Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
1.
Stem Cells Transl Med ; 13(3): 278-292, 2024 Mar 15.
Article in English | MEDLINE | ID: mdl-38217535

ABSTRACT

Automated technologies are attractive for enhancing the robust manufacturing of tissue-engineered products for clinical translation. In this work, we present an automation strategy using a robotics platform for media changes, and imaging of cartilaginous microtissues cultured in static microwell platforms. We use an automated image analysis pipeline to extract microtissue displacements and morphological features as noninvasive quality attributes. As a result, empty microwells were identified with a 96% accuracy, and dice coefficient of 0.84 for segmentation. Design of experiment are used for the optimization of liquid handling parameters to minimize empty microwells during long-term differentiation protocols. We found no significant effect of aspiration or dispension speeds at and beyond manual speed. Instead, repeated media changes and time in culture were the driving force or microtissue displacements. As the ovine model is the preclinical model of choice for large skeletal defects, we used ovine periosteum-derived cells to form cartilage-intermediate microtissues. Increased expression of COL2A1 confirms chondrogenic differentiation and RUNX2 shows no osteogenic specification. Histological analysis shows an increased secretion of cartilaginous extracellular matrix and glycosaminoglycans in larger microtissues. Furthermore, microtissue-based implants are capable of forming mineralized tissues and bone after 4 weeks of ectopic implantation in nude mice. We demonstrate the development of an integrated bioprocess for culturing and manipulation of cartilaginous microtissues and anticipate the progressive substitution of manual operations with automated solutions for the manufacturing of microtissue-based living implants.


Subject(s)
Cartilage , Tissue Engineering , Mice , Animals , Sheep , Tissue Engineering/methods , Mice, Nude , Cell Differentiation , Osteogenesis , Chondrogenesis
2.
Hellenic J Cardiol ; 2023 Oct 05.
Article in English | MEDLINE | ID: mdl-37805173

ABSTRACT

BACKGROUND: Clinical characteristics and outcomes of patients with transthyretin amyloidosis cardiomyopathy (ATTR-CM) vary by region, necessitating the acquisition of country-specific evidence for proper management. METHODS: This is an observational study including sequential patients presenting in the Amyloidosis Reference Center of Greece, from 01/2014 to 12/2022. ATTR-CM was diagnosed by positive scintigraphy and exclusion of light-chain amyloidosis or positive biopsy typing. Genetic testing was performed in all cases. RESULTS: One-hundred and nine ATTR-CM patients were included (median age, 81 years) of which 15 carried TTR mutations (27% Val30Met). Most patients (82%) presented with heart failure and 59% with atrial fibrillation, while 10% had aortic stenosis. Importantly, 78 (71.6%) had clinically significant extracardiac manifestations (45% musculoskeletal disorder, 40% peripheral neuropathy and 33% gastrointestinal symptoms). Sixty-five (60%) received disease-specific treatment with tafamidis. Estimated median survival was 48 months; advanced NYHA class, National Amyloidosis Center stage, eGFR<45 ml/kg/1.73m2, NT-pro-BNP>5000 pg/mL were associated with worse survival, while tafamidis treatment was associated with improved survival in patients with IVS≥ 12 mm. DISCUSSION: These are the first data describing the characteristics, management, and outcomes of patients with ATTR-CM in Greece, which could influence local guidelines. SHORT TITLE: Transthyretin cardiomyopathy in Greece.

3.
Bioeng Transl Med ; 8(3): e10468, 2023 May.
Article in English | MEDLINE | ID: mdl-37206246

ABSTRACT

Cartilage microtissues are promising tissue modules for bottom up biofabrication of implants leading to bone defect regeneration. Hitherto, most of the protocols for the development of these cartilaginous microtissues have been carried out in static setups, however, for achieving higher scales, dynamic process needs to be investigated. In the present study, we explored the impact of suspension culture on the cartilage microtissues in a novel stirred microbioreactor system. To study the effect of the process shear stress, experiments with three different impeller velocities were carried out. Moreover, we used mathematical modeling to estimate the magnitude of shear stress on the individual microtissues during dynamic culture. Identification of appropriate mixing intensity allowed dynamic bioreactor culture of the microtissues for up to 14 days maintaining microtissue suspension. Dynamic culture did not affect microtissue viability, although lower proliferation was observed as opposed to the statically cultured ones. However, when assessing cell differentiation, gene expression values showed significant upregulation of both Indian Hedgehog (IHH) and collagen type X (COLX), well known markers of chondrogenic hypertrophy, for the dynamically cultured microtissues. Exometabolomics analysis revealed similarly distinct metabolic profiles between static and dynamic conditions. Dynamic cultured microtissues showed a higher glycolytic profile compared with the statically cultured ones while several amino acids such as proline and aspartate exhibited significant differences. Furthermore, in vivo implantations proved that microtissues cultured in dynamic conditions are functional and able to undergo endochondral ossification. Our work demonstrated a suspension differentiation process for the production of cartilaginous microtissues, revealing that shear stress resulted to an acceleration of differentiation towards hypertrophic cartilage.

4.
Carbohydr Polym ; 312: 120790, 2023 Jul 15.
Article in English | MEDLINE | ID: mdl-37059530

ABSTRACT

In this work a dual crosslinked network based on sodium alginate graft copolymer, bearing poly(N-isopropylacrylamide-co-N-tert-butylacrylamide) P(NIPAM-co-NtBAM) side chains was developed and examined as a shear thinning soft gelating bioink. The copolymer was found to undergo a two-step gelation mechanism; in the first step a three-dimensional (3D) network is formed through ionic interactions between the negatively ionized carboxylic groups of the alginate backbone and the positive charges of Ca2+ divalent cations, according to the "egg-box" mechanism. The second gelation step occurs upon heating which triggers the hydrophobic association of the thermoresponsive P(NIPAM-co-NtBAM) side chains, increasing the network crosslinking density in a highly cooperative manner. Interestingly, the dual crosslinking mechanism resulted in a five-to-eight-fold improvement of the storage modulus implying reinforced hydrophobic crosslinking above the critical thermo-gelation temperature which is further boosted by the ionic crosslinking of the alginate backbone. The proposed bioink could form arbitrary geometries under mild 3D printing conditions. Last, it is demonstrated that the proposed developed bioink can be further utilized as bioprinting ink and showcased its ability to promote human periosteum derived cells (hPDCs) growth in 3D and their capacity to form 3D spheroids. In conclusion, the bioink, owing its ability to reverse thermally the crosslinking of its polymer network, can be further utilized for the facile recovery of the cell spheroids, implying its promising potential use as cell spheroid-forming template bionk for applications in 3D biofabrication.


Subject(s)
Alginates , Hydrogels , Humans , Hydrogels/chemistry , Alginates/chemistry , Cell Proliferation , Printing, Three-Dimensional , Polymers , Tissue Engineering , Tissue Scaffolds/chemistry
5.
Acta Biomater ; 165: 111-124, 2023 Jul 15.
Article in English | MEDLINE | ID: mdl-36283613

ABSTRACT

Bone fractures are one of the most common traumatic large-organ injuries and although many fractures can heal on their own, 2-12% of fractures are slow healing or do not heal (non-unions). Autologous grafts are currently used for treatment of non-unions but are associated with limited healthy bone tissue. Tissue engineered cell-based products have promise for an alternative treatment method. It was previously demonstrated that cartilaginous microspheroids of periosteum-derived cells could be assembled into scaffold-free constructs and heal murine critically-sized long bone defects (non-unions). However, the handleability of such scaffold-free implants can be compromised when scaling-up. In this work, cartilaginous spheroids were combined with melt electrowritten (MEW) meshes to create an engineered cell-based implant, able to induce in vivo bone formation. MEW polycaprolactone meshes were tailored to contain pores (116 ± 28 µm) of a size that captured microspheroids (180 ± 15 µm). Periosteum-derived microspheroids pre-cultured for 4 days, were seeded on MEW meshes and gene expression analysis demonstrated up-regulation of chondrogenic (SOX9, COL2) and prehypertrophic (VEGF) gene markers after 14 days, creating a biohybrid sheet. When implanted subcutaneously (4 weeks), the biohybrid sheets mineralized (23 ± 3% MV/TV) and formed bone and bone marrow. Bone formation was also observed when implanted in a murine critically-sized long bone defect, though a high variation between samples was detected. The high versatility of this biofabrication approach lies in the possibility to tailor the scaffolds to shape and dimensions corresponding to the large bone defects and the individual patient using robust bone forming building blocks. These strategies are instrumental in the development of personalized regenerative therapies with predictive clinical outcomes. STATEMENT OF SIGNIFICANCE: Successful treatments for healing of large long bone defects are still limited and 2-12% of fractures do not heal properly. We combined a novel biofabrication technique: melt electrowriting (MEW), with robust biology: bone forming cartilaginous spheroids to create biohybrid sheets able to form bone upon implantation. MEW enabled the fabrication of scaffolds with micrometer-sized fibers in defined patterns which allowed the capturing of and merging with cartilaginous spheroids which had the potency to mature into bone via the developmental process of endochondral ossification. The present study contributes to the rapidly growing field of "Biofabrication with Spheroid and Organoid Materials'' and demonstrates design considerations that are of great importance for biofabrication of functional tissues through the assembly of cellular spheroids.


Subject(s)
Cartilage , Fractures, Bone , Humans , Mice , Animals , Tissue Engineering/methods , Osteogenesis , Wound Healing , Periosteum , Tissue Scaffolds
6.
Adv Mater ; 35(13): e2206110, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36461812

ABSTRACT

Surface curvature both emerges from, and influences the behavior of, living objects at length scales ranging from cell membranes to single cells to tissues and organs. The relevance of surface curvature in biology is supported by numerous experimental and theoretical investigations in recent years. In this review, first, a brief introduction to the key ideas of surface curvature in the context of biological systems is given and the challenges that arise when measuring surface curvature are discussed. Giving an overview of the emergence of curvature in biological systems, its significance at different length scales becomes apparent. On the other hand, summarizing current findings also shows that both single cells and entire cell sheets, tissues or organisms respond to curvature by modulating their shape and their migration behavior. Finally, the interplay between the distribution of morphogens or micro-organisms and the emergence of curvature across length scales is addressed with examples demonstrating these key mechanistic principles of morphogenesis. Overall, this review highlights that curved interfaces are not merely a passive by-product of the chemical, biological, and mechanical processes but that curvature acts also as a signal that co-determines these processes.


Subject(s)
Mechanical Phenomena , Cell Membrane , Morphogenesis
7.
Front Bioeng Biotechnol ; 10: 946992, 2022.
Article in English | MEDLINE | ID: mdl-36091464

ABSTRACT

Spheroids are widely applied as building blocks for biofabrication of living tissues, where they exhibit spontaneous fusion toward an integrated structure upon contact. Tissue fusion is a fundamental biological process, but due to a lack of automated monitoring systems, the in-depth characterization of this process is still limited. Therefore, a quantitative high-throughput platform was developed to semi-automatically select doublet candidates and automatically monitor their fusion kinetics. Spheroids with varying degrees of chondrogenic maturation (days 1, 7, 14, and 21) were produced from two different cell pools, and their fusion kinetics were analyzed via the following steps: (1) by applying a novel spheroid seeding approach, the background noise was decreased due to the removal of cell debris while a sufficient number of doublets were still generated. (2) The doublet candidates were semi-automatically selected, thereby reducing the time and effort spent on manual selection. This was achieved by automatic detection of the microwells and building a random forest classifier, obtaining average accuracies, sensitivities, and precisions ranging from 95.0% to 97.4%, from 51.5% to 92.0%, and from 66.7% to 83.9%, respectively. (3) A software tool was developed to automatically extract morphological features such as the doublet area, roundness, contact length, and intersphere angle. For all data sets, the segmentation procedure obtained average sensitivities and precisions ranging from 96.8% to 98.1% and from 97.7% to 98.8%, respectively. Moreover, the average relative errors for the doublet area and contact length ranged from 1.23% to 2.26% and from 2.30% to 4.66%, respectively, while the average absolute errors for the doublet roundness and intersphere angle ranged from 0.0083 to 0.0135 and from 10.70 to 13.44°, respectively. (4) The data of both cell pools were analyzed, and an exponential model was used to extract kinetic parameters from the time-series data of the doublet roundness. For both cell pools, the technology was able to characterize the fusion rate and quality in an automated manner and allowed us to demonstrate that an increased chondrogenic maturity was linked with a decreased fusion rate. The platform is also applicable to other spheroid types, enabling an increased understanding of tissue fusion. Finally, our approach to study spheroid fusion over time will aid in the design of controlled fabrication of "assembloids" and bottom-up biofabrication of living tissues using spheroids.

8.
Front Med (Lausanne) ; 9: 913287, 2022.
Article in English | MEDLINE | ID: mdl-35733863

ABSTRACT

CAR-T cell therapy is a promising treatment for acute leukemia and lymphoma. CAR-T cell therapies take a pioneering role in autologous gene therapy with three EMA-approved products. However, the chance of clinical success remains relatively low as the applicability of CAR-T cell therapy suffers from long, labor-intensive manufacturing and a lack of comprehensive insight into the bioprocess. This leads to high manufacturing costs and limited clinical success, preventing the widespread use of CAR-T cell therapies. New manufacturing approaches are needed to lower costs to improve manufacturing capacity and shorten provision times. Semi-automated devices such as the Miltenyi Prodigy® were developed to reduce hands-on production time. However, these devices are not equipped with the process analytical technology necessary to fully characterize and control the process. An automated AI-driven CAR-T cell manufacturing platform in smart manufacturing hospitals (SMH) is being developed to address these challenges. Automation will increase the cost-effectiveness and robustness of manufacturing. Using Artificial Intelligence (AI) to interpret the data collected on the platform will provide valuable process insights and drive decisions for process optimization. The smart integration of automated CAR-T cell manufacturing platforms into hospitals enables the independent manufacture of autologous CAR-T cell products. In this perspective, we will be discussing current challenges and opportunities of the patient-specific but highly automated, AI-enabled CAR-T cell manufacturing. A first automation concept will be shown, including a system architecture based on current Industry 4.0 approaches for AI integration.

9.
Biofabrication ; 14(3)2022 04 20.
Article in English | MEDLINE | ID: mdl-35354130

ABSTRACT

Microvasculature is essential for the exchange of gas and nutrient for most tissues in our body. Some tissue structures such as the meniscus presents spatially confined blood vessels adjacent to non-vascularized regions. In biofabrication, mimicking the spatial distribution of such vascular components is paramount, as capillary ingrowth into non-vascularized tissues can lead to tissue matrix alterations and subsequent pathology. Multi-material three-dimensional (3D) bioprinting strategies have the potential to resolve anisotropic tissue features, although building complex constructs comprising stable vascularized and non-vascularized regions remains a major challenge to date. In this study, we developed endothelial cell-laden pro- and anti-angiogenic bioinks, supplemented with bioactive matrix-derived microfibers (MFs) that were created from type I collagen sponges (col-1) and cartilage decellularized extracellular matrix (CdECM), respectively. Human umbilical vein endothelial cell (HUVEC)-driven capillary networks started to form 2 d after bioprinting. Supplementing cartilage-derived MFs to endothelial-cell laden bioinks reduced the total length of neo-microvessels by 29%, and the number of microvessel junctions by 37% after 14 d, compared to bioinks with pro-angiogenic col-1 MFs. As a proof of concept, the bioinks were bioprinted into an anatomical meniscus shape with a biomimetic vascularized outer and non-vascularized inner region, using a gellan gum microgel suspension bath. These 3D meniscus-like constructs were cultured up to 14 d, with in the outer zone the HUVEC-, mural cell-, and col-1 MF-laden pro-angiogenic bioink, and in the inner zone a meniscus progenitor cell (MPC)- and CdECM MF-laden anti-angiogenic bioink, revealing successful spatial confinement of the nascent vascular network only in the outer zone. Further, to co-facilitate both microvessel formation and MPC-derived matrix formation, we formulated cell culture medium conditions with a temporal switch. Overall, this study provides a new strategy that could be applied to develop zonal biomimetic meniscal constructs. Moreover, the use of ECM-derived MFs to promote or inhibit capillary networks opens new possibilities for the biofabrication of tissues with anisotropic microvascular distribution. These have potential for many applications includingin vitromodels of vascular-to-avascular tissue interfaces, cancer progression, and for testing anti-angiogenic therapies.


Subject(s)
Bioprinting , Tissue Engineering , Bioprinting/methods , Cartilage , Extracellular Matrix , Human Umbilical Vein Endothelial Cells , Humans , Printing, Three-Dimensional , Tissue Engineering/methods , Tissue Scaffolds/chemistry
10.
Biofabrication ; 13(4)2021 09 21.
Article in English | MEDLINE | ID: mdl-34450613

ABSTRACT

Spheroids have become essential building blocks for biofabrication of functional tissues. Spheroid formats allow high cell-densities to be efficiently engineered into tissue structures closely resembling the native tissues. In this work, we explore the assembly capacity of cartilaginous spheroids (d∼ 150µm) in the context of endochondral bone formation. The fusion capacity of spheroids at various degrees of differentiation was investigated and showed decreased kinetics as well as remodeling capacity with increased spheroid maturity. Subsequently, design considerations regarding the dimensions of engineered spheroid-based cartilaginous mesotissues were explored for the corresponding time points, defining critical dimensions for these type of tissues as they progressively mature. Next, mesotissue assemblies were implanted subcutaneously in order to investigate the influence of spheroid fusion parameters on endochondral ossification. Moreover, as a step towards industrialization, we demonstrated a novel automated image-guided robotics process, based on targeting and registering single-spheroids, covering the range of spheroid and mesotissue dimensions investigated in this work. This work highlights a robust and automated high-precision biomanufacturing roadmap for producing spheroid-based implants for bone regeneration.


Subject(s)
Osteogenesis , Tissue Engineering , Bone Regeneration , Cartilage , Robotics , Spheroids, Cellular , Tissue Scaffolds
11.
Soft Matter ; 17(27): 6603-6615, 2021 Jul 21.
Article in English | MEDLINE | ID: mdl-34142683

ABSTRACT

The mechanical microenvironment of cells has been associated with phenotypic changes that cells undergo in three-dimensional spheroid culture formats. Radial asymmetry in mechanical stress - with compression in the core and tension at the periphery - has been analyzed by representing tissue spheroids as homogeneous visco-elastic droplets under surface tension. However, the influence of the granular microstructure of tissue spheroids in the distribution of mechanical stress in tissue spheroids has not been accounted for in a generic manner. Here, we quantify the distribution and propagation of mechanical forces in structurally heterogeneous multicellular assemblies. For this, we perform numerical simulations of a deformable cell model, which represents cells as elastic, contractile shells surrounding a liquid incompressible cytoplasm, interacting by means of non-specific adhesion. Using this model, we show how cell-scale properties such as cortical stiffness, active tension and cell-cell adhesive tension influence the distribution of mechanical stress in simulated tissue spheroids. Next, we characterize the transition at the tissue-scale from a homogeneous liquid droplet to a heterogeneous packed granular assembly.


Subject(s)
Mechanical Phenomena , Spheroids, Cellular , Pressure , Stress, Mechanical , Surface Tension
12.
Front Bioeng Biotechnol ; 9: 624890, 2021.
Article in English | MEDLINE | ID: mdl-34109163

ABSTRACT

An increasing need toward a more efficient expansion of adherent progenitor cell types arises with the advancements of cell therapy. The use of a dynamic expansion instead of a static planar expansion could be one way to tackle the challenges of expanding adherent cells at a large scale. Microcarriers are often reported as a biomaterial for culturing cells in suspension. However, the type of microcarrier has an effect on the cell expansion. In order to find an efficient expansion process for a specific adherent progenitor cell type, it is important to investigate the effect of the type of microcarrier on the cell expansion. Human periosteum-derived progenitor cells are extensively used in skeletal tissue engineering for the regeneration of bone defects. Therefore, we evaluated the use of different microcarriers on human periosteum-derived progenitor cells. In order to assess the potency, identity and viability of these cells after being cultured in the spinner flasks, this study performed several in vitro and in vivo analyses. The novelty of this work lies in the combination of screening different microcarriers for human periosteum-derived progenitor cells with in vivo assessments of the cells' potency using the microcarrier that was selected as the most promising one. The results showed that expanding human periosteum-derived progenitor cells in spinner flasks using xeno-free medium and Star-Plus microcarriers, does not affect the potency, identity or viability of the cells. The potency of the cells was assured with an in vivo evaluation, where bone formation was achieved. In summary, this expansion method has the potential to be used for large scale cell expansion with clinical relevance.

13.
Biomaterials ; 273: 120820, 2021 06.
Article in English | MEDLINE | ID: mdl-33872857

ABSTRACT

Tissue engineered constructs have the potential to respond to the unmet medical need of treating deep osteochondral defects. However, current tissue engineering strategies struggle in the attempt to create patterned constructs with biologically distinct functionality. In this work, a developmentally-inspired modular approach is proposed, whereby distinct cartilaginous organoids are used as living building blocks. First, a hierarchical construct was created, composed of three layers of cartilaginous tissue intermediates derived from human periosteum-derived cells: (i) early (SOX9), (ii) mature (COL2) and (iii) (pre)hypertrophic (IHH, COLX) phenotype. Subcutaneous implantation in nude mice generated a hybrid tissue containing one mineralized and one non-mineralized part. However, the non-mineralized part was represented by a collagen type I positive fibrocartilage-like tissue. To engineer a more stable articular cartilage part, iPSC-derived cartilage microtissues (SOX9, COL2; IHH neg) were generated. Subcutaneous implantation of assembled iPSC-derived cartilage microtissues resulted in a homogenous cartilaginous tissue positive for collagen type II but negative for osteocalcin. Finally, iPSC-derived cartilage microtissues in combination with the pre-hypertrophic cartilage organoids (IHH, COLX) could form dual tissues consisting of i) a cartilaginous safranin O positive and ii) a bony osteocalcin positive region upon subcutaneous implantation, corresponding to the pre-engineered zonal pattern. The assembly of functional building blocks, as presented in this work, opens possibilities for the production of complex tissue engineered implants by embedding zone-specific functionality through the use of pre-programmed living building blocks.


Subject(s)
Cartilage, Articular , Organoids , Animals , Collagen Type II , Mice , Mice, Nude , Tissue Engineering , Tissue Scaffolds
14.
Adv Drug Deliv Rev ; 169: 22-39, 2021 02.
Article in English | MEDLINE | ID: mdl-33290762

ABSTRACT

A decade after the term developmental engineering (DE) was coined to indicate the use of developmental processes as blueprints for the design and development of engineered living implants, a myriad of proof-of-concept studies demonstrate the potential of this approach in small animal models. This review provides an overview of DE work, focusing on applications in bone regeneration. Enabling technologies allow to quantify the distance between in vitro processes and their developmental counterpart, as well as to design strategies to reduce that distance. By embedding Nature's robust mechanisms of action in engineered constructs, predictive large animal data and subsequent positive clinical outcomes can be gradually achieved. To this end, the development of next generation biofabrication technologies should provide the necessary scale and precision for robust living bone implant biomanufacturing.


Subject(s)
Bone and Bones , Prostheses and Implants , Tissue Engineering , Animals , Computer Simulation , Humans
15.
Bioengineering (Basel) ; 7(3)2020 Jul 20.
Article in English | MEDLINE | ID: mdl-32698462

ABSTRACT

Implementing a personalised feeding strategy for each individual batch of a bioprocess could significantly reduce the unnecessary costs of overfeeding the cells. This paper uses lactate measurements during the cell culture process as an indication of cell growth to adapt the feeding strategy accordingly. For this purpose, a model predictive control is used to follow this a priori determined reference trajectory of cumulative lactate. Human progenitor cells from three different donors, which were cultivated in 12-well plates for five days using six different feeding strategies, are used as references. Each experimental set-up is performed in triplicate and for each run an individualised model-based predictive control (MPC) controller is developed. All process models exhibit an accuracy of 99.80% ± 0.02%, and all simulations to reproduce each experimental run, using the data as a reference trajectory, reached their target with a 98.64% ± 0.10% accuracy on average. This work represents a promising framework to control the cell growth through adapting the feeding strategy based on lactate measurements.

16.
Article in English | MEDLINE | ID: mdl-32411692

ABSTRACT

Stem cell expansion on 3D porous scaffolds cultured in bioreactor systems has been shown to be beneficial for maintenance of the original cell functionality in tissue engineering strategies (TE). However, the production of extracellular matrix (ECM) makes harvesting the progenitor cell population from 3D scaffolds a challenge. Medium composition plays a role in stimulating cell proliferation over extracellular matrix (ECM) production. In this regard, a computational model describing tissue growth inside 3D scaffolds can be a great tool in designing optimal experimental conditions. In this study, a computational model describing cell and ECM growth in a perfusion bioreactor is developed, including a description of the effect of a (generic) growth factor on the biological processes taking place inside the 3D scaffold. In the model, the speed of cell and ECM growth depends on the flow-induced shear stress, curvature and the concentrations of oxygen, glucose, lactate, and growth factor. The effect of the simulated growth factor is to differentially enhance cell proliferation over ECM production. After model calibration with historic in-house data, a multi-objective optimization procedure is executed aiming to minimize the total experimental cost whilst maximizing cell growth during culture. The obtained results indicate there are multiple optimum points for the medium refreshment regime and the initial growth factor concentration where a trade-off is made between the final amount of cells and the culture cost. Finally, the model is applied to experiments reported in the literature studying the effects of perfusion-based cell culture and/or growth factor supplementation on cell expansion. The qualitative similarities between the simulation and experimental results, even in the absence of proper model calibration, reinforces the generic character of the proposed modeling framework. The model proposed in this study can contribute to the cost efficient production of cell-based TE products, ultimately contributing to their affordability and accessibility.

17.
Materials (Basel) ; 13(6)2020 Mar 12.
Article in English | MEDLINE | ID: mdl-32178317

ABSTRACT

In the current study, a new approach for surface modification and surface hardening of aluminum alloys is developed. The method is based on the logic of in-situ reinforcing FSP strategies. The novelty of the proposed process is the application of a bulk reinforcing metallic material instead of metallic powders. The FSP was carried out on aluminum alloy AA5083-thick plates. A thin sheet of pure copper (cross-section 4 × 0.8 mm2) was placed in a machined groove on the upper surface of the aluminum plate, and both materials were FSPed together. Samples with one, two and three FSP passes were manufactured respectively. Results indicate that the copper thin sheet was successfully integrated in the AA5083 stir zone. By increasing the FSP passes, almost all copper was integrated in the stir zone, mainly in the form of coper-based micron-sized intermetallic particles, and secondly, by copper diffusion in the AA5083 matrix. Due to the presence of complex intermetallic compounds created by the high heat input and intense plastic deformation, the hardness inside the stir-zone was found highly increased from 77 to 138 HV.

18.
Cytotherapy ; 22(2): 82-90, 2020 02.
Article in English | MEDLINE | ID: mdl-31987754

ABSTRACT

BACKGROUND: Human mesenchymal stromal cells (hMSCs) have become attractive candidates for advanced medical cell-based therapies. An in vitro expansion step is routinely used to reach the required clinical quantities. However, this is influenced by many variables including donor characteristics, such as age and gender, and culture conditions, such as cell seeding density and available culture surface area. Computational modeling in general and machine learning in particular could play a significant role in deciphering the relationship between the individual donor characteristics and their growth dynamics. METHODS: In this study, hMSCs obtained from 174 male and female donors, between 3 and 64 years of age with passage numbers ranging from 2 to 27, were studied. We applied a Random Forests (RF) technique to model the cell expansion procedure by predicting the population doubling time (PDT) for each passage, taking into account individual donor-related characteristics. RESULTS: Using the RF model, the mean absolute error between model predictions and experimental results for the PDT in passage 1 to 4 is significantly lower compared with the errors obtained with theoretical estimates or historical data. Moreover, statistical analysis indicate that the PD and PDT in different age categories are significantly different, especially in the youngest group (younger than 10 years of age) compared with the other age groups. DISCUSSION: In summary, we introduce a predictive computational model describing in vitro cell expansion dynamics based on individual donor characteristics, an approach that could greatly assist toward automation of a cell expansion culture process.


Subject(s)
Cell Proliferation/physiology , Cell- and Tissue-Based Therapy/methods , Computer Simulation , Mesenchymal Stem Cells/cytology , Adolescent , Adult , Cell Count , Cell Differentiation , Child , Child, Preschool , Female , Humans , Machine Learning , Male , Middle Aged , Tissue Donors , Young Adult
19.
Adv Sci (Weinh) ; 7(2): 1902295, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31993293

ABSTRACT

Clinical translation of cell-based products is hampered by their limited predictive in vivo performance. To overcome this hurdle, engineering strategies advocate to fabricate tissue products through processes that mimic development and regeneration, a strategy applicable for the healing of large bone defects, an unmet medical need. Natural fracture healing occurs through the formation of a cartilage intermediate, termed "soft callus," which is transformed into bone following a process that recapitulates developmental events. The main contributors to the soft callus are cells derived from the periosteum, containing potent skeletal stem cells. Herein, cells derived from human periosteum are used for the scalable production of microspheroids that are differentiated into callus organoids. The organoids attain autonomy and exhibit the capacity to form ectopic bone microorgans in vivo. This potency is linked to specific gene signatures mimicking those found in developing and healing long bones. Furthermore, callus organoids spontaneously bioassemble in vitro into large engineered tissues able to heal murine critical-sized long bone defects. The regenerated bone exhibits similar morphological properties to those of native tibia. These callus organoids can be viewed as a living "bio-ink" allowing bottom-up manufacturing of multimodular tissues with complex geometric features and inbuilt quality attributes.

20.
Stem Cells Transl Med ; 8(8): 810-821, 2019 08.
Article in English | MEDLINE | ID: mdl-31038850

ABSTRACT

Xenogeneic-free media are required for translating advanced therapeutic medicinal products to the clinics. In addition, process efficiency is crucial for ensuring cost efficiency, especially when considering large-scale production of mesenchymal stem cells (MSCs). Human platelet lysate (HPL) has been increasingly adopted as an alternative for fetal bovine serum (FBS) for MSCs. However, its therapeutic and regenerative potential in vivo is largely unexplored. Herein, we compare the effects of FBS and HPL supplementation for a scalable, microcarrier-based dynamic expansion of human periosteum-derived cells (hPDCs) while assessing their bone forming capacity by subcutaneous implantation in small animal model. We observed that HPL resulted in faster cell proliferation with a total fold increase of 5.2 ± 0.61 in comparison to 2.7 ± 02.22-fold in FBS. Cell viability and trilineage differentiation capability were maintained by HPL, although a suppression of adipogenic differentiation potential was observed. Differences in mRNA expression profiles were also observed between the two on several markers. When implanted, we observed a significant difference between the bone forming capacity of cells expanded in FBS and HPL, with HPL supplementation resulting in almost three times more mineralized tissue within calcium phosphate scaffolds. FBS-expanded cells resulted in a fibrous tissue structure, whereas HPL resulted in mineralized tissue formation, which can be classified as newly formed bone, verified by µCT and histological analysis. We also observed the presence of blood vessels in our explants. In conclusion, we suggest that replacing FBS with HPL in bioreactor-based expansion of hPDCs is an optimal solution that increases expansion efficiency along with promoting bone forming capacity of these cells. Stem Cells Translational Medicine 2019;8:810&821.


Subject(s)
Batch Cell Culture Techniques/methods , Bone Regeneration , Culture Media/pharmacology , Primary Cell Culture/methods , Stem Cells/drug effects , Adipogenesis , Animals , Batch Cell Culture Techniques/instrumentation , Bioreactors , Blood Platelets/metabolism , Cell Differentiation , Cell Proliferation , Cells, Cultured , Culture Media/chemistry , Humans , Mice , Mice, Nude , Osteogenesis , Periosteum/cytology , Primary Cell Culture/instrumentation , Stem Cell Transplantation/methods , Stem Cells/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...