Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 141
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Small ; 20(26): e2310731, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38247187

RESUMEN

The development of robust adhesive, conductive, and flexible materials has garnered significant attention in the realm of human-machine interface and electronic devices. Conventional preparation methods to achieve these exceptional properties rely on incorporating highly polar raw materials, multiple components, or solvents. However, the overexposure of functional groups and the inherent toxicity of organic solvents often render gels non-stick or potentially biocompatible making them unsuitable for human-contact devices. In this study, a straightforward three-step strategy is devised for preparing responsive adhesive gels without complex components. Structurally conductive poly(N-(2-hydroxyethyl)-acrylamide-co-p-styrene sulfonate hydrate) (PHEAA-NaSS) gels are synthesized by integrating ionic and hydrophilic networks with distinct solvent effects. Initially, the in-suit formed PHEAA-NaSS networks are activated by dimethyl sulfoxide, which substantially increases intramolecular hydrogen bonding and enhances the matrix stretchability and interfacial adhesion. Subsequently, ethanol exchange reduced solvent impact and led to a compact network that limited surface exposure of ionic and hydrophilic groups, resulting in nonstick, robust for convenient storage. Finally, upon contacting with water, the network demonstrates rehydration, resulting in favorable adhesion, biocompatibility, and conductivity. The proposed PHEAA-NaSS/W gels can stably and reliably capture joint motion and electrophysiological signals. Furthermore, this uncomplicated gel preparation method is also applicable to other electrolyte monomers.


Asunto(s)
Materiales Biocompatibles , Conductividad Eléctrica , Geles , Solventes , Dispositivos Electrónicos Vestibles , Solventes/química , Materiales Biocompatibles/química , Geles/química , Humanos , Adhesivos/química
2.
Small ; 18(48): e2204837, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36207286

RESUMEN

Janus porous biomaterials are gaining increasing attention and there are considerable efforts to develop simple, rapid, and scalable methods capable of tuning micro- and macro-structures. Here, a single-step electro-fabrication method to create a Janus porous film by the electrodeposition of the amino-polysaccharide chitosan is reported. Specifically, a Janus structure emerges spontaneously when electrodeposition is performed at sub-ambient temperature (0-5 °C). Sub-ambient temperature electrodeposition experiments show that: a Janus microstructure emerges (potentially as the result of a subtle alteration of the intermolecular interactions responsible for self-assembly); important microstructural features (pore size, porosity, and thicknesses) can be tuned by conditions; and this method is readily scalable (vs serial printing) and can yield complex tubular structures with Janus faces. In vitro studies demonstrate anisotropic cell guidance, and in vivo studies using a rat calvarial defect model further confirm the beneficial features of such Janus porous film for guided bone regeneration. In summary, these results further demonstrate that electro-fabrication provides a simple and scalable platform technology for the controlled functional structures of soft matter for applications in regenerative medicine.


Asunto(s)
Materiales Biocompatibles , Galvanoplastia , Animales , Ratas , Porosidad , Temperatura , Medicina Regenerativa
3.
Small ; 18(36): e2106824, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35060321

RESUMEN

The development of programmable functional biomaterials makes 4D printing add a new dimension, time (t), based on 3D structures (x, y, z), therefore, 4D printed constructs could transform their morphology or function over time in response to environmental stimuli. Nowadays, highly efficient bone defect repair remains challenging in clinics. Combining programmable biomaterials, living cells, and bioactive factors, 4D bioprinting provides greater potential for constructing dynamic, personalized, and precise bone tissue engineering scaffolds by complex structure formation and functional maturation. Therefore, 4D bioprinting has been regarded as the next generation of bone repair technology. This review focuses on 4D printing and its advantages in orthopedics. The applications of different smart biomaterials and 4D printing strategies are briefly introduced. Furthermore, one summarizes the recent advancements of 4D printing in bone tissue engineering, uncovering the addressed and unaddressed medical requirements. In addition, current challenges and future perspectives are further discussed, which will offer more inspiration about the clinical transformation of this emerging 4D bioprinting technology in bone regeneration.


Asunto(s)
Bioimpresión , Ortopedia , Materiales Biocompatibles/química , Impresión Tridimensional , Ingeniería de Tejidos/métodos
4.
Biomacromolecules ; 23(3): 1376-1391, 2022 03 14.
Artículo en Inglés | MEDLINE | ID: mdl-35195006

RESUMEN

Hydrogels with inherent antibacterial activities have been attracting increasing attention, particularly for biomedical applications. Biology provides a range of materials and mechanisms to meet diverse requirements for bacterial combating. Lysozyme after fibrillation (LZMF) has a much superior antibacterial ability than globular native lysozyme due to its decreased positive charges and increased hydrophobic ß-sheet component. Here, we propose to design a poly(ethylene glycol) (PEG) cross-linked LZMF composite antibacterial hydrogel by utilizing the nucleophilic substitution reaction between LZMF and N-hydroxysuccinimide end groups on four-arm PEG-NHS. The generated PEG-LZMF hydrogel is bacteria-resistant both in vitro and in vivo as expected and has good biocompatibility. Moreover, the volume expansion of PEG can be significantly inhibited due to the presence of hydrophobic lysozyme amyloid fibrils. In addition, the relatively fast cross-linking reaction can make PEG-LZMF both injectable and shape-compatible. The simultaneous reaction with tissue-exposed -NH2 or -SH also confers a tissue-adhesive ability. We envision that this hydrophobic lysozyme amyloid fibril-integrated PEG composite hydrogel can effectively adhere/protect open wounds and internal incisions and suppress pathogen infection through a biomimetic antibacterial mechanism. Considering the simple fabrication process, this multifunctional PEG-LZMF antibacterial hydrogel is promising for clinical transformation.


Asunto(s)
Adhesivos , Muramidasa , Amiloide , Antibacterianos/química , Antibacterianos/farmacología , Materiales Biocompatibles/química , Hidrogeles/química , Hidrogeles/farmacología , Muramidasa/química , Polietilenglicoles/química
5.
Int J Mol Sci ; 23(6)2022 Mar 16.
Artículo en Inglés | MEDLINE | ID: mdl-35328601

RESUMEN

Delayed surface endothelialization is a bottleneck that restricts the further application of cardiovascular stents. It has been reported that the nature-inspired extracellular matrix (ECM) secreted by the hyaluronic acid (HA) micro-patterned smooth muscle cells (SMC) and endothelial cells (EC) can significantly promote surface endothelialization. However, this ECM coating obtained by decellularized method (dECM) is difficult to obtain directly on the surface of degradable magnesium (Mg) alloy. In this study, the method of obtaining bionic dECM by micro-patterning SMC/EC was further improved, and the nature-inspired ECM was prepared onto the Mg-Zn-Y-Nd (ZE21B) alloy surface by self-assembly. The results showed that the ECM coating not only improved surface endothelialization of ZE21B alloy, but also presented better blood compatibility, anti-hyperplasia, and anti-inflammation functions. The innovation and significance of the study is to overcome the disadvantage of traditional dECM coating and further expand the application of dECM coating to the surface of degradable materials and materials with different shapes.


Asunto(s)
Aleaciones , Células Endoteliales , Aleaciones/farmacología , Materiales Biocompatibles Revestidos/farmacología , Matriz Extracelular , Magnesio/farmacología , Miocitos del Músculo Liso
6.
Nano Lett ; 20(10): 7716-7721, 2020 10 14.
Artículo en Inglés | MEDLINE | ID: mdl-32946240

RESUMEN

Implant loosening is still the major form of the failure of artificial joints. Herein, inspired by the operculum of the river snail, we prepared a novel bionic micro/nanoscale topography on a titanium surface. This bionic topography promoted early cell adhesion through up-regulating the expression of ITG α5ß1 and thus accelerated the following cell spreading, proliferation, and differentiation. Moreover, a miR-21 coating, which promoted the angiogenic differentiation of MSCs, was fabricated on the bionic topography. Benefiting from both bionic micro/nanoscale topography and miR-21, blood vessel growth and bone formation and mineralization around the implant, as well as bone-implant bonding strength, were significantly improved. Collectively, the present study highlights the combination of the bionic micro/nanoscale topography and miR-21 on promoting cell adhesion and angiogenic differentiation and improving in vivo angiogenesis and bone-implant osseointegration. This work provides a new train of thought propelling the development of implants for potential application in the orthopedics field.


Asunto(s)
Biónica , MicroARNs , Adhesión Celular , Diferenciación Celular , Materiales Biocompatibles Revestidos , MicroARNs/genética , Oseointegración , Propiedades de Superficie , Titanio
7.
Chem Rev ; 117(5): 4376-4421, 2017 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-28221776

RESUMEN

Although the biological functions of cell and tissue can be regulated by biochemical factors (e.g., growth factors, hormones), the biophysical effects of materials on the regulation of biological activity are receiving more attention. In this Review, we systematically summarize the recent progress on how biomaterials with controllable properties (e.g., compositional/degradable dynamics, mechanical properties, 2D topography, and 3D geometry) can regulate cell behaviors (e.g., cell adhesion, spreading, proliferation, cell alignment, and the differentiation or self-maintenance of stem cells) and tissue/organ functions. How the biophysical features of materials influence tissue/organ regeneration have been elucidated. Current challenges and a perspective on the development of novel materials that can modulate specific biological functions are discussed. The interdependent relationship between biomaterials and biology leads us to propose the concept of "materiobiology", which is a scientific discipline that studies the biological effects of the properties of biomaterials on biological functions at cell, tissue, organ, and the whole organism levels. This Review highlights that it is more important to develop ECM-mimicking biomaterials having a self-regenerative capacity to stimulate tissue regeneration, instead of attempting to recreate the complexity of living tissues or tissue constructs ex vivo. The principles of materiobiology may benefit the development of novel biomaterials providing combinative bioactive cues to activate the migration of stem cells from endogenous reservoirs (i.e., cell niches), stimulate robust and scalable self-healing mechanisms, and unlock the body's innate powers of regeneration.


Asunto(s)
Materiales Biocompatibles , Células , Ingeniería de Tejidos , Adhesión Celular , Linaje de la Célula , Movimiento Celular , Andamios del Tejido
8.
J Mater Sci Mater Med ; 28(10): 146, 2017 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-28823088

RESUMEN

Wound dressings play important roles in the management of wounds, and calcium cross-linked alginate (Ca2+-Alg) is a commonly used hydrogel that is adapted for wound treatment. However, conventional methods for fabricating Ca2+-Alg hydrogels can be tedious and difficult to control because of the rapid Ca2+-induced gelation of alginate. In this study, An electrodeposition method was used to rapidly and controllably fabricate Ca2+-Alg films for wound treatment. Several measures of film growth (e.g., thickness and mass) are shown to linearly correlate to the imposed charge transfer at the electrode. Similarly, this charge transfer was also observed to control important physicochemical wound healing properties such as water uptake and retention capacity. Furthermore, a wound healing animal test was performed to evaluate the performance of this electro-fabricated calcium alginate film for wound treatment. This in vivo study demonstrated that wounds dressed with an electro-fabricated Ca2+-Alg film closed faster than that of untreated wounds. Further, the new dermis tissue that formed was composed of reorganized and stratified epithelial layer, with fully developed connective tissue, hair follicle, sebaceous glands as well as aligned collagen. Therefore, our study indicates that this electrofabrication method for the rapid and controlled preparation of alginate film could provide exciting opportunities for wound treatment. More broadly, this study demonstrates the potential of electrochemistry for the fabrication of high performance polymeric materials. Here we report a rapid and controllable fabrication of free-standing alginate films by coupling anodic electrodeposition with subsequent peeling of deposited materials for wound dressing.


Asunto(s)
Alginatos/química , Vendajes , Calcio/química , Electricidad , Membranas Artificiales , Heridas y Lesiones/terapia , Animales , Ácido Glucurónico/química , Ácidos Hexurónicos/química , Masculino , Ensayo de Materiales , Ratones , Ratones Endogámicos BALB C , Agua , Cicatrización de Heridas
9.
Exp Hematol ; 133: 104192, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38432427

RESUMEN

Hematopoietic stem cell transplantation remains the preferred treatment for a variety of hematopoietic function disorders. To address the issue of limited numbers of hematopoietic stem/progenitor cells (HSPCs), significant progress has been made in the technology for ex vivo expansion of HSPCs. In addition, biomaterial-assisted in vivo production technology for therapeutic cells, including HSPCs, is gradually gaining attention. With the aid of specifically functional biomaterials, researchers can construct bone-like tissues exhibiting typical bone marrow-like structures (termed in vivo osteo-organoids in this article) for the production of therapeutic cells. These in vivo osteo-organoids mimic the native bone marrow niche and provide a microenvironment conducive to the expansion and differentiation of HSPCs. In this perspective article, we systematically summarize the history of in vivo osteo-organoids as a model for studying hematopoiesis and cancer metastasis and propose the challenges faced by the in vivo osteo-organoid production platform for therapeutic cells in terms of clinical translation. Ultimately, we hope to achieve functional customization of in vivo osteo-organoid-derived cells through continuously developed material design methods, so as to meet the treatment needs of different types of diseases and bring hope for life to more people.


Asunto(s)
Materiales Biocompatibles , Células Madre Hematopoyéticas , Humanos , Animales , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Trasplante de Células Madre Hematopoyéticas , Organoides/citología , Hematopoyesis , Diferenciación Celular
10.
Adv Mater ; 36(21): e2311459, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38346345

RESUMEN

Hair loss is characterized by the inability of hair follicles (HFs) to enter the telogen-anagen transition (TAT) and lack of de novo HFs. Current pharmaceutical therapies and surgical modalities have been largely limited to regulating hair regrowth efficiently without side effects and lacking treatment compliance. Here, this work proposes a materiobiomodulation therapy (MBMT), wherein polydopamine (PDA) nanoparticles with redox activity can be modulated to have a stoichiometric ROS (H2O2) donating ability. These nanoparticles can intracellularly deliver ROS with high-efficiency via the clathrin-dependent endocytosis process. Utilizing homozygote transgenic HyPerion (a genetically-encoded H2O2 biosensor) mice, this work also achieves in vivo dynamic monitoring of intracellular H2O2 elevation induced by ROS donators. Subcutaneous administration with ROS donators results in rapid onset of TAT and subsequent hair regrowth with a specific ROS "hormesis effect." Mechanistically, ROS activate ß-catenin-dependent Wnt signaling, upregulating hair follicle stem cell expression. This work further develops a microneedles patch for transdermal ROS delivery, demonstrating long-term, low-dose ROS release. Unlike photobiomodulation therapy (PBMT), MBMT requires no external stimuli, providing a convenient and efficient approach for clinical hair loss treatment. This material-HF communication implicates new avenues in HF-related diseases, achieving targeted ROS delivery with minimal side effects.


Asunto(s)
Folículo Piloso , Indoles , Nanopartículas , Polímeros , Especies Reactivas de Oxígeno , Animales , Folículo Piloso/metabolismo , Folículo Piloso/efectos de los fármacos , Ratones , Indoles/química , Nanopartículas/química , Polímeros/química , Especies Reactivas de Oxígeno/metabolismo , Peróxido de Hidrógeno/metabolismo , Alopecia/terapia , Cabello/crecimiento & desarrollo , Cabello/efectos de los fármacos , Vía de Señalización Wnt/efectos de los fármacos , Humanos , Ratones Transgénicos
11.
Adv Sci (Weinh) ; 11(13): e2305756, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38189598

RESUMEN

Currently available guided bone regeneration (GBR) films lack active immunomodulation and sufficient osteogenic ability- in the treatment of periodontitis, leading to unsatisfactory treatment outcomes. Challenges remain in developing simple, rapid, and programmable manufacturing methods for constructing bioactive GBR films with tailored biofunctional compositions and microstructures. Herein, the controlled electroassembly of collagen under the salt effect is reported, which enables the construction of porous films with precisely tunable porous structures (i.e., porosity and pore size). In particular, bioactive salt species such as the anti-inflammatory drug diclofenac sodium (DS) can induce and customize porous structures while enabling the loading of bioactive salts and their gradual release. Sequential electro-assembly under pre-programmed salt conditions enables the manufacture of a Janus composite film with a dense and DS-containing porous layer capable of multiple functions in periodontitis treatment, which provides mechanical support, guides fibrous tissue growth, and acts as a barrier preventing its penetration into bone defects. The DS-containing porous layer delivers dual bio-signals through its morphology and the released DS, inhibiting inflammation and promoting osteogenesis. Overall, this study demonstrates the potential of electrofabrication as a customized manufacturing platform for the programmable assembly of collagen for tailored functions to adapt to specific needs in regenerative medicine.


Asunto(s)
Periodontitis , Andamios del Tejido , Humanos , Andamios del Tejido/química , Porosidad , Osteogénesis , Colágeno/química , Periodontitis/tratamiento farmacológico
12.
J Mater Sci Mater Med ; 24(7): 1627-37, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23563980

RESUMEN

Premixed injectable calcium phosphate cement (p-ICPC) pastes have advantages over aqueous injectable calcium phosphate cement (a-ICPC) because p-ICPC remain stable during storage and harden only after placement into the defect. This paper focused on the suspension stability of p-ICPC paste by using fumed silica as a stabilizing agent and propylene glycol (PEG) as a continuous phase. Multiple light scanning techniques were first applied to evaluate the suspension stability. The results indicated that fumed silica effectively enhanced the suspension stability of p-ICPC pastes. The stabilizing effect of fumed silica results from the network structure formed in PEG because of its thixotropy. The p-ICPC could be eventually hydrated to form hydroxyapatite under aqueous circumstances by the unique replacement between water and PEG. p-ICPC (1) not only possesses proper thixotropy and compressive strength but has good injectability as well. p-ICPC (1) was cytocompatible and had no adverse effect on the attachment and proliferation of MG-63 cells in vitro. These observations may have applicability to the development of other nonaqueous injectable biomaterials for non-immediate filling and long-term storage.


Asunto(s)
Cementos para Huesos/química , Fosfatos de Calcio/administración & dosificación , Fosfatos de Calcio/química , Animales , Líquidos Corporales/fisiología , Cementos para Huesos/farmacocinética , Fosfatos de Calcio/farmacocinética , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Precipitación Química , Fuerza Compresiva , Estabilidad de Medicamentos , Inyecciones , Ensayo de Materiales/métodos , Ratones , Osteoblastos/efectos de los fármacos , Osteoblastos/fisiología , Reología , Suspensiones , Sustancias Viscoelásticas/química
13.
Macromol Biosci ; 23(4): e2200507, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36645702

RESUMEN

Biodegradable materials are pivotal in the biomedical field, where how to precisely control their structure and performance is critical for their translational application. In this study, poly(L-lactide-b-ε-caprolactone) block copolymers (bPLCL) with well-defined segment structure are obtained by a first synthesis of poly(ε-caprolactone) soft block, followed by ring opening polymerization of lactide to form poly(L-lactide acid)  hard block. The pre-polymerization allows for fabrication of bPLCL with the definite compositions of soft/hard segment while preserving the individual segment of their special soft or hard segment. These priorities make the bPLCL afford biodegradable polymer with better mechanical and biodegradable controllability than the random poly(L-lactide-co-ε-caprolactone) (rPLCL) synthesized via traditional one-pot polymerization. 10 mol% ε-caprolactone introduction can result in a formation of an elastic polymer with elongation at break of 286.15% ± 55.23%. Also, bPLCL preserves the unique crystalline structure of the soft and hard segments to present a more sustainable biodegradability than the rPLCL. The combinative merits make the pre-polymerization technique a promising strategy for a scalable production of PLCL materials for potential biomedical application.


Asunto(s)
Poliésteres , Polímeros , Polímeros/química , Poliésteres/química , Lactonas/química , Caproatos/química
14.
Adv Healthc Mater ; 12(27): e2301264, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37341519

RESUMEN

Macrophages play a crucial role in the complete processes of tissue repair and regeneration, and the activation of M2 polarization is an effective approach to provide a pro-regenerative immune microenvironment. Natural extracellular matrix (ECM) has the capability to modulate macrophage activities via its molecular, physical, and mechanical properties. Inspired by this, an ECM-mimetic hydrogel strategy to modulate macrophages via its dynamic structural characteristics and bioactive cell adhesion sites is proposed. The LZM-SC/SS hydrogel is in situ formed through the amidation reaction between lysozyme (LZM), 4-arm-PEG-SC, and 4-arm-PEG-SS, where LZM provides DGR tripeptide for cell adhesion, 4-arm-PEG-SS provides succinyl ester for dynamic hydrolysis, and 4-arm-PEG-SC balances the stability and dynamics of the network. In vitro and subcutaneous tests indicate the dynamic structural evolution and cell adhesion capacity promotes macrophage movement and M2 polarization synergistically. Comprehensive bioinformatic analysis further confirms the immunomodulatory ability, and reveals a significant correlation between M2 polarization and cell adhesion. A full-thickness wound model is employed to validate the induced M2 polarization, vessel development, and accelerated healing by LZM-SC/SS. This study represents a pioneering exploration of macrophage modulation by biomaterials' structures and components rather than drug or cytokines and provides new strategies to promote tissue repair and regeneration.


Asunto(s)
Hidrogeles , Cicatrización de Heridas , Hidrogeles/química , Macrófagos/metabolismo , Materiales Biocompatibles/química , Matriz Extracelular/química
15.
Biomed Microdevices ; 14(2): 259-70, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22124885

RESUMEN

Due to the unique physicochemical properties and membrane-permeable capacity, mesoporous silica nanoparticles (MSNs) are considered as an ideal carrier for intracellular delivery. Herein, we endeavored to address the size effect of MSNs on the cellular uptake, endosomal escape and controlled release, the key steps for the intracellular delivery. The well-ordered MSNs in the range from 55-nm to 440-nm with similar pore texture were prepared by modified base-catalyzed sol-gel method. With MC3T3-E1 model cell line, the in vitro results indicated that after 12 h cultivation, MSNs within 55 ~ 440 nm could all be internalized into the cells, and further escaped out of the endosomal compartment. The efficiency of the cellular uptake and endosomal escape strongly depended on the particle size, with the best efficiencies from 100-nm MSNs. Furthermore, the MTT results indicated that these MSNs materials were all biocompatible. The controlled release experiments with hydrophobic dexamethasone and hydrophilic vitamin C as models showed that for these small-molecular drugs, the loading amount all mainly determined by the surface area of the MSNs, and the subsequent release of the drug dramatically decreased with the increasing of the particle size. By contrast, the release rate of vitamin C was much quicker than that of the dexamethasone. These findings presented here could provide new means to tailor the size of MSNs and thus to guide the design of MSNs-based intracellular delivery system. Due to the good cell biocompatibility, high cellular uptake and endosomal escape, we conjectured that the 100-nm MSNs are more favorable for the intracellular delivery of drugs in live cells.


Asunto(s)
Preparaciones de Acción Retardada/farmacología , Endocitosis , Nanopartículas/química , Dióxido de Silicio/química , Células 3T3 , Animales , Ácido Ascórbico/metabolismo , Materiales Biocompatibles/química , Dexametasona/metabolismo , Citometría de Flujo , Interacciones Hidrofóbicas e Hidrofílicas , Ratones , Tamaño de la Partícula
16.
J Mater Sci Mater Med ; 23(3): 697-710, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22311074

RESUMEN

A hydrogel which will undergo macroscopic transition responding to redox stimuli is prepared. Mercapto precursors are prepared from 4-armed polyethylene glycol and after deprotection of thiolate anions, they can transform into disulfide crosslinked hydrogels within 3 min by responding to oxidant H(2)O(2). Desirable elasticity is exhibited with a wide range of storage modulus from 50 Pa to 14 kPa through rheological investigation. In addition, the hydrogels are found to be hydrolytically stable but degrade within 75 days when exposed to reductant such as glutathione (GSH). So gelation time and degradation behavior can be regulated by concentrations of precursor, oxidant, reductant, temperature, and pH value. Notably, interest arises from the long-period degradation under low GSH concentration of 0.01 mM that is similar to extracellular level, but not the fast disintegration under high concentration intracellular, providing the possibility of "smart" degradation responding to those cell-secreted biomacromolecules during the process of tissue regeneration. Furthermore, both hydrogels and their degradation products show cell viability above 90% culturing with C2C12 cells, representing nontoxic properties. Such a stimuli-responsive degradation strategy will give promising application in tissue repair and regeneration; especially enable the achievement of matching the degradation kinetics with physiological environment.


Asunto(s)
Hidrogeles , Polietilenglicoles/química , Animales , Línea Celular , Espectroscopía de Resonancia Magnética , Ratones , Oxidación-Reducción , Reología
17.
Adv Healthc Mater ; 11(4): e2101590, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34797950

RESUMEN

A proper pH microenvironment is crucial to mobilizing regeneration function of biomaterials. Neutralizing the acidity in bone defects with alkaline substances is a promising strategy to create favorable environments for cell proliferation and bone repair. In this study, to neutralize the acidity and reduce the inflammation caused by the rapid release of citric acid, a novel citrate-based biodegradable elastomeric poly(citric acid-1,8-octanediol-1,4-bis(2-hydroxyethyl)piperazine (BHEp)) (POPC) is synthesized with the introduction of the alkaline fragment BHEp, and then POPC/ß-tricalcium phosphate (ß-TCP) porous scaffolds are fabricated by 3D printing technique. The results reveal that the alkaline fragment BHEp effectively corrects the acid environment and improves the biocompatibility, cells affinity and promoted cell adhesion, and proliferation of POPC. Furthermore, the improved pH of POPC15/ß-TCP (PTCP15) enhances the adhesion and the proliferation of rabbit bone marrow mesenchymal stem cells, and the expression of osteogenesis-related genes. Moreover, PTCP15 scaffolds relieve inflammatory response and switch RAW 264.7 toward a prohealing extreme. The rat femoral defect model further demonstrates good biocompatibility and enhanced bone regeneration of PTCP15. In conclusion, the results offer a promising approach for biodegradable polymers to address the degradation acidity issue. Meanwhile, a positive regulation strategy is provided for biopolymer to enhance cell proliferation, osteogenic differentiation, and bone repair.


Asunto(s)
Materiales Biocompatibles , Osteogénesis , Animales , Materiales Biocompatibles/farmacología , Regeneración Ósea , Fosfatos de Calcio/química , Fosfatos de Calcio/farmacología , Diferenciación Celular , Citratos/química , Ácido Cítrico , Impresión Tridimensional , Conejos , Ratas , Andamios del Tejido/química
18.
ACS Appl Mater Interfaces ; 14(35): 39746-39758, 2022 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-36006024

RESUMEN

Timely restoration of blood supply following ischemia is critical to rescue damaged tissue. However, clinical efficacy is hampered by the inflammatory response after ischemia. Whether inflammation fine tunes the angiogenesis and the function of blood vessels via the heterogeneity of neutrophils remain poorly understood. Herein, the objective of this work is to incorporate the growth factors secreted by neutrophils into a porous gelatin methacrylate (GelMA) hydrogel, which subsequently is used as a novel regenerative scaffold with defined architecture for ischemia. We demonstrate that anti-inflammatory neutrophils (N2-polarized neutrophils) play an important role in promoting the migration of human umbilical vein endothelial cells (HUVECs) and formation of capillary-like networks in vitro. More importantly, vascular anastomosis can be achieved by modulating the neutrophils to N2 phenotype. In addition, N2-polarized composite hydrogel scaffolds can regulate inflammation, maintain the survival of exogenous cells, and promote angiogenesis in vivo. Notably, the composite hydrogel scaffolds promote neovascularization during exogenous introduction of endothelial cells by anastomosis. Taken together, this study highlights N2-polarized neutrophils composite hydrogels can achieve vascularization rapidly by regulating inflammation and promoting vascular anastomosis. This work lays the foundation for research into the treatment of ischemia and may inspire further research into novel treatment options.


Asunto(s)
Hidrogeles , Neutrófilos , Materiales Biocompatibles , Gelatina/farmacología , Células Endoteliales de la Vena Umbilical Humana , Humanos , Hidrogeles/farmacología , Inflamación , Isquemia , Neovascularización Patológica , Neovascularización Fisiológica , Ingeniería de Tejidos
19.
J Mater Chem B ; 10(34): 6464-6471, 2022 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-35960152

RESUMEN

Biodegradable microspheres have been widely applied as cell carriers for tissue engineering and regenerative medicine. However, most cell carriers only have a simple planar structure and show poor biological activity and cell adherence, resulting in low cell density and unfavorable application effect. How to develop size-controllable microspheres with an open-porous structure remains a challenge, and is a key factor to extend their employment as cell/drug delivery vehicles to boost regeneration of tissues (e.g., bone). Herein, well-defined open porous microspheres of poly(lactic-co-glycolic acid) (PLGA with good biocompatibility approved by the Food and Drug Administration (FDA)) were developed by using a gas-assisted-emulsion and surface-alkalization-treatment technology (GEST). The gas-assisted-emulsion strategy enables the formation of microspheres with a large size of 200-300 µm, meanwhile, the microspheres have a large amount of micropores with diameter in the range of 10-60 µm. The following alkalization-treatment on the surface makes the microspheres form a good porous interconnectivity throughout both the surface and the interior of the microspheres. The good porous interconnectivity endows the microspheres with a highly open pore structure and a large specific surface area for nutrient exchange and cell attachment, thus promoting cell proliferation and nutrient transportation, promising their potential as an ideal cell carrier to increase cell density and bioactivity for cell therapy-based tissue engineering.


Asunto(s)
Ácido Poliglicólico , Ingeniería de Tejidos , Emulsiones , Ácido Láctico/química , Microesferas , Ácido Poliglicólico/química , Copolímero de Ácido Poliláctico-Ácido Poliglicólico , Porosidad , Ingeniería de Tejidos/métodos , Estados Unidos
20.
ACS Appl Mater Interfaces ; 14(39): 44111-44124, 2022 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-36137506

RESUMEN

Bone regeneration is a well-orchestrated process involving electrical, biochemical, and mechanical multiple physiological cues. Electrical signals play a vital role in the process of bone repair. The endogenous potential will spontaneously form on defect sites, guide the cell behaviors, and mediate bone healing when the bone fracture occurs. However, the mechanism on how the surface charges of implant potentially guides osteogenesis and osteoimmunology has not been clearly revealed yet. In this study, piezoelectric BaTiO3/ß-TCP (BTCP) ceramics are prepared by two-step sintering, and different surface charges are established by polarization. In addition, the cell osteogenesis and osteoimmunology of BMSCs and RAW264.7 on different surface charges were explored. The results showed that the piezoelectric constant d33 of BTCP was controllable by adjusting the sintering temperature and rate. The polarized BTCP with a negative surface charge (BTCP-) promoted protein adsorption and BMSC extracellular Ca2+ influx. The attachment, spreading, migration, and osteogenic differentiation of BMSCs were enhanced on BTCP-. Additionally, the polarized BTCP ceramics with a positive surface charge (BTCP+) significantly inhibited M1 polarization of macrophages, affecting the expression of the M1 marker in macrophages and changing secretion of proinflammatory cytokines. It in turn enhanced osteogenic differentiation of BMSCs, suggesting that positive surface charges could modulate the bone immunoregulatory properties and shift the immune microenvironment to one that favored osteogenesis. The result provides an alternative method of synergistically modulating cellular immunity and the osteogenesis function and enhancing the bone regeneration by fabricating piezoelectric biomaterials with electrical signals.


Asunto(s)
Materiales Biocompatibles , Osteogénesis , Materiales Biocompatibles/farmacología , Fosfatos de Calcio/química , Diferenciación Celular , Citocinas , Propiedades de Superficie
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA