Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
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.
Math Biosci ; 359: 108997, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36996999

RESUMEN

Dysregulated inflammation underlies various diseases. Specialized pro-resolving mediators (SPMs) like Resolvin D1 (RvD1) have been shown to resolve inflammation and halt disease progression. Macrophages, key immune cells that drive inflammation, respond to the presence of RvD1 by polarizing to an anti-inflammatory type (M2). However, RvD1's mechanisms, roles, and utility are not fully understood. This paper introduces a gene-regulatory network (GRN) model that contains pathways for RvD1 and other SPMs and proinflammatory molecules like lipopolysaccharides. We couple this GRN model to a partial differential equation-agent-based hybrid model using a multiscale framework to simulate an acute inflammatory response with and without the presence of RvD1. We calibrate and validate the model using experimental data from two animal models. The model reproduces the dynamics of key immune components and the effects of RvD1 during acute inflammation. Our results suggest RvD1 can drive macrophage polarization through the G protein-coupled receptor 32 (GRP32) pathway. The presence of RvD1 leads to an earlier and increased M2 polarization, reduced neutrophil recruitment, and faster apoptotic neutrophil clearance. These results support a body of literature that suggests that RvD1 is a promising candidate for promoting the resolution of acute inflammation. We conclude that once calibrated and validated on human data, the model can identify critical sources of uncertainty, which could be further elucidated in biological experiments and assessed for clinical use.


Asunto(s)
Inflamación , Macrófagos , Animales , Humanos , Ácidos Docosahexaenoicos/farmacología , Ácidos Docosahexaenoicos/metabolismo
3.
Math Biosci ; 358: 108995, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36924879

RESUMEN

Nanoparticles (NPs) coated with peptide-major histocompatibility complexes (pMHCs) can be used as a therapy to treat autoimmune diseases. They do so by inducing the differentiation and expansion of disease-suppressing T regulatory type 1 (Tr1) cells by binding to their T cell receptors (TCRs) expressed as TCR-nanoclusters (TCRnc). Their efficacy can be controlled by adjusting NP size and number of pMHCs coated on them (referred to as valence). The binding of these NPs to TCRnc on T cells is thus polyvalent and occurs at three levels: the TCR-pMHC, NP-TCRnc and T cell levels. In this study, we explore how this polyvalent interaction is manifested and examine if it can facilitate T cell activation downstream. This is done by developing a multiscale biophysical model that takes into account the three levels of interactions and the geometrical complexity of the binding. Using the model, we quantify several key parameters associated with this interaction analytically and numerically, including the insertion probability that specifies the number of remaining pMHC binding sites in the contact area between T cells and NPs, the dwell time of interaction between NPs and TCRnc, carrying capacity of TCRnc, the distribution of covered and bound TCRs, and cooperativity in the binding of pMHCs within the contact area. The model was fit to previously published dose-response curves of interferon-γ obtained experimentally by stimulating a population of T cells with increasing concentrations of NPs at various valences and NP sizes. Exploring the parameter space of the model revealed that for an appropriate choice of the contact area angle, the model can produce moderate jumps between dose-response curves at low valences. This suggests that the geometry and kinetics of NP binding to TCRnc can act in synergy to facilitate T cell activation.


Asunto(s)
Nanopartículas , Receptores de Antígenos de Linfocitos T , Receptores de Antígenos de Linfocitos T/química , Receptores de Antígenos de Linfocitos T/metabolismo , Péptidos/metabolismo , Linfocitos T , Complejo Mayor de Histocompatibilidad , Unión Proteica
4.
Math Biosci Eng ; 19(11): 10941-10962, 2022 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-36124576

RESUMEN

Tumor hypoxia is commonly recognized as a condition stimulating the progress of the aggressive phenotype of tumor cells. Hypoxic tumor cells inhibit the delivery of cytotoxic drugs, causing hypoxic areas to receive insufficient amounts of anticancer agents, which results in adverse treatment responses. Being such an obstruction to conventional therapies for cancer, hypoxia might be considered a target to facilitate the efficacy of treatments in the resistive environment of tumor sites. In this regard, benefiting from prodrugs that selectively target hypoxic regions remains an effective approach. Additionally, combining hypoxia-activated prodrugs (HAPs) with conventional chemotherapeutic drugs has been used as a promising strategy to eradicate hypoxic cells. However, determining the appropriate sequencing and scheduling of the combination therapy is also of great importance in obtaining favorable results in anticancer therapy. Here, benefiting from a modeling approach, we study the efficacy of HAPs in combination with chemotherapeutic drugs on tumor growth and the treatment response. Different treatment schedules have been investigated to see the importance of determining the optimal schedule in combination therapy. The effectiveness of HAPs in varying hypoxic conditions has also been explored in the study. The model provides qualitative conclusions about the treatment response, as the maximal benefit is obtained from combination therapy with greater cell death for highly hypoxic tumors. It has also been observed that the antitumor effects of HAPs show a hypoxia-dependent profile.


Asunto(s)
Antineoplásicos , Neoplasias , Profármacos , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Humanos , Hipoxia/tratamiento farmacológico , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Profármacos/farmacología , Hipoxia Tumoral
5.
J Theor Biol ; 547: 111173, 2022 08 21.
Artículo en Inglés | MEDLINE | ID: mdl-35644484

RESUMEN

Solid tumour growth depends on a host of factors which affect the cell life cycle and extracellular matrix vascularization that leads to a favourable environment. The whole solid tumour can either grow or wither in response to the action of the immune system and therapeutics. A personalised mathematical model of such behaviour must consider both the intra- and inter-cellular dynamics and the mechanics of the solid tumour and its microenvironment. However, such wide range of spatial and temporal scales can hardly be modelled in a single model, and require the so-called multiscale models, defined as orchestrations of single-scale component models, connected by relation models that transform the data for one scale to another. While multiscale models are becoming common, there is a well-established engineering approach to the definition of the scale separation, e.g., how the spatiotemporal continuum is split in the various component models. In most studies scale separation is defined as natural, linked to anatomical concepts such as organ, tissue, or cell; but these do not provide reliable definition of scales: for examples skeletal organs can be as large as 500 mm (femur), or as small as 3 mm (stapes). Here we apply a recently proposed scale-separation approach based on the actual experimental and computational limitations to a patient-specific model of the growth of neuroblastoma. The resulting multiscale model can be properly informed with the available experimental data and solved in a reasonable timeframe with the available computational resources.


Asunto(s)
Modelos Biológicos , Neoplasias , Fenómenos Fisiológicos Celulares , Simulación por Computador , Matriz Extracelular/metabolismo , Humanos , Neoplasias/patología , Neovascularización Patológica/patología , Microambiente Tumoral
6.
Biomech Model Mechanobiol ; 21(2): 471-511, 2022 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-35000016

RESUMEN

Potts shunt (PS) was suggested as palliation for patients with suprasystemic pulmonary arterial hypertension (PAH) and right ventricular (RV) failure. PS, however, can result in poorly understood mortality. Here, a patient-specific geometrical multiscale model of PAH physiology and PS is developed for a paediatric PAH patient with stent-based PS. In the model, 7.6mm-diameter PS produces near-equalisation of the aortic and PA pressures and [Formula: see text] (oxygenated vs deoxygenated blood flow) ratio of 0.72 associated with a 16% decrease of left ventricular (LV) output and 18% increase of RV output. The flow from LV to aortic arch branches increases by 16%, while LV contribution to the lower body flow decreases by 29%. Total flow in the descending aorta (DAo) increases by 18% due to RV contribution through the PS with flow into the distal PA branches decreasing. PS induces 18% increase of RV work due to its larger stroke volume pumped against lower afterload. Nonetheless, larger RV work does not lead to increased RV end-diastolic volume. Three-dimensional flow assessment demonstrates the PS jet impinging with a high velocity and wall shear stress on the opposite DAo wall with the most of the shunt flow being diverted to the DAo. Increasing the PS diameter from 5mm up to 10mm results in a nearly linear increase in post-operative shunt flow and a nearly linear decrease in shunt pressure-drop. In conclusion, this model reasonably represents patient-specific haemodynamics pre- and post-creation of the PS, providing insights into physiology of this complex condition, and presents a predictive tool that could be useful for clinical decision-making regarding suitability for PS in PAH patients with drug-resistant suprasystemic PAH.


Asunto(s)
Hipertensión Pulmonar , Arteria Pulmonar , Niño , Hemodinámica , Humanos , Cuidados Paliativos , Stents
7.
Biomech Model Mechanobiol ; 20(6): 2179-2202, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34476656

RESUMEN

The lymphatics maintain fluid balance by returning interstitial fluid to veins via contraction/compression of vessel segments with check valves. Disruption of lymphatic pumping can result in a condition called lymphedema with interstitial fluid accumulation. Lymphedema treatments are often ineffective, which is partially attributable to insufficient understanding of specialized lymphatic muscle lining the vessels. This muscle exhibits cardiac-like phasic contractions and smooth muscle-like tonic contractions to generate and regulate flow. To understand the relationship between this sub-cellular contractile machinery and organ-level pumping, we have developed a multiscale computational model of phasic and tonic contractions in lymphatic muscle and coupled it to a lymphangion pumping model. Our model uses the sliding filament model (Huxley in Prog Biophys Biophys Chem 7:255-318, 1957) and its adaptation for smooth muscle (Mijailovich in Biophys J 79(5):2667-2681, 2000). Multiple structural arrangements of contractile components and viscoelastic elements were trialed but only one provided physiologic results. We then coupled this model with our previous lumped parameter model of the lymphangion to relate results to experiments. We show that the model produces similar pressure, diameter, and flow tracings to experiments on rat mesenteric lymphatics. This model provides the first estimates of lymphatic muscle contraction energetics and the ability to assess the potential effects of sub-cellular level phenomena such as calcium oscillations on lymphangion outflow. The maximum efficiency value predicted (40%) is at the upper end of estimates for other muscle types. Spontaneous calcium oscillations during diastole were found to increase outflow up to approximately 50% in the range of frequencies and amplitudes tested.


Asunto(s)
Sistema Linfático/fisiología , Modelos Biológicos , Animales , Calcio/metabolismo , Células Musculares/fisiología , Contracción Muscular/fisiología , Miosinas/metabolismo , Presión , Ratas , Troponina C/metabolismo
8.
AAPS J ; 23(5): 102, 2021 08 25.
Artículo en Inglés | MEDLINE | ID: mdl-34435271

RESUMEN

Mathematical modeling has been an important tool in pharmaceutical research for 50 + years and there is increased emphasis over the last decade on using modeling to improve the efficiency and effectiveness of drug development. In an earlier commentary, we applied a multiscale model linking 6 scales (whole body, tumor, vasculature, cell, spatial location, time), together with literature data on nanoparticle and tumor properties, to demonstrate the effects of nanoparticle particles on systemic disposition. The current commentary used a 4-scale model (cell membrane, intracellular organelles, spatial location, time) together with literature data on the intracellular processing of membrane receptors and transporters to demonstrate disruption of transporter homeostasis can lead to drug-drug interaction (DDI) between victim drug (VD) and perpetrator drug (PD), including changes in the area-under-concentration-time-curve of VD in cells that are considered significant by the US Food and Drug Administration (FDA). The model comprised 3 computational components: (a) intracellular transporter homeostasis, (b) pharmacokinetics of extracellular and intracellular VD/PD concentrations, and (c) pharmacodynamics of PD-induced stimulation or inhibition of an intracellular kinetic process. Model-based simulations showed that (a) among the five major endocytic processes, perturbation of transporter internalization or recycling led to the highest incidence and most extensive DDI, with minor DDI for perturbing transporter synthesis and early-to-late endosome and no DDI for perturbing transporter degradation and (b) three experimental conditions (spatial transporter distribution in cells, VD/PD co-incubation time, extracellular PD concentrations) were determinants of DDI detection. We propose modeling is a useful tool for hypothesis generation and for designing experiments to identify potential DDI; its application further aligns with the model-informed drug development paradigm advocated by FDA.


Asunto(s)
Desarrollo de Medicamentos/métodos , Interacciones Farmacológicas , Transportador 1 de Anión Orgánico Específico del Hígado/metabolismo , Modelos Biológicos , Miembro 1B3 de la Familia de los Transportadores de Solutos de Aniones Orgánicos/metabolismo , Área Bajo la Curva , Simulación por Computador , Hepatocitos/metabolismo , Homeostasis , Humanos
9.
Front Physiol ; 12: 503687, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33613304

RESUMEN

Clinically, fractional flow reserve (FFR)-guided coronary artery bypass grafting (CABG) is more effective than CABG guided by coronary angiography alone. However, no scholars have explained the mechanism from the perspective of hemodynamics. Two patients were clinically selected; their angiography showed 70% coronary stenosis, and the FFRs were 0.7 (patient 1) and 0.95 (patient 2). The FFR non-invasive computational model of the two patients was constructed by a 0-3D coupled multiscaled model, in order to verify that the model can accurately calculate the FFR results. Virtual bypass surgery was performed on these two stenoses, and a CABG multiscaled model was constructed. The flow rate of the graft and the stenosis coronary artery, as well as the wall shear stress (WSS) and the oscillatory shear index (OSI) in the graft were calculated. The non-invasive calculation results of FFR are 0.67 and 0.91, which are close to the clinical results, which proves that our model is accurate. According to the CABG model, the flow ratios of the stenosis coronary artery to the graft of patient 1 and patient 2 were 0.12 and 0.42, respectively. The time-average wall shear stress (TAWSS) results of patient 1 and patient 2 grafts were 2.09 and 2.16 Pa, respectively, and WSS showed uniform distribution on the grafts. The OSI results of patients 1 and 2 grafts were 0.0375 and 0.1264, respectively, and a significantly high OSI region appeared at the anastomosis of patient 2. The FFR value of the stenosis should be considered when performing bypass surgery. When the stenosis of high FFR values is grafted, a high OSI region is created at the graft, especially at the anastomosis. In the long term, this can cause anastomotic blockage and graft failure.

10.
In Silico Biol ; 14(3-4): 71-88, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-35001886

RESUMEN

Vascular endothelial growth factor (VEGF) has been known as a key mediator of angiogenesis in cancer. Bevacizumab is anti-VEGF monoclonal antibody that has been approved by the FDA as a first-line treatment in many types of cancer. In this paper, we extend a previously validated multiscale tumor model to comprehensively include the multiple roles of VEGF during the course of angiogenesis and its binding mechanism with bevacizumab. We use the model to simulate tumor system response under various bevacizumab concentrations, both in stand-alone treatment and in combination with chemotherapy. Our simulation indicates that periodic administration of bevacizumab with lower concentration can achieve greater efficacy than a single treatment with higher concentration. The simulation of the combined therapy also shows that the continuous administration of bevacizumab during the maintenance phase can lead to antitumor activity which further suppresses its growth. Agreement with experimental results indicates the potential of the model in predicting the efficacy of anti-VEGF therapies and could therefore contribute to developing prospective clinical trials.

11.
J Infect Public Health ; 13(10): 1438-1445, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32773211

RESUMEN

OBJECTIVE: This study retrospectively examined the health and social determinants of the COVID-19 outbreak in 175 countries from a spatial epidemiological approach. METHODS: We used spatial analysis to examine the cross-national determinants of confirmed cases of COVID-19 based on the World Health Organization official COVID-19 data and the World Bank Indicators of Interest to the COVID-19 outbreak. All models controlled for COVID-19 government measures. RESULTS: The percentage of the population age between 15-64 years (Age15-64), percentage smokers (SmokTot.), and out-of-pocket expenditure (OOPExp) significantly explained global variation in the current COVID-19 outbreak in 175 countries. The percentage population age group 15-64 and out of pocket expenditure were positively associated with COVID-19. Conversely, the percentage of the total population who smoke was inversely associated with COVID-19 at the global level. CONCLUSIONS: This study is timely and could serve as a potential geospatial guide to developing public health and epidemiological surveillance programs for the outbreak in multiple countries. Removal of catastrophic medical expenditure, smoking cessation, and observing public health guidelines will not only reduce illness related to COVID-19 but also prevent unecessary deaths.


Asunto(s)
Infecciones por Coronavirus/epidemiología , Infecciones por Coronavirus/prevención & control , Pandemias/prevención & control , Neumonía Viral/epidemiología , Neumonía Viral/prevención & control , Adolescente , Adulto , Factores de Edad , Betacoronavirus , COVID-19 , Bases de Datos Factuales , Gastos en Salud/estadística & datos numéricos , Humanos , Persona de Mediana Edad , Modelos Estadísticos , Estudios Retrospectivos , SARS-CoV-2 , Fumar/epidemiología , Regresión Espacial , Adulto Joven
12.
Front Immunol ; 11: 620716, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33613551

RESUMEN

Germinal centers play a key role in the adaptive immune system since they are able to produce memory B cells and plasma cells that produce high affinity antibodies for an effective immune protection. The mechanisms underlying cell-fate decisions are not well understood but asymmetric division of antigen, B-cell receptor affinity, interactions between B-cells and T follicular helper cells (triggering CD40 signaling), and regulatory interactions of transcription factors have all been proposed to play a role. In addition, a temporal switch from memory B-cell to plasma cell differentiation during the germinal center reaction has been shown. To investigate if antigen affinity-based Tfh cell help recapitulates the temporal switch we implemented a multiscale model that integrates cellular interactions with a core gene regulatory network comprising BCL6, IRF4, and BLIMP1. Using this model we show that affinity-based CD40 signaling in combination with asymmetric division of B-cells result in switch from memory B-cell to plasma cell generation during the course of the germinal center reaction. We also show that cell fate division is unlikely to be (solely) based on asymmetric division of Ag but that BLIMP1 is a more important factor. Altogether, our model enables to test the influence of molecular modulations of the CD40 signaling pathway on the production of germinal center output cells.


Asunto(s)
Linfocitos B/inmunología , Antígenos CD40/inmunología , Simulación por Computador , Centro Germinal/inmunología , Memoria Inmunológica/inmunología , Linfopoyesis/inmunología , Modelos Inmunológicos , Células Plasmáticas/inmunología , Células T Auxiliares Foliculares/inmunología , División Celular Asimétrica , Linfocitos B/citología , Linaje de la Célula , Redes Reguladoras de Genes , Centro Germinal/citología , Humanos , Factores Reguladores del Interferón/genética , Factores Reguladores del Interferón/fisiología , Células Plasmáticas/citología , Factor 1 de Unión al Dominio 1 de Regulación Positiva/genética , Factor 1 de Unión al Dominio 1 de Regulación Positiva/fisiología , Proteínas Proto-Oncogénicas c-bcl-6/genética , Proteínas Proto-Oncogénicas c-bcl-6/fisiología , Transducción de Señal , Factores de Tiempo
13.
Biomech Model Mechanobiol ; 19(2): 577-590, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-31571083

RESUMEN

Mathematical models can provide a quantitatively sophisticated description of tumor cell (TC) behaviors under mechanical microenvironment and help us better understand the role of specific biophysical factors based on their influences on the TC behaviors. To this end, we propose an off-lattice cell-based multiscale mathematical model to describe the dynamic growth-induced solid stress during tumor progression and investigate the influence of the mechanical microenvironment on TC invasion. At the cellular level, cell-cell and cell-matrix interactive forces depend on the mechanical properties of the cells and the cancer-associated fibroblasts in the stroma, respectively. The constitutive relationship between the interactive forces and cell migrations obeys the Hooke's law and damping effects. At the tissue level, the integrated growth-induced forces caused by proliferating cells within the simulation region are balanced by the external forces applied by the surrounding host tissues. Then, the cell movements are calculated according to the Newton's second law of motion, and the morphology of TC invasion is updated. The simulation results reveal the continuous changes of the macroscopic mechanical forces due to the interactions among the structural components and the microscopic environmental factors. Moreover, the simulation results demonstrate the adverse effect of the stiffness of tumor tissue on tumor growth and invasion. A decrease in the stiffness of tumor and matrix can promote TCs to proliferate at a much faster rate and invade into the surrounding healthy tissue more easily, whereas an increase in the stiffness can lead to an aggressive morphology of tumor invasion. We envision that the proposed model can be served as a quantitative theoretical platform to study the underlying biophysical role of the mechanical microenvironmental factors during tumor invasion and metastasis.


Asunto(s)
Neoplasias/patología , Estrés Mecánico , Microambiente Tumoral , Algoritmos , Fenómenos Biomecánicos , Comunicación Celular , Movimiento Celular , Proliferación Celular , Matriz Extracelular/metabolismo , Humanos , Modelos Biológicos , Invasividad Neoplásica
14.
Int J Mol Sci ; 20(23)2019 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-31801239

RESUMEN

Many biological processes are triggered or driven by mechanical forces in the cytoskeletal network, but these transducing forces have rarely been assessed. Striated muscle, with its well-organized structure provides an opportunity to assess intracellular forces using small-angle X-ray fiber diffraction. We present a new methodology using Monte Carlo simulations of muscle contraction in an explicit 3D sarcomere lattice to predict the fiber deformations and length changes along thin filaments during contraction. Comparison of predicted diffraction patterns to experimental meridional X-ray reflection profiles allows assessment of the stepwise changes in intermonomer spacings and forces in the myofilaments within living muscle cells. These changes along the filament length reflect the effect of forces from randomly attached crossbridges. This approach enables correlation of the molecular events, such as the current number of attached crossbridges and the distributions of crossbridge forces to macroscopic measurements of force and length changes during muscle contraction. In addition, assessments of fluctuations in local forces in the myofilaments may reveal how variations in the filament forces acting on signaling proteins in the sarcomere M-bands and Z-discs modulate gene expression, protein synthesis and degradation, and as well to mechanisms of adaptation of muscle in response to changes in mechanical loading.


Asunto(s)
Citoesqueleto de Actina/fisiología , Actinas/fisiología , Contracción Isométrica/fisiología , Músculo Estriado/fisiología , Miosinas/fisiología , Sarcómeros/fisiología , Citoesqueleto de Actina/ultraestructura , Actinas/ultraestructura , Animales , Simulación por Computador , Conectina/fisiología , Conectina/ultraestructura , Modelos Biológicos , Método de Montecarlo , Músculo Estriado/diagnóstico por imagen , Miosinas/ultraestructura , Rana catesbeiana/fisiología , Sarcómeros/ultraestructura , Dispersión del Ángulo Pequeño , Técnicas de Cultivo de Tejidos , Difracción de Rayos X
15.
Math Biosci Eng ; 16(6): 6257-6273, 2019 07 04.
Artículo en Inglés | MEDLINE | ID: mdl-31698561

RESUMEN

Tumour hypoxia has been associated with increased resistance to various cancer treatments, particularly radiation therapy. Conversely, tumour hypoxia is a validated and ideal target for guided cancer drug delivery. For this reason, hypoxia-activated prodrugs (HAPs) have been developed, which remain inactive in the body until in the presence of tissue hypoxia, allowing for an activation tendency in hypoxic regions. We present here an experimentally motivated mathematical model predicting the effectiveness of HAPs in a variety of clinical settings. We first examined HAP effectiveness as a function of the amount of tumour hypoxia and showed that the drugs have a larger impact on tumours with high levels of hypoxia. We then combined HAP treatment with radiation to examine the effects of combination therapies. Our results showed radiation-HAP combination therapies to be more effective against highly hypoxic tumours. The analysis of combination therapies was extended to consider schedule sequencing of the combination treatments. These results suggested that administering HAPs before radiation was most effective in reducing total cell number. Finally, a sensitivity analysis of the drug-related parameters was done to examine the effect of drug diffusivity and enzyme abundance on the overall effectiveness of the drug. Altogether, the results highlight the importance of the knowledge of tumour hypoxia levels before administration of HAPs in order to ensure positive results.


Asunto(s)
Quimioradioterapia/métodos , Neoplasias/tratamiento farmacológico , Neoplasias/radioterapia , Profármacos/farmacología , Hipoxia Tumoral , Animales , Calibración , Carcinoma de Pulmón de Células no Pequeñas/terapia , Línea Celular Tumoral , Simulación por Computador , Humanos , Neoplasias Pulmonares/terapia , Ratones , Ratones Desnudos , Modelos Teóricos , Trasplante de Neoplasias , Ratas , Rabdomiosarcoma/terapia , Programas Informáticos
16.
In Silico Biol ; 13(1-2): 1-20, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-29226860

RESUMEN

Angiogenesis, a formation of blood vessels from an existing vasculature, plays a key role in tumor growth and its progression into cancer. The lining of blood vessels consists of endothelial cells (ECs) which proliferate and migrate, allowing the capillaries to sprout towards the tumor to deliver the needed oxygen. Various treatments aiming to suppress or even inhibit angiogenesis have been explored. Mesenchymal stem cells (MSCs) have recently been undergoing development in cell-based therapy for cancer due to their ability to migrate towards the capillaries and induce the apoptosis of the ECs, causing capillary degeneration. However, further investigations in this direction are needed as it is usually difficult to preclinically assess the efficacy of such therapy. We develop a hybrid multiscale model that integrates molecular, cellular, tissue and extracellular components of tumor system to investigate angiogenesis and tumor growth under MSC-mediated therapy. Our simulations produce angiogenesis and vascular tumor growth profiles as observed in the experiments. Furthermore, the simulations show that the effectiveness of MSCs in inducing EC apoptosis is density dependent and its full effect is reached within several days after MSCs application. Quantitative agreements with experimental data indicate the predictive potential of our model for evaluating the efficacy of cell-based therapies targeting angiogenesis.


Asunto(s)
Tratamiento Basado en Trasplante de Células y Tejidos , Modelos Biológicos , Neoplasias/patología , Neoplasias/terapia , Neovascularización Patológica/terapia , Algoritmos , Animales , Apoptosis , Diferenciación Celular , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Células Cultivadas , Simulación por Computador , Células Endoteliales/citología , Células Endoteliales/metabolismo , Humanos , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Neoplasias/etiología , Fenotipo , Transducción de Señal
17.
J Theor Biol ; 454: 253-267, 2018 10 07.
Artículo en Inglés | MEDLINE | ID: mdl-29909142

RESUMEN

Tumour recurrence post chemotherapy is an established clinical problem and many cancer types are often observed to be increasingly drug resistant subsequent to chemotherapy treatments. Drug resistance in cancer is a multipart phenomenon which can be derived from several origins and in many cases it has been observed that cancer cells have the ability to possess, acquire and communicate drug resistant traits. Here, an in silico framework is developed in order to study drug resistance and drug response in cancer cell populations exhibiting various drug resistant features. The framework is based on an on-lattice hybrid multiscale mathematical model and is equipped to simulate multiple mechanisms on different scales that contribute towards chemotherapeutic drug resistance in cancer. This study demonstrates how drug resistant tumour features may depend on the interplay amongst intracellular, extracelluar and intercellular factors. On a cellular level, drug resistant cell phenotypes are here derived from inheritance or mutations that are spontaneous, drug-induced or communicated via exosomes. Furthermore intratumoural heterogeneity and spatio-temporal drug dynamics heavily influences drug delivery and the development of drug resistant cancer cell subpopulations. Chemotherapy treatment strategies are here optimised for various in silico tumour scenarios and treatment objectives. We demonstrate that optimal chemotherapy treatment strategies drastically depend on which drug resistant mechanisms are activated, and that furthermore suboptimal chemotherapy administration may promote drug resistance.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Resistencia a Antineoplásicos , Modelos Teóricos , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Recuento de Células , Ciclo Celular/efectos de los fármacos , Muerte Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Simulación por Computador , Progresión de la Enfermedad , Relación Dosis-Respuesta a Droga , Exosomas/fisiología , Humanos , Modelos Biológicos , Células Tumorales Cultivadas , Microambiente Tumoral/fisiología
18.
Med Eng Phys ; 57: 40-50, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29753628

RESUMEN

Part of clinically applicable bone graft substitutes are developed by using mechanical stimulation of flow-perfusion into cell-seeded scaffolds. The role of fluid flow is crucial in driving the nutrient to seeded cells and in stimulating cell colonization. A common numerical approach is to use a multiscale model to link some physical quantities (wall shear stress and inlet flow rate) that act at different scales. In this study, a multiscale model is developed in order to determine the optimal inlet flow rate to cultivate osteoblast-like cells seeded in a controlled macroporous biomaterial inside a perfusion bioreactor system. We focus particularly on the influence of Wall Shear Stress on cell colonization to predict cell colonization at the macroscale. Results obtained at the microscale are interpolated at the macroscale to determine the optimal flow rate. For a macroporous scaffold made of interconnected pores with pore diameters of above 350 µm and interconnection diameters of 150 µm, the model predicts a cell colonization of 325% after a 7-day-cell culture with a constant inlet flow rate of 0.69 mL·min-1. Furthermore, the strength of this protocol is the possibility to adapt it to most porous biomaterials and dynamic cell culture systems.


Asunto(s)
Reactores Biológicos , Trasplante Óseo , Modelos Biológicos , Materiales Biocompatibles , Proliferación Celular , Hidrodinámica , Perfusión , Porosidad , Estrés Mecánico
19.
J Theor Biol ; 446: 87-100, 2018 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-29524441

RESUMEN

If improvements are to be made in tuberculosis (TB) treatment, an increased understanding of disease in the lung is needed. Studies have shown that bacteria in a less metabolically active state, associated with the presence of lipid bodies, are less susceptible to antibiotics, and recent results have highlighted the disparity in concentration of different compounds into lesions. Treatment success therefore depends critically on the responses of the individual bacteria that constitute the infection. We propose a hybrid, individual-based approach that analyses spatio-temporal dynamics at the cellular level, linking the behaviour of individual bacteria and host cells with the macroscopic behaviour of the microenvironment. The individual elements (bacteria, macrophages and T cells) are modelled using cellular automaton (CA) rules, and the evolution of oxygen, drugs and chemokine dynamics are incorporated in order to study the effects of the microenvironment in the pathological lesion. We allow bacteria to switch states depending on oxygen concentration, which affects how they respond to treatment. This is the first multiscale model of its type to consider both oxygen-driven phenotypic switching of the Mycobacterium tuberculosis and antibiotic treatment. Using this model, we investigate the role of bacterial cell state and of initial bacterial location on treatment outcome. We demonstrate that when bacteria are located further away from blood vessels, less favourable outcomes are more likely, i.e. longer time before infection is contained/cleared, treatment failure or later relapse. We also show that in cases where bacteria remain at the end of simulations, the organisms tend to be slower-growing and are often located within granulomas, surrounded by caseous material.


Asunto(s)
Antibacterianos/uso terapéutico , Granuloma , Modelos Biológicos , Mycobacterium tuberculosis/metabolismo , Tuberculosis Pulmonar , Granuloma/tratamiento farmacológico , Granuloma/metabolismo , Granuloma/microbiología , Humanos , Tuberculosis Pulmonar/tratamiento farmacológico , Tuberculosis Pulmonar/metabolismo
20.
Math Models Methods Appl Sci ; 28(1): 61-93, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-29353950

RESUMEN

Cancer results from a complex interplay of different biological, chemical, and physical phenomena that span a wide range of time and length scales. Computational modeling may help to unfold the role of multiple evolving factors that exist and interact in the tumor microenvironment. Understanding these complex multiscale interactions is a crucial step towards predicting cancer growth and in developing effective therapies. We integrate different modeling approaches in a multiscale, avascular, hybrid tumor growth model encompassing tissue, cell, and sub-cell scales. At the tissue level, we consider the dispersion of nutrients and growth factors in the tumor microenvironment, which are modeled through reaction-diffusion equations. At the cell level, we use an agent based model (ABM) to describe normal and tumor cell dynamics, with normal cells kept in homeostasis and cancer cells differentiated apoptotic, hypoxic, and necrotic states. Cell movement is driven by the balance of a variety of forces according to Newton's second law, including those related to growth-induced stresses. Phenotypic transitions are defined by specific rule of behaviors that depend on microenvironment stimuli. We integrate in each cell/agent a branch of the epidermal growth factor receptor (EGFR) pathway. This pathway is modeled by a system of coupled nonlinear differential equations involving the mass laws of 20 molecules. The rates of change in the concentration of some key molecules trigger proliferation or migration advantage response. The bridge between cell and tissue scales is built through the reaction and source terms of the partial differential equations. Our hybrid model is built in a modular way, enabling the investigation of the role of different mechanisms at multiple scales on tumor progression. This strategy allows representating both the collective behavior due to cell assembly as well as microscopic intracellular phenomena described by signal transduction pathways. Here, we investigate the impact of some mechanisms associated with sustained proliferation on cancer progression. Specifically, we focus on the intracellular proliferation/migration-advantage-response driven by the EGFR pathway and on proliferation inhibition due to accumulation of growth-induced stresses. Simulations demonstrate that the model can adequately describe some complex mechanisms of tumor dynamics, including growth arrest in avascular tumors. Both the sub-cell model and growth-induced stresses give rise to heterogeneity in the tumor expansion and a rich variety of tumor behaviors.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA