Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
Biomater Adv ; 164: 213959, 2024 Jul 18.
Article in English | MEDLINE | ID: mdl-39083876

ABSTRACT

Biomaterial-based approaches for bone regeneration seek to explore alternative strategies to repair non-healing fractures and critical-sized bone defects. Fracture non-union occurs due to a number of factors resulting in the formation of bone defects. Rigorous evaluation of the biomaterials in relevant models and assessment of their potential to translate towards clinical use is vital. Large animal experimentation can be used to model fracture non-union while scaling-up materials for clinical use. Growth factors modulate cell phenotype, behaviour and initiate signalling pathways leading to changes in matrix deposition and tissue formation. Bone morphogenetic protein-2 (BMP-2) is a potent osteogenic growth factor, with a rapid clearance time in vivo necessitating clinical use at a high dose, with potential deleterious side-effects. The current studies have examined the potential for Laponite® nanoclay coated poly(caprolactone) trimethacrylate (PCL-TMA900) scaffolds to bind BMP-2 for enhanced osteoinduction in a large animal critical-sized bone defect. An ovine femoral condyle defect model confirmed PCL-TMA900 scaffolds coated with Laponite®/BMP-2 produced significant bone formation compared to the uncoated PCL-TMA 900 scaffold in vivo, assessed by micro-computed tomography (µCT) and histology. This indicated the ability of Laponite® to deliver the bioactive BMP-2 on the PCL-TMA900 scaffold. Bone formed around the Laponite®/BMP-2 coated PCL-TMA900 scaffold, with no erroneous bone formation observed away from the scaffold material confirming localisation of BMP-2 delivery. The current studies demonstrate the ability of a nanoclay to localise and deliver bioactive BMP-2 within a tailored octet-truss scaffold for efficacious bone defect repair in a large animal model with significant implications for translation to the clinic.

2.
Biodes Manuf ; 7(2): 121-136, 2024.
Article in English | MEDLINE | ID: mdl-38497056

ABSTRACT

Autograft or metal implants are routinely used in skeletal repair. However, they fail to provide long-term clinical resolution, necessitating a functional biomimetic tissue engineering alternative. The use of native human bone tissue for synthesizing a biomimetic material ink for three-dimensional (3D) bioprinting of skeletal tissue is an attractive strategy for tissue regeneration. Thus, human bone extracellular matrix (bone-ECM) offers an exciting potential for the development of an appropriate microenvironment for human bone marrow stromal cells (HBMSCs) to proliferate and differentiate along the osteogenic lineage. In this study, we engineered a novel material ink (LAB) by blending human bone-ECM (B) with nanoclay (L, Laponite®) and alginate (A) polymers using extrusion-based deposition. The inclusion of the nanofiller and polymeric material increased the rheology, printability, and drug retention properties and, critically, the preservation of HBMSCs viability upon printing. The composite of human bone-ECM-based 3D constructs containing vascular endothelial growth factor (VEGF) enhanced vascularization after implantation in an ex vivo chick chorioallantoic membrane (CAM) model. The inclusion of bone morphogenetic protein-2 (BMP-2) with the HBMSCs further enhanced vascularization and mineralization after only seven days. This study demonstrates the synergistic combination of nanoclay with biomimetic materials (alginate and bone-ECM) to support the formation of osteogenic tissue both in vitro and ex vivo and offers a promising novel 3D bioprinting approach to personalized skeletal tissue repair. Supplementary Information: The online version contains supplementary material available at 10.1007/s42242-023-00265-z.

3.
Nanoscale ; 15(45): 18457-18472, 2023 Nov 23.
Article in English | MEDLINE | ID: mdl-37941481

ABSTRACT

Clay nanoparticles, in particular synthetic smectites, have generated interest in the field of tissue engineering and regenerative medicine due to their utility as cross-linkers for polymers in biomaterial design and as protein release modifiers for growth factor delivery. In addition, recent studies have suggested a direct influence on the osteogenic differentiation of responsive stem and progenitor cell populations. Relatively little is known however about the mechanisms underlying nanoclay bioactivity and in particular the cellular processes involved in nanoclay-stem cell interactions. In this study we employed confocal microscopy, inductively coupled plasma mass spectrometry and transmission electron microscopy to track the interactions between clay nanoparticles and human bone marrow stromal cells (hBMSCs). In particular we studied nanoparticle cellular uptake mechanisms and uptake kinetics, intracellular trafficking pathways and the fate of endocytosed nanoclay. We found that nanoclay particles present on the cell surface as µm-sized aggregates, enter hBMSCs through clathrin-mediated endocytosis, and their uptake kinetics follow a linear increase with time during the first week of nanoclay addition. The endocytosed particles were observed within the endosomal/lysosomal compartments and we found evidence for both intracellular degradation of nanoclay and exocytosis as well as an increase in autophagosomal activity. Inhibitor studies indicated that endocytosis was required for nanoclay upregulation of alkaline phosphatase activity but a similar dependency was not observed for autophagy. This study into the nature of nanoclay-stem cell interactions, in particular the intracellular processing of nanosilicate, may provide insights into the mechanisms underlying nanoclay bioactivity and inform the successful utilisation of clay nanoparticles in biomaterial design.


Subject(s)
Mesenchymal Stem Cells , Nanoparticles , Humans , Osteogenesis , Clay , Tissue Engineering , Biocompatible Materials , Nanoparticles/chemistry
4.
Adv Mater ; 35(48): e2304461, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37658732

ABSTRACT

Self-assembly, the spontaneous ordering of components into patterns, is widespread in nature and fundamental to generating function across length scales. Morphogen gradients in biological development are paradigmatic as both products and effectors of self-assembly and various attempts have been made to reproduce such gradients in biomaterial design. To date, approaches have typically utilized top-down fabrication techniques that, while allowing high-resolution control, are limited by scale and require chemical cross-linking steps to stabilize morphogen patterns in time. Here, a bottom-up approach to protein patterning is developed based on a novel binary reaction-diffusion process where proteins function as diffusive reactants to assemble a nanoclay-protein composite hydrogel. Using this approach, it is possible to generate scalable and highly stable 3D patterns of target proteins down to sub-cellular resolution through only physical interactions between clay nanoparticles and the proteins and ions present in blood. Patterned nanoclay gels are able to guide cell behavior to precisely template bone tissue formation in vivo. These results demonstrate the feasibility of stabilizing 3D gradients of biological signals through self-assembly processes and open up new possibilities for morphogen-based therapeutic strategies and models of biological development and repair.


Subject(s)
Nanoparticles , Hydrogels , Clay
5.
Bioengineering (Basel) ; 9(7)2022 Jul 21.
Article in English | MEDLINE | ID: mdl-35877383

ABSTRACT

Musculoskeletal disorders are a significant burden on the global economy and public health. Hydrogels have significant potential for enhancing the repair of damaged and injured musculoskeletal tissues as cell or drug delivery systems. Hydrogels have unique physicochemical properties which make them promising platforms for controlling cell functions. Gelatin methacryloyl (GelMA) hydrogel in particular has been extensively investigated as a promising biomaterial due to its tuneable and beneficial properties and has been widely used in different biomedical applications. In this review, a detailed overview of GelMA synthesis, hydrogel design and applications in regenerative medicine is provided. After summarising recent progress in hydrogels more broadly, we highlight recent advances of GelMA hydrogels in the emerging fields of musculoskeletal drug delivery, involving therapeutic drugs (e.g., growth factors, antimicrobial molecules, immunomodulatory drugs and cells), delivery approaches (e.g., single-, dual-release system), and material design (e.g., addition of organic or inorganic materials, 3D printing). The review concludes with future perspectives and associated challenges for developing local drug delivery for musculoskeletal applications.

6.
Bone ; 159: 116389, 2022 06.
Article in English | MEDLINE | ID: mdl-35301163

ABSTRACT

The past decade has seen a growing appreciation for the role of the innate immune response in mediating repair and biomaterial directed tissue regeneration. The long-held view of the host immune/inflammatory response as an obstacle limiting stem cell regenerative activity, has given way to a fresh appreciation of the pivotal role the macrophage plays in orchestrating the resolution of inflammation and launching the process of remodelling and repair. In the context of bone, work over the past decade has established an essential coordinating role for macrophages in supporting bone repair and sustaining biomaterial driven osteogenesis. In this review evidence for the role of the macrophage in bone regeneration and repair is surveyed before discussing recent biomaterial and drug-delivery based approaches that target macrophage modulation with the goal of accelerating and enhancing bone tissue regeneration.


Subject(s)
Biocompatible Materials , Bone Regeneration , Macrophages/physiology , Osteogenesis , Wound Healing
7.
ACS Appl Mater Interfaces ; 13(28): 32624-32639, 2021 Jul 21.
Article in English | MEDLINE | ID: mdl-34228435

ABSTRACT

Organ dysfunction is a major cause of morbidity and mortality. Transplantation is typically the only definitive cure, challenged by the lack of sufficient donor organs. Tissue engineering encompasses the development of biomaterial scaffolds to support cell attachment, proliferation, and differentiation, leading to tissue regeneration. For efficient clinical translation, the forming technology utilized must be suitable for mass production. Herein, uniaxial polyhydroxyalkanoate scaffolds manufactured by pressurized gyration, a hybrid scalable spinning technique, are successfully used in bone, nerve, and cardiovascular applications. Chorioallantoic membrane and in vivo studies provided evidence of vascularization, collagen deposition, and cellular invasion for bone tissue engineering. Highly efficient axonal outgrowth was observed in dorsal root ganglion-based 3D ex vivo models. Human induced pluripotent stem cell derived cardiomyocytes exhibited a mature cardiomyocyte phenotype with optimal calcium handling. This study confirms that engineered polyhydroxyalkanoate-based gyrospun fibers provide an exciting and unique toolbox for the development of scalable scaffolds for both hard and soft tissue regeneration.


Subject(s)
Cells/metabolism , Polyhydroxyalkanoates/chemistry , Tissue Engineering/methods , Tissue Scaffolds/chemistry , Animals , Cell Differentiation/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Chickens , Elastic Modulus , Ganglia, Spinal/metabolism , Humans , Induced Pluripotent Stem Cells/metabolism , Mesenchymal Stem Cells/metabolism , Mice , Myocytes, Cardiac/metabolism , Porosity , Pressure , Rats , Rotation , Schwann Cells/metabolism
8.
ACS Nano ; 15(7): 11202-11217, 2021 Jul 27.
Article in English | MEDLINE | ID: mdl-34180656

ABSTRACT

Synthetic nanostructured materials incorporating both organic and inorganic components offer a unique, powerful, and versatile class of materials for widespread applications due to the distinct, yet complementary, nature of the intrinsic properties of the different constituents. We report a supramolecular system based on synthetic nanoclay (Laponite, Lap) and peptide amphiphiles (PAs, PAH3) rationally designed to coassemble into nanostructured hydrogels with high structural integrity and a spectrum of bioactivities. Spectroscopic and scattering techniques and molecular dynamic simulation approaches were harnessed to confirm that PAH3 nanofibers electrostatically adsorbed and conformed to the surface of Lap nanodisks. Electron and atomic force microscopies also confirmed an increase in diameter and surface area of PAH3 nanofibers after coassembly with Lap. Dynamic oscillatory rheology revealed that the coassembled PAH3-Lap hydrogels displayed high stiffness and robust self-healing behavior while gas adsorption analysis confirmed a hierarchical and heterogeneous porosity. Furthermore, this distinctive structure within the three-dimensional (3D) matrix provided spatial confinement for the nucleation and hierarchical organization of high-aspect ratio hydroxyapatite nanorods into well-defined spherical clusters within the 3D matrix. Applicability of the organic-inorganic PAH3-Lap hydrogels was assessed in vitro using human bone marrow-derived stromal cells (hBMSCs) and ex vivo using a chick chorioallantoic membrane (CAM) assay. The results demonstrated that the organic-inorganic PAH3-Lap hydrogels promote human skeletal cell proliferation and, upon mineralization, integrate with the CAM, are infiltrated by blood vessels, stimulate extracellular matrix production, and facilitate extensive mineral deposition relative to the controls.


Subject(s)
Mesenchymal Stem Cells , Nanofibers , Humans , Hydrogels/chemistry , Durapatite/chemistry , Nanofibers/chemistry , Rheology
9.
ACS Biomater Sci Eng ; 7(6): 2716-2722, 2021 06 14.
Article in English | MEDLINE | ID: mdl-33825442

ABSTRACT

Synthetic clays are promising biomaterials for delivery of therapeutic molecules in regenerative medicine. However, before their use can be translated into clinical applications, their safety must be assessed in human volunteers. The aim of this study was to test the hypothesis that a synthetic nanoclay (LAPONITE) does not cause irritation to the human skin. To achieve this, a nanoclay gel at two different concentrations (1.5 and 3% w/v) was applied on the forearm of healthy volunteers for 24 h. 1% sodium lauryl sulfate (SLS) and 3% (w/v) polyacrylic acid were used as the positive and negative controls, respectively. The compromise in the skin barrier function was measured by trans-epidermal water loss (TEWL), erythema by spectroscopic measurements, and skin inflammatory biomarkers (IL-1α and IL-1RA) by the enzyme-linked immunosorbent assay. We found that the nanoclay caused no prolonged increase in TEWL, erythema, or induction of inflammatory cytokines. This was in contrast to 1% SLS, a known irritant, which induced significant increases in both skin erythema and TEWL. We conclude that the nanoclay is not an irritant and is thus suitable for therapeutic interventions at the skin surface.


Subject(s)
Dermatitis, Irritant , Dermatitis, Irritant/etiology , Gels , Healthy Volunteers , Humans , Sodium Dodecyl Sulfate/adverse effects , Water Loss, Insensible
10.
Biomater Sci ; 9(8): 3150-3161, 2021 Apr 21.
Article in English | MEDLINE | ID: mdl-33730142

ABSTRACT

LAPONITE® clay nanoparticles are known to exert osteogenic effects on human bone marrow stromal cells (HBMSCs), most characteristically, an upregulation in alkaline phosphatase activity and increased calcium deposition. The specific properties of LAPONITE® that impart its bioactivity are not known. In this study the role of lithium, a LAPONITE® degradation product, was investigated through the use of lithium salts and lithium modified LAPONITE® formulations. In contrast to intact particles, lithium ions applied at concentrations equivalent to that present in LAPONITE®, failed to induce any significant increase in alkaline phosphatase (ALP) activity. Furthermore, no significant differences were observed in ALP activity with modified clay structures and the positive effect on osteogenic gene expression did not correlate with the lithium content of modified clays. These results suggest that other properties of LAPONITE® nanoparticles, and not their lithium content, are responsible for their bioactivity.


Subject(s)
Lithium , Nanoparticles , Cell Differentiation , Cells, Cultured , Clay , Humans , Osteogenesis
11.
Methods Mol Biol ; 2147: 63-72, 2021.
Article in English | MEDLINE | ID: mdl-32840811

ABSTRACT

Biofabrication is revolutionizing substitute tissue manufacturing. Skeletal stem cells (SSCs) can be blended with hydrogel biomaterials and printed to form three-dimensional structures that can closely mimic tissues of interest. Our bioink formulation takes into account the potential for cell printing including a bioink nanocomposite that contains low fraction polymeric content to facilitate cell encapsulation and survival, while preserving hydrogel integrity and mechanical properties following extrusion. Clay inclusion to the nanocomposite strengthens the alginate-methylcellulose network providing a biopaste with unique shear-thinning properties that can be easily prepared under sterile conditions. SSCs can be mixed with the clay-based paste, and the resulting bioink can be printed in 3D structures ready for implantation. In this chapter, we provide the methodology for preparation, encapsulation, and printing of SSCs in a unique clay-based bioink.


Subject(s)
Bioprinting/methods , Bone Regeneration/physiology , Clay/chemistry , Nanocomposites/chemistry , Tissue Engineering/methods , Tissue Scaffolds/chemistry , Biocompatible Materials/chemical synthesis , Biocompatible Materials/chemistry , Bone Substitutes/chemical synthesis , Bone Substitutes/chemistry , Bone and Bones/cytology , Bone and Bones/physiology , Cell Culture Techniques/instrumentation , Cell Culture Techniques/methods , Cells, Cultured , Humans , Ink , Microtechnology/methods , Printing, Three-Dimensional , Silicates/chemistry , Stem Cells/cytology , Stem Cells/physiology , Tissue Engineering/instrumentation
12.
Mater Sci Eng C Mater Biol Appl ; 118: 111440, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33255033

ABSTRACT

Functionalized scaffolds hold promise for stem cell therapy by controlling stem cell fate and differentiation potential. Here, we have examined the potential of a 2-dimensional (2D) scaffold to stimulate bone regeneration. Solubilized extracellular matrix (ECM) from human bone tissue contains native extracellular cues for human skeletal cells that facilitate osteogenic differentiation. However, human bone ECM displays limited mechanical strength and degradation stability under physiological conditions, necessitating modification of the physical properties of ECM before it can be considered for tissue engineering applications. To increase the mechanical stability of ECM, we explored the potential of synthetic Laponite® (LAP) clay as a counter material to prepare a 2D scaffold using Layer-by-Layer (LbL) self-assembly. The LAP and ECM multilayer nanofilms (ECM/LAP film) were successfully generated through electrostatic and protein-clay interactions. Furthermore, to enhance the mechanical properties of the ECM/LAP film, application of a NaCl solution wash step, instead of deionized water following LAP deposition resulted in the generation of stable, multi-stacked LAP layers which displayed enhanced mechanical properties able to sustain human skeletal progenitor cell growth. The ECM/LAP films were not cytotoxic and, critically, showed enhanced osteogenic differentiation potential as a consequence of the synergistic effects of ECM and LAP. In summary, we demonstrate the fabrication of a novel ECM/LAP nanofilm layer material with potential application in hard tissue engineering.


Subject(s)
Extracellular Matrix , Osteogenesis , Bone and Bones , Cell Differentiation , Humans , Silicates , Stem Cells , Tissue Scaffolds
13.
Biomacromolecules ; 21(6): 2096-2103, 2020 06 08.
Article in English | MEDLINE | ID: mdl-32267672

ABSTRACT

Because nitric oxide (NO) gas is an endogenously produced signaling molecule related to numerous physiological functions, manystudies have been conducted to develop NO delivery systems for potential biomedical applications. However, NO is a reactive radical gas molecule that has a very short life-time and readily transforms into nitrogen oxide species via reaction with oxygen species. Therefore, it is necessary to develop an NO delivery carrier that allows local release of the NO gas at the site of application. In this study, Laponite (LP) nanoclay was used to fabricate an NO delivery carrier through the formation of Laponite-polyamine (LP-PAn) composites. The Laponite clay and pentaethylenehexamine (PEHA) formed a macromolecular structure by electrostatic interaction and the nitric oxide donor, N-diazeniumdiolate (NONOates), was synthesized into the LP-PAn composite. We investigated the conformation of the LP-PAn composite structure and the NO donor formation by ζ potential, X-ray diffraction, and UV-vis and Fourier transform infrared (FT-IR) spectroscopies and also by analyzing the NO release profile. Additionally, we confirmed the applicability in biomedical applications via a cell viability and in vitro endothelial cell tube formation assay.


Subject(s)
Hydrogels , Nitric Oxide , Polyamines , Silicates , Spectroscopy, Fourier Transform Infrared
14.
Biofabrication ; 12(3): 035010, 2020 05 12.
Article in English | MEDLINE | ID: mdl-32259804

ABSTRACT

Acellular soft hydrogels are not ideal for hard tissue engineering given their poor mechanical stability, however, in combination with cellular components offer significant promise for tissue regeneration. Indeed, nanocomposite bioinks provide an attractive platform to deliver human bone marrow stromal cells (HBMSCs) in three dimensions producing cell-laden constructs that aim to facilitate bone repair and functionality. Here we present the in vitro, ex vivo and in vivo investigation of bioprinted HBMSCs encapsulated in a nanoclay-based bioink to produce viable and functional three-dimensional constructs. HBMSC-laden constructs remained viable over 21 d in vitro and immediately functional when conditioned with osteogenic media. 3D scaffolds seeded with human umbilical vein endothelial cells (HUVECs) and loaded with vascular endothelial growth factor (VEGF) implanted ex vivo into a chick chorioallantoic membrane (CAM) model showed integration and vascularisation after 7 d of incubation. In a pre-clinical in vivo application of a nanoclay-based bioink to regenerate skeletal tissue, we demonstrated bone morphogenetic protein-2 (BMP-2) absorbed scaffolds produced extensive mineralisation after 4 weeks (p < 0.0001) compared to the drug-free and alginate controls. In addition, HBMSC-laden 3D printed scaffolds were found to significantly (p < 0.0001) support bone tissue formation in vivo compared to acellular and cast scaffolds. These studies illustrate the potential of nanoclay-based bioink, to produce viable and functional constructs for clinically relevant skeletal tissue regeneration.


Subject(s)
Bone and Bones/blood supply , Clay/chemistry , Minerals/metabolism , Nanocomposites/chemistry , Neovascularization, Physiologic , Printing, Three-Dimensional , Tissue Engineering , Tissue Scaffolds/chemistry , Animals , Bone Morphogenetic Protein 2/pharmacology , Bone and Bones/drug effects , Calcification, Physiologic/drug effects , Cell Differentiation/drug effects , Cell Survival/drug effects , Chickens , Chorioallantoic Membrane/drug effects , Humans , Implants, Experimental , Mice , Models, Animal , Neovascularization, Physiologic/drug effects , Osteogenesis/drug effects , Silicates/chemistry , Subcutaneous Tissue/drug effects
15.
Nat Commun ; 11(1): 1365, 2020 03 13.
Article in English | MEDLINE | ID: mdl-32170076

ABSTRACT

Nanoclays have generated interest in biomaterial design for their ability to enhance the mechanics of polymeric materials and impart biological function. As well as their utility as physical cross-linkers, clays have been explored for sustained localization of biomolecules to promote in vivo tissue regeneration. To date, both biomolecule-clay and polymer-clay nanocomposite strategies have utilised the negatively charged clay particle surface. As such, biomolecule-clay and polymer-clay interactions are set in competition, potentially limiting the functional enhancements achieved. Here, we apply specific bisphosphonate interactions with the positively charged clay particle edge to develop self-assembling hydrogels and functionalized clay nanoparticles with preserved surface exchange capacity. Low concentrations of nanoclay are applied to cross-link hyaluronic acid polymers derivatised with a pendant bisphosphonate to generate hydrogels with enhanced mechanical properties and preserved protein binding able to sustain, for over six weeks in vivo, the localized activity of the clinically licensed growth factor BMP-2.


Subject(s)
Diphosphonates/metabolism , Hydrogels/chemistry , Intercellular Signaling Peptides and Proteins/metabolism , Nanocomposites/chemistry , Nanoparticles/chemistry , Animals , Bone Morphogenetic Protein 2/metabolism , Clay , Drug Delivery Systems , Female , Materials Testing , Mice , Polymers/chemistry , Protein Binding , Silicates
16.
Acta Biomater ; 100: 378-387, 2019 12.
Article in English | MEDLINE | ID: mdl-31541735

ABSTRACT

The retention and sustained activity of therapeutic proteins at delivery sites are goals of regenerative medicine. Vascular endothelial growth factor (VEGF) has significant potential in promoting the growth and regeneration of blood vessels but is intrinsically labile. This is exacerbated by the inflammatory microenvironments at sites requiring regeneration. For VEGF to be efficacious, it may require a carrier that stabilises it, protects it from degradation and retains it at the site of interest. In this study, we tested the hypothesis that injectable nanoclay gels comprising Laponite™ XLG (a synthetic hectorite clay) can stabilise VEGF and retain it in the active form for therapeutic delivery. To achieve this, VEGF was incorporated in Laponite gels and its activity tested at a range of concentrations using in vitro cell culture tubulogenesis assays and in vivo angiogenesis assays. We found that VEGF-Laponite gels enhanced tubulogenesis in a dose-dependent manner in vitro. When administered subcutaneously in vivo, Laponite was retained at the injection site for up to a period of three weeks and promoted a 4-fold increase in blood vessel formation compared with that of alginate or vehicle controls as confirmed by CD31 staining. Notably, as compared to alginate, Laponite gels did not release VEGF, indicating a strong interaction between the growth factor and the nanoclay and suggesting that Laponite enhancement of VEGF efficacy is due to its retention at the implantation site for a prolonged period. Our approach provides a robust method for the delivery of bioactive recombinant VEGF without the necessity for complex hydrogel or protein engineering. STATEMENT OF SIGNIFICANCE: In medicine, it is important to deliver drugs to a particular location in the body. Often, however, the drugs are quickly broken down and carried away in the blood before they can exert their effect. In this study, we used a type of synthetic clay, called Laponite™, to preserve a molecule, named VEGF, that stimulates the growth of blood vessels. Previously, we have been able to bind VEGF to the surface of clays, but the clay is not effective when injected or applied as a gel. Herein, we show that we can mix VEGF with the clay and that it strongly stimulates blood vessel growth. We speculate that this would be a useful material for skin wound healing.


Subject(s)
Gels/chemistry , Injections , Nanoparticles/chemistry , Neovascularization, Physiologic , Silicates/chemistry , Animals , Blood Vessels/physiology , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Male , Mice , Vascular Endothelial Growth Factor A/metabolism
17.
Biofabrication ; 11(3): 035027, 2019 06 12.
Article in English | MEDLINE | ID: mdl-30991370

ABSTRACT

Bioprinting of living cells is rapidly developing as an advanced biofabrication approach to engineer tissues. Bioinks can be extruded in three-dimensions (3D) to fabricate complex and hierarchical constructs for implantation. However, a lack of functionality can often be attributed to poor bioink properties. Indeed, advanced bioinks encapsulating living cells should: (i) present optimal rheological properties and retain 3D structure post fabrication, (ii) promote cell viability and support cell differentiation, and (iii) localise proteins of interest (e.g. vascular endothelial growth factor (VEGF)) to stimulate encapsulated cell activity and tissue ingrowth upon implantation. In this study, we present the results of the inclusion of a synthetic nanoclay, Laponite® (LPN) together with a gelatin methacryloyl (GelMA) bioink and the development of a functional cell-instructive bioink. A nanocomposite bioink displaying enhanced shape fidelity retention and interconnected porosity within extrusion-bioprinted fibres was observed. Human bone marrow stromal cell (HBMSC) viability within the nanocomposite showed no significant changes over 21 days of culture in LPN-GelMA (85.60 ± 10.27%), compared to a significant decrease in GelMA from 7 (95.88 ± 2.90%) to 21 days (55.54 ± 14.72%) (p < 0.01). HBMSCs were observed to proliferate in LPN-GelMA with a significant increase in cell number over 21 days (p < 0.0001) compared to GelMA alone. HBMSC-laden LPN-GelMA scaffolds supported osteogenic differentiation evidenced by mineralised nodule formation, including in the absence of the osteogenic drug dexamethasone. Ex vivo implantation in a chick chorioallantoic membrane model, demonstrated excellent integration of the bioink constructs in the vascular chick embryo after 7 days of incubation. VEGF-loaded LPN-GelMA constructs demonstrated significantly higher vessel penetration than GelMA-VEGF (p < 0.0001) scaffolds. Integration and vascularisation was directly related to increased drug absorption and retention by LPN-GelMA compared to LPN-free GelMA. In summary, a novel light-curable nanocomposite bioink for 3D skeletal regeneration supportive of cell growth and growth factor retention and delivery, evidenced by ex vivo vasculogenesis, was developed with potential application in hard and soft tissue reparation.


Subject(s)
Gelatin/chemistry , Ink , Nanocomposites/chemistry , Neovascularization, Physiologic , Osteogenesis , Silicates/chemistry , Animals , Bioprinting , Cattle , Cell Proliferation , Cell Survival , Chickens , Chorioallantoic Membrane/metabolism , Humans , Hydrogels/chemistry , Mesenchymal Stem Cells/cytology , Methacrylates/chemistry , Muramidase/metabolism , Porosity , Serum Albumin, Bovine/metabolism , Swine , Vascular Endothelial Growth Factor A/metabolism
18.
Adv Healthc Mater ; 7(15): e1800331, 2018 08.
Article in English | MEDLINE | ID: mdl-29911340

ABSTRACT

Laponite nanoparticles have attracted attention in the tissue engineering field for their protein interactions, gel-forming properties, and, more recently, osteogenic bioactivity. Despite growing interest in the osteogenic properties of Laponite, the application of Laponite colloidal gels to host the osteogenic differentiation of responsive stem cell populations remains unexplored. Here, the potential to harness the gel-forming properties of Laponite to generate injectable bioactive microenvironments for osteogenesis is demonstrated. A diffusion/dialysis gelation method allows the rapid formation of stable transparent gels from injectable, thixotropic Laponite suspensions in physiological fluids. Upon contact with buffered saline or blood serum, nanoporous gel networks exhibiting, respectively, fivefold and tenfold increases in gel stiffness are formed due to the reorganization of nanoparticle interactions. Laponite diffusion gels are explored as osteogenic microenvironments for skeletal stem cell containing populations. Laponite films support cell adhesion, proliferation, and differentiation of human bone marrow stromal cells in 2D. Laponite gel encapsulation significantly enhances osteogenic protein expression compared with 3D pellet culture controls. In both 2D and 3D conditions, cell associated mineralization is strongly enhanced. This study demonstrates that Laponite diffusion gels offer considerable potential as biologically active and clinically relevant bone tissue engineering scaffolds.


Subject(s)
Gels/chemistry , Osteogenesis/physiology , Silicates/chemistry , Cell Adhesion/physiology , Cell Differentiation/physiology , Cell Proliferation/physiology , Cells, Cultured , Cellular Microenvironment/physiology , Humans , Mesenchymal Stem Cells/cytology , Tissue Engineering/methods
19.
Biomaterials ; 159: 204-214, 2018 03.
Article in English | MEDLINE | ID: mdl-29331807

ABSTRACT

Clay nanoparticles, composites and hydrogels are emerging as a new class of biomaterial with exciting potential for tissue engineering and regenerative medicine applications. Clay particles have been extensively explored in polymeric nanocomposites for self-assembly and enhanced mechanical properties as well as for their potential as drug delivery modifiers. In recent years, a cluster of studies have explored cellular interactions with clay nanoparticles alone or in combination with polymeric matrices. These pioneering studies have suggested new and unforeseen utility for certain clays as bioactive additives able to enhance cellular functions including adhesion, proliferation and differentiation, most notably for osteogenesis. This review examines the recent literature describing the potential effects of clay-based nanomaterials on cell function and examines the potential role of key clay physicochemical properties in influencing such interactions and their exciting possibilities for regenerative medicine.


Subject(s)
Biocompatible Materials/chemistry , Nanoparticles/chemistry , Animals , Cell Adhesion/physiology , Cell Differentiation/physiology , Humans , Nanocomposites/chemistry , Regenerative Medicine/methods , Stem Cells/cytology , Tissue Engineering/methods
20.
J Tissue Eng Regen Med ; 10(3): 187-98, 2016 Mar.
Article in English | MEDLINE | ID: mdl-25491789

ABSTRACT

This review explores the application of hydrogels in orthopaedic clinical situations which may benefit from enhanced growth factor delivery and improved osteogenesis of bone graft material. Hydrogels are defined, and in vivo evidence supporting their application in these clinical areas is explored. Our focus is on clinically pertinent properties, such as the chemistry of formation, biocompatibility, efficacy of cell and growth factor delivery, ability to withstand mechanical loading and potential to be delivered via an injection. Naturally derived hydrogels, such as gelatin, hyaluronic acid and fibroin, together with a number of synthetic polyethylene glycol-based gels combined with protease-sensitive domains, have shown excellent biocompatibility. There is significant literature evidence supporting the ability of hydrogels to facilitate growth factor and cell delivery. Burst release of the selected growth factor remains a consistent challenge, which has been overcome in some studies with chemical modifications of the hydrogel. Interestingly, a number of studies detail percutaneous delivery with hydrogels combined with calcium-based minerals to enhance osteogenicity, with mixed results. Few of the studies explored the biomechanical properties of the materials, and none of the studies reviewed demonstrated the ability of a hydrogel/graft material to withstand mechanical loading in a clinically relevant segmental bone defect model.


Subject(s)
Bone Regeneration/drug effects , Fracture Healing/drug effects , Hydrogel, Polyethylene Glycol Dimethacrylate/pharmacology , Animals , Humans , Hydrogel, Polyethylene Glycol Dimethacrylate/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL