Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Front Immunol ; 14: 1231329, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38130715

RESUMEN

Bone fracture healing is a well-orchestrated but complex process that involves numerous regulations at different scales. This complexity becomes particularly evident during the inflammatory stage, as immune cells invade the healing region and trigger a cascade of signals to promote a favorable regenerative environment. Thus, the emergence of criticalities during this stage might hinder the rest of the process. Therefore, the investigation of the many interactions that regulate the inflammation has a primary importance on the exploration of the overall healing progression. In this context, an in silico model named COMMBINI (COmputational Model of Macrophage dynamics in the Bone INjury Immunoresponse) has been developed to investigate the mechano-biological interactions during the early inflammatory stage at the tissue, cellular and molecular levels. An agent-based model is employed to simulate the behavior of immune cells, inflammatory cytokines and fracture debris as well as their reciprocal multiscale biological interactions during the development of the early inflammation (up to 5 days post-injury). The strength of the computational approach is the capacity of the in silico model to simulate the overall healing process by taking into account the numerous hidden events that contribute to its success. To calibrate the model, we present an in silico immunofluorescence method that enables a direct comparison at the cellular level between the model output and experimental immunofluorescent images. The combination of sensitivity analysis and a Genetic Algorithm allows dynamic cooperation between these techniques, enabling faster identification of the most accurate parameter values, reducing the disparity between computer simulation and histological data. The sensitivity analysis showed a higher sensibility of the computer model to the macrophage recruitment ratio during the early inflammation and to proliferation in the late stage. Furthermore, the Genetic Algorithm highlighted an underestimation of macrophage proliferation by in vitro experiments. Further experiments were conducted using another externally fixated murine model, providing an independent validation dataset. The validated COMMBINI platform serves as a novel tool to deepen the understanding of the intricacies of the early bone regeneration phases. COMMBINI aims to contribute to designing novel treatment strategies in both the biological and mechanical domains.


Asunto(s)
Curación de Fractura , Modelos Biológicos , Ratones , Animales , Simulación por Computador , Macrófagos , Inflamación
2.
Front Bioeng Biotechnol ; 9: 703725, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34660547

RESUMEN

In silico modeling is a powerful strategy to investigate the biological events occurring at tissue, cellular and subcellular level during bone fracture healing. However, most current models do not consider the impact of the inflammatory response on the later stages of bone repair. Indeed, as initiator of the healing process, this early phase can alter the regenerative outcome: if the inflammatory response is too strongly down- or upregulated, the fracture can result in a non-union. This review covers the fundamental information on fracture healing, in silico modeling and experimental validation. It starts with a description of the biology of fracture healing, paying particular attention to the inflammatory phase and its cellular and subcellular components. We then discuss the current state-of-the-art regarding in silico models of the immune response in different tissues as well as the bone regeneration process at the later stages of fracture healing. Combining the aforementioned biological and computational state-of-the-art, continuous, discrete and hybrid modeling technologies are discussed in light of their suitability to capture adequately the multiscale course of the inflammatory phase and its overall role in the healing outcome. Both in the establishment of models as in their validation step, experimental data is required. Hence, this review provides an overview of the different in vitro and in vivo set-ups that can be used to quantify cell- and tissue-scale properties and provide necessary input for model credibility assessment. In conclusion, this review aims to provide hands-on guidance for scientists interested in building in silico models as an additional tool to investigate the critical role of the inflammatory phase in bone regeneration.

3.
Biomech Model Mechanobiol ; 20(4): 1627-1644, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-34047890

RESUMEN

Critical-sized bone defects are critical healing conditions that, if left untreated, often lead to non-unions. To reduce the risk, critical-sized bone defects are often treated with recombinant human BMP-2. Although enhanced bone tissue formation is observed when BMP-2 is administered locally to the defect, spatial and temporal distribution of callus tissue often differs from that found during regular bone healing or in defects treated differently. How this altered tissue patterning due to BMP-2 treatment is linked to mechano-biological principles at the cellular scale remains largely unknown. In this study, the mechano-biological regulation of BMP-2-treated critical-sized bone defect healing was investigated using a multiphysics multiscale in silico approach. Finite element and agent-based modeling techniques were combined to simulate healing within a critical-sized bone defect (5 mm) in a rat femur. Computer model predictions were compared to in vivo microCT data outcome of bone tissue patterning at 2, 4, and 6 weeks postoperation. In vivo, BMP-2 treatment led to complete healing through periosteal bone bridging already after 2 weeks postoperation. Computer model simulations showed that the BMP-2 specific tissue patterning can be explained by the migration of mesenchymal stromal cells to regions with a specific concentration of BMP-2 (chemotaxis). This study shows how computational modeling can help us to further understand the mechanisms behind treatment effects on compromised healing conditions as well as to optimize future treatment strategies.


Asunto(s)
Proteína Morfogenética Ósea 2/química , Regeneración Ósea/efectos de los fármacos , Quimiotaxis/efectos de los fármacos , Colágeno/química , Osteogénesis/efectos de los fármacos , Factor de Crecimiento Transformador beta/química , Cicatrización de Heridas/fisiología , Animales , Callo Óseo , Diferenciación Celular , Simulación por Computador , Fémur/efectos de los fármacos , Análisis de Elementos Finitos , Humanos , Técnicas In Vitro , Células Madre Mesenquimatosas/metabolismo , Ratas , Proteínas Recombinantes/química , Riesgo , Microtomografía por Rayos X
4.
OTA Int ; 4(2 Suppl)2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-37608858

RESUMEN

This manuscript summarizes presentations of a symposium on key considerations in design of biomechanical models at the 2019 Basic Science Focus Forum of the Orthopaedic Trauma Association. The first section outlines the most important characteristics of a high-quality biomechanical study. The second section considers choices associated with designing experiments using finite element modeling versus synthetic bones versus human specimens. The third section discusses appropriate selection of experimental protocols and finite element analyses. The fourth section considers the pros and cons of use of biomechanical research for implant design. Finally, the fifth section examines how results from biomechanical studies can be used when clinical evidence is lacking or contradictory. When taken together, these presentations emphasize the critical importance of biomechanical research and the need to carefully consider and optimize models when designing a biomechanical study.

5.
J Bone Miner Res ; 34(10): 1923-1937, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31121071

RESUMEN

Increasing age is associated with a reduced bone regeneration potential and increased risk of morbidities and mortality. A reduced bone formation response to mechanical loading has been shown with aging, and it remains unknown if the interplay between aging and mechanical stimuli during regeneration is similar to adaptation. We used a combined in vivo/in silico approach to investigate age-related alterations in the mechanical regulation of bone healing and identified the relative impact of altered cellular function on tissue patterns during the regenerative cascade. To modulate the mechanical environment, femoral osteotomies in adult and elderly mice were stabilized using either a rigid or a semirigid external fixator, and the course of healing was evaluated using histomorphometric and micro-CT analyses at 7, 14, and 21 days post-surgery. Computer models were developed to investigate the influence of the local mechanical environment within the callus on tissue formation patterns. The models aimed to identify the key processes at the cellular level that alter the mechanical regulation of healing with aging. Fifteen age-related biological alterations were investigated on two levels (adult and elderly) with a design of experiments setup. We show a reduced response to changes in fixation stability with age, which could be explained by reduced cellular mechanoresponse, simulated as alteration of the ranges of mechanical stimuli driving mesenchymal stem cell differentiation. Cellular mechanoresponse has been so far widely ignored as a therapeutic target in aged patients. Our data hint to mechanotherapeutics as a potential treatment to enhance bone healing in the elderly. © 2019 American Society for Bone and Mineral Research.


Asunto(s)
Envejecimiento/metabolismo , Fracturas del Fémur/metabolismo , Curación de Fractura , Mecanotransducción Celular , Envejecimiento/patología , Animales , Modelos Animales de Enfermedad , Femenino , Fracturas del Fémur/diagnóstico por imagen , Fracturas del Fémur/patología , Ratones , Microtomografía por Rayos X
6.
PLoS One ; 12(9): e0183755, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28873093

RESUMEN

Today, different implant designs exist in the market; however, there is not a clear understanding of which are the best implant design parameters to achieve mechanical optimal conditions. Therefore, the aim of this project was to investigate if the geometry of a commercial short stem hip prosthesis can be further optimized to reduce stress shielding effects and achieve better short-stemmed implant performance. To reach this aim, the potential of machine learning techniques combined with parametric Finite Element analysis was used. The selected implant geometrical parameters were: total stem length (L), thickness in the lateral (R1) and medial (R2) and the distance between the implant neck and the central stem surface (D). The results show that the total stem length was not the only parameter playing a role in stress shielding. An optimized implant should aim for a decreased stem length and a reduced length of the surface in contact with the bone. The two radiuses that characterize the stem width at the distal cross-section in contact with the bone were less influential in the reduction of stress shielding compared with the other two parameters; but they also play a role where thinner stems present better results.


Asunto(s)
Artroplastia de Reemplazo de Cadera/métodos , Artroplastia de Reemplazo/métodos , Prótesis de Cadera , Aprendizaje Automático , Algoritmos , Simulación por Computador , Toma de Decisiones , Fémur/cirugía , Análisis de Elementos Finitos , Humanos , Imagenología Tridimensional , Diseño de Prótesis , Programas Informáticos , Estrés Mecánico , Soporte de Peso
7.
Front Physiol ; 8: 287, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28533757

RESUMEN

Bone is a living part of the body that can, in most situations, heal itself after fracture. However, in some situations, healing may fail. Compromised conditions, such as large bone defects, aging, immuno-deficiency, or genetic disorders, might lead to delayed or non-unions. Treatment strategies for those conditions remain a clinical challenge, emphasizing the need to better understand the mechanisms behind endogenous bone regeneration. Bone healing is a complex process that involves the coordination of multiple events at different length and time scales. Computer models have been able to provide great insights into the interactions occurring within and across the different scales (organ, tissue, cellular, intracellular) using different modeling approaches [partial differential equations (PDEs), agent-based models, and finite element techniques]. In this review, we summarize the latest advances in computer models of bone healing with a focus on multiscale approaches and how they have contributed to understand the emergence of tissue formation patterns as a result of processes taking place at the lower length scales.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...