Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31.389
Filter
1.
PLoS One ; 19(8): e0308207, 2024.
Article in English | MEDLINE | ID: mdl-39110684

ABSTRACT

Neurofibromatosis Type 1 (NF1) is a complex genetic disorder characterized by the development of benign neurofibromas, which can cause significant morbidity in affected individuals. While the molecular mechanisms underlying NF1 pathogenesis have been extensively studied, the development of effective therapeutic strategies remains a challenge. This paper presents the development and validation of a novel biomaterial testing model to enhance our understanding of NF1 pathophysiology, disease mechanisms and evaluate potential therapeutic interventions. Our long-term goal is to develop an invitro model of NF1 to evaluate drug targets. We have developed an in vitro system to test the cellular behavior of NF1 patient derived cells on electroconductive aligned nanofibrous biomaterials with electrical stimulatory cues. We hypothesized that cells cultured on electroconductive biomaterial will undergo morphological changes and variations in cell proliferation that could be further enhanced with the combination of exogenous electrical stimulation (ES). In this study, we developed electrospun Hyaluronic Acid-Carbon Nanotube (HA-CNT) nanofiber scaffolds to mimic the axon's topographical and bioelectrical cues that influence neurofibroma growth and development. The cellular behavior was qualitatively and quantitively analyzed through immunofluorescent stains, Alamar blue assays and ELISA assays. Schwann cells from NF1 patients appear to have lost their ability to respond to electrical stimulation in the development and regeneration range, which was seen through changes in morphology, proliferation and NGF release. Without stimulation, the conductive material enhances NF1 SC behavior. Wild-type SC respond to electrical stimulation with increased cell proliferation and NGF release. Using this system, we can better understand the interaction between axons and SC that lead to tumor formation, homeostasis and regeneration.


Subject(s)
Cell Proliferation , Electric Stimulation , Hyaluronic Acid , Nanotubes, Carbon , Schwann Cells , Schwann Cells/metabolism , Nanotubes, Carbon/chemistry , Humans , Hyaluronic Acid/chemistry , Nanofibers/chemistry , Neurofibromatosis 1/pathology , Neurofibromatosis 1/metabolism , Tissue Scaffolds/chemistry , Cells, Cultured , Biocompatible Materials/chemistry
2.
Theranostics ; 14(11): 4438-4461, 2024.
Article in English | MEDLINE | ID: mdl-39113795

ABSTRACT

The high incidence of bone defect-related diseases caused by trauma, infection, and tumor resection has greatly stimulated research in the field of bone regeneration. Generally, bone healing is a long and complicated process wherein manipulating the biological activity of interventional scaffolds to support long-term bone regeneration is significant for treating bone-related diseases. It has been reported that some physical cues can act as growth factor substitutes to promote osteogenesis through continuous activation of endogenous signaling pathways. This review focuses on the latest progress in bone repair by remote actuation and on-demand activation of biomaterials pre-incorporated with physical cues (heat, electricity, and magnetism). As an alternative method to treat bone defects, physical cues show many advantages, including effectiveness, noninvasiveness, and remote manipulation. First, we introduce the impact of different physical cues on bone repair and potential internal regulatory mechanisms. Subsequently, biomaterials that mediate various physical cues in bone repair and their respective characteristics are summarized. Additionally, challenges are discussed, aiming to provide new insights and suggestions for developing intelligent biomaterials to treat bone defects and promote clinical translation.


Subject(s)
Biocompatible Materials , Bone Regeneration , Tissue Scaffolds , Bone Regeneration/drug effects , Biocompatible Materials/chemistry , Humans , Animals , Tissue Scaffolds/chemistry , Osteogenesis/drug effects , Tissue Engineering/methods , Bone and Bones/metabolism
3.
Clin Oral Investig ; 28(9): 476, 2024 Aug 09.
Article in English | MEDLINE | ID: mdl-39120764

ABSTRACT

OBJECTIVES: To synthesize casein enzymatic hydrolysate (CEH)-laden gelatin methacryloyl (GelMA) fibrous scaffolds and evaluate the cytocompatibility and anti-inflammatory effects on dental pulp stem cells (DPSCs). MATERIALS AND METHODS: GelMA fibrous scaffolds with 10%, 20%, and 30% CEH (w/w) and without CEH (control) were obtained via electrospinning. Chemo-morphological, degradation, and mechanical analyses were conducted to evaluate the morphology and composition of the fibers, mass loss, and mechanical properties, respectively. Adhesion/spreading and viability of DPSCs seeded on the scaffolds were also assessed. The anti-inflammatory potential on DPSCs was tested after the chronic challenge of cells with lipopolysaccharides (LPS), followed by treatment with extracts obtained after immersing the scaffolds in α-MEM. The synthesis of the pro-inflammatory cytokines IL-6, IL-1α, and TNF-α was measured by ELISA. Data were analyzed by ANOVA/post-hoc tests (α = 5%). RESULTS: CEH-laden electrospun fibers had a larger diameter than pure GelMA (p ≤ 0.036). GelMA scaffolds laden with 20% and 30% CEH had a greater mass loss. Tensile strength was reduced for the 10% CEH fibers (p = 0.0052), whereas no difference was observed for the 20% and 30% fibers (p ≥ 0.6736) compared to the control. Young's modulus decreased with CEH (p < 0.0001). Elongation at break increased for the 20% and 30% CEH scaffolds (p ≤ 0.0038). Over time, DPSCs viability increased across all groups, indicating cytocompatibility, with CEH-laden scaffolds exhibiting greater cell viability after seven days (p ≤ 0.0166). Also, 10% CEH-GelMA scaffolds decreased the IL-6, IL-1α, and TNF-α synthesis (p ≤ 0.035). CONCLUSION: CEH-laden GelMA scaffolds facilitated both adhesion and proliferation of DPSCs, and 10% CEH provided anti-inflammatory potential after chronic LPS challenge. CLINICAL RELEVANCE: CEH incorporated in GelMA fibrous scaffolds demonstrated the potential to be used as a cytocompatible and anti-inflammatory biomaterial for vital pulp therapy.


Subject(s)
Anti-Inflammatory Agents , Caseins , Cell Survival , Dental Pulp , Gelatin , Tissue Scaffolds , Gelatin/chemistry , Dental Pulp/cytology , Dental Pulp/drug effects , Tissue Scaffolds/chemistry , Humans , Anti-Inflammatory Agents/pharmacology , Cell Survival/drug effects , Methacrylates/chemistry , Materials Testing , Enzyme-Linked Immunosorbent Assay , Tensile Strength , Cells, Cultured , Stem Cells/drug effects , Cell Adhesion/drug effects , Biocompatible Materials/pharmacology , Biocompatible Materials/chemistry , Cytokines/metabolism , Surface Properties
4.
Biomed Mater ; 19(5)2024 Aug 08.
Article in English | MEDLINE | ID: mdl-39114907

ABSTRACT

(+)4-cholesten-3-one has been proved to have potential wound healing effect in the process of wound regeneration. This study aimed to evaluate the effect of (+)4-cholesten-3-one/sodium alginate/gelatin on skin injury and reveal its potential molecular mechanism. First, we prepared sodium alginate/gelatin hydrogel (SA/Gel hydrogel) with different ratios and tested their characteristics. Based on these results, different concentrations of (+)4-cholesten-3-one were added into SA/Gel hydrogel. A full-thickness skin injury model was successfully established to evaluate wound healing activityin vivo. HE staining and Masson staining were used to evaluate the thickness of granulation tissue and collagen deposition level. Immunohistochemical staining and immunofluorescence staining were applied to detect the level of revascularization and proliferation in each group of wounds. Western blot, quantitative-PCR and immunofluorescence staining were used to detect the expression of proteins related to Wnt/ß-catenin signaling pathway in each group of wounds.In vitroresults showed that the hydrogel not only created a 3D structure for cell adhesion and growth, but also exhibited good swelling ability, excellent degradability and favorable bio-compatibility. Most importantly,in vivoexperiments further indicated that (+)4-cholesten-3-one/SA/Gel hydrogel effectively enhanced wound healing. The effectiveness is due to its superior abilities in accelerating healing process, granulation tissue regeneration, collagen deposition, promoting angiogenesis, tissue proliferation, as well as fibroblast activation and differentiation. The underlying mechanism was related to the Wnt/ß-catenin signaling pathway. This study highlighted that (+)4-cholesten-3-one/SA/Gel hydrogel holds promise as a wound healing dressing in future clinical applications.


Subject(s)
Alginates , Gelatin , Hydrogels , Regeneration , Skin , Wound Healing , Wound Healing/drug effects , Alginates/chemistry , Animals , Gelatin/chemistry , Hydrogels/chemistry , Hydrogels/pharmacology , Skin/injuries , Skin/drug effects , Skin/metabolism , Regeneration/drug effects , Cell Proliferation/drug effects , Male , Mice , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Rats , Collagen/chemistry , Wnt Signaling Pathway/drug effects , Humans
5.
BMC Biotechnol ; 24(1): 53, 2024 Aug 06.
Article in English | MEDLINE | ID: mdl-39107760

ABSTRACT

Chemotherapy as a cornerstone of cancer treatment is slowly being edged aside owing to its severe side effects and systemic toxicity. In this case, nanomedicine has emerged as an effective tool to address these drawbacks. Herein, a biocompatible carrier based on bovine serum albumin (BSA) coated gadolinium oxide nanoparticles (Gd2O3@BSA) was fabricated for curcumin (CUR) delivery and its physicochemical features along with its potential anticancer activity against nasal squamous cell carcinoma were also investigated. It was found that the fabricated Gd2O3@BSA containing CUR (Gd2O3@BSA-CUR) had spherical morphology with hydrodynamic size of nearly 26 nm, zeta-potential of -36 mV and high drug (CUR) loading capacity. Drug release profile disclosed that the release of CUR from the prepared Gd2O3@BSA-CUR nanoparticles occurred in a sustained- and pH-dependent manner. Also, in vitro cytotoxicity analysis revealed that the fabricated Gd2O3@BSA nanoparticles possessed excellent biosafety toward HFF2 normal cells, while Gd2O3@BSA-CUR appeared to display the greatest anticancer potential against RPMI 2650 and CNE-1 cancer cell lines. The results also show that the Gd2O3@BSA nanoparticles were compatible with the blood cells with minor hemolytic effect (< 3%). The manufactured NPs were found to be completely safe for biological applications in an in vivo subacute toxicity study. Taken together, these finding substantiate the potential anticancer activity of Gd2O3@BSA-CUR nanoparticles against nasal squamous cell carcinoma, but the results obtained demand further studies to assess their full potential.


Subject(s)
Antineoplastic Agents , Carcinoma, Squamous Cell , Gadolinium , Serum Albumin, Bovine , Gadolinium/chemistry , Gadolinium/pharmacology , Humans , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Carcinoma, Squamous Cell/drug therapy , Serum Albumin, Bovine/chemistry , Cell Line, Tumor , Animals , Curcumin/pharmacology , Curcumin/chemistry , Nose Neoplasms/drug therapy , Nanoparticles/chemistry , Metal Nanoparticles/chemistry , Cell Survival/drug effects , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Drug Liberation , Hemolysis/drug effects
6.
Commun Biol ; 7(1): 941, 2024 Aug 03.
Article in English | MEDLINE | ID: mdl-39097626

ABSTRACT

Extracellular vesicles (EVs) are membrane-enclosed bio-nanoparticles secreted by cells and naturally evolved to transport various bioactive molecules between cells and even organisms. These cellular objects are considered one of the most promising bio-nanovehicles for the delivery of native and exogenous molecular cargo. However, many challenges with state-of-the-art EV-based candidates as drug carriers still exist, including issues with scalability, batch-to-batch reproducibility, and cost-sustainability of the final therapeutic formulation. Microalgal extracellular vesicles, which we named nanoalgosomes, are naturally released by various microalgal species. Here, we evaluate the innate biological properties of nanoalgosomes derived from cultures of the marine microalgae Tetraselmis chuii, using an optimized manufacturing protocol. Our investigation of nanoalgosome biocompatibility in preclinical models includes toxicological analyses, using the invertebrate model organism Caenorhabditis elegans, hematological and immunological evaluations ex vivo and in mice. We evaluate nanoalgosome cellular uptake mechanisms in C. elegans at cellular and subcellular levels, and study their biodistribution in mice with accurate space-time resolution. Further examination highlights the antioxidant and anti-inflammatory bioactivities of nanoalgosomes. This holistic approach to nanoalgosome functional characterization demonstrates that they are biocompatible and innate bioactive effectors with unique bone tropism. These findings suggest that nanoalgosomes have significant potential for future therapeutic applications.


Subject(s)
Anti-Inflammatory Agents , Antioxidants , Extracellular Vesicles , Microalgae , Extracellular Vesicles/metabolism , Animals , Microalgae/metabolism , Mice , Anti-Inflammatory Agents/pharmacology , Antioxidants/metabolism , Antioxidants/pharmacology , Caenorhabditis elegans/metabolism , Biocompatible Materials/chemistry , Chlorophyta/metabolism , Bone and Bones/metabolism , Tropism
7.
Int J Nanomedicine ; 19: 7751-7773, 2024.
Article in English | MEDLINE | ID: mdl-39099796

ABSTRACT

Endogenous stem cell homing refers to the transport of endogenous mesenchymal stem cells (MSCs) to damaged tissue. The paradigm of using well-designed biomaterials to induce resident stem cells to home in to the injured site while coordinating their behavior and function to promote tissue regeneration is known as endogenous regenerative medicine (ERM). ERM is a promising new avenue in regenerative therapy research, and it involves the mobilizing of endogenous stem cells for homing as the principal means through which to achieve it. Comprehending how mesenchymal stem cells home in and grasp the influencing factors of mesenchymal stem cell homing is essential for the understanding and design of tissue engineering. This review summarizes the process of MSC homing, the factors influencing the homing process, analyses endogenous stem cell homing studies of interest in the field of skin tissue repair, explores the integration of endogenous homing promotion strategies with cellular therapies and details tissue engineering strategies that can be used to modulate endogenous homing of stem cells. In addition to providing more systematic theories and ideas for improved materials for endogenous tissue repair, this review provides new perspectives to explore the complex process of tissue remodeling to enhance the rational design of biomaterial scaffolds and guide tissue regeneration strategies.


Subject(s)
Biocompatible Materials , Mesenchymal Stem Cells , Tissue Engineering , Wound Healing , Humans , Mesenchymal Stem Cells/cytology , Wound Healing/drug effects , Wound Healing/physiology , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Tissue Engineering/methods , Animals , Regenerative Medicine/methods , Tissue Scaffolds/chemistry , Cell Movement/drug effects , Skin , Mesenchymal Stem Cell Transplantation/methods
8.
Biomed Mater ; 19(5)2024 Aug 06.
Article in English | MEDLINE | ID: mdl-39105245

ABSTRACT

Bioresorbable chitosan scaffolds have shown potential for osteochondral repair applications. Thein vivodegradation of chitosan, mediated by lysozyme and releasing glucosamine, enables progressive replacement by ingrowing tissue. Here the degradation process of a chitosan-nHA based bioresorbable scaffold was investigated for mass loss, mechanical properties and degradation products released from the scaffold when subjected to clinically relevant enzyme concentrations. The scaffold showed accelerated mass loss during the early stages of degradation but without substantial reduction in mechanical strength or structure deterioration. Although not cytotoxic, the medium in which the scaffold was degraded for over 2 weeks showed a transient decrease in mesenchymal stem cell viability, and the main degradation product (glucosamine) demonstrated a possible adverse effect on viability when added at its peak concentration. This study has implications for the design and biomedical application of chitosan scaffolds, underlining the importance of modelling degradation products to determine suitability for clinical translation.


Subject(s)
Cell Survival , Chitosan , Materials Testing , Mesenchymal Stem Cells , Tissue Engineering , Tissue Scaffolds , Chitosan/chemistry , Cell Survival/drug effects , Tissue Scaffolds/chemistry , Mesenchymal Stem Cells/cytology , Animals , Tissue Engineering/methods , Biocompatible Materials/chemistry , Cells, Cultured , Glucosamine/chemistry , Humans , Muramidase/chemistry , Absorbable Implants
9.
Methods Mol Biol ; 2835: 29-37, 2024.
Article in English | MEDLINE | ID: mdl-39105903

ABSTRACT

The application of adult mesenchymal stem cells (MSCs) in the field of tissue regeneration is of increasing interest to the scientific community. In particular, scaffolds and/or hydrogel based on glycosaminoglycans (GAGs) play a pivotal role due to their ability to support the in vitro growth and differentiation of MSCs toward a specific phenotype. Here, we describe different possible approaches to develop GAGs-based biomaterials, hydrogel, and polymeric viscous solutions in order to assess/develop a suitable biomimetic environment. To sustain MSCs viability and promote their differentiation for potential therapeutic applications.


Subject(s)
Cell Differentiation , Glycosaminoglycans , Mesenchymal Stem Cells , Glycosaminoglycans/metabolism , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Humans , Hydrogels/chemistry , Cell Culture Techniques/methods , Adult Stem Cells/cytology , Adult Stem Cells/metabolism , Cell Survival , Tissue Scaffolds/chemistry , Cells, Cultured , Animals , Tissue Engineering/methods , Cell Proliferation , Biocompatible Materials/chemistry , Adult
10.
Methods Mol Biol ; 2835: 269-275, 2024.
Article in English | MEDLINE | ID: mdl-39105922

ABSTRACT

Three-dimensional (3D) scaffolds provide cell support while improving tissue regeneration through amplified cellular responses between implanted materials and native tissues. So far, highly conductive cardiac, nerve, and muscle tissues have been engineered by culturing stem cells on electrically inert scaffolds. These scaffolds, even though suitable, may not be very useful compared to the results shown by cells when cultured on conductive scaffolds. Noticing the mature phenotype the stem cells develop over time when cultured on conductive scaffolds, scientists have been trying to impart conductivity to traditionally nonconductive scaffolds. One way to achieve this goal is to blend conductive polymers (polyaniline, polypyrrole, PEDOT:PSS) with inert biomaterials and produce a 3D scaffold using various fabrication techniques. One such technique is projection micro-stereolithography, which is an additive manufacturing technique. It uses a photosensitive solution blended with conductive polymers and uses visible/UV light to crosslink the solution. 3D scaffolds with complex architectural features down to microscale resolution can be printed with this technique promptly. This chapter reports a protocol to fabricate electrically conductive scaffolds using projection micro-stereolithography.


Subject(s)
Cell Culture Techniques , Electric Conductivity , Polymers , Tissue Engineering , Tissue Scaffolds , Tissue Scaffolds/chemistry , Tissue Engineering/methods , Polymers/chemistry , Cell Culture Techniques/methods , Pyrroles/chemistry , Animals , Humans , Biocompatible Materials/chemistry , Cells, Cultured , Stem Cells/cytology , Aniline Compounds/chemistry , Myocytes, Cardiac/cytology , Stereolithography
11.
Methods Mol Biol ; 2835: 289-300, 2024.
Article in English | MEDLINE | ID: mdl-39105924

ABSTRACT

Hydrogels are a class of biomaterials that can provide a three-dimensional (3D) environment capable of mimicking the extracellular matrix of native tissues. In this chapter, we present a method to generate electrospun nanofibers for the purpose of reinforcing hydrogels. The addition of electrospun fibers can be used to improve the mechanical properties of hydrogels and broaden their range of applications. First, the polymer for making the electrospun fibers is formulated using chloroform/ethanol, polycaprolactone (PCL), polyethylene glycol (PEG), and polyethylene glycol diacrylate (PEGDA). Second, the polymer is used to generate thin electrospun nanofibers by an electrospinning technique using aluminum foil as a collector, which acts as the conductive substrate that collects the charged fibers. Third, the resulting electrospun fibers undergo a filtration process using nylon membrane filters, followed by lyophilization, ensuring complete removal of water from the sample.


Subject(s)
Hydrogels , Nanofibers , Polyethylene Glycols , Nanofibers/chemistry , Polyethylene Glycols/chemistry , Hydrogels/chemistry , Biocompatible Materials/chemistry , Humans , Cell- and Tissue-Based Therapy/methods , Polyesters/chemistry , Tissue Scaffolds/chemistry , Tissue Engineering/methods
12.
ACS Biomater Sci Eng ; 10(8): 4645-4661, 2024 Aug 12.
Article in English | MEDLINE | ID: mdl-39086282

ABSTRACT

Graphite carbon nitride (g-C3N4) is a two-dimensional conjugated polymer with a unique energy band structure similar to graphene. Due to its outstanding analytical advantages, such as relatively small band gap (2.7 eV), low-cost synthesis, high thermal stability, excellent photocatalytic ability, and good biocompatibility, g-C3N4 has attracted the interest of researchers and industry, especially in the medical field. This paper summarizes the latest research on g-C3N4-based composites in various biomedical applications, including therapy, diagnostic imaging, biosensors, antibacterial, and wearable devices. In addition, the application prospects and possible challenges of g-C3N4 in nanomedicine are also discussed in detail. This review is expected to inspire emerging biomedical applications based on g-C3N4.


Subject(s)
Biosensing Techniques , Graphite , Nitrogen Compounds , Graphite/chemistry , Humans , Nitrogen Compounds/chemistry , Biocompatible Materials/chemistry , Animals , Nitriles/chemistry , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Wearable Electronic Devices , Nanomedicine/methods
13.
Nat Commun ; 15(1): 6739, 2024 Aug 07.
Article in English | MEDLINE | ID: mdl-39112538

ABSTRACT

The minimally invasive injection of tissue engineering scaffolds is of interest as it requires a smaller incision and quickens recovery. However, the engineering of scaffolds capable of injection remains a challenge. Here, we report on a shrunken scaffold inspired by the shrinking of puffed food in a humid environment. A scaffold is freeze-dried to remove water then placed in a humid atmosphere. The humidity causes the dry scaffold to shrink by up to 90%. In addition, the humidity treatment reduces the scaffolds modulus minimizing the foreign body response after implantation. The scaffolds can rapidly swell into their original size and shape after application. A tool for the delivery of the minimally invasive scaffolds is developed and we demonstrate the potential for minimally invasive delivery using this shrinking technique.


Subject(s)
Tissue Engineering , Tissue Scaffolds , Tissue Scaffolds/chemistry , Tissue Engineering/methods , Animals , Humidity , Freeze Drying/methods , Minimally Invasive Surgical Procedures/methods , Mice , Biocompatible Materials/chemistry
14.
Sci Rep ; 14(1): 18311, 2024 08 07.
Article in English | MEDLINE | ID: mdl-39112669

ABSTRACT

Finding a novel drug delivery system (DDS) represents one of the most challenging endeavors in cancer therapy. Hence, in this study, we developed a new biocompatible and biodegradable zinc-based nanoscale metal-organic framework (Zn-NMOF) coated with folic acid (FA) functionalized chitosan (CS) to facilitate targeted delivery of doxorubicin (D), a standard chemotherapeutic agent, into breast cancer cells. The synthesis of the NMOF-CS-FA-D nanocomposite preceded its comprehensive characterization via FT-IR, DLS, XRD, SEM, and TEM analyses. Subsequent in vitro studies were conducted on MCF-7 breast cancer cells and HFF-1 normal cells, encompassing assessments of cell viability, expression levels of apoptotic and autophagy genes, cell cycle arrest, and apoptotic analyses. The size of the NMOF-CS-FA-D particles was determined to be less than 80 nm, with a drug loading efficiency of 72 ± 5%. The release kinetics of DOX from the nanocomposite were investigated, revealing controlled release behavior at pH 7.4 and accelerated release at pH 5.0, which is conducive to drug delivery into cancer cells. In vitro results indicated a 17.39% ± 6.34 cell viability after 24 h of treatment with a 40 nM concentration of the NMOF-CS-FA-D nanocomposite. Furthermore, the expression levels of Caspase-9 and BAX, key apoptotic genes, along with BECLIN1, an autophagy gene, were found to increase by two-fold, four-fold, and two-fold, respectively, following 5 h of treatment with the nanocomposite. Additionally, analysis of cell cycle distribution revealed 15.4 ± 2% of cells in the sub-G1 phase, indicative of apoptotic cells, and 31.9% of cells undergoing early and late apoptosis in MCF-7 cells. Collectively, these findings underscore the potential of the NMOF-CS-FA-D nanocomposite in inhibiting cancer cell proliferation with low side effects.


Subject(s)
Apoptosis , Breast Neoplasms , Chitosan , Doxorubicin , Metal-Organic Frameworks , Nanocomposites , Zinc , Humans , Nanocomposites/chemistry , Metal-Organic Frameworks/chemistry , Metal-Organic Frameworks/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Breast Neoplasms/metabolism , MCF-7 Cells , Zinc/chemistry , Zinc/pharmacology , Chitosan/chemistry , Female , Doxorubicin/pharmacology , Doxorubicin/chemistry , Doxorubicin/administration & dosage , Apoptosis/drug effects , Cell Survival/drug effects , Folic Acid/chemistry , Folic Acid/pharmacology , Drug Delivery Systems , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Drug Liberation , Drug Carriers/chemistry , Caspase 9/metabolism , Caspase 9/genetics , Autophagy/drug effects
15.
Proc Natl Acad Sci U S A ; 121(35): e2322418121, 2024 Aug 27.
Article in English | MEDLINE | ID: mdl-39159377

ABSTRACT

The growing world population and increasing life expectancy are driving the need to improve the quality of blood transfusion, organ transplantation, and preservation. Here, to improve the ability of red blood cells (RBCs) for normothermic machine perfusion, a biocompatible blood silicification approach termed "shielding-augmenting RBC-in-nanoscale amorphous silica (SARNAS)" has been developed. The key to RBC surface engineering and structure augmentation is the precise control of the hydrolysis form of silicic acid to realize stabilization of RBC within conformal nanoscale silica-based exoskeletons. The formed silicified RBCs (Si-RBCs) maintain membrane/structural integrity, normal cellular functions (e.g., metabolism, oxygen-carrying capability), and enhance resistance to external stressors as well as tunable mechanical properties, resulting in nearly 100% RBC cryoprotection. In vivo experiments confirm their excellent biocompatibility. By shielding RBC surface antigens, the Si-RBCs provide universal blood compatibility, the ability for allogeneic mechanical perfusion, and more importantly, the possibility for cross-species transfusion. Being simple, reliable, and easily scalable, the SARNAS strategy holds great promise to revolutionize the use of engineered blood for future clinical applications.


Subject(s)
Biocompatible Materials , Erythrocytes , Silicon Dioxide , Erythrocytes/metabolism , Silicon Dioxide/chemistry , Biocompatible Materials/chemistry , Animals , Humans , Perfusion/methods , Blood Preservation/methods , Blood Transfusion/methods , Mice
16.
Nat Commun ; 15(1): 6820, 2024 Aug 09.
Article in English | MEDLINE | ID: mdl-39122702

ABSTRACT

Biomaterial wound dressings, such as hydrogels, interact with host cells to regulate tissue repair. This study investigates how crosslinking of gelatin-based hydrogels influences immune and stromal cell behavior and wound healing in female mice. We observe that softer, lightly crosslinked hydrogels promote greater cellular infiltration and result in smaller scars compared to stiffer, heavily crosslinked hydrogels. Using single-cell RNA sequencing, we further show that heavily crosslinked hydrogels increase inflammation and lead to the formation of a distinct macrophage subpopulation exhibiting signs of oxidative activity and cell fusion. Conversely, lightly crosslinked hydrogels are more readily taken up by macrophages and integrated within the tissue. The physical properties differentially affect macrophage and fibroblast interactions, with heavily crosslinked hydrogels promoting pro-fibrotic fibroblast activity that drives macrophage fusion through RANKL signaling. These findings suggest that tuning the physical properties of hydrogels can guide cellular responses and improve healing, offering insights for designing better biomaterials for wound treatment.


Subject(s)
Fibroblasts , Hydrogels , Macrophages , Wound Healing , Animals , Hydrogels/chemistry , Wound Healing/drug effects , Fibroblasts/metabolism , Fibroblasts/drug effects , Macrophages/metabolism , Macrophages/drug effects , Mice , Female , Cell Communication/drug effects , Biocompatible Materials/chemistry , RANK Ligand/metabolism , Mice, Inbred C57BL , Cross-Linking Reagents/chemistry , Gelatin/chemistry , Inflammation/metabolism , Inflammation/pathology
17.
Molecules ; 29(15)2024 Jul 25.
Article in English | MEDLINE | ID: mdl-39124887

ABSTRACT

This article presents a new method for preparing multifunctional composite biomaterials with applications in advanced biomedical fields. The biomaterials consist of dicalcium phosphate (DCPD) and bioactive silicate glasses (SiO2/Na2O and SiO2/K2O), containing the antibiotic streptomycin sulfate. Materials were deeply characterized by X-ray diffraction and attenuated total reflectance Fourier transform infrared spectroscopy, and zeta potential analysis, UV-visible spectrophotometry, and ion-exchange measurement were applied in a simulating body fluid (SBF) solution. The main results include an in situ chemical transformation of dicalcium phosphate into an apatitic phase under the influence of silicate solutions and the incorporation of the antibiotic. The zeta potential showed a decrease in surface charge from ζ = -24.6 mV to ζ = -16.5 mV. In addition, a controlled and prolonged release of antibiotics was observed over a period of 37 days, with a released concentration of up to 755 ppm. Toxicity tests in mice demonstrated good tolerance of the biomaterials, with no significant adverse effects. Moreover, these biomaterials have shown potent antibacterial activity against various bacterial strains, including Listeria monocytogenes, Staphylococcus aureus, Escherichia coli, and Pseudomonas aeruginosa, suggesting their potential use in tissue engineering, drug delivery, and orthopedic and dental implants. By integrating the antibiotic into the biomaterial composites, we achieved controlled release and prolonged antibacterial efficacy. This research contributes to advancing biomaterials by exploring innovative synthetic routes and showcasing their promise in regenerative medicine and controlled drug delivery.


Subject(s)
Anti-Bacterial Agents , Biocompatible Materials , Regenerative Medicine , Biocompatible Materials/chemistry , Biocompatible Materials/chemical synthesis , Regenerative Medicine/methods , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/chemical synthesis , Animals , Mice , Drug Delivery Systems , X-Ray Diffraction , Microbial Sensitivity Tests , Delayed-Action Preparations/pharmacology , Spectroscopy, Fourier Transform Infrared , Calcium Phosphates/chemistry , Calcium Phosphates/chemical synthesis , Drug Liberation , Streptomycin/pharmacology , Silicon Dioxide/chemistry
18.
Biomed Mater ; 19(5)2024 Aug 14.
Article in English | MEDLINE | ID: mdl-39105493

ABSTRACT

Bone is a dynamic tissue that can always regenerate itself through remodeling to maintain biofunctionality. This tissue performs several vital physiological functions. However, bone scaffolds are required for critical-size damages and fractures, and these can be addressed by bone tissue engineering. Bone tissue engineering (BTE) has the potential to develop scaffolds for repairing critical-size damaged bone. BTE is a multidisciplinary engineered scaffold with the desired properties for repairing damaged bone tissue. Herein, we have provided an overview of the common carbohydrate polymers, fundamental structural, physicochemical, and biological properties, and fabrication techniques for bone tissue engineering. We also discussed advanced biofabrication strategies and provided the limitations and prospects by highlighting significant issues in bone tissue engineering. There are several review articles available on bone tissue engineering. However, we have provided a state-of-the-art review article that discussed recent progress and trends within the last 3-5 years by emphasizing challenges and future perspectives.


Subject(s)
Biocompatible Materials , Bone and Bones , Carbohydrates , Ceramics , Tissue Engineering , Tissue Scaffolds , Tissue Engineering/methods , Ceramics/chemistry , Humans , Bone and Bones/metabolism , Tissue Scaffolds/chemistry , Animals , Carbohydrates/chemistry , Biocompatible Materials/chemistry , Bone Regeneration , Bone Substitutes/chemistry , Polymers/chemistry
19.
Int J Nanomedicine ; 19: 8285-8308, 2024.
Article in English | MEDLINE | ID: mdl-39161362

ABSTRACT

The endometrium is an extremely important component of the uterus and is crucial for individual health and human reproduction. However, traditional methods still struggle to ideally repair the structure and function of damaged endometrium and restore fertility. Therefore, seeking and developing innovative technologies and materials has the potential to repair and regenerate damaged or diseased endometrium. The emergence and functionalization of various nanomedicine and biomaterials, as well as the proposal and development of regenerative medicine and tissue engineering techniques, have brought great hope for solving these problems. In this review, we will summarize various nanomedicine, biomaterials, and innovative technologies that contribute to endometrial regeneration, including nanoscale exosomes, nanomaterials, stem cell-based materials, naturally sourced biomaterials, chemically synthesized biomaterials, approaches and methods for functionalizing biomaterials, as well as the application of revolutionary new technologies such as organoids, organ-on-chips, artificial intelligence, etc. The diverse design and modification of new biomaterials endow them with new functionalities, such as microstructure or nanostructure, mechanical properties, biological functions, and cellular microenvironment regulation. It will provide new options for the regeneration of endometrium, bring new hope for the reconstruction and recovery of patients' reproductive abilities.


Subject(s)
Biocompatible Materials , Endometrium , Nanomedicine , Regeneration , Regenerative Medicine , Tissue Engineering , Humans , Endometrium/drug effects , Endometrium/physiology , Nanomedicine/methods , Female , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Tissue Engineering/methods , Regeneration/drug effects , Regenerative Medicine/methods , Nanostructures/chemistry , Animals , Exosomes/chemistry , Stem Cells/drug effects , Stem Cells/cytology
20.
Int J Nanomedicine ; 19: 8309-8336, 2024.
Article in English | MEDLINE | ID: mdl-39161358

ABSTRACT

Purpose: The treatment of craniofacial bone defects caused by trauma, tumors, and infectious and degenerative diseases is a significant issue in current clinical practice. Following the rapid development of bone tissue engineering (BTE) in the last decade, bioactive scaffolds coupled with multifunctional properties are in high demand with regard to effective therapy for bone defects. Herein, an innovative bone scaffold consisting of GO/Cu nanoderivatives and GelMA-based organic-inorganic hybrids was reported for repairing full-thickness calvarial bone defect. Methods: In this study, motivated by the versatile biological functions of nanomaterials and synthetic hydrogels, copper nanoparticle (CuNP)-decorated graphene oxide (GO) nanosheets (GO/Cu) were combined with methacrylated gelatin (GelMA)-based organic-inorganic hybrids to construct porous bone scaffolds that mimic the extracellular matrix (ECM) of bone tissues by photocrosslinking. The material characterizations, in vitro cytocompatibility, macrophage polarization and osteogenesis of the biohybrid hydrogel scaffolds were investigated, and two different animal models (BALB/c mice and SD rats) were established to further confirm the in vivo neovascularization, macrophage recruitment, biocompatibility, biosafety and bone regenerative potential. Results: We found that GO/Cu-functionalized GelMA/ß-TCP hydrogel scaffolds exhibited evidently promoted osteogenic activities, M2 type macrophage polarization, increased secretion of anti-inflammatory factors and excellent cytocompatibility, with favorable surface characteristics and sustainable release of Cu2+. Additionally, improved neovascularization, macrophage recruitment and tissue integration were found in mice implanted with the bioactive hydrogels. More importantly, the observations of microCT reconstruction and histological analysis in a calvarial bone defect model in rats treated with GO/Cu-incorporated hydrogel scaffolds demonstrated significantly increased bone morphometric values and newly formed bone tissues, indicating accelerated bone healing. Conclusion: Taken together, this BTE-based bone repair strategy provides a promising and feasible method for constructing multifunctional GO/Cu nanocomposite-incorporated biohybrid hydrogel scaffolds with facilitated osteogenesis, angiogenesis and immunoregulation in one system, with the optimization of material properties and biosafety, it thereby demonstrates great application potential for correcting craniofacial bone defects in future clinical scenarios.


Subject(s)
Bone Regeneration , Copper , Graphite , Hydrogels , Rats, Sprague-Dawley , Skull , Tissue Engineering , Tissue Scaffolds , Animals , Bone Regeneration/drug effects , Tissue Scaffolds/chemistry , Copper/chemistry , Copper/pharmacology , Graphite/chemistry , Hydrogels/chemistry , Hydrogels/pharmacology , Skull/drug effects , Skull/injuries , Rats , Mice , Tissue Engineering/methods , Osteogenesis/drug effects , Mice, Inbred BALB C , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Male , Metal Nanoparticles/chemistry , Nanostructures/chemistry , Gelatin/chemistry , RAW 264.7 Cells
SELECTION OF CITATIONS
SEARCH DETAIL