Skip to main content
Erschienen in: Journal of Neuroinflammation 1/2016

Open Access 01.12.2016 | Research

Metabolic consequences of inflammatory disruption of the blood-brain barrier in an organ-on-chip model of the human neurovascular unit

verfasst von: Jacquelyn A. Brown, Simona G. Codreanu, Mingjian Shi, Stacy D. Sherrod, Dmitry A. Markov, M. Diana Neely, Clayton M. Britt, Orlando S. Hoilett, Ronald S. Reiserer, Philip C. Samson, Lisa J. McCawley, Donna J. Webb, Aaron B. Bowman, John A. McLean, John P. Wikswo

Erschienen in: Journal of Neuroinflammation | Ausgabe 1/2016

Abstract

Background

Understanding blood-brain barrier responses to inflammatory stimulation (such as lipopolysaccharide mimicking a systemic infection or a cytokine cocktail that could be the result of local or systemic inflammation) is essential to understanding the effect of inflammatory stimulation on the brain. It is through the filter of the blood-brain barrier that the brain responds to outside influences, and the blood-brain barrier is a critical point of failure in neuroinflammation. It is important to note that this interaction is not a static response, but one that evolves over time. While current models have provided invaluable information regarding the interaction between cytokine stimulation, the blood-brain barrier, and the brain, these approaches—whether in vivo or in vitro—have often been only snapshots of this complex web of interactions.

Methods

We utilize new advances in microfluidics, organs-on-chips, and metabolomics to examine the complex relationship of inflammation and its effects on blood-brain barrier function ex vivo and the metabolic consequences of these responses and repair mechanisms. In this study, we pair a novel dual-chamber, organ-on-chip microfluidic device, the NeuroVascular Unit, with small-volume cytokine detection and mass spectrometry analysis to investigate how the blood-brain barrier responds to two different but overlapping drivers of neuroinflammation, lipopolysaccharide and a cytokine cocktail of IL-1β, TNF-α, and MCP1,2.

Results

In this study, we show that (1) during initial exposure to lipopolysaccharide, the blood-brain barrier is compromised as expected, with increased diffusion and reduced presence of tight junctions, but that over time, the barrier is capable of at least partial recovery; (2) a cytokine cocktail also contributes to a loss of barrier function; (3) from this time-dependent cytokine activation, metabolic signature profiles can be obtained for both the brain and vascular sides of the blood-brain barrier model; and (4) collectively, we can use metabolite analysis to identify critical pathways in inflammatory response.

Conclusions

Taken together, these findings present new data that allow us to study the initial effects of inflammatory stimulation on blood-brain barrier disruption, cytokine activation, and metabolic pathway changes that drive the response and recovery of the barrier during continued inflammatory exposure.
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​s12974-016-0760-y) contains supplementary material, which is available to authorized users.
Abkürzungen
ANOVA
Analysis of variance
BBB
Blood-brain barrier
CNS
Central nervous system
ELISA
Enzyme-linked immunosorbent assay
FITC-dextran
Fluorescein isothiocyanate-dextran
GM-CSF
Granulocyte-macrophage colony-stimulating factor
hBMVEC
Human brain-derived microvascular endothelial cells
HDMSE
High-definition data-independent mass spectrometry acquisition with simultaneous analysis of molecular fragmentation
hiPSC
Human induced pluripotent stem cells
IM-MS
Ion mobility-mass spectrometry
LC
Liquid chromatography
LPS
Lipopolysaccharide
MS
Mass spectrometry
MSE
Data-independent MS acquisition with simultaneous analysis of molecular fragmentation
NVU
NeuroVascular Unit
PCA
Principal component analysis
PDMS
Polydimethylsiloxane
ROCK
Rho-associated coiled-coil kinase
TEER
Transendothelial electrical resistance
UPLC
Ultraperformance liquid chromatography
VIIBRE
Vanderbilt Institute for Integrative Biosystems Research and Education

Background

Recent research has shown that systemic infection and inflammation not only affect multiple organs in the body but also the central nervous system (CNS). An excellent example of this is maternal immune activation, which increases the risk of neurological disorders in the gestating fetus [17]. Previous studies have determined that traumatic brain injuries and cancer can activate the immune system and affect the CNS [8, 9]. These reports support the idea that substances created by peripheral immune responses are crossing the blood-brain barrier (BBB) and affecting CNS function. Furthermore, immune activators such as lipopolysaccharide (LPS) have been shown to impair intestinal barrier function, and in neural tissue culture, LPS has been reported to induce cytokine activation and cell damage [1012]. Recent animal studies have illustrated that LPS exposure in pregnant mice elevates fetal IL-6 and perturbs fetal brain development [7, 13]. Other data collected in rodents suggest that the BBB is relatively impermeable to LPS [14]. While the rodent work is compelling, it can be difficult to reconcile with human postmortem studies [15, 16] and observations made in cell culture, which seem to indicate a much more pronounced effect of LPS on the BBB [17]. One limitation of these studies is that due to the high cost of sample collection from both animals and human patients, often only a single time point after a single, short-duration exposure is analyzed. Systemic immune activation is a process that develops over time [18, 19], and thus, BBB function should be assessed in a dose- and time-dependent manner. In addition to LPS directly crossing the human BBB, the cytokines it induces in the vasculature that comprise the BBB certainly can cross the barrier and affect its function [2022].
To aid our further understanding of how the BBB responds to immune activation, we have developed a novel dual-chamber microfluidic device that models BBB function. It utilizes human primary cells and enables flow and sample collection from both compartments (brain and vascular side) separated by a barrier [23]. By using this novel technology, we are able to continually perfuse the vascular side of the BBB model with LPS or a cytokine cocktail and to collect effluent samples from before and during the course of exposure. In these studies, our data suggest that LPS has a time-dependent effect on BBB permeability, cytokine activation, and metabolic activity, and we observe a robust metabolic pathway activation response using a mixture of TNF-α, IL-1β, and MCP1,2.

Methods

NeuroVascular Unit microfluidic device

The NeuroVascular Unit (NVU) was fabricated by the Vanderbilt Institute for Integrative Biosystems Research and Education (VIIBRE) Microfabrication Core. The basic design is a two-chamber system wherein the chambers are constructed of polydimethylsiloxane (PDMS) and divided by a porous 0.2-μm polycarbonate membrane (Sigma-Aldrich, St. Louis, MO, USA). Each chamber has its own inlet and outlet for perfusion. The device can be perfused in any orientation, so that cells seeded into a chamber can be induced by gravity to adhere to one side of the porous membrane and by inverting the device and seeding the opposite side with a different cell type, they can be grown in opposition to one another to form the BBB [23]. As previously described [23], the bottom/vascular chamber is 2.9 μL in volume, with uniform shear forces across the chamber, while the top/brain chamber is 18 μL in volume. The top perfusion layer is attached to the brain chamber and allows a mild media exchange without the introduction of shear stress onto the neurons and co-differentiating astrocytes (cultured within the collagen) and the astrocytes and pericytes that are adhered to the barrier.

Cell culture

Cell culture was carried out as previously described [23]. In brief, cells used to establish our NVU BBB model include primary human brain-derived microvascular endothelial cells (hBMVEC) from Applied Cell Biology (Kirkland, WA, USA) and pericytes and astrocytes from ScienCell (Carlsbad, CA, USA) and ATCC (Manassas, VA, USA), respectively. Before endothelial cells were introduced into the lower vascular chamber of the NVU, the device was coated with laminin at 9.6 μg/mL for 24 h at 37°C. On day 0, hBMVECs were loaded into the vascular chamber (1 × 106 cells/mL), followed by device inversion to allow cell attachment to the membrane. At day 1, media was perfused at 2 μl/min, and the hBMVECs were allowed to grow for 12 days to reach confluence and establish tight junctions. On day 12, the device was returned to its original orientation and a 2:1 mix of primary human astrocytes and pericytes was loaded into the upper brain chamber and allowed 1–2 days to reach confluence. On day 14, collagen gel containing 4 million human induced pluripotent stem cell (hiPSC)-derived human cortical neurons and co-differentiating astrocytes per milliliter was loaded on top of the astrocytes and pericytes. The hiPSC-derived neurons and co-differentiating astrocytes were differentiated to days 95–200 via dual-SMAD inhibition followed by terminal differentiation by adapting methods described elsewhere, except that LDN was used at 4 μM [2427]. The collagen gel was given 2 h to solidify before restarting perfusion of the brain chamber. For the first 3 days after the neurons were added, the brain chamber was perfused with media containing Rho-associated coiled-coil kinase (ROCK) inhibitor (10 μM, Tocris) Y-27632 (Sigma-Aldrich, St. Louis, MO, USA) to help the neurons survive the stress of replating [2831]. Once the ROCK inhibitor was no longer needed, we used media without it, and the NVU was ready for testing.

LPS and cytokine cocktail exposure and sample collection

LPS was purchased from Sigma-Aldrich (St. Louis, MO, USA) and applied to the vascular chamber at 100 μg/mL, which is a concentration that has previously been shown to disrupt tight junctions and increase intestinal permeability [32]. Effluent samples were collected from both the vascular and brain chambers of the NVU at three time points: before exposure (denoted as 0), 6 h after exposure, and 24 h after exposure. For the cytokine cocktail, 100 ng/ml of TNF-α, IL-1β, and MCP1,2 (Sigma-Aldrich, St. Louis, MO, USA) were diluted in vascular media [23] and applied to the vascular chamber only for 24 h.

Live/dead evaluation

To evaluate the DNA integrity of the cells, a live/dead assay kit (Cat. No. L3224, Fisher, Waltham, MA, USA) was used according to the manufacturer’s instructions using 1 μM of calcein AM (i.e., live stain indicator) and 2 μM of ethidium homodimer-1 (i.e., dead stain indicator), as previously described [33]. Cells that are stained with red are considered unhealthy. All live/dead images were taken from the same frame with different excitation wavelengths.

FITC-dextran diffusion and transendothelial electrical resistance

Fluorescein isothiocyanate-dextran (FITC-dextran) of 10 kDa (Sigma-Aldrich, St. Louis, MO, USA) was prepared at 1 μg/mL (100 nM) in cell culture media for diffusion testing. As previously described [23], the vascular compartment of the NVU was perfused with 10 kDa solution for 23 h. In our original protocol [23] at 23 h, the flow on both sides (vascular and brain chambers) was stopped for 1 h, allowing the dextran to diffuse across the BBB and accumulate in the brain compartment. After 1 h, perfusion of both chambers was restarted and individual effluents were collected for fluorescent intensity analysis using a plate reader (TECAN M1000). We have shown that the stop-flow was not necessary and now collect fluid for 1 h of continuous perfusion with FITC-dextran. By measuring FITC-dextran diffusion across the membrane, we were able to evaluate the effectiveness of the BBB [34].
Transendothelial electrical resistance (TEER) measurements were performed using our custom-built multi-frequency impedance analyzer based on an AD5933 chip (Analog Devices, Nashua, NH, USA) utilizing a four-probe approach [35]. The largest changes in impedance as a function of BBB maturation were observed at 15 kHz, and all TEER measurements used in this study to evaluate the effect of LPS on BBB function are reported at that frequency.

Tight junction staining

Fluorescent labeling of the tight junctions was evaluated using a Zeiss Axiovert 200 automated microscope equipped with a CoolSnap CCD camera. Collected images were analyzed with ImageJ. Tight junction staining was conducted as detailed in [36], using ZO-1 and claudin-5 (Invitrogen, Grand Island, NY, USA) directly conjugated to Alexa 488 [37]. Briefly, greyscale measurements of the border between cell plasma membranes were taken for ten different cell-to-cell junctions in four different plates, and an average intensity was derived for each culture condition [23].

Cytokine ELISA

Effluent samples taken before 6 h and after 24 h of LPS exposure were collected and diluted 1:3 for cytokine analysis using either the V-Plex Human Cytokine Kit (Meso Scale Discovery, Rockville, MD, USA) or Quantikine enzyme-linked immunosorbent assay (ELISA) for TNF-α and IL-1β (R&D Systems, Inc., Minneapolis, MN, USA). Sample preparation was carried out as described previously [38].

Data analysis

The statistical analysis of the mass spectrometry data is described within the section on metabolite data processing and analysis. All other data were analyzed in a blinded fashion using GraphPad software. Analysis of variance (ANOVA) models were used to analyze the data and contained one between-subjects variable, such as “treatment” and “trial number” (e.g., before and after drug treatment). The appropriateness of ANOVA models was evaluated by considering the distributional properties of the variables studied and by the adequacy of the homogeneity of variance assumption. The Greenhouse-Geisser (or Huynh-Feldt) adjustment was used for all within-subjects effects containing more than two levels in order to protect against violations of the sphericity/compound symmetry assumptions when a repeated measure ANOVA was used.

Metabolite extraction

All solvents used for metabolite extraction (methanol, water, acetonitrile, and formic acid) were liquid chromatography (LC)-mass spectrometry (MS) grade (Fisher Scientific, Fair Lawn, NJ, USA). Metabolites were extracted from NVU media using a MeOH:H2O (80:20, v:v) solvent extraction mixture. A volume of 500 μL of cold (−20°C) extraction mixture was added to each 60 μL aliquot of media, vortexed for 30 s, and incubated at −80°C overnight to precipitate proteins. After incubation, samples were cleared by centrifugation at 15,000 rpm for 15 min, and the resulting supernatant was removed and evaporated to dryness in a vacuum concentrator. Dried extracts were reconstituted in 60 μL of C18 reconstitution solvent mixture containing 98:2 (v:v) H2O:ACN with 0.1% formic acid for reverse phase analysis, followed by centrifugation for 5 min at 15,000 rpm to remove insoluble debris. Quality control samples were prepared by combining equal volumes (10 μL) of each sample type (experimental design and sample workflow are shown in Additional file 1).

Mass spectrometry

Ultraperformance liquid chromatography-ion mobility-mass spectrometry (UPLC-IM-MS) and data-independent MS acquisition with simultaneous analysis of molecular fragmentation (MSE) were performed on a Waters Synapt G2 HDMS (Milford, MA, USA) mass spectrometer equipped with a Waters nanoAcquity UPLC system and autosampler (Milford, MA, USA), as previously described [39]. Metabolites were separated on a reverse phase 1 mm × 100 mm HSS T3 C18 column packed with 1.8-μm particles (Waters, Milford, MA, USA) held at 45°C. Liquid chromatography was performed using a 30-min gradient at a flow rate of 75 μL min−1 using mobile phase A (0.1% formic acid in H2O) and mobile phase B (0.1% formic acid in ACN). The following elution gradient was used for analysis: 0 min, 99% A; 1 min, 99% A; 10 min, 40% A; 20 min, 1% A; 22 min, 1% A; 25 min, 99% A.
High-definition MSE (HDMSE) analyses were run using resolution mode, with a capillary voltage of 2.75 kV, source temperature at 100°C, sample cone voltage at 30 V, extraction cone voltage at 5 V, source gas flow of 400 mL min−1, desolvation gas temperature of 325°C, He cell flow of 180 mL min−1, and an ion mobility (IM) gas flow of 90 mL min−1. The data were acquired in positive ion mode from 50 to 2000 Da with a 1-s scan time; leucine enkephalin was used as the lock mass (m/z 556.2771). All analytes were analyzed using MSE with an energy ramp from 10 to 40 eV and an injection volume of 5 μL [40]. (For the work flow, see Additional file 1.)

Metabolite data processing and analysis

The acquired UPLC-IM-MSE data were imported, processed, normalized, and interpreted in Progenesis QI v.2.1 (Nonlinear Dynamics, Newcastle, UK). Each UPLC-IM-MSE data file was imported as an ion intensity map (used for visualization in both m/z and retention time dimensions), followed by retention time alignment and peak picking. Peak picking was performed on individual aligned runs by matching peaks in an aggregate data set that was created from all aligned runs. Following peak picking, the features (retention time and m/z pairs) were reduced using both adduct ([M + H]+, [M + Na]+, [M + K]+, etc.) and isotope deconvolution. Data were normalized to all compounds as an abundance ratio between the run being normalized and a reference run. Statistically significant changes were identified using multivariate statistical analysis, including principal component analysis (PCA), and p values were generated using ANOVA or pairwise comparison. Volcano plots (log2 fold change vs. −log10 p value) were generated for basal conditions (no LPS treatment) vs. 100 μg/mL LPS stimulation after either 6 or 24 h. Four biological replicates (NVU) and two technical replicates from each sample type were used to calculate the fold change and p value, and features were considered differentially expressed only if they met both criteria of fold change ≥|2| and significance (p ≤ 0.05); we have termed this list “prioritized metabolites”. Feature lists generated from different individual comparisons were visually compared using Venn diagrams generated by the Venny software package [41]. Statistically significant metabolites or compounds were assigned tentative structural identifications using accurate mass measurements (<10 ppm error) and isotope distribution by searching the Human Metabolome Database (HMDB) [40], METLIN [42], MassBank [43], and the NIST 14 Tandem Database and Search Program of the National Institute of Standards and Technology [44]. Following tentative structural identifications, further data processing was performed by removing metabolites associated with drugs, plants, food, and microbial origin. Metabolite peak identifications were putatively assigned using product ions observed in the fragment ion spectra analyzed in HDMSE mode by searching the aforementioned databases. Ion mobility separations were used to isolate precursor ions and correlate product ions [45].

Metabolic activity network mummichog analysis

Metabolomics pathway analysis was performed by mummichog software 1.0.5 using default parameters. Compound ions measurement files exported from Progenesis QI analysis software were used to generate the mummichog input files. mummichog tested the enrichment of input metabolites against random data resampled from the list of compounds by permutations and produced an empirical p value for known biological pathways. Input metabolites in the significant pathways (p value ≤0.05) were linked in a network figure by known metabolic pathways [46].

Results

Inflammatory signals and cell viability

Although it has been well established that exposure to LPS induces cytokine responses [16, 32], and in the case of other organ systems that LPS exposure has been linked to reduced tight junction protein expression [17], relatively little is known about its effects on the BBB and how this compares to the direct cytokine exposure that LPS is supposed to induce. To study the effects of inflammatory signals on BBB function, we leveraged novel microfluidic technology in a dual-chambered device, creating a system that contains the relevant cell types for BBB formation and enables these cells to form the barrier in the presence of flow and a differential serum concentration from the vascular to the brain side of the device [23]. Once the brain-derived endothelial cells, astrocytes, and pericytes have had a chance to form the basis for the BBB, hiPSC-derived cortical neurons and co-differentiating astrocytes are suspended in a collagen gel and loaded on top of the astrocytes and pericytes. This entire component is what we call the NeuroVascular Unit (NVU), which previously has been shown to restrict diffusion of small molecules and facilitate active transport [23]. Having established our cellular model of the BBB (Fig. 1a), we then sought a concentration of LPS and cytokine cocktail that, while activating the system, would not cause cell death. We wanted the highest concentration that did not increase cell death from control. Over the dose ranges tested (25 to 100 μg/ml), we found that 100 μg/mL showed no cell death in either the vascular chamber containing endothelial cells (Fig. 1b) or in the brain chamber containing pericytes, astrocytes, and neurons (Fig. 1c), as assessed by live/dead staining after 24 h of LPS or cocktail exposure.

Blood-brain barrier transport of inflammatory signals

The custom microfluidic device used to generate the organ-on-chip model of the BBB and the NVU consists primarily of PDMS, a polymer with many desirable features for cell culture, being easily molded, cell-compatible, and gas-permeable, to name a few. We note, however, that it also has been shown in tests to absorb small hydrophobic molecules [4749]. To know the actual dose of inflammatory signals reaching our NVU BBB, we first looked at absorption in an empty device (Fig. 2a), and from these findings, we observed that while we started with relatively high concentrations, final exposure was much closer to physiologic ranges seen in patients at the onset of severe sepsis (~300 pg/ml) [50], with the greatest loss being LPS itself. When we next examined the transport of LPS vs. cocktail across the BBB in the NVU, we saw that the percentage of LPS transport was 46% (Fig. 2b) and that of TNF-α and IL-1β was 31 and 35%, respectively (Fig. 2c,d). These data indicate that both LPS and cytokines it often stimulates cross the barrier with relatively high efficacy.

Inflammatory stimulation effects on blood-brain barrier integrity

To measure directly how BBB permeability was changing over time in response to LPS, we determined the diffusion of 10-kDa FITC-dextran across the NVU BBB. As was expected, at time 0, before LPS exposure, diffusion was extremely low; however, after 6 h of exposure to LPS introduced on the vascular side, diffusion was significantly increased four times over the control at time 0 (p = .0001, N = 7). Interestingly, at 24 h, diffusion was reduced from its level at 6 h but had not returned to pre-exposure levels (Fig. 3a). Having thus established a time-course effect of LPS on membrane resistance and barrier diffusion, we then assessed how this compared to direct cytokine stimulation and found the effect on BBB permeability to be similar (Fig. 3b). We also considered how transendothelial resistance changed as a function of LPS and time. During early exposures to LPS for six continuous hours, TEER measurements suggest that the permeability of the NVU BBB does increase as a function of time and dose, as observed by a reduction in resistance. An inverse response is observed after 24 h of exposure to LPS, however; i.e., TEER measurements show an increase in membrane resistance as a function of time and dose (Fig. 3c).
Additional experiments showed the expression and localization of tight junction proteins as a response to dose and time of LPS exposure. For both claudin-5 and ZO-1, we saw a dose-dependent reduction in levels of expression at 6 h of LPS exposure and an increase after 24 h (Fig. 3d, e). When the staining intensity was quantified over multiple samples, we observed a significant decrease in both tight junction proteins at 6 h (−12% ZO-1, −18% Clad-5 (p ≤ .03, N = 10)) and a significant increase at 24 h (31% ZO-1, Clad-5 22% (p ≤ .03, N = 10)) for all concentrations tested (Fig. 3f shows the quantitation of the staining). These results indicate that the changes in the NVU BBB as a result of LPS exposure directly correlate with tight junction protein expression. Taken together, these findings argue that although the BBB may initially become more permeable when exposed to this foreign immunogen, over time it begins to block LPS passage—this is related to tight junction protein expression. These results also showed similar disruption of the BBB by cytokines, as was seen with LPS, suggesting overlapping mechanisms.

Cytokine activation in BBB model as a result of LPS stimulation

The link between LPS exposure and cytokine activation has been well established in both cell culture and animal models. Despite this wealth of data, few studies have investigated cytokine response as a function of time or changes in barrier permeability. Using 50 μL of effluent collected from the vascular and brain side of our NVU devices at 0, 6, and 24 h of LPS exposure, we ran ELISA detection for a battery of cytokines (GM-CSF, IL12-p40, IL-15, IL-16, IL-1a, IL-5, IL-17A, TNF-b, VEGF, TNF-α, IL-1β ). Interestingly, of the 11 cytokines investigated, over half exhibited significant changes in one or more of the chambers and time points under investigation; however, different patterns of cytokine release and membrane permeability were observed when individual cytokines were studied alone. For example, the cytokine granulocyte-macrophage colony-stimulating factor (GM-CSF) known to be active in BBB disruption [51] showed a pattern of release in both vascular and brain chambers very similar to that seen in FITC-dextran diffusion, wherein release was highest at 6 h and markedly reduced at 24 h (Fig. 4a). However, if we look at a cytokine such as IL-17A, which is known to stimulate neurite outgrowth [52], its release levels in the vascular chamber were significantly reduced at 6 and 24 h, but increased at these time points in the brain chamber (Fig. 4b). Finally, the canonical cytokine TNF-α, which is known to be part of the inflammatory response to BBB disruption [53, 54], showed an increase in release over time in both the vascular and brain chambers (Fig. 4c). (For a complete list of all the cytokines and their fold changes, see Additional file 2.) From these findings, we perceive a complex array of cytokine changes that occur over time in the BBB, and these changes may result in the observed alterations of barrier permeability.

Metabolic response of BBB model to LPS and cytokine cocktails

In addition to looking at the traditional response of our BBB model to inflammatory stimulation in terms of cytokine activation, we also used novel technology to gain a better understanding of how the BBB metabolically responds to these challenges. To investigate the metabolic response of our BBB to inflammatory stimulation over time, UPLC-IM-MS was used to determine if the global molecular metabolic profiles change throughout the course of LPS exposure (see Additional file 1). Principal component analysis plots of the UPLC-IM-MS/MS for LPS samples illustrate distinct separations between the control and treated sample types and between the 6- and the 24-h time points (Fig. 5a, b). Volcano plots for the vascular chamber (Fig. 5c) (p ≤ 0.05 and fold changes ≥│2│) illustrate that critical metabolites are released over time (6 h of LPS stimulation; see colored points). We observe the same trend in the brain chamber after 6 h of LPS stimulation (Fig. 5d). Moreover, after 24 h of LPS stimulation, more metabolites that met the significance criteria were released (Fig. 5c, d, right). These global metabolic profile data indicate that metabolic changes occurred after 6 h and before 24 h of stimulation (Fig. 5c, d) in both chambers.
In an IM-MS analysis of response to the cytokine cocktail, we observe two distinct groups (0 h treatment and 24 h treatment) using PCA (Fig. 6a). Volcano plots for the vascular chamber (Fig. 6b) (p ≤ 0.05 and fold changes ≥│2│) illustrate that both time and treatment have a significant effect on metabolic activity, with treatment causing the biggest impact. We see the same trend in the brain chamber (Fig. 6c). Venn diagrams of the global metabolite profiles show that while a significant number of metabolites are observed after 6 and 24 hours of LPS stimulation, greater than 50% of the compounds observed that met the significance criteria are time-specific (Fig. 7a, b). We do note more significant changes in the global metabolic profiles during the 24-h LPS stimulation when compared to 6 h of stimulation. These observations may be attributed to the cumulative effect of continuous LPS response (Fig. 7c, d). These same trends were consistent with our observations following cytokine cocktail activation (Fig. 7e, f), however, and the inflammatory stimulation was likely a bit stronger given less absorption by the PDMS (Fig. 2a). We found many more metabolites significantly affected by treatment (Fig. 7g, h). The observed global metabolic response to LPS between the vascular and brain chambers (across the BBB) after 6 h of LPS stimulation is similar (see Fig. 8a, b). In contrast, the observed global metabolite profile data for 24 h of LPS stimulation suggest that more significantly changing metabolites were observed in the vascular chamber compared to the brain chamber (Fig. 8c, d). In addition to the global analysis of metabolic trends in response to LPS, a number of significantly changed metabolite compounds were identified through database correlation (see the “Metabolite data processing and analysis” section above, and the tentative structural identifications listed in Additional file 3). These preliminary identifications allow us to prioritize that fatty acid and protein degradation pathways may be affected by LPS stimulation. When examining cytokine cocktail treatment after 24 h of exposure, we see that the overlap between the vascular and brain sides is increased with treatment (Fig. 8e, f), and treatment with cytokine cocktail nearly doubles the number of metabolites changed in both chambers (Fig. 8g, h). Collectively, these two data sets reflecting inflammatory drive show similar but not identical metabolic signatures with regard to the BBB response.

Pathway identifications of metabolic response to inflammatory stimulation

Using biologically driven computational analysis (mummichog), metabolites observed in these studies were used to predict metabolic network activity. In determining the BBB response to cytokine cocktail response, network activity analysis (or mummichog) was used to observe metabolites affected either by consumption or production in response to inflammatory stimulation (Fig. 9). These activity network analyses allow our data to be grouped in pathways in an effort to identify network relationships between global metabolic profiles (in this case, treated vs. untreated). In the predictive activity network analysis for the vascular side of the BBB, glutathione, CoA, and tryptophan are highlighted as central nodes (Fig. 9a). On the brain side, CoA and tryptophan are shared central nodes, as are dopamine and glutamate (Fig. 9b). In addition to identifying central nodes of metabolic interconnection, these analyses can also prioritize the significance of specific metabolites in a known pathway and identify specific pathways that are affected by our studies. For these analyses, we used both LPS and cytokine cocktail data sets, and we observed multiple pathways that were continuously highlighted regardless of barrier side or inflammatory stimulation (see Fig. 10, red rows). Glycine, serine, alanine, and threonine metabolism and aspartate and asparagine metabolism indicate that protein synthesis utilizes critical pathways that are changed upon inflammatory stimulation.
We also observed activated pathways specific to each side of the BBB (see Fig. 10, yellow rows). For example, pyrimidine metabolism is a pathway known for being involved with systemic inflammation such as gout and arthritis, as well as some neurodevelopmental disorders [55, 56], and our data suggest its activation in the vascular chamber. These data suggest a novel role for pyrimidine metabolism in BBB response to inflammatory stimulation. In the case of the brain side, the most prominent pathway implicated in our network pathway analysis was drug metabolism involving a canonical cytochrome P450 pathway for mediating the processing of antidepressants, antipsychotics, drugs of abuse, endogenous neurochemicals such as serotonin and dopamine, neurotoxins, and carcinogens [57] and neuroinflammation. There were also several pathways that were treatment-specific (Fig. 10, purple rows). For example, glutathione metabolism was observed for LPS but was not present in the cytokine cocktail data, whereas for the cytokine cocktail treatment, tryptophan metabolism was strongly indicated. It is known that glutathione plays important roles in antioxidant defense, nutrient metabolism, and regulation of cellular events, including cytokine production and immune response, as well as gene expression, DNA and protein synthesis, cell proliferation and apoptosis, and signal transduction. In these data, we observe a broad spectrum of metabolic activity consistent with what we would expect for a compound such as LPS. We also observed some interesting findings, such as the potential activation of tryptophan metabolism, which is critical as an essential amino acid and a key player in serotonin production, but is less known for its effect on inflammation [58].
Network activity analysis also allows us to prioritize the further exploration of networks or metabolites of interest, for example, the purine and pyrimidine pathways (Fig. 10). If we compare activation in the brain side to the vascular side of our BBB model, we observe numerous pathways that are significantly changed in both sides but in opposite directions (Figs. 10 and 11). The vascular chamber is mostly up-regulating the pro-inflammatory pathway, while at the same time, the brain chamber is mostly down-regulating this pathway. By combining a novel micro-organ of a BBB with cutting-edge metabolomics, we have been able to drastically increase our understanding of the metabolic consequences of inflammatory stimulation on the brain microvasculature, as well as the neurons themselves.

Discussion

In the present study, we report that exposure of the vascular chamber of the NVU to the inflammatory stimulation of either LPS or a cytokine cocktail will induce time-dependent changes in BBB function and cytokine activation, as well as a global change of the metabolomics profile on both the vascular and brain chambers. Our data suggest that the initial response to LPS stimulation is characterized by reduced tight junction formation and increased membrane permeability. While it has been shown that LPS disrupts tight junctions in numerous organ systems, including the gut and lung [59, 60], very few studies have considered its effects on the BBB [61], and none have reported the spontaneous recovery we found: after an increase in exposure time (between 6 and 24 h), a recovery from the initial exposure results in an increase in the formation of tight junctions and a decrease in membrane permeability from the 6-h time point, but not back to the levels before exposure.
In addition to being able to analyze BBB changes in response to LPS and cytokine cocktail, this unique microfluidic model has sufficient cell mass and low enough volume to allow us to conduct cytokine analysis of the effluent from both the vascular and brain sides at the time points chosen. Previous studies have already demonstrated that cytokines are often released in an oscillating fashion and may have both pro- and anti-inflammatory properties, depending on their release profiles [6264]. The 3D cell culture made possible by our NVU [23] enabled us to see and profile cytokine activation based not only on when cytokines were elevated but also on whether they were differentially elevated in terms of vascular vs. brain compartment. Taken collectively, these data argue for a cytokine activation model whereby pro-inflammatory cytokines are activated in both chambers at initial exposure, but at later time points, only a subset remains activated in both compartments. The data also suggest that at these later time points, pro-repair cytokine activation is up-regulated in the brain compartment as the BBB begins to rebound, whereas the vascular compartment remains more pro-inflammatory.
Notable advances in identifying metabolites and understanding the significance of these measurements are demonstrated in these studies. By leveraging IM-MS with our organ-on-chip model of the BBB, we were able to obtain a more thorough investigation of metabolite changes due to neuroinflammation. In these studies, our data suggest that (1) inflammation involving the BBB is closely linked to protein synthesis, (2) different sides of the barrier use different proteomic and metabolic pathways to respond to inflammatory signals, and (3) even when the same pathway is involved, the vasculature could be driving pro-inflammatory processes while the brain is ramping down inflammation.

Conclusions

This work integrated several new technologies, including microfluidic organs-on-chips as in vitro models, IM-MS metabolomics, and pathway identification, and it is from this integration that we have gained new insights into BBB response to inflammatory stimulation. We have shown inflammatory disruption of the BBB and transport of the inflammatory signals across the BBB, mapped changes in cytokine to barrier disruption, and created a detailed analysis of the metabolic signature and metabolic pathways associated with inflammatory stimulation.

Acknowledgements

We thank Allison Price for her editorial assistance and guidance. We thank Dr. Kevin Ess for collaborative assistance in obtaining iPSCs.

Funding

This work was funded in part by the National Institutes of Health’s National Center for Advancing Translational Sciences under Award Number 5UH3TR000491-04 (JPW) and National Institute of Environmental Health Sciences under Award Number R01 ES016931 (ABB) and by the Vanderbilt Institute for Integrative Biosystems Research and Education and the Center for Innovative Technology of Vanderbilt University. The content is solely the responsibility of the authors and does not necessarily represent the official views of the NIH.

Availability of data and materials

The datasets analyzed during the current study (beyond the lists in the supplementary information files) are available from the corresponding author on reasonable request.

Authors’ contributions

JAB designed and conducted the NVU experiments, analyzed results, and drafted the manuscript. MS and DJW conducted experiments with the NVU and helped establish experimental protocols. SGC and SDS designed and conducted metabolomics-based UPLC-IM-MS protocols, sample preparation, methods, and experiments and performed data processing and analysis for mass spectrometry experiments. DAM developed microfluidic components and guided their testing and optimization. LJM contributed to the optimization of the NVU and experimental design. MDN and ABB developed the protocols to create iPSC-derived glutamatergic neurons and provided all neurons used in the NVUs. CMB was responsible for NVU production, testing, and optimization. OSH and RSR were responsible for the design, fabrication, testing, and use of the TEER device. PCS developed the original NVU, contributed to its refinement, and guided its production. JAM and JPW designed experiments and analyzed, integrated, and interpreted data. All authors assisted with preparation of the manuscript and approved the final version.

Competing interests

The authors declare that they have no competing interests.
Not applicable.
Neurons were differentiated from de-identified human induced pluripotent stem cells (hiPSC) reprogrammed from primary dermal fibroblasts that had been obtained by skin biopsy from healthy adult subjects with no known family history of neurodegenerative disease after appropriate patient consent/assent under the guidelines of an approved IRB protocol (Vanderbilt #080369, Kevin Ess, principal investigator; Aaron Bowman, investigator). We use a de-identified alphanumeric nomenclature to designate independent hiPSC lines.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.
Literatur
1.
Zurück zum Zitat Garbett KA, Hsiao EY, Kalman S, Patterson PH, Mirnics K. Effects of maternal immune activation on gene expression patterns in the fetal brain. Transl Psychiatry. 2012;2, e98.CrossRefPubMedPubMedCentral Garbett KA, Hsiao EY, Kalman S, Patterson PH, Mirnics K. Effects of maternal immune activation on gene expression patterns in the fetal brain. Transl Psychiatry. 2012;2, e98.CrossRefPubMedPubMedCentral
2.
Zurück zum Zitat Hsiao EY, Patterson PH. Activation of the maternal immune system induces endocrine changes in the placenta via IL-6. Brain Behav Immun. 2011;25:604–15.CrossRefPubMed Hsiao EY, Patterson PH. Activation of the maternal immune system induces endocrine changes in the placenta via IL-6. Brain Behav Immun. 2011;25:604–15.CrossRefPubMed
3.
Zurück zum Zitat Patterson PH. Immune involvement in schizophrenia and autism: etiology, pathology and animal models. Behav Brain Res. 2009;204:313–21.CrossRefPubMed Patterson PH. Immune involvement in schizophrenia and autism: etiology, pathology and animal models. Behav Brain Res. 2009;204:313–21.CrossRefPubMed
4.
Zurück zum Zitat Shelton RC, Claiborne J, Sidoryk-Wegrzynowicz M, Reddy R, Aschner M, Lewis DA, Mirnics K. Altered expression of genes involved in inflammation and apoptosis in frontal cortex in major depression. Mol Psychiatry. 2010;16:751–62.CrossRefPubMedPubMedCentral Shelton RC, Claiborne J, Sidoryk-Wegrzynowicz M, Reddy R, Aschner M, Lewis DA, Mirnics K. Altered expression of genes involved in inflammation and apoptosis in frontal cortex in major depression. Mol Psychiatry. 2010;16:751–62.CrossRefPubMedPubMedCentral
5.
Zurück zum Zitat Shi L, Fatemi SH, Sidwell RW, Patterson PH. Maternal influenza infection causes marked behavioral and pharmacological changes in the offspring. J Neurosci. 2003;23:297–302.PubMed Shi L, Fatemi SH, Sidwell RW, Patterson PH. Maternal influenza infection causes marked behavioral and pharmacological changes in the offspring. J Neurosci. 2003;23:297–302.PubMed
6.
Zurück zum Zitat Shi L, Tu N, Patterson PH. Maternal influenza infection is likely to alter fetal brain development indirectly: the virus is not detected in the fetus. Int J Dev Neurosci. 2005;23:299–305.CrossRefPubMed Shi L, Tu N, Patterson PH. Maternal influenza infection is likely to alter fetal brain development indirectly: the virus is not detected in the fetus. Int J Dev Neurosci. 2005;23:299–305.CrossRefPubMed
7.
Zurück zum Zitat Smith SE, Li J, Garbett K, Mirnics K, Patterson PH. Maternal immune activation alters fetal brain development through interleukin-6. J Neurosci. 2007;27:10695–702.CrossRefPubMedPubMedCentral Smith SE, Li J, Garbett K, Mirnics K, Patterson PH. Maternal immune activation alters fetal brain development through interleukin-6. J Neurosci. 2007;27:10695–702.CrossRefPubMedPubMedCentral
8.
Zurück zum Zitat Deeken JF, Loscher W. The blood-brain barrier and cancer: transporters, treatment, and Trojan horses. Clin Cancer Res. 2007;13:1663–74.CrossRefPubMed Deeken JF, Loscher W. The blood-brain barrier and cancer: transporters, treatment, and Trojan horses. Clin Cancer Res. 2007;13:1663–74.CrossRefPubMed
9.
Zurück zum Zitat Thal SC, Neuhaus W. The blood-brain barrier as a target in traumatic brain injury treatment. Arch Med Res. 2014;45:698–710.CrossRefPubMed Thal SC, Neuhaus W. The blood-brain barrier as a target in traumatic brain injury treatment. Arch Med Res. 2014;45:698–710.CrossRefPubMed
10.
Zurück zum Zitat Liu J, Huang D, Xu J, Tong J, Wang Z, Huang L, Yang Y, Bai X, Wang P, Suo H, et al. Tiagabine protects dopaminergic neurons against neurotoxins by inhibiting microglial activation. Sci Rep. 2015;5:15720.CrossRefPubMedPubMedCentral Liu J, Huang D, Xu J, Tong J, Wang Z, Huang L, Yang Y, Bai X, Wang P, Suo H, et al. Tiagabine protects dopaminergic neurons against neurotoxins by inhibiting microglial activation. Sci Rep. 2015;5:15720.CrossRefPubMedPubMedCentral
11.
Zurück zum Zitat Ren Y, Xie Y, Jiang G, Fan J, Yeung J, Li W, Tam PK, Savill J. Apoptotic cells protect mice against lipopolysaccharide-induced shock. J Immunol. 2008;180:4978–85.CrossRefPubMed Ren Y, Xie Y, Jiang G, Fan J, Yeung J, Li W, Tam PK, Savill J. Apoptotic cells protect mice against lipopolysaccharide-induced shock. J Immunol. 2008;180:4978–85.CrossRefPubMed
12.
Zurück zum Zitat Suzumura A. Microglia in pathophysiology of neuroimmunological disorders. Nihon Rinsho. 2013;71:801–6.PubMed Suzumura A. Microglia in pathophysiology of neuroimmunological disorders. Nihon Rinsho. 2013;71:801–6.PubMed
13.
Zurück zum Zitat Urakubo A, Jarskog LF, Lieberman JA, Gilmore JH. Prenatal exposure to maternal infection alters cytokine expression in the placenta, amniotic fluid, and fetal brain. Schizophr Res. 2001;47:27–36.CrossRefPubMed Urakubo A, Jarskog LF, Lieberman JA, Gilmore JH. Prenatal exposure to maternal infection alters cytokine expression in the placenta, amniotic fluid, and fetal brain. Schizophr Res. 2001;47:27–36.CrossRefPubMed
14.
Zurück zum Zitat Jin L, Nation RL, Li J, Nicolazzo JA. Species-dependent blood-brain barrier disruption of lipopolysaccharide: amelioration by colistin in vitro and in vivo. Antimicrob Agents Chemother. 2013;57:4336–42.CrossRefPubMedPubMedCentral Jin L, Nation RL, Li J, Nicolazzo JA. Species-dependent blood-brain barrier disruption of lipopolysaccharide: amelioration by colistin in vitro and in vivo. Antimicrob Agents Chemother. 2013;57:4336–42.CrossRefPubMedPubMedCentral
15.
Zurück zum Zitat Blom MA, van Twillert MG, de Vries SC, Engels F, Finch CE, Veerhuis R, Eikelenboom P. NSAIDS inhibit the IL-1 beta-induced IL-6 release from human post-mortem astrocytes: the involvement of prostaglandin E2. Brain Res. 1997;777:210–8.CrossRefPubMed Blom MA, van Twillert MG, de Vries SC, Engels F, Finch CE, Veerhuis R, Eikelenboom P. NSAIDS inhibit the IL-1 beta-induced IL-6 release from human post-mortem astrocytes: the involvement of prostaglandin E2. Brain Res. 1997;777:210–8.CrossRefPubMed
16.
Zurück zum Zitat Gayle DA, Beloosesky R, Desai M, Amidi F, Nunez SE, Ross MG. Maternal LPS induces cytokines in the amniotic fluid and corticotropin releasing hormone in the fetal rat brain. Am J Physiol Regul Integr Comp Physiol. 2004;286:R1024–9.CrossRefPubMed Gayle DA, Beloosesky R, Desai M, Amidi F, Nunez SE, Ross MG. Maternal LPS induces cytokines in the amniotic fluid and corticotropin releasing hormone in the fetal rat brain. Am J Physiol Regul Integr Comp Physiol. 2004;286:R1024–9.CrossRefPubMed
17.
Zurück zum Zitat Xaio H, Banks WA, Niehoff ML, Morley JE. Effect of LPS on the permeability of the blood-brain barrier to insulin. Brain Res. 2001;896:36–42.CrossRefPubMed Xaio H, Banks WA, Niehoff ML, Morley JE. Effect of LPS on the permeability of the blood-brain barrier to insulin. Brain Res. 2001;896:36–42.CrossRefPubMed
18.
Zurück zum Zitat Ai M, Curran MA. Immune checkpoint combinations from mouse to man. Cancer Immunol Immunother. 2015;64:885–92.CrossRefPubMed Ai M, Curran MA. Immune checkpoint combinations from mouse to man. Cancer Immunol Immunother. 2015;64:885–92.CrossRefPubMed
19.
Zurück zum Zitat Niebauer J, Volk HD, Kemp M, Dominguez M, Schumann RR, Rauchhaus M, Poole-Wilson PA, Coats AJ, Anker SD. Endotoxin and immune activation in chronic heart failure: a prospective cohort study. Lancet. 1999;353:1838–42.CrossRefPubMed Niebauer J, Volk HD, Kemp M, Dominguez M, Schumann RR, Rauchhaus M, Poole-Wilson PA, Coats AJ, Anker SD. Endotoxin and immune activation in chronic heart failure: a prospective cohort study. Lancet. 1999;353:1838–42.CrossRefPubMed
20.
Zurück zum Zitat Banks WA, Kastin AJ, Broadwell RD. Passage of cytokines across the blood-brain barrier. Neuroimmunomodulation. 1995;2:241–8.CrossRefPubMed Banks WA, Kastin AJ, Broadwell RD. Passage of cytokines across the blood-brain barrier. Neuroimmunomodulation. 1995;2:241–8.CrossRefPubMed
21.
Zurück zum Zitat Nilsen EM, Johansen FE, Jahnsen FL, Lundin KE, Scholz T, Brandtzaeg P, Haraldsen G. Cytokine profiles of cultured microvascular endothelial cells from the human intestine. Gut. 1998;42:635–42.CrossRefPubMedPubMedCentral Nilsen EM, Johansen FE, Jahnsen FL, Lundin KE, Scholz T, Brandtzaeg P, Haraldsen G. Cytokine profiles of cultured microvascular endothelial cells from the human intestine. Gut. 1998;42:635–42.CrossRefPubMedPubMedCentral
22.
Zurück zum Zitat Yarlagadda A, Alfson E, Clayton AH. The blood brain barrier and the role of cytokines in neuropsychiatry. Psychiatry (Edgmont). 2009;6:18–22. Yarlagadda A, Alfson E, Clayton AH. The blood brain barrier and the role of cytokines in neuropsychiatry. Psychiatry (Edgmont). 2009;6:18–22.
23.
Zurück zum Zitat Brown JA, Pensabene V, Markov DA, Allwardt V, Neely MD, Shi M, Britt CM, Hoilett OS, Yang Q, Brewer BM, et al. Recreating blood-brain barrier physiology and structure on chip: a novel neurovascular microfluidic bioreactor. Biomicrofluidics. 2015;9:054124.CrossRefPubMedPubMedCentral Brown JA, Pensabene V, Markov DA, Allwardt V, Neely MD, Shi M, Britt CM, Hoilett OS, Yang Q, Brewer BM, et al. Recreating blood-brain barrier physiology and structure on chip: a novel neurovascular microfluidic bioreactor. Biomicrofluidics. 2015;9:054124.CrossRefPubMedPubMedCentral
24.
Zurück zum Zitat Neely MD, Litt MJ, Tidball AM, Li GG, Aboud AA, Hopkins CR, Chamberlin R, Hong CC, Ess KC, Bowman AB. DMH1, a highly selective small molecule BMP inhibitor promotes neurogenesis of hiPSCs: comparison of PAX6 and SOX1 expression during neural induction. ACS Chem Neurosci. 2012;3:482–91.CrossRefPubMedPubMedCentral Neely MD, Litt MJ, Tidball AM, Li GG, Aboud AA, Hopkins CR, Chamberlin R, Hong CC, Ess KC, Bowman AB. DMH1, a highly selective small molecule BMP inhibitor promotes neurogenesis of hiPSCs: comparison of PAX6 and SOX1 expression during neural induction. ACS Chem Neurosci. 2012;3:482–91.CrossRefPubMedPubMedCentral
25.
Zurück zum Zitat Chambers SM, Fasano CA, Papapetrou EP, Tomishima M, Sadelain M, Studer L. Highly efficient neural conversion of human ES and iPS cells by dual inhibition of SMAD signaling. Nat Biotechnol. 2009;27:275–80.CrossRefPubMedPubMedCentral Chambers SM, Fasano CA, Papapetrou EP, Tomishima M, Sadelain M, Studer L. Highly efficient neural conversion of human ES and iPS cells by dual inhibition of SMAD signaling. Nat Biotechnol. 2009;27:275–80.CrossRefPubMedPubMedCentral
26.
Zurück zum Zitat Shi Y, Kirwan P, Livesey FJ. Directed differentiation of human pluripotent stem cells to cerebral cortex neurons and neural networks. Nat Protoc. 2012;7:1836–46.CrossRefPubMed Shi Y, Kirwan P, Livesey FJ. Directed differentiation of human pluripotent stem cells to cerebral cortex neurons and neural networks. Nat Protoc. 2012;7:1836–46.CrossRefPubMed
27.
Zurück zum Zitat Shi Y, Kirwan P, Smith J, Robinson HP, Livesey FJ. Human cerebral cortex development from pluripotent stem cells to functional excitatory synapses. Nat Neurosci. 2012;15:477–86. S1.CrossRefPubMed Shi Y, Kirwan P, Smith J, Robinson HP, Livesey FJ. Human cerebral cortex development from pluripotent stem cells to functional excitatory synapses. Nat Neurosci. 2012;15:477–86. S1.CrossRefPubMed
28.
Zurück zum Zitat Cucullo L, Hossain M, Puvenna V, Marchi N, Janigro D. The role of shear stress in blood-brain barrier endothelial physiology. BMC Neurosci. 2011;12:40.CrossRefPubMedPubMedCentral Cucullo L, Hossain M, Puvenna V, Marchi N, Janigro D. The role of shear stress in blood-brain barrier endothelial physiology. BMC Neurosci. 2011;12:40.CrossRefPubMedPubMedCentral
29.
Zurück zum Zitat Germann B, Neuhaus W, Hofer-Warbinek R, Noe CR. Applying blood-brain barrier in vitro models to study the influence of drugs on endothelial cells—effects of selected COX-inhibitors. Pharmazie. 2008;63:303–7.PubMed Germann B, Neuhaus W, Hofer-Warbinek R, Noe CR. Applying blood-brain barrier in vitro models to study the influence of drugs on endothelial cells—effects of selected COX-inhibitors. Pharmazie. 2008;63:303–7.PubMed
30.
Zurück zum Zitat Molino Y, Jabes F, Lacassagne E, Gaudin N, Khrestchatisky M. Setting-up an in vitro model of rat blood-brain barrier (BBB): a focus on BBB impermeability and receptor-mediated transport. J Vis Exp. 2014;88:e51278. Molino Y, Jabes F, Lacassagne E, Gaudin N, Khrestchatisky M. Setting-up an in vitro model of rat blood-brain barrier (BBB): a focus on BBB impermeability and receptor-mediated transport. J Vis Exp. 2014;88:e51278.
31.
Zurück zum Zitat Vandenhaute E, Dehouck L, Boucau MC, Sevin E, Uzbekov R, Tardivel M, Gosselet F, Fenart L, Cecchelli R, Dehouck MP. Modelling the neurovascular unit and the blood-brain barrier with the unique function of pericytes. Curr Neurovasc Res. 2011;8:258–69.CrossRefPubMed Vandenhaute E, Dehouck L, Boucau MC, Sevin E, Uzbekov R, Tardivel M, Gosselet F, Fenart L, Cecchelli R, Dehouck MP. Modelling the neurovascular unit and the blood-brain barrier with the unique function of pericytes. Curr Neurovasc Res. 2011;8:258–69.CrossRefPubMed
32.
Zurück zum Zitat Guo S, Al-Sadi R, Said HM, Ma TY. Lipopolysaccharide causes an increase in intestinal tight junction permeability in vitro and in vivo by inducing enterocyte membrane expression and localization of TLR-4 and CD14. Am J Pathol. 2013;182:375–87.CrossRefPubMedPubMedCentral Guo S, Al-Sadi R, Said HM, Ma TY. Lipopolysaccharide causes an increase in intestinal tight junction permeability in vitro and in vivo by inducing enterocyte membrane expression and localization of TLR-4 and CD14. Am J Pathol. 2013;182:375–87.CrossRefPubMedPubMedCentral
33.
Zurück zum Zitat Markov DA, Lu JQ, Samson PC, Wikswo JP, McCawley LJ. Thick-tissue bioreactor as a platform for long-term organotypic culture and drug delivery. Lab Chip. 2012;12:4560–8.CrossRefPubMed Markov DA, Lu JQ, Samson PC, Wikswo JP, McCawley LJ. Thick-tissue bioreactor as a platform for long-term organotypic culture and drug delivery. Lab Chip. 2012;12:4560–8.CrossRefPubMed
35.
Zurück zum Zitat Sarro E, Lecina M, Fontova A, Sola C, Godia F, Cairo JJ, Bragos R. Electrical impedance spectroscopy measurements using a four-electrode configuration improve on-line monitoring of cell concentration in adherent animal cell cultures. Biosens Bioelectron. 2012;31:257–63.CrossRefPubMed Sarro E, Lecina M, Fontova A, Sola C, Godia F, Cairo JJ, Bragos R. Electrical impedance spectroscopy measurements using a four-electrode configuration improve on-line monitoring of cell concentration in adherent animal cell cultures. Biosens Bioelectron. 2012;31:257–63.CrossRefPubMed
36.
Zurück zum Zitat Cao X, Lin H, Muskhelishvili L, Latendresse J, Richter P, Heflich RH. Tight junction disruption by cadmium in an in vitro human airway tissue model. Respir Res. 2015;16:30.CrossRefPubMedPubMedCentral Cao X, Lin H, Muskhelishvili L, Latendresse J, Richter P, Heflich RH. Tight junction disruption by cadmium in an in vitro human airway tissue model. Respir Res. 2015;16:30.CrossRefPubMedPubMedCentral
37.
Zurück zum Zitat Wen J, Qian S, Yang Q, Deng L, Mo Y, Yu Y. Overexpression of netrin-1 increases the expression of tight junction-associated proteins, claudin-5, occludin, and ZO-1, following traumatic brain injury in rats. Exp Ther Med. 2014;8:881–6.PubMedPubMedCentral Wen J, Qian S, Yang Q, Deng L, Mo Y, Yu Y. Overexpression of netrin-1 increases the expression of tight junction-associated proteins, claudin-5, occludin, and ZO-1, following traumatic brain injury in rats. Exp Ther Med. 2014;8:881–6.PubMedPubMedCentral
38.
Zurück zum Zitat Fiorentino M, Levine MM, Sztein MB, Fasano A. Effect of wild-type Shigella species and attenuated Shigella vaccine candidates on small intestinal barrier function, antigen trafficking, and cytokine release. PLoS One. 2014;9, e85211.CrossRefPubMedPubMedCentral Fiorentino M, Levine MM, Sztein MB, Fasano A. Effect of wild-type Shigella species and attenuated Shigella vaccine candidates on small intestinal barrier function, antigen trafficking, and cytokine release. PLoS One. 2014;9, e85211.CrossRefPubMedPubMedCentral
39.
Zurück zum Zitat Brown JA, Sherrod SD, Goodwin CR, Brewer B, Yang L, Garbett KA, Li D, McLean JA, Wikswo JP, Mirnics K. Metabolic consequences of interleukin-6 challenge in developing neurons and astroglia. J Neuroinflammation. 2014;11:183.CrossRefPubMedPubMedCentral Brown JA, Sherrod SD, Goodwin CR, Brewer B, Yang L, Garbett KA, Li D, McLean JA, Wikswo JP, Mirnics K. Metabolic consequences of interleukin-6 challenge in developing neurons and astroglia. J Neuroinflammation. 2014;11:183.CrossRefPubMedPubMedCentral
40.
Zurück zum Zitat Wishart DS, Jewison T, Guo AC, Wilson M, Knox C, Liu Y, Djoumbou Y, Mandal R, Aziat F, Dong E, et al. HMDB 3.0—The Human Metabolome Database in 2013. Nucleic Acids Res. 2013;41:D801–7.CrossRefPubMed Wishart DS, Jewison T, Guo AC, Wilson M, Knox C, Liu Y, Djoumbou Y, Mandal R, Aziat F, Dong E, et al. HMDB 3.0—The Human Metabolome Database in 2013. Nucleic Acids Res. 2013;41:D801–7.CrossRefPubMed
41.
Zurück zum Zitat Hulsen T, de Vlieg J, Alkema W. BioVenn—a web application for the comparison and visualization of biological lists using area-proportional Venn diagrams. BMC Genomics. 2008;9:488.CrossRefPubMedPubMedCentral Hulsen T, de Vlieg J, Alkema W. BioVenn—a web application for the comparison and visualization of biological lists using area-proportional Venn diagrams. BMC Genomics. 2008;9:488.CrossRefPubMedPubMedCentral
42.
Zurück zum Zitat Smith CA, O’Maille G, Want EJ, Qin C, Trauger SA, Brandon TR, Custodio DE, Abagyan R, Siuzdak G. METLIN: a metabolite mass spectral database. Ther Drug Monit. 2005;27:747–51.CrossRefPubMed Smith CA, O’Maille G, Want EJ, Qin C, Trauger SA, Brandon TR, Custodio DE, Abagyan R, Siuzdak G. METLIN: a metabolite mass spectral database. Ther Drug Monit. 2005;27:747–51.CrossRefPubMed
43.
Zurück zum Zitat Horai H, Arita M, Kanaya S, Nihei Y, Ikeda T, Suwa K, Ojima Y, Tanaka K, Tanaka S, Aoshima K, et al. MassBank: a public repository for sharing mass spectral data for life sciences. J Mass Spectrom. 2010;45:703–14.CrossRefPubMed Horai H, Arita M, Kanaya S, Nihei Y, Ikeda T, Suwa K, Ojima Y, Tanaka K, Tanaka S, Aoshima K, et al. MassBank: a public repository for sharing mass spectral data for life sciences. J Mass Spectrom. 2010;45:703–14.CrossRefPubMed
45.
Zurück zum Zitat Sherrod SD, McLean JA. Systems-wide high-dimensional data acquisition and informatics using structural mass spectrometry strategies. Clin Chem. 2016;62:77–83.CrossRefPubMed Sherrod SD, McLean JA. Systems-wide high-dimensional data acquisition and informatics using structural mass spectrometry strategies. Clin Chem. 2016;62:77–83.CrossRefPubMed
46.
Zurück zum Zitat Schrimpe-Rutledge AC, Codreanu SG, Sherrod SD, McLean JA. Untargeted metabolomics strategies—challenges and emerging directions. J Am Soc Mass Spectrom. 2016;27:1897–905. Schrimpe-Rutledge AC, Codreanu SG, Sherrod SD, McLean JA. Untargeted metabolomics strategies—challenges and emerging directions. J Am Soc Mass Spectrom. 2016;27:1897–905.
47.
Zurück zum Zitat Regehr KJ, Domenech M, Koepsel JT, Carver KC, Ellison-Zelski SJ, Murphy WL, Schuler LA, Alarid ET, Beebe DJ. Biological implications of polydimethylsiloxane-based microfluidic cell culture. Lab Chip. 2009;9:2132–9.CrossRefPubMedPubMedCentral Regehr KJ, Domenech M, Koepsel JT, Carver KC, Ellison-Zelski SJ, Murphy WL, Schuler LA, Alarid ET, Beebe DJ. Biological implications of polydimethylsiloxane-based microfluidic cell culture. Lab Chip. 2009;9:2132–9.CrossRefPubMedPubMedCentral
48.
Zurück zum Zitat Berthier E, Young EWK, Beebe D. Engineers are from PDMS-land, biologists are from polystyrenia. Lab Chip. 2012;12:1224–37.CrossRefPubMed Berthier E, Young EWK, Beebe D. Engineers are from PDMS-land, biologists are from polystyrenia. Lab Chip. 2012;12:1224–37.CrossRefPubMed
49.
Zurück zum Zitat Toepke MW, Beebe DJ. PDMS absorption of small molecules and consequences in microfluidic applications. Lab Chip. 2006;6:1484–6.CrossRefPubMed Toepke MW, Beebe DJ. PDMS absorption of small molecules and consequences in microfluidic applications. Lab Chip. 2006;6:1484–6.CrossRefPubMed
50.
Zurück zum Zitat Opal SM, Scannon PJ, Vincent JL, White M, Carroll SF, Palardy JE, Parejo NA, Pribble JP, Lemke JH. Relationship between plasma levels of lipopolysaccharide (LPS) and LPS-binding protein in patients with severe sepsis and septic shock. J Infect Dis. 1999;180:1584–9.CrossRefPubMed Opal SM, Scannon PJ, Vincent JL, White M, Carroll SF, Palardy JE, Parejo NA, Pribble JP, Lemke JH. Relationship between plasma levels of lipopolysaccharide (LPS) and LPS-binding protein in patients with severe sepsis and septic shock. J Infect Dis. 1999;180:1584–9.CrossRefPubMed
51.
Zurück zum Zitat Penkowa M, Moos T, Carrasco J, Hadberg H, Molinero A, Bluethmann H, Hidalgo J. Strongly compromised inflammatory response to brain injury in interleukin-6-deficient mice. Glia. 1999;25:343–57.CrossRefPubMed Penkowa M, Moos T, Carrasco J, Hadberg H, Molinero A, Bluethmann H, Hidalgo J. Strongly compromised inflammatory response to brain injury in interleukin-6-deficient mice. Glia. 1999;25:343–57.CrossRefPubMed
52.
Zurück zum Zitat Chisholm SP, Cervi AL, Nagpal S, Lomax AE. Interleukin-17A increases neurite outgrowth from adult postganglionic sympathetic neurons. J Neurosci. 2012;32:1146–55.CrossRefPubMed Chisholm SP, Cervi AL, Nagpal S, Lomax AE. Interleukin-17A increases neurite outgrowth from adult postganglionic sympathetic neurons. J Neurosci. 2012;32:1146–55.CrossRefPubMed
53.
Zurück zum Zitat Osburg B, Peiser C, Domling D, Schomburg L, Ko YT, Voigt K, Bickel U. Effect of endotoxin on expression of TNF receptors and transport of TNF-alpha at the blood-brain barrier of the rat. Am J Physiol Endocrinol Metab. 2002;283:E899–908.CrossRefPubMed Osburg B, Peiser C, Domling D, Schomburg L, Ko YT, Voigt K, Bickel U. Effect of endotoxin on expression of TNF receptors and transport of TNF-alpha at the blood-brain barrier of the rat. Am J Physiol Endocrinol Metab. 2002;283:E899–908.CrossRefPubMed
54.
Zurück zum Zitat Wang W, Lv S, Zhou Y, Fu J, Li C, Liu P. Tumor necrosis factor-alpha affects blood-brain barrier permeability in acetaminophen-induced acute liver failure. Eur J Gastroenterol Hepatol. 2011;23:552–8.CrossRefPubMed Wang W, Lv S, Zhou Y, Fu J, Li C, Liu P. Tumor necrosis factor-alpha affects blood-brain barrier permeability in acetaminophen-induced acute liver failure. Eur J Gastroenterol Hepatol. 2011;23:552–8.CrossRefPubMed
55.
Zurück zum Zitat Micheli V, Camici M, Tozzi MG, Ipata PL, Sestini S, Bertelli M, Pompucci G. Neurological disorders of purine and pyrimidine metabolism. Curr Top Med Chem. 2011;11:923–47.CrossRefPubMed Micheli V, Camici M, Tozzi MG, Ipata PL, Sestini S, Bertelli M, Pompucci G. Neurological disorders of purine and pyrimidine metabolism. Curr Top Med Chem. 2011;11:923–47.CrossRefPubMed
56.
58.
Zurück zum Zitat Anderson G, Maes M, Berk M. Inflammation-related disorders in the tryptophan catabolite pathway in depression and somatization. Adv Protein Chem Struct Biol. 2012;88:27–48.CrossRefPubMed Anderson G, Maes M, Berk M. Inflammation-related disorders in the tryptophan catabolite pathway in depression and somatization. Adv Protein Chem Struct Biol. 2012;88:27–48.CrossRefPubMed
59.
Zurück zum Zitat Bein A, Zilbershtein A, Golosovsky M, Davidov D, Schwartz B. LPS induces hyper-permeability of intestinal epithelial cells. J Cell Physiol. 2016;232:381–90. Bein A, Zilbershtein A, Golosovsky M, Davidov D, Schwartz B. LPS induces hyper-permeability of intestinal epithelial cells. J Cell Physiol. 2016;232:381–90.
60.
Zurück zum Zitat Lin X, Barravecchia M, Kothari P, Young JL, Dean DA. beta1-Na(+), K(+)-ATPase gene therapy upregulates tight junctions to rescue lipopolysaccharide-induced acute lung injury. Gene Ther. 2016;23:489–99.CrossRefPubMed Lin X, Barravecchia M, Kothari P, Young JL, Dean DA. beta1-Na(+), K(+)-ATPase gene therapy upregulates tight junctions to rescue lipopolysaccharide-induced acute lung injury. Gene Ther. 2016;23:489–99.CrossRefPubMed
61.
Zurück zum Zitat Hu M, Liu B. Resveratrol attenuates lipopolysaccharide-induced dysfunction of blood-brain barrier in endothelial cells via AMPK activation. Korean J Physiol Pharmacol. 2016;20:325–32.CrossRefPubMedPubMedCentral Hu M, Liu B. Resveratrol attenuates lipopolysaccharide-induced dysfunction of blood-brain barrier in endothelial cells via AMPK activation. Korean J Physiol Pharmacol. 2016;20:325–32.CrossRefPubMedPubMedCentral
62.
Zurück zum Zitat Etna MP, Giacomini E, Severa M, Coccia EM. Pro- and anti-inflammatory cytokines in tuberculosis: a two-edged sword in TB pathogenesis. Semin Immunol. 2014;26:543–51.CrossRefPubMed Etna MP, Giacomini E, Severa M, Coccia EM. Pro- and anti-inflammatory cytokines in tuberculosis: a two-edged sword in TB pathogenesis. Semin Immunol. 2014;26:543–51.CrossRefPubMed
63.
Zurück zum Zitat Mocchegiani E, Costarelli L, Giacconi R, Malavolta M, Basso A, Piacenza F, Ostan R, Cevenini E, Gonos ES, Monti D. Micronutrient-gene interactions related to inflammatory/immune response and antioxidant activity in ageing and inflammation. A systematic review. Mech Ageing Dev. 2014;136-137:29–49.CrossRefPubMed Mocchegiani E, Costarelli L, Giacconi R, Malavolta M, Basso A, Piacenza F, Ostan R, Cevenini E, Gonos ES, Monti D. Micronutrient-gene interactions related to inflammatory/immune response and antioxidant activity in ageing and inflammation. A systematic review. Mech Ageing Dev. 2014;136-137:29–49.CrossRefPubMed
64.
Zurück zum Zitat Nakagawa Y, Chiba K. Diversity and plasticity of microglial cells in psychiatric and neurological disorders. Pharmacol Ther. 2015;154:21–35.CrossRefPubMed Nakagawa Y, Chiba K. Diversity and plasticity of microglial cells in psychiatric and neurological disorders. Pharmacol Ther. 2015;154:21–35.CrossRefPubMed
Metadaten
Titel
Metabolic consequences of inflammatory disruption of the blood-brain barrier in an organ-on-chip model of the human neurovascular unit
verfasst von
Jacquelyn A. Brown
Simona G. Codreanu
Mingjian Shi
Stacy D. Sherrod
Dmitry A. Markov
M. Diana Neely
Clayton M. Britt
Orlando S. Hoilett
Ronald S. Reiserer
Philip C. Samson
Lisa J. McCawley
Donna J. Webb
Aaron B. Bowman
John A. McLean
John P. Wikswo
Publikationsdatum
01.12.2016
Verlag
BioMed Central
Erschienen in
Journal of Neuroinflammation / Ausgabe 1/2016
Elektronische ISSN: 1742-2094
DOI
https://doi.org/10.1186/s12974-016-0760-y

Weitere Artikel der Ausgabe 1/2016

Journal of Neuroinflammation 1/2016 Zur Ausgabe

Reviewer acknowledgement

Reviewer acknowledgement

Neu in den Fachgebieten Neurologie und Psychiatrie

Akuter Schwindel: Wann lohnt sich eine MRT?

28.04.2024 Schwindel Nachrichten

Akuter Schwindel stellt oft eine diagnostische Herausforderung dar. Wie nützlich dabei eine MRT ist, hat eine Studie aus Finnland untersucht. Immerhin einer von sechs Patienten wurde mit akutem ischämischem Schlaganfall diagnostiziert.

Niedriger diastolischer Blutdruck erhöht Risiko für schwere kardiovaskuläre Komplikationen

25.04.2024 Hypotonie Nachrichten

Wenn unter einer medikamentösen Hochdrucktherapie der diastolische Blutdruck in den Keller geht, steigt das Risiko für schwere kardiovaskuläre Ereignisse: Darauf deutet eine Sekundäranalyse der SPRINT-Studie hin.

Frühe Alzheimertherapie lohnt sich

25.04.2024 AAN-Jahrestagung 2024 Nachrichten

Ist die Tau-Last noch gering, scheint der Vorteil von Lecanemab besonders groß zu sein. Und beginnen Erkrankte verzögert mit der Behandlung, erreichen sie nicht mehr die kognitive Leistung wie bei einem früheren Start. Darauf deuten neue Analysen der Phase-3-Studie Clarity AD.

Viel Bewegung in der Parkinsonforschung

25.04.2024 Parkinson-Krankheit Nachrichten

Neue arznei- und zellbasierte Ansätze, Frühdiagnose mit Bewegungssensoren, Rückenmarkstimulation gegen Gehblockaden – in der Parkinsonforschung tut sich einiges. Auf dem Deutschen Parkinsonkongress ging es auch viel um technische Innovationen.