Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 78
Filter
1.
Osteoarthritis Cartilage ; 31(4): 534-542, 2023 04.
Article in English | MEDLINE | ID: mdl-36623637

ABSTRACT

OBJECTIVE: To examine the association between 3D patellar shape and 1) isolated magnetic resonance imaging (MRI)-based patellofemoral osteoarthritis (PFOA), 2) the morphological features of PFOA, and 3) the clinical symptoms of PFOA. DESIGN: MRI data from 66 women with isolated MRI-based PFOA and 66 age- and BMI-matched healthy women were selected from a cohort study. The patellae were manually segmented from MRI scans and used to create a 3D statistical shape model (SSM) of the patella. Structural abnormalities were semi-standardized scored on MRI using MRI osteoarthritis knee score (MOAKS). Regression analyses were applied to determine the associations between the shape parameters retrieved from the SSM, group status, clinical symptoms, and structural abnormalities. RESULTS: Four shape variants showed a statistically significant (<0.05) association with the group status. The mode responsible for most of the shape variations showed participants with PFOA possess a relatively thicker dorsal bump on the articular part of the patella, compared to patellae of control participants. Three of these variants showed an association with the presence of osteophytes and cartilage loss on the patella. Multiple associations were found between patellar shape and the clinical symptoms of PFOA. CONCLUSIONS: Patellar shape is associated with the prevalence of MRI-based PFOA in women. Some shape variants were also associated with clinical symptoms. Interestingly, one particular shape variant associated with the presence of MRI-based PFOA was earlier shown to be associated with structural abnormalities associated with OA in a population aged under 40. This may suggest that patellar shape may be an early detectable risk factor for PFOA.


Subject(s)
Osteoarthritis, Knee , Patellofemoral Joint , Humans , Female , Aged , Patella/diagnostic imaging , Patella/pathology , Cohort Studies , Patellofemoral Joint/diagnostic imaging , Patellofemoral Joint/pathology , Radiography , Osteoarthritis, Knee/diagnostic imaging , Osteoarthritis, Knee/pathology , Magnetic Resonance Imaging/methods
2.
Nanotechnology ; 29(43): 43LT02, 2018 Oct 26.
Article in English | MEDLINE | ID: mdl-30152409

ABSTRACT

Nanopillar arrays that are bactericidal but not cytotoxic against the host cells could be used in implantable medical devices to prevent implant-associated infections. It is, however, unclear what heights, widths, interspacing, and shape should be used for the nanopillars to achieve the desired antibacterial effects while not hampering the integration of the device in the body. Here, we present an in-silico approach based on finite element modeling of the interactions between Staphylococcus aureus and nanopatterns on the one hand and osteoblasts and nanopatterns on the other hand to find the best design parameters. We found that while the height of the nanopillars seems to have little impact on the bactericidal behavior, shorter widths and larger interspacings substantially increase the bactericidal effects. The same combination of parameters could, however, also cause cytotoxicity. Our results suggest that a specific combination of height (120 nm), width (50 nm), and interspacing (300 nm) offers the bactericidal effects without cytotoxicity.


Subject(s)
Anti-Bacterial Agents/chemistry , Computer Simulation , Models, Biological , Nanostructures/chemistry , Nanostructures/ultrastructure , Osteoblasts/cytology , Prostheses and Implants/microbiology , Anti-Bacterial Agents/toxicity , Cell Survival , Computer-Aided Design , Finite Element Analysis , Humans , Microbial Viability , Nanostructures/toxicity , Staphylococcal Infections/prevention & control , Staphylococcus aureus/physiology , Surface Properties
3.
Osteoarthritis Cartilage ; 24(12): 2108-2115, 2016 12.
Article in English | MEDLINE | ID: mdl-27495945

ABSTRACT

OBJECTIVE: The etiology of osteochondral defects (OCDs), for which the ankle (talocrural) joint is one of the common sites, is not yet fully understood. In this study, we hypothesized that bone shape plays a role in development of OCDs. Therefore, we quantitatively compared the morphology of the talus and the distal tibia between an OCD group and a control group. METHODS: The shape variations of the talus and distal tibia were described separately by constructing two statistical shape models (SSMs) based on the segmentation of the bones from ankle computed tomography (CT) scans obtained from control (i.e., 35 CT scans) and OCD (i.e., 37 CT scans) groups. The first five modes of shape variation for the SSM corresponding to each bone were statistically compared between control and OCD groups using an analysis of variance (ANOVA) corrected with the Bonferroni for multiple comparisons. RESULTS: The first five modes of variation in the SSMs respectively represented 49% and 40% of the total variance of talus and tibia. Less than 5% of the variance per mode was described by the higher modes. Mode 5 of the talus (P = 0.004) primarily describing changes in the vertical neck angle and Mode 1 of the tibia (P < 0.0001) representing variations at the medial malleolus, showed statistically significant difference between the control and OCD groups. CONCLUSION: Shape differences exist between control and OCD groups. This indicates that a geometry modulated biomechanical behavior of the talocrural joint may be a risk factor for OCD.


Subject(s)
Intra-Articular Fractures , Ankle Joint , Humans , Talus , Tibia , Tomography, X-Ray Computed
4.
Eur Cell Mater ; 29: 141-53; discussion 153-4, 2015 Mar 04.
Article in English | MEDLINE | ID: mdl-25738583

ABSTRACT

Regeneration of load-bearing segmental bone defects is a major challenge in trauma and orthopaedic surgery. The ideal bone graft substitute is a biomaterial that provides immediate mechanical stability, while stimulating bone regeneration to completely bridge defects over a short period. Therefore, selective laser melted porous titanium, designed and fine-tuned to tolerate full load-bearing, was filled with a physiologically concentrated fibrin gel loaded with bone morphogenetic protein-2 (BMP-2). This biomaterial was used to graft critical-sized segmental femoral bone defects in rats. As a control, porous titanium implants were either left empty or filled with a fibrin gels without BMP-2. We evaluated bone regeneration, bone quality and mechanical strength of grafted femora using in vivo and ex vivo µCT scanning, histology, and torsion testing. This biomaterial completely regenerated and bridged the critical-sized bone defects within eight weeks. After twelve weeks, femora were anatomically re-shaped and revealed open medullary cavities. More importantly, new bone was formed throughout the entire porous titanium implants and grafted femora regained more than their innate mechanical stability: torsional strength exceeded twice their original strength. In conclusion, combining porous titanium implants with a physiologically concentrated fibrin gels loaded with BMP-2 improved bone regeneration in load-bearing segmental defects. This material combination now awaits its evaluation in larger animal models to show its suitability for grafting load-bearing defects in trauma and orthopaedic surgery.


Subject(s)
Bone Morphogenetic Protein 2/pharmacology , Fibrin/pharmacology , Fractures, Bone/therapy , Prostheses and Implants , Titanium , Animals , Biomechanical Phenomena , Bone Regeneration , Bone Substitutes/pharmacology , Femur/drug effects , Femur/injuries , Femur/surgery , Fractures, Bone/diagnostic imaging , Fractures, Bone/physiopathology , Gels , Male , Microscopy, Electron, Scanning , Porosity , Rats, Wistar , Weight-Bearing , X-Ray Microtomography
5.
J Biomech Eng ; 137(7)2015 Jul.
Article in English | MEDLINE | ID: mdl-25790039

ABSTRACT

Transport of solutes through diffusion is an important metabolic mechanism for the avascular cartilage tissue. Three types of interconnected physical phenomena, namely mechanical, electrical, and chemical, are all involved in the physics of transport in cartilage. In this study, we use a carefully designed experimental-computational setup to separate the effects of mechanical and chemical factors from those of electrical charges. Axial diffusion of a neutral solute Iodixanol into cartilage was monitored using calibrated microcomputed tomography micro-CT images for up to 48 hr. A biphasic-solute computational model was fitted to the experimental data to determine the diffusion coefficients of cartilage. Cartilage was modeled either using one single diffusion coefficient (single-zone model) or using three diffusion coefficients corresponding to superficial, middle, and deep cartilage zones (multizone model). It was observed that the single-zone model cannot capture the entire concentration-time curve and under-predicts the near-equilibrium concentration values, whereas the multizone model could very well match the experimental data. The diffusion coefficient of the superficial zone was found to be at least one order of magnitude larger than that of the middle zone. Since neutral solutes were used, glycosaminoglycan (GAG) content cannot be the primary reason behind such large differences between the diffusion coefficients of the different cartilage zones. It is therefore concluded that other features of the different cartilage zones such as water content and the organization (orientation) of collagen fibers may be enough to cause large differences in diffusion coefficients through the cartilage thickness.


Subject(s)
Cartilage, Articular/metabolism , Models, Biological , Biological Transport , Cartilage, Articular/diagnostic imaging , Diffusion , Hydrogen-Ion Concentration , Image Processing, Computer-Assisted , X-Ray Microtomography
6.
Osteoarthritis Cartilage ; 22(12): 2074-82, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25241242

ABSTRACT

OBJECTIVE: A cam-type deformity drastically increases the risk of hip osteoarthritis (OA). Since this type of skeletal anomaly is more prevalent among young active adults, it is hypothesized that the loading conditions experienced during certain types of vigorous physical activities stimulates formation of cam-type deformity. We further hypothesize that the growth plate shape modulates the influence of mechanical factors on the development of cam-type deformity. DESIGN: We used finite element (FE) models of the proximal femur with an open growth plate to study whether mechanical factors could explain the development of cam-type deformity in adolescents. Four different loading conditions (representing different types of physical activities) and three different levels of growth plate extension towards the femoral neck were considered. Mechanical stimuli at the tissue level were calculated by means of the osteogenic index (OI) for all loading conditions and growth plate shape variations. RESULTS: Loading conditions and growth plate shape influence the distribution of OI in hips with an open growth plate, thereby driving the development of cam-type deformity. In particular, specific types of loads experienced during physical activities and a larger growth plate extension towards the femoral neck increase the chance of cam-type deformity. CONCLUSIONS: Specific loading patterns seem to stimulate the development of cam-type deformity by modifying the distribution of the mechanical stimulus. This is in line with recent clinical studies and reveals mechanobiological mechanisms that trigger the development of cam-type deformity. Avoiding these loading patterns during skeletal growth might be a potential preventative strategy for future hip OA.


Subject(s)
Femoracetabular Impingement/complications , Femur Neck/abnormalities , Biomechanical Phenomena , Child , Growth Plate/abnormalities , Humans , Male , Weight-Bearing
7.
Nat Commun ; 14(1): 7919, 2023 Dec 12.
Article in English | MEDLINE | ID: mdl-38086804

ABSTRACT

Durable interfacing of hard and soft materials is a major design challenge caused by the ensuing stress concentrations. In nature, soft-hard interfaces exhibit remarkable mechanical performance, with failures rarely happening at the interface. Here, we mimic the strategies observed in nature to design efficient soft-hard interfaces. We base our geometrical designs on triply periodic minimal surfaces (i.e., Octo, Diamond, and Gyroid), collagen-like triple helices, and randomly distributed particles. A combination of computational simulations and experimental techniques, including uniaxial tensile and quad-lap shear tests, are used to characterize the mechanical performance of the interfaces. Our analyses suggest that smooth interdigitated connections, compliant gradient transitions, and either decreasing or constraining strain concentrations lead to simultaneously strong and tough interfaces. We generate additional interfaces where the abovementioned toughening mechanisms work synergistically to create soft-hard interfaces with strengths approaching the upper achievable limit and enhancing toughness values by 50%, as compared to the control group.

8.
J Mech Behav Biomed Mater ; 148: 106173, 2023 12.
Article in English | MEDLINE | ID: mdl-37866280

ABSTRACT

Orthopedic plates are commonly used after osteotomies for temporary fixation of bones. Patient-specific plates have recently emerged as a promising fixation device. However, it is unclear how various strategies used for the design of such plates perform in comparison with each other. Here, we compare the biomechanical performance of 3D printed patient-specific bone plates designed using conventional computer-aided design (CAD) techniques with those designed with the help of topology optimization (TO) algorithms, focusing on cases involving slipped capital femoral epiphysis (SCFE). We established a biomechanical testing protocol to experimentally assess the performance of the designed plates while measuring the full-field strain using digital image correlation. We also created an experimentally validated finite element model to analyze the performance of the plates under physiologically relevant loading conditions. The results indicated that the TO construct exhibited higher ultimate load and biomechanical performance as compared to the CAD construct, suggesting that TO is a viable approach for the design of such patient-specific bone plates. The TO plate also distributed stress more evenly over the screws, likely resulting in more durable constructs and improved anatomical conformity while reducing the risk of screw and plate failure during cyclic loading. Although differences existed between finite element analysis and experimental testing, this study demonstrated that finite element modelling can be used as a reliable method for evaluating and optimizing plates for SCFE patients. In addition to enhancing the mechanical performance of patient-specific fixation plates, the utilization of TO in plate design may also improve the surgical outcome and decrease the recovery time by reducing the plate and incision sizes.


Subject(s)
Slipped Capital Femoral Epiphyses , Humans , Slipped Capital Femoral Epiphyses/surgery , Bone Plates , Osteotomy , Bone Screws , Printing, Three-Dimensional , Biomechanical Phenomena , Finite Element Analysis , Fracture Fixation, Internal
9.
Acta Biomater ; 169: 589-604, 2023 10 01.
Article in English | MEDLINE | ID: mdl-37536493

ABSTRACT

Additively manufactured (AM) degradable porous metallic biomaterials offer unique opportunities for satisfying the design requirements of an ideal bone substitute. Among the currently available biodegradable metals, iron has the highest elastic modulus, meaning that it would benefit the most from porous design. Given the successful preclinical applications of such biomaterials for the treatment of cardiovascular diseases, the moderate compatibility of AM porous iron with osteoblast-like cells, reported in earlier studies, has been surprising. This may be because, as opposed to static in vitro conditions, the biodegradation products of iron in vivo are transported away and excreted. To better mimic the in situ situations of biodegradable biomaterials after implantation, we compared the biodegradation behavior and cytocompatibility of AM porous iron under static conditions to the conditions with dynamic in situ-like fluid flow perfusion in a bioreactor. Furthermore, the compatibility of these scaffolds with four different cell types was evaluated to better understand the implications of these implants for the complex process of natural wound healing. These included endothelial cells, L929 fibroblasts, RAW264.7 macrophage-like cells, and osteoblastic MG-63 cells. The biodegradation rate of the scaffolds was significantly increased in the perfusion bioreactor as compared to static immersion. Under either condition, the compatibility with L929 cells was the best. Moreover, the compatibility with all the cell types was much enhanced under physiomimetic dynamic flow conditions as compared to static biodegradation. Our study highlights the importance of physiomimetic culture conditions and cell type selection when evaluating the cytocompatibility of degradable biomaterials in vitro. STATEMENT OF SIGNIFICANCE: Additively manufactured (AM) degradable porous metals offer unique opportunities for the treatment of large bony defects. Despite the successful preclinical applications of biodegradable iron in the cardiovascular field, the moderate compatibility of AM porous iron with osteoblast-like cells was reported. To better mimic the in vivo condition, we compared the biodegradation behavior and cytocompatibility of AM porous iron under static condition to dynamic perfusion. Furthermore, the compatibility of these scaffolds with various cell types was evaluated to better simulate the process of natural wound healing. Our study suggests that AM porous iron holds great promise for orthopedic applications, while also highlighting the importance of physio-mimetic culture conditions and cell type selection when evaluating the cytocompatibility of degradable biomaterials in vitro.


Subject(s)
Endothelial Cells , Iron , Iron/pharmacology , Porosity , Biocompatible Materials/pharmacology , Metals
10.
3D Print Med ; 9(1): 16, 2023 Jun 09.
Article in English | MEDLINE | ID: mdl-37294496

ABSTRACT

Custom-made triflange acetabular implants are increasingly used in complex revision surgery where supporting bone stock is diminished. In most cases these triflange cups induce stress-shielding. A new concept for the triflange is introduced that uses deformable porous titanium to redirect forces from the acetabular rim to the bone stock behind the implant and thereby reduces further stress-shielding. This concept is tested for deformability and primary stability.Three different designs of highly porous titanium cylinders were tested under compression to determine their mechanical properties. The most promising design was used to design five acetabular implants either by incorporating a deformable layer at the back of the implant or by adding a separate generic deformable mesh behind the implant. All implants were inserted into sawbones with acetabular defects followed by a cyclic compression test of 1800N for 1000 cycles.The design with a cell size of 4 mm and 0.2 mm strut thickness performed the best and was applied for the design of the acetabular implants. An immediate primary fixation was realized in all three implants with an incorporated deformable layer. One of the two implants with a separate deformable mesh needed fixation with screws. Cyclic tests revealed an average additional implant subsidence of 0.25 mm that occurred in the first 1000 cycles with minimal further subsidence thereafter.It is possible to realize primary implant fixation and stability in simulated large acetabular revision surgery using a deformable titanium layer behind the cup. Additional research is needed for further implementation of such implants in the clinic.

11.
J Mech Behav Biomed Mater ; 146: 106097, 2023 10.
Article in English | MEDLINE | ID: mdl-37678107

ABSTRACT

OBJECTIVE: Mandibular reconstruction using patient-specific cage implants is a promising alternative to the vascularized free flap reconstruction for nonirradiated patients with adequate soft tissues, or for patients whose clinical condition is not conducive to microsurgical reconstruction. This study aimed to assess the biomechanical performance of 3D printed patient-specific cage implants designed with a semi-automated workflow in a combined cadaveric and retrospective case series study. METHODS: We designed cage implants for two human cadaveric mandibles using our previously developed design workflow. The biomechanical performance of the implants was assessed with the finite element analysis (FEA) and quasi-static biomechanical testing. Digital image correlation (DIC) was used to measure the full-field strains and validate the FE models by comparing the distribution of maximum principal strains within the bone. The retrospective study of a case series involved three patients, each of whom was treated with a cage implant of similar design. The biomechanical performance of these implants was evaluated using the experimentally validated FEA under the scenarios of both mandibular union and nonunion. RESULTS: No implant or screw failure was observed prior to contralateral bone fracture during the quasi-static testing of both cadaveric mandibles. The FEA and DIC strain contour plots indicated a strong linear correlation (r = 0.92) and a low standard error (SE=29.32µÎµ), with computational models yielding higher strain values by a factor of 2.7. The overall stresses acting on the case series' implants stayed well below the yield strength of additively manufactured (AM) commercially pure titanium, when simulated under highly strenuous chewing conditions. Simulating a full union between the graft and remnant mandible yielded a substantial reduction (72.7±1.5%) in local peak stresses within the implants as compared to a non-bonded graft. CONCLUSIONS: This study shows the suitability of the developed semi-automated workflow in designing patient-specific cage implants with satisfactory mechanical functioning under demanding chewing conditions. The proposed workflow can aid clinical engineers in creating reconstruction systems and streamlining pre-surgical planning. Nevertheless, more research is still needed to evaluate the osteogenic potential of bone graft insertions.


Subject(s)
Bone Screws , Mandible , Humans , Retrospective Studies , Workflow , Mandible/surgery , Cadaver
12.
Acta Biomater ; 162: 182-198, 2023 05.
Article in English | MEDLINE | ID: mdl-36972809

ABSTRACT

The development of biodegradable Fe-based bone implants has rapidly progressed in recent years. Most of the challenges encountered in developing such implants have been tackled individually or in combination using additive manufacturing technologies. Yet not all the challenges have been overcome. Herein, we present porous FeMn-akermanite composite scaffolds fabricated by extrusion-based 3D printing to address the unmet clinical needs associated with Fe-based biomaterials for bone regeneration, including low biodegradation rate, MRI-incompatibility, mechanical properties, and limited bioactivity. In this research, we developed inks containing Fe, 35 wt% Mn, and 20 or 30 vol% akermanite powder mixtures. 3D printing was optimized together with the debinding and sintering steps to obtain scaffolds with interconnected porosity of 69%. The Fe-matrix in the composites contained the γ-FeMn phase as well as nesosilicate phases. The former made the composites paramagnetic and, thus, MRI-friendly. The in vitro biodegradation rates of the composites with 20 and 30 vol% akermanite were respectively 0.24 and 0.27 mm/y, falling within the ideal range of biodegradation rates for bone substitution. The yield strengths of the porous composites stayed within the range of the values of the trabecular bone, despite in vitro biodegradation for 28 d. All the composite scaffolds favored the adhesion, proliferation, and osteogenic differentiation of preosteoblasts, as revealed by Runx2 assay. Moreover, osteopontin was detected in the extracellular matrix of cells on the scaffolds. Altogether, these results demonstrate the remarkable potential of these composites in fulfilling the requirements of porous biodegradable bone substitutes, motivating future in vivo research. STATEMENT OF SIGNIFICANCE: We developed FeMn-akermanite composite scaffolds by taking advantage of the multi-material capacity of extrusion-based 3D printing. Our results demonstrated that the FeMn-akermanite scaffolds showed an exceptional performance in fulfilling all the requirements for bone substitution in vitro, i.e., a sufficient biodegradation rate, having mechanical properties in the range of trabecular bone even after 4 weeks biodegradation, paramagnetic, cytocompatible and most importantly osteogenic. Our results encourage further research on Fe-based bone implants in in vivo.


Subject(s)
Bone Substitutes , Bone Substitutes/pharmacology , Porosity , Osteogenesis , Printing, Three-Dimensional , Tissue Scaffolds/chemistry
13.
Front Bioeng Biotechnol ; 11: 1289299, 2023.
Article in English | MEDLINE | ID: mdl-38356932

ABSTRACT

The currently available treatments for inner ear disorders often involve systemic drug administration, leading to suboptimal drug concentrations and side effects. Cochlear implants offer a potential solution by providing localized and sustained drug delivery to the cochlea. While the mechanical characterization of both the implants and their constituent material is crucial to ensure functional performance and structural integrity during implantation, this aspect has been mostly overlooked. This study proposes a novel methodology for the mechanical characterization of our recently developed cochlear implant design, namely, rectangular and cylindrical, fabricated using two-photon polymerization (2 PP) with a novel photosensitive resin (IP-Q™). We used in silico computational models and ex silico experiments to study the mechanics of our newly designed implants when subjected to torsion mimicking the foreseeable implantation procedure. Torsion testing on the actual-sized implants was not feasible due to their small size (0.6 × 0.6 × 2.4 mm³). Therefore, scaled-up rectangular cochlear implants (5 × 5 × 20 mm³, 10 × 10 × 40 mm³, and 20 × 20 × 80 mm³) were fabricated using stereolithography and subjected to torsion testing. Finite element analysis (FEA) accurately represented the linear behavior observed in the torsion experiments. We then used the validated Finite element analysis models to study the mechanical behavior of real-sized implants fabricated from the IP-Q resin. Mechanical characterization of both implant designs, with different inner porous structures (pore size: 20 µm and 60 µm) and a hollow version, revealed that the cylindrical implants exhibited approximately three times higher stiffness and mechanical strength as compared to the rectangular ones. The influence of the pore sizes on the mechanical behavior of these implant designs was found to be small. Based on these findings, the cylindrical design, regardless of the pore size, is recommended for further research and development efforts.

14.
Biomater Adv ; 139: 212993, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35882142

ABSTRACT

Modulation of the immune response following the implantation of biomaterials can have beneficial effects on bone regeneration. This involves complex interactions between the inflammatory and osteogenic cells. Therefore, the study of cell-cell interactions using direct co-culture models integrated with biomaterials is of great interest. This research aimed to study the viability, morphology, and osteogenic activity of preosteoblasts (OBs) co-cultured with pro-inflammatory macrophages (M1s) on the 3D printed (non)patterned surfaces. OBs and M1s remained alive and proliferated actively for 14 days in the mixture of Dulbecco's Modified Eagle's Medium (DMEM) and alpha Minimum Essential Medium (α-MEM) (1:1), regardless of the cell ratio in the co-cultures. The spatial organization of the two types of cells changed with the time of culture from an initially uniform cell distribution to the formation of a thick layer of OBs covered by clusters of M1s. On day 7, the expression of PGE2 and TNF-α were upregulated in the co-culture relative to the mono-culture of OBs and M1s. The inflammation decreased differentiation and matrix mineralization of OBs after 28 days of culture. Interestingly, the incorporation of 3D printed submicron pillars into the direct co-culture model enhanced the differentiation of preosteoblasts, as shown by relatively higher RUNX2 expression, thereby revealing the osteoimmunomodulatory potential of such surface patterns.


Subject(s)
Biocompatible Materials , Osteogenesis , Cell Differentiation , Coculture Techniques , Printing, Three-Dimensional
15.
Acta Biomater ; 138: 398-409, 2022 01 15.
Article in English | MEDLINE | ID: mdl-34763109

ABSTRACT

The fatigue performance of additively manufactured auxetic meta-biomaterials made from commercially pure titanium has been studied only recently. While certain assumptions have been made regarding the mechanisms underlying their fatigue failure, the exact mechanisms are not researched yet. Here, we studied the mechanisms of crack formation and propagation in cyclically loaded auxetic meta-biomaterials. Twelve different designs were subjected to compression-compression fatigue testing while performing full-field strain measurement using digital image correlation (DIC). The fatigue tests were stopped at different points before complete specimen failure to study the evolution of damage in the micro-architecture of the specimens using micro-computed tomography (micro-CT). Furthermore, finite element models were made to study the presence of stress concentrations. Structural weak spots were found in the inverted nodes and the vertical struts located along the outer rim of the specimens, matching the maximum principal strain concentrations and fracture sites in the DIC and micro-CT data. Cracks were often found to originate from internal void spaces or from sites susceptible to mode-I cracking. Many specimens maintained their structural integrity and exhibited no signs of rapid strain accumulation despite the presence of substantial crack growth. This observation underlines the importance of such microscale studies to identify accumulated damage that otherwise goes unnoticed. The potential release of powder particles from damaged lattices could elicit a foreign body response, adversely affecting the implant success. Finding the right failure criterion, therefore, requires more data than only those pertaining to macroscopic measurements and should always include damage assessment at the microscale. STATEMENT OF SIGNIFICANCE: The negative Poisson's ratio of auxetic meta-biomaterials makes them expand laterally in response to axial tension. This extraordinary property has great potential in the field of orthopedics, where it could enhance bone-implant contact. The fatigue performance of additively manufactured auxetic meta-biomaterials has only recently been studied and was found to be superior to many other bending- and stretch-dominated micro-architectures. In this study, we go beyond these macroscopic measurements and focus on the crack initiation and propagation. Full-field strain measurements and 3D imaging are used to paint a detailed picture of the mechanisms underlying fatigue. Using these data, specific aspects of the design and/or printing process can be targeted to improve the performance of auxetic meta-biomaterials in load-bearing applications.


Subject(s)
Biocompatible Materials , Titanium , Materials Testing , Stress, Mechanical , X-Ray Microtomography
16.
Biomater Adv ; 133: 112617, 2022 Feb.
Article in English | MEDLINE | ID: mdl-35525731

ABSTRACT

Additively manufacturing of porous iron offers a unique opportunity to increase its biodegradation rate by taking advantage of arbitrarily complex porous structures. Nevertheless, achieving the required biodegradation profile remains challenging due to the natural passivation of iron that decrease the biodegradation rate. Moreover, the biocompatibility of iron is reported to be limited. Here, we address both challenges by applying poly(2-ethyl-2-oxazoline) coating to extrusion-based 3D printed porous iron. We characterized the specimens by performing in vitro biodegradation, electrochemical measurements, time-dependent mechanical tests, and in vitro cytocompatibility assays. The coated porous iron exhibited a biodegradation rate that was 2.6× higher than that of non-coated counterpart and maintained the bone-mimicking mechanical properties throughout biodegradation. Despite the formation of dense biodegradation products, the coating ensured a relatively stable biodegradation (i.e., 17% reduction in the degradation rate between days 14 and 28) as compared to that of non-coated specimens (i.e., 43% drop). Furthermore, the coating could be identified even after biodegradation, demonstrating the longevity of the coating. Finally, the coated specimens significantly increased the viability and supported the attachment and growth of preosteoblasts. Our results demonstrate the great potential of poly(2-ethyl-2-oxazoline) coating for addressing the multiple challenges associated with the clinical adoption of porous iron.


Subject(s)
Iron , Polyamines , Iron/pharmacology , Porosity
17.
Acta Biomater ; 148: 355-373, 2022 08.
Article in English | MEDLINE | ID: mdl-35690326

ABSTRACT

Advanced additive manufacturing techniques have been recently used to tackle the two fundamental challenges of biodegradable Fe-based bone-substituting materials, namely low rate of biodegradation and insufficient bioactivity. While additively manufactured porous iron has been somewhat successful in addressing the first challenge, the limited bioactivity of these biomaterials hinder their progress towards clinical application. Herein, we used extrusion-based 3D printing for additive manufacturing of iron-matrix composites containing silicate-based bioceramic particles (akermanite), thereby addressing both of the abovementioned challenges. We developed inks that carried iron and 5, 10, 15, or 20 vol% of akermanite powder mixtures for the 3D printing process and optimized the debinding and sintering steps to produce geometrically-ordered iron-akermanite composites with an open porosity of 69-71%. The composite scaffolds preserved the designed geometry and the original α-Fe and akermanite phases. The in vitro biodegradation rates of the composites were improved as much as 2.6 times the biodegradation rate of geometrically identical pure iron. The yield strengths and elastic moduli of the scaffolds remained within the range of the mechanical properties of the cancellous bone, even after 28 days of biodegradation. The composite scaffolds (10-20 vol% akermanite) demonstrated improved MC3T3-E1 cell adhesion and higher levels of cell proliferation. The cellular secretion of collagen type-1 and the alkaline phosphatase activity on the composite scaffolds (10-20 vol% akermanite) were, respectively higher than and comparable to Ti6Al4V in osteogenic medium. Taken together, these results clearly show the potential of 3D printed porous iron-akermanite composites for further development as promising bone substitutes. STATEMENT OF SIGNIFICANCE: Porous iron matrix composites containing akermanite particles were produced by means of multi-material additive manufacturing to address the two fundamental challenges associated with biodegradable iron-based biomaterials, namely very low rate of biodegradation and insufficient bioactivity. Our porous iron-akermanite composites exhibited enhanced biodegradability and superior bioactivity compared to porous monolithic iron scaffolds. The murine bone cells proliferated on the composite scaffolds, and secreted the collagen type-1 matrix that stimulated bony-like mineralization. The results show the exceptional potential of the developed porous iron-based composite scaffolds for application as bone substitutes.


Subject(s)
Bone Substitutes , Animals , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Bone Regeneration , Ceramics , Collagen , Iron/chemistry , Iron/pharmacology , Mice , Porosity , Printing, Three-Dimensional , Tissue Scaffolds/chemistry
18.
Acta Biomater ; 151: 628-646, 2022 10 01.
Article in English | MEDLINE | ID: mdl-35940565

ABSTRACT

The treatment of femoral nonunion with large segmental bone defect is still challenging. Although magnesium alloys have been considered potential materials for such a treatment, their application is limited by their fast degradation. Adding bioceramic particles into magnesium to form Mg-matrix composites is a promising strategy to adjust their biodegradation rates and to improve their mechanical properties and cytocompatibility further. Here, we developed an extrusion-based additive manufacturing technique to fabricate biodegradable Mg-Zn/bioceramic composite scaffolds ex-situ. Inks carrying a Mg-Zn powder and 5, 10 and 15% ß-tricalcium phosphate (TCP) powder particles were investigated regarding the dispersion of ß-TCP particles in the inks and viscoelastic properties. Optimally formulated inks were then employed for subsequent 3D printing of porous composite scaffolds. The in vitro biodegradation rate of the scaffolds containing 5% ß-TCP decreased to 0.5 mm/y, which falls within the range desired for critical-sized bone substitution. As compared to the monolithic Mg-Zn scaffolds, the elastic moduli and yield strengths of the composite scaffolds were much enhanced, which remained in the range of the cancellous bone properties even after 28 d of in vitro degradation. The Mg-Zn/5TCP and Mg-Zn/10TCP scaffolds also exhibited improved biocompatibility when cultured with preosteoblasts, as compared to Mg-Zn scaffolds. In addition, the ALP activity and mineralization level of the composite scaffolds were much enhanced in the extracts of the composite scaffolds. Taken together, this research marks a great breakthrough in fabricating porous Mg-matrix composite scaffolds that meet several design criteria in terms of appropriate biodegradation rate, mechanical properties, and bioactivity. STATEMENT OF SIGNIFICANCE: The treatment of posttraumatic femoral nonunion with large segmental bone defect is still challenging. In this study, we developed a multi-material extrusion-based additive technique to fabricate porous Mg/bioceramic composite scaffolds for such a treatment. The technique allowed for the fine-tuning of printable inks to optimize the dispersion of micro-sized particles. The relative densities of the struts of the fabricated composite scaffolds reached 99%. The added bioceramic particles (ß-TCP) exhibited proper interfacial bonding with the Mg alloy matrix. The porous Mg-based composite possessed desired biodegradability, bone-mimicking mechanical properties throughout the in vitro biodegradation period and improved bioactivity to bone cells. These results demonstrated great prospects of extrusion-based 3D printed porous Mg materials to be developed further as ideal biodegradable bone-substituting materials.


Subject(s)
Magnesium , Tissue Scaffolds , Alloys/pharmacology , Biocompatible Materials , Calcium Phosphates , Magnesium/pharmacology , Porosity , Powders , Printing, Three-Dimensional , Zinc
19.
J Mech Behav Biomed Mater ; 132: 105291, 2022 08.
Article in English | MEDLINE | ID: mdl-35660552

ABSTRACT

The reconstruction of large mandibular defects with optimal aesthetic and functional outcomes remains a major challenge for maxillofacial surgeons. The aim of this study was to design patient-specific mandibular reconstruction implants through a semi-automated digital workflow and to assess the effects of topology optimization on the biomechanical performance of the designed implants. By using the proposed workflow, a fully porous implant (LA-implant) and a topology-optimized implant (TO-implant) both made of Ti-6Al-4V ELI were designed and additively manufactured using selective laser melting. The mechanical performance of the implants was predicted by performing finite element analysis (FEA) and was experimentally assessed by conducting quasi-static and cyclic biomechanical tests. Digital image correlation (DIC) was used to validate the FE model by comparing the principal strains predicted by the FEM model with the measured distribution of the same type of strain. The numerical predictions were in good agreement with the DIC measurements and the predicted locations of specimen failure matched the actual ones. No statistically significant differences (p < 0.05) in the mean stiffness, mean ultimate load, or mean ultimate displacement were detected between the LA- and TO-implant groups. No implant failures were observed during quasi-static or cyclic testing under masticatory loads that were substantially higher (>1000 N) than the average maximum biting force of healthy individuals. Given its relatively lower weight (16.5%), higher porosity (17.4%), and much shorter design time (633.3%), the LA-implant is preferred for clinical application. This study clearly demonstrates the capability of the proposed workflow to develop patient-specific implants with high precision and superior mechanical performance, which will greatly facilitate cost- and time-effective pre-surgical planning and is expected to improve the surgical outcome.


Subject(s)
Mandibular Reconstruction , Biomechanical Phenomena , Finite Element Analysis , Humans , Stress, Mechanical , Titanium , Workflow
20.
Proc Inst Mech Eng H ; 225(12): 1121-35, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22320052

ABSTRACT

Several mass-spring-damper models have been developed to study the response of the human body to the collision with the ground during hopping, trotting, or running. The mass, spring, and damper elements represent the masses, stiffness properties, and damping properties of hard and soft tissues. The masses that models are composed of are connected to each other via springs and dampers. The present paper reviews the various types of mass-spring-damper models including one-body and multi-body models. The models are further categorized as being either passive or active. In passive models, the mechanical properties (stiffness and damping) of soft tissues remain constant regardless of the type of footwear, ground stiffness, etc. In active models, the mechanical properties adapt to external loads. The governing equations of motion of all models as well as their parameters are presented. The specific ways that the models take account of the shoe-ground interactions are discussed as well. The methods used for determination of different modelling parameters are briefly surveyed. The advantages and disadvantages of the different types of mass-spring-damper models are also discussed. The paper concludes with a brief discussion of possible future research trends in the area of mass-spring-damper modelling.


Subject(s)
Models, Biological , Muscle, Skeletal/physiology , Running/physiology , Algorithms , Biomechanical Phenomena/physiology , Body Weight/physiology , Elasticity/physiology , Extremities/physiology , Humans , Nonlinear Dynamics , Shoes , Vibration
SELECTION OF CITATIONS
SEARCH DETAIL