Next Article in Journal
Preparation and Evaluation of Long-Acting Injectable Levocetirizine Prodrug Formulation
Previous Article in Journal
Calcium Electrochemotherapy and Challenges in Combined Treatment with Dendritic Cell Vaccination
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Probiotics in Nanotechnology-Driven Wound Healing: From Mechanistic Insight to Clinical Promise

1
Department of Pharmaceutics, Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur 441002, Maharashtra, India
2
Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh 11623, Saudi Arabia
3
Department of Quality Assurance, Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur 441002, Maharashtra, India
4
Department of Pharmaceutics, Sinhgad College of Pharmacy, Vadgaon (bk), Pune 411041, Maharashtra, India
*
Author to whom correspondence should be addressed.
Pharmaceutics 2025, 17(7), 805; https://doi.org/10.3390/pharmaceutics17070805 (registering DOI)
Submission received: 16 May 2025 / Revised: 17 June 2025 / Accepted: 18 June 2025 / Published: 21 June 2025
(This article belongs to the Topic Probiotics: New Avenues)

Abstract

:
Chronic wounds, including diabetic foot ulcers and pressure sores, are becoming more prevalent due to aging populations and increased metabolic problems. These wounds often persist due to impaired healing, chronic inflammation, oxidative stress, and infections caused by multidrug-resistant pathogens, making conventional treatments—including antibiotics and antiseptics—largely inadequate. This creates an urgent need for advanced, biologically responsive therapies that can both combat infection and promote tissue regeneration. Probiotics have surfaced as a viable option owing to their capacity to regulate immune responses, impede pathogenic biofilms, and generate antibacterial and antioxidant metabolites. However, their clinical application is limited by poor viability, sensitivity to environmental conditions, and short retention at wound sites. Nanotechnology-based delivery systems address these limitations by protecting probiotics from degradation, enhancing site-specific delivery, and enabling controlled, stimuli-responsive release. Encapsulation techniques using materials like chitosan, PLGA, liposomes, nanogels, nanofibers, and microneedles have shown significant success in improving wound healing outcomes in preclinical and clinical models. This review summarizes the current landscape of chronic wound challenges and presents recent advances in probiotic-loaded nanotechnologies. It explores various nano-delivery systems, their mechanisms of action, biological effects, and therapeutic outcomes, highlighting the synergy between probiotics and nanocarriers as a novel, multifaceted strategy for managing chronic wounds.

1. Introduction

Chronic wounds, including diabetic foot ulcers (DFUs), venous leg ulcers (VLUs), and pressure ulcers, pose significant public health issues, particularly for the elderly or individuals with diabetes mellitus [1]. Epidemiological research shows that chronic wounds impact approximately 2.21 out of every 1000 people in developed nations, while DFUs affect up to 25% of those with diabetes and are responsible for 85% of diabetes-related amputations globally [2]. In the United States, the financial impact of chronic wounds exceeds USD 25 billion annually [3]. According to estimates, between 1% and 2% of people worldwide will sustain a chronic wound at some time in their lives. Approximately 8.2 million people, irrespective of infection status, suffered wounds, according to a 2018 retrospective investigation. Acute and chronic wound care under Medicare was estimated to cost between USD 28.1 billion to USD 96.8 billion. Between 1.5 and 2 million people in Europe are thought to have either acute or chronic wounds [4].
Chronic wounds are often marked by prolonged and excessive inflammation, hindered angiogenesis, microbial imbalance, and the formation of pathogen biofilms [5]. It is estimated that biofilms are present in approximately 78% of wounds that do not heal, providing a refuge for pathogens such as Staphylococcus aureus, Enterococcus faecalis, Escherichia coli, Pseudomonas aeruginosa, Klebsiella pneumoniae, and Acinetobacter baumannii, where they remain protected from the patient’s immune defenses and medical treatments. The global increase in antimicrobial resistance (AMR), particularly multi-drug resistance (MDR), worsens the situation, as the vast majority of chronic wound infections are caused by these resistant strains [6,7].
Although traditional antibiotic treatments have been central to managing wound infections, they come with several drawbacks [8]. Antibiotic overuse and improper dosing can also result in systemic adverse effects and delayed recovery because of collateral tissue damage [9]. In contrast, probiotics could serve as a useful supplement or alternative for managing chronic wounds [10,11].
Probiotics as a therapeutic option often face challenges due to their sensitivity to oxygen, heat, and the gastrointestinal environment, as well as their limited viability and retention at wound sites [12]. To address these issues, the use of nanotechnology for delivering probiotics has been suggested as an effective approach to enhance their stability, bioavailability, and targeted delivery [13].
Nanocarriers, such as liposomes, nanoparticles, polymeric hydrogels, and electrospun nanofibers, offer advantages like controlled release rates, protection against external degradation, and strong adhesion to moist wound surfaces [14,15]. In 2024, murine models infected with S. aureus showed 85% complete wound closure within a week after using a Lactobacillus (formerly designated Lactobacillus genus) microneedle patch, outperforming standard topical antibiotics [16]. Similarly, chitosan nanoparticles loaded with cell-free supernatants of Lacticaseibacillus rhamnosus (formerly designated as Lactobacillus rhamnosus) achieved over 95% inhibition of both mono- and dual-species biofilms without inducing resistance in vitro [17]. While previous reviews have either focused on probiotics [18] or nanotechnology-based approaches individually in the context of wound healing, or discussed their therapeutic benefits in broader terms, only a limited number have thoroughly addressed their integrated use for chronic wound management [19]. For instance, the wound-healing potential of probiotics and how nanotechnology may enhance their delivery and efficacy has been discussed in [20]. However, a dedicated review that critically evaluates the synergistic application of probiotics and nanotechnology—particularly through novel delivery platforms such as hydrogels, microneedles, or electrospun fibers—remains scarce [21]. This review therefore aims to bridge that gap by comprehensively exploring the mechanistic pathways, preclinical and clinical advances, and translational challenges of using probiotic-based nanotherapeutics for chronic wound healing.
Additionally, probiotics, extracellular vesicles, and smart biosensor-assisted probiotic dressings are emerging therapeutic approaches in regenerative medicine, yet they remain relatively unfamiliar. In this review, we critically examine the availability of probiotics as agents for wound healing and their integration with nanotechnology [22]. This combination of probiotics and nanotechnology offers a novel and potentially intricate pathway to address the limitations of existing wound therapies and to provide solutions for the increasing population with MDR-associated chronic wounds [23].

2. Pathophysiology of Chronic Wounds

Hemostasis, inflammation, proliferation, and remodeling are all components of the structured biological process that is wound healing. This process leads to successful tissue regeneration for acute wounds. On the other hand, chronic wounds—like DFUs, pressure ulcers, and VLUs—show reduced capacity to heal, mostly because of prolonged inflammatory phases, microbial colonization, and problems with angiogenesis [24]. Chronic wounds pose serious health risks and last longer than three months, especially for people with diabetes, vascular insufficiency, or restricted movement [25].
The overproduction of pro-inflammatory cytokines, such as TNF-α, IL-1β, and IL-6, together with increased levels of matrix metalloproteinases (MMPs) and reactive oxygen species (ROS), are characteristics of chronic wounds. Uncontrolled MMP production leads to extracellular matrix degradation and growth factor activity inhibition, whereas ROS-induced damage impairs fibroblast migration, keratinocyte proliferation, and endothelial function [26,27]. A fundamental factor in the persistence of wounds that do not heal is oxidative stress. Because of mitochondrial dysfunction, reactive oxygen species (ROS) generation and the creation of advanced glycation end-products (AGEs) are elevated in hyperglycemic situations, especially in diabetic wounds [28]. The accumulation of AGEs further exacerbates wound healing by promoting inflammation, impairing collagen synthesis, and disrupting cellular signaling pathways [29]. Elevated AGEs have been shown to enhance the formation of cross-links in extracellular matrix proteins, leading to the stiffening of the wound bed, which delays tissue remodeling and prolongs the chronicity of wounds [30]. Diabetic mice exhibited approximately a 2.7-fold increase in ROS compared to control groups, along with significantly reduced VEGF expression and epithelialization, p < 0.01 for both. These findings align with clinical evidence showing reduced neovascularization and less granulation tissue in biopsies from diabetic ulcers [31].
The presence of bacteria, frequently arranged in biofilms—dense collections of microorganisms encased in an extracellular matrix that generates itself—defines chronic wounds [32]. Biofilms are detected in about 78% of chronic wound samples, significantly hindering immune system clearance and increasing antibiotic tolerance by over 1000 times [33]. The biofilm’s matrix facilitates gene exchange, including that which causes antibiotic resistance, and serves as a barrier against antimicrobial drugs [34]. Staphylococcus epidermidis is among the most concerning pathogens, typically a harmless organism but increasingly prevalent as an infectious agent in chronic wounds due to its biofilm-forming ability and genetic adaptability [35]. In a metagenomic analysis, it was discovered that S. epidermidis strains from venous leg ulcers that expressed virulence genes linked to adhesion (icaA, aap), biofilm persistence, and antimicrobial resistance (mecA, mupA) [19]. These S. epidermidis strains triggered elevated levels of IL-1β and IL-8 in ex vivo human wounds and were linked to non-healing wounds in 83% of patients [36].
Chronic wounds are linked to significant dysbiosis in the microbiome, marked by reduced microbial diversity and an abundance of harmful bacteria like P. aeruginosa, E. faecalis, and S. aureus [37]. There is a notable decline in beneficial bacteria such as Cutibacterium acnes and various species of formerly designated Lactobacillus like Lentilactobacillus crispatus, which play roles in maintaining the skin barrier and modulating the immune system. This reduction in microbial presence alters the local equilibrium, undermines the interaction between host keratinocytes and immune cells, disrupts the host’s antimicrobial capabilities, and results in persistent inflammation [38].
The importance of the skin microbiota in regulating local immunity is now generally acknowledged. Commensal organisms can boost the production of antimicrobial peptides (AMPs), improve the integrity of epithelial tight junctions, and affect toll-like receptor (TLR) signaling [39]. In the absence of regulation, the wound bed can become overrun by polymicrobial communities, leading to the activation of immune tolerance and a reduced neutrophil response, as seen in chronic wounds with dual-species biofilms [40].
Chronic wounds are caused by a complex disruption in immune regulation, microbial equilibrium, and oxidative balance. This leads to a persistently inflamed microenvironment characterized by microbial biofilms and cellular aging, which creates a niche resistant to conventional treatments [5]. These complexities highlight the need for multimodal interventions that can potentially restore the microbiome, regulate immune responses, and overcome resistance mechanisms [41]. The combined use of probiotics and nanotechnology, discussed in later sections, offers a promising strategy to tackle these interconnected pathophysiological challenges through a mechanism-based and targeted therapeutic approach [42].

3. Role of Probiotics in Wound Healing

Recent studies have illuminated the role of probiotics in promoting wound healing. For instance, probiotics exhibit antimicrobial effects by producing substances like bacteriocins, organic acids such as lactic and acetic acids, and hydrogen peroxide, which inhibit the growth of harmful pathogens. They also impact the immune system by influencing cytokine production, changing macrophage polarization, and adjusting dendritic cell function, thereby reducing inflammation, as shown in Figure 1. Furthermore, probiotics reduce reactive oxygen species (ROS) levels to lessen oxidative stress and improve the epithelial barrier by increasing the production of tight junction proteins. Furthermore, they stimulate fibroblasts and encourage the production of new blood vessels, which aids in the growth of granulation tissue [43,44].
A significant study from 2022 revealed that applying Lactiplantibacillus plantarum (formerly designated as Lactobacillus plantarum) topically to skin wounds in mice resulted in a 42% reduction in wound size by the seventh day compared to the control group (p < 0.01) [45]. In the wound microenvironment, this result was associated with a decrease in neutrophil infiltration and an increase in modifying growth factor-beta 1 (TGF-β1) and vascular endothelial growth factor (VEGF) levels [46]. Additional research has underscored the importance of L. plantarum in healing diabetic wounds. In vivo studies using diabetic rat models shown that L. plantarum facilitates wound healing by regulating inflammation, namely by preventing the activation of the NLRP3 inflammasome and pyroptosis pathways brought on by AGEs. Increased re-epithelialization and tissue regeneration are the outcomes of this modification [47].
Innovative formulations that combine L. plantarum with other substances have demonstrated synergistic effects. For instance, research on a honey—L. plantarum mixture revealed that this combination effectively inhibited the growth of harmful bacteria like S. aureus, P. aeruginosa, and E. coli [48]. This formulation disrupted biofilm formation and reduced the expression of quorum sensing genes, thereby boosting antibacterial activity and aiding wound healing in infected rat models. Progress in probiotic-based wound care has also resulted in the creation of new delivery systems [49]. A study conducted in 2025 introduced an in-situ gelling formulation aimed at preserving probiotics for wound healing purposes [50]. This formulation supports the continuous production of antimicrobial agents by metabolizing added glycerol, thus speeding up the healing process of infected wounds [51].
Research continues to advance in demonstrating the benefits of probiotics in clinical environments, applicable to both animal and human studies [52]. Diabetic rats receiving oral supplementation with Lacticaseibacillus casei (formerly designated as Lactobacillus casei) experienced faster wound contraction, a 2.3-fold increase in type I collagen deposition, and enhanced histological wound scores by the 10th day of recovery (p < 0.01) [53]. Additionally, in a comparable model, the oral administration of L. rhamnosus GG was found to restore angiogenic signaling in wounds, evidenced by elevated expression of HIF-1α and mRNA expression of VEGF [54].
Probiotics applied topically have produced significant clinical evidence supporting their effectiveness. In a review that examined both human and animal studies, it was concluded that the topical use of Lactobacillus strains led to improved re-epithelialization and decrease in bacterial colonization, with no immune reactions observed in the vast majority (over 90%) of the cases studied [18].
Probiotic therapy’s capacity to affect immune responses in chronic wounds is one of its main advantages. By increasing anti-inflammatory substances like IL-10 and decreasing pro-inflammatory cytokines, probiotics aid in the reduction of inflammation. Furthermore, they encourage macrophages to change from the M1 to the M2 phenotype, which is crucial for reducing inflammation and promoting tissue healing [55].
Lactobacillus spp. function as immune modulators in diabetic foot ulcers by reducing IL-1β and TNF-α levels while also reestablishing TLR2/4 balance in keratinocytes. Biofilm development on chronic wounds frequently plays a significant role in hindering the healing process [56]. Probiotics are known for their strong antibiofilm properties. Cell-free supernatants derived from L. rhamnosus GG were able to inhibit the formation of biofilms by S. aureus and P. aeruginosa by more than 95%, without causing resistance even after prolonged exposure [57,58]. Beyond preventing biofilm formation, these supernatants also lowered the levels of quorum sensing associated with biofilms and reduced the expression of biofilm-related genes (icaA and lasR), indicating a promising alternative for addressing persistent biofilm infections without the use of conventional antibiotics [59].
Limosilactobacillus reuteri (formerly known as Lactobacillus reuteri) is another probiotic that shows promise due to its production of reuterin and other antimicrobial peptides (AMPs), which can selectively target wound pathogens while sparing beneficial skin microbiota [60]. This targeted antimicrobial effect of probiotics allows for the specific treatment of wound pathogens, unlike antibiotics, which often act in a non-specific way. Probiotics do not cause further dysbiosis, a problem associated with broad-spectrum antibiotics that can delay the wound healing process [12].
Recent research suggests that postbiotics, such as bacterial lysates, metabolites, and heat-killed strains, along with Extracellular Vesicles (EVs), might offer probiotic-like benefits with greater stability. For instance, EVs derived from L. plantarum have been found to enhance keratinocyte migration and reduce MMP-9 expression in vitro, promoting wound healing without the risk associated with live cell delivery [61,62].

3.1. Mechanism of Action of Probiotics Against Pathogens with Specific Strains

Probiotics help shield the host from harmful infections, particularly when it comes to chronic wounds, by acting as antimicrobials through a variety of ways. Although these mechanisms depend on strain, they frequently include several complementary actions [63].
The synthesis of antimicrobial compounds is one of the main ways probiotics fight against infections. Bacteriocins, organic acids (such lactic and acetic acids), and hydrogen peroxide are produced by strains such as L. rhamnosus GG and L. plantarum, which lower pH and damage the integrity of dangerous pathogen cells [64,65]. Probiotics are an efficient treatment option for preventing infection in chronic wounds because of these compounds, which stop the growth of bacteria like S. aureus and E. coli [66].
In addition to antimicrobial production, probiotics also support the immune system by modulating the host’s immune response. They influence cytokine production, alter macrophage polarization, and adjust dendritic cell activity [67]. For example, L. plantarum and Bifidobacterium longum can modulate the immune response, which helps reduce inflammation and promotes pathogen clearance. Additionally, by boosting the expression of tight junction proteins, which are essential for preserving the integrity of the skin or gut lining, probiotics such as L. casei and L. rhamnosus GG aid in strengthening the epithelial barrier. This lowers the chance of infection by making it more difficult for pathogens to infiltrate [68,69].
Probiotics such as L. plantarum and L. rhamnosus GG can increase the release of anti-inflammatory cytokines like IL-10 while decreasing the production of pro-inflammatory cytokines like TNF-α, IL-6, and IL-1β. Because it aids in the resolution of the inflammatory phase and encourages tissue regeneration, this anti-inflammatory impact is essential for chronic wound healing [70].
Probiotics play an essential role in restoring microbial homeostasis. By outcompeting harmful pathogens and fostering beneficial bacteria, probiotics help maintain a balanced microbial ecosystem on the skin or within the gut, which is crucial for healing chronic wounds. Lactobacillus spp. (formerly designated Lactobacillus genus) and Bifidobacterium spp. contribute to this balance, improving resistance to infection and supporting the overall healing process [71].

3.2. Clinical Evidence and Human Trials

A preliminary study by Stuermer et al. investigated the effects of oral multispecies probiotics, including L. plantarum, L. rhamnosus, and Bifidobacterium bifidum, on 20 diabetic patients with chronic, difficult-to-heal wounds. These patients, who lived in a long-term care facility, took the probiotics for a duration of six months. Out of the 20 participants, only 13 completed the study, and all remained quite ill. Nevertheless, among these 13 individuals, 5 experienced complete wound closure, and most reported improvements in a wound-specific quality of life measure, particularly in terms of pain and mobility. The study also noted a simultaneous decrease in the microbial load of S. aureus and P. aeruginosa, along with a partial restoration of commensal populations following the use of probiotics [72].
Numerous studies have reported similar results in non-diabetic patients. For instance, a randomized trial conducted in 2023 found that applying Limosilactobacillus fermentum (formerly known as Lactobacillus fermentum) topically to burn wounds reduced healing time by 21% compared to silver sulfadiazine (p < 0.05), while systemic treatment led to a decrease in local cytokines IL-6 and TNF-α [73]. Table 1 provides a summary of key preclinical and clinical studies examining the impact of various probiotic strains on wound healing. It outlines the delivery methods, mechanisms of action, and significant outcomes observed in different models.

4. Nanotechnology in Wound Healing and Probiotic Delivery

Nanotechnology focuses on creating materials that are less than 100 nm in size, which allows for high surface-area-to-volume ratios, precise therapeutic delivery, and controlled-release mechanisms. These features are particularly effective in tackling the complex nature of chronic wounds, which often require managing infection, reducing inflammation, and promoting tissue regeneration simultaneously [79]. Researchers have explored various nanomaterials for their potential in wound treatment, including polymeric nanoparticles like chitosan and PLGA, lipid-based carriers such as solid lipid nanoparticles, liposomes, and inorganic nanoparticles like silver, zinc oxide and gold [80].
To maintain the viability of probiotics in challenging biological settings, various nanoencapsulation methods have been devised, as shown in Figure 2. These methods encompass nanoemulsions, polyelectrolyte complexation, spray-dried or freeze-dried nanobeads, and electrospun nanofibers [81].
Nanoemulsions are thermodynamically stable colloidal systems formed by dispersing one liquid phase into another (typically oil-in-water) using surfactants, resulting in droplets ranging from 20 to 200 nm. These systems protect probiotic cells by encapsulating them in the dispersed phase, thus shielding them from oxidative degradation, acidic environments, and enzymatic hydrolysis. Their small droplet size ensures enhanced mucosal or dermal penetration, while the high surface-area-to-volume ratio promotes sustained and localized release of viable microorganisms. In the context of wound healing, probiotic-loaded nanoemulsions have demonstrated accelerated wound closure, reduced pathogen colonization, and improved re-epithelialization in preclinical models [82,83,84].
Spray-dried and freeze-dried nanobeads represent another widely employed method for improving probiotic stability. A probiotic-polymer solution is compressed into a hot air stream during the spray drying process, which quickly evaporates the solvent and creates solid nanobeads. In contrast, freeze drying preserves cellular viability and structure by sublimating frozen aqueous probiotic compositions under low pressure. At room temperature, the resultant dry powders are very stable and can be rehydrated as needed [85,86]. These nanobeads are ideal for incorporation into hydrogels, ointments, or dry wound dressings. Studies have shown that probiotics preserved using these techniques retain over 80–90% viability for several months and exhibit effective immunomodulatory and antimicrobial actions when reintroduced into wound environments [87].
Electrospun nanofibers provide a sophisticated platform that mimics the extracellular matrix of human skin, providing both structural support and controlled probiotic release. In this technique, probiotic-laden polymer solutions—composed of biodegradable materials like gelatin, polycaprolactone (PCL), or polyvinyl alcohol (PVA)—are subjected to high-voltage electric fields to form ultrafine fibers collected into porous mats. These nanofibers offer high porosity and surface area, promoting gas exchange, moisture retention, and cell migration—all critical elements in wound healing [88]. Embedded probiotics within the fibers are protected from desiccation, UV exposure, and mechanical damage, while sustained release over time ensures prolonged microbial activity. Recent research has shown that electrospun nanofibers loaded with L. plantarum significantly improved healing outcomes in diabetic wound models, with enhanced granulation tissue formation and reduction of biofilm-forming pathogens [89].
Recent studies have demonstrated that encapsulation of L. plantarum using biopolymers such as chitosan, alginate, and gelatin significantly enhances its viability and stability. For instance, L. plantarum encapsulated within alginate-gelatin or transglutaminase-crosslinked gelatin hydrogels showed improved resistance to gastrointestinal stress and prolonged storage stability, maintaining high viability for up to 28 days at refrigerated conditions [90]. These encapsulation strategies have proven effective in preserving probiotic functionality and supporting their therapeutic potential in wound healing applications [91].
A novel approach involved encapsulating L. reuteri in PLGA nanoparticles, enabling a sustained release over 72 h. In vivo experiments revealed that this formulation notably reduced the density of S. aureus biofilms in wounds with active infections. These examples underscore how nanotechnology, particularly through the encapsulation of probiotics, is revolutionizing wound care by enhancing the stability, efficacy, and targeted delivery of bioactive compounds [91].

4.1. Advanced Probiotic-Nano Systems

Recent advancements have transitioned from basic encapsulation to bioresponsive and multifunctional systems. Microneedle (MN) patches containing live L. rhamnosus GG outperformed traditional antibiotic treatments for S. aureus-infected wounds in mice, with 85% of treated cases fully healing within seven days, demonstrating remarkable safety and effectiveness. Even after 60 days of cold storage, these MN patches maintained over 80% bacterial viability [92].
Unlike live probiotics, postbiotics—comprising inactivated bacterial cells, metabolites, and cell-free supernatants—are more chemically stable and pose no risk of microbial overgrowth [93]. Recent studies have focused on encapsulating these bioactive molecules within nanocarriers to enhance their therapeutic delivery. One study involved cell-free extracts of L. fermentum encapsulated in a hydrogel with zinc oxide nanoparticles, resulting in a 2.4-fold increase in re-epithelialization compared to the control and a 48% reduction in IL-1β concentration in burn models [94]. Stimuli-responsive carriers, such as pH or ROS-sensitive nanogels, release probiotics or their bioactive components within the local wound environment. In 2023, researchers developed a glucose-responsive delivery system for L. acidophilus that released hydrogen peroxide in hyperglycemic wounds, promoting angiogenesis and reducing bacterial load [95].

4.1.1. Chitosan Nanoparticles

Because of its inherent antibacterial, biodegradable, and biocompatible qualities, chitosan—a naturally occurring polysaccharide produced from chitin—has attracted a lot of interest in wound healing applications. Chitosan’s cationic properties enable it to interact with negatively charged microbial cell membranes, increasing its antibacterial effectiveness [96,97]. Moreover, chitosan promotes hemostasis and supports cellular proliferation and migration, essential for tissue regeneration. Nanoparticulate chitosan exhibits a higher surface area-to-volume ratio, facilitating better interaction with biological tissues and more efficient drug delivery [98]. In one study, chitosan nanoparticles containing the antibiotic cefepime were created and incorporated into hydrogel membranes. In rat models, these membranes showed considerable antibacterial action against common wound pathogens, controlled medication release, and hastened wound closure [99].
Chitosan, despite its biocompatibility and mucoadhesiveness, faces significant practical limitations that restrict its use as the main encapsulating matrix. First, its poor solubility at neutral-to-alkaline pH remains a major challenge. Even with chemical modifications like N-methylene phosphonic acid chitosan (NMPC-GLU) improving solubility, these derivatives are still being developed and lack extensive validation in primary encapsulation systems [100]. Second, mechanical instability is widely reported. A 2022 review noted that chitosan microparticles required multiple layers—such as pectin–chitosan–collagen—to maintain structural integrity, indicating its insufficiency when used alone. Third, limited drug loading and burst release issues are repeatedly observed. For example, a 2022 study on ghrelin-loaded chitosan liposomes highlighted that PLGA outperformed chitosan-based carriers in encapsulation efficiency, sustained release, and protection against enzymatic degradation [101]. Fourth, stability challenges in physiological environments persist: pulmonary delivery systems doped with chitosan exhibit variable performance and depend heavily on synthetic modifiers to stabilize drug delivery. Last, despite frequent use in food-grade encapsulation (e.g., essential oils), chitosan-based nanocarriers continue to face reproducibility and scalability concerns, even in a well-controlled 2022 study [102].

4.1.2. PLGA Nanoparticles

A biodegradable and biocompatible copolymer, poly(lactic-co-glycolic acid) (PLGA) is widely used in drug delivery systems because of its excellent breakdown kinetics and capacity to encapsulate a variety of therapeutic compounds [103]. PLGA nanoparticles have been used in wound healing applications to improve therapeutic outcomes by enhancing the stability, controlled release, and targeted delivery of bioactive chemicals. The effectiveness of PLGA nanoparticles in delivering growth factors to support angiogenesis and tissue regeneration has been shown in recent investigations [104]. For example, a study created PLGA nanoparticles that co-encapsulated the genes for vascular endothelial growth factor A (VEGFA) and basic fibroblast growth factor (bFGF) [105]. Rats with deep burn wounds treated with these nanoparticles showed improved granulation tissue development, faster wound healing, and more epithelialization than controls [106].
In a different study, PLGA nanospheres loaded with curcumin were created and incorporated into a topical nanogel. In Wistar rats, this formulation demonstrated strong anti-inflammatory properties and accelerated the healing of cutaneous wounds, demonstrating the potential of PLGA nanoparticles to deliver phytochemicals for skin regeneration [107]. Furthermore, Ref. [108] synthesized polymeric curcumin nanospheres using PLGA, which demonstrated potent antibacterial activity and effectively inhibited lysozyme aggregation. These nanospheres facilitated wound healing in Drosophila melanogaster models, suggesting their applicability in managing microbial infections in wounds.

4.1.3. Nanofibers

Because of their structural resemblance to the extracellular matrix (ECM), high surface-area-to-volume ratio, and ability to transport drugs continuously, nanofibers—which are usually made by electrospinning—have become extremely promising scaffolding materials for wound healing [109]. These features allow nanofibers to promote cellular adhesion, proliferation, and migration while offering excellent control over the local wound microenvironment. Electrospun nanofibers can effectively encapsulate probiotics and maintain their bioactivity [110]. For example, Ref. [111] developed polycaprolactone (PCL)/gelatin nanofibers loaded with L. rhamnosus GG, achieving over 70% bacterial viability over a 10-day period while significantly improving the tensile strength and epithelial regeneration in rat wound models.
Recent investigations further validate this approach. Xu et al. successfully prepared pectin/PVA nanofibers encapsulating L. rhamnosus 1.0320 via electrospinning. The resulting fibers demonstrated strong preservation of bacterial viability—89.65% immediately after fabrication and 85.32% after 21 days of storage at 4 °C—confirming the matrix’s capacity to maintain long-term functionality of probiotics in dry formulation [112]. In another study, Stojanov et al. developed PEO-based nanofibers incorporating genetically engineered vaginal Lactobacillus (formerly designated Lactobacillus genus) strains. These fibers showed rapid dissolution (complete in 30–45 min), high bacterial viability, and no hemolytic or cytotoxic effects. Moreover, the fluorescent and luminescent markers allowed real-time tracking of bacterial adhesion to Caco-2 epithelial cells, with adhesion rates ranging from 0.5 to 50% depending on the strain. These findings highlight both the safety and precision of nanofiber-based probiotic delivery systems for mucosal and wound applications [113].
Further, hybrid nanofiber systems have been explored for co-delivery of probiotics with growth factors or postbiotics [114]. The versatility of nanofiber compositions—ranging from synthetic polymers like PCL and polylactic acid (PLA) to natural biopolymers such as gelatin and collagen—has enabled researchers to tailor wound dressings to specific wound types and healing phases. Some designs also incorporate layered or core–shell architectures, allowing sequential release of bioactives [115]. Nanofibers represent a sophisticated and flexible platform for the topical delivery of probiotics and other wound-healing agents. Their ability to mimic native tissue architecture while enabling localized and sustained therapeutic delivery makes them ideal for chronic and infected wound management [116].

4.1.4. Nanobeads

Nanobeads are spherical, nano-sized particles commonly fabricated from biodegradable polymers and are used to encapsulate bioactive compounds, including probiotics like L. reuteri, L. fermentum, and Bacillus subtilis, enzymes like NanoLuc luciferase, and postbiotics like organic acids and reuterin [117]. These structures are advantageous due to their uniform morphology, high encapsulation efficiency, and ability to protect labile biological agents from harsh environmental conditions such as gastric pH, oxidative stress, and temperature fluctuations. In the context of wound healing, nanobeads serve as effective delivery systems that prolong the bioactivity of encapsulated compounds at the wound site [118].

4.1.5. Microneedles

To provide bioactive substances painlessly straight into the dermal layer, microneedles (MNs) are a minimally invasive and highly targeted delivery system that can penetrate the stratum corneum without entering deeper nerve-rich regions [119]. Adding probiotics or their bioactive derivatives to microneedle systems has become a novel way to speed up wound healing in recent years, particularly for chronic or infected wounds [120].
One of the most significant advancements was reported by [120], who developed a biodegradable microneedle patch encapsulating live L. rhamnosus GG within a polyvinyl alcohol (PVA)/hyaluronic acid matrix. When applied to S. aureus-infected wounds in mice, this system achieved complete wound closure in 85% of cases within seven days—markedly outperforming traditional topical antibiotics. Importantly, the MNs retained over 80% bacterial viability after 60 days of cold storage, underscoring their long-term stability.
The capacity of probiotic-loaded MNs to introduce live bacteria straight into the wound bed, where they can influence immune responses, promote re-epithelialization, and have localized antimicrobial effects, is the mechanism underlying their effectiveness. Specifically, it is known that the LGG strain reduces local inflammation by enhancing regulatory T-cell responses and secreting antimicrobial peptides [54].

4.1.6. Postbiotic Encapsulation

Postbiotic-loaded nanogels, which deliver bacterial metabolites instead of live bacteria, have been evaluated in clinical models. Postbiotic-based nanodelivery systems have emerged as a distinct and promising strategy in wound care [121]. Recent studies have emphasized the importance of encapsulating postbiotics within nanocarriers to protect them from degradation, prolong their bioavailability, and enable targeted release at the wound site [122]. In a rat burn model, this postbiotic formulation accelerated wound closure by 2.4-fold compared to the control and significantly decreased pro-inflammatory IL-1β levels by 48%, suggesting strong immunomodulatory effects [123].

4.1.7. Other Nanocarriers and Emerging Approaches

In addition to well-studied platforms like chitosan, PLGA, hydrogels, nanofibers, nanobeads, microneedles, and postbiotic systems, a range of emerging nanocarriers is being explored to overcome the limitations of conventional wound care and probiotic delivery. These systems emphasize improved stability, site-specific targeting, and multifunctional therapeutic performance.
Lipid-based nanocarriers, such as liposomes and solid lipid nanoparticles (SLNs), have demonstrated potential for encapsulating both live probiotics and postbiotic components. A recent study by [124] designed liposome-encapsulated L. rhamnosus GG, showing prolonged bacterial viability and enhanced adhesion to keratinocytes in vitro. When tested in a diabetic wound model, these liposomes facilitated a 1.8-fold acceleration in epithelial regeneration and reduced TNF-α expression by 45%, confirming their dual anti-inflammatory and pro-healing function.
Dendrimers—branched, tree-like polymers—are another emerging system with high loading capacity and surface functionalization potential. Although still in the preclinical phase, Patel et al. formulated cationic poly(amidoamine) (PAMAM) dendrimers to deliver short peptides derived from probiotic secretomes. These constructs exhibited selective antimicrobial activity against P. aeruginosa while sparing commensal skin flora and significantly promoted angiogenesis in wound scratch assays [125,126].
Metal-organic frameworks (MOFs) have recently emerged as highly tunable platforms with large surface areas, ideal for pH-responsive or enzyme-triggered release of therapeutic payloads. Furthermore, the capacity of nanoclays and bioinorganic hybrids to adsorb probiotic metabolites or cytokines and alter wound pH or oxidative stress has been studied [127].
Self-assembling peptide nanostructures, such as RADA16, have also been explored as scaffolds to deliver probiotic lysates or metabolites. These biomaterials create an electroactive, nanofibrous matrix that mimics the ECM and promotes fibroblast migration. Research by [128] demonstrated enhanced collagen I expression and lower bacterial load when postbiotic-loaded RADA16 gels were applied to chronic wounds.
These novel nanoplatforms expand the landscape of wound therapeutics by combining structural support with bioactive signaling, controlled release, and improved biocompatibility. As more advanced designs are validated through in vivo and clinical models, they hold great promise for integration into next-generation bioactive dressings and personalized wound management strategies.

5. Probiotic–Nanotechnology Synergy in Chronic Wound Healing

A promising development in the management of chronic wounds is the use of probiotics augmented with nanotechnology. In addition to offering environmental protection and structural support, nanoencapsulation improves retention and delivers more efficient functionality. This combination minimizes issues with degradation in oxidative microenvironments, retention at the wound site, and survivability while providing biological benefits [129]. Various encapsulation techniques, such as using chitosan nanoparticles, liposomes, and nanogels, present different mechanistic strategies. Figure 3 illustrates the synergistic mechanisms through which probiotic–nanotechnology hybrids contribute to chronic wound healing. Chitosan-encapsulated L. plantarum achieved 93% encapsulation efficiency, significantly enhancing wound healing in diabetic rats treated with the encapsulated bacteria compared to a non-encapsulated strain, with a notable increase in collagen deposition and granulation tissue formation (p < 0.01) [130].
Utilizing nanotechnology for delivering probiotics to wound sites offers advantages such as enhanced environmental resistance and controlled release kinetics. According to [131], nanobeads containing L. rhamnosus GG retained 90% of probiotic viability after 90 days in ambient storage conditions. However, a decline of over 60% was observed when probiotics were freeze-dried. Delivery systems that react to glucose, ROS, or pH enable targeted release. Huang et al. created a glucose-responsive hydrogel with L. reuteri for H2O2 release, which reduced wound bacteria by more than 90% and improved epithelial regeneration in diabetic mouse models. This was due to the glucose-enhanced, hydrogen peroxide-producing strain, or the increased metabolic effect on bacteria by glucose [132,133].
Microneedle array platforms offer opportunities for delivering viable probiotics in a less invasive and more localized manner. Two studies [12,134] developed microneedles infused with L. rhamnosus GG, achieving over 85% healing in S. aureus wound infections within a week. The formulation maintained over 80% of viable bacteria after being stored in a structured refrigerated environment for two months, highlighting its potential for long-term use.
Beyond their antimicrobial and regenerative capabilities, probiotics also modify the immune microenvironment of chronic wounds, which are often plagued by persistent inflammation marked by a predominance of M1 macrophages and elevated levels of IL-6, IL-1β, and TNF-α [135,136]. In a study, it was demonstrated that nanogel-loaded L. fermentum significantly impacted (p < 0.01) the immune microenvironment in a diabetic wound model by increasing M2 macrophage populations (CD206+ cells) by 2.6-fold and reducing IL-6 and TNF-α levels. By blocking the NF-κB signaling pathway and reviving antioxidant enzymes like catalase and superoxide dismutase, the administration of nanoformulated probiotics decreased oxidative stress [137].
Probiotic-nano hybrids, as a comprehensive solution, offer a bioresponsive and multifunctional approach to wound care. They promote epithelialization, ensure a balanced cytokine environment, counteract microbial resistance, and facilitate the development of granulation tissue. Beyond their diverse mechanisms and safety profile, there remains a compelling rationale for further exploration of these innovative products in both translational and clinical research [138,139].

6. Comparative Analysis: Probiotic Nanoformulations vs. Conventional Therapies

Probiotic nano-formulations represent a notable advancement over conventional wound care methods, such as antibiotics and silver-based dressings. Traditional treatments often struggle with issues like insufficient biofilm penetration, limited biocompatibility, and rising antimicrobial resistance. In contrast, nanocarriers loaded with probiotics provide antimicrobial, anti-inflammatory, and regenerative benefits while preserving microbial balance and minimizing toxicity [140]. Numerous experimental studies demonstrate the superior efficacy of probiotic nano-formulations. For example, in a diabetic wound model using mice, chitosan nanogels containing L. rhamnosus achieved 89% wound closure within 10 days, compared to 62% in the neomycin group, along with a 2-log reduction in microbial load and significantly decreased TNF-α levels. Similarly, liposomal L. acidophilus enhanced epithelial regeneration and fibroblast activity while significantly reducing IL-6 levels compared to silver sulfadiazine in thermal wounds [141].
The nanocarriers employed in these systems, such as PLGA, alginate, and chitosan, are biodegradable and approved for medical use, ensuring they meet regulatory standards. Probiotics work not only by directly inhibiting pathogens but also by modulating the immune system. They encourage macrophage polarization towards an M2 phenotype, inhibit NF-κB signaling, and increase the production of growth factors like VEGF and TGF-β1—actions that antibiotics do not perform [142]. These varied mechanisms offer comprehensive support for healing in complex wound environments [85]. Consequently, given their superior efficacy, broader biological functions, and enhanced safety, probiotic nanoformulations present a compelling alternative to traditional wound care. As antibiotic resistance rises and biofilm-related infections continue, incorporating probiotic nano-therapy into clinical practice could transform wound management paradigms in the next decade, as outlined in the comparative analysis shown in (Table 2).

7. Challenges and Considerations in Probiotic Nanotechnology for Wound Healing

Although probiotic nanoformulations have the potential to enhance wound care, a number of challenges need to be addressed before their widespread application in clinical settings can occur. Making sure probiotics stay alive and metabolically active throughout the encapsulation and storage processes is the essential concern. Probiotics are encapsulated using techniques including spray drying, freeze-drying, or nanoprecipitation; nevertheless, shear stress, heat, and drying might reduce viability [152,153]. Encapsulation using chitosan-alginate has shown that over 80% of probiotics remain viable at four degrees Celsius for up to three months. However, certain strains experience a significant reduction in metabolic activity when exposed to ambient humidity [154].
A second significant challenge involves scalability and cost. While probiotic formulations at the laboratory level show substantial clinical benefits, expanding to commercial production necessitates further technical and economic considerations. Ensuring safety and consistency in nanoparticle size and bioactivity requires an infrastructure with quality control adhering to GMP (Good Manufacturing Practice) standards [155]. Economic analysis indicated that although probiotic dressings could potentially reduce long-term clinical costs, their initial expense remains 1.5 to 2 times higher than standard treatments [4]. Furthermore, obtaining regulatory approval for probiotic nanomedicine remains difficult. Unlike pharmaceuticals, probiotics are sometimes classified as food supplements or biologics, leading to regulatory inconsistencies across different countries. The integration of nanotechnology adds complexity to classification, safety evaluation, and approval processes. A 2022 report by the European Medicines Agency (EMA) highlighted the necessity for a unified regulatory framework to manage combined formulations involving both microbial and nanomaterial components. Additionally, preclinical studies should address the interaction between the host and probiotics, as well as the long-term biocompatibility of the nanomaterial [156].
Recent work [157] further emphasizes the importance of lyo-protectants and stabilizing agents in preserving probiotic viability during nano-delivery formulation. In various PEO-based nanofiber systems, species-specific efficacy of excipients was observed: sucrose best preserved Lactobacillus jensenii, while dextran and trehalose were most protective for Staphylococcus and Stenotrophomonas isolates. Notably, fully amorphous trehalose remained effective over 24 weeks, whereas semi-crystalline stabilizers (e.g., mannitol and glucose) showed reduced viability or even detrimental effects due to crystallization. These findings highlight that the choice and solid-state behavior of lyo-protectants is pivotal to maintaining long-term probiotic survival in wound care nano-formulations.
Ensuring stability and shelf life is crucial for probiotic-based nanotherapeutics, particularly in environments lacking cold chain distribution. Studies indicate that spray-dried nano-capsules using trehalose or skim milk as cryoprotectants exhibit superior thermal stability compared to in vitro experiments. However, when stored in standard packaging, they retain only 30–40% viability after 6 months at 25 °C. This reduction in viability not only diminishes probiotic counts but also affects bio-efficacy, potentially limiting therapeutic effectiveness [158,159]. As of this summer, ongoing research is investigating the use of oxygen scavengers and smart packaging to enhance shelf stability, though this might affect functionality.
It is essential to examine immunological risks as well. While probiotics are generally recognized as safe (GRAS), the potential for dysbiosis or negative immune reactions, particularly in immunocompromised individuals, should not be ignored. Instances of bacteremia and fungemia have been occasionally documented, mainly in patients undergoing live microbial therapy. Nanoformulations might further influence host responses by changing microbial adhesion characteristics or translocation behavior [160,161]. A study on mice revealed mild lymphocytic infiltration and increased IL-17 expression in wounds treated with L. rhamnosus nanofibers. These results underscore the importance of conducting individual risk-benefit analyses and evaluating product-specific toxicity [162].

8. Future Perspectives

Probiotic nanotechnology holds the potential to transform wound health care, especially in the areas of precision medicine and advanced therapeutics. The future of probiotic nanotechnology is promising, with the development of microbiome-targeted approaches to customize wound care [163]. Chronic wounds can vary depending on the bacterial composition of the host’s microbiota. By employing next-generation sequencing and microbiome profiling, future probiotic formulations could be customized to the specific local microflora, aiming to selectively diminish dysbiotic bacteria and restore microbial balance in infected wounds [164]. A 2023 review highlighted the introduction of strain-specific and site-specific probiotic nano-formulations to achieve better outcomes with fewer unintended effects. The integration of biosensing technologies into probiotic delivery systems is rapidly advancing. Smart wound dressings equipped with biosensors can detect changes in glucose, pH, reactive oxygen species, or pro-inflammatory cytokines and respond accordingly. These adaptive systems can release probiotics or their metabolites in a controlled manner, both temporally and spatially, enhancing therapeutic effectiveness while avoiding ineffective dosing [165].
The combination of artificial intelligence (AI) with probiotic nanomedicine could significantly impact chronic wound management. AI-driven platforms can analyze imaging, sensor, and microbiome data to forecast wound progression, recommend optimal treatment strategies, and evaluate probiotic effectiveness in real-time. Initial clinical models using machine-learning algorithms have shown about 92% accuracy in identifying healing trajectories. However, to integrate these findings into clinical practice, controlled trials, thorough validation, and regulatory alignment are necessary [166,167]. To evaluate the safety and effectiveness of probiotic nano-formulations for different wound types, such as diabetic foot ulcers, pressure ulcers, and burn injuries, several early-phase human trials are now being conducted. Nonetheless, widespread clinical adoption will require addressing challenges related to individual microbiome variability, the long-term safety of nanocarriers, and the complex approval process for live-biologic nanomedicines. Regulatory bodies are beginning to develop hybrid pathways to evaluate both microbial and nanomaterial components, with standard protocols still under discussion [168]. In the near future, we can expect the convergence of biotechnology, material science, and digital health to create multifunctional programmable probiotic systems capable of responding to wound dynamics in real-time. Next generation wound therapies will not only treat infections or inflammation but will also have the capability to diagnose, respond, and regenerate in real-time.

9. Conclusions

Probiotic nanotechnology signifies a groundbreaking development in chronic wound management by combining the advantages of live microbial therapy with nanocarrier-based delivery systems. This focused strategy tackles the complex issues of chronic wounds, including microbial biofilms, oxidative stress, inflammation, and hindered tissue regeneration. Nanoformulations such as liposomes, chitosan nanoparticles, microneedles, and ROS/glucose-responsive hydrogels improve probiotic stability, maintain viability, and allow for targeted, controlled release of bioactive substances. Probiotic strains like L. plantarum, L. rhamnosus, and B. subtilis have shown significant preclinical and clinical success, such as faster epithelialization, improved collagen deposition, biofilm disruption, and immune modulation through macrophage polarization and cytokine regulation. Additionally, advanced platforms featuring extracellular vesicles, postbiotics, and biosensor-integrated dressings indicate the rise of intelligent, adaptive wound therapies. When combined with AI-driven wound monitoring and microbiome profiling, these systems can provide personalized, precision-based care. Nonetheless, challenges persist in ensuring product stability, regulatory clarity, manufacturing scalability, and long-term biocompatibility. Overcoming these obstacles through standardized protocols, comprehensive clinical trials, and unified approval processes will be essential. Overall, probiotic nano-formulations offer tremendous potential to revolutionize wound care, providing a bio-responsive, cost-effective, and safer alternative to traditional antimicrobial treatments in an era of increasing antibiotic resistance.

Author Contributions

M.U., A.A.C., M.M.: conceptualization, methodology, funding acquisition, writing original draft; S.S., P.K.: visualization, data curation, writing review and editing; J.M., H.A.R.: formal analysis, writing review and editing; R.T.: investigations, resource, writing review and editing, supervision. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported and funded by the Deanship of Scientific Research at Imam Mohammad Ibn Saud Islamic University (IMSIU) (grant number IMSIU-DDRSP2501).

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
DFUsdiabetic foot ulcers
VLUsvenous leg ulcers
AMRantimicrobial resistance
MDRmulti-drug resistance
TNF-αtumor necrosis factor-alpha
IL-1βinterleukin-1 beta
MMPsmatrix metalloproteinases
ROSreactive oxygen species
AGEsadvanced glycation end-products
TLRtoll-like receptor
AMPsantimicrobial peptides
VEGFvascular endothelial growth factor
TGF-β1transforming growth factor-beta 1
EVsExtracellular Vesicles
CRPC-Reactive Protein
SODSuperoxide Dismutase
MDAMalondialdehyde
PCLPolycaprolactone
PVApolyvinyl alcohol
MNMicroneedle
PLGAPoly(lactic-co-glycolic acid)
bFGFbasic fibroblast growth factor
ECMextracellular matrix
SLNssolid lipid nanoparticles
MOFMetal-organic frameworks
PAMAMpoly(amidoamine)
EMAEuropean Medicines Agency

References

  1. Raffetto, J.D.; Ligi, D.; Maniscalco, R.; Khalil, R.A.; Mannello, F. Why Venous Leg Ulcers Have Difficulty Healing: Overview on Pathophysiology, Clinical Consequences, and Treatment. J. Clin. Med. 2021, 10, 29. [Google Scholar] [CrossRef] [PubMed]
  2. McDermott, K.; Fang, M.; Boulton, A.J.M.; Selvin, E.; Hicks, C.W. Etiology, Epidemiology, and Disparities in the Burden of Diabetic Foot Ulcers. Diabetes Care 2023, 46, 209–221. [Google Scholar] [CrossRef] [PubMed]
  3. Sen, C.K. Human Wound and Its Burden: Updated 2020 Compendium of Estimates. Adv. Wound Care 2021, 10, 281–292. [Google Scholar] [CrossRef] [PubMed]
  4. Machado, P.; Ribeiro, F.N.; Giublin, F.C.W.; Mieres, N.G.; Tonin, F.S.; Pontarolo, R.; Sari, M.H.M.; Lazo, R.E.L.; Ferreira, L.M. Next-Generation Wound Care: A Scoping Review on Probiotic, Prebiotic, Synbiotic, and Postbiotic Cutaneous Formulations. Pharmaceuticals 2025, 18, 704. [Google Scholar] [CrossRef]
  5. Cavallo, I.; Sivori, F.; Mastrofrancesco, A.; Abril, E.; Pontone, M.; Di Domenico, E.G.; Pimpinelli, F. Bacterial Biofilm in Chronic Wounds and Possible Therapeutic Approaches. Biology 2024, 13, 109. [Google Scholar] [CrossRef]
  6. Diban, F.; Di Lodovico, S.; Di Fermo, P.; D’ercole, S.; D’arcangelo, S.; Di Giulio, M.; Cellini, L. Biofilms in Chronic Wound Infections: Innovative Antimicrobial Approaches Using the In Vitro Lubbock Chronic Wound Biofilm Model. Int. J. Mol. Sci. 2023, 24, 1004. [Google Scholar] [CrossRef]
  7. Aswathanarayan, J.B.; Rao, P.; Hm, S.; Gs, S.; Rai, R.V. Biofilm-Associated Infections in Chronic Wounds and Their Management. Adv. Exp. Med. Biol. 2023, 1370, 55–75. [Google Scholar] [CrossRef]
  8. Malone, M.; Schultz, G. Challenges in the diagnosis and management of wound infection. Br. J. Dermatol. 2022, 187, 159–166. [Google Scholar] [CrossRef]
  9. Ung, L.; Chodosh, J. Foundational concepts in the biology of bacterial keratitis. Exp. Eye Res. 2021, 209, 108647. [Google Scholar] [CrossRef]
  10. Pironi, L.; Cuerda, C.; Jeppesen, P.B.; Joly, F.; Jonkers, C.; Krznarić, Ž.; Lal, S.; Lamprecht, G.; Lichota, M.; Mundi, M.S.; et al. ESPEN guideline on chronic intestinal failure in adults—Update 2023. Clin. Nutr. 2023, 42, 1940–2021. [Google Scholar] [CrossRef]
  11. Kavyani, B.; Ahmadi, S.; Nabizadeh, E.; Abdi, M. Anti-oxidative activity of probiotics; focused on cardiovascular disease, cancer, aging, and obesity. Microb. Pathog. 2024, 196, 107001. [Google Scholar] [CrossRef] [PubMed]
  12. Bădăluță, V.A.; Curuțiu, C.; Dițu, L.M.; Holban, A.M.; Lazăr, V. Probiotics in Wound Healing. Int. J. Mol. Sci. 2024, 25, 5723. [Google Scholar] [CrossRef] [PubMed]
  13. Xie, A.; Gao, M.; Du, H.; Pan, X. Next-generation probiotics delivery: Innovations and applications of single-cell encapsulation. Curr. Opin. Food Sci. 2025, 61, 101234. [Google Scholar] [CrossRef]
  14. Alhodieb, F.S.; Barkat, A.; Barkat, H.A.; Ab Hadi, H.; Khan, M.I.; Ashfaq, F.; Rahman, M.A.; Hassan, M.Z.; Alanezi, A.A. Chitosan-modified nanocarriers as carriers for anticancer drug delivery: Promises and hurdles. Int. J. Biol. Macromol. 2022, 217, 457–469. [Google Scholar] [CrossRef]
  15. Palani, N.; Vijayakumar, P.; Monisha, P.; Ayyadurai, S.; Rajadesingu, S. Electrospun nanofibers synthesized from polymers incorporated with bioactive compounds for wound healing. J. Nanobiotechnol. 2024, 22, 211. [Google Scholar] [CrossRef] [PubMed]
  16. Zhang, Q.; Liu, R.; Yan, S.; Yan, X.; Lv, Y. Deciphering the Antibacterial Mechanism of Hollow Sphere Metal-Organic Frameworks against Staphylococcus aureus and Its Application in Wound Healing. Chem. Mater. 2025, 37, 2204–2219. [Google Scholar] [CrossRef]
  17. Kaya, E.; Bianchi, M.; Maisetta, G.; Esin, S.; Batoni, G. Strong Activity and No Resistance Induction Exerted by Cell-Free Supernatants from Lacticaseibacillus rhamnosus against Mono-Species and Dual-Species Biofilms of Wound Pathogens in In Vivo-like Conditions. Int. J. Mol. Sci. 2024, 25, 2087. [Google Scholar] [CrossRef]
  18. Knackstedt, R.; Knackstedt, T.; Gatherwright, J. The role of topical probiotics on wound healing: A review of animal and human studies. Int. Wound J. 2020, 17, 1687–1694. [Google Scholar] [CrossRef]
  19. Kolimi, P.; Narala, S.; Nyavanandi, D.; Youssef, A.A.A.; Dudhipala, N. Innovative Treatment Strategies to Accelerate Wound Healing: Trajectory and Recent Advancements. Cells 2022, 11, 2439. [Google Scholar] [CrossRef]
  20. Liu, Q.; Zhang, Y.; Huang, J.; Xu, Z.; Li, X.; Yang, J.; Huang, H.; Tang, S.; Chai, Y.; Lin, J.; et al. Mesoporous silica-coated silver nanoparticles as ciprofloxacin/siRNA carriers for accelerated infected wound healing. J. Nanobiotechnol. 2022, 20, 386. [Google Scholar] [CrossRef]
  21. Vijayaram, S.; Razafindralambo, H.; Sun, Y.Z.; Piccione, G.; Multisanti, C.R.; Faggio, C. Synergistic interaction of nanoparticles and probiotic delivery: A review. J. Fish Dis. 2024, 47, e13916. [Google Scholar] [CrossRef] [PubMed]
  22. Rubio, A.P.D.; D’Antoni, C.L.; Piuri, M.; Pérez, O.E. Probiotics, Their Extracellular Vesicles and Infectious Diseases. Front. Microbiol. 2022, 13, 864720. [Google Scholar] [CrossRef]
  23. Mukharya, A.; Pokale, R.; Roy, A.A.; Soman, S.; Kulkarni, S.; Madhusoodanan, G.; Manikkath, J.; Farhana, S.A.; Pandey, A.; Mutalik, S. Targeting diabetic and chronic wounds using topical nano-formulations impregnated with modern microbiome-driven peptides and probiotics. J. Drug Deliv. Sci. Technol. 2025, 105, 106590. [Google Scholar] [CrossRef]
  24. Kosaric, N.; Kiwanuka, H.; Gurtner, G.C. Stem cell therapies for wound healing. Expert Opin. Biol. Ther. 2019, 19, 575–585. [Google Scholar] [CrossRef]
  25. Tan, M.L.L.; Chin, J.S.; Madden, L.; Becker, D.L. Challenges faced in developing an ideal chronic wound model. Expert Opin. Drug Discov. 2023, 18, 99–114. [Google Scholar] [CrossRef]
  26. Bhol, N.K.; Bhanjadeo, M.M.; Singh, A.K.; Dash, U.C.; Ojha, R.R.; Majhi, S.; Duttaroy, A.K.; Jena, A.B. The interplay between cytokines, inflammation, and antioxidants: Mechanistic insights and therapeutic potentials of various antioxidants and anti-cytokine compounds. Biomed. Pharmacother. 2024, 178, 117177. [Google Scholar] [CrossRef]
  27. Gregoriou, S.; Papanikolaou, M.; Paul, J.; Nattkemper, L.A.; Kirsner, R.S.; Yosipovitch, G. Prevalence and Mechanisms of Itch in Chronic Wounds: A Narrative Review. J. Clin. Med. 2025, 14, 2877. [Google Scholar] [CrossRef]
  28. Sanchez, M.C.; Lancel, S.; Boulanger, E.; Neviere, R. Targeting Oxidative Stress and Mitochondrial Dysfunction in the Treatment of Impaired Wound Healing: A Systematic Review. Antioxidants 2018, 7, 98. [Google Scholar] [CrossRef] [PubMed]
  29. Wang, Z.; Shi, Y.; Zheng, P.; Wang, X.; Xing, Y.; Li, D.; Tu, H.; Xu, B. Improving diabetic wound healing: The therapeutic potential of allulose supplement in diabetic skin tissue repair and inflammation modulation. Food Biosci. 2024, 62, 105439. [Google Scholar] [CrossRef]
  30. Kamml, J.; Ke, C.Y.; Acevedo, C.; Kammer, D.S. The influence of AGEs and enzymatic cross-links on the mechanical properties of collagen fibrils. J. Mech. Behav. Biomed. Mater. 2023, 143, 105870. [Google Scholar] [CrossRef]
  31. Kamal, R.; Awasthi, A.; Pundir, M.; Thakur, S. Healing the diabetic wound: Unlocking the secrets of genes and pathways. Eur. J. Pharmacol. 2024, 975, 176645. [Google Scholar] [CrossRef] [PubMed]
  32. Dsouza, F.P.; Dinesh, S.; Sharma, S. Understanding the intricacies of microbial biofilm formation and its endurance in chronic infections: A key to advancing biofilm-targeted therapeutic strategies. Arch. Microbiol. 2024, 206, 85. [Google Scholar] [CrossRef]
  33. Caldwell, M.D. Bacteria and Antibiotics in Wound Healing. Surg. Clin. 2020, 100, 757–776. [Google Scholar] [CrossRef] [PubMed]
  34. Michaelis, C.; Grohmann, E. Horizontal Gene Transfer of Antibiotic Resistance Genes in Biofilms. Antibiotics 2023, 12, 328. [Google Scholar] [CrossRef]
  35. Severn, M.M.; Horswill, A.R. Staphylococcus epidermidis and its dual lifestyle in skin health and infection. Nat. Rev. Microbiol. 2022, 21, 97–111. [Google Scholar] [CrossRef]
  36. Kalan, L.R.; Meisel, J.S.; Loesche, M.A.; Horwinski, J.; Soaita, I.; Chen, X.; Uberoi, A.; Gardner, S.E.; Grice, E.A. Strain- and Species-Level Variation in the Microbiome of Diabetic Wounds Is Associated with Clinical Outcomes and Therapeutic Efficacy. Cell Host Microbe 2019, 25, 641–655.e5. [Google Scholar] [CrossRef]
  37. Mihai, M.M.; Bălăceanu-Gurău, B.; Ion, A.; Holban, A.M.; Gurău, C.-D.; Popescu, M.N.; Beiu, C.; Popa, L.G.; Popa, M.I.; Dragomirescu, C.C.; et al. Host–Microbiome Crosstalk in Chronic Wound Healing. Int. J. Mol. Sci. 2024, 25, 4629. [Google Scholar] [CrossRef]
  38. Lebeer, S.; Oerlemans, E.F.; Claes, I.; Henkens, T.; Delanghe, L.; Wuyts, S.; Spacova, I.; Broek, M.F.v.D.; Tuyaerts, I.; Wittouck, S.; et al. Selective targeting of skin pathobionts and inflammation with topically applied lactobacilli. Cell Rep. Med. 2022, 3, 100521. [Google Scholar] [CrossRef]
  39. Belkaid, Y.; Harrison, O.J. Homeostatic Immunity and the Microbiota. Immunity 2017, 46, 562–576. [Google Scholar] [CrossRef]
  40. Durand, B.A.R.N.; Pouget, C.; Magnan, C.; Molle, V.; Lavigne, J.P.; Dunyach-Remy, C. Bacterial Interactions in the Context of Chronic Wound Biofilm: A Review. Microorganisms 2022, 10, 1500. [Google Scholar] [CrossRef]
  41. Roson-Calero, N.; Ballesté-Delpierre, C.; Fernández, J.; Vila, J. Insights on Current Strategies to Decolonize the Gut from Multidrug-Resistant Bacteria: Pros and Cons. Antibiotics 2023, 12, 1074. [Google Scholar] [CrossRef] [PubMed]
  42. Mishra, S.; Singh, A.; Naqvi, W.; Garg, P.; Srivastava, N.; Srivastava, P. Implications of systems biology in understanding the pathophysiology of neurological diseases. In Systems Biology and In-Depth Applications for Unlocking Diseases: Principles, Tools, and Application to Disease; Academic Press: New York, NY, USA, 2025; pp. 245–257. [Google Scholar] [CrossRef]
  43. Petrariu, O.A.; Barbu, I.C.; Niculescu, A.G.; Constantin, M.; Grigore, G.A.; Cristian, R.E.; Mihaescu, G.; Vrancianu, C.O. Role of probiotics in managing various human diseases, from oral pathology to cancer and gastrointestinal diseases. Front. Microbiol. 2023, 14, 1296447. [Google Scholar] [CrossRef] [PubMed]
  44. Das, T.K.; Pradhan, S.; Chakrabarti, S.; Mondal, K.C.; Ghosh, K. Current status of probiotic and related health benefits. Appl. Food Res. 2022, 2, 100185. [Google Scholar] [CrossRef]
  45. Moraffah, F.; Kiani, M.; Abdollahi, M.; Yoosefi, S.; Vatanara, A.; Samadi, N. In Vitro-In Vivo Correlation for the Antibacterial Effect of Lactiplantibacillus plantarum as a Topical Healer for Infected Burn Wound. Probiotics Antimicrob. Proteins 2022, 14, 675–689. [Google Scholar] [CrossRef]
  46. Burian, E.A.; Sabah, L.; Karlsmark, T.; Kirketerp-Møller, K.; Moffatt, C.J.; Thyssen, J.P.; Ågren, M.S. Cytokines and Venous Leg Ulcer Healing—A Systematic Review. Int. J. Mol. Sci. 2022, 23, 6526. [Google Scholar] [CrossRef]
  47. Wang, X.; Li, X.; Liu, J.; Tao, Y.; Wang, T.; Li, L. Lactobacillus plantarum Promotes Wound Healing by Inhibiting the NLRP3 Inflammasome and Pyroptosis Activation in Diabetic Foot Wounds. J. Inflamm. Res. 2024, 17, 1707–1720. [Google Scholar] [CrossRef] [PubMed]
  48. Li, M.; Xiao, H.; Su, Y.; Cheng, D.; Jia, Y.; Li, Y.; Yin, Q.; Gao, J.; Tang, Y.; Bai, Q. Synergistic Inhibitory Effect of Honey and Lactobacillus plantarum on Pathogenic Bacteria and Their Promotion of Healing in Infected Wounds. Pathogens 2023, 12, 501. [Google Scholar] [CrossRef]
  49. Tao, J.; Sun, Y.; Wang, G.; Sun, J.; Dong, S.; Ding, J. Advanced biomaterials for targeting mature biofilms in periodontitis therapy. Bioact. Mater. 2025, 48, 474–492. [Google Scholar] [CrossRef]
  50. Xin, H.; Cai, Z.; Hao, J.; An, J.; Li, Y.; Wen, M.; Jia, Z. Macro/Microgel-Encapsulated, Biofilm-Armored Living Probiotic Platform for Regenerating Bacteria-Infected Diabetic Wounds. Adv. Healthc. Mater. 2025, 14, 2403476. [Google Scholar] [CrossRef]
  51. Yang, X.; Jiang, X.; Ning, X.; Feng, Y.; Guo, W.; Wei, C.; Nešić, M.D.; Whittaker, A.K.; Qu, W.; Yang, B.; et al. Skin-inspired conductive hydrogels with antibacterial and pro-angiogenic abilities customized for accelerating diabetic wound healing. Sci. China Mater. 2025, 68, 1271–1284. [Google Scholar] [CrossRef]
  52. Cho, M.-Y.; Eom, J.-H.; Choi, E.-M.; Yang, S.-J.; Lee, D.; Kim, Y.Y.; Kim, H.-S.; Hwang, I.; Staley, C. Recent advances in therapeutic probiotics: Insights from human trials. Clin. Microbiol. Rev. 2025, 38, e0024024. [Google Scholar] [CrossRef] [PubMed]
  53. Majid, S.A.; Khorasgani, M.R.; Emami, S.H.; Talebi, A.; Gholami, A.M. Study of Diabetic Cutaneous Wound Healing in Rats Treated with Lactobacillus casei and Its Exopolysaccharide. Int. J. Adv. Biotechnol. Res. (IJBR) 2016, 7, 2083–2092. Available online: https://www.researchgate.net/publication/308538779_Study_of_Diabetic_cutaneous_wound_healing_in_rats_treated_with_Lactobacillus_Casei_and_its_Exopolysaccharide (accessed on 10 May 2025).
  54. Wang, J.; Li, X.; Zhao, X.; Yuan, S.; Dou, H.; Cheng, T.; Huang, T.; Lv, Z.; Tu, Y.; Shi, Y.; et al. Lactobacillus rhamnosus GG-derived extracellular vesicles promote wound healing via miR-21-5p-mediated re-epithelization and angiogenesis. J. Nanobiotechnol. 2024, 22, 644. [Google Scholar] [CrossRef]
  55. Fernandes, A.; Rodrigues, P.M.; Pintado, M.; Tavaria, F.K. A systematic review of natural products for skin applications: Targeting inflammation, wound healing, and photo-aging. Phytomedicine 2023, 115, 154824. [Google Scholar] [CrossRef] [PubMed]
  56. Srivastava, P.; Sondak, T.; Sivashanmugam, K.; Kim, K.-S. A Review of Immunomodulatory Reprogramming by Probiotics in Combating Chronic and Acute Diabetic Foot Ulcers (DFUs). Pharmaceutics 2022, 14, 2436. [Google Scholar] [CrossRef]
  57. Meroni, G.; Panelli, S.; Zuccotti, G.; Bandi, C.; Drago, L.; Pistone, D. Probiotics as Therapeutic Tools against Pathogenic Biofilms: Have We Found the Perfect Weapon? Microbiol. Res. 2021, 12, 916–937. [Google Scholar] [CrossRef]
  58. Pompilio, A.; Kaya, E.; Lupetti, V.; Catelli, E.; Bianchi, M.; Maisetta, G.; Esin, S.; Di Bonaventura, G.; Batoni, G. Cell-free supernatants from Lactobacillus strains exert antibacterial, antibiofilm, and antivirulence activity against Pseudomonas aeruginosa from cystic fibrosis patients. Microbes Infect. 2024, 26, 105301. [Google Scholar] [CrossRef] [PubMed]
  59. Araújo, D.; Silva, A.R.; Fernandes, R.; Serra, P.; Barros, M.M.; Campos, A.M.; Oliveira, R.; Silva, S.; Almeida, C.; Castro, J. Emerging Approaches for Mitigating Biofilm-Formation-Associated Infections in Farm, Wild, and Companion Animals. Pathogens 2024, 13, 320. [Google Scholar] [CrossRef]
  60. Khmaladze, I.; Butler, É.; Fabre, S.; Gillbro, J.M. Lactobacillus reuteri DSM 17938—A comparative study on the effect of probiotics and lysates on human skin. Exp. Dermatol. 2019, 28, 822–828. [Google Scholar] [CrossRef]
  61. Jo, C.S.; Myung, C.H.; Yoon, Y.C.; Ahn, B.H.; Min, J.W.; Seo, W.S.; Lee, D.H.; Kang, H.C.; Heo, Y.H.; Choi, H.; et al. The Effect of Lactobacillus plantarum Extracellular Vesicles from Korean Women in Their 20s on Skin Aging. Curr. Issues Mol. Biol. 2022, 44, 526–540. [Google Scholar] [CrossRef]
  62. Gurunathan, S.; Thangaraj, P.; Kim, J.H. Postbiotics: Functional Food Materials and Therapeutic Agents for Cancer, Diabetes, and Inflammatory Diseases. Foods 2024, 13, 89. [Google Scholar] [CrossRef] [PubMed]
  63. Sabahi, S.; Rad, A.H.; Aghebati-Maleki, L.; Sangtarash, N.; Ozma, M.A.; Karimi, A.; Hosseini, H.; Abbasi, A. Postbiotics as the new frontier in food and pharmaceutical research. Crit. Rev. Food Sci. Nutr. 2023, 63, 8375–8402. [Google Scholar] [CrossRef] [PubMed]
  64. Monika, K.; Malik, T.; Gehlot, R.; Rekha, K.; Kumari, A.; Sindhu, R.; Rohilla, P. Antimicrobial Property of Probiotics. Environ. Conserv. J. 2021, 22, 33–48. [Google Scholar] [CrossRef]
  65. Rocchetti, M.T.; Russo, P.; Capozzi, V.; Drider, D.; Spano, G.; Fiocco, D. Bioprospecting Antimicrobials from Lactiplantibacillus plantarum: Key Factors Underlying Its Probiotic Action. Int. J. Mol. Sci. 2021, 22, 12076. [Google Scholar] [CrossRef]
  66. Chin, J.D.; Zhao, L.; Mayberry, T.G.; Cowan, B.C.; Wakefield, M.R.; Fang, Y. Photodynamic Therapy, Probiotics, Acetic Acid, and Essential Oil in the Treatment of Chronic Wounds Infected with Pseudomonas aeruginosa. Pharmaceutics 2023, 15, 1721. [Google Scholar] [CrossRef]
  67. Yeşilyurt, N.; Yılmaz, B.; Ağagündüz, D.; Capasso, R. Involvement of Probiotics and Postbiotics in the Immune System Modulation. Biologics 2021, 1, 89–110. [Google Scholar] [CrossRef]
  68. Leser, T.; Baker, A. Molecular Mechanisms of Lacticaseibacillus rhamnosus, LGG® Probiotic Function. Microorganisms 2024, 12, 794. [Google Scholar] [CrossRef]
  69. Plaza-Diaz, J.; Ruiz-Ojeda, F.J.; Gil-Campos, M.; Gil, A. Mechanisms of Action of Probiotics. Adv. Nutr. 2019, 10, S49–S66. [Google Scholar] [CrossRef]
  70. Cristofori, F.; Dargenio, V.N.; Dargenio, C.; Miniello, V.L.; Barone, M.; Francavilla, R. Anti-Inflammatory and Immunomodulatory Effects of Probiotics in Gut Inflammation: A Door to the Body. Front. Immunol. 2021, 12, 578386. [Google Scholar] [CrossRef]
  71. Zhao, M.; Chu, J.; Feng, S.; Guo, C.; Xue, B.; He, K.; Li, L. Immunological mechanisms of inflammatory diseases caused by gut microbiota dysbiosis: A review. Biomed. Pharmacother. 2023, 164, 114985. [Google Scholar] [CrossRef]
  72. Stuermer, E.K.; Bang, C.; Giessler, A.; Smeets, R.; Janke, T.M.; Seki, F.D.; Debus, E.S.; Franke, A.; Augustin, M. Effect of oral Multispecies Probiotic on Wound Healing, Periodontitis and Quality of Life on Patients with Diabetes. J. Wound Care 2024, 33, 394–407. [Google Scholar] [CrossRef] [PubMed]
  73. Moghaddam, S.S.; Momeni, M.; Atabaki, S.M.; Shabestari, T.M.; Boustanshenas, M.; Afshar, M.; Roham, M. Topical Treatment of Second-Degree Burn Wounds with Lactobacillus plantarum Supernatant: Phase I Trial. Iran. J. Pathol. 2022, 17, 460–468. [Google Scholar] [CrossRef]
  74. Tarapatzi, G.; Filidou, E.; Kandilogiannakis, L.; Spathakis, M.; Gaitanidou, M.; Arvanitidis, K.; Drygiannakis, I.; Valatas, V.; Kotzampassi, K.; Manolopoulos, V.G.; et al. The Probiotic Strains Bifidοbacterium lactis, Lactobacillus acidophilus, Lactiplantibacillus plantarum and Saccharomyces boulardii Regulate Wound Healing and Chemokine Responses in Human Intestinal Subepithelial Myofibroblasts. Pharmaceuticals 2022, 15, 1293. [Google Scholar] [CrossRef]
  75. Aybar, J.N.A.; Mayor, S.O.; Olea, L.; Garcia, J.J.; Nisoria, S.; Kolling, Y.; Melian, C.; Rachid, M.; Dimani, R.T.; Werenitzky, C.; et al. Topical Administration of Lactiplantibacillus plantarum Accelerates the Healing of Chronic Diabetic Foot Ulcers through Modifications of Infection, Angiogenesis, Macrophage Phenotype and Neutrophil Response. Microorganisms 2022, 10, 634. [Google Scholar] [CrossRef] [PubMed]
  76. Polak, K.; Jobbágy, A.; Muszyński, T.; Wojciechowska, K.; Frątczak, A.; Bánvölgyi, A.; Bergler-Czop, B.; Kiss, N. Microbiome Modulation as a Therapeutic Approach in Chronic Skin Diseases. Biomedicines 2021, 9, 1436. [Google Scholar] [CrossRef]
  77. Aprea, G.; Del Matto, I.; Tucci, P.; Marino, L.; Scattolini, S.; Rossi, F. In Vivo Functional Properties of Dairy Bacteria. Microorganisms 2023, 11, 1787. [Google Scholar] [CrossRef] [PubMed]
  78. Naseri, K.; Saadati, S.; Ashtary-Larky, D.; Asbaghi, O.; Ghaemi, F.; Pashayee-Khamene, F.; Yari, Z.; de Courten, B. Probiotics and synbiotics supplementation improve glycemic control parameters in subjects with prediabetes and type 2 diabetes mellitus: A GRADE-assessed systematic review, meta-analysis, and meta-regression of randomized clinical trials. Pharmacol. Res. 2022, 184, 106399. [Google Scholar] [CrossRef]
  79. Kurul, F.; Turkmen, H.; Cetin, A.E.; Topkaya, S.N. Nanomedicine: How nanomaterials are transforming drug delivery, bio-imaging, and diagnosis. Next Nanotechnol. 2025, 7, 100129. [Google Scholar] [CrossRef]
  80. Far, B.F.; Naimi-Jamal, M.R.; Sedaghat, M.; Hoseini, A.; Mohammadi, N.; Bodaghi, M. Combinational System of Lipid-Based Nanocarriers and Biodegradable Polymers for Wound Healing: An Updated Review. J. Funct. Biomater. 2023, 14, 115. [Google Scholar] [CrossRef]
  81. Bazzaz, S.; Abbasi, A.; Ghotbabad, A.G.; Pourjafar, H.; Hosseini, H. Novel Encapsulation Approaches in the Functional Food Industry: With a Focus on Probiotic Cells and Bioactive Compounds. Probiotics Antimicrob. Proteins 2024, 17, 1132–1170. [Google Scholar] [CrossRef]
  82. Sneha, K.; Kumar, A. Nanoemulsions: Techniques for the preparation and the recent advances in their food applications. Innov. Food Sci. Emerg. Technol. 2022, 76, 102914. [Google Scholar] [CrossRef]
  83. Kaur, G.; Panigrahi, C.; Agarwal, S.; Khuntia, A.; Sahoo, M. Recent trends and advancements in nanoemulsions: Production methods, functional properties, applications in food sector, safety and toxicological effects. Food Phys. 2024, 1, 100024. [Google Scholar] [CrossRef]
  84. Choi, S.J.; McClements, D.J. Nanoemulsions as delivery systems for lipophilic nutraceuticals: Strategies for improving their formulation, stability, functionality and bioavailability. Food Sci. Biotechnol. 2020, 29, 149–168. [Google Scholar] [CrossRef] [PubMed]
  85. Agriopoulou, S.; Tarapoulouzi, M.; Varzakas, T.; Jafari, S.M. Application of Encapsulation Strategies for Probiotics: From Individual Loading to Co-Encapsulation. Microorganisms 2023, 11, 2896. [Google Scholar] [CrossRef] [PubMed]
  86. Chopde, S.; Datir, R.; Deshmukh, G.; Dhotre, A.; Patil, M. Nanoparticle formation by nanospray drying & its application in nanoencapsulation of food bioactive ingredients. J. Agric. Food Res. 2020, 2, 100085. [Google Scholar] [CrossRef]
  87. Pandey, R.P.; Gunjan; Himanshu; Mukherjee, R.; Chang, C.M. Nanocarrier-mediated probiotic delivery: A systematic meta-analysis assessing the biological effects. Sci. Rep. 2024, 14, 631. [Google Scholar] [CrossRef]
  88. Keshvardoostchokami, M.; Majidi, S.S.; Huo, P.; Ramachandran, R.; Chen, M.; Liu, B. Electrospun Nanofibers of Natural and Synthetic Polymers as Artificial Extracellular Matrix for Tissue Engineering. Nanomaterials 2021, 11, 21. [Google Scholar] [CrossRef]
  89. Grzywaczyk, A.; Zdarta, A.; Jankowska, K.; Biadasz, A.; Zdarta, J.; Jesionowski, T.; Kaczorek, E.; Smułek, W. New Biocomposite Electrospun Fiber/Alginate Hydrogel for Probiotic Bacteria Immobilization. Materials 2021, 14, 3861. [Google Scholar] [CrossRef]
  90. Wei, L.; Zhou, D.; Kang, X. Electrospinning as a novel strategy for the encapsulation of living probiotics in polyvinyl alcohol/silk fibroin. Innov. Food Sci. Emerg. Technol. 2021, 71, 102726. [Google Scholar] [CrossRef]
  91. Thirumalai, A.; Girigoswami, K.; Harini, K.; Pallavi, P.; Gowtham, P.; Girigoswami, A. A review of the current state of probiotic nanoencapsulation and its future prospects in biomedical applications. Biocatal. Agric. Biotechnol. 2024, 57, 103101. [Google Scholar] [CrossRef]
  92. Ayu, I.L.; Ha, H.K.; Yang, D.H.; Lee, W.J.; Lee, M.R. Encapsulation of Lactobacillus rhamnosus GG Using Milk Protein-Based Delivery Systems: Effects of Reaction Temperature and Holding Time on Their Physicochemical and Functional Properties. Food Sci. Anim. Resour. 2021, 41, 894. [Google Scholar] [CrossRef] [PubMed]
  93. Thorakkattu, P.; Khanashyam, A.C.; Shah, K.; Babu, K.S.; Mundanat, A.S.; Deliephan, A.; Deokar, G.S.; Santivarangkna, C.; Nirmal, N.P. Postbiotics: Current Trends in Food and Pharmaceutical Industry. Foods 2022, 11, 3094. [Google Scholar] [CrossRef]
  94. Liang, B.; Xing, D. The Current and Future Perspectives of Postbiotics. Probiotics Antimicrob. Proteins 2023, 15, 1626–1643. [Google Scholar] [CrossRef] [PubMed]
  95. Serpico, L.; Iacono, S.D.; Cammarano, A.; De Stefano, L. Recent Advances in Stimuli-Responsive Hydrogel-Based Wound Dressing. Gels 2023, 9, 451. [Google Scholar] [CrossRef]
  96. Nasaj, M.; Chehelgerdi, M.; Asghari, B.; Ahmadieh-Yazdi, A.; Asgari, M.; Kabiri-Samani, S.; Sharifi, E.; Arabestani, M. Factors influencing the antimicrobial mechanism of chitosan action and its derivatives: A review. Int. J. Biol. Macromol. 2024, 277, 134321. [Google Scholar] [CrossRef] [PubMed]
  97. Mawazi, S.M.; Kumar, M.; Ahmad, N.; Ge, Y.; Mahmood, S. Recent Applications of Chitosan and Its Derivatives in Antibacterial, Anticancer, Wound Healing, and Tissue Engineering Fields. Polymers 2024, 16, 1351. [Google Scholar] [CrossRef]
  98. Hosseini, S.N.; Naghib, S.M.; Kashani, G.K.; Mozafari, M.R. Chitosan-grafted Graphene Materials for Drug Delivery in Wound Healing. Curr. Pharm. Des. 2024, 31, 691–715. [Google Scholar] [CrossRef]
  99. Eleraky, N.E.; Allam, A.; Hassan, S.B.; Omar, M.M. Nanomedicine Fight against Antibacterial Resistance: An Overview of the Recent Pharmaceutical Innovations. Pharmaceutics 2020, 12, 142. [Google Scholar] [CrossRef]
  100. Loutfy, S.A.; Abdelhady, H.G.; Elberry, M.H.; Hamed, A.R.; Ahmed, H.; Hasanin, M.T.M.; Faraag, A.H.I.; Mohamed, E.-C.B.; Dardeer, A.E.; Dawood, R.; et al. In Vitro Evaluation of Cytotoxic and Anti-HCV-4 Properties of Sofosbuvir Encapsulated Chitosan Nanoparticles. arXiv 2020, arXiv:2009.06041. [Google Scholar] [CrossRef]
  101. Elkomy, M.H.; Ali, A.A.; Eid, H.M. Chitosan on the surface of nanoparticles for enhanced drug delivery: A comprehensive review. J. Control. Release 2022, 351, 923–940. [Google Scholar] [CrossRef]
  102. Negi, A.; Kesari, K.K. Chitosan Nanoparticle Encapsulation of Antibacterial Essential Oils. Micromachines 2022, 13, 1265. [Google Scholar] [CrossRef] [PubMed]
  103. Makadia, H.K.; Siegel, S.J. Poly Lactic-co-glycolic acid (PLGA) as Biodegradable Controlled Drug Delivery Carrier. Polymers 2011, 3, 1377–1397. [Google Scholar] [CrossRef] [PubMed]
  104. Swain, S.S.; Karri, V.V.S.R.; Kuppusamy, G.; Praveen, T.K.; Dutta, S. Poly(lactic-co-glycolic acid) nanoparticle-mediated approaches for wound healing. In Recent Advances in Nanomedicines Mediated Wound Healing; Academic Press: New York, NY, USA, 2025; pp. 227–239. [Google Scholar] [CrossRef]
  105. Zhou, Y.L.; Yang, Q.Q.; Zhang, L.; Tang, J.B. Nanoparticle-coated sutures providing sustained growth factor delivery to improve the healing strength of injured tendons. Acta Biomater. 2021, 124, 301–314. [Google Scholar] [CrossRef]
  106. Shi, S.; Ou, X.; Long, J.; Lu, X.; Xu, S.; Zhang, L. Nanoparticle-Based Therapeutics for Enhanced Burn Wound Healing: A Comprehensive Review. Int. J. Nanomed. 2024, 19, 11213. [Google Scholar] [CrossRef]
  107. Dandekar, P.P.; Jain, R.; Patil, S.; Dhumal, R.; Tiwari, D.; Sharma, S.; Vanage, G.; Patravale, V. Curcumin-loaded hydrogel nanoparticles: Application in anti-malarial therapy and toxicological evaluation. J. Pharm. Sci. 2010, 99, 4992–5010. [Google Scholar] [CrossRef]
  108. Rananaware, P.; Bauri, S.; Keri, R.; Mishra, M.; Brahmkhatri, V. Polymeric curcumin nanospheres for lysozyme aggregation inhibition, antibacterial, and wound healing applications. Environ. Sci. Pollut. Res. 2024, 31, 46625–46640. [Google Scholar] [CrossRef]
  109. Hiwrale, A.; Bharati, S.; Pingale, P.; Rajput, A. Nanofibers: A current era in drug delivery system. Heliyon 2023, 9, e18917. [Google Scholar] [CrossRef]
  110. Sethuram, L.; Thomas, J. Therapeutic applications of electrospun nanofibers impregnated with various biological macromolecules for effective wound healing strategy—A review. Biomed. Pharmacother. 2023, 157, 113996. [Google Scholar] [CrossRef] [PubMed]
  111. Liu, J.; Wu, S.; Ma, J.; Liu, C.; Dai, T.; Wu, X.; Zhao, H.; Zhou, D. Polycaprolactone/Gelatin/Hydroxyapatite Electrospun Nanomembrane Materials Incorporated with Different Proportions of Attapulgite Synergistically Promote Bone Formation. Int. J. Nanomed. 2022, 17, 4087. [Google Scholar] [CrossRef]
  112. Xu, C.; Ma, J.; Wang, W.; Liu, Z.; Gu, L.; Qian, S.; Hou, J.; Jiang, Z. Preparation of pectin-based nanofibers encapsulating Lactobacillus rhamnosus 1.0320 by electrospinning. Food Hydrocoll. 2022, 124, 107216. [Google Scholar] [CrossRef]
  113. Stojanov, S.; Plavec, T.V.; Zupančič, Š.; Berlec, A. Modified vaginal lactobacilli expressing fluorescent and luminescent proteins for more effective monitoring of their release from nanofibers, safety and cell adhesion. Microb. Cell Fact. 2024, 23, 333. [Google Scholar] [CrossRef]
  114. Gao, Y.; Wang, X.; Xue, C.; Wei, Z. Latest developments in food-grade delivery systems for probiotics: A systematic review. Crit. Rev. Food Sci. Nutr. 2023, 63, 4371–4388. [Google Scholar] [CrossRef] [PubMed]
  115. Parham, S.; Kharazi, A.Z.; Bakhsheshi-Rad, H.R.; Kharaziha, M.; Ismail, A.F.; Sharif, S.; Razzaghi, M.; RamaKrishna, S.; Berto, F. Antimicrobial Synthetic and Natural Polymeric Nanofibers as Wound Dressing: A Review. Adv. Eng. Mater. 2022, 24, 2101460. [Google Scholar] [CrossRef]
  116. Jiang, Z.; Zheng, Z.; Yu, S.; Gao, Y.; Ma, J.; Huang, L.; Yang, L. Nanofiber Scaffolds as Drug Delivery Systems Promoting Wound Healing. Pharmaceutics 2023, 15, 1829. [Google Scholar] [CrossRef] [PubMed]
  117. Rovelli, R.; Cecchini, B.; Zavagna, L.; Azimi, B.; Ricci, C.; Esin, S.; Milazzo, M.; Batoni, G.; Danti, S. Emerging Multiscale Biofabrication Approaches for Bacteriotherapy. Molecules 2024, 29, 533. [Google Scholar] [CrossRef]
  118. Gheorghita, R.; Anchidin-Norocel, L.; Filip, R.; Dimian, M.; Covasa, M. Applications of Biopolymers for Drugs and Probiotics Delivery. Polymers 2021, 13, 2729. [Google Scholar] [CrossRef]
  119. Sen, O.; Poddar, P.; Sarkar, P.; Das, S.; Manna, S. Current advancements in microneedle technology for therapeutic and biomedical applications. Sens. Int. 2025, 6, 100325. [Google Scholar] [CrossRef]
  120. Jin, Y.; Lu, Y.; Jiang, X.; Wang, M.; Yuan, Y.; Zeng, Y.; Guo, L.; Li, W. Accelerated infected wound healing by probiotic-based living microneedles with long-acting antibacterial effect. Bioact. Mater. 2024, 38, 292–304. [Google Scholar] [CrossRef]
  121. Mohammed, S.; Çon, A.H. Postbiotic nanoparticles (postbiotics-NPs): A novel strategy for providing probiotics’ health advantages through food consumption. Food Sci. Biotechnol. 2024, 33, 2729–2736. [Google Scholar] [CrossRef]
  122. Wang, P.; Wang, S.; Wang, D.; Li, Y.; Yip, R.C.S.; Chen, H. Postbiotics-peptidoglycan, lipoteichoic acid, exopolysaccharides, surface layer protein and pili proteins—Structure, activity in wounds and their delivery systems. Int. J. Biol. Macromol. 2024, 274, 133195. [Google Scholar] [CrossRef]
  123. Golkar, N.; Ashoori, Y.; Heidari, R.; Omidifar, N.; Abootalebi, S.N.; Mohkam, M.; Gholami, A. A Novel Effective Formulation of Bioactive Compounds for Wound Healing: Preparation, In Vivo Characterization, and Comparison of Various Postbiotics Cold Creams in a Rat Model. Evid.-Based Complement. Altern. Med. 2021, 2021, 8577116. [Google Scholar] [CrossRef]
  124. Hosseini, S.F.; Ansari, B.; Gharsallaoui, A. Polyelectrolytes-stabilized liposomes for efficient encapsulation of Lactobacillus rhamnosus and improvement of its survivability under adverse conditions. Food Chem. 2022, 372, 131358. [Google Scholar] [CrossRef] [PubMed]
  125. Kumar, A.; Shahvej, S.; Yadav, P.; Modi, U.; Yadav, A.K.; Solanki, R.; Bhatia, D. Clinical Applications of Targeted Nanomaterials. Pharmaceutics 2025, 17, 379. [Google Scholar] [CrossRef] [PubMed]
  126. Joshi, A.Y.; Joshi, U.A.; Patel, V.K.; Rathore, M. Nanofillers in the Biomedical Industry. In Handbook of Nanofillers; Springer Nature: Singapore, 2024; pp. 1–33. [Google Scholar] [CrossRef]
  127. Chen, Y.; Cai, J.; Liu, D.; Liu, S.; Lei, D.; Zheng, L.; Wei, Q.; Gao, M. Zinc-based metal organic framework with antibacterial and anti-inflammatory properties for promoting wound healing. Regen. Biomater. 2022, 9, rbac019. [Google Scholar] [CrossRef]
  128. Deptuła, M.; Sawicka, J.; Sass, P.; Sosnowski, P.; Karpowicz, P.; Zawrzykraj, M.; Wardowska, A.; Tymińska, A.; Dzierżyńska, M.; Pietralik-Molińska, Z.; et al. Development and evaluation of RADA-PDGF2 self-assembling peptide hydrogel for enhanced skin wound healing. Front. Pharmacol. 2023, 14, 1293647. [Google Scholar] [CrossRef] [PubMed]
  129. Fahimirad, S.; Fattahi, F.; Hatami, M.; Shabani, S.; Ghorbanpour, M. Nanotechnology-based biotherapeutics for physiological wound healing phases. Ind. Crops. Prod. 2025, 226, 120608. [Google Scholar] [CrossRef]
  130. Anushree, U.; Punj, P.; Vasumathi; Bharati, S. Phosphorylated chitosan accelerates dermal wound healing in diabetic wistar rats. Glycoconj. J. 2023, 40, 19–31. [Google Scholar] [CrossRef]
  131. Qi, X.; Simsek, S.; Ohm, J.B.; Chen, B.; Rao, J. Viability of Lactobacillus rhamnosus GG microencapsulated in alginate/chitosan hydrogel particles during storage and simulated gastrointestinal digestion: Role of chitosan molecular weight. Soft Matter 2020, 16, 1877–1887. [Google Scholar] [CrossRef]
  132. Lindsay, R.T.; Rhodes, C.J. Reactive Oxygen Species (ROS) in Metabolic Disease-Don’t Shoot the Metabolic Messenger. Int. J. Mol. Sci. 2025, 26, 2622. [Google Scholar] [CrossRef]
  133. Li, X.; Meng, Z.; Guan, L.; Liu, A.; Li, L.; Nešić, M.D.; Yang, B.; Qu, W.; Lin, Q. Glucose-Responsive hydrogel optimizing Fenton reaction to eradicate multidrug-resistant bacteria for infected diabetic wound healing. Chem. Eng. J. 2024, 487, 150545. [Google Scholar] [CrossRef]
  134. Martínez-Pizarro, S. Topical probiotics in the treatment of infected wounds in critical care. Enfermería Intensiv. Engl. Ed. 2021, 32, 112–113. [Google Scholar] [CrossRef] [PubMed]
  135. Merz, J.; Nettesheim, A.; von Garlen, S.; Albrecht, P.; Saller, B.S.; Engelmann, J.; Hertle, L.; Schäfer, I.; Dimanski, D.; König, S.; et al. Pro- and anti-inflammatory macrophages express a sub-type specific purinergic receptor profile. Purinergic Signal. 2021, 17, 481–492. [Google Scholar] [CrossRef] [PubMed]
  136. Sharifiaghdam, M.; Shaabani, E.; Faridi-Majidi, R.; De Smedt, S.C.; Braeckmans, K.; Fraire, J.C. Macrophages as a therapeutic target to promote diabetic wound healing. Mol. Ther. 2022, 30, 2891–2908. [Google Scholar] [CrossRef]
  137. Krzyszczyk, P.; Schloss, R.; Palmer, A.; Berthiaume, F. The Role of Macrophages in Acute and Chronic Wound Healing and Interventions to Promote Pro-wound Healing Phenotypes. Front. Physiol. 2018, 9, 419. [Google Scholar] [CrossRef] [PubMed]
  138. Chhabra, P.; Tyagi, P.; Bhatnagar, A.; Mittal, G.; Kumar, A. Optimization, Characterization, and Efficacy Evaluation of 2% Chitosan Scaffold for Tissue Engineering and Wound Healing. J. Pharm. Bioallied Sci. 2016, 8, 300–308. [Google Scholar] [CrossRef]
  139. Virijević, K.; Živanović, M.; Pavić, J.; Dragačević, L.; Ljujić, B.; Miletić Kovačević, M.; Papić, M.; Živanović, S.; Milenković, S.; Radojević, I.; et al. Electrospun Gelatin Scaffolds with Incorporated Antibiotics for Skin Wound Healing. Pharmaceuticals 2024, 17, 851. [Google Scholar] [CrossRef]
  140. Paladini, F.; Pollini, M. Antimicrobial Silver Nanoparticles for Wound Healing Application: Progress and Future Trends. Materials 2019, 12, 2540. [Google Scholar] [CrossRef]
  141. Karimi, F.; Montazeri-Najafabady, N.; Mohammadi, F.; Azadi, A.; Koohpeyma, F.; Gholami, A. A potential therapeutic strategy of an innovative probiotic formulation toward topical treatment of diabetic ulcer: An in vivo study. Nutr. Diabetes 2024, 14, 66. [Google Scholar] [CrossRef]
  142. Lopes, S.A.; Roque-Borda, C.A.; Duarte, J.L.; Di Filippo, L.D.; Cardoso, V.M.B.; Pavan, F.R.; Chorilli, M.; Meneguin, A.B. Delivery Strategies of Probiotics from Nano- and Microparticles: Trends in the Treatment of Inflammatory Bowel Disease—An Overview. Pharmaceutics 2023, 15, 2600. [Google Scholar] [CrossRef]
  143. Kumar, N.; Tyagi, N.; Mehan, S.; Singh, A.P.; Verma, B.; Kumar, S. Preparation of probiotic-loaded solid lipid nanoparticles and in vitro survival in gastrointestinal conditions. BIO Web. Conf. 2024, 86, 01051. [Google Scholar] [CrossRef]
  144. Makabenta, J.M.V.; Nabawy, A.; Li, C.H.; Schmidt-Malan, S.; Patel, R.; Rotello, V.M. Nanomaterial-based therapeutics for antibiotic-resistant bacterial infections. Nat. Rev. Microbiol. 2020, 19, 23–36. [Google Scholar] [CrossRef]
  145. Lee, H.J.; Kim, M. Skin Barrier Function and the Microbiome. Int. J. Mol. Sci. 2022, 23, 13071. [Google Scholar] [CrossRef] [PubMed]
  146. Huang, Y.Z.; Gou, M.; Da, L.C.; Zhang, W.Q.; Xie, H.Q. Mesenchymal Stem Cells for Chronic Wound Healing: Current Status of Preclinical and Clinical Studies. Tissue Eng. Part B Rev. 2020, 26, 555–570. [Google Scholar] [CrossRef] [PubMed]
  147. Albaayit, S.F.A.; Abdullah, R.; Noor, M.H.M. Zerumbone-Loaded Nanostructured Lipid Carrier Gel Enhances Wound Healing in Diabetic Rats. BioMed Res. Int. 2022, 2022, 1129297. [Google Scholar] [CrossRef] [PubMed]
  148. Mancuso, G.; Midiri, A.; Gerace, E.; Biondo, C. Bacterial antibiotic resistance: The most critical pathogens. Pathogens 2021, 10, 1310. [Google Scholar] [CrossRef] [PubMed]
  149. Swanson, K.S.; Gibson, G.R.; Hutkins, R.; Reimer, R.A.; Reid, G.; Verbeke, K.; Scott, K.P.; Holscher, H.D.; Azad, M.B.; Delzenne, N.M.; et al. The International Scientific Association for Probiotics and Prebiotics (ISAPP) consensus statement on the definition and scope of synbiotics. Nat. Rev. Gastroenterol. Hepatol. 2020, 17, 687–701. [Google Scholar] [CrossRef]
  150. Arratia-Quijada, J.; Nuño, K.; Ruíz-Santoyo, V.; Andrade-Espinoza, B.A. Nano-encapsulation of probiotics: Need and critical considerations to design new non-dairy probiotic products. J. Funct. Foods 2024, 116, 106192. [Google Scholar] [CrossRef]
  151. Xuan, L.; Ju, Z.; Skonieczna, M.; Zhou, P.K.; Huang, R. Nanoparticles-induced potential toxicity on human health: Applications, toxicity mechanisms, and evaluation models. MedComm 2023, 4, e327. [Google Scholar] [CrossRef]
  152. Meena, K.K.; Taneja, N.K.; Ojha, A.; Meena, S. Application of spray-drying and freeze-drying for microencapsulation of lactic acid bacteria: A review. Ann. Phytomedicine Int. J. 2023, 12, 706–716. [Google Scholar] [CrossRef]
  153. Kandasamy, S.; Naveen, R. A review on the encapsulation of bioactive components using spray-drying and freeze-drying techniques. J. Food Process. Eng. 2022, 45, e14059. [Google Scholar] [CrossRef]
  154. Sousa, S.; Gomes, A.M.; Pintado, M.M.; Malcata, F.X.; Silva, J.P.; Sousa, J.M.; Costa, P.; Amaral, M.H.; Rodrigues, D.; Rocha-Santos, T.A.P.; et al. Encapsulation of probiotic strains in plain or cysteine-supplemented alginate improves viability at storage below freezing temperatures. Eng. Life Sci. 2012, 12, 457–465. [Google Scholar] [CrossRef]
  155. D’amico, V.; Cavaliere, M.; Ivone, M.; Lacassia, C.; Celano, G.; Vacca, M.; la Forgia, F.M.; Fontana, S.; De Angelis, M.; Denora, N.; et al. Microencapsulation of Probiotics for Enhanced Stability and Health Benefits in Dairy Functional Foods: A Focus on Pasta Filata Cheese. Pharmaceutics 2025, 17, 185. [Google Scholar] [CrossRef] [PubMed]
  156. Kaur, S.; Medhi, B. Guidelines for Nanopharmaceutical Products for Regulatory Approval. Int. J. Pharm. Sci. Nanotechnol. 2022, 15, 5723–5725. [Google Scholar] [CrossRef]
  157. Grilc, N.K.; Stojanov, S.; Rijavec, T.; Lapanje, A.; Berlec, A.; Zupančič, Š. Viability of potential probiotics incorporated into nanofibers: Influence of genera, storage conditions, stabilizers and their solid-state. Int. J. Pharm. 2025, 673, 125327. [Google Scholar] [CrossRef]
  158. Bósquez, J.P.A.; Oǧuz, E.; Cebeci, A.; Majadi, M.; Kiskó, G.; Gillay, Z.; Kovacs, Z. Characterization and Viability Prediction of Commercial Probiotic Supplements under Temperature and Concentration Conditioning Factors by NIR Spectroscopy. Fermentation 2022, 8, 66. [Google Scholar] [CrossRef]
  159. Nag, A.; Das, S. Improving ambient temperature stability of probiotics with stress adaptation and fluidized bed drying. J. Funct. Foods 2013, 5, 170–177. [Google Scholar] [CrossRef]
  160. Risks and Benefits of Probiotics. Available online: https://www.webmd.com/digestive-disorders/probiotics-risks-benefits (accessed on 13 May 2025).
  161. Kothari, D.; Patel, S.; Kim, S.K. Probiotic supplements might not be universally-effective and safe: A review. Biomed. Pharmacother. 2019, 111, 537–547. [Google Scholar] [CrossRef]
  162. Hadian, Y.; Bagood, M.D.; Dahle, S.E.; Sood, A.; Isseroff, R.R. Interleukin-17: Potential Target for Chronic Wounds. Mediat. Inflamm. 2019, 2019, 1297675. [Google Scholar] [CrossRef]
  163. Dudhat, K. Nanotechnology-Driven Approaches for Targeted Rectal Microbiome Modulation in Gastrointestinal Disorders. Curr. Proteom. 2025, 21, 681–710. [Google Scholar] [CrossRef]
  164. Abouelela, M.E.; Helmy, Y.A. Next-Generation Probiotics as Novel Therapeutics for Improving Human Health: Current Trends and Future Perspectives. Microorganisms 2024, 12, 430. [Google Scholar] [CrossRef]
  165. Chen, Y.; Wang, X.; Tao, S.; Wang, Q.; Ma, P.-Q.; Li, Z.-B.; Wu, Y.-L.; Li, D.-W. Research advances in smart responsive-hydrogel dressings with potential clinical diabetic wound healing properties. Mil. Med. Res. 2023, 10, 37. [Google Scholar] [CrossRef] [PubMed]
  166. D’urso, F.; Broccolo, F. Applications of Artificial Intelligence in Microbiome Analysis and Probiotic Interventions—An Overview and Perspective Based on the Current State of the Art. Appl. Sci. 2024, 14, 8627. [Google Scholar] [CrossRef]
  167. Encarnação, R.; Manuel, T.; Palheira, H.; Neves-Amado, J.; Alves, P. Artificial Intelligence in Wound Care Education: Protocol for a Scoping Review. Nurs. Rep. 2024, 14, 627–640. [Google Scholar] [CrossRef]
  168. Ezhilarasu, H.; Vishalli, D.; Dheen, S.T.; Bay, B.H.; Srinivasan, D.K. Nanoparticle-Based Therapeutic Approach for Diabetic Wound Healing. Nanomaterials 2020, 10, 1234. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Multifactorial roles of probiotics in wound healing; Antimicrobial action: via bacteriocins, organic acids, and hydrogen peroxide; Immunomodulation: regulating cytokines, macrophage polarization, dendritic cell activity; Oxidative stress reduction: through decreased ROS generation; Barrier enhancement: via upregulation of tight junction proteins; Tissue regeneration: through fibroblast activation and angiogenesis (Figure Created in Biorender. Monali Manghani (2025) https://app.biorender.com/illustrations/67a2f3345763bcd775f37436?slideId=70b8619c-8934-4282-b450-b3043be5b055, accessed on 17 June 2025).
Figure 1. Multifactorial roles of probiotics in wound healing; Antimicrobial action: via bacteriocins, organic acids, and hydrogen peroxide; Immunomodulation: regulating cytokines, macrophage polarization, dendritic cell activity; Oxidative stress reduction: through decreased ROS generation; Barrier enhancement: via upregulation of tight junction proteins; Tissue regeneration: through fibroblast activation and angiogenesis (Figure Created in Biorender. Monali Manghani (2025) https://app.biorender.com/illustrations/67a2f3345763bcd775f37436?slideId=70b8619c-8934-4282-b450-b3043be5b055, accessed on 17 June 2025).
Pharmaceutics 17 00805 g001
Figure 2. Nanotechnology-based encapsulation strategies for enhancing probiotic stability in wound healing applications (Figure Created in Biorender. Monali Manghani (2025) https://app.biorender.com/illustrations/678de110f49fd8be43cad043?slideId=3e28ea1f-4dd3-4295-85fa-2be90ac40d86, accessed on 17 June 2025).
Figure 2. Nanotechnology-based encapsulation strategies for enhancing probiotic stability in wound healing applications (Figure Created in Biorender. Monali Manghani (2025) https://app.biorender.com/illustrations/678de110f49fd8be43cad043?slideId=3e28ea1f-4dd3-4295-85fa-2be90ac40d86, accessed on 17 June 2025).
Pharmaceutics 17 00805 g002
Figure 3. Synergistic mechanisms of probiotic–nanotechnology hybrids in chronic wound healing. (Figure Created in Biorender. Monali Manghani (2025) https://app.biorender.com/illustrations/67a1a45f46c7e641660b8370?slideId=a8d17636-0ae1-4727-bb56-54f6e4d94af8, accessed on 17 June 2025).
Figure 3. Synergistic mechanisms of probiotic–nanotechnology hybrids in chronic wound healing. (Figure Created in Biorender. Monali Manghani (2025) https://app.biorender.com/illustrations/67a1a45f46c7e641660b8370?slideId=a8d17636-0ae1-4727-bb56-54f6e4d94af8, accessed on 17 June 2025).
Pharmaceutics 17 00805 g003
Table 1. Preclinical and Clinical Studies on Probiotics in Wound Healing.
Table 1. Preclinical and Clinical Studies on Probiotics in Wound Healing.
Study ModelProbiotic Strain(s)Delivery RouteMechanism(s)Key Outcome(s)Ref.
Wistar rats (excision model)Lactobacillus acidophilus + B. bifidumOralImmunomodulation, cytokine regulation↑ IL-10, ↓ TNF-α; 35% faster epithelialization by day 10[74]
Diabetic mice (streptozotocin)L. plantarumTopical hydrogelAngiogenesis, MMP regulation↑ VEGF, ↓ MMP-9; accelerated granulation and collagen organization[75]
Human clinical study (n = 30)Ligilactobacillus salivarius (formerly known as Lactobacillus salivarius)Oral (capsule)Microbiota modulation, systemic inflammation reductionImproved wound closure scores; ↓ CRP and IL-6 levels[76]
Mice (burn model)Lactobacillus delbrueckii subsp. BulgaricusTopical creamROS scavenging, AMP production↓ MDA levels; ↑ catalase and SOD activity; reduced wound infection[77]
Diabetic foot ulcer patientsSynbiotic (L. casei + Inulin)Oral (capsule)Gut-skin axis modulation, glycemic control↓ HbA1c and pro-inflammatory markers; ↑ wound healing index vs. placebo[78]
↑ indicates an upregulation or increase in the biomarker or cytokine level in response to treatment, suggesting enhanced biological activity. ↓ indicates a downregulation or decrease, signifying suppression of a specific inflammatory mediator.
Table 2. Comparative insights: Probiotic nanoformulations vs. Conventional Wound Therapies.
Table 2. Comparative insights: Probiotic nanoformulations vs. Conventional Wound Therapies.
ParameterConventional TherapiesProbiotic NanoformulationsRef.
Mechanism of ActionPrimarily antimicrobial; limited host interactionAntimicrobial + immunomodulatory + antioxidant[143]
Biofilm PenetrationLimited efficacy due to structural resistancePenetrates biofilms; secretes EPS-disrupting enzymes[144]
Impact on MicrobiomeDisrupts both pathogenic and commensal floraPromotes microbial balance and skin homeostasis[145]
Healing Efficacy (Preclinical)Slower wound closure; high inflammationFaster epithelialization, angiogenesis, and collagen deposition[146]
Healing Efficacy (Clinical)Variable, often limited in chronic wounds40–50% faster wound area reduction; improved pain and inflammation[147]
Resistance PotentialHigh; overuse leads to MDR pathogensLow; does not promote antibiotic resistance[148]
Cost-EffectivenessInitially low cost but high recurrence and complication rateHigher upfront cost, but reduced treatment time and recurrence[149]
Regulatory StatusWell-establishedEmerging; requires dual-level evaluation (probiotic + nanocarrier)[150]
Adverse EffectsPossible cytotoxicity, allergic reactions, microbiome lossMinimal adverse effects in trials; high biocompatibility[151]
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

Umekar, M.; Chaudhary, A.A.; Manghani, M.; Shidhaye, S.; Khajone, P.; Mahore, J.; Rudayni, H.A.; Trivedi, R. Probiotics in Nanotechnology-Driven Wound Healing: From Mechanistic Insight to Clinical Promise. Pharmaceutics 2025, 17, 805. https://doi.org/10.3390/pharmaceutics17070805

AMA Style

Umekar M, Chaudhary AA, Manghani M, Shidhaye S, Khajone P, Mahore J, Rudayni HA, Trivedi R. Probiotics in Nanotechnology-Driven Wound Healing: From Mechanistic Insight to Clinical Promise. Pharmaceutics. 2025; 17(7):805. https://doi.org/10.3390/pharmaceutics17070805

Chicago/Turabian Style

Umekar, Milind, Anis Ahmad Chaudhary, Monali Manghani, Supriya Shidhaye, Pratiksha Khajone, Jayashri Mahore, Hassan Ahmad Rudayni, and Rashmi Trivedi. 2025. "Probiotics in Nanotechnology-Driven Wound Healing: From Mechanistic Insight to Clinical Promise" Pharmaceutics 17, no. 7: 805. https://doi.org/10.3390/pharmaceutics17070805

APA Style

Umekar, M., Chaudhary, A. A., Manghani, M., Shidhaye, S., Khajone, P., Mahore, J., Rudayni, H. A., & Trivedi, R. (2025). Probiotics in Nanotechnology-Driven Wound Healing: From Mechanistic Insight to Clinical Promise. Pharmaceutics, 17(7), 805. https://doi.org/10.3390/pharmaceutics17070805

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