ArticlePublisher preview available

Microenvironment‐Responsive Hydrogels with Detachable Skin Adhesion and Mild‐Temperature Photothermal Property for Chronic Wound Healing

Wiley
Advanced Functional Materials
Authors:
To read the full-text of this research, you can request a copy directly from the authors.

Abstract and Figures

Chronic wounds have emerged as a global healthcare burden that have inability to heal within an expected time frame owing to the complex pathophysiological microenvironment, bacterial infection, multidrug resistance, and fragile skin tissue. Tailored to the features of chronic wounds, a multifunctional hydrogel (GA) composed of ethylenediamine‐modified gelatin (Gela‐amino) and oxidized sodium alginate (Alg─CHO) is reported. The thermosensitivity of gelatin and Schiff base bonds endow GA hydrogels with physiological temperature‐enhanced nonirritating tissue adhesion and low‐temperature‐triggered nondestructive separation, beneficial for the management of vulnerable and sensitive wounds. In addition, GA hydrogels exhibit pH‐responsive degradation that adapt to the acidic wound microenvironment. Under near‐infrared (NIR) irradiation, DNase I enzyme and indocyanine green‐loaded GA hydrogels (DI@GA) efficiently eradicate drug‐resistant bacteria biofilm and subsequently exert an antibacterial effect attributed to the bioactity of GA hydrogels and mild‐temperature photothermal therapy (≈45 °C). Remarkably, DI@GA/NIR hydrogels accelerate diabetic wound recovery and skin regeneration by dispersing biofilm, killing bacteria, inhibiting inflammation, promoting collagen deposition, and improving angiogenesis. The advanced hydrogels without the addition of antibiotics are promising to act as skin‐friendly wound dressings for rescuing infectious and stalled chronic wounds.
This content is subject to copyright. Terms and conditions apply.
RESEARCH ARTICLE
www.afm-journal.de
Microenvironment-Responsive Hydrogels with Detachable
Skin Adhesion and Mild-Temperature Photothermal
Property for Chronic Wound Healing
Huimin Geng, Xiaoyue Zheng, Yulin Zhang, Xiaomiao Cui, Zhiwei Li, Xunhui Zhang,
Jiwei Cui, Fangang Meng,* Lei Sun,* and Shilei Ni*
Chronic wounds have emerged as a global healthcare burden that have
inability to heal within an expected time frame owing to the complex
pathophysiological microenvironment, bacterial infection, multidrug
resistance, and fragile skin tissue. Tailored to the features of chronic wounds,
a multifunctional hydrogel (GA) composed of ethylenediamine-modified
gelatin (Gela-amino) and oxidized sodium alginate (AlgCHO) is reported.
The thermosensitivity of gelatin and Schiff base bonds endow GA hydrogels
with physiological temperature-enhanced nonirritating tissue adhesion and
low-temperature-triggered nondestructive separation, beneficial for the
management of vulnerable and sensitive wounds. In addition, GA hydrogels
exhibit pH-responsive degradation that adapt to the acidic wound
microenvironment. Under near-infrared (NIR) irradiation, DNase I enzyme
and indocyanine green-loaded GA hydrogels (DI@GA) efficiently eradicate
drug-resistant bacteria biofilm and subsequently exert an antibacterial effect
attributed to the bioactity of GA hydrogels and mild-temperature
photothermal therapy (45 °C). Remarkably, DI@GA/NIR hydrogels
accelerate diabetic wound recovery and skin regeneration by dispersing
biofilm, killing bacteria, inhibiting inflammation, promoting collagen
deposition, and improving angiogenesis. The advanced hydrogels without the
addition of antibiotics are promising to act as skin-friendly wound dressings
for rescuing infectious and stalled chronic wounds.
H. Geng, Y. Zhang, Z. Li, S. Ni
Department of Neurosurgery
Qilu Hospital of Shandong University and Institute of Brain and
Brain-Inspired Science
Cheeloo College of Medicine
Shandong University
Jinan, Shandong , China
E-mail: nishilei@sdu.edu.cn
H. Geng, X. Cui, X. Zhang, J. Cui
Key Laboratory of Colloid and Interface Chemistry of the Ministry of Educa-
tion
School of Chemistry and Chemical Engineering
Shandong University
Jinan, Shandong , China
The ORCID identification number(s) for the author(s) of this article
can be found under https://doi.org/./adfm.
DOI: 10.1002/adfm.202305154
1. Introduction
Chronic wounds including venous leg ul-
cers, diabetic foot ulcers, and pressure ul-
cers have become a major challenge to
worldwide healthcare systems and bring
an enormous socioeconomic burden.[1,2 ]
The complex molecular and cellular de-
ficiencies (e.g., excessive levels of proin-
flammatory cytokines, proteases, reactive
oxygen species, and a deficiency of stem
cells) in chronic wounds lead to their in-
ability to heal within an expected time
frame.[3,4 ] In addition, most of chronic
wounds are accompanied by bacterial con-
tamination and contain more than one
bacterial species (e.g., Staphylococcus,Pseu-
domonas,andEnterobacter).[5] Generally, the
microbial colonization in chronic wounds
can form robust biofilms with the aid
of self-produced extracellular polymeric
substances (i.e., polysaccharide, protein,
and external DNA) and organic acids.[6,7 ]
Biofilm can protect the bacteria from the
attack of conventional antibiotics and the
host’s immune clearance, eventually result-
ing in multidrug resistance.[2] Over 78%
X. Zheng, L. Sun
Department of Endocrinology
Qilu Hospital of Shandong University
Cheeloo College of Medicine
Shandong University
Jinan, Shandong , China
E-mail: Lei.Sun@email.sdu.edu.cn
F. Men g
Beijing Neurosurgical Institute
Beijing Tiantan Hospital
CapitalMedicalUniversity
Beijing , China
E-mail: fgmeng@ccmu.edu.cn
Adv. Funct. Mater. 2023,33,  ©  Wiley-VCH GmbH
2305154 (1 of 17)
... Compared with excisional infected wounds, burns are more likely to have heavier bacterial load and different healing outcomes requiring multi-modal therapies to meet the wound conditions. Several hydrogels have already been developed as drug delivery systems and can release antibacterial agents depending on environmental conditions using either light-triggered therapy [28] or environment stimuli-responsive systems [29]. However, the clinical translation of wound dressings relies heavily on effective release mechanisms to control highly resistant infections, ensure safety, modulate critical phases of burn healing, including, inflammation, and offer ease of application with high patient compliance. ...
... Enhanced intensities of peaks at 1629, 1559, and 1423 cm −1 in the HGO spectrum, corresponding to amide I, amide II, and amide III bands respectively, confirm the formation of Schiff base bonds. [53] ...
Article
Full-text available
An asymmetrical wound dressing functions akin to human skin by serving as a protective barrier between a wound and its immediate environment. However, significant challenges persist concerning the robust adhesion and asymmetrical adhesion properties of hydrogels, particularly when applied in emergency hemostasis and wound healing contexts. Herein, the study has successfully synthesized hydrogel patches with Janus asymmetric‐adhesion, denoted as HGO‐C, exclusively comprised of natural polymers. This achievement is realized through the assembly of adhesive hydrogel (HGO) and non‐adhesive hydrogel (CGC), thereby amalgamating their distinct functionalities. The non‐adhesive hydrogel component served as a physical shield and safeguarding the wound against contamination, while the adhesive hydrogel, when in contacted with the wound surface, firmly adhered to it, swiftly arresting bleeding and facilitating wound healing. Cytocompatibility tests, hemolysis tests, antibacterial assays, and coagulation assays demonstrated excellent biocompatibility, antibacterial, and hemostatic properties of HGO‐C. Finally, the in vivo experiments, including a liver hemorrhage assay and a wound healing assay, unequivocally showed the rapid hemostatic and enhanced wound healing capabilities of HGO‐C. Consequently, these distinctive hydrogel patches, derived from natural polymers and characterized by their asymmetric adhesion properties, may have great potential for real‐life usage in clinical patients.
Article
Chronic wounds constitute an increasingly prevalent global healthcare issue, characterized by recurring bacterial infections, pronounced oxidative stress, compromised functionality of immune cells, unrelenting inflammatory reactions, and deficits in angiogenesis. In response to these multifaceted challenges, the study introduced a stimulus‐responsive glycopeptide hydrogel constructed by oxidized Bletilla striata polysaccharide (OBSP), gallic acid‐grafted ε‐Polylysine (PLY‐GA), and paeoniflorin‐loaded micelles (MIC@Pae), called OBPG&MP. The hydrogel emulates the structure of glycoprotein fibers of the extracellular matrix (ECM), exhibiting exceptional injectability, self‐healing, and biocompatibility. It adapts responsively to the inflammatory microenvironment of chronic wounds, sequentially releasing therapeutic agents to eradicate bacterial infection, neutralize reactive oxygen species (ROS), modulate macrophage polarization, suppress inflammation, and encourage vascular regeneration and ECM remodeling, playing a critical role across the inflammatory, proliferative, and remodeling phases of wound healing. Both in vitro and in vivo studies confirmed the efficacy of OBPG&MP hydrogel in regulating the wound microenvironment and enhancing the regeneration and remodeling of chronic wound skin tissue. This research supports the vast potential for herb‐derived multifunctional hydrogels in tissue engineering and regenerative medicine.
Article
Full-text available
Excessive inflammation, bacterial infection, and blocked angiogenesis make diabetic wound healing challenging. Multifunctional wound dressings have several advantages in diabetic wound healing. In addition, the pH regulation of the microenvironment is shown to be a key factor that promotes skin regeneration through cellular immune regulation. However, few reports have focused on the development of functional dressings with the ability to regulate the pH microenvironment and promote diabetic wound healing. This study presents a novel approach for regulating the pH microenvironment of diabetic wound sites using a glycopeptide‐based hydrogel consisting of modified hyaluronic acid and poly(6‐aminocaproic acid). This hydrogel forms a network through Schiff base interactions and metal complexation, which suppresses inflammation and accelerates angiogenesis during wound healing. Hydrogels not only have adequate mechanical properties and self‐healing ability but can also support tissue adhesion. They can also promote the secretion of inducible cAMP early repressor, which promotes the polarization of macrophages toward the M2 type. The in vivo results confirm that hydrogel promotes diabetic wound repair and skin regeneration by exerting rapid anti‐inflammatory effects and promoting angiogenesis. Therefore, this hydrogel system represents an effective strategy for treating diabetic wounds.
Article
Full-text available
Wound with drug‐resistant bacterial infections has become a serious challenge for the healthcare system, and designing wound dressing to self‐adapt to the need of different stage of wound healing remains challenging. Herein, self‐adaptive wound dressings with multiple stimuli‐responsiveness and antibacterial activity are developed. Specifically, MoS2 carrying a reactive oxygen species (ROS) responsive nitric oxide (NO) release precursor L‐arginine (MSPA) is designed and incorporated into carboxymethyl chitosan/poly(N‐isopropylacrylamide) based cryogels (CMCS/PNIPAM) with multiple responsiveness (pH, near infrared (NIR), and temperature) to form self‐adaptive antibacterial cryogels that adapt to the therapeutic needs of different stages in infected wound healing. In response to the slightly acidic environment of bacterial infection, the cryogels assist the bacterial capture capacity through acid‐triggered protonation behavior, and effectively enhance the photodynamic antibacterial efficiency. Controllable on‐demand delivery of ROS, NO, and remote management of infected biofluid are achieved with NIR light as a trigger switch. The multiple stimuli‐responsive nanozyme‐based cryogels efficiently eliminate MRSA bacterial biofilm through NO assisted photodynamicand photothermal therapy (PDT&PTT). The multiple enzyme‐like activities of the cryogels effectively relieved oxidative damage. Notably, these cryogels effectively reduce wound infection, alleviated oxidative stress, and accelerate collagen deposition and angiogenesis in infected wounds, indicating that multiple stimuli‐responsive self‐adaptive wound dressings provide new ideas for infected wound treatment.
Article
Full-text available
Due to protection of extracellular polymeric substances, the therapeutic efficiency of conventional antimicrobial agents is often impeded by their poor infiltration and accumulation in biofilm. Herein, one type of surface charge adaptable nitric oxide (NO) nanogenerator was developed for biofilm permeation, retention and eradication. This nanogenerator (PDG@Au-NO/PBAM) is composed of a core-shell structure: thermo-sensitive NO donor conjugated AuNPs on cationic poly(dopamine-co-glucosamine) nanoparticle (PDG@Au-NO) served as core, and anionic phenylboronic acid-acryloylmorpholine (PBAM) copolymer was employed as a shell. The NO nanogenerator featured long circulation and good biocompatibility. Once the nanogenerator reached acidic biofilm, its surface charge would be switched to positive after shell dissociation and cationic core exposure, which was conducive for the nanogenerator to infiltrate and accumulate in the depth of biofilm. In addition, the nanogenerator could sustainably generate NO to disturb the integrity of biofilm at physiological temperature, then generate hyperthermia and explosive NO release upon NIR irradiation to efficiently eradicate drug-resistant bacteria biofilm. Such rational design offers a promising approach for developing nanosystems against biofilm-associated infections.
Article
Full-text available
Tissue adhesives have become vital candidates for the treatment of injuries, working as hemostatic agents for wound-healing and tissue-sealing. The most accessible commercial adhesives are based on cyanoacrylate and fibrin glue; however, they suffer from drawbacks such as cytotoxicity and poor adhesive strength, which limit their bioapplications. Therefore, the development of an adhesive system that improves tissue repair and closure while exhibiting low cytotoxicity is an important but challenging task. Herein, to overcome the limitations of commercial adhesives, we fabricated biocompatible hydrogels based on amino-functionalized polyaspartamide crosslinked with naturally occurring dextran aldehyde via Schiff base chemistry and investigated their adhesive properties. The crosslink density is affected by the ratio between the amine and aldehyde functional groups of each polymer, as well as by the polymer concentrations. We evaluated the adhesive characteristics of the hydrogels using rheometer and lap shear tests. The polyaspartamide-based adhesive hydrogels showed good mechanical strength and self-healing properties and higher adhesive properties than fibrin glue, demonstrating their potential for tissue adhesion applications.
Article
Full-text available
Bacterial infection, excessive inflammation and damaging blood vessels network are the major factors to delay the healing of diabetic ulcer. At present, most of wound repair materials are passive and can't response to the wound microenvironment, resulting in a low utilization of bioactive substances and hence a poor therapeutic effect. Therefore, it's essential to design an intelligent wound dressing responsive to the wound microenvironment to achieve the release of drugs on-demand on the basis of multifunctionality. In this work, metformin-laden CuPDA NPs composite hydrogel (Met@ CuPDA NPs/HG) was fabricated by dynamic phenylborate bonding of gelatin modified by dopamine (Gel-DA), Cu-loaded polydopamine nanoparticles (CuPDA NPs) with hyaluronic acid modified by phenyl boronate acid (HA-PBA), which possessed good injectability, self-healing, adhesive and DPPH scavenging performance. The slow release of metformin was achieved by the interaction with CuPDA NPs, boric groups (B-N coordination) and the constraint of hydrogel network. Metformin had a pH and glucose responsive release behavior to treat different wound microenvironment intelligently. Moreover, CuPDA NPs endowed the hydrogel excellent photothermal responsiveness to kill bacteria of >95% within 10 min and also the slow release of Cu2+ to protect wound from infection for a long time. Met@ CuPDA NPs/HG also recruited cells to a certain direction and promoted vascularization by releasing Cu2+. More importantly, Met@CuPDA NPs/HG effectively decreased the inflammation by eliminating ROS and inhibiting the activation of NF-κB pathway. Animal experiments demonstrated that Met@CuPDA NPs/HG significantly promoted wound healing of diabetic SD rats by killing bacteria, inhibiting inflammation, improving angiogenesis and accelerating the deposition of ECM and collagen. Therefore, Met@CuPDA NPs/HG had a great application potential for diabetic wound healing.
Article
Full-text available
Due to the continuous rise in biofilm‐related infections, biofilms seriously threaten human health. The formation of biofilms makes conventional antibiotics ineffective and dampens immune clearance. Therefore, it is important to understand the mechanisms of biofilm formation and develop novel strategies to treat biofilms more effectively. This review article begins with an introduction to biofilm formation in various clinical scenarios and their corresponding therapy. Established biofilm models used in research are then summarized. The potential targets which may assist in the development of new strategies for combating biofilms are further discussed. The novel technologies developed recently for the prevention and treatment of biofilms including antimicrobial surface coatings, physical removal of biofilms, development of new antimicrobial molecules, and delivery of antimicrobial agents are subsequently presented. Finally, directions for future studies are pointed out.
Article
Hydrogels, as one of the most feasible soft biomaterials, have gained considerable attention in therapeutic applications by virtue of their tunable properties including superior patient compliance, good biocompatibility and biodegradation, and high cargo-loading efficiency. However, hydrogel application is still limited by some challenges like inefficient encapsulation, easy leakage of loaded cargoes, and the lack of controllability. Recently, nanoarchitecture-integrated hydrogel systems were found to be therapeutics with optimized properties, extending their bioapplication. In this review, we briefly presented the category of hydrogels according to their synthetic materials and further discussed the advantages in bioapplication. Additionally, various applications of nanoarchitecture hybrid hydrogels in biomedical engineering are systematically summarized, including cancer therapy, wound healing, cardiac repair, bone regeneration, diabetes therapy, and obesity therapy. Last, the current challenges, limitations, and future perspectives in the future development of nanoarchitecture-integrated flexible hydrogels are addressed.
Article
Diabetic wounds have a complex microenvironment, due to local hypoxia, bacterial infection, and the accumulation of reactive oxygen species. Multiple adverse factors contribute to the non-healing state. Without effective treatment, diabetic wounds can lead to serious systemic complications. The current clinical treatments of enhanced glycemic control and anti-infection are unable to meet the needs of diabetic wound management. Here, we constructed a nanoparticle with catalase-like functionality (EGAP) and encapsulated it in a thermosensitive hydrogel to yield a versatile hydrogel formulation ([email protected]). The drug can form a protective physical barrier over the wound and create a moist environment. First, the EGAP converts H2O2 to O2 at the wound site, thereby alleviating local hypoxia. Then, the released EGF can promote the proliferation of epidermal cells. Furthermore, the gallic acid released from the outer layer of EGAP can exert anti-inflammatory and antioxidant effects. Finally, the silver ions released from the inner templates (APs) can kill bacteria. In vivo and in vitro experiments have shown that our treatment system can promote wound healing from multiple perspectives. This strategy by improving the local microenvironment of diabetic wounds provides a new theoretical basis and practical directions for chronic diabetic wound repair.
Article
The biofilm matrix can be considered to be a shared space for the encased microbial cells, comprising a wide variety of extracellular polymeric substances (EPS), such as polysaccharides, proteins, amyloids, lipids and extracellular DNA (eDNA), as well as membrane vesicles and humic-like microbially derived refractory substances. EPS are dynamic in space and time and their components interact in complex ways, fulfilling various functions: to stabilize the matrix, acquire nutrients, retain and protect eDNA or exoenzymes, or offer sorption sites for ions and hydrophobic substances. The retention of exoenzymes effectively renders the biofilm matrix an external digestion system influencing the global turnover of biopolymers, considering the ubiquitous relevance of biofilms. Physico-chemical and biological interactions and environmental conditions enable biofilm systems to morph into films, microcolonies and macrocolonies, films, ridges, ripples, columns, pellicles, bubbles, mushrooms and suspended aggregates — in response to the very diverse conditions confronting a particular biofilm community. Assembly and dynamics of the matrix are mostly coordinated by secondary messengers, signalling molecules or small RNAs, in both medically relevant and environmental biofilms. Fully deciphering how bacteria provide structure to the matrix, and thus facilitate and benefit from extracellular reactions, remains the challenge for future biofilm research. In this Review, Flemming et al. revisit our understanding of the biofilm matrix, focusing on the diversity of the extracellular polymeric substance components and novel aspects of mechanisms and consequences of their functional interactions.