Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 111
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Nature ; 579(7797): 111-117, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32103177

RESUMEN

The avascular nature of cartilage makes it a unique tissue1-4, but whether and how the absence of nutrient supply regulates chondrogenesis remain unknown. Here we show that obstruction of vascular invasion during bone healing favours chondrogenic over osteogenic differentiation of skeletal progenitor cells. Unexpectedly, this process is driven by a decreased availability of extracellular lipids. When lipids are scarce, skeletal progenitors activate forkhead box O (FOXO) transcription factors, which bind to the Sox9 promoter and increase its expression. Besides initiating chondrogenesis, SOX9 acts as a regulator of cellular metabolism by suppressing oxidation of fatty acids, and thus adapts the cells to an avascular life. Our results define lipid scarcity as an important determinant of chondrogenic commitment, reveal a role for FOXO transcription factors during lipid starvation, and identify SOX9 as a critical metabolic mediator. These data highlight the importance of the nutritional microenvironment in the specification of skeletal cell fate.


Asunto(s)
Huesos/citología , Microambiente Celular , Condrogénesis , Metabolismo de los Lípidos , Factor de Transcripción SOX9/metabolismo , Células Madre/citología , Células Madre/metabolismo , Animales , Huesos/irrigación sanguínea , Condrocitos/citología , Condrocitos/metabolismo , Ácidos Grasos/metabolismo , Femenino , Privación de Alimentos , Factores de Transcripción Forkhead/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Osteogénesis , Oxidación-Reducción , Factor de Transcripción SOX9/genética , Transducción de Señal , Cicatrización de Heridas
2.
BMC Biol ; 20(1): 253, 2022 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-36352408

RESUMEN

BACKGROUND: Without the availability of disease-modifying drugs, there is an unmet therapeutic need for osteoarthritic patients. During osteoarthritis, the homeostasis of articular chondrocytes is dysregulated and a phenotypical transition called hypertrophy occurs, leading to cartilage degeneration. Targeting this phenotypic transition has emerged as a potential therapeutic strategy. Chondrocyte phenotype maintenance and switch are controlled by an intricate network of intracellular factors, each influenced by a myriad of feedback mechanisms, making it challenging to intuitively predict treatment outcomes, while in silico modeling can help unravel that complexity. In this study, we aim to develop a virtual articular chondrocyte to guide experiments in order to rationalize the identification of potential drug targets via screening of combination therapies through computational modeling and simulations. RESULTS: We developed a signal transduction network model using knowledge-based and data-driven (machine learning) modeling technologies. The in silico high-throughput screening of (pairwise) perturbations operated with that network model highlighted conditions potentially affecting the hypertrophic switch. A selection of promising combinations was further tested in a murine cell line and primary human chondrocytes, which notably highlighted a previously unreported synergistic effect between the protein kinase A and the fibroblast growth factor receptor 1. CONCLUSIONS: Here, we provide a virtual articular chondrocyte in the form of a signal transduction interactive knowledge base and of an executable computational model. Our in silico-in vitro strategy opens new routes for developing osteoarthritis targeting therapies by refining the early stages of drug target discovery.


Asunto(s)
Cartílago Articular , Osteoartritis , Humanos , Ratones , Animales , Cartílago Articular/metabolismo , Osteoartritis/tratamiento farmacológico , Osteoartritis/genética , Osteoartritis/metabolismo , Condrocitos/metabolismo , Hipertrofia/metabolismo , Transducción de Señal
3.
Adv Funct Mater ; 31(42)2021 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-34924912

RESUMEN

Bioengineering of tissues and organs has the potential to generate functional replacement organs. However, achieving the full-thickness vascularization that is required for long-term survival of living implants has remained a grand challenge, especially for clinically sized implants. During the pre-vascular phase, implanted engineered tissues are forced to metabolically rely on the diffusion of nutrients from adjacent host-tissue, which for larger living implants results in anoxia, cell death, and ultimately implant failure. Here it is reported that this challenge can be addressed by engineering self-oxygenating tissues, which is achieved via the incorporation of hydrophobic oxygen-generating micromaterials into engineered tissues. Self-oxygenation of tissues transforms anoxic stresses into hypoxic stimulation in a homogenous and tissue size-independent manner. The in situ elevation of oxygen tension enables the sustained production of high quantities of angiogenic factors by implanted cells, which are offered a metabolically protected pro-angiogenic microenvironment. Numerical simulations predict that self-oxygenation of living tissues will effectively orchestrate rapid full-thickness vascularization of implanted tissues, which is empirically confirmed via in vivo experimentation. Self-oxygenation of tissues thus represents a novel, effective, and widely applicable strategy to enable the vascularization living implants, which is expected to advance organ transplantation and regenerative medicine applications.

4.
Eur Heart J ; 41(48): 4556-4564, 2020 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-32128588

RESUMEN

Providing therapies tailored to each patient is the vision of precision medicine, enabled by the increasing ability to capture extensive data about individual patients. In this position paper, we argue that the second enabling pillar towards this vision is the increasing power of computers and algorithms to learn, reason, and build the 'digital twin' of a patient. Computational models are boosting the capacity to draw diagnosis and prognosis, and future treatments will be tailored not only to current health status and data, but also to an accurate projection of the pathways to restore health by model predictions. The early steps of the digital twin in the area of cardiovascular medicine are reviewed in this article, together with a discussion of the challenges and opportunities ahead. We emphasize the synergies between mechanistic and statistical models in accelerating cardiovascular research and enabling the vision of precision medicine.


Asunto(s)
Inteligencia Artificial , Cardiología , Algoritmos , Humanos , Medicina de Precisión
5.
Cytotherapy ; 22(2): 82-90, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31987754

RESUMEN

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.


Asunto(s)
Proliferación Celular/fisiología , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Simulación por Computador , Células Madre Mesenquimatosas/citología , Adolescente , Adulto , Recuento de Células , Diferenciación Celular , Niño , Preescolar , Femenino , Humanos , Aprendizaje Automático , Masculino , Persona de Mediana Edad , Donantes de Tejidos , Adulto Joven
6.
Hum Mutat ; 40(10): 1760-1767, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31066482

RESUMEN

Neurofibromatosis type 1 (NF1) is an autosomal dominant disorder affecting approximately 1 in 2,000 newborns. Up to 5% of NF1 patients suffer from pseudarthrosis of a long bone (NF1-PA). Current treatments are often unsatisfactory, potentially leading to amputation. To gain more insight into the pathogenesis we cultured cells from PA tissue and normal-appearing periosteum of the affected bone for NF1 mutation analysis. PA cells were available from 13 individuals with NF1. Biallelic NF1 inactivation was identified in all investigated PA cells obtained during the first surgery. Three of five cases sampled during a later intervention showed biallelic NF1 inactivation. Also, in three individuals, we examined periosteum-derived cells from normal-appearing periosteum proximal and distal to the PA. We identified the same biallelic NF1 inactivation in the periosteal cells outside the PA region. These results indicate that NF1 inactivation is required but not sufficient for the development of NF1-PA. We observed that late-onset NF1-PA occurs and is not always preceded by congenital bowing. Furthermore, the failure to identify biallelic inactivation in two of five later interventions and one reintervention with a known somatic mutation indicates that NF1-PA can persist after the removal of most NF1 negative cells.


Asunto(s)
Neurofibromatosis 1/complicaciones , Seudoartrosis/diagnóstico , Seudoartrosis/etiología , Alelos , Biopsia , Preescolar , Análisis Mutacional de ADN , Exones , Femenino , Silenciador del Gen , Humanos , Masculino , Mutación , Neurofibromatosis 1/diagnóstico , Neurofibromatosis 1/genética , Neurofibromina 1/genética
7.
J Acoust Soc Am ; 145(2): 1048, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30823826

RESUMEN

Bone healing process is a complicated phenomenon regulated by biochemical and mechanical signals. Experimental studies have shown that ultrasound (US) accelerates bone ossification and has a multiple influence on cell differentiation and angiogenesis. In a recent work of the authors, a bioregulatory model for providing bone-healing predictions was addressed, taking into account for the first time the salutary effect of US on the involved angiogenesis. In the present work, a mechanobioregulatory model of bone solidification under the US presence incorporating also the mechanical environment on the regeneration process, which is known to affect cellular processes, is presented. An iterative procedure is adopted, where the finite element method is employed to compute the mechanical stimuli at the linear elastic phases of the poroelastic callus region and a coupled system of partial differential equations to simulate the enhancement by the US cell angiogenesis process and thus the oxygen concentration in the fractured area. Numerical simulations with and without the presence of US that illustrate the influence of progenitor cells' origin in the healing pattern and the healing rate and simultaneously demonstrate the salutary effect of US on bone repair are presented and discussed.


Asunto(s)
Fenómenos Biomecánicos/efectos de la radiación , Huesos , Curación de Fractura/efectos de la radiación , Modelos Biológicos , Ondas Ultrasónicas , Animales , Huesos/citología , Huesos/efectos de la radiación , Simulación por Computador , Curación de Fractura/fisiología , Fracturas Óseas/fisiopatología , Osteogénesis/efectos de la radiación
8.
Biotechnol Bioeng ; 115(3): 617-629, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29205280

RESUMEN

In regenerative medicine, computer models describing bioreactor processes can assist in designing optimal process conditions leading to robust and economically viable products. In this study, we started from a (3D) mechanistic model describing the growth of neotissue, comprised of cells, and extracellular matrix, in a perfusion bioreactor set-up influenced by the scaffold geometry, flow-induced shear stress, and a number of metabolic factors. Subsequently, we applied model reduction by reformulating the problem from a set of partial differential equations into a set of ordinary differential equations. Comparing the reduced model results to the mechanistic model results and to dedicated experimental results assesses the reduction step quality. The obtained homogenized model is 105 fold faster than the 3D version, allowing the application of rigorous optimization techniques. Bayesian optimization was applied to find the medium refreshment regime in terms of frequency and percentage of medium replaced that would maximize neotissue growth kinetics during 21 days of culture. The simulation results indicated that maximum neotissue growth will occur for a high frequency and medium replacement percentage, a finding that is corroborated by reports in the literature. This study demonstrates an in silico strategy for bioprocess optimization paying particular attention to the reduction of the associated computational cost.


Asunto(s)
Reactores Biológicos , Técnicas de Cultivo de Célula/métodos , Modelos Biológicos , Periostio/citología , Periostio/crecimiento & desarrollo , Ingeniería de Tejidos/métodos , Técnicas de Cultivo de Célula/instrumentación , Células Cultivadas , Humanos , Ingeniería de Tejidos/instrumentación
9.
PLoS Comput Biol ; 12(9): e1005108, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-27658116

RESUMEN

Perfusion bioreactors regulate flow conditions in order to provide cells with oxygen, nutrients and flow-associated mechanical stimuli. Locally, these flow conditions can vary depending on the scaffold geometry, cellular confluency and amount of extra cellular matrix deposition. In this study, a novel application of the immersed boundary method was introduced in order to represent a detailed deformable cell attached to a 3D scaffold inside a perfusion bioreactor and exposed to microscopic flow. The immersed boundary model permits the prediction of mechanical effects of the local flow conditions on the cell. Incorporating stiffness values measured with atomic force microscopy and micro-flow boundary conditions obtained from computational fluid dynamics simulations on the entire scaffold, we compared cell deformation, cortical tension, normal and shear pressure between different cell shapes and locations. We observed a large effect of the precise cell location on the local shear stress and we predicted flow-induced cortical tensions in the order of 5 pN/µm, at the lower end of the range reported in literature. The proposed method provides an interesting tool to study perfusion bioreactors processes down to the level of the individual cell's micro-environment, which can further aid in the achievement of robust bioprocess control for regenerative medicine applications.

10.
J Theor Biol ; 365: 247-64, 2015 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-25452136

RESUMEN

A timely restoration of the ruptured blood vessel network in order to deliver oxygen and nutrients to the fracture zone is crucial for successful bone healing. Indeed, oxygen plays a key role in the aerobic metabolism of cells, in the activity of a myriad of enzymes as well as in the regulation of several (angiogenic) genes. In this paper, a previously developed model of bone fracture healing is further improved with a detailed description of the influence of oxygen on various cellular processes that occur during bone fracture healing. Oxygen ranges of the cell-specific oxygen-dependent processes were established based on the state-of-the art experimental knowledge through a rigorous literature study. The newly developed oxygen model is compared with previously published experimental and in silico results. An extensive sensitivity analysis was also performed on the newly introduced oxygen thresholds, indicating the robustness of the oxygen model. Finally, the oxygen model was applied to the challenging clinical case of a critical sized defect (3mm) where it predicted the formation of a fracture non-union. Further model analyses showed that the harsh hypoxic conditions in the central region of the callus resulted in cell death and disrupted bone healing thereby indicating the importance of a timely vascularization for the successful healing of a large bone defect. In conclusion, this work demonstrates that the oxygen model is a powerful tool to further unravel the complex spatiotemporal interplay of oxygen delivery, diffusion and consumption with the several healing steps, each occurring at distinct, optimal oxygen tensions during the bone repair process.


Asunto(s)
Curación de Fractura/efectos de los fármacos , Fracturas Óseas/patología , Modelos Biológicos , Oxígeno/farmacología , Callo Óseo/efectos de los fármacos , Callo Óseo/patología , Simulación por Computador , Matriz Extracelular/metabolismo , Humanos , Factores de Tiempo
11.
PLoS Comput Biol ; 10(11): e1003888, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25375821

RESUMEN

Although bone has a unique restorative capacity, i.e., it has the potential to heal scarlessly, the conditions for spontaneous bone healing are not always present, leading to a delayed union or a non-union. In this work, we use an integrative in vivo-in silico approach to investigate the occurrence of non-unions, as well as to design possible treatment strategies thereof. The gap size of the domain geometry of a previously published mathematical model was enlarged in order to study the complex interplay of blood vessel formation, oxygen supply, growth factors and cell proliferation on the final healing outcome in large bone defects. The multiscale oxygen model was not only able to capture the essential aspects of in vivo non-unions, it also assisted in understanding the underlying mechanisms of action, i.e., the delayed vascularization of the central callus region resulted in harsh hypoxic conditions, cell death and finally disrupted bone healing. Inspired by the importance of a timely vascularization, as well as by the limited biological potential of the fracture hematoma, the influence of the host environment on the bone healing process in critical size defects was explored further. Moreover, dependent on the host environment, several treatment strategies were designed and tested for effectiveness. A qualitative correspondence between the predicted outcomes of certain treatment strategies and experimental observations was obtained, clearly illustrating the model's potential. In conclusion, the results of this study demonstrate that due to the complex non-linear dynamics of blood vessel formation, oxygen supply, growth factor production and cell proliferation and the interactions thereof with the host environment, an integrative in silico-in vivo approach is a crucial tool to further unravel the occurrence and treatments of challenging critical sized bone defects.


Asunto(s)
Curación de Fractura/fisiología , Fracturas Óseas/fisiopatología , Modelos Biológicos , Animales , Cartílago/metabolismo , Células Cultivadas , Simulación por Computador , Masculino , Ratones , Ratones Endogámicos C57BL , Oxígeno/metabolismo , Periostio/citología , Ingeniería de Tejidos , Andamios del Tejido
12.
Biotechnol Bioeng ; 111(12): 2560-70, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24902541

RESUMEN

Perfusion bioreactors have shown great promise for tissue engineering applications providing a homogeneous and consistent distribution of nutrients and flow-induced shear stresses throughout tissue-engineered constructs. However, non-uniform fluid-flow profiles found in the perfusion chamber entrance region have been shown to affect tissue-engineered construct quality characteristics during culture. In this study a whole perfusion and construct, three dimensional (3D) computational fluid dynamics approach was used in order to optimize a critical design parameter such as the location of the regular pore scaffolds within the perfusion bioreactor chamber. Computational studies were coupled to bioreactor experiments for a case-study flow rate. Two cases were compared in the first instance seeded scaffolds were positioned immediately after the perfusion chamber inlet while a second group was positioned at the computationally determined optimum distance were a steady state flow profile had been reached. Experimental data showed that scaffold location affected significantly cell content and neo-tissue distribution, as determined and quantified by contrast enhanced nanoCT, within the constructs both at 14 and 21 days of culture. However, gene expression level of osteopontin and osteocalcin was not affected by the scaffold location. This study demonstrates that the bioreactor chamber environment, incorporating a scaffold and its location within it, affects the flow patterns within the pores throughout the scaffold requiring therefore dedicated optimization that can lead to bone tissue engineered constructs with improved quality attributes.


Asunto(s)
Reactores Biológicos , Periostio/citología , Ingeniería de Tejidos/métodos , Andamios del Tejido , Células Cultivadas , ADN/análisis , Humanos , Perfusión , Células Madre/citología , Tomografía Computarizada por Rayos X
13.
Int Orthop ; 38(9): 1771-8, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24984594

RESUMEN

In silico, defined in analogy to in vitro and in vivo as those studies that are performed on a computer, is an essential step in problem-solving and product development in classical engineering fields. The use of in silico models is now slowly easing its way into medicine. In silico models are already used in orthopaedics for the planning of complicated surgeries, personalised implant design and the analysis of gait measurements. However, these in silico models often lack the simulation of the response of the biological system over time. In silico models focusing on the response of the biological systems are in full development. This review starts with an introduction into in silico models of orthopaedic processes. Special attention is paid to the classification of models according to their spatiotemporal scale (gene/protein to population) and the information they were built on (data vs hypotheses). Subsequently, the review focuses on the in silico models used in regenerative orthopaedics research. Contributions of in silico models to an enhanced understanding and optimisation of four key elements-cells, carriers, culture and clinics-are illustrated. Finally, a number of challenges are identified, related to the computational aspects but also to the integration of in silico tools into clinical practice.


Asunto(s)
Regeneración Ósea , Ortopedia/métodos , Medicina Regenerativa/métodos , Simulación por Computador , Humanos , Técnicas In Vitro , Ortopedia/tendencias , Medicina Regenerativa/tendencias , Ingeniería de Tejidos/métodos , Ingeniería de Tejidos/tendencias
14.
Comput Struct Biotechnol J ; 23: 2453-2464, 2024 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38882677

RESUMEN

The kinetics of the protein elongation cycle by the ribosome depends on intertwined factors. One of these factors is the electrostatic interaction of the nascent protein with the ribosome exit tunnel. In this computational biology theoretical study, we focus on the rate of the peptide bond formation and its dependence on the ribosome exit tunnel electrostatic potential profile. We quantitatively predict how oligopeptides of variable lengths can affect the peptide bond formation rate. We applied the Michaelis-Menten model as previously extended to incorporate the mechano-biochemical effects of forces on the rate of reaction at the catalytic site of the ribosome. For a given pair of carboxy-terminal amino acid substrate at the P- and an aminoacyl-tRNA at the A-sites, the relative time courses of the peptide bond formation reaction can be reversed depending on the oligopeptide sequence embedded in the tunnel and their variable lengths from the P-site. The reversal is predicted to occur from a shift in positions of charged amino acids upstream in the oligopeptidyl-tRNA at the P-site. The position shift must be adjusted by clever design of the oligopeptide probes using the electrostatic potential profile along the exit tunnel axial path. These predicted quantitative results bring strong evidence of the importance and relative contribution of the electrostatic interaction of the ribosome exit tunnel with the nascent peptide chain during elongation.

15.
Mater Today Bio ; 26: 101100, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38854953

RESUMEN

Objectives: Calcium phosphate-based biomaterials (CaP) are the most widely used biomaterials to enhance bone regeneration in the treatment of alveolar bone deficiencies, cranio-maxillofacial and periodontal infrabony defects, with positive preclinical and clinical results reported. This systematic review aimed to assess the influence of the physicochemical properties of CaP biomaterials on the performance of bone regeneration in preclinical animal models. Methods: The PubMed, EMBASE and Web of Science databases were searched to retrieve the preclinical studies investigating physicochemical characteristics of CaP biomaterials. The studies were screened for inclusion based on intervention (physicochemical characterization and in vivo evaluation) and reported measurable outcomes. Results: A total of 1532 articles were retrieved and 58 studies were ultimately included in the systematic review. A wide range of physicochemical characteristics of CaP biomaterials was found to be assessed in the included studies. Despite a high degree of heterogeneity, the meta-analysis was performed on 39 studies and evidenced significant effects of biomaterial characteristics on their bone regeneration outcomes. The study specifically showed that macropore size, Ca/P ratio, and compressive strength exerted significant influence on the formation of newly regenerated bone. Moreover, factors such as particle size, Ca/P ratio, and surface area were found to impact bone-to-material contact during the regeneration process. In terms of biodegradability, the amount of residual graft was determined by macropore size, particle size, and compressive strength. Conclusion: The systematic review showed that the physicochemical characteristics of CaP biomaterials are highly determining for scaffold's performance, emphasizing its usefulness in designing the next generation of bone scaffolds to target higher rates of regeneration.

16.
Gels ; 10(2)2024 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-38391470

RESUMEN

While available treatments have addressed a variety of complications in the dentoalveolar region, associated challenges have resulted in exploration of tissue engineering techniques. Often, scaffold biomaterials with specific properties are required for such strategies to be successful, development of which is an active area of research. This study focuses on the development of a copolymer of poly (N-isopropylacrylamide) (pNIPAM) and chitosan, used for 3D printing of scaffolds for dentoalveolar regeneration. The synthesized material was characterized by Fourier transform infrared spectroscopy, and the possibility of printing was evaluated through various printability tests. The rate of degradation and swelling was analyzed through gravimetry, and surface morphology was characterized by scanning electron microscopy. Viability of dental pulp stem cells seeded on the scaffolds was evaluated by live/dead analysis and DNA quantification. The results demonstrated successful copolymerization, and three formulations among various synthesized formulations were successfully 3D printed. Up to 35% degradability was confirmed within 7 days, and a maximum swelling of approximately 1200% was achieved. Furthermore, initial assessment of cell viability demonstrated biocompatibility of the developed scaffolds. While further studies are required to achieve the tissue engineering goals, the present results tend to indicate that the proposed hydrogel might be a valid candidate for scaffold fabrication serving dentoalveolar tissue engineering through 3D printing.

17.
Stem Cells Transl Med ; 13(3): 278-292, 2024 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-38217535

RESUMEN

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.


Asunto(s)
Cartílago , Ingeniería de Tejidos , Ratones , Animales , Ovinos , Ingeniería de Tejidos/métodos , Ratones Desnudos , Diferenciación Celular , Osteogénesis , Condrogénesis
18.
PLoS Comput Biol ; 8(10): e1002724, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23071433

RESUMEN

The healing of a fracture depends largely on the development of a new blood vessel network (angiogenesis) in the callus. During angiogenesis tip cells lead the developing sprout in response to extracellular signals, amongst which vascular endothelial growth factor (VEGF) is critical. In order to ensure a correct development of the vasculature, the balance between stalk and tip cell phenotypes must be tightly controlled, which is primarily achieved by the Dll4-Notch1 signaling pathway. This study presents a novel multiscale model of osteogenesis and sprouting angiogenesis, incorporating lateral inhibition of endothelial cells (further denoted MOSAIC model) through Dll4-Notch1 signaling, and applies it to fracture healing. The MOSAIC model correctly predicted the bone regeneration process and recapitulated many experimentally observed aspects of tip cell selection: the salt and pepper pattern seen for cell fates, an increased tip cell density due to the loss of Dll4 and an excessive number of tip cells in high VEGF environments. When VEGF concentration was even further increased, the MOSAIC model predicted the absence of a vascular network and fracture healing, thereby leading to a non-union, which is a direct consequence of the mutual inhibition of neighboring cells through Dll4-Notch1 signaling. This result was not retrieved for a more phenomenological model that only considers extracellular signals for tip cell migration, which illustrates the importance of implementing the actual signaling pathway rather than phenomenological rules. Finally, the MOSAIC model demonstrated the importance of a proper criterion for tip cell selection and the need for experimental data to further explore this. In conclusion, this study demonstrates that the MOSAIC model creates enhanced capabilities for investigating the influence of molecular mechanisms on angiogenesis and its relation to bone formation in a more mechanistic way and across different time and spatial scales.


Asunto(s)
Células Endoteliales/fisiología , Modelos Biológicos , Neovascularización Fisiológica/fisiología , Osteogénesis/fisiología , Animales , Callo Óseo/metabolismo , Biología Computacional , Simulación por Computador , Células Endoteliales/metabolismo , Curación de Fractura/fisiología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas de la Membrana/metabolismo , Ratones , Ratas , Receptor Notch1/metabolismo , Transducción de Señal , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
19.
J Mech Behav Biomed Mater ; 147: 106120, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37757617

RESUMEN

In fracture fixation, biodegradable implant materials are an interesting alternative to conventional non-biodegradable materials as the latter often require a second implant removal surgery to avoid long-term complications. In this study, we present an in silico strategy to design/study biodegradable metal implants focusing on mandibular fracture fixation plates of WE43 (Mg alloy). The in silico strategy is composed of an orchestrated interaction between three separate computational models. The first model simulates the mass loss of the degradable implant based on the chemistry of Mg biodegradation. A second model estimates the loading on the jaw plate in the physiological environment, incorporating a phenomenological dynamic bone regeneration process. The third model characterizes the mechanical behavior of the jaw plate and the influence of material degradation on the mechanical behavior. A sensitivity analysis was performed on parameters related to choices regarding numerical implementation and parameter dependencies were implemented to guarantee robust and correct results. Different clinical scenarios were tested, related to the amount of screws used to fix the plate. The results showed a lower initial strength when more screw holes were left open, as well as a faster decrease over time in strength due to the increased area available for surface degradation. The obtained degradation results were found to be in accordance with previously reported data of in vivo studies with biodegradable plates. The combination of these three models allows for the design of patient-specific biodegradable fixation implants able to deliver the desired mechanical behavior tuned to the bone regeneration process.


Asunto(s)
Fijación Interna de Fracturas , Mandíbula , Humanos , Fijación Interna de Fracturas/métodos , Fenómenos Biomecánicos , Mandíbula/cirugía , Placas Óseas , Tornillos Óseos , Implantes Absorbibles
20.
Artículo en Inglés | MEDLINE | ID: mdl-37819828

RESUMEN

The idea of a systematic digital representation of the entire known human pathophysiology, which we could call the Virtual Human Twin, has been around for decades. To date, most research groups focused instead on developing highly specialised, highly focused patient-specific models able to predict specific quantities of clinical relevance. While it has facilitated harvesting the low-hanging fruits, this narrow focus is, in the long run, leaving some significant challenges that slow the adoption of digital twins in healthcare. This position paper lays the conceptual foundations for developing the Virtual Human Twin (VHT). The VHT is intended as a distributed and collaborative infrastructure, a collection of technologies and resources (data, models) that enable it, and a collection of Standard Operating Procedures (SOP) that regulate its use. The VHT infrastructure aims to facilitate academic researchers, public organisations, and the biomedical industry in developing and validating new digital twins in healthcare solutions with the possibility of integrating multiple resources if required by the specific context of use. Healthcare professionals and patients can also use the VHT infrastructure for clinical decision support or personalised health forecasting. As the European Commission launched the EDITH coordination and support action to develop a roadmap for the development of the Virtual Human Twin, this position paper is intended as a starting point for the consensus process and a call to arms for all stakeholders.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA