Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.180
Filter
1.
Gan To Kagaku Ryoho ; 51(6): 671-673, 2024 Jun.
Article in Japanese | MEDLINE | ID: mdl-39009529

ABSTRACT

A hydrogel spacer injection between the prostate and rectum is reported to reduce the risk of rectal toxicity in radiotherapy for prostate cancer. We present a case of an ectopic injection of hydrogel spacer. The patient was a 77-year-old male with intermediate-risk prostate cancer. It was planned that he would receive intensity modulated radiation therapy(IMRT), and a hydrogel spacer was inserted. Three days after insertion, the patient had a fever of 38.6℃ and presented frequent urination and perineal pain. Swelling and heat sensation were observed in the perineum. CRP was 12.00 mg/dL and the white blood cell count was as high as 9,300/µL. T2-weighted images showed a 5.3×1.9 cm high-intensity area around the lower urethra. Ectopic injection of hydrogel spacer and concomitant infection were diagnosed. Upon administering antibiotic treatment, his symptoms and inflammation improved immediately. Four months after hydrogel spacer insertion, T2-weighted images showed a high-intensity area in the lower urethra and around the ischial bone, which was attributed to the remaining hydrogel spacer. The hydrogel spacer and his symptoms completely disappeared at 9 months after hydrogel spacer insertion.


Subject(s)
Hydrogels , Prostatic Neoplasms , Radiotherapy, Intensity-Modulated , Humans , Male , Aged , Prostatic Neoplasms/radiotherapy , Radiotherapy, Intensity-Modulated/methods , Hydrogels/administration & dosage , Injections
2.
Biomater Sci ; 12(13): 3458-3470, 2024 Jun 25.
Article in English | MEDLINE | ID: mdl-38836321

ABSTRACT

Current treatment strategies for infection of chronic wounds often result in compromised healing and necrosis due to antibiotic toxicity, and underlying biomarkers affected by treatments are not fully known. Here, a multifunctional dressing was developed leveraging the unique wound-healing properties of chitosan, a natural polysaccharide known for its numerous benefits in wound care. The dressing consists of an oxygenating perfluorocarbon functionalized methacrylic chitosan (MACF) hydrogel incorporated with antibacterial polyhexamethylene biguanide (PHMB). A non-healing diabetic infected wound model with emerging metabolomics tools was used to explore the anti-infective and wound healing properties of the resultant multifunctional dressing. Direct bacterial bioburden assessment demonstrated superior antibacterial properties of hydrogels over a commercial dressing. However, wound tissue quality analyses confirmed that sustained PHMB for 21 days resulted in tissue necrosis and disturbed healing. Therefore, a follow-up comparative study investigated the best treatment course for antiseptic application ranging from 7 to 21 days, followed by the oxygenating chitosan-based MACF treatment for the remainder of the 21 days. Bacterial counts, tissue assessments, and lipidomics studies showed that 14 days of application of MACF-PHMB dressings followed by 7 days of MACF dressings provides a promising treatment for managing infected non-healing diabetic skin ulcers.


Subject(s)
Anti-Bacterial Agents , Bandages , Chitosan , Hydrogels , Wound Healing , Chitosan/chemistry , Chitosan/pharmacology , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/administration & dosage , Hydrogels/chemistry , Hydrogels/pharmacology , Hydrogels/administration & dosage , Wound Healing/drug effects , Animals , Biguanides/chemistry , Biguanides/pharmacology , Biguanides/administration & dosage , Wound Infection/drug therapy , Wound Infection/microbiology , Male , Oxygen/chemistry , Chronic Disease , Fluorocarbons/chemistry , Fluorocarbons/pharmacology , Fluorocarbons/administration & dosage
3.
J Control Release ; 371: 570-587, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38852624

ABSTRACT

Myocardial infarction (MI)-induced impaired cardiomyocyte (CM) mitochondrial function and microenvironmental inflammatory cascades severely accelerate the progression of heart failure for compromised myocardial repair. Modulation of the crosstalk between CM mitochondrial DNA (mtDNA) and STING has been recently identified as a robust strategy in enhancing MI treatment, but remains seldom explored. To develop a novel approach that can address persistent myocardial injury using this crosstalk, we report herein construction of a biomimetic hydrogel system, Rb1/PDA-hydrogel comprised of ginsenoside Rb1/polydopamine nanoparticles (Rb1/PDA NPs)-loaded carboxylated chitosan, 4-arm-PEG-phenylboronic acid (4-arm-PEG-PBA), and 4-arm-PEG-dopamine (4-arm-PEG-DA) crosslinked networks. An optimized hydrogel formulation presents not only desired adhesion properties to the surface of the myocardium, but also adaptability for deep myocardial injection, resulting in ROS scavenging, CM mitochondrial function protection, M1 macrophage polarization inhibition through the STING pathway, and angiogenesis promotion via an internal-external spatial combination. The enhanced therapeutic efficiency is supported by the histological analysis of the infarcted area, which shows that the fibrotic area of the MI rats decreases from 58.4% to 5.5%, the thickness of the left ventricular wall increases by 1-fold, and almost complete recovery of cardiac function after 28 days of treatment. Overall, this study reported the first use of a strong adhesive and injectable hydrogel with mtDNA and STING signaling characteristics for enhanced MI treatment via an internal-external spatial combination strategy.


Subject(s)
DNA, Mitochondrial , Hydrogels , Myocardial Infarction , Myocytes, Cardiac , Polymers , Animals , Myocardial Infarction/drug therapy , Myocardial Infarction/metabolism , Hydrogels/administration & dosage , Hydrogels/chemistry , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Male , Polymers/chemistry , Polymers/administration & dosage , Indoles/administration & dosage , Indoles/chemistry , Nanoparticles/administration & dosage , Nanoparticles/chemistry , Chitosan/chemistry , Chitosan/administration & dosage , Rats, Sprague-Dawley , Polyethylene Glycols/chemistry , Polyethylene Glycols/administration & dosage , Rats , Membrane Proteins , Reactive Oxygen Species/metabolism , Boronic Acids , Ginsenosides
4.
Int J Pharm ; 660: 124348, 2024 Jul 20.
Article in English | MEDLINE | ID: mdl-38885776

ABSTRACT

Skin and soft tissue infections (SSTIs) represent a significant healthcare challenge, particularly in the context of increasing antibiotic resistance. This study investigates the efficacy of a novel therapeutic approach combining bacteriophage (phage) therapy with a gum Karaya (GK)-based hydrogel delivery system in a porcine model of deep staphylococcal SSTIs. The study exploits the lytic activity and safety of the Staphylococcus phage 812K1/420 of the Kayvirus genus, which is active against methicillin-resistant Staphylococcus aureus (MRSA). The GK injectable hydrogels and hydrogel films, developed by our research group, serve as effective, non-toxic, and easy-to-apply delivery systems, supporting moist wound healing and re-epithelialization. In the porcine model, the combined treatment showed asynergistic effect, leading to a significant reduction in bacterial load (2.5 log CFU/gram of tissue) within one week. Local signs of inflammation were significantly reduced by day 8, with clear evidence of re-epithelialization and wound contraction. Importantly, no adverse effects of the GK-based delivery system were observed throughout the study. The results highlight the potential of this innovative therapeutic approach to effectively treat deep staphylococcal SSTIs, providing a promising avenue for further research and clinical application in the field of infections caused by antibiotic-resistant bacteria.


Subject(s)
Disease Models, Animal , Hydrogels , Methicillin-Resistant Staphylococcus aureus , Phage Therapy , Staphylococcal Infections , Wound Infection , Animals , Methicillin-Resistant Staphylococcus aureus/drug effects , Hydrogels/administration & dosage , Hydrogels/chemistry , Phage Therapy/methods , Swine , Staphylococcal Infections/therapy , Staphylococcal Infections/drug therapy , Wound Infection/therapy , Wound Infection/microbiology , Wound Infection/drug therapy , Wound Healing/drug effects , Staphylococcus Phages , Female , Plant Gums/chemistry
5.
J Neural Eng ; 21(4)2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38885674

ABSTRACT

Objective.To develop a clinically relevant injectable hydrogel derived from decellularized porcine peripheral nerves and with mechanical properties comparable to native central nervous system (CNS) tissue to be used as a delivery vehicle for Schwann cell transplantation to treat spinal cord injury (SCI).Approach.Porcine peripheral nerves (sciatic and peroneal) were decellularized by chemical decellularization using a sodium deoxycholate and DNase (SDD) method previously developed by our group. The decellularized nerves were delipidated using dichloromethane and ethanol solvent and then digested using pepsin enzyme to form injectable hydrogel formulations. Genipin was used as a crosslinker to enhance mechanical properties. The injectability, mechanical properties, and gelation kinetics of the hydrogels were further analyzed using rheology. Schwann cells encapsulated within the injectable hydrogel formulations were passed through a 25-gauge needle and cell viability was assessed using live/dead staining. The ability of the hydrogel to maintain Schwann cell viability against an inflammatory milieu was assessedin vitrousing inflamed astrocytes co-cultured with Schwann cells.Mainresults. The SDD method effectively removes cells and retains extracellular matrix in decellularized tissues. Using rheological studies, we found that delipidation of decellularized porcine peripheral nerves using dichloromethane and ethanol solvent improves gelation kinetics and mechanical strength of hydrogels. The delipidated and decellularized hydrogels crosslinked using genipin mimicked the mechanical strength of CNS tissue. The hydrogels were found to have shear thinning properties desirable for injectable formulations and they also maintained higher Schwann cell viability during injection compared to saline controls. Usingin vitroco-culture experiments, we found that the genipin-crosslinked hydrogels also protected Schwann cells from astrocyte-mediated inflammation.Significance. Injectable hydrogels developed using delipidated and decellularized porcine peripheral nerves are a potential clinically relevant solution to deliver Schwann cells, and possibly other therapeutic cells, at the SCI site by maintaining higher cellular viability and increasing therapeutic efficacy for SCI treatment.


Subject(s)
Hydrogels , Peripheral Nerves , Schwann Cells , Spinal Cord Injuries , Animals , Schwann Cells/physiology , Schwann Cells/drug effects , Hydrogels/chemistry , Hydrogels/administration & dosage , Swine , Spinal Cord Injuries/therapy , Peripheral Nerves/physiology , Peripheral Nerves/drug effects , Spinal Cord Regeneration/physiology , Spinal Cord Regeneration/drug effects , Cells, Cultured , Cell Survival/physiology , Cell Survival/drug effects
6.
J Control Release ; 371: 406-428, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38849093

ABSTRACT

Preventing the recurrence of melanoma after surgery and accelerating wound healing are among the most challenging aspects of melanoma management. Photothermal therapy has been widely used to treat tumors and bacterial infections and promote wound healing. Owing to its efficacy and specificity, it may be used for postoperative management of tumors. However, its use is limited by the uncontrollable distribution of photosensitizers and the likelihood of damage to the surrounding normal tissue. Hydrogels provide a moist environment with strong biocompatibility and adhesion for wound healing owing to their highly hydrophilic three-dimensional network structure. In addition, these materials serve as excellent drug carriers for tumor treatment and wound healing. It is possible to combine the advantages of both of these agents through different loading modalities to provide a powerful platform for the prevention of tumor recurrence and wound healing. This review summarizes the design strategies, research progress and mechanism of action of hydrogels used in photothermal therapy and discusses their role in preventing tumor recurrence and accelerating wound healing. These findings provide valuable insights into the postoperative management of melanoma and may guide the development of promising multifunctional hydrogels for photothermal therapy.


Subject(s)
Hydrogels , Melanoma , Photothermal Therapy , Wound Healing , Hydrogels/chemistry , Hydrogels/administration & dosage , Humans , Melanoma/therapy , Photothermal Therapy/methods , Animals , Wound Healing/drug effects , Photosensitizing Agents/administration & dosage , Photosensitizing Agents/therapeutic use , Drug Carriers/chemistry , Neoplasm Recurrence, Local/prevention & control
7.
Int J Pharm ; 659: 124281, 2024 Jun 25.
Article in English | MEDLINE | ID: mdl-38802026

ABSTRACT

Hydrogel drug delivery systems (DDSs) for treating ulcerative colitis (UC) have garnered attention. However, there is a lack of meta-analysis summarizing their effectiveness. Therefore, this study aimed to conduct a meta-analysis of pre-clinical evidence comparing hydrogel DDSs with free drug administration. Subgroup analyses were performed based on hydrogel materials (polysaccharide versus non-polysaccharide) and administration routes of the hydrogel DDSs (rectal versus oral). The outcome indicators included colon length, histological scores, tumor necrosis factor-α (TNF-α), zonula occludens protein 1(ZO-1), and area under the curve (AUC). The results confirmed the therapeutic enhancement of the hydrogel DDSs for UC compared with the free drug group. Notably, no significant differences were found between polysaccharide and non-polysaccharide materials, however, oral administration was found superior regarding TNF-α and AUC. In conclusion, oral hydrogel DDSs can serve as potential excellent dosage forms in oral colon -targeting DDSs, and in the design of colon hydrogel delivery systems, polysaccharides do not show advantages compared with other materials.


Subject(s)
Colitis, Ulcerative , Drug Delivery Systems , Hydrogels , Colitis, Ulcerative/drug therapy , Hydrogels/chemistry , Hydrogels/administration & dosage , Drug Delivery Systems/methods , Animals , Tumor Necrosis Factor-alpha , Humans , Administration, Oral , Colon/metabolism , Colon/drug effects , Polysaccharides/chemistry , Polysaccharides/administration & dosage , Administration, Rectal , Area Under Curve
8.
Int J Pharm ; 659: 124263, 2024 Jun 25.
Article in English | MEDLINE | ID: mdl-38815639

ABSTRACT

Periodontitis is a multifactorial inflammatory disease characterized by severe alveolar bone damage and attachment loss. The imbalance of T help 17 (Th17) / regulatory T cells (Treg) induces excessive interleukin (IL)-17, which leads to alveolar bone damage and aggravates the development of periodontitis. Therefore, we proposed a therapeutic strategy to restore Th17/Treg homeostasis by interfering reactive oxygen species (ROS)-macrophage polarization cascade using active targeting microemulsions-based thermosensitive hydrogel. Folic acid-modified quercetin-loaded microemulsions (FA-Qu-MEs) were dispersed in poloxamer 407 and poly(N-isopropylacrylamide) matrix of hydrogel (FA-Qu-MEs@Gel). FA-Qu-MEs@Gel could be locally injected into the periodontal pocket and sustainedly release drugs. FA-Qu-MEs exhibited excellent ROS scavenging potency by targeting macrophages, resulting M1 phenotype macrophage from to M2 phenotype macrophage. Subsequently, the phenotypic changes of macrophages lead to decreased expression of IL-6 and tumor necrosis factor-α, which inhibited activated Th17, while IL-10 secreted by M2 macrophages promoted Treg differentiation. Finally, the restored Th17/Treg homeostasis reduced the level of IL-17 to accelerate alveolar bone regeneration. This study deigns a novel system that promote alveolar bone regeneration by remodeling Th17/Treg homeostasis via regulating ROS-macrophages polarization cascade for periodontitis treatment.


Subject(s)
Emulsions , Homeostasis , Hydrogels , Macrophages , Periodontitis , Reactive Oxygen Species , T-Lymphocytes, Regulatory , Th17 Cells , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , Reactive Oxygen Species/metabolism , Periodontitis/drug therapy , Periodontitis/immunology , Animals , Th17 Cells/drug effects , Th17 Cells/immunology , Hydrogels/administration & dosage , Homeostasis/drug effects , Macrophages/drug effects , Macrophages/immunology , Mice , Male , Poloxamer/chemistry , RAW 264.7 Cells , Acrylic Resins/chemistry , Bone Regeneration/drug effects , Mice, Inbred C57BL
9.
Biomater Sci ; 12(13): 3389-3400, 2024 Jun 25.
Article in English | MEDLINE | ID: mdl-38804911

ABSTRACT

Rheumatoid arthritis (RA) is a chronic autoimmune inflammatory joint disorder affecting nearly 1% of the global population. In RA, synovial joints are infiltrated by inflammatory mediators and enzymes, leading to articular cartilage deterioration, joint damage, and bone erosion. Herein, the 9-aminoacridine-6-O-stearoyl-L-ascorbic acid hydrogel (9AA-SAA hydrogel) was formulated by the heat-cool method and further characterized for surface charge, surface morphology, rheology, and cytocompatibility. Furthermore, we evaluated the therapeutic efficacy of the 9AA-SAA hydrogel, an enzyme-responsive drug delivery system with on-and-off switching capabilities based on disease severity against collagen-induced experimental arthritis in Wistar rats. The anti-inflammatory action of the US FDA-approved drug 9-aminoacridine (9AA) was revealed which acted through nuclear receptor subfamily 4 group A member 1 (NR4A1), an anti-inflammatory orphan nuclear receptor that inhibits nuclear factor kappa-light-chain-enhancer of activated B cells (NF-kB). Furthermore, we have explored the role of ascorbic acid, an active moiety of 6-O-stearoyl-L-ascorbic acid (SAA), in promoting the production of collagen production through ten-eleven translocation-2 (TET2) upregulation. Targeting through NR4A1 and TET2 could be the probable mechanism for the treatment of experimental arthritis. The combination of 9AA and ascorbic acid demonstrated enhanced therapeutic efficacy in the 9AA-SAA hydrogel, significantly reducing the severity of experimental arthritis. This approach, in contrast to existing treatments with limited effectiveness, presents a promising and more effective strategy for RA treatment by mitigating inflammation in experimental arthritis.


Subject(s)
Arthritis, Experimental , Ascorbic Acid , Hydrogels , Rats, Wistar , Animals , Ascorbic Acid/pharmacology , Ascorbic Acid/chemistry , Ascorbic Acid/administration & dosage , Hydrogels/chemistry , Hydrogels/administration & dosage , Hydrogels/pharmacology , Rats , Arthritis, Experimental/drug therapy , Arthritis, Experimental/pathology , Male , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/administration & dosage , Mice , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Biocompatible Materials/administration & dosage , Injections
10.
Biomater Sci ; 12(12): 3100-3111, 2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38712522

ABSTRACT

In this study, we developed a ROS-responsive thermosensitive poly(ethylene glycol)-polypeptide hydrogel loaded with a chemotherapeutic drug, doxorubicin (Dox), an antiviral imidazoquinoline, resiquimod (R848), and antibody targeting programmed cell death protein 1 (aPD-1) for local chemoimmunotherapy. The hydrogel demonstrated controllable degradation and sustained drug release behavior according to the concentration of ROS in vitro. Following intratumoral injection into mice bearing B16F10 melanoma, the Dox/R848/aPD-1 co-loaded hydrogel effectively inhibited tumor growth, prolonged animal survival time and promoted anti-tumor immune responses with low systemic toxicity. In the postoperative model, the Dox/R848/aPD-1 co-loaded hydrogel exhibited enhanced tumor recurrence prevention and long-term immune memory effects. Thus, the hydrogel-based local chemoimmunotherapy system demonstrates potential for effective anti-tumor treatment and suppression of tumor recurrence.


Subject(s)
Doxorubicin , Hydrogels , Immunotherapy , Peptides , Reactive Oxygen Species , Animals , Hydrogels/chemistry , Hydrogels/administration & dosage , Doxorubicin/chemistry , Doxorubicin/administration & dosage , Doxorubicin/pharmacology , Peptides/chemistry , Peptides/administration & dosage , Peptides/pharmacology , Mice , Reactive Oxygen Species/metabolism , Melanoma, Experimental/drug therapy , Melanoma, Experimental/pathology , Melanoma, Experimental/therapy , Melanoma, Experimental/immunology , Mice, Inbred C57BL , Polyethylene Glycols/chemistry , Cell Line, Tumor , Temperature , Antineoplastic Agents/chemistry , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Drug Delivery Systems , Drug Liberation , Drug Carriers/chemistry
11.
J Control Release ; 370: 583-599, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38729435

ABSTRACT

Stem cells are recognized as an important target and tool in regenerative engineering. In this study, we explored the feasibility of engineering amniotic fluid-derived mesenchymal stem cell-secreted molecules (afMSC-SMs) as a versatile bioactive material for skin regenerative medicine applications in a time- and cost-efficient and straightforward manner. afMSC-SMs, obtained in powder form through ethanol precipitation, effectively contributed to preserving the self-renewal capacity and differentiation potential of primary human keratinocytes (pKCs) in a xeno-free environment, offering a potential alternative to traditional culture methods for their long-term in vitro expansion, and allowed them to reconstitute a fully stratified epithelium sheet on human dermal fibroblasts. Furthermore, we demonstrated the flexibility of afMSC-SMs in wound healing and hair regrowth through injectable hydrogel and nanogel-mediated transdermal delivery systems, respectively, expanding the pool of regenerative applications. This cell-free approach may offer several potential advantages, including streamlined manufacturing processes, scalability, controlled formulation, longer shelf lives, and mitigation of risks associated with living cell transplantation. Accordingly, afMSC-SMs could serve as a promising therapeutic toolbox for advancing cell-free regenerative medicine, simplifying their broad applicability in various clinical settings.


Subject(s)
Keratinocytes , Mesenchymal Stem Cells , Regenerative Medicine , Skin , Humans , Regenerative Medicine/methods , Keratinocytes/cytology , Animals , Mesenchymal Stem Cells/cytology , Skin/metabolism , Cells, Cultured , Amniotic Fluid/cytology , Wound Healing/drug effects , Cell Differentiation , Fibroblasts/metabolism , Fibroblasts/cytology , Tissue Engineering/methods , Hydrogels/chemistry , Hydrogels/administration & dosage
12.
J Control Release ; 370: 543-555, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38729434

ABSTRACT

Hyaluronic acid (HA)-based biopolymer hydrogels are promising therapeutic dressings for various wounds but still underperform in treating diabetic wounds. These wounds are extremely difficult to heal and undergo a prolonged and severe inflammatory process due to bacterial infection, overexpression of reactive oxygen species (ROS), and insufficient synthesis of NO. In this study, a dynamic crosslinked hyaluronic acid (HA) hydrogel dressing (Gel-HAB) loaded with allomelanin (AMNP)-N, N'-dis-sec-butyl-N, N'-dinitroso-1, 4-phenylenediamine (BNN6) nanoparticles (AMNP-BNN6) was developed for healing diabetic wounds. The dynamic acylhydrazone bond formed between hydrazide-modified HA (HA-ADH) and oxidized HA (OHA) makes the hydrogel injectable, self-healing, and biocompatible. The hydrogel, loaded with AMNP-BNN6 nanoparticles, exhibits promising ROS scavenging ability and on-demand release of nitric oxide (NO) under near-infrared (NIR) laser irradiation to achieve mild photothermal antibacterial therapy (PTAT) (∼ 48 °C). Notably, the Gel-HAB hydrogel effectively reduced the oxidative stress level, controlled infections, accelerated vascular regeneration, and promoted angiogenesis, thereby achieving rapid healing of diabetic wounds. The injectable self-healing nanocomposite hydrogel could serve as a mild photothermal-enhanced antibacterial, antioxidant, and nitric oxide release platform for the treatment of diabetic wounds.


Subject(s)
Anti-Bacterial Agents , Antioxidants , Hyaluronic Acid , Hydrogels , Nanoparticles , Nitric Oxide , Wound Healing , Hyaluronic Acid/chemistry , Wound Healing/drug effects , Anti-Bacterial Agents/administration & dosage , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Hydrogels/chemistry , Hydrogels/administration & dosage , Nitric Oxide/administration & dosage , Animals , Antioxidants/pharmacology , Antioxidants/administration & dosage , Antioxidants/chemistry , Nanoparticles/chemistry , Mice , Diabetes Mellitus, Experimental , Reactive Oxygen Species/metabolism , Photothermal Therapy/methods , Male , Bandages , Humans
13.
J Neural Eng ; 21(3)2024 Jun 27.
Article in English | MEDLINE | ID: mdl-38806019

ABSTRACT

Objective.Severe traumatic brain injury (sTBI) induced neuronal loss and brain atrophy contribute significantly to long-term disabilities. Brain extracellular matrix (ECM) associated chondroitin sulfate (CS) glycosaminoglycans promote neural stem cell (NSC) maintenance, and CS hydrogel implants have demonstrated the ability to enhance neuroprotection, in preclinical sTBI studies. However, the ability of neuritogenic chimeric peptide (CP) functionalized CS hydrogels in promoting functional recovery, after controlled cortical impact (CCI) and suction ablation (SA) induced sTBI, has not been previously demonstrated. We hypothesized that neuritogenic (CS)CP hydrogels will promote neuritogenesis of human NSCs, and accelerate brain tissue repair and functional recovery in sTBI rats.Approach.We synthesized chondroitin 4-Osulfate (CS-A)CP, and 4,6-O-sulfate (CS-E)CP hydrogels, using strain promoted azide-alkyne cycloaddition (SPAAC), to promote cell adhesion and neuritogenesis of human NSCs,in vitro; and assessed the ability of (CS-A)CP hydrogels in promoting tissue and functional repair, in a novel CCI-SA sTBI model,in vivo. Main results.Results indicated that (CS-E)CP hydrogels significantly enhanced human NSC aggregation and migration via focal adhesion kinase complexes, when compared to NSCs in (CS-A)CP hydrogels,in vitro. In contrast, NSCs encapsulated in (CS-A)CP hydrogels differentiated into neurons bearing longer neurites and showed greater spontaneous activity, when compared to those in (CS-E)CP hydrogels. The intracavitary implantation of (CS-A)CP hydrogels, acutely after CCI-SA-sTBI, prevented neuronal and axonal loss, as determined by immunohistochemical analyses. (CS-A)CP hydrogel implanted animals also demonstrated the significantly accelerated recovery of 'reach-to-grasp' function when compared to sTBI controls, over a period of 5-weeks.Significance.These findings demonstrate the neuritogenic and neuroprotective attributes of (CS)CP 'click' hydrogels, and open new avenues for the development of multifunctional glycomaterials that are functionalized with biorthogonal handles for sTBI repair.


Subject(s)
Brain Injuries, Traumatic , Hydrogels , Neural Stem Cells , Neurites , Rats, Sprague-Dawley , Recovery of Function , Hydrogels/administration & dosage , Animals , Rats , Recovery of Function/drug effects , Recovery of Function/physiology , Humans , Neural Stem Cells/drug effects , Neurites/drug effects , Neurites/physiology , Male , Chondroitin Sulfates/administration & dosage , Chondroitin Sulfates/pharmacology , Glycosaminoglycans/administration & dosage , Cells, Cultured , Neurogenesis/drug effects , Neurogenesis/physiology
14.
J Control Release ; 371: 111-125, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38782064

ABSTRACT

In esophageal cancer (EC), clinical specimen testing has uncovered a significant increase in BTB and CNC homolog 1 (BACH1) expression and a shift towards an immunosuppressive environment, alongside a notable decrease in p53 protein expression. Therefore, therapeutic strategies focusing on BACH1 inhibition and p53 upregulation appear promising. Traditional oral treatments for EC lack precision and efficacy. Here, we propose a novel approach employing tumor-targeted nanoparticles (NPs) for drug delivery. However, the formation of a drug reservoir at the esophageal site, crucial for the sustained release of therapeutics, presents significant challenges in nano-delivery systems for EC treatment. To address this, we developed a thermosensitive hydrogel composed of F127 and tannic acid, serving as a vehicle for NP loading. These NPs, synthesized through the emulsion/volatization methods of mPEG-PLGA-PLL-cRGD, facilitate in situ drug delivery. Upon contacting esophageal tissue, the hydrogel transitions to a gel, adhering to the lining and enabling sustained release of encapsulated therapeutics. The formulation encompasses NPs laden with small interfering RNA targeting BACH1 (siBACH1) and the p53 activator PRIMA-1, creating a cohesive gel-nano system. Preliminary biological assessments demonstrate that this injectable, thermosensitive gel-nano system adheres effectively to esophageal tissue and targets EC cells. For better modeling clinical outcomes, a patient-derived organoid xenograft (PDOX) model was innovated, involving transplantation of EC-derived organoids into humanized mice, reconstructed with peripheral blood mononuclear cells (PBMCs). Post-treatment analysis showed substantial EC growth inhibition (89.51% tumor inhibition rate), significant BACH1 level reduction, restored anti-tumor immune responses, and pronounced tumor apoptosis. In summary, our study introduces a thermosensitive gel-nano system for EC treatment via restoring p53 activity and boosting T-cell immunity, with potential for clinical application.


Subject(s)
Esophageal Neoplasms , Nanoparticles , Tumor Suppressor Protein p53 , Esophageal Neoplasms/drug therapy , Esophageal Neoplasms/immunology , Animals , Humans , Nanoparticles/administration & dosage , Nanoparticles/chemistry , Cell Line, Tumor , Hydrogels/administration & dosage , Hydrogels/chemistry , T-Lymphocytes/immunology , T-Lymphocytes/drug effects , Female , Mice , Temperature , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/therapeutic use , Drug Delivery Systems
15.
J Control Release ; 371: 258-272, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38815704

ABSTRACT

Currently, exosomes showed appropriate potential in the repair of skin injury. However, the functions of the exosomes could be compromised rapidly due to their short half-life and high clearance rate in vivo. In addition, the controlled release of effective concentrations of exosomes could increase the utilization efficiency of exosomes in wound healing. Accordingly, the design of an effective system for the controlled delivery of exosomes during the wound treatment period was necessary. In this contribution, we designed a novel exosome-based multifunctional nanocomposite platform with photothermal-controlled release performance for the repair of skin injury. Based on the agarose hydrogel, two-dimensional Ti3C2 (Ti3C2 MXene) and human umbilical cord mesenchymal stem cell (hucMSC)-derived exosomes, the as-prepared platform (i.e., hucMSC-derived exosome/Ti3C2 MXene hydrogel) was synthesized for the first time. Apart from possessing injectability, the hucMSC-derived exosome/Ti3C2 MXene hydrogel utilized the excellent photothermal effect of Ti3C2 MXene and proper phase transition performance of agarose hydrogel to provide a photothermal-controlled release system for the hucMSC-derived exosomes, which was beneficial for the personalized on-demand drug delivery. Importantly, the hucMSC-derived exosomes maintained their inherent structure and activity after being released from the Ti3C2 MXene hydrogel. Additionally, the as-prepared hydrogel with multifunctional performance also presented remarkable biocompatibility and photothermal-antibacterial property, and could efficiently accelerate wound healing by promoting cell proliferation, angiogenesis, collagen deposition, and reducing the level of inflammation at the wound site. The results suggested that the exosome-based multifunctional nanocomposite platform with great potential for wound healing would make significant advances in the revolution of traditional treatment methods in skin injury.


Subject(s)
Delayed-Action Preparations , Exosomes , Hydrogels , Mesenchymal Stem Cells , Nanocomposites , Skin , Wound Healing , Humans , Wound Healing/drug effects , Animals , Nanocomposites/administration & dosage , Nanocomposites/chemistry , Hydrogels/administration & dosage , Hydrogels/chemistry , Skin/injuries , Skin/metabolism , Titanium/chemistry , Mice , Male , Anti-Bacterial Agents/administration & dosage , Drug Delivery Systems
16.
J Control Release ; 370: 747-762, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38740094

ABSTRACT

Rheumatoid arthritis (RA) is a progressive autoimmune disease and drug therapy has been restricted due to poor therapeutic efficacy and adverse effects. In RA synovium, dendritic cells present self-antigens to activate cascade immune pathway. Furthermore, downstream macrophages secrete high levels of pro-inflammatory cytokines; Hyperplasia of activated synovial fibroblasts (FLS) is responsible for hypoxic synovium microenvironment, secretion of cytokines/chemokines and erosion of bone/cartilage tissues. Positive feedback loop of inflammation between macrophages and FLS independent of antigen-presentation is constructed. Herein, an injectable pH-sensitive peptide hydrogel encapsulating siRNA/Methotrexate-polyethyleneimine (siMP, including sip65MP, sip38MP, siCD86MP) and Bismuthene nanosheet/Methotrexate-polyethyleneimine (BiMP) is successfully developed. Among them, siCD86MP reduces protein level of co-stimulatory molecule CD86 while sip65MP and sip38MP separately inhibit NF-κB and MAPK-p38 pathways of macrophages and FLS to suppress secretion of cytokines and MMPs. Meanwhile, reduction in anti-apoptotic property of FLS induced by inhibition of NF-κB pathway has a synergistic effect with photodynamic therapy (PDT) and photothermal therapy (PTT) mediated by BiMP for FLS elimination, effectively ameliorating hypoxic synovium microenvironment. After being injected into synovium, hydrogel responds to acidic microenvironment and serves as a reservoir for sustained drug release and inherent retention capacity of which enables cationic nanoparticles to bypass tissue barrier for precise synovium targeting. This brand-new drug delivery system combines modulating cascade immune pathway from beginning to end by RNAi and eliminating FLS for improving synovium microenvironment by phototherapy together, providing a robust strategy for clinical RA treatment.


Subject(s)
Arthritis, Rheumatoid , Fibroblasts , Hydrogels , Methotrexate , Synovial Membrane , Fibroblasts/drug effects , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/therapy , Hydrogels/administration & dosage , Synovial Membrane/immunology , Animals , Methotrexate/administration & dosage , Methotrexate/pharmacology , RNA, Small Interfering/administration & dosage , Photochemotherapy/methods , Mice , Humans , Macrophages/drug effects , Macrophages/immunology , RAW 264.7 Cells , Cytokines/metabolism , Antirheumatic Agents/administration & dosage , Cellular Microenvironment/drug effects , NF-kappa B/metabolism , Phototherapy/methods , Peptides/administration & dosage
17.
J Control Release ; 370: 643-652, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38744344

ABSTRACT

Neonatal hypoglycemia is a common disease in newborns, which can precipitate energy shortage and follow by irreversible brain and neurological injury. Herein, we present a novel approach for treating neonatal hypoglycemia involving an adhesive polyvinylpyrrolidone/gallic acid (PVP/GA) film loading glucose. The PVP/GA film with loose cross-linking can be obtained by mixing their ethanol solution and drying complex. When depositing this soft film onto wet tissue, it can absorb interfacial water to form a hydrogel with a rough surface, which facilitates tight contact between the hydrogel and tissue. Meanwhile, the functional groups in the hydrogels and tissues establish both covalent and non-covalent bonds, leading to robust bioadhesion. Moreover, the adhered PVP/GA hydrogel can be detached without damaging tissue as needed. Furthermore, the PVP/GA films exhibit excellent antibacterial properties and biocompatibility. Notably, these films effectively load glucose and deliver it to the sublingual tissue of newborn rabbits, showcasing a compelling therapeutic effect against neonatal hypoglycemia. The strengths of the PVP/GA film encompass excellent wet adhesion in the wet and highly dynamic environment of the oral cavity, on-demand detachment, antibacterial efficacy, biocompatibility, and straightforward preparation. Consequently, this innovative film holds promise for diverse biomedical applications, including but not limited to wearable devices, sealants, and drug delivery systems.


Subject(s)
Animals, Newborn , Glucose , Hypoglycemia , Povidone , Animals , Rabbits , Glucose/administration & dosage , Glucose/chemistry , Povidone/chemistry , Infant, Newborn , Humans , Hydrogels/administration & dosage , Hydrogels/chemistry , Adhesives/administration & dosage , Adhesives/chemistry , Anti-Bacterial Agents/administration & dosage , Drug Delivery Systems
18.
Int J Nanomedicine ; 19: 4045-4060, 2024.
Article in English | MEDLINE | ID: mdl-38736656

ABSTRACT

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.


Subject(s)
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
19.
Int J Nanomedicine ; 19: 4081-4101, 2024.
Article in English | MEDLINE | ID: mdl-38736654

ABSTRACT

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.


Subject(s)
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
20.
J Orthop Surg Res ; 19(1): 274, 2024 May 02.
Article in English | MEDLINE | ID: mdl-38698396

ABSTRACT

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.


Subject(s)
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
SELECTION OF CITATIONS
SEARCH DETAIL
...