Abstract
Neurodegenerative diseases (NDDs) include more than 600 types of nervous system disorders in humans that impact tens of millions of people worldwide. Estimates by the World Health Organization (WHO) suggest NDDs will increase by nearly 50% by 2030. Hence, development of advanced models for research on NDDs is needed to explore new therapeutic strategies and explore the pathogenesis of these disorders. Different approaches have been deployed in order to investigate nervous system disorders, including two-and three-dimensional (2D and 3D) cell cultures and animal models. However, these models have limitations, such as lacking cellular tension, fluid shear stress, and compression analysis; thus, studying the biochemical effects of therapeutic molecules on the biophysiological interactions of cells, tissues, and organs is problematic. The microfluidic “organ-on-a-chip” is an inexpensive and rapid analytical technology to create an effective tool for manipulation, monitoring, and assessment of cells, and investigating drug discovery, which enables the culture of various cells in a small amount of fluid (10−9 to 10−18 L). Thus, these chips have the ability to overcome the mentioned restrictions of 2D and 3D cell cultures, as well as animal models. Stem cells (SCs), particularly neural stem cells (NSCs), induced pluripotent stem cells (iPSCs), and embryonic stem cells (ESCs) have the capability to give rise to various neural system cells. Hence, microfluidic organ-on-a-chip and SCs can be used as potential research tools to study the treatment of central nervous system (CNS) and peripheral nervous system (PNS) disorders. Accordingly, in the present review, we discuss the latest progress in microfluidic brain-on-a-chip as a powerful and advanced technology that can be used in basic studies to investigate normal and abnormal functions of the nervous system.
Keywords: Nervous system, Brain, Neurodegenerative diseases, Microfluidic brain-on-a-chip, Stem cells
Introduction
Investigation into the central nervous system (CNS) is pivotal to develop novel therapies for neurodegenerative diseases (NDDs). NDDs include more than 600 types of nervous system disorders in humans such as Alzheimer’s disease (AD), Parkinson’s disease (PD), Huntington’s disease (HD), amyotrophic lateral sclerosis (ALS or Lou Gehrig’s), multiple sclerosis (MS), and impact tens of millions of people worldwide of which 15 million suffer from AD [1–4]. According to the World Health Organization (WHO), NDDs will increase by nearly 50% by 2030, and more than twofold by 2050. Therefore, there is an essential necessity to establish effective models of NDDs and efficacious therapeutic strategies to inhibit or slow the progression of these disorders [5, 6]. The United States (US) “Brain Research through Advancing Innovative Neurotechnologies” (BRAIN) initiative has proposed to encourage the development of devices to image and assess the function of the brain, and the European Union (EU) Human Brain Project (HBP) has also tried to construct computational models of the brain. These organizations have allocated more than US $1 billion and EU €1 billion to promote BRAIN and HBP, respectively [7]. Moreover, the Defense Advanced Research Projects Agency (DARPA), National Science Foundation (NSF), and National Institutes of Health (NIH) have contributed to fund the BRAIN initiative to further understand the human brain and it’s diseases [8].
The Challenges Faced by Current Researches into Neural System Disorders
There are several models of neural systems that have been used to study the physiological and pathological functions of the CNS in NDDs, including animal models, 2D and 3D cell cultures, and clinical studies. However, investigators have to face different challenges that limit the various models. For example, in randomized clinical trials (RCT), adherence to the Consolidated Standards of Reporting Trials (CONSORT) guidelines requires consideration be given to patient diagnosis and selection process, alongside other ethical issues [9]. Animal models are important to study the function of the CNS and NDDs, although they often cannot be generalized to humans. For instance, rodents have a less complex brain structure and different cognitive functions. Some transgenic animal models of human diseases may not provide the physiopathological features of the human neural system and NDDs. As such, significant differences at the molecular and cellular levels exist between rodents and humans [10]. It has been reported that many clinical trials in humans have failed despite promising results that were achieved in pre-clinical studies on animal models [11, 12].
Classical two-dimensional (2D) monolayer cell cultures have been widely utilized to discover therapeutic molecules, including nano-drug delivery, and to predict their side effects [13–15]. However, these models have a poor ability to assess drug responses in complex diseases, and they cannot fully simulate the physiological condition of tissue architecture and the microenvironment as they are altered during the disease (Table 1) [17–19].
Table 1.
Some advantages and disadvantages of 2D and 3D cell cultures
| Models | Advantages | Disadvantages | Reference |
|---|---|---|---|
| 2D cell cultures | ► Economical ►Well established and fast for primary assessment ► Easy and convenient for the analysis setup ► High-throughput capacity: feasible for mass screening |
► Stiff, plastic substrate ► Lacking cell-cell and cell-matrix interaction ►Unable to mimic in vivo microenvironment ► Homogenous drug distribution ►Unable to assess the tissue penetration ability and bystander killing efficiency of drugs, or the drug resistance of cells ► High requirement for design–in vitro–in vivo–redesign cycle: overall high cost |
(https://www.creative-biolabs.com/adc/2d-vs-3d.htm) [16] |
| 3D cell cultures | ► Substrates mimic the natural extracellular matrix ► Mimic the in vivo microenvironment, as well as cell–cell and cell–matrix interactions ► Enable the assessment of tissue penetration ability and bystander killing efficiency of drugs, as well as the drug resistance of cells ► Low design-in vitro-in vivo-redesign requirement: reduce overall cost for the pre-clinical in vivo test |
► More expensive and laborious for the establishment of culture models and analyzing experiments ► Relatively low throughput capacity |
(https://www.creative-biolabs.com/adc/2d-vs-3d.htm) [16] |
Three-dimensional (3D) models comprising bioprinted organ cell cultures, self-organized spherical organoids, and microfabricated organ-on-a-chip systems are used to assess drug delivery, drug discovery, and toxicity in neural systems for therapeutic purposes [20–24]. Organoids are one of the most important 3D culture techniques, which by utilizing self-regenerating SCs enable recapitulating the normal function of organs such as breast, liver, stomach, eye, kidney, lung, gut, pancreas, prostate, and specially the brain, which is the most complex organ in animals and humans [25–27]. Nevertheless, these 3D cell culture models can be very variable structures, and it is hard to predict the exact positions of different neural cells for evaluating the biological activities of neural networks [28, 29]. Furthermore, the genetic and biochemical assessment of the seeded cells in the 3D cell culture models such as the production, entrapment, absorption, secretion, and trans- and intracellular transportation of a vast range of molecules is difficult. In addition, these models are generally unable to account for cellular tension, fluid shear stress, and compression analysis. As well, they are unable to recapitulate the normal microenvironment and tissue architecture including blood supply and circulation of immune cells, and also tissue-tissue interaction between parenchymal cells, connective tissue, and vascular endothelium that are pivotal to organ function in health and disease [30, 31]. A large number of molecules and genes govern cell expansion, tissue regeneration, and organogenesis under the influence of mechanobiological stimuli and epigenetic regulator elements [32, 33]. It is essential to establish a model that recapitulates the in vivo structure of diseases to understand the causes and mechanisms of these illnesses and enables the discovery of new therapeutic strategies for NDDs [34–36]. Although in vivo models have many advantages to understand disease mechanisms, they have some disadvantages like being costly, time consuming, and exhibit uncertain translation of their results to humans. Therefore, in vitro models have attracted a lot of attention due to lower costs, time saving, and the greater simplicity of experiments. Recently, 3D cell culture in vitro models, which use human brain cells, have been developed to study the structure and physiological properties of CNS diseases in greater detail [37]. Recent progress suggests that organoids can be used as a preclinical model of human diseases, including NDDs [38].
As mentioned above, there are several limitations to the current models for research on neural systems. Hence, advanced models are needed to overcome these limitations and improve experimental models. As a result, investigators have put forward human organ-on-a-chip devices to substitute for current models as a safe and applicable strategy for drug discovery (Fig. 1).
Fig. 1.

Schematic illustrations of SCs in a brain-on-a-chip technology for NDDs investigations
Microfluidic Technology
Microfluidic systems can create a powerful microengineered scaffold-free or scaffold-based tool for the manipulation, monitoring, and assessment of cells, and for use in drug discovery with high specificity and resolution [39–46]. This technology was developed in the early 1990s as a very small-scale vehicle, which was usually constructed of microchambers, microchannels, and functionalized microdomains whose dimensions range from tens to hundreds of micrometers, used for culturing various cells in a small 10−9 to 10−18 L volume of fluid, that is constantly under flow conditions to evaluate the function of cells, tissues, and organs (Fig. 2) [39, 44, 47]. Currently, a vast range of microfluidic systems have been fabricated using various materials and methods. These systems are able to be applied in the manufacture of microfluidic brain-on-a-chip devices for brain activities evaluation. Microfluidic chips are usually fabricated by “soft lithography, photolithography, contact printing, laser patterning, and 3D printing” techniques using several materials such as polydimethylsi-loxane (PDMS), polycarbonate (PC), polyetherimide (PEI), silicon, glass, hyaluronic acid, matrigel, collagen, silk protein, agarose, etc. [48, 49]. Although the most commonly used material for microfluidic device is PDMS, there are several other potential candidate materials that can be used for microfluidic devices (Table 2). Microfluidic brain-on-a-chip systems can be manufactured using a combination of soft lithography and photolithography as well as 3D printing technologies. These microfluidic brain-on-a-chip systems have used an optically transparent, highly flexible, nontoxic, and air-permeable polymer called PDMS, which provides the ability for high-resolution optical imaging [44, 47, 51–54]. More detailed discussion of the production methods for various microfluidic brain-on-a-chip systems can be obtained elsewhere [51] and are summarized in Table 3. In comparison with 2D cell cultures and 3D organoids, and also animal models, microfluidic systems consume only a very small amount of samples and materials. They also have the ability to allow detection of cell, tissue, and organ activities in an inexpensive, rapid, highly accurate, and precise analytical method [45, 55, 56]. Moreover, microfluidic brain-on-a-chip systems can potentially improve the drug evaluation process in an in vitro environment. Selected human-derived nerve cells are able to be proliferated, differentiated, and organized in brain-on-a-chip models [57]. Moreover, these platforms are able to create a uniform profile of controlled flow of nutrients, establish individual cellular activity, and provide a platform for monitoring and excitation of neuronal cells. In addition, this technology can allow the study of mechanical, physiological, pharmacological, and biochemical aspects, such as dynamic scaffold alteration and the occurrence of stress seen when nervous diseases undergo progression, and treatments for these diseases are able to be investigated and monitored in real time [37, 58, 59].
Fig. 2.

Schematic illustration of a microfluidic brain-on-a-chip device. human induced pluripotent stem cells (hiPSC)-based can be produced from adult somatic cells using a nanoliposome-based-clustered regularly interspaced palindromic repeats (CRISPR) system. The hiPSCs can differentiate into many cell types such as (A)) astrocytes (ASTs), (B) neurons, and (C) oligodendrocytes. Co-culture of theses neural cells (A, B, and C) in a microfluidic brain-on-a-chip device can be used to evaluate the molecular, cellular, and structural connections between neural cells such as ASTs, neurons, and oligodendrocytes for NDDs researches
Table 2.
Potential materials used in microfluidics
| Material | Relevant property | Proposed application |
|---|---|---|
| Collagen or chitosan | Biocompatibility, versatile control of structure and chemistry | Bio-sensing, film assembly |
| Silkworm (Bombyx mori) | Biocompatibility, mechanically robust, flexibility, high mechanical modulus, and toughness | Fabrication of microfluidic channel |
| Agarose hydrogel | Lox cytotoxicity, biodegradability, mechanical stability at low solid fractions | Cell culture, sensors, and actuators |
| Teflon | Ease of fabrication with maximum chemical resistance | High-precision assay, super clean tools, valves, and pumps fabrication |
| Acrylonitrile butadiene styrene (ABS) | High resolution, excellent surface finish | Making of the master mold, microfluidics interface (MI), pathogen detection, biological assay |
| Photocurable resin/polymer | Very high resolution with small features | Biology observation of cell growth |
| ABS, polycarbonate, polyphenylsulfone, elastomers | Cheap material, ease of support removal | Pathogen detection of bacteria and viruses |
| Polyamide | Fast build speed, multi-material printing, very durable and high-temperature stable material | Making of the master mold |
| Hydrogels | Swelling and contraction, act as sensors and actuators | Self-regulating valves, microlens arrays, drug release systems, binding of antigens and proteins and glucose. Flow sensors pH regulators, flooding cooling devices |
| Polyurethane-methacrylate (PUMA) | Economical to manufacture, biocompatible, non-toxic, strong electro-osmotic mobility | High-aspect-ratio microstructures |
| Polyethylene glycols (PEGs) | Relatively inexpensive, available in a wide variety of molecular weights, biocompatible, negligible cytotoxicity | Microfluidic valves, channel cover to improve the microfluidic lifetime |
| Polyhydroxyalkanoates (PHAs) | Biocompatibility, tunable biodegradability | Microfilm barrier for vapor and oxygen |
| Gelatin methacrylate (gel-MA) | Photopolymerizable, porous membrane | Mechanistic vascular and valvular biology cell support matrix |
| Polylactic acid (PLA) and polyglycolic acid (PGA) | Tunable biodegradation | Porous scaffold for cell culture with better adhesion |
| Poly(polyol sebacate) (PPS) | Biocompatibility, design adaptability, mechanical compliance, low cytotoxicity, degradability | 3D microfluidic system, microbioreactor |
| Poly(ethylene glycol) diacrylate (PEGDA) and gelatin methacryloyl (GelMA) | Biocompatibility, neovascularization potential, multi-material fabrication capability at a high spatial resolution | Tissue engineering, regenerative medicine, and bio-sensing |
| Poly(methyl methacrylate) | Favorable mechanical and thermal resistance, chemical compatibility | Genomic analysis |
| Styrene ethylene butylene styrene (SEBS) | Biocompatibility, rheological characteristics | Fabrication of complex and more sophisticated microfluidic networks (pFNs) |
| SEBS | Electrical surface properties, stable and relatively high zeta potential magnitude | Microdevices for electrokinetic applications |
| SEBS | Reduced drug absorption, optical transmittance, mechanical performance | Cell culture |
Reprinted with permission from ref. [50], Copyright 2018, Multidisciplinary Digital Publishing Institute (MDPI)
Table 3.
Manufacturing approaches for mcrofluidic brain-on-a-chip
| Manufacturing | The most used materials | Advantages | Disadvantages |
|---|---|---|---|
| Photolithography | Silicon | ► Highly precise designs with computer-aided manufacturing (CAM) | ► During the platform fabrication, the manufacturing procedure does not support simultaneous integration with biology because such steps like material removal and photoresist curing typically require extreme pH, high temperature, and exposure to radiation |
| Soft lithography | PDMS | ► Highly precise designs due to the use of photolithography for master creation and compatibility with computer-aided design (CAD)resources | ► The disadvantages of photolithography are applied to soft lithography because this method requires photolithography for master creation. Hence, the manufacturing procedure does not support simultaneous integration with biology |
| Contact printing | PDMS | ►Similar advantages to soft lithography (e.g., compatibility with CAD and precise designs) ► Compatibility with a wide range of analytes ► Creation of periodic functionalized microdomains across macroscopic length scales |
► This method can only be used to deposit small molecules on the thin material layers ► There are some challenges for repeated contact printing on the same substrate because the mechanical contact step is usually manual |
| Laser patterning | PEGDA, agarose, silicon | ► Unique advantages for directing cell growth in silicon hydrogel NSCs due to the ability to spatially control 3D hydrogel chemistry | ► Limits material availability because in this method only hydrogels and biomolecules functionalized with photoreactive groups can be used |
| 3D Printing | Alginate, agarose, Polycaprolactone (PCL), gellan gum | ► This method has been used in tissue engineering ► 3D Printing is a digital model and printer path gellan gum information can be derived from medical imaging data ► This method is a CAM process that affords repeatability and robustness in multi-layer and multi-material assembly ► A diverse material set such as thermosets, thermoplastics, composites, hydrogels, and solutions can be used in certain types of 3D printing such as micro-extrusion printing |
► Because 3D printing is a serial processing technique, throughput can be limited for large parts that contain intricate path geometries |
Rewritten with modifications from ref. [51], 2017, Springer Nature
The so-called homo chippiens was conceived as a laboratory-on-a-chip (LOC) project to emulate the functions of the entire human body by a collection of different microfluidic organ-on-a-chip systems [60]. The US National Center for Advancing Translational Sciences (NCATS) has also awarded grants to construct synthetic tissues and organs for drug development. The reliability of microfluidic brain-on-a-chip systems can be improved by selecting human-derived nervous cells, and can be more appropriate than conventional animal models used for drug assessment and disease monitoring. The US Food and Drug Administration (FDA) has proposed to encourage this technology to predict results of novel drug testing before they are tested in animal models and in human clinical trials [57, 61–63].
Hence, microfluidic systems have been developed to improve research into NDDs in vitro. In this regard, several studies have been performed based on microfluidic chips for analysis of CNS axon propagation [64], dopaminergic neurons [65, 66], creation of a neurovascular unit-on-a-chip [67], studies on neural stem cells (NSCs) [68], blood–brain barrier (BBB) function [69–74], AD [37, 75], brain tumors [76, 77], and neurotransmitter function (Table 4) [83]. In order to show whether microfluidic brain-on-a-chip devices can overcome these restrictions, in the current article, we review novel reports about the applications of microfluidic brain-on-a-chip devices in studies of neural system disorders.
Table 4.
A summary of studies on microfluidic systems in NDDs
| Subject | Study aims | Experimental design | Main findings | Conclusion | Reference |
|---|---|---|---|---|---|
| NSC-based microfluidic system | ► Development of a 3D ECM-based microfluidic system for NSC differentiation and neural tissue regeneration | ►NSCs isolated from the cortex of Sprague-Dawley rats ► The microfluidic device fabricated by a PDMS microfluidic chip and two PDMS-coated glass slides as the cover ► Five types of NSC microenvironments, including 2D cellular monolayer culture, 2D cellular monolayer culture on the ECM, dispersed cells in the ECM, 3D spheroid aggregates, and 3D spheroids cultured in the ECM, were constructed within an integrated microfluidic chip ► The effect of perfusion and static culture on NSCs was evaluated ► The NSC viability, self-renewal, proliferation, and differentiation into neurons, astrocytes, or oligodendrocytes were evaluated |
► The NSCs in the microfluidic device established good viability ► The 3D culture in the ECM accelerated NSC self-renewal and proliferation ► The static 2D culture allowed NSC differentiation into the neuron lineage ► The spheroid culture on the ECM under perfusion and the monolayer culture in static state allowed NSCs differentiation into glial cells |
► This microfluidic device could provide a NSC-based model for NDD therapy | [68] |
| ► Development of a reproducible encapsulation of NSCs and dental pulp stem cells (DPSCs) using microcapsules | ► Spinal cord tissues, DPSCs and NSCs were isolated from C57BL/6 and the cortex of E14 C57BL/6 mice, respectively C57BL/6 mice, respectively ►NSCs and DPSCs were individually encapsulated within alginate-collagen microcapsules ► Microcapsules containing undifferentiated NSCs, undifferentiated DPSCs, and neuronal pre-differentiated DPSCs transplanted in ex vivo spinal cord injury (SCI) model |
► Both DPSCs and NSCs maintained their multipotency and survived in the microcapsules ► In the SCI model, the microcapsules efficiently maintained the transplanted DPSCs and NSCs at the site of implantation ► After transplantation, the cells survived and differentiated to a neural lineage |
► This microfluidic device provides an effective method to study SCs behavior ► This technique provides a useful method for the encapsulation of DPSCs and NSCs within alginate-collagen microcapsules |
[78] | |
| hiPSC-based microfluidic system | ► Assessment of a new technology to develop a hiPSC-derived 3D brain organoids with organs-on-a-chip method | ► The brain organoid-on-a-chip device was made of PDMS using soft lithography procedure ► Sodium fluorescein permeation assay was performed to validate drug distribution on chip ► Embryoid bodies (EBs) were generated via 3D culture of the hiPSCs that derived from skin fibroblasts, then perfused into hydrogel channel of the chip ► To evaluate this brain organ-on-a-chip model, immunohistochemical staining using pluripotent markers of hiPSCs (TUJ1, PAX6, PAX2, SOX2, Nestin, CTIP2, TBR1, CD133, and ISL1) were performed ► TUNEL assay and real-time PCR were conducted to evaluate apoptosis of cells within EBs and the expressions of brain regional markers (PAX2, PAX6and ISL1), cortical layer markers (CTIP2 and TBR1), and neural markers (SOX2, TUJ1, and Nestin), respectively |
► The brain organoids under perfused culture demonstrated the key features of early human brain development, including neural differentiation, regionalization, and cortical organization ► The brain organoids displayed an upregulation of cortical layer markers (CTIP2 and TBR1) |
► This technology created a robust and simple brain organoid-on-a-chip that can be used for studies of NDDs | [79] |
| ► Development of a brain organ-on-a-chip as to study of prenatal nicotine exposure | ► The microfluidic chip was constructed of PDMS using soft lithography procedure ► Embryoid bodies mast be deleted and added only (EB) were generated via 3D culture of the hiPSCs derived from skin fibroblasts of a healthy individual, then perfused into hydrogel channels of the chip and exposed to different doses of nicotine ► Immunohistochemical staining, TUNEL assay, and real-time PCR were performed to validate the efficacy of this chip |
► Gene expression showed a high expression of PAX2, PAX6, and ISL1 markers ► The PAX2 or ISL1 positive cells were located either together or separately in PAX6 sites ► TUNEL assay revealed nearly 40% cell death within organoids in a static culture condition ► Exposure to nicotine enhanced premature neuronal differentiation with upregulation of TUJ1 marker ► Impairment of brain regionalization and cortical development were found in the nicotine-exposed organoids |
► This technology provides a model for NDDs under environmental exposure ► These findings showed that nicotine exposure impairs neurogenesis in early fetal brain development |
[80] | |
| Microfluidic 3D chip models for NDD studies | ► Design a neurospheroids 3D microfluidic chip mimics the brain microenvironment to study NDDs ► Investigate the influence of flow on neurospheroid size, neural differentiation, and neural network formation ► Evaluation of the toxicity of β-amyloid on neurospheroids |
► Prenatal cortical neurons were isolated from the rat embryos, seeded in chip according to different conditions: group I, static conditions (neurospheroids cultured without flow); group IA, static conditions with amyloid-β treatment; group II, dynamic conditions (neurospheroids cultured with flow); group IIA, dynamic conditions with amyloid-β treatment ► Cell viability assay, immunocytochemistry, and scanning electron microscopy (SEM) were evaluated for validation of this chip |
► In comparison with static conditions, neurospheroids with flow condition were more robust, larger, and formed complex neural networks, suggested a pivotal effect of flow on oxygen, cytokine transport, and removal of metabolic wastes In comparison with static conditions, amyloid-β treatment significantly decreased the viability of neurospheroids and impaired neural networks more than the flow condition |
► This neurospheroid 3D microfluidic chip can be used as an in vitro model for NDDs studies | [37] |
| ► Development of a microfluidic platform for the characterization of CNS active compounds based on calcium imaging readouts | ► Microfluidic devices were fabricated using soft-lithography. ► Primary hippocampal cultures were obtained from the Sprague-Dawley rat pups and cultured on the microfluidic devices ► Immunocytochemistry and calcium imaging were conducted |
► Using this microfluidic device, pharmacological investigations on glutamatergic receptor antagonist can be performed. | ► This microfluidic platform can be used to study CNS active compounds. | [81] | |
| ► Engineering a 3D microfluidic platform of vascular and neuronal networks to study motor neuron disease (MND). | ► Microfluidic devices were made of PDMS using soft lithography ► Co-culture of human ES-derived MN spheroids and endothelial cells were performed in microfluidic devices ► immunostaining of Tuj1, vascular endothelial (VE)-cadherin, HB9, and islet1 and enzyme-linked immunosorbent assay (ELISA) to measure growth factor concentrations (BMP-2 and BDNF) were conducted ► Ca2+ oscillation imaging, perfusability of vascular networks, and vessel permeability coefficient were evaluated as well |
► Co-culture with endothelial cells enhances neuronal connectivity and neurite elongation as measured by Ca2+ oscillation ► Bi-directional signaling was detected in the neural networks that affected vascular network formation |
► This in vitro model enables investigations of neurovascular coupling, which is essential to understanding the pathogenesis of NDDs, including ALS | [82] | |
| Blood–brain barrier-on-a--chip | ► Development of a microfluidic device that models BBB function to analyze the influence of LPS and cytokine cocktail of inflammatory reactions | ► Neurovascular unit microfluidic was fabricated in a two-chamber system; the device was coated with laminin and constructed of PDMS ► Human brain-derived microvascular endothelial cells introduced into the lower chamber and the pericytes and astrocytes were loaded into the upper chamber ► LPS and cytokine cocktail were delivered to neural cells in the vascular chamber ► Different assays, including live/dead evaluation, transendothelial electrical resistance (TEER), FITC-dextran diffusion, tight junction staining, and cytokine measurement were performed to examine the effects of inflammation on BBB function |
► The initial exposure to LPS compromised the BBB function, reduced presence of tight junctions, and increased diffusion ► Cytokine cocktail compromised the BBB function as well |
► This microfluidic device provides a suitable BBB model that can be used to study the effects of inflammatory stimulation on BBB function | [70] |
| ► Design a 3D microfluidic neurovascular chip (NVC) to study BBB function | ► Astrocytes were prepared from the primary cortical neurons of rat embryos ► Human umbilical vein endothelial cell (HUVEC) human cerebral microvascular endothelial cell (hCMEC/D3) cultured on collagen coated cell culture dishes ► The microfluidic device was made of PDMS using soft lithography procedure ► Endothelial cells, neurons, and astrocytes were seeded into the NVC ► Different assays, including immunocytochemistry and confocal imaging, calcium imaging, permeability assay, quantification of permeability coefficient, and neurite analysis, were conducted to evaluate validation of this microfluidic neurovascular chip |
► The results have shown distinct cell type–specific morphological and functional analysis of neurons ► Monolayer cells with size-selective permeability were formed from human cerebral endothelial cells similar to existing in vitro BBB models |
3D microfluidic NVC can be a useful device for neurovascular studies | [69] | |
| ► Development of a microfluidic BBB model for in vitro drug permeability assays | ► Brain microvascular endothelial cells (BMECs) were derived from hiPSCs and co-cultured with primary rat astrocytes on the two sides of a porous membrane on a pumpless microfluidic platform ► The microfluidic device was designed according to the human brain blood residence time mimicking in vivo-like BBB model ► Permeability capacity of the BBB model was conducted after the trans-endothelial electrical resistance (TEER) aiming to evaluate drug permeability of large molecules fluorescein isothiocyanate (FITC)-dextrans |
► The results revealed that this microfluidic BBB model formed continuous tight junction and a suitable TEER resistance ► The permeability level were comparable to in vivo values |
► This microfluidic BBB model mimics physiological BBB function and can be applied as a screening tool for of drug assessments | [74] | |
| ► Engineering a microfluidic 3D BBB chip and study of inflammatory reactions | ► Human brain microvascular endothelial cells (hBMVECs), human brain pericytes, and human astrocytes were used ► The microfluidic device was made of PDMS using soft lithography procedure ► The human ASTs were inserted into the gel and incubated for 18 h. Then, the pericytes and hBMVECs were seeded to form cylindrical lumen using these two cell types ► Permeability assay was performed by the permeability coefficient of small molecular (3 kDa) fluorescent dextran ► Transwell cell culture was conducted on 24-well Transwell coated with rat-tail collagen I; then, the inserts were inverted and pericytes or astrocytes were seeded. After incubation, the inserts were placed in 24-well plates and seeded with hBMVEC. TEER values were measured after 120 h of culture ► Inflammatory stimulation of microfluidic chips was performed with TNF-α and the cytokine release profile was assayed ► Immunocytochemistry staining was conducted using rabbit anti-glial fibrillary acidic protein (GFAP), mouse VE-cadherin, mouse anti-PECAM, mouse anti-zonula occludens-1 (ZO-1), rabbit anti-alpha-smooth muscle actin (SMA), and mouse anti-collagen IV |
► The chip exhibited barrier permeability similar to BBB ► Different secretion profiles of interleukin-6 (IL-6) and granulocyte colony-stimulating factor (G-CSF) were observed after inflammatory trigger with TNF-α ► The level of inflammatory reactions was significantly higher in the 3D BBB chip than the same cells that co-cultured in static Transwell plates |
► This microfluidic 3D BBB chip can be use to study human neurovascular function and inflammatory reactions in vitro. | [72] | |
| ► Design of a microfluidic BBB model which enable to provide both optical transparency and physiological shear stress. | ► The device were by designed by using sandwiching of an optically transparent nanoporous membrane between two PDMS micromolded channels instead of polyester membrane. ► ASTs were encapsulated into collagen hydrogels and inserted into basolateral compartment of the device. The brain ECs were grown on the surface of separating transparent membrane in the cell culture area of a microfluidic to form a ECs layer. ► Immunofluorescence staining for claudin-5 and phalloidin staining for actin were performed. |
► The results demonstrated that the model was capable to simulate the physiological fluid shear stress and have optical transparency to study the function of the BBB model | ► This BBB model can used to study of BBB function with optical transparency and physiological shear stress | [73] | |
| Brain cancer-on-a--chip | ► Development of a 3D brain cancer chip for drug screening | ► The chip composed of polymerizable poly (ethylene) glycol diacrylate (PEGDA) hydrogel and made of PDMS ► Glioblastoma multiforme (GBM) cells were cultured in the chip and treated with pitavastatin and irinotecan |
► The finding demonstrated that this brain cancer-on-a-chip is able to generate a potent GBM cancer model to evaluate the efficacy, screening, and release of drugs | ► This chip is a suitable model of brain cancer for drug screening | [76] |
| ► Design of a microfluidic platform for single-cell proteomic analysis of cancer cells | ► Human glioblastoma cell line and brain tumor cells isolated from patients to analyze the clinical application of the microfluidic system ► A PDMS-based microfluidic attached to poly-L-lysine was designed ► The microfluidic chip was validated by immunohistochemistry and confirmed the striking intertumoral and intratumoral heterogeneity characteristic of glioblastoma |
► Their results and bioinformatics analysis revealed that the microfluidic data enable to quantify molecular signatures and predict tumor progression | ► This microfluidic platform enables the study of single-cell proteomic analysis of cancer cells | [77] |
Microfluidic SC-Based Neural Tissue Engineering
SCs have the capability of continuous self-renewal, unrestricted proliferation, and the capacity to differentiate into various different cell types depending on external cues [84, 85]. SCs can be potentially used for drug delivery, cell therapies, tissue remodeling, organ regeneration, and for disease models [86]. This ability is due to transcriptional factors, which are pivotal regulatory molecules governing the maintenance of these cells [87]. NSCs, hiPSCs derived from somatic cells, embryonic stem cells (ESCs), and also embryonic germ cells (EGCs) can all be encouraged to differentiate into astrocytes (ASTs), oligodendrocytes, and neurons (Fig. 3). Hence, the creation of models using SC-based neural systems might facilitate drug discovery and the elucidation of NDD mechanisms [88–93]. The ability to differentiate into different cells makes SCs suitable for autologous cell transplantation [94]. NSCs, ESCs, and hiPSCs have been employed as potential neuro-regenerative cells in neural system including CNS and peripheral nervous system (PNS) in LOC devices [95–98].
Fig. 3.

Schematic illustrations of SCs in a microfluidic brain-on-a-chip for NDD studies. SCs; particularly NSCs, hiPSCs, and ESCs have the capability to give rise to various neural system cells. Hence, the combination of SCs and microfluidic brain on-a-chip is able to be used as a potential strategy for the investigation of NDDs
NSC-Based Microfluidic Systems
Microfluidic devices have produced a powerful technology for investigation of NSC differentiation. NSCs are known as neural progenitor cells (NPCs) or neural stem/progenitor cells (NSPCs) that are multipotent cells capable of generating both neuronal and glial cells [99, 100]. Glial cells include four main cell types including microglia, ASTs, oligodendrocytes, and their progenitors, NG2-glia [101]. NSC transplantation, which leads to differentiation and expansion of various nervous cells, is pivotal for the restoration of the damaged nervous system and for the treatment of NDDs [102]. In addition, autologous NSCs have also been used to treat non-acute severe traumatic brain injury (TBI) [103]. NSCs express non-specific lineage molecules including stage-specific embryonic antigen (SSEA)-1 and cluster of differentiation (CD)133 (Prominin-1), which can be used to isolate and characterize these cells for research and therapeutic application [104]. Neuronal cells derived from NSCs are able to mature and integrate into the host neural system tissue and recover damage after stroke. Hence, NSCs are therapeutically valuable [105]. Neuronal cells differentiated from NSCs are highly sensitive to physicochemical agents present in their microenvironment, thus these cells must be cultured and evaluated in controlled conditions like those achieved in microfluidic organ-on-a-chip devices [106, 107].
Extracellular matrix (ECM) glycoproteins play important roles in the nervous system and are widely expressed in constituent cells [108, 109]. Early changes in the ECM have been observed in several neurological disorders, including ischemic stroke, MS, AD [109, 110], and schizophrenia [111]. Hence, ECM is important in the study of neural systems and NDDs. In this regard, Wang et al. [68] developed a 3D ECM-based microfluidic system for NSC differentiation and regeneration. NSCs isolated from the fetuses were cultured on a microfluidic device fabricated using PDMS. Then, these cells were investigated under two conditions consisting of conventional static 2D cell culture and a dynamic microfluidic engineered ECM-based 3D model. Their findings showed the dynamic 3D cell culture, using ECM accelerated NSC self-renewal and proliferation, whereas the static 2D cell culture only allowed NSC differentiation into the neuron lineage. Spheroid culture on ECM under perfusion allowed NSCs to differentiate into glial cells [68].
Localization and delivery of stem cells to the target site is one of the major challenges in cell transplantation therapies. SCs microencapsulation has several advantages, such as allowing localization of the cells to a single area, and sustaining cell viability through exchange of nutrients and waste products between the surrounding tissue and the encapsulated cells [78, 112]. Murine cortex cells were differentiated to neurospheres from floating cells. These aggregated neurosphere cells were separated by accutase to single NSCs, loaded into alginate–collagen microcapsules, and then they were placed in a control-lable microfluidic device. Ultimately, their results revealed that the 3D-based microfluidic microenvironment had the ability to improve the viability, expansion, and differentiation of NSCs [78].
hiPSC-Based Microfluidic Systems
hiPSCs are a useful cell types for analyzing brain development and NDDs [113]. Neural cells can be derived from hiPSCs and can be used as an important dynamic model to evaluate the molecular, cellular, and structural activities of the human neural system [114–116]. Implementing these cells in drug discovery devices allows the development of novel investigative models for studying treatment of NDDs [117–119]. Wang et al. [79] developed hiPSC-derived 3D brain organoids using an organ-on-a-chip method prepared from PDMS and soft lithography (Table 2). The results demonstrated that the brain organoids possessed the key features of early human brain development, including neural differentiation, regionalization, and cortical organization [79]. These authors [80] also developed a brain organ-on-a-chip as a microfluidic model to assess neuronal dysfunction caused by nicotine. They found that exposure to nicotine enhanced premature neuronal differentiation and the human pluripotent (hPSCs) based microfluidic organ-on-a-chip device could be a model for the nervous system damaged by nicotine [80].
Microfluidic 3D Chip Models for NDDs Studies
NDDs are generally age-dependent disorders, which are characterized by a slow decrease of the number and function of CNS neural cells [120]. AD is the most common type of NDD seen in elderly people [121, 122]. The limited number of relevant animal models for AD is one of the major challenges hindering development of new AD therapies because studies in rodents and non-human primates often do not translate to humans [123]. Hence, new tools to study AD are needed. Park et al. [37] designed a neurospheroid 3D in vitro model based on a microfluidic chip for AD researchers to evaluate the toxicity of β-amyloid on neurospheroids (Figs. 4 and 5). They found that amyloid-β treatment impaired formation of neural networks and significantly decreased the viability of neurospheroids. Indeed, the neural networks became larger and more complex when neurospheroids were cultured in the presence of interstitial flow when compared to those cultured under static conditions. Such difference could be due to cytokines, better access to nutrients and oxygen (O2), and clearance of metabolic wastes in the flow conditions (Fig. 6) [37].
Fig. 4.

Formation of neural networks in a microfluidic device to develop an in vitro model of Alzheimer’s disease (AD). Formation of neural networks has a critical role for communication of neurons and brain function. In this study, formation of neural networks was compared in two patterns; group I (with static condition) and group II (with dynamic condition), (a and c). Figs 4a and c are scanning electron microscopy (SEM) images of neurospheroids that demonstrates the greater neurite extension in group with the dynamic condition (4a) than the static condition group (4c). Consequently, greater neurite extension in group with dynamic condition leading to the formation of a more robust neural network than the group with the static condition. Figs 4b and d illustrate optical images of the chip with the static and dynamic conditions, respectively, in which demonstrate more active neural network formation in group with dynamic condition (d) compare with the static condition (b). Figs 4e and f reveals a quantitative analysis of the optical images. As shown in the beneath sections of the figs 4b and d, the chip was divided into ten sections by column that each column contains five microwells from inlet to outlet. For comparative analysis between the groups I and II, the average size of neurospheroids and the total number of neurites that extended from microwells was analyzed in each section. The results demonstrate that no neurites were distinguished from the static group, whereas in the dynamic group, a high number of neurites extending from microwells was detected near the inlet and declined towards the outlet of the microchip (Fig 4e). Fig 4f have shown that the sizes of neurospheroids in the static group were almost the same throughout the microchip, conversely, neurospheroids were larger near the inlet and become smaller toward the outlet in the dynamic group.Reprinted with permission from ref. [37], Copyright 2015, Royal Society of Chemistry
Fig. 5.

Comparative immunostaining of neurospheroids for the synaptic marker between the static and dynamic groups. The neurospheroids were immunostained for the synapsinIIa and nestin. SynapsinIIa was increased in the dynamic model compared with the static model (a) indicating that interstitial flow augments synapse formation that leads to the formation of a complex neural network. Fig 5a also illustrates that the levels of synapsinIIa and nestine were less intense in the groups IA (static, medium + amyloid-β (Aβ)) and IIA dynamic, medium + Aβ), which indicates greater destruction of neural networks in the groups that treated with Aβ. The quantitative analysis of the intensity of synapsinIIa and nestine are illustrates in the Figs 5b and c. Reprinted with permission from ref. [37], Copyright 2015, Royal Society of Chemistry
Fig. 6.

Comparative immunofluorescence imaging of neurospheroids stained with thioflavin S (green) and immunostained against neural marker β-III tubulin (red). The fluorescence intensity of neurospheroids that stained with thioflavin S (green) was increased after treatment with AB. In contrast, lower intensity of β-III tubulin was detected in the groups that treated with amyloid-β (section a, groups IA and IIA). The quantitative analysis of the intensity of immunofluorescence images are illustrates in the Figs 6b and c. Reprinted with permission from ref. [37], Copyright 2015, Royal Society of Chemistry
MacKerron et al. [81] developed a microfluidic platform for the characterization of CNS active compounds. The pharmacological properties of a glutaminergic receptor antagonist could be evaluated using this method [81].
Motor neuron disease (MND), also called amyotrophic ALS, is a NDD that affects motor neurons with an unknown etiology. This disease has few experimental models for laboratory investigation [124]. Osaki and colleagues [82] studied MND by designing a 3D microfluidic platform containing vascular and neuronal networks. They found that this model enabled study of neurovascular coupling, which is essential to understanding the pathogenesis of NDDs, such as ALS [82].
BBB-on-a-Chip
Blood circulates throughout the body via blood vessels to deliver O2, nutrients, and hormones to tissues, and to remove CO2 and metabolic wastes from tissues. Circulating blood in the brain is separated from the parenchyma by a highly selective semipermeable membrane known as the BBB designed to exclude pathogens and toxins from the brain [125]. The BBB is made up of four cell types consisting of pericytes, neurons, ASTs, and brain endothelial cells [126]. The BBB is composed of non-fenestrated endothelial cells with tight intercellular junctions, which regulate the movement of molecules and cells between blood and the CNS [125, 127]. Some special compounds such as O2, CO2, and lipid-soluble molecules can generally diffuse freely across the BBB. Essential nutrients like glucose and larger molecules are actively transported across BBB through molecular transporters and receptor-mediated endocytosis, respectively [128]. In addition, harmful agents such as lipophilic agents are effluxed from the endothelial cells by specific membrane efflux pumps like P-glycoprotein (Pgp) [129, 130]. Animal and in vitro models have been used to study the effect of various treatments or drugs on the BBB at cellular, tissue, and systemic levels. Animal models do provide the complexity of the BBB environment for the study of immunology, pharmacodynamics, and pharmacokinetics [131]. However, animal models have less complex brain structures than humans and may not be completely suitable for study of the BBB [96, 132, 133]. The neurovascular unit on-a-chip is an in vitro model, which is reproducible, cost-effective, and time saving, allowing for accelerated evaluation of emerging new drugs [134].
Brown et al. [70] designed a microfluidic device to mimic the BBB function to analyze the influence of a cytokine cocktail and lipopolysaccharide (LPS) on inflammatory reactions. The results revealed that initial exposure to LPS compromised the BBB function and had effects on both physical and metabolic properties of the BBB. Inflammatory cytokines compromised the BBB function as well as LPS [70].
Adriani et al. [69] developed a new 3D microfluidic device containing ASTs, neuron, and human cerebral microvascular endothelial cell (hCMEC/D3) model the permeability properties of the BBB in an in vitro system (Figs. 7 and 8). The immunostaining results showed that endothelial cells had formed a monolayer with intercellular junctions in a monolayer (Fig. 9). In addition, permeability testing revealed that the endothelial cells monolayer acted as a size-selective barrier similar to the BBB in vitro [69, 135].
Fig. 7.

A schematic illustration of 3D model of neurovascular system in a microfluidic device.(a) a designed model of a microfluidic device stained with a food dye (left) and a schematic design of the 3D neurovascular chip (NVC) with focus on their channels: ASTs and neurons are cultures in two central hydrogel channels (blue and orange channels, respectively), endothelial cells (ECs) and the media are hosted in two side channels (green and red, respectively). (b) experiment timeline. (c) Phase contrast imaging of the growth and development of primary neurons, primary ASTs, and ECs (HUVEC and hCMEC/D3) in their respective microfluidic channels over time. Reprinted with permission from ref. [69], Copyright 2017, Royal Society of Chemistry
Fig. 8.

Immunocytochemical staining of initial neurons, primary ASTs, and ECs in 3D neurovascular microfluidic model. (a) The side and top views of the three types of cells in the 3D microfluidic devices: neuronsidentified by doublecortin (DCX) (red), ASTs are positive for GFAP (white), and HUVEC are GFP labeled (green). (b) a 3D view of the neuron in the gel regions. (c) Representative images from the left to right showing immature neurons identified by DCX, AST susing GFAP which exhibited star-shaped morphology, HUVEC and hCMEC/D3 express ingzonula occuldens-1 (ZO-1)(which is a tight junction protein), and GFAP positive ASTs (red) residing to GFP-marked ECs (green) in the NVC. Reprinted with permission from ref. [69], Copyright 2017, Royal Society of Chemistry
Fig. 9.

The characterization of the endothelial barrier in neurovascular microfluidic 3D model. Monolayers of HUVEC and (a) hCMEC/D3 (b) were stained with Hoechst for nucleus, rhodamine phalloidin for F-actin, and monoclonal antibody and NucBlue against VE-cadherin. Images c,d and e,f represent the 3D visualization and sections of the endothelial walls for HUVEC and hCMEC/D3, respectively. Permeability coefficients of 10kDa and 70kDa dextrans in monoculture (with endothelial cells only) and triple co-culture of HUVEC and hCMEC/D3 are calculated in the Graph g and h, respectively 7 days and 4 days in vitro as the permeability time point were chosen for hCMEC/D3 and HUVEC, respectively. Reprinted with permission from ref. [69], Copyright 2017, Royal Society of Chemistry
Wang et al. [74] designed a microfluidic BBB model for in vitro drug permeability assays. The results showed that this model mimicked the physiological BBB function while the measured permeability coefficients for large molecules (caffeine, fluorescein-isothiocyanate (FITC)-dextrans, cimetidine, and doxorubicin) were comparable to those found in in vivo models [74]. Indeed, this model formed continuous tight junctions and had a high barrier integrity with values of trans-endothelial electrical resistance (TEER) above 2000 Ω.cm2 that were similar to in vivo models [74]. Neuroinflammation is known to be involved in the pathophysiology of several neurological and psychiatric disorders and study of neural inflammatory markers can help to discover new treatment options for these disorders [136, 137]. For the study of neural inflammatory processes, Herland et al. [72] engineered a 3D BBB in vitro model using a microfluidic chip containing primary human brain pericytes and microvascular endothelial cells (hBMVECs). Then the cells were stimulated with the inflammatory cytokine tumor necrosis factor alpha (TNF)-α and the cytokine release profile (interleukin (IL)-6 and granulocyte colony-stimulating factor (G-CSF)) was measured. The findings revealed that the chip had a barrier permeability similar to the BBB and the level of inflammatory mediators (IL-6 and G-CSF) was significantly higher in the 3D BBB chip than that for the same cells co-cultured in static trans-well plates [72].
Microfluidic devices are commonly fabricated using PDMS, which has good optical transparency, high flexibility, and high gas permeability that allows fabrication of microtissue models [138]. Sellgren et al. [73] designed a 3D microfluidic BBB model with two PDMS micromolded channels instead of a polyester membrane and found that this model was able to provide optical transparency with suitable physiological fluid shear stress to study the function of the BBB model [73].
Brain Cancer-on-a-Chip
The cellular microenvironment in tumors is heterogeneous and complex with varying levels of vascularization and restricted mass transport that may limit the efficiency of therapeutics [139]. Commonly, cell culture on 2D surfaces is used as an in vitro model to characterize the cell biology of tumors and to evaluate newly developed drugs [140]. However, there is a need for 3D cell culture, which claimed to be more similar to the in vivo situation than 2D cell culture, to study cell-to-cell and cell-to-matrix interactions [139, 141–143]. 3D cell culture have also been used to identify signaling molecules involved in cell–cell and cell–matrix interactions as well as to develop new drugs. Thus, LOC microfluidic technology as a 3D in vitro tissue model could improve the screening of personalized drugs [144, 145].
Glioblastoma multiforme (GBM) is a highly malignant brain tumor and is one of the most challenging types of cancer to treat. Standard treatment regimens for GBM consist of a combination of maximal surgical resection followed by chemotherapy and radiotherapy [146]. The development of advanced models for studying drug delivery to brain tumors could help to discover new treatment options for brain tumors. Fan et al. [76] designed a 3D brain cancer chip to mimic GBM tumors for drug screening. They delivered two different drugs, pitavastatin and irinotecan, to cancerous spheroid cells growing in a PEGDA hydrogel in a PDMS microfluidic device. PEGDA is a hydrophilic long-chain monomer that is suitable as a carrier for drug delivery and biomedical applications. The findings revealed that this 3D brain cancer-on-a-chip was able to generate a useful GBM cancer model for drug screening and drug release assays [76]. Prediction of tumor progression is a challenging issue in brain cancer. Sun and co-workers [77], designed a microfluidic platform for single-cell proteomic analysis of GBM cells. They reported that this microfluidic platform enabled accurate prediction of tumor progression. In another study, Altemus et al. [147] developed a microfluidic BBB model to investigate breast tumor metastasis to the brain. The results revealed that the chip had the capability to model the molecular mechanisms of brain metastasis as well as help in the development of new drugs [147].
The Technical and Biological Challenges of Microfluidic Brain-on-a-Chip Systems
Several technical challenges remain to be overcome in microfluidic brain-on-a-chip systems. One of the remaining major technical challenges is the tendency of drugs and chemicals to undergo non-specific binding to PDMS. As well, PDMS has a structure, which is incompatible with a many organic solvents. Keng et al. [148] developed a compatible type of 2D microscale platform, which was called “electrowetting-on-dielectric” (EWOD). The EWOD device was made of inorganic materials, which were coated with a perfluoropolymer layer. It was manufactured as a typical device including two parallel plates and electrodes that were coated with conductive, dielectric non-wetting layers [148].
Other technical restrictions of microfluidic brain-on-a-chip systems are providing sterile conditions during manufacture, avoiding bubbles, different flow rates between platforms, creating ideal hemoglobin-based oxygenation and nutrient levels, and inclusion of biosensors [149]. Furthermore, the modeling of cell–matrix or cell–cell interactions, cell migration, and 3D cell growth by microfluidic brain-on-a-chip platforms present difficulties. As well, in surface-based microfluidic brain-on-a-chip platforms, cell growth and migration are geometrically restricted in comparison with bulk-based microfluidic brain-on-a-chip systems.
Another important limitation in 3D-based microfluidic brain-on-a-chip platforms is the use of electroactive systems constructed of microelectrode arrays (MEAs) and fluidic channels to simultaneously monitor different microenvironmental factors. More recently, Haehnel et al. [150] utilized a magnetic force method to construct a microelectrode-microfluidic device. This device detected and analyzed many microbiological, chemical, and environmental factors relevant to microfluidic LOC technologies [150].
Most microfluidic systems, with the exception of 3D-printed microfluidic organ-on-a-chip platforms, have not yet used a fully automated technology to create and control their activities [51, 151]. More recently, Kane et al. [152] claimed to have fabricated the first example of an automated microfluidic organ-on-a-chip using SC-derived dopaminergic neurons to evaluate PD. Their automated microfluidic device had the potential ability to allow individual investigation for this neural disease [152]. It is likely that more fully automated biomanufacturing processes for microfluidic brain-on-a-chip systems will be developed in the near future.
However, there are also several biological challenges remaining in brain-on-a-chip systems. The design and creation of these complex systems is not a simple process as the overall size becomes increasingly smaller. In humans and animals, there are complex inter-connections between different organs that exert influences on each other. For instance, the endocrine and immune systems exert an influence on many different organs. In organ-on-a-chip systems, providing these interactions is challenging and complex [153, 154].
Another important biological and technical challenge is the reconstruction of the entire brain including the simulation of vasculogenesis and angiogenesis in a micro-scale platform. Thus, to better mimic the properties and activities of human brain in vitro, it is essential to create a microvasculature 3D-based microfluidic model of the brain. In comparison with 2D-based models, the endothelial cells taking part in vasculogenesis and angiogenesis in 3D-based ECM microfluidic brain-ob-a-chip systems grow inside ECM with a self-assembled mechanism. Hence, 3D-based ECM microfluidic brain-on-a-chip systems are able to generate a more natural vascular structure of the brain, compared to 2D cell culture.
PDMS-based microfluidic brain-on-a-chip systems have a single layer of microchannels with the diameter ranging from 60 to 200 μm, which allows more precise control of these brain-like microenvironments. Nevertheless, endothelial cells in 3D-based ECM microfluidic brain-on-a-chip systems are adherent to a basement sheet with 40–120 nm thickness using proteins such as collagen IV, laminin, and fibronectin for cell adherence. However, monitoring this small system is an important problem [82, 155, 156].
More recently, studies have shown that creating a 3D microenvironment using scaffolds composed of ECM, hydrogels etc. (Table 3) is almost able to mimic the BBB and neural tissues similar to the human brain [153, 154]. As noted in BBB-on-a-Chip section, Adriani et al. [69] constructed a 3D neurovascular microfluidic BBB system including cerebral endothelial cells, neurons, and ASTs, which mimicked the human BBB [69].
Whole Body-on-a-Chip
In fact, a major challenge of microfluidic brain-on-a-chip technology is the nature of the method, which may be unable to provide mimicry of entire human organs. Many laboratory models lack the ability for pharmacokinetic studies including absorption, distribution, metabolism, and excretion (ADME) [157, 158]. The microfluidic organ-on-a-chip can emulate the biological function of organs such as bone marrow, spleen, gut, brain, and liver. Nevertheless, to predict the effects and toxicity of therapeutic agents, it is necessary to design and manufacture multi-organ-on-a-chip devices using microtechnology. Hence, researchers have designed and generated the “human-on-a-chip” that functions as whole body-on-a-chip. This has been called “homo chippiens.” These whole-body models are able to accelerate the pharmacokinetic and pharmacodynamic studies of experimental drugs using the multi-organ-on-a-chip in technology in comparison with microfluidic single-organ-on-chip devices [61, 159–163]. Thus, the whole body-on-a-chip has been designed to emulate multiple different organs such as brain, liver, lung, kidney, adipose tissue, bone marrow, and the heart [164–166].
Conclusions and Future Directions
NDDs affect tens of millions of people worldwide. Therefore, there is an essential need to establish advanced models and develop new therapeutic strategies to inhibit or slow the progression of these disorders. The use of 2D and 3D cell cultures and animal models cannot completely recapitulate the etiology and pathophysiology of NDDs because researchers need to study the biochemical effects of molecules on the microenvironment and architecture of neural tissues. 3D-based ECM microfluidic brain-on-a-chip devices can create a potential solution to allow the controlled manipulation, monitoring, and assessment of cells. The enablement of drug discovery on a microscale can facilitate high-throughput screening of large drug libraries. More recently, SC-based microfluidic brain-on-a-chip system have given rise to many neural system structures in the laboratory. Hence, the microfluidic brain-on-a-chip system is a novel and advanced technology, which can be utilized for NDD modeling in order to evaluate both normal and abnormal conditions of the CNS and the PNS for basic medical investigations and can also be used for therapeutic aims in clinical applications and personalized medicine.
Acknowledgments
Funding
M.R.H. was supported by US NIH Grants R01AI050875 and R21AI121700.
Abbreviations
- 2D
two-dimensional
- 3D
three-dimensional
- AD
Alzheimer’s disease
- ADME
adsorption, distribution, metabolism, excretion
- ALT
amyotrophic lateral sclerosis
- ASTs
astrocytes
- BBB
blood–brain barrier
- BECs
brain endothelial cells
- bFGF
basic fibroblast growth factor
- BMECs
brain microvascular endothelial cells
- BRAIN
Brain Research through Advancing Innovative Neurotechnologies
- CD
cluster of differentiation
- CNS
central nervous system
- CTIP2
chicken ovalbumin upstream promoter transcription factor-interacting protein 2
- DARPA
Defense Advanced Research Projects Agency
- DCX
doublecortin
- DOX
doxorubicin
- ECM
extracellular matrix
- ECs
endothelial cells
- EGCs
embryonic germ cells
- EGFR
epidermal growth factor receptor
- EGFR
epidermal growth factor
- ESCs
embryonic stem cells
- FDA
Food and Drug Administration
- FITC
fluorescein isothiocyanate
- FOXG1
forkhead box protein G1
- GBM
glioblastoma multiforme
- G-CSF
granulocyte colony-stimulating factor
- GFAP
glial fibrillary acidic protein
- hBMVECs
human brain microvascular endothelial cells
- HBP
Human Brain Project
- HD
Huntington’s disease
- hiPSCs
human-induced pluripotent stem cells
- HUVEC
human umbilical vein endothelial cells
- ISL1
insulin gene enhancer protein 1
- ITSS
insulin-transferrin–sodium selenite supplement
- IL-6
interleukin-6
- KROX20
early growth response 2 (egr2)
- LOC
laboratory-on-a-chip
- LPS
lipopolysaccharide
- MS
multiple sclerosis
- NCATS
National Center for Advancing Translational Sciences
- NDDs
neurodegenerative diseases
- NIH
National Institut6es of Health
- NPCs
neural progenitor cells
- NSCs
neural stem cells
- NSF
National Science Foundation
- NG2
neuron glial antigen 2
- NSPCs
neural stem/progenitor cells
- NVC
neurovascular chip
- PD
Parkinson’s disease
- PAX2/6
paired box gene 2/6
- PDMS
polydimethylsiloxane
- PEGDA
poly(ethylene) glycol diacrylate
- Pgp
P-glycoprotein
- PNS
peripheral nervous system
- PTEF
polytetrafluoroethylene
- PTEN
phosphatase and tensin homolog
- RT-PCR
real-time polymerase chain reaction
- SCs
stem cells
- SCZ
schizophrenia
- SEM
scanning electron microscopy
- SOX2
sex determining region Y-box 2
- SSEA
stage-specific embryonic antigen
- TBI
traumatic brain injury
- TBR1
T-box brain 1
- TEER
trans-endothelial electrical resistance
- TNF-α
tumor necrosis factor-alpha
- TUJ1
neuron-specific class III beta-tubulin
- TUNEL
terminal deoxynucleotidyl transferase (TdT) dUTP nick-end labeling
- ZO-1
zonula occludens-1
- GFP
green fluorescent protein
- hCMEC/D3
human cerebral microvascular endothelial cell
- HUVEC
human umbilical veinendothelial cell
Footnotes
Competing Interests M.R.H. is on the following Scientific Advisory Boards:
Transdermal Cap Inc., Cleveland, OH.
BeWell Global Inc., Wan Chai, Hong Kong.
Hologenix Inc., Santa Monica, CA.
LumiThera Inc., Poulsbo, WA.
Vielight, Toronto, Canada.
Bright Photomedicine, Sao Paulo, Brazil.
Quantum Dynamics LLC, Cambridge, MA.
Global Photon Inc., Bee Cave, TX.
Medical Coherence, Boston MA.
NeuroThera, Newark DE.
JOOVV Inc., Minneapolis-St. Paul, MN.
AIRx Medical, Pleasanton, CA.
FIR Industries, Inc. Ramsey, NJ.
UVLRx Therapeutics, Oldsmar, FL.
Ultralux UV Inc., Lansing MI.
Illumiheal & Petthera, Shoreline, WA.
MB Lasertherapy, Houston, TX.
ARRC LED, San Clemente, CA.
Varuna Biomedical Corp., Incline Village, NV.
Niraxx Light Therapeutics, Inc., Boston, MA.
M.R.H. has been a consultant for:
Lexington Int, Boca Raton, FL.
USHIO Corp, Japan.
Merck KGaA, Darmstadt, Germany.
Philips Electronics Nederland B.V.
Johnson & Johnson Inc., Philadelphia, PA.
Sanofi-Aventis Deutschland GmbH, Frankfurt am Main, Germany.
M.R.H. is a stockholder in:
Global Photon Inc., Bee Cave, TX.
Mitonix, Newark, DE.
References
- 1.Heemels MT (2016) Neurodegenerative diseases. Nature 539(7628):179 10.1038/539179a [DOI] [PubMed] [Google Scholar]
- 2.Matilla-Duenas A, Corral-Juan M, Rodriguez-Palmero Seuma A, Vilas D, Ispierto L, Morais S, Sequeiros J, Alonso I et al. (2017) Rare neurodegenerative diseases: clinical and genetic update. Adv Exp Med Biol 1031:443–496. 10.1007/978-3-319-67144-4_25 [DOI] [PubMed] [Google Scholar]
- 3.McColgan P, Tabrizi SJ (2018) Huntington’s disease: a clinical review. Eur J Neurol 25(1):24–34. 10.1111/ene.13413 [DOI] [PubMed] [Google Scholar]
- 4.Trovato Salinaro A, Pennisi M, Di Paola R, Scuto M, Crupi R, Cambria MT, Ontario ML, Tomasello M et al. (2018) Neuroinflammation and neurohormesis in the pathogenesis of Alzheimer’s disease and Alzheimer-linked pathologies: modulation by nutritional mushrooms. Immun Ageing 15:8 10.1186/s12979-017-0108-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Marsh SE, Blurton-Jones M (2017) Neural stem cell therapy for neurodegenerative disorders: the role of neurotrophic support. Neurochem Int 106:94–100. 10.1016/j.neuint.2017.02.006 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Noble W, Burns MP (2010) Challenges in neurodegeneration research. Front Psychiatry 1:7 10.3389/fpsyt.2010.00007 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Reardon S (2014) Brain-mapping projects to join forces. Nature:18 [Google Scholar]
- 8.Samuel A, Levine H, Blagoev KB (2013) Scientific priorities for the BRAIN initiative. Nat Methods 10(8):713–714. 10.1038/nmeth.2565 [DOI] [PubMed] [Google Scholar]
- 9.Logroscino G, Capozzo R, Tortelli R, Marin B (2016) Current issues in randomized clinical trials of neurodegenerative disorders at enrolment and reporting: diagnosis, recruitment, representativeness of patients, ethnicity, and quality of reporting In: The right therapy for neurological disorders, vol 39 Karger Publishers, pp. 24–36. 10.1159/000445410 [DOI] [PubMed] [Google Scholar]
- 10.Vagaska B, Ferretti P (2017) Toward modeling the human nervous system in a dish: recent progress and outstanding challenges. Regen Med 12(1):15–23 [DOI] [PubMed] [Google Scholar]
- 11.Bracken MB (2009) Why animal studies are often poor predictors of human reactions to exposure. J R Soc Med 102(3):120–122 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Mak IW, Evaniew N, Ghert M (2014) Lost in translation: animal models and clinical trials in cancer treatment. Am J Transl Res 6(2):114–118 [PMC free article] [PubMed] [Google Scholar]
- 13.Karimi M, Zare H, Bakhshian Nik A, Yazdani N, Hamrang M, Mohamed E, Sahandi Zangabad P, Moosavi Basri SM et al. (2016) Nanotechnology in diagnosis and treatment of coronary artery disease. Nanomedicine (London) 11(5):513–530. 10.2217/nnm.16.3 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Malekzad H, Mirshekari H, Sahandi Zangabad P, Moosavi Basri SM, Baniasadi F, Sharifi Aghdam M, Karimi M, Hamblin MR (2018) Plant protein-based hydrophobic fine and ultrafine carrier particles in drug delivery systems. Crit Rev Biotechnol 38(1):47–67. 10.1080/07388551.2017.1312267 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Sambale F, Lavrentieva A, Stahl F, Blume C, Stiesch M, Kasper C, Bahnemann D, Scheper T (2015) Three dimensional spheroid cell culture for nanoparticle safety testing. J Biotechnol 205:120–129. https://doi.Org/10.1016/j.jbiotec.2015.01.001 [DOI] [PubMed] [Google Scholar]
- 16.Kapałczyńska M, Kolenda T, Przybyła W, Zajączkowska M, Teresiak A, Filas V, Ibbs M, Bliźniak R et al. (2018) 2D and 3D cell cultures—a comparison of different types of cancer cell cultures. Arch Med Sci 14(4):910 10.5114/aoms.2016.63743 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Fang Y, Eglen RM (2017) Three-dimensional cell cultures in drug discovery and development. SLAS Discov 22(5):456–472. 10.1177/1087057117696795 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Hoarau-Véchot J, Rafii A, Touboul C, Pasquier J (2018) Halfway between 2D and animal models: are 3D cultures the ideal tool to study cancer–microenvironment interactions? Int J Mol Sci 19(1):181. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Jabbarzadegan M, Rajayi H, Mofazzal Jahromi MA, Yeganeh H, Yousefi M, Muhammad Hassan Z, Majidi J (2017) Application of arteether-loaded polyurethane nanomicelles to induce immune response in breast cancer model. Artif Cells Nanomed Biotechnol 45(4):808–816. 10.1080/21691401.2016.1178131 [DOI] [PubMed] [Google Scholar]
- 20.Farjadian F, Moghoofei M, Mirkiani S, Ghasemi A, Rabiee N, Hadifar S, Beyzavi A, Karimi M, Hamblin MR (2018) Bacterial components as naturally inspired nano-carriers for drug/gene delivery and immunization: set the bugs to work? Biotechnology Advances [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Garreta E, Oria R, Tarantino C, Pla-Roca M, Prado P, Fernández-Avilés F, Campistol JM, Samitier J et al. (2017) Tissue engineering by decellularization and 3D bioprinting. Mater Today 20(4):166–178. 10.1016/j.mattod.2016.12.005 [DOI] [Google Scholar]
- 22.Hong N, Yang GH, Lee J, Kim G (2018) 3D bioprinting and its in vivo applications. J Biomed Mater Res B Appl Biomater 106(1):444–459. 10.1002/jbm.b33826 [DOI] [PubMed] [Google Scholar]
- 23.Karimi M, M Moosavi Basri S, Vossoughi M, S Pakchin P, Mirshekari H, R Hamblin M (2016) Redox-sensitive smart nanosystems for drug and gene delivery. Curr Org Chem 20(28):2949–2959 [Google Scholar]
- 24.Tsutsui K, Taira M, Sakata H (2005) Neural mechanisms of three-dimensional vision. Neurosci Res 51(3):221–229. 10.1016/j.neures.2004.11.006 [DOI] [PubMed] [Google Scholar]
- 25.Dutta D, Heo I, Clevers H (2017) Disease modeling in stem cell-derived 3D organoid systems. Trends Mol Med 23(5):393–410. 10.1016/j.molmed.2017.02.007 [DOI] [PubMed] [Google Scholar]
- 26.Wang Z, Wang SN, Xu TY, Miao ZW, Su DF, Miao CY (2017) Organoid technology for brain and therapeutics research. CNS Neurosci Ther 23(10):771–778. 10.1111/cns.12754 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Willyard C (2015) The boom in mini stomachs, brains, breasts, kidneys and more. Nature 523(7562):520–522. 10.1038/523520a [DOI] [PubMed] [Google Scholar]
- 28.Aebersold MJ, Dermutz H, Forró C, Weydert S, Thompson-Steckel G, Vörös J, Demkó L (2016) “Brains on a chip”: towards engineered neural networks. TrAC Trends Anal Chem 78:60–69 [Google Scholar]
- 29.Park J, Wetzel I, Dreau D, Cho H (2018) 3D miniaturization of human organs for drug discovery. Adv healthc Mater 7(2). 10.1002/adhm.201700551 [DOI] [PubMed] [Google Scholar]
- 30.Bhatia SN, Ingber DE (2014) Microfluidic organs-on-chips. Nat Biotechnol 32(8):760–772. 10.1038/nbt.2989 [DOI] [PubMed] [Google Scholar]
- 31.Jahromi MAM, Zangabad PS, Basri SMM, Zangabad KS, Ghamarypour A, Aref AR, Karimi M, Hamblin MR (2017) Nanomedicine and advanced technologies for burns: preventing infection and facilitating wound healing. Advanced Drug Delivery Reviews [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Gjorevski N, Nelson CM (2010) The mechanics of development: models and methods for tissue morphogenesis. Birth Defects Res C Embryo Today 90(3):193–202. 10.1002/bdrc.20185 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Mammoto T, Ingber DE (2010) Mechanical control of tissue and organ development. Development 137(9):1407–1420. 10.1242/dev.024166 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Auluck PK, Chan HE, Trojanowski JQ, Lee VM-Y, Bonini NM (2002) Chaperone suppression of α-synuclein toxicity in a Drosophila model for Parkinson’s disease. Science 295(5556):865–868 [DOI] [PubMed] [Google Scholar]
- 35.Becker LA, Huang B, Bieri G, Ma R, Knowles DA, Jafar-Nejad P, Messing J, Kim HJ et al. (2017) Therapeutic reduction of ataxin-2 extends lifespan and reduces pathology in TDP-43 mice. Nature 544(7650):367–371 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Link CD (1995) Expression of human beta-amyloid peptide in transgenic Caenorhabditis elegans. Proc Natl Acad Sci 92(20):9368–9372 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Park J, Lee BK, Jeong GS, Hyun JK, Lee CJ, Lee S-H (2015) Three-dimensional brain-on-a-chip with an interstitial level of flow and its application as an in vitro model of Alzheimer’s disease. Lab Chip 15(1):141–150 [DOI] [PubMed] [Google Scholar]
- 38.Li M, Izpisua Belmonte JC (2019) Organoids—preclinical models of human disease. N Engl J Med 380(6):569–579. 10.1056/NEJMra1806175 [DOI] [PubMed] [Google Scholar]
- 39.El-Ali J, Sorger PK, Jensen KF (2006) Cells on chips. Nature 442(7101):403–411 [DOI] [PubMed] [Google Scholar]
- 40.Esch EW, Bahinski A, Huh D (2015) Organs-on-chips at the frontiers of drug discovery. Nat Rev Drug Discov 14(4):248–260 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Kilic O, Pamies D, Lavell E, Schiapparelli P, Feng Y, Hartung T, Bal-Price A, Hogberg HT et al. (2016) Brain-on-a-chip model enables analysis of human neuronal differentiation and chemotaxis. Lab Chip 16(21):4152–4162 [DOI] [PubMed] [Google Scholar]
- 42.Liu Y, Gill E, Shery Huang YY (2017) Microfluidic on-chip biomimicry for 3D cell culture: a fit-for-purpose investigation from the end user standpoint. Future Sci OA 3(2):FSO173 10.4155/fsoa-2016-0084 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Perez-Toralla K, Mottet G, Tulukcuoglu-Guneri E, Champ J, Bidard FC, Pierga JY, Klijanienko J, Draskovic I et al. (2017) FISH-in-CHIPS: a microfluidic platform for molecular typing of cancer cells. Methods Mol Biol 1547:211–220. 10.1007/978-1-4939-6734-6_16 [DOI] [PubMed] [Google Scholar]
- 44.Temiz Y, Lovchik RD, Kaigala GV, Delamarche E (2015) Lab-on-a-chip devices: how to close and plug the lab? Microelectron Eng 132:156–175 [Google Scholar]
- 45.Whitesides GM (2006) The origins and the future of microfluidics. Nature 442(7101):368–373. 10.1038/nature05058 [DOI] [PubMed] [Google Scholar]
- 46.Zheng F, Fu F, Cheng Y, Wang C, Zhao Y, Gu Z (2016) Organ-on-a-chip systems: microengineering to biomimic living systems. Small 12(17):2253–2282. 10.1002/smll.201503208 [DOI] [PubMed] [Google Scholar]
- 47.Logun M, Zhao W, Mao L, Karumbaiah L (2018) Microfluidics in malignant glioma research and precision medicine. Adv Biosyst 2(5):1700221. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Sosa-Hernández JE, Villalba-Rodríguez AM, Romero-Castillo KD, Aguilar-Aguila-Isaías MA, Garcia-Reyes IE, Hernández-Antonio A, Ahmed I, Sharma A et al. (2018) Organs-on-a-chip module: a review from the development and applications perspective. Micromachines 9(10):536. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Yu Y, Shang L, Guo J, Wang J, Zhao Y (2018) Design of capillary microfluidics for spinning cell-laden microfibers. Nat Protoc:1 [DOI] [PubMed] [Google Scholar]
- 50.Sosa-Hernandez JE, Villalba-Rodriguez AM, Romero-Castillo KD, Aguilar-Aguila-Isaias MA, Garcia-Reyes IE, Hernandez-Antonio A, Ahmed I, Sharma A et al. (2018) Organs-on-a-chip module: a review from the development and applications perspective. Micromachines (Basel) 9(10). 10.3390/mi9100536 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Haring AP, Sontheimer H, Johnson BN (2017) Microphysiological human brain and neural systems-on-a-chip: potential alternatives to small animal models and emerging platforms for drug discovery and personalized medicine. Stem Cell Rev 13(3):381–406.Error! Hyperlink reference not valid. 10.1007/s12015-017-9738-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Qin D, Xia Y, Whitesides GM (2010) Soft lithography for micro- and nanoscale patterning. Nat Protoc 5(3):491–502 [DOI] [PubMed] [Google Scholar]
- 53.Whitesides GM, Ostuni E, Takayama S, Jiang X, Ingber DE (2001) Soft lithography in biology and biochemistry. Annu Rev Biomed Eng 3(1):335–373 [DOI] [PubMed] [Google Scholar]
- 54.Xia Y, Whitesides GM (1998) Soft lithography. Angew Chem Int Ed 37(5):550–575 [DOI] [PubMed] [Google Scholar]
- 55.Huh D, Kim HJ, Fraser JP, Shea DE, Khan M, Bahinski A, Hamilton GA, Ingber DE (2013) Microfabrication of human organs-on-chips. Nat Protoc 8(11):2135–2157. 10.1038/nprot.2013.137 [DOI] [PubMed] [Google Scholar]
- 56.Mancera-Andrade EI, Parsaeimehr A, Arevalo-Gallegos A, Ascencio-Favela G, Parra Saldivar R (2018) Microfluidics technology for drug delivery: a review. Front Biosci (Elite Ed) 10:74–91 [DOI] [PubMed] [Google Scholar]
- 57.Miccoli B, Braeken D, Ethan Li Y-C (2018) Brain-on-a-chip devices for drug screening and disease modeling applications. Curr Pharm Des [DOI] [PubMed] [Google Scholar]
- 58.Musick K, Khatami D, Wheeler BC (2009) Three-dimensional micro-electrode array for recording dissociated neuronal cultures. Lab Chip 9(14):2036–2042 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Queval A, Ghattamaneni NR, Perrault CM, Gill R, Mirzaei M, McKinney RA, Juncker D (2010) Chamber and microfluidic probe for microperfusion of organotypic brain slices. Lab Chip 10(3):326–334 [DOI] [PubMed] [Google Scholar]
- 60.Reardon S (2015) Scientists seek ‘Homo chippiens. Nature 518(7539):285–286 [DOI] [PubMed] [Google Scholar]
- 61.Baker M (2011) Tissue models: a living system on a chip. Nature 471(7340):661–665. 10.1038/471661a [DOI] [PubMed] [Google Scholar]
- 62.Reardon S (2015) Organs-on-chips’ go mainstream. Nature 523(7560):266 10.1038/523266a [DOI] [PubMed] [Google Scholar]
- 63.Zhang B, Korolj A, Lai BFL, Radisic M (2018) Advances in organ-on-a-chip engineering. Nat Rev Mater:1 [Google Scholar]
- 64.Park J, Kim S, Park SI, Choe Y, Li J,Han A (2014) A microchip for quantitative analysis of CNS axon growth under localized biomolecular treatments. J Neurosci Methods 221:166–174 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Lu X, Kim-Han JS, O’Malley KL, Sakiyama-Elbert SE (2012) A microdevice platform for visualizing mitochondrial transport in aligned dopaminergic axons. J Neurosci Methods 209(1):35–39 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Moreno EL, Hachi S, Hemmer K, Trietsch SJ, Baumuratov AS, Hankemeier T, Vulto P, Schwamborn JC et al. (2015) Differentiation of neuroepithelial stem cells into functional dopaminergic neurons in 3D microfluidic cell culture. Lab Chip 15(11 ):2419–2428 [DOI] [PubMed] [Google Scholar]
- 67.Achyuta AKH, Conway AJ, Crouse RB, Bannister EC, Lee RN, Katnik CP, Behensky AA, Cuevas J et al. (2013) A modular approach to create a neurovascular unit-on-a-chip. Lab Chip 13(4):542–553 [DOI] [PubMed] [Google Scholar]
- 68.Wang Y, Ma J, Li N, Wang L, Shen L, Sun Y, Wang Y, Zhao J et al. (2017) Microfluidic engineering of neural stem cell niches for fate determination. Biomicrofluidics 11(1):014106 10.1063/1.4974902 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Adriani G, Ma D, Pavesi A, Kamm RD, Goh EL (2017) A 3D neurovascular microfluidic model consisting of neurons, astrocytes and cerebral endothelial cells as a blood–brain barrier. Lab Chip 17(3):448–459 [DOI] [PubMed] [Google Scholar]
- 70.Brown JA, Codreanu SG, Shi M, Sherrod SD, Markov DA, Neely MD, Britt CM, Hoilett OS et al. (2016) Metabolic consequences of inflammatory disruption of the blood–brain barrier in an organ-on-chip model of the human neurovascular unit. J Neuroinflammation 13(1):306. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Griep L, Wolbers F, De Wagenaar B, ter Braak PM, Weksler B, Romero IA, Couraud P, Vermes I et al. (2013) BBB on chip: microfluidic platform to mechanically and biochemically modulate blood–brain barrier function. Biomed Microdevices 15(1):145–150 [DOI] [PubMed] [Google Scholar]
- 72.Herland A, van der Meer AD, FitzGerald EA, Park T-E, Sleeboom JJ, Ingber DE (2016) Distinct contributions of astrocytes and pericytes to neuroinflammation identified in a 3D human blood–brain barrier on a chip. PLoS One 11(3):e0150360. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Sellgren KL, Hawkins BT, Grego S (2015) An optically transparent membrane supports shear stress studies in a three-dimensional microfluidic neurovascular unit model. Biomicrofluidics 9(6):061102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Wang YI, Abaci HE, Shuler ML (2017) Microfluidic blood–brain barrier model provides in vivo-like barrier properties for drug permeability screening. Biotechnol Bioeng 114(1):184–194 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Ruiz A, Joshi P, Mastrangelo R, Francolini M, Verderio C, Matteoli M (2014) Testing Aβ toxicity on primary CNS cultures using drugscreening microfluidic chips. Lab Chip 14(15):2860–2866 [DOI] [PubMed] [Google Scholar]
- 76.Fan Y, Nguyen DT, Akay Y, Xu F, Akay M (2016) Engineering a brain cancer chip for high-throughput drug screening. Sci Rep 6:25062. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Sun J, Masterman-Smith MD, Graham NA, Jiao J, Mottahedeh J, Laks DR, Ohashi M, DeJesus J et al. (2010) A microfluidic platform for systems pathology: multiparameter single-cell signaling measurements of clinical brain tumor specimens. Cancer Res 70(15):6128–6138 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Hidalgo San Jose L, Stephens P, Song B, Barrow D (2018) Microfluidic encapsulation supports stem cell viability, proliferation, and neuronal differentiation. Tissue Eng Part C Methods 24:158–170. 10.1089/ten.TEC.2017.0368 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Wang Y, Wang L, Guo Y, Zhu Y, Qin J (2018) Engineering stem cell-derived 3D brain organoids in a perfusable organ-on-a-chip system. RSC Adv 8(3):1677–1685 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Wang Y, Wang L, Zhu Y, Qin J (2018) Human brain organoid-on-a-chip to model prenatal nicotine exposure. Lab Chip 18(6):851–860. 10.1039/c7lc01084b [DOI] [PubMed] [Google Scholar]
- 81.MacKerron C, Robertson G, Zagnoni M, Bushell TJ (2017) A microfluidic platform for the characterisation of CNS active compounds. Sci Rep 7(1):15692. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Osaki T, Sivathanu V, Kamm RD (2018) Engineered 3D vascular and neuronal networks in a microfluidic platform. Sci Rep 8(1):5168. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Sandlin ZD, Shou M, Shackman JG, Kennedy RT (2005) Microfluidic electrophoresis chip coupled to microdialysis for in vivo monitoring of amino acid neurotransmitters. Anal Chem 77(23):7702–7708 [DOI] [PubMed] [Google Scholar]
- 84.Kelava I, Lancaster MA (2016) Stem cell models of human brain development. Cell Stem Cell 18(6):736–748. 10.1016/j.stem.2016.05.022 [DOI] [PubMed] [Google Scholar]
- 85.Qian T, Shusta EV, Palecek SP (2015) Advances in microfluidic platforms for analyzing and regulating human pluripotent stem cells. Curr Opin Genet Dev 34:54–60 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Zhang J, Wei X, Zeng R, Xu F, Li X (2017) Stem cell culture and differentiation in microfluidic devices toward organ-on-a-chip. Future Sci OA 3(2):FSO187 10.4155/fsoa-2016-0091 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Mammoto A, Mammoto T, Ingber DE (2012) Mechanosensitive mechanisms in transcriptional regulation. J Cell Sci 125 (Pt 13:3061–3073. 10.1242/jcs.093005 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Adegbola A, Bury LA, Fu C, Zhang M, Wynshaw-Boris A (2017) Concise review: induced pluripotent stem cell models for neuropsychiatric diseases. Stem Cells Transl Med 6(12):2062–2070. 10.1002/sctm.17-0150 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Alvarez CV, Garcia-Lavandeira M, Garcia-Rendueles ME, Diaz-Rodriguez E, Garcia-Rendueles AR, Perez-Romero S, Vila TV, Rodrigues JS et al. (2012) Defining stem cell types: understanding the therapeutic potential of ESCs, ASCs, and iPS cells. J Mol Endocrinol 49(2):R89–R111. 10.1530/JME-12-0072 [DOI] [PubMed] [Google Scholar]
- 90.De Filippis L, Zalfa C, Ferrari D (2017) Neural stem cells and human induced pluripotent stem cells to model rare CNS diseases. CNS Neurol Disord Drug Targets 16(8):915–926. 10.2174/1871527316666170615121753 [DOI] [PubMed] [Google Scholar]
- 91.Fong ELS, Yu H (2017) Organs-on-chips: filtration enabled by differentiation. Nat Biomed Eng 1(5):0074 [Google Scholar]
- 92.Kornblum HI (2007) Introduction to neural stem cells. Stroke 38(2 Suppl):810–816. 10.1161/01.STR.0000255757.12198.0f [DOI] [PubMed] [Google Scholar]
- 93.Wang Y, Zhao C, Hou Z, Yang Y, Bi Y, Wang H, Zhang Y, Gao S (2018) Unique molecular events during reprogramming of human somatic cells to induced pluripotent stem cells (iPSCs) at naive state. Elife 7 10.7554/eLife.29518 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Wang S, Wu J, Liu GH (2018) First stem cell transplantation to regenerate human lung. Protein Cell 9(3):244–245. 10.1007/s13238-017-0498-z [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Karimi M, Bahrami S, Mirshekari H, Basri SM, Nik AB, Aref AR, Akbari M, Hamblin MR (2016) Microfluidic systems for stem cell-based neural tissue engineering. Lab Chip 16(14):2551–2571. 10.1039/c6lc00489j [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Pamies D, Hartung T, Hogberg HT (2014) Biological and medical applications of a brain-on-a-chip. Exp Biol Med (Maywood) 239(9):1096–1107. 10.1177/1535370214537738 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Prajumwongs P, Weeranantanapan O, Jaroonwitchawan T, Noisa P (2016) Human embryonic stem cells: a model for the study of neural development and neurological diseases. Stem Cells Int 2016:2958210 10.1155/2016/2958210 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Takayama Y, Kida YS (2016) In vitro reconstruction of neuronal networks derived from human iPS cells using microfabricated devices. PLoS One 11(2):e0148559 10.1371/joumal.pone.0148559 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Lupo G, Nisi PS, Esteve P, Paul YL, Novo CL, Sidders B, Khan MA, Biagioni S et al. (2018) Molecular profiling of aged neural progenitors identifies Dbx2 as a candidate regulator of age-associated neurogenic decline. Aging Cell 17 10.1111/acel.12745 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Shu P, Fu H, Zhao X, Wu C, Ruan X, Zeng Y, Liu W, Wang M et al. (2017) MicroRNA-214 modulates neural progenitor cell differentiation by targeting quaking during cerebral cortex development. Sci Rep 7(1):8014 10.1038/s41598-017-08450-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Jakel S, Dimou L (2017) Glial cells and their function in the adult brain: a journey through the history of their ablation. Front Cell Neurosci 11:24. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Sacco R, Cacci E, Novarino G (2018) Neural stem cells in neuropsychiatric disorders. Curr Opin Neurobiol 48:131–138. 10.1016/j.conb.2017.12.005 [DOI] [PubMed] [Google Scholar]
- 103.Wang Z, Luo Y, Chen L, Liang W (2017) Safety of neural stem cell transplantation in patients with severe traumatic brain injury. Exp Ther Med 13(6):3613–3618. 10.3892/etm.2017.4423 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Kim WT, Ryu CJ (2017) Cancer stem cell surface markers on normal stem cells. BMB Rep 50(6):285–298 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Kopach O, Rybachuk O, Krotov V, Kyryk V, Voitenko N, Pivneva T (2018) Maturation of neural stem cells and integration into hippocampal circuits: functional study in post-ischemia in situ. J Cell Sci 131 :jcs. 210989. [DOI] [PubMed] [Google Scholar]
- 106.Jessberger S (2016) Stem cell-mediated regeneration of the adult brain. Transfus Med Hemother 43(5):321–326. 10.1159/000447646 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Wang B, Jedlicka S, Cheng X (2014) Maintenance and neuronal cell differentiation of neural stem cells C17. 2 correlated to medium availability sets design criteria in microfluidic systems. PLoS One 9(10):e109815. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Barros CS, Franco SJ, Muller U (2011) Extracellular matrix: functions in the nervous system. Cold Spring Harb Perspect Biol 3(1):a005108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Bonneh-Barkay D, Wiley CA (2009) Brain extracellular matrix in neurodegeneration. Brain Pathol 19(4):573–585 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Lepelletier FX, Mann D, Robinson A, Pinteaux E, Boutin H (2017) Early changes in extracellular matrix in Alzheimer’s disease. Neuropathol Appl Neurobiol 43(2):167–182 [DOI] [PubMed] [Google Scholar]
- 111.Berretta S (2012) Extracellular matrix abnormalities in schizophrenia. Neuropharmacology 62(3):1584–1597 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Leslie SK, Kinney RC, Schwartz Z, Boyan BD (2017) Microencapsulation of stem cells for therapy In: Cell Microencapsulation. Springer, pp. 251–259 [DOI] [PubMed] [Google Scholar]
- 113.Vitrac A, Cloez-Tayarani I (2018) Induced pluripotent stem cells as a tool to study brain circuits in autism-related disorders. Stem Cell Res Ther 9(1):226. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Koo Y, Hawkins BT, Yun Y (2018) Three-dimensional (3D) tetra-culture brain on chip platform for organophosphate toxicity screening. Sci Rep 8(1):2841 10.1038/s41598-018-20876-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Parr CJC, Yamanaka S, Saito H (2017) An update on stem cell biology and engineering for brain development. Mol Psychiatry 22(6):808–819. 10.1038/mp.2017.66 [DOI] [PubMed] [Google Scholar]
- 116.Qian X, Jacob F, Song MM, Nguyen HN, Song H, Ming GL (2018) Generation of human brain region-specific organoids using a miniaturized spinning bioreactor. Nat Protoc 13(3):565–580. 10.1038/nprot.2017.152 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Hartley BJ, Brennand KJ (2017) Neural organoids for disease phenotyping, drug screening and developmental biology studies. Neurochem Int 106:85–93. 10.1016/j.neuint.2016.10.004 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Hunsberger JG, Efthymiou AG, Malik N, Behl M, Mead IL, Zeng X, Simeonov A, Rao M (2015) Induced pluripotent stem cell models to enable in vitro models for screening in the central nervous system. Stem Cells Dev 24(16):1852–1864. 10.1089/scd.2014.0531 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Tong G, Izquierdo P, Raashid RA (2017) Human induced pluripotent stem cells and the modelling of Alzheimer’s disease: the human brain outside the dish. Open Neurol J 11:27–38. 10.2174/1874205X01711010027 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Wei T-Y, Fu Y, Chang K-H, Lin K-J, Lu Y-J, Cheng C-M (2017) Point-of-care devices using disease biomarkers to diagnose neurodegenerative disorders. Trends Biotechnol [DOI] [PubMed] [Google Scholar]
- 121.Fyfe I (2018) Alzheimer disease: epigenetics links ageing with Alzheimer disease. Nat Rev Neurol. 10.1038/nmeurol.2018.36 [DOI] [PubMed] [Google Scholar]
- 122.Goldman JS, Hahn SE, Catania JW, LaRusse-Eckert S, Butson MB, Rumbaugh M, Strecker MN, Roberts JS et al. (2011) Genetic counseling and testing for Alzheimer disease: joint practice guidelines of the American College of Medical Genetics and the National Society of Genetic Counselors. Genet Med 13(6):597–605. 10.1097/GIM.0b013e31821d69b8 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.De Felice FG, Munoz DP (2016) Opportunities and challenges in developing relevant animal models for Alzheimer’s disease. Ageing Res Rev 26:112–114 [DOI] [PubMed] [Google Scholar]
- 124.Talbot K (2002) Motor neurone disease. Postgrad Med J 78(923):513–519 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Abbott NJ (2013) Blood–brain barrier structure and function and the challenges for CNS drug delivery. J Inherit Metab Dis 36(3):437–449 [DOI] [PubMed] [Google Scholar]
- 126.Yamamizu K, Iwasaki M, Takakubo H, Sakamoto T, Ikuno T, Miyoshi M, Kondo T, Nakao Y et al. (2017) In vitro modeling of blood–brain barrier with human iPSC-derived endothelial cells, pericytes, neurons, and astrocytes via notch signaling. Stem Cell Reports 8(3):634–647. 10.1016/j.stemcr.2017.01.023 [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 127.Daneman R (2012) The blood–brain barrier in health and disease. Ann Neurol 72(5):648–672 [DOI] [PubMed] [Google Scholar]
- 128.Ballabh P, Braun A, Nedergaard M (2004) The blood–brain barrier: an overview: structure, regulation, and clinical implications. NeurobiolDis 16(1):1–13 [DOI] [PubMed] [Google Scholar]
- 129.Chernykh I, Yakusheva E, Shulkin A (2015) P-Glycoprotein expression in blood–brain barrier in bilateral occlusion of the common carotid artery. Nauchnye vedomosti Belgorodskogo gosudarstvennogo universiteta Seriya: Medicina Farmaciya 29(4):91–95 [Google Scholar]
- 130.van der Helm MW, van der Meer AD, Eijkel JC, van den Berg A, Segerink LI (2016) Microfluidic organ-on-chip technology for blood–brain barrier research. Tissue barriers 4(1):e1142493. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Wolff A, Antfolk M, Brodin B, Tenje M (2015) In vitro blood–brain barrier models—an overview of established models and new microfluidic approaches. J Pharm Sci 104(9):2727–2746 [DOI] [PubMed] [Google Scholar]
- 132.Huh D, Torisawa Y-s, Hamilton GA, Kim HJ, Ingber DE (2012) Microengineered physiological biomimicry: organs-on-chips. Lab Chip 12(12):2156–2164 [DOI] [PubMed] [Google Scholar]
- 133.Perrin S (2014) Preclinical research: make mouse studies work. Nature 507(7493):423–425 [DOI] [PubMed] [Google Scholar]
- 134.Alcendor DJ, Block FE 3rd, Cliffel DE, Daniels JS, Ellacott KL, Goodwin CR, Hofmeister LH, Li D et al. (2013) Neurovascular unit on a chip: implications for translational applications. Stem Cell Res Ther 4(Suppl 1):S18 10.1186/scrt379 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Adriani G, Ma D, Pavesi A, Goh E, Kamm R (2015) Modeling the blood–brain barrier in a 3D triple co-culture microfluidic system. In: Engineering in medicine and biology society (EMBC), 2015 37th annual international conference of the IEEE IEEE, pp. 338–341 [DOI] [PubMed] [Google Scholar]
- 136.Bergink V, Gibney SM, Drexhage HA (2014) Autoimmunity, inflammation, and psychosis: a search for peripheral markers. Biol Psychiatry 75(4):324–331 [DOI] [PubMed] [Google Scholar]
- 137.Khandaker GM, Cousins L, Deakin J, Lennox BR, Yolken R, Jones PB (2015) Inflammation and immunity in schizophrenia: implications for pathophysiology and treatment. Lancet Psychiatry 2(3):258–270 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Yesil-Celiktas O, Hassan S, Miri AK, Maharjan S, Al-kharboosh R, Quiñones-Hinojosa A, Zhang YS (2018) Mimicking human pathophysiology in organ-on-chip devices. Adv Biosyst 2:1800109 10.1002/adbi.201800109 [DOI] [Google Scholar]
- 139.Fischbach C, Chen R, Matsumoto T, Schmelzle T, Brugge JS, Polverini PJ, Mooney DJ (2007) Engineering tumors with 3D scaffolds. Nat Methods 4(10):855–860 [DOI] [PubMed] [Google Scholar]
- 140.Unger C, Kramer N, Walzl A, Scherzer M, Hengstschlager M, Dolznig H (2014) Modeling human carcinomas: physiologically relevant 3D models to improve anti-cancer drug development. Adv Drug Deliv Rev 79:50–67 [DOI] [PubMed] [Google Scholar]
- 141.Lancaster MA, Renner M, Martin CA, Wenzel D, Bicknell LS, Hurles ME, Homfray T, Penninger JM et al. (2013) Cerebral organoids model human brain development and microcephaly. Nature 501(7467):373–379. 10.1038/nature12517 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Pickl M, Ries C (2009) Comparison of 3D and 2D tumor models reveals enhanced HER2 activation in 3D associated with an increased response to trastuzumab. Oncogene 28(3):461–468 [DOI] [PubMed] [Google Scholar]
- 143.Tanner K, Gottesman MM (2015) Beyond 3D culture models of cancer. Sci Transl Med 7(283):283ps289–283ps289 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Cosson S, Lutolf M (2014) Hydrogel microfluidics for the patterning of pluripotent stem cells. Sci Rep 4 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Loessner D, Stok KS, Lutolf MP, Hutmacher DW, Clements JA, Rizzi SC (2010) Bioengineered 3D platform to explore cell-ECM interactions and drug resistance of epithelial ovarian cancer cells. Biomaterials 31(32):8494–8506 [DOI] [PubMed] [Google Scholar]
- 146.Laquintana V, Trapani A, Denora N, Wang F, Gallo JM, Trapani G (2009) New strategies to deliver anticancer drugs to brain tumors. Expert Opinion On Drug Delivery 6(10):1017–1032 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Altemus M, Leung B, Morikawa A, Dziubinski M, Castro M, Merajver S (2016) Novel microfluidic blood–brain niche to study breast cancer metastasis to the brain. AACR [Google Scholar]
- 148.Keng PY, Chen S, Ding H, Sadeghi S, Shah GJ, Dooraghi A, Phelps ME, Satyamurthy N et al. (2012) Micro-chemical synthesis of molecular probes on an electronic microfluidic device. Proc Natl Acad Sci U S A 109(3):690–695. 10.1073/pnas.1117566109 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Low LA, Tagle DA (2017) Organs-on-chips: progress, challenges, and future directions. Exp Biol Med 242(16):1573–1578 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Haehnel V, Khan FZ, Mutschke G, Cierpka C, Uhlemann M, Fritsch I (2019) Combining magnetic forces for contactless manipulation of fluids in microelectrode-microfluidic systems. Sci Rep 9(1):5103 10.1038/s41598-019-41284-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Obien MEJ, Deligkaris K, Bullmann T, Bakkum DJ, Frey U (2015) Revealing neuronal function through microelectrode array recordings. Front Neurosci 8:423. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Kane KIW, Moreno EL, Hachi S, Walter M, Jarazo J, Oliveira MAP, Hankemeier T, Vulto P et al. (2019) Automated microfluidic cell culture of stem cell derived dopaminergic neurons. Sci Rep 9(1):1796 10.1038/s41598-018-34828-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Kim S, Kim W, Lim S, Jeon JS (2017) Vasculature-on-a-chip for in vitro disease models. Bioengineering (Basel) 4(1). 10.3390/bioengineering4010008 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.Sontheimer-Phelps A, Hassell BA, Ingber DE (2019) Modelling cancer in microfluidic human organs-on-chips. Nat Rev Cancer 19(2):65–81. 10.1038/s41568-018-0104-6 [DOI] [PubMed] [Google Scholar]
- 155.Blinder YJ, Freiman A, Raindel N, Mooney DJ, Levenberg S (2015) Vasculogenic dynamics in 3D engineered tissue constructs. Sci Rep 5:17840 10.1038/srep17840 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Wang X, Sun Q, Pei J (2018) Microfluidic-based 3D engineered microvascular networks and their applications in vascularized microtumor models. Micromachines (Basel) 9(10). 10.3390/mi9100493 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157.Lee SH, Sung JH (2017) Microtechnology-based multi-organ models. Bioengineering (Basel) 4(2). 10.3390/bioengineering4020046 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158.Lee SH, Ha SK, Choi I, Choi N, Park TH, Sung JH (2016) Microtechnology-based organ systems and whole-body models for drug screening. Biotechnol J 11(6):746–756. 10.1002/biot.201500551 [DOI] [PubMed] [Google Scholar]
- 159.An F, Qu Y, Liu X, Zhong R, Luo Y (2015) Organ-on-a-chip: new platform for biological analysis. Anal Chem Insights 10:39–45. 10.4137/ACI.S28905 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160.Karimi M, Zangabad PS, Mehdizadeh F, Malekzad H, Ghasemi A, Bahrami S, Zare H, Moghoofei M et al. (2017) Nanocaged platforms: modification, drug delivery and nanotoxicity. Opening synthetic cages to release the tiger. Nanoscale 9(4):1356–1392. 10.1039/c6nr07315h [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161.Kimura H, Sakai Y, Fujii T (2018) Organ/body-on-a-chip based on microfluidic technology for drug discovery. Drug Metab Pharmacokinet 33(1):43–48. 10.1016/j.dmpk.2017.11.003 [DOI] [PubMed] [Google Scholar]
- 162.Lee SH, Sung JH (2018) Organ-on-a-chip technology for reproducing multiorgan physiology. Adv healthc Mater 7(2). 10.1002/adhm.201700419 [DOI] [PubMed] [Google Scholar]
- 163.Yum K, Hong SG, Healy KE, Lee LP (2014) Physiologically relevant organs on chips. Biotechnol J 9(1):16–27. 10.1002/biot.201300187 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164.Perestrelo AR, Águas AC, Rainer A, Forte G (2015) Microfluidic organ/body-on-a-chip devices at the convergence of biology and microengineering. Sensors 15(12):31142–31170 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165.Skardal A, Murphy SV, Devarasetty M, Mead I, Kang HW, Seol YJ, Shrike Zhang Y, Shin SR et al. (2017) Multi-tissue interactions in an integrated three-tissue organ-on-a-chip platform. Sci Rep 7(1):8837 10.1038/s41598-017-08879-x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166.Sung JH, Esch MB, Prot J- M, Long CJ, Smith A, Hickman JJ, Shuler ML (2013) Microfabricated mammalian organ systems and their integration into models of whole animals and humans. Lab Chip 13(7):1201–1212 [DOI] [PMC free article] [PubMed] [Google Scholar]
