Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
1.
Carbohydr Polym ; 291: 119550, 2022 Sep 01.
Article in English | MEDLINE | ID: mdl-35698380

ABSTRACT

Bacterial exopolysaccharides (EPS) are water-soluble polymers consisting of repeating sugar moieties that serve a wide range of functions for the bacterial species that produce them. Their functions include biofilm matrix constituent, nutrient retention, protection from environmental threats and even pathogenicity. EPS have also been exploited for use in various applications in the biomedical field: most notably as viscosupplements, drug delivery vehicles and in tissue engineering constructs. The use of EPS in bone tissue engineering has increased in recent years due to the wide range of compounds available, low cost, and ease of production on an industrial scale. This review discusses the extraction and purification methods employed to produce bacterial EPS. A particular focus is on bone-related tissue engineering applications where EPS is the primary active agent, or as a scaffold matrix, as well as a carrier for osteopromotive agents.


Subject(s)
Biocompatible Materials , Polysaccharides, Bacterial , Bacteria , Biocompatible Materials/pharmacology , Bone Regeneration
2.
Mater Today Bio ; 10: 100110, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33997761

ABSTRACT

Novel approaches, combining technology, biomaterial design, and cutting-edge cell culture, have been increasingly considered to advance the field of tissue engineering and regenerative medicine. Within this context, acoustic manipulation to remotely control spatial cellular organization within a carrier matrix has arisen as a particularly promising method during the last decade. Acoustic or sound-induced manipulation takes advantage of hydrodynamic forces exerted on systems of particles within a liquid medium by standing waves. Inorganic or organic particles, cells, or organoids assemble within the nodes of the standing wave, creating distinct patterns in response to the applied frequency and amplitude. Acoustic manipulation has advanced from micro- or nanoparticle arrangement in 2D to the assembly of multiple cell types or organoids into highly complex in vitro tissues. In this review, we discuss the past research achievements in the field of acoustic manipulation with particular emphasis on biomedical application. We survey microfluidic, open chamber, and high throughput devices for their applicability to arrange non-living and living units in buffer or hydrogels. We also investigate the challenges arising from different methods, and their prospects to gain a deeper understanding of in vitro tissue formation and application in the field of biomedical engineering.

3.
Eur Cell Mater ; 41: 40-51, 2021 01 07.
Article in English | MEDLINE | ID: mdl-33411938

ABSTRACT

The repair of focal cartilage defects remains one of the foremost issues in the field of orthopaedics. Chondral defects may arise from a variety of joint pathologies and left untreated, will likely progress to osteoarthritis. Current repair techniques, such as microfracture, result in short-term clinical improvements but have poor long-term outcomes. Emerging scaffold-based repair strategies have reported superior outcomes compared to microfracture and motivate the development of new biomaterials for this purpose. In this study, unique composite implants consisting of a base porous reinforcing component (woven poly(ε-caprolactone)) infiltrated with 1 of 2 hydrogels (self-assembling peptide or thermo-gelling hyaluronan) or bone marrow aspirate were evaluated. The objective was to evaluate cartilage repair with composite scaffold treatment compared to the current standard of care (microfracture) in a translationally relevant large animal model, the Yucatan minipig. While many cartilage-repair studies have shown some success in vivo, most are short term and not clinically relevant. Informed by promising 6-week findings, a 12-month study was carried out and those results are presented here. To aid in comparisons across platforms, several structural and functionally relevant outcome measures were performed. Despite positive early findings, the long-term results indicated less than optimal structural and mechanical results with respect to cartilage repair, with all treatment groups performing worse than the standard of care. This study is important in that it brings much needed attention to the importance of performing translationally relevant long-term studies in an appropriate animal model when developing new clinical cartilage repair approaches.


Subject(s)
Cartilage, Articular , Animals , Biocompatible Materials , Cartilage, Articular/surgery , Disease Models, Animal , Hyaluronic Acid , Swine , Swine, Miniature
4.
Mater Today Bio ; 7: 100058, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32613184

ABSTRACT

Biofabrication is providing scientists and clinicians the ability to produce engineered tissues with desired shapes and gradients of composition and biological cues. Typical resolutions achieved with extrusion-based bioprinting are at the macroscopic level. However, for capturing the fibrillar nature of the extracellular matrix (ECM), it is necessary to arrange ECM components at smaller scales, down to the micron and the molecular level. Herein, we introduce a bioink containing the tyramine derivative of hyaluronan (HA; henceforth known as THA) and collagen (Col) type 1. In this bioink, similar to connective tissues, Col is present in the fibrillar form, and HA functions as a viscoelastic space filler. THA was enzymatically cross-linked under mild conditions allowing simultaneous Col fibrillogenesis, thus achieving a homogeneous distribution of Col fibrils within the viscoelastic HA-based matrix. The THA-Col composite displayed synergistic properties in terms of storage modulus and shear thinning, translating into good printability. Shear-induced alignment of the Col fibrils along the printing direction was achieved and quantified via immunofluorescence and second-harmonic generation. Cell-free and cell-laden constructs were printed and characterized, analyzing the influence of the controlled microscopic anisotropy on human bone marrow-derived mesenchymal stromal cell (hMSC) migration. Anisotropic HA-Col showed cell-instructive properties modulating hMSC adhesion, morphology, and migration from micropellets stimulated by the presence and the orientation of Col fibers. Actin filament staining showed that hMSCs embedded in aligned constructs displayed increased cytoskeleton alignment along the fibril direction. Based on gene expression of cartilage/bone markers and ECM production, hMSCs embedded in the isotropic bioink displayed chondrogenic differentiation comparable with standard pellet culture by means of proteoglycan production (safranin O staining and proteoglycan quantification). The possibility of printing matrix components with control over microscopic alignment brings biofabrication one step closer to capturing the complexity of native tissues.

5.
Biofabrication ; 12(3): 032001, 2020 05 28.
Article in English | MEDLINE | ID: mdl-32259809

ABSTRACT

Biofabrication is enriching the tissue engineering field with new ways of producing structurally organized complex tissues. Among the numerous bioinks under investigation, hyaluronic acid (HA) and its derivatives stand out for their biological relevance, cytocompatibility, shear-thinning properties, and potential to fine-tune the desired properties with chemical modification. In this paper, we review the recent advances on bioinks containing HA. The available literature is presented based on subjects including the rheological properties in connection with printability, the chemical strategies for endowing HA with the desired properties, the clinical application, the most advanced preclinical studies, the advantages and limitations in comparison with similar biopolymer-based bioinks, and future perspectives.


Subject(s)
Bioprinting , Hyaluronic Acid/chemistry , Ink , Printing, Three-Dimensional , Cross-Linking Reagents/chemistry , Rheology
6.
Eur Cell Mater ; 39: 1-17, 2020 01 03.
Article in English | MEDLINE | ID: mdl-31899537

ABSTRACT

Appropriate cell sources, bioactive factors and biomaterials for generation of functional and integrated annulus fibrosus (AF) tissue analogues are still an unmet need. In the present study, the AF cell markers, collagen type I, cluster of differentiation 146 (CD146), mohawk (MKX) and smooth muscle protein 22α (SM22α) were found to be suitable indicators of functional AF cell induction. In vitro 2D culture of human AF cells showed that transforming growth factor ß1 (TGF-ß1) upregulated the expression of the functional AF markers and increased cell contractility, indicating that TGF-ß1-pre-treated AF cells were an appropriate cell source for AF tissue regeneration. Furthermore, a tissue engineered construct, composed of polyurethane (PU) scaffold with a TGF-ß1-supplemented collagen type I hydrogel and human AF cells, was evaluated with in vitro 3D culture and ex vivo preclinical bioreactor-loaded organ culture models. The collagen type I hydrogel helped maintaining the AF functional phenotype. TGF-ß1 supplement within the collagen I hydrogel further promoted cell proliferation and matrix production of AF cells within in vitro 3D culture. In the ex vivo IVD organ culture model with physiologically relevant mechanical loading, TGF-ß1 supplement in the transplanted constructs induced the functional AF cell phenotype and enhanced collagen matrix synthesis. In conclusion, TGF-ß1-containing collagen-PU constructs can induce the functional cell phenotype of human AF cells in vitro and in situ. This combined cellular, biomaterial and bioactive agent therapy has a great potential for AF tissue regeneration and rupture repair.


Subject(s)
Annulus Fibrosus/pathology , Collagen/pharmacology , Polyurethanes/pharmacology , Tissue Scaffolds/chemistry , Transforming Growth Factor beta1/pharmacology , Wound Healing/drug effects , Adult , Animals , Annulus Fibrosus/drug effects , Biomarkers/metabolism , Cattle , Cells, Cultured , Female , Gene Expression Regulation/drug effects , Humans , Male , Middle Aged , Organ Culture Techniques , Phenotype , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rupture , Wound Healing/genetics
7.
Hernia ; 24(6): 1201-1210, 2020 12.
Article in English | MEDLINE | ID: mdl-31781966

ABSTRACT

PURPOSE: Mesh-related infection is a critical outcome for patients with hernia defect stabilized with synthetic or biological meshes. Even though bioactive meshes loaded with antibiotics or antiseptics are slowly emerging in the market, the available solutions still lack versatility. Here, we proposed a polymer solution, i.e., hyaluronic acid-poly(N-isopropylacrylamide) (HApN), which forms a hydrogel to be used as coating for meshes only when it reaches body temperature. METHODS: We assessed how the gelation of HApN was influenced by the incorporation of different antibiotic and antiseptic formulations, and how this gel can be used to coat several mesh types. The impact of the coating on the elastic behavior of a macroporous mesh was tested under cyclic elongation condition. Finally, we selected two different coating formulations, one based on antibiotics (gentamicin + rifampicin) and one based on antiseptic (chlorhexidine) and tested in vitro their antimicrobial efficacies. RESULTS: HApN can be used as carrier for different antimicrobial agents, without having a strong influence on its gelation behavior. Porous or dense meshes can be coated with this polymer, even though the stability was not optimal on macroporous meshes such as Optilene when pores are too large. HApN loaded with drugs inhibited in vitro the growth of several Gram-positive and Gram-negative bacteria. CONCLUSION: Compared to the available technologies developed to endow meshes with antibacterial activity, the proposed HApN offers further versatility with potential to prevent mesh-related infection in hernioplasty.


Subject(s)
Anti-Infective Agents/therapeutic use , Hernia/drug therapy , Herniorrhaphy/methods , Hyaluronic Acid/therapeutic use , Surgical Mesh/microbiology , Animals , Anti-Infective Agents/pharmacology , Female , Humans , Hyaluronic Acid/pharmacology , Male
8.
Acta Biomater ; 101: 293-303, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31726249

ABSTRACT

Biomaterials play a pivotal role in cell-free cartilage repair approaches, where cells must migrate through the scaffold, fill the defect, and then proliferate and differentiate facilitating tissue remodeling. Here we used multiple assays to test the influence of chemokines and growth factors on cell migration and cartilage repair in two different hyaluronan (HA)-based hydrogels. We first investigated bone marrow Mesenchymal Stromal Cells (BMSC) migration in vitro, in response to different concentrations of platelet-derived growth factor-BB (PDGF-BB), chemokine ligand 5 (CCL5/RANTES) and stromal cell-derived factor 1 (SDF-1), using a 3D spheroid-based assay. PDGF-BB was selected as most favourable chemotactic agent, and MSC migration was assessed in the context of physical impediment to cell recruitment by testing Fibrin-HA and HA-Tyramine hydrogels of different cross-linking densities. Supplementation of PDGF-BB stimulated progressive migration of MSC through the gels over time. We then investigated in situ cell migration into the hydrogels with and without PDGF-BB, using a cartilage-bone explant model implanted subcutaneously in athymic mice. In vivo studies show that when placed into an osteochondral defect, both hydrogels supported endogenous cell infiltration and provided an amenable microenvironment for cartilage production. These processes were best supported in Fibrin-HA hydrogel in the absence of PDGF-BB. This study used an advanced preclinical testing platform to select an appropriate microenvironment provided by implanted hydrogels, demonstrating that HA-based hydrogels can promote the initial and critical step of endogenous cell recruitment and circumvent some of the clinical challenges in cartilage tissue repair. STATEMENT OF SIGNIFICANCE: The challenge of articular cartilage repair arises from its complex structure and architecture, which confers the unique mechanical behavior of the extracellular matrix. The aim of our research is to identify biomaterials for implants that can support migration of endogenous stem and progenitor cell populations from cartilage and bone tissue, in order to permanently replace damaged cartilage with the original hyaline structure. Here, we present an in vitro 3D spheroid-based migration assay and an osteochondral defect model, which provide the opportunity to assess biomaterials and biomolecules, and to get stronger experimental evidence of the not well-characterized dynamic process of endogenous cells colonization in an osteochondral defect. Furthermore, the delicate step of early cell migration into biomaterials towards functional tissue engineering is reproduced. These tests can be used for pre-clinical testing of newly developed material designs in the field of scaffold engineering.


Subject(s)
Biomimetic Materials/pharmacology , Cartilage, Articular/metabolism , Extracellular Matrix/metabolism , Hyaluronic Acid/pharmacology , Hydrogels/pharmacology , Adolescent , Aged , Animals , Becaplermin/pharmacology , Cartilage, Articular/drug effects , Cattle , Cell Movement/drug effects , Chondrogenesis/drug effects , Cross-Linking Reagents/pharmacology , Extracellular Matrix/drug effects , Humans , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Middle Aged , Spheroids, Cellular/drug effects , Tyramine/pharmacology , Wound Healing/drug effects
9.
Eur Cell Mater ; 36: 200-217, 2018 10 25.
Article in English | MEDLINE | ID: mdl-30370912

ABSTRACT

Numerous studies show promise for cell-based tissue engineering strategies aiming to repair painful intervertebral disc (IVD) degeneration. However, clinical translation to human IVD repair is slow. In the present study, the regenerative potential of an autologous nucleus pulposus (NP)-cell-seeded thermoresponsive hyaluronic acid hydrogel in human lumbar IVDs was assessed under physiological conditions. First, agarose-encased in vitro constructs were developed, showing greater than 90 % NP cell viability and high proteoglycan deposition within HA-pNIPAM hydrogels following 3 weeks of dynamic loading. Second, a bovine-induced IVD degeneration model was used to optimise and validate T1ρ magnetic resonance imaging (MRI) for detection of changes in proteoglycan content in isolated intact IVDs. Finally, isolated intact human lumbar IVDs were pre-scanned using the established MRI sequence. Then, IVDs were injected with HA-pNIPAM hydrogel alone or autologous NP-cell-seeded. Next, the treated IVDs were cultured under cyclic dynamic loading for 5 weeks. Post-treatment T1ρ values were significantly higher as compared to pre-treatment scans within the same IVD and region of interest. Histological evaluation of treated human IVDs showed that the implanted hydrogel alone accumulated proteoglycans, while those that contained NP cells also displayed neo-matrix-surrounded cells within the gel. The study indicated a clinical potential for repairing early degenerative human IVDs using autologous cells/hydrogel suspensions. This unique IVD culture set-up, combined with the long-term physiological culture of intact human IVDs, allowed for a more clinically relevant evaluation of human tissue repair and regeneration, which otherwise could not be replicated using the available in vitro and in vivo models.


Subject(s)
Hyaluronic Acid/chemistry , Hydrogels/chemistry , Nucleus Pulposus/transplantation , Organ Culture Techniques , Regeneration , Temperature , Acrylic Resins/chemistry , Animals , Bioreactors , Cattle , Collagen Type I/metabolism , Collagen Type II/metabolism , Compressive Strength , Elastic Modulus , Female , Humans , Magnetic Resonance Imaging , Male , Middle Aged , Nucleus Pulposus/diagnostic imaging , Proteoglycans/metabolism , Transplantation, Autologous , Wound Healing
10.
Biofabrication ; 10(4): 044104, 2018 09 25.
Article in English | MEDLINE | ID: mdl-30188324

ABSTRACT

Extrusion-based three-dimensional bioprinting relies on bioinks engineered to combine viscoelastic properties for extrusion and shape retention, and biological properties for cytocompatibility and tissue regeneration. To satisfy these conflicting requirements, bioinks often utilize either complex mixtures or complex modifications of biopolymers. In this paper we introduce and characterize a bioink exploiting a dual crosslinking mechanism, where an enzymatic reaction forms a soft gel suitable for cell encapsulation and extrusion, while a visible light photo-crosslinking allows shape retention of the printed construct. The influence of cell density and cell type on the rheological and printability properties was assessed correlating the printing outcomes with the damping factor, a rheological characteristic independent of the printing system. Stem cells, chondrocytes and fibroblasts were encapsulated, and their viability was assessed up to 14 days with live/dead, alamar blue and trypan blue assays. Additionally, the impact of the printing parameters on cell viability was investigated. Owing to its straightforward preparation, low modification, presence of two independent crosslinking mechanisms for tuning shear-thinning independently of the final shape fixation, the use of visible green instead of UV light, the possibility of encapsulating and sustaining the viability of different cell types, the hyaluronan bioink here presented is a valid biofabrication tool for producing 3D printed tissue-engineered constructs.


Subject(s)
Bioprinting , Cross-Linking Reagents/chemistry , Hyaluronic Acid/chemistry , Ink , Light , Printing, Three-Dimensional , Animals , Cattle , Cell Count , Cell Survival , Elasticity , Humans , Viscosity
11.
Hernia ; 22(6): 961-974, 2018 12.
Article in English | MEDLINE | ID: mdl-30168006

ABSTRACT

BACKGROUND: Infectious complications following mesh implantation for abdominal wall repair appear in 0.7 up to 26.6% of hernia repairs and can have a detrimental impact for the patient. To prevent or to treat mesh-related infection, the scientific community is currently developing a veritable arsenal of antibacterial meshes. The numerous and increasing reports published every year describing new technologies indicate a clear clinical need, and an academic interest in solving this problem. Nevertheless, to really appreciate, to challenge, to compare and to optimize the antibacterial properties of next generation meshes, it is important to know which models are available and to understand them. PURPOSE: We proposed for the first time, a complete overview focusing only on the in vitro and in vivo models which have been employed specifically in the field of antibacterial meshes for hernia repair. RESULTS AND CONCLUSION: From this investigation, it is clear that there has been vast progress and breadth in new technologies and models to test them. However, it also shows that standardization or adoption of a more restricted number of models would improve comparability and be a benefit to the field of study.


Subject(s)
Anti-Infective Agents/administration & dosage , Herniorrhaphy , Models, Animal , Models, Biological , Surgical Mesh , Surgical Wound Infection/prevention & control , Animals , Bacterial Adhesion , Bacteriolysis , Biofilms , Disk Diffusion Antimicrobial Tests , Humans , Materials Testing
12.
Acta Biomater ; 77: 201-211, 2018 09 01.
Article in English | MEDLINE | ID: mdl-30030175

ABSTRACT

Insufficient vascularization is a major cause for the development of non-unions. To overcome this problem, adipose tissue-derived microvascular fragments (MVF) may serve as vascularization units. However, their application into bone defects needs a carrier system. Herein, we analyzed whether this is achieved by a thermoresponsive hydrogel (TRH). MVF were isolated from CD-1 mice and cultivated after incorporation into TRH, while non-incorporated MVF served as controls. Viability of MVF was assessed immunohistochemically over a 7-day period. Moreover, osteotomies were induced in femurs of CD-1 mice. The osteotomy gaps were filled with MVF-loaded TRH (TRH + MVF), unloaded TRH (TRH) or no material (control). Bone healing was evaluated 14 and 35 days postoperatively. MVF incorporated into TRH exhibited less apoptotic cells and showed a stable vessel morphology compared to controls. Micro-computed tomography revealed a reduced bone volume in TRH + MVF femurs. Histomorphometry showed less bone and more fibrous tissue after 35 days in TRH + MVF femurs compared to controls. Accordingly, TRH + MVF femurs exhibited a lower osseous bridging score and a reduced bending stiffness. Histology and Western blot analysis revealed an increased vascularization and CD31 expression, whereas vascular endothelial growth factor (VEGF) expression was reduced in TRH + MVF femurs. Furthermore, the callus of TRH + MVF femurs showed increased receptor activator of NF-κB ligand expression and higher numbers of osteoclasts. These findings indicate that TRH is an appropriate carrier system for MVF. Application of TRH + MVF increases the vascularization of bone defects. However, this impairs bone healing, most likely due to lower VEGF expression during the early course of bone healing. STATEMENT OF SIGNIFICANCE: In the present study we analyzed for the first time the in vivo performance of a thermoresponsive hydrogel (TRH) as a delivery system for bioactive microvascular fragments (MVF). We found that TRH represents an appropriate carrier for MVF as vascularization units and maintains their viability. Application of MVF-loaded TRH impaired bone formation in an established murine model of bone healing, although vascularization was improved. This unexpected outcome was most likely due to a reduced VEGF expression in the early phase bone healing.


Subject(s)
Adipose Tissue/cytology , Bone Regeneration , Hydrogels/chemistry , Microcirculation , Microvessels/growth & development , Animals , Bony Callus/pathology , Elasticity , Femur/pathology , Fracture Healing , Male , Mice , Neovascularization, Physiologic , Osteoclasts/metabolism , Osteotomy , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Shear Strength , Vascular Endothelial Growth Factor A/metabolism , Viscosity , X-Ray Microtomography
13.
Biomaterials ; 167: 15-31, 2018 06.
Article in English | MEDLINE | ID: mdl-29554478

ABSTRACT

The incidence of mesh-related infection after abdominal wall hernia repair is low, generally between 1 and 4%; however, worldwide, this corresponds to tens of thousands of difficult cases to treat annually. Adopting best practices in prevention is one of the keys to reduce the incidence of mesh-related infection. Once the infection is established, however, only a limited number of options are available that provides an efficient and successful treatment outcome. Over the past few years, there has been a tremendous amount of research dedicated to the functionalization of prosthetic meshes with antimicrobial properties, with some receiving regulatory approval and are currently available for clinical use. In this context, it is important to review the clinical importance of mesh infection, its risk factors, prophylaxis and pathogenicity. In addition, we give an overview of the main functionalization approaches that have been applied on meshes to confer anti-bacterial protection, the respective benefits and limitations, and finally some relevant future directions.


Subject(s)
Abdominal Wall/surgery , Anti-Infective Agents/therapeutic use , Biocompatible Materials/therapeutic use , Herniorrhaphy/adverse effects , Surgical Mesh/adverse effects , Surgical Wound Infection/etiology , Surgical Wound Infection/prevention & control , Animals , Anti-Infective Agents/administration & dosage , Antibiotic Prophylaxis/methods , Biocompatible Materials/administration & dosage , Herniorrhaphy/methods , Humans , Wound Healing/drug effects
14.
Eur Cell Mater ; 35: 151-164, 2018 03 02.
Article in English | MEDLINE | ID: mdl-29498410

ABSTRACT

Antibiotic-loaded biomaterials (ALBs) have emerged as a potential useful adjunctive antimicrobial measure for the prevention of infection in open fracture care. A biodegradable thermo-responsive poly(N-isopropylacrylamide) grafted hyaluronic acid (HApN) hydrogel loaded with gentamicin has recently been shown to prevent implant-related infection in a rabbit osteosynthesis model. The primary aim of this study was to determine the influence of this HApN hydrogel on bone healing at an early stage (4 weeks). A rabbit humeral osteotomy model with plating osteosynthesis was used to compare fracture healing in rabbits receiving the hydrogel as compared with control animals. The secondary aim was to observe fracture healing in groups treated with and without antibiotic-loaded hydrogel in the presence of bacterial contamination. In all groups, outcome measures were mechanical stability and histological score, with additional quantitative bacteriology in the inoculated groups. Application of the HApN hydrogel in non-inoculated rabbits did not significantly influence humeral stiffness or histological scores for fracture healing in comparison to controls. In the inoculated groups, animals receiving the bacterial inoculum without hydrogel were culture-positive at euthanasia and found to display lower humeral stiffness values and higher histopathological scores for bacterial presence in comparison with equivalents receiving the gentamicin-loaded HApN hydrogel, which were also infection-free. In summary, our data showed that HApN was an effective antibiotic carrier that did not affect fracture healing. This data supported its suitability for application in fracture care. Addition of osteopromotive compounds could provide further support for accelerating fracture healing in addition to successful infection prophylaxis.


Subject(s)
Bacterial Load/drug effects , Fracture Healing/drug effects , Gentamicins/pharmacology , Hydrogels/chemistry , Staphylococcus aureus/physiology , Temperature , Acrylic Resins/chemistry , Animals , Biomechanical Phenomena , Disease Models, Animal , Female , Humerus/diagnostic imaging , Humerus/drug effects , Humerus/pathology , Humerus/surgery , Hyaluronic Acid/chemistry , Rabbits , Staphylococcus aureus/drug effects
15.
J Control Release ; 269: 88-99, 2018 01 10.
Article in English | MEDLINE | ID: mdl-29127000

ABSTRACT

The systemic administration of drugs to treat bone diseases is often associated with poor uptake of the drug in the targeted tissue, potential systemic toxicity and suboptimal efficacy. In order to overcome these limitations, many micro- and nano-sized drug carriers have been developed for the treatment of bone pathologies that exhibit specific affinity for bone. Drug carriers can be functionalized with bone mineral seekers (BMS), creating a targeted drug delivery system (DDS) which is able to bind to bone and release therapeutics directly at the site of interest. This class of advanced DDS is of tremendous interest due to their strong affinity to bone, with great expectation to treat life-threatening bone disorders such as osteomyelitis, osteosarcoma or even osteoporosis. In this review, we first explain the mechanisms behind the affinity of several well-known BMS to bone, and then we present several effective approaches allowing the incorporation BMS into advanced DDS. Finally, we report the therapeutic applications of BMS based DDS under development or already established. Understanding the mechanisms behind the biological activity of recently developed BMS and their integration into advanced therapeutic delivery systems are essential prerequisites for further development of bone-targeting therapies with optimal efficacy.


Subject(s)
Bone and Bones/metabolism , Calcification, Physiologic , Drug Delivery Systems , Animals , Humans
16.
Acta Biomater ; 65: 1-20, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29128537

ABSTRACT

Articular cartilage is commonly described as a tissue that is made of up to 80% water, is devoid of blood vessels, nerves, and lymphatics, and is populated by only one cell type, the chondrocyte. At first glance, an easy tissue for clinicians to repair and for scientists to reproduce in a laboratory. Yet, chondral and osteochondral defects currently remain an open challenge in orthopedics and tissue engineering of the musculoskeletal system, without considering osteoarthritis. Why do we fail in repairing and regenerating articular cartilage? Behind its simple and homogenous appearance, articular cartilage hides a heterogeneous composition, a high level of organisation and specific biomechanical properties that, taken together, make articular cartilage a unique material that we are not yet able to repair or reproduce with high fidelity. This review highlights the available therapies for cartilage repair and retraces the research on different biomaterials developed for tissue engineering strategies. Their potential to recreate the structure, including composition and organisation, as well as the function of articular cartilage, intended as cell microenvironment and mechanically competent replacement, is described. A perspective of the limitations of the current research is given in the light of the emerging technologies supporting tissue engineering of articular cartilage. STATEMENT OF SIGNIFICANCE: The mechanical properties of articular tissue reflect its functionally organised composition and the recreation of its structure challenges the success of in vitro and in vivo reproduction of the native cartilage. Tissue engineering and biomaterials science have revolutionised the way scientists approach the challenge of articular cartilage repair and regeneration by introducing the concept of the interdisciplinary approach. The clinical translation of the current approaches are not yet fully successful, but promising results are expected from the emerging and developing new generation technologies.


Subject(s)
Biocompatible Materials , Cartilage, Articular/physiology , Tissue Engineering , Animals , Biomechanical Phenomena , Cartilage, Articular/growth & development , Humans , Regeneration
17.
Osteoarthritis Cartilage ; 26(2): 264-275, 2018 02.
Article in English | MEDLINE | ID: mdl-29169959

ABSTRACT

OBJECTIVE: The application of adjunctive mediators in Autologous chondrocyte implantation (ACI) techniques might be useful for improving the dedifferentiated chondrocyte phenotype, to support neocartilage formation and inhibit post-traumatic cartilage destruction. In this study we examined if (a) interleukin 10 treatment can cause chondrogenic phenotype stabilization and matrix preservation in mechanically injured cartilage and if (b) IL-10 can promote chondrogenesis in a clinically applied collagen scaffold for ACI treatment. MATERIALS AND METHODS: For (a) bovine articular cartilage was harvested, subjected to an axial unconfined injury and treated with bovine IL-10 (1-10,000 pg/ng/ml). For (b) a post-operatively remaining ACI graft was treated with human IL-10. Expression levels of type I/II/X collagen, SOX9 and aggrecan were measured by qPCR (a,b). After 3 weeks cell death was analyzed (nuclear blebbing and TUNEL assay) and matrix composition was determined by GAG measurements and immunohistochemistry (aggrecan, type I/II collagen, hyaluronic acid). STATISTICS: One way ANOVA analysis with Bonferroni's correction. RESULTS: (a) IL-10 stabilized the chondrogenic phenotype after injurious compression and preserved matrix integrity. This was indicated by elevated expression of chondrogenic markers COL2A1, ACAN, SOX9, while COL1A1 and COL10A1 were reduced. An increased GAG content paralleled this and histological staining of type 2 collagen, aggrecan and toluidine blue were enhanced after 3 weeks. (b) IL-10 [100 pg/ml] improved the chondrogenic differentiation of human chondrocytes, which was accompanied by cartilaginous matrix formation after 3 weeks of incubation. CONCLUSION: Interleukin-10 is a versatile adjuvant candidate to control the post-injurious environment in cartilage defects and promote chondrogenesis in ACI grafts.


Subject(s)
Cartilage, Articular/injuries , Chondrogenesis/drug effects , Interleukin-10/pharmacology , Animals , Apoptosis/drug effects , Cartilage, Articular/drug effects , Cartilage, Articular/metabolism , Cartilage, Articular/pathology , Cattle , Cells, Cultured , Chondrocytes/drug effects , Chondrocytes/pathology , Chondrocytes/transplantation , Collagen/metabolism , Extracellular Matrix/metabolism , Glycosaminoglycans/metabolism , Humans , Tissue Scaffolds
18.
Acta Biomater ; 54: 386-398, 2017 05.
Article in English | MEDLINE | ID: mdl-28286037

ABSTRACT

Fabrication of composite scaffolds using stereolithography (SLA) for bone tissue engineering has shown great promises. However, in order to trigger effective bone formation and implant integration, exogenous growth factors are commonly combined to scaffold materials. In this study, we fabricated biodegradable composite scaffolds using SLA and endowed them with osteopromotive properties in the absence of biologics. First we prepared photo-crosslinkable poly(trimethylene carbonate) (PTMC) resins containing 20 and 40wt% of hydroxyapatite (HA) nanoparticles and fabricated scaffolds with controlled macro-architecture. Then, we conducted experiments to investigate how the incorporation of HA in photo-crosslinked PTMC matrices improved human bone marrow stem cells osteogenic differentiation in vitro and kinetic of bone healing in vivo. We observed that bone regeneration was significantly improved using composite scaffolds containing as low as 20wt% of HA, along with difference in terms of osteogenesis and degree of implant osseointegration. Further investigations revealed that SLA process was responsible for the formation of a rich microscale layer of HA corralling scaffolds. To summarize, this work is of substantial importance as it shows how the fabrication of hierarchical biomaterials via surface-enrichment of functional HA nanoparticles in composite polymer stereolithographic structures could impact in vitro and in vivo osteogenesis. STATEMENT OF SIGNIFICANCE: This study reports for the first time the enhance osteopromotion of composite biomaterials, with controlled macro-architecture and microscale distribution of hydroxyapatite particles, manufactured by stereolithography. In this process, the hydroxyapatite particles are not only embedded into an erodible polymer matrix, as reported so far in the literature, but concentrated at the surface of the structures. This leads to robust in vivo bone formation at low concentration of hydroxyapatite. The reported 3D self-corralling composite architecture provides significant opportunities to develop functional biomaterials for bone repair and tissue engineering.


Subject(s)
Bone Marrow Cells/pathology , Bone Regeneration/drug effects , Durapatite , Mesenchymal Stem Cells/metabolism , Nanoparticles/chemistry , Osteogenesis/drug effects , Skull , Tissue Scaffolds/chemistry , Animals , Bone Marrow Cells/metabolism , Durapatite/chemistry , Durapatite/pharmacology , Female , Humans , Mesenchymal Stem Cells/pathology , Rabbits , Skull/injuries , Skull/metabolism , Skull/pathology
19.
J Mater Sci Mater Med ; 27(9): 144, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27552808

ABSTRACT

The European Society for Biomaterials 2015 Translational Research Symposium focused on 'Innovating in the Medical Device Industry - Challenges & Opportunities' from different perspectives, i.e., from a non-profit research organisation to a syndicate of small and medium-sized companies and large companies. Lecturers from regulatory consultants, industry and research institutions described the innovation process and regulatory processes (e.g., 510K, PMA, combination product) towards market approval. The aim of the present article is to summarise and explain the main statements made during the symposium, in terms of challenges and opportunities for medical device industries, in a constantly changing customer and regulatory environment.


Subject(s)
Equipment and Supplies , Translational Research, Biomedical/methods , Translational Research, Biomedical/trends , Animals , Biocompatible Materials , Clinical Trials as Topic , Congresses as Topic , Diffusion of Innovation , Europe , Humans , Societies, Medical
20.
Eur Cell Mater ; 32: 74-86, 2016 07 08.
Article in English | MEDLINE | ID: mdl-27386841

ABSTRACT

The seeding of scaffolds with adipose tissue-derived microvascular fragments represents a promising strategy to establish a sufficient blood supply in tissue constructs. Herein, we analysed whether a single application of macrophage-activating lipopeptide-2 (MALP-2) at the implantation site further improves the early vascularisation of such microvessel-seeded constructs. Microvascular fragments were isolated from epididymal fat pads of C57BL/6 mice. The fragments were seeded on polyurethane scaffolds, which were implanted into mouse dorsal skinfold chambers exposed to MALP-2 or vehicle (control). The inflammatory host tissue response and the vascularisation of the scaffolds were analysed using intravital fluorescence microscopy, histology and immunohistochemistry. We found that the numbers of microvascular adherent leukocytes were significantly increased in MALP-2-treated chambers during the first 3 days after scaffold implantation when compared to controls. This temporary inflammation resulted in an improved vascularisation of the host tissue surrounding the implants, as indicated by a higher density of CD31-positive microvessels at day 14. However, the MALP-2-exposed scaffolds themselves presented with a lower functional microvessel density in their centre. In addition, in vitro analyses revealed that MALP-2 promotes apoptotic cell death of endothelial and perivascular cells in isolated microvascular fragments. Hence, despite the beneficial pro-angiogenic properties of MALP-2 at the implantation site, the herein evaluated approach may not be recommended to improve the vascularisation capacity of microvascular fragments in tissue engineering applications.


Subject(s)
Lipopeptides/pharmacology , Microvessels/physiology , Neovascularization, Physiologic/drug effects , Polyurethanes/pharmacology , Tissue Scaffolds/chemistry , Animals , Caspase 3/metabolism , Cell Survival/drug effects , Hemodynamics/drug effects , Immunohistochemistry , Implants, Experimental , Inflammation/pathology , Male , Mice, Inbred C57BL , Microscopy, Fluorescence , Microvessels/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL