultrasonic treatment. This nanoemulsion is further processed through medium-pressure homogenization and high-pressure homogenization to obtain NPs, which are then dispensed into vials and freeze-dried to obtain a freeze-dried powder. The hydrogel dressing is effective in promoting healing due to the oxygen-dissolving capacity and good moisture retention and breathability of perfluorodecalin | [310] |
5 Future directions and conclusion
The process of wound healing may be complicated by both external and internal factors, with microbial contamination of wounds being the prime factor. Development of several topical products has been tried by researchers for achieving a good recovery with the scarring being minimal. Such products make provision for a moist environment and exhibit activity against bacteria. However, certainly, a matter of concern is chronic wound, which is still challenging due to the involvement of microbes that are resistant to various drugs and biofilms of bacteria. The ratio of surface-to-volume in the case of NPs is high and such products can be employed efficiently in wound therapy. Many metal NPs are very less toxic and possess antimicrobicidal activity for which they are found to be the perfect candidates for integrating into the dressing of wounds [311,312,313,314,315]. Furthermore, progress has been made in developing nanocompounds encapsulating growth factors, stem cells, or genes. This will offer advanced treatment modalities in the coming time. However, more studies are the need of the hour to disclose the full potential and the risk factors associated with the application of nanomaterials.
This area of nanotechnology-based platforms, with its impressive effect and many publications, has many potential applications in wound healing. Numerous researchers have developed and proved the wound-healing characteristics of various inorganic and metal NPs (including magnetic, silica silver, nano-graphene oxide scaffolds, copper and gold terbium oxide, cerium and titanium dioxide, etc.). Many organic materials show considerable promise for the treatment of wounds and tissue healing since they play a critical role in tissue adhesion and differentiation, as well as in the distribution of therapeutics, growth factors, stem cells, and others. Other than antimicrobial activity, these nano-delivery systems’ therapeutic and technological advantages include the administration of various drugs (including lipophilic ones) with reduced cytotoxicity and increased stability, improved skin penetration, and controlled release upon topical application, stimulation of fibroblast proliferation, and reduced tissue inflammation. The composition and features (morphology, biodegradability, and other functions) of each nanomaterial must be suited for the unique circumstances of wound healing at various phases to influence the selection process. As a result of this cooperation, new effective and safe wound healing, and tissue repair and regeneration agents with clinical use can be developed using these nanotechnology-based platforms. The molecular processes of wound healing and the function of nanostructural characteristics must yet be thoroughly studied. In addition to the long-term safety of nanomaterials and the scaling up of high-purity NPs legally allowed as standards, consideration should be given to skin pigmentation after prolonged contact and the recurrence of resistant microorganisms.
Developing and approving nanoformulations for wound healing require rigorous examination and adherence to certain standards from a regulatory standpoint. Prior to being made accessible to patients, regulatory bodies play a crucial role in guaranteeing the safety, effectiveness, and quality of these products. Important regulatory considerations are given in the following paragraphs.
Based on their function, nanoformulations for wound healing are either classified as medical devices or as combination goods for regulatory purposes. Guidance on the proper categorization and regulatory process for such goods is provided by relevant regulatory bodies like the Food and Drug Administration in the United States, the European Medicines Agency in Europe, or other national regulatory authorities [316,317]. Manufacturers are responsible for determining the applicable regulatory framework and complying with any applicable approval procedures.
Clinical trials cannot begin without first conducting a thorough preclinical evaluation of a nanoformulation to determine its safety and efficacy. Researching the nanomaterials’ biocompatibility, toxicity, and possible dangers is a necessary step. It is important to show that nanoformulation is beneficial in accelerating wound healing, decreasing infection rates, or delivering some other therapeutic advantage in preclinical studies. The International Organization for Standardization provides excellent recommendations and criteria for study design that should be followed [318].
Consistent production under strict quality control measures is essential for the success of nanoformulations. To assure the quality, safety, and dependability of these items, regulatory bodies mandate that producers use good manufacturing practices. To maintain consistency and long-term stability from batch to batch, manufacturers should disclose information on their production methods, nanomaterial specifications, and characterization and control techniques. Regulatory authorities need thorough risk evaluations for nanoformulations that take into account both the formulation’s intended use and any possible dangers posed by the NPs. The onus is on producers to assess the dangers and provide solutions that are supported by evidence. This may include taking measures to lessen NP release, stop their systemic absorption, and deal with any possible adverse consequences.
To ensure the safety and efficacy of nanoformulations for wound healing in actual clinical settings, regulatory bodies need post-market monitoring after product authorization. Post-market monitoring mechanisms should be set up by manufacturers to track side effects, patient outcomes, and new safety issues. These data are essential for evaluating products over time and making any necessary adjustments to packaging or regulations.
In conclusion, nanoformulations have emerged as a promising approach for wound healing due to their unique properties and ability to deliver therapeutic agents to the wound site. They offer several advantages over traditional wound-healing approaches, such as enhanced drug delivery, controlled release, and improved stability of therapeutic agents. Various types of nanoformulations, such as NPs, liposomes, and hydrogels, have been developed and tested in preclinical and clinical studies for wound-healing applications. Despite the promising results obtained from these studies, there are still several challenges that need to be addressed, including toxicity, biocompatibility, and scalability of production. Further research and development are required to overcome these challenges and translate nanoformulations for wound healing into clinical practice. Nonetheless, nanoformulations hold great potential to revolutionize wound healing and improve patient outcomes in the future.
Acknowledgments
The authors would like to extend their sincere appreciation to their respective institutions for providing a suitable ambiance for developing this manuscript. Y.K.M. is highly indebted and sincerely thanks SERB-DST, Government of India, for providing support to his Nano-biotechnology and Translational Knowledge Laboratory through research Grant No. SRG/2022/000641.
-
Funding information: This study is supported via funding from Prince Sattam bin Abdulaziz University project number (PSAU/2023/R/1445).
-
Author contributions: H. C., Y. K. M.: conceptualization, design, material collection, writting – original draft; S.M.: image preparation; T.K.M., I.S., S.K. A.: software, visualization, manuscript editing; S.P.M., A.A.R., H.A., A.A., M.A., H.R.A., B.A., M. S. A., S.C., K. D., I.B.S.: writing and editing of the manuscript. All authors have accepted responsibility for the entire content of this manuscript and approved its submission.
-
Conflict of interest: The authors state no conflict of interest.
References
[1] Chopra H, Kumar S, Singh I. Bioadhesive hydrogels and their applications. Bioadhesives Drug Deliv. 2020;1:147–70. 10.1002/9781119640240.CH6.Search in Google Scholar
[2] Chopra H, Kumar S, Singh I. Biopolymer-based scaffolds for tissue engineering applications. Curr Drug Targets. 2020;22:282–95. 10.2174/1389450121999201102140408.Search in Google Scholar PubMed
[3] Chopra H, Singh I, Kumar S, Bhattacharya T, Rahman MH, Akter R, et al. Comprehensive review on hydrogels. Curr Drug Deliv. 2022;19(6):658–75. 10.2174/1567201818666210601155558.Search in Google Scholar PubMed
[4] Chopra H, Gandhi S, Gautam RK, Kamal MA. Bacterial nanocellulose based wound dressings: current and future prospects. Curr Pharm Des. 2022;28(7):570–80. 10.2174/1381612827666211021162828.Search in Google Scholar PubMed
[5] Sajid M. Nanomaterials: types, properties, recent advances, and toxicity concerns. Curr Opin Environ Sci Heal. 2022;25. 10.1016/j.coesh.2021.100319.Search in Google Scholar
[6] Li T, Lu XM, Zhang MR, Hu K, Li Z. Peptide-based nanomaterials: Self-assembly, properties and applications. Bioact Mater. 2022;11:268–82. 10.1016/j.bioactmat.2021.09.029.Search in Google Scholar PubMed PubMed Central
[7] Su S, Kang PM. Systemic review of biodegradable nanomaterials in nanomedicine. Nanomaterials. 2020;10(4):656. 10.3390/nano10040656.Search in Google Scholar PubMed PubMed Central
[8] Yang J, Hou B, Wang J, Tian B, Bi J, Wang N, et al. Nanomaterials for the removal of heavy metals from wastewater. Nanomaterials. 2019;9(3):424. 10.3390/nano9030424.Search in Google Scholar PubMed PubMed Central
[9] Baig N, Kammakakam I, Falath W, Kammakakam I. Nanomaterials: A review of synthesis methods, properties, recent progress, and challenges. Mater Adv. 2021;2:1821–71. 10.1039/d0ma00807a.Search in Google Scholar
[10] Fytianos G, Rahdar A, Kyzas GZ. Nanomaterials in cosmetics: Recent updates. Nanomaterials. 2020;10:1–16. 10.3390/nano10050979.Search in Google Scholar PubMed PubMed Central
[11] Nanotechnologies: 1. What are nanomaterials? https://ec.europa.eu/health/scientific_committees/opinions_layman/nanomaterials/en/l-2/1.htm#:∼:text=Nanomaterials%20are%20usually%20considered%20to,%2C%20tubes%2C%20rods%20or%20fibres.Search in Google Scholar
[12] Bhadauria SS, Malviya R. Advancement in nanoformulations for the management of diabetic wound healing. Endocr Metab Immune Disord – Drug Targets. 2022;22:911–26. 10.2174/1871530322666220304214106.Search in Google Scholar PubMed
[13] Chakrabarti S, Chattopadhyay P, Islam J, Ray S, Raju PS, Mazumder B. Aspects of nanomaterials in wound healing. Curr Drug Deliv. 2018;16:26–41. 10.2174/1567201815666180918110134.Search in Google Scholar PubMed
[14] Nqakala ZB, Sibuyi NRS, Fadaka AO, Meyer M, Onani MO, Madiehe AM. Advances in nanotechnology towards development of silver nanoparticle-based wound-healing agents. Int J Mol Sci. 2021;22:11272. 10.3390/IJMS222011272.Search in Google Scholar PubMed PubMed Central
[15] Ingle AP, Paralikar P, Pandit R, Anasane N, Gupta I, Rai M, et al. Nanoformulations for wound infections. Nanotechnol Appl Pharm Technol. 2017;1:223–46. 10.1007/978-3-319-70299-5_9/COVER.Search in Google Scholar
[16] Salehi B, Rodrigues CF, Peron G, Dall’Acqua S, Sharifi-Rad J, Azmi L, et al. Curcumin nanoformulations for antimicrobial and wound healing purposes. Phyther Res. 2021;35:2487–99. 10.1002/PTR.6976.Search in Google Scholar PubMed
[17] Singh Bakshi I, Chopra H, Sharma M, Kaushik D, Pahwa R. Herbal bioactives for wound healing application. Herb Bioact Drug Deliv Syst. 2022;1:259–82. 10.1016/b978-0-12-824385-5.00003-0.Search in Google Scholar
[18] Chopra H, Kumar S, Safi SZ, Singh I, Emran TB. Wound dressings: Recent updates. Int J Surg. 2022;104:106793. 10.1016/j.ijsu.2022.106793.Search in Google Scholar PubMed
[19] Landén NX, Li D, Ståhle M. Transition from inflammation to proliferation: a critical step during wound healing. Cell Mol Life Sci. 2016;73(20):3861–85. 10.1007/s00018-016-2268-0.Search in Google Scholar PubMed PubMed Central
[20] Strbo N, Yin N, Stojadinovic O. Innate and adaptive immune responses in wound epithelialization. Adv Wound Care. 2014;3(7):492–501. 10.1089/wound.2012.0435.Search in Google Scholar PubMed PubMed Central
[21] Takeuchi O, Akira S. Pattern recognition receptors and inflammation. Cell. 2010;140(6):805–20. 10.1016/j.cell.2010.01.022.Search in Google Scholar PubMed
[22] Sinno H, Prakash S. Complements and the wound healing cascade: an updated review. Plast Surg Int. 2013;2013:146764. 10.1155/2013/146764.Search in Google Scholar PubMed PubMed Central
[23] Vestweber D. How leukocytes cross the vascular endothelium. Nat Rev Immunol. 2015;15(11):692–704. 10.1038/nri3908.Search in Google Scholar PubMed
[24] Eming SA, Martin P, Tomic-Canic M. Wound repair and regeneration: Mechanisms, signaling, and translation. Sci Transl Med. 2014;6(265):265sr6. 10.1126/scitranslmed.3009337.Search in Google Scholar PubMed PubMed Central
[25] Werner S, Grose R. Regulation of wound healing by growth factors and cytokines. Physiol Rev. 2003;83(3):835–70. 10.1152/physrev.2003.83.3.835.Search in Google Scholar PubMed
[26] Nosbaum A, Prevel N, Truong H-A, Mehta P, Ettinger M, Scharschmidt TC, et al. Cutting edge: regulatory T cells facilitate cutaneous wound healing. J Immunol. 2016;196(5):2010–4. 10.4049/jimmunol.1502139.Search in Google Scholar PubMed PubMed Central
[27] Sindrilaru A, Scharffetter-Kochanek K. Disclosure of the culprits: macrophages – versatile regulators of wound healing. Adv Wound Care. 2013;2(7):357–68. 10.1089/wound.2012.0407.Search in Google Scholar PubMed PubMed Central
[28] Lucas T, Waisman A, Ranjan R, Roes J, Krieg T, Müller W, et al. Differential roles of macrophages in diverse phases of skin repair. J Immunol. 2010;184(7):3964–77. 10.4049/jimmunol.0903356.Search in Google Scholar PubMed
[29] Galli SJ, Borregaard N, Wynn TA. Phenotypic and functional plasticity of cells of innate immunity: Macrophages, mast cells and neutrophils. Nat Immunol. 2011;12(11):1035–44. 10.1038/ni.2109.Search in Google Scholar PubMed PubMed Central
[30] Brancato SK, Albina JE. Wound macrophages as key regulators of repair. Am J Pathol. 2011;178(1):19–25. 10.1016/j.ajpath.2010.08.003.Search in Google Scholar PubMed PubMed Central
[31] Fadok VA, Bratton DL, Konowal A, Freed PW, Westcott JY, Henson PM. Macrophages that have ingested apoptotic cells in vitro inhibit proinflammatory cytokine production through autocrine/paracrine mechanisms involving TGF-β, PGE2, and PAF. J Clin Invest. 1998;101(4):890–8. 10.1172/JCI1112.Search in Google Scholar PubMed PubMed Central
[32] Sindrilaru A, Peters T, Wieschalka S, Baican C, Baican A, Peter H, et al. An unrestrained proinflammatory M1 macrophage population induced by iron impairs wound healing in humans and mice. J Clin Invest. 2011;121(3):985–97. 10.1172/JCI44490.Search in Google Scholar PubMed PubMed Central
[33] Mirza RE, Fang MM, Novak ML, Urao N, Sui A, Ennis WJ, et al. Macrophage PPARγ and impaired wound healing in type 2 diabetes. J Pathol. 2015;236(4):433–44. 10.1002/path.4548.Search in Google Scholar PubMed PubMed Central
[34] Khanna S, Biswas S, Shang Y, Collard E, Azad A, Kauh C, et al. Macrophage dysfunction impairs resolution of inflammation in the wounds of diabetic mice. PLoS One. 2010;5(3):e9539. 10.1371/journal.pone.0009539.Search in Google Scholar PubMed PubMed Central
[35] Martin P, D’Souza D, Martin J, Grose R, Cooper L, Maki R, et al. Wound healing in the PU.1 null mouse – Tissue repair is not dependent on inflammatory cells. Curr Biol. 2003;13(13):1122–8. 10.1016/S0960-9822(03)00396-8.Search in Google Scholar
[36] Volk SW, Bohling MW. Comparative wound healing – Are the small animal veterinarian’s clinical patients an improved translational model for human wound healing research? Wound Repair Regen. 2013;21(3):372–81. 10.1111/wrr.12049.Search in Google Scholar PubMed
[37] Knobloch K, Vogt PM. The reconstructive sequence in the 21st century. A reconstructive clockwork. Chirurg. 2010;81(5):441–6. 10.1007/s00104-010-1917-3.Search in Google Scholar PubMed
[38] Rittié L. Cellular mechanisms of skin repair in humans and other mammals. J Cell Commun Signal. 2016;10(2):103–20. 10.1007/s12079-016-0330-1.Search in Google Scholar PubMed PubMed Central
[39] Krawczyk WS. A pattern of epidermal cell migration during wound healing. J Cell Biol. 1971;49(2):247–63. 10.1083/jcb.49.2.247.Search in Google Scholar PubMed PubMed Central
[40] Gov NS. Collective cell migration patterns: Follow the leader. Proc Natl Acad Sci U S A. 2007;104(41):15970–1. 10.1073/pnas.0708037104.Search in Google Scholar PubMed PubMed Central
[41] Omelchenko T, Vasiliev JM, Gelfand IM, Feder HH, Bonder EM. Rho-dependent formation of epithelial “leader” cells during wound healing. Proc Natl Acad Sci U S A. 2003;100(19):10788–93. 10.1073/pnas.1834401100.Search in Google Scholar PubMed PubMed Central
[42] Farooqui R, Fenteany G. Multiple rows of cells behind an epithelial wound edge extend cryptic lamellipodia to collectively drive cell-sheet movement. J Cell Sci. 2005;118:51–63. 10.1242/jcs.01577.Search in Google Scholar PubMed
[43] Stone RC, Pastar I, Ojeh N, Chen V, Liu S, Garzon KI, et al. Epithelial-mesenchymal transition in tissue repair and fibrosis. Cell Tissue Res. 2016;365(3):495–506. 10.1007/s00441-016-2464-0.Search in Google Scholar PubMed PubMed Central
[44] Jacinto A, Martinez-Arias A, Martin P. Mechanisms of epithelial fusion and repair. Nat Cell Biol. 2001;3(5):E117–23. 10.1038/35074643.Search in Google Scholar PubMed
[45] Santoro MM, Gaudino G. Cellular and molecular facets of keratinocyte reepithelization during wound healing. Exp Cell Res. 2005;304(1):274–86. 10.1016/j.yexcr.2004.10.033.Search in Google Scholar PubMed
[46] Pastar I, Stojadinovic O, Yin NC, Ramirez H, Nusbaum AG, Sawaya A, et al. Epithelialization in wound healing: a comprehensive review. Adv Wound Care. 2014;3(7):445–64. 10.1089/wound.2013.0473.Search in Google Scholar PubMed PubMed Central
[47] Donati G, Watt FM. Stem cell heterogeneity and plasticity in epithelia. Cell Stem Cell. 2015;16(5):465–76. 10.1016/j.stem.2015.04.014.Search in Google Scholar PubMed
[48] Blanpain C, Fuchs E. Plasticity of epithelial stem cells in tissue regeneration. Science. 2014;344(6189):1242281. 10.1126/science.1242281.Search in Google Scholar PubMed PubMed Central
[49] Stojadinovic O, Pastar I, Nusbaum AG, Vukelic S, Krzyzanowska A, Tomic-Canic M. Deregulation of epidermal stem cell niche contributes to pathogenesis of nonhealing venous ulcers. Wound Repair Regen. 2014;22(2):220–27. 10.1111/wrr.12142.Search in Google Scholar PubMed PubMed Central
[50] Yang M, Li Q, Sheng L, Li H, Weng R, Zan T. Bone marrow-derived mesenchymal stem cells transplantation accelerates tissue expansion by promoting skin regeneration during expansion. Ann Surg. 2011;253(1):202–9. 10.1097/SLA.0b013e3181f9ba1ah.Search in Google Scholar PubMed
[51] Tenenhaus M, Rennekampff HO. Current concepts in tissue engineering: Skin and wound. Plast Reconstr Surg. 2016;138(3 Suppl):42S–50S. 10.1097/PRS.0000000000002685.Search in Google Scholar PubMed
[52] Chen D, Hao H, Fu X, Han W. Insight into reepithelialization: How do mesenchymal stem cells perform? Stem Cells Int. 2016;2016:6120173. 10.1155/2016/6120173.Search in Google Scholar PubMed PubMed Central
[53] McCarty SM, Percival SL. Proteases and delayed wound healing. Adv Wound Care. 2013;2(8):438–47. 10.1089/wound.2012.0370.Search in Google Scholar PubMed PubMed Central
[54] Krzyszczyk P, Schloss R, Palmer A, Berthiaume F. The role of macrophages in acute and chronic wound healing and interventions to promote pro-wound healing phenotypes. Front Physiol. 2018;9:419. 10.3389/fphys.2018.00419.Search in Google Scholar PubMed PubMed Central
[55] Martin P, Nunan R. Cellular and molecular mechanisms of repair in acute and chronic wound healing. Br J Dermatol. 2015;173(2):370–8. 10.1111/bjd.13954.Search in Google Scholar PubMed PubMed Central
[56] Mihai MM, Preda M, Lungu I, Gestal MC, Popa MI, Holban AM. Nanocoatings for chronic wound repair – modulation of microbial colonization and biofilm formation. Int J Mol Sci. 2018;19(4):1179. 10.3390/ijms19041179.Search in Google Scholar PubMed PubMed Central
[57] Lebrun I, Marques-Porto R, Pereira A, Pereira A, Perpetuo E. Bacterial toxins: an overview on bacterial proteases and their action as virulence factors. Mini-Reviews Med Chem. 2009;9(7):820–8. 10.2174/138955709788452603.Search in Google Scholar PubMed
[58] McCarty SM, Cochrane CA, Clegg PD, Percival SL. The role of endogenous and exogenous enzymes in chronic wounds: A focus on the implications of aberrant levels of both host and bacterial proteases in wound healing. Wound Repair Regen. 2012;20(2):125–36. 10.1111/j.1524-475X.2012.00763.x.Search in Google Scholar PubMed
[59] Vollmer P, Walev I, Rose-John S, Bhakdi S. Novel pathogenic mechanism of microbial metalloproteinases: Liberation of membrane-anchored molecules in biologically active form exemplified by studies with the human interleukin-6 receptor. Infect Immun. 1996;64(9):3646–51. 10.1128/iai.64.9.3646-3651.1996.Search in Google Scholar PubMed PubMed Central
[60] Lantz MS. Are bacterial proteases important virulence factors? J Periodontal Res. 1997;32(1):126–32. 10.1111/j.1600-0765.1997.tb01393.x.Search in Google Scholar PubMed
[61] Supuran CT, Scozzafava A, Mastrolorenzo A. Bacterial proteases: Current therapeutic use and future prospects for the development of new antibiotics. Expert Opin Ther Pat. 2001;11(2):221–59. 10.1517/13543776.11.2.221.Search in Google Scholar
[62] Supuran CT, Scozzafava A, Clare BW. Bacterial protease inhibitors. Med Res Rev. 2002;22(4):329–72. 10.1002/med.10007.Search in Google Scholar PubMed
[63] Pires-Bouças PD, Izumi E, Furlaneto-Maia L, LeonardoSturion L, Suzart S. Effects of environmental and nutritional factors on gelatinolytic activity by enterococcus faecalis strains isolated from clinical sources. African J Microbiol Res. 2010;4(10):969–76.Search in Google Scholar
[64] Smoot LM, Smoot JC, Graham MR, Somerville GA, Sturdevant DE, Lux Migliaccio CA, et al. Global differential gene expression in response to growth temperature alteration in group A Streptococcus. Proc Natl Acad Sci U S A. 2001;98(18):10416–21. 10.1073/pnas.191267598.Search in Google Scholar PubMed PubMed Central
[65] Podbielski A, Leonard BAB. The group a streptococcal dipeptide permease (Dpp) is involved in the uptake of essential amino acids and affects the expression of cysteine protease. Mol Microbiol. 1998;28(6):1323–34. 10.1046/j.1365-2958.1998.00898.x.Search in Google Scholar PubMed
[66] Podbielski A, Pohl B, Woischnik M, Körner C, Schmidt KH, Rozdzlnski E, et al. Molecular characterization of group A streptococcal (GAS) oligopeptide permease (Opp) and its effect on cysteine protease production. Mol Microbiol. 1996;21(5):1087–99. 10.1046/j.1365-2958.1996.661421.x.Search in Google Scholar PubMed
[67] Lyon WR, Madden JC, Levin JC, Stein JL, Caparon MG. Mutation of luxS affects growth and virulence factor expression in Streptococcus pyogenes. Mol Microbiol. 2001;42(1):145–57. 10.1046/j.1365-2958.2001.02616.x.Search in Google Scholar PubMed
[68] Chaussee MS, Phillips ER, Ferretti JJ. Temporal production of streptococcal erythrogenic toxin B (streptococcal cysteine proteinase) in response to nutrient depletion. Infect Immun. 1997;65(5):1956–9. 10.1128/iai.65.5.1956-1959.1997.Search in Google Scholar PubMed PubMed Central
[69] Wretlind B, Pavlovskis OR. Pseudomonas aeruginosa elastase and its role in pseudomonas infections. Rev Infect Dis. 1983;5(Suppl 5):S998–1004. 10.1093/clinids/5.supplement_5.s998.Search in Google Scholar PubMed
[70] Kon Y, Tsukada H, Hasegawa T, Igarashi K, Wada K, Suzuki E, et al. The role of Pseudomonas aeruginosa elastase as a potent inflammatory factor in a rat air pouch inflammation model. FEMS Immunol Med Microbiol. 1999;25(3):313–21. 10.1016/S0928-8244(99)00101-7.Search in Google Scholar
[71] Peters JE, Park SJ, Darzins A, Freck LC, Saulnier JM, Wallach JM, et al. Further studies on Pseudomonas aeruginosa LasA: analysis of specificity. Mol Microbiol. 1992;6(9):1155–62. 10.1111/j.1365-2958.1992.tb01554.x.Search in Google Scholar PubMed
[72] Sorsa T, Ingman T, Suomalainen K, Haapasalo M, Konttinen YT, Lindy O, et al. Identification of proteases from periodontopathogenic bacteria as activators of latent human neutrophil and fibroblast-type interstitial collagenases. Infect Immun. 1992;60(11):4491–5. 10.1128/iai.60.11.4491-4495.1992.Search in Google Scholar PubMed PubMed Central
[73] Romanelli M, Schipani E, Piaggesi A, Barachini P. Evaluation of surface pH on venous leg ulcers under Allevyn dressings. Int Congr Symp Ser – R Soc Med. 1996.Search in Google Scholar
[74] Wilson IAI, Henry M, Quill RD, Byrne PJ. The pH of varicose ulcer surfaces and its relationship to healing. Vasa – J Vasc Dis. 1979;8(4):339–42.Search in Google Scholar
[75] Leveen HH, Falk G, Borek B, Diaz C, Lynfield Y, Wynkoop BJ, et al. Chemical acidification of wounds. An adjuvant to healing and the unfavorable action of alkalinity and ammonia. Ann Surg. 1973;178(6):745–53. 10.1097/00000658-197312000-00011.Search in Google Scholar PubMed PubMed Central
[76] Kaufman T, Eichenlaub EH, Angel MF, Levin M, Futrell JW. Topical acidification promotes healing of experimental deep partial thickness skin burns: a randomized double-blind preliminary study. Burns. 1985;12(2):84–90. 10.1016/0305-4179(85)90032-4.Search in Google Scholar PubMed
[77] Frykberg RG. Topical wound oxygen therapy in the treatment of chronic diabetic foot ulcers. Medicina. 2021;57(9):917. 10.3390/MEDICINA57090917.Search in Google Scholar PubMed PubMed Central
[78] Greener B, Hughes AA, Bannister NP, Douglass J. Proteases and pH in chronic wounds. J Wound Care. 2005;14(2):59–61. 10.12968/jowc.2005.14.2.26739.Search in Google Scholar PubMed
[79] Kaiser P, Wächter J, Windbergs M. Therapy of infected wounds: overcoming clinical challenges by advanced drug delivery systems. Drug Deliv Transl Res. 2021;11(4):1545–67. 10.1007/s13346-021-00932-7.Search in Google Scholar PubMed PubMed Central
[80] Niska K, Zielinska E, Radomski MW, Inkielewicz-Stepniak I. Metal nanoparticles in dermatology and cosmetology: Interactions with human skin cells. Chem Biol Interact. 2018;295:38–51. 10.1016/j.cbi.2017.06.018.Search in Google Scholar PubMed
[81] Sood A, Granick MS, Tomaselli NL. Wound dressings and comparative effectiveness data. Adv Wound Care. 2014;3(8):511–529. 10.1089/wound.2012.0401.Search in Google Scholar PubMed PubMed Central
[82] Gao Y, Song N, Liu W, Dong A, Wang YJ, Yang YW. Construction of antibacterial N-halamine polymer nanomaterials capable of bacterial membrane disruption for efficient anti-infective wound therapy. Macromol Biosci. 2019;19(4):e1800453. 10.1002/mabi.201800453.Search in Google Scholar PubMed
[83] Dai M, Zhang J, Liu N, Zhang XH. Precise engineering of lignin incorporated dextran/glycol nanomaterials for wound dressings for the care of anorectal surgery. J Polym Environ. 2022;30(1):1–11. 10.1007/s10924-021-02175-6.Search in Google Scholar
[84] Lu Y, Jia C, Gong C, Wang H, Xiao Q, Guo J, et al. A hydrogel system containing molybdenum-based nanomaterials for wound healing. Nano Res. 2023;16:5368–75. 10.1007/s12274-022-5255-9.Search in Google Scholar
[85] Liu T, Lu Y, Zhan R, Qian W, Luo G. Nanomaterials and nanomaterials-based drug delivery to promote cutaneous wound healing. Adv Drug Deliv Rev. 2023;193:114670. 10.1016/j.addr.2022.114670.Search in Google Scholar PubMed
[86] Mihai MM, Dima MB, Dima B, Holban AM. Nanomaterials for wound healing and infection control. Materials (Basel). 2019;12(13):2176. 10.3390/ma12132176.Search in Google Scholar PubMed PubMed Central
[87] El-Naggar ME, Shalaby ES, Abd-Al-Aleem AH, Youssef MA. Nanomaterials and nanofibers as wound dressing mats: an overview of the fundamentals, properties and applications. Egypt J Chem. 2021;64(12):7447–73. 10.21608/EJCHEM.2021.91351.4345.Search in Google Scholar
[88] Wang C, Wang J, Zeng L, Qiao Z, Liu X, Liu H, et al. Fabrication of electrospun polymer nanofibers with diverse morphologies. Molecules. 2019;24(5):834. 10.3390/molecules24050834.Search in Google Scholar PubMed PubMed Central
[89] Aidana Y, Wang Y, Li J, Chang S, Wang K, Yu D-G. Fast dissolution electrospun medicated nanofibers for effective delivery of poorly water-soluble drug. Curr Drug Deliv. 2021;19(4):422–35. 10.2174/1567201818666210215110359.Search in Google Scholar PubMed
[90] Nauman S, Lubineau G, Alharbi HF. Post processing strategies for the enhancement of mechanical properties of enms (Electrospun nanofibrous membranes): A review. Membranes (Basel). 2021;11(1):39. 10.3390/membranes11010039.Search in Google Scholar PubMed PubMed Central
[91] Ma H, Burger C, Chu B, Hsiao BS. Electrospun nanofibers for environmental protection. Handb Fibrous Mater. 2020;11(10):770. 10.1002/9783527342587.ch28.Search in Google Scholar
[92] Zhao K, Kang SX, Yang YY, Yu DG. Electrospun functional nanofiber membrane for antibiotic removal in water: Review. Polymers (Basel). 2021;13(2):226. 10.3390/polym13020226.Search in Google Scholar PubMed PubMed Central
[93] Wang M, Yu D-G, Li X, Williams GR. The development and bio-applications of multifluid electrospinning. Mater Highlights. 2020;1(1–2):1–13. 10.2991/mathi.k.200521.001.Search in Google Scholar
[94] Wang Y, Tian L, Zhu T, Mei J, Chen Z, Yu DG. Electrospun aspirin/eudragit/lipid hybrid nanofibers for colon-targeted delivery using an energy-saving process. Chem Res Chinese Univ. 2021;37(3):443–9. 10.1007/s40242-021-1006-9.Search in Google Scholar PubMed PubMed Central
[95] Li Y, Zhu J, Cheng H, Li G, Cho H, Jiang M, et al. Developments of advanced electrospinning techniques: a critical review. Adv Mater Technol. 2021;6(11):2100410. 10.1002/admt.202100410.Search in Google Scholar
[96] Xin R, Ma H, Venkateswaran S, Hsiao BS. Electrospun nanofibrous adsorption membranes for wastewater treatment: mechanical strength enhancement. Chem Res Chinese Univ. 2021;37:355–65. 10.1007/s40242-021-1095-5.Search in Google Scholar
[97] Samadian H, Zamiri S, Ehterami A, Farzamfar S, Vaez A, Khastar H, et al. Electrospun cellulose acetate/gelatin nanofibrous wound dressing containing berberine for diabetic foot ulcer healing: in vitro and in vivo studies. Sci Rep. 2020;10:8312. 10.1038/s41598-020-65268-7.Search in Google Scholar PubMed PubMed Central
[98] Chen J, Huang Z, Zhang H, Zhang Z, Wang D, Xia D, et al. Three-dimensional layered nanofiber sponge with in situ grown silver-metal organic framework for enhancing wound healing. Chem Eng J. 2022;443:4041711. 10.2139/ssrn.4041711.Search in Google Scholar
[99] Shadman-Manesh V, Gholipour-Kanani A, Najmoddin N, Rabbani S. Preclinical evaluation of the polycaprolactone-polyethylene glycol electrospun nanofibers containing egg-yolk oil for acceleration of full thickness burns healing. Sci Reports. 2023;13:1–12. 10.1038/s41598-023-28065-6.Search in Google Scholar PubMed PubMed Central
[100] Bouhajeb R, Abreu AC, Selmi S, Gerke C, Bellalah A, Alvear-Jiménez A, et al. Implantable electrospun nanofibers with wound-healing capabilities in the reduction of pressure ulcers. ACS Appl Polym Mater. 2023;5(1):429–40. 10.1021/ACSAPM.2C01585/SUPPL_FILE/AP2C01585_SI_001.PDF.Search in Google Scholar
[101] Anaya Mancipe JM, Boldrini Pereira LC, de Miranda Borchio PG, Dias ML, da Silva Moreira Thiré RM. Novel polycaprolactone (PCL)-type I collagen core-shell electrospun nanofibers for wound healing applications. J Biomed Mater Res Part B Appl Biomater. 2023;111:366–81. 10.1002/JBM.B.35156.Search in Google Scholar PubMed
[102] Koohzad F, Asoodeh A. Cross-linked electrospun pH-sensitive nanofibers adsorbed with Temporin-Ra for promoting wound healing. ACS Appl Mater Interfaces. 2023;15(12):15172–84. 10.1021/ACSAMI.2C23268.Search in Google Scholar PubMed
[103] Sarhan WA, Azzazy HME, El-Sherbiny IM. Honey/chitosan nanofiber wound dressing enriched with allium sativum and cleome droserifolia: enhanced antimicrobial and wound healing activity. ACS Appl Mater Interfaces. 2016;8:6379–90. 10.1021/acsami.6b00739.Search in Google Scholar PubMed
[104] Ahmed R, Tariq M, Ali I, Asghar R, Noorunnisa Khanam P, Augustine R, et al. Novel electrospun chitosan/polyvinyl alcohol/zinc oxide nanofibrous mats with antibacterial and antioxidant properties for diabetic wound healing. Int J Biol Macromol. 2018;120(Pt A):385–93. 10.1016/j.ijbiomac.2018.08.057.Search in Google Scholar PubMed
[105] Malafatti JOD, Bernardo MP, Moreira FKV, Ciol H, Inada NM, Mattoso LHC, et al. Electrospun poly(lactic acid) nanofibers loaded with silver sulfadiazine/[Mg–Al]-layered double hydroxide as an antimicrobial wound dressing. Polym Adv Technol. 2020;31(6):1377–87. 10.1002/pat.4867.Search in Google Scholar
[106] Liakos I, Rizzello L, Hajiali H, Brunetti V, Carzino R, Pompa PP, et al. Fibrous wound dressings encapsulating essential oils as natural antimicrobial agents. J Mater Chem B. 2015;3:1583–9. 10.1039/c4tb01974a.Search in Google Scholar PubMed
[107] Topman G, Lin FH, Gefen A. The natural medications for wound healing – Curcumin, Aloe-Vera and Ginger – do not induce a significant effect on the migration kinematics of cultured fibroblasts. J Biomech. 2013;46(1):170–4. 10.1016/j.jbiomech.2012.09.015.Search in Google Scholar PubMed
[108] Karami Z, Rezaeian I, Zahedi P, Abdollahi M. Preparation and performance evaluations of electrospun poly(ε-caprolactone), poly(lactic acid), and their hybrid (50/50) nanofibrous mats containing thymol as an herbal drug for effective wound healing. J Appl Polym Sci. 2013;129:756–66. 10.1002/app.38683.Search in Google Scholar
[109] Zahiri M, Khanmohammadi M, Goodarzi A, Ababzadeh S, Sagharjoghi Farahani M, Mohandesnezhad S, et al. Encapsulation of curcumin loaded chitosan nanoparticle within poly (ε-caprolactone) and gelatin fiber mat for wound healing and layered dermal reconstitution. Int J Biol Macromol. 2020;153:1241–50. 10.1016/j.ijbiomac.2019.10.255.Search in Google Scholar PubMed
[110] Rath G, Hussain T, Chauhan G, Garg T, Goyal AK. Collagen nanofiber containing silver nanoparticles for improved wound-healing applications. J Drug Target. 2016;24(6):520–9. 10.3109/1061186X.2015.1095922.Search in Google Scholar PubMed
[111] El-Aassar MR, Ibrahim OM, Fouda MMG, El-Beheri NG, Agwa MM. Wound healing of nanofiber comprising Polygalacturonic/Hyaluronic acid embedded silver nanoparticles: In-vitro and in-vivo studies. Carbohydr Polym. 2020;238:116175. 10.1016/j.carbpol.2020.116175.Search in Google Scholar PubMed
[112] Li CW, Wang Q, Li J, Hu M, Shi SJ, Li ZW, et al. Silver nanoparticles/chitosan oligosaccharide/poly(vinyl alcohol) nanofiber promotes wound healing by activating TGFβ1/Smad signaling pathway. Int J Nanomed. 2016;11:373–86. 10.2147/IJN.S91975.Search in Google Scholar PubMed PubMed Central
[113] Song DW, Kim SH, Kim HH, Lee KH, Ki CS, Park YH. Multi-biofunction of antimicrobial peptide-immobilized silk fibroin nanofiber membrane: Implications for wound healing. Acta Biomater. 2016;39:146–55. 10.1016/j.actbio.2016.05.008.Search in Google Scholar PubMed
[114] Parin FN, Terzioğlu P, Sicak Y, Yildirim K, Öztürk M. Pine honey–loaded electrospun poly (vinyl alcohol)/gelatin nanofibers with antioxidant properties. J Text Inst. 2021;112(8):1–8. 10.1080/00405000.2020.1773199.Search in Google Scholar
[115] Ahmadi S, Hivechi A, Bahrami SH, Milan PB, Ashraf SS. Cinnamon extract loaded electrospun chitosan/gelatin membrane with antibacterial activity. Int J Biol Macromol. 2021;173:580–90. 10.1016/j.ijbiomac.2021.01.156.Search in Google Scholar PubMed
[116] Salehi M, Niyakan M, Ehterami A, Haghi-Daredeh S, Nazarnezhad S, Abbaszadeh-Goudarzi G, et al. Porous electrospun poly(ε-caprolactone)/gelatin nanofibrous mat containing cinnamon for wound healing application: in vitro and in vivo study. Biomed Eng Lett. 2020;10(1):149–61. 10.1007/s13534-019-00138-4.Search in Google Scholar PubMed PubMed Central
[117] Yao CH, Yeh JY, Chen YS, Li MH, Huang CH. Wound-healing effect of electrospun gelatin nanofibres containing Centella asiatica extract in a rat model. J Tissue Eng Regen Med. 2017;11(3):905–915. 10.1002/term.1992.Search in Google Scholar PubMed
[118] Hadisi Z, Nourmohammadi J, Nassiri SM. The antibacterial and anti-inflammatory investigation of Lawsonia Inermis-gelatin-starch nano-fibrous dressing in burn wound. Int J Biol Macromol. 2018;107(Pt B):2008–19. 10.1016/j.ijbiomac.2017.10.061.Search in Google Scholar PubMed
[119] Adeli-Sardou M, Yaghoobi MM, Torkzadeh-Mahani M, Dodel M. Controlled release of lawsone from polycaprolactone/gelatin electrospun nano fibers for skin tissue regeneration. Int J Biol Macromol. 2019;124:478–491. 10.1016/j.ijbiomac.2018.11.237.Search in Google Scholar PubMed
[120] Farahani H, Barati A, Arjomandzadegan M, Vatankhah E. Nanofibrous cellulose acetate/gelatin wound dressing endowed with antibacterial and healing efficacy using nanoemulsion of Zataria multiflora. Int J Biol Macromol. 2020;162:762–773. 10.1016/j.ijbiomac.2020.06.175.Search in Google Scholar PubMed
[121] Mahmood K, Kamilah H, Alias AK, Ariffin F, Mohammadi Nafchi A. Functionalization of electrospun fish gelatin mats with bioactive agents: Comparative effect on morphology, thermo-mechanical, antioxidant, antimicrobial properties, and bread shelf stability. Food Sci Nutr. 2022;10(2):584–596. 10.1002/fsn3.2676.Search in Google Scholar PubMed PubMed Central
[122] Akşit NN, Gürdap S, İşoğlu SD, İşoğlu İA. Preparation of antibacterial electrospun poly(D, L-lactide-co-glycolide)/gelatin blend membranes containing Hypericum capitatum var. capitatum. Int J Polym Mater Polym Biomater. 2021;70(11):797–809. 10.1080/00914037.2020.1765354.Search in Google Scholar
[123] Unalan I, Endlein SJ, Slavik B, Buettner A, Goldmann WH, Detsch R, et al. Evaluation of electrospun poly(ε-caprolactone)/gelatin nanofiber mats containing clove essential oil for antibacterial wound dressing. Pharmaceutics. 2019;11(11):570. 10.3390/pharmaceutics11110570.Search in Google Scholar PubMed PubMed Central
[124] Ahlawat J, Kumar V, Gopinath P. Carica papaya loaded poly (vinyl alcohol)-gelatin nanofibrous scaffold for potential application in wound dressing. Mater Sci Eng C. 2019;103:109834. 10.1016/j.msec.2019.109834.Search in Google Scholar PubMed
[125] El Fawal G, Hong H, Mo X, Wang H. Fabrication of scaffold based on gelatin and polycaprolactone (PCL) for wound dressing application. J Drug Deliv Sci Technol. 2021;63:102501. 10.1016/j.jddst.2021.102501.Search in Google Scholar
[126] Pezeshki-Modaress M, Mirzadeh H, Zandi M, Rajabi-Zeleti S, Sodeifi N, Aghdami N, et al. Gelatin/chondroitin sulfate nanofibrous scaffolds for stimulation of wound healing: In-vitro and in-vivo study. J Biomed Mater Res – Part A. 2017;105(7):2020–34. 10.1002/jbm.a.35890.Search in Google Scholar PubMed
[127] Shi R, Geng H, Gong M, Ye J, Wu C, Hu X, et al. Long-acting and broad-spectrum antimicrobial electrospun poly (ε-caprolactone)/gelatin micro/nanofibers for wound dressing. J Colloid Interface Sci. 2018;509:275–84. 10.1016/j.jcis.2017.08.092.Search in Google Scholar PubMed
[128] Shan YH, Peng LH, Liu X, Chen X, Xiong J, Gao JQ. Silk fibroin/gelatin electrospun nanofibrous dressing functionalized with astragaloside IV induces healing and anti-scar effects on burn wound. Int J Pharm. 2015;479(2):291–301. 10.1016/j.ijpharm.2014.12.067.Search in Google Scholar PubMed
[129] Zhang D, Li L, Shan Y, Xiong J, Hu Z, Zhang Y, et al. In vivo study of silk fibroin/gelatin electrospun nanofiber dressing loaded with astragaloside IV on the effect of promoting wound healing and relieving scar. J Drug Deliv Sci Technol. 2019;52:272–81. 10.1016/j.jddst.2019.04.021.Search in Google Scholar
[130] Baek S, Park H, Kim M, Lee D. Preparation of PCL/(+)-catechin/gelatin film for wound healing using air-jet spinning. Appl Surf Sci. 2020;509:145033. 10.1016/j.apsusc.2019.145033.Search in Google Scholar
[131] Basar AO, Castro S, Torres-Giner S, Lagaron JM, Turkoglu Sasmazel H. Novel poly(ε-caprolactone)/gelatin wound dressings prepared by emulsion electrospinning with controlled release capacity of Ketoprofen anti-inflammatory drug. Mater Sci Eng C. 2017;81:459–68. 10.1016/j.msec.2017.08.025.Search in Google Scholar PubMed
[132] Farzamfar S, Naseri-Nosar M, Samadian H, Mahakizadeh S, Tajerian R, Rahmati M, et al. Taurine-loaded poly (ε-caprolactone)/gelatin electrospun mat as a potential wound dressing material: In vitro and in vivo evaluation. J Bioact Compat Polym. 2018;33(3):282–94. 10.1177/0883911517737103.Search in Google Scholar
[133] Xia L, Lu L, Liang Y, Cheng B. Fabrication of centrifugally spun prepared poly(lactic acid)/gelatin/ciprofloxacin nanofibers for antimicrobial wound dressing. RSC Adv. 2019;9:35328–35. 10.1039/c9ra07826f.Search in Google Scholar PubMed PubMed Central
[134] Foroutan Koudehi M, Zibaseresht R. Synthesis of molecularly imprinted polymer nanoparticles containing gentamicin drug as wound dressing based polyvinyl alcohol/gelatin nanofiber. Mater Technol. 2020;35(1):21–30. 10.1080/10667857.2019.1649888.Search in Google Scholar
[135] Yang J, Liu CL, Ding YN, Sun TC, Bai XH, Cao ZK, et al. Synergistic antibacterial polyacrylonitrile/gelatin nanofibers coated with metal-organic frameworks for accelerating wound repair. Int J Biol Macromol. 2021;189:698–704. 10.1016/j.ijbiomac.2021.08.175.Search in Google Scholar PubMed
[136] Bakhsheshi-Rad HR, Ismail AF, Aziz M, Akbari M, Hadisi Z, Daroonparvar M, et al. Antibacterial activity and in vivo wound healing evaluation of polycaprolactone-gelatin methacryloyl-cephalexin electrospun nanofibrous. Mater Lett. 2019;256:126618. 10.1016/j.matlet.2019.126618.Search in Google Scholar
[137] Razzaq A, Khan ZU, Saeed A, Shah KA, Khan NU, Menaa B, et al. Development of cephradine-loaded gelatin/polyvinyl alcohol electrospun nanofibers for effective diabetic wound healing: In-vitro and in-vivo assessments. Pharmaceutics. 2021;13(3):349. 10.3390/pharmaceutics13030349.Search in Google Scholar PubMed PubMed Central
[138] Safdari M, Shakiba E, Kiaie SH, Fattahi A. Preparation and characterization of Ceftazidime loaded electrospun silk fibroin/gelatin mat for wound dressing. Fibers Polym. 2016;17:744 -50. 10.1007/s12221-016-5822-3.Search in Google Scholar
[139] Yu M, Huang J, Zhu T, Lu J, Liu J, Li X, et al. Liraglutide-loaded PLGA/gelatin electrospun nanofibrous mats promote angiogenesis to accelerate diabetic wound healing: Via the modulation of miR-29b-3p. Biomater Sci. 2020;8:4225–38. 10.1039/d0bm00442a.Search in Google Scholar PubMed
[140] Heo DN, Yang DH, Lee JB, Bae MS, Kim JH, Moon SH, et al. Burn-wound healing effect of gelatin/polyurethane nanofiber scaffold containing silver-sulfadiazine. J Biomed Nanotechnol. 2013;9(3):511–5. 10.1166/jbn.2013.1509.Search in Google Scholar PubMed
[141] Felciya SJG, Devi MV, Ramanathan G, Poornima V, Sivagnanam UT. Fabrication of polyhydroxy butyric acid–Gelatin blended nanofibrous matrix integrated with silver sulfadiazine as an alternate wound dressing for treating burns. Mater Lett. 2021;282:128541. 10.1016/j.matlet.2020.128541.Search in Google Scholar
[142] Li H, Wang M, Williams GR, Wu J, Sun X, Lv Y, et al. Electrospun gelatin nanofibers loaded with vitamins A and e as antibacterial wound dressing materials. RSC Adv. 2016;6:50267–77. 10.1039/c6ra05092a.Search in Google Scholar
[143] Xu F, Wang H, Zhang J, Jiang L, Zhang W, Hu Y. A facile design of EGF conjugated PLA/gelatin electrospun nanofibers for nursing care of in vivo wound healing applications. J Ind Text. 2022;51(1_suppl):420S–440S. 10.1177/1528083720976348.Search in Google Scholar
[144] Joshi A, Xu Z, Ikegami Y, Yoshida K, Sakai Y, Joshi A, et al. Exploiting synergistic effect of externally loaded bFGF and endogenous growth factors for accelerated wound healing using heparin functionalized PCL/gelatin co-spun nanofibrous patches. Chem Eng J. 2021;404:126518. 10.1016/j.cej.2020.126518.Search in Google Scholar
[145] Kandhasamy S, Perumal S, Madhan B, Umamaheswari N, Banday JA, Perumal PT, et al. Synthesis and Fabrication of Collagen-Coated Ostholamide Electrospun Nanofiber Scaffold for Wound Healing. ACS Appl Mater Interfaces. 2017;9(10):8556–68. 10.1021/acsami.6b16488.Search in Google Scholar PubMed
[146] Bagheri M, Validi M, Gholipour A, Makvandi P, Sharifi E. Chitosan nanofiber biocomposites for potential wound healing applications: Antioxidant activity with synergic antibacterial effect. Bioeng Transl Med. 2022;7(1):e10254. 10.1002/btm2.10254.Search in Google Scholar PubMed PubMed Central
[147] Ahn S, Chantre CO, Gannon AR, Lind JU, Campbell PH, Grevesse T, et al. Soy protein/cellulose nanofiber scaffolds mimicking skin extracellular matrix for enhanced wound healing. Adv Healthc Mater. 2018; 7(9):e1701175. 10.1002/adhm.201701175.Search in Google Scholar PubMed PubMed Central
[148] Jung SM, Min SK, Lee HC, Kwon YS, Jung MH, Shin HS. Spirulina-PCL nanofiber wound dressing to improve cutaneous wound healing by enhancing antioxidative mechanism. J Nanomater. 2016;2016:6135727. 10.1155/2016/6135727.Search in Google Scholar
[149] Guo X, Liu Y, Bera H, Zhang H, Chen Y, Cun D, et al. α-Lactalbumin-based nanofiber dressings improve burn wound healing and reduce scarring. ACS Appl Mater Interfaces. 2020;12(41):45702–13. 10.1021/acsami.0c05175.Search in Google Scholar PubMed
[150] Robson MC. Wound infection: A failure of wound healing caused by an imbalance of bacteria. Surg Clin North Am. 1997;77(3):637–50. 10.1016/S0039-6109(05)70572-7.Search in Google Scholar PubMed
[151] Boroumand Z, Golmakani N, Boroumand S. Clinical trials on silver nanoparticles for wound healing (review). Nanomed J. 2018;5(4):186–91.Search in Google Scholar
[152] Gravante G, Montone A. A retrospective analysis of ambulatory burn patients: Focus on wound dressings and healing times. Ann R Coll Surg Engl. 2010;92(2):118–23. 10.1308/003588410X12518836439001.Search in Google Scholar PubMed PubMed Central
[153] Thirumurugan G, Satya Veni V, Ramachandran S, Seshagiri Rao JVLN, Dhanaraju MD. Superior wound healing effect of topically delivered silver nanoparticle formulation using eco-friendly potato plant pathogenic fungus: Synthesis and characterization. J Biomed Nanotechnol. 2011;7(5):659–66. 10.1166/jbn.2011.1336.Search in Google Scholar PubMed
[154] Vijayakumar V, Samal SK, Mohanty S, Nayak SK. Recent advancements in biopolymer and metal nanoparticle-based materials in diabetic wound healing management. Int J Biol Macromol. 2019;122:137–48. 10.1016/j.ijbiomac.2018.10.120.Search in Google Scholar PubMed
[155] Lu MM, Bai J, Shao D, Qiu J, Li M, Zheng X, et al. Antibacterial and biodegradable tissue nano-adhesives for rapid wound closure. Int J Nanomed. 2018;13:5849–63. 10.2147/IJN.S177109.Search in Google Scholar PubMed PubMed Central
[156] Lu S, Gao W, Gu HY. Construction, application and biosafety of silver nanocrystalline chitosan wound dressing. Burns. 2008;34(5):623–8. 10.1016/j.burns.2007.08.020.Search in Google Scholar PubMed
[157] Ahamed MIN, Sankar S, Kashif PM, Basha SKH, Sastry TP. Evaluation of biomaterial containing regenerated cellulose and chitosan incorporated with silver nanoparticles. Int J Biol Macromol. 2015;72:680–6. 10.1016/j.ijbiomac.2014.08.055.Search in Google Scholar PubMed
[158] Ghosh Auddy R, Abdullah MF, Das S, Roy P, Datta S, Mukherjee A. New guar biopolymer silver nanocomposites for wound healing applications. Biomed Res Int. 2013;2013:912458. 10.1155/2013/912458.Search in Google Scholar PubMed PubMed Central
[159] Bhuvaneswari T, Thiyagarajan M, Geetha N, Venkatachalam P. Bioactive compound loaded stable silver nanoparticle synthesis from microwave irradiated aqueous extracellular leaf extracts of Naringi crenulata and its wound healing activity in experimental rat model. Acta Trop. 2014;135:55–61. 10.1016/j.actatropica.2014.03.009.Search in Google Scholar PubMed
[160] Lin YH, Hsu WS, Chung WY, Ko TH, Lin JH. Evaluation of various silver-containing dressing on infected excision wound healing study. J Mater Sci Mater Med. 2014;25(5):1375–86. 10.1007/s10856-014-5152-1.Search in Google Scholar PubMed
[161] Neibert K, Gopishetty V, Grigoryev A, Tokarev I, Al-Hajaj N, Vorstenbosch J, et al. Wound-healing with mechanically robust and biodegradable hydrogel fibers loaded with silver nanoparticles. Adv Healthc Mater. 2012;1(5):621–30. 10.1002/adhm.201200075.Search in Google Scholar PubMed
[162] Tian J, Wong KKY, Ho CM, Lok CN, Yu WY, Che CM, et al. Topical delivery of silver nanoparticles promotes wound healing. ChemMedChem. 2007;2:129–36. 10.1002/cmdc.200600171.Search in Google Scholar PubMed
[163] Im AR, Kim JY, Kim HS, Cho S, Park Y, Kim YS. Wound healing and antibacterial activities of chondroitin sulfate- and acharan sulfate-reduced silver nanoparticles. Nanotechnology. 2013;24(9):395102. 10.1088/0957-4484/24/39/395102.Search in Google Scholar PubMed
[164] Depan D, Misra RDK. Hybrid nanoscale architecture of wound dressing with super hydrophilic, antimicrobial, and ultralow fouling attributes. J Biomed Nanotechnol. 2015;11(2):306–18. 10.1166/jbn.2015.1908.Search in Google Scholar PubMed
[165] Dai X, Guo Q, Zhao Y, Zhang P, Zhang T, Zhang X, et al. Functional silver nanoparticle as a benign antimicrobial agent that eradicates antibiotic-resistant bacteria and promotes wound healing. ACS Appl Mater Interfaces. 2016;8:39. 10.1021/acsami.6b09267.Search in Google Scholar PubMed
[166] Gunasekaran T, Nigusse T, Dhanaraju MD. Silver nanoparticles as real topical bullets for wound healing. J Am Coll Clin Wound Spec. 2011;3(4):82–96. 10.1016/j.jcws.2012.05.001.Search in Google Scholar PubMed PubMed Central
[167] Kumar SSD, Rajendran NK, Houreld NN, Abrahamse H. Recent advances on silver nanoparticle and biopolymer-based biomaterials for wound healing applications. Int J Biol Macromol. 2018;115:165–75. 10.1016/j.ijbiomac.2018.04.003.Search in Google Scholar PubMed
[168] Cameron SJ, Hosseinian F, Willmore WG. A current overview of the biological and cellular effects of nanosilver. Int J Mol Sci. 2018;19(7):2030. 10.3390/ijms19072030.Search in Google Scholar PubMed PubMed Central
[169] Gong CP, Li SC, Wang RY. Development of biosynthesized silver nanoparticles based formulation for treating wounds during nursing care in hospitals. J Photochem Photobiol B Biol. 2018;183:137–41. 10.1016/j.jphotobiol.2018.04.030.Search in Google Scholar PubMed
[170] Adibhesami M, Ahmadi M, Farshid AA, Sarrafzadeh-Rezaei F, Dalir-Naghadeh B. Effects of silver nanoparticles on Staphylococcus aureus contaminated open wounds healing in mice: An experimental study. Vet Res Forum an Int Q J. 2017;8(1):23–8.Search in Google Scholar
[171] Singla R, Soni S, Patial V, Kulurkar PM, Kumari A, Mahesh S, et al. In vivo diabetic wound healing potential of nanobiocomposites containing bamboo cellulose nanocrystals impregnated with silver nanoparticles. Int J Biol Macromol. 2017;105(pt1):45–55. 10.1016/j.ijbiomac.2017.06.109.Search in Google Scholar PubMed
[172] Nam G, Rangasamy S, Purushothaman B, Song JM. The application of bactericidal silver nanoparticles in wound treatment. Nanomater Nanotechnol. 2015;5:23. 10.5772/60918.Search in Google Scholar
[173] Liu X, Lee PY, Ho CM, Lui VCH, Chen Y, Che CM, et al. Silver nanoparticles mediate differential responses in keratinocytes and fibroblasts during skin wound healing. ChemMedChem. 2010;5(3):468–75. 10.1002/cmdc.200900502.Search in Google Scholar PubMed
[174] Cadinoiu AN, Rata DM, Daraba OM, Ichim DL, Popescu I, Solcan C, et al. Silver Nanoparticles Biocomposite Films with Antimicrobial Activity: In vitro and In vivo Tests. Int J Mol Sci. 2022;23(18):10671. 10.3390/IJMS231810671.Search in Google Scholar
[175] Barroso A, Mestre H, Ascenso A, Simões S, Reis C. Nanomaterials in wound healing: From material sciences to wound healing applications. Nano Sel. 2020;1(5):443–460. 10.1002/nano.202000055.Search in Google Scholar
[176] Volkova N, Yukhta M, Pavlovich O, Goltsev A. Application of cryopreserved fibroblast culture with Au nanoparticles to treat burns. Nanoscale Res Lett. 2016;11:22. 10.1186/s11671-016-1242-y.Search in Google Scholar PubMed PubMed Central
[177] 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. 10.1002/lsm.20206.Search in Google Scholar PubMed
[178] Matei AM, Caruntu C, Tampa M, Georgescu SR, Matei C, Constantin MM, et al. Applications of nanosized-lipid-based drug delivery systems in wound care. Appl Sci. 2021;11(11):4915. 10.3390/app11114915.Search in Google Scholar
[179] Fatima F, Aleemuddin M, Ahmed MM, Anwer MK, Aldawsari MF, Soliman GA, et al. Design and evaluation of solid lipid nanoparticles loaded topical gels: repurpose of fluoxetine in diabetic wound healing. Gels. 2023;9(1):21. 10.3390/gels9010021.Search in Google Scholar PubMed PubMed Central
[180] Bonferoni MC, Rossi S, Cornaglia AI, Mannucci B, Grisoli P, Vigani B. Essential oil-loaded lipid nanoparticles for wound healing. Int J Nanomed. 2018;13(13):175–86.Search in Google Scholar
[181] de Souza ML, dos Santos WM, de Sousa ALMD, de Albuquerque Wanderley Sales V, Nóbrega FP, de Oliveira MVG, et al. Lipid nanoparticles as a skin wound healing drug delivery system: discoveries and advances. Curr Pharm Des. 2020;26(36):4536–50. 10.2174/1381612826666200417144530.Search in Google Scholar PubMed
[182] Hosny KM, Naveen NR, Kurakula M, Sindi AM, Sabei FY, Al Fatease A, et al. Design and development of neomycin sulfate gel loaded with solid lipid nanoparticles for buccal mucosal wound healing. Gels. 2022;8(6):385. 10.3390/gels8060385.Search in Google Scholar PubMed PubMed Central
[183] Sandhu SK, Kumar S, Raut J, Singh M, Kaur S, Sharma G, et al. Systematic development and characterization of novel, high drug-loaded, photostable, curcumin solid lipid nanoparticle hydrogel for wound healing. Antioxidants. 2021;10(5):725. 10.3390/antiox10050725.Search in Google Scholar PubMed PubMed Central
[184] Saporito F, Sandri G, Bonferoni MC, Rossi S, Boselli C, Cornaglia AI, et al. Essential oil-loaded lipid nanoparticles for wound healing. Int J Nanomedicine. 2018;13:175–86. 10.2147/IJN.S152529.Search in Google Scholar PubMed PubMed Central
[185] Ataide JA, Zanchetta B, Santos ÉM, Fava ALM, Alves TFR, Cefali LC, et al. Nanotechnology-based dressings for wound management. Pharmaceuticals. 2022;15(10):1286. 10.3390/ph15101286.Search in Google Scholar PubMed PubMed Central
[186] 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. 10.1089/tea.2007.0143.Search in Google Scholar PubMed
[187] Webber MJ, Tongers J, Renault MA, Roncalli JG, Losordo DW, Stupp SI. Development of bioactive peptide amphiphiles for therapeutic cell delivery. Acta Biomater. 2010;6(1):3–11. 10.1016/j.actbio.2009.07.031.Search in Google Scholar PubMed PubMed Central
[188] Webber MJ, Kessler JA, Stupp SI. Emerging peptide nanomedicine to regenerate tissues and organs. J Intern Med. 2010;267(1):71–88. 10.1111/j.1365-2796.2009.02184.x.Search in Google Scholar PubMed PubMed Central
[189] Jayawarna V, Smith A, Gough JE, Ulijn RV. Three-dimensional cell culture of chondrocytes on modified di-phenylalanine scaffolds. Biochem Soc Trans. 2007;35(pt3):535–7. 10.1042/BST0350535.Search in Google Scholar PubMed
[190] Smith AM, Williams RJ, Tang C, Coppo P, Collins RF, Turner ML, et al. Fmoc-diphenylalanine self assembles to a hydrogel via a novel architecture based on π–π interlocked β-sheets. Adv Mater. 2008;20(1):37–41. 10.1002/ADMA.200701221.Search in Google Scholar
[191] You C, Li Q, Wang X, Wu P, Ho JK, Jin R, et al. Silver nanoparticle loaded collagen/chitosan scaffolds promote wound healing via regulating fibroblast migration and macrophage activation. Sci Rep. 2017;7:10489. 10.1038/S41598-017-10481-0.Search in Google Scholar
[192] Zhang S, Ou Q, Xin P, Yuan Q, Wang Y, Wu J. Polydopamine/puerarin nanoparticle-incorporated hybrid hydrogels for enhanced wound healing. Biomater Sci. 2019;7:4230–6. 10.1039/c9bm00991d.Search in Google Scholar PubMed
[193] Augustine R, Hasan A, Patan NK, Dalvi YB, Varghese R, Antony A, et al. Cerium oxide nanoparticle incorporated electrospun poly(3-hydroxybutyrate-co-3-hydroxyvalerate) membranes for diabetic wound healing applications. ACS Biomater Sci Eng. 2020;6(1):58–70. 10.1021/acsbiomaterials.8b01352.Search in Google Scholar PubMed
[194] Laghezza Masci V, Taddei AR, Courant T, Tezgel O, Navarro F, Giorgi F, et al. Characterization of collagen/lipid nanoparticle–curcumin cryostructurates for wound healing applications. Macromol Biosci. 2019;19:1800446. 10.1002/mabi.201800446.Search in Google Scholar PubMed
[195] Masood N, Ahmed R, Tariq M, Ahmed Z, Masoud MS, Ali I, et al. Silver nanoparticle impregnated chitosan-PEG hydrogel enhances wound healing in diabetes induced rabbits. Int J Pharm. 2019;559:23–36. 10.1016/j.ijpharm.2019.01.019.Search in Google Scholar PubMed
[196] Cao L, Shao G, Ren F, Yang M, Nie Y, Peng Q, et al. Cerium oxide nanoparticle-loaded polyvinyl alcohol nanogels delivery for wound healing care systems on surgery. Drug Deliv. 2021;28(1):390–9. 10.1080/10717544.2020.1858998.Search in Google Scholar PubMed PubMed Central
[197] Lee YH, Hong YL, Wu TL. Novel silver and nanoparticle-encapsulated growth factor co-loaded chitosan composite hydrogel with sustained antimicrobility and promoted biological properties for diabetic wound healing. Mater Sci Eng C. 2021;118:111385. 10.1016/j.msec.2020.111385.Search in Google Scholar PubMed
[198] Bhubhanil S, Talodthaisong C, Khongkow M, Namdee K, Wongchitrat P, Yingmema W, et al. Enhanced wound healing properties of guar gum/curcumin-stabilized silver nanoparticle hydrogels. Sci Rep. 2021;11(1):21836. 10.1038/s41598-021-01262-x.Search in Google Scholar PubMed PubMed Central
[199] Pérez-Rafael S, Ivanova K, Stefanov I, Puiggalí J, del Valle LJ, Todorova K, et al. Nanoparticle-driven self-assembling injectable hydrogels provide a multi-factorial approach for chronic wound treatment. Acta Biomater. 2021;134:131–43. 10.1016/j.actbio.2021.07.020.Search in Google Scholar PubMed
[200] Kırbaş OK, Bozkurt BT, Taşlı PN, Hayal TB, Özkan İ, Bülbül B, et al. Effective scarless wound healing mediated by erbium borate nanoparticles. Biol Trace Elem Res. 2021;199:3262–71. 10.1007/s12011-020-02458-4.Search in Google Scholar PubMed
[201] Al-Omari A, Cameron DW, Lee C, Corrales-Medina VF. Oral antibiotic therapy for the treatment of infective endocarditis: A systematic review. BMC Infect Dis. 2014;14:140. 10.1186/1471-2334-14-140.Search in Google Scholar PubMed PubMed Central
[202] Chopra H, Islam MA, Sharun K, Emran TB, Al-Tawfiq JA, Dhama K. Recent advances in the treatment of biofilms induced surgical site infections. Int J Surg. 2023;109(1):65–7.10.1097/JS9.0000000000000036Search in Google Scholar PubMed PubMed Central
[203] Bordi C, de Bentzmann S. Hacking into bacterial biofilms: a new therapeutic challenge. Ann Intensive Care. 2011;1:19. 10.1186/2110-5820-1-19.Search in Google Scholar PubMed PubMed Central
[204] Zhang L, Pornpattananangkul D, Hu C-M, Huang C-M. Development of nanoparticles for antimicrobial drug delivery. Curr Med Chem. 2010;17(6):585–94. 10.2174/092986710790416290.Search in Google Scholar PubMed
[205] Allen TM, Cullis PR. Liposomal drug delivery systems: From concept to clinical applications. Adv Drug Deliv Rev. 2013;65(1):36–48. 10.1016/j.addr.2012.09.037.Search in Google Scholar PubMed
[206] Malam Y, Loizidou M, Seifalian AM. Liposomes and nanoparticles: nanosized vehicles for drug delivery in cancer. Trends Pharmacol Sci. 2009;30(11):592–9. 10.1016/j.tips.2009.08.004.Search in Google Scholar PubMed
[207] Nacucchio MC, Gatto Bellora MJ, Sordelli DO, D’Aquino M. Enhanced liposome-mediated activity of piperacillin against staphylococci. Antimicrob Agents Chemother. 1985;27(1):137–9. 10.1128/AAC.27.1.137.Search in Google Scholar PubMed PubMed Central
[208] Mugabe C, Azghani AO, Omri A. Liposome-mediated gentamicin delivery: Development and activity against resistant strains of Pseudomonas aeruginosa isolated from cystic fibrosis patients. J Antimicrob Chemother. 2005;55(2):269–71. 10.1093/jac/dkh518.Search in Google Scholar PubMed
[209] Bala I, Hariharan S, Kumar MNVR. PLGA nanoparticles in drug delivery: The state of the art. Crit Rev Ther Drug Carrier Syst. 2004;21(5):387–422. 10.1615/CritRevTherDrugCarrierSyst.v21.i5.20.Search in Google Scholar
[210] Esmaeili F, Hosseini-Nasr M, Rad-Malekshahi M, Samadi N, Atyabi F, Dinarvand R. Preparation and antibacterial activity evaluation of rifampicin-loaded poly lactide-co-glycolide nanoparticles. Nanomed Nanotechnol Biol Med. 2007;3(2):161–7. 10.1016/j.nano.2007.03.003.Search in Google Scholar PubMed
[211] Cheow WS, Chang MW, Hadinoto K. Antibacterial efficacy of inhalable levofloxacin-loaded polymeric nanoparticles against E. coli biofilm cells: The effect of antibiotic release profile. Pharm Res. 2010;27:1597–609. 10.1007/s11095-010-0142-6.Search in Google Scholar PubMed
[212] Hadinoto K, Sundaresan A, Cheow WS. Lipid-polymer hybrid nanoparticles as a new generation therapeutic delivery platform: A review. Eur J Pharm Biopharm. 2013;85(3):427–43. 10.1016/j.ejpb.2013.07.002.Search in Google Scholar PubMed
[213] Cheow WS, Hadinoto K. Factors affecting drug encapsulation and stability of lipid-polymer hybrid nanoparticles. Colloids Surfaces B Biointerfaces. 2011;85(2):214–20. 10.1016/j.colsurfb.2011.02.033.Search in Google Scholar PubMed
[214] Krishnamurthy S, Vaiyapuri R, Zhang L, Chan JM. Lipid-coated polymeric nanoparticles for cancer drug delivery. Biomater Sci. 2015;3:923–36. 10.1039/c4bm00427b.Search in Google Scholar PubMed
[215] Palmer BC, Phelan-Dickenson SJ, Delouise LA. Multi-walled carbon nanotube oxidation dependent keratinocyte cytotoxicity and skin inflammation. Part Fibre Toxicol. 2019;16:3. 10.1186/s12989-018-0285-x.Search in Google Scholar PubMed PubMed Central
[216] Lai X, Wang M, Zhu Y, Feng X, Liang H, Wu J, et al. ZnO NPs delay the recovery of psoriasis-like skin lesions through promoting nuclear translocation of p-NFκB p65 and cysteine deficiency in keratinocytes. J Hazard Mater. 2021;410:124566. 10.1016/j.jhazmat.2020.124566.Search in Google Scholar PubMed
[217] Xiao H, Zhang H. Skin inflammation and psoriasis may be linked to exposure of ultrafine carbon particles. J Environ Sci (China). 2020;96:206–8. 10.1016/j.jes.2020.06.028.Search in Google Scholar PubMed
[218] Wang M, Lai X, Shao L, Li L. Evaluation of immunoresponses and cytotoxicity from skin exposure to metallic nanoparticles. Int J Nanomed. 2018;13:4445–59. 10.2147/IJN.S170745.Search in Google Scholar PubMed PubMed Central
[219] Hashempour S, Ghanbarzadeh S, Maibach HI, Ghorbani M, Hamishehkar H. Skin toxicity of topically applied nanoparticles. Ther Deliv. 2019;10(6):383–96. 10.4155/tde-2018-0060.Search in Google Scholar PubMed
[220] Pan Y, Paschoalino WJ, Szuchmacher Blum A, Mauzeroll J. Recent advances in bio-templated metallic nanomaterial synthesis and electrocatalytic applications. ChemSusChem. 2021;14(3):758–91. 10.1002/cssc.202002532.Search in Google Scholar PubMed
[221] Vijayakumar G, Kim HJ, Rangarajulu SK. In vitro antibacterial and wound healing activities evoked by silver nanoparticles synthesized through probiotic bacteria. Antibiotics. 2023;12(1):141. 10.3390/ANTIBIOTICS12010141/S1.Search in Google Scholar
[222] Blinov AV, Kachanov MD, Gvozdenko AA, Nagdalian AA, Blinova AA, Rekhman ZA, et al. Synthesis and characterization of zinc oxide nanoparticles stabilized with biopolymers for application in wound-healing mixed gels. Gels. 2023;9(1):57. 10.3390/GELS9010057.Search in Google Scholar
[223] Balaure PC, Holban AM, Grumezescu AM, Mogoşanu GD, Bălşeanu TA, Stan MS, et al. In vitro and in vivo studies of novel fabricated bioactive dressings based on collagen and zinc oxide 3D scaffolds. Int J Pharm. 2019;557:199–207. 10.1016/j.ijpharm.2018.12.063.Search in Google Scholar PubMed
[224] 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;164:214–21. 10.1016/j.carbpol.2017.01.061.Search in Google Scholar PubMed
[225] Khan MI, Behera SK, Paul P, Das B, Suar M, Jayabalan R, 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. 2019;208(5):609–29. 10.1007/s00430-018-0564-z.Search in Google Scholar PubMed
[226] Mirjalili F, Mahmoodi M. Controlled release of protein from gelatin/chitosan hydrogel containing platelet-rich fibrin encapsulated in chitosan nanoparticles for accelerated wound healing in an animal model. Int J Biol Macromol. 2023;225:588–604. 10.1016/j.ijbiomac.2022.11.117.Search in Google Scholar PubMed
[227] Liu H, Ding M, Wang H, Chen Y, Liu Y, Wei L, et al. Silver nanoparticles modified hFGF2-linking camelina oil bodies accelerate infected wound healing. Colloids Surf B Biointerfaces. 2023;222:113089. 10.1016/j.colsurfb.2022.113089.Search in Google Scholar PubMed
[228] Liu T, Feng Z, Li Z, Lin Z, Chen L, Li B, et al. Carboxymethyl chitosan/sodium alginate hydrogels with polydopamine coatings as promising dressings for eliminating biofilm and multidrug-resistant bacteria induced wound healing. Int J Biol Macromol. 2023;225:923–37. 10.1016/j.ijbiomac.2022.11.156.Search in Google Scholar PubMed
[229] Zhao X, Sun X, Yildirimer L, Lang Q, Lin ZY, Zheng R, et al. Cell infiltrative hydrogel fibrous scaffolds for accelerated wound healing. Acta Biomater. 2017;49:66–77. 10.1016/j.actbio.2016.11.017.Search in Google Scholar PubMed PubMed Central
[230] Wang S, Wei Y, Wang Y, Cheng Y. Cyclodextrin regulated natural polysaccharide hydrogels for biomedical applications-a review. Carbohydr Polym. 2023;313:120760. 10.1016/J.CARBPOL.2023.120760.Search in Google Scholar
[231] Sari MHM, Cobre A, de F, Pontarolo R, Ferreira LM. Status and future scope of soft nanoparticles-based hydrogel in wound healing. Pharmaceutics. 2023;15(3):874. 10.3390/pharmaceutics15030874.Search in Google Scholar PubMed PubMed Central
[232] Chopra H, Bibi S, Kumar S, Khan MS, Kumar P, Singh I. Preparation and evaluation of chitosan/PVA based hydrogel films loaded with honey for wound healing application. Gels. 2022;8(2):111. 10.3390/gels8020111.Search in Google Scholar PubMed PubMed Central
[233] Chopra H, Kumar S, Singh I. Strategies and therapies for wound healing: a review. Curr Drug Targets. 2021;23(1):87–98. 10.2174/1389450122666210415101218.Search in Google Scholar PubMed
[234] Chopra H, Kumar S, Singh I. Bioinks for 3D printing of artificial extracellular matrices. Advanced 3D-Printed Systems and Nanosystems for Drug Delivery and Tissue Engineering. United Kingdom: Elsevier; 2020. p. 1–37. 10.1016/b978-0-12-818471-4.00001-7.Search in Google Scholar
[235] Chopra H, Bibi S, Mohanta YK, Kumar Mohanta T, Kumar S, Singh I, et al. In vitro and in silico characterization of curcumin-loaded chitosan–PVA hydrogels: antimicrobial and potential wound healing activity. Gels. 2023;9(5):394. 10.3390/gels9050394.Search in Google Scholar PubMed PubMed Central
[236] Li X, Cho B, Martin R, Seu M, Zhang C, Zhou Z, et al. Nanofiber-hydrogel composite–mediated angiogenesis for soft tissue reconstruction. Sci Transl Med. 2019;11(490):eaau6210. 10.1126/scitranslmed.aau6210.Search in Google Scholar PubMed
[237] Liang Y, Zhao X, Hu T, Chen B, Yin Z, Ma PX, et al. Adhesive hemostatic conducting injectable composite hydrogels with sustained drug release and photothermal antibacterial activity to promote full-thickness skin regeneration during wound healing. Small. 2019;15(12):e1900046. 10.1002/smll.201900046.Search in Google Scholar PubMed
[238] Zhao X, Wu H, Guo B, Dong R, Qiu Y, Ma PX. Antibacterial anti-oxidant electroactive injectable hydrogel as self-healing wound dressing with hemostasis and adhesiveness for cutaneous wound healing. Biomaterials. 2017;122:34–47. 10.1016/j.biomaterials.2017.01.011.Search in Google Scholar PubMed
[239] Tonsomboon K, Butcher AL, Oyen ML. Strong and tough nanofibrous hydrogel composites based on biomimetic principles. Mater Sci Eng C. 2017;72:220–7. 10.1016/j.msec.2016.11.025.Search in Google Scholar PubMed
[240] Dimatteo R, Darling NJ, Segura T. In situ forming injectable hydrogels for drug delivery and wound repair. Adv Drug Deliv Rev. 2018;127:167–84. 10.1016/j.addr.2018.03.007.Search in Google Scholar PubMed PubMed Central
[241] Khorasani MT, Joorabloo A, Moghaddam A, Shamsi H, MansooriMoghadam Z. Incorporation of ZnO nanoparticles into heparinised polyvinyl alcohol/chitosan hydrogels for wound dressing application. Int J Biol Macromol. 2018;114:1203–15. 10.1016/j.ijbiomac.2018.04.010.Search in Google Scholar PubMed
[242] Ghavaminejad A, Park CH, Kim CS. In situ synthesis of antimicrobial silver nanoparticles within antifouling zwitterionic hydrogels by catecholic redox chemistry for wound healing application. Biomacromolecules. 2016;17(3):1213–23. 10.1021/acs.biomac.6b00039.Search in Google Scholar PubMed
[243] Arafa MG, El-Kased RF, Elmazar MM. Thermoresponsive gels containing gold nanoparticles as smart antibacterial and wound healing agents. Sci Rep. 2018;8:13674. 10.1038/s41598-018-31895-4.Search in Google Scholar PubMed PubMed Central
[244] Jones N, Ray B, Ranjit KT, Manna AC. Antibacterial activity of ZnO nanoparticle suspensions on a broad spectrum of microorganisms. FEMS Microbiol Lett. 2008;279(1):71–6. 10.1111/j.1574-6968.2007.01012.x.Search in Google Scholar PubMed
[245] Gordon T, Perlstein B, Houbara O, Felner I, Banin E, Margel S. Synthesis and characterization of zinc/iron oxide composite nanoparticles and their antibacterial properties. Colloids Surfaces A Physicochem Eng Asp. 2011;374(1–3):1–8. 10.1016/j.colsurfa.2010.10.015.Search in Google Scholar
[246] Mahmoud NN, Hikmat S, Abu Ghith D, Hajeer M, Hamadneh L, Qattan D, et al. Gold nanoparticles loaded into polymeric hydrogel for wound healing in rats: Effect of nanoparticles’ shape and surface modification. Int J Pharm. 2019;565:174–86. 10.1016/j.ijpharm.2019.04.079.Search in Google Scholar PubMed
[247] Mahmoud NN, Alkilany AM, Khalil EA, Al-Bakri AG. Antibacterial activity of gold nanorods against staphylococcus aureus and propionibacterium acnes: Misinterpretations and artifacts. Int J Nanomed. 2017;12:7311–22. 10.2147/IJN.S145531.Search in Google Scholar PubMed PubMed Central
[248] Bagher Z, Ehterami A, Safdel MH, Khastar H, Semiari H, Asefnejad A, et al. Wound healing with alginate/chitosan hydrogel containing hesperidin in rat model. J Drug Deliv Sci Technol. 2020;55:101379. 10.1016/j.jddst.2019.101379.Search in Google Scholar
[249] Koneru A, Dharmalingam K, Anandalakshmi R. Cellulose based nanocomposite hydrogel films consisting of sodium carboxymethylcellulose–grapefruit seed extract nanoparticles for potential wound healing applications. Int J Biol Macromol. 2020;148:833–42. 10.1016/j.ijbiomac.2020.01.018.Search in Google Scholar PubMed
[250] Zhou L, Xu T, Yan J, Li X, Xie Y, Chen H. Fabrication and characterization of matrine-loaded konjac glucomannan/fish gelatin composite hydrogel as antimicrobial wound dressing. Food Hydrocoll. 2020;104:105702. 10.1016/j.foodhyd.2020.105702.Search in Google Scholar
[251] Huang L, Zhu Z, Wu D, Gan W, Zhu S, Li W, et al. Antibacterial poly (ethylene glycol) diacrylate/chitosan hydrogels enhance mechanical adhesiveness and promote skin regeneration. Carbohydr Polym. 2019;225:115110. 10.1016/j.carbpol.2019.115110.Search in Google Scholar PubMed
[252] Annabi N, Rana D, Shirzaei Sani E, Portillo-Lara R, Gifford JL, Fares MM, et al. Engineering a sprayable and elastic hydrogel adhesive with antimicrobial properties for wound healing. Biomaterials. 2017;139:229–43. 10.1016/j.biomaterials.2017.05.011.Search in Google Scholar PubMed
[253] Liu Y, Li F, Guo Z, Xiao Y, Zhang Y, Sun X, et al. Silver nanoparticle-embedded hydrogel as a photothermal platform for combating bacterial infections. Chem Eng J. 2020;382:122990. 10.1016/j.cej.2019.122990.Search in Google Scholar
[254] Liang Y, Zhao X, Hu T, Han Y, Guo B. Mussel-inspired, antibacterial, conductive, antioxidant, injectable composite hydrogel wound dressing to promote the regeneration of infected skin. J Colloid Interface Sci. 2019;556:514–28. 10.1016/j.jcis.2019.08.083.Search in Google Scholar PubMed
[255] He J, Shi M, Liang Y, Guo B. Conductive adhesive self-healing nanocomposite hydrogel wound dressing for photothermal therapy of infected full-thickness skin wounds. Chem Eng J. 2020;394:124888. 10.1016/j.cej.2020.124888.Search in Google Scholar
[256] Fu YJ, Shi YF, Wang LY, Zhao YF, Wang RK, Li K, et al. All-natural immunomodulatory bioadhesive hydrogel promotes angiogenesis and diabetic wound healing by regulating macrophage heterogeneity. Adv Sci. 2023;10:2206771. 10.1002/ADVS.202206771.Search in Google Scholar PubMed PubMed Central
[257] Zhang X, Kang X, Jin L, Bai J, Liu W, Wang Z. Stimulation of wound healing using bioinspired hydrogels with basic fibroblast growth factor (bFGF. Int J Nanomed. 2018;13:3897–906. 10.2147/IJN.S168998.Search in Google Scholar PubMed PubMed Central
[258] Luo W, Hu B, Zhang HL, Li C, Shi Y, Li X, et al. Antibacterial, photothermal and stable Ag-titanium-oxo-clusters hydrogel designed for wound healing. Mater Des. 2023;226:111674. 10.1016/J.MATDES.2023.111674.Search in Google Scholar
[259] Hu N, Cai Z, Jiang X, Wang C, Tang T, Xu T, et al. Hypoxia-pretreated ADSC-derived exosome-embedded hydrogels promote angiogenesis and accelerate diabetic wound healing. Acta Biomater. 2023;157:175–86. 10.1016/J.ACTBIO.2022.11.057.Search in Google Scholar
[260] Yue X, Zhao S, Qiu M, Zhang J, Zhong G, Huang C, et al. Physical dual-network photothermal antibacterial multifunctional hydrogel adhesive for wound healing of drug-resistant bacterial infections synthesized from natural polysaccharides. Carbohydr Polym. 2023;312:120831. 10.1016/J.CARBPOL.2023.120831.Search in Google Scholar
[261] Ge P, Chang S, Wang T, Zhao Q, Wang G, He B. An antioxidant and antibacterial polydopamine-modified thermo-sensitive hydrogel dressing for Staphylococcus aureus-infected wound healing. Nanoscale. 2023;15:644–56. 10.1039/D2NR04908B.Search in Google Scholar
[262] Cheng H, Yu Q, Chen Q, Feng L, Zhao W, Zhao C. Biomass-derived ultrafast cross-linked hydrogels with double dynamic bonds for hemostasis and wound healing. Biomater Sci. 2023;11:931–48. 10.1039/D2BM00907B.Search in Google Scholar PubMed
[263] Stoica AE, Chircov C, Grumezescu AM. Nanomaterials for wound dressings: An Up-to-Date overview. Molecules. 2020;25(11):2699. 10.3390/molecules25112699.Search in Google Scholar PubMed PubMed Central
[264] Rajendran NK, Kumar SSD, Houreld NN, Abrahamse H. A review on nanoparticle based treatment for wound healing. J Drug Deliv Sci Technol. 2018;44:421–30. 10.1016/j.jddst.2018.01.009.Search in Google Scholar
[265] Gethin G. The significance of surface pH in chronic wounds. Wounds UK. 2007;3(3):52–6.Search in Google Scholar
[266] Percival SL, McCarty S, Hunt JA, Woods EJ. The effects of pH on wound healing, biofilms, and antimicrobial efficacy. Wound Repair Regen. 2014;22(2):174–86. 10.1111/wrr.12125.Search in Google Scholar PubMed
[267] Ninan N, Forget A, Shastri VP, Voelcker NH, Blencowe A. Antibacterial and anti-inflammatory pH-responsive tannic acid-carboxylated agarose composite hydrogels for wound healing. ACS Appl Mater Interfaces. 2016;8:42. 10.1021/acsami.6b10491.Search in Google Scholar PubMed
[268] Kiaee G, Mostafalu P, Samandari M, Sonkusale S. A pH-mediated electronic wound dressing for controlled drug delivery. Adv Healthc Mater. 2018;7(18):e1800396. 10.1002/adhm.201800396.Search in Google Scholar PubMed
[269] Ahmadian Z, Correia A, Hasany M, Figueiredo P, Dobakhti F, Eskandari MR, et al. A hydrogen-bonded extracellular matrix-mimicking bactericidal hydrogel with radical scavenging and hemostatic function for pH-responsive wound healing acceleration. Adv Healthc Mater. 2021;10(3):2001122. 10.1002/adhm.202001122.Search in Google Scholar PubMed
[270] Ding L, Shan X, Zhao X, Zha H, Chen X, Wang J, et al. Spongy bilayer dressing composed of chitosan–Ag nanoparticles and chitosan–Bletilla striata polysaccharide for wound healing applications. Carbohydr Polym. 2017;157:1538–47. 10.1016/j.carbpol.2016.11.040.Search in Google Scholar PubMed
[271] He J, Zhang Z, Yang Y, Ren F, Li J, Zhu S, et al. Injectable self-healing adhesive pH-responsive hydrogels accelerate gastric hemostasis and wound healing. Nano-Micro Lett. 2021;13:80. 10.1007/s40820-020-00585-0.Search in Google Scholar PubMed PubMed Central
[272] Liu H, Zhu X, Guo H, Huang H, Huang S, Huang S, et al. Nitric oxide released injectable hydrogel combined with synergistic photothermal therapy for antibacterial and accelerated wound healing. Appl Mater Today. 2020;20:100781. 10.1016/j.apmt.2020.100781.Search in Google Scholar
[273] Zhao L, Niu L, Liang H, Tan H, Liu C, Zhu F. PH and glucose dual-responsive injectable hydrogels with insulin and fibroblasts as bioactive dressings for diabetic wound healing. ACS Appl Mater Interfaces. 2017;9:43. 10.1021/acsami.7b09395.Search in Google Scholar PubMed
[274] Wang M, Wang C, Chen M, Xi Y, Cheng W, Mao C, et al. Efficient angiogenesis-based diabetic wound healing/skin reconstruction through bioactive antibacterial adhesive ultraviolet shielding nanodressing with exosome release. ACS Nano. 2019;13(9):10279–93. 10.1021/acsnano.9b03656.Search in Google Scholar PubMed
[275] Hettich BF, Ben-Yehuda Greenwald M, Werner S, Leroux JC. Exosomes for wound healing: purification optimization and identification of bioactive components. Adv Sci. 2020;7(23):2002596. 10.1002/advs.202002596.Search in Google Scholar PubMed PubMed Central
[276] Tao S-C, Guo S-C, Li M, Ke Q-F, Guo Y-P, Zhang C-Q. Chitosan wound dressings incorporating exosomes derived from MicroRNA-126-overexpressing synovium mesenchymal stem cells provide sustained release of exosomes and heal full-thickness skin defects in a diabetic rat model. Stem Cells Transl Med. 2017;6(3):736–47. 10.5966/sctm.2016-0275.Search in Google Scholar PubMed PubMed Central
[277] Vu NB, Nguyen HT, Palumbo R, Pellicano R, Fagoonee S, Pham PV. Stem cell-derived exosomes for wound healing: Current status and promising directions. Minerva Med. 2021;112(3):384–400. 10.23736/S0026-4806.20.07205-5.Search in Google Scholar PubMed
[278] Chen MC, Ling MH, Lai KY, Pramudityo E. Chitosan microneedle patches for sustained transdermal delivery of macromolecules. Biomacromolecules. 2012;13(12):4022–31. 10.1021/bm301293d.Search in Google Scholar PubMed
[279] Biesman BS, Cohen JL, Dibernardo BE, Emer JJ, Geronemus RG, Gold MH, et al. Treatment of atrophic facial acne scars with microneedling followed by polymethylmethacrylate-collagen gel dermal filler. Dermatologic Surg. 2019;45(12):1570–9. 10.1097/DSS.0000000000001872.Search in Google Scholar PubMed
[280] Frydman GH, Olaleye D, Annamalai D, Layne K, Yang I, Kaafarani HMA, et al. Manuka honey microneedles for enhanced wound healing and the prevention and/or treatment of Methicillin-resistant Staphylococcus aureus (MRSA) surgical site infection. Sci Rep. 2020;10:13229. 10.1038/s41598-020-70186-9.Search in Google Scholar PubMed PubMed Central
[281] Yao S, Luo Y, Wang Y. Engineered microneedles arrays for wound healing. Eng Regen. 2022;3(3):232–40. 10.1016/j.engreg.2022.05.003.Search in Google Scholar
[282] Ibrahim ZA, El-Ashmawy AA, Shora OA. Therapeutic effect of microneedling and autologous platelet-rich plasma in the treatment of atrophic scars: A randomized study. J Cosmet Dermatol. 2017;16(3):388–99. 10.1111/jocd.12356.Search in Google Scholar PubMed
[283] Park JH, Allen MG, Prausnitz MR. Polymer microneedles for controlled-release drug delivery. Pharm Res. 2006;23:97–113. 10.1007/s11095-006-0028-9.Search in Google Scholar PubMed
[284] Yao S, Chi J, Wang Y, Zhao Y, Luo Y, Wang Y. Zn-MOF encapsulated antibacterial and degradable microneedles array for promoting wound healing. Adv Healthc Mater. 2021;10(12):e2100056. 10.1002/adhm.202100056.Search in Google Scholar PubMed
[285] Zeng Z, Jiang G, Sun Y, Aharodnikau UE, Yunusov KE, Gao X, et al. Rational design of flexible microneedles coupled with CaO2@PDA-loaded nanofiber films for skin wound healing on diabetic rats. Biomater Sci. 2022;10:5326–39. 10.1039/D2BM00861K.Search in Google Scholar PubMed
[286] Xu J, Xu D, Xuan X, He H. Advances of microneedles in biomedical applications. Molecules. 2021;26(19):5912. 10.3390/MOLECULES26195912.Search in Google Scholar PubMed PubMed Central
[287] Du G, Zhang Z, He P, Zhang Z, Sun X. Determination of the mechanical properties of polymeric microneedles by micromanipulation. J Mech Behav Biomed Mater. 2021;117:104384. 10.1016/j.jmbbm.2021.104384.Search in Google Scholar PubMed
[288] Gao S, Zhang W, Zhai X, Zhao X, Wang J, Weng J, et al. An antibacterial and proangiogenic double-layer drug-loaded microneedle patch for accelerating diabetic wound healing. Biomater Sci. 2023;11:533–41. 10.1039/D2BM01588A.Search in Google Scholar PubMed
[289] Grangier A, Branchu J, Volatron J, Piffoux M, Gazeau F, Wilhelm C, et al. Technological advances towards extracellular vesicles mass production. Adv Drug Deliv Rev. 2021;176:113843. 10.1016/j.addr.2021.113843.Search in Google Scholar PubMed
[290] Ma W, Zhang X, Liu Y, Fan L, Gan J, Liu W, et al. Polydopamine decorated microneedles with Fe-MSC-derived nanovesicles encapsulation for wound healing. Adv Sci. 2022;9(13):2103317. 10.1002/advs.202103317.Search in Google Scholar PubMed PubMed Central
[291] Liu P, Yang X, Han J, Zhao M, Guo J, Si R, et al. Tazarotene-loaded PLGA nanoparticles potentiate deep tissue pressure injury healing via VEGF-Notch signaling. Mater Sci Eng C. 2020;114:111027. 10.1016/j.msec.2020.111027.Search in Google Scholar PubMed
[292] Zhu Z, Liu Y, Xue Y, Cheng X, Zhao W, Wang J, et al. Tazarotene released from aligned electrospun membrane facilitates cutaneous wound healing by promoting angiogenesis. ACS Appl Mater Interfaces. 2019;11(39):36141–53. 10.1021/acsami.9b13271.Search in Google Scholar PubMed
[293] Zeng Y, Wang C, Lei K, Xiao C, Jiang X, Zhang W, et al. Multifunctional MOF-based microneedle patch with synergistic chemo-photodynamic antibacterial effect and sustained release of growth factor for chronic wound healing. Adv Healthc Mater. 2023;12(19):2300250. 10.1002/ADHM.202300250.Search in Google Scholar
[294] Fu C, Shi S, Wei N, Fan Y, Gu H, Liu P, et al. Biocompatible triple-helical recombinant collagen dressings for accelerated wound healing in microneedle-injured and photodamaged skin. Cosmetics. 2023;10(1):31. 10.3390/COSMETICS10010031.Search in Google Scholar
[295] Wang P, Wu J, Yang H, Liu H, Yao T, Liu C, et al. Intelligent microneedle patch with prolonged local release of hydrogen and magnesium ions for diabetic wound healing. Bioact Mater. 2023;24:463–76. 10.1016/J.BIOACTMAT.2023.01.001.Search in Google Scholar
[296] Sheng L, Zhang Z, Zhang Y, Wang E, Ma B, Xu Q, et al. A novel “hot spring”-mimetic hydrogel with excellent angiogenic properties for chronic wound healing. Biomaterials. 2021;264:120414. 10.1016/j.biomaterials.2020.120414.Search in Google Scholar PubMed
[297] Yuan Y, Fan D, Shen S, Ma X. An M2 macrophage-polarized anti-inflammatory hydrogel combined with mild heat stimulation for regulating chronic inflammation and impaired angiogenesis of diabetic wounds. Chem Eng J. 2022;433:133859. 10.1016/j.cej.2021.133859.Search in Google Scholar
[298] Miyauchi T, Miyata M, Ikeda Y, Akasaki Y, Hamada N, Shirasawa T, et al. Waon therapy upregulates Hsp90 and leads to angiogenesis through the Akt-endothelial nitric oxide synthase pathway in mouse hindlimb ischemia. Circ J. 2012;76(7):1712–21. 10.1253/circj.CJ-11-0915.Search in Google Scholar PubMed
[299] Rattan SIS, Fernandes RA, Demirovic D, Dymek B, Lima CF. Heat stress and hormetin-induced hormesis in human cells: Effects on aging, wound healing, angiogenesis, and differentiation. Dose-Response. 2009;7(1):90–103. 10.2203/dose-response.08-014.Rattan.Search in Google Scholar PubMed PubMed Central
[300] Kang K, Lim DH, Choi IH, Kang T, Lee K, Moon EY, et al. Vascular tube formation and angiogenesis induced by polyvinylpyrrolidone-coated silver nanoparticles. Toxicol Lett. 2011;205(3):227–34. 10.1016/j.toxlet.2011.05.1033.Search in Google Scholar PubMed
[301] Zhao Y, Liu Y, Tian C, Liu Z, Wu K, Zhang C, et al. Construction of antibacterial photothermal PCL/AgNPs/BP nanofibers for infected wound healing. Mater Des. 2023;226:111670. 10.1016/J.MATDES.2023.111670.Search in Google Scholar
[302] Cho SK, Mattke S, Gordon H, Sheridan M, Ennis W. Development of a model to predict healing of chronic wounds within 12 weeks. Adv Wound Care. 2020;9(9):516–24. 10.1089/WOUND.2019.1091.Search in Google Scholar PubMed PubMed Central
[303] Anderson C, Bekele Z, Qiu Y, Tschannen D, Dinov ID. Modeling and prediction of pressure injury in hospitalized patients using artificial intelligence. BMC Med Inform Decis Mak. 2021;21:1–13. 10.1186/S12911-021-01608-5.Search in Google Scholar PubMed PubMed Central
[304] VanDelden J. Nano-enhanced wound dressing. 2018.Search in Google Scholar
[305] Fu L, Long M, Ouyang J, Yang H, Zhang Y. Nano-oxide/kaolin composite hemostatic antibacterial material, hemostatic healing-promoting dressing and preparation method thereof, US Pat., 20200230283, 2020.Search in Google Scholar
[306] Ling X, Ying Z, Maolin Z. Nano-silver antibacterial hydrogel and preparation method thereof, US Pat., CN102698313, 2014.Search in Google Scholar
[307] Zhongru G, Xianyan Y, Xiaoyi C. Medical nano-fiber sponge material and preparation method and application thereof, US Pat., CN102657893, 2014.Search in Google Scholar
[308] Yufei T, Yani S, Kang Z, Lei C, Qian L. Self-powered wound patch and preparation method thereof, US Pat., CN113274539, 2022.Search in Google Scholar
[309] Haowen H, Ziqi W, Jiayi Z, Keqin D, Xiumei Y. Preparation method and application of carbon-based bimetallic nanocluster with antibacterial activity and wound healing accelerating capacity, US Pat., CN114533947, 2022.Search in Google Scholar
[310] Yuhao C, Zhongtao Z, Hongwei Y, Zhengyang Y, Peizheng Y, Kairui Y, et al. Nano oxygenated hydrogel healing-promoting dressing containing perfluorodecalin, preparation method therefor and application thereof, US Pat., WO2022184149A1, 2022.Search in Google Scholar
[311] Correa MG, Martínez FB, Vidal CP, Streitt C, Escrig J, de Dicastillo CL. Antimicrobial metal-based nanoparticles: A review on their synthesis, types and antimicrobial action. Beilstein J Nanotechnol. 2020;11:1450–69. 10.3762/BJNANO.11.129.Search in Google Scholar
[312] Dobrucka R, Ankiel M. Possible applications of metal nanoparticles in antimicrobial food packaging. J Food Saf. 2019;39(2):e12617. 10.1111/jfs.12617.Search in Google Scholar
[313] Makowski M, Silva ÍC, Do Amaral CP, Gonçalves S, Santos NC. Advances in lipid and metal nanoparticles for antimicrobial peptide delivery. Pharmaceutics. 2019;11(11):588. 10.3390/pharmaceutics11110588.Search in Google Scholar PubMed PubMed Central
[314] Marinescu L, Ficai D, Oprea O, Marin A, Ficai A, Andronescu E, et al. Optimized Synthesis Approaches of Metal Nanoparticles with Antimicrobial Applications. J Nanomater. 2020;2020:6651207. 10.1155/2020/6651207.Search in Google Scholar
[315] Elena S, Gomes D, Esteruelas G, Bonilla L, Lopez-machado AL, Galindo R, et al. Metal-based nanoparticles as antimicrobial agents: an overview. Nanomaterials (Basel). 2020;10(2):292.10.3390/nano10020292Search in Google Scholar PubMed PubMed Central
[316] Nanotechnology Guidance Documents. FDA. 2022.Search in Google Scholar
[317] Multidisciplinary: nanomedicines. European Medicines Agency. 2022.Search in Google Scholar
[318] ISO/TC 229. Nanotechnologies. 2005.Search in Google Scholar
© 2023 the author(s), published by De Gruyter
This work is licensed under the Creative Commons Attribution 4.0 International License.
Articles in the same Issue
- Research Articles
- Preparation of CdS–Ag2S nanocomposites by ultrasound-assisted UV photolysis treatment and its visible light photocatalysis activity
- Significance of nanoparticle radius and inter-particle spacing toward the radiative water-based alumina nanofluid flow over a rotating disk
- Aptamer-based detection of serotonin based on the rapid in situ synthesis of colorimetric gold nanoparticles
- Investigation of the nucleation and growth behavior of Ti2AlC and Ti3AlC nano-precipitates in TiAl alloys
- Dynamic recrystallization behavior and nucleation mechanism of dual-scale SiCp/A356 composites processed by P/M method
- High mechanical performance of 3-aminopropyl triethoxy silane/epoxy cured in a sandwich construction of 3D carbon felts foam and woven basalt fibers
- Applying solution of spray polyurea elastomer in asphalt binder: Feasibility analysis and DSR study based on the MSCR and LAS tests
- Study on the chronic toxicity and carcinogenicity of iron-based bioabsorbable stents
- Influence of microalloying with B on the microstructure and properties of brazed joints with Ag–Cu–Zn–Sn filler metal
- Thermohydraulic performance of thermal system integrated with twisted turbulator inserts using ternary hybrid nanofluids
- Study of mechanical properties of epoxy/graphene and epoxy/halloysite nanocomposites
- Effects of CaO addition on the CuW composite containing micro- and nano-sized tungsten particles synthesized via aluminothermic coupling with silicothermic reduction
- Cu and Al2O3-based hybrid nanofluid flow through a porous cavity
- Design of functional vancomycin-embedded bio-derived extracellular matrix hydrogels for repairing infectious bone defects
- Study on nanocrystalline coating prepared by electro-spraying 316L metal wire and its corrosion performance
- Axial compression performance of CFST columns reinforced by ultra-high-performance nano-concrete under long-term loading
- Tungsten trioxide nanocomposite for conventional soliton and noise-like pulse generation in anomalous dispersion laser cavity
- Microstructure and electrical contact behavior of the nano-yttria-modified Cu-Al2O3/30Mo/3SiC composite
- Melting rheology in thermally stratified graphene-mineral oil reservoir (third-grade nanofluid) with slip condition
- Re-examination of nonlinear vibration and nonlinear bending of porous sandwich cylindrical panels reinforced by graphene platelets
- Parametric simulation of hybrid nanofluid flow consisting of cobalt ferrite nanoparticles with second-order slip and variable viscosity over an extending surface
- Chitosan-capped silver nanoparticles with potent and selective intrinsic activity against the breast cancer cells
- Multi-core/shell SiO2@Al2O3 nanostructures deposited on Ti3AlC2 to enhance high-temperature stability and microwave absorption properties
- Solution-processed Bi2S3/BiVO4/TiO2 ternary heterojunction photoanode with enhanced photoelectrochemical performance
- Electroporation effect of ZnO nanoarrays under low voltage for water disinfection
- NIR-II window absorbing graphene oxide-coated gold nanorods and graphene quantum dot-coupled gold nanorods for photothermal cancer therapy
- Nonlinear three-dimensional stability characteristics of geometrically imperfect nanoshells under axial compression and surface residual stress
- Investigation of different nanoparticles properties on the thermal conductivity and viscosity of nanofluids by molecular dynamics simulation
- Optimized Cu2O-{100} facet for generation of different reactive oxidative species via peroxymonosulfate activation at specific pH values to efficient acetaminophen removal
- Brownian and thermal diffusivity impact due to the Maxwell nanofluid (graphene/engine oil) flow with motile microorganisms and Joule heating
- Appraising the dielectric properties and the effectiveness of electromagnetic shielding of graphene reinforced silicone rubber nanocomposite
- Synthesis of Ag and Cu nanoparticles by plasma discharge in inorganic salt solutions
- Low-cost and large-scale preparation of ultrafine TiO2@C hybrids for high-performance degradation of methyl orange and formaldehyde under visible light
- Utilization of waste glass with natural pozzolan in the production of self-glazed glass-ceramic materials
- Mechanical performance of date palm fiber-reinforced concrete modified with nano-activated carbon
- Melting point of dried gold nanoparticles prepared with ultrasonic spray pyrolysis and lyophilisation
- Graphene nanofibers: A modern approach towards tailored gypsum composites
- Role of localized magnetic field in vortex generation in tri-hybrid nanofluid flow: A numerical approach
- Intelligent computing for the double-diffusive peristaltic rheology of magneto couple stress nanomaterials
- Bioconvection transport of upper convected Maxwell nanoliquid with gyrotactic microorganism, nonlinear thermal radiation, and chemical reaction
- 3D printing of porous Ti6Al4V bone tissue engineering scaffold and surface anodization preparation of nanotubes to enhance its biological property
- Bioinspired ferromagnetic CoFe2O4 nanoparticles: Potential pharmaceutical and medical applications
- Significance of gyrotactic microorganisms on the MHD tangent hyperbolic nanofluid flow across an elastic slender surface: Numerical analysis
- Performance of polycarboxylate superplasticisers in seawater-blended cement: Effect from chemical structure and nano modification
- Entropy minimization of GO–Ag/KO cross-hybrid nanofluid over a convectively heated surface
- Oxygen plasma assisted room temperature bonding for manufacturing SU-8 polymer micro/nanoscale nozzle
- Performance and mechanism of CO2 reduction by DBD-coupled mesoporous SiO2
- Polyarylene ether nitrile dielectric films modified by HNTs@PDA hybrids for high-temperature resistant organic electronics field
- Exploration of generalized two-phase free convection magnetohydrodynamic flow of dusty tetra-hybrid Casson nanofluid between parallel microplates
- Hygrothermal bending analysis of sandwich nanoplates with FG porous core and piezomagnetic faces via nonlocal strain gradient theory
- Design and optimization of a TiO2/RGO-supported epoxy multilayer microwave absorber by the modified local best particle swarm optimization algorithm
- Mechanical properties and frost resistance of recycled brick aggregate concrete modified by nano-SiO2
- Self-template synthesis of hollow flower-like NiCo2O4 nanoparticles as an efficient bifunctional catalyst for oxygen reduction and oxygen evolution in alkaline media
- High-performance wearable flexible strain sensors based on an AgNWs/rGO/TPU electrospun nanofiber film for monitoring human activities
- High-performance lithium–selenium batteries enabled by nitrogen-doped porous carbon from peanut meal
- Investigating effects of Lorentz forces and convective heating on ternary hybrid nanofluid flow over a curved surface using homotopy analysis method
- Exploring the potential of biogenic magnesium oxide nanoparticles for cytotoxicity: In vitro and in silico studies on HCT116 and HT29 cells and DPPH radical scavenging
- Enhanced visible-light-driven photocatalytic degradation of azo dyes by heteroatom-doped nickel tungstate nanoparticles
- A facile method to synthesize nZVI-doped polypyrrole-based carbon nanotube for Ag(i) removal
- Improved osseointegration of dental titanium implants by TiO2 nanotube arrays with self-assembled recombinant IGF-1 in type 2 diabetes mellitus rat model
- Functionalized SWCNTs@Ag–TiO2 nanocomposites induce ROS-mediated apoptosis and autophagy in liver cancer cells
- Triboelectric nanogenerator based on a water droplet spring with a concave spherical surface for harvesting wave energy and detecting pressure
- A mathematical approach for modeling the blood flow containing nanoparticles by employing the Buongiorno’s model
- Molecular dynamics study on dynamic interlayer friction of graphene and its strain effect
- Induction of apoptosis and autophagy via regulation of AKT and JNK mitogen-activated protein kinase pathways in breast cancer cell lines exposed to gold nanoparticles loaded with TNF-α and combined with doxorubicin
- Effect of PVA fibers on durability of nano-SiO2-reinforced cement-based composites subjected to wet-thermal and chloride salt-coupled environment
- Effect of polyvinyl alcohol fibers on mechanical properties of nano-SiO2-reinforced geopolymer composites under a complex environment
- In vitro studies of titanium dioxide nanoparticles modified with glutathione as a potential drug delivery system
- Comparative investigations of Ag/H2O nanofluid and Ag-CuO/H2O hybrid nanofluid with Darcy-Forchheimer flow over a curved surface
- Study on deformation characteristics of multi-pass continuous drawing of micro copper wire based on crystal plasticity finite element method
- Properties of ultra-high-performance self-compacting fiber-reinforced concrete modified with nanomaterials
- Prediction of lap shear strength of GNP and TiO2/epoxy nanocomposite adhesives
- A novel exploration of how localized magnetic field affects vortex generation of trihybrid nanofluids
- Fabrication and physicochemical characterization of copper oxide–pyrrhotite nanocomposites for the cytotoxic effects on HepG2 cells and the mechanism
- Thermal radiative flow of cross nanofluid due to a stretched cylinder containing microorganisms
- In vitro study of the biphasic calcium phosphate/chitosan hybrid biomaterial scaffold fabricated via solvent casting and evaporation technique for bone regeneration
- Insights into the thermal characteristics and dynamics of stagnant blood conveying titanium oxide, alumina, and silver nanoparticles subject to Lorentz force and internal heating over a curved surface
- Effects of nano-SiO2 additives on carbon fiber-reinforced fly ash–slag geopolymer composites performance: Workability, mechanical properties, and microstructure
- Energy bandgap and thermal characteristics of non-Darcian MHD rotating hybridity nanofluid thin film flow: Nanotechnology application
- Green synthesis and characterization of ginger-extract-based oxali-palladium nanoparticles for colorectal cancer: Downregulation of REG4 and apoptosis induction
- Abnormal evolution of resistivity and microstructure of annealed Ag nanoparticles/Ag–Mo films
- Preparation of water-based dextran-coated Fe3O4 magnetic fluid for magnetic hyperthermia
- Statistical investigations and morphological aspects of cross-rheological material suspended in transportation of alumina, silica, titanium, and ethylene glycol via the Galerkin algorithm
- Effect of CNT film interleaves on the flexural properties and strength after impact of CFRP composites
- Self-assembled nanoscale entities: Preparative process optimization, payload release, and enhanced bioavailability of thymoquinone natural product
- Structure–mechanical property relationships of 3D-printed porous polydimethylsiloxane films
- Nonlinear thermal radiation and the slip effect on a 3D bioconvection flow of the Casson nanofluid in a rotating frame via a homotopy analysis mechanism
- Residual mechanical properties of concrete incorporated with nano supplementary cementitious materials exposed to elevated temperature
- Time-independent three-dimensional flow of a water-based hybrid nanofluid past a Riga plate with slips and convective conditions: A homotopic solution
- Lightweight and high-strength polyarylene ether nitrile-based composites for efficient electromagnetic interference shielding
- Review Articles
- Recycling waste sources into nanocomposites of graphene materials: Overview from an energy-focused perspective
- Hybrid nanofiller reinforcement in thermoset and biothermoset applications: A review
- Current state-of-the-art review of nanotechnology-based therapeutics for viral pandemics: Special attention to COVID-19
- Solid lipid nanoparticles for targeted natural and synthetic drugs delivery in high-incidence cancers, and other diseases: Roles of preparation methods, lipid composition, transitional stability, and release profiles in nanocarriers’ development
- Critical review on experimental and theoretical studies of elastic properties of wurtzite-structured ZnO nanowires
- Polyurea micro-/nano-capsule applications in construction industry: A review
- A comprehensive review and clinical guide to molecular and serological diagnostic tests and future development: In vitro diagnostic testing for COVID-19
- Recent advances in electrocatalytic oxidation of 5-hydroxymethylfurfural to 2,5-furandicarboxylic acid: Mechanism, catalyst, coupling system
- Research progress and prospect of silica-based polymer nanofluids in enhanced oil recovery
- Review of the pharmacokinetics of nanodrugs
- Engineered nanoflowers, nanotrees, nanostars, nanodendrites, and nanoleaves for biomedical applications
- Research progress of biopolymers combined with stem cells in the repair of intrauterine adhesions
- Progress in FEM modeling on mechanical and electromechanical properties of carbon nanotube cement-based composites
- Antifouling induced by surface wettability of poly(dimethyl siloxane) and its nanocomposites
- TiO2 aerogel composite high-efficiency photocatalysts for environmental treatment and hydrogen energy production
- Structural properties of alumina surfaces and their roles in the synthesis of environmentally persistent free radicals (EPFRs)
- Nanoparticles for the potential treatment of Alzheimer’s disease: A physiopathological approach
- Current status of synthesis and consolidation strategies for thermo-resistant nanoalloys and their general applications
- Recent research progress on the stimuli-responsive smart membrane: A review
- Dispersion of carbon nanotubes in aqueous cementitious materials: A review
- Applications of DNA tetrahedron nanostructure in cancer diagnosis and anticancer drugs delivery
- Magnetic nanoparticles in 3D-printed scaffolds for biomedical applications
- An overview of the synthesis of silicon carbide–boron carbide composite powders
- Organolead halide perovskites: Synthetic routes, structural features, and their potential in the development of photovoltaic
- Recent advancements in nanotechnology application on wood and bamboo materials: A review
- Application of aptamer-functionalized nanomaterials in molecular imaging of tumors
- Recent progress on corrosion mechanisms of graphene-reinforced metal matrix composites
- Research progress on preparation, modification, and application of phenolic aerogel
- Application of nanomaterials in early diagnosis of cancer
- Plant mediated-green synthesis of zinc oxide nanoparticles: An insight into biomedical applications
- Recent developments in terahertz quantum cascade lasers for practical applications
- Recent progress in dielectric/metal/dielectric electrodes for foldable light-emitting devices
- Nanocoatings for ballistic applications: A review
- A mini-review on MoS2 membrane for water desalination: Recent development and challenges
- Recent updates in nanotechnological advances for wound healing: A narrative review
- Recent advances in DNA nanomaterials for cancer diagnosis and treatment
- Electrochemical micro- and nanobiosensors for in vivo reactive oxygen/nitrogen species measurement in the brain
- Advances in organic–inorganic nanocomposites for cancer imaging and therapy
- Advancements in aluminum matrix composites reinforced with carbides and graphene: A comprehensive review
- Modification effects of nanosilica on asphalt binders: A review
- Decellularized extracellular matrix as a promising biomaterial for musculoskeletal tissue regeneration
- Review of the sol–gel method in preparing nano TiO2 for advanced oxidation process
- Micro/nano manufacturing aircraft surface with anti-icing and deicing performances: An overview
- Cell type-targeting nanoparticles in treating central nervous system diseases: Challenges and hopes
- An overview of hydrogen production from Al-based materials
- A review of application, modification, and prospect of melamine foam
- A review of the performance of fibre-reinforced composite laminates with carbon nanotubes
- Research on AFM tip-related nanofabrication of two-dimensional materials
- Advances in phase change building materials: An overview
- Development of graphene and graphene quantum dots toward biomedical engineering applications: A review
- Nanoremediation approaches for the mitigation of heavy metal contamination in vegetables: An overview
- Photodynamic therapy empowered by nanotechnology for oral and dental science: Progress and perspectives
- Biosynthesis of metal nanoparticles: Bioreduction and biomineralization
- Current diagnostic and therapeutic approaches for severe acute respiratory syndrome coronavirus-2 (SARS-COV-2) and the role of nanomaterial-based theragnosis in combating the pandemic
- Application of two-dimensional black phosphorus material in wound healing
- Special Issue on Advanced Nanomaterials and Composites for Energy Conversion and Storage - Part I
- Helical fluorinated carbon nanotubes/iron(iii) fluoride hybrid with multilevel transportation channels and rich active sites for lithium/fluorinated carbon primary battery
- The progress of cathode materials in aqueous zinc-ion batteries
- Special Issue on Advanced Nanomaterials for Carbon Capture, Environment and Utilization for Energy Sustainability - Part I
- Effect of polypropylene fiber and nano-silica on the compressive strength and frost resistance of recycled brick aggregate concrete
- Mechanochemical design of nanomaterials for catalytic applications with a benign-by-design focus
Articles in the same Issue
- Research Articles
- Preparation of CdS–Ag2S nanocomposites by ultrasound-assisted UV photolysis treatment and its visible light photocatalysis activity
- Significance of nanoparticle radius and inter-particle spacing toward the radiative water-based alumina nanofluid flow over a rotating disk
- Aptamer-based detection of serotonin based on the rapid in situ synthesis of colorimetric gold nanoparticles
- Investigation of the nucleation and growth behavior of Ti2AlC and Ti3AlC nano-precipitates in TiAl alloys
- Dynamic recrystallization behavior and nucleation mechanism of dual-scale SiCp/A356 composites processed by P/M method
- High mechanical performance of 3-aminopropyl triethoxy silane/epoxy cured in a sandwich construction of 3D carbon felts foam and woven basalt fibers
- Applying solution of spray polyurea elastomer in asphalt binder: Feasibility analysis and DSR study based on the MSCR and LAS tests
- Study on the chronic toxicity and carcinogenicity of iron-based bioabsorbable stents
- Influence of microalloying with B on the microstructure and properties of brazed joints with Ag–Cu–Zn–Sn filler metal
- Thermohydraulic performance of thermal system integrated with twisted turbulator inserts using ternary hybrid nanofluids
- Study of mechanical properties of epoxy/graphene and epoxy/halloysite nanocomposites
- Effects of CaO addition on the CuW composite containing micro- and nano-sized tungsten particles synthesized via aluminothermic coupling with silicothermic reduction
- Cu and Al2O3-based hybrid nanofluid flow through a porous cavity
- Design of functional vancomycin-embedded bio-derived extracellular matrix hydrogels for repairing infectious bone defects
- Study on nanocrystalline coating prepared by electro-spraying 316L metal wire and its corrosion performance
- Axial compression performance of CFST columns reinforced by ultra-high-performance nano-concrete under long-term loading
- Tungsten trioxide nanocomposite for conventional soliton and noise-like pulse generation in anomalous dispersion laser cavity
- Microstructure and electrical contact behavior of the nano-yttria-modified Cu-Al2O3/30Mo/3SiC composite
- Melting rheology in thermally stratified graphene-mineral oil reservoir (third-grade nanofluid) with slip condition
- Re-examination of nonlinear vibration and nonlinear bending of porous sandwich cylindrical panels reinforced by graphene platelets
- Parametric simulation of hybrid nanofluid flow consisting of cobalt ferrite nanoparticles with second-order slip and variable viscosity over an extending surface
- Chitosan-capped silver nanoparticles with potent and selective intrinsic activity against the breast cancer cells
- Multi-core/shell SiO2@Al2O3 nanostructures deposited on Ti3AlC2 to enhance high-temperature stability and microwave absorption properties
- Solution-processed Bi2S3/BiVO4/TiO2 ternary heterojunction photoanode with enhanced photoelectrochemical performance
- Electroporation effect of ZnO nanoarrays under low voltage for water disinfection
- NIR-II window absorbing graphene oxide-coated gold nanorods and graphene quantum dot-coupled gold nanorods for photothermal cancer therapy
- Nonlinear three-dimensional stability characteristics of geometrically imperfect nanoshells under axial compression and surface residual stress
- Investigation of different nanoparticles properties on the thermal conductivity and viscosity of nanofluids by molecular dynamics simulation
- Optimized Cu2O-{100} facet for generation of different reactive oxidative species via peroxymonosulfate activation at specific pH values to efficient acetaminophen removal
- Brownian and thermal diffusivity impact due to the Maxwell nanofluid (graphene/engine oil) flow with motile microorganisms and Joule heating
- Appraising the dielectric properties and the effectiveness of electromagnetic shielding of graphene reinforced silicone rubber nanocomposite
- Synthesis of Ag and Cu nanoparticles by plasma discharge in inorganic salt solutions
- Low-cost and large-scale preparation of ultrafine TiO2@C hybrids for high-performance degradation of methyl orange and formaldehyde under visible light
- Utilization of waste glass with natural pozzolan in the production of self-glazed glass-ceramic materials
- Mechanical performance of date palm fiber-reinforced concrete modified with nano-activated carbon
- Melting point of dried gold nanoparticles prepared with ultrasonic spray pyrolysis and lyophilisation
- Graphene nanofibers: A modern approach towards tailored gypsum composites
- Role of localized magnetic field in vortex generation in tri-hybrid nanofluid flow: A numerical approach
- Intelligent computing for the double-diffusive peristaltic rheology of magneto couple stress nanomaterials
- Bioconvection transport of upper convected Maxwell nanoliquid with gyrotactic microorganism, nonlinear thermal radiation, and chemical reaction
- 3D printing of porous Ti6Al4V bone tissue engineering scaffold and surface anodization preparation of nanotubes to enhance its biological property
- Bioinspired ferromagnetic CoFe2O4 nanoparticles: Potential pharmaceutical and medical applications
- Significance of gyrotactic microorganisms on the MHD tangent hyperbolic nanofluid flow across an elastic slender surface: Numerical analysis
- Performance of polycarboxylate superplasticisers in seawater-blended cement: Effect from chemical structure and nano modification
- Entropy minimization of GO–Ag/KO cross-hybrid nanofluid over a convectively heated surface
- Oxygen plasma assisted room temperature bonding for manufacturing SU-8 polymer micro/nanoscale nozzle
- Performance and mechanism of CO2 reduction by DBD-coupled mesoporous SiO2
- Polyarylene ether nitrile dielectric films modified by HNTs@PDA hybrids for high-temperature resistant organic electronics field
- Exploration of generalized two-phase free convection magnetohydrodynamic flow of dusty tetra-hybrid Casson nanofluid between parallel microplates
- Hygrothermal bending analysis of sandwich nanoplates with FG porous core and piezomagnetic faces via nonlocal strain gradient theory
- Design and optimization of a TiO2/RGO-supported epoxy multilayer microwave absorber by the modified local best particle swarm optimization algorithm
- Mechanical properties and frost resistance of recycled brick aggregate concrete modified by nano-SiO2
- Self-template synthesis of hollow flower-like NiCo2O4 nanoparticles as an efficient bifunctional catalyst for oxygen reduction and oxygen evolution in alkaline media
- High-performance wearable flexible strain sensors based on an AgNWs/rGO/TPU electrospun nanofiber film for monitoring human activities
- High-performance lithium–selenium batteries enabled by nitrogen-doped porous carbon from peanut meal
- Investigating effects of Lorentz forces and convective heating on ternary hybrid nanofluid flow over a curved surface using homotopy analysis method
- Exploring the potential of biogenic magnesium oxide nanoparticles for cytotoxicity: In vitro and in silico studies on HCT116 and HT29 cells and DPPH radical scavenging
- Enhanced visible-light-driven photocatalytic degradation of azo dyes by heteroatom-doped nickel tungstate nanoparticles
- A facile method to synthesize nZVI-doped polypyrrole-based carbon nanotube for Ag(i) removal
- Improved osseointegration of dental titanium implants by TiO2 nanotube arrays with self-assembled recombinant IGF-1 in type 2 diabetes mellitus rat model
- Functionalized SWCNTs@Ag–TiO2 nanocomposites induce ROS-mediated apoptosis and autophagy in liver cancer cells
- Triboelectric nanogenerator based on a water droplet spring with a concave spherical surface for harvesting wave energy and detecting pressure
- A mathematical approach for modeling the blood flow containing nanoparticles by employing the Buongiorno’s model
- Molecular dynamics study on dynamic interlayer friction of graphene and its strain effect
- Induction of apoptosis and autophagy via regulation of AKT and JNK mitogen-activated protein kinase pathways in breast cancer cell lines exposed to gold nanoparticles loaded with TNF-α and combined with doxorubicin
- Effect of PVA fibers on durability of nano-SiO2-reinforced cement-based composites subjected to wet-thermal and chloride salt-coupled environment
- Effect of polyvinyl alcohol fibers on mechanical properties of nano-SiO2-reinforced geopolymer composites under a complex environment
- In vitro studies of titanium dioxide nanoparticles modified with glutathione as a potential drug delivery system
- Comparative investigations of Ag/H2O nanofluid and Ag-CuO/H2O hybrid nanofluid with Darcy-Forchheimer flow over a curved surface
- Study on deformation characteristics of multi-pass continuous drawing of micro copper wire based on crystal plasticity finite element method
- Properties of ultra-high-performance self-compacting fiber-reinforced concrete modified with nanomaterials
- Prediction of lap shear strength of GNP and TiO2/epoxy nanocomposite adhesives
- A novel exploration of how localized magnetic field affects vortex generation of trihybrid nanofluids
- Fabrication and physicochemical characterization of copper oxide–pyrrhotite nanocomposites for the cytotoxic effects on HepG2 cells and the mechanism
- Thermal radiative flow of cross nanofluid due to a stretched cylinder containing microorganisms
- In vitro study of the biphasic calcium phosphate/chitosan hybrid biomaterial scaffold fabricated via solvent casting and evaporation technique for bone regeneration
- Insights into the thermal characteristics and dynamics of stagnant blood conveying titanium oxide, alumina, and silver nanoparticles subject to Lorentz force and internal heating over a curved surface
- Effects of nano-SiO2 additives on carbon fiber-reinforced fly ash–slag geopolymer composites performance: Workability, mechanical properties, and microstructure
- Energy bandgap and thermal characteristics of non-Darcian MHD rotating hybridity nanofluid thin film flow: Nanotechnology application
- Green synthesis and characterization of ginger-extract-based oxali-palladium nanoparticles for colorectal cancer: Downregulation of REG4 and apoptosis induction
- Abnormal evolution of resistivity and microstructure of annealed Ag nanoparticles/Ag–Mo films
- Preparation of water-based dextran-coated Fe3O4 magnetic fluid for magnetic hyperthermia
- Statistical investigations and morphological aspects of cross-rheological material suspended in transportation of alumina, silica, titanium, and ethylene glycol via the Galerkin algorithm
- Effect of CNT film interleaves on the flexural properties and strength after impact of CFRP composites
- Self-assembled nanoscale entities: Preparative process optimization, payload release, and enhanced bioavailability of thymoquinone natural product
- Structure–mechanical property relationships of 3D-printed porous polydimethylsiloxane films
- Nonlinear thermal radiation and the slip effect on a 3D bioconvection flow of the Casson nanofluid in a rotating frame via a homotopy analysis mechanism
- Residual mechanical properties of concrete incorporated with nano supplementary cementitious materials exposed to elevated temperature
- Time-independent three-dimensional flow of a water-based hybrid nanofluid past a Riga plate with slips and convective conditions: A homotopic solution
- Lightweight and high-strength polyarylene ether nitrile-based composites for efficient electromagnetic interference shielding
- Review Articles
- Recycling waste sources into nanocomposites of graphene materials: Overview from an energy-focused perspective
- Hybrid nanofiller reinforcement in thermoset and biothermoset applications: A review
- Current state-of-the-art review of nanotechnology-based therapeutics for viral pandemics: Special attention to COVID-19
- Solid lipid nanoparticles for targeted natural and synthetic drugs delivery in high-incidence cancers, and other diseases: Roles of preparation methods, lipid composition, transitional stability, and release profiles in nanocarriers’ development
- Critical review on experimental and theoretical studies of elastic properties of wurtzite-structured ZnO nanowires
- Polyurea micro-/nano-capsule applications in construction industry: A review
- A comprehensive review and clinical guide to molecular and serological diagnostic tests and future development: In vitro diagnostic testing for COVID-19
- Recent advances in electrocatalytic oxidation of 5-hydroxymethylfurfural to 2,5-furandicarboxylic acid: Mechanism, catalyst, coupling system
- Research progress and prospect of silica-based polymer nanofluids in enhanced oil recovery
- Review of the pharmacokinetics of nanodrugs
- Engineered nanoflowers, nanotrees, nanostars, nanodendrites, and nanoleaves for biomedical applications
- Research progress of biopolymers combined with stem cells in the repair of intrauterine adhesions
- Progress in FEM modeling on mechanical and electromechanical properties of carbon nanotube cement-based composites
- Antifouling induced by surface wettability of poly(dimethyl siloxane) and its nanocomposites
- TiO2 aerogel composite high-efficiency photocatalysts for environmental treatment and hydrogen energy production
- Structural properties of alumina surfaces and their roles in the synthesis of environmentally persistent free radicals (EPFRs)
- Nanoparticles for the potential treatment of Alzheimer’s disease: A physiopathological approach
- Current status of synthesis and consolidation strategies for thermo-resistant nanoalloys and their general applications
- Recent research progress on the stimuli-responsive smart membrane: A review
- Dispersion of carbon nanotubes in aqueous cementitious materials: A review
- Applications of DNA tetrahedron nanostructure in cancer diagnosis and anticancer drugs delivery
- Magnetic nanoparticles in 3D-printed scaffolds for biomedical applications
- An overview of the synthesis of silicon carbide–boron carbide composite powders
- Organolead halide perovskites: Synthetic routes, structural features, and their potential in the development of photovoltaic
- Recent advancements in nanotechnology application on wood and bamboo materials: A review
- Application of aptamer-functionalized nanomaterials in molecular imaging of tumors
- Recent progress on corrosion mechanisms of graphene-reinforced metal matrix composites
- Research progress on preparation, modification, and application of phenolic aerogel
- Application of nanomaterials in early diagnosis of cancer
- Plant mediated-green synthesis of zinc oxide nanoparticles: An insight into biomedical applications
- Recent developments in terahertz quantum cascade lasers for practical applications
- Recent progress in dielectric/metal/dielectric electrodes for foldable light-emitting devices
- Nanocoatings for ballistic applications: A review
- A mini-review on MoS2 membrane for water desalination: Recent development and challenges
- Recent updates in nanotechnological advances for wound healing: A narrative review
- Recent advances in DNA nanomaterials for cancer diagnosis and treatment
- Electrochemical micro- and nanobiosensors for in vivo reactive oxygen/nitrogen species measurement in the brain
- Advances in organic–inorganic nanocomposites for cancer imaging and therapy
- Advancements in aluminum matrix composites reinforced with carbides and graphene: A comprehensive review
- Modification effects of nanosilica on asphalt binders: A review
- Decellularized extracellular matrix as a promising biomaterial for musculoskeletal tissue regeneration
- Review of the sol–gel method in preparing nano TiO2 for advanced oxidation process
- Micro/nano manufacturing aircraft surface with anti-icing and deicing performances: An overview
- Cell type-targeting nanoparticles in treating central nervous system diseases: Challenges and hopes
- An overview of hydrogen production from Al-based materials
- A review of application, modification, and prospect of melamine foam
- A review of the performance of fibre-reinforced composite laminates with carbon nanotubes
- Research on AFM tip-related nanofabrication of two-dimensional materials
- Advances in phase change building materials: An overview
- Development of graphene and graphene quantum dots toward biomedical engineering applications: A review
- Nanoremediation approaches for the mitigation of heavy metal contamination in vegetables: An overview
- Photodynamic therapy empowered by nanotechnology for oral and dental science: Progress and perspectives
- Biosynthesis of metal nanoparticles: Bioreduction and biomineralization
- Current diagnostic and therapeutic approaches for severe acute respiratory syndrome coronavirus-2 (SARS-COV-2) and the role of nanomaterial-based theragnosis in combating the pandemic
- Application of two-dimensional black phosphorus material in wound healing
- Special Issue on Advanced Nanomaterials and Composites for Energy Conversion and Storage - Part I
- Helical fluorinated carbon nanotubes/iron(iii) fluoride hybrid with multilevel transportation channels and rich active sites for lithium/fluorinated carbon primary battery
- The progress of cathode materials in aqueous zinc-ion batteries
- Special Issue on Advanced Nanomaterials for Carbon Capture, Environment and Utilization for Energy Sustainability - Part I
- Effect of polypropylene fiber and nano-silica on the compressive strength and frost resistance of recycled brick aggregate concrete
- Mechanochemical design of nanomaterials for catalytic applications with a benign-by-design focus