Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Front Bioeng Biotechnol ; 11: 1145550, 2023.
Article in English | MEDLINE | ID: mdl-37362221

ABSTRACT

Sprouting angiogenesis is a core biological process critical to vascular development. Its accurate simulation, relevant to multiple facets of human health, is of broad, interdisciplinary appeal. This study presents an in-silico model replicating a microfluidic assay where endothelial cells sprout into a biomimetic extracellular matrix, specifically, a large-pore, low-concentration fibrin-based porous hydrogel, influenced by chemotactic factors. We introduce a novel approach by incorporating the extracellular matrix and chemotactic factor effects into a unified term using a single parameter, primarily focusing on modelling sprouting dynamics and morphology. This continuous model naturally describes chemotactic-induced sprouting with no need for additional rules. In addition, we extended our base model to account for matrix sensing and degradation, crucial aspects of angiogenesis. We validate our model via a hybrid in-silico experimental method, comparing the model predictions with experimental results derived from the microfluidic setup. Our results underscore the intricate relationship between the extracellular matrix structure and angiogenic sprouting, proposing a promising method for predicting the influence of the extracellular matrix on angiogenesis.

2.
J Nanobiotechnology ; 21(1): 115, 2023 Mar 29.
Article in English | MEDLINE | ID: mdl-36978078

ABSTRACT

BACKGROUND: The lack of predictive models that mimic the blood-brain barrier (BBB) hinders the development of effective drugs for neurodegenerative diseases. Animal models behave differently from humans, are expensive and have ethical constraints. Organ-on-a-chip (OoC) platforms offer several advantages to resembling physiological and pathological conditions in a versatile, reproducible, and animal-free manner. In addition, OoC give us the possibility to incorporate sensors to determine cell culture features such as trans-endothelial electrical resistance (TEER). Here, we developed a BBB-on-a-chip (BBB-oC) platform with a TEER measurement system in close distance to the barrier used for the first time for the evaluation of the permeability performance of targeted gold nanorods for theranostics of Alzheimer's disease. GNR-PEG-Ang2/D1 is a therapeutic nanosystem previously developed by us consisting of gold nanorods (GNR) functionalized with polyethylene glycol (PEG), angiopep-2 peptide (Ang2) to overcome the BBB and the D1 peptide as beta amyloid fibrillation inhibitor, finally obtaining GNR-PEG-Ang2/D1 which showed to be useful for disaggregation of the amyloid in in vitro and in vivo models. In this work, we evaluated its cytotoxicity, permeability, and some indications of its impact on the brain endothelium by employing an animal-free device based on neurovascular human cells. RESULTS: In this work, we fabricated a BBB-oC with human astrocytes, pericytes and endothelial cells and a TEER measuring system (TEER-BBB-oC) integrated at a micrometric distance of the endothelial barrier. The characterization displayed a neurovascular network and the expression of tight junctions in the endothelium. We produced GNR-PEG-Ang2/D1 and determined its non-cytotoxic range (0.05-0.4 nM) for plated cells included in the BBB-oC and confirmed its harmless effect at the highest concentration (0.4 nM) in the microfluidic device. The permeability assays revealed that GNR-PEG-Ang2/D1 cross the BBB and this entry is facilitated by Ang2 peptide. Parallel to the permeability analysis of GNR-PEG-Ang2/D1, an interesting behavior of the TJs expression was observed after its administration probably related to the ligands on the nanoparticle surface. CONCLUSIONS: BBB-oC with a novel TEER integrated setup which allow a correct read-out and cell imaging monitoring was proven as a functional and throughput platform to evaluate the brain permeability performance of nanotherapeutics in a physiological environment with human cells, putting forward a viable alternative to animal experimentation.


Subject(s)
Alzheimer Disease , Blood-Brain Barrier , Humans , Blood-Brain Barrier/metabolism , Endothelial Cells/metabolism , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Electric Impedance , Gold/pharmacology , Astrocytes/metabolism , Amyloid beta-Peptides/metabolism , Endothelium/metabolism , Permeability , Lab-On-A-Chip Devices
3.
Acta Biomater ; 151: 264-277, 2022 10 01.
Article in English | MEDLINE | ID: mdl-35981686

ABSTRACT

Most of the conventional in vitro models to test biomaterial-driven vascularization are too simplistic to recapitulate the complex interactions taking place in the actual cell microenvironment, which results in a poor prediction of the in vivo performance of the material. However, during the last decade, cell culture models based on microfluidic technology have allowed attaining unprecedented levels of tissue biomimicry. In this work, we propose a microfluidic-based 3D model to evaluate the effect of bioactive biomaterials capable of releasing signaling cues (such as ions or proteins) in the recruitment of endogenous endothelial progenitor cells, a key step in the vascularization process. The usability of the platform is demonstrated using experimentally-validated finite element models and migration and proliferation studies with rat endothelial progenitor cells (rEPCs) and bone marrow-derived rat mesenchymal stromal cells (BM-rMSCs). As a proof of concept of biomaterial evaluation, the response of rEPCs to an electrospun composite made of polylactic acid with calcium phosphates nanoparticles (PLA+CaP) was compared in a co-culture microenvironment with BM-rMSC to a regular PLA control. Our results show a significantly higher rEPCs migration and the upregulation of several pro-inflammatory and proangiogenic proteins in the case of the PLA+CaP. The effects of osteopontin (OPN) on the rEPCs migratory response were also studied using this platform, suggesting its important role in mediating their recruitment to a calcium-rich microenvironment. This new tool could be applied to screen the capacity of a variety of bioactive scaffolds to induce vascularization and accelerate the preclinical testing of biomaterials. STATEMENT OF SIGNIFICANCE: For many years researchers have used neovascularization models to evaluate bioactive biomaterials both in vitro, with low predictive results due to their poor biomimicry and minimal control over cell cues such as spatiotemporal biomolecule signaling, and in vivo models, presenting drawbacks such as being highly costly, time-consuming, poor human extrapolation, and ethically controversial. We describe a compact microphysiological platform designed for the evaluation of proangiogenesis in biomaterials through the quantification of the level of sprouting in a mimicked endothelium able to react to gradients of biomaterial-released signals in a fibrin-based extracellular matrix. This model is a useful tool to perform preclinical trustworthy studies in tissue regeneration and to better understand the different elements involved in the complex process of vascularization.


Subject(s)
Endothelial Progenitor Cells , Animals , Biocompatible Materials/metabolism , Biocompatible Materials/pharmacology , Calcium/metabolism , Calcium Phosphates/pharmacology , Fibrin/pharmacology , Humans , Microfluidics , Neovascularization, Physiologic , Osteopontin/metabolism , Polyesters/pharmacology , Rats , Tissue Engineering , Tissue Scaffolds
4.
Biofabrication ; 13(3)2021 06 01.
Article in English | MEDLINE | ID: mdl-33962409

ABSTRACT

The creation of cardiac tissue models for preclinical testing is still a non-solved problem in drug discovery, due to the limitations related to thein vitroreplication of cardiac tissue complexity. Among these limitations, the difficulty of mimicking the functional properties of the myocardium due to the immaturity of the used cells hampers the obtention of reliable results that could be translated into human patients.In vivomodels are the current gold standard to test new treatments, although it is widely acknowledged that the used animals are unable to fully recapitulate human physiology, which often leads to failures during clinical trials. In the present work, we present a microfluidic platform that aims to provide a range of signaling cues to immature cardiac cells to drive them towards an adult phenotype. The device combines topographical electrospun nanofibers with electrical stimulation in a microfabricated system. We validated our platform using a co-culture of neonatal mouse cardiomyocytes and cardiac fibroblasts, showing that it allows us to control the degree of anisotropy of the cardiac tissue inside the microdevice in a cost-effective way. Moreover, a 3D computational model of the electrical field was created and validated to demonstrate that our platform is able to closely match the distribution obtained with the gold standard (planar electrode technology) using inexpensive rod-shaped biocompatible stainless-steel electrodes. The functionality of the electrical stimulation was shown to induce a higher expression of the tight junction protein Cx-43, as well as the upregulation of several key genes involved in conductive and structural cardiac properties. These results validate our platform as a powerful tool for the tissue engineering community due to its low cost, high imaging compatibility, versatility, and high-throughput configuration capabilities.


Subject(s)
Electric Stimulation , Animals , Anisotropy , Humans , Mice , Myocytes, Cardiac , Nanofibers , Tissue Engineering
5.
J Mech Behav Biomed Mater ; 63: 456-469, 2016 10.
Article in English | MEDLINE | ID: mdl-27475947

ABSTRACT

PURPOSE: This work shows an effective methodology to characterize the creep-recovery behavior of silicones before their application in podiatry. The aim is to characterize, model and compare the creep-recovery properties of different types of silicone used in podiatry orthotics. METHODS: Creep-recovery phenomena of silicones used in podiatry orthotics is characterized by dynamic mechanical analysis (DMA). Silicones provided by Herbitas are compared by observing their viscoelastic properties by Functional Data Analysis (FDA) and nonlinear regression. The relationship between strain and time is modeled by fixed and mixed effects nonlinear regression to compare easily and intuitively podiatry silicones. RESULTS: Functional ANOVA and Kohlrausch-Willians-Watts (KWW) model with fixed and mixed effects allows us to compare different silicones observing the values of fitting parameters and their physical meaning. The differences between silicones are related to the variations of breadth of creep-recovery time distribution and instantaneous deformation-permanent strain. Nevertheless, the mean creep-relaxation time is the same for all the studied silicones. Silicones used in palliative orthoses have higher instantaneous deformation-permanent strain and narrower creep-recovery distribution. CONCLUSIONS: The proposed methodology based on DMA, FDA and nonlinear regression is an useful tool to characterize and choose the proper silicone for each podiatry application according to their viscoelastic properties.


Subject(s)
Materials Testing , Podiatry , Silicones/analysis , Elasticity , Orthotic Devices , Stress, Mechanical , Viscosity
SELECTION OF CITATIONS
SEARCH DETAIL
...