Next Article in Journal
Microstructure and Photocatalytic Performance of BaTi5O11 Nanocrystals Synthesized via Sol-Gel Method Mediated by Organic Solvents
Previous Article in Journal
Advances in Preparation and Biomedical Applications of Sodium Alginate-Based Electrospun Nanofibers
Previous Article in Special Issue
Amygdalin-Doped Biopolymer Composites as Potential Wound Dressing Films: In Vitro Study on E. coli and S. aureus
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Biological Models for Evaluating Hydrogel-Based Formulations in Wound Healing

by
Ioana Baldea
1,
Ioana Georgeta Grosu
2,*,
Sahar Ghafury
1,*,
Cristian Golat
1,
Doriane Doubali
1,
Ana-Maria Vestemean
1,
Aris Nicolas Cedorge
1,
Ilinca Florian
1,
Michael Yiannoulatos
1,
Muhammad Mudassir Wajahat
1,
Lorenzo Raoul Silli
1,
Thesseus Stavrou
1 and
Daniela Rodica Mitrea
1
1
Department of Physiology, University of Medicine and Pharmacy, Clinicilor 1, 400006 Cluj-Napoca, Romania
2
National Institute for Research and Development of Isotopic and Molecular Technologies, Donat 67-103, 400293 Cluj-Napoca, Romania
*
Authors to whom correspondence should be addressed.
Gels 2025, 11(9), 705; https://doi.org/10.3390/gels11090705
Submission received: 31 July 2025 / Revised: 29 August 2025 / Accepted: 1 September 2025 / Published: 3 September 2025
(This article belongs to the Special Issue Novel Functional Gels for Biomedical Applications (2nd Edition))

Abstract

Skin, the largest organ of the human body, serves as a critical physico-chemical barrier against environmental insults and plays essential roles in hydration, thermoregulation, immune defense, and metabolic functions. Wound healing is a complex, multistage biological process involving hemostasis, inflammation, proliferation, and remodeling. Hydrogels have emerged as a promising class of wound dressings due to their high moisture retention, biocompatibility, and ability to mimic the extracellular matrix, thereby supporting accelerated healing and controlled drug delivery. This review provides a comprehensive overview of current hydrogel types—classified by origin, crosslinking mechanisms, and responsiveness to stimuli—and evaluates their use in experimental research on in vitro, ex vivo, and in vivo wound healing models. Furthermore, clinical applications of hydrogels in wound therapy are discussed. Advances in semisynthetic and stimuli-responsive hydrogels, along with improved testing models, offer enhanced therapeutic potential and underscore the need for continued innovation to optimize wound care outcomes and alleviate healthcare burdens.

Graphical Abstract

1. Introduction

Skin is the largest human body organ, providing the initial physico-chemical barrier against saprophytes and pathogens, radiation, such as ultraviolet irradiation, and chemical and mechanical injuries. Besides the barrier function, skin is an active part of many other homeostatic mechanisms. It maintains the body’s hydration state, thermoregulation, immune recognition and defense, synthesis of vitamin D precursors, and excretion of some catabolites and drugs [1].
Millions of skin injuries caused by physical or thermal agents occur annually, and if not properly treated, they can develop into chronic wounds. Wounds are defined as any damage or disruption of the body’s tissues, typically the epidermis. They can be classified into open (laceration) and closed (hematomas). According to the wound age, onset mechanism, and healing capacity, they can be acute or chronic [2].
The wound healing process is a physiological reaction immediately initiated after tissue injury. It is a dynamic and multistage biological response comprising four overlapping phases: hemostasis (immediate), inflammation (days 1–4), proliferation (days 4–21), and remodeling or maturation (day 21 to up to 2 years).
In the initial phase, characterized by loss of lymphatic fluid and/or blood, hemostasis is crucial, leading to the formation of a platelet plug and eventually a stable clot. These also lead to initiation of the inflammatory reaction triggered by serotonin and histamine released from thrombocytes and local leukocytes. Inside the fibrin network there are scattered neutrophils and leukocytes that decontaminate the wound and phagocytose bacteria and cellular debris. The wound contracts due to increased fibroblast and myofibroblast activity. By days five to seven, fibroblasts start collagen and glycosaminoglycans synthesis to form the core of the granulation tissue. Migrating cells, mainly keratinocytes from the outskirts of the plaque, move towards the wound and start re-epithelialization of the wound surface. This is sustained by the ongoing angiogenesis from the existing vessel network, where the endothelial cells proliferate and start migrating. Their proliferation leads to new vascular sprouts that replace and heal the wounded vascular network. However, the scar tissue does not bear the same strength as the initial tissue. In the maturation phase, the newly formed scar tissue is remodeled to reduce thickness and increase elasticity and strength as a response to mechanical stress. Also, pigmentogenesis is activated due to melanocyte activity and migration, particularly as a response to inflammation and ultraviolet exposure [2,3].
This intricate process can be disrupted by various physiological factors such as aging and infection or by pre-existing medical conditions like diabetes and obesity. This may lead to delayed healing or development of chronic wounds, often accompanied by complications including severe inflammation, infection, hemorrhage, and, in extreme cases, limb amputation [4,5].
To support optimal healing, appropriate wound management is essential, and the choice of dressing plays a central role. An ideal wound dressing should maintain a moist environment to promote cell migration and tissue regeneration, manage exudate to prevent maceration, permit gas exchange, and provide a barrier against microbial contamination. Furthermore, dressing must be sterile, non-toxic, nonallergenic, and comfortable, while also being easy to apply and remove without causing further tissue damage [6]. Traditional dressings such as gauze and cotton pads serve primarily as passive barriers. They often fail to address the complex physiological requirements of healing, especially in chronic or infected wounds [7,8]. Modern wound dressings such as semi-permeable films and foams, hydrocolloids, and hydrogels [9] include interactive and bioactive materials designed to actively participate in the healing process [10]. These advanced formulations aim to control infection, modulate inflammation, promote cell migration, and deliver therapeutic agents directly to the wound site [11,12,13].
Despite their promising properties and biological effects, evaluating the optimal hydrogel formulation for wound healing is challenging due to the complexity of both the hydrogels themselves and the diverse types of wounds. Hydrogels need to perform multiple functions—such as aiding debridement, maintaining moisture, promoting cell proliferation, enhancing drug delivery, and stimulating epithelialization—throughout different stages of healing. Additionally, the wide variety of wound types—acute, chronic, infected, burn, ischemic, superficial, or deep—further complicates the selection of an appropriate testing model.
Numerous models are used to assess wound healing, ranging from in vitro systems like 2D skin cultures to more complex 3D reconstructed skin models and ex vivo human or porcine skin. Common in vivo models include surgical wounds with or without induced diabetes mellitus, burn wounds of varying depth, and, less commonly, ischemic or radiation-induced wounds on different animal species, predominantly rats and mice.
The sheer variety of models, each with advantages and disadvantages linked to complexity, costs, technical requirements, standardization issues, ethical concerns, particularly for animal and human studies, and the difficulties of clinical translation of the obtained data make it difficult to select and standardize a testing method that allows for consistent comparisons across studies.
In view of these facts, the manuscript aims to summarize current hydrogel types used in wound healing and to present an overview of the biological efficacy of various hydrogels evaluated through in vitro, ex vivo, in vivo, and clinical models. It also highlights the advantages and limitations of each approach, aiming to support more informed and effective model selection for testing hydrogel-based wound healing therapies.

2. Relevant Sections and Discussion

2.1. Hydrogels

Among the various types of wound dressings, hydrogels stand out due to their superior moisture-retention capacity, biodegradability, biocompatibility, and ability to facilitate natural debridement processes. Hydrogels are three-dimensional, hydrophilic, porous polymeric networks that exhibit high water retention capacity. They received significant attention in topical wound therapy due to their ability to mimic the native extracellular matrix (ECM), maintain a moist wound environment, and deliver bioactive agents in a controlled manner. Their structure, formed through either physical (reversible ionic or hydrogen bonds) or chemical (covalent) crosslinking, can be engineered to respond to various stimuli such as pH [14,15], temperature [16,17], light [18], chemical triggers [19], or enzymatic activity [20,21], which allows dynamic interaction with the wound microenvironment [22,23,24]. They can absorb exudate, promote oxygen diffusion, soften necrotic tissue, and provide a soothing, non-adhesive interface that enhances patient comfort and compliance. Hydrogels can replicate both the physical structure and biological functions of the ECM, supporting cell adhesion, signaling, and matrix deposition, all critical for tissue regeneration [25,26]. Hydrogels, particularly those based on biopolymers such as hyaluronic acid [27,28], collagen [29,30], alginate [31,32], or synthetic polymers like polyvinyl alcohol (PVA) [33], have emerged as promising tools in skin regeneration due to their reduced toxicity and manufacturing variability. They can act as dermal substitutes that prevent scarring by mimicking the mechanical properties of healthy skin. Also, they develop an optimal microclimate between the wound bed and the dressing [34]. Therefore, hydrogels play a fundamental role in advanced wound care strategies, offering both functional adaptability and therapeutic efficacy across a wide range of wound types.

2.2. Hydrogel Classification in the Context of Topical Wound Therapy

Hydrogels can be systematically classified according to several key parameters: origin or source (natural, synthetic, or semi-synthetic), crosslinking mechanism (physical or chemical), responsiveness to external stimuli (such as pH, temperature, or enzymatic activity in the case of stimuli-sensitive hydrogels), and therapeutic agent delivered. This classification approach facilitates a comprehensive understanding of the functional properties of hydrogels and supports the rational selection of suitable hydrogel systems for specific wound types and experimental biological models.

2.2.1. Origin of Polymeric Scaffold

Based on the origin of their polymeric scaffold, hydrogels can be classified into natural, synthetic, and semi-synthetic [35,36].
Natural hydrogels are derived from biopolymers obtained from plant, animal, or microbial sources. Biopolymers commonly used for hydrogel fabrication include polysaccharides such as cellulose [37,38], alginate [39,40,41], chitosan [42,43,44], and hyaluronic acid [45,46,47], as well as proteins like collagen [48,49,50], gelatin [51,52,53], and fibrin [54].
Due to their structural similarity to native soft tissues and their bioactivity, natural hydrogels can support cellular adhesion, proliferation, and differentiation. Moreover, they exhibit excellent biocompatibility, biodegradability, and low toxicity, making them suitable for biomedical applications such as wound dressings, drug delivery systems, and tissue engineering scaffolds [55]. However, due to their heterogeneous molecular structures and limited molecular tunability, natural polymer-based hydrogels typically exhibit weaker and less stable cross-linked networks. This results in lower tensile strength, elasticity, and overall mechanical performance [56,57].
Synthetic hydrogels are three-dimensional polymer networks formed through the covalent or ionic crosslinking of hydrophilic homopolymers or copolymers. They offer precise control over molecular architecture due to a well-defined structure, which can be adjusted to enhance their properties. This allows tailored mechanical, chemical, and degradation properties for specific biomedical or industrial requirements [58]. Commonly used synthetic polymers include polyethylene glycol (PEG) [59,60], polyvinyl alcohol (PVA) [61,62], polyacrylamide (PAAm) [63,64], poly(hydroxyethyl methacrylate) (PHEMA) [65,66], and polylactic-co-glycolic acid (PLGA) [67,68].
Semi-synthetic hydrogels are engineered by combining natural and synthetic polymers. The aim is to integrate the biocompatibility and biological activity of natural materials with the mechanical strength, tunability, and structural stability of synthetic polymers. This leads to advanced biomaterials with enhanced mechanical performance, controlled degradation profiles, and superior biological functionality for applications in regenerative medicine, drug delivery, and tissue engineering [69,70].

2.2.2. Cross-Linking Mechanism

Natural and synthetic polymers contain various hydrophilic functional groups, such as –COOH, –OH, –NH2, –CONH, –CONH2, and –SO3H, that facilitate the formation of three-dimensional hydrogel networks through either physical interactions or chemical crosslinking. Based on these crosslinking mechanisms, hydrogels are broadly categorized into two types: chemically and physically crosslinked hydrogels. Chemically crosslinked hydrogels are characterized by stable covalent bonds between polymer chains. Physically crosslinked hydrogels are formed through reversible interactions such as hydrogen bonding, ionic interactions, hydrophobic associations, and other non-covalent intermolecular forces [71].
Chemically crosslinked hydrogels exhibit robust three-dimensional networks with enhanced mechanical strength, elasticity, structural integrity, and prolonged degradation profiles. A variety of strategies can be employed to achieve covalent crosslinking. Common approaches include reactions between complementary functional groups (e.g., –COOH, –OH, –NH2), such as amide bond formation via carbodiimide chemistry [72,73], isocyanate-based addition reactions [74,75], and Schiff base reactions involving aldehydes and amines [76,77,78]. Typical crosslinkers used include glutaraldehyde [79,80], epichlorohydrin [81,82], and divinyl sulfone [83,84]. Free radical polymerization is another widely used method, where monomers bearing unsaturated double bonds undergo rapid polymerization initiated by thermal, UV, or redox initiators, allowing fast network formation under mild conditions [85,86,87]. Condensation reactions, such as those forming polyesters or polyamides, are another approach to hydrogel formation, particularly when using carboxylic acid and amine or hydroxyl groups [88,89]. Additionally, enzymatic crosslinking—such as transglutaminase-catalyzed amide bond formation—offers a biofriendly alternative, suitable for in situ applications [90,91].
Physically crosslinked hydrogels are formed through inter- and/or intramolecular non-covalent interactions between polymer chains, such as hydrogen bonding, ionic interactions, hydrophobic associations, van der Waals forces, crystallization, and protein-based interactions [92,93]. These reversible interactions allow the formation of three-dimensional networks without the need for chemical crosslinkers, making them especially attractive for biomedical applications due to their biocompatibility, injectability, and absence of toxic residues. For example, hydrogen bonds form between complementary functional groups like hydroxyl, carboxyl, and amino moieties; ionic interactions occur between oppositely charged polymers (e.g., alginate with Ca2+); and hydrophobic interactions drive self-assembly in amphiphilic block copolymers [92,94,95]. Additionally, crystallization (e.g., freeze–thaw treatment of PVA) [96,97] and protein–protein interactions (e.g., in gelatin or silk fibroin) [98,99] can establish physically crosslinked networks. While these hydrogels offer stimuli-responsiveness and self-healing capabilities, they often exhibit lower mechanical strength and limited stability compared to chemically crosslinked hydrogels.

2.2.3. Stimuli-Responsive Hydrogels

Stimuli-responsive hydrogels can be systematically classified based on the specific external stimuli to which they respond. There is a broad spectrum of physical, chemical, and biological triggers: pH, temperature, light, electric and magnetic fields, and biological signals.
pH-responsive hydrogels, the most common type of stimuli-sensitive hydrogels, swell or shrink in response to environmental pH changes due to ionizable groups like carboxyl or amine moieties, making them particularly valuable in wound healing by enabling targeted drug release and responsive behavior in the acidic microenvironment of infected or inflamed tissues [100].
Temperature-responsive hydrogels, which undergo sol–gel transitions near body temperature, are important in wound healing, as they enable injectable, in situ forming systems that adapt to the wound site. This provides sustained drug release, improves local drug concentration, and minimizes the systemic side effects associated with intravenous or oral administration [101,102,103]. Photo-responsive hydrogels contain light-sensitive groups like o-nitrobenzyl [104], coumarin [105,106], and azobenzene [107,108] that trigger structural changes under specific wavelengths through photoisomerization, photocleavage, or photodimerization [109], enabling precise, light-controlled drug release and dynamic modulation of the wound environment, promoting targeted therapy [110]. Electro- and magneto-responsive hydrogels [111,112] react to electrical or magnetic fields, enabling controlled deformation or drug release. Glucose- and enzyme-responsive hydrogels [113,114] respond to biochemical triggers, making them ideal for targeted therapies.

2.2.4. Type of Therapeutic Agent Delivered

Hydrogels have attracted significant interest as versatile carriers in targeted therapeutic delivery, bioadhesive systems, and controlled release platforms. Due to their inherent biocompatibility, moisture retention, and structural similarity to the extracellular matrix, hydrogels, in particular polysaccharide-based, are highly effective for wound healing applications, even in carrier-free formulations. They support tissue regeneration and maintain an optimal healing environment with no need for additional therapeutic agents [115,116]. Hydrogel-based wound dressings have been extensively explored for the delivery of therapeutics such as antimicrobials, growth factors, nucleic acids, metal nanoparticles, and exosomes. These systems enhance drug stability in the intracellular environment and prolong therapeutic activity at the wound site, even for periods exceeding 10 days.
Growth factors and peptides promote cell proliferation and angiogenesis but suffer from rapid degradation, while hydrogels protect and sustain their release [117,118,119]. Nucleic acid therapeutics undergo enzymatic breakdown and exhibit poor uptake, yet hydrogels improve their stability and transfection efficiency [120,121]. Exosomes boost immune modulation and regeneration; hydrogel incorporation extends their retention and therapeutic effect [122,123]. Antimicrobials are essential for infection control but often suffer from low bioavailability, burst release, and potential toxicity. Incorporation into hydrogels enhances their bioavailability through localized, sustained release. It minimizes systemic exposure and reduces side effects and resistance [124]. Metallic and non-metallic nanoparticles offer antimicrobial, antioxidant, and healing benefits, while hydrogels ensure their even distribution and prolonged activity, addressing complex wound care challenges [125,126].
Based on these facts, the use of hydrogels in wound healing care, regenerative medicine, or drug delivery presents clear advantages. However, their biocompatibility, biological activity, or efficacy strongly depends on the hydrogel interactions with the biological tissues. Choosing the right model is essential for the testing of the hydrogel properties, since their use might be beneficial in a certain wound type or a phase of the healing process, while it might be detrimental in another step.
For instance, a hydrogel that exerts anti-fibrotic properties, such as alginate, has a negative impact in the proliferation step but can be beneficial in the maturation phase or in chronic wounds when fibroblast overactivation leads to epithelialization inhibition [127]. Alginate is widely used in wound care in forms such as hydrogels, films, nanofibers, microspheres, and topical formulations [128,129,130,131]. It is a biocompatible, linear anionic polysaccharide derived from brown algae (e.g., Sargassum) and some bacteria. Alginate hydrogels can be formed via various crosslinking methods—ionic, covalent, thermal, or cell-mediated—allowing customization of their physical and chemical properties for biomedical applications [132].
In the inflammatory phase of wound healing, a non-absorbent hydrogel, like naturally derived polymers such as collagen or alginate-based formulations, is less useful due to abundant exudate but is beneficial in the proliferation and migration phase, when it provides an ECM-resembling scaffold that diminishes healing time and improves scar formation. Alginate films could also be unsuitable for heavily exuding wounds due to poor thickness [129]. Aloe vera-enhanced alginate films improved mechanical strength and thermal stability without altering chemical properties [133]. Alginate microspheres can be used as delivery systems for drugs, growth factors, and cells [134,135,136], or to enhance the absorbance properties of the polymer while still maintaining moisture in the wound bed [127,131].
From a translational perspective, natural polymers such as alginate, chitosan, hyaluronic acid, collagen, and gelatin are attractive due to their biocompatibility, biodegradability, and intrinsic bioactivity. However, they also exhibit disadvantages such as poor mechanical strength and the risk of immunogenicity, which limit direct clinical translation unless combined with reinforcing networks or antibacterial agents. In contrast, synthetic systems such as polyvinyl alcohol (PVA) hydrogels provide robust mechanical stability, moisture retention, and oxygen permeability. These properties make them suitable for protecting fragile wounds and promoting re-epithelialization, despite their lack of inherent antibacterial activity, which requires functional modification. Several reviews have comprehensively examined the advantages and limitations of both natural and synthetic polymer-based hydrogels for wound healing applications as well as clinically available hydrogel wound dressings [137,138,139].
Hybrid hydrogels bridge these gaps by integrating bioactive natural polymers with mechanically resilient synthetic backbones or responsive moieties, enabling multifunctional dressings that can release antimicrobials, respond dynamically to wound microenvironmental cues, and support cellular proliferation in vivo. Such adaptability, coupled with a high potential for scalable and low-cost manufacturing, positions hybrid and engineered synthetic hydrogels as the most clinically translatable platforms for wound care applications [140].
Hybrid hydrogels containing polyvinyl alcohol (PVA), with chitosan with or without collagen, can be used in all healing phases due to their dual effects of absorbing and moisture preservation, which regulates the humidity of the wound bioenvironment [141]. Nanocomposite sponges using sodium alginate, graphene oxide, and PVA showed high porosity, good mechanical properties, stimulated cell proliferation in vitro, and enhanced wound healing in vivo [142]. The use of hybrid hydrogels in wound healing is still limited by mechanical properties, biocompatibility, and the formation of degradation byproducts, such as acids. For instance, polylactic acid (PLA)-containing gels decrease tissue pH by freeing lactic acid, which is detrimental for cell survival and proliferation [143].
Cellulose also proved beneficial due to its mechanical strength and absorptive ability. Carboxymethyl cellulose and electrospun cellulose extracts with polylactide and polydioxanone nanofibers increased skin regeneration and aided angiogenesis, reviewed in [143].
Polyurethane-based wound dressings, incorporating natural polymers (e.g., collagen, chitosan, hyaluronic acid), synthetic polymers (e.g., PEG, PVA, polyacrylamide), and bioactive agents (e.g., LL37 peptide, platelet lysate, exosomes), have been reviewed for their physicochemical and biological modifications, applications in wound repair and regeneration, and potential for the development of advanced functional dressings [144]. Bilayer dressings combining a dense polyurethane membrane with a polycaprolactone/gelatin scaffold control wound moisture, provide antibacterial activity, and promote cellular adhesion and proliferation, supporting multiple stages of wound healing [145].
Hydrocolloid dressings were prepared from sodium carboxymethyl cellulose or sodium alginate, styrene–isoprene–styrene, and either silk fibroin nanoparticles or petroleum hydrocarbon resin, optionally loaded with bioactive agents. These hybrid hydrocolloids exhibit improved mechanical properties, water uptake, and swelling. They promote dermal regeneration and epithelialization, enhance wound structural integrity, reduce wound size, and increase collagen fiber density [140].
Another issue in wound healing is the presence of bacteria, leading to inflammation and cell death, with delayed or impaired healing eventually leading to the development of chronic wounds or defective scars. Composite hydrogel dressings with carboxymethyl chitosan, gelatin microspheres, and tetracycline hydrochloride showed sustained drug release and effective inhibition of E. coli and S. aureus [146]. Innovative blends, such as silk fibroin/sodium alginate membranes loaded with strontium, promoted angiogenesis and exerted antibacterial effects by disrupting bacterial functions [147].
Nanoparticle incorporation further enhanced antibacterial activity [148]. Mercy et al. synthesized combined hydrogels containing alginate/gelatin hydrogels with nanosilver with or without plant extracts such as aloe vera, curcumin, plantain, and Calendula. The hydrogels showed low cytotoxicity, strong antibacterial activity against E. coli and S. aureus, and enhanced wound closure in vitro on fibroblastic cells (V79). Adding the plant extracts increased efficacy [149].
Microencapsulated plantain extract in alginate promoted antioxidant and anti-inflammatory effects, boosted collagen synthesis in dermal fibroblasts (BJ), and accelerated in vitro wound closure [131]. Alginate microspheres containing white lily extract showed increased in vitro wound healing effects by antioxidant and anti-inflammatory activity and the modulation of matrix metalloproteases, while boosting collagen synthesis on a co-culture model of senescent dermal fibroblasts (BJ) and endothelial cells (HUVEC) exposed to LPS. The model mimicked a chronic, infected wound [127]. Sodium alginate beads, containing resveratrol with or without graphene nanoplatelets, were synthesized and tested for physicochemical properties, resveratrol release, and biocompatibility in human cell culture. The gel beads showed good biocompatibility against dermal fibroblasts (BJ) and endothelial cells (HUVEC) and provided an antioxidant effect by long, pH-dependent resveratrol release [146]. Moringa oleifera leaf (MOL) extract, rich in antimicrobial and anti-inflammatory compounds, was incorporated into polyvinyl alcohol (PVA) and graphene oxide (GO) hydrogels. The PVA/MOL/GO hydrogel exhibited strong antibacterial activity against S. aureus and E. coli and cytocompatibility in 3T3L1 cells and shortened in vitro wound healing, as confirmed by scratch assay [150]. SA-based hydrogels—sulfonated, quaternized, and chlorogenic acid (CGA)-modified—were coated onto Eucommia ulmoides rubber (EUR) films. Biocompatibility was confirmed using L929 fibroblasts. The dressings showed good mechanical strength, water vapor permeability, and controlled drug release. Antibacterial tests against E. coli and S. aureus and an in vivo mouse full-thickness wound model revealed accelerated healing, confirmed by histological and immunohistochemical analyses [151].
There is a growing need for suitable biological models that are specifically designed for the testing of hydrogel properties in the bioenvironment, which would allow the fine-tuning of their physico-chemical structure to the intended use. These models have to mimic the clinical steps of wound healing, depending on the wound types and the behavior of different cell types from the wound area.
In view of the clinical use of hydrogel formulations, their biocompatibility and biological activities have to be thoroughly tested on human cell-derived models, closely mimicking the wound structure. The employed models involve in vitro 2D skin cell cultures and possibly more complex human models, such as 3D reconstructed human epithelium or full-thickness skin models, ex vivo human or porcine explants, or standardized in vivo models created on lab animals, predominantly mice and rats.

2.3. Biological Models Employed in the Evaluation of the Wound Healing Properties of Hydrogels

2.3.1. In Vitro Models

In vitro wound healing models (Table 1 and Table 2) play a vital role in improving our understanding of tissue repair by providing a simplified, controlled environment, enabling systematic testing of therapeutic interventions under reproducible conditions.
A comparative presentation of the advantages/disadvantages, limitations, and research applications of the in vitro models is presented in Table 1 (2D models) and Table 2 (3D models).
Monolayer Cultures
Fibroblast and/or keratinocyte monolayer models are simpler models, easier to manipulate, and provide cost-effective benefits. Cells are cultured cells on flat and rigid surfaces generally made of transparent glass or plastic, so microscopic cellular observation is possible while maintaining cellular attachment [152]. One of its shortcomings is that the monolayer lacks three-dimensional structure and therefore complex cell–cell interactions and ECM dynamics in actual tissues, restricting its physiological interactions and diminishing the possibility for direct clinical transfer. However, the 2D in vitro models (Figure 1) are frequently used for basic research into cellular mechanisms such as preliminary drug screening and toxicity assessments [153], cell migration and basic proliferation studies, and cell signaling pathways in wound repair analysis [152].
An interesting approach to wound healing modeling was the development of a human-relevant in vitro scab model composed of HaCaT keratinocytes and fresh human blood, maintained for over two weeks. The model expressed all TGF-β isoforms and ECM-related target genes, reflecting early wound healing activity. TGF-β1 stimulation promoted matrix remodeling and Smad7 expression, while pathway inhibition led to scab condensation and degranulation. This model provides a viable platform to study early wound healing events involving blood–skin cell interactions [161].
Scratch Wound Assays
The scratch assay is a 2D in vitro technique derived from the monolayer model, wherein a uniform “wound” is introduced by mechanically disrupting a confluent layer of cells, typically epidermal fibroblasts or keratinocytes. Cell migration into the wound area is tracked over time to assess wound closure. Some of the most common uses of this method are in wound healing and cancer assays.
Another wound creation technique comes from the use of inserts, including silicone or bioprinted, to avoid the mechanical trauma inflicted on neighboring cells [154]. The scratch wound assay is valued for its reproducibility and simplicity and can be quantified by imaging software, including available open-source analytical platforms [155]. However, it lacks the structural and functional complexity of the in vivo tissue, offering limited insight into ECM dynamics, dermal-epidermal interactions, and immune responses [152]. The method was also developed for 3D models. Initially a 3D fibrin clot scaffold was used for cell migration; then collagen or other matrices and fibroblast spheroids were employed to test fibroblast and neutrophil migration in the presence of inhibitors or activators [156]. A more precise scratch assay method was developed based on photosensitized culture substrates. Photosensitizers were engulfed into a polystyrene matrix, which was coated in a thin layer on the bottom of the cell culture dish. After the confluent culture was grown on the dish surface, the photosensitizer was activated by irradiation, which produced superoxide anion that killed the cells precisely in the exposed area, leaving a precise wound. The method can be applied to monolayers, 3D cultures, or even microfluidics to generate the wounds [157].
Reconstructed Human Epidermis (RHE), 3D Models
Researchers have developed more advanced models, such as the 3D reconstructed human epidermis (RHE) or full dermal-epidermal reconstructs (FDE), to overcome the 2D tissue architecture limitations [152].
RHE models consist of stratified keratinocytes cultured on a porous membrane at the air-liquid interface, frequently supported by a collagen matrix to mimic the basal membrane. Full dermal-epidermal reconstructions also include the reconstructed dermis by using human fibroblasts for the dermal part, on which the keratinocytes are further stratified to make the upper epidermis. These models allow for a multilayered epithelium with structural and functional similarities to native human skin, including barrier function and ECM deposition [152]. The air-liquid interface promotes keratinocyte differentiation and stratification, resulting in a tissue architecture more suitable to represent in vivo conditions. As a general use, these models allow research focused on tissue engineering and regenerative medicine, drug efficacy testing, or studies involving complex cell behaviors within a physiologically relevant environment. RHE and FDE models have high immediacy in studies focused on epidermal development, barrier stability, and preclinical testing of topical agents [162]. However, these benefits come with increased complexity, higher expenses, and the need for specialized equipment and expertise (Table 2 and Table 3). Additionally, their viability is limited to shorter timeframes, and analytical procedures may be more challenging due to their 3D nature [163].
An in vitro thermal wound infection model was developed using human skin equivalents (HSE) and biofilm-forming MRSA to evaluate antimicrobial agents. Thermal injury, induced by a liquid nitrogen-cooled device, caused keratinocyte death and epidermal detachment, triggering inflammation and antimicrobial responses. MRSA exposure led to rapid colonization and upregulation of TLR2, IL-6, IL-8, and defensins. Topical mupirocin reduced MRSA levels by over 99.9% within 24 h. This model effectively mimics early wound infection and is suitable for testing topical antimicrobials [164].
Hydrogels, particularly collagen, are useful tools for the design of the dermal matrix or as a bioink to support the development of the 3D RHE or full-thickness skin models. A full-thickness human skin model (FTM) was developed using chitosan and collagen hydrogel (CC-FTM). The CC-FTMs supported epidermal differentiation, with reduced keratinocyte activation resembling normal human skin. It also induced an improved lipid matrix structure, ceramide profile, and lower transepidermal water loss, which indicated enhanced barrier function. These features make CC-FTMs a more physiologically relevant model for studying epidermal barrier properties [164]. A multicellular wound-on-chip model was developed to mimic the paracrine signaling of early inflammation during wound healing. Using collagen or Matrigel coatings, dermal fibroblasts and HUVECs were cultured in a tri-channel microfluidic system. Inflammation was induced by TNF-α or macrophage triculture, both significantly increasing IL-1β, IL-6, and IL-8 levels, which were reduced by dexamethasone. M2 macrophages enhanced cytokine production and vascular structure formation. This model effectively replicated early wound inflammation [158].
An overview of the wound healing processes in 2D and 3D in vitro models is provided in Table 3.

2.3.2. Ex Vivo Models—Human or Porcine Skin Explants

Ex vivo skin models involve culturing excised human or animal skin to replicate the complex structure and microenvironment of native tissue. They are widely used to study wound healing mechanisms and inflammation, and to assess the biological effects of therapeutic interventions. A key advantage is the preservation of the original skin architecture and cell-to-cell communication. It presents the stratified epithelial organization while also maintaining the epidermis, dermis, and basement membrane layers. Ex vivo models also show communication between fibroblasts, keratinocytes, and extracellular matrix proteins. This allows for more physiologically relevant observations compared to in vitro cultures. However, these models lack vascularization, innervation, and immune cell migration, limiting their ability to accurately replicate the dynamic immune responses seen in vivo, such as the recruitment of circulating leukocytes, cytokine signaling cascades, and the resolution of inflammation. Furthermore, variability in tissue sources and preparation methods may limit reproducibility and long-term applications [159].
In a human skin explant model mimicking superficial wounds via tape-stripping, the topical application of octenidine dihydrochloride-based hydrogel promoted wound healing by lowering inflammatory and proteolytic activity, shown by reduced cytokine and matrix metalloproteinase expression [160]. Similarly, an ex vivo porcine skin model treated with a thyme-oil-loaded PLGA (polylactic-co-glycolic acid) nanoparticle hydrogel increased healing by antioxidant mechanisms [165]. A more complex human skin model simulating deep burn wounds under tension revealed that p407/p188 poloxamer hydrogel slowed epidermal regeneration [166]. In another study using partial-thickness human skin wounds, a polydopamine-based hydrogel with in situ generated silver nanoparticles promoted epidermal regeneration, improved biocompatibility, reduced inflammatory markers, and demonstrated broad-spectrum antimicrobial activity [167].
In an ex vivo porcine skin model treated with adamantane-based collagen mimetic peptide (CMP) hydrogel, the hydrogel showed satisfactory permeation for the peptide, leading to its localization in the epidermis and dermis [168]. Complex alginate hydrogels loaded with curcumin-synthesized cerium oxide nanoparticles (Cur-CeO) showed good biocompatibility to cultivate L929 fibroblasts and antioxidant properties ex vivo. The hydrogel-nanocomposites accelerated circular deep wound repair by reducing oxidative stress and inflammation and increasing keratinocyte and fibroblast migration onto the wound bed to promote faster tissue recovery in rats [169].
In vivo models involve various animal species such as mice, rats, rabbits, and guinea pigs. Common in vivo models include surgical wounds (incisional or excisional), diabetic non-healing wounds (induced via chemical methods), burn wounds of varying depth, and, less commonly, ischemic or radiation-induced wounds.

2.3.3. In Vivo Wound Healing Models

Several models can be employed to create a standardized wound in vivo (Figure 2, Table 4, Table 5 and Table 6). Surgical wounds can be performed as full-thickness excisional wounds or incisional wounds. One of the most used is the full-thickness excisional wound, when a circular or square wound is excised down to the panniculus carnosus (in rodents) or subcutaneous tissue. It is easy to perform, standardize, and monitor throughout the healing phases. Incisional wounds consist of linear cuts, sometimes sutured, to mimic surgical incisions and test healing strength and scar formation. The wound size is usually standardized, e.g., 6–10 mm diameter for mice [170,171].
Another type of wound is the burn wound, which is a thermal injury induced to study scarring, delayed healing, or infection. Special types of models can be created using ischemia or diabetic wounds when the animals, usually mice or rats, have a previously induced diabetes mellitus condition (e.g., streptozocin STZ -induced diabetic mice). These models mimic chronic or non-healing wounds [171].
Surgical Wounds
The in vivo surgical wound model is a widely used technique to study wound healing processes under controlled biological conditions. Surgical wounds can be developed in animal subjects (mice, rats, guinea pigs, rabbits, pigs, or larger animals) and simulate different types of wounds to assess tissue regeneration, epithelialization, healing mechanisms, therapeutic interventions, scarring, and inflammation. Lab animals typically used for these kinds of models are mice and rats due to the low costs, easy handling in the lab, and well-characterized phenotype, including the immune response (Table 4, Figure 2).
The efficacy of different gel formulations containing naturally derived antioxidants [173,175,176,177,183], sodium thiosulfate, a ferroptosis inhibitor [174], sildenafil cream [179], or nanoliposomes containing spirulina-derived peptides [193] on mouse or rat models is presented in Table 4.
Diabetic wounds heal poorly due to peripheric angiopathy and neuropathy-induced changes and chronic inflammation [197,198], (Figure 3). Some studies showed emerging promising therapies that address these specific pathological barriers and promote functional tissue repair by using paeoniflorin-loaded hyaluronic acid (HA-PF) hydrogel [187] or N-acetyl cysteine [186].
In nonhealing surgical wounds, an efficient approach was the local therapy with extracellular vesicles, cellular fractions, and ECM embedded in gel formulations. The advantages are multiple: low immunogenicity, since they contain no intact cells; well-preserved vesicles, which allow transport of the active compounds through the wound bed; and direct cell stimulation by growth factors [178,188,189,192].
Developing an antibacterial hydrogel for the therapy of surgical wounds, particularly efficient against Staphylococcus aureus, a common pathogen found in the wound bed of humans and animals, was attempted by incorporation of ZnO nanoparticles in a double-layer alginate dressing [199], silver nanoparticle-coated alginate nanofibers [200], or lavender oil-infused nanofiber matrices [201].
Another issue that needs to be solved when using hydrogels for local therapy on in vivo animals or humans is their mechanical properties, particularly stiffness, adhesion, and moisture retention characteristics. These properties are required for the initial application of the hydrogel and afterwards for the persistence of the gel on the wound. An interesting approach was the development of an injectable, self-healing hydrogel composite (OGN-7) using oxidized alginate, gelatin, and 5 mg/mL PCL:gelatin nanofibers (7:3 ratio). Dual Schiff-base crosslinking and nanofiber integration enabled in situ gelation without additional crosslinkers. OGN-7 demonstrated enhanced stiffness, faster gelation, excellent remendability, and biocompatibility. It fully fused within 1 h, maintained mechanical integrity under stress, and outperformed standard alginate dressings in a full-thickness wound model. Histological analysis confirmed improved re-epithelialization and collagen deposition [202].
Another approach is the patch application directly on the wound bed because of the adhesive, hydration, drug delivery, and antimicrobial properties of these complex formulations, such as chitosan with doxycycline, zinc, and selenium nanoparticle patches [184], chitosan membranes [185], or silk wraps with epidermal growth factor or silver sulfadiazine [194] (Table 4).
Stimulation of wound bed cell proliferation can be performed by local physical therapies such as laser [190], photodynamic therapy (PDT) [196], electrical current [195] (Figure 2).
Moreover, mutations can be induced in mice to generate adequate models for the study of healing mechanisms. Systemic interleukin-10 administration improved wound healing, reduced inflammation, and enhanced scar quality in KO mice with double deletion of IL-10 and IL-4 genes, proving the important role of IL-10, an anti-inflammatory cytokine, in the normal healing process [191].
The use of rabbits for the generation of surgical wounds is beneficial since it allows a larger wound area, which is more useful for the efficacy and biocompatibility of dressings or biomaterials. In a recent study, a nanoemulsion hydrogel with Hypericum perforatum upregulated growth factors, increased vessel growth, and showed antimicrobial characteristics in rabbits [172]. A similar antibacterial effect was obtained in rat and rabbit models; using Aloe ferox Miller and Aloe arborescens Miller promoted healing, reduced inflammation, and displayed selective antimicrobial activity [181].
Burn Wounds
Burn wounds represent a model mainly used to study the scarring in different conditions in a chronic or nonhealing/infected wound (Figure 3). However, a significant problem with the burn wound model is the standardization, which makes comparison between different therapies difficult. Although each study has its own control group, the burn depth and wound surface are important features that influence the healing process. For instance, a full-thickness burn heals slower and with scarring, due to the lack of hair follicle stem cells, when compared to a second-degree burn. Also, some of the studies do not offer complete data regarding the wound dimensions, such as depth, diameter, closure timeline, and scab formation. When compared to burn wounds, surgical wounds can be standardized, and the follow-up can be performed easier, leading to more comparable results. Despite these issues, the advantages of the burn wounds when following a strict protocol are significant: they can be easily created, the method is quick and reproducible, it is less stressful, and the survival of the animals is very good when compared to a diabetic model with a surgical wound that mimics a similar chronic nonhealing wound.
There are several studies that used this model to evaluate the results of local hydrogel therapies such as chitosan gel with epidermal growth factor (EGF) [203], zinc alginate hydrogel [204], verapamil-loaded hydrogel with EGF and verapamil nanofibers alone [205], and Zein/pectin/Vitamin C scaffold crosslinked to mimic hydrogel [206]. The results are presented in Table 5. All formulations showed improved healing time and led to better scar formation when compared to controls. Adding the epidermal growth factor was beneficial when combined with chitosan but not with verapamil, while the presence of the vitamin C enhanced collagen 1 expression. An interesting approach was the use of Clostridium Perfringens Wound Healing Substance to increase collagen production and capillary formation [207]. The natural antioxidants embedded in ointment [208] or administered as a spray [209] also seemed to improve healing, particularly by anti-inflammatory effects (Table 5).
Table 5. In vivo thermal burn wound models used for experimental research.
Table 5. In vivo thermal burn wound models used for experimental research.
No.Localization/Species Treatment Main Results
1Dorsal area—ratsZinc alginate hydrogelFaster wound healing, increased angiogenesis, and fibroblast migration [204]
2Dorsal area—rats Gel containing verapamil HCl-loaded nanofibers composed with alginate Controlled release of verapamil leading to smooth, scar-free wound closure [205]
3Dorsal area—miceZein/pectin/vitamin C scaffolds crosslinked to mimic hydrogelAntioxidant effects, faster re-epithelialization, anti-inflammatory activity [206]
4Second-degree burn on dorsal area—ratsChitosan gel with epidermal growth factor Increased cell proliferation and epithelialization, improved scar tissue formation [203]
5Dorsal area—ratsCream containing Clostridium Perfringens-derived wound-healing substance Enhanced skin collagen formation and increased capillary formation [207]
6Dorsal area—BALB/c mice Ointment containing Aloe emodin and resveratrol Faster wound healing by increasing blood vessel growth, upregulating IL-1β and VEGF, and stimulating immune cell response [208]
7Second-degree burn on dorsal area—rats Spray formulation of Olea europaea and Aloe vera leaves, Cocus nucifera fruit, and Chamomilla recutita flower plant extracts Antioxidant and anti-inflammatory effects, increased angiogenesis, faster wound healing [209]
Other Wounds
Research on complex wounds (ischemic, irradiated, deep, axial flap) showed improved healing by using topical application of platelet-derived [210] or tooth pulp stem cell vesicles [203], respectively, and local physical therapies such as photodynamic therapy and pulsed therapeutic ultrasound [211], the main findings are presented in Table 6.
Table 6. In vivo ischemic or irradiation wound models used for experimental research.
Table 6. In vivo ischemic or irradiation wound models used for experimental research.
Model/Species Treatment Main Results
1Ischemic wound in rabbitsTA Platelet-derived exosome product incorporated into a surgical fibrin sealant biogelBoosted deep tissue repair by enhancing new vessel growth, structural rebuilding, and skin appendage renewal [210]
2Fasciocutaneous flap on rats exposed to ischemia/revascularization of the vascular pedicleLT photodynamic therapy mediated by Photofrin and 630 nm light Inhibited revascularization in axial flaps, showing reduced blood flow [212]
3Ischemic wound by using the Matrigel plug model on nu/nu mice TA apoptotic extracellular vesicles from tooth pulp stem cells Enhanced angiogenesis by collagen 1 delivery form vesicles and stimulation of the PI3K/AKT/VEGF pathway leading to faster wound healing [205]
4X-ray irradiation on mice LT-pulsed therapeutic ultrasound No beneficial effects on wound closure rate were reported [211]
Angiotensin (1–7) with systemic administration accelerated wound healing in multiple models, including rats, diabetic mice, and guinea pigs—by maintaining flap viability and improving tissue regeneration and growth, without hypertensive effects [180].
Limitations: species differences, ethical concerns
While animal models remain a cornerstone of wound healing research [213], several key limitations affect their translational relevance.
Rodents, commonly used in experimental models, differ significantly from humans in skin architecture, immune responses, and wound healing mechanisms [214]. For example, they primarily repair wounds through contraction, unlike the re-epithelialization process predominant in human skin, which limits the applicability of findings to clinical settings [215]. Also, the presence of fur increases the number of skin hair follicles, the source of the follicular stem cells, leading to quicker healing, limited inflammation, and less scarring. These properties also make it difficult to generate a standardized chronic nonhealing wound in a rodent model.
In addition, ethical concerns regarding animal use require strict regulatory oversight and justification of experimental design, in line with the 3Rs principle—Replacement, Reduction, and Refinement [216].
Model selection must also account for practical considerations such as complexity, time, cost, and the researcher’s expertise. Ethical concerns, especially with in vivo animal models and human tissue use, are significant factors as well.
Another major challenge lies in translating results from laboratory models to clinical practice. Data from in vitro studies often fail to replicate in vivo due to the intricacies of living organisms, including immune responses, hormonal regulation, and blood circulation. Furthermore, outcomes from animal studies may not accurately reflect human biology, leading to differing therapeutic effects in clinical settings compared to preclinical models.

2.4. Hydrogels in Clinical Practice

Hydrogels used in the treatment of different wound types present important characteristics that may help in healing [217]: adherence to the lesion but with slight removal, transparency [218]; hydration or siccative network [219]; stimulators of keratinocyte and fibroblast proliferation and migration, angiogenesis trigger [220], immune cell modulators [221]; antibacterial properties [222]; non-immunogenic structure [223]; and many other advantages if compared with classical treatments.
Nowadays, skin wounds (burns, diabetic ulcers, venous ulcers, accidents, etc.) can be treated with hydrogels applied on the lesion area as solid sheets, semi-liquid (amorphic) layers [222], foams, or sprays, including different substances that help the healing. Many polysaccharide-based hydrogels were developed with alginate, starch, cellulose, chitosan, hyaluronic acid, and carboxymethyl chitosan, but with low antibacterial activity. The usage of essential oil extracts increased the antibacterial properties of natural hydrogels, with the ginger, cumin, or eucalyptus essential oils showing the best antibacterial activities against S. aureus and E. coli, compared to other studied natural oils [224]. For antibacterial effects, metallic oxides were also introduced in some hydrogel formulas: zinc oxide, titanium dioxide, or nickel oxide [225]. The best results in wound protection were obtained with the antibiotic inclusion, such as ciprofloxacin against S. aureus, E. coli, and Klebsiella pneumoniae [226]; gentamicin against Staphylococcus saprophyticus [227]; ampicillin sodium against G+ and G− bacteria [228]; levofloxacin for the infection with P. aeruginosa or S. aureus [225]; cephalosporins against E. coli [225,229]; and vancomycin as the final choice for antibiotic treatment [230].
Different adjuvants of skin wound healing were introduced in hydrogels, growth factors like platelet-derived growth factor (PDGF) [231], vascular endothelial growth factor (VEGF) [232], or fibroblast growth factor (FGF) [233]. Several hydrogel formulas were developed with collagen or hyaluronic acids, molecules that induce immune reactions in human patients [234].
Nanoparticle-loaded hydrogels (metal nanoparticle hybrid hydrogels) were developed for acute (surgical, burns, etc.) and for chronic (diabetic, dermatological, etc.) wounds, patches that are enriched with silver, gold, or zinc oxide nanoparticles in hydrogel formulations that may help in tissue regeneration through their antibacterial action and drug delivery enhancement [235]. Different combinations were developed, like silver nanoparticles with epigallocatechin gallate that enhance the growth factors expression at the lesion area [236] or natural hydrogels with gold nanoparticles that can stimulate the fibroblast migration and the inflammation decrease [237].
Hybrid nanogels may adjust their characteristics (hydrophobic or hydrophilic enhancements, volume variations) to different body changes (glucose level, tissue or blood pH, temperature, biomarkers, etc.) or to environmental modifications (electrical field, magnetic field, light, etc.), features that make them efficient materials in wound treatment [58,238].
Nanoconfined hydrogels with different nanosheets are developed, with improved properties that permit the reorganization of their structure when damaged, synthetic skin that may be used in quick repairs of human skin wounds [239].
Some hybrid hydrogels are already used in patients, but there are concerns regarding their toxicity, the nanoparticles’ migration from the wound level, and their tissue accumulation (skin, liver, kidneys, etc.) [240].

2.4.1. Hydrogels in Preclinical Studies

The hydrogels’ effects on wounds are investigated in many different animal experimental models. To evaluate their antibacterial activity, in vivo studies used quaternized chitosan–matrigel polyacrylamide [241], chitosan–polyvinyl alcohol [242], collagen-hyaluronic acid [243], agar-fumaric acid-silver nanoparticles [244], poly(lactic-co-glycolic acid)/polyethylene glycol/silver [245], or quaternized chitosan/curcumin/benzaldehyde terminated. The antioxidant activity of these innovative products was also investigated in animal models using gels with different compositions like PGE2/chitosan [246], pearl peptides/selenium-containing block-functionalized PEG/polypropylene glycol [247], or resveratrol/(collagen-laminin-based dermal matrix)/hyaluronic acid-dipalmitoylphosphatidylcholine [248].
Hydrogels able to accelerate the tissue reorganization during the healing process were developed and studied in vivo: hyaluronic acid-paeoniflorin [187], collagen-hyaluronic acid [243], feruloyl-modified peptide/glycol chitosan [249], tannic acid/PVA (polyvinyl alcohol)/PEG (polyethylene glycol)/chitosan carboxylate/hyaluronic acid [250], dopamine-substituted multidomain peptide [251], and chitosan/alginate/vitamin E [252].

2.4.2. Hydrogels in Clinical Trials

The beneficial or noxious effects of hydrogels must also be studied through clinical trials, and among the experimented formulas, the following have to be mentioned: hydrogel/nano-silver-based dressing [253] or Olea europaea leaf extract hydrogel (EHO-85) [254] used for increasing re-epithelialization, hydrogel-urea-papain with efficacy on different lesion types [255], hydrogel enriched with sodium alginate and Vitamins A and E that showed inconclusive results [256], and Triticum vulgare extract-polyhexanide (Fitostimoline®) with good effects on inflammation but without complete healing of lesions [257]. For skin ulcers (diabetic, venous), good results were recorded when collagen with fibroblasts and keratinocytes was used as gauze applied on wounds [258]. A double-blind clinical trial conducted on 316 patients, aged above 75 years, with chronic nonhealing venous insufficiency ulcers and without diabetes mellitus or cardiovascular-associated diseases, showed significant improved responses when human-derived adipose tissue mesenchymal stem cells were used, in comparison with the control group that was treated with saline. The authors hypothesized that the immune response inhibition induced by delayed maturation of dendritic cells and natural killer (NK) cells could be responsible for the improved results of the treatment on these wounds, reviewed in [259]. A recent study, performed in patients with chronic deep wounds, compared the standard treatment with acellular dermal matrix (ADM), applied topically as a paste. The study was conducted for 12 weeks in 86 patients aged above 19 years. Patients with complicated wounds (infections, tunneling), diabetes mellitus, or superficial wounds were excluded. The results showed wound healing in 76.3% of cases in the experimental group, compared to 30.6% in the control group. These outcomes represent a promising therapy option for chronic nonhealing wounds [260].

2.4.3. Approved and Commercialized Gels

Since hydrogels contain different types of polymers, the US FDA (Food and Drug Administration) and MDA (Medical Device Amendments) incorporated these innovative treatments in class I, II, and III of risk for developing diseases related to the drug compounds.
Synthetic Wound Dressings
For dry skin lesions, several synthetic wound dressings are recommended: 3M™ Tegaderm™ hydrogel wound filler (propylene glycol), DermaSyn or Cutimed gel (carbomer 940), Woun’Dres (carbomer, collagen), DermaGauze™ (acrylate polymer), and Simpurity™ Hydrogel (polyethylene oxide, polyvinyl alcohol, acrylate, polyurethane). The management of ulcers and burns may be performed with Suprasorb G (CMC polymer, propylene glycol) or Simpurity™ Hydrogel (polyethylene oxide, polyvinyl alcohol, acrylate, polyurethane), and of radiation skin lesions with AquaDerm™ (2-Acrylamido-2 methyl-1-propanesulfonic acid sodium, propylene glycol, poly(ethylene glycol) dimethacrylate, 2-hydroxy-2-methylpropiophenone).
Natural Wound Dressings
Natural lesion-covering hydrogels are seen as wound dressings that can promote wound healing with fewer adverse reactions. Dry skin wounds may be treated with natural hydrogels like Purilon® Gel (sodium carboxymethylcellulose, calcium alginate) or ActivHeal Hydrogel (alginate), while ulcers and burns are treated with Prontosan wound gel (glycerol, hydroxyethyl cellulose, polyhexamethylene biguanide, undecylenamidopropyl betaine), Algicell Ag with antimicrobial silver alginate (calcium alginate with silver), or SoloSite Gel (sodium carboxymethylcellulose). The exudative lesions can be improved by Hyalofill (hyaluronic acid) when used as a wound dressing [261].

3. Conclusions

Chronic and acute wounds continue to pose major clinical and economic challenges, emphasizing the need for more effective wound care solutions. Hydrogels represent a promising class of wound dressings due to their biocompatibility, adaptability, and capacity to support the healing environment. Emerging hydrogel technologies, such as semisynthetic and stimuli-responsible hydrogels, offer enhanced functionality, including biocompatibility, antimicrobial action, tissue regeneration, drug delivery, and lower local pain. The strategic selection of appropriate in vitro and in vivo models remains critical to evaluating their therapeutic potential. While animal models have historically advanced our understanding of wound healing, their limitations highlight the growing importance of human in vitro skin models for preclinical testing. Continued innovation in hydrogel design and testing platforms holds great promise for improving patient outcomes, shortening healing times, and reducing the burden on healthcare systems.

4. Future Directions

The current research gaps in the development of hydrogels for wound healing are due to two main issues: developing specific wound-type hydrogels and biological evaluation.
In order to develop efficient hydrogels, there is a need for interdisciplinary collaboration between different specialists, such as chemists, physicians, biologists, and medical practitioners, to adapt the gels’ mechanical and chemical properties and biocompatibility to the biorequirements of each step of the healing process or wound type. These interactions will strongly influence five key cellular mechanisms, such as cell–cell communication, growth, migration, mechanic sensing, and tissue elaboration, reviewed in [262]. For instance, a modular hydrogel design, starting from synthetic polymers alone or combined with naturally derived ones to support biocompatibility, involves the specific formulation for a certain wound healing type; thus, it can be readily tested and translated in the clinic [263].
Hybrid hydrogels address these challenges by combining bioactive natural polymers with durable synthetic frameworks or responsive components. This enables the development of multifunctional dressings capable of adapting to the wound microenvironment, releasing antimicrobials, anti-inflammatories, or antioxidants, and promoting cell growth and differentiation. Their versatility, along with scalable potential and cost-effective production, makes hybrid and engineered synthetic hydrogels among the most promising platforms for clinical translation.
Additionally, computational modeling and artificial intelligence could help predict wound healing outcomes and optimize hydrogel formulations. Artificial intelligence (AI) and machine learning (ML) are increasingly recognized as transformative tools in hydrogel and wound dressing research, offering significant improvements over traditional trial-and-error approaches. Key properties of hydrogels, such as swelling behavior, mechanical strength, fluid handling, adhesion, tissue conformability, and biocompatibility, can be predicted with accuracy [264,265]. In wound care, AI-powered image analysis can automatically assess wound characteristics to guide material selection, predict healing outcomes, and personalize treatment strategies [266,267].
Future research in wound healing should prioritize the advancement of more accurate and standardized models that better mimic human physiology. A good assessment of hydrogels would require a standardized testing protocol that involves in vitro, in vivo, and human preclinical trials, eventually leading to clinical translation.
Existent in vitro and in vivo systems often fail to replicate the complexity of human wounds, particularly the immune response, vascularization, and local homeostatic imbalances leading to non-healing wounds. Given the limitations of animal models and increasing ethical concerns, there is a pressing need to adopt alternative testing strategies that reduce reliance on animal testing while still meeting regulatory standards.
Emerging technologies such as full-thickness skin, organ-on-a-chip, microfluidic platforms, and 3D bioprinted skin equivalents offer promise for the creation of dynamic, human-relevant models. These models can simulate critical wound conditions such as hypoxia, infection, and inflammation and enable more predictive testing of the hydrogel performance to facilitate a better understanding of their interaction with human tissues. With the advancing of personalized medicine care, tailored hydrogels, particularly stimuli-responsive types, designed to match a specific wound type and comorbidities (e.g., diabetes, vascular disease), will likely become a key focus. To support this, wound healing models will need to incorporate patient-derived cells and tissue samples to allow for personalized testing.

Author Contributions

Conceptualization, I.B., I.G.G. and D.R.M.; methodology, S.G., I.B. and I.G.G.; software, A.-M.V.; validation, I.B. and I.G.G.; formal analysis I.B., I.G.G. and D.R.M., investigation, C.G., D.D., A.-M.V., A.N.C., I.F., M.Y., L.R.S. and T.S.; data curation, I.B. and I.G.G.; writing—original draft preparation, C.G., D.D., A.-M.V., A.N.C., I.F., M.M.W., M.Y., L.R.S. and T.S.; writing—review and editing, I.B. and I.G.G.; visualization, I.B. and I.G.G.; supervision, I.B.; project administration, I.B.; funding acquisition, I.G.G. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported through the “Nucleu” Program within the National Research Development and Innovation Plan 2022–2027, Romania, carried out with the support of MEC, project no. 27N/03.01.2023, component project code PN 23 24 01 05.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data is contained within the article.

Acknowledgments

During the preparation of this manuscript, the author(s) used ChatGPT, version -4.5 (OpenAI, San Francisco, CA, USA), for the purpose of graphical abstract design. The authors have reviewed and edited the output and take full responsibility for the content of this publication.

Conflicts of Interest

The authors declare no conflicts of interest. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript; or in the decision to publish the results.

References

  1. Yousef, H.; Alhajj, M.; Fakoya, A.O.; Sharma, S. Anatomy, Skin (Integument), Epidermis; StatPearls Publishing: Treasure Island, FL, USA, 2025. [Google Scholar]
  2. Wallace, H.A.; Basehore, B.M.; Zito, P.M. Wound Healing Phases; StatPearls Publishing: Treasure Island, FL, USA, 2025. [Google Scholar]
  3. Yazarlu, O.; Iranshahi, M.; Kashani, H.R.K.; Reshadat, S.; Habtemariam, S.; Iranshahy, M.; Hasanpour, M. Perspective on the Application of Medicinal Plants and Natural Products in Wound Healing: A Mechanistic Review. Pharmacol. Res. 2021, 174, 105841. [Google Scholar] [CrossRef]
  4. Brown, J.A. The Role of Hyaluronic Acid in Wound Healing’s Proliferative Phase. J. Wound Care 2004, 13, 48–51. [Google Scholar] [CrossRef] [PubMed]
  5. van Koppen, C.J.; Hartmann, R.W. Advances in the Treatment of Chronic Wounds: A Patent Review. Expert. Opin. Ther. Pat. 2015, 25, 931–937. [Google Scholar] [CrossRef]
  6. Kus, K.J.B.; Ruiz, E.S. Wound Dressings—A Practical Review. Curr. Dermatol. Rep. 2020, 9, 298–308. [Google Scholar] [CrossRef]
  7. Borda, L.J.; Macquhae, F.E.; Kirsner, R.S. Wound Dressings: A Comprehensive Review. Curr. Dermatol. Rep. 2016, 5, 287–297. [Google Scholar] [CrossRef]
  8. Ferraz, M.P. Wound Dressing Materials: Bridging Material Science and Clinical Practice. Appl. Sci. 2025, 15, 1725. [Google Scholar] [CrossRef]
  9. Brumberg, V.; Astrelina, T.; Malivanova, T.; Samoilov, A. Modern Wound Dressings: Hydrogel Dressings. Biomedicines 2021, 9, 1235. [Google Scholar] [CrossRef] [PubMed]
  10. Moradifar, F.; Sepahdoost, N.; Tavakoli, P.; Mirzapoor, A. Multi-Functional Dressings for Recovery and Screenable Treatment of Wounds: A Review. Heliyon 2025, 11, e41465. [Google Scholar] [CrossRef] [PubMed]
  11. Vowden, K.; Vowden, P. Wound Dressings: Principles and Practice. Surgery 2017, 35, 489–494. [Google Scholar] [CrossRef]
  12. Alberts, A.; Tudorache, D.-I.; Niculescu, A.-G.; Grumezescu, A.M. Advancements in Wound Dressing Materials: Highlighting Recent Progress in Hydrogels, Foams, and Antimicrobial Dressings. Gels 2025, 11, 123. [Google Scholar] [CrossRef]
  13. Boateng, J.S.; Matthews, K.H.; Stevens, H.N.E.; Eccleston, G.M. Wound Healing Dressings and Drug Delivery Systems: A Review. J. Pharm. Sci. 2008, 97, 2892–2923. [Google Scholar] [CrossRef]
  14. Thambi, T.; Jung, J.M.; Lee, D.S. Recent Strategies to Develop PH-Sensitive Injectable Hydrogels. Biomater. Sci. 2023, 11, 1948–1961. [Google Scholar] [CrossRef]
  15. Rizwan, M.; Yahya, R.; Hassan, A.; Yar, M.; Azzahari, A.; Selvanathan, V.; Sonsudin, F.; Abouloula, C. PH Sensitive Hydrogels in Drug Delivery: Brief History, Properties, Swelling, and Release Mechanism, Material Selection and Applications. Polymers 2017, 9, 137. [Google Scholar] [CrossRef] [PubMed]
  16. Boon-in, S.; Theerasilp, M.; Crespy, D. Temperature-Responsive Double-Network Cooling Hydrogels. ACS Appl. Polym. Mater. 2023, 5, 2562–2574. [Google Scholar] [CrossRef]
  17. Guleria, S.; Chopra, L. Manikanika Temperature Responsive Hydrogels for Biomedical Applications. Mater. Today Proc. 2023, 92, 356–363. [Google Scholar] [CrossRef]
  18. Xing, Y.; Zeng, B.; Yang, W. Light Responsive Hydrogels for Controlled Drug Delivery. Front. Bioeng. Biotechnol. 2022, 10, 1075670. [Google Scholar] [CrossRef]
  19. Zhu, J.; Wu, H.; Li, Z.; Xu, X.; Xing, H.; Wang, M.; Jia, H.; Liang, L.; Li, C.; Sun, L.; et al. Responsive Hydrogels Based on Triggered Click Reactions for Liver Cancer. Adv. Mater. 2022, 34, e2201651. [Google Scholar] [CrossRef] [PubMed]
  20. Zhao, Y.; Tan, Y.; Zeng, C.; Pan, W. Ultrafast Enzyme-Responsive Hydrogel for Real-Time Assessment and Treatment Optimization in Infected Wounds. J. Nanobiotechnol. 2025, 23, 9. [Google Scholar] [CrossRef]
  21. Sobczak, M. Enzyme-Responsive Hydrogels as Potential Drug Delivery Systems—State of Knowledge and Future Prospects. Int. J. Mol. Sci. 2022, 23, 4421. [Google Scholar] [CrossRef]
  22. Li, Z.; Zhou, Y.; Li, T.; Zhang, J.; Tian, H. Stimuli-responsive Hydrogels: Fabrication and Biomedical Applications. VIEW 2022, 3, 20200112. [Google Scholar] [CrossRef]
  23. Protsak, I.S.; Morozov, Y.M. Fundamentals and Advances in Stimuli-Responsive Hydrogels and Their Applications: A Review. Gels 2025, 11, 30. [Google Scholar] [CrossRef]
  24. Neumann, M.; di Marco, G.; Iudin, D.; Viola, M.; van Nostrum, C.F.; van Ravensteijn, B.G.P.; Vermonden, T. Stimuli-Responsive Hydrogels: The Dynamic Smart Biomaterials of Tomorrow. Macromolecules 2023, 56, 8377–8392. [Google Scholar] [CrossRef]
  25. Qiao, L.; Liang, Y.; Chen, J.; Huang, Y.; Alsareii, S.A.; Alamri, A.M.; Harraz, F.A.; Guo, B. Antibacterial Conductive Self-Healing Hydrogel Wound Dressing with Dual Dynamic Bonds Promotes Infected Wound Healing. Bioact. Mater. 2023, 30, 129–141. [Google Scholar] [CrossRef] [PubMed]
  26. Kang, J.I.; Park, K.M. Advances in Gelatin-Based Hydrogels for Wound Management. J. Mater. Chem. B 2021, 9, 1503–1520. [Google Scholar] [CrossRef]
  27. Yang, X.; Wang, B.; Peng, D.; Nie, X.; Wang, J.; Yu, C.-Y.; Wei, H. Hyaluronic Acid-Based Injectable Hydrogels for Wound Dressing and Localized Tumor Therapy: A Review. Adv. Nanobiomed Res. 2022, 2, 2200124. [Google Scholar] [CrossRef]
  28. Wu, L.; Zhou, Y.; Zhang, Y.; Hu, J.; Ikegami, Y.; Aishima, S.; Ijima, H. Fast Wound Healing with a New Functional Hyaluronic Acid Dual Network Hydrogel. Gels 2025, 11, 266. [Google Scholar] [CrossRef]
  29. Gu, H.; Li, H.; Wei, L.; Lu, J.; Wei, Q. Collagen-Based Injectable and Self-Healing Hydrogel with Multifunction for Regenerative Repairment of Infected Wounds. Regen. Biomater. 2023, 10, rbad018. [Google Scholar] [CrossRef]
  30. Zhang, Y.; Wang, Y.; Li, Y.; Yang, Y.; Jin, M.; Lin, X.; Zhuang, Z.; Guo, K.; Zhang, T.; Tan, W. Application of Collagen-Based Hydrogel in Skin Wound Healing. Gels 2023, 9, 185. [Google Scholar] [CrossRef]
  31. El-Sayed, N.S.; Helmy, N.M.; Kamel, S. Dual-Adhesive and Self-Healing Alginate-Based Hydrogel for Wound Healing. Chem. Pap. 2024, 78, 1021–1031. [Google Scholar] [CrossRef]
  32. Ishfaq, B.; Khan, I.U.; Khalid, S.H.; Asghar, S. Design and Evaluation of Sodium Alginate-Based Hydrogel Dressings Containing Betula Utilis Extract for Cutaneous Wound Healing. Front. Bioeng. Biotechnol. 2023, 11, 1042077. [Google Scholar] [CrossRef] [PubMed]
  33. Bahadoran, M.; Shamloo, A.; Nokoorani, Y.D. Development of a Polyvinyl Alcohol/Sodium Alginate Hydrogel-Based Scaffold Incorporating BFGF-Encapsulated Microspheres for Accelerated Wound Healing. Sci. Rep. 2020, 10, 7342. [Google Scholar] [CrossRef]
  34. Miguel, S.P.; Ribeiro, M.P.; Brancal, H.; Coutinho, P.; Correia, I.J. Thermoresponsive Chitosan–Agarose Hydrogel for Skin Regeneration. Carbohydr. Polym. 2014, 111, 366–373. [Google Scholar] [CrossRef] [PubMed]
  35. Ullah, F.; Othman, M.B.H.; Javed, F.; Ahmad, Z.; Akil, H.M. Classification, Processing and Application of Hydrogels: A Review. Mater. Sci. Eng. C 2015, 57, 414–433. [Google Scholar] [CrossRef] [PubMed]
  36. Ghanem, A.M.; Ashour, S.A.; Hussien, R.M. A review on recent advances in hydrogels as drug delivery system. Int. J. Appl. Pharm. 2025, 17, 39–47. [Google Scholar] [CrossRef]
  37. Kundu, R.; Mahada, P.; Chhirang, B.; Das, B. Cellulose Hydrogels: Green and Sustainable Soft Biomaterials. Curr. Res. Green Sustain. Chem. 2022, 5, 100252. [Google Scholar] [CrossRef]
  38. Kushwaha, J.; Singh, R. Cellulose Hydrogel and Its Derivatives: A Review of Application in Heavy Metal Adsorption. Inorg. Chem. Commun. 2023, 152, 110721. [Google Scholar] [CrossRef]
  39. Ren, Y.; Wang, Q.; Xu, W.; Yang, M.; Guo, W.; He, S.; Liu, W. Alginate-Based Hydrogels Mediated Biomedical Applications: A Review. Int. J. Biol. Macromol. 2024, 279, 135019. [Google Scholar] [CrossRef]
  40. Savić Gajić, I.M.; Savić, I.M.; Svirčev, Z. Preparation and Characterization of Alginate Hydrogels with High Water-Retaining Capacity. Polymers 2023, 15, 2592. [Google Scholar] [CrossRef]
  41. Tan, J.; Luo, Y.; Guo, Y.; Zhou, Y.; Liao, X.; Li, D.; Lai, X.; Liu, Y. Development of Alginate-Based Hydrogels: Crosslinking Strategies and Biomedical Applications. Int. J. Biol. Macromol. 2023, 239, 124275. [Google Scholar] [CrossRef]
  42. Mishra, A.; Omoyeni, T.; Singh, P.K.; Anandakumar, S.; Tiwari, A. Trends in Sustainable Chitosan-Based Hydrogel Technology for Circular Biomedical Engineering: A Review. Int. J. Biol. Macromol. 2024, 276, 133823. [Google Scholar] [CrossRef]
  43. Farasati Far, B.; Omrani, M.; Naimi Jamal, M.R.; Javanshir, S. Multi-Responsive Chitosan-Based Hydrogels for Controlled Release of Vincristine. Commun. Chem. 2023, 6, 28. [Google Scholar] [CrossRef]
  44. Hong, F.; Qiu, P.; Wang, Y.; Ren, P.; Liu, J.; Zhao, J.; Gou, D. Chitosan-Based Hydrogels: From Preparation to Applications, a Review. Food Chem. X 2024, 21, 101095. [Google Scholar] [CrossRef]
  45. An, S.; Choi, S.; Min, S.; Cho, S.-W. Hyaluronic Acid-Based Biomimetic Hydrogels for Tissue Engineering and Medical Applications. Biotechnol. Bioprocess Eng. 2021, 26, 503–516. [Google Scholar] [CrossRef]
  46. Gholamali, I.; Vu, T.T.; Jo, S.-H.; Park, S.-H.; Lim, K.T. Exploring the Progress of Hyaluronic Acid Hydrogels: Synthesis, Characteristics, and Wide-Ranging Applications. Materials 2024, 17, 2439. [Google Scholar] [CrossRef] [PubMed]
  47. Grieco, M.; Ursini, O.; Palamà, I.E.; Gigli, G.; Moroni, L.; Cortese, B. HYDRHA: Hydrogels of Hyaluronic Acid. New Biomedical Approaches in Cancer, Neurodegenerative Diseases, and Tissue Engineering. Mater. Today Bio 2022, 17, 100453. [Google Scholar] [CrossRef]
  48. Guan, D.; Li, W.; Zhao, S.; Tao, Z.; Liu, K.; Liu, Q.; Kong, W.; Zhu, S.; Sun, Y. Carboxymethyl Cellulose-Enhanced Recombinant Human Collagen Hydrogel Promotes Hemostasis and Wound Healing. Int. J. Biol. Macromol. 2025, 315, 144458. [Google Scholar] [CrossRef] [PubMed]
  49. Alberts, A.; Bratu, A.G.; Niculescu, A.-G.; Grumezescu, A.M. Collagen-Based Wound Dressings: Innovations, Mechanisms, and Clinical Applications. Gels 2025, 11, 271. [Google Scholar] [CrossRef]
  50. She, J.; Liu, J.; Mu, Y.; Lv, S.; Tong, J.; Liu, L.; He, T.; Wang, J.; Wei, D. Recent Advances in Collagen-Based Hydrogels: Materials, Preparation and Applications. React. Funct. Polym. 2025, 207, 106136. [Google Scholar] [CrossRef]
  51. Xiang, L.; Cui, W. Biomedical Application of Photo-Crosslinked Gelatin Hydrogels. J. Leather Sci. Eng. 2021, 3, 3. [Google Scholar] [CrossRef]
  52. Mohanto, S.; Narayana, S.; Merai, K.P.; Kumar, J.A.; Bhunia, A.; Hani, U.; Al Fatease, A.; Gowda, B.H.J.; Nag, S.; Ahmed, M.G.; et al. Advancements in Gelatin-Based Hydrogel Systems for Biomedical Applications: A State-of-the-Art Review. Int. J. Biol. Macromol. 2023, 253, 127143. [Google Scholar] [CrossRef]
  53. Andreazza, R.; Morales, A.; Pieniz, S.; Labidi, J. Gelatin-Based Hydrogels: Potential Biomaterials for Remediation. Polymers 2023, 15, 1026. [Google Scholar] [CrossRef]
  54. Li, S.; Dan, X.; Chen, H.; Li, T.; Liu, B.; Ju, Y.; Li, Y.; Lei, L.; Fan, X. Developing Fibrin-Based Biomaterials/Scaffolds in Tissue Engineering. Bioact. Mater. 2024, 40, 597–623. [Google Scholar] [CrossRef]
  55. Catoira, M.C.; Fusaro, L.; Di Francesco, D.; Ramella, M.; Boccafoschi, F. Overview of Natural Hydrogels for Regenerative Medicine Applications. J. Mater. Sci. Mater. Med. 2019, 30, 115. [Google Scholar] [CrossRef] [PubMed]
  56. Lin, X.; Zhao, X.; Xu, C.; Wang, L.; Xia, Y. Progress in the Mechanical Enhancement of Hydrogels: Fabrication Strategies and Underlying Mechanisms. J. Polym. Sci. 2022, 60, 2525–2542. [Google Scholar] [CrossRef]
  57. Shukla, A.; Syaifie, P.H.; Rochman, N.T.; Jaya Syaifullah, S.; Jauhar, M.M.; Mardliyati, E. A Recent Study of Natural Hydrogels: Improving Mechanical Properties for Biomedical Applications. Biomed. Mater. 2025, 20, 022010. [Google Scholar] [CrossRef] [PubMed]
  58. Cai, M.-H.; Chen, X.-Y.; Fu, L.-Q.; Du, W.-L.; Yang, X.; Mou, X.-Z.; Hu, P.-Y. Design and Development of Hybrid Hydrogels for Biomedical Applications: Recent Trends in Anticancer Drug Delivery and Tissue Engineering. Front. Bioeng. Biotechnol. 2021, 9, 630943. [Google Scholar] [CrossRef]
  59. Sun, S.; Cui, Y.; Yuan, B.; Dou, M.; Wang, G.; Xu, H.; Wang, J.; Yin, W.; Wu, D.; Peng, C. Drug Delivery Systems Based on Polyethylene Glycol Hydrogels for Enhanced Bone Regeneration. Front. Bioeng. Biotechnol. 2023, 11, 1117647. [Google Scholar] [CrossRef] [PubMed]
  60. Bakaic, E.; Smeets, N.M.B.; Hoare, T. Injectable Hydrogels Based on Poly(Ethylene Glycol) and Derivatives as Functional Biomaterials. RSC Adv. 2015, 5, 35469–35486. [Google Scholar] [CrossRef]
  61. Wu, S.; Hua, M.; Alsaid, Y.; Du, Y.; Ma, Y.; Zhao, Y.; Lo, C.; Wang, C.; Wu, D.; Yao, B.; et al. Poly(Vinyl Alcohol) Hydrogels with Broad-Range Tunable Mechanical Properties via the Hofmeister Effect. Adv. Mater. 2021, 33, 2007829. [Google Scholar] [CrossRef]
  62. Bercea, M. Recent Advances in Poly(Vinyl Alcohol)-Based Hydrogels. Polymers 2024, 16, 2021. [Google Scholar] [CrossRef]
  63. Penkavova, V.; Spalova, A.; Tomas, J.; Tihon, J. Polyacrylamide Hydrogels Prepared by Varying Water Content during Polymerization: Material Characterization, Reswelling Ability, and Aging Resistance. Polym. Eng. Sci. 2022, 62, 901–916. [Google Scholar] [CrossRef]
  64. Cresens, C.; Aytekin, S.; Shaghaghi, B.; Gerrits, L.; Montero-Calle, A.; Barderas, R.; Kouwer, P.; Rocha, S. Synthesis and Mechanical Characterization of Polyacrylamide (PAAm) Hydrogels with Different Stiffnesses for Large-Batch Cell Culture Applications. bioRxiv 2024. [Google Scholar] [CrossRef]
  65. Zare, M.; Bigham, A.; Zare, M.; Luo, H.; Rezvani Ghomi, E.; Ramakrishna, S. PHEMA: An Overview for Biomedical Applications. Int. J. Mol. Sci. 2021, 22, 6376. [Google Scholar] [CrossRef] [PubMed]
  66. Halpenny, G.M.; Steinhardt, R.C.; Okialda, K.A.; Mascharak, P.K. Characterization of PHEMA-Based Hydrogels That Exhibit Light-Induced Bactericidal Effect via Release of NO. J. Mater. Sci. Mater. Med. 2009, 20, 2353–2360. [Google Scholar] [CrossRef] [PubMed]
  67. Visan, A.I.; Negut, I. Development and Applications of PLGA Hydrogels for Sustained Delivery of Therapeutic Agents. Gels 2024, 10, 497. [Google Scholar] [CrossRef] [PubMed]
  68. Rahmani, F.; Atabaki, R.; Behrouzi, S.; Mohamadpour, F.; Kamali, H. The Recent Advancement in the PLGA-Based Thermo-Sensitive Hydrogel for Smart Drug Delivery. Int. J. Pharm. 2023, 631, 122484. [Google Scholar] [CrossRef]
  69. Hipwood, L.; Clegg, J.; Weekes, A.; Davern, J.W.; Dargaville, T.R.; Meinert, C.; Bock, N. Semi-Synthetic Click-Gelatin Hydrogels as Tunable Platforms for 3D Cancer Cell Culture. Gels 2022, 8, 821. [Google Scholar] [CrossRef]
  70. Lambrecht, S.; Gazizova, A.; Kara, S.; Meyer, J.; Jopp, S. Antimicrobial Properties and Biocompatibility of Semi-Synthetic Carbohydrate-Based Ionic Hydrogels. RSC Adv. 2024, 14, 30719–30731. [Google Scholar] [CrossRef]
  71. Alavarse, A.C.; Frachini, E.C.G.; da Silva, R.L.C.G.; Lima, V.H.; Shavandi, A.; Petri, D.F.S. Crosslinkers for Polysaccharides and Proteins: Synthesis Conditions, Mechanisms, and Crosslinking Efficiency, a Review. Int. J. Biol. Macromol. 2022, 202, 558–596. [Google Scholar] [CrossRef]
  72. Khunmanee, S.; Jeong, Y.; Park, H. Crosslinking Method of Hyaluronic-Based Hydrogel for Biomedical Applications. J. Tissue Eng. 2017, 8, 2041731417726464. [Google Scholar] [CrossRef]
  73. Saeedi, M.; Moghbeli, M.R.; Vahidi, O. Chitosan/Glycyrrhizic Acid Hydrogel: Preparation, Characterization, and Its Potential for Controlled Release of Gallic Acid. Int. J. Biol. Macromol. 2023, 231, 123197. [Google Scholar] [CrossRef]
  74. de Siqueira, E.C.; de França, J.A.A.; de Souza, R.F.M.; Leoterio, D.M.d.S.; Cordeiro, J.N.; Doboszewski, B. Mecanisms of the Chemical Crosslinking to Obtain the Hydrogels: Synthesis, Conditions of Crosslinking and Biopharmaceutical Applications. Res. Soc. Dev. 2023, 12, e18312943072. [Google Scholar] [CrossRef]
  75. Cansu Tarakci, E.; Nihal Gevrek, T. Isocyanate Group Containing Reactive Hydrogels: Facile Synthesis and Efficient Biofunctionalization. Eur. Polym. J. 2022, 175, 111338. [Google Scholar] [CrossRef]
  76. You, Y.; Xie, Y.; Jiang, Z. Injectable and Biocompatible Chitosan-Alginic Acid Hydrogels. Biomed. Mater. 2019, 14, 025010. [Google Scholar] [CrossRef]
  77. Pappalardo, R.; Boffito, M.; Cassino, C.; Caccamo, V.; Chiono, V.; Ciardelli, G. Schiff-Base Cross-Linked Hydrogels Based on Properly Synthesized Poly(Ether Urethane)s as Potential Drug Delivery Vehicles in the Biomedical Field: Design and Characterization. ACS Omega 2024, 9, 45774–45788. [Google Scholar] [CrossRef] [PubMed]
  78. Hegazy, H.A.; Moon, H.H.; Lee, D.-H.; Bhang, S.H.; Kim, Y.-C.; Song, C.; Kim, J.-H. Preparation of Polyaspartamide-Based Adhesive Hydrogels via Schiff Base Reaction with Aldehyde-Functionalized Dextran. Mater. Adv. 2023, 4, 1989–1997. [Google Scholar] [CrossRef]
  79. Buhus, G.; Popa, M.; Desbrieres, J. Hydrogels Based on Carboxymethylcellulose and Gelatin for Inclusion and Release of Chloramphenicol. J. Bioact. Compat. Polym. 2009, 24, 525–545. [Google Scholar] [CrossRef]
  80. Akakuru, O.U.; Isiuku, B.O. Chitosan Hydrogels and Their Glutaraldehyde-Crosslinked Counterparts as Potential Drug Release and Tissue Engineering Systems—Synthesis, Characterization, Swelling Kinetics and Mechanism. J. Phys. Chem. Biophys. 2017, 7, 1–7. [Google Scholar] [CrossRef]
  81. Demirbilek, C.; Dinç, C.Ö. Diethylaminoethyl Dextran/Epichlorohydrin (DEAE-D/ECH) Hydrogel as Adsorbent for Murexide. Desalination Water Treat. 2016, 57, 6884–6893. [Google Scholar] [CrossRef]
  82. Sarmah, D.; Borah, M.; Mandal, M.; Karak, N. Swelling Induced Mechanically Tough Starch–Agar Based Hydrogel as a Control Release Drug Vehicle for Wound Dressing Applications. J. Mater. Chem. B 2023, 11, 2927–2936. [Google Scholar] [CrossRef]
  83. Andrade del Olmo, J.; Alonso, J.M.; Sáez Martínez, V.; Ruiz-Rubio, L.; Pérez González, R.; Vilas-Vilela, J.L.; Pérez-Álvarez, L. Biocompatible Hyaluronic Acid-Divinyl Sulfone Injectable Hydrogels for Sustained Drug Release with Enhanced Antibacterial Properties against Staphylococcus Aureus. Mater. Sci. Eng. C 2021, 125, 112102. [Google Scholar] [CrossRef]
  84. Gomez, I.; Alesanco, Y.; Blázquez, J.A.; Viñuales, A.; Colmenares, L.C. Room-Temperature Self-Standing Cellulose-Based Hydrogel Electrolytes for Electrochemical Devices. Polymers 2020, 12, 2686. [Google Scholar] [CrossRef]
  85. Irvine, G.; Dawson, F.; George, A.; Kopeć, M. Are Polymer Gels Synthesized by Free Radical Polymerization with Cleavable Crosslinkers Really Degradable? Eur. Polym. J. 2024, 213, 113089. [Google Scholar] [CrossRef]
  86. Tran, H.D.N.; Park, K.D.; Ching, Y.C.; Huynh, C.; Nguyen, D.H. A Comprehensive Review on Polymeric Hydrogel and Its Composite: Matrices of Choice for Bone and Cartilage Tissue Engineering. J. Ind. Eng. Chem. 2020, 89, 58–82. [Google Scholar] [CrossRef]
  87. Shantha, K.L.; Harding, D.R.K. Synthesis and Evaluation of Sucrose-containing Polymeric Hydrogels for Oral Drug Delivery. J. Appl. Polym. Sci. 2002, 84, 2597–2604. [Google Scholar] [CrossRef]
  88. de Nooy, A.E.J.; Capitani, D.; Masci, G.; Crescenzi, V. Ionic Polysaccharide Hydrogels via the Passerini and Ugi Multicomponent Condensations: Synthesis, Behavior and Solid-State NMR Characterization. Biomacromolecules 2000, 1, 259–267. [Google Scholar] [CrossRef] [PubMed]
  89. Zhang, R.; Tang, M.; Bowyer, A.; Eisenthal, R.; Hubble, J. A Novel PH- and Ionic-Strength-Sensitive Carboxy Methyl Dextran Hydrogel. Biomaterials 2005, 26, 4677–4683. [Google Scholar] [CrossRef]
  90. Cai, X.; Hu, S.; Yu, B.; Cai, Y.; Yang, J.; Li, F.; Zheng, Y.; Shi, X. Transglutaminase-Catalyzed Preparation of Crosslinked Carboxymethyl Chitosan/Carboxymethyl Cellulose/Collagen Composite Membrane for Postsurgical Peritoneal Adhesion Prevention. Carbohydr. Polym. 2018, 201, 201–210. [Google Scholar] [CrossRef] [PubMed]
  91. Lai, E.; Bao, B.; Zhu, Y.; Lin, H. Transglutaminase-Catalyzed Bottom-Up Synthesis of Polymer Hydrogel. Front. Bioeng. Biotechnol. 2022, 10, 824747. [Google Scholar] [CrossRef]
  92. Priya, A.S.; Premanand, R.; Ragupathi, I.; Bhaviripudi, V.R.; Aepuru, R.; Kannan, K.; Shanmugaraj, K. Comprehensive Review of Hydrogel Synthesis, Characterization, and Emerging Applications. J. Compos. Sci. 2024, 8, 457. [Google Scholar] [CrossRef]
  93. Zhang, P.; Qi, J.; Zhang, R.; Zhao, Y.; Yan, J.; Gong, Y.; Liu, X.; Zhang, B.; Wu, X.; Wu, X.; et al. Recent Advances in Composite Hydrogels: Synthesis, Classification, and Application in the Treatment of Bone Defects. Biomater. Sci. 2024, 12, 308–329. [Google Scholar] [CrossRef]
  94. Pita-López, M.L.; Fletes-Vargas, G.; Espinosa-Andrews, H.; Rodríguez-Rodríguez, R. Physically Cross-Linked Chitosan-Based Hydrogels for Tissue Engineering Applications: A State-of-the-Art Review. Eur. Polym. J. 2021, 145, 110176. [Google Scholar] [CrossRef]
  95. Taaca, K.L.M.; Prieto, E.I.; Vasquez, M.R. Current Trends in Biomedical Hydrogels: From Traditional Crosslinking to Plasma-Assisted Synthesis. Polymers 2022, 14, 2560. [Google Scholar] [CrossRef]
  96. Chen, Y.; Li, J.; Lu, J.; Ding, M.; Chen, Y. Synthesis and Properties of Poly(Vinyl Alcohol) Hydrogels with High Strength and Toughness. Polym. Test. 2022, 108, 107516. [Google Scholar] [CrossRef]
  97. Hu, W.; Wang, Z.; Xiao, Y.; Zhang, S.; Wang, J. Advances in Crosslinking Strategies of Biomedical Hydrogels. Biomater. Sci. 2019, 7, 843–855. [Google Scholar] [CrossRef]
  98. Li, D.; Liang, R.; Wang, Y.; Zhou, Y.; Cai, W. Preparation of Silk Fibroin-derived Hydrogels and Applications in Skin Regeneration. Health Sci. Rep. 2024, 7, e2295. [Google Scholar] [CrossRef]
  99. Madappura, A.P.; Madduri, S. A Comprehensive Review of Silk-Fibroin Hydrogels for Cell and Drug Delivery Applications in Tissue Engineering and Regenerative Medicine. Comput. Struct. Biotechnol. J. 2023, 21, 4868–4886. [Google Scholar] [CrossRef]
  100. Liaqat, H.; Badshah, S.F.; Minhas, M.U.; Barkat, K.; Khan, S.A.; Hussain, M.D.; Kazi, M. PH-Sensitive Hydrogels Fabricated with Hyaluronic Acid as a Polymer for Site-Specific Delivery of Mesalamine. ACS Omega 2024, 9, 28827–28840. [Google Scholar] [CrossRef]
  101. Wang, Y.; He, C.; Chen, C.; Dong, W.; Yang, X.; Wu, Y.; Kong, Q.; Yan, B. Thermoresponsive Self-Healing Zwitterionic Hydrogel as an In Situ Gelling Wound Dressing for Rapid Wound Healing. ACS Appl. Mater. Interfaces 2022, 14, 55342–55353. [Google Scholar] [CrossRef] [PubMed]
  102. Wu, J.; Wang, W.; Shen, J.; Zhou, N.; Li, Y.; Tang, B.Z.; Zhang, M. A Thermosensitive Hydrogel with Efficient NIR Photothermal Conversion as Injectable Wound Dressing for Accelerating Skin Wound Healing. Adv. Funct. Mater. 2024, 34, 2312374. [Google Scholar] [CrossRef]
  103. Chen, H.; Xu, J.; Sun, J.; Jiang, Y.; Zheng, W.; Hu, W.; Qian, H. Recent Advances on Thermosensitive Hydrogels-Mediated Precision Therapy. Asian J. Pharm. Sci. 2024, 19, 100911. [Google Scholar] [CrossRef]
  104. Yang, Y.; Zhu, W.; Dong, Z.; Chao, Y.; Xu, L.; Chen, M.; Liu, Z. 1D Coordination Polymer Nanofibers for Low-Temperature Photothermal Therapy. Adv. Mater. 2017, 29, 1703588. [Google Scholar] [CrossRef]
  105. Ye, Y.; Wang, W.; Liu, X.; Chen, Y.; Tian, S.; Fu, P. A Sol–Gel Transition and Self-Healing Hydrogel Triggered via Photodimerization of Coumarin. Gels 2023, 10, 21. [Google Scholar] [CrossRef]
  106. Qiu, W.; Gehre, C.; Nepomuceno, J.P.; Bao, Y.; Li, Z.; Müller, R.; Qin, X. Coumarin-Based Photodegradable Hydrogels Enable Two-Photon Subtractive Biofabrication at 300 Mm s−1. Angew. Chem. Int. Ed. 2024, 63, e202404599. [Google Scholar] [CrossRef] [PubMed]
  107. Wang, J.; Bian, Y.; Bai, J.; Gao, L. Visible-Light Responsive Hydrogel Based on Methoxy Azobenzene Amphiphilic Small Molecule. J. Photochem. Photobiol. A Chem. 2024, 457, 115893. [Google Scholar] [CrossRef]
  108. Homma, K.; Chang, A.C.; Yamamoto, S.; Tamate, R.; Ueki, T.; Nakanishi, J. Design of Azobenzene-Bearing Hydrogel with Photoswitchable Mechanics Driven by Photo-Induced Phase Transition for in Vitro Disease Modeling. Acta Biomater. 2021, 132, 103–113. [Google Scholar] [CrossRef]
  109. Tomatsu, I.; Peng, K.; Kros, A. Photoresponsive Hydrogels for Biomedical Applications. Adv. Drug Deliv. Rev. 2011, 63, 1257–1266. [Google Scholar] [CrossRef]
  110. Ge, Z.; Wu, H.; Wu, J.; He, Y.; Tan, R.; Wang, Y.; Xiao, T.; Dong, G.; Zhou, P.; Xing, Z. Photoresponsive Hydrogel Dressing Containing Nanoparticles with Excellent Synergetic Photodynamic, Photothermal, and Chemodynamic Therapies for Effective Infected Wound Healing. ACS Appl. Bio Mater. 2024, 7, 6970–6984. [Google Scholar] [CrossRef]
  111. Castrejón-Comas, V.; Mataró, N.; Resina, L.; Zanuy, D.; Nuñez-Aulina, Q.; Sánchez-Morán, J.; Enshaei, H.; Arnau, M.; Muñoz-Galán, H.; Worch, J.C.; et al. Electro-Responsive Hyaluronic Acid-Based Click-Hydrogels for Wound Healing. Carbohydr. Polym. 2025, 348, 122941. [Google Scholar] [CrossRef] [PubMed]
  112. Ding, H.; Hao, L.; Mao, H. Magneto-Responsive Biocomposites in Wound Healing: From Characteristics to Functions. J. Mater. Chem. B 2024, 12, 7463–7479. [Google Scholar] [CrossRef] [PubMed]
  113. Huang, Z.; Wang, M.; Chai, L.; Chen, H.; Chen, D.; Li, Y.; Liu, H.; Wu, Y.; Yang, X.; He, L.; et al. Glucose-Responsive, Self-Healing, Wet Adhesive and Multi-Biofunctional Hydrogels for Diabetic Wound Healing. Mater. Today Bio 2024, 27, 101159. [Google Scholar] [CrossRef]
  114. Cheng, Q.; Zhu, J.; Tu, S.; Fan, X. Enzyme-Crosslinked Hyaluronic Acid-Based Multifunctional Hydrogel Promotes Diabetic Wound Healing. Macromol. Res. 2025, 33, 779–788. [Google Scholar] [CrossRef]
  115. Maaz Arif, M.; Khan, S.M.; Gull, N.; Tabish, T.A.; Zia, S.; Ullah Khan, R.; Awais, S.M.; Arif Butt, M. Polymer-Based Biomaterials for Chronic Wound Management: Promises and Challenges. Int. J. Pharm. 2021, 598, 120270. [Google Scholar] [CrossRef]
  116. Hussain, Z.; Thu, H.E.; Shuid, A.N.; Katas, H.; Hussain, F. Recent Advances in Polymer-Based Wound Dressings for the Treatment of Diabetic Foot Ulcer: An Overview of State-of-the-Art. Curr. Drug Targets 2018, 19, 527–550. [Google Scholar] [CrossRef]
  117. Heremans, J.; Ballet, S.; Martin, C. The Versatility of Peptide Hydrogels: From Self-assembly to Drug Delivery Applications. J. Pept. Sci. 2025, 31, e3662. [Google Scholar] [CrossRef]
  118. Bakhtiary, N.; Ghalandari, B.; Ghorbani, F.; Varma, S.N.; Liu, C. Advances in Peptide-Based Hydrogel for Tissue Engineering. Polymers 2023, 15, 1068. [Google Scholar] [CrossRef]
  119. Cauwenbergh, T.; Ballet, S.; Martin, C. Peptide Hydrogel-Drug Conjugates for Tailored Disease Treatment. Mater. Today Bio 2025, 31, 101423. [Google Scholar] [CrossRef] [PubMed]
  120. Liu, J.; Xi, Z.; Fan, C.; Mei, Y.; Zhao, J.; Jiang, Y.; Zhao, M.; Xu, L. Hydrogels for Nucleic Acid Drugs Delivery. Adv. Healthc. Mater. 2024, 13, e2401895. [Google Scholar] [CrossRef] [PubMed]
  121. Wu, R.; Li, W.; Yang, P.; Shen, N.; Yang, A.; Liu, X.; Ju, Y.; Lei, L.; Fang, B. DNA Hydrogels and Their Derivatives in Biomedical Engineering Applications. J. Nanobiotechnology 2024, 22, 518. [Google Scholar] [CrossRef]
  122. Zhang, Y.; Yan, W.; Wu, L.; Yu, Z.; Quan, Y.; Xie, X. Different Exosomes Are Loaded in Hydrogels for the Application in the Field of Tissue Repair. Front. Bioeng. Biotechnol. 2025, 13, 1545636. [Google Scholar] [CrossRef] [PubMed]
  123. Zheng, Q.; Yao, J.; Sun, Z.; Mao, Y.; Wei, J.; Xie, Y.; Hu, X.K.; Li, X. Hydrogels as Carriers Deliver Stem Cells/Exosomes for Liver Injury. Mater. Adv. 2024, 5, 3587–3601. [Google Scholar] [CrossRef]
  124. Sille, I.E.; Pissinis, D.E.; Fagali, N.S.; Ghilini, F.; Urrutia, M.N.; Schilardi, P.L. Antimicrobial-Loaded Polyacrylamide Hydrogels Supported on Titanium as Reservoir for Local Drug Delivery. Pathogens 2023, 12, 202. [Google Scholar] [CrossRef]
  125. Ahmad, N.; Bukhari, S.N.A.; Hussain, M.A.; Ejaz, H.; Munir, M.U.; Amjad, M.W. Nanoparticles Incorporated Hydrogels for Delivery of Antimicrobial Agents: Developments and Trends. RSC Adv. 2024, 14, 13535–13564. [Google Scholar] [CrossRef]
  126. Moorcroft, S.C.T.; Roach, L.; Jayne, D.G.; Ong, Z.Y.; Evans, S.D. Nanoparticle-Loaded Hydrogel for the Light-Activated Release and Photothermal Enhancement of Antimicrobial Peptides. ACS Appl. Mater. Interfaces 2020, 12, 24544–24554. [Google Scholar] [CrossRef]
  127. Bâldea, I.; Soran, M.-L.; Stegarescu, A.; Opriș, O.; Kacso, I.; Tripon, S.; Adascalitei, A.; Fericel, I.G.; Decea, R.; Lung, I. Lilium Candidum Extract Loaded in Alginate Hydrogel Beads for Chronic Wound Healing. Gels 2025, 11, 22. [Google Scholar] [CrossRef]
  128. Homaeigohar, S.; Boccaccini, A.R. Antibacterial Biohybrid Nanofibers for Wound Dressings. Acta Biomater. 2020, 107, 25–49. [Google Scholar] [CrossRef]
  129. Aderibigbe, B.; Buyana, B. Alginate in Wound Dressings. Pharmaceutics 2018, 10, 42. [Google Scholar] [CrossRef]
  130. Zhang, M.; Zhao, X. Alginate Hydrogel Dressings for Advanced Wound Management. Int. J. Biol. Macromol. 2020, 162, 1414–1428. [Google Scholar] [CrossRef] [PubMed]
  131. Bâldea, I.; Lung, I.; Opriş, O.; Stegarescu, A.; Kacso, I.; Soran, M.-L. Antioxidant, Anti-Inflammatory Effects and Ability to Stimulate Wound Healing of a Common-Plantain Extract in Alginate Gel Formulations. Gels 2023, 9, 901. [Google Scholar] [CrossRef] [PubMed]
  132. Zhang, H.; Cheng, J.; Ao, Q. Preparation of Alginate-Based Biomaterials and Their Applications in Biomedicine. Mar. Drugs 2021, 19, 264. [Google Scholar] [CrossRef] [PubMed]
  133. Pereira, R.; Carvalho, A.; Vaz, D.C.; Gil, M.H.; Mendes, A.; Bártolo, P. Development of Novel Alginate Based Hydrogel Films for Wound Healing Applications. Int. J. Biol. Macromol. 2013, 52, 221–230. [Google Scholar] [CrossRef] [PubMed]
  134. Serra, M.; Correia, C.; Malpique, R.; Brito, C.; Jensen, J.; Bjorquist, P.; Carrondo, M.J.T.; Alves, P.M. Microencapsulation Technology: A Powerful Tool for Integrating Expansion and Cryopreservation of Human Embryonic Stem Cells. PLoS ONE 2011, 6, e23212. [Google Scholar] [CrossRef]
  135. Supramaniam, J.; Adnan, R.; Mohd Kaus, N.H.; Bushra, R. Magnetic Nanocellulose Alginate Hydrogel Beads as Potential Drug Delivery System. Int. J. Biol. Macromol. 2018, 118, 640–648. [Google Scholar] [CrossRef]
  136. Buket Basmanav, F.; Kose, G.T.; Hasirci, V. Sequential Growth Factor Delivery from Complexed Microspheres for Bone Tissue Engineering. Biomaterials 2008, 29, 4195–4204. [Google Scholar] [CrossRef]
  137. Ho, T.T.; Tran, H.A.; Doan, V.K.; Maitz, J.; Li, Z.; Wise, S.G.; Lim, K.S.; Rnjak-Kovacina, J. Natural Polymer-Based Materials for Wound Healing Applications. Adv. Nanobiomed Res. 2024, 4, 2300131. [Google Scholar] [CrossRef]
  138. Alberts, A.; Bratu, A.G.; Niculescu, A.-G.; Grumezescu, A.M. New Perspectives of Hydrogels in Chronic Wound Management. Molecules 2025, 30, 686. [Google Scholar] [CrossRef]
  139. Hodge, J.G.; Zamierowski, D.S.; Robinson, J.L.; Mellott, A.J. Evaluating Polymeric Biomaterials to Improve next Generation Wound Dressing Design. Biomater. Res. 2022, 26, 50. [Google Scholar] [CrossRef]
  140. Aderibigbe, B.A. Hybrid-Based Wound Dressings: Combination of Synthetic and Biopolymers. Polymers 2022, 14, 3806. [Google Scholar] [CrossRef]
  141. Vasile, C.; Pamfil, D.; Stoleru, E.; Baican, M. New Developments in Medical Applications of Hybrid Hydrogels Containing Natural Polymers. Molecules 2020, 25, 1539. [Google Scholar] [CrossRef] [PubMed]
  142. Ma, R.; Wang, Y.; Qi, H.; Shi, C.; Wei, G.; Xiao, L.; Huang, Z.; Liu, S.; Yu, H.; Teng, C.; et al. Nanocomposite Sponges of Sodium Alginate/Graphene Oxide/Polyvinyl Alcohol as Potential Wound Dressing: In Vitro and in Vivo Evaluation. Compos. B Eng. 2019, 167, 396–405. [Google Scholar] [CrossRef]
  143. Rana, M.M.; De la Hoz Siegler, H. Evolution of Hybrid Hydrogels: Next-Generation Biomaterials for Drug Delivery and Tissue Engineering. Gels 2024, 10, 216. [Google Scholar] [CrossRef]
  144. Liang, W.; Ni, N.; Huang, Y.; Lin, C. An Advanced Review: Polyurethane-Related Dressings for Skin Wound Repair. Polymers 2023, 15, 4301. [Google Scholar] [CrossRef]
  145. Eskandarinia, A.; Kefayat, A.; Agheb, M.; Rafienia, M.; Amini Baghbadorani, M.; Navid, S.; Ebrahimpour, K.; Khodabakhshi, D.; Ghahremani, F. A Novel Bilayer Wound Dressing Composed of a Dense Polyurethane/Propolis Membrane and a Biodegradable Polycaprolactone/Gelatin Nanofibrous Scaffold. Sci. Rep. 2020, 10, 3063. [Google Scholar] [CrossRef]
  146. Chen, H.; Xing, X.; Tan, H.; Jia, Y.; Zhou, T.; Chen, Y.; Ling, Z.; Hu, X. Covalently Antibacterial Alginate-Chitosan Hydrogel Dressing Integrated Gelatin Microspheres Containing Tetracycline Hydrochloride for Wound Healing. Mater. Sci. Eng. C 2017, 70, 287–295. [Google Scholar] [CrossRef] [PubMed]
  147. Li, S.; Li, L.; Guo, C.; Qin, H.; Yu, X. A Promising Wound Dressing Material with Excellent Cytocompatibility and Proangiogenesis Action for Wound Healing: Strontium Loaded Silk Fibroin/Sodium Alginate (SF/SA) Blend Films. Int. J. Biol. Macromol. 2017, 104, 969–978. [Google Scholar] [CrossRef]
  148. Mokhena, T.C.; Luyt, A.S. Electrospun Alginate Nanofibres Impregnated with Silver Nanoparticles: Preparation, Morphology and Antibacterial Properties. Carbohydr. Polym. 2017, 165, 304–312. [Google Scholar] [CrossRef] [PubMed]
  149. Jessy Mercy, D.; Thirumalai, A.; Udayakumar, S.; Deepika, B.; Janani, G.; Girigoswami, A.; Girigoswami, K. Enhancing Wound Healing with Nanohydrogel-Entrapped Plant Extracts and Nanosilver: An In Vitro Investigation. Molecules 2024, 29, 5004. [Google Scholar] [CrossRef] [PubMed]
  150. Ningrum, D.R.; Hanif, W.; Mardhian, D.F.; Asri, L.A.T.W. In Vitro Biocompatibility of Hydrogel Polyvinyl Alcohol/Moringa Oleifera Leaf Extract/Graphene Oxide for Wound Dressing. Polymers 2023, 15, 468. [Google Scholar] [CrossRef]
  151. Guo, C.; Zhou, Z.; Zhang, S.; Peng, P.; Liu, J.; Yan, Y.; Dong, J. Sulfonated, Quaternized, and Chlorogenic Acid Composited Sodium Alginate Hydrogels/Eucommia Ulmoides Rubber Films as in Vitro Antibacterial Wound Dressings for Accelerating Wound Healing. Ind. Crops Prod. 2022, 190, 115885. [Google Scholar] [CrossRef]
  152. Hofmann, E.; Fink, J.; Pignet, A.-L.; Schwarz, A.; Schellnegger, M.; Nischwitz, S.P.; Holzer-Geissler, J.C.J.; Kamolz, L.-P.; Kotzbeck, P. Human In Vitro Skin Models for Wound Healing and Wound Healing Disorders. Biomedicines 2023, 11, 1056. [Google Scholar] [CrossRef]
  153. Yadav, P.D.; Londhe, P.V.; Chavan, S.S.; Mohite, D.D.; Firame, G.B.; Kadam, S.S.; Patil, M.J.; Ansari, M.I. Electrospun Composite Nanofibers for Wound Healing: Synthesis, Characterization, and Clinical Potential of Biopolymer-Based Materials. Discov. Mater. 2024, 4, 99. [Google Scholar] [CrossRef]
  154. Joshi, A.S.; Madhusudanan, M.; Mijakovic, I. 3D Printed Inserts for Reproducible High Throughput Screening of Cell Migration. Front. Cell Dev. Biol. 2023, 11, 1256250. [Google Scholar] [CrossRef] [PubMed]
  155. Joshi, A.S. Advancing in Vitro Cell Migration Studies: A Review of Open-Source Analytical Platforms for Cancer and Wound Healing Research. Cell Adhes. Migr. 2025, 19, 2488116. [Google Scholar] [CrossRef] [PubMed]
  156. Nandi, S.; Brown, A.C. Characterizing Cell Migration Within Three-Dimensional In Vitro Wound Environments. J. Vis. Exp. 2017, 126, e56099. [Google Scholar] [CrossRef]
  157. Azzam, S.; Tomasova, L.; Danner, C.; Skiba, M.; Klein, M.; Guttenberg, Z.; Michaelis, S.; Wegener, J. A High-Precision Wound Healing Assay Based on Photosensitized Culture Substrates. Sci. Rep. 2024, 14, 9103. [Google Scholar] [CrossRef]
  158. Biglari, S.; Le, T.Y.L.; Tan, R.P.; Wise, S.G.; Zambon, A.; Codolo, G.; De Bernard, M.; Warkiani, M.; Schindeler, A.; Naficy, S.; et al. Simulating Inflammation in a Wound Microenvironment Using a Dermal Wound-on-a-Chip Model. Adv. Healthc. Mater. 2019, 8, e1801307. [Google Scholar] [CrossRef]
  159. Flynn, K.; Mahmoud, N.N.; Sharifi, S.; Gould, L.J.; Mahmoudi, M. Chronic Wound Healing Models. ACS Pharmacol. Transl. Sci. 2023, 6, 783–801. [Google Scholar] [CrossRef]
  160. Seiser, S.; Janker, L.; Zila, N.; Mildner, M.; Rakita, A.; Matiasek, J.; Bileck, A.; Gerner, C.; Paulitschke, V.; Elbe-Bürger, A. Octenidine-Based Hydrogel Shows Anti-Inflammatory and Protease-Inhibitory Capacities in Wounded Human Skin. Sci. Rep. 2021, 11, 32. [Google Scholar] [CrossRef]
  161. Liu, C.; Rinderknecht, H.; Histing, T.; Kolbenschlag, J.; Nussler, A.K.; Ehnert, S. Establishment of an In Vitro Scab Model for Investigating Different Phases of Wound Healing. Bioengineering 2022, 9, 191. [Google Scholar] [CrossRef]
  162. Kapałczyńska, M.; Kolenda, T.; Przybyła, W.; Zajączkowska, M.; Teresiak, A.; Filas, V.; Ibbs, M.; Bliźniak, R.; Łuczewski, Ł.; Lamperska, K. 2D and 3D Cell Cultures—A Comparison of Different Types of Cancer Cell Cultures. Arch. Med. Sci. 2016, 14, 910. [Google Scholar] [CrossRef]
  163. Haisma, E.M.; Rietveld, M.H.; de Breij, A.; van Dissel, J.T.; El Ghalbzouri, A.; Nibbering, P.H. Inflammatory and Antimicrobial Responses to Methicillin-Resistant Staphylococcus Aureus in an In Vitro Wound Infection Model. PLoS ONE 2013, 8, e82800. [Google Scholar] [CrossRef]
  164. Mieremet, A.; Rietveld, M.; Absalah, S.; van Smeden, J.; Bouwstra, J.A.; El Ghalbzouri, A. Improved Epidermal Barrier Formation in Human Skin Models by Chitosan Modulated Dermal Matrices. PLoS ONE 2017, 12, e0174478. [Google Scholar] [CrossRef]
  165. Folle, C.; Díaz-Garrido, N.; Mallandrich, M.; Suñer-Carbó, J.; Sánchez-López, E.; Halbaut, L.; Marqués, A.M.; Espina, M.; Badia, J.; Baldoma, L.; et al. Hydrogel of Thyme-Oil-PLGA Nanoparticles Designed for Skin Inflammation Treatment. Gels 2024, 10, 149. [Google Scholar] [CrossRef]
  166. Gross-Amat, O.; Guillen, M.; Salmon, D.; Nataf, S.; Auxenfans, C. Characterization of a Topically Testable Model of Burn Injury on Human Skin Explants. Int. J. Mol. Sci. 2020, 21, 6956. [Google Scholar] [CrossRef]
  167. O’Connor, N.A.; Syed, A.; Kastrat, E.; Cheng, H.-P. Antibacterial Silver Nanoparticle Containing Polydopamine Hydrogels That Enhance Re-Epithelization. Gels 2024, 10, 363. [Google Scholar] [CrossRef]
  168. Singh, V.; Marimuthu, T.; Lesotho, N.F.; Makatini, M.M.; Ntombela, T.; Van Eyk, A.; Choonara, Y.E. Synthesis of a Retro -GFOGER Adamantane-Based Collagen Mimetic Peptide Imbibed in a Hyaluronic Acid Hydrogel for Enhanced Wound Healing. ACS Appl. Bio Mater. 2025, 8, 4657–4672. [Google Scholar] [CrossRef] [PubMed]
  169. Zhao, R.; Zhao, C.; Wan, Y.; Majid, M.; Abbas, S.Q.; Wang, Y. In Vitro and in Vivo Evaluation of Alginate Hydrogel-Based Wound Dressing Loaded with Green Chemistry Cerium Oxide Nanoparticles. Front. Chem. 2023, 11, 1298808. [Google Scholar] [CrossRef] [PubMed]
  170. Gurtner, G.C.; Werner, S.; Barrandon, Y.; Longaker, M.T. Wound Repair and Regeneration. Nature 2008, 453, 314–321. [Google Scholar] [CrossRef]
  171. Grada, A.; Mervis, J.; Falanga, V. Research Techniques Made Simple: Animal Models of Wound Healing. J. Investig. Dermatol. 2018, 138, 2095–2105.e1. [Google Scholar] [CrossRef]
  172. Kurt, A.A.; Ibrahim, B.; Çınar, H.; Atsü, A.N.; Bursalıoğlu, E.O.; Bayır, İ.; Özmen, Ö.; Aslan, İ. Nanoemulsion Hydrogel Delivery System of Hypericum Perforatum L.: In Silico Design, In Vitro Antimicrobial–Toxicological Profiling, and In Vivo Wound-Healing Evaluation. Gels 2025, 11, 431. [Google Scholar] [CrossRef]
  173. Gagliardi, A.; Giuliano, E.; Voci, S.; Costa, N.; Bulotta, S.; Salvatici, M.C.; Ambrosio, N.; Paolino, D.; Siddique, F.; Majid, M.; et al. Rutin-Loaded Zein Gel as a Green Biocompatible Formulation for Wound Healing Application. Int. J. Biol. Macromol. 2024, 269, 132071. [Google Scholar] [CrossRef] [PubMed]
  174. Grover, P.; Khanna, K.; Bhatnagar, A.; Purkayastha, J. In Vivo-Wound Healing Studies of Sodium Thiosulfate Gel in Rats. Biomed. Pharmacother. 2021, 140, 111797. [Google Scholar] [CrossRef] [PubMed]
  175. Okur, M.E.; Karadağ, A.E.; Üstündağ Okur, N.; Özhan, Y.; Sipahi, H.; Ayla, Ş.; Daylan, B.; Demirci, B.; Demirci, F. In Vivo Wound Healing and In Vitro Anti-Inflammatory Activity Evaluation of Phlomis Russeliana Extract Gel Formulations. Molecules 2020, 25, 2695. [Google Scholar] [CrossRef]
  176. Akbari, F.; Azadbakht, M.; Bagheri, A.; Vahedi, L. In Vitro and In Vivo Wound Healing Activity of Astragalus Floccosus Boiss. (Fabaceae). Adv. Pharmacol. Pharm. Sci. 2022, 2022, 1–15. [Google Scholar] [CrossRef]
  177. Jabbar, A.A.; Abdul-Aziz Ahmed, K.; Abdulla, M.A.; Abdullah, F.O.; Salehen, N.A.; Mothana, R.A.; Houssaini, J.; Hassan, R.R.; Hawwal, M.F.; Fantoukh, O.I.; et al. Sinomenine Accelerate Wound Healing in Rats by Augmentation of Antioxidant, Anti-Inflammatory, Immunuhistochemical Pathways. Heliyon 2024, 10, e23581. [Google Scholar] [CrossRef] [PubMed]
  178. Li, H.; Ziemer, M.; Stojanovic, I.; Saksida, T.; Maksimovic-Ivanic, D.; Mijatovic, S.; Djmura, G.; Gajic, D.; Koprivica, I.; Krajnovic, T.; et al. Mesenchymal Stem Cells From Mouse Hair Follicles Reduce Hypertrophic Scarring in a Murine Wound Healing Model. Stem Cell Rev. Rep. 2022, 18, 2028–2044. [Google Scholar] [CrossRef]
  179. Tunç, A.S.; Ercan, N. Effect of Topical Sildenafil on Wound Healing and Oxidative Stress in Rats. Injury 2024, 55, 111525. [Google Scholar] [CrossRef]
  180. Rodgers, K.; Xiong, S.; Felix, J.; Roda, N.; Espinoza, T.; Maldonado, S.; Dizerega, G. Development of Angiotensin (1-7) as an Agent to Accelerate Dermal Repair. Wound Repair Regen. 2001, 9, 238–247. [Google Scholar] [CrossRef]
  181. Jia, Y.; Zhao, G.; Jia, J. Preliminary Evaluation: The Effects of Aloe Ferox Miller and Aloe Arborescens Miller on Wound Healing. J. Ethnopharmacol. 2008, 120, 181–189. [Google Scholar] [CrossRef]
  182. Choi, J.; Park, Y.-G.; Yun, M.-S.; Seol, J.-W. Effect of Herbal Mixture Composed of Alchemilla Vulgaris and Mimosa on Wound Healing Process. Biomed. Pharmacother. 2018, 106, 326–332. [Google Scholar] [CrossRef]
  183. Oladejo, O.W.; Imosemi, I.O.; Osuagwu, F.C.; Oyedele, O.O.; Oluwadara, O.O.; Ekpo, O.E.; Aiku, A.; Adewoyin, O.; Akang, E.E.U. A Comparative Study of the Wound Healing Properties of Honey and Ageratum Conyzoides. Afr. J. Med. Med. Sci. 2003, 32, 193–196. [Google Scholar]
  184. Khosravian, P.; Motamedi, N.; Javdani, M.; Khorasgani, E.M. Histopathologic Evaluation of Skin Wound Healing Due to Local Application of Transdermal Chitosan Patch in Combination with Doxycycline, Zinc Nanoparticles, and Selenium Nanoparticles in Mice. BMC Vet. Res. 2025, 21, 221. [Google Scholar] [CrossRef]
  185. Nordback, P.H.; Miettinen, S.; Kääriäinen, M.; Haaparanta, A.-M.; Kellomäki, M.; Kuokkanen, H.; Seppänen, R. Chitosan Membranes in a Rat Model of Full-Thickness Cutaneous Wounds: Healing and IL-4 Levels. J. Wound Care 2015, 24, 245–251. [Google Scholar] [CrossRef]
  186. Ozkaya, H.; Omma, T.; Bag, Y.M.; Uzunoglu, K.; Isildak, M.; Duymus, M.E.; Kismet, K.; Senes, M.; Fidanci, V.; Celepli, P.; et al. Topical and Systemic Effects of N-Acetyl Cysteine on Wound Healing in a Diabetic Rat Model. Wounds 2019, 31, 91–96. [Google Scholar]
  187. Yang, H.; Song, L.; Sun, B.; Chu, D.; Yang, L.; Li, M.; Li, H.; Dai, Y.; Yu, Z.; Guo, J. Modulation of Macrophages by a Paeoniflorin-Loaded Hyaluronic Acid-Based Hydrogel Promotes Diabetic Wound Healing. Mater. Today Bio 2021, 12, 100139. [Google Scholar] [CrossRef] [PubMed]
  188. Ru, J.; Zhang, Q.; Zhu, S.; Cai, J.; He, Y.; Lu, F. Delivery of Adipose-Derived Growth Factors from Heparinized Adipose Acellular Matrix Accelerates Wound Healing. Front. Bioeng. Biotechnol. 2023, 11, 1270618. [Google Scholar] [CrossRef] [PubMed]
  189. Li, R.; Wang, H.; Wang, X.; Yang, Y.; Zhong, K.; Zhang, X.; Li, H. MSC-EVs and UCB-EVs Promote Skin Wound Healing and Spatial Transcriptome Analysis. Sci. Rep. 2025, 15, 4006. [Google Scholar] [CrossRef]
  190. Hong, S.E.; Hong, M.K.; Kang, S.R.; Young Park, B. Effects of Neodymium–Yttrium–Aluminum Garnet (Nd:YAG) Pulsed High-Intensity Laser Therapy on Full Thickness Wound Healing in an Experimental Animal Model. J. Cosmet. Laser Ther. 2016, 18, 432–437. [Google Scholar] [CrossRef]
  191. Kieran, I.; Knock, A.; Bush, J.; So, K.; Metcalfe, A.; Hobson, R.; Mason, T.; O’Kane, S.; Ferguson, M. Interleukin-10 Reduces Scar Formation in Both Animal and Human Cutaneous Wounds: Results of Two Preclinical and Phase II Randomized Control Studies. Wound Repair Regen. 2013, 21, 428–436. [Google Scholar] [CrossRef] [PubMed]
  192. Deng, C.; He, Y.; Feng, J.; Dong, Z.; Yao, Y.; Mok, H.; Lin, M.; Feng, L. Extracellular Matrix/Stromal Vascular Fraction Gel Conditioned Medium Accelerates Wound Healing in a Murine Model. Wound Repair Regen. 2017, 25, 923–932. [Google Scholar] [CrossRef]
  193. Ebrahimi, A.; Reza Farahpour, M.; Amjadi, S.; Mohammadi, M.; Hamishehkar, H. Nanoliposomal Peptides Derived from Spirulina Platensis Protein Accelerate Full-Thickness Wound Healing. Int. J. Pharm. 2023, 630, 122457. [Google Scholar] [CrossRef]
  194. Gil, E.S.; Panilaitis, B.; Bellas, E.; Kaplan, D.L. Functionalized Silk Biomaterials for Wound Healing. Adv. Healthc. Mater. 2013, 2, 206–217. [Google Scholar] [CrossRef]
  195. Huang, Y.; Qin, P.; Zhou, P.; Long, B.; Zhang, S.; Gao, R.; Zhu, B.; Li, Y.; Li, Q. Non-Pharmacological Interventions of Intermittent Fasting and Pulsed Radiofrequency Energy (PRFE) Combination Therapy Promote Diabetic Wound Healing. Nutr. Diabetes 2024, 14, 83. [Google Scholar] [CrossRef]
  196. Mehmandoust, F.G.; Torkaman, G.; Firoozabadi, M.; Talebi, G. Anodal and Cathodal Pulsed Electrical Stimulation on Skin Wound Healing in Guinea Pigs. J. Rehabil. Res. Dev. 2007, 44, 611. [Google Scholar] [CrossRef] [PubMed]
  197. Burgess, J.L.; Wyant, W.A.; Abdo Abujamra, B.; Kirsner, R.S.; Jozic, I. Diabetic Wound-Healing Science. Medicina 2021, 57, 1072. [Google Scholar] [CrossRef]
  198. Zhang, W.; Liu, L.; Cheng, H.; Zhu, J.; Li, X.; Ye, S.; Li, X. Hydrogel-Based Dressings Designed to Facilitate Wound Healing. Mater. Adv. 2024, 5, 1364–1394. [Google Scholar] [CrossRef]
  199. Wang, T.; Wang, J.; Wang, R.; Yuan, P.; Fan, Z.; Yang, S. Preparation and Properties of ZnO/Sodium Alginate Bi-Layered Hydrogel Films as Novel Wound Dressings. New J. Chem. 2019, 43, 8684–8693. [Google Scholar] [CrossRef]
  200. Nešović, K.; Mišković-Stanković, V. A Comprehensive Review of the Polymer-based Hydrogels with Electrochemically Synthesized Silver Nanoparticles for Wound Dressing Applications. Polym. Eng. Sci. 2020, 60, 1393–1419. [Google Scholar] [CrossRef]
  201. Hajiali, H.; Summa, M.; Russo, D.; Armirotti, A.; Brunetti, V.; Bertorelli, R.; Athanassiou, A.; Mele, E. Alginate–Lavender Nanofibers with Antibacterial and Anti-Inflammatory Activity to Effectively Promote Burn Healing. J. Mater. Chem. B 2016, 4, 1686–1695. [Google Scholar] [CrossRef] [PubMed]
  202. Hong, C.; Chung, H.; Lee, G.; Kim, D.; Jiang, Z.; Kim, S.-H.; Lee, K. Remendable Cross-Linked Alginate/Gelatin Hydrogels Incorporating Nanofibers for Wound Repair and Regeneration. Biomacromolecules 2024, 25, 4344–4357. [Google Scholar] [CrossRef]
  203. Alemdaroğlu, C.; Değim, Z.; Çelebi, N.; Zor, F.; Öztürk, S.; Erdoğan, D. An Investigation on Burn Wound Healing in Rats with Chitosan Gel Formulation Containing Epidermal Growth Factor. Burns 2006, 32, 319–327. [Google Scholar] [CrossRef] [PubMed]
  204. Niculescu, A.-G.; Bîrcă, A.C.; Mogoşanu, G.D.; Rădulescu, M.; Holban, A.M.; Manuc, D.; Alberts, A.; Grumezescu, A.M.; Mogoantă, L. Zinc Alginate Hydrogel-Coated Wound Dressings: Fabrication, Characterization, and Evaluation of Anti-Infective and In Vivo Performance. Gels 2025, 11, 427. [Google Scholar] [CrossRef]
  205. Barakat, H.S.; Freag, M.S.; Gaber, S.M.; Al Oufy, A.; Abdallah, O.Y. Development of Verapamil Hydrochloride-Loaded Biopolymer-Based Composite Electrospun Nanofibrous Mats: In Vivo Evaluation of Enhanced Burn Wound Healing without Scar Formation. Drug Des. Devel Ther. 2023, 17, 1211–1231. [Google Scholar] [CrossRef] [PubMed]
  206. Fiorentini, F.; Suarato, G.; Summa, M.; Miele, D.; Sandri, G.; Bertorelli, R.; Athanassiou, A. Plant-Based, Hydrogel-like Microfibers as an Antioxidant Platform for Skin Burn Healing. ACS Appl. Bio Mater. 2023, 6, 3103–3116. [Google Scholar] [CrossRef]
  207. Takahashi, M.; Morimoto, Y.; Kon, M.; Saga, K. A Histological Study of Cutaneous Thermal Wounds Using a Clostridium Perfringens-Derived Wound Healing Substance with Wound Healing Stimulation Activity. J. Dermatol. 1995, 22, 98–106. [Google Scholar] [CrossRef] [PubMed]
  208. Lin, L.-X.; Wang, P.; Wang, Y.-T.; Huang, Y.; Jiang, L.; Wang, X.-M. Aloe Vera and Vitis Vinifera Improve Wound Healing in an in Vivo Rat Burn Wound Model. Mol. Med. Rep. 2016, 13, 1070–1076. [Google Scholar] [CrossRef]
  209. Askin, S.; Kaynarpinar, M. Efficacy of Formulation With Potential as Herbal Medicine on Second Degree Burn Wound: Biochemical and Molecular Evaluation. J. Cosmet. Dermatol. 2025, 24, e70122. [Google Scholar] [CrossRef]
  210. Fei, Y.; Ling, Z.; Tong, Q.; Wang, J. Apoptotic Extracellular Vesicles from Supernumerary Tooth-Derived Pulp Stem Cells Transfer COL1A1 to Promote Angiogenesis via PI3K/Akt/VEGF Pathway. Int. J. Nanomed. 2024, 19, 6811–6828. [Google Scholar] [CrossRef]
  211. Lowe, A.S.; Walker, M.D.; Cowan, R.; Baxter, G.D. Therapeutic Ultrasound and Wound Closure: Lack of Healing Effect on x-Ray Irradiated Wounds in Murine Skin. Arch. Phys. Med. Rehabil. 2001, 82, 1507–1511. [Google Scholar] [CrossRef]
  212. Belmont, M.J.; Marabelle, N.; Mang, T.S.; Hall, R.; Wax, M.K. Effect of Photodynamic Therapy on Revascularization of Fasciocutaneous Flaps. Laryngoscope 2000, 110, 942–945. [Google Scholar] [CrossRef]
  213. Sami, D.G.; Heiba, H.H.; Abdellatif, A. Wound Healing Models: A Systematic Review of Animal and Non-Animal Models. Wound Med. 2019, 24, 8–17. [Google Scholar] [CrossRef]
  214. Zomer, H.D.; Trentin, A.G. Skin Wound Healing in Humans and Mice: Challenges in Translational Research. J. Dermatol. Sci. 2018, 90, 3–12. [Google Scholar] [CrossRef]
  215. Parnell, L.K.S.; Volk, S.W. The Evolution of Animal Models in Wound Healing Research: 1993–2017. Adv. Wound Care 2019, 8, 692–702. [Google Scholar] [CrossRef] [PubMed]
  216. Pasupuleti, M.; Molahally, S.; Salwaji, S. Ethical Guidelines, Animal Profile, Various Animal Models Used in Periodontal Research with Alternatives and Future Perspectives. J. Indian. Soc. Periodontol. 2016, 20, 360. [Google Scholar] [CrossRef] [PubMed]
  217. Gounden, V.; Singh, M. Hydrogels and Wound Healing: Current and Future Prospects. Gels 2024, 10, 43. [Google Scholar] [CrossRef] [PubMed]
  218. Tavakoli, S.; Klar, A.S. Advanced Hydrogels as Wound Dressings. Biomolecules 2020, 10, 1169. [Google Scholar] [CrossRef]
  219. Shu, W.; Wang, Y.; Zhang, X.; Li, C.; Le, H.; Chang, F. Functional Hydrogel Dressings for Treatment of Burn Wounds. Front. Bioeng. Biotechnol. 2021, 9, 788461. [Google Scholar] [CrossRef]
  220. Fan, F.; Saha, S.; Hanjaya-Putra, D. Biomimetic Hydrogels to Promote Wound Healing. Front. Bioeng. Biotechnol. 2021, 9, 718377. [Google Scholar] [CrossRef] [PubMed]
  221. He, Y.; Cen, Y.; Tian, M. Immunomodulatory Hydrogels for Skin Wound Healing: Cellular Targets and Design Strategy. J. Mater. Chem. B 2024, 12, 2435–2458. [Google Scholar] [CrossRef]
  222. Surowiecka, A.; Strużyna, J.; Winiarska, A.; Korzeniowski, T. Hydrogels in Burn Wound Management—A Review. Gels 2022, 8, 122. [Google Scholar] [CrossRef]
  223. Murphy, S.V.; Skardal, A.; Atala, A. Evaluation of Hydrogels for Bio-printing Applications. J. Biomed. Mater. Res. A 2013, 101A, 272–284. [Google Scholar] [CrossRef]
  224. Wang, H.; Liu, Y.; Cai, K.; Zhang, B.; Tang, S.; Zhang, W.; Liu, W. Antibacterial Polysaccharide-Based Hydrogel Dressing Containing Plant Essential Oil for Burn Wound Healing. Burns Trauma. 2021, 9, tkab041. [Google Scholar] [CrossRef]
  225. Li, S.; Dong, S.; Xu, W.; Tu, S.; Yan, L.; Zhao, C.; Ding, J.; Chen, X. Antibacterial Hydrogels. Adv. Sci. 2018, 5, 1700527. [Google Scholar] [CrossRef]
  226. Manju, S.; Antony, M.; Sreenivasan, K. Synthesis and Evaluation of a Hydrogel That Binds Glucose and Releases Ciprofloxacin. J. Mater. Sci. 2010, 45, 4006–4012. [Google Scholar] [CrossRef]
  227. Sa, Y.; Wang, M.; Deng, H.; Wang, Y.; Jiang, T. Beneficial Effects of Biomimetic Nano-Sized Hydroxyapatite/Antibiotic Gentamicin Enriched Chitosan–Glycerophosphate Hydrogel on the Performance of Injectable Polymethylmethacrylate. RSC Adv. 2015, 5, 91082–91092. [Google Scholar] [CrossRef]
  228. Kamoun, E.A.; Kenawy, E.-R.S.; Tamer, T.M.; El-Meligy, M.A.; Mohy Eldin, M.S. Poly (Vinyl Alcohol)-Alginate Physically Crosslinked Hydrogel Membranes for Wound Dressing Applications: Characterization and Bio-Evaluation. Arab. J. Chem. 2015, 8, 38–47. [Google Scholar] [CrossRef]
  229. Lai, P.-L.; Hong, D.-W.; Ku, K.-L.; Lai, Z.-T.; Chu, I.-M. Novel Thermosensitive Hydrogels Based on Methoxy Polyethylene Glycol-Co-Poly(Lactic Acid-Co-Aromatic Anhydride) for Cefazolin Delivery. Nanomedicine 2014, 10, 553–560. [Google Scholar] [CrossRef] [PubMed]
  230. Gustafson, C.T.; Boakye-Agyeman, F.; Brinkman, C.L.; Reid, J.M.; Patel, R.; Bajzer, Z.; Dadsetan, M.; Yaszemski, M.J. Controlled Delivery of Vancomycin via Charged Hydrogels. PLoS ONE 2016, 11, e0146401. [Google Scholar] [CrossRef] [PubMed]
  231. Sawadkar, P.; Lali, F.; Garcia-Gareta, E.; Garrido, B.G.; Chaudhry, A.; Matharu, P.; Kyriakidis, C.; Greco, K. Innovative Hydrogels in Cutaneous Wound Healing: Current Status and Future Perspectives. Front. Bioeng. Biotechnol. 2025, 13, 1454903. [Google Scholar] [CrossRef]
  232. Li, Z.; Qu, T.; Ding, C.; Ma, C.; Sun, H.; Li, S.; Liu, X. Injectable Gelatin Derivative Hydrogels with Sustained Vascular Endothelial Growth Factor Release for Induced Angiogenesis. Acta Biomater. 2015, 13, 88–100. [Google Scholar] [CrossRef]
  233. Xiao, Z.; Zheng, X.; An, Y.; Wang, K.; Zhang, J.; He, H.; Wu, J. Zwitterionic Hydrogel for Sustained Release of Growth Factors to Enhance Wound Healing. Biomater. Sci. 2021, 9, 882–891. [Google Scholar] [CrossRef] [PubMed]
  234. Bitterman-Deutsch, O.; Kogan, L.; Nasser, F. Delayed Immune Mediated Adverse Effects to Hyaluronic Acid Fillers: Report of Five Cases and Review of the Literature. Dermatol. Rep. 2015, 7, 5851. [Google Scholar] [CrossRef]
  235. Dam, P.; Celik, M.; Ustun, M.; Saha, S.; Saha, C.; Kacar, E.A.; Kugu, S.; Karagulle, E.N.; Tasoglu, S.; Buyukserin, F.; et al. Wound Healing Strategies Based on Nanoparticles Incorporated in Hydrogel Wound Patches. RSC Adv. 2023, 13, 21345–21364. [Google Scholar] [CrossRef] [PubMed]
  236. Kar, A.K.; Singh, A.; Dhiman, N.; Purohit, M.P.; Jagdale, P.; Kamthan, M.; Singh, D.; Kumar, M.; Ghosh, D.; Patnaik, S. Polymer-Assisted In Situ Synthesis of Silver Nanoparticles with Epigallocatechin Gallate (EGCG) Impregnated Wound Patch Potentiate Controlled Inflammatory Responses for Brisk Wound Healing. Int. J. Nanomed. 2019, 14, 9837–9854. [Google Scholar] [CrossRef]
  237. Zhang, B.; Lv, Y.; Yu, C.; Zhang, W.; Song, S.; Li, Y.; Chong, Y.; Huang, J.; Zhang, Z. Au–Pt Nanozyme-Based Multifunctional Hydrogel Dressing for Diabetic Wound Healing. Biomater. Adv. 2022, 137, 212869. [Google Scholar] [CrossRef] [PubMed]
  238. Sahiner, N.; Godbey, W.T.; McPherson, G.L.; John, V.T. Microgel, Nanogel and Hydrogel–Hydrogel Semi-IPN Composites for Biomedical Applications: Synthesis and Characterization. Colloid. Polym. Sci. 2006, 284, 1121–1129. [Google Scholar] [CrossRef]
  239. Liang, C.; Dudko, V.; Khoruzhenko, O.; Hong, X.; Lv, Z.-P.; Tunn, I.; Umer, M.; Timonen, J.V.I.; Linder, M.B.; Breu, J.; et al. Stiff and Self-Healing Hydrogels by Polymer Entanglements in Co-Planar Nanoconfinement. Nat. Mater. 2025, 24, 599–606. [Google Scholar] [CrossRef]
  240. Wang, Y.; Zhang, M.; Yan, Z.; Ji, S.; Xiao, S.; Gao, J. Metal Nanoparticle Hybrid Hydrogels: The State-of-the-Art of Combining Hard and Soft Materials to Promote Wound Healing. Theranostics 2024, 14, 1534–1560. [Google Scholar] [CrossRef]
  241. Xue, H.; Hu, L.; Xiong, Y.; Zhu, X.; Wei, C.; Cao, F.; Zhou, W.; Sun, Y.; Endo, Y.; Liu, M.; et al. Quaternized Chitosan-Matrigel-Polyacrylamide Hydrogels as Wound Dressing for Wound Repair and Regeneration. Carbohydr. Polym. 2019, 226, 115302. [Google Scholar] [CrossRef]
  242. Lee, Y.-H.; Hong, Y.-L.; Wu, T.-L. 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. [Google Scholar] [CrossRef]
  243. Ying, H.; Zhou, J.; Wang, M.; Su, D.; Ma, Q.; Lv, G.; Chen, J. In Situ Formed Collagen-Hyaluronic Acid Hydrogel as Biomimetic Dressing for Promoting Spontaneous Wound Healing. Mater. Sci. Eng. C 2019, 101, 487–498. [Google Scholar] [CrossRef]
  244. Basha, S.I.; Ghosh, S.; Vinothkumar, K.; Ramesh, B.; Kumari, P.H.P.; Mohan, K.V.M.; Sukumar, E. Fumaric Acid Incorporated Ag/Agar-Agar Hybrid Hydrogel: A Multifunctional Avenue to Tackle Wound Healing. Mater. Sci. Eng. C 2020, 111, 110743. [Google Scholar] [CrossRef]
  245. Daristotle, J.L.; Lau, L.W.; Erdi, M.; Hunter, J.; Djoum, A.; Srinivasan, P.; Wu, X.; Basu, M.; Ayyub, O.B.; Sandler, A.D.; et al. Sprayable and Biodegradable, Intrinsically Adhesive Wound Dressing with Antimicrobial Properties. Bioeng. Transl. Med. 2020, 5, 10149. [Google Scholar] [CrossRef]
  246. Zhang, S.; Liu, Y.; Zhang, X.; Zhu, D.; Qi, X.; Cao, X.; Fang, Y.; Che, Y.; Han, Z.-C.; He, Z.-X.; et al. Prostaglandin E 2 Hydrogel Improves Cutaneous Wound Healing via M2 Macrophages Polarization. Theranostics 2018, 8, 5348–5361. [Google Scholar] [CrossRef]
  247. Liu, M.; Chen, Y.; Zhu, Q.; Tao, J.; Tang, C.; Ruan, H.; Wu, Y.; Loh, X.J. Antioxidant Thermogelling Formulation for Burn Wound Healing. Chem. Asian J. 2022, 17, e202200396. [Google Scholar] [CrossRef]
  248. Gokce, E.H.; Tuncay Tanrıverdi, S.; Eroglu, I.; Tsapis, N.; Gokce, G.; Tekmen, I.; Fattal, E.; Ozer, O. Wound Healing Effects of Collagen-Laminin Dermal Matrix Impregnated with Resveratrol Loaded Hyaluronic Acid-DPPC Microparticles in Diabetic Rats. Eur. J. Pharm. Biopharm. 2017, 119, 17–27. [Google Scholar] [CrossRef]
  249. Ioniță, A.C. In vitro effects of some synthesized aminoacetanilide N’-substituted on human leukocytes separated from peripheral blood. Farmacia 2019, 67, 684–690. [Google Scholar] [CrossRef]
  250. Li, Y.; Fu, R.; Zhu, C.; Fan, D. An Antibacterial Bilayer Hydrogel Modified by Tannic Acid with Oxidation Resistance and Adhesiveness to Accelerate Wound Repair. Colloids Surf. B Biointerfaces 2021, 205, 111869. [Google Scholar] [CrossRef] [PubMed]
  251. Hussain, M.; Suo, H.; Xie, Y.; Wang, K.; Wang, H.; Hou, Z.; Gao, Y.; Zhang, L.; Tao, J.; Jiang, H.; et al. Dopamine-Substituted Multidomain Peptide Hydrogel with Inherent Antimicrobial Activity and Antioxidant Capability for Infected Wound Healing. ACS Appl. Mater. Interfaces 2021, 13, 29380–29391. [Google Scholar] [CrossRef] [PubMed]
  252. Ehterami, A.; Salehi, M.; Farzamfar, S.; Samadian, H.; Vaez, A.; Ghorbani, S.; Ai, J.; Sahrapeyma, H. Chitosan/Alginate Hydrogels Containing Alpha-Tocopherol for Wound Healing in Rat Model. J. Drug Deliv. Sci. Technol. 2019, 51, 204–213. [Google Scholar] [CrossRef]
  253. Yahia, E.A.; El-Sharkawey, A.E.; Bayoumi, M.M. Quantitative Evaluation of Diabetic Foot Wound Healing Using Hydrogel Nanosilver Based Dressing Vs. Traditional Dressing: A Prospective Randomized Control Study. Pak. J. Med. Health Sci. 2021, 15, 1571–1574. [Google Scholar] [CrossRef]
  254. Verdú-Soriano, J.; Casado-Díaz, A.; de Cristino-Espinar, M.; Luna-Morales, S.; Dios-Guerra, C.; Moreno-Moreno, P.; Dorado, G.; Quesada-Gómez, J.M.; Rodríguez-Mañas, L.; Lázaro-Martínez, J.L. Hard-to-Heal Wound Healing: Superiority of Hydrogel EHO-85 (Containing Olea Europaea Leaf Extract) vs. a Standard Hydrogel. A Randomized Controlled Trial. Gels 2023, 9, 962. [Google Scholar] [CrossRef]
  255. dos Santos, T.M.; de Sousa, D.S.F.; Di Piero, K.C.; Todeschini, A.R.; Dias, W.B.; Oliveira, C.A.; dos Santos, E.P.; Monteiro, M.S.d.S.B.; Gomes, M.K.; Moura, C.d.R.; et al. Development and Clinical Application of Hydrogel Formulations Containing Papain and Urea for Wound Healing. Braz. J. Pharm. Sci. 2023, 59, e201090. [Google Scholar] [CrossRef]
  256. Barbosa, M.; Carvalho, V.; Paggiaro, A. Hydrogel Enriched With Sodium Alginate and Vitamins A and E for Diabetic Foot Ulcer: A Randomized Controlled Trial. Wounds 2022, 34, 229–235. [Google Scholar] [CrossRef]
  257. Della Pepa, G.; Lombardi, G.; Gianfrancesco, S.; Piccolo, R.; Chirico, G.; Pellegrino, M.; Santella, L.; Tecce, N.; Volpicelli, A.; Sollo, E.; et al. Triticum Vulgare Extract and Polyhexanide (Fitostimoline® Hydrogel/Fitostimoline® Plus Gauze) versus Saline Gauze Dressing in Patients with Diabetic Foot Ulcers: Results of a Randomized Controlled Trial. J. Clin. Med. 2023, 12, 3596. [Google Scholar] [CrossRef]
  258. Edmonds, M. Apligraf in the Treatment of Neuropathic Diabetic Foot Ulcers. Int. J. Low. Extrem. Wounds 2009, 8, 11–18. [Google Scholar] [CrossRef]
  259. Elsharkawi, M.; Ghoneim, B.; Westby, D.; Jones, D.; Tawfick, W.; Walsh, S.R. Adipose-Derived Stem Cells in Patients with Venous Ulcers: Systematic Review. Vascular 2023, 31, 989–993. [Google Scholar] [CrossRef]
  260. Kim, Y.H.; Shim, H.S.; Lee, J.; Kim, S.W. A Prospective Randomized Controlled Multicenter Clinical Trial Comparing Paste-Type Acellular Dermal Matrix to Standard Care for the Treatment of Chronic Wounds. J. Clin. Med. 2022, 11, 2203. [Google Scholar] [CrossRef] [PubMed]
  261. Clegg, J.R.; Adebowale, K.; Zhao, Z.; Mitragotri, S. Hydrogels in the Clinic: An Update. Bioeng. Transl. Med. 2024, 9, e10680. [Google Scholar] [CrossRef] [PubMed]
  262. Vernerey, F.J.; Lalitha Sridhar, S.; Muralidharan, A.; Bryant, S.J. Mechanics of 3D Cell–Hydrogel Interactions: Experiments, Models, and Mechanisms. Chem. Rev. 2021, 121, 11085–11148. [Google Scholar] [CrossRef] [PubMed]
  263. Richbourg, N.; Wechsler, M.E.; Rodriguez-Cruz, J.J.; Peppas, N.A. Model-Based Modular Hydrogel Design. Nat. Rev. Bioeng. 2024, 2, 575–587. [Google Scholar] [CrossRef]
  264. Li, Z.; Song, P.; Li, G.; Han, Y.; Ren, X.; Bai, L.; Su, J. AI Energized Hydrogel Design, Optimization and Application in Biomedicine. Mater. Today Bio 2024, 25, 101014. [Google Scholar] [CrossRef] [PubMed]
  265. Islamkulov, M.; Karakuş, S.; Özeroğlu, C. Design Artificial Intelligence-Based Optimization and Swelling Behavior of Novel Crosslinked Polymeric Network Hydrogels Based on Acrylamide-2-Hydroxyethyl Methacrylate and Acrylamide-N-Isopropylacrylamide. Colloid. Polym. Sci. 2023, 301, 259–272. [Google Scholar] [CrossRef]
  266. Rippon, M.G.; Fleming, L.; Chen, T.; Rogers, A.A.; Ousey, K. Artificial Intelligence in Wound Care: Diagnosis, Assessment and Treatment of Hard-to-Heal Wounds: A Narrative Review. J. Wound Care 2024, 33, 229–242. [Google Scholar] [CrossRef]
  267. Chen, M.-Y. Progress in the Application of Artificial Intelligence in Skin Wound Assessment and Prediction of Healing Time. Am. J. Transl. Res. 2024, 16, 2765–2776. [Google Scholar] [CrossRef]
Figure 1. The most relevant in vitro wound healing models. Two-dimensional models (left panel), represented by monolayer cultures of fibroblasts/keratinocytes with or without adding other cell types such as endothelial cells (for vasculature) and immune cells (for inflammation studies), and the scratch assay represent useful tools for assessment of wound healing, with multiple advantages, such as high reproducibility, simplicity, and cost-effectiveness, at the expense of limited complexity. Three-dimensional models (right panel), represented by stratified keratinocytes, reconstructed human epithelium (RHE), and full-thickness skin models (FTM), mimic the complexity of skin epidermal layers with or without the dermal layer and add other skin cells, such as endothelial, immune, or neuronal cells, to increase the physiological relevance at the expense of increased costs and enhanced production and assessment difficulties.
Figure 1. The most relevant in vitro wound healing models. Two-dimensional models (left panel), represented by monolayer cultures of fibroblasts/keratinocytes with or without adding other cell types such as endothelial cells (for vasculature) and immune cells (for inflammation studies), and the scratch assay represent useful tools for assessment of wound healing, with multiple advantages, such as high reproducibility, simplicity, and cost-effectiveness, at the expense of limited complexity. Three-dimensional models (right panel), represented by stratified keratinocytes, reconstructed human epithelium (RHE), and full-thickness skin models (FTM), mimic the complexity of skin epidermal layers with or without the dermal layer and add other skin cells, such as endothelial, immune, or neuronal cells, to increase the physiological relevance at the expense of increased costs and enhanced production and assessment difficulties.
Gels 11 00705 g001
Figure 2. Schematic diagram of the design of surgical wound models in vivo for the study of healing efficacy of different types of chemical and physical treatment options. Created with www.BioRender.com, accessed on the 9 July 2025.
Figure 2. Schematic diagram of the design of surgical wound models in vivo for the study of healing efficacy of different types of chemical and physical treatment options. Created with www.BioRender.com, accessed on the 9 July 2025.
Gels 11 00705 g002
Figure 3. Comparative wound healing timeline for different types of in vivo models. All wound models show healing and/or scarring on day 9, except for the diabetic wound. Created with www.BioRender.com, accessed on the 9 July 2025.
Figure 3. Comparative wound healing timeline for different types of in vivo models. All wound models show healing and/or scarring on day 9, except for the diabetic wound. Created with www.BioRender.com, accessed on the 9 July 2025.
Gels 11 00705 g003
Table 1. In vitro 2D models for wound healing studies.
Table 1. In vitro 2D models for wound healing studies.
Model TypeCharacteristics Advantages Limitations Typical
Applications
Monolayer Culture [152,153,154] single layer of cells on a flat surface simple, cost-effective, and easy to analyze limited cell–cell and ECM interactions basic research, drug screening
Scratch Assay [152,155,156]mechanical wound in monolayer reproducible, widely used not representative of tissue architecture migration studies, wound closure assays
Transwell Assay [131,152,157] migration through a porous membrane quantitative, adaptable no structural tissue context chemotaxis, migration analysis
Co-culture System (2D) [127,131]multiple cell types in a monolayer setup intercellular communication, easy setup hard to dissect specific pathways interaction studies, signaling analysis
Table 3. Monitoring of the wound healing main processes on 2D and 3D in vitro models.
Table 3. Monitoring of the wound healing main processes on 2D and 3D in vitro models.
Process2D Models 3D Models
Inflammation inducible by pro-inflammatory stimuli
assessment: inflammatory markers
+: reproducible
layered cells with immune-like behavior
assessment: inflammatory markers
+: better simulate real inflammatory responses
Oxidative Stress inducible by prooxidant stimuli −: source identification of the marker, donor/time-dependent
Cell migration assessment: imaging/oxidative stress markers
+: reproducible
central hypoxia increases ROS
+: reflects wound zone stress, assessment: oxidative stress markers
−: source identification of the marker, donor/time dependent
Collagen Synthesis scratch assay shows cell migration
assessment: keratinocyte or fibroblast movement over time analyzed by imaging software, in dynamics
reconstructed skin models with epidermis with or without dermis
−: difficult wound creation
−: assessment by harvesting, HP imaging for re-epithelialization at different time points, and epithelial barrier function tests
Apoptosis fibroblast cultures
assessment: protein output/imaging
−: collagen deposited in scaffold or matrix
assessment: imaging/biochemical assays
+ = advantages, − = disadvantages.
Table 4. In vivo surgical models used for experimental wound healing research.
Table 4. In vivo surgical models used for experimental wound healing research.
No.Model/SpeciesTreatmentMain Results
1FTEW dorsal area—rabbitsNanoemulsion hydrogel with Hypericum perforatum extractUpregulated growth factors, increased vessel growth, and anti-microbial properties [172]
2FTEW dorsal area—ratsRutin-loaded zein gelPromoted movement of cells towards the wound site, increased inflammation [173]
3FTEW dorsal area—ratsSodium thiosulfate gelIncreased fibroblast movement towards the wound bed, increased antioxidant enzymes [174]
4FTEW dorsal area—miceGel containing P. russeliana extract Low antioxidant effects, anti-inflammatory activity, promoted collagen formation, analgesic and pro-angiogenesis properties [175]
5FTEW dorsal area—ratsAlginate hydrogel loaded with cerium oxide nanoparticlesAccelerated wound repair by reducing oxidative stress and inflammation, faster tissue recovery [169]
6FTEW dorsal area—ratsCream containing Astragalus floccosus extractAccelerated wound healing by increased fibroblast proliferation, collagen synthesis, and re-epithelialization [176]
7FTEW dorsal neck area—on Sprague Dawley rats Sinomenine alkaloid solution extracted from Sinomenum Acutum Faster wound closure with increased fibroblast proliferation and collagen deposition, antioxidant and anti-inflammatory effects [177]
8FTEW (bilaterally punched dorsum wound) on C57BL/6 mice mouse mesenchymal stem cells from hair follicles/dermal fibroblasts/growth factorsReduced hypertrophic scarring and enhanced wound healing/combination therapy of MSCs, fibroblasts, and growth factors was the most effective [178]
9FTEW (punched dorsum wound-6 mm) on ratsSildenafil cream Faster wound closure, reduced oxidative stress [179]
10FTEW on rat and diabetic mouse, rat random flap,
partial-thickness thermal injury on guinea pig
Systemic therapy with angiotensin (1–7) Faster wound healing, increased cell proliferation without hypertensive effect, promoted regeneration, and flap survival [180]
11SIW on rats and rabbits Aloe ferox miller and Aloe Arborescens Miller whole leaf extracts Faster healing, reduced inflammation, and antibacterial and antifungal activity with no skin toxicity [181]
12FTEW dorsal area—BALB/c miceAlchemilla vulgaris and Mimosa extract mixture Rapid wound repair through improved skin re-epithelialization, higher cell proliferation, collagen synthesis, angiogenesis, and skin appendages [182]
13FTEW dorsolateral flanks—Wistar rats Ointment containing honey and Ageratum conyzoides leaf extract Increased wound contraction, faster healing, and Ageratum extract reduced inflammation but increased fibrosis despite lower fibroblast count [183]
14FTEW dorsal area—mice chitosan patch with doxycycline/zinc/selenium nanoparticlesAnti-inflammatory and hemostatic effects, enhanced blood vessel formation [184]
15Circular full-thickness skin excisions on a rat’s scalpChitosan membrane Accelerated healing, anti-inflammatory effects by reduced leukocyte counts, and increased IL-4 levels [185]
16FTEW dorsal area—diabetic Wistar rats N-acetyl cysteine
local and systemic
Reduced wound size and oxidative stress by both topical and systemic administration [186]
17FTEW dorsal area—diabetic Wistar rats Paeoniflorin-loaded hyaluronic acid hydrogel Accelerated wound repair by anti-inflammatory effects mediated by modulation of macrophage populations (M1 towards M2 phenotype), enhanced angiogenesis, and collagen production [187]
18FTEW dorsal area—nude mice Heparinized adipose-derived scaffolds enriched with growth factors Improved re-epithelialization, angiogenesis, and skin appendage regeneration through enhanced fibroblast migration and blood vessel growth [188]
19FTEW dorsal area—mice injection of mesenchymal Stem cell-derived extracellular vesicles/umbilical cord blood-derived extracellular vesicles in the wound margins Enhanced wound healing by increased tissue growth and reduced scar formation [189]
20FTEW dorsal area—ratsNeodymium–yttrium–aluminum garnet (Nd:YAG) pulsed high-intensity laser Improved wound healing by increasing fibroblast proliferation, collagen synthesis, and thickness of the granular layer [190]
21SI dorsal area—KO mice (double deletion of IL-10 and IL-4 genes)Interleukin-10, local application Improved wound healing, reduced inflammation, and enhanced scarring quality [191]
22FTEW dorsal area—mice Extracellular matrix/stromal vascular fraction gel conditioned medium Faster wound closure, higher collagen deposition, increased growth factor secretion, and higher fibroblast and keratinocyte count and activity [192]
23FTEW dorsal area—mice Encapsulated Spirulina protein hydrolysates with nanoliposomes Increased wound closure in mice by boosting fibroblast growth, skin regrowth, and enhanced markers of angiogenesis and collagen deposition [193]
24FTEW dorsal area—mice Silk protein-biomaterial wound dressings with epidermal growth factor and silver sulfadiazine Better wound closure rate, less scarring, and collagen production [194]
25FTEW dorsal area—C57 diabetic mice Intermittent fasting and pulsed radiofrequency energy Combined therapy exhibited antioxidant effects and induced faster wound healing by boosting cell migration, angiogenesis, and nerve growth [195]
26Full-thickness SIW dorsal area—albino guinea pigs Pulsed electrical stimulation Both anodal and cathodal electrical stimulation boosted wound recovery in guinea pigs, improving closure and scar strength, regardless of polarity sequence [196]
FTEW = Full-thickness excisional wound, SIW = surgical incision wound.
Table 2. In vitro 3D models for wound healing studies.
Table 2. In vitro 3D models for wound healing studies.
Model Type Characteristics Advantages Limitations Typical
Applications
3D Tissue Constructs [152]multiple cell layers in the ECM better tissue architecture, more realistic higher complexity, longer setup advanced research, tissue engineering
Co-culture System (3D) [155,156]different cell types embedded in or layered within a 3D matrix mimics natural skin structure and function technically demanding ECM remodeling, inflammatory studies
Hydrogel-based Models [30,131,158]cells in/around gels like collagen or
Matrigel
ECM simulation, tunable stiffness batch variability, limited duration ECM interaction, scaffold testing
Organotypic Culture [158]stratified epidermis + fibroblast-populated dermis closest to in vivo skin time-consuming, resource-intensive preclinical drug testing, epidermal healing
Explant Culture [159,160] full-thickness skin cultured ex vivo preserves native structure short-term viability, donor variability re-epithelialization, tissue remodeling
Microfluidic Skin-on-a-Chip [156,158]skin model in a microfluidic device with a controlled environment real-time imaging, dynamic flow conditions expensive, specialized equipment needed high-resolution studies, mechanistic modeling
Bioprinted Skin Constructs [161]layered printing of cells and ECM components reproducible, customizable architecture high cost, needs specialized skills personalized modeling, drug/cosmetic testing
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Baldea, I.; Grosu, I.G.; Ghafury, S.; Golat, C.; Doubali, D.; Vestemean, A.-M.; Cedorge, A.N.; Florian, I.; Yiannoulatos, M.; Wajahat, M.M.; et al. Biological Models for Evaluating Hydrogel-Based Formulations in Wound Healing. Gels 2025, 11, 705. https://doi.org/10.3390/gels11090705

AMA Style

Baldea I, Grosu IG, Ghafury S, Golat C, Doubali D, Vestemean A-M, Cedorge AN, Florian I, Yiannoulatos M, Wajahat MM, et al. Biological Models for Evaluating Hydrogel-Based Formulations in Wound Healing. Gels. 2025; 11(9):705. https://doi.org/10.3390/gels11090705

Chicago/Turabian Style

Baldea, Ioana, Ioana Georgeta Grosu, Sahar Ghafury, Cristian Golat, Doriane Doubali, Ana-Maria Vestemean, Aris Nicolas Cedorge, Ilinca Florian, Michael Yiannoulatos, Muhammad Mudassir Wajahat, and et al. 2025. "Biological Models for Evaluating Hydrogel-Based Formulations in Wound Healing" Gels 11, no. 9: 705. https://doi.org/10.3390/gels11090705

APA Style

Baldea, I., Grosu, I. G., Ghafury, S., Golat, C., Doubali, D., Vestemean, A.-M., Cedorge, A. N., Florian, I., Yiannoulatos, M., Wajahat, M. M., Silli, L. R., Stavrou, T., & Mitrea, D. R. (2025). Biological Models for Evaluating Hydrogel-Based Formulations in Wound Healing. Gels, 11(9), 705. https://doi.org/10.3390/gels11090705

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop