Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.527
Filter
1.
Carbohydr Polym ; 339: 122232, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-38823905

ABSTRACT

In this study, new types of hybrid double-network (DN) hydrogels composed of polyvinyl alcohol (PVA), chitosan (CH), and sodium alginate (SA) are introduced, with the hypothesis that this combination and incorporating multi-walled carbon nanotubes (MWCNTs) and graphene nanoplatelets (GNPs) will enhance osteogenetic differentiation and the structural and mechanical properties of scaffolds for bone tissue engineering applications. Initially, the impact of varying mass ratios of the PVA/CH/SA mixture on mechanical properties, swelling ratio, and degradability was examined. Based on this investigation, a mass ratio of 4:6:6 was determined to be optimal. At this ratio, the hydrogel demonstrated a Young's modulus of 47.5 ± 5 kPa, a swelling ratio of 680 ± 6 % after 3 h, and a degradation rate of 46.5 ± 5 % after 40 days. In the next phase, following the determination of the optimal mass ratio, CNTs and GNPs were incorporated into the 4:6:6 composite resulting in a significant enhancement in the electrical conductivity and stiffness of the scaffolds. The introduction of CNTs led to a notable increase of 36 % in the viability of MG63 osteoblast cells. Additionally, the inhibition zone test revealed that GNPs and CNTs increased the diameter of the inhibition zone by 49.6 % and 52.6 %, respectively.


Subject(s)
Alginates , Bone Regeneration , Chitosan , Hydrogels , Polyvinyl Alcohol , Tissue Engineering , Tissue Scaffolds , Chitosan/chemistry , Alginates/chemistry , Alginates/pharmacology , Polyvinyl Alcohol/chemistry , Tissue Scaffolds/chemistry , Humans , Bone Regeneration/drug effects , Hydrogels/chemistry , Hydrogels/pharmacology , Tissue Engineering/methods , Nanotubes, Carbon/chemistry , Osteoblasts/drug effects , Osteoblasts/cytology , Graphite/chemistry , Graphite/pharmacology , Biomimetic Materials/chemistry , Biomimetic Materials/pharmacology , Cell Survival/drug effects , Cell Line
2.
Drug Deliv ; 31(1): 2354687, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38823413

ABSTRACT

Hepatocellular carcinoma (HCC) is the fourth leading cause of cancer-associated death worldwide. Beside early detection, early diagnosis, and early surgery, it is urgent to try new strategies for the treatment of HCC. Triptolide (TPL) has been employed to treat HCC. However, its clinical applications were restricted by the narrow therapeutic window, severe toxicity, and poor water-solubility. In this study, we developed cancer cell membrane-camouflaged biomimetic PLGA nanoparticles loading TPL (TPL@mPLGA) with the homologous targeting property for the treatment of HCC. The TPL@mPLGA was successfully prepared with particle size of 195.5 ± 7.5 nm and zeta potential at -21.5 ± 0.2 mV with good stability. The drug loading (DL) of TPL@mPLGA was 2.94%. After Huh-7 cell membrane coating, the natural Huh-7 cell membrane proteins were found to be retained on TPL@mPLGA, thus endowing the TPL@mPLGA with enhanced accumulation at tumor site, and better anti-tumor activity in vitro and in vivo when compared with TPL or TPL@PLGA. The TPL@mPLGA showed enhanced anti-tumor effects and reduced toxicity of TPL, which could be adopted for the treatment of HCC.


Subject(s)
Carcinoma, Hepatocellular , Diterpenes , Epoxy Compounds , Liver Neoplasms , Nanoparticles , Phenanthrenes , Polylactic Acid-Polyglycolic Acid Copolymer , Diterpenes/administration & dosage , Diterpenes/pharmacology , Diterpenes/chemistry , Diterpenes/pharmacokinetics , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/pathology , Epoxy Compounds/chemistry , Epoxy Compounds/administration & dosage , Epoxy Compounds/pharmacology , Phenanthrenes/administration & dosage , Phenanthrenes/pharmacology , Phenanthrenes/chemistry , Phenanthrenes/pharmacokinetics , Liver Neoplasms/drug therapy , Liver Neoplasms/pathology , Humans , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Nanoparticles/chemistry , Animals , Cell Line, Tumor , Mice , Cell Membrane/drug effects , Particle Size , Drug Carriers/chemistry , Mice, Nude , Biomimetic Materials/chemistry , Biomimetic Materials/pharmacology , Mice, Inbred BALB C
3.
Int J Nanomedicine ; 19: 3957-3972, 2024.
Article in English | MEDLINE | ID: mdl-38711614

ABSTRACT

Purpose: Current treatment approaches for Prostate cancer (PCa) often come with debilitating side effects and limited therapeutic outcomes. There is urgent need for an alternative effective and safe treatment for PCa. Methods: We developed a nanoplatform to target prostate cancer cells based on graphdiyne (GDY) and a copper-based metal-organic framework (GDY-CuMOF), that carries the chemotherapy drug doxorubicin (DOX) for cancer treatment. Moreover, to provide GDY-CuMOF@DOX with homotypic targeting capability, we coated the PCa cell membrane (DU145 cell membrane, DCM) onto the surface of GDY-CuMOF@DOX, thus obtaining a biomimetic nanoplatform (DCM@GDY-CuMOF@DOX). The nanoplatform was characterized by using transmission electron microscope, atomic force microscope, X-ray diffraction, etc. Drug release behavior, antitumor effects in vivo and in vitro, and biosafety of the nanoplatform were evaluated. Results: We found that GDY-CuMOF exhibited a remarkable capability to load DOX mainly through π-conjugation and pore adsorption, and it responsively released DOX and generated Cu+ in the presence of glutathione (GSH). In vivo experiments demonstrated that this nanoplatform exhibits remarkable cell-killing efficiency by generating lethal reactive oxygen species (ROS) and mediating cuproptosis. In addition, DCM@GDY-CuMOF@DOX effectively suppresses tumor growth in vivo without causing any apparent side effects. Conclusion: The constructed DCM@GDY-CuMOF@DOX nanoplatform integrates tumor targeting, drug-responsive release and combination with cuproptosis and chemodynamic therapy, offering insights for further biomedical research on efficient PCa treatment.


Subject(s)
Copper , Doxorubicin , Graphite , Metal-Organic Frameworks , Prostatic Neoplasms , Male , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Doxorubicin/pharmacology , Doxorubicin/chemistry , Animals , Humans , Cell Line, Tumor , Copper/chemistry , Copper/pharmacology , Graphite/chemistry , Graphite/pharmacology , Metal-Organic Frameworks/chemistry , Metal-Organic Frameworks/pharmacology , Mice , Drug Liberation , Reactive Oxygen Species/metabolism , Biomimetic Materials/chemistry , Biomimetic Materials/pharmacology , Mice, Nude , Nanoparticles/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Drug Carriers/chemistry , Xenograft Model Antitumor Assays
4.
Molecules ; 29(9)2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38731407

ABSTRACT

The problem of bacterial resistance has become more and more common with improvements in health care. Worryingly, the misuse of antibiotics leads to an increase in bacterial multidrug resistance and the development of new antibiotics has virtually stalled. These challenges have prompted the need to combat bacterial infections with the use of radically different approaches. Taking lessons from the exciting properties of micro-/nano-natural-patterned surfaces, which can destroy cellular integrity, the construction of artificial surfaces to mimic natural functions provides new opportunities for the innovation and development of biomedicine. Due to the diversity of natural surfaces, functional surfaces inspired by natural surfaces have a wide range of applications in healthcare. Nature-inspired surface structures have emerged as an effective and durable strategy to prevent bacterial infection, opening a new way to alleviate the problem of bacterial drug resistance. The present situation of bactericidal and antifouling surfaces with natural and biomimetic micro-/nano-structures is briefly reviewed. In addition, these innovative nature-inspired methods are used to manufacture a variety of artificial surfaces to achieve extraordinary antibacterial properties. In particular, the physical antibacterial effect of nature-inspired surfaces and the functional mechanisms of chemical groups, small molecules, and ions are discussed, as well as the wide current and future applications of artificial biomimetic micro-/nano-surfaces. Current challenges and future development directions are also discussed at the end. In the future, controlling the use of micro-/nano-structures and their subsequent functions will lead to biomimetic surfaces offering great potential applications in biomedicine.


Subject(s)
Anti-Bacterial Agents , Nanostructures , Surface Properties , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Nanostructures/chemistry , Biomimetic Materials/chemistry , Biomimetic Materials/pharmacology , Humans , Bacteria/drug effects , Bacterial Infections/drug therapy , Bacterial Infections/prevention & control
5.
J Nanobiotechnology ; 22(1): 250, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38750519

ABSTRACT

The complexity of repairing large segment defects and eradicating residual tumor cell puts the osteosarcoma clinical management challenging. Current biomaterial design often overlooks the crucial role of precisely regulating innervation in bone regeneration. Here, we develop a Germanium Selenium (GeSe) co-doped polylactic acid (PLA) nanofiber membrane-coated tricalcium phosphate bioceramic scaffold (TCP-PLA/GeSe) that mimics the bone-periosteum structure. This biomimetic scaffold offers a dual functionality, combining piezoelectric and photothermal conversion capabilities while remaining biodegradable. When subjected to ultrasound irradiation, the US-electric stimulation of TCP-PLA/GeSe enables spatiotemporal control of neurogenic differentiation. This feature supports early innervation during bone formation, promoting early neurogenic differentiation of Schwann cells (SCs) by increasing intracellular Ca2+ and subsequently activating the PI3K-Akt and Ras signaling pathways. The biomimetic scaffold also demonstrates exceptional osteogenic differentiation potential under ultrasound irradiation. In rabbit model of large segment bone defects, the TCP-PLA/GeSe demonstrates promoted osteogenesis and nerve fibre ingrowth. The combined attributes of high photothermal conversion capacity and the sustained release of anti-tumor selenium from the TCP-PLA/GeSe enable the synergistic eradication of osteosarcoma both in vitro and in vivo. This strategy provides new insights on designing advanced biomaterials of repairing large segment bone defect and osteosarcoma.


Subject(s)
Bone Regeneration , Calcium Phosphates , Osteogenesis , Osteosarcoma , Tissue Scaffolds , Osteosarcoma/drug therapy , Osteosarcoma/pathology , Animals , Bone Regeneration/drug effects , Tissue Scaffolds/chemistry , Rabbits , Calcium Phosphates/chemistry , Calcium Phosphates/pharmacology , Osteogenesis/drug effects , Polyesters/chemistry , Humans , Cell Differentiation/drug effects , Bone Neoplasms/pathology , Bone Neoplasms/drug therapy , Bone Neoplasms/therapy , Cell Line, Tumor , Biomimetic Materials/chemistry , Biomimetic Materials/pharmacology , Schwann Cells/drug effects , Nanofibers/chemistry , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Selenium/chemistry , Selenium/pharmacology
6.
ACS Appl Mater Interfaces ; 16(19): 25101-25112, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38691046

ABSTRACT

The evolution of nano-drug delivery systems addresses the limitations of conventional cancer treatments with stimulus-responsive nanomaterial-based delivery systems presenting temporal and spatial advantages. Among various nanomaterials, boron nitride nanoparticles (BNNs) demonstrate significant potential in drug delivery and cancer treatment, providing a high drug loading capacity, multifunctionality, and low toxicity. However, the challenge lies in augmenting nanomaterial accumulation exclusively within tumors while preserving healthy tissues. To address this, we introduce a novel approach involving cancer cell membrane-functionalized BNNs (CM-BIDdT) for the codelivery of doxorubicin (Dox) and indocyanine green to treat homologous tumor. The cancer cell membrane biomimetic CM-BIDdT nanoparticles possess highly efficient homologous targeting capabilities toward tumor cells. The surface modification with acylated TAT peptides (dTAT) further enhances the nanoparticle intracellular accumulation. Consequently, CM-BIDdT nanoparticles, responsive to the acidic tumor microenvironment, hydrolyze amide bonds, activate the transmembrane penetrating function, and achieve precise targeting with substantial accumulation at the tumor site. Additionally, the photothermal effect of CM-BIDdT under laser irradiation not only kills cells through thermal ablation but also destroys the membrane on the surface of the nanoparticles, facilitating Dox release. Therefore, the fabricated CM-BIDdT nanoparticles orchestrate chemo-photothermal combination therapy and effectively inhibit tumor growth with minimal adverse effects, holding promise as a new modality for synergistic cancer treatment.


Subject(s)
Boron Compounds , Doxorubicin , Indocyanine Green , Nanoparticles , Doxorubicin/chemistry , Doxorubicin/pharmacology , Indocyanine Green/chemistry , Indocyanine Green/pharmacology , Boron Compounds/chemistry , Boron Compounds/pharmacology , Animals , Humans , Mice , Nanoparticles/chemistry , Cell Line, Tumor , Photothermal Therapy , Biomimetic Materials/chemistry , Biomimetic Materials/pharmacology , Neoplasms/drug therapy , Neoplasms/pathology , Neoplasms/therapy , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , tat Gene Products, Human Immunodeficiency Virus/chemistry , Mice, Inbred BALB C , Drug Carriers/chemistry , Drug Delivery Systems
7.
Carbohydr Polym ; 338: 122204, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-38763712

ABSTRACT

This study presents the development and characterization of a novel double-network self-healing hydrogel based on N-carboxyethyl chitosan (CEC) and oxidized dextran (OD) with the incorporation of crosslinked collagen (CEC-OD/COL-GP) to enhance its biological and physicochemical properties. The hydrogel formed via dynamic imine bond formation exhibited efficient self-healing within 30 min, and a compressive modulus recovery of 92 % within 2 h. In addition to its self-healing ability, CEC-OD/COL-GP possesses unique physicochemical characteristics including transparency, injectability, and adhesiveness to various substrates and tissues. Cell encapsulation studies confirmed the biocompatibility and suitability of the hydrogel as a cell-culture scaffold, with the presence of a collagen network that enhances cell adhesion, spreading, long-term cell viability, and proliferation. Leveraging their unique properties, we engineered assemblies of self-healing hydrogel modules for controlled spatiotemporal drug delivery and constructed co-culture models that simulate angiogenesis in tumor microenvironments. Overall, the CEC-OD/COL-GP hydrogel is a versatile and promising material for biomedical applications, offering a bottom-up approach for constructing complex structures with self-healing capabilities, controlled drug release, and support for diverse cell types in 3D environments. This hydrogel platform has considerable potential for advancements in tissue engineering and therapeutic interventions.


Subject(s)
Cell Adhesion , Chitosan , Dextrans , Hydrogels , Hydrogels/chemistry , Hydrogels/pharmacology , Chitosan/chemistry , Dextrans/chemistry , Humans , Cell Adhesion/drug effects , Cell Survival/drug effects , Collagen/chemistry , Animals , Drug Liberation , Cell Proliferation/drug effects , Cell Encapsulation/methods , Biomimetic Materials/chemistry , Biomimetic Materials/pharmacology , Mice , Biomimetics/methods , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Tissue Scaffolds/chemistry
8.
Acta Biomater ; 181: 391-401, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38704114

ABSTRACT

Potassium ion transport across myocardial cell membrane is essential for type 2 long QT syndrome (LQT2). However, the dysfunction of potassium ion transport due to genetic mutations limits the therapeutic effect in treating LQT2. Biomimetic ion channels that selectively and efficiently transport potassium ions across the cellular membranes are promising for the treatment of LQT2. To corroborate this, we synthesized a series of foldamer-based ion channels with different side chains, and found a biomimetic ion channel of K+ (BICK) with the highest transport activity among them. The selected BICK can restore potassium ion transport and increase transmembrane potassium ion current, thus shortening phase 3 of action potential (AP) repolarization and QT interval in LQT2. Moreover, BICK does not affect heart rate and cardiac rhythm in treating LQT2 model induced by E4031 in isolated heart as well as in guinea pigs. By restoring ion transmembrane transport tactic, biomimetic ion channels, such as BICK, will show great potential in treating diseases related to ion transport blockade. STATEMENT OF SIGNIFICANCE: Type 2 long QT syndrome (LQT2) is a disease caused by K+ transport disorder, which can cause malignant arrhythmia and even death. There is currently no radical cure, so it is critical to explore ways to improve K+ transmembrane transport. In this study, we report that a small-molecule biomimetic ion channel BICK can efficiently simulate natural K+ channel proteins on the cardiomyocyte and cure E4031-induced LQT2 in guinea pig by restoring K+ transport function for the first time. This study found that the potassium transmembrane transport by BICK significantly reduced the QT interval, which provides a conceptually new strategy for the treatment of LQT2 disease.


Subject(s)
Long QT Syndrome , Potassium , Long QT Syndrome/metabolism , Animals , Potassium/metabolism , Guinea Pigs , Humans , Action Potentials/drug effects , Ion Transport/drug effects , Male , Biomimetic Materials/chemistry , Biomimetic Materials/pharmacology , Potassium Channels/metabolism , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Heart Rate/drug effects
9.
ACS Appl Bio Mater ; 7(5): 2862-2871, 2024 May 20.
Article in English | MEDLINE | ID: mdl-38699864

ABSTRACT

Mosquito-borne viruses are a major worldwide health problem associated with high morbidity and mortality rates and significant impacts on national healthcare budgets. The development of antiviral drugs for both the treatment and prophylaxis of these diseases is thus of considerable importance. To address the need for therapeutics with antiviral activity, a library of heparan sulfate mimetic polymers was screened against dengue virus (DENV), Yellow fever virus (YFV), Zika virus (ZIKV), and Ross River virus (RRV). The polymers were prepared by RAFT polymerization of various acidic monomers with a target MW of 20 kDa (average Mn ∼ 27 kDa by GPC). Among the polymers, poly(SS), a homopolymer of sodium styrenesulfonate, was identified as a broad spectrum antiviral with activity against all the tested viruses and particularly potent inhibition of YFV (IC50 = 310 pM). Our results further uncovered that poly(SS) exhibited a robust inhibition of ZIKV infection in both mosquito and human cell lines, which points out the potential functions of poly(SS) in preventing mosquito-borne viruses associated diseases by blocking viral transmission in their mosquito vectors and mitigating viral infection in patients.


Subject(s)
Antiviral Agents , Heparitin Sulfate , Polymers , Antiviral Agents/pharmacology , Antiviral Agents/chemistry , Antiviral Agents/chemical synthesis , Heparitin Sulfate/chemistry , Heparitin Sulfate/pharmacology , Animals , Humans , Polymers/chemistry , Polymers/pharmacology , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Biocompatible Materials/chemical synthesis , Culicidae/drug effects , Culicidae/virology , Microbial Sensitivity Tests , Materials Testing , Particle Size , Cell Line , Molecular Structure , Chlorocebus aethiops , Biomimetic Materials/chemistry , Biomimetic Materials/pharmacology , Zika Virus/drug effects
10.
J Nanobiotechnology ; 22(1): 263, 2024 May 17.
Article in English | MEDLINE | ID: mdl-38760755

ABSTRACT

The prevalence of cardiovascular diseases continues to be a challenge for global health, necessitating innovative solutions. The potential of high-density lipoprotein (HDL) mimetic nanotherapeutics in the context of cardiovascular disease and the intricate mechanisms underlying the interactions between monocyte-derived cells and HDL mimetic showing their impact on inflammation, cellular lipid metabolism, and the progression of atherosclerotic plaque. Preclinical studies have demonstrated that HDL mimetic nanotherapeutics can regulate monocyte recruitment and macrophage polarization towards an anti-inflammatory phenotype, suggesting their potential to impede the progression of atherosclerosis. The challenges and opportunities associated with the clinical application of HDL mimetic nanotherapeutics, emphasize the need for additional research to gain a better understanding of the precise molecular pathways and long-term effects of these nanotherapeutics on monocytes and macrophages to maximize their therapeutic efficacy. Furthermore, the use of nanotechnology in the treatment of cardiovascular diseases highlights the potential of nanoparticles for targeted treatments. Moreover, the concept of theranostics combines therapy and diagnosis to create a selective platform for the conversion of traditional therapeutic medications into specialized and customized treatments. The multifaceted contributions of HDL to cardiovascular and metabolic health via highlight its potential to improve plaque stability and avert atherosclerosis-related problems. There is a need for further research to maximize the therapeutic efficacy of HDL mimetic nanotherapeutics and to develop targeted treatment approaches to prevent atherosclerosis. This review provides a comprehensive overview of the potential of nanotherapeutics in the treatment of cardiovascular diseases, emphasizing the need for innovative solutions to address the challenges posed by cardiovascular diseases.


Subject(s)
Cardiovascular Diseases , Lipoproteins, HDL , Macrophages , Monocytes , Humans , Lipoproteins, HDL/chemistry , Lipoproteins, HDL/metabolism , Lipoproteins, HDL/pharmacology , Macrophages/drug effects , Macrophages/metabolism , Animals , Cardiovascular Diseases/drug therapy , Monocytes/drug effects , Nanoparticles/chemistry , Atherosclerosis/drug therapy , Plaque, Atherosclerotic/drug therapy , Nanomedicine/methods , Biomimetic Materials/chemistry , Biomimetic Materials/pharmacology
11.
Int J Biol Macromol ; 270(Pt 1): 132359, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38754678

ABSTRACT

The objective of this study was to evaluate the synergistic effect of eggshell-derived nanohydroxyapatite (EnHA) and carboxymethyl chitosan (CMC) in remineralizing artificially induced dentinal lesions. EnHA and CMC were synthesized using simple chemical processes and characterized using FTIR, XRD, HRSEM-EDX, TEM, DLS and TGA/DTA analyses. A total of 64 pre-demineralized coronal dentin specimens were randomly subjected to following treatments (n = 16):artificial saliva (AS), EnHA, CMC, and EnHA-CMC, followed by pH cycling for 7 days. HRSEM-EDX, Vickers-indenter, and micro-Raman analyses were used to assess surface-topography, microhardness, and chemical analysis, respectively. All tested materials demonstrated non-cytotoxicity when assessed on hDPSCs using MTT assay. FTIR, XRD and thermal analyses confirmed the characteristics of both EnHA and CMC. EnHA showed irregular rod-shaped nanoparticles (30-70 nm) with the presence of Ca,P,Na, and Mg ions. Dentin treated with EnHA-CMC exhibited complete tubular occlusion and highest microhardness whereas the AS group revealed the least mineral deposits (p < 0.05). No significant differences were observed between EnHA and CMC groups (p > 0.05). In addition, molecular conformation analysis revealed peak intensities in collagen's polypeptide chains in dentin treated with CMC and EnHA-CMC, whereas other groups showed poor collagen stability. The results highlighted that EnHA-CMC aided in rapid and effective biomineralization, suggesting its potential as a therapeutic solution for treating dentin caries.


Subject(s)
Chitosan , Dentin , Durapatite , Egg Shell , Chitosan/analogs & derivatives , Chitosan/chemistry , Chitosan/pharmacology , Durapatite/chemistry , Durapatite/pharmacology , Dentin/chemistry , Dentin/drug effects , Egg Shell/chemistry , Animals , Humans , Tooth Remineralization/methods , Nanoparticles/chemistry , Biomimetic Materials/pharmacology , Biomimetic Materials/chemistry , Hydrogen-Ion Concentration
12.
ACS Appl Mater Interfaces ; 16(21): 27187-27201, 2024 May 29.
Article in English | MEDLINE | ID: mdl-38747985

ABSTRACT

Development of theranostic nanomedicines to tackle glioma remains to be challenging. Here, we present an advanced blood-brain barrier (BBB)-crossing nanovaccine based on cancer cell membrane-camouflaged poly(N-vinylcaprolactam) (PVCL) nanogels (NGs) incorporated with MnO2 and doxorubicin (DOX). We show that the disulfide bond-cross-linked redox-responsive PVCL NGs can be functionalized with dermorphin and imiquimod R837 through cell membrane functionalization. The formed functionalized PVCL NGs having a size of 220 nm are stable, can deplete glutathione, and responsively release both Mn2+ and DOX under the simulated tumor microenvironment to exert the chemo/chemodynamic therapy mediated by DOX and Mn2+, respectively. The combined therapy induces tumor immunogenic cell death to maturate dendritic cells (DCs) and activate tumor-killing T cells. Further, the nanovaccine composed of cancer cell membranes as tumor antigens, R837 as an adjuvant with abilities of DC maturation and macrophages M1 repolarization, and MnO2 with Mn2+-mediated stimulator of interferon gene activation of tumor cells can effectively act on both targets of tumor cells and immune cells. With the dermorphin-mediated BBB crossing, cell membrane-mediated homologous tumor targeting, and Mn2+-facilitated magnetic resonance (MR) imaging property, the designed NG-based theranostic nanovaccine enables MR imaging and combination chemo-, chemodynamic-, and imnune therapy of orthotopic glioma with a significantly decreased recurrence rate.


Subject(s)
Glioma , Magnetic Resonance Imaging , Manganese Compounds , Theranostic Nanomedicine , Glioma/diagnostic imaging , Glioma/drug therapy , Glioma/therapy , Glioma/pathology , Animals , Mice , Humans , Manganese Compounds/chemistry , Manganese Compounds/pharmacology , Doxorubicin/chemistry , Doxorubicin/pharmacology , Doxorubicin/therapeutic use , Cancer Vaccines/chemistry , Immunotherapy , Oxides/chemistry , Oxides/pharmacology , Cell Line, Tumor , Brain Neoplasms/diagnostic imaging , Brain Neoplasms/drug therapy , Brain Neoplasms/therapy , Brain Neoplasms/pathology , Biomimetic Materials/chemistry , Biomimetic Materials/pharmacology , Blood-Brain Barrier/metabolism , Nanogels/chemistry , Imiquimod/chemistry , Imiquimod/pharmacology , Nanovaccines
13.
ACS Nano ; 18(22): 14726-14741, 2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38778025

ABSTRACT

The management of diabetic wound healing remains a severe clinical challenge due to the complicated wound microenvironments, including abnormal immune regulation, excessive reactive oxygen species (ROS), and repeated bacterial infections. Herein, we report an extracellular matrix (ECM)-mimetic coating derived from scallop byssal protein (Sbp9Δ), which can be assembled in situ within 30 min under the trigger of Ca2+ driven by strong coordination interaction. The biocompatible Sbp9Δ coating and genetically programmable LL37-fused coating exhibit outstanding antioxidant, antibacterial, and immune regulatory properties in vitro. Proof-of-concept applications demonstrate that the coating can reliably promote wound healing in animal models, including diabetic mice and rabbits, ex vivo human skins, and Staphylococcus aureus-infected diabetic mice. In-depth mechanism investigation indicates that improved wound microenvironments accelerated wound repair, including alleviated bacterial infection, lessened inflammation, appearance of abundant M2-type macrophages, removal of ROS, promoted angiogenesis, and re-epithelialization. Collectively, our investigation provides an in situ, convenient, and effective approach for diabetic wound repair.


Subject(s)
Extracellular Matrix , Wound Healing , Animals , Wound Healing/drug effects , Mice , Rabbits , Extracellular Matrix/metabolism , Extracellular Matrix/drug effects , Extracellular Matrix/chemistry , Humans , Diabetes Mellitus, Experimental , Staphylococcus aureus/drug effects , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Biomimetic Materials/chemistry , Biomimetic Materials/pharmacology , Reactive Oxygen Species/metabolism , Coated Materials, Biocompatible/chemistry , Coated Materials, Biocompatible/pharmacology
14.
Int J Biol Macromol ; 269(Pt 2): 132124, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38723802

ABSTRACT

Bacterial cellulose (BC) hydrogel is renowned in the field of tissue engineering for its high biocompatibility, excellent mechanical strength, and eco-friendliness. Herein, we present a biomimetic mineralization method for preparing BC/hydroxyapatite (HAP) composite hydrogel scaffolds with different mineralization time and ion concentration of the mineralized solution. Spherical HAP reinforcement enhanced bone mineralization, thereby imparting increased bioactivity to BC matrix materials. Subsequently, platelet-rich plasma (PRP) was introduced into the scaffold. The PRP-loaded hydrogel enhanced the release of growth factors, which promoted cell adhesion, growth, and bone healing. After 3 weeks of MC3T3-E1 cell-induced osteogenesis, PRP positively affected cell differentiation in BC/HAP@PRP scaffolds. Overall, these scaffolds exhibited excellent biocompatibility, mineralized nodule formation, and controlled release in vitro, demonstrating great potential for application in bone tissue repair.


Subject(s)
Cellulose , Durapatite , Hydrogels , Osteogenesis , Platelet-Rich Plasma , Tissue Engineering , Tissue Scaffolds , Platelet-Rich Plasma/chemistry , Tissue Engineering/methods , Durapatite/chemistry , Durapatite/pharmacology , Cellulose/chemistry , Cellulose/pharmacology , Animals , Mice , Tissue Scaffolds/chemistry , Hydrogels/chemistry , Osteogenesis/drug effects , Bone and Bones/drug effects , Bone and Bones/metabolism , Delayed-Action Preparations/pharmacology , Cell Differentiation/drug effects , Biomimetics/methods , Biomimetic Materials/chemistry , Biomimetic Materials/pharmacology , Cell Line , Bone Regeneration/drug effects
15.
Carbohydr Polym ; 336: 122124, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-38670755

ABSTRACT

Sulfated polysaccharides play important roles in tissue engineering applications because of their high growth factor preservation ability and their native-like biological features. There are different sulfated polysaccharides based on different repeating units in the carbohydrate backbone, the position of the sulfate group, and the sulfation degree of the polysaccharide. These led to various sulfated polymers with different negative charge densities and resultant structure-property relationships. Since numerous reports are presented related to sulfated polysaccharide applications in tissue engineering, it is crucial to review the role of effective physicochemical and biological parameters in their usage; as well as their structure-property relationships. Within this review, we focused on the effect of naturally occurring and synthetic sulfated polysaccharides in tissue engineering applications reported in the last years, highlighting the challenges of the scaffold fabrication process, the position, and the degree of sulfate on biomedical activity. Additionally, we discussed their use in numerous in vitro and in vivo model systems.


Subject(s)
Biomimetic Materials , Polysaccharides , Sulfates , Tissue Engineering , Tissue Scaffolds , Tissue Engineering/methods , Polysaccharides/chemistry , Polysaccharides/pharmacology , Tissue Scaffolds/chemistry , Humans , Animals , Sulfates/chemistry , Biomimetic Materials/chemistry , Biomimetic Materials/pharmacology , Biopolymers/chemistry , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology
16.
Acta Biomater ; 180: 183-196, 2024 May.
Article in English | MEDLINE | ID: mdl-38604465

ABSTRACT

The utilization of biodegradable magnesium (Mg) alloys in the fabrication of temporary non-vascular stents is an innovative trend in biomedical engineering. However, the heterogeneous degradation profiles of these biomaterials, together with potential bacterial colonization that could precipitate infectious or stenotic complications, are critical obstacles precluding their widespread clinical application. In pursuit of overcoming these limitations, this study applies the principles of biomimicry, particularly the hydrophobic and anti-fouling characteristics of lotus leaves, to pioneer the creation of nanocomposite coatings. These coatings integrate poly-trimethylene carbonate (PTMC) with covalent organic frameworks (COFs), to modify the stent's surface property. The strategic design of the coating's topography, porosity, and self-polishing capabilities collectively aims to decelerate degradation processes and minimize biological adhesion. The protective qualities of the coatings were substantiated through rigorous testing in both in vitro dynamic bile tests and in vivo New Zealand rabbit choledochal models. Empirical findings from these trials confirmed that the implementation of COF-based nanocomposite coatings robustly fortifies Mg implantations, conferring heightened resistance to both biocorrosion and biofouling as well as improved biocompatibility within bodily environments. The outcomes of this research elucidate a comprehensive framework for the multifaceted strategies against stent corrosion and fouling, thereby charting a visionary pathway toward the systematic conception of a new class of reliable COF-derived surface modifications poised to amplify the efficacy of Mg-based stents. STATEMENT OF SIGNIFICANCE: Biodegradable magnesium (Mg) alloys are widely utilized in temporary stents, though their rapid degradation and susceptibility to bacterial infection pose significant challenges. Our research has developed a nanocomposite coating inspired by the lotus, integrating poly-trimethylene carbonate with covalent organic frameworks (COF). The coating achieved self-polishing property and optimal surface energy on the Mg substrate, which decelerates stent degradation and reduces biofilm formation. Comprehensive evaluations utilizing dynamic bile simulations and implantation in New Zealand rabbit choledochal models reveal that the coating improves the durability and longevity of the stent. The implications of these findings suggest the potential COF-based Mg alloy stent surface treatments and a leap forward in advancing stent performance and endurance in clinical applications.


Subject(s)
Absorbable Implants , Coated Materials, Biocompatible , Magnesium , Nanocomposites , Stents , Animals , Rabbits , Magnesium/chemistry , Magnesium/pharmacology , Nanocomposites/chemistry , Corrosion , Coated Materials, Biocompatible/chemistry , Coated Materials, Biocompatible/pharmacology , Metal-Organic Frameworks/chemistry , Metal-Organic Frameworks/pharmacology , Biofouling/prevention & control , Dioxanes/chemistry , Biomimetic Materials/chemistry , Biomimetic Materials/pharmacology , Polymers/chemistry , Polymers/pharmacology , Alloys/chemistry , Alloys/pharmacology
17.
J Colloid Interface Sci ; 667: 624-639, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38663278

ABSTRACT

Quick scarless healing remains a key issue for diabetic wounds. Here, a stretchable elastomeric hydrogel dressing composed of hydroxyethylcellulose (HEC), silk nano fiber-magnesium ion complex (Mg2+-SNF) and glycerol (Gly) was developed to optimize mechanical niche, anti-inflammatory and angiogenic behavior simultaneously. The composite hydrogel dressing exhibited skin-like elasticity (175.1 ± 23.9 %) and modulus (156.7 ± 2.5 KPa) while Mg2+-SNF complex endowed the dressing with angiogenesis, both favoring quick scarless skin regeneration. In vitro cell studies revealed that the hydrogel dressing stimulated fibroblast proliferation, endothelial cell migration and vessel-like tube formation, and also induced anti-inflammatory behavior of macrophages. In vivo results revealed accelerated healing of diabetic wounds. The improved granulation ingrowth and collagen deposition suggested high quality repair. Both thinner epidermal layer and low collagen I/III ratio of the regenerated skin confirmed scarless tissue formation. This bioactive hydrogel dressing has promising potential to address the multifaceted challenges of diabetic wound management.


Subject(s)
Glycerol , Magnesium , Wound Healing , Wound Healing/drug effects , Animals , Glycerol/chemistry , Glycerol/pharmacology , Magnesium/chemistry , Magnesium/pharmacology , Mice , Silk/chemistry , Hydrogels/chemistry , Hydrogels/pharmacology , Biomimetic Materials/chemistry , Biomimetic Materials/pharmacology , Diabetes Mellitus, Experimental/drug therapy , Bandages , Humans , Rats , Nanofibers/chemistry , Cell Proliferation/drug effects , Neovascularization, Physiologic/drug effects , Male , Human Umbilical Vein Endothelial Cells , Cellulose/chemistry , Cellulose/pharmacology , Cellulose/analogs & derivatives
18.
Colloids Surf B Biointerfaces ; 238: 113909, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38599076

ABSTRACT

Nasopharyngeal carcinoma (NPC) is a common head and neck malignancy, which is characterized by high incidence and aggression with poor diagnosis and limited therapeutic opportunity. The innovative strategy for achieving precise NPC active-targeting drug delivery has emerged as a prominent focus in clinical research. Here, a minimalist cancer cell membrane (CCM) shielded biomimetic nanoparticle (NP) was designed for NPC active-targeting therapy. Chemotherapeutant model drug doxorubicin (DOX) was loaded in polyamidoamine (PAMAM) dendrimer. The PAMAM/DOX (PD) NP was further shielded by human CNE-2 NPC CCM. Characterization results verified that the biomimetic PAMAM/DOX@CCM (abbreviated as PDC) NPs had satisfactory physical properties with high DOX-loading and excellent stability. Cell experiments demonstrated that the CNE-2 membrane-cloaked PDC NPs presented powerful cellular uptake in the sourcing cells by homologous targeting and adhesive interaction. Further in vivo results confirmed that this biomimetic nanoplatform had extended circulation and remarkable tumor-targeting capability, and the PDC NPs effectively suppressed the progression of CNE-2 tumors by systemic administration. This CCM-shielded biomimetic NP displayed a minimalist paradigm nanoplatform for precise NPC therapy, and the strategy of CCM-shielded biomimetic drug delivery system (DDS) has great potential for extensive cancer active-targeting therapy.


Subject(s)
Biomimetic Materials , Cell Membrane , Doxorubicin , Nanoparticles , Nasopharyngeal Carcinoma , Nasopharyngeal Neoplasms , Humans , Doxorubicin/pharmacology , Doxorubicin/chemistry , Nanoparticles/chemistry , Nasopharyngeal Carcinoma/drug therapy , Nasopharyngeal Carcinoma/pathology , Biomimetic Materials/chemistry , Biomimetic Materials/pharmacology , Cell Membrane/chemistry , Cell Membrane/metabolism , Cell Membrane/drug effects , Animals , Nasopharyngeal Neoplasms/drug therapy , Nasopharyngeal Neoplasms/pathology , Dendrimers/chemistry , Mice , Cell Line, Tumor , Drug Delivery Systems , Antibiotics, Antineoplastic/pharmacology , Antibiotics, Antineoplastic/chemistry , Antibiotics, Antineoplastic/administration & dosage , Cell Proliferation/drug effects , Mice, Nude , Mice, Inbred BALB C , Biomimetics , Particle Size
19.
ACS Nano ; 18(18): 11753-11768, 2024 May 07.
Article in English | MEDLINE | ID: mdl-38649866

ABSTRACT

The association between dysfunctional microglia and amyloid-ß (Aß) is a fundamental pathological event and increases the speed of Alzheimer's disease (AD). Additionally, the pathogenesis of AD is intricate and a single drug may not be enough to achieve a satisfactory therapeutic outcome. Herein, we reported a facile and effective gene therapy strategy for the modulation of microglia function and intervention of Aß anabolism by ROS-responsive biomimetic exosome-liposome hybrid nanovesicles (designated as TSEL). The biomimetic nanovesicles codelivery ß-site amyloid precursor protein cleaving enzyme-1 (BACE1) siRNA (siBACE1) and TREM2 plasmid (pTREM2) gene drug efficiently penetrate the blood-brain barrier and enhance the drug accumulation at AD lesions with the help of exosomes homing ability and angiopep-2 peptides. Specifically, an upregulation of TREM2 expression can reprogram microglia from a pro-inflammatory M1 phenotype to an anti-inflammatory M2 phenotype while also restoring its capacity to phagocytose Aß and its nerve repair function. In addition, siRNA reduces the production of Aß plaques at the source by knocking out the BACE1 gene, which is expected to further enhance the therapeutic effect of AD. The in vivo study suggests that TSEL through the synergistic effect of two gene drugs can ameliorate APP/PS1 mice cognitive impairment by regulating the activated microglial phenotype, reducing the accumulation of Aß, and preventing the retriggering of neuroinflammation. This strategy employs biomimetic nanovesicles for the delivery of dual nucleic acids, achieving synergistic gene therapy for AD, thus offering more options for the treatment of AD.


Subject(s)
Alzheimer Disease , Amyloid Precursor Protein Secretases , Aspartic Acid Endopeptidases , Biomimetic Materials , Genetic Therapy , Alzheimer Disease/therapy , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Alzheimer Disease/metabolism , Animals , Amyloid Precursor Protein Secretases/metabolism , Amyloid Precursor Protein Secretases/genetics , Mice , Biomimetic Materials/chemistry , Biomimetic Materials/pharmacology , Aspartic Acid Endopeptidases/genetics , Aspartic Acid Endopeptidases/metabolism , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/chemistry , Gene Transfer Techniques , Microglia/metabolism , Microglia/drug effects , Microglia/pathology , RNA, Small Interfering/chemistry , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacology , Humans , Liposomes/chemistry , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Biomimetics , Exosomes/metabolism , Exosomes/chemistry , Receptors, Immunologic/metabolism , Receptors, Immunologic/genetics
20.
Nanoscale ; 16(18): 8708-8738, 2024 May 09.
Article in English | MEDLINE | ID: mdl-38634521

ABSTRACT

Cancer immunotherapy, a burgeoning modality for cancer treatment, operates by activating the autoimmune system to impede the growth of malignant cells. Although numerous immunotherapy strategies have been employed in clinical cancer therapy, the resistance of cancer cells to immunotherapeutic medications and other apprehensions impede the attainment of sustained advantages for most patients. Recent advancements in nanotechnology for drug delivery hold promise in augmenting the efficacy of immunotherapy. However, the efficacy is currently constrained by the inadequate specificity of delivery, low rate of response, and the intricate immunosuppressive tumor microenvironment. In this context, the investigation of cell membrane coated nanoparticles (CMNPs) has revealed their ability to perform targeted delivery, immune evasion, controlled release, and immunomodulation. By combining the advantageous features of natural cell membranes and nanoparticles, CMNPs have demonstrated their unique potential in the realm of cancer immunotherapy. This review aims to emphasize recent research progress and elucidate the underlying mechanisms of CMNPs as an innovative drug delivery platform for enhancing cancer immunotherapy. Additionally, it provides a comprehensive overview of the current immunotherapeutic strategies involving different cell membrane types of CMNPs, with the intention of further exploration and optimization.


Subject(s)
Cell Membrane , Immunotherapy , Nanoparticles , Neoplasms , Humans , Neoplasms/therapy , Neoplasms/drug therapy , Neoplasms/immunology , Cell Membrane/metabolism , Cell Membrane/chemistry , Nanoparticles/chemistry , Drug Delivery Systems , Biomimetic Materials/chemistry , Biomimetic Materials/pharmacology , Animals , Tumor Microenvironment/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...