Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 26
1.
J Orthop Res ; 2024 Mar 24.
Article En | MEDLINE | ID: mdl-38522018

Segmental bone defects, often clinically treated with nondegradable poly(methylmethacrylate) (PMMA) in multistage surgeries, present a significant clinical challenge. Our study investigated the efficacy of 3D printed biodegradable polycaprolactone fumarate (PCLF)/PCL spacers in a one-stage surgical intervention for these defects, focusing on early bone regeneration influenced by spacer porosities. We compared nonporous PCLF/PCL and PMMA spacers, conventionally molded into cylinders, with porous PCLF/PCL spacers, 3D printed to structurally mimic segmental defects in rat femurs for a 4-week implantation study. Histological analysis, including tissue staining and immunohistochemistry with bone-specific antibodies, was conducted for histomorphometry evaluation. The PCLF/PCL spacers demonstrated compressive properties within 6 ± 0.5 MPa (strength) and 140 ± 15 MPa (modulus). Both porous PCLF/PCL and Nonporous PMMA formed collagen-rich membranes (PCLF/PCL: 92% ± 1.3%, PMMA: 86% ± 1.5%) similar to those induced in the Masquelet technique, indicating PCLF/PCL's potential for one-stage healing. Immunohistochemistry confirmed biomarkers for tissue regeneration, underscoring PCLF/PCL's regenerative capabilities. This research highlights PCLF/PCL scaffolds' ability to induce membrane formation in critical-sized segmental bone defects, supporting their use in one-stage surgery. Both solid and porous PCLF/PCL spacers showed adequate compressive properties, with the porous variants exhibiting BMP-2 expression and woven bone formation, akin to clinical standard PMMA. Notably, the early ossification of the membrane into the pores of porous scaffolds suggests potential for bone interlocking and regeneration, potentially eliminating the need for a second surgery required for PMMA spacers. The biocompatibility and biodegradability of PCLF/PCL make them promising alternatives for treating critical bone defects, especially in vulnerable patient groups.

2.
Macromol Biosci ; 24(4): e2300414, 2024 Apr.
Article En | MEDLINE | ID: mdl-38035771

This study focuses to develop a unique hybrid hydrogel bioink formulation that incorporates poly(ethylene glycol) dimethacrylate (PEGDMA), gelatin (Gel), and methylcellulose (MC). This formulation achieves the necessary viscosity for extrusion-based three-dimensional (3D) printing of scaffolds intended for bone regeneration. After thorough optimization of the hybrid bioink system with Gel, three distinct scaffold groups are investigated in vitro: 0%, 3%, and 6% (w/v) Gel. These scaffold groups are examined for their morphology, mechanical strength, biodegradation, in vitro cell proliferation and differentiation, and in vivo bone formation using a rat cranial defect model. Among these scaffold compositions, the 3% Gel scaffold exhibits the most favorable characteristics, prompting further evaluation as a rat mesenchymal stem cell (rMSC) carrier in a critical-size cranial defect within a Lewis rat model. The compressive strength of all three scaffold groups range between 1 and 2 MPa. Notably, the inclusion of Gel in the scaffolds leads to enhanced bioactivity and cell adhesion. The Gel-containing scaffolds notably amplify osteogenic differentiation, as evidenced by alkaline phosphatase (ALP) and Western blot analyses. The in vivo results, as depicted by microcomputed tomography, showcase augmented osteogenesis within cell-seeded scaffolds, thus validating this innovative PEGDMA-based scaffold system as a promising candidate for cranial bone defect healing.


Methacrylates , Tissue Engineering , Tissue Scaffolds , Rats , Animals , Tissue Engineering/methods , Osteogenesis , X-Ray Microtomography , Rats, Inbred Lew , Polyethylene Glycols/pharmacology , Bone Regeneration , Hydrogels/pharmacology , Cell Differentiation , Printing, Three-Dimensional
3.
J Biomed Mater Res A ; 112(5): 672-684, 2024 05.
Article En | MEDLINE | ID: mdl-37971074

Polycaprolactone fumarate (PCLF) is a cross-linkable PCL derivative extensively considered for tissue engineering applications. Although injection molding has been widely used to develop PCLF scaffolds, platforms developed using such technique lack precise control on architecture, design, and porosity required to ensure adequate cellular and tissue responses. In particular, the scaffolds should provide a suitable surface for cell attachment and proliferation, and facilitate cell-cell communication and nutrient flow. 3D printing technologies have led to new architype for biomaterial development with micro-architecture mimicking native tissue. Here, we developed a method for 3D printing of PCLF structures using the extrusion printing technique. The crosslinking property of PCLF enabled the unique post-processing of 3D printed scaffolds resulting in highly porous and flexible PCLF scaffolds with compressive properties imitating natural features of cancellous bone. Generated scaffolds supported excellent attachment and proliferation of mesenchymal stem cells (MSC). The high porosity of PCLF scaffolds facilitated vascularized membrane formation demonstrable with the stringency of the ex ovo chicken chorioallantoic membrane (CAM) implantation. Furthermore, upon implantation to rat calvarium defects, PCLF scaffolds enabled an exceptional new bone formation with a bone mineral density of newly formed bone mirroring native bone tissue. These studies suggest that the 3D-printed highly porous PCLF scaffolds may serve as a suitable biomaterial platform to significantly expand the utility of the PCLF biomaterial for bone tissue engineering applications.


Fumarates , Tissue Scaffolds , Rats , Animals , Tissue Scaffolds/chemistry , Fumarates/pharmacology , Fumarates/chemistry , Biocompatible Materials/chemistry , Polyesters/pharmacology , Polyesters/chemistry , Tissue Engineering/methods , Bone Regeneration , Porosity , Printing, Three-Dimensional
4.
Biomater Adv ; 153: 213539, 2023 Oct.
Article En | MEDLINE | ID: mdl-37429047

Pre-formed hydrogel scaffolds have emerged as favorable vehicles for tissue regeneration, promoting minimally invasive treatment of native tissue. However, due to the high degree of swelling and inherently poor mechanical properties, development of complex structural hydrogel scaffolds at different dimensional scales has been a continuous challenge. Herein, we take a novel approach at the intersections of engineering design and bio-ink chemistry to develop injectable pre-formed structural hydrogel scaffolds fabricated via visible light (VL) induced digital light processing (DLP). In this study, we first determined the minimum concentration of poly(ethylene glycol) diacrylate (PEGDA) to be added to the gelatin methacrylate (GelMA) bio-ink in order to achieve scalable and high printing-fidelity with desired cell adhesion, viability, spreading, and osteogenic differentiation characteristics. Despite the advantages of hybrid GelMA-PEGDA bio-ink in improving scalability and printing-fidelity, compressibility, shape-recovery, and injectability of the 3D bioprinted scaffolds were compromised. To restore these needed characteristics for minimally invasive tissue regeneration applications, we performed topological optimization to design highly compressible and injectable pre-formed (i.e., 3D bioprinted) microarchitectural scaffolds. The designed injectable pre-formed microarchitectural scaffolds showed a great capacity to retain the viability of the encapsulated cells (>72 % after 10 cycles of injection). Lastly, ex ovo chicken chorioallantoic membrane (CAM) studies revealed that the optimized injectable pre-formed hybrid hydrogel scaffold is biocompatible and supports angiogenic growth.


Osteogenesis , Tissue Scaffolds , Tissue Scaffolds/chemistry , Hydrogels , Light , Gelatin/chemistry
5.
Bioact Mater ; 27: 216-230, 2023 Sep.
Article En | MEDLINE | ID: mdl-37122896

Three-dimensional (3D) printing technology is driving forward the progresses of various engineering fields, including tissue engineering. However, the pristine 3D-printed scaffolds usually lack robust functions in stimulating desired activity for varied regeneration applications. In this study, we combined the two-dimensional (2D) hetero-nanostructures and immuno-regulative interleukin-4 (IL-4) cytokines for the functionalization of 3D-printed scaffolds to achieve a pro-healing immuno-microenvironment for optimized bone injury repair. The 2D hetero-nanostructure consists of graphene oxide (GO) layers, for improved cell adhesion, and black phosphorous (BP) nanosheets, for the continuous release of phosphate ions to stimulate cell growth and osteogenesis. In addition, the 2D hetero-nanolayers facilitated the adsorption of large content of immuno-regulative IL-4 cytokines, which modulated the polarization of macrophages into M2 phenotype. After in vivo implantation in rat, the immuno-functioned 3D-scaffolds achieved in vivo osteo-immunomodulation by building a pro-healing immunological microenvironment for better angiogenesis and osteogenesis in the defect area and thus facilitated bone regeneration. These results demonstrated that the immuno-functionalization of 3D-scaffolds with 2D hetero-nanostructures with secondary loading of immuno-regulative cytokines is an encouraging strategy for improving bone regeneration.

6.
ACS Biomater Sci Eng ; 9(3): 1585-1597, 2023 03 13.
Article En | MEDLINE | ID: mdl-36854041

Current bone cement systems often demand free radical or metal-related initiators and/or catalysts for the crosslinking process, which may cause serious toxicity to the human body. In addition, the resultant dense scaffolds may have a prolonged degradation time and are difficult for cells to infiltrate and form new tissue. In this study, we developed a porous "click" organic-inorganic nanohybrid (PO-click-ON) cement that crosslinks via metal-free biorthogonal click chemistry and forms porous structures mimicking the native bone tissue via particulate leaching. Strain-promoted click reaction enables fast and efficient crosslinking of polymer chains with the exclusion of any toxic initiator or catalyst. The resulting PO-click-ON implants supported exceptional in vitro stem cell adhesion and osteogenic differentiation with a large portion of stem cells infiltrated deep into the scaffolds. In vivo study using a rat cranial defect model demonstrated that the PO-click-ON system achieved outstanding cell adsorption, neovascularization, and bone formation. The porous click cement developed in this study serves as a promising platform with multifunctionality for bone and other tissue engineering applications.


Bone Cements , Osteogenesis , Humans , Rats , Animals , Bone Cements/chemistry , Tissue Engineering/methods , Bone and Bones , Stem Cells
7.
IEEE Trans Biomed Eng ; 70(3): 841-852, 2023 03.
Article En | MEDLINE | ID: mdl-36049008

OBJECTIVE: Hydrogel scaffolds have attracted attention to develop cellular therapy and tissue engineering platforms for regenerative medicine applications. Among factors, local mechanical properties of scaffolds drive the functionalities of cell niche. Dynamic mechanical analysis (DMA), the standard method to characterize mechanical properties of hydrogels, restricts development in tissue engineering because the measurement provides a single elasticity value for the sample, requires direct contact, and represents a destructive evaluation preventing longitudinal studies on the same sample. We propose a novel technique, acoustic force elastography microscopy (AFEM), to evaluate elastic properties of tissue engineering scaffolds. RESULTS: AFEM can resolve localized and two-dimensional (2D) elastic properties of both transparent and opaque materials with advantages of being non-contact and non-destructive. Gelatin hydrogels, neat synthetic oligo[poly(ethylene glycol)fumarate] (OPF) scaffolds, OPF hydroxyapatite nanocomposite scaffolds and ex vivo biological tissue were examined with AFEM to evaluate the elastic modulus. These measurements of Young's modulus range from approximately 2 kPa to over 100 kPa were evaluated and are in good agreement with finite element simulations, surface wave measurements, and DMA tests. CONCLUSION: The AFEM can resolve localized and 2D elastic properties of hydrogels, scaffolds and thin biological tissues. These materials can either be transparent or non-transparent and their evaluation can be done in a non-contact and non-destructive manner, thereby facilitating longitudinal evaluation. SIGNIFICANCE: AFEM is a promising technique to quantify elastic properties of scaffolds for tissue engineering and will be applied to provide new insights for exploring elastic changes of cell-laden scaffolds for tissue engineering and material science.


Elasticity Imaging Techniques , Tissue Scaffolds , Tissue Engineering/methods , Microscopy, Atomic Force , Hydrogels
8.
Biomaterials ; 284: 121507, 2022 05.
Article En | MEDLINE | ID: mdl-35421800

The nanomaterials research spectrum has seen the continuous emergence of two-dimensional (2D) materials over the years. These highly anisotropic and ultrathin materials have found special attention in developing biomedical platforms for therapeutic applications, biosensing, drug delivery, and regenerative medicine. Three-dimensional (3D) printing and bioprinting technologies have emerged as promising tools in medical applications. The convergence of 2D nanomaterials with 3D printing has extended the application dynamics of available biomaterials to 3D printable inks and bioinks. Furthermore, the unique properties of 2D nanomaterials have imparted multifunctionalities to 3D printed constructs applicable to several biomedical applications. 2D nanomaterials such as graphene and its derivatives have long been the interest of researchers working in this area. Beyond graphene, a range of emerging 2D nanomaterials, such as layered silicates, black phosphorus, transition metal dichalcogenides, transition metal oxides, hexagonal boron nitride, and MXenes, are being explored for the multitude of biomedical applications. Better understandings on both the local and systemic toxicity of these materials have also emerged over the years. This review focuses on state-of-art 3D fabrication and biofabrication of biomedical platforms facilitated by 2D nanomaterials, with the comprehensive summary of studies focusing on the toxicity of these materials. We highlight the dynamism added by 2D nanomaterials in the printing process and the functionality of printed constructs.


Bioprinting , Graphite , Nanostructures , Bioprinting/methods , Oxides , Printing, Three-Dimensional , Tissue Engineering/methods , Tissue Scaffolds
9.
ACS Nano ; 16(2): 2741-2755, 2022 02 22.
Article En | MEDLINE | ID: mdl-35072461

Scaffold-free spheroids offer great potential as a direct supply of cells for bottom-up bone tissue engineering. However, the building of functional spheroids with both cells and bioactive signals remains challenging. Here, we engineered functional spheroids with mesenchymal stem cells (MSCs) and two-dimensional heteronano-layers (2DHNL) that consisted of black phosphorus (BP) and graphene oxide (GO) to create a 3D cell-instructive microenvironment for large defect bone repair. The effects of the engineered 2D materials on the proliferation, osteogenic differentiation of stem cells was evaluated in an in vitro 3D spheroidal microenvironment. Excellent in vivo support of osteogenesis of MSCs, neovascularization, and bone regeneration was achieved after transplanting these engineered spheroids into critical-sized rat calvarial defects. Further loading of osteogenic factor dexamethasone (DEX) on the 2DHNL showed outstanding in vivo osteogenic induction and bone regrowth without prior in vitro culture in osteogenic medium. The shortened overall culture time would be advantageous for clinical translation. These functional spheroids impregnated with engineered 2DHNL enabling stem cell and osteogenic factor codelivery could be promising functional building blocks to provide cells and differential clues in an all-in-one system to create large tissues for time-effective in vivo bone repair.


Mesenchymal Stem Cells , Osteogenesis , Animals , Cell Differentiation , Rats , Stem Cells , Tissue Engineering/methods , Tissue Scaffolds
10.
Stem Cells Int ; 2021: 1938819, 2021.
Article En | MEDLINE | ID: mdl-34434236

In situ cell recruitment is a promising regenerative medicine strategy with the purpose of tissue regeneration without stem cell transplantation. This chemotaxis-based strategy is aimed at ensuring a restorative environment through the release of chemokines that promote site-specific migration of healing cell populations. Stromal cell-derived factor-1α (SDF-1α) is a critical chemokine that can regulate the migration of mesenchymal stem cells (MSCs). Accordingly, here, SDF-1α-loaded microporous oligo[poly(ethylene glycol) fumarate]/bis[2-(methacryloyloxy)ethyl] phosphate composites (SDF-1α/OPF/BP) were engineered and probed. SDF-1α/OPF/BP composites were loaded with escalating SDF-1α concentrations, namely, 0 ng/ml, 50 ng/ml, 100 ng/ml, and 200 ng/ml, and were cocultured with MSC. Scratching assay, Transwell assay, and three-dimensional migration model were utilized to assess the migration response of MSCs. Immunofluorescence staining of Runx2 and osteopontin (OPN), ELISA assay of osteocalcin (OCN) and alkaline phosphatase (ALP), and Alizarin Red S staining were conducted to assess the osteogenesis of MSCs. All SDF-1α/OPF/BP composites engendered a release of SDF-1α (>80%) during the first four days. SDF-1α released from the composites significantly promoted migration and osteogenic differentiation of MSCs documented by upregulated expression of osteogenic-related proteins, ALP, Runx2, OCN, and OPN. SDF-1α at 100 ng/ml was optimal for enhanced migration and osteogenic proficiency. Thus, designed SDF-1α/OPF/BP composites were competent in promoting the homing and osteogenesis of MSCs and thus offer a promising bioactive scaffold candidate for on-demand bone tissue regeneration.

11.
Biomaterials ; 276: 121014, 2021 09.
Article En | MEDLINE | ID: mdl-34280821

Injectable polymers have attracted intensive attention in tissue engineering and drug delivery applications. Current injectable polymer systems often require free-radical or heavy-metal initiators and catalysts for the crosslinking process, which may be extremely toxic to the human body. Here, we report a novel polyhedral oligomeric silsesquioxane (POSS) based strain-promoted alkyne-azide cycloaddition (SPAAC) "click" organic-inorganic nanohybrids (click-ON) system that can be click-crosslinked without any toxic initiators or catalysts. The click-ON scaffolds supported excellent adhesion, proliferation, and osteogenesis of stem cells. In vivo evaluation using a rat cranial defect model showed outstanding bone formation with minimum cytotoxicity. Essential osteogenic alkaline phosphatase (ALP) and vascular CD31 marker expression were detected on the defect site, indicating excellent support of in vivo osteogenesis and vascularization. Using salt leaching techniques, an injectable porous click-ON cement was developed to create porous structures and support better in vivo bone regeneration. Beyond defect filling, the click-ON cement also showed promising application for spinal fusion using rabbits as a model. Compared to the current clinically used poly (methyl methacrylate) (PMMA) cement, this click-ON cement showed great advantages of low heat generation, better biocompatibility and biodegradability, and thus has great potential for bone and related tissue engineering applications.


Bone Cements , Tissue Engineering , Animals , Bone Regeneration , Hydrogels , Osteogenesis , Rabbits , Rats , Tissue Scaffolds
12.
Mater Sci Eng C Mater Biol Appl ; 127: 112252, 2021 Aug.
Article En | MEDLINE | ID: mdl-34225891

Bone morphogenetic proteins (BMPs) are well known as enhancers and facilitators of osteogenesis during bone regeneration. The use of recombinant BMP-2 (rhBMP-2) in bone defect healing has drawbacks, which has driven the scouting for alternatives, such as recombinant BMP-9 (rhBMP-9), to provide comparable new bone formation. However, the dosage of rhBMP-9 is quintessential for the facilitation of adequate bone defect healing. Therefore, this study has been designed to evaluate the optimal dosage of BMP-9 by comparing the bone defect healing induced by rhBMP-9 over rhBMP-2. The chitosan (CS) microparticles (MPs), coated with BMPs, were embedded in a thermoresponsive methylcellulose (MC) and calcium alginate (Alg) based injectable delivery system containing a dosage of either 0.5 µg or 1.5 µg of the respective rhBMP per bone defect. A 5 mm critical-sized cranial defect rat model has been used in this study, and bone tissues were harvested at eight weeks post-surgery. The standard tools for comparing the new bone regeneration included micro computerized tomography (micro-CT) and histological analysis. A novel perspective of analyzing the new bone quality and crystallinity was employed by using Raman spectroscopy, along with its elastic modulus quantified through Atomic Force Microscopy (AFM). Results showed that the rhBMP-9 administered at a dosage of 1.5 µg per bone defect, using this delivery system, can adequately facilitate the bone void filling with ample new bone mineralization and crystallinity as compared to rhBMP-2, thus approving the hypothesis for a viable rhBMP-2 alternative.


Bone Morphogenetic Protein 2 , Growth Differentiation Factor 2 , Animals , Bone Morphogenetic Proteins , Bone Regeneration , Calcification, Physiologic , Growth Differentiation Factor 2/pharmacology , Osteogenesis , Rats , Recombinant Proteins , Transforming Growth Factor beta
13.
Mater Sci Eng C Mater Biol Appl ; 124: 112075, 2021 May.
Article En | MEDLINE | ID: mdl-33947567

Most of the synthetic polymer-based hydrogels lack the intrinsic properties needed for tissue engineering applications. Here, we describe a biomimetic approach to induce the mineralization and vascularization of poly(ethylene glycol) (PEG)-based hydrogel to template the osteogenic activities. The strategy involves the covalent functionalization of oligo[poly(ethylene glycol) fumarate] (OPF) with phosphate groups and subsequent treatment of phosphorylated-OPF (Pi-OPF) hydrogels with alkaline phosphatase enzyme (ALP) and calcium. Unlike previously reported studies for ALP induced mineralization, in this study, the base polymer itself was modified with the phosphate groups for uniform mineralization of hydrogels. In addition to improvement of mechanical properties, enhancement of MC3T3-E1 cell attachment and proliferation, and promotion of mesenchymal stem cells (MSC) differentiation were observed as the intrinsic benefits of such mineralization. Current bone tissue engineering (BTE) research endeavors are also extensively focused on vascular tissue regeneration due to its inherent advantages in bone regeneration. Taking this into account, we further functionalized the mineralized hydrogels with FG-4592, small hypoxia mimicking molecule. The functionalized hydrogels elicited upregulated in vitro angiogenic activities of human umbilical vein endothelial cells (HUVEC). In addition, when implanted subcutaneously in rats, enhanced early vascularization activities around the implantation site were observed as demonstrated by the immunohistochemistry results. This further leveraged the formation of calcified tissues at the implantation site at later time points evident through X-ray imaging. The overall results here show the perspectives of bifunctional OPF hydrogels for vascularized BTE.


Endothelial Cells , Hydrogels , Animals , Bone Regeneration , Bone and Bones , Hydrogels/pharmacology , Osteogenesis , Polyethylene Glycols , Rats , Tissue Engineering
14.
Mater Sci Eng C Mater Biol Appl ; 121: 111812, 2021 Feb.
Article En | MEDLINE | ID: mdl-33579456

Mesenchymal stem cell (MSC)-spheroids have sparked significant interest in bone tissue engineering due to their resemblance to natural bone tissue, especially in terms of cell-cell and cell-extracellular matrix interactions. Many biomaterials or biomolecules have been incorporated into MSC-spheroids to enhance their osteogenic abilities. In this respect, we assessed the osteogenic responses of MSC spheroids leveraged through the unique combination of collagen and black phosphorus (BP). The MSC spheroids were successfully constructed with 6 µg/mL collagen and/or a concentration gradient (0 µg/mL, 4 µg/mL, 8 µg/mL, and 16 µg/mL) of BP and were evaluated for MSC viability and their osteogenic differentiation over a time period of 14 days. Improved MSC viability and osteogenic ability were observed for the spheroids with collagen and BP at the concentration of 4 µg/mL and 8 µg/mL. Next, blank spheroids (Control) or the optimized MSC spheroids with 6 µg/mL collagen and 4 µg/mL BP (Col+BP4) were further encapsulated into two types of hydrogel scaffolds: porous oligo[poly(ethylene glycol) fumarate] (OPF) hydrogel and hydroxyapatite-collagen I scaffold (HE-COL). The osteogenic abilities of these four groups were evaluated after 14 and 21 days of osteogenic induction. The MSC spheroids incorporated with collagen and BP implanted into OPF porous hydrogel (Col+BP/OPF) elicited a higher expression of Runx2, osteopontin, and alkaline phosphatase than blank spheroids implanted into OPF porous hydrogel (Control/OPF). Enhanced osteogenesis was also observed in the Col+BP/HE-COL group as compared to Control/HE-COL. Taken together, the results from this study showed the perspectives of collagen and BP incorporated MSC spheroids for the development of injectable cellular therapies for bone regeneration.


Mesenchymal Stem Cells , Osteogenesis , Cell Differentiation , Collagen , Hydrogels/pharmacology , Phosphorus , Tissue Engineering
15.
Biomater Sci ; 9(8): 2768-2803, 2021 Apr 21.
Article En | MEDLINE | ID: mdl-33620047

Phosphorene, also known as black phosphorus (BP), is a two-dimensional (2D) material that has gained significant attention in several areas of current research. Its unique properties such as outstanding surface activity, an adjustable bandgap width, favorable on/off current ratios, infrared-light responsiveness, good biocompatibility, and fast biodegradation differentiate this material from other two-dimensional materials. The application of BP in the biomedical field has been rapidly emerging over the past few years. This article aimed to provide a comprehensive review of the recent progress on the unique properties and extensive medical applications for BP in bone, nerve, skin, kidney, cancer, and biosensing related treatment. The details of applications of BP in these fields were summarized and discussed.


Nanotubes, Carbon , Neoplasms , Quantum Dots , Bone and Bones , Humans , Phosphorus
16.
J Biomed Mater Res A ; 109(2): 193-206, 2021 02.
Article En | MEDLINE | ID: mdl-32441388

Conduits that promote nerve regeneration are currently of great medical concern, particularly when gaps exist between nerve endings. To address this issue, our laboratory previously developed a nerve conduit from biodegradable poly(caprolactone fumarate) (PCLF) that supports peripheral nerve regeneration. The present study improves upon this work by further developing an electrically conductive, positively charged PCLF scaffold through the incorporation of graphene, carbon nanotubes (CNTs), and [2-(methacryloyloxy)ethyl]trimethylammonium chloride (MTAC) (PCLF-Graphene-CNT-MTAC) using ultraviolet (UV) induced photocrosslinking. Scanning electron microscopy, transmission electron microscopy, and atomic force microscopy were used to assess the incorporation of CNTs and graphene into PCLF-Graphene-CNT-MTAC scaffolds, which displayed enhanced surface roughness and reduced electrochemical impedance when compared to neat PCLF. Scaffolds with these surface modifications also showed improved growth and differentiation of rat pheochromocytoma 12 cells in vitro, with enhanced cell growth, neurite extension, and cellular migration. Furthermore, an increased number of neurite protrusions were observed when the conduit was electrically stimulated. These results show that the electrically conductive PCLF-Graphene-CNT-MTAC nerve scaffolds presented here support the cellular behaviors that are critical for nerve regeneration, ultimately making this material an attractive candidate for regenerative medicine applications.


Cell Differentiation/drug effects , Cell Proliferation/drug effects , Graphite/pharmacology , Nanotubes, Carbon , Neurons/drug effects , Tissue Scaffolds , Animals , Electric Conductivity , Electric Impedance , Electric Stimulation , Nerve Regeneration/drug effects , Neurites/drug effects , PC12 Cells , Rats , Surface Properties , Ultraviolet Rays
17.
J Biomed Mater Res A ; 109(1): 6-17, 2021 01.
Article En | MEDLINE | ID: mdl-32418273

3D bioprinting is a promising new tissue restoration technique that enables the precise deposition of cells and growth factors in order to more closely mimic the structure and function of native organs. In this study, we report the development of a new bioink using oligo(poly[ethylene glycol] fumarate) (OPF), a photo-crosslinkable, and biodegradable polymer, for 3D bioprinting. In addition to OPF, a small portion of gelatin was also incorporated into the bioink to make it bio-printable. After immersion in the cell medium, gelatin was eluted away to create a bioprinted scaffold of pure OPF. Excellent cell viability, spreading, and long-term proliferation of encapsulated cells was observed using both bone and nerve cells as examples. These results demonstrate that OPF bioink has great potential in future 3D bioprinting applications that aim to replicate complex, layered tissues, and/or organs.


Bone Regeneration/drug effects , Fumarates/chemistry , Nerve Regeneration/drug effects , Polyethylene Glycols/chemistry , Printing, Three-Dimensional , Tissue Engineering/methods , 3T3 Cells , Animals , Bioprinting , Bone and Bones/drug effects , Cell Proliferation/drug effects , Cell Survival , Cross-Linking Reagents , Gelatin , Hydrogels , Mice , Nerve Tissue/drug effects , Neurons/drug effects , Osteocytes/drug effects , Tissue Scaffolds
18.
Acta Biomater ; 111: 129-140, 2020 07 15.
Article En | MEDLINE | ID: mdl-32428680

Three-dimensional (3D) printing is a promising technology for tissue engineering. However, 3D-printing methods are limited in their ability to produce desired microscale features or electrochemical properties in support of robust cell adhesion, proliferation, and differentiation. This study addresses this deficiency by proposing an integrated, one-step, method to increase the cytocompatibility of 3D-printed scaffolds through functionalization leveraging conductive carbon nanotubes (CNTs). To this end, CNTs were first sonicated with water-soluble single-stranded deoxyribonucleic acid (ssDNA) to generate a negatively charged ssDNA@CNT nano-complex. Concomitantly, 3D-printed poly(propylene fumarate) (PPF) scaffolds were ammonolyzed to introduce free amine groups, which can take on a positive surface charge in water. The ssDNA@CNT nano-complex was then applied to 3D-printed scaffolds through a simple one-step coating utilizing electric-static force. This fast and facile functionalization step resulted in a homogenous and non-toxic coating of CNTs to the surface, which significantly improved the adhesion, proliferation, and differentiation of pre-osteoblast cells. In addition, the CNT based conductive coating layer enabled modulation of cell behavior through electrical stimuli (ES) leading to cellular proliferation and osteogenic gene marker expression, including alkaline phosphatase (ALP), osteocalcin (OCN), and osteopontin (OPN). Collectively, these data provide the foundation for a one-step functionalization method for simple, fast, and effective functionalization of 3D printed scaffolds that support enhanced cell adhesion, proliferation, and differentiation, especially when employed in conjunction with ES. STATEMENT OF SIGNIFICANCE: Three-dimensional (3D) printing is a promising technology for tissue engineering. However, 3D-printing methods have limited ability to produce desired features or electrochemical properties in support of robust cell behavior. To address this deficiency, the current study proposed an integrated, one-step method to increase the cytocompatibility of 3D-printed scaffolds through functionalization leveraging conductive carbon nanotubes (CNTs). This fast and facile functionalization resulted in a homogenous and non-toxic coating of CNTs to the surface, which significantly improved the adhesion, proliferation, and differentiation of cells on the 3D-printed scaffolds.


Nanotubes, Carbon , Tissue Engineering , Bone and Bones , Cell Differentiation , Cell Proliferation , Osteogenesis , Printing, Three-Dimensional , Tissue Scaffolds
19.
ACS Biomater Sci Eng ; 6(8): 4653-4665, 2020 08 10.
Article En | MEDLINE | ID: mdl-33455193

Injectable hydrogels have unique advantages for the repair of irregular tissue defects. In this study, we report a novel injectable carbon nanotube (CNT) and black phosphorus (BP) gel with enhanced mechanical strength, electrical conductivity, and continuous phosphate ion release for tissue engineering. The gel utilized biodegradable oligo(poly(ethylene glycol) fumarate) (OPF) polymer as the cross-linking matrix, with the addition of cross-linkable CNT-poly(ethylene glycol)-acrylate (CNTpega) to grant mechanical support and electric conductivity. Two-dimensional (2D) black phosphorus nanosheets were also infused to aid in tissue regeneration through the steady release of phosphate that results from environmental oxidation of phosphorus in situ. This newly developed BP-CNTpega-gel was found to enhance the adhesion, proliferation, and osteogenic differentiation of MC3T3 preosteoblast cells. With electric stimulation, the osteogenesis of preosteoblast cells was further enhanced with elevated expression of several key osteogenic pathway genes. As monitored with X-ray imaging, the BP-CNTpega-gel demonstrated excellent in situ gelation and cross-linking to fill femur defects, vertebral body cavities, and posterolateral spinal fusion sites in the rabbit. Together, these results indicate that this newly developed injectable BP-CNTpega-gel owns promising potential for future bone and broad types of tissue engineering applications.


Nanotubes, Carbon , Tissue Engineering , Animals , Electric Conductivity , Osteogenesis , Phosphates , Phosphorus , Rabbits
20.
ACS Biomater Sci Eng ; 5(9): 4587-4600, 2019 Sep 09.
Article En | MEDLINE | ID: mdl-33448832

Bone morphogenetic protein-9 (BMP-9) has been shown to be the most osteogenic BMP. Most of these experiments, however, involve an adenovirus-transfection strategy. Here, we used the scaffold-based strategy to study the bone forming ability of recombinant BMP-9 combined with vascular endothelial growth factor (VEGF). A robust, injectable, multicomponent-releasing scaffold in the form of a composite gel was developed by combining chitosan microparticles (MPs) with thermosensitive gel (MPs-gel). The MPs acted as the carriers for BMP-9 and the gel was loaded with VEGF. The developed gel consisted of hydrophobic chains of methyl cellulose (MC) and the cross-linked structures of alginate (Alg) and calcium. Gelation was achieved at physiological temperature and thus facilitated the injection and localization of MPs enabling an increased efficacy of incorporated growth factors at the target site. A release profile of incorporated growth factors over a two-week period showed higher release of VEGF at each time point compared to that of BMP-9. Human mesenchymal stem cells (hMSCs) encapsulated within the MPs-gel maintained their viability. BMP-9 enhanced the proliferation of hMSCs along the surface of MPs. Furthermore, BMP-9 potently induced the osteogenic differentiation of encapsulated hMSCs elucidated by the increased alkaline phosphatase (ALP) activity and the higher expression of ALP, collagen 1, and osteocalcin genes. In addition, in vivo experiments demonstrated that MPs-gel with the combination of BMP-9-VEGF could significantly enhance both subcutaneous and cranial bone formation (p < 0.05). Taken together, the results here strongly suggest that BMP-9-VEGF incorporated MPs-gel holds promise as an injectable bone tissue engineering platform.

...