Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 1.153
1.
Int J Nanomedicine ; 19: 4045-4060, 2024.
Article En | MEDLINE | ID: mdl-38736656

Purpose: Dry eye disease (DED) is a multifactorial ocular surface disease with a rising incidence. Therefore, it is urgent to construct a reliable and efficient drug delivery system for DED treatment. Methods: In this work, we loaded C-dots nanozyme into a thermosensitive in situ gel to create C-dots@Gel, presenting a promising composite ocular drug delivery system to manage DED. Results: This composite ocular drug delivery system (C-dots@Gel) demonstrated the ability to enhance adherence to the corneal surface and extend the ocular surface retention time, thereby enhancing bioavailability. Furthermore, no discernible ocular surface irritation or systemic toxicity was observed. In the DED mouse model induced by benzalkonium chloride (BAC), it was verified that C-dots@Gel effectively mitigated DED by stabilizing the tear film, prolonging tear secretion, repairing corneal surface damage, and augmenting the population of conjunctival goblet cells. Conclusion: Compared to conventional dosage forms (C-dots), the C-dots@Gel could prolong exhibited enhanced retention time on the ocular surface and increased bioavailability, resulting in a satisfactory therapeutic outcome for DED.


Antioxidants , Carbon , Cornea , Dry Eye Syndromes , Hydrogels , Animals , Dry Eye Syndromes/drug therapy , Mice , Carbon/chemistry , Antioxidants/chemistry , Antioxidants/pharmacokinetics , Antioxidants/pharmacology , Antioxidants/administration & dosage , Hydrogels/chemistry , Hydrogels/administration & dosage , Hydrogels/pharmacokinetics , Cornea/drug effects , Drug Delivery Systems/methods , Disease Models, Animal , Biological Availability , Tears/drug effects , Tears/chemistry , Benzalkonium Compounds/chemistry , Benzalkonium Compounds/administration & dosage , Benzalkonium Compounds/pharmacokinetics , Female , Male , Temperature , Quantum Dots/chemistry
2.
Int J Nanomedicine ; 19: 4081-4101, 2024.
Article En | MEDLINE | ID: mdl-38736654

Purpose: Spinal cord injury (SCI) is an incurable and disabling event that is accompanied by complex inflammation-related pathological processes, such as the production of excessive reactive oxygen species (ROS) by infiltrating inflammatory immune cells and their release into the extracellular microenvironment, resulting in extensive apoptosis of endogenous neural stem cells. In this study, we noticed the neuroregeneration-promoting effect as well as the ability of the innovative treatment method of FTY720-CDs@GelMA paired with NSCs to increase motor function recovery in a rat spinal cord injury model. Methods: Carbon dots (CDs) and fingolimod (FTY720) were added to a hydrogel created by chemical cross-linking GelMA (FTY720-CDs@GelMA). The basic properties of FTY720-CDs@GelMA hydrogels were investigated using TEM, SEM, XPS, and FTIR. The swelling and degradation rates of FTY720-CDs@GelMA hydrogels were measured, and each group's ability to scavenge reactive oxygen species was investigated. The in vitro biocompatibility of FTY720-CDs@GelMA hydrogels was assessed using neural stem cells. The regeneration of the spinal cord and recovery of motor function in rats were studied following co-treatment of spinal cord injury using FTY720-CDs@GelMA hydrogel in combination with NSCs, utilising rats with spinal cord injuries as a model. Histological and immunofluorescence labelling were used to determine the regeneration of axons and neurons. The recovery of motor function in rats was assessed using the BBB score. Results: The hydrogel boosted neurogenesis and axonal regeneration by eliminating excess ROS and restoring the regenerative environment. The hydrogel efficiently contained brain stem cells and demonstrated strong neuroprotective effects in vivo by lowering endogenous ROS generation and mitigating ROS-mediated oxidative stress. In a follow-up investigation, we discovered that FTY720-CDs@GelMA hydrogel could dramatically boost NSC proliferation while also promoting neuronal regeneration and synaptic formation, hence lowering cavity area. Conclusion: Our findings suggest that the innovative treatment of FTY720-CDs@GelMA paired with NSCs can effectively improve functional recovery in SCI patients, making it a promising therapeutic alternative for SCI.


Fingolimod Hydrochloride , Hydrogels , Neural Stem Cells , Rats, Sprague-Dawley , Spinal Cord Injuries , Animals , Spinal Cord Injuries/drug therapy , Spinal Cord Injuries/therapy , Fingolimod Hydrochloride/pharmacology , Fingolimod Hydrochloride/chemistry , Fingolimod Hydrochloride/administration & dosage , Neural Stem Cells/drug effects , Hydrogels/chemistry , Hydrogels/pharmacology , Hydrogels/administration & dosage , Rats , Recovery of Function/drug effects , Reactive Oxygen Species/metabolism , Quantum Dots/chemistry , Disease Models, Animal , Female , Spinal Cord/drug effects
3.
J Orthop Surg Res ; 19(1): 274, 2024 May 02.
Article En | MEDLINE | ID: mdl-38698396

OBJECTIVE: There are few effective osteoarthritis (OA) therapies. A novel injectable polyacrylamide hydrogel (iPAAG) previously demonstrated efficacy and safety up to week 26 in an open-label study of knee OA. Here we report longer-term effectiveness and safety data. METHODS: This multi-centre, open-label study included patients with symptomatic and radiographic knee OA. Primary outcome was WOMAC pain (0-100 scale) at 13 weeks, and patients continued to 26 weeks before entering a further 26-week extension phase. Secondary efficacy outcomes included WOMAC stiffness and function subscales, Patient Global Assessment (PGA) and proportion of OMERACT-OARSI responders. Safety outcomes were adverse events (AEs). RESULTS: 49 participants (31 women, mean age 70) received an ultrasound-guided, intra-articular injection of 6 ml iPAAG; 46 completed the extension phase to 52 weeks. There was a significant reduction in the WOMAC pain score from baseline to 52 weeks (- 17.7 points (95% CI - 23.1; - 12.4); p < 0.0001). Similar sustained improvements were observed for WOMAC stiffness (11.0 points; 95% CI - 17.0; - 4.9), physical function (18.0 points; 95% CI - 19.1; - 10.6), and PGA (16.3 points; 95% CI - 23.1; - 9.4). At 52 weeks 62.2% of patients were OMERACT-OARSI responders. From 26 to 52 weeks, 8 adverse effects (AE), including 1 serious AE (cerebrovascular accident) were reported in 5 subjects. None of the new adverse events were thought to be device related. CONCLUSION: This open-label study suggests persistent benefits and safety of iPAAG through 52 weeks after a single injection. TRIAL REGISTRATION: Clinicaltrials.gov NCT04179552.


Acrylic Resins , Osteoarthritis, Knee , Humans , Female , Osteoarthritis, Knee/drug therapy , Acrylic Resins/administration & dosage , Male , Aged , Middle Aged , Treatment Outcome , Follow-Up Studies , Injections, Intra-Articular , Time Factors , Hydrogels/administration & dosage , Aged, 80 and over
4.
J Control Release ; 369: 591-603, 2024 May.
Article En | MEDLINE | ID: mdl-38582336

Ischemia stroke is one of the leading causes of death and disability worldwide. Owing to the limited delivery efficiency to the brain caused by the blood-brain barrier (BBB) and off-target effects of systemic treatment, it is crucial to develop an in situ drug delivery system to improve the therapeutic effect in ischemic stroke. Briefly, we report a multifunctional in situ hydrogel delivery system for the co-delivery of reactive oxygen species (ROS)-responsive nanoparticles loaded with atorvastatin calcium (DSPE-se-se-PEG@AC NPs) and ß-nerve growth factor (NGF), which is expected to remodel pathological microenvironment for improving cerebral ischemia injury. The in vitro results exhibited the multifunctional hydrogel scavenged oxygen-glucose deprivation (OGD)-induced free radical, rescued the mitochondrial function, and maintained the survival and function of neurons, hence reducing neuronal apoptosis and neuroinflammation, consequently relieving ischemia injury in hippocampal neurons cell line (HT22). In the rat ischemia stroke model, the hydrogel significantly minified cerebral infarction by regulating inflammatory response, saving apoptotic neurons, and promoting angiogenesis and neurogenesis. Besides, the hydrogel distinctly improved the rats' neurological deficits after cerebral ischemia injury over the long-term observation. In conclusion, the in-situ hydrogel platform has demonstrated promising therapeutic effects in both in vitro and in vivo studies, indicating its potential as a new and effective therapy.


Atorvastatin , Brain Ischemia , Hydrogels , Rats, Sprague-Dawley , Animals , Hydrogels/administration & dosage , Brain Ischemia/drug therapy , Male , Atorvastatin/administration & dosage , Atorvastatin/therapeutic use , Atorvastatin/pharmacology , Cell Line , Reactive Oxygen Species/metabolism , Nanoparticles/administration & dosage , Brain/drug effects , Brain/pathology , Brain/metabolism , Nerve Growth Factor/administration & dosage , Mice , Neurons/drug effects , Neurons/pathology , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/therapeutic use , Neuroprotective Agents/pharmacology , Rats , Apoptosis/drug effects , Polyethylene Glycols/chemistry , Polyethylene Glycols/administration & dosage , Drug Delivery Systems , Ischemic Stroke/drug therapy , Ischemic Stroke/pathology
5.
J Control Release ; 369: 604-616, 2024 May.
Article En | MEDLINE | ID: mdl-38582337

Corneal stromal fibrosis is a common cause of visual impairment resulting from corneal injury, inflammation and surgery. Therefore, there is an unmet need for inhibiting corneal stromal fibrosis. However, bioavailability of topical eye drops is very low due to the tear and corneal barriers. In situ delivery offers a unique alternative to improve efficacy and minimize systemic toxicity. Herein, a drug delivery platform based on thermoresponsive injectable hydrogel/nano-micelles composite with in situ drug-controlled release and long-acting features is developed to prevent corneal scarring and reduce corneal stromal fibrosis in lamellar keratoplasty. The in-situ gelation hydrogels enabled direct delivery of celastrol to the corneal stroma. In vivo evaluation with a rabbit anterior lamellar keratoplasty model showed that hydrogel/micelles platform could effectively inhibit corneal stromal fibrosis. This strategy achieves controlled and prolonged release of celastrol in the corneal stroma of rabbit. Following a single corneal interlamellar injection, celastrol effectively alleviated fibrosis via mTORC1 signal promoting autophagy and inhibiting TGF-ß1/Smad2/3 signaling pathway. Overall, this strategy demonstrates promise for the clinical application of celastrol in preventing corneal scarring and reducing corneal stromal fibrosis post-lamellar keratoplasty, highlighting the potential benefits of targeted drug delivery systems in ocular therapeutics.


Corneal Transplantation , Hydrogels , Pentacyclic Triterpenes , Animals , Rabbits , Pentacyclic Triterpenes/administration & dosage , Hydrogels/administration & dosage , Corneal Transplantation/methods , Cicatrix/prevention & control , Cicatrix/drug therapy , Delayed-Action Preparations , Fibrosis , Drug Delivery Systems , Cornea/drug effects , Cornea/metabolism , Triterpenes/administration & dosage , Drug Liberation , Corneal Stroma/drug effects , Humans
6.
J Control Release ; 369: 545-555, 2024 May.
Article En | MEDLINE | ID: mdl-38588825

Severe burn injuries with massive dermal loss are often underestimated despite their significant impact on morbidity and mortality. Resembling the natural extracellular matrix (ECM), hyaluronic acid (HA)-based dressings have been extensively explored as suitable candidates for burn wound treatment. However, native HA hydrogel's limitations, such as low mechanical strength, rapid degradation, and uncontrollable drug delivery, hinder its efficacy, especially for full-thickness burns requiring injectable hydrogels with robust antibacterial and angiogenic capabilities. Herein, we present a novel multifunctional sequential dual-curing hydrogel system, combining hyperbranched poly(DMA-DMAPMA-PEGDA) (DDP) polymer with thiolated hyaluronic acid (HA-SH). The DDP copolymer, featuring multi-vinyls and catechol functionalities, facilitates two curing reactions taking place sequentially with HA-SH under physiological conditions, balancing convenient injection with the mechanical strength essential for effective wound management. Furthermore, the resulting DDP/HA hydrogels demonstrate enhanced therapeutic attributes, including intrinsic angiogenic and antimicrobial effects, setting them as promising dressing options for deep burn wound therapy.


Burns , Catechols , Hyaluronic Acid , Hydrogels , Wound Healing , Hyaluronic Acid/chemistry , Hyaluronic Acid/administration & dosage , Hydrogels/administration & dosage , Hydrogels/chemistry , Burns/drug therapy , Burns/therapy , Animals , Catechols/administration & dosage , Catechols/chemistry , Wound Healing/drug effects , Humans , Male , Bandages , Anti-Bacterial Agents/administration & dosage , Anti-Bacterial Agents/chemistry , Regeneration/drug effects , Cross-Linking Reagents/chemistry , Injections , Mice
7.
ACS Biomater Sci Eng ; 10(5): 3164-3172, 2024 May 13.
Article En | MEDLINE | ID: mdl-38671385

Intestinal adhesion is one of the complications that occurs more frequently after abdominal surgery. Postsurgical intestinal adhesion (PIA) can lead to a series of health problems, including abdominal pain, intestinal obstruction, and female infertility. Currently, hydrogels and nanofibrous films as barriers are often used for preventing PIA formation; however, these kinds of materials have their intrinsic disadvantages. Herein, we developed a dual-structure drug delivery patch consisting of poly lactic-co-glycolic acid (PLGA) nanofibers and a chitosan hydrogel (NHP). PLGA nanofibers loaded with deferoxamine mesylate (DFO) were incorporated into the hydrogel; meanwhile, the hydrogel was loaded with anti-inflammatory drug dexamethasone (DXMS). The rapid degradation of the hydrogel facilitated the release of DXMS at the acute inflammatory stage of the early injury and provided effective anti-inflammatory effects for wound sites. Moreover, PLGA composite nanofibers could provide sustained and stable release of DFO for promoting the peritoneal repair by the angiogenesis effects of DFO. The in vivo results indicated that NHP can effectively prevent PIA formation by restraining inflammation and vascularization, promoting peritoneal repair. Therefore, we believe that our NHP has a great potential application in inhibition of PIA.


Dexamethasone , Drug Delivery Systems , Hydrogels , Nanofibers , Polylactic Acid-Polyglycolic Acid Copolymer , Nanofibers/chemistry , Nanofibers/therapeutic use , Hydrogels/chemistry , Hydrogels/pharmacology , Hydrogels/administration & dosage , Tissue Adhesions/prevention & control , Animals , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Dexamethasone/pharmacology , Dexamethasone/administration & dosage , Dexamethasone/therapeutic use , Chitosan/chemistry , Chitosan/pharmacology , Intestines/drug effects , Anti-Inflammatory Agents/administration & dosage , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/therapeutic use , Postoperative Complications/prevention & control , Rats, Sprague-Dawley , Mice , Female , Rats
8.
Biomater Sci ; 12(10): 2460-2479, 2024 May 14.
Article En | MEDLINE | ID: mdl-38578143

Chronic wounds have gradually evolved into a global health challenge, comprising long-term non-healing wounds, local tissue necrosis, and even amputation in severe cases. Accordingly, chronic wounds place a considerable psychological and economic burden on patients and society. Chronic wounds have multifaceted pathogenesis involving excessive inflammation, insufficient angiogenesis, and elevated reactive oxygen species levels, with bacterial infection playing a crucial role. Hydrogels, renowned for their excellent biocompatibility, moisture retention, swelling properties, and oxygen permeability, have emerged as promising wound repair dressings. However, hydrogels with singular functions fall short of addressing the complex requirements associated with chronic wound healing. Hence, current research emphasises the development of multifunctional antibacterial hydrogels. This article reviews chronic wound characteristics and the properties and classification of antibacterial hydrogels, as well as their potential application in chronic wound management.


Anti-Bacterial Agents , Hydrogels , Wound Healing , Hydrogels/chemistry , Hydrogels/pharmacology , Hydrogels/administration & dosage , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Humans , Wound Healing/drug effects , Animals , Chronic Disease , Bandages
9.
Biomater Sci ; 12(10): 2561-2578, 2024 May 14.
Article En | MEDLINE | ID: mdl-38602364

The targeted delivery of pharmacologically active molecules, metabolites, and growth factors to the brain parenchyma has become one of the major challenges following the onset of neurodegeneration and pathological conditions. The therapeutic effect of active biomolecules is significantly impaired after systemic administration in the central nervous system (CNS) because of the blood-brain barrier (BBB). Therefore, the development of novel therapeutic approaches capable of overcoming these limitations is under discussion. Exosomes (Exo) are nano-sized vesicles of endosomal origin that have a high distribution rate in biofluids. Recent advances have introduced Exo as naturally suitable bio-shuttles for the delivery of neurotrophic factors to the brain parenchyma. In recent years, many researchers have attempted to regulate the delivery of Exo to target sites while reducing their removal from circulation. The encapsulation of Exo in natural and synthetic hydrogels offers a valuable strategy to address the limitations of Exo, maintaining their integrity and controlling their release at a desired site. Herein, we highlight the current and novel approaches related to the application of hydrogels for the encapsulation of Exo in the field of CNS tissue engineering.


Drug Delivery Systems , Exosomes , Hydrogels , Exosomes/chemistry , Exosomes/metabolism , Hydrogels/chemistry , Hydrogels/administration & dosage , Humans , Animals , Central Nervous System/metabolism , Central Nervous System/drug effects , Blood-Brain Barrier/metabolism , Tissue Engineering , Drug Carriers/chemistry
10.
J Cosmet Dermatol ; 23(6): 2125-2134, 2024 Jun.
Article En | MEDLINE | ID: mdl-38590107

OBJECTIVE: Salicylic acid (SA) has been used for treatment of acne of different severity levels. However, there are few researches about the safety and efficacy for treatment of mild to moderate acne, and the improvement of the skin condition by using 2% supramolecular salicylic acid (SSA) compared to Davuwen Adapaline gel. METHODS: A multicenter, randomized, assessor-blind and parallel-controlled study was conducted. A total of 500 patients (trial group: 249, control group: 251) with mild to moderate (grade I-II) facial acne vulgaris were recruited in this study over a 16-week trial period. Patients in the trial group were treated with Broda 2% SSA hydrogel, while control group treated with Davuwen Adapaline gel once a day. The number of inflammatory papules, comedones, and pustules were counted and the rate of lesion reduction was calculated pre- and post-treatment. Then, the skin physiological indicators, including L*a*b*, TEWL, skin sebum and hydration were measured. Statistical analysis was conducted using SAS 9.4. Significance was set at p = 0.05. RESULTS: At the end of 12 weeks' therapy, the regression and markedly improvement rate of the trail group and the control group were 51.01% and 43.10% respectively, and there was no significant difference in the improvement rate between two groups (p = 0.0831). Although, there was no difference in adverse events rate between two groups, the adverse events rate of the trail group was 0.40%, a little lower than the control group (0.80%). Moreover, there was a significant difference in the numbers of pores at T1 between two groups. CONCLUSION: Both 2% SSA and Adapaline gel were equally effective in the treatment of mild to moderate acne vulgaris. 2% SSA is worth the clinical promotion and application in mild to moderate acne vulgaris.


Acne Vulgaris , Gels , Hydrogels , Salicylic Acid , Severity of Illness Index , Humans , Acne Vulgaris/drug therapy , Female , Male , Salicylic Acid/administration & dosage , Salicylic Acid/adverse effects , Salicylic Acid/therapeutic use , Young Adult , Adolescent , Adult , Single-Blind Method , Hydrogels/administration & dosage , Treatment Outcome , Dermatologic Agents/administration & dosage , Dermatologic Agents/adverse effects , Administration, Cutaneous , Adapalene/administration & dosage , Adapalene/adverse effects
11.
Expert Opin Drug Deliv ; 21(4): 573-591, 2024 Apr.
Article En | MEDLINE | ID: mdl-38588553

INTRODUCTION: Endotracheal intubation is a common procedure to maintain an open airway with risks for traumatic injury. Pathological changes resulting from intubation can cause upper airway complications, including vocal fold scarring, laryngotracheal stenosis, and granulomas and present with symptoms such as dysphonia, dysphagia, and dyspnea. Current intubation-related laryngotracheal injury treatment approaches lack standardized guidelines, relying on individual clinician experience, and surgical and medical interventions have limitations and carry risks. AREAS COVERED: The clinical and preclinical therapeutics for wound healing in the upper airway are described. This review discusses the current developments on local drug delivery systems in the upper airway utilizing particle-based delivery systems, including nanoparticles and microparticles, and bulk-based delivery systems, encompassing hydrogels and polymer-based approaches. EXPERT OPINION: Complex laryngotracheal diseases pose challenges for effective treatment, struggling due to the intricate anatomy, limited access, and recurrence. Symptomatic management often requires invasive surgical procedures or medications that are unable to achieve lasting effects. Recent advances in nanotechnology and biocompatible materials provide potential solutions, enabling precise drug delivery, personalization, and extended treatment efficacy. Combining these technologies could lead to groundbreaking treatments for upper airways diseases, significantly improving patients' quality of life. Research and innovation in this field are crucial for further advancements.


Drug Delivery Systems , Wound Healing , Humans , Wound Healing/drug effects , Animals , Intubation, Intratracheal/methods , Quality of Life , Nanoparticles , Hydrogels/administration & dosage , Biocompatible Materials/administration & dosage , Biocompatible Materials/chemistry , Polymers/chemistry , Nanotechnology , Laryngeal Diseases/drug therapy , Trachea/injuries
12.
Int J Pharm ; 656: 124099, 2024 May 10.
Article En | MEDLINE | ID: mdl-38614431

Diabetic wounds (DWs) pose a significant health burden worldwide, with their management presenting numerous challenges. Biopolymeric formulations have recently gained attention as promising therapeutic approaches for diabetic wound healing. These formulations, composed of biocompatible and biodegradable polymers, offer unique properties such as controlled drug release, enhanced wound closure, and reduced scarring. In this review, we aim to provide a comprehensive overview of the current state of research and future prospects regarding the application of biopolymeric formulations for diabetic wound healing. The review begins by highlighting the underlying pathophysiology of DWs, including impaired angiogenesis, chronic inflammation, and compromised extracellular matrix (ECM) formation. It further explores the key characteristics of biopolymeric materials, such as their biocompatibility, biodegradability, and tunable physicochemical properties, which make them suitable for diabetic wound healing applications. The discussion further delves into the types of biopolymeric formulations utilized in the treatment of DWs. These include hydrogels, nanoparticles (NP), scaffolds, films, and dressings. Furthermore, the review addresses the challenges associated with biopolymeric formulations for diabetic wound healing. In conclusion, biopolymeric formulations present a promising avenue for diabetic wound healing. Their unique properties and versatility allow for tailored approaches to address the specific challenges associated with DWs. However, further research and developments are required to optimize their therapeutic efficacy, stability, manufacturing processes, and regulatory considerations. With continued advancements in biopolymeric formulations, the future holds great promise for improving the management and outcomes of DWs.


Wound Healing , Wound Healing/drug effects , Humans , Biopolymers/chemistry , Biopolymers/administration & dosage , Animals , Diabetes Mellitus/drug therapy , Hydrogels/chemistry , Hydrogels/administration & dosage , Bandages , Biocompatible Materials/chemistry , Biocompatible Materials/administration & dosage , Nanoparticles/chemistry
13.
J Control Release ; 369: 573-590, 2024 May.
Article En | MEDLINE | ID: mdl-38554773

Postoperative abdominal adhesions are a common clinical problem after surgery and can cause many serious complications. Current most commonly used antiadhesion products are less effective due to their short residence time and focus primary on barrier function. Herein, we developed a sprayable hydrogel barrier (sHA-ADH/OHA-E) with self-regulated drug release based on ROS levels at the trauma site, to serve as a smart inflammatory microenvironment modulator and GATA6+ macrophages trap for non-adherent recovery from abdominal surgery. Sulfonated hyaluronic acid (HA) conjugates modified with adipic dihydrazide (sHA-ADH), and oxidized HA conjugates grafted with epigallocatechin-3-gallate (EGCG) via ROS-cleavable boronate bonds (OHA-E) were synthesized. sHA-ADH/OHA-E hydrogel was facilely fabricated within 5 s after simply mixing sHA-ADH and OHA-E through forming dynamic covalent acylhydrazones. With good biocompatibility, appropriate mechanical strength, tunable shear-thinning, self-healing, asymmetric adhesion, and reasonable in vivo retention time, sHA-ADH/OHA-E hydrogel meets the requirements of a perfect physical barrier. Intriguingly, sulfonic acid groups endowed the hydrogel with satisfactory anti-fibroblast and macrophage attachment capability, and were demonstrated for the first time to act as polyanion traps to prevent GATA6+ macrophages aggregation. Importantly, EGCG could be intelligently released by ROS triggering to alleviate oxidative stress and promote proinflammatory M1 macrophage polarize to antiinflammatory M2 phenotype. Further, the fibrinolytic system balance was restored to reduce fibrosis. Thanks to the above advantages, the sHA-ADH/OHA-E hydrogel exhibited excellent anti-adhesion effects in a rat sidewall defect-cecum abrasion model and is expected to be a promising and clinically translatable antiadhesion barrier.


GATA6 Transcription Factor , Hyaluronic Acid , Hydrogels , Macrophages , Postoperative Complications , Reactive Oxygen Species , Tissue Adhesions/prevention & control , Animals , Hydrogels/chemistry , Hydrogels/administration & dosage , Macrophages/drug effects , Macrophages/metabolism , Reactive Oxygen Species/metabolism , Hyaluronic Acid/chemistry , Postoperative Complications/prevention & control , GATA6 Transcription Factor/metabolism , Catechin/analogs & derivatives , Catechin/chemistry , Catechin/administration & dosage , Catechin/pharmacology , Rats, Sprague-Dawley , Mice , Adipates/chemistry , Male , Abdomen/surgery , RAW 264.7 Cells , Free Radical Scavengers/administration & dosage , Free Radical Scavengers/chemistry , Free Radical Scavengers/pharmacology , Drug Liberation
14.
Biomater Sci ; 12(9): 2312-2320, 2024 Apr 30.
Article En | MEDLINE | ID: mdl-38497434

Postsurgical treatment comprehensively benefits from the application of tissue-adhesive injectable hydrogels, which reduce postoperative complications by promoting wound closure and tissue regeneration. Although various hydrogels have been employed as clinical tissue adhesives, many exhibit deficiencies in adhesive strength under wet conditions or in immunomodulatory functions. Herein, we report the development of reactive oxygen species (ROS) scavenging and tissue-adhesive injectable hydrogels composed of polyamine-modified gelatin crosslinked with the 4-arm poly (ethylene glycol) crosslinker. Polyamine-modified gelatin was particularly potent in suppressing the secretion of proinflammatory cytokines from stimulated primary macrophages. This effect is attributed to its ability to scavenge ROS and inhibit the nuclear translocation of nuclear factor kappa-B. Polyamine-modified gelatin-based hydrogels exhibited ROS scavenging abilities and enhanced tissue adhesive strength on collagen casing. Notably, the hydrogel demonstrated exceptional tissue adhesive properties in a wet environment, as evidenced by its performance using porcine small intestine tissue. This approach holds significant promise for designing immunomodulatory hydrogels with superior tissue adhesion strength compared to conventional medical materials, thereby contributing to advancements in minimally invasive surgical techniques.


Gelatin , Hydrogels , Reactive Oxygen Species , Tissue Adhesives , Hydrogels/chemistry , Hydrogels/administration & dosage , Hydrogels/pharmacology , Animals , Tissue Adhesives/chemistry , Tissue Adhesives/pharmacology , Tissue Adhesives/administration & dosage , Reactive Oxygen Species/metabolism , Mice , Swine , Gelatin/chemistry , Polyethyleneimine/chemistry , Free Radical Scavengers/chemistry , Free Radical Scavengers/pharmacology , Free Radical Scavengers/administration & dosage , Macrophages/drug effects , Macrophages/metabolism , Polyethylene Glycols/chemistry , Injections , Cytokines/metabolism , Intestine, Small/metabolism , Intestine, Small/drug effects
15.
Biomater Sci ; 12(9): 2356-2368, 2024 Apr 30.
Article En | MEDLINE | ID: mdl-38497791

Corneal transplantation is the gold standard treatment for corneal-related blindness; however, this strategy faces challenges such as limited donor cornea, graft rejection, suture-related complications, and the need for specialized equipment and advanced surgical skills. Development of tissue adhesives for corneal regeneration is of great clinical value. However, currently available corneal tissue sealants pose challenges, such as lack of safety, biocompatibility, and desired mechanical properties. To meet these requirements simultaneously, a bovine stromal corneal extracellular matrix (dCor) was used to design a bioadhesive photocurable hydrogel based on gelatin methacrylate (GelMA) and polyethylene glycol diacrylate (PEGDA) hydrogels (dCor/Gel-PEG). Integration of dCor into the dual networks of GelMA and PEGDA (Gel-PEG) led to a bioadhesive hydrogel for curing corneal defects, which could be crosslinked by Irgacure 2959 within 5 min ultraviolet irradiation. The viability of corneal stromal stem cells (CSSCs) was improved on the dCor/Gel-PEG hydrogel in comparison to the Gel-PEG hydrogel. The gene expression profile supported the keratocyte differentiation of CSSCs seeded on dCor/Gel-PEG via increased KERA and ALDH, with inhibited myofibroblast transdifferentiation via decreased α-SMA due to the presence of dCor. Interestingly, the dCor/Gel-PEG hydrogel exhibited favorable mechanical performance in terms of elasticity and bioadherence to the host corneal stroma. Ex vivo and in vivo examinations proved the feasibility of this hydrogel for the sutureless reconstruction of deep anterior corneal defects with promising histopathological results.


Extracellular Matrix , Gelatin , Hydrogels , Polyethylene Glycols , Animals , Hydrogels/chemistry , Hydrogels/pharmacology , Hydrogels/administration & dosage , Cattle , Polyethylene Glycols/chemistry , Gelatin/chemistry , Extracellular Matrix/chemistry , Tissue Adhesives/chemistry , Tissue Adhesives/pharmacology , Tissue Adhesives/administration & dosage , Methacrylates/chemistry , Cornea , Stem Cells/cytology , Stem Cells/drug effects
16.
Int J Pharm ; 656: 124029, 2024 May 10.
Article En | MEDLINE | ID: mdl-38527566

α-Bisabolol (αBIS), a plant-derived compound with anti-inflammatory properties, is potentially a therapeutic agent for Atopic dermatitis. However, its poor water solubility and photoinstability limit its topical application. Therefore, the present study, aimed to develop cationic polymeric nanocapsules of αBIS to improve its skin delivery, photostability, and therapeutic efficacy. The αBIS-loaded nanocapsules were prepared using the solvent displacement technique. A Box-Behnken (BB) design was employed to statistically optimize formulation variables and αBIS-loaded nanocapsules characterized by particle size, surface charge and encapsulation efficiency. The optimal formulation was selected, and the spherical shape of the nanocapsules was confirmed by scanning electron microscopy (SEM). Furthermore, hydrogel containing αBIS-loaded nanocapsules was prepared by thickening of nanocapsule suspension with Carbopol 934 and evaluated for rheology, in vitro drug release and skin permeation. Furthermore, a mice model of atopic dermatitis was used to evaluate the anti-inflammatory potential of the hydrogels. The optimal formulation displayed a spherical morphology under scanning electron microscopy (SEM) with an optimum particle size of 133.00 nm, polydispersity index (PDI) of 0.12, high EE% of 93 %, and improved optical stability of αBIS in the prepared nanocapsules compared to the free drug. The nano-based hydrogels demonstrated non-Newtonian pseudoplastic behavior and an increased αBIS in vitro release profile without causing skin irritation in rabbits. Drug retention within the dermis and epidermis layers significantly surpassed that of drug-free hydrogel. Moreover, in vivo histopathological studies and myeloperoxidase (MPO) enzyme activity, revealed that hydrogel containing bisabolol nanocapsules exhibited The best anti-inflammatory effect. The results showed that hydrogels containing bisabolol nanocapsules markedly alleviated dermatitis-related inflammation and reduced skin thickness in Balb/c mice. Our findings support nanocapsules as an effective drug delivery system to enhance αBIS stability, bioavailability, and therapeutic efficacy in AD treatment.


Anti-Inflammatory Agents , Dermatitis, Atopic , Drug Liberation , Hydrogels , Mice, Inbred BALB C , Monocyclic Sesquiterpenes , Nanocapsules , Animals , Hydrogels/chemistry , Hydrogels/administration & dosage , Nanocapsules/chemistry , Dermatitis, Atopic/drug therapy , Monocyclic Sesquiterpenes/administration & dosage , Anti-Inflammatory Agents/administration & dosage , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/pharmacology , Skin Absorption/drug effects , Particle Size , Disease Models, Animal , Mice , Administration, Cutaneous , Male , Skin/drug effects , Skin/metabolism , Skin/pathology , Sesquiterpenes/administration & dosage , Sesquiterpenes/chemistry , Sesquiterpenes/pharmacology , Sesquiterpenes/pharmacokinetics , Female
17.
Int J Biol Macromol ; 266(Pt 1): 131175, 2024 May.
Article En | MEDLINE | ID: mdl-38552696

Myocardial ischemia-reperfusion injury (MIRI) significantly contributes to the high incidence of complications and mortality associated with acute myocardial infarction. Recently, injectable electroconductive hydrogels (IECHs) have emerged as promising tools for replicating the mechanical, electroconductive, and physiological characteristics of cardiac tissue. Herein, we aimed to develop a novel IECH by incorporating irbesartan as a drug delivery system (DDS) for cardiac repair. Our approach involved merging a conductive poly-thiophene derivative (PEDOT: PSS) with an injectable dual-network adhesive hydrogel (DNAH) comprising a catechol-branched polyacrylamide network and a chitosan-hyaluronic acid covalent network. The resulting P-DNAH hydrogel, benefitting from a high conducting polymer content, a chemically crosslinked network, a robust dissipative matrix, and dynamic oxidation of catechol to quinone exhibited superior mechanical strength, desirable conductivity, and robust wet-adhesiveness. In vitro experiments with the P-DNAH hydrogel carrying irbesartan (P-DNAH-I) demonstrated excellent biocompatibility by cck-8 kit on H9C2 cells and a rapid initial release of irbesartan. Upon injection into the infarcted hearts of MIRI mouse models, the P-DNAH-I hydrogel effectively inhibited the inflammatory response and reduced the infarct size. In conclusion, our results suggest that the P-DNAH hydrogel, possessing suitable mechanical properties and electroconductivity, serves as an ideal IECH for DDS, delivering irbesartan to promote heart repair.


Acrylic Resins , Chitosan , Hydrogels , Myocardial Reperfusion Injury , Irbesartan/administration & dosage , Myocardial Reperfusion Injury/drug therapy , Chitosan/administration & dosage , Chitosan/chemistry , Acrylic Resins/administration & dosage , Acrylic Resins/chemistry , Hydrogels/administration & dosage , Hydrogels/chemistry , Hydrogels/toxicity , Electric Conductivity , Elasticity , Injections , Cell Line , Animals , Rats , Disease Models, Animal , Mice , Male , Mice, Inbred C57BL , Cell Survival/drug effects
18.
J Control Release ; 369: 296-308, 2024 May.
Article En | MEDLINE | ID: mdl-38301925

Immunosuppression caused by incomplete radiofrequency ablation (iRFA) is a crucial factor affecting the effectiveness of RFA for solid tumors. However, little is known about the changes iRFA induces in the tumor immune microenvironment (TIME) of hepatocellular carcinoma (HCC), the primary application area for RFA. In this study, we found iRFA promotes a suppressive TIME in residual HCC tumors, characterized by M2 macrophage polarization, inhibited antigen presentation by dendritic cells (DCs), and reduced infiltration of cytotoxic T lymphocytes (CTLs). Interestingly, the STING agonist MSA-2 was able to reorganize M2-like tumor-promoting macrophages into M1-like anti-tumor states and enhance antigen presentation by DCs. To optimize the therapeutic effect of MSA-2, we used a calcium ion (Ca2+) responsive sodium alginate (ALG) as a carrier, forming an injectable hydrogel named ALG@MSA-2. This hydrogel can change from liquid to gel, maintaining continuous drug release in situ. Our results suggested that ALG@MSA-2 effectively activated anti-tumor immunity, as manifested by increased M1-like macrophage polarization, enhanced antigen presentation by DCs, increased CTL infiltration, and inhibited residual tumor growth. ALG@MSA-2 also resulted in a complete regression of contralateral tumors and widespread liver metastases in vivo. In addition, the excellent biosafety of ALG@MSA-2 was also proved by blood biochemical analysis and body weight changes in mice. In summary, this study demonstrated that the immune cascade of ALG@MSA-2 mediated the STING pathway activation and promoted a favorable TIME which might provide novel insights for the RFA treatment of HCC.


Alginates , Carcinoma, Hepatocellular , Hydrogels , Liver Neoplasms , Membrane Proteins , Mice, Inbred C57BL , Radiofrequency Ablation , Animals , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/therapy , Liver Neoplasms/drug therapy , Liver Neoplasms/immunology , Liver Neoplasms/therapy , Hydrogels/administration & dosage , Radiofrequency Ablation/methods , Alginates/chemistry , Alginates/administration & dosage , Dendritic Cells/drug effects , Dendritic Cells/immunology , Cell Line, Tumor , Macrophages/drug effects , Macrophages/immunology , Tumor Microenvironment/drug effects , Mice , Male , T-Lymphocytes, Cytotoxic/drug effects , T-Lymphocytes, Cytotoxic/immunology , Humans
19.
Nature ; 623(7985): 58-65, 2023 Nov.
Article En | MEDLINE | ID: mdl-37914945

To construct tissue-like prosthetic materials, soft electroactive hydrogels are the best candidate owing to their physiological mechanical modulus, low electrical resistance and bidirectional stimulating and recording capability of electrophysiological signals from biological tissues1,2. Nevertheless, until now, bioelectronic devices for such prostheses have been patch type, which cannot be applied onto rough, narrow or deep tissue surfaces3-5. Here we present an injectable tissue prosthesis with instantaneous bidirectional electrical conduction in the neuromuscular system. The soft and injectable prosthesis is composed of a biocompatible hydrogel with unique phenylborate-mediated multiple crosslinking, such as irreversible yet freely rearrangeable biphenyl bonds and reversible coordinate bonds with conductive gold nanoparticles formed in situ by cross-coupling. Closed-loop robot-assisted rehabilitation by injecting this prosthetic material is successfully demonstrated in the early stage of severe muscle injury in rats, and accelerated tissue repair is achieved in the later stage.


Biocompatible Materials , Hydrogels , Prostheses and Implants , Wounds and Injuries , Animals , Rats , Biocompatible Materials/administration & dosage , Biocompatible Materials/chemistry , Biocompatible Materials/therapeutic use , Electric Conductivity , Gold/chemistry , Hydrogels/administration & dosage , Hydrogels/chemistry , Hydrogels/therapeutic use , Metal Nanoparticles/chemistry , Muscles/injuries , Muscles/innervation , Robotics , Wounds and Injuries/rehabilitation , Wounds and Injuries/surgery
20.
ACS Biomater Sci Eng ; 9(9): 5332-5346, 2023 09 11.
Article En | MEDLINE | ID: mdl-37642176

Periodontitis is an inflammatory disease characterized by tooth loss and alveolar bone resorption. Bacteria are the original cause of periodontitis, and excess reactive oxygen species (ROS) encourage and intensify inflammation. In this study, a mussel-inspired and MnO2 NPs-reinforced adhesive hydrogel capable of alleviating periodontitis with improved antibacterial and antioxidant abilities was developed. The hydrogel was created by combining polyvinyl alcohol (PVA), 3,4-dihydroxy-d-phenylalanine (DOPA), and MnO2 nanoparticles (NPs) (named PDMO hydrogel). The hydrogel was demonstrated to be able to scavenge various free radicals (including total ROS─O2•- and OH•) and relieve the hypoxia in an inflammatory microenvironment by scavenging excess ROS and generating O2 due to its superoxide dismutase (SOD)/catalase (CAT)-like activity. Besides, under 808 nm near-infrared (NIR) light, the photothermal performance of the PDMO hydrogel displayed favorable antibacterial and antibiofilm effects toward Escherichia coli, Staphylococcus aureus, and Porphyromonas gingivalis (up to nearly 100% antibacterial rate). Furthermore, the PDMO hydrogel exhibited favorable therapeutic efficacy in alleviating gingivitis in Sprague-Dawley rats, even comparable to or better than the commercial PERIO. In addition, in the periodontitis models, the PDMO2 group showed the height of the residual alveolar bone and the smallest shadow area of low density among other groups, indicating the positive role of the PDMO2 hydrogel in bone regeneration. Finally, the biosafety of the PDMO hydrogel was comprehensively investigated, and the hydrogel was demonstrated to have good biocompatibility. Therefore, the developed PDMO hydrogel provided an effective solution to resolve biofilm recolonization and oxidative stress in periodontitis and could be a superior candidate for local drug delivery system in the clinical management of periodontitis with great potential for future clinical translation.


Hydrogels , Periodontitis , Periodontitis/drug therapy , Hydrogels/administration & dosage , Hydrogels/chemical synthesis , Hydrogels/pharmacology , Anti-Bacterial Agents/administration & dosage , Anti-Bacterial Agents/pharmacology , Antioxidants/administration & dosage , Antioxidants/pharmacology , Bacteria/drug effects , Animals , Rats , Rats, Sprague-Dawley , Bone Regeneration/drug effects , Biofilms/drug effects , Reactive Oxygen Species/metabolism
...