Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
Add more filters











Publication year range
1.
J Control Release ; 374: 154-170, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39127448

ABSTRACT

To realize high-quality vascularized bone regeneration, we developed a multifunctional hydrogel (SHPP-ZB) by incorporating BMP-2@ZIF-8/PEG-NH2 nanoparticles (NPs) into a sodium alginate/hydroxyapatite/polyvinyl alcohol hydrogel loaded with PDGF-BB, allowing for the sequential release of angiogenic and osteogenic growth factors (GFs) during bone repair. ZIF-8 served as a protective host for BMP-2 from degradation, ensuring high encapsulation efficiency and long-term bioactivity. The SHPP-ZB hydrogel exhibited enhanced mechanical strength and injectability, making it suitable for complex bone defects. It provided a swelling interface for tissue interlocking and the early release of Zn2+ and tannin acid (TA) to exert antioxidant and antibacterial effects, followed by the sequential release of angiogenic and osteogenic GFs to promote high-quality vascularized bone regeneration. In vitro experiments demonstrated the superior angiogenic and osteogenic properties of SHPP-ZB compared to other groups. In vivo experiments indicated that the sequential delivery of GFs via SHPP-ZB hydrogel could improve vascularized bone regeneration. Further, RNA sequencing analysis of regenerative bone tissue revealed that SHPP-ZB hydrogel promoted vascularized bone regeneration by regulating JUN, MAPK, Wnt, and calcium signaling pathways in vivo. This study presented a promising approach for efficient vascularized bone regeneration in large-scale bone defects.


Subject(s)
Alginates , Becaplermin , Bone Morphogenetic Protein 2 , Bone Regeneration , Hydrogels , Osteogenesis , Bone Regeneration/drug effects , Animals , Hydrogels/chemistry , Hydrogels/administration & dosage , Osteogenesis/drug effects , Bone Morphogenetic Protein 2/administration & dosage , Alginates/chemistry , Becaplermin/administration & dosage , Nanoparticles/chemistry , Durapatite/chemistry , Durapatite/administration & dosage , Angiogenesis Inducing Agents/administration & dosage , Angiogenesis Inducing Agents/pharmacology , Angiogenesis Inducing Agents/chemistry , Male , Polyvinyl Alcohol/chemistry , Polyethylene Glycols/chemistry , Tannins/chemistry , Tannins/administration & dosage , Tannins/pharmacology , Neovascularization, Physiologic/drug effects , Humans , Rats, Sprague-Dawley , Mice
2.
Int J Biol Macromol ; 272(Pt 1): 132763, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38821311

ABSTRACT

In this paper, a heart-shaped nanocomposite (MXenen@Cu-MOF, MC) has been prepared by hydrothermal method. This material can effectively prevent the accumulation of MXene, improve the material's electrical conductivity and antibacterial properties. In addition, it is loaded into Polyvinyl alcohol/Poly-dopamine hydrogel wound dressings (PPMC), which can effectively destroy bacterial biofilms and provide a new pathway for internal electrical currents, helping to repair internal electric fields and promote wound healing. Through the concentration gradient experiments of hydrogel such as antibacterial, conductive, hemolysis and cell migration, we believe that the addition of MC can improve the basic properties of hydrogel. Among them, PPMC0.2 is the hydrogel with the best performance under the premise of meeting bio-compatibility. Its resistance, between 500 and 1000 Ω, is lower than the skin resistance at the wound site and provides the basis for the passage of current through the body. In addition, the cell mobility (24h) of PPMC0.2 reached 58%, and the wound healing rate (6day) was 81.84%, which was much higher than that of other experimental groups. The experimental results proved that PPMC hydrogel can promote wound healing, and this study also provided a new therapeutic idea for chronic wound healing.


Subject(s)
Hydrogels , Nanocomposites , Polyvinyl Alcohol , Wound Healing , Wound Healing/drug effects , Polyvinyl Alcohol/chemistry , Nanocomposites/chemistry , Hydrogels/chemistry , Hydrogels/pharmacology , Animals , Electric Conductivity , Indoles/chemistry , Indoles/pharmacology , Polymers/chemistry , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Humans , Cell Movement/drug effects , Mice , Electricity , Hemolysis/drug effects
3.
Int J Biol Macromol ; 264(Pt 2): 130768, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38467228

ABSTRACT

Lanthanide luminescent hydrogels have broad application prospects in various fields. However, most of lanthanide hydrogels possess relatively simple functions, which is not conducive to practical applications. Therefore, it is becoming increasingly urgent to develop multifunctional hydrogels. Herein, a multifunctional chitosan-based lanthanide luminescent hydrogel with ultra-stretchability, multi-adhesion, excellent self-healing, emission color tunability, and good antibacterial ability was prepared by a simple one-step free radical polymerization. In this work, our designed lanthanide complexes [Ln(4-VDPA)3] contain three reaction sites, which can be copolymerized with N-[tris(hydroxymethyl) methyl] acrylamide (THMA), acrylamide (AM), and diacryloyl poly(ethylene glycol) (DPEG) to form the first chemical crosslinking network, while hydroxypropyltrimethyl ammonium chloride chitosan (HACC) interacts with the hydroxyl and amino groups derived from the chemical crosslinking network through hydrogen bonds to form the second physical crosslinking network. The structure of the double network as well as the dynamic hydrogen bond and lanthanide coordination endow the hydrogel with excellent stretchability, adhesion and self-healing properties. Moreover, the introduction of lanthanide complexes and chitosan makes the hydrogel exhibit outstanding luminescence and antibacterial performances. This research not only realizes the simple synthesis of multifunctional luminescent hydrogels, but also provides a new idea for the fabrication of biomass-based hydrogels as intelligent and sustainable materials.


Subject(s)
Chitosan , Lanthanoid Series Elements , Prunella , Hydrogels , Luminescence , Acrylamide , Anti-Bacterial Agents/pharmacology , Tissue Adhesions
4.
J Biomed Mater Res A ; 112(7): 973-987, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38308554

ABSTRACT

The degeneration of intervertebral disc (IVD) is a disease of the entire joint between two vertebrae in the spine caused by loss of extracellular matrix (ECM) integrity, to date with no cure. The various regenerative approaches proposed so far have led to very limited successes. An emerging opportunity arises from the use of decellularized ECM as a scaffolding material that, directly or in combination with other materials, has greatly facilitated the advancement of tissue engineering. Here we focused on the decellularized matrix obtained from human umbilical cord Wharton's jelly (DWJ) which retains several structural and bioactive molecules very similar to those of the IVD ECM. However, being a viscous gel, DWJ has limited ability to retain ordered structural features when considered as architecture scaffold. To overcome this limitation, we produced DWJ-based multifunctional hydrogels, in the form of 3D millicylinders containing different percentages of alginate, a seaweed-derived polysaccharide, and gelatin, denatured collagen, which may impart mechanical integrity to the biologically active DWJ. The developed protocol, based on a freezing step, leads to the consolidation of the entire polymeric dispersion mixture, followed by an ionic gelation step and a freeze-drying process. Finally, a porous, stable, easily storable, and suitable matrix for ex vivo experiments was obtained. The properties of the millicylinders (Wharton's jelly millicylinders [WJMs]) were then tested in culture of degenerated IVD cells isolated from disc tissues of patients undergoing surgical discectomy. We found that WJMs with the highest percentage of DWJ were effective in supporting cell migration, restoration of the IVD phenotype (increased expression of Collagen type 2, aggrecan, Sox9 and FOXO3a), anti-inflammatory action, and stem cell activity of resident progenitor/notochordal cells (increased number of CD24 positive cells). We are confident that the DWJ-based formulations proposed here can provide adequate stimuli to the cells present in the degenerated IVD to restart the anabolic machinery.


Subject(s)
Hydrogels , Intervertebral Disc , Regeneration , Wharton Jelly , Humans , Wharton Jelly/cytology , Hydrogels/chemistry , Hydrogels/pharmacology , Intervertebral Disc Degeneration/therapy , Intervertebral Disc Degeneration/pathology , Tissue Scaffolds/chemistry , Cells, Cultured
5.
ACS Nano ; 18(4): 3468-3479, 2024 Jan 30.
Article in English | MEDLINE | ID: mdl-38227490

ABSTRACT

Chronic wounds have imposed a severe physical and economic burden on the global healthcare system, which are usually treated by the delivery of drugs or bioactive molecules to the wound bed through wound dressings. In this work, we have demonstrated a hydrogel-functionalized bandage with Janus wettability in a bilayer structure to achieve unidirectional drug delivery and multifunctional wound care. The Janus patterned bandage with porous gradient wetting channels on the upper layer is responsible for the unidirectional transport of the drug from the outside to the wound bed (up to 90% drug transport efficiency) while preventing drug diffusion in unwanted directions (<8%). The hydrogel composed of chitosan quaternary ammonium salt (HACC), poly(vinyl alcohol) (PVA), and poly(acrylic acid) (PAA) at the bottom layer further functionalized such a bandage with biocompatibility, excellent antibacterial properties, and hemostatic ability to promote wound healing. Especially, the hydrogel-functionalized bandage with Janus wettability exhibits excellent mechanical flexibility (∼198% strain), which can comply well with skin deformation (stretching, bending, or twisting) and maintain unidirectional drug delivery behavior without any leakage. The in vivo full-thickness skin wound model confirms that the hydrogel-functionalized bandage can significantly facilitate epithelialization and collagen deposition and improve drug delivery efficiency, thus promoting wound closure and healing (the wound healing ratio was 98.10% at day 15). Such a synergistic strategy of unidirectional drug delivery and multifunctional wound care provides a more efficient, economical, and direct method to promote wound healing, which could be used as a potential high-performance wound dressing for clinical application.


Subject(s)
Chitosan , Wound Healing , Humans , Wettability , Skin , Hydrogels/chemistry , Bandages , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Chitosan/chemistry
6.
Adv Healthc Mater ; 13(3): e2302328, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37824839

ABSTRACT

Diabetic wound is one of the chronic wounds that is difficult to heal, and effective treatment of it still confronts a great challenge. Monitoring the variation of diabetic wound microenvironment (such as hydrogen peroxide (H2 O2 )) can understand the wound state and guide the wound management. Herein, a multifunctional hydrogel with the abilities of monitoring the H2 O2 concentration, alleviating oxidative stress and promoting wound healing is developed, which is prepared by encapsulating manganese-containing bioactive glass (MnBG) and CePO4 :Tb in biocompatible gelatin methacryloyl (GelMA) hydrogel (CPT-MnBG-Gel). On the one hand, the H2 O2 -dependent fluorescence quenching effect of the CePO4 :Tb contributes to visible monitoring of the H2 O2 concentration of wounds via smartphone imaging, and the CPT-MnBG-Gel hydrogel can effectively monitor the H2 O2 level of 10.35-200 µmol L-1 . On the other hand, MnBG can alleviate oxidative stress and promote the proliferation, migration and differentiation of fibroblasts and endothelial cells in vitro owing to the bioactive Mn and Si ions, and in vivo evaluation also demonstrates that the CPT-MnBG-Gel hydrogels can effectively accelerate wound healing. Hence, such multifunctional hydrogel is promising for diabetic wound management and accelerating wound healing.


Subject(s)
Diabetes Mellitus , Hydrogels , Hydrogels/pharmacology , Endothelial Cells , Cell Differentiation , Fibroblasts , Wound Healing , Anti-Bacterial Agents
7.
Adv Healthc Mater ; : e2303537, 2023 Dec 07.
Article in English | MEDLINE | ID: mdl-38060436

ABSTRACT

The synergistic strategy of nanozyme-based catalytic therapy and photothermal therapy holds great potential for combating bacterial infection. However, challenges such as single and limited enzyme catalytic property, unfavorable catalytic environment, ineffective interaction between nanozymes and bacteria, unsafe laser irradiation ranges, and failed trauma fluid management impede their antibacterial capability and wound healing speed. Herein, for the first time, a PNMn hydrogel is fabricated with multi-enzyme activities and excellent near-infrared (NIR)-II photothermal performance for self-enhanced NIR-II photothermal-catalytic capabilities to efficiently eradicate bacteria. This hydrogel triggers parallel and cascade reactions to generate •OH, •O2 - , and 1 O2 radicals from H2 O2 and O2 without external energy input. Notably, it provides a suitable catalytic environment while capturing bacteria (≈30.1% of Escherichia coli and ≈29.3% of Staphylococcus aureus) to reinforce antibacterial activity. Furthermore, the PNMn hydrogel expedites skin wound healing by managing excess fluid (swelling rate up to ≈7299%). The PNMn hydrogel possesses remarkable stretching, elasticity, toughness, and adhesive characteristics under any shape of the wound, thus making it suitable for wound dressing. Therefore, the PNMn hydrogel has great potential to be employed as a next-generation wound dressing in the clinical context, providing a non-antibiotic strategy to improve the antibacterial performance and promote wound healing.

8.
Polymers (Basel) ; 15(19)2023 Sep 28.
Article in English | MEDLINE | ID: mdl-37835967

ABSTRACT

The development of biological macromolecule hydrogel dressings with fatigue resistance, sufficient mechanical strength, and versatility in clinical treatment is critical for accelerating full-thickness healing of skin wounds. Therefore, in this study, multifunctional, biological macromolecule hydrogels based on a recombinant type I collagen/chitosan scaffold incorporated with a metal-polyphenol structure were fabricated to accelerate wound healing. The resulting biological macromolecule hydrogel possesses sufficient mechanical strength, fatigue resistance, and healing properties, including antibacterial, antioxygenic, self-healing, vascularization, hemostatic, and adhesive abilities. Chitosan and recombinant type I collagen formed the scaffold network, which was the first covalent crosslinking network of the hydrogel. The second physical crosslinking network comprised the coordination of a metal-polyphenol structure, i.e., Cu2+ with the catechol group of dopamine methacrylamide (DMA) and stacking of DMA benzene rings. Double-crosslinked networks are interspersed and intertwined in the hydrogel to reduce the mechanical strength and increase its fatigue resistance, making it more suitable for clinical applications. Moreover, the biological macromolecule hydrogel can continuously release Cu2+, which provides strong antibacterial and vascularization properties. An in vivo full-thickness skin defect model confirmed that multifunctional, biological macromolecule hydrogels based on a recombinant type I collagen/chitosan scaffold incorporated with a metal-polyphenol structure can facilitate the formation of granulation tissue and collagen deposition for a short period to promote wound healing. This study highlights that this biological macromolecule hydrogel is a promising acute wound-healing dressing for biomedical applications.

9.
Acta Biomater ; 172: 330-342, 2023 12.
Article in English | MEDLINE | ID: mdl-37806374

ABSTRACT

Flexible epidermal sensors based on conductive hydrogels hold great promise for various applications, such as wearable electronics and personal healthcare monitoring. However, the integration of conductive hydrogel epidermal sensors into multiple applications remains challenging. In this study, a multifunctional PAAm/PEG/hydrolyzed keratin (Hereinafter referred to as HK)/MXene conductive hydrogel (PPHM hydrogel) was designed as a high-performance therapeutic all-in-one epidermal sensor. This sensor not only accelerates wound healing but also provides wearable human-computer interaction. The developed sensor possesses highly sensitive sensing properties (Gauge Factor = 4.82 at high strain), strong mechanical tensile properties (capable of achieving a maximum elongation at break of 600 %), rapid self-healing capability, stable self-adhesive capability, biocompatibility, freeze resistance at -20 °C, and adjustable photo-thermal conversion capability. This therapeutic all-in-one sensor can sensitively monitor human movements, enabling the detection of small electrophysiological signals for diagnosing relevant activities and diseases. Furthermore, using a rat frostbite model, we demonstrated that the composite hydrogel sensor can serve as an effective wound dressing to accelerate the healing process. This study serves as a valuable reference for the development of multifunctional flexible epidermal sensors for personal smart health monitoring. STATEMENT OF SIGNIFICANCE: Accelerated wound healing reduces the risk of wound infection, and conductive hydrogel-based sensors can monitor physiological signals. The multifunctional application of conductive hydrogel sensors combined with wound diagnostic and therapeutic capabilities can meet personalized medical requirements for wound healing and sensor monitoring. The aim of this study is to develop a multifunctional hydrogel patch. The multifunctional hydrogel can be assembled into a flexible wearable high-performance diagnostic and therapeutic integrated sensor that can effectively accelerate the healing of frostbite wounds and satisfy the real-time monitoring of multi-application scenarios. We expect that this study will inform efforts to integrate wound therapy and sensor monitoring.


Subject(s)
Frostbite , Humans , Animals , Rats , Frostbite/therapy , Bandages , Cytoskeleton , Electric Conductivity , Hydrogels/pharmacology
10.
Biomater Adv ; 154: 213615, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37716334

ABSTRACT

Hyaluronic acid/silk fibroin (HA/SF or HS) hydrogels with remarkable mechanical characteristics have been reported as tissue engineering biomaterials. Herein, the addition of dopamine/polydopamine (DA/PDA) to HS hydrogels to develop multifunctional HA/PDA/SF (or HDS) hydrogels for the delivery of drugs such as N-acetyl-L-cysteine (NAC) from nasal to brain tissue is examined. Herein, DA-dependent functions of HDS hydrogels with highly adhesive forces, photothermal response (PTR) effects generated by near infrared (NIR) irradiation, and anti-oxidative effects were demonstrated. An in-vitro study shows that the HDS/NAC hydrogels could open tight junctions in the RPMI 2650 cell line, a model cell of the nasal mucosa, as demonstrated by the decreased values of transepithelial electrical resistance (TEER) and more discrete ZO-1 staining than those for the control group. This effect was markedly enhanced by NIR irradiation of the HDS/NAC-NIR hydrogels. Compared to the results obtained using NAC solution, an in-vivo imaging study (IVIS) in rats showed an approximately nine-fold increase in the quantity of NAC delivered from the nasal cavity to the brain tissue in the span of 2 h through the PTR effect generated by the NIR irradiation of the nasal tissue and administration of the HDS/NAC hydrogels. Herein, dopamine-dependent multifunctional HDS hydrogels were studied, and the nasal administration of HDS/NAC-NIR hydrogels with PTR effects generated by NIR irradiation was found to have significantly enhanced NAC delivery to brain tissues.


Subject(s)
Fibroins , Rats , Animals , Acetylcysteine/pharmacology , Hyaluronic Acid/pharmacology , Dopamine/pharmacology , Hydrogels/pharmacology , Nasal Cavity , Brain
11.
Environ Res ; 238(Pt 1): 117087, 2023 12 01.
Article in English | MEDLINE | ID: mdl-37716390

ABSTRACT

Hydrogels represent intricate three-dimensional polymeric structures, renowned for their compatibility with living systems and their ability to naturally degrade. These networks stand as promising and viable foundations for a range of biomedical uses. The practical feasibility of employing hydrogels in clinical trials has been well-demonstrated. Among the prevalent biomedical uses of hydrogels, a significant application arises in the context of wound healing. This intricate progression involves distinct phases of inflammation, proliferation, and remodeling, often triggered by trauma, skin injuries, and various diseases. Metabolic conditions like diabetes have the potential to give rise to persistent wounds, leading to delayed healing processes. This current review consolidates a collection of experiments focused on the utilization of hydrogels to expedite the recovery of wounds. Hydrogels have the capacity to improve the inflammatory conditions at the wound site, and they achieve this by diminishing levels of reactive oxygen species (ROS), thereby exhibiting antioxidant effects. Hydrogels have the potential to enhance the growth of fibroblasts and keratinocytes at the wound site. They also possess the capability to inhibit both Gram-positive and Gram-negative bacteria, effectively managing wounds infected by drug-resistant bacteria. Hydrogels can trigger angiogenesis and neovascularization processes, while also promoting the M2 polarization of macrophages, which in turn mitigates inflammation at the wound site. Intelligent and versatile hydrogels, encompassing features such as pH sensitivity, reactivity to reactive oxygen species (ROS), and responsiveness to light and temperature, have proven advantageous in expediting wound healing. Furthermore, hydrogels synthesized using environmentally friendly methods, characterized by high levels of biocompatibility and biodegradability, hold the potential for enhancing the wound healing process. Hydrogels can facilitate the controlled discharge of bioactive substances. More recently, there has been progress in the creation of conductive hydrogels, which, when subjected to electrical stimulation, contribute to the enhancement of wound healing. Diabetes mellitus, a metabolic disorder, leads to a slowdown in the wound healing process, often resulting in the formation of persistent wounds. Hydrogels have the capability to expedite the healing of diabetic wounds, facilitating the transition from the inflammatory phase to the proliferative stage. The current review sheds light on the biological functionalities of hydrogels, encompassing their role in modulating diverse mechanisms and cell types, including inflammation, oxidative stress, macrophages, and bacteriology. Additionally, this review emphasizes the significance of smart hydrogels with responsiveness to external stimuli, as well as conductive hydrogels for promoting wound healing. Lastly, the discussion delves into the advancement of environmentally friendly hydrogels with high biocompatibility, aimed at accelerating the wound healing process.


Subject(s)
Diabetes Mellitus , Hydrogels , Humans , Hydrogels/chemistry , Hydrogels/pharmacology , Reactive Oxygen Species , Anti-Bacterial Agents/pharmacology , Precision Medicine , Gram-Negative Bacteria , Gram-Positive Bacteria , Wound Healing , Inflammation
12.
Adv Healthc Mater ; 12(30): e2301696, 2023 12.
Article in English | MEDLINE | ID: mdl-37669499

ABSTRACT

Postoperative peritoneal adhesion is a serious clinical complication. Various hydrogel barriers have been developed to prevent peritoneal adhesion. However, it remains a challenge to design a hydrogel with desirable physicochemical properties and bioactivities. In this study, a zwitterionic polysaccharide-based multifunctional hydrogel is developed using epigallocatechin-3-gallate (EGCG) to prevent postoperative abdominal adhesion. This hydrogel is simple to use and has desirable properties, such as excellent injectability, self-healing, and non-swelling properties. The hydrogel also has ultralow fouling capabilities, such as superior bactericidal performance, cell and protein adhesion, and low immunogenicity resistance. Moreover, the hydrogel exhibits good antioxidant activity, which is attributed to the integration of EGCG. Furthermore, the detailed mechanism from in vivo and in vitro experimental studies illustrates that hydrogel compositions can synergistically prevent adhesion formation through multiple pathways, including anti-inflammatory and antioxidant capabilities and inhibition effects on the mesothelial-mesenchymal transition (MMT) process induced by transforming growth factor (TGF-ß). In summary, this zwitterionic multifunctional hydrogel has great potential to prevent postoperative adhesion formation in the clinical setting.


Subject(s)
Hydrogels , Peritoneum , Hydrogels/chemistry , Peritoneum/metabolism , Peritoneum/surgery
13.
Adv Healthc Mater ; 12(28): e2301370, 2023 11.
Article in English | MEDLINE | ID: mdl-37437207

ABSTRACT

Continuous oxidative stress and cellular dysfunction caused by hyperglycemia are distinguishing features of diabetic wounds. It has been a great challenge to develop a smart dressing that can accelerate diabetic wound healing through regulating abnormal microenvironments. In this study, a platelet rich plasma (PRP) loaded multifunctional hydrogel with reactive oxygen species (ROS) and glucose dual-responsive property is reported. It can be conveniently prepared with PRP, dopamine (DA) grafted alginate (Alg-DA), and 6-aminobenzo[c][1,2]oxaborol-1(3H)-ol (ABO) conjugated hyaluronic acid (HA-ABO) through ionic crosslinks, hydrogen-bond interactions, and boronate ester bonds. The hydrogel possesses injectability, moldability, tissue adhesion, self-healing, low hemolysis, and hemostasis performances. Its excellent antioxidant property can create a low oxidative stress microenvironment for other biological events. Under an oxidative stress and/or hyperglycemia state, the hydrogel can degrade at an accelerated rate to release a variety of cytokines derived from activated blood platelets. The result is a series of positive changes that are favorable for diabetic wound healing, including fast anti-inflammation, activated macrophage polarization toward M2 phenotype, promoted migration and proliferation of fibroblasts, as well as expedited angiogenesis. This work provides an efficient strategy for chronic diabetic wound management and offers an alternative for developing a new-type PRP-based bioactive wound dressing.


Subject(s)
Diabetes Mellitus , Hyperglycemia , Platelet-Rich Plasma , Humans , Hydrogels/pharmacology , Alginates , Dopamine , Anti-Bacterial Agents
14.
Regen Biomater ; 10: rbad046, 2023.
Article in English | MEDLINE | ID: mdl-37287896

ABSTRACT

Autologous and allogeneic bone grafts remain the gold standard for repairing bone defects. However, donor shortages and postoperative infections contribute to unsatisfactory treatment outcomes. Tissue engineering technology that utilizes biologically active composites to accelerate the healing and reconstruction of segmental bone defects has led to new ideas for in situ bone repair. Multifunctional nanocomposite hydrogels were constructed by covalently binding silver (Ag+) core-embedded mesoporous silica nanoparticles (Ag@MSN) to bone morphogenetic protein-2 (BMP-2), which was encapsulated into silk fibroin methacryloyl (SilMA) and photo-crosslinked to form an Ag@MSN-BMP-2/SilMA hydrogel to preserve the biological activity of BMP-2 and slow its release. More importantly, multifunctional Ag+-containing nanocomposite hydrogels showed antibacterial properties. These hydrogels possessed synergistic osteogenic and antibacterial effects to promote bone defect repair. Ag@MSN-BMP-2/SilMA exhibited good biocompatibility in vitro and in vivo owing to its interconnected porosity and improved hydrophilicity. Furthermore, the multifunctional nanocomposite hydrogel showed controllable sustained-release activity that promoted bone regeneration in repairing rat skull defects by inducing osteogenic differentiation and neovascularization. Overall, Ag@MSN-BMP-2/SilMA hydrogels enrich bone regeneration strategies and show great potential for bone regeneration.

15.
Small ; 19(28): e2300111, 2023 07.
Article in English | MEDLINE | ID: mdl-37191242

ABSTRACT

A photoactivated bone scaffold integrated with minimally invasive implantation and mild thermal-stimulation capability shows great promise in the repair and regeneration of irregularly damaged bone tissues. Developing multifunctional photothermal biomaterials that can simultaneously serve as both controllable thermal stimulators and biodegradable engineering scaffolds for integrated immunomodulation, infection therapy, and impaired bone repair remains an enormous challenge. Herein, an injectable and photocurable hydrogel therapeutic platform (AMAD/MP) based on alginate methacrylate, alginate-graft-dopamine, and polydopamine (PDA)-functionalized Ti3C2 MXene (MXene@PDA) nanosheets is rationally designed for near-infrared (NIR)-mediated bone regeneration synergistic immunomodulation, osteogenesis, and bacterial elimination. The optimized AMAD/MP hydrogel exhibits favorable biocompatibility, osteogenic activity, and immunomodulatory functions in vitro. The proper immune microenvironment provided by AMAD/MP could further modulate the balance of M1/M2 phenotypes of macrophages, thereby suppressing reactive oxygen species-induced inflammatory status. Significantly, this multifunctional hydrogel platform with mild thermal stimulation efficiently attenuates local immune reactions and further promotes new bone formation without the addition of exogenous cells, cytokines, or growth factors. This work highlights the potential application of an advanced multifunctional hydrogel providing photoactivated on-demand thermal cues for bone tissue engineering and regenerative medicine.


Subject(s)
Hydrogels , Osteogenesis , Hydrogels/pharmacology , Bone Regeneration , Biocompatible Materials , Tissue Engineering , Tissue Scaffolds
16.
Gels ; 9(4)2023 Mar 27.
Article in English | MEDLINE | ID: mdl-37102889

ABSTRACT

Chitosan is a promising naturally derived polysaccharide to be used in hydrogel forms for pharmaceutical and biomedical applications. The multifunctional chitosan-based hydrogels have attractive properties such as the ability to encapsulate, carry, and release the drug, biocompatibility, biodegradability, and non-immunogenicity. In this review, the advanced functions of the chitosan-based hydrogels are summarized, with emphasis on fabrications and resultant properties reported in literature from the recent decade. The recent progress in the applications of drug delivery, tissue engineering, disease treatments, and biosensors are reviewed. Current challenges and future development direction of the chitosan-based hydrogels for pharmaceutical and biomedical applications are prospected.

17.
Adv Sci (Weinh) ; 10(17): e2206306, 2023 06.
Article in English | MEDLINE | ID: mdl-37078785

ABSTRACT

Chronic inflammatory diseases, such as intervertebral disc degeneration (IVDD), which affect the lives of hundreds of millions of people, still lack effective and precise treatments. In this study, a novel hydrogel system with many extraordinary properties is developed for gene-cell combination therapy of IVDD. Phenylboronic acid-modified G5 PAMAM (G5-PBA) is first synthesized, and therapeutic siRNA silencing the expression of P65 mixed with G5-PBA (siRNA@G5-PBA) is then embedded into the hydrogel (siRNA@G5-PBA@Gel) based on multi-dynamic bonds including acyl hydrazone bonds, imine linkage, π-π stacking, and hydrogen bonding interactions. Local and acidic inflammatory microenvironment-responsive gene-drug release can achieve spatiotemporal regulation of gene expression. In addition, gene-drug release from the hydrogel can be sustained for more than 28 days in vitro and in vivo, greatly inhibiting the secretion of inflammatory factors and the subsequent degeneration of nucleus pulposus (NP) cells induced by lipopolysaccharide (LPS). Through prolonged inhibition of the P65/NLRP3 signaling pathway, the siRNA@G5-PBA@Gel is verified to relieve inflammatory storms, which can significantly enhance the regeneration of IVD when combined with cell therapy. Overall, this study proposes an innovative system for gene-cell combination therapy and a precise and minimally invasive treatment method for IVD regeneration.


Subject(s)
Intervertebral Disc Degeneration , Intervertebral Disc , Humans , Intervertebral Disc Degeneration/genetics , Intervertebral Disc Degeneration/therapy , Hydrogels/chemistry , Intervertebral Disc/metabolism , RNA, Small Interfering/metabolism , Cell- and Tissue-Based Therapy
18.
Int J Biol Macromol ; 227: 505-523, 2023 Feb 01.
Article in English | MEDLINE | ID: mdl-36495992

ABSTRACT

Hydrogel is a three-dimensional network polymer material rich in water. It is widely used in the biomedical field because of its unique physical and chemical properties and good biocompatibility. In recent years, the incidence of inflammatory bowel disease (IBD) has gradually increased, and the disadvantages caused by traditional drug treatment of IBD have emerged. Therefore, there is an urgent need for new treatments to alleviate IBD. Hydrogel has become a potential therapeutic platform. However, there is a lack of comprehensive review of functional hydrogels for IBD treatment. This paper first summarizes the pathological changes in IBD sites. Then, the action mechanisms of hydrogels prepared from chitosan, sodium alginate, hyaluronic acid, functionalized polyethylene glycol, cellulose, pectin, and γ-polyglutamic acid on IBD were described from aspects of drug delivery, peptide and protein delivery, biologic therapies, loading probiotics, etc. In addition, the advanced functions of IBD treatment hydrogels were summarized, with emphasis on adhesion, synergistic therapy, pH sensitivity, particle size, and temperature sensitivity. Finally, the future development direction of IBD treatment hydrogels has been prospected.


Subject(s)
Chitosan , Inflammatory Bowel Diseases , Humans , Chitosan/chemistry , Hydrogels/chemistry , Hyaluronic Acid/chemistry , Drug Delivery Systems , Inflammatory Bowel Diseases/drug therapy
19.
Bioact Mater ; 24: 96-111, 2023 Jun.
Article in English | MEDLINE | ID: mdl-36582346

ABSTRACT

Wound healing has become one of the basic issues faced by the medical community because of the susceptibility of skin wounds to bacterial infection. As such, it is highly desired to design a nanocomposite hydrogel with excellent antibacterial activity to achieve high wound closure effectiveness. Here, based on ultrasound-triggered piezocatalytic therapy, a multifunctional hydrogel is designed to promote bacteria-infected wound healing. Under ultrasonic vibration, the surface of barium titanate (BaTiO3, BT) nanoparticles embedded in the hydrogel rapidly generate reactive oxygen species (ROS) owing to the established strong built-in electric field, endowing the hydrogel with superior antibacterial efficacy. This modality shows intriguing advantages over conventional photodynamic therapy, such as prominent soft tissue penetration ability and the avoidance of serious skin phototoxicity after systemic administration of photosensitizers. Moreover, the hydrogel based on N-[tris(hydroxymethyl)methyl]acrylamide (THM), N-(3-aminopropyl)methacrylamide hydrochloride (APMH) and oxidized hyaluronic acid (OHA) exhibits outstanding self-healing and bioadhesive properties able to accelerate full-thickness skin wound healing. Notably, compared with the widely reported mussel-inspired adhesive hydrogels, OHA/THM-APMH hydrogel due to the multiple hydrogen bonds from unique tri-hydroxyl structure overcomes the shortage that catechol groups are easily oxidized, giving it long-term and repeatable adhesion performance. Importantly, this hybrid hydrogel confines BT nanoparticles to wound area and locally induced piezoelectric catalysis under ultrasound to eradicate bacteria, markedly improving the therapeutic biosafety and exhibits great potential for harmless treatment of bacteria-infected tissues.

20.
Polymers (Basel) ; 14(23)2022 Dec 05.
Article in English | MEDLINE | ID: mdl-36501708

ABSTRACT

Ionic conductive hydrogels used as flexible wearable sensor devices have attracted considerable attention because of their easy preparation, biocompatibility, and macro/micro mechanosensitive properties. However, developing an integrated conductive hydrogel that combines high mechanical stability, strong adhesion, and excellent mechanosensitive properties to meet practical requirements remains a great challenge owing to the incompatibility of properties. Herein, we prepare a multifunctional ionic conductive hydrogel by introducing high-modulus bacterial cellulose (BC) to form the skeleton of double networks, which exhibit great mechanical properties in both tensile (83.4 kPa, 1235.9% strain) and compressive (207.2 kPa, 79.9% strain) stress-strain tests. Besides, the fabricated hydrogels containing high-concentration Ca2+ show excellent anti-freezing (high ionic conductivities of 1.92 and 0.36 S/m at room temperature and -35 ∘C, respectively) properties. Furthermore, the sensing mechanism based on the conductive units and applied voltage are investigated to the benefit of the practical applications of prepared hydrogels. Therefore, the designed and fabricated hydrogels provide a novel strategy and can serve as candidates in the fields of sensors, ionic skins, and soft robots.

SELECTION OF CITATIONS
SEARCH DETAIL