Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
1.
Biotechnol Bioeng ; 118(10): 3787-3798, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34110009

RESUMEN

Restoration of a wound is a common surgical procedure in clinic. Currently, the skin required for clinical use is taken from the patient's own body. However, it can be difficult to obtain enough skin sources for large-sized wounds and thus surgeons have started using commercial skin substitutes. The current commercial skin, which includes epidermis substitute, dermis substitute, and bilateral skin substitute, has been popularized in clinic. However, the application is limited by the occurrence of ischemia necrosis after transplantation. Recent studies suggest the use of pre-vascularized skin substitutes for wound healing is a promising area in the research field of skin tissue engineering. Pre-vascularization can be induced by changes in cultivation periods, exertion of mechanical stimuli, or coculture with endothelial cells and various factors. However, few methods could control the formation of vascular branches in engineering tissue in a self-assembly way. In this study, we use three-dimensional (3D) printing technology to confirm that a mechanical force can control the growth of blood vessels in the direction of mechanical stimulation with no branches, and that Yes-associated protein activity is involved in the regulatory progress. In vivo experiments verified that the blood vessels successfully function for blood circulation, and maintain the same direction. Results provide a theoretical basis for products of pre-vascularized skin tissues and other organs created by 3D bioprinting.


Asunto(s)
Bioimpresión , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Hidrogeles/química , Mecanotransducción Celular , Neovascularización Fisiológica , Impresión Tridimensional , Andamios del Tejido/química , Humanos , Piel/irrigación sanguínea , Piel/metabolismo , Cicatrización de Heridas
2.
J Mater Sci Mater Med ; 30(12): 135, 2019 Dec 04.
Artículo en Inglés | MEDLINE | ID: mdl-31802280

RESUMEN

The major challenge to treat the clinical adverse effects of long-segment urethra is in achieving viable tissue substitution. The substituted construct's properties-such as its resilience, contraction, and ability to minimize scar-stenosis formation should be considered. In the present work, a unique polyurethane-urea (PUU) fibrous membrane is fabricated by electrospinning. Then PUU was coated by collagen and formed the elasticity hydrogel after immersed in collagen solution. Meanwhile, the cPUU hydrogel exhibited a fibrous microstructure. This cPUU hydrogel had outstanding stretching property with 404 ± 40% elongation at break compared with traditional hydrogels, which satisfied the requirement of urethra. The cPUU hydrogel also supported the adhesion and growth of bladder smooth-muscle cells (BSMCs) in natural state cell morphology. Urethral defects in New Zealand male rabbits were repaired with cPUU seeded with BSMCs in vivo. After three months, more smooth-surface area of reconstructed urethral tissues was observed in the cPUU hydrogel-BMSCs groups compared with that of the control group. The luminal patency and the incidence of complications-including calculus formation, urinary fistula, and urethral-stricture occurrence were significantly lower in the cPUU group compared with that of the control group. Hence, cPUU fibrous hydrogels are promising scaffolds for application in urological tissue engineering.


Asunto(s)
Colágeno/química , Hidrogeles/química , Miocitos del Músculo Liso/fisiología , Poliuretanos/química , Urea/química , Enfermedades Uretrales/terapia , Animales , Materiales Biocompatibles , Masculino , Ensayo de Materiales , Conejos , Ingeniería de Tejidos , Andamios del Tejido , Vejiga Urinaria/citología
3.
Nano Lett ; 18(4): 2475-2484, 2018 04 11.
Artículo en Inglés | MEDLINE | ID: mdl-29565139

RESUMEN

Photodynamic therapy (PDT) is an oxygen-dependent light-triggered noninvasive therapeutic method showing many promising aspects in cancer treatment. For effective PDT, nanoscale carriers are often needed to realize tumor-targeted delivery of photosensitizers, which ideally should further target specific cell organelles that are most vulnerable to reactive oxygen species (ROS). Second, as oxygen is critical for PDT-induced cancer destruction, overcoming hypoxia existing in the majority of solid tumors is important for optimizing PDT efficacy. Furthermore, as PDT is a localized treatment method, achieving systemic antitumor therapeutic outcomes with PDT would have tremendous clinical values. Aiming at addressing the above challenges, we design a unique type of enzyme-encapsulated, photosensitizer-loaded hollow silica nanoparticles with rationally designed surface engineering as smart nanoreactors. Such nanoparticles with pH responsive surface coating show enhanced retention responding to the acidic tumor microenvironment and are able to further target mitochondria, the cellular organelle most sensitive to ROS. Meanwhile, decomposition of tumor endogenous H2O2 triggered by those nanoreactors would lead to greatly relieved tumor hypoxia, further favoring in vivo PDT. Moreover, by combining our nanoparticle-based PDT with check-point-blockade therapy, systemic antitumor immune responses could be achieved to kill nonirradiated tumors 1-2 cm away, promising for metastasis inhibition.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Preparaciones de Acción Retardada/química , Sistemas de Liberación de Medicamentos , Nanopartículas/química , Fármacos Fotosensibilizantes/administración & dosificación , Animales , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/terapia , Línea Celular Tumoral , Femenino , Peróxido de Hidrógeno/metabolismo , Concentración de Iones de Hidrógeno , Inmunoterapia/métodos , Ratones , Mitocondrias/metabolismo , Fotoquimioterapia/métodos , Fármacos Fotosensibilizantes/uso terapéutico , Especies Reactivas de Oxígeno/metabolismo , Dióxido de Silicio/química , Hipoxia Tumoral/efectos de los fármacos , Microambiente Tumoral/efectos de los fármacos
4.
Int J Mol Sci ; 20(9)2019 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-31027323

RESUMEN

Poly(methyl methacrylate) (PMMA) bone cements have been widely used in clinical practices. In order to enhance PMMA's imaging performance to facilitate surgical procedures, a supplementation of radiopaque agent is needed. However, PMMA bone cements are still facing problems of loosening and bacterial infection. In this study, a multifunctional coating to simultaneously encapsulate drug and prevent the infection of radiopaque agent has been developed. Barium sulfate (BaSO4), a common radiopaque agent, is used as a substrate material. We successfully fabricated porous BaSO4 microparticles, then modified with hexakis-(6-iodo-6-deoxy)-alpha-cyclodextrin (I-CD) and silver (Ag) to obtain porous BaSO4@PDA/I-CD/Ag microparticles. The porous nature and presence of PDA coating and I-CD on the surface of microparticles result in efficient loading and release of drugs such as protein. Meanwhile, the radiopacity of BaSO4@PDA/I-CD/Ag microparticles is enhanced by this multifunctional coating containing Ba, I and Ag. PMMA bone cements containing BaSO4@PDA/I-CD/Ag microparticles show 99% antibacterial rate against both Staphylococcus aureus (S. aureus) and Escherichia Coli (E. coli), yet without apparently affecting its biocompatibility. Together, this multifunctional coating possessing enhanced radiopacity, controlled drug delivery capability and exceptional antibacterial performance, may be a new way to modify radiopaque agents for bone cements.


Asunto(s)
Antibacterianos/química , Antibacterianos/farmacología , Sulfato de Bario/química , Cementos para Huesos/química , Polimetil Metacrilato/química , Materiales Biocompatibles/química , Materiales Biocompatibles/farmacología , Escherichia coli/efectos de los fármacos , Staphylococcus aureus/efectos de los fármacos
5.
Molecules ; 24(8)2019 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-31027249

RESUMEN

Fibrous hydrogel scaffolds have recently attracted increasing attention for tissue engineering applications. While a number of approaches have been proposed for fabricating microfibers, it remains difficult for current methods to produce materials that meet the essential requirements of being simple, flexible and bio-friendly. It is especially challenging to prepare cell-laden microfibers which have different structures to meet the needs of various applications using a simple device. In this study, we developed a facile two-flow microfluidic system, through which cell-laden hydrogel microfibers with various structures could be easily prepared in one step. Aiming to meet different tissue engineering needs, several types of microfibers with different structures, including single-layer, double-layer and hollow microfibers, have been prepared using an alginate-methacrylated gelatin composite hydrogel by merely changing the inner and outer fluids. Cell-laden single-layer microfibers were obtained by subsequently seeding mouse embryonic osteoblast precursor cells (MC3T3-E1) cells on the surface of the as-prepared microfibers. Cell-laden double-layer and hollow microfibers were prepared by directly encapsulating MC3T3-E1 cells or human umbilical vein endothelial cells (HUVECs) in the cores of microfibers upon their fabrication. Prominent proliferation of cells happened in all cell-laden single-layer, double-layer and hollow microfibers, implying potential applications for them in tissue engineering.


Asunto(s)
Hidrogeles/química , Microfluídica/métodos , Ingeniería de Tejidos/métodos , Animales , Células Endoteliales de la Vena Umbilical Humana/citología , Humanos , Ratones
6.
Int J Mol Sci ; 19(6)2018 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-29895809

RESUMEN

Poly (methyl methacrylate) (PMMA)-based bone cements are the most commonly used injectable orthopedic materials due to their excellent injectability and mechanical properties. However, their poor biocompatibility and excessive stiffness may cause complications such as aseptic implant loosening and stress shielding. In this study, we aimed to develop a new type of partially biodegradable composite bone cement by incorporating magnesium (Mg) microspheres, known as "Mg sacrifices" (MgSs), in the PMMA matrix. Being sensitive to the physiological environment, the MgSs in PMMA could gradually degrade to produce bioactive Mg ions and, meanwhile, result in an interconnected macroporous structure within the cement matrix. The mechanical properties, solidification, and biocompatibility, both in vitro and in vivo, of PMMA⁻Mg bone cement were characterized. Interestingly, the incorporation of Mg microspheres did not markedly affect the mechanical strength of bone cement. However, the maximum temperature upon setting of bone cement decreased. This partially biodegradable composite bone cement showed good biocompatibility in vitro. In the in vivo study, considerable bony ingrowth occurred in the pores upon MgS degradation. Together, the findings from this study indicate that such partially biodegradable PMMA⁻Mg composite may be ideal bone cement for minimally invasive orthopedic surgeries such as vertebroplasty and kyphoplasty.


Asunto(s)
Cementos para Huesos/química , Polimetil Metacrilato/química , Magnesio/química , Oseointegración/fisiología , Temperatura
7.
Int J Mol Sci ; 17(8)2016 Aug 09.
Artículo en Inglés | MEDLINE | ID: mdl-27517902

RESUMEN

Tissue engineering-based urethral replacement holds potential for repairing large segmental urethral defects, which remains a great challenge at present. This study aims to explore the potential of combining biodegradable poly(l-lactide) (PLLA)/poly(ethylene glycol) (PEG) scaffolds and human amniotic mesenchymal cells (hAMSCs) for repairing urethral defects. PLLA/PEG fibrous scaffolds with various PEG fractions were fabricated via electrospinning. The scaffolds were then seeded with hAMSCs prior to implantation in New Zealand male rabbits that had 2.0 cm-long defects in the urethras. The rabbits were randomly divided into three groups. In group A, hAMSCs were grown on PLLA/PEG scaffolds for two days and then implanted to the urethral defects. In group B, only the PLLA/PEG scaffolds were used to rebuild the rabbit urethral defect. In group C, the urethral defect was reconstructed using a regular urethral reparation technique. The repair efficacy was compared among the three groups by examining the urethral morphology, tissue reconstruction, luminal patency, and complication incidence (including calculus formation, urinary fistula, and urethral stricture) using histological evaluation and urethral radiography methods. Findings from this study indicate that hAMSCs-loaded PLLA/PEG scaffolds resulted in the best urethral defect repair in rabbits, which predicts the promising application of a tissue engineering approach for urethral repair.


Asunto(s)
Lactatos/química , Células Madre Mesenquimatosas/citología , Polietilenglicoles/química , Ingeniería de Tejidos/métodos , Andamios del Tejido/química , Animales , Materiales Biocompatibles/química , Humanos , Inmunohistoquímica , Masculino , Poliésteres/química , Conejos
8.
J Mater Sci Mater Med ; 26(4): 160, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25786398

RESUMEN

A layered gelatin-chitosan hydrogel with graded composition was prepared via photocrosslinking to simulate the polysaccharide/collagen composition of the natural tissue and mimic the multi-layered gradient structure of the cartilage-bone interface tissue. Firstly, gelatin and carboxymethyl chitosan were reacted with glycidyl methacrylate (GMA) to obtain methacrylated gelatin (Gtn-GMA) and carboxymethyl chitosan (CS-GMA). Then, the mixed solutions of Gtn-GMA in different methacrylation degrees with CS-GMA were prepared to form the superficial, transitional and deep layers of the hydrogel, respectively under the irradiation of ultraviolet light, while polyhedral oligomeric silsesquioxane was introduced in the deep layer to improve the mechanical properties. Results suggested that the pore sizes of the superficial, transitional and deep layers of the layered hydrogel were 115 ± 30, 94 ± 34, 51 ± 12 µm, respectively and their porosities were all higher than 80 %. The compressive strengths of them were 165 ± 54, 565 ± 50 and 993 ± 108 kPa, respectively and the strain of the gradient hydrogel decreased along the thickness direction, similar to the natural tissue. The in vitro cytotoxicity results showed that the hydrogel had good cytocompatibility and the in vivo repair results of osteochondral defect demonstrated remarkable recovery by using the gradient gelatin-chitosan hydrogel, especially when the hydrogel loading transforming growth factor-ß1. Therefore, it was suggested that the prepared layered gelatin-chitosan hydrogel in this study could be potentially used to promote cartilage-bone interface tissue repair.


Asunto(s)
Materiales Biomiméticos/química , Quitosano/química , Quitosano/uso terapéutico , Fracturas del Cartílago/terapia , Gelatina/química , Hidrogeles/química , Animales , Materiales Biomiméticos/uso terapéutico , Quitosano/efectos de la radiación , Fuerza Compresiva , Reactivos de Enlaces Cruzados/química , Reactivos de Enlaces Cruzados/efectos de la radiación , Fracturas del Cartílago/patología , Gelatina/efectos de la radiación , Gelatina/uso terapéutico , Hidrogeles/efectos de la radiación , Hidrogeles/uso terapéutico , Luz , Ensayo de Materiales , Fotoquímica , Porosidad , Conejos , Resultado del Tratamiento
9.
Adv Mater ; 36(2): e2305468, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37681640

RESUMEN

Intervertebral disc degeneration (IVDD) is a global public health issue. The injury of annulus fibrosus (AF) caused by acupuncture or discectomy can trigger IVDD again. However, there is currently no suitable method for treating AF injury. In this study, the high-strength smart microneedles (MNs) which can penetrate the AF tissue through a local and minimally invasive method, and achieve remote control of speeded-up release of the drug and hyperthermia by the Near Infrared is developed. The PDA/GelMA composite MNs loaded with diclofenac sodium are designed to extracellularly "offend" the inflammatory microenvironment and mitigate damage to cells, and intracellularly increase the level of cytoprotective heat shock proteins to enhance the defense against the hostile microenvironment, achieving "offensive and defensive" effects. In vitro experiments demonstrate that the synergistic treatment of photothermal therapy and anti-inflammation effectively reduces inflammation, inhibits cell apoptosis, and promotes the synthesis of the extracellular matrix (ECM). In vivo experiments show that the MNs mitigate the inflammatory response, promote ECM deposition, reduce the level of apoptosis, and restore the biomechanical properties of the intervertebral disc (IVD) in rats. Overall, this high-strength smart MNs display promising "offensive and defensive" effects that can provide a new strategy for IVD repair.


Asunto(s)
Degeneración del Disco Intervertebral , Disco Intervertebral , Ratas , Animales , Disco Intervertebral/metabolismo , Degeneración del Disco Intervertebral/metabolismo , Degeneración del Disco Intervertebral/cirugía , Matriz Extracelular/metabolismo , Inflamación/metabolismo , Antiinflamatorios/metabolismo
10.
J Orthop Translat ; 47: 132-143, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-39027342

RESUMEN

Background: A pivotal determinant for the success of tissue regeneration lies in the establishment of sufficient vasculature. Utilizing autologous tissue grafts from donors offers the dual advantage of mitigating the risk of disease transmission and circumventing the necessity for post-transplant immunosuppression, rendering it an exemplary vascularization strategy. Among the various potential autologous donors, adipose tissue emerges as a particularly auspicious source, being both widely available and compositionally rich. Notably, adipose-derived microvascular fragments (ad-MVFs) are a promising candidate for vascularization. ad-MVFs can be isolated from adipose tissue in a short period of time and show high vascularized capacity. In this study, we extracted ad-MVFs from adipose tissue and utilized their strong angiogenic ability to accelerate bone repair by promoting vascularization. Methods: ad-MVFs were extracted from the rat epididymis using enzymatic hydrolysis. To preserve the integrity of the blood vessels, gelatin methacryloyl (GelMA) hydrogel was chosen as the carrier for ad-MVFs in three-dimensional (3D) culture. The ad-MVFs were cultured directly on the well plates for two-dimensional (2D) culture as a control. The morphology of ad-MVFs was observed under both 2D and 3D cultures, and the release levels of vascular endothelial growth factor (VEGF) and bone morphogenetic protein 2 (BMP-2) were assessed under both culture conditions. In vitro studies investigated the impact of ad-MVFs/GelMA hydrogel on the toxicity, osteoblastic activity, and mineralization of rat bone marrow mesenchymal stem cells (rBMSCs), along with the examination of osteogenic gene and protein expression. In vivo experiments involved implanting the ad-MVFs/GelMA hydrogel into critical-size skull defects in rats, and its osteogenic ability was evaluated through radiographic and histological methods. Results: ad-MVFs were successfully isolated from rat adipose tissue. When cultured under 2D conditions, ad-MVFs exhibited a gradual disintegration and loss of their original vascular morphology. Compared with 2D culture, ad-MVFs can not only maintain the original vascular morphology, but also connect into a network in hydrogel under 3D culture condition. Moreover, the release levels of VEGF and BMP-2 were significantly higher than those in 2D culture. Moreover, the ad-MVFs/GelMA hydrogel exhibited superior osteoinductive activity. After implanting into the skull defect of rats, the ad-MVFs/GelMA hydrogel showed obvious effects for angiogenesis and osteogenesis. The translational potential of this article: The utilization of autologous adipose tissue as a donor presents a more direct route toward clinical translation. Anticipated future clinical applications envision the transformation of discarded adipose tissue into a valuable resource for personalized tissue repair, thereby realizing a paradigm shift in the utilization of this abundant biological material.

11.
Gels ; 10(5)2024 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-38786250

RESUMEN

Intervertebral disc degeneration (IVDD) is a worldwide disease that causes low back pain and reduces quality of life. Biotherapeutic strategies based on tissue engineering alternatives, such as intervertebral disc scaffolds, supplemented by drug-targeted therapy have brought new hope for IVDD. In this study, to explore the role and mechanism of MnO2/GelMA composite hydrogels in alleviating IVDD, we prepared composite hydrogels with MnO2 and methacrylate gelatin (GelMA) and characterized them using compression testing and transmission electron microscopy (TEM). Annulus fibrosus cells (AFCs) were cultured in the composite hydrogels to verify biocompatibility by live/dead and cytoskeleton staining. Cell viability assays and a reactive oxygen species (ROS) probe were used to analyze the protective effect of the composite hydrogels under oxidative damage. To explore the mechanism of improving the microenvironment, we detected the expression levels of antioxidant and autophagy-related genes and proteins by qPCR and Western blotting. We found that the MnO2/GelMA composite hydrogels exhibited excellent biocompatibility and a porous structure, which promoted cell proliferation. The addition of MnO2 nanoparticles to GelMA cleared ROS in AFCs and induced the expression of antioxidant and cellular autophagy through the common SIRT1/NRF2 pathway. Therefore, the MnO2/GelMA composite hydrogels, which can improve the disc microenvironment through scavenging intracellular ROS and resisting oxidative damage, have great application prospects in the treatment of IVDD.

12.
Biomater Sci ; 12(5): 1239-1248, 2024 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-38231128

RESUMEN

The regeneration of tendon and bone junctions (TBJs), a fibrocartilage transition zone between tendons and bones, is a challenge due to the special triphasic structure. In our study, a silk fibroin (SF)-based triphasic scaffold consisting of aligned type I collagen (Col I), transforming growth factor ß (TGF-ß), and hydroxyapatite (HA) was fabricated to mimic the compositional gradient feature of the native tendon-bone architecture. Rat tendon-derived stem cells (rTDSCs) were loaded on the triphasic SF scaffold, and the high cell viability suggested that the scaffold presents good biocompatibility. Meanwhile, increased expressions of tenogenic-, chondrogenic-, and osteogenic-related genes in the TBJs were observed. The in vivo studies of the rTDSC-seeded scaffold in a rat TBJ rupture model showed tendon tissue regeneration with a clear transition zone within 8 weeks of implantation. These results indicated that the biomimetic triphasic SF scaffolds seeded with rTDSCs have great potential to be applied in TBJ regeneration.


Asunto(s)
Fibroínas , Ratas , Animales , Fibroínas/química , Andamios del Tejido/química , Biomimética , Tendones , Células Madre , Ingeniería de Tejidos/métodos , Seda/química
13.
Front Bioeng Biotechnol ; 11: 1205792, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37469449

RESUMEN

The incidence of tissue and organ damage caused by various diseases is increasing worldwide. Tissue engineering is a promising strategy of tackling this problem because of its potential to regenerate or replace damaged tissues and organs. The biochemical and biophysical cues of biomaterials can stimulate and induce biological activities such as cell adhesion, proliferation and differentiation, and ultimately achieve tissue repair and regeneration. Micro/nano materials are a special type of biomaterial that can mimic the microstructure of tissues on a microscopic scale due to its precise construction, further providing scaffolds with specific three-dimensional structures to guide the activities of cells. The study and application of biomimetic micro/nano-materials have greatly promoted the development of tissue engineering. This review aims to provide an overview of the different types of micro/nanomaterials, their preparation methods and their application in tissue regeneration.

14.
Bioact Mater ; 19: 581-593, 2023 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-35600980

RESUMEN

Annulus fibrosus (AF) repair remains a challenge because of its limited self-healing ability. Endogenous repair strategies combining scaffolds and growth factors show great promise in AF repair. Although the unique and beneficial characteristics of decellularized extracellular matrix (ECM) in tissue repair have been demonstrated, the poor mechanical property of ECM hydrogels largely hinders their applications in tissue regeneration. In the present study, we combined polyethylene glycol diacrylate (PEGDA) and decellularized annulus fibrosus matrix (DAFM) to develop an injectable, photocurable hydrogel for AF repair. We found that the addition of PEGDA markedly improved the mechanical strength of DAFM hydrogels while maintaining their porous structure. Transforming growth factor-ß1 (TGF-ß1) was further incorporated into PEGDA/DAFM hydrogels, and it could be continuously released from the hydrogel. The in vitro experiments showed that TGF-ß1 facilitated the migration of AF cells. Furthermore, PEGDA/DAFM/TGF-ß1 hydrogels supported the adhesion, proliferation, and increased ECM production of AF cells. In vivo repair performance of the hydrogels was assessed using a rat AF defect model. The results showed that the implantation of PEGDA/DAFM/TGF-ß1 hydrogels effectively sealed the AF defect, prevented nucleus pulposus atrophy, retained disc height, and partially restored the biomechanical properties of disc. In addition, the implanted hydrogel was infiltrated by cells resembling AF cells and well integrated with adjacent AF tissue. In summary, findings from this study indicate that TGF-ß1-supplemented DAFM hydrogels hold promise for AF repair.

15.
Macromol Biosci ; 23(10): e2300105, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37247409

RESUMEN

The present study describes a silk microfiber reinforced meniscus scaffold (SMRMS) with hierarchical fibrous and porous structure made from silk fibroin (SF) and wool keratin (WK) using electrospinning and freeze-drying technology. This study focuses on the morphology, secondary structure, mechanical properties, and water absorption properties of the scaffold. The cytotoxicity and biocompatibility of SMRMS are assessed in vivo and in vitro. The scaffold shows hierarchical fibrous and porous structure, hierarchical pore size distribution (ranges from 50 to 650 µm), robust mechanical properties (compression strength can reach at 2.8 MPa), and stable biodegradability. A positive growth condition revealed by in vitro cytotoxicity testing indicates that the scaffold is not hazardous to cells. In vivo assessments of biocompatibility reveal that only a mild inflammatory reaction is present in implanted rat tissue. Meniscal scaffold made of SF/WK composite shows a potential application prospect in the meniscal repair engineering field with its development.

16.
Biomater Transl ; 4(1): 27-40, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37206307

RESUMEN

Cell sheet-based scaffold-free technology holds promise for tissue engineering applications and has been extensively explored during the past decades. However, efficient harvest and handling of cell sheets remain challenging, including insufficient extracellular matrix content and poor mechanical strength. Mechanical loading has been widely used to enhance extracellular matrix production in a variety of cell types. However, currently, there are no effective ways to apply mechanical loading to cell sheets. In this study, we prepared thermo-responsive elastomer substrates by grafting poly(N-isopropyl acrylamide) (PNIPAAm) to poly(dimethylsiloxane) (PDMS) surfaces. The effect of PNIPAAm grafting yields on cell behaviours was investigated to optimize surfaces suitable for cell sheet culturing and harvesting. Subsequently, MC3T3-E1 cells were cultured on the PDMS-g-PNIPAAm substrates under mechanical stimulation by cyclically stretching the substrates. Upon maturation, the cell sheets were harvested by lowering the temperature. We found that the extracellular matrix content and thickness of cell sheet were markedly elevated upon appropriate mechanical conditioning. Reverse transcription quantitative polymerase chain reaction and Western blot analyses further confirmed that the expression of osteogenic-specific genes and major matrix components were up-regulated. After implantation into the critical-sized calvarial defects of mice, the mechanically conditioned cell sheets significantly promoted new bone formation. Findings from this study reveal that thermo-responsive elastomer, together with mechanical conditioning, can potentially be applied to prepare high-quality cell sheets for bone tissue engineering.

17.
Research (Wash D C) ; 6: 0021, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37040486

RESUMEN

The critical factor determining the in vivo effect of bone repair materials is the microenvironment, which greatly depends on their abilities to promote vascularization and bone formation. However, implant materials are far from ideal candidates for guiding bone regeneration due to their deficient angiogenic and osteogenic microenvironments. Herein, a double-network composite hydrogel combining vascular endothelial growth factor (VEGF)-mimetic peptide with hydroxyapatite (HA) precursor was developed to build an osteogenic microenvironment for bone repair. The hydrogel was prepared by mixing acrylated ß-cyclodextrins and octacalcium phosphate (OCP), an HA precursor, with gelatin solution, followed by ultraviolet photo-crosslinking. To improve the angiogenic potential of the hydrogel, QK, a VEGF-mimicking peptide, was loaded in acrylated ß-cyclodextrins. The QK-loaded hydrogel promoted tube formation of human umbilical vein endothelial cells and upregulated the expression of angiogenesis-related genes, such as Flt1, Kdr, and VEGF, in bone marrow mesenchymal stem cells. Moreover, QK could recruit bone marrow mesenchymal stem cells. Furthermore, OCP in the composite hydrogel could be transformed into HA and release calcium ions facilitating bone regeneration. The double-network composite hydrogel integrated QK and OCP showed obvious osteoinductive activity. The results of animal experiments showed that the composite hydrogel enhanced bone regeneration in skull defects of rats, due to perfect synergistic effects of QK and OCP on vascularized bone regeneration. In summary, improving the angiogenic and osteogenic microenvironments by our double-network composite hydrogel shows promising prospects for bone repair.

18.
ACS Nano ; 17(8): 7645-7661, 2023 04 25.
Artículo en Inglés | MEDLINE | ID: mdl-37022700

RESUMEN

The excessive reactive oxygen species (ROS) level, inflammation, and weak tissue regeneration ability after annulus fibrosus (AF) injury constitute an unfavorable microenvironment for AF repair. AF integrity is crucial for preventing disc herniation after discectomy; however, there is no effective way to repair the AF. Herein, a composite hydrogel integrating properties of antioxidant, anti-inflammation, and recruitment of AF cells is developed through adding mesoporous silica nanoparticles modified by ceria and transforming growth factor ß3 (TGF-ß3) to the hydrogels. The nanoparticle loaded gelatin methacrylate/hyaluronic acid methacrylate composite hydrogels eliminate ROS and induce anti-inflammatory M2 type macrophage polarization. The released TGF-ß3 not only plays a role in recruiting AF cells but is also responsible for promoting extracellular matrix secretion. The composite hydrogels can be solidified in situ in the defect area to effectively repair AF in rats. The strategies targeting endogenous ROS removal and improving the regenerative microenvironment by the nanoparticle-loaded composite hydrogels have potential applications in AF repair and intervertebral disc herniation prevention.


Asunto(s)
Anillo Fibroso , Ratas , Animales , Especies Reactivas de Oxígeno/metabolismo , Factor de Crecimiento Transformador beta3/metabolismo , Factor de Crecimiento Transformador beta3/farmacología , Matriz Extracelular/metabolismo , Hidrogeles/farmacología
19.
Biomater Transl ; 4(2): 104-114, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38283921

RESUMEN

There is a high demand for bespoke grafts to replace damaged or malformed bone and cartilage tissue. Three-dimensional (3D) printing offers a method of fabricating complex anatomical features of clinically relevant sizes. However, the construction of a scaffold to replicate the complex hierarchical structure of natural tissues remains challenging. This paper reports a novel biofabrication method that is capable of creating intricately designed structures of anatomically relevant dimensions. The beneficial properties of the electrospun fibre meshes can finally be realised in 3D rather than the current promising breakthroughs in two-dimensional (2D). The 3D model was created from commercially available computer-aided design software packages in order to slice the model down into many layers of slices, which were arrayed. These 2D slices with each layer of a defined pattern were laser cut, and then successfully assembled with varying thicknesses of 100 µm or 200 µm. It is demonstrated in this study that this new biofabrication technique can be used to reproduce very complex computer-aided design models into hierarchical constructs with micro and nano resolutions, where the clinically relevant sizes ranging from a simple cube of 20 mm dimension, to a more complex, 50 mm-tall human ears were created. In-vitro cell-contact studies were also carried out to investigate the biocompatibility of this hierarchal structure. The cell viability on a micromachined electrospun polylactic-co-glycolic acid fibre mesh slice, where a range of hole diameters from 200 µm to 500 µm were laser cut in an array where cell confluence values of at least 85% were found at three weeks. Cells were also seeded onto a simpler stacked construct, albeit made with micromachined poly fibre mesh, where cells can be found to migrate through the stack better with collagen as bioadhesives. This new method for biofabricating hierarchical constructs can be further developed for tissue repair applications such as maxillofacial bone injury or nose/ear cartilage replacement in the future.

20.
Sci Adv ; 9(21): eadf8645, 2023 05 26.
Artículo en Inglés | MEDLINE | ID: mdl-37235658

RESUMEN

Implant-associated infection is a major threat affecting the success of orthopedic surgeries. Although various materials scavenge bacteria by generating reactive oxygen species (ROS), the intrinsic inability of ROS to distinguish bacteria from cells notably limits the therapeutic effects. Here, we found that the arginine carbon dots (Arg-CDs) that were transformed from arginine exhibited supreme antibacterial and osteoinductive activity. We further designed the Schiff base bond between Arg-CDs and aldehyde hyaluronic acid/gelatin methacryloyl (HG) hydrogel to release Arg-CDs in response to the acidic bone injury microenvironment. The free Arg-CDs could selectively kill bacteria by generating excessive ROS. Furthermore, the Arg-CD-loaded HG composite hydrogel showed excellent osteoinductive activity through inducing the M2 polarization of macrophages by up-regulating interleukin-10 (Il10) expression. Together, our findings revealed that transformation of the arginine into zero-dimensional Arg-CDs could endow the material with exceptional antibacterial and osteoinductive activity, favoring the regeneration of infectious bone.


Asunto(s)
Arginina , Nanoestructuras , Especies Reactivas de Oxígeno , Arginina/farmacología , Antibacterianos/química , Hidrogeles/farmacología , Hidrogeles/uso terapéutico , Carbono/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA