Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
Add more filters










Publication year range
1.
Adv Sci (Weinh) ; : e2305555, 2024 Apr 18.
Article in English | MEDLINE | ID: mdl-38634605

ABSTRACT

Bioprinting technology offers unprecedented opportunities to construct in vitro tissue models that recapitulate the 3D morphology and functionality of native tissue. Yet, it remains difficult to obtain adequate functional readouts from such models. In particular, it is challenging to position sensors in desired locations within pre-fabricated 3D bioprinted structures. At the same time, bioprinting tissue directly onto a sensing device is not feasible due to interference with the printer head. As such, a multi-sensing platform inspired by origami that overcomes these challenges by "folding" around a separately fabricated 3D tissue structure is proposed, allowing for the insertion of electrodes into precise locations, which are custom-defined using computer-aided-design software. The multi-sensing origami platform (MSOP) can be connected to a commercial multi-electrode array (MEA) system for data-acquisition and processing. To demonstrate the platform, how integrated 3D MEA electrodes can record neuronal electrical activity in a 3D model of a neurovascular unit is shown. The MSOP also enables a microvascular endothelial network to be cultured separately and integrated with the 3D tissue structure. Accordingly, how impedance-based sensors in the platform can measure endothelial barrier function is shown. It is further demonstrated the device's versatility by using it to measure neuronal activity in brain organoids.

2.
APL Bioeng ; 7(3): 031506, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37781727

ABSTRACT

Implantable sensors have revolutionized the way we monitor biophysical and biochemical parameters by enabling real-time closed-loop intervention or therapy. These technologies align with the new era of healthcare known as healthcare 5.0, which encompasses smart disease control and detection, virtual care, intelligent health management, smart monitoring, and decision-making. This review explores the diverse biomedical applications of implantable temperature, mechanical, electrophysiological, optical, and electrochemical sensors. We delve into the engineering principles that serve as the foundation for their development. We also address the challenges faced by researchers and designers in bridging the gap between implantable sensor research and their clinical adoption by emphasizing the importance of careful consideration of clinical requirements and engineering challenges. We highlight the need for future research to explore issues such as long-term performance, biocompatibility, and power sources, as well as the potential for implantable sensors to transform healthcare across multiple disciplines. It is evident that implantable sensors have immense potential in the field of medical technology. However, the gap between research and clinical adoption remains wide, and there are still major obstacles to overcome before they can become a widely adopted part of medical practice.

3.
Biomed Opt Express ; 14(10): 5223-5237, 2023 Oct 01.
Article in English | MEDLINE | ID: mdl-37854575

ABSTRACT

The development of organs-on-a-chip platforms has revolutionized in-vitro cellular culture by allowing cells to be grown in an environment that better mimics human physiology. However, there is still a challenge in integrating those platforms with advanced imaging technology. This is extremely important when we want to study molecular changes and subcellular processes on the level of a single molecule using super-resolution microscopy (SRM), which has a resolution beyond the diffraction limit of light. Currently, existing platforms that include SRM have certain limitations, either as they only support 2D monocultures, without flow or as they demand a lot of production and handling. In this study, we developed a Super-Res-Chip platform, consisting of a 3D-printed chip and a porous membrane, that could be used to co-culture cells in close proximity either in 2D or in 3D while allowing SRM on both sides of the membrane. To demonstrate the functionality of the device, we co-cultured in endothelial and epithelial cells and used direct stochastic optical reconstruction microscopy (dSTORM) to investigate how glioblastoma cells affect the expression of the gap-junction protein Connexin43 in endothelial cells grown in 2D and in 3D. Cluster analysis of Connexin43 distribution revealed no difference in the number of clusters, their size, or radii, but did identify differences in their density. Furthermore, the spatial resolution was high also when the cells were imaged through the membrane (20-30 nm for x-y) and 10-20 nm when imaged directly both for 2D and 3D conditions. Overall, this chip allows to characterize of complex cellular processes on a molecular scale in an easy manner and improved the capacity for imaging in a single molecule resolution complex cellular organization.

4.
Adv Mater ; 35(51): e2304654, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37753928

ABSTRACT

Monoclonal antibodies (mAbs) hold promise in treating Parkinson's disease (PD), although poor delivery to the brain hinders their therapeutic application. In the current study, it is demonstrated that brain-targeted liposomes (BTL) enhance the delivery of mAbs across the blood-brain-barrier (BBB) and into neurons, thereby allowing the intracellular and extracellular treatment of the PD brain. BTL are decorated with transferrin to improve brain targeting through overexpressed transferrin-receptors on the BBB during PD. BTL are loaded with SynO4, a mAb that inhibits alpha-synuclein (AS) aggregation, a pathological hallmark of PD. It is shown that 100-nm BTL cross human BBB models intact and are taken up by primary neurons. Within neurons, SynO4 is released from the nanoparticles and bound to its target, thereby reducing AS aggregation, and enhancing neuronal viability. In vivo, intravenous BTL administration results in a sevenfold increase in mAbs in brain cells, decreasing AS aggregation and neuroinflammation. Treatment with BTL also improve behavioral motor function and learning ability in mice, with a favorable safety profile. Accordingly, targeted nanotechnologies offer a valuable platform for drug delivery to treat brain neurodegeneration.


Subject(s)
Parkinson Disease , Animals , Humans , Mice , alpha-Synuclein/metabolism , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Behavioral Symptoms , Brain/metabolism , Liposomes/metabolism , Parkinson Disease/drug therapy , Transferrins
5.
Neurotrauma Rep ; 4(1): 560-572, 2023.
Article in English | MEDLINE | ID: mdl-37636339

ABSTRACT

Traumatic brain injury (TBI), which is characterized by damage to the brain resulting from a sudden traumatic event, is a major cause of death and disability worldwide. It has short- and long-term effects, including neuroinflammation, cognitive deficits, and depression. TBI consists of multiple steps that may sometimes have opposing effects or mechanisms, making it challenging to investigate and translate new knowledge into effective therapies. In order to better understand and address the underlying mechanisms of TBI, we have developed an in vitro platform that allows dynamic simulation of TBI conditions by applying external magnetic forces to induce acceleration and deceleration injury, which is often observed in human TBI. Endothelial and neuron-like cells were successfully grown on magnetic gels and applied to the platform. Both cell types showed an instant response to the TBI model, but the endothelial cells were able to recover quickly-in contrast to the neuron-like cells. In conclusion, the presented in vitro model mimics the mechanical processes of acceleration/deceleration injury involved in TBI and will be a valuable resource for further research on brain injury.

6.
Adv Sci (Weinh) ; 10(27): e2207498, 2023 09.
Article in English | MEDLINE | ID: mdl-37485582

ABSTRACT

Despite significant advancements in in vitro cardiac modeling approaches, researchers still lack the capacity to obtain in vitro measurements of a key indicator of cardiac function: contractility, or stroke volume under specific loading conditions-defined as the pressures to which the heart is subjected prior to and during contraction. This work puts forward a platform that creates this capability, by providing a means of dynamically controlling loading conditions in vitro. This dynamic tissue loading platform consists of a thin magnetoresponsive hydrogel cantilever on which 2D engineered myocardial tissue is cultured. Exposing the cantilever to an external magnetic field-generated by positioning magnets at a controlled distance from the cantilever-causes the hydrogel film to stretch, creating tissue load. Next, cell contraction is induced through electrical stimulation, and the force of the contraction is recorded, by measuring the cantilever's deflection. Force-length-based measurements of contractility are then derived, comparable to clinical measurements. In an illustrative application, the platform is used to measure contractility both in untreated myocardial tissue and in tissue exposed to an inotropic agent. Clear differences are observed between conditions, suggesting that the proposed platform has significant potential to provide clinically relevant measurements of contractility.


Subject(s)
Heart , Myocardial Contraction , Myocardial Contraction/physiology , Heart/physiology , Myocardium , Hydrogels , Magnetic Phenomena
7.
Microbiol Spectr ; 11(3): e0123323, 2023 06 15.
Article in English | MEDLINE | ID: mdl-37222614

ABSTRACT

Understanding the pathogenesis of bacterial infections is critical for combatting them. For some infections, animal models are inadequate and functional genomic studies are not possible. One example is bacterial meningitis, a life-threatening infection with high mortality and morbidity. Here, we used the newly developed, physiologically relevant, organ-on-a-chip platform integrating the endothelium with neurons, closely mimicking in vivo conditions. Using high-magnification microscopy, permeability measurements, electrophysiological recordings, and immunofluorescence staining, we studied the dynamic by which the pathogens cross the blood-brain barrier and damage the neurons. Our work opens up possibilities for performing large-scale screens with bacterial mutant libraries for identifying the virulence genes involved in meningitis and determining the role of these genes, including various capsule types, in the infection process. These data are essential for understanding and therapy of bacterial meningitis. Moreover, our system offers possibilities for the study of additional infections-bacterial, fungal, and viral. IMPORTANCE The interactions of newborn meningitis (NBM) with the neurovascular unit are very complex and are hard to study. This work presents a new platform to study NBM in a system that enables monitoring of multicellular interactions and identifies processes that were not observed before.


Subject(s)
Meningitis, Bacterial , Animals , Meningitis, Bacterial/microbiology , Blood-Brain Barrier , Neurons , Lab-On-A-Chip Devices
8.
Neurotrauma Rep ; 4(1): 255-266, 2023.
Article in English | MEDLINE | ID: mdl-37095852

ABSTRACT

Traumatic brain injury (TBI) is a major health problem that affects millions of persons worldwide every year among all age groups, mainly young children, and elderly persons. It is the leading cause of death for children under the age of 16 and is highly correlated with a variety of neuronal disorders, such as epilepsy, and neurodegenerative disease, such as Alzheimer's disease or amyotrophic lateral sclerosis. Over the past few decades, our comprehension of the molecular pathway of TBI has improved, yet despite being a major public health issue, there is currently no U.S. Food and Drug Administration-approved treatment for TBI, and a gap remains between these advances and their application to the clinical treatment of TBI. One of the major hurdles for pushing TBI research forward is the accessibility of TBI models and tools. Most of the TBI models require costume-made, complex, and expensive equipment, which often requires special knowledge to operate. In this study, we present a modular, three-dimensional printed TBI induction device, which induces, by the pulse of a pressure shock, a TBI-like injury on any standard cell-culture tool. Moreover, we demonstrate that our device can be used on multiple systems and cell types and can induce repetitive TBIs, which is very common in clinical TBI. Further, we demonstrate that our platform can recapitulate the hallmarks of TBI, which include cell death, decrease in neuronal functionality, axonal swelling (for neurons), and increase permeability (for endothelium). In addition, in view of the continued discussion on the need, benefits, and ethics of the use of animals in scientific research, this in vitro, high-throughput platform will make TBI research more accessible to other labs that prefer to avoid the use of animals yet are interested in this field. We believe that this will enable us to push the field forward and facilitate/accelerate the availability of novel treatments.

9.
Burns ; 49(5): 1170-1180, 2023 Aug.
Article in English | MEDLINE | ID: mdl-36195492

ABSTRACT

The use of in-vitro and ex-vivo models for the study of burn wound injuries is encouraged to reduce the animal burden in experimental burn research. However, few existing platforms enable the production of reproducible, locally confined thermal injuries at short durations in a high-throughput manner for both in-vitro and ex-vivo models. To address this gap, we established an automated high-throughput burn platform (HTBP) that provided accurate control over burn temperature, exposure time, and pressure application. This platform was built by fabricating an aluminum heat block with 96 pins and positioning a high-resolution actuator below the block. By activating the actuator, 96-well cell culture plates and skin samples were pressed against the heat block's pins. We demonstrated the applicability of the HTBP for studying in-vitro burn injuries by investigating the effects of burn temperature and contact duration on cell viability and migration in human umbilical vein endothelial cells and NIH-3T3 fibroblasts. We showed that higher temperatures and a longer contact duration decreased cellular viability and increased the area of the burn. Moreover, we found that even a short exposure time of 200 msec caused a severe burn wound at 75 °C in a cell monolayer. In addition, we used the HTBP to generate burn injuries at different burn durations in ex-vivo porcine skin and showed that dermis discoloration was present in histologic sections after exposure to 100 °C for a short duration of 500 msec. Our work demonstrates that the HTBP can constitute an important tool for both in-vitro and ex-vivo research of mild and severe burn injuries in a tightly controlled setting and high-throughput manner.


Subject(s)
Burns , Swine , Animals , Humans , Burns/pathology , Endothelial Cells , Temperature , Hot Temperature , Fibroblasts/pathology , Skin/pathology
10.
Pharmaceutics ; 14(11)2022 Nov 17.
Article in English | MEDLINE | ID: mdl-36432688

ABSTRACT

Fluorescent organic nanoparticles (FONs) are a large family of nanostructures constituted by organic components that emit light in different spectral regions upon excitation, due to the presence of organic fluorophores. FONs are of great interest for numerous biological and medical applications, due to their high tunability in terms of composition, morphology, surface functionalization, and optical properties. Multifunctional FONs combine several functionalities in a single nanostructure (emission of light, carriers for drug-delivery, functionalization with targeting ligands, etc.), opening the possibility of using the same nanoparticle for diagnosis and therapy. The preparation, characterization, and application of these multifunctional FONs require a multidisciplinary approach. In this review, we present FONs following a tutorial approach, with the aim of providing a general overview of the different aspects of the design, preparation, and characterization of FONs. The review encompasses the most common FONs developed to date, the description of the most important features of fluorophores that determine the optical properties of FONs, an overview of the preparation methods and of the optical characterization techniques, and the description of the theoretical approaches that are currently adopted for modeling FONs. The last part of the review is devoted to a non-exhaustive selection of some recent biomedical applications of FONs.

11.
Int J Mol Sci ; 23(4)2022 Feb 12.
Article in English | MEDLINE | ID: mdl-35216162

ABSTRACT

Fragile X syndrome (FXS), the most common form of inherited intellectual disability, is caused by a developmentally regulated silencing of the FMR1 gene, but its effect on human neuronal network development and function is not fully understood. Here, we isolated isogenic human embryonic stem cell (hESC) subclones-one with a full FX mutation and one that is free of the mutation (control) but shares the same genetic background-differentiated them into induced neurons (iNs) by forced expression of NEUROG-1, and compared the functional properties of the derived neuronal networks. High-throughput image analysis demonstrates that FX-iNs have significantly smaller cell bodies and reduced arborizations than the control. Both FX- and control-neurons can discharge repetitive action potentials, and FX neuronal networks are also able to generate spontaneous excitatory synaptic currents with slight differences from the control, demonstrating that iNs generate more mature neuronal networks than the previously used protocols. MEA analysis demonstrated that FX networks are hyperexcitable with significantly higher spontaneous burst-firing activity compared to the control. Most importantly, cross-correlation analysis enabled quantification of network connectivity to demonstrate that the FX neuronal networks are significantly less synchronous than the control, which can explain the origin of the development of intellectual dysfunction associated with FXS.


Subject(s)
Fragile X Syndrome/metabolism , Membrane Potentials , Transcriptome , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cells, Cultured , Fragile X Mental Retardation Protein/genetics , Human Embryonic Stem Cells/cytology , Human Embryonic Stem Cells/metabolism , Humans , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neural Stem Cells/physiology , Neurogenesis , Rats
12.
Cell Tissue Res ; 387(3): 493-498, 2022 Mar.
Article in English | MEDLINE | ID: mdl-34850274

ABSTRACT

Blood coagulation factors can enter the brain under pathological conditions that affect the blood-brain interface. Besides their contribution to pathological brain states, such as neural hyperexcitability, neurodegeneration, and scar formation, coagulation factors have been linked to several physiological brain functions. It is for example well established that the coagulation factor thrombin modulates synaptic plasticity; it affects neural excitability and induces epileptic seizures via activation of protease-activated receptors in the brain. However, major limitations of current experimental and clinical approaches have prevented us from obtaining a profound mechanistic understanding of "neuro-coagulation" in health and disease. Here, we present how novel human relevant models, i.e., Organ-on-Chips equipped with advanced sensors, can help overcoming some of the limitations in the field, thus providing a perspective toward a better understanding of neuro-coagulation in brain homeostasis.


Subject(s)
Receptor, PAR-1 , Thrombin , Brain/metabolism , Homeostasis , Humans , Receptor, PAR-1/metabolism , Technology , Thrombin/metabolism , Thrombin/pharmacology
13.
Lab Chip ; 22(1): 71-79, 2021 12 21.
Article in English | MEDLINE | ID: mdl-34878455

ABSTRACT

Transepithelial/transendothelial electrical resistance (TEER) is a label-free assay that is commonly used to assess tissue barrier integrity. TEER measurement systems have been embedded in organ-on-a-chip devices to provide live readouts of barrier functionality. Yet, these systems commonly provide the impedance values which correspond to the highest level of permeability throughout the chip and cannot provide localized information on specific regions of interest. This work introduces a system that provides this essential information: a spatial-TEER (S-TEER) organ-on-a-chip platform, which incorporates moving (scanning) electrodes that can measure electrical resistance at any desired location along the chip. We demonstrate the system's capacity to obtain localized measurements of permeability in selected regions of a cell sample. We show how, in a layer with non-uniform levels of cell coverage, permeability is higher in areas with lower cell density-suggesting that the system can be used to monitor local cellular growth in vitro. To demonstrate the applicability of the chip in studies of barrier function, we characterize tissue response to TNF-α and to EGTA, agents known to harm tissue barrier integrity.


Subject(s)
Lab-On-A-Chip Devices , Electric Impedance , Electrodes , Permeability
14.
APL Bioeng ; 5(3): 030902, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34368601

ABSTRACT

The complexity of the human brain creates significant, almost insurmountable challenges for neurological drug development. Advanced in vitro platforms are increasingly enabling researchers to overcome these challenges, by mimicking key features of the brain's composition and functionality. Many of these platforms are called "Brains-on-a-Chip"-a term that was originally used to refer to microfluidics-based systems containing miniature engineered tissues, but that has since expanded to describe a vast range of in vitro central nervous system (CNS) modeling approaches. This Perspective seeks to refine the definition of a Brain-on-a-Chip for the next generation of in vitro platforms, identifying criteria that determine which systems should qualify. These criteria reflect the extent to which a given platform overcomes the challenges unique to in vitro CNS modeling (e.g., recapitulation of the brain's microenvironment; inclusion of critical subunits, such as the blood-brain barrier) and thereby provides meaningful added value over conventional cell culture systems. The paper further outlines practical considerations for the development and implementation of Brain-on-a-Chip platforms and concludes with a vision for where these technologies may be heading.

15.
Annu Rev Biomed Eng ; 23: 359-384, 2021 07 13.
Article in English | MEDLINE | ID: mdl-34255993

ABSTRACT

The blood-brain barrier (BBB) is one of the most selective endothelial barriers. An understanding of its cellular, morphological, and biological properties in health and disease is necessary to develop therapeutics that can be transported from blood to brain. In vivo models have provided some insight into these features and transport mechanisms adopted at the brain, yet they have failed as a robust platform for the translation of results into clinical outcomes. In this article, we provide a general overview of major BBB features and describe various models that have been designed to replicate this barrier and neurological pathologies linked with the BBB. We propose several key parameters and design characteristics that can be employed to engineer physiologically relevant models of the blood-brain interface and highlight the need for a consensus in the measurement of fundamental properties of this barrier.


Subject(s)
Blood-Brain Barrier , Brain , Biological Transport , Biology , Humans
16.
ACS Nano ; 15(7): 11087-11098, 2021 Jul 27.
Article in English | MEDLINE | ID: mdl-34137606

ABSTRACT

Loss of tactile sensation is a common occurrence in patients with traumatic peripheral nerve injury or soft tissue loss, but as yet, solutions for restoring such sensation are limited. Implanted neuro-prosthetics are a promising direction for tactile sensory restoration, but available technologies have substantial shortcomings, including complexity of use and of production and the need for an external power supply. In this work, we propose, fabricate, and demonstrate the use of a triboelectric nanogenerator (TENG) as a relatively simple, self-powered, biocompatible, sensitive, and flexible device for restoring tactile sensation. This integrated tactile TENG (TENG-IT) device is implanted under the skin and translates tactile pressure into electrical potential, which it relays via cuff electrodes to healthy sensory nerves, thereby stimulating them, to mimic tactile sensation. We show that the device elicits electrical activity in sensory neurons in vitro, and that the extent of this activity is dependent on the level of tactile pressure applied to the device. We subsequently demonstrate the TENG-IT in vivo, showing that it provides tactile sensation capabilities (as measured by a von Frey test) to rats in which sensation in the hindfoot was blocked through transection of the distal tibial nerve. These findings point to the substantial potential of self-powered TENG-based implanted devices as a means of restoring tactile sensation.


Subject(s)
Electric Power Supplies , Nanotechnology , Rats , Animals , Electrodes , Electricity , Touch/physiology
17.
APL Bioeng ; 5(2): 026103, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33948527

ABSTRACT

Organ-on-a-Chip platforms provide rich opportunities to observe interactions between different cell types under in vivo-like conditions, i.e., in the presence of flow. Yet, the costs and know-how required for the fabrication and implementation of these platforms restrict their accessibility. This study introduces and demonstrates a novel Insert-Chip: a microfluidic device that provides the functionality of an Organ-on-a-Chip platform, namely, the capacity to co-culture cells, expose them to flow, and observe their interactions-yet can easily be integrated into standard culture systems (e.g., well plates or multi-electrode arrays). The device is produced using stereolithograpy 3D printing and is user-friendly and reusable. Moreover, its design features overcome some of the measurement and imaging challenges characterizing standard Organ-on-a-Chip platforms. We have co-cultured endothelial and epithelial cells under flow conditions to demonstrate the functionality of the device. Overall, this novel microfluidic device is a promising platform for the investigation of biological functions, cell-cell interactions, and response to therapeutics.

18.
Sensors (Basel) ; 21(1)2021 Jan 01.
Article in English | MEDLINE | ID: mdl-33401414

ABSTRACT

During hundreds of millions of years of evolution, insects have evolved some of the most efficient and robust sensing organs, often far more sensitive than their man-made equivalents. In this study, we demonstrate a hybrid bio-technological approach, integrating a locust tympanic ear with a robotic platform. Using an Ear-on-a-Chip method, we manage to create a long-lasting miniature sensory device that operates as part of a bio-hybrid robot. The neural signals recorded from the ear in response to sound pulses, are processed and used to control the robot's motion. This work is a proof of concept, demonstrating the use of biological ears for robotic sensing and control.


Subject(s)
Grasshoppers , Robotics , Animals , Ear, Middle , Lab-On-A-Chip Devices , Sound
19.
Brain ; 143(11): 3181-3213, 2020 12 05.
Article in English | MEDLINE | ID: mdl-33020798

ABSTRACT

The complexity of the human brain poses a substantial challenge for the development of models of the CNS. Current animal models lack many essential human characteristics (in addition to raising operational challenges and ethical concerns), and conventional in vitro models, in turn, are limited in their capacity to provide information regarding many functional and systemic responses. Indeed, these challenges may underlie the notoriously low success rates of CNS drug development efforts. During the past 5 years, there has been a leap in the complexity and functionality of in vitro systems of the CNS, which have the potential to overcome many of the limitations of traditional model systems. The availability of human-derived induced pluripotent stem cell technology has further increased the translational potential of these systems. Yet, the adoption of state-of-the-art in vitro platforms within the CNS research community is limited. This may be attributable to the high costs or the immaturity of the systems. Nevertheless, the costs of fabrication have decreased, and there are tremendous ongoing efforts to improve the quality of cell differentiation. Herein, we aim to raise awareness of the capabilities and accessibility of advanced in vitro CNS technologies. We provide an overview of some of the main recent developments (since 2015) in in vitro CNS models. In particular, we focus on engineered in vitro models based on cell culture systems combined with microfluidic platforms (e.g. 'organ-on-a-chip' systems). We delve into the fundamental principles underlying these systems and review several applications of these platforms for the study of the CNS in health and disease. Our discussion further addresses the challenges that hinder the implementation of advanced in vitro platforms in personalized medicine or in large-scale industrial settings, and outlines the existing differentiation protocols and industrial cell sources. We conclude by providing practical guidelines for laboratories that are considering adopting organ-on-a-chip technologies.


Subject(s)
Models, Neurological , Nervous System Physiological Phenomena , Translational Research, Biomedical , Animals , Engineering , Humans , Models, Animal
20.
Biomolecules ; 10(9)2020 09 10.
Article in English | MEDLINE | ID: mdl-32927777

ABSTRACT

The human-relevance of an in vitro model is dependent on two main factors-(i) an appropriate human cell source and (ii) a modeling platform that recapitulates human in vivo conditions. Recent years have brought substantial advancements in both these aspects. In particular, mesenchymal stem cells (MSCs) have emerged as a promising cell source, as these cells can differentiate into multiple cell types, yet do not raise the ethical and practical concerns associated with other types of stem cells. In turn, advanced bioengineered in vitro models such as microfluidics, Organs-on-a-Chip, scaffolds, bioprinting and organoids are bringing researchers ever closer to mimicking complex in vivo environments, thereby overcoming some of the limitations of traditional 2D cell cultures. This review covers each of these advancements separately and discusses how the integration of MSCs into novel in vitro platforms may contribute enormously to clinical and fundamental research.


Subject(s)
Cell Lineage/physiology , Cell- and Tissue-Based Therapy/methods , Mesenchymal Stem Cells/physiology , Models, Biological , Organoids/physiology , Tissue Engineering/methods , Bioartificial Organs , Biomimetic Materials/therapeutic use , Bioprinting/methods , Cell Culture Techniques , Cell Differentiation , Humans , Lab-On-A-Chip Devices , Mesenchymal Stem Cells/cytology , Organoids/cytology , Tissue Scaffolds
SELECTION OF CITATIONS
SEARCH DETAIL
...