Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
ACS Nano ; 18(10): 7580-7595, 2024 Mar 12.
Article in English | MEDLINE | ID: mdl-38422400

ABSTRACT

The lack of both digital light processing (DLP) compatible and biocompatible photopolymers, along with inappropriate material properties required for wearable sensor applications, substantially hinders the employment of DLP 3D printing in the fabrication of multifunctional hydrogels. Herein, we discovered and implemented a photoreactive poloxamer derivative, Pluronic F-127 diacrylate, which overcomes these limitations and is optimized to achieve DLP 3D printed micelle-based hydrogels with high structural complexity, resolution, and precision. In addition, the dehydrated hydrogels exhibit a shape-memory effect and are conformally attached to the geometry of the detection point after rehydration, which implies the 4D printing characteristic of the fabrication process and is beneficial for the storage and application of the device. The excellent cytocompatibility and in vivo biocompatibility further strengthen the potential application of the poloxamer micelle-based hydrogels as a platform for multifunctional wearable systems. After processing them with a lithium chloride (LiCl) solution, multifunctional conductive ionic hydrogels with antifreezing and antiswelling properties along with good transparency and water retention are easily prepared. As capacitive flexible sensors, the DLP 3D printed micelle-based hydrogel devices exhibit excellent sensitivity, cycling stability, and durability in detecting multimodal deformations. Moreover, the DLP 3D printed conductive hydrogels are successfully applied as real-time human motion and tactile sensors with satisfactory sensing performances even in a -20 °C low-temperature environment.


Subject(s)
Micelles , Wearable Electronic Devices , Humans , Poloxamer , Electric Conductivity , Hydrogels , Printing, Three-Dimensional
2.
Acta Biomater ; 168: 78-112, 2023 09 15.
Article in English | MEDLINE | ID: mdl-37516417

ABSTRACT

As one of the long-established and necessary medical devices, surgical sutures play an essentially important role in the closing and healing of damaged tissues and organs postoperatively. The recent advances in multiple disciplines, like materials science, engineering technology, and biomedicine, have facilitated the generation of various innovative surgical sutures with humanization and multi-functionalization. For instance, the application of numerous absorbable materials is assuredly a marvelous progression in terms of surgical sutures. Moreover, some fantastic results from recent laboratory research cannot be ignored either, ranging from the fiber generation to the suture structure, as well as the suture modification, functionalization, and even intellectualization. In this review, the suture materials, including natural or synthetic polymers, absorbable or non-absorbable polymers, and metal materials, were first introduced, and then their advantages and disadvantages were summarized. Then we introduced and discussed various fiber fabrication strategies for the production of surgical sutures. Noticeably, advanced nanofiber generation strategies were highlighted. This review further summarized a wide and diverse variety of suture structures and further discussed their different features. After that, we covered the advanced design and development of surgical sutures with multiple functionalizations, which mainly included surface coating technologies and direct drug-loading technologies. Meanwhile, the review highlighted some smart and intelligent sutures that can monitor the wound status in a real-time manner and provide on-demand therapies accordingly. Furthermore, some representative commercial sutures were also introduced and summarized. At the end of this review, we discussed the challenges and future prospects in the field of surgical sutures in depth. This review aims to provide a meaningful reference and guidance for the future design and fabrication of innovative surgical sutures. STATEMENT OF SIGNIFICANCE: This review article introduces the recent advances of surgical sutures, including material selection, fiber morphology, suture structure and construction, as well as suture modification, functionalization, and even intellectualization. Importantly, some innovative strategies for the construction of multifunctional sutures with predetermined biological properties are highlighted. Moreover, some important commercial suture products are systematically summarized and compared. This review also discusses the challenges and future prospects of advanced sutures in a deep manner. In all, this review is expected to arouse great interest from a broad group of readers in the fields of multifunctional biomaterials and regenerative medicine.


Subject(s)
Biocompatible Materials , Regenerative Medicine , Biocompatible Materials/chemistry , Wound Healing , Sutures , Polymers/chemistry , Suture Techniques
3.
ACS Nano ; 17(4): 3847-3864, 2023 02 28.
Article in English | MEDLINE | ID: mdl-36779870

ABSTRACT

Postoperative abdominal adhesions are a common problem after surgery and can produce serious complications. Current antiadhesive strategies focus mostly on physical barriers and are unsatisfactory and inefficient. In this study, we designed and synthesized advanced injectable cream-like hydrogels with multiple functionalities, including rapid gelation, self-healing, antioxidation, anti-inflammation, and anti-cell adhesion. The multifunctional hydrogels were facilely formed by the conjugation reaction of epigallocatechin-3-gallate (EGCG) and hyaluronic acid (HA)-based microgels and poly(vinyl alcohol) (PVA) based on the dynamic boronic ester bond. The physicochemical properties of the hydrogels including antioxidative and anti-inflammatory activities were systematically characterized. A mouse cecum-abdominal wall adhesion model was implemented to investigate the efficacy of our microgel-based hydrogels in preventing postoperative abdominal adhesions. The hydrogels, with a high molecular weight HA, significantly decreased the inflammation, oxidative stress, and fibrosis and reduced the abdominal adhesion formation, compared to the commercial Seprafilm group or Injury-only group. Label-free quantitative proteomics analysis demonstrated that S100A8 and S100A9 expressions were associated with adhesion formation; the microgel-containing hydrogels inhibited these expressions. The microgel-containing hydrogels with multifunctionality decreased the formation of postoperative intra-abdominal adhesions in a murine model, demonstrating promise for clinical applications.


Subject(s)
Abdominal Wall , Microgels , Mice , Animals , Hydrogels/chemistry , Abdominal Wall/pathology , Abdominal Wall/surgery , Tissue Adhesions/prevention & control , Tissue Adhesions/pathology , Inflammation/pathology
4.
Appl Mater Today ; 27: 101473, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35434263

ABSTRACT

The pandemic of the coronavirus disease 2019 (COVID-19) has made biotextiles, including face masks and protective clothing, quite familiar in our daily lives. Biotextiles are one broad category of textile products that are beyond our imagination. Currently, biotextiles have been routinely utilized in various biomedical fields, like daily protection, wound healing, tissue regeneration, drug delivery, and sensing, to improve the health and medical conditions of individuals. However, these biotextiles are commonly manufactured with fibers with diameters on the micrometer scale (> 10 µm). Recently, nanofibrous materials have aroused extensive attention in the fields of fiber science and textile engineering because the fibers with nanoscale diameters exhibited obviously superior performances, such as size and surface/interface effects as well as optical, electrical, mechanical, and biological properties, compared to microfibers. A combination of innovative electrospinning techniques and traditional textile-forming strategies opens a new window for the generation of nanofibrous biotextiles to renew and update traditional microfibrous biotextiles. In the last two decades, the conventional electrospinning device has been widely modified to generate nanofiber yarns (NYs) with the fiber diameters less than 1000 nm. The electrospun NYs can be further employed as the primary processing unit for manufacturing a new generation of nano-textiles using various textile-forming strategies. In this review, starting from the basic information of conventional electrospinning techniques, we summarize the innovative electrospinning strategies for NY fabrication and critically discuss their advantages and limitations. This review further covers the progress in the construction of electrospun NY-based nanotextiles and their recent applications in biomedical fields, mainly including surgical sutures, various scaffolds and implants for tissue engineering, smart wearable bioelectronics, and their current and potential applications in the COVID-19 pandemic. At the end, this review highlights and identifies the future needs and opportunities of electrospun NYs and NY-based nanotextiles for clinical use.

5.
Biomed Microdevices ; 23(2): 22, 2021 04 05.
Article in English | MEDLINE | ID: mdl-33821331

ABSTRACT

Alginate hydrogel beads are a common platform for generating 3D cell cultures in biomedical research. Simple methods for bead generation using a manual pipettor or syringe are low-throughput and produce beads showing high variability in size and shape. To address these challenges, we designed a 3D printed bead generator that uses an airflow to cleave beads from a stream of hydrogel solution. The performance of the proposed alginate bead generator was evaluated by changing the volume flow rates of alginate (QAlg) and air (QA), the diameter of device nozzle (d) and the concentration of alginate gel solution (C). We identified that the diameter of beads (D = 0.9 -2.8 mm) can be precisely controlled by changing QA and d. Also the bead generation frequency (f) can be tuned by changing QAlg. Finally, we demonstrated that viability and biological function (pericellular matrix deposition) of chondrocytes were not adversely affected by high f using this bead generator. Because 3D printing is becoming a more accessible technique, our unique design will allow greater access to average biomedical research laboratories, STEM education and industries in cost- and time-effective manner.


Subject(s)
Alginates , Cell Culture Techniques , Hydrogels , Printing, Three-Dimensional
6.
Adv Healthc Mater ; 10(12): e2100180, 2021 06.
Article in English | MEDLINE | ID: mdl-33890428

ABSTRACT

Despite significant progress in understanding the disease mechanism of traumatic brain injury (TBI), promising preclinical therapeutics have seldom been translated into successful clinical outcomes, partially because the model animals have physiological and functional differences in the central nervous system (CNS) compared to humans. Human relevant models are thus urgently required. Here, an in vitro mild TBI (mTBI) modeling system is reported based on 3D cultured human induced pluripotent stem cells (iPSC) derived neural progenitor cells (iPSC-NPCs) to evaluate consequences of single and repetitive mTBI using a 3D printed mini weight-drop impact device. Computational simulation is performed to understand the single/cumulative effects of weight-drop impact on the NPC differentiated neurospheres. Experimental results reveal that neurospheres show reactive astrogliosis and glial scar formation after repetitive (10 hits) mild impacts, while no astrocyte activation is found after one or two mild impacts. A 3D co-culture model of human microglia cells with neurospheres is further developed. It is found that astrocyte response is promoted even after two mild impacts, possibly caused by the chronic neuroinflammation after microglia activation. The in vitro mTBI modeling system recapitulates several hallmarks of the brain impact injury and might serve as a good platform for future drug screening.


Subject(s)
Brain Concussion , Induced Pluripotent Stem Cells , Neural Stem Cells , Animals , Humans , Neural Stem Cells/transplantation , Neurogenesis , Printing, Three-Dimensional
7.
J Mater Chem B ; 9(35): 7182-7195, 2021 09 15.
Article in English | MEDLINE | ID: mdl-33651063

ABSTRACT

Due to their intrinsic injectable and self-healing characteristics, dynamic hydrogels, based on dynamic covalent bonds, have gained a great attention. In this study, a novel dynamic hydrogel based on the boronic ester dynamic covalent bond is facilely developed using phenylboronic acid-modified hyaluronic acid (HA-PBA) and plant-derived polyphenol-tannic acid (TA). The dynamic hydrogel gelated quickly under mild conditions and had favorable viscoelastic properties with good self-healing and shear-thinning capabilities. Moreover, the simultaneous utilization of TA as a reductant for the green synthesis of silver nanoparticles (AgNP) inspired the preparation of a TA-reduced AgNP hybrid dynamic hydrogel with potent and broad-spectrum antibacterial activities. The dynamic hydrogels could also be applied for pH- and reactive oxygen species (ROS)-responsive release of loaded protein molecules without showing evident cytotoxicity and hemolysis in vitro. In addition, the dynamic hydrogels showed the anti-oxidative properties of high free radical and ROS scavenging capacity, which was verified by the DPPH (2,2-diphenyl-1-picryl-hydrazyl-hydrate) free radical assay and ROS fluorescence staining. Overall, this novel class of cytocompatible, self-healing, dual stimuli responsive, antibacterial, anti-oxidative, and injectable hydrogels could be promising as a wound dressing for chronic wound healing.


Subject(s)
Anti-Bacterial Agents/pharmacology , Antioxidants/pharmacology , Biocompatible Materials/pharmacology , Pseudomonas aeruginosa/drug effects , Tannins/pharmacology , Wound Healing/drug effects , Animals , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/chemistry , Antioxidants/chemical synthesis , Antioxidants/chemistry , Biocompatible Materials/chemical synthesis , Biocompatible Materials/chemistry , Biphenyl Compounds/antagonists & inhibitors , Boronic Acids/chemistry , Boronic Acids/pharmacology , Cells, Cultured , Erythrocytes/drug effects , Hemolysis/drug effects , Hyaluronic Acid/chemistry , Hyaluronic Acid/pharmacology , Hydrogels/chemical synthesis , Hydrogels/chemistry , Hydrogels/pharmacology , Materials Testing , Mice , Microbial Sensitivity Tests , Molecular Conformation , Particle Size , Picrates/antagonists & inhibitors , Polyphenols/chemistry , Polyphenols/pharmacology , Tannins/chemistry
8.
Biomaterials ; 240: 119832, 2020 05.
Article in English | MEDLINE | ID: mdl-32113114

ABSTRACT

The small intestine (SI) is difficult to regenerate or reconstruct due to its complex structure and functions. Recent developments in stem cell research, advanced engineering technologies, and regenerative medicine strategies bring new hope of solving clinical problems of the SI. This review will first summarize the structure, function, development, cell types, and matrix components of the SI. Then, the major cell sources for SI regeneration are introduced, and state-of-the-art biofabrication technologies for generating engineered SI tissues or models are overviewed. Furthermore, in vitro models and in vivo transplantation, based on intestinal organoids and tissue engineering, are highlighted. Finally, current challenges and future perspectives are discussed to help direct future applications for SI repair and regeneration.


Subject(s)
Bioprinting , Intestine, Small , Regeneration , Tissue Engineering , Tissue Scaffolds
9.
Carbohydr Polym ; 233: 115803, 2020 Apr 01.
Article in English | MEDLINE | ID: mdl-32059877

ABSTRACT

In this study, an injectable and self-healing hydrogel based on the boronic ester dynamic covalent bond between phenylboronic acid modified hyaluronic acid (HA-PBA) and the commercially available poly (vinyl alcohol) (PVA) is prepared and should have multi-functions for biomedical applications. The hydrogels were rapidly formed under mild conditions, and the rheological properties and in vitro degradation were systematically characterized. The HA-based hydrogels possessed good injectability and self-healing properties because of the dynamic bond. Moreover, due to the sensitivity of boronic ester to the biologically relevant concentration of hydrogen peroxide (H2O2), a major reactive oxygen species (ROS), the injectable hydrogel could be used as a H2O2/ROS responsive drug delivery system. The hydrogels supported good viability of encapsulated neural progenitor cells (NPC) and protected NPC from ROS induced damage in vitro when H2O2 was present in the media. The dynamic hydrogels were further applied as bio-inks for 3D printing/bioprinting. Overall, this facilely prepared dynamic hydrogel based on HA-PBA and PVA may have many potential biomedical applications, including drug delivery, 3D culture of cells, and 3D bioprinting.

10.
ACS Appl Mater Interfaces ; 11(13): 12298-12307, 2019 Apr 03.
Article in English | MEDLINE | ID: mdl-30855125

ABSTRACT

Craniotomy involves the removal of a skull fragment to access the brain, such as during tumor or epilepsy surgery, which is immediately replaced intraoperatively. The infection incidence after craniotomy ranges from 0.8 to 3%, with approximately half caused by Staphylococcus aureus ( S. aureus). To mitigate infectious complications following craniotomy, we engineered a three-dimensional (3D) bioprinted bone scaffold to harness the potent antibacterial activity of macrophages (MΦs) together with antibiotics using a mouse S. aureus craniotomy-associated biofilm model that establishes a persistent infection on the bone flap, subcutaneous galea, and brain. The 3D scaffold contained rifampin and daptomycin printed in a composite slurry, with viable MΦs incorporated into a hydrogel-based bioink, which was assessed for both the treatment and prevention of craniotomy-associated infections in the mouse model. For the treatment paradigm, the bone flap was removed at day 7 post infection after a mature biofilm had formed and was replaced with a 3D printed antibiotic scaffold, with or without MΦ incorporation. Bacterial burdens in the galea and brain were reduced by at least 100-fold at early time points, which was potentiated by bioprinting viable MΦs into the 3D antibiotic scaffold. We also examined a prevention paradigm, where the scaffolds were placed at the time of surgery and challenged with S. aureus one day later at the surgical site. Interestingly, unlike the treatment paradigm, the incorporation of viable MΦs into the 3D antibiotic scaffold did not enhance bacterial clearance compared to that of antibiotic alone. With further refinement, our 3D bioprinted scaffold represents a potential treatment modality, as it delivers therapeutic antibiotic levels more rapidly than systemic administration, based on its proximity to the infection site. In addition, the incorporation of viable MΦs into the 3D scaffold is an important advance, which demonstrated an improved therapeutic benefit for the treatment of established biofilms that represent the most clinically challenging scenario.


Subject(s)
Anti-Bacterial Agents , Craniotomy , Macrophages , Printing, Three-Dimensional , Staphylococcal Infections , Staphylococcus aureus/physiology , Wound Infection , Animals , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Biofilms/drug effects , Biofilms/growth & development , Cell Line , Female , Humans , Macrophages/metabolism , Macrophages/pathology , Male , Mice , Staphylococcal Infections/metabolism , Staphylococcal Infections/pathology , Staphylococcal Infections/therapy , Wound Infection/metabolism , Wound Infection/microbiology , Wound Infection/pathology , Wound Infection/therapy
11.
ACS Biomater Sci Eng ; 5(10): 5284-5294, 2019 Oct 14.
Article in English | MEDLINE | ID: mdl-33455233

ABSTRACT

Nerve guidance conduit (NGC)-infilling substrates have been reported to facilitate the regeneration of injured peripheral nerves (PNs), especially for large nerve gaps. In this study, longitudinally oriented electrospun core-sheath nanoyarns (csNYs), consisting of a polylactic acid microfiber core and an electrospun nanofiber sheath, were fabricated for potential PN tissue engineering applications. Our novel csNY displayed a well-aligned nanofibrous surface topography, resembling the ultrastructure of axons and fascicles of a native PN system, and it also provided a mechanically stable structure. The biological results showed that the csNY significantly enhanced the attachment, growth, and proliferation of human adipose derived mesenchymal stem cells (hADMSC) and also promoted the migration, proliferation, and phenotype maintenance of rabbit Schwann cells (rSCs). Our csNY notably increased the differentiation capability of hADMSC into SC-like cells (hADMSC-SC), in comparison with a 2D tissue culture polystyrene plate. More importantly, when combined with the appropriate induction medium, our csNY promoted hADMSC-SC to express high levels of myelination-associated markers. Overall, this study demonstrates that our csNYs have great potential to serve as not only ideal in vitro culture models for understanding SC-axon interaction and SC myelination but also as promising NGC-infilling substrates for PN regeneration applications.

12.
ACS Appl Mater Interfaces ; 10(26): 21825-21835, 2018 Jul 05.
Article in English | MEDLINE | ID: mdl-29897225

ABSTRACT

The blood-brain barrier (BBB) is an active and complex diffusion barrier that separates the circulating blood from the brain and extracellular fluid, regulates nutrient transportation, and provides protection against various toxic compounds and pathogens. Creating an in vitro microphysiological BBB system, particularly with relevant human cell types, will significantly facilitate the research of neuropharmaceutical drug delivery, screening, and transport, as well as improve our understanding of pathologies that are due to BBB damage. Currently, most of the in vitro BBB models are generated by culturing rodent astrocytes and endothelial cells, using commercially available transwell membranes. Those membranes are made of plastic biopolymers that are nonbiodegradable, porous, and stiff. In addition, distinct from rodent astrocytes, human astrocytes possess unique cell complexity and physiology, which are among the few characteristics that differentiate human brains from rodent brains. In this study, we established a novel human BBB microphysiologocal system, consisting of a three-dimensionally printed holder with a electrospun poly(lactic- co-glycolic) acid (PLGA) nanofibrous mesh, a bilayer coculture of human astrocytes, and endothelial cells, derived from human induced pluripotent stem cells (hiPSCs), on the electrospun PLGA mesh. This human BBB model achieved significant barrier integrity with tight junction protein expression, an effective permeability to sodium fluorescein, and higher transendothelial electrical resistance (TEER) comparing to electrospun mesh-based counterparts. Moreover, the coculture of hiPSC-derived astrocytes and endothielial cells promoted the tight junction protein expression and the TEER value. We further verified the barrier functions of our BBB model with antibrain tumor drugs (paclitaxel and bortezomib) and a neurotoxic peptide (amyloid ß 1-42). The human microphysiological system generated in this study will potentially provide a new, powerful tool for research on human BBB physiology and pathology.


Subject(s)
Induced Pluripotent Stem Cells , Amyloid beta-Peptides , Astrocytes , Blood-Brain Barrier , Cells, Cultured , Coculture Techniques , Humans , Nanofibers
13.
Acta Biomater ; 74: 131-142, 2018 07 01.
Article in English | MEDLINE | ID: mdl-29842971

ABSTRACT

Bioengineered adipose tissues have gained increased interest as a promising alternative to autologous tissue flaps and synthetic adipose fillers for soft tissue augmentation and defect reconstruction in clinic. Although many scaffolding materials and biofabrication methods have been investigated for adipose tissue engineering in the last decades, there are still challenges to recapitulate the appropriate adipose tissue microenvironment, maintain volume stability, and induce vascularization to achieve long-term function and integration. In the present research, we fabricated cryogels consisting of methacrylated gelatin, methacrylated hyaluronic acid, and 4arm poly(ethylene glycol) acrylate (PEG-4A) by using cryopolymerization. The cryogels were repeatedly injectable and stretchable, and the addition of PEG-4A improved the robustness and mechanical properties. The cryogels supported human adipose progenitor cell (HWA) and adipose derived mesenchymal stromal cell adhesion, proliferation, and adipogenic differentiation and maturation, regardless of the addition of PEG-4A. The HWA laden cryogels facilitated the co-culture of human umbilical vein endothelial cells (HUVEC) and capillary-like network formation, which in return also promoted adipogenesis. We further combined cryogels with 3D bioprinting to generate handleable adipose constructs with clinically relevant size. 3D bioprinting enabled the deposition of multiple bioinks onto the cryogels. The bioprinted flap-like constructs had an integrated structure without delamination and supported vascularization. STATEMENT OF SIGNIFICANCE: Adipose tissue engineering is promising for reconstruction of soft tissue defects, and also challenging for restoring and maintaining soft tissue volume and shape, and achieving vascularization and integration. In this study, we fabricated cryogels with mechanical robustness, injectability, and stretchability by using cryopolymerization. The cryogels promoted cell adhesion, proliferation, and adipogenic differentiation and maturation of human adipose progenitor cells and adipose derived mesenchymal stromal cells. Moreover, the cryogels also supported 3D bioprinting on top, forming vascularized adipose constructs. This study demonstrates the potential of the implementation of cryogels for generating volume-stable adipose tissue constructs and provides a strategy to fabricate vascularized flap-like constructs for complex soft tissue regeneration.


Subject(s)
Adipogenesis , Adipose Tissue/metabolism , Cell Proliferation , Cryogels , Mesenchymal Stem Cells/metabolism , Tissue Engineering , Adipose Tissue/cytology , Bioprinting , Cell Adhesion , Cell Line, Transformed , Cryogels/chemical synthesis , Cryogels/chemistry , Female , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Mesenchymal Stem Cells/cytology , Printing, Three-Dimensional
14.
J Biomed Mater Res B Appl Biomater ; 106(5): 1788-1798, 2018 07.
Article in English | MEDLINE | ID: mdl-28901689

ABSTRACT

Vascularization is a fundamental prerequisite for large bone construct development and remains one of the main challenges of bone tissue engineering. Our current study presents the combination of 3D printing technique with a hydrogel-based prevascularization strategy to generate prevascularized bone constructs. Human adipose derived mesenchymal stem cells (ADMSC) and human umbilical vein endothelial cells (HUVEC) were encapsulated within our bioactive hydrogels, and the effects of culture conditions on in vitro vascularization were determined. We further generated composite constructs by forming 3D printed polycaprolactone/hydroxyapatite scaffolds coated with cell-laden hydrogels and determined how the co-culture affected vascularization and osteogenesis. It was demonstrated that 3D co-cultured ADMSC-HUVEC generated capillary-like networks within the porous 3D printed scaffold. The co-culture systems promoted in vitro vascularization, but had no significant effects on osteogenesis. The prevascularized constructs were subcutaneously implanted into nude mice to evaluate the in vivo vascularization capacity and the functionality of engineered vessels. The hydrogel systems facilitated microvessel and lumen formation and promoted anastomosis of vascular networks of human origin with host murine vasculature. These findings demonstrate the potential of prevascularized 3D printed scaffolds with anatomical shape for the healing of larger bone defects. © 2017 Wiley Periodicals, Inc. J Biomed Mater Res Part B: Appl Biomater, 106B: 1788-1798, 2018.


Subject(s)
Human Umbilical Vein Endothelial Cells , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Neovascularization, Physiologic , Osteogenesis , Printing, Three-Dimensional , Tissue Scaffolds/chemistry , Animals , Coculture Techniques , Durapatite/chemistry , Female , Heterografts , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/transplantation , Humans , Hydrogels/chemistry , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Mice, Nude , Polyesters/chemistry , Tissue Engineering
15.
RSC Adv ; 7(47): 29312-29320, 2017 Jun 05.
Article in English | MEDLINE | ID: mdl-28670447

ABSTRACT

Reconstruction of complex, craniofacial bone defects often requires autogenous vascularized bone grafts, and still remains a challenge today. In order to address this issue, we isolated the stromal vascular fraction (SVF) from adipose tissues and maintained the phenotypes and the growth of endothelial lineage cells within SVF derived cells (SVFC) by incorporating an endothelial cell medium. We 3D bioprinted SVFC within our hydrogel bioinks and conditioned the constructs in either normoxia or hypoxia. We found that short-term hypoxic conditioning promoted vascularization-related gene expression, whereas long-term hypoxia impaired cell viability and vascularization. 3D bioprinted bone constructs composed of polycaprolactone/hydroxyapatite (PCL/HAp) and SVFC-laden hydrogel bioinks were then implanted into athymic mice, after conditioning in normoxic or short-term hypoxic environments, in order to determine the in vitro and in vivo vascularization and osteogenic differentiation of the constructs. Short-term hypoxic conditioning promoted microvessel formation in vitro and in vivo and promoted integration with existing host vasculature, but did not affect osteogenic differentiation of SVFC. These findings demonstrate the benefit of short-term hypoxia and the potential for utilization of SVFC and 3D bioprinting for generating prevascularized 3D bioprinted bone constructs. Furthermore, the ability to custom design complex anatomical shapes has promising applications for the regeneration of both large and small craniofacial bone defects.

16.
ACS Biomater Sci Eng ; 3(5): 826-835, 2017 May 08.
Article in English | MEDLINE | ID: mdl-33440487

ABSTRACT

Hydrogel-based cartilage tissue engineering strategies require the induction and long-term maintenance of adipose derived mesenchymal stem cells (ADMSC) into a stable chondrogenic phenotype. However, ADMSC exhibit the tendency to undergo hypertrophic differentiation, rather than forming permanent hyaline cartilage phenotype changes. This may hinder their implementation in articular cartilage regeneration, but may allow the possibility for bone and bone to soft tissue interface repair. In this study, we examined the effects of hydroxyapatite (HAp) on the chondrogenesis and hypertrophy of ADMSC within bioprinted hyaluronic acid (HA)-based hydrogels. We found that a small amount of HAp (∼10% of polymer concentration) promoted both chondrogenic and hypertrophic differentiation of ADMSC. Increased HAp contents promoted hypertrophic conversion and early osteogenic differentiation of encapsulated ADMSC. Subsequently, ADMSC-laden, stratified constructs with nonmineralized and mineralized layers (i.e., HA based and HA-HAp based) were 3D bioprinted. The constructs were conditioned in chondrogenic medium in either a normoxic or hypoxic environment for 8 weeks to assess the effects of oxygen tension on ADMSC differentiation and interface integration. We further implanted the bioprinted constructs subcutaneously into nude mice for 4 weeks. It was found that hypoxia partially inhibited hypertrophic differentiation by significantly down-regulating the expression of COL10A1, ALP, and MMP13. In addition, hypoxia also suppressed spontaneous calcification of ADMSC and promoted interface integration. This study demonstrates that both HAp content and hypoxia are important to mediate chondrogenesis, hypertrophy, and endochondral ossification of ADMSC. An optimized recipe and condition will allow for 3D bioprinting of multizonal grafts with integrated hard tissue and soft tissue interfaces for the treatment of complex orthopedic defects.

SELECTION OF CITATIONS
SEARCH DETAIL
...