Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 214
Filter
1.
J Mater Chem B ; 2024 Aug 20.
Article in English | MEDLINE | ID: mdl-39161280

ABSTRACT

Colloidal photonic crystals (CPCs), fabricated from the assembly of micro-/nano-particles, have attracted considerable interest due to their unique properties, such as structural color, slow-photon effect, and high specific surface area (SSA). Benefiting from these properties, significant progress has been made in the biological applications of CPCs. In this perspective, these properties and relative manipulation strategies are firstly discussed, building bridges between properties and biological applications of CPCs. Structural color endows CPCs with naked-eye sensing capability, which can be applied to physiological state assessment and diagnosis, as well as self-report of CPC-based diagnostic and therapeutic devices. The slow-photon effect contributes to enhanced fluorescence, surface-enhanced Raman scattering, and efficacy of photodynamic/photothermal therapy, when CPCs are combined with corresponding functional materials. High SSA provides CPCs with abundant binding sites and superior capabilities for loading, adsorption, delivery, etc. These properties can be utilized individually or synergistically to grant CPCs superior performance in biological applications. Next, the recent advancements of CPCs towards biological applications are summarized, including biosensors, wound dressings, cells-on-a-chip, and phototherapy. Finally, a perspective on the challenges and future development of CPCs for biological applications is presented.

2.
Adv Sci (Weinh) ; : e2405764, 2024 Aug 21.
Article in English | MEDLINE | ID: mdl-39166390

ABSTRACT

In treating infectious diseases, achieving selective bacterial inhibition is crucial for preserving the microecological equilibrium. The current approaches predominantly rely on synthetic materials tailored to specific bacteria, considering their cell walls or oxygen requirements. Herein, inspired by intricate bacterial communication, a natural implant is proposed coating utilizing bacterial outer membrane vesicles (OMVs), essential components in bacterial signaling, integrated onto diverse implant surfaces through a universal poly (tannic acid) bridging layer. This coating is homogenous and stable, unexpectedly promoting the proliferation of parental bacteria while inhibiting heterologous bacteria both in vitro and in vivo. Through high-throughput sequencing and bioinformatics analysis, the selective bacteriostatic ability arises from OMVs, upregulating anti-oxidative stress genes in heterologous bacteria and activating biofilm-related genes in parental bacteria. This study positions OMVs as an appealing biomaterial for selective bacterial inhibition through a biological approach, showcasing their potential in regulating the microecological balance through a natural interface modification strategy.

3.
Mater Horiz ; 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-39143938

ABSTRACT

Osteoarthritis (OA), which disables articular cartilage, affects millions of people. The self-healing capacity is inhibited by internal oxidative stress and external lubrication deficiency and enzymatic degradation. To overcome these challenges, a tailored cartilage-armor is designed to ameliorate the inflamed cartilage, which is implemented by a novel collagen type II (Col II)-binding peptide conjugated zwitterionic polymer (PSB-b-PColBP, PSP). By mimicking natural lubricin, PSP specifically targets the cartilage surface and forms an in situ hydration armor. This engineered cartilage-armor can prevent enzymatic cartilage degradation (nearly 100% resistance to catabolic enzymes) and provide durable lubrication properties (COF < 0.013 for 500 cycles). An autophagy-activation process, absent in previous biomimetic lubricants, enhances the enzymatic activity of the tailored cartilage-armor, offering effective anti-oxidant properties to suppress oxidative stress. By inhibiting the PI3K-Akt/NF-κB signaling pathway, chondrocytes protected by the tailored armor can secrete a cartilage matrix even in inflammatory microenvironments. In OA rat models, osteophyte formation and the inflammatory response have been inhibited by the cartilage-armor, demonstrating a therapeutic effect comparable to most drug-loaded systems. This study underscores the potential of tailoring cartilage-armor with the cartilage targeting and autophagy-activating properties in integrating offensive-defensive mechanisms for cartilage remodeling. This represents an alternative strategy for clinical OA therapy.

4.
ACS Appl Mater Interfaces ; 16(32): 41881-41891, 2024 Aug 14.
Article in English | MEDLINE | ID: mdl-39092619

ABSTRACT

Dental caries, the most prevalent chronic disease across all age groups, has a high prevalence, particularly among children. However, there is no specific and effective treatment for the prevention of caries in primary teeth (Pr.T.), which stems from a lack of knowledge regarding the basic nature of the tooth surface. Herein, we observed that the adhesion energies of the caries-related bacteria Streptococcus mutans and Streptococcus sanguinis to Pr.T were approximately 10 and 5.5 times higher than those to permanent teeth (Pe.T). A lower degree of mineralization and more hydrophilic characteristics of the Pr.T enamel account for this discrepancy. Accordingly, we proposed that the on-target modification of both hydroxyapatite and organic components on Pr.T by dual modification would render a sufficient hydration layer. This resulted in an approximately 11-time decrease in bacterial adhesion energy after treatment. In contrast, a single hydroxyapatite modification on Pe.T and young permanent teeth (Y.Pe.T) was sufficient to achieve a similar effect. Theoretical simulation further verified the rationality of the approach. Our findings may help understand the reason for Pr.T being caries-prone and provide references for treatment using resin restorations. This strategy offers valuable insights into daily oral hygiene and dental prophylactic treatment in children.


Subject(s)
Bacterial Adhesion , Dental Caries , Durapatite , Streptococcus mutans , Streptococcus sanguis , Tooth, Deciduous , Dental Caries/prevention & control , Dental Caries/microbiology , Streptococcus mutans/drug effects , Humans , Bacterial Adhesion/drug effects , Streptococcus sanguis/drug effects , Durapatite/chemistry , Dental Enamel/chemistry , Dental Enamel/drug effects
5.
Front Nutr ; 11: 1401035, 2024.
Article in English | MEDLINE | ID: mdl-39176026

ABSTRACT

This study aims to investigate the influence of psychological ownership of nutritional products on the purchase intention of high-pressure working groups, as well as the underlying mechanisms and boundary conditions of this influence. This study aims to investigate the impact of psychological ownership of nutritional products on the purchase intention of high-pressure working groups, as well as the underlying mechanisms and boundary conditions of this influence. The research contributes through the use of variance analysis, mediation models, and moderation models on data from adult participants over the age of 18, across three experiments. Experiment 1, conducted on the Credamo platform, randomly recruited 285 participants, with 148 males (51.9%) and 137 females (48.1%), and the results indicated a direct impact of psychological ownership on purchase intention. Building upon this, Experiment 2, which also utilized the Credamo platform and recruited 280 participants consisting of 136 males (48.6%) and 144 females (51.4%), further revealed the mediating role of perceived value. Experiment 3, with 265 participants randomly recruited on the Credamo platform, including 131 males (49.4%) and 134 females (50.6%), identified the moderating effect of nutritional awareness. The theoretical contribution of this study lies primarily in its in-depth exploration of the impact of psychological ownership of nutritional products. By integrating the factors of perceived value and nutritional awareness, it provides a systematic explanation for better understanding the purchase intentions of high-pressure working groups. Additionally, this study offers valuable strategies for merchants to enhance the purchase intentions of high-pressure living groups.

6.
Adv Mater ; : e2405953, 2024 Aug 05.
Article in English | MEDLINE | ID: mdl-39101293

ABSTRACT

Implant-associated infections (IAIs) are the main cause of prosthetic implant failure. Bacterial biofilms prevent antibiotic penetration, and the unique metabolic conditions in hypoxic biofilm microenvironment may limit the efficacy of conventional antibiotic treatment. Escaping survival bacteria may not be continually eradicated, resulting in the recurrence of IAIs. Herein, a sonosensitive metal-organic framework of Cu-TCPP (tetrakis(4-carboxyphenyl) porphyrin) nanosheets and tinidazole doped probiotic-derived membrane vesicles (OMVs) with high-penetration sonodynamic therapy (SDT), bacterial metabolic state interference, and bacterial cuproptosis-like death to eradicate IAIs is proposed. The Cu-TCPP can convert O2 to toxic 1O2 through SDT in the normoxic conditions, enhancing the hypoxic microenvironment and activating the antibacterial activity of tinidazole. The released Cu(II) under ultrasound can be converted to Cu(I) by exogenous poly(tannic acid) (pTA) and endogenous glutathione. The disruption of the bacterial membrane by SDT can enhance the Cu(I) transporter activity. Transcriptomics indicate that the SDT-enhanced Cu(I) overload and hypoxia-activated therapy hinder the tricarboxylic acid cycle (TCA), leading to bacterial cuproptosis-like death. Moreover, the OMVs-activated therapy can polarize macrophages to a M2-like phenotype and facilitate bone repair. The sonodynamic biofilm microenvironment modulation strategy, whereby the hypoxia-enhanced microenvironment is potentiated to synergize SDT with OMVs-activated therapy, provides an effective strategy for antibacterial and osteogenesis performance.

7.
Mater Horiz ; 2024 Jul 11.
Article in English | MEDLINE | ID: mdl-38990315

ABSTRACT

Photothermal therapy (PTT) encounters challenges in addressing deep tissue infections, characterized by limited penetration or potential hyperthermal damage to surrounding tissues, initiating undesirable inflammatory cascades. Inspired by polar bear thermal regulation, we present a "bio-based endogenic thermal-adaptive booster" implant coating. This coating integrates a photothermal poly(tannic acid) (pTA) layer, mimicking the "polar bear dark skin", securely linked with anti-inflammatory dexamethasone (Dex), resembling the "secretion", and a red blood cell membrane (RBCM) layer, forming the insulating "transparent fur". The RBCM "fur" demonstrates unexpectedly superior local heat storage, amplifying the photothermal effect of the pTA "skin" by 1.30 times and boosting localized photothermal antibacterial efficiency by 1.30-fold (approximately 99%) compared to those without RBCM. Furthermore, RBCM sustains Dex release and offers additional protection against thermal inflammation, releasing Dex 1.90 times more under NIR irradiation than under non-photothermal conditions. In a rat infectious bone model, the photothermal-boosting implant coating provides a favorable biological interface and achieves a 99.97% photothermal antibacterial ratio, enhancing osseointegration without evident tissue harm, evidenced by a 2.47-fold increase in bone volume fraction and a 2.24-fold reduction in pro-inflammatory cytokines compared to those lacking a RBCM. Insights derived from cell membrane-based thermal-adaptive coatings herald a paradigm shift in efficient and safe PTT.

8.
Biomater Sci ; 12(15): 3745-3764, 2024 Jul 23.
Article in English | MEDLINE | ID: mdl-38959069

ABSTRACT

Hydrogels, as an emerging biomaterial, have found extensive use in the healing of wounds due to their distinctive physicochemical structure and functional properties. Moreover, hydrogels can be made to match a range of therapeutic requirements for materials used in wound healing through specific functional modifications. This review provides a step-by-step explanation of the processes involved in cutaneous wound healing, including hemostasis, inflammation, proliferation, and reconstitution, along with an investigation of the factors that impact these processes. Furthermore, a thorough analysis is conducted on the various stages of the wound healing process at which functional hydrogels are implemented, including hemostasis, anti-infection measures, encouraging regeneration, scar reduction, and wound monitoring. Next, the latest progress of multifunctional hydrogels for wound healing and the methods to achieve these functions are discussed in depth and categorized for elucidation. Finally, perspectives and challenges associated with the clinical applications of multifunctional hydrogels are discussed.


Subject(s)
Hydrogels , Skin , Wound Healing , Wound Healing/drug effects , Hydrogels/chemistry , Hydrogels/pharmacology , Humans , Animals , Skin/drug effects , Skin/injuries , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Hemostasis/drug effects
9.
PLoS One ; 19(7): e0306476, 2024.
Article in English | MEDLINE | ID: mdl-38968246

ABSTRACT

OBJECTIVE: This study investigates the relationship between cardiovascular health (CVH), as quantified by the American Heart Association's Life's Essential 8 (LE8) metric, and female infertility, utilizing data from the National Health and Nutrition Examination Survey (NHANES) spanning 2013-2018. METHODS: We encompassed females aged 20-49 years and above from the NHANES in this cross-sectional analysis. We assessed CVH using the LE8 score, encompassing eight domains: dietary pattern, physical activity, nicotine exposure, sleep duration, body mass index (BMI), lipid profile, fasting blood glucose, and blood pressure levels. Logistic regression models were applied to explore the association between CVH scores and reported infertility, adjusting for potential confounders including age, race/ethnicity, and socioeconomic status. RESULTS: Findings revealed a notable inverse association between CVH scores (per 10 scores) and female infertility [OR = 0.93, 95%CI: 0.90-0.96], Participants with higher CVH levels were 41% less likely to had female infertility compared to those with lower levels [OR = 0.59, 95%CI: 0.41-0.84]. Higher overall CVH scores, particularly in physical activity, BMI, and blood glucose, were associated with lower odds of infertility. This trend remained consistent across various demographic subgroups. CONCLUSION: Our findings underscore the significance of maintaining optimal cardiovascular health, as evidenced by higher LE8 scores, in mitigating the risk of female infertility. These insights advocate for the integration of CVH improvement strategies within the broader framework of reproductive health care, emphasizing the dual benefits of cardiovascular and reproductive health optimization.


Subject(s)
Body Mass Index , Cardiovascular Diseases , Infertility, Female , Humans , Female , Adult , Middle Aged , Infertility, Female/epidemiology , Cross-Sectional Studies , Cardiovascular Diseases/epidemiology , Nutrition Surveys , Young Adult , Exercise , United States/epidemiology , Blood Glucose/analysis , Risk Factors
10.
Adv Healthc Mater ; : e2400163, 2024 Jul 29.
Article in English | MEDLINE | ID: mdl-39075811

ABSTRACT

Micro/nanomotors represent a promising class of drug delivery carriers capable of converting surrounding chemical or external energy into mechanical power, enabling autonomous movement. Their distinct autonomous propulsive force distinguishes them from other carriers, offering significant potential for enhancing drug penetration across cellular and tissue barriers. A comprehensive understanding of micro/nanomotor dynamics with various power sources is crucial to facilitate their transition from proof-of-concept to clinical application. In this review, micro/nanomotors are categorized into three classes based on their energy sources: endogenously stimulated, exogenously stimulated, and live cell-driven. The review summarizes the mechanisms governing micro/nanomotor movements under these energy sources and explores factors influencing autonomous motion. Furthermore, it discusses methods for controlling micro/nanomotor movement, encompassing aspects related to their structure, composition, and environmental factors. The remarkable propulsive force exhibited by micro/nanomotors makes them valuable for significant biomedical applications, including tumor therapy, bio-detection, bacterial infection therapy, inflammation therapy, gastrointestinal disease therapy, and environmental remediation. Finally, the review addresses the challenges and prospects for the application of micro/nanomotors. Overall, this review emphasizes the transformative potential of micro/nanomotors in overcoming biological barriers and enhancing therapeutic efficacy, highlighting their promising clinical applications across various biomedical fields.

11.
J Mater Chem B ; 12(29): 7001-7019, 2024 Jul 24.
Article in English | MEDLINE | ID: mdl-38919030

ABSTRACT

The properties of nanomaterials make them promising and advantageous for use in drug delivery systems, but challenges arise from the immune system's recognition of exogenous nanoparticles, leading to their clearance and reduced targeting efficiency. Drawing inspiration from nature, this paper explores biomimetic strategies to transform recognizable nanomaterials into a "camouflaged state." The focal point of this paper is the exploration of bionic nanoparticles, with a focus on cell membrane-coated nanoparticles. These biomimetic structures, particularly those mimicking red blood cells (RBCs), white blood cells (WBCs), platelets, and cancer cells, demonstrate enhanced drug delivery efficiency and prolonged circulation. This article underscores the versatility of these biomimetic structures across diverse diseases and explores the use of hybrid cell membrane-coated nanoparticles as a contemporary trend. This review also investigated exosomes and protein bionic nanoparticles, emphasizing their potential for specific targeting, immune evasion, and improved therapeutic outcomes. We expect that this continued development based on biomimetic nanomaterials will contribute to the efficiency and safety of disease treatment.


Subject(s)
Biomimetic Materials , Drug Delivery Systems , Humans , Biomimetic Materials/chemistry , Animals , Nanostructures/chemistry , Nanoparticles/chemistry , Biomimetics/methods
12.
Nanomicro Lett ; 16(1): 224, 2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38888701

ABSTRACT

Organic photovoltaics (OPVs) need to overcome limitations such as insufficient thermal stability to be commercialized. The reported approaches to improve stability either rely on the development of new materials or on tailoring the donor/acceptor morphology, however, exhibiting limited applicability. Therefore, it is timely to develop an easy method to enhance thermal stability without having to develop new donor/acceptor materials or donor-acceptor compatibilizers, or by introducing another third component. Herein, a unique approach is presented, based on constructing a polymer fiber rigid network with a high glass transition temperature (Tg) to impede the movement of acceptor and donor molecules, to immobilize the active layer morphology, and thereby to improve thermal stability. A high-Tg one-dimensional aramid nanofiber (ANF) is utilized for network construction. Inverted OPVs with ANF network yield superior thermal stability compared to the ANF-free counterpart. The ANF network-incorporated active layer demonstrates significantly more stable morphology than the ANF-free counterpart, thereby leaving fundamental processes such as charge separation, transport, and collection, determining the device efficiency, largely unaltered. This strategy is also successfully applied to other photovoltaic systems. The strategy of incorporating a polymer fiber rigid network with high Tg offers a distinct perspective addressing the challenge of thermal instability with simplicity and universality.

13.
Adv Sci (Weinh) ; 11(29): e2404143, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38785180

ABSTRACT

Commencing with the breakdown of the diabetic osteoimmune microenvironment, multiple pathogenic factors, including hyperglycemia, inflammation, hypoxia, and deleterious cytokines, are conjointly involved in the progression of diabetic periodontal bone regeneration. Based on the challenge of periodontal bone regeneration treatment and the absence of real-time feedback of blood oxygen fluctuation in diabetes mellitus, a novel self-adaptive hyperthermia supramolecular cascade nano-reactor ACFDG is constructed via one-step supramolecular self-assembly strategy to address multiple factors in diabetic periodontal bone regeneration. Hyperthermia supramolecular ACFDG possesses high photothermal conversion efficiency (32.1%), and it can effectively inhibit the vicious cycle of ROS-inflammatory cascade through catalytic cascade reactions, up-regulate the expression of heat shock proteins (HSPs) under near-infrared (NIR) irradiation, which promotes periodontal bone regeneration. Remarkably, ACFDG can provide real-time non-invasive diagnosis of blood oxygen changes during periodontal bone regeneration through photoacoustic (PA) imaging, thus can timely monitor periodontal hypoxia status. In conclusion, this multifunctional supramolecular nano-reactor combined with PA imaging for real-time efficacy monitoring provides important insights into the biological mechanisms of diabetic periodontal bone regeneration and potential clinical theranostics.


Subject(s)
Bone Regeneration , Photoacoustic Techniques , Photoacoustic Techniques/methods , Bone Regeneration/physiology , Animals , Diabetes Mellitus, Experimental/therapy , Hyperthermia, Induced/methods , Disease Models, Animal , Rats , Humans , Mice
14.
Small ; : e2311967, 2024 May 07.
Article in English | MEDLINE | ID: mdl-38712482

ABSTRACT

Intracellular bacteria pose a great challenge to antimicrobial therapy due to various physiological barriers at both cellular and bacterial levels, which impede drug penetration and intracellular targeting, thereby fostering antibiotic resistance and yielding suboptimal treatment outcomes. Herein, a cascade-target bacterial-responsive drug delivery nanosystem, MM@SPE NPs, comprising a macrophage membrane (MM) shell and a core of SPE NPs. SPE NPs consist of phenylboronic acid-grafted dendritic mesoporous silica nanoparticles (SP NPs) encapsulated with epigallocatechin-3-gallate (EGCG), a non-antibiotic antibacterial component, via pH-sensitive boronic ester bonds are introduced. Upon administration, MM@SPE NPs actively home in on infected macrophages due to the homologous targeting properties of the MM shell, which is subsequently disrupted during cellular endocytosis. Within the cellular environment, SPE NPs expose and spontaneously accumulate around intracellular bacteria through their bacteria-targeting phenylboronic acid groups. The acidic bacterial microenvironment further triggers the breakage of boronic ester bonds between SP NPs and EGCG, allowing the bacterial-responsive release of EGCG for localized intracellular antibacterial effects. The efficacy of MM@SPE NPs in precisely eliminating intracellular bacteria is validated in two rat models of intracellular bacterial infections. This cascade-targeting responsive system offers new solutions for treating intracellular bacterial infections while minimizing the risk of drug resistance.

15.
Adv Healthc Mater ; 13(20): e2400514, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38652681

ABSTRACT

Gliomas, the most prevalent primary brain tumors, pose considerable challenges due to their heterogeneity, intricate tumor microenvironment (TME), and blood-brain barrier (BBB), which restrict the effectiveness of traditional treatments like surgery and chemotherapy. This review provides an overview of engineered cell membrane technologies in glioma therapy, with a specific emphasis on targeted drug delivery and modulation of the immune microenvironment. This study investigates the progress in engineered cell membranes, encompassing physical, chemical, and genetic alterations, to improve drug delivery across the BBB and effectively target gliomas. The examination focuses on the interaction of engineered cell membrane-coated nanoparticles (ECM-NPs) with the TME in gliomas, emphasizing their potential to modulate glioma cell behavior and TME to enhance therapeutic efficacy. The review further explores the involvement of ECM-NPs in immunomodulation techniques, highlighting their impact on immune reactions. While facing obstacles related to membrane stability and manufacturing scalability, the review outlines forthcoming research directions focused on enhancing membrane performance. This review underscores the promise of ECM-NPs in surpassing conventional therapeutic constraints, proposing novel approaches for efficacious glioma treatment.


Subject(s)
Brain Neoplasms , Cell Membrane , Glioma , Nanoparticles , Tumor Microenvironment , Glioma/drug therapy , Glioma/therapy , Glioma/metabolism , Glioma/immunology , Glioma/pathology , Humans , Nanoparticles/chemistry , Cell Membrane/metabolism , Tumor Microenvironment/drug effects , Brain Neoplasms/drug therapy , Brain Neoplasms/pathology , Brain Neoplasms/therapy , Brain Neoplasms/metabolism , Brain Neoplasms/immunology , Animals , Blood-Brain Barrier/metabolism , Drug Delivery Systems/methods , Immunomodulation/drug effects
16.
Regen Biomater ; 11: rbae025, 2024.
Article in English | MEDLINE | ID: mdl-38605853

ABSTRACT

Wound repair is a complex physiological process that often leads to bacterial infections, which significantly threaten human health. Therefore, developing wound-healing materials that promote healing and prevent bacterial infections is crucial. In this study, the coordination interaction between sulfhydryl groups on dithiothreitol (DTT) and MoS2 nanosheets is investigated to synthesize a MoS2-DTT nanozyme with photothermal properties and an improved free-radical scavenging ability. Double-bond-modified hyaluronic acid is used as a monomer and is cross-linked with a PF127-DA agent. PHMoD is prepared in coordination with MoS2-DTT as the functional component. This hydrogel exhibits antioxidant and antibacterial properties, attributed to the catalytic activity of catalase-like enzymes and photothermal effects. Under the near-infrared (NIR), it exhibits potent antibacterial effects against gram-positive (Staphylococcus aureus) and gram-negative bacteria (Escherichia coli), achieving bactericidal rates of 99.76% and 99.42%, respectively. Furthermore, the hydrogel exhibits remarkable reactive oxygen species scavenging and antioxidant capabilities, effectively countering oxidative stress in L929 cells. Remarkably, in an animal model, wounds treated with the PHMoD(2.0) and NIR laser heal the fastest, sealing completely within 10 days. These results indicate the unique biocompatibility and bifunctionality of the PHMoD, which make it a promising material for wound-healing applications.

17.
Carbohydr Polym ; 334: 122011, 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38553212

ABSTRACT

Injectable hydrogels have wide applications in clinical practice. However, the development of tough and bioadhesive ones based on biopolymers, along with biofriendly and robust crosslinking strategies, still represents a great challenge. Herein, we report an injectable hydrogel composed of maleimidyl alginate and pristine gelatin, for which the precursor solutions could self-crosslink via mild Michael-type addition without any catalyst or external energy upon mixing. This hydrogel is tough and bioadhesive, which can maintain intactness as well as adherence to the defect of porcine skin under fierce bending and twisting, warm water bath, and boiling water shower. Besides, it is biocompatible, bioactive and biodegradable, which could support the growth and remodeling of cells by affording an extracellular matrix-like environment. As a proof of application, we demonstrate that this hydrogel could significantly accelerate diabetic skin wound healing, thereby holding great potential in healthcare.


Subject(s)
Biocompatible Materials , Gelatin , Animals , Swine , Biocompatible Materials/pharmacology , Hydrogels , Alginates , Water
18.
ACS Appl Mater Interfaces ; 16(7): 8484-8498, 2024 Feb 21.
Article in English | MEDLINE | ID: mdl-38334265

ABSTRACT

Malignant tumors are still one of the most deadly diseases that threaten human life and health. However, developing new drugs is challenging due to lengthy trials, funding constraints, and regulatory approval procedures. Consequently, researchers have devoted themselves to transforming some clinically approved old drugs into antitumor drugs with certain active ingredients, which have become an attractive alternative. Disulfiram (DSF), an antialcohol medication, can rapidly metabolize in the physiological environment into diethyldithiocarbamate (DTC) which can readily react with Cu2+ ions in situ to form the highly toxic bis(N,N-diethyldithiocarbamate)-copper(II) (CuET) complex. In this study, DSF is loaded into mesoporous dopamine nanocarriers and surface-chelated with tannin and Cu2+ to construct M-MDTC nanoprodrugs under the camouflage of K7 tumor cell membranes. After intravenous injection, M-MDTC nanoprodrugs successfully reach the tumor sites with the help of mediated cell membranes. Under slightly acidic pH and photothermal stimulation conditions, DSF and Cu2+ are simultaneously released, forming a highly toxic CuET to kill tumor cells in situ. The generated CuET can also induce immunogenic cell death of tumor cells, increase the proportion of CD86+ CD80+ cells, and promote dendritic cell maturation. In vitro and in vivo studies of M-MDTC nanoprodrugs have shown excellent tumor-cell-killing ability and solid tumor suppression. This approach enables in situ amplification of chemotherapy in the tumor microenvironment, achieving an effective antitumor treatment.


Subject(s)
Cadaverine/analogs & derivatives , Copper , Neoplasms , Humans , Cell Line, Tumor , Copper/pharmacology , Copper/therapeutic use , Tumor Microenvironment , Biomimetics , Disulfiram/pharmacology , Ditiocarb/pharmacology , Ditiocarb/therapeutic use , Neoplasms/drug therapy , Neoplasms/pathology
19.
Small ; 20(24): e2307628, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38191883

ABSTRACT

Injectable bioadhesives are attractive for managing gastric ulcers through minimally invasive procedures. However, the formidable challenge is to develop bioadhesives that exhibit high injectability, rapidly adhere to lesion tissues with fast gelation, provide reliable protection in the harsh gastric environment, and simultaneously ensure stringent standards of biocompatibility. Here, a natural bioadhesive with tunable cohesion is developed based on the facile and controllable gelation between silk fibroin and tannic acid. By incorporating a hydrogen bond disruptor (urea or guanidine hydrochloride), the inherent network within the bioadhesive is disturbed, inducing a transition to a fluidic state for smooth injection (injection force <5 N). Upon injection, the fluidic bioadhesive thoroughly wets tissues, while the rapid diffusion of the disruptor triggers instantaneous in situ gelation. This orchestrated process fosters the formed bioadhesive with durable wet tissue affinity and mechanical properties that harmonize with gastric tissues, thereby bestowing long-lasting protection for ulcer healing, as evidenced through in vitro and in vivo verification. Moreover, it can be conveniently stored (≥3 m) postdehydration. This work presents a promising strategy for designing highly injectable bioadhesives utilizing natural feedstocks, avoiding any safety risks associated with synthetic materials or nonphysiological gelation conditions, and offering the potential for minimally invasive application.


Subject(s)
Hydrogen Bonding , Stomach Ulcer , Animals , Stomach Ulcer/drug therapy , Injections , Tissue Adhesives/chemistry , Adhesives/chemistry , Fibroins/chemistry , Tannins/chemistry , Rats, Sprague-Dawley
20.
Macromol Biosci ; 24(4): e2300416, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38215472

ABSTRACT

Osteoporotic bone defects cannot withstand surgery with more significant trauma due to bone fragility, while systemic drug therapy has formidable adverse effects. Consequently, the present study introduces an innovatively devised injectable double-crosslinked hydrogel, as a potential therapeutic avenue for addressing varied shapes of osteoporotic bone defects via a minimally invasive approach. The injectable hydrogel is formed by the formation of Schiff base bonds between oxidized sodium alginate (OSA) and carboxymethyl chitosan, and the polymerization of gelatin methacrylate by UV light crosslinking. Additionally, alendronate sodium (ALN) is loaded into the hydrogel through Schiff base formation with OSA, and nanohydroxyapatite (nHA) is incorporated into the hydrogel via blending. The hydrogel demonstrates excellent injectability, and the nHA improves the mechanical properties of hydrogel and can promote bone formation. In addition, the hydrogel can sustain the release of ALN, which has the effect of inhibiting osteoclasts. Cell studies indicate that the hydrogel can promote the differentiation of osteoblasts and inhibit the activity of osteoclast, so as to obtain better osteogenic effect. Therefore, the injectable hydrogel can be used to repair osteoporotic bone defects through a minimally invasive, simple treatment modality.


Subject(s)
Osteogenesis , Osteoporosis , Humans , Osteoclasts , Hydrogels/pharmacology , Hydrogels/chemistry , Schiff Bases , Osteoporosis/drug therapy
SELECTION OF CITATIONS
SEARCH DETAIL