This article provides researchers, scientists, and drug development professionals with a comprehensive overview of microfluidic platforms for modeling vascular inflammation, with a focus on Transendothelial Electrical Resistance (TEER) as a...
This article provides researchers, scientists, and drug development professionals with a comprehensive overview of microfluidic platforms for modeling vascular inflammation, with a focus on Transendothelial Electrical Resistance (TEER) as a key quantitative metric. It covers the foundational biology of vascular inflammation and the principle of TEER, explores the design and fabrication of vascularized microfluidic chips, details methodological protocols for establishing and applying these models in drug screening and disease studies, and addresses critical troubleshooting and validation strategies. By synthesizing current methodologies and applications, this guide aims to serve as a vital resource for advancing the use of these sophisticated in vitro models in preclinical research.
The vasculature system is not merely a passive network of tubes for blood transport; it is a dynamic, active interface that plays a critical role in the body's inflammatory processes [1]. Vascular endothelial cells provide a selective barrier between the bloodstream and surrounding tissues, regulating the movement of fluids, solutes, and immune cells [2]. During inflammation, this barrier function is deliberately modulatedâinitially to enable immune cell recruitment for host defense, but when dysregulated, leading to chronic inflammatory diseases and pathological tissue damage [1] [2].
Understanding vascular inflammation requires models that accurately recapitulate the complexity of endothelial barrier function and its disruption. Recent advances in microfluidic organ-on-a-chip technology have enabled the development of sophisticated in vitro models that mimic the physiological conditions of vascular inflammation, allowing for real-time, quantitative assessment of barrier integrity through methods such as Transendothelial Electrical Resistance (TEER) measurements [1] [3]. These models are particularly valuable for drug discovery, providing scalable platforms for screening potential therapeutics targeting vascular inflammatory components [1] [4].
In inflammatory states, endothelial cells undergo activation characterized by increased expression of adhesion molecules and changes in cell-cell junctions [1] [5]. Key pro-inflammatory cytokines such as Tumor Necrosis Factor-alpha (TNF-α) and Interferon-gamma (IFN-γ) destabilize vascular endothelial (VE)-cadherin, the main component of endothelial adherens junctions, leading to increased vascular permeability [1]. Simultaneously, endothelial activation promotes increased expression of intercellular adhesion molecule-1 (ICAM-1) and vascular cell adhesion molecule-1 (VCAM-1), facilitating immune cell attachment and transmigration [1] [5].
The integrity of the endothelial barrier is maintained through complex interactions between tight junctions, adherens junctions, and the cytoskeleton. During inflammation, pro-inflammatory mediators trigger intracellular signaling pathways that lead to the phosphorylation of junctional proteins, their internalization, and subsequent breakdown of barrier function [1]. This process is characterized by a shift from a restrictive to a permeable endothelial phenotype, allowing for increased paracellular flux [2].
The inflammatory response in vasculature involves multiple overlapping signaling pathways. The vascular endothelial growth factor (VEGF) signaling pathway plays a central role, with VEGF-A binding to VEGFR-2 promoting angiogenesis and vascular permeability [2]. Simultaneously, the Angiopoietin-Tie2 system regulates vascular stability, with Ang-2 acting as an antagonist to Tie2 signaling and promoting vascular destabilization during inflammation [2]. In lymphatic vessels, VEGF-C and VEGF-D binding to VEGFR-3 drive lymphangiogenesis, which participates in the resolution of inflammation [2].
Additional inflammatory mediators include chemokines of the CCL and CXCL families, which attract immune cells to sites of inflammation [5]. Activated vascular cells also secrete cytokines including IL-6, IL-8, and MCP-1, further amplifying the inflammatory response [5]. Pattern recognition receptors such as Toll-like receptors (TLRs) and the receptor for advanced glycation endproducts (RAGE) enhance the inflammatory response within the vasculature when activated [5].
Figure 1: Signaling Pathways in Vascular Inflammation. This diagram illustrates the key molecular events in endothelial activation and barrier dysfunction during inflammation, highlighting the roles of pro-inflammatory cytokines, junctional disruption, adhesion molecule expression, and growth factor signaling.
Transepithelial/Transendothelial Electrical Resistance (TEER) is a widely accepted quantitative technique to measure the integrity of tight junction dynamics in cell culture models of endothelial and epithelial monolayers [3]. TEER reflects the ionic conductance of the paracellular pathway in the endothelial monolayer, providing a sensitive, non-invasive method to monitor live cells during various stages of growth, differentiation, and inflammatory challenge [3]. The technique can be performed in real-time without cell damage and is based on measuring ohmic resistance or impedance across a wide spectrum of frequencies [3].
The fundamental principle of TEER measurement applies Ohm's law to cellular monolayers. In practice, an alternating current (AC) voltage signal is applied across the endothelial layer, and the resulting current is measured to calculate resistance [3]. TEER values are typically reported in Ω·cm² and are calculated by subtracting the blank resistance of the membrane support and medium from the total resistance, then multiplying by the effective surface area of the membrane [3]. This normalization allows for comparison across different experimental setups and platforms.
Traditional TEER measurements have been performed using culture inserts with semipermeable membranes, but recent advances have adapted this technique for microfluidic organ-on-a-chip platforms [1] [3]. The OrganoPlate 3-lane 64 platform enables the formation of 64 perfused human umbilical vein endothelial cell (HUVEC) tubules in a standard 384-well plate format, compatible with automated TEER measurement devices [1] [4]. This system allows for continuous TEER monitoring inside an incubator for long-term permeability studies under flow conditions, more closely mimicking the physiological environment than static cultures [1].
Microfluidic TEER platforms offer significant advantages for vascular inflammation studies, including the ability to sensitively detect low barrier resistance characteristic of endothelial models, perform parallel measurements across multiple replicates, and combine TEER with other readouts such as high-content imaging and immunofluorescence [1] [4]. The platform's design incorporates extracellular matrix (ECM) gels in microfluidic channels using capillary barriers (phaseguides), creating independently accessible microfluidic channels on either side of an ECM scaffold where endothelial tubules form [1].
Table 1: Factors Affecting TEER Measurements in Vascular Models
| Factor Category | Specific Factors | Impact on TEER | Optimization Strategies |
|---|---|---|---|
| Biological Factors | Cell type and origin [6] | HUVECs typically show lower baseline resistance than epithelial cells | Select appropriate cell type for physiological context |
| Cell confluency and culture duration [6] | Incomplete monolayers significantly reduce TEER | Ensure full confluency before measurements | |
| Passage number and cell health [3] | Higher passages may exhibit barrier dysfunction | Use consistent, low-passage cells | |
| Environmental Factors | Temperature fluctuations [6] | TEER sensitive to temperature changes | Maintain 37°C during measurements |
| Medium composition and serum [3] [6] | Serum components can affect junction formation | Standardize medium formulation | |
| Contamination (mycoplasma, bacteria) [6] | Dramatically reduces barrier function | Routine contamination checks | |
| Technical Factors | Electrode design and placement [3] [6] | Affects current density distribution | Use appropriate electrodes; standardize placement |
| Measurement frequency [3] | Prevents electrode polarization | Standardize AC frequency (e.g., 12.5 Hz) | |
| Membrane properties and coatings [6] | Affects cell attachment and junction formation | Consistent membrane selection |
The following protocol outlines the methodology for establishing and analyzing vascular inflammation models in microfluidic platforms, with integrated TEER measurements as a primary readout of barrier function [1].
Figure 2: Experimental Workflow for Vascular Inflammation-on-a-Chip. This workflow outlines the key steps in establishing, perturbing, and analyzing vascular inflammation models in microfluidic platforms with integrated TEER measurements.
Day 1: Platform Preparation and ECM Seeding
Day 2: Endothelial Tubule Formation
Day 4: Baseline Assessment and Inflammation Induction
Days 4-6: Continuous Monitoring and Endpoint Analysis
TEER data should be normalized to baseline values and expressed as percentage change from baseline or as absolute TEER values (Ω·cm²) [3]. Statistical analysis should include appropriate replicates (n ⥠3 per condition) and account for multiple comparisons when testing various inflammatory stimuli or drug treatments.
In parallel, quantitative analysis of immunofluorescence images should assess:
Table 2: Quantitative Responses in Vascular Inflammation Models
| Experimental Condition | TEER Response | ICAM-1 Expression | VE-Cadherin Organization | Immune Cell Migration |
|---|---|---|---|---|
| Baseline (No inflammation) | Stable high TEER (>50 Ω·cm²) [3] | Low baseline expression | Continuous junctional staining | Minimal adhesion |
| TNF-α (10 ng/mL) | ~40-60% decrease in 24h [1] | Moderate increase | Discontinuous, internalized | Increased adhesion |
| IFN-γ (50 ng/mL) | ~20-40% decrease in 24h [1] | Moderate increase | Mild disruption | Moderate adhesion |
| TNF-α + IFN-γ | ~60-80% decrease in 24h [1] | Strong increase | Severe disruption | Extensive adhesion/transmigration |
| PBMC Co-culture | ~50-70% decrease in 24h [1] | Strong increase | Severe disruption | Extensive transmigration |
Table 3: Essential Research Reagents for Vascular Inflammation Studies
| Reagent Category | Specific Examples | Function in Vascular Research | Application Notes |
|---|---|---|---|
| Endothelial Cells | HUVECs (Lonza C2519AS) [1] | Primary model for human vascular endothelium | Use passages 3-5 for optimal barrier function |
| Culture Media | EGM-2 Medium (Lonza CC-3162) [1] | Supports endothelial growth and function | Contains growth factors for endothelial maintenance |
| Pro-inflammatory Cytokines | TNF-α, IFN-γ [1] | Induce endothelial activation and barrier disruption | Typical working concentrations: 10-100 ng/mL |
| Extracellular Matrix | Rat tail collagen I (4 mg/mL) [1] | Provides physiological substrate for tubule formation | Polymerization conditions critical for channel stability |
| Adhesion Molecule Antibodies | Anti-ICAM-1, Anti-VE-cadherin [1] | Detect endothelial activation and junction disruption | Essential for immunofluorescence endpoint analysis |
| Immune Cells | Peripheral Blood Mononuclear Cells (PBMCs) [1] | Model leukocyte-endothelial interactions | Isolate from buffy coat; can pre-stimulate with CD3/CD28 |
| Viability Dyes | CellTracker Orange CMRA [1] | Label immune cells for tracking | Use working concentration of 2.5 μM |
| Flow Markers | Fluorescent dextrans [1] | Alternative permeability assessment | Complementary to TEER measurements |
Low Baseline TEER Values
Inconsistent TEER Measurements
Variable Inflammatory Responses
The basic vascular inflammation model can be extended to study more complex physiological scenarios:
These advanced applications leverage the core principles and protocols outlined above while incorporating additional cellular complexity or experimental endpoints to address specific research questions in vascular inflammation.
The vascular endothelium, a single layer of cells lining the blood vessels, serves as a critical selective barrier between the circulation and surrounding tissues. Endothelial dysfunction is a pivotal event in the initiation and progression of inflammatory diseases, characterized by compromised barrier integrity, enhanced leukocyte adhesion, and aberrant signaling [7] [8]. Inflammatory conditions disrupt the delicate balance of endothelial cell function, leading to increased permeability and the pathological transmigration of immune cells. Understanding these mechanisms is essential for developing novel therapeutic strategies for cardiovascular diseases, inflammatory bowel disease, and other conditions where vascular inflammation plays a central role [7] [8].
The emergence of advanced microfluidic platform models has revolutionized the study of vascular inflammation by providing more physiologically relevant human in vitro systems. These platforms, particularly those enabling real-time, high-throughput measurement of Trans-Endothelial Electrical Resistance (TEER), allow for the precise quantification of endothelial barrier function under controlled inflammatory stimuli [1] [9]. This application note details the integration of these platforms and associated protocols to model endothelial dysfunction, providing researchers with a robust framework for investigating the molecular pathways underlying vascular barrier disruption.
Research utilizing microfluidic models has yielded critical quantitative data on endothelial barrier responses. The following tables summarize key findings regarding the impact of inflammatory cytokines on barrier integrity and the expression of associated adhesion molecules.
Table 1: Impact of Inflammatory Cytokines on Endothelial Barrier Integrity (TEER) in Microfluidic Models
| Cytokine Stimulus | Concentration Range | Exposure Time | Effect on TEER | Reported Significance | Source Model |
|---|---|---|---|---|---|
| TNFα | 10â100 ng/mL | 24â48 hours | Significant Decrease | Dose-dependent reduction in barrier function [10] | 3D HUVEC tubules in OrganoPlate [1] |
| IFN-γ | 10â100 ng/mL | 24â48 hours | Significant Decrease | Dose-dependent reduction; synergistic effect with TNFα [1] | 3D HUVEC tubules in OrganoPlate [1] |
| TNFα + IFN-γ | 10â100 ng/mL each | 24â48 hours | Largest Decrease | Synergistic, dose-dependent effect on barrier disruption [1] | 3D HUVEC tubules in OrganoPlate [1] |
Table 2: Inflammatory Marker Expression in 3D Microvessel Models Following TNFα Stimulation
| Parameter Measured | Baseline Expression | Post-TNFα Stimulation | Effect of NFκB Inhibition (BAY 11-7082) | Reference |
|---|---|---|---|---|
| Monocyte Adhesion | Low | Marked Induction | Significantly abrogated | [10] |
| VCAM-1 Expression | Low | Marked Induction | Significantly abrogated | [10] |
| IL-6 Production | Low | Marked Induction | Significantly abrogated | [10] |
| VE-Cadherin Localization | Intact junctions | Disruption | Partial restoration of junction integrity | [10] |
This protocol outlines the steps for culturing endothelial cells in a microfluidic device and inducing inflammation using cytokines or immune cells [1].
Key Research Reagent Solutions:
Procedure:
This protocol describes the specific methodology for performing continuous TEER measurements in the OrganoPlate platform to quantify barrier function [1] [9].
Procedure:
Following functional assessments, the same microvessels can be fixed and stained for phenotypic and molecular analysis [1] [11].
Procedure:
A key pathway mediating inflammatory responses in the endothelium is the NFκB pathway, activated by cytokines like TNFα.
Diagram 1: TNFα-NFκB Signaling in Endothelial Dysfunction. This pathway shows how TNFα binding triggers NFκB activation, leading to the expression of pro-inflammatory mediators and disruption of endothelial barrier integrity, a process that can be inhibited by BAY 11-7082 [10].
In addition to NFκB signaling, metabolic reprogramming of endothelial cells towards aerobic glycolysis (the Warburg effect) and the induction of oxidative stress are critical mechanisms that disrupt barrier integrity by impairing junctional stability and nitric oxide bioavailability [7] [8].
A comprehensive study of endothelial dysfunction involves a multi-step process, from model establishment to data analysis.
Diagram 2: Integrated Workflow for Vascular Inflammation Studies. This flowchart outlines the sequential steps for modeling and analyzing endothelial dysfunction in a microfluidic platform, combining functional TEER measurements with molecular endpoint analyses [1] [11] [10].
Table 3: Key Research Reagent Solutions for Vascular Inflammation-on-a-Chip Models
| Item Category | Specific Product/Model | Critical Function in the Protocol |
|---|---|---|
| Microfluidic Platform | OrganoPlate 3-lane 64 | Provides a scalable, high-throughput platform with 64 independent, perfusable microfluidic circuits for 3D cell culture [1]. |
| Endothelial Cells | Primary HUVECs (Lonza, C2519AS) | Standard human endothelial cells for forming the vascular tubule. HBMECs are used for brain-specific models [1] [11]. |
| Cell Culture Medium | EGM-2 BulletKit (Lonza, CC-3162) / EGM-2MV | Optimized medium with growth factors (VEGF, FGF) for the maintenance and proliferation of microvascular endothelial cells [1] [11]. |
| Extracellular Matrix | Rat Tail Collagen-I (4 mg/mL) | Forms the 3D hydrogel scaffold that supports the structure of the endothelial tubule and provides a physiological microenvironment [1]. |
| Inflammatory Cytokines | Recombinant Human TNFα & IFN-γ | Key stimuli to experimentally induce endothelial activation, disrupt junctions, and create an pro-inflammatory state [1] [10]. |
| NFκB Pathway Inhibitor | BAY 11-7082 | A pharmacological inhibitor used to validate the specific role of the NFκB signaling pathway in the inflammatory response [10]. |
| Immunostaining Antibodies | Anti-VE-Cadherin, Anti-ICAM-1, Phalloidin (F-actin) | Essential reagents for endpoint immunofluorescence analysis of junctional integrity, endothelial activation, and cytoskeletal remodeling [1] [10]. |
| TEER Measurement Device | OrganoTEER for OrganoPlate 3-lane 64 | Enables sensitive, real-time, and label-free quantification of endothelial barrier integrity across all 64 chips in the platform [1] [9]. |
| Psma-alb-56 | Psma-alb-56, MF:C66H95N11O18, MW:1330.5 g/mol | Chemical Reagent |
| m-PEG12-Hydrazide | m-PEG12-Hydrazide, MF:C26H54N2O13, MW:602.7 g/mol | Chemical Reagent |
Transepithelial/Transendothelial Electrical Resistance (TEER) is a quantitative, non-invasive technique used to measure the integrity and function of cellular barriers formed by epithelial and endothelial cell monolayers in vitro [12] [3]. These barriers are critical for controlling the movement of ions, nutrients, and other molecules throughout the body, and their dysfunction is implicated in numerous disease states [13].
The measurement is based on a fundamental electrophysiological principle: the tight junctions between adjacent cells form a paracellular pathway that restricts the passive flow of ions. When these junctions are intact, they create a high-resistance electrical barrier. TEER directly measures this electrical resistance, providing a functional readout of barrier health [12] [14].
TEER's primary advantage lies in its ability to provide real-time, quantitative data without damaging the cells, allowing researchers to monitor the same culture throughout an experimentâfrom barrier formation and maturation to the application of experimental treatments [12] [13].
The most common method for determining TEER uses Ohm's law (V=IR). In this setup, two electrodes are placed on either side of a cellular monolayer grown on a semi-permeable filter insert [3] [14]. A small, alternating current (AC) is applied across the cell layer, and the resulting voltage drop is measured. The resistance (R) is calculated from the current (I) and voltage (V) [12] [13].
The final TEER value is normalized to the surface area of the membrane to allow for comparisons across different experimental setups, using the formula:
TEER (Ω·cm²) = (RTOTAL - RBLANK) à Membrane Area (cm²) [3]
Where:
Commercial systems like the EVOM series (World Precision Instruments) typically use a low-frequency AC square wave (e.g., 12.5 Hz) to avoid electrode polarization and cell damage that can be caused by direct current [3].
A more advanced technique, Electrochemical Impedance Spectroscopy (EIS), measures the complex impedance of a cell layer across a wide spectrum of frequencies [16] [14]. Unlike single-frequency measurements, EIS can distinguish between the resistive and capacitive properties of the tissue.
The following diagram illustrates the generalized workflow for planning and executing a TEER experiment, from initial setup to data interpretation.
The application of TEER is particularly powerful in the context of vascular inflammation research using advanced microfluidic "organ-on-a-chip" models. These devices aim to recapitulate the physiological and pathological conditions of human barriers more accurately than traditional static cultures [3] [14].
In vascular models, the endothelium forms a critical barrier between the blood and surrounding tissues. During inflammation, pro-inflammatory cytokines (e.g., TNF-α, IL-1β) disrupt endothelial tight junctions, leading to increased vascular permeability. TEER provides a sensitive, quantitative means to monitor this barrier breakdown in real-time and to evaluate the efficacy of potential therapeutic compounds [13].
However, integrating TEER into microfluidic devices presents unique challenges and considerations:
TEER measurement is an indispensable tool in pharmaceutical research, applied across multiple stages of drug development.
The table below lists typical TEER values for different cell types as reported in scientific literature, providing a reference for researchers assessing their in vitro models.
| Cell Type / Tissue Model | Representative TEER Value (Ω·cm²) | Significance and Context |
|---|---|---|
| Blood-Brain Barrier (BBB) Endothelium (in vitro) | 500 - 600+ [17] [16] | High resistance is critical for protecting the brain; models aim to achieve high values to be physiologically relevant. |
| Retinal Pigment Epithelium (RPE) | ~500 - 600 [17] | Considered mature and functionally intact at this range. |
| Intestinal Epithelium (e.g., Caco-2) | Varies widely | Model-dependent; used for predicting oral drug absorption. |
| Bronchiolar Epithelium (16HBE) | 955 - 1034 [16] | Represents a tight barrier in the pulmonary system. |
| Generic "Healthy" Barrier | High | Indicates well-formed tight junctions and intact monolayer [12]. |
| Generic "Compromised" Barrier | Low | Suggests leaky junctions due to toxicity, disease, or inflammation [12]. |
This protocol details the standard method for measuring TEER in Transwell-style cultures using a manual voltohmmeter and "chopstick" electrodes.
Table: Essential Materials for TEER Measurement
| Item | Function | Notes |
|---|---|---|
| TEER Voltohmmeter (e.g., EVOM2) | Instrument that applies AC and measures voltage/current. | Uses a low-frequency AC signal (e.g., 12.5 Hz) to prevent cell damage and electrode polarization [3]. |
| Ag/AgCl "Chopstick" Electrodes (e.g., STX2) | Paired electrodes for current application and voltage sensing. | Silver/Silver Chloride electrodes are standard for stable potential [3] [14]. Handle carefully to avoid distortion. |
| Cell Culture Inserts | Permeable support for growing cell monolayers. | Various sizes (e.g., 6, 12, 24-well format). Material (e.g., polycarbonate, polyester) may vary. |
| Cell Culture Medium | Maintains cell viability during measurement. | Should be equilibrated to experimental temperature and CO2 levels. |
System Calibration and Blank Measurement:
Sample Preparation:
Resistance Measurement:
Data Calculation:
Post-Measurement Handling:
For researchers requiring more detailed electrical characterization of their barrier models, the following protocol outlines the use of Electrochemical Impedance Spectroscopy (EIS).
The relationship between the biological structure and the electrical model used in EIS is illustrated below.
TEER remains the gold standard technique for the functional assessment of in vitro barrier models due to its quantitative, non-invasive, and real-time nature. Its application is vital for validating the integrity of microfluidic vascular models before they are used in inflammation studies, drug permeability screening, or toxicity testing. By following standardized protocols and understanding the critical factors that influence the measurement, researchers can reliably use TEER to generate robust, reproducible data that advances our understanding of barrier function in health and disease.
Transepithelial/transendothelial electrical resistance (TEER) is a gold-standard, quantitative technique used to measure the integrity and permeability of cellular monolayers in vitro [3]. This non-invasive method is paramount for assessing the functionality of biological barriersâsuch as the blood-brain barrier, intestinal epithelium, and vascular endotheliumâby reporting on the tightness of the tight junctions between adjacent cells [18] [19]. In the specific context of microfluidic platform modeling of vascular inflammation, TEER provides a real-time, sensitive readout of endothelial barrier integrity, which is crucial for studying disease mechanisms and screening potential therapeutics [20] [9].
The core electrical principle of TEER is founded on measuring the resistance to the flow of ions across a cellular monolayer. Tight junctions form a seal that controls the paracellular pathway, the route between cells. When these junctions are intact and functional, they restrict the passive flow of ions, resulting in a high electrical resistance. Conversely, compromised barrier function, a hallmark of vascular inflammation, leads to increased ion permeability and a measurable drop in electrical resistance [3] [19]. TEER, therefore, serves as a direct proxy for the dynamic state of tight junctions and the overall health of the cellular barrier.
The measurement of TEER is a direct application of Ohm's law (V = IR), where the electrical resistance (R) is calculated from a known applied alternating current (AC) and the resulting voltage drop across the cell layer [3]. The use of AC voltage is critical to avoid damaging the cells and electrodes, which can occur with direct current (DC) [3]. The total measured resistance ((R{TOTAL})) across a porous membrane and the cell monolayer is a composite value. To isolate the component specific to the cell layer ((R{TISSUE})), the resistance of a blank membrane ((R_{BLANK})) is measured and subtracted [3]:
[R{TISSUE}(Ω)=R{TOTAL}âR_{BLANK}]
This cell-specific resistance is inversely proportional to the effective membrane area. To enable comparisons across different experimental setups, the TEER value is normalized to the surface area and reported in the standardized units of Ω·cm² [3]:
[TEER{REPORTED} = R{TISSUE}(Ω) \times M_{AREA}(cm^2)]
The paracellular pathway is the primary route for ion flow measured by TEER. This pathway is gated by tight junctions, multiprotein complexes that form a selective seal between endothelial or epithelial cells [21]. The transmembrane proteins claudins and occludin are the primary architects of these junctional strands, creating a barrier that dictates the selective permeability of ions and small molecules [21]. The integrity of this protein network is dynamically regulated and can be disrupted by inflammatory mediators.
It is important to distinguish that TEER specifically reflects the ionic conductance of the paracellular pathway. This is different from the permeability to non-electrolyte tracer molecules (e.g., dextrans), which indicates paracellular water flow and effective pore size [3]. This makes TEER an exceptionally sensitive tool for detecting subtle, real-time changes in barrier function that may not be immediately apparent with tracer-based methods.
In vascular inflammation, pro-inflammatory cytokines such as Tumor Necrosis Factor-alpha (TNFα) and Interferon-gamma (IFN-γ) directly target endothelial tight junctions, leading to barrier disruption [20]. These cytokines trigger intracellular signaling pathways that result in the phosphorylation and internalization of key junctional proteins, most notably VE-Cadherin, which is the main component of endothelial adherens junctions and is essential for barrier regulation [20]. The destabilization of VE-Cadherin and associated tight junction proteins like Zonula Occludens-1 (ZO-1) increases paracellular permeability, an effect that is robustly quantified by a decrease in TEER.
Concurrently, endothelial activation occurs, characterized by the increased surface expression of adhesion proteins like ICAM-1 [20]. This facilitates the adhesion and transendothelial migration of immune cells, such as peripheral blood mononuclear cells (PBMCs), which can further exacerbate barrier disruption. The relationship between immune cell migration and vascular leakage is complex and not fully understood, highlighting the need for sophisticated in vitro models that can concurrently monitor TEER and immune cell behavior [20].
Advanced organ-on-a-chip platforms, such as the OrganoPlate, have been developed to model vascular inflammation and integrate continuous, high-throughput TEER measurements [20] [9]. These models demonstrate the precise utility of TEER in capturing barrier dynamics. The following table summarizes typical TEER responses to inflammatory stimuli in such a system:
Table 1: TEER Response to Inflammatory Stimuli in a Vascular Inflammation-on-a-Chip Model [20] [9]
| Inflammatory Stimulus | Observed Effect on TEER | Correlated Biological Events |
|---|---|---|
| TNFα & IFN-γ (Cytokines) | Significant, dose-dependent decrease in TEER values over 24-48 hours. | Destabilization of VE-Cadherin, increased ICAM-1 expression, changes in endothelial cell morphology. |
| Activated PBMCs | Decrease in TEER values. | Immune cell adhesion to endothelium via ICAM-1, followed by transendothelial migration. |
| Control (No Stimulus) | Stable, high TEER values. | Intact VE-Cadherin localization, low ICAM-1 expression, confluent endothelial monolayer. |
This data confirms that TEER is a highly sensitive and reliable metric for quantifying the impact of both biochemical (cytokines) and cellular (immune cells) inflammatory triggers on endothelial barrier function.
This protocol details the steps for culturing endothelial tubules in an OrganoPlate 3-lane 64 and performing real-time TEER measurements to assess barrier integrity under inflammatory conditions [20].
Research Reagent Solutions Table 2: Essential Materials for Vascular Inflammation TEER Assay [20]
| Item | Function/Description | Example |
|---|---|---|
| OrganoPlate 3-lane 64 | A microfluidic 384-well plate format with 64 independent chips, each with three channels, enabling high-throughput perfusion culture. | Mimetas |
| Human Umbilical Vein Endothelial Cells (HUVECs) | Primary endothelial cells used to form the vascular tubules. | Lonza, C2519AS |
| EGM-2 Medium | Specialized growth medium for HUVEC culture and maintenance. | Lonza, CC-3162 |
| Extracellular Matrix (ECM) Gel | A hydrogel scaffold (e.g., collagen) injected into the middle channel against which endothelial tubules form. | Corning Matrigel |
| Inflammatory Cytokines | Used to induce endothelial activation and barrier disruption. | Recombinant Human TNFα & IFN-γ |
| Peripheral Blood Mononuclear Cells (PBMCs) | Human immune cells used to model immune-endothelial interactions. | Isolated from buffy coat |
| OrganoTEER Instrument | Automated device for performing parallel TEER measurements across all 64 chips in the OrganoPlate. | Mimetas |
Procedure:
TEER Calculation:
Interpretation:
The following diagram illustrates the key signaling pathways involved in inflammatory disruption of endothelial tight junctions, leading to a decrease in TEER.
Diagram 1: Inflammatory disruption of endothelial barrier.
This workflow outlines the key steps in performing a vascular inflammation assay with integrated TEER measurement.
Diagram 2: TEER assay workflow.
Microfluidic technology has emerged as a transformative tool for modeling vascular inflammation, offering significant advantages over conventional 2D cell culture and animal models. These platforms enable the replication of physiological microenvironments with precise control over biochemical and mechanical cues, providing researchers with more human-relevant data for drug development [22]. By mimicking the 3D architecture and hemodynamic forces of human vasculature, microfluidic systems bridge the critical gap between oversimplified traditional in vitro models and species-divergent in vivo studies [23].
The integration of real-time monitoring capabilities such as Trans-Endothelial Electrical Resistance (TEER) measurements further enhances the value of these platforms for assessing vascular barrier function during inflammatory processes [20] [24]. This combination of physiological relevance and advanced readout technologies makes microfluidics particularly valuable for studying complex processes like immune cell migration and endothelial dysfunction in vascular inflammation research.
Table 1: Performance Comparison of Vascular Inflammation Models
| Parameter | Traditional 2D Models | Animal Models | Microfluidic Systems |
|---|---|---|---|
| Analysis Time | Days [25] | Weeks to months [23] | <90 minutes to days [25] [26] |
| Sample Volume | mL range | Not applicable | Microliter range [25] |
| Throughput | Low to moderate | Low | High (40-96 parallel chips) [26] [27] [20] |
| TEER Measurement Sensitivity | Limited by electrode design | Not directly measurable | High (detects early toxicity) [27] |
| Shear Stress Control | Limited or absent | Physiological but not controllable | Precise control (0.01-0.70 dyn/cm²) [27] |
| Species Relevance | Human cells but simplified | Species differences | Human cells in physiological context [22] |
Table 2: Key Functional Advantages of Microfluidic Vascular Models
| Feature | Advantage | Research Impact |
|---|---|---|
| Unidirectional Flow | Mimics physiological blood flow [26] | Promotes endothelial cell alignment; enables study of flow-dependent inflammation |
| Integrated TEER | Real-time, non-invasive barrier monitoring [20] [24] | Detects subtle changes in vascular permeability before cell death occurs [27] |
| Co-culture Capability | Endothelial, smooth muscle, and immune cells [26] [27] | Recapitulates cellular crosstalk in vascular inflammation |
| Inflammatory Response Modeling | Cytokine exposure and immune cell migration [20] | Enables study of TNFα/IFN-γ induced barrier dysfunction and leukocyte transmigration |
| High-Content Imaging | Optical clarity and accessibility [20] | Facilitates morphological analysis and immunofluorescence alongside functional TEER data |
Purpose: To create a microfluidic model of vascular inflammation for TEER-based assessment of endothelial barrier function.
Materials:
Procedure:
Day 1: Device Preparation and Cell Seeding
Endothelial Cell Seeding:
Medium Perfusion:
Day 4-5: Inflammatory Stimulation and TEER Monitoring
Inflammatory Challenge:
Optional Immune Cell Recruitment:
Day 5-7: Endpoint Analysis
Purpose: To continuously monitor vascular barrier integrity during inflammatory challenge using integrated electrodes.
Materials:
Procedure:
Chip Preparation and Electrode Integration:
Cell Culture and Barrier Formation:
TEER Measurement Protocol:
Inflammatory Stimulation:
Data Interpretation:
Figure 1: Signaling Pathways in Microfluidic Vascular Inflammation Models. This diagram illustrates the key molecular events in cytokine-induced endothelial barrier dysfunction that can be monitored in real-time using TEER measurements in microfluidic platforms.
Figure 2: Integrated Workflow for Vascular Inflammation and TEER Studies. This workflow demonstrates the sequential steps for establishing, challenging, and analyzing vascular inflammation in microfluidic platforms with integrated TEER measurements.
Table 3: Essential Research Reagents for Microfluidic Vascular Inflammation Studies
| Reagent/Cell Type | Function/Application | Example Source/Product |
|---|---|---|
| HUVECs | Primary endothelial cells for vascular tubule formation | Lonza (C2519AS) [20] |
| HCAECs | Coronary artery-specific endothelial cells | Promocell (C-12221) [26] |
| OrganoPlate Platform | Microfluidic device for vessel culture | Mimetas (2-lane-48 UF, 3-lane-64) [26] [20] |
| OrganoTEER System | Automated TEER measurement | Mimetas [20] |
| TNFα & IL-1β | Pro-inflammatory cytokines for barrier challenge | Recombinant human proteins [26] [20] |
| Fibrinogen/Thrombin | ECM hydrogel for 3D support | Sigma-Aldrich [20] |
| Anti-VE-Cadherin | Immunofluorescence of junctions | Various suppliers [20] |
| Anti-ICAM-1 | Marker for endothelial activation | Various suppliers [20] |
| CellTracker Dyes | Immune cell labeling for migration studies | Invitrogen (C34551) [20] |
| PREDICT96 Platform | High-throughput microfluidic system | Nortis [27] |
The development of physiologically relevant in vitro models of the vasculature is a critical endeavor in biomedical research, particularly for the study of vascular inflammation and the assessment of drug efficacy and toxicity. Microfluidic technology has emerged as a powerful tool for creating advanced vascular models, often referred to as vasculature-on-a-chip, that overcome the limitations of conventional two-dimensional (2D) cell cultures and animal studies [22] [29]. These limitations include an inability to replicate the three-dimensional (3D) cellular microenvironment, cell-cell interactions, and dynamic flow conditions present in vivo [22]. Among the various fabrication strategies for engineering microvascular networks in microfluidic devices, two primary approaches have gained prominence: microvessel patterning and vasculogenesis/angiogenesis-based self-assembly [22] [30]. Microvessel patterning involves the direct engraving of vascular-like microchannels into cell-laden hydrogel scaffolds using microfabrication techniques, followed by endothelial cell (EC) seeding [22]. In contrast, the self-assembly approach leverages the innate ability of ECs to form 3D vascular networks spontaneously within hydrogels through vasculogenic and angiogenic processes [22] [30]. This Application Note provides a detailed comparison of these two fabrication strategies, with a specific focus on their application in modeling vascular inflammation and quantifying endothelial barrier integrity via Trans-Endothelial Electrical Resistance (TEER) measurements. We include standardized protocols to facilitate the adoption of these techniques, along with a comparative analysis of their respective strengths and limitations for vascular inflammation research.
The choice between patterning and self-assembly strategies is fundamental, as it dictates the model's architecture, physiological relevance, and applicability to specific research questions, such as those involving vascular inflammation.
Table 1: Comparison of Microvessel Patterning and Self-Assembly Fabrication Strategies
| Feature | Microvessel Patterning | Self-Assembly (Vasculogenesis/Angiogenesis) |
|---|---|---|
| Fundamental Principle | Direct engraving of microchannels into hydrogels using microfabrication; subsequent EC seeding [22] [30] | Spontaneous formation of vascular networks by ECs encapsulated in hydrogels [22] [30] |
| Key Characteristics | Pre-defined geometry; high reproducibility; direct control over vessel size and architecture [22] | Complex, biologically relevant networks; lumen formation; branching morphogenesis [30] [31] |
| Vessel Architecture | Single, straight vessels or simple networks; often rectangular cross-section [31] | Interconnected, capillary-like networks with circular cross-sections [30] |
| Throughput | Moderate; suitable for medium-scale studies | High; scalable for drug screening (e.g., 64-96 chips per plate) [1] [32] |
| Barrier Function Assessment | Compatible with permeability assays; TEER measurement can be challenging due to 3D gel configuration | Highly compatible with real-time TEER monitoring in perfused tubules (e.g., in OrganoPlate platform) [1] [9] |
| Primary Applications | Fundamental studies of shear stress, leukocyte adhesion, molecule transport [22] | Disease modeling (e.g., vascular inflammation), drug screening, organ-specific phenotyping [1] [30] |
This protocol details the creation of a single, perfusable microvessel within a 3D collagen gel using a needle-based sacrificial molding technique [22] [31].
Materials
Procedure
This protocol describes the formation of a self-assembled, capillary-like network by co-culturing ECs with supporting stromal cells in a fibrin hydrogel within a microfluidic device [30].
Materials
Procedure
This protocol is optimized for measuring TEER in self-assembled endothelial tubules within the OrganoPlate platform to quantify inflammatory activation and barrier dysfunction [1] [9].
Materials
Procedure
Successful implementation of the protocols above requires a set of core reagents and specialized equipment.
Table 2: Key Research Reagent Solutions for Vascular-on-Chip Models
| Item | Function/Description | Example Use Case |
|---|---|---|
| OrganoPlate | Microfluidic platform with 40, 64, or 96 chips in a standard microtiter plate format; enables high-throughput, perfused 3D cell culture [1] [9]. | Foundation for self-assembled network formation and continuous TEER measurement. |
| OrganoTEER | Automated instrument for continuous, non-invasive Trans-Endothelial Electrical Resistance measurement in the OrganoPlate [1] [9]. | Gold-standard quantification of endothelial barrier integrity in response to inflammatory stimuli. |
| Type I Collagen | Natural hydrogel derived from rat tail; provides a biocompatible 3D scaffold for cell growth and morphogenesis. | The primary matrix for the sacrificial molding protocol (Protocol 1). |
| Fibrin Gel | Natural hydrogel formed from fibrinogen and thrombin; highly conducive to endothelial cell sprouting and network formation. | The preferred matrix for the self-assembly vasculogenesis protocol (Protocol 2) [30]. |
| HUVECs | Primary Human Umbilical Vein Endothelial Cells; a standard and widely used model for studying vascular endothelium. | The core cellular component for forming the endothelial lumen in both patterning and self-assembly strategies. |
| TNFα & IFN-γ | Pro-inflammatory cytokines that synergistically activate endothelial cells, disrupting junctions and upregulating adhesion molecules [1]. | Used to induce a controlled and reproducible state of vascular inflammation in the model. |
| Anti-VE-Cadherin Antibody | Target for immunofluorescence staining to visualize and quantify the integrity of endothelial adherens junctions. | Endpoint analysis to correlate TEER drop with structural disassembly of the barrier. |
| Anti-ICAM-1 Antibody | Target for immunofluorescence staining to visualize and quantify endothelial activation in response to inflammation [1]. | Endpoint analysis to confirm the pro-inflammatory phenotype of the endothelium. |
| DSPE-PEG46-Folate | DSPE-PEG46-Folate|Folate-Targeted PEG Lipid | DSPE-PEG46-Folate is a high-affinity ligand for targeted drug delivery to folate receptor-rich cancer cells. For Research Use Only. Not for human use. |
| FAK inhibitor 6 | FAK inhibitor 6, MF:C25H24FN5O2S, MW:477.6 g/mol | Chemical Reagent |
Within the context of vascular inflammation research using microfluidic platforms, the ability to quantify endothelial barrier integrity is paramount. The Transendothelial Electrical Resistance (TEER) assay is a gold-standard, non-invasive, and quantitative method for real-time assessment of this barrier function. This application note details a integrated protocol for establishing a functional Human Umbilical Vein Endothelial Cell (HUVEC) tubule network on a basement membrane matrix within a microfluidic device, followed by its validation through TEER measurement. This methodology is essential for researchers and drug development professionals modeling diseases like atherosclerosis or thrombosis, where barrier dysfunction is a critical pathological event [33] [34] [35].
The following diagrams outline the core experimental workflow and the key signaling pathways modulating endothelial barrier function, providing a conceptual map for the protocol.
Diagram 1: A sequential workflow for establishing HUVEC tubules and measuring TEER.
Diagram 2: Key signaling pathways regulating endothelial barrier integrity and TEER.
Table 1: Essential research reagents and materials for the HUVEC tubule formation and TEER assay.
| Item | Function / Description | Example & Notes |
|---|---|---|
| Endothelial Cells | Primary cell source for tubule formation. | Primary HUVEC or HUVEC-CS immortalized line [36]. Use low-passage cells (P2-P8) [37]. |
| Basement Membrane Extract (BME) | 3D substrate that supports tube formation. | Geltrex or Matrigel [37] [38]. Use a reduced-growth factor formulation; concentration is critical (>10 mg/mL) [38]. |
| Endothelial Cell Medium | Base culture medium. | Medium 200PRF, supplemented appropriately [37] [34]. |
| Low Serum Growth Supplement (LSGS) | Provides essential pro-angiogenic factors. | Contains FBS, bFGF, EGF, hydrocortisone, and heparin [37] [34]. |
| cAMP-Elevating Agents | Pharmacological enhancers of barrier function. | 8-CPT-cAMP (250 µM) and Ro-20-1724 (17.5 µM) as positive control [34]. |
| Pro-Inflammatory Cytokines | Inducers of barrier dysfunction for disease modeling. | TNF-α and IL-1β (e.g., 2 ng/mL) to simulate vascular inflammation [33]. |
| Cell Viability Stain | For fluorescent visualization of tubular networks. | Calcein AM (2 µg/mL final concentration) [37] [38]. |
| Microfluidic Chip | Platform with microchannels and integrated electrodes. | PDMS-based chips with or without embedded electrodes for TEER [33]. |
| TEER Measurement System | Instrument for real-time, label-free impedance monitoring. | Electric Cell-substrate Impedance Sensing (ECIS) or similar systems [34]. |
Table 2: Key quantitative parameters for HUVEC tube formation and TEER response to stimuli.
| Parameter | Typical Value / Range | Context and Notes |
|---|---|---|
| Cell Seeding Density | 35,000 - 45,000 cells/cm² [37] | Must be optimized for specific microfluidic device geometry. |
| BME Coating Volume | 50 - 100 µL/cm² [37] | Thicker gels (100 µL/cm²) for smaller wells to prevent drying. |
| Time to Initial Tube Formation | 1 - 2 hours [38] | Endothelial cells begin to align and form cords. |
| Peak Tube Formation | 4 - 6 hours (HUVEC) [37] | Well-defined, lumen-containing tubules are visible. |
| Tube Network Apoptosis | ~18 - 24 hours [37] [38] | Networks begin to deteriorate after this period. |
| Baseline TEER (Confluent HUVEC) | 600 - 1200 Ω [34] | Measured using ECIS system; baseline for healthy monolayer. |
| ICâ â of RAP in 3D Pathological Model | 55.36 µM [33] | Highlights altered drug efficacy in a 3D perfused AS model vs. 2D (32.69 µM). |
Within vascular biology and drug development research, robust in vitro models of inflammation are crucial. This application note provides detailed protocols for inducing and quantifying inflammatory responses in microfluidic vascular platforms, specifically tailored for research on Transendothelial Electrical Resistance (TEER) within a thesis context. We focus on the application of key effector cytokinesâTumor Necrosis Factor-alpha (TNF-α) and Interferon-gamma (IFN-γ)âand Peripheral Blood Mononuclear Cells (PBMCs) to model vascular inflammation, assess barrier integrity, and investigate underlying mechanobiological phenomena [39] [40].
The tables below summarize standard concentrations and key quantitative data for inducing inflammation with cytokines and PBMCs, providing a reference for experimental design.
Table 1: Standard Cytokine Concentrations for Inflammatory Induction
| Cytokine | Typical Working Concentration | Key Functions in Vascular Inflammation | Primary Source |
|---|---|---|---|
| TNF-α | 10-100 ng/mL [39] | Promotes endothelial activation, adhesion molecule (ICAM-1, VCAM-1) expression, and pro-inflammatory cytokine secretion. | Activated macrophages, T cells [39] |
| IFN-γ | 10-100 ng/mL [39] | Synergizes with TNF-α, enhances antigen presentation, and drives Th1-type immune responses. | Activated T cells (Th1), NK cells [39] |
Table 2: Key Quantitative Data from Cytokine and PBMC Assays
| Parameter | Measured Value / Observation | Experimental Context | Significance |
|---|---|---|---|
| PBMC Proliferation (with PHA) | Varies significantly between donor lots [41] | One-way MLR assay; 0.5-1.0 µg/mL PHA [41] | Highlights need for PBMC lot quality control. |
| IFN-γ & TNF-α in PBMC Supernatant | High levels correlate with robust MSC immunosuppressive potency assays [41] | PHA-stimulated PBMCs (0.2-1.0 µg/mL) [41] | Serves as a quality marker for effective PBMC responses. |
| Duration of IFN-γ Neutralization | Required prolonged intervention (>6 days) to attenuate kidney injury [39] | Established anti-MPO glomerulonephritis model in mice [39] | Suggests sustained cytokine presence is critical in chronic models. |
This protocol describes the direct activation of endothelial cells using recombinant cytokines to model vascular inflammation and monitor barrier dysfunction via TEER.
Materials:
Procedure:
This protocol leverages the activation of PBMCs to induce a more complex, cellular model of inflammation, culminating in immune cell transmigration.
Materials:
Procedure:
The following diagrams, created using the specified color palette, illustrate the core signaling pathways and experimental workflows.
Cytokine Signaling in Endothelial Cells
Vascular Inflammation Assay Workflow
Table 3: Essential Materials for Vascular Inflammation Assays
| Item | Function / Application | Example / Note |
|---|---|---|
| Recombinant TNF-α & IFN-γ | Direct induction of endothelial inflammation and barrier dysfunction. | Use high-purity, carrier-free proteins. Aliquot and store at -80°C [39]. |
| PBMCs | Provide a complex, physiologically relevant immune challenge. | Quality varies by donor lot; pre-screen for consistent inflammatory response (high IFN-γ/TNF-α production) [41]. |
| PHA (Phytohemagglutinin) | Mitogen used to activate T-cells within PBMC populations. | Optimal concentration for activation is typically 0.5-1.0 µg/mL [41]. |
| Microfluidic Platform | Provides a dynamic, shear-stress-enabled environment for realistic vascular models. | Devices should allow for TEER measurement and real-time imaging [22] [40]. |
| TEER Measurement System | Quantitative, non-invasive monitoring of endothelial barrier integrity. | Key metric for assessing inflammatory insult and therapeutic intervention [40]. |
| Anti-inflammatory mAbs | Tool for validating specific cytokine pathways (e.g., anti-TNF-α, anti-IFN-γ). | Used as positive controls to confirm mechanism [39]. |
| Collagen I / IV, Fibronectin | ECM coatings for the vascular channel to support endothelial cell adhesion and function. | Mimics the native basement membrane [22]. |
| ACP-5862-d4 | ACP-5862-d4, MF:C26H23N7O3, MW:485.5 g/mol | Chemical Reagent |
| Inosine-2,8-d2 | Inosine-2,8-d2, MF:C10H12N4O5, MW:270.24 g/mol | Chemical Reagent |
Transepithelial/transendothelial electrical resistance (TEER) is a widely accepted quantitative technique to measure the integrity of tight junction dynamics in cell culture models of endothelial and epithelial monolayers [3]. In the context of drug development, TEER values are strong indicators of the integrity of cellular barriers before they are evaluated for transport of drugs or chemicals [3]. The capability to perform TEER measurements in real-time without cell damage makes it particularly valuable for preclinical screening, enabling researchers to monitor barrier integrity throughout the course of an experiment and identify subtle, dynamic responses to compounds [3] [27]. This application note details the implementation of TEER within microfluidic platforms, specifically framing its utility for screening drug efficacy and toxicity in models of vascular inflammation and kidney function.
TEER is a direct measure of the ionic conductance of the paracellular pathway in an epithelial or endothelial monolayer [3]. It reflects the integrity of tight junctionsâthe specialized cell-cell adhesions that regulate paracellular transport. A higher TEER value indicates a tighter, more selective barrier, while a decrease in TEER signifies increased paracellular permeability and a potential loss of barrier integrity [3] [27].
In toxicity and efficacy screening, this is critical because many drug-induced injuries and inflammatory conditions manifest through the disruption of these cellular barriers. TEER serves as a sensitive, non-invasive, and label-free metric to quantify this disruption. Unlike endpoint assays that require cell lysis or fluorescent labels, TEER allows for the repeated assessment of the same tissue replicate over time, enabling the tracking of the onset, progression, and potential recovery from toxic or therapeutic insults [27]. This can reveal early indicators of toxicity before cell death occurs, providing a more nuanced understanding of a compound's mechanism of action [27].
Table 1: Advantages of TEER for Drug Screening Applications
| Feature | Benefit in Drug Screening |
|---|---|
| Non-invasive & Label-free | Permits real-time, longitudinal monitoring of the same tissue; avoids interference from dyes or labels. |
| Quantitative & Sensitive | Provides a numerical value (Ω·cm²) for high-resolution tracking of subtle barrier changes. |
| Early Indicator of Toxicity | Can detect barrier disruption prior to the onset of cell death, offering an earlier readout. |
| Functional Readout | Directly measures a key physiological propertyâbarrier functionârather than a surrogate marker. |
| Compatibility with Microfluidics | Enables integration into high-throughput, physiologically relevant organ-on-a-chip platforms. |
Recent studies demonstrate the practical application of TEER in high-throughput screening environments. The following tables summarize key quantitative findings from seminal research in nephrotoxicity and vascular inflammation.
Table 2: TEER as a Metric for Nephrotoxicity Screening in a High-Throughput Microfluidic Platform (PREDICT96) [27]
| Experimental Condition | Baseline TEER (Ω·cm²) | Response to Cisplatin (40 µM) | Correlation with Other Assays |
|---|---|---|---|
| PT (hRPTEC) / Microvascular (hMVEC) Co-culture | 5 - 10 Ω·cm² | Dose-dependent decrease in TEER. | Well-correlated with LDH cytotoxicity and loss of tight junction coverage. |
| PT (hRPTEC) Mono-culture | 5 - 10 Ω·cm² | No dose-dependent TEER change observed. | Higher baseline LDH release; less differentiated phenotype. |
| Key Finding | Co-culture model is essential for TEER to function as a reliable metric of toxicity. | TEER decrease was detectable prior to increases in cell death, highlighting its early-indicator capability. |
Table 3: TEER for Monitoring Vascular Inflammation in a Scalable Organ-on-a-Chip Platform (OrganoPlate) [20]
| Treatment Condition | Effect on TEER (HUVEC Tubules) | Additional Phenotypic Observations |
|---|---|---|
| Inflammatory Cytokines (TNF-α & IFN-γ) | Significant decrease in TEER values. | Increased ICAM expression; changes in endothelial morphology; destabilization of VE-Cadherin. |
| Human Peripheral Blood Mononuclear Cells (PBMCs) | Decrease in TEER values. | Increased immune cell adhesion and migration. |
| Key Finding | Platform allows parallel TEER measurements in 64 perfused tubules, demonstrating suitability for high-throughput screening of anti-inflammatory compounds. |
Another study utilizing a tissue-engineered venule-like model demonstrated that perfusion with aged human serum (average donor age 65) resulted in increased paracellular permeability and transcriptomic signatures of aging compared to young serum (average donor age 25), further underscoring TEER's role in modeling age-related vascular dysfunction and compound sensitivity [43].
This protocol is adapted from a study using the PREDICT96 platform to evaluate cisplatin-induced toxicity in a human primary kidney proximal tubule (PT) model [27].
4.1.1 Research Reagent Solutions
Table 4: Essential Materials for Nephrotoxicity Screening
| Item | Function / Description | Example |
|---|---|---|
| Primary Cells | Forms the functional kidney barrier. | Human Renal Proximal Tubule Epithelial Cells (hRPTEC). |
| Support Cells | Co-culture to improve physiological relevance and model differentiation. | Human Microvascular Endothelial Cells (hMVEC). |
| Microfluidic Platform | High-throughput culture and measurement. | PREDICT96 system (96 independent microfluidic tissues). |
| TEER Measurement System | Integrated, rapid TEER measurement capability. | PREDICT96 TEER system. |
| Culture Medium | Cell-specific growth medium. | EGM-2 for hMVEC; PT-specific medium for hRPTEC. |
| Toxicant | Model nephrotoxic compound. | Cisplatin. |
| Validation Assays | Endpoint assays to correlate with TEER data. | Lactate Dehydrogenase (LDH) Cytotoxicity Assay; Immunofluorescence for ZO-1. |
4.1.2 Methodological Workflow
4.1.3 Procedure
This protocol is based on a scalable platform for modeling vascular inflammation in the OrganoPlate, combining TEER with immune cell migration assays [20].
4.2.1 Research Reagent Solutions
Table 5: Essential Materials for Vascular Inflammation Screening
| Item | Function / Description | Example |
|---|---|---|
| Endothelial Cells | Forms the vascular tubule barrier. | Human Umbilical Vein Endothelial Cells (HUVECs). |
| Immune Cells | To model immune cell interaction and migration. | Human Peripheral Blood Mononuclear Cells (PBMCs). |
| Microfluidic Platform | Membrane-free, ECM-supported 3D culture. | OrganoPlate 3-lane 64. |
| Automated TEER Reader | For high-throughput TEER measurements. | OrganoTEER measurement system. |
| Extracellular Matrix (ECM) | Physiological scaffold for tubule formation. | Fibrin or Collagen I gel. |
| Inflammatory Cytokines | To induce endothelial activation and barrier disruption. | TNF-α and IFN-γ. |
| Cell Tracking Dye | To visualize and quantify immune cell migration. | CellTracker Orange CMRA. |
4.2.2 Methodological Workflow
4.2.3 Procedure
Successful implementation of TEER for screening requires careful control of experimental conditions:
Integrating TEER measurement into microfluidic organ-on-a-chip platforms transforms it from a simple integrity check into a powerful, dynamic tool for drug discovery. Its ability to provide non-invasive, real-time, and functional data on barrier health makes it ideal for identifying subtle toxicities and efficacious compounds early in the development pipeline. The protocols and data outlined herein provide a framework for deploying TEER in robust, high-throughput screening campaigns for nephrotoxicity and vascular inflammation, ultimately contributing to the development of safer and more effective therapeutics.
Within microfluidic platform modeling of vascular inflammation, the accurate assessment of tissue barrier integrity is paramount. Transepithelial/transendothelial electrical resistance (TEER) has emerged as a gold-standard, non-invasive technique to quantify the integrity of cellular monolayers in real-time [3]. For researchers and drug development professionals, selecting the appropriate measurement system is a critical step in experimental design. This application note provides a detailed comparison between the widely used EVOM2 volt-ohmmeter and the more comprehensive approach of impedance spectroscopy, framing their application within the context of vascular inflammation research on microfluidic platforms. We present structured quantitative data, detailed experimental protocols, and essential toolkits to guide this decision-making process.
The choice between an EVOM2 system and an impedance spectroscopy-based approach hinges on the specific requirements of the experimental workflow, including throughput, data richness, and compatibility with dynamic culture systems.
Table 1: System Comparison for Vascular Inflammation Research
| Feature | EVOM2 (Volt-Ohmmeter) | Impedance Spectroscopy |
|---|---|---|
| Core Measurement | Ohmic resistance at a single, low frequency (e.g., 12.5 Hz) [3] | Complex impedance (magnitude and phase) across a frequency spectrum [44] |
| Primary Output | TEER value (Ω·cm²) | Nyquist plots, Bode plots, equivalent circuit parameters (e.g., TEER, membrane capacitance) [44] |
| Information Depth | Primarily reflects paracellular ionic conductance (tight junction integrity) [3] | Distinguishes paracellular (TEER) and transcellular (cell membrane capacitance) pathways [45] |
| Throughput | EVOM2 Auto: ~96 wells in <3.5 minutes [46] | Varies by system; typically slower than single-frequency readers |
| Key Advantage | Speed, simplicity, and established workflows for standard Transwells [46] | Richer data for mechanistic studies, detects subtle barrier changes [47] [44] |
| Best Suited For | High-throughput integrity screening in standard static or dynamic cultures [46] | Advanced R&D, investigating barrier formation kinetics, and complex co-culture models [47] |
Table 2: Microfluidic Integration and Data Output
| Aspect | EVOM2 | Impedance Spectroscopy |
|---|---|---|
| Integration with Microfluidics | Often uses external ("chopstick") electrodes or custom-integrated wires [17] [45] | Often features electrodes embedded within the microfluidic device itself [47] |
| Spatial Resolution | Low; provides an average resistance for the entire membrane area [3] | Can be very high with HD-MEA chips, enabling impedance mapping of tissue heterogeneity [47] |
| Real-Time Monitoring | Possible with custom add-ons or auto-sampling systems [48] | Inherent capability; standard for many commercial systems [44] |
This protocol adapts the standard EVOM2 for use in a dual-flow milli-fluidic bioreactor model of vascular inflammation, based on established methods in the literature [45].
Research Reagent Solutions & Materials
Procedure
The workflow for this protocol is summarized in the following diagram:
This protocol utilizes a High-Density Microelectrode Array (HD-MEA) chip to monitor endothelial barrier function with high spatiotemporal resolution before and after an inflammatory stimulus [47].
Research Reagent Solutions & Materials
Procedure
The workflow for this advanced protocol is as follows:
Table 3: Essential Research Reagent Solutions for TEER Studies
| Item | Function & Application |
|---|---|
| EVOM2 with EVOM Auto System | Automated TEER measurement system for high-throughput screening in 96-well plates, minimizing handling errors and contamination [46]. |
| High-Density Microelectrode Array (HD-MEA) Chip | A CMOS-integrated chip with thousands of microelectrodes for impedance mapping of epithelial/endothelial tissues with subcellular spatial resolution [47]. |
| Dual-Flow Milli-fluidic Bioreactor | A cell culture system that allows independent perfusion of apical and basal channels, subjecting cells to physiological shear stress while enabling integrated TEER measurements [45]. |
| Ag/AgCl Electrodes | The standard electrode type for TEER, used in EVOM2 "chopstick" electrodes or for custom integration into bioreactors due to their stable and non-polarizable properties [3] [45]. |
| Collagen Type I Coating | A common extracellular matrix protein used to coat microfluidic channels and solid substrates (like HD-MEA chips) to promote endothelial cell adhesion and spreading [47]. |
| Csf1R-IN-6 | Csf1R-IN-6, MF:C20H18N8O3, MW:418.4 g/mol |
| 3CPLro-IN-1 | 3CPLro-IN-1, MF:C29H25NO3, MW:435.5 g/mol |
In the field of microfluidic platform modeling of vascular inflammation, the accurate assessment of barrier integrity is paramount. Transepithelial/transendothelial electrical resistance (TEER) serves as a gold-standard, quantitative technique for measuring the integrity of tight junction dynamics in endothelial and epithelial monolayers [3]. The reliability of TEER data, however, is highly dependent on the method used for electrical measurement. The integration of either two-point or four-point probe methods represents a critical methodological choice, with significant implications for minimizing measurement error and enhancing data accuracy. This application note details the principles, advantages, and limitations of both probe configurations. It provides structured experimental protocols tailored for researchers developing robust models for drug development and vascular inflammation studies, where sensitive and accurate barrier function assessment is essential.
The core difference between the two methods lies in the separation of current injection and voltage measurement functions.
Two-Point Probe Method: This configuration uses two electrodes to perform both current injection and voltage measurement simultaneously [49]. The measured resistance (Rmeasured) is the sum of the true sample resistance (Rsample) and the contact resistances (Rc1, Rc2) at the electrode-sample interfaces, such that Rmeasured = Rsample + Rc1 + Rc2 [50]. This introduces significant error, especially when the sample resistance is low.
Four-Point Probe Method: This system employs four distinct electrodes: the two outer electrodes act as current (or driving) electrodes for current injection, while the two inner electrodes function as voltage (or sensing) electrodes to determine the resulting potential [51]. A key advantage is that the voltage electrodes do not carry significant current, which effectively eliminates the influence of electrode polarization and contact impedance from the measurement [51] [49]. This allows for the determination of the absolute resistance value of the sample itself.
The following table summarizes the key performance differences between the two methods, particularly in the context of biological and materials testing.
Table 1: Comparative Analysis of Two-Point and Four-Point Probe Methods
| Aspect | Two-Point Probe | Four-Point Probe |
|---|---|---|
| Measured Value | Total system resistance (sample + contact resistances) [50] | Absolute sample resistance [50] |
| Contact Resistance | Included in measurement, causing significant error [49] | Effectively eliminated from measurement [51] [49] |
| Typical Resistivity Trend | Higher measured values (Single probe > Two-point > Four-point) [50] | Lower, more accurate absolute values [50] |
| Ideal Use Case | Measuring high-resistance samples; assessing total through-thickness resistance [49] | Measuring low-resistance samples; determining intrinsic material resistivity [49] |
| Data Stability | Challenging for low-resistance samples due to contact noise [49] | High accuracy for low-resistance samples; requires high instrument precision [49] |
| Practical Application | Suitable for testing all types of battery electrodes and supercapacitors [49] | Can fail to distinguish coating influence on low-resistance foil-based electrodes [49] |
The choice of method is heavily influenced by the sample's electrical properties. For high-resistance samples, the contact resistance may be negligible compared to the sample resistance, making the two-point method acceptable. However, for low-resistance samples, such as many biological tissues or conductive foils, the contact resistance can constitute the majority of the measured signal, leading to substantial overestimation of the true sample resistance [50] [49]. The four-point probe method is superior in these scenarios as it directly measures the sample's resistance without the confounding effect of contact impedance [51].
This protocol outlines the use of manual two-point probes, such as the widely used STX2 or STX4 electrodes, often coupled with an Epithelial Voltohmmeter (EVOM) [3] [52].
Materials:
Procedure:
This protocol is modeled on integrated four-electrode systems, such as the ECIS TEER96 platform, which are designed for continuous, automated measurement within a cell culture incubator [53].
Materials:
Procedure:
Table 2: Essential Materials and Reagents for TEER and Vascular Inflammation Studies
| Item | Function/Application |
|---|---|
| EVOM Meter & STX/STX-HTS Electrodes [52] | A manual system for reliable two-point TEER measurement in standard well plate formats (e.g., 24-well). |
| EVOM Auto [52] | Automated two-point TEER system for high-throughput screening, eliminating user placement variability in 24- and 96-well formats. |
| ECIS TEER96 [53] | An automated platform for continuous, four-electrode TEER measurement of 96 samples simultaneously in a standard incubator. |
| OrganoPlate & OrganoTEER [20] [4] | A microfluidic organ-on-a-chip platform for 3D vessel culture, with a compatible TEER measurement apparatus for real-time, perfused barrier assessment. |
| Inflammatory Cytokines (TNF-α, INF-γ) [20] | Used to stimulate endothelial cells and induce a controlled inflammatory response, mimicking vascular inflammation in vitro. |
| Human Umbilical Vein Endothelial Cells (HUVECs) [20] [4] | A primary cell line commonly used to establish in vitro models of the vascular endothelium. |
| FITC-labeled Dextrans [32] | A fluorescent tracer for parallel validation of barrier integrity via macromolecular permeability assays. |
The decision-making process for integrating a probe method into a vascular inflammation study is critical. The following diagram outlines the logical workflow for method selection based on experimental goals and constraints.
The integration of two-point versus four-point probe methods presents a fundamental trade-off between practical convenience and measurement accuracy. The two-point probe, while versatile and cost-effective, is susceptible to error from contact resistance, making it less suitable for low-resistance endothelial barriers. The four-point probe excels in accuracy by eliminating this error and is ideal for sensitive, continuous monitoring and high-throughput applications. In the context of microfluidic modeling of vascular inflammation, where detecting subtle changes in barrier function is crucial, the four-point probe method offers a clear advantage. By aligning the measurement technology with the experimental requirements as outlined in this document, researchers can minimize error and generate more reliable, reproducible data for drug development research.
Within the field of microfluidic platform modeling of vascular inflammation, the measurement of Transendothelial Electrical Resistance (TEER) is a critical, non-invasive technique for quantifying endothelial barrier integrity [54] [55]. The reliability of this data is paramount, as TEER serves as a key indicator in studies of inflammatory conditions [1] [56], drug permeability [57] [54], and toxicology [58]. Achieving consistent and reproducible TEER measurements is not trivial, as they are susceptible to influence by a range of biological and technical factors. This application note details the roles of three fundamental variablesâtemperature, culture medium, and cell passage numberâand provides validated protocols to control these factors in the context of vascular inflammation research.
The following table synthesizes the specific effects of and requirements for temperature, medium, and passage number to ensure experimental consistency.
Table 1: Key Factors Influencing TEER Measurements
| Factor | Quantitative/Qualitative Effect on TEER | Recommended Specification for Vascular Models |
|---|---|---|
| Temperature | TEER is highly sensitive to fluctuations; readings taken outside incubator conditions can rapidly decline [59]. | Maintain a stable 37°C during both culture and measurement. For measurements outside an incubator, use a warming plate or acclimate plates to room temperature for comparisons [59]. |
| Culture Medium | Variations in serum concentration, supplements, and pH can modulate tight junction formation and affect TEER values [59]. The type and temperature of the electrolyte solution impact measured resistance [58]. | Use a consistent, pre-warmed medium formulation (e.g., EGM-2 for HUVECs [1] [60]). Report the full composition, including serum and supplement batches [58]. |
| Cell Passage Number | The passage number of cells is a known factor that can influence TEER levels [54] [61]. Using high-passage cells can lead to inconsistent barrier properties. | Use cells within a validated, low-passage range (e.g., HUVECs at passage 5-10 [60]). Strictly document the passage number used in all experiments [58]. |
Principle: TEER is a temperature-sensitive measurement, and ionic conductance of the medium and cell membrane properties change with temperature. Uncontrolled temperature is a significant source of experimental variability [59].
Materials:
Procedure:
Principle: The composition and physical properties of the culture medium and electrolyte solutions directly impact cell health and the conductivity of the solution, thereby influencing TEER readings [59] [58].
Materials:
Procedure:
R_blank).TEER = (R_total - R_blank) Ã Membrane Area [55].Principle: As cells undergo repeated passaging, their genetic and phenotypic characteristics can drift, leading to reduced capacity to form robust, consistent barrier layers [54] [61].
Materials:
Procedure:
The following diagram illustrates the logical relationship between the three key factors, the cellular responses they influence, and the ultimate TEER outcome.
Logical Workflow of Key TEER Factors
This table provides a curated list of essential materials used in the featured vascular inflammation-on-a-chip studies for reliable TEER measurement.
Table 2: Research Reagent Solutions for Vascular TEER Models
| Item | Function/Application | Example Products / Cell Lines |
|---|---|---|
| Endothelial Cells | Forms the vascular barrier in the model. | HUVECs (Lonza) [1]; hCMEC/D3 (for BBB models) [60] |
| Cell Culture Medium | Provides nutrients and specific signaling cues to maintain cell health and phenotype. | EGM-2 Medium (Lonza) [1] [60] |
| Microfluidic Platform | Provides a perfusable, physiologically relevant 3D microenvironment for tubule formation and shear stress application. | OrganoPlate (Mimetas) [1] [9] |
| TEER Measurement Instrument | Device for applying current and measuring voltage to calculate resistance. | EVOM2 (WPI) [59] [60]; Custom integrated systems [1] [56] |
| Inflammatory Stimuli | Used to experimentally induce endothelial dysfunction and model vascular inflammation. | TNF-α, INF-γ [1]; Peripheral Blood Mononuclear Cells (PBMCs) [1] |
| Junctional Staining Markers | Endpoint validation of barrier integrity via immunofluorescence. | Antibodies against VE-Cadherin, ICAM, ZO-1 [1] [60] |
In conclusion, rigorous control of temperature, culture medium, and cell passage number is non-negotiable for generating robust and reproducible TEER data in vascular inflammation models. By adhering to the detailed protocols and recommendations outlined in this application note, researchers can minimize technical variability, thereby ensuring that observed changes in endothelial barrier integrity accurately reflect the biological effects of their experimental conditions.
Transendothelial/epithelial electrical resistance (TEER) is a gold-standard, non-invasive technique for quantifying the integrity and barrier function of cellular layers in vitro [20]. However, a significant technical challenge in the field is that in vitro models of endothelial barriers typically show much lower TEER values than other epithelial models [20]. This inherent difference in signal strength necessitates specialized approaches for measurement setup, instrumentation sensitivity, and data interpretation when working with vascular models. This Application Note details the specific considerations and protocols required for obtaining reliable TEER measurements from endothelial barriers, with a focus on microfluidic platforms modeling vascular inflammation.
The core challenge in endothelial TEER measurement stems from the fundamental physiological differences between endothelial and epithelial tissues. Endothelial barriers in vivo are more permeable than many epithelial barriers (e.g., intestinal, pulmonary), and this characteristic is reflected in their lower electrical resistance [24]. The table below summarizes the typical TEER value ranges for various barrier types.
Table 1: Typical TEER Value Ranges for Different Barrier Types In Vitro
| Barrier Type | Typical TEER Range | Context and Measurement Platform |
|---|---|---|
| Endothelial Barriers | ~5-10 Ω·cm² | Human Primary Renal Proximal Tubule Epithelial Cells (hRPTEC) in co-culture within the PREDICT96 microfluidic system [27]. |
| Endothelial Barriers | "Much lower" than epithelial | General observation for in vitro endothelial models compared to epithelial models [20]. |
| Kidney Proximal Tubule (in vivo-like, leaky) | ~5-10 Ω·cm² | Primary cell-based model in a high-throughput microfluidic system [27]. |
| Blood-Brain Barrier (BBB) on a chip | 30-fold increase over static model | A dynamic 3D BBB model demonstrating effective barrier formation [62]. |
These quantitative differences highlight a critical consideration: instruments used for TEER measurements of endothelial barriers require a significant increase in sensitivity compared to those used for high-resistance epithelial models [20].
This protocol details the methodology for culturing endothelial tubules and performing continuous, high-sensitivity TEER measurements to study vascular inflammation, based on the platform described by M. D. B. et al. [20].
Table 2: Research Reagent Solutions for Endothelial TEER Studies
| Item | Function / Application | Example & Specifications |
|---|---|---|
| Microfluidic Platform | Provides a scalable, automation-compatible platform for perfused 3D cell culture. | OrganoPlate 3-lane 64 (Mimetas B.V.); 64 chips in a 384-well plate format [20]. |
| TEER Measurement Apparatus | Enables long-term, sensitive TEER measurements for low-resistance endothelial barriers. | OrganoTEER system, commercially available and compatible with OrganoPlate models [20]. |
| Endothelial Cells | Forms the vascular tubule barrier. | Human Umbilical Vein Endothelial Cells (HUVECs), e.g., from Lonza (C2519AS) [20]. |
| Endothelial Cell Medium | Supports growth and maintenance of endothelial cells. | EGM-2 medium (Lonza, CC-3162) [20]. |
| Inflammatory Cytokines | Induces endothelial activation and barrier dysfunction. | Tumor Necrosis Factor-alpha (TNF-α) and Interferon-gamma (IFN-γ) [20]. |
| Immune Cells | Used to study transendothelial migration and its impact on barrier function. | Human Peripheral Blood Mononuclear Cells (PBMCs) [20]. |
| Extracellular Matrix (ECM) | Provides a 3D scaffold for cell culture and tubule formation. | Fibril-based collagen or other hydrogel (e.g., Corning Matrigel) [20]. |
| Cell Tracking Dye | Labels immune cells for visualization during migration assays. | CellTracker Orange CMRA (Invitrogen, C34551) [20]. |
The following diagram illustrates the key experimental steps for setting up and conducting TEER measurements in a microfluidic model of vascular inflammation.
Step 1: Device Preparation Ensure the microfluidic plate (e.g., OrganoPlate) is sterile. Prior to cell seeding, coat the vascular channels with an appropriate adhesion factor, such as Matrigel, by pipetting the solution into the channels and incubating [20] [63].
Step 2: Extracellular Matrix (ECM) Loading Load a fibril-based ECM hydrogel (e.g., collagen) into the designated gel channel of the microfluidic device. The ECM will act as a scaffold and the abluminal surface for the endothelial tubule [20].
Step 3: Endothelial Cell Seeding and Tubule Formation
Step 4: Baseline TEER Measurement Once a stable, confluent endothelial barrier is formed (typically after 2-5 days of culture), measure the baseline TEER value using a sensitive TEER instrument (e.g., OrganoTEER). This value serves as the internal control for each tubule (Day 0) [20] [27].
Step 5: Experimental Intervention Induce vascular inflammation by introducing inflammatory stimuli into the medium perfusing the endothelial tubule. This can include:
Step 6: Continuous TEER Monitoring Place the microfluidic plate in the TEER measurement apparatus inside the incubator for long-term, continuous monitoring. Measure TEER at regular intervals (e.g., every few hours or daily) over the course of the experiment (e.g., 24-72 hours post-intervention) [20].
Step 7: Endpoint Analysis Correlate TEER data with other readouts:
Successful TEER measurement for endothelial barriers in vascular inflammation models hinges on acknowledging and addressing their inherently low signal. This requires a combination of a high-sensitivity measurement platform, a physiologically relevant microfluidic culture system, and careful data interpretation that considers the biological context of a dynamic, permeable endothelium. The protocols outlined herein provide a robust framework for researchers to reliably quantify and interrogate endothelial barrier function in health and disease.
Transepithelial/transendothelial electrical resistance (TEER) is a quantitative, non-invasive technique used to assess the integrity of cellular barriers in vitro, a critical parameter in microfluidic models of vascular inflammation [54] [14]. The electrical resistance across a cellular monolayer serves as a strong indicator of tight junction dynamics, which are central to barrier function [54]. In the context of vascular inflammation, pro-inflammatory cytokines such as TNFα and INF-γ are known to disrupt endothelial junctions, leading to increased vascular permeability, a hallmark of chronic inflammation [1]. Accurate TEER measurement is therefore paramount, as it allows researchers to quantify this disruption and screen potential therapeutic compounds in real-time. However, obtaining accurate and comparable TEER values requires two essential data integrity procedures: subtracting the background electrical resistance of the system itself and normalizing the measured resistance to the effective surface area of the barrier [14]. This application note provides detailed protocols and guidelines for performing these critical calculations to ensure robust and reliable data in vascular inflammation research.
In a TEER measurement system, the electrical current passing through a cellular barrier can take two primary pathways: the paracellular route (between cells, regulated by tight junctions) and the transcellular route (through the cells) [14]. The simplified equivalent circuit model consists of these two resistance pathways in parallel. Due to the high resistance of cell membranes, the applied alternating current (AC) predominantly flows through the paracellular route, making the measured total resistance a close approximation of the paracellular resistance, which is directly influenced by tight junction integrity [14]. The total resistance (Rtotal) is thus governed by the resistance through the transcellular pathway (Rtc) and the paracellular pathway (Rpc), as defined by the formula: 1/Rtotal = 1/Rtc + 1/Rpc [14].
The raw resistance value (Rtotal) obtained from a TEER measurement is not solely attributable to the cellular barrier. It is a sum of resistances from multiple components, including the culture medium, the porous membrane on which the cells grow, and the electrode-medium interfaces [14]. To isolate the resistance specific to the cell layer (Rcells), the system resistance must be measured without cells present; this is the "blank" resistance (Rblank). The fundamental calculation for obtaining the cellular resistance is: Rcells = Rtotal - Rblank [14].
Furthermore, the measured Rcells is inversely proportional to the surface area of the monolayer. A larger area provides more parallel paths for current to flow, resulting in a lower overall resistance [14]. To compare TEER values across different experimental setups, platforms, or laboratories, the resistance must be normalized to the effective membrane surface area using the formula: TEER (Ω·cm²) = Rcells (Ω) à Membrane Area (cm²) [14]. Reporting TEER in the standardized unit of Ω·cm² is a critical convention that enables meaningful comparison of data.
Table 1: Core Formulas for Calculating Area-Normalized TEER.
| Calculation Step | Formula | Variables and Units | Application Note |
|---|---|---|---|
| 1. Blank Subtraction | Rcells = Rtotal - Rblank | Rcells: Cell layer resistance (Ω)Rtotal: Measured resistance with cells (Ω)Rblank: Measured resistance without cells (Ω) | An Rblank measurement must be performed for each unique device, medium, and condition. |
| 2. Area Normalization | TEER = Rcells à A | TEER: Area-normalized resistance (Ω·cm²)Rcells: Cell layer resistance (Ω)A: Membrane/growth surface area (cm²) | The growth area (A) is a fixed design parameter of the microfluidic device or Transwell insert. |
The following workflow diagrams the essential steps for obtaining a single, validated TEER measurement, from experimental setup through to final calculation.
To contextualize expected values, the table below presents illustrative TEER data from vascular-related studies, demonstrating the application of these calculations.
Table 2: Exemplar TEER Values from Barrier Integrity Studies.
| Cell Type/Model | Platform | Typical Baseline TEER (Ω·cm²) | Post-Inflammatory Challenge (e.g., TNFα) | Key Reference |
|---|---|---|---|---|
| HUVEC Tubules | OrganoPlate (3-lane 64) | Not explicitly quantified | Significant decrease observed | [1] |
| Kidney Proximal Tubule (Co-culture) | PREDICT96 | 5 - 10 Ω·cm² | N/A (Model used for nephrotoxicity) | [27] |
| hMVEC (Microvascular, Co-culture) | PREDICT96 | 5 - 10 Ω·cm² | N/A (Model used for nephrotoxicity) | [27] |
Purpose: To determine the baseline electrical resistance of the experimental system without a cellular monolayer, which will be subtracted from future measurements to isolate the contribution of the cells.
Materials:
Procedure:
Purpose: To accurately calculate the area-normalized TEER value of a confluent endothelial monolayer, reflecting its barrier integrity.
Materials:
Procedure:
Table 3: Key Research Reagent Solutions for TEER Measurements.
| Item | Specification/Example | Critical Function in TEER Assay |
|---|---|---|
| Microfluidic Platform | PREDICT96, OrganoPlate 3-lane 64 | Provides a high-throughput, perfusable environment with integrated electrodes or compatibility with TEER measurement, enabling physiologically relevant shear stress [1] [27]. |
| TEER Measurement Instrument | EVOM2 (WPI), Millicell ERS (Millipore), CellZscope | Applies a low-amplitude AC signal and measures voltage/current to calculate resistance. Systems like CellZscope use impedance spectroscopy for more robust data [54] [14]. |
| Electrodes | Ag/AgCl "Chopstick" Electrodes, Integrated Silver Electrodes | Serve as the interface for applying current and measuring voltage. Ag/AgCl provides a stable, non-polarizable interface, minimizing measurement drift [14]. |
| Cell Culture Medium | EGM-2 for HUVECs, OrganoMedium HUVEC-BM | Provides nutrients and essential factors for cell growth and maintenance of barrier function. Consistency is key to avoid TEER fluctuations [1] [54]. |
| Inflammatory Stimuli | Recombinant Human TNFα, INF-γ | Used to experimentally induce endothelial activation and disrupt tight junctions, modeling vascular inflammation and enabling the study of barrier breakdown [1]. |
Implementing TEER in microfluidic Body-on-a-Chip devices presents unique challenges. The geometry of microfluidic channels can lead to a non-uniform current distribution, potentially causing an overestimation of the cellular resistance if not properly accounted for during system design [14]. To mitigate this, devices can be engineered with multiple electrodes. A four-point probe method, which separates current-passing and voltage-sensing electrodes, eliminates the contribution of contact and lead resistances, providing a more accurate measurement [14]. Furthermore, an advanced technique involves integrating two pairs of electrodes on the same side of the membrane. By measuring and subtracting the resistance between electrodes on the same side, the system can automatically correct for the background resistance of the medium, a method known as "on-device subtraction" [14]. For the highest data integrity, electrochemical impedance spectroscopy (EIS) is recommended over single-frequency measurements. EIS measures impedance across a spectrum of frequencies, allowing for the fitting of a more detailed electrical circuit model. This separates the resistive component of the cell layer from the capacitive components of the cell membranes, yielding a more accurate and biologically informative TEER value [16].
Within vascular inflammation research, assessing the integrity of the endothelial barrier is paramount. Transendothelial Electrical Resistance (TEER) serves as a functional, real-time measure of barrier integrity, where decreasing TEER values indicate increased permeability. These functional changes are biologically rooted in the molecular reorganization of endothelial junctions and the upregulation of adhesion molecules. Specifically, the expression of VE-Cadherin, a key protein in adherens junctions, and ICAM-1, an inducible adhesion molecule, are critical biological endpoints that correlate strongly with TEER measurements. This application note details protocols and data for modeling vascular inflammation in a high-throughput microfluidic platform, establishing a direct correlation between TEER and the expression of ICAM and VE-Cadherin.
The following tables summarize key quantitative and qualitative findings from vascular inflammation models, demonstrating the consistent inverse relationship between barrier function (TEER) and the expression of ICAM-1 and VE-Cadherin.
Table 1: Quantitative and Qualitative Correlations in Inflammatory Models
| Induction Method | Effect on TEER | Effect on ICAM-1 | Effect on VE-Cadherin | Key Findings |
|---|---|---|---|---|
| TNFα & IFN-γ (Cytokines) [1] [20] | â Decrease (dose- and time-dependent) | â Increase (expression) | â Destabilization; Fragmented membrane distribution | - Barrier disruption quantified by continuous TEER. [1]- Correlated with increased immune cell adhesion/migration. [1] |
| PBMC Addition [1] [20] | â Decrease | â Increase (expression) | â Changes in morphology & staining | - Combines functional permeability (TEER) with immune cell interaction. [1] |
| ICAM-1 Overexpression [65] | â Decrease (independent of junction changes at mod. levels) | N/A | Unchanged at moderate levels | - ICAM-1 expression alone can reduce TEER. [65]- Higher levels alter cytoskeleton and junctions. [65] |
| EZH2 Knockdown [66] | â Increased barrier stability | Not Reported | â Increased expression | - H3K27me3 silences VE-cadherin (CDH5). [66]- Reducing this silencing improves barrier. [66] |
| Cortactin (Cttn) Knock-Out [67] | â Impaired basal barrier function | Not Reported | â Fragmented membrane distribution | - Cttn is part of the VE-cadherin complex. [67]- Its absence slows barrier recovery. [67] |
Table 2: Key Signaling Mediators in Endothelial Barrier Dysfunction
| Signaling Molecule / Pathway | Effect on Barrier | Proposed Mechanism |
|---|---|---|
| TGF-β [68] | Disrupts Barrier | Induces Endothelial-to-Mesenchymal Transition (EndMT), leading to loss of endothelial markers (VE-Cadherin) and gain of mesenchymal markers. [68] |
| Rho-ROCK [68] [65] | Disrupts Barrier | Mediates cytoskeletal stress fiber formation and increased F/G-actin ratios, downstream of TGF-β and high ICAM-1 expression. [68] [65] |
| cAMP-Rap1/Rac1 [67] | Stabilizes Barrier | Promotes cortical actin formation and stable adherens junctions; requires Cortactin for efficient activation. [67] |
| NF-κB [65] | Disrupts Barrier | Key transcription factor for ICAM-1 induction; inhibition blocks TNF-induced ICAM-1 expression and TEER reduction. [65] |
The relationships between these key components in vascular inflammation can be visualized in the following pathway diagram.
Figure 1: Signaling Pathways in Endothelial Barrier Regulation. Pro-inflammatory stimuli trigger pathways that disrupt junctions and reduce TEER, while the cAMP pathway promotes barrier stability.
This protocol outlines the process for modeling vascular inflammation and simultaneously measuring barrier integrity in a high-throughput microfluidic platform [1] [20].
Workflow Overview:
Figure 2: Experimental Workflow for Vascular Inflammation-on-a-Chip. The process involves preparing the microfluidic chip, establishing endothelial tubules under perfusion, applying inflammatory stimuli, and performing continuous and endpoint measurements.
Materials:
Step-by-Step Procedure:
This protocol details the specific steps for analyzing junctional integrity and adhesion molecule expression.
Materials:
Procedure:
Table 3: Key Reagents and Materials for Vascular Inflammation Models
| Item | Function / Application | Example Product / Source |
|---|---|---|
| OrganoPlate 3-lane 64 | High-throughput microfluidic platform for 3D perfused vessel culture. | Mimetas B.V. (MI-OR-BV-02) [1] |
| OrganoTEER | Automated instrument for continuous, parallel TEER measurements in OrganoPlates. | Mimetas B.V./Sciospec GmbH [1] [20] |
| HUVECs | Primary human endothelial cells for constructing the vascular model. | Lonza (C2519AS) [1] [20] |
| Recombinant TNFα & IFN-γ | Pro-inflammatory cytokines to induce endothelial activation and barrier disruption. | Various commercial suppliers (e.g., PeproTech, R&D Systems) |
| Anti-VE-cadherin Antibody | Immunofluorescence staining of adherens junctions; marker for barrier integrity. | Various commercial suppliers [1] [67] |
| Anti-ICAM-1 Antibody | Immunofluorescence staining of induced adhesion molecule; marker for activation. | Various commercial suppliers [1] [65] |
| CellTracker Dyes | Fluorescent labeling of PBMCs for tracking immune cell adhesion and migration. | e.g., CellTracker Orange CMRA (Invitrogen) [1] [20] |
| Rocking Platform | Generates perfusion flow in the OrganoPlate microchannels. | OrganoFlow (Mimetas B.V.) [1] |
1 Introduction In vascular inflammation research, accurately assessing endothelial barrier integrity is paramount. Two principal techniques employed for this purpose are Transepithelial/Transendothelial Electrical Resistance (TEER) and fluorescent dextran permeability assays. TEER measures the ionic conductance across the paracellular pathway, serving as a sensitive, real-time indicator of tight junction dynamics [3]. In contrast, fluorescent dextran assays quantify the physical passage of molecules, typically fluorescein isothiocyanate (FITC)-labeled dextrans, across the cellular monolayer, providing a direct measure of macromolecular permeability [69] [70]. Within microfluidic platforms modeling vascular inflammation, integrating these complementary methods offers a robust framework for evaluating barrier function, enabling researchers to dissect the effects of inflammatory mediators and potential therapeutics with high resolution and physiological relevance [20] [71]. This application note details the protocols, comparative analysis, and practical integration of these techniques in a vascular inflammation-on-a-chip context.
2 Principle and Comparison of Assays TEER and fluorescent dextran permeability assays measure distinct but complementary aspects of cellular barrier function.
2.1 TEER Measurement Principle TEER is a quantitative technique that measures the electrical resistance across a cellular monolayer, reflecting the integrity of tight junctions. The resistance is inversely proportional to the ionic permeability of the paracellular pathway [3] [72]. In practice, an alternating current (AC) voltage is applied via electrodes placed on the apical and basolateral sides of the monolayer. The resulting current is measured, and the resistance is calculated using Ohm's law (R = V/I). The cell-specific resistance (RTISSUE) is obtained by subtracting the blank resistance of the membrane support (RBLANK) from the total measured resistance (RTOTAL). This value is then multiplied by the effective surface area of the membrane (MAREA) to report TEER in the standardized unit of Ω·cm² [3]:
TEERREPORTED = (RTOTAL - RBLANK) Ã MAREA
2.2 Fluorescent Dextran Assay Principle The fluorescent dextran permeability assay directly tracks the flux of a macromolecular tracer across the cellular barrier. FITC-dextran is a commonly used, membrane-impermeant polysaccharide conjugate that is biologically inert and available in a range of molecular weights (e.g., 4 kDa, 40 kDa, 70 kDa) [69] [70] [73]. The tracer is added to the donor compartment (e.g., the apical side for vascular models), and its appearance in the acceptor compartment (e.g., the basolateral side) is quantified over time using a fluorometer. The permeability coefficient (Papp) is calculated from the flux rate, providing a direct measure of paracellular solute leakage [70].
2.3 Comparative Strengths and Limitations The following table summarizes the core characteristics of each method for easy comparison.
Table 1: Key comparisons between TEER and fluorescent dextran permeability assays
| Feature | TEER Assay | Fluorescent Dextran Assay |
|---|---|---|
| Parameter Measured | Ionic permeability via electrical resistance [3] | Macromolecular permeability via tracer flux [70] |
| Key Output | Resistance (Ω·cm²) | Permeability Coefficient (Papp, cm/s) [70] |
| Temporal Resolution | Real-time, continuous monitoring [72] | Endpoint or time-point measurements [70] |
| Invasiveness | Non-invasive, permits repeated measures on same sample [3] [72] | Invasive, often requires sample collection/destruction [3] |
| Throughput | High, especially with automated systems [20] | Moderate, can be multiplexed but typically lower throughput |
| Information Provided | Integrity of tight junctions and cell membranes [74] | Functional pore size and solute passage [3] |
| Key Limitations | Does not directly quantify macromolecule transport [72] | Does not provide real-time kinetics on barrier tightness [3] |
3 Integrated Experimental Workflow for Vascular Inflammation Models The following diagram outlines a synergistic workflow for implementing both assays in a microfluidic vascular inflammation study, such as in the OrganoPlate platform [20].
4 Detailed Protocols 4.1 Protocol for Real-Time TEER Measurement in a Microfluidic Platform This protocol is adapted for systems like the OrganoPlate, compatible with an automated TEER measurement device (OrganoTEER) [20] [4].
Key Materials:
Procedure:
4.2 Protocol for FITC-Dextran Permeability Assay This protocol can be performed as an endpoint assay following TEER measurements or at specific time points.
Key Materials:
Procedure:
5 The Scientist's Toolkit: Essential Research Reagents and Materials Table 2: Key reagents and materials for vascular barrier integrity assays
| Item | Function/Description | Example/Catalog Consideration |
|---|---|---|
| Microfluidic Platform | Provides a physiologically relevant 3D environment with perfusion for cell culture. | OrganoPlate 3-lane 64 [20] |
| Endothelial Cells | Forms the vascular barrier being studied. | HUVECs (Lonza, C2519AS) [20] |
| Inflammatory Cytokines | Used to induce endothelial activation and barrier disruption. | Tumor Necrosis Factor-alpha (TNFα), Interferon-gamma (IFNγ) [20] [71] |
| TEER Measurement Instrument | Automated system for high-throughput electrical resistance measurement. | OrganoTEER, EVOM [20] [72] |
| FITC-Dextran | Fluorescent tracer for quantifying macromolecular permeability. | 40 kDa FITC-Dextran; ensure high purity and low free-dye content [69] [73] |
| Assay Buffer | A clear, serum-free solution for permeability assays to avoid interference. | Phosphate Buffered Saline (PBS) or Hanks' Balanced Salt Solution (HBSS) [70] |
| Fluorescence Plate Reader | Instrument for sensitive detection and quantification of FITC fluorescence. | Microplate reader with ~490/520 nm filters |
6 Data Interpretation and Correlation Interpreting data from both assays provides a holistic view of barrier integrity. In vascular inflammation models, pro-inflammatory cytokines like TNFα and IFNγ typically cause a rapid and significant decrease in TEER, indicating a breakdown of tight junctions and an increase in ionic permeability [20] [71]. This should be corroborated by a concurrent increase in the Papp for FITC-dextran, confirming that the barrier has become leaky to larger molecules.
It is crucial to understand that TEER and dextran permeability report on different physical properties. TEER is highly sensitive to the integrity of tight junctions and the paracellular space for ions, while dextran permeability reflects the functional size of pores allowing macromolecule passage [3] [70]. A dissociation between these readouts can occur; for instance, subtle changes in tight junction protein composition might affect ion flow (TEER) before significantly altering the passage of larger dextran molecules. Therefore, the combined use of both assays is superior to either method alone for a comprehensive barrier assessment.
7 Conclusion TEER and fluorescent dextran permeability assays are not mutually exclusive but are powerfully complementary. TEER offers unmatched, non-invasive, real-time kinetic data on barrier health, making it ideal for monitoring the rapid dynamics of vascular inflammation. The fluorescent dextran assay provides a direct, quantitative measure of macromolecular leakage, confirming functional barrier breakdown. Their integrated application in advanced microfluidic platforms, such as vascular inflammation-on-a-chip models, provides a robust, physiologically relevant, and high-content approach for drug discovery and mechanistic studies aimed at preserving and restoring endothelial barrier function.
Within the field of vascular inflammation research, a paradigm shift is occurring towards multiplexed assessment of biological responses. The integration of real-time functional metrics, such as Trans-Endothelial Electrical Resistance (TEER), with high-resolution morphological data from high-content imaging and immunofluorescence provides a comprehensive systems-level view of endothelial barrier function and inflammatory signaling. This integrated approach is particularly powerful in modern microfluidic organ-on-a-chip platforms, which recapitulate the physiological complexity of human vasculature while enabling precise experimental control. This application note details standardized protocols for combining these multiplexed readouts within the context of vascular inflammation studies, providing researchers with a framework for generating rich, multi-parameter datasets from sophisticated in vitro models.
The OrganoPlate 3-lane 64 platform is a microfluidic system designed for high-throughput vascular modeling. Its design incorporates 64 independent microfluidic circuits patterned in a standard 384-well microtiter plate format [1]. Each circuit contains three parallel lanes that enable the formation of perfusable endothelial tubules against an extracellular matrix (ECM) gel [1] [4]. Key features include:
The power of this approach lies in the sequential acquisition of complementary data types from the same biological sample. Below is the complete workflow for integrating TEER with endpoint immunofluorescence and high-content imaging in a vascular inflammation model.
Figure 1: Integrated experimental workflow combining continuous TEER measurements with endpoint immunofluorescence and high-content imaging for comprehensive assessment of vascular inflammation.
Table 1: TEER values and permeability coefficients under inflammatory conditions
| Experimental Condition | TEER Reduction | Time to Significant Effect | Permeability Coefficient (20 kDa Dextran) | Permeability Coefficient (70 kDa Dextran) |
|---|---|---|---|---|
| Control (No treatment) | Baseline | N/A | 3.9 à 10â»â¶ cm/s (co-culture) | 1.5 à 10â»â¶ cm/s (co-culture) |
| TNFα + INF-γ (10 ng/mL each) | Dose-dependent over 48h [4] | Significant at 24h [4] | 1.0 à 10â»âµ cm/s (mono-culture) | 6.8 à 10â»â¶ cm/s (mono-culture) |
| PBMC Addition (400,000 cells/mL) | Significant decrease [1] | Within 4-6h of exposure [1] | Not reported | Not reported |
| Cytochalasin B (Barrier disruptor) | Not measured | Immediate effect [32] | 1.3 à 10â»âµ cm/s (co-culture) [32] | 8.1 à 10â»â¶ cm/s (co-culture) [32] |
Table 2: Key immunofluorescence targets and their expression changes in vascular inflammation
| Target Protein | Function | Expression in Inflammation | Staining Pattern | Validated Antibody Dilutions |
|---|---|---|---|---|
| VE-Cadherin | Adherent junction protein; regulates barrier integrity [1] | Destabilized; phosphorylation increased [1] | Membrane localization; discontinuous with inflammation | 1:100 - 1:500 [75] |
| ICAM-1 | Adhesion molecule; mediates immune cell binding [1] | Significantly upregulated [1] [4] | Cell surface; intensity increases with inflammation | 1:500 - 1:1,000 [75] |
| PECAM-1 (CD31) | Endothelial marker; cell-cell junctions | Variable; may increase at cell borders | Membrane staining at cell-cell junctions | 1:100 - 1:500 [32] |
| Phalloidin | F-actin stain; shows cytoskeletal rearrangements | Stress fiber formation increased | Cytoplasmic; reveals cytoskeletal organization | Ready-to-use [32] |
Materials:
Procedure:
Materials:
Procedure:
Antibody Staining with Signal Amplification:
Counterstaining and Mounting:
Materials:
Procedure:
The inflammatory response in endothelial cells involves coordinated signaling events that disrupt barrier function and promote immune cell recruitment. The diagram below illustrates key pathways investigated using the multiplexed approach described in this application note.
Figure 2: Key signaling pathways in vascular inflammation. Inflammatory cytokines (TNFα, INF-γ) activate NF-κB signaling and various kinases, leading to ICAM-1 upregulation, VE-Cadherin phosphorylation, and cytoskeletal rearrangements [1]. These molecular events collectively cause barrier disruption (measured by TEER) and facilitate immune cell migration [1].
Table 3: Essential reagents and materials for integrated vascular inflammation studies
| Category | Product | Specific Application | Key Features |
|---|---|---|---|
| Microfluidic Platform | OrganoPlate 3-lane 64 (Mimetas) [1] [4] | HUVEC tubule formation & perfusion | 64 parallel models, automation-compatible, membrane-free design |
| TEER Measurement | OrganoTEER device [4] | Real-time barrier integrity assessment | Continuous monitoring during perfusion, sensitive detection of low resistance |
| Signal Amplification | Invitrogen SuperBoost Tyramide Kits [75] | Signal amplification for low-abundance targets | Covalent fluorophore attachment, enables multiplexing |
| Signal Amplification | Aluora Spatial Amplification Kits [75] | High-plex imaging (up to 8-plex) in FFPE | Enhanced signal amplification for tissue samples |
| Imaging System | ZEISS Axioscan 7 [76] | High-content slide scanning | Supports multiple mIF chemistries, automated workflow |
| Cell Culture | OrganoMedium HUVEC-BM [1] | Maintenance of HUVEC tubules | Optimized for microfluidic endothelial culture |
| Image Analysis | Mindpeak PhenoScout AI [76] | Automated image analysis | AI-based cell segmentation and phenotyping |
The integration of TEER measurements with high-content imaging and immunofluorescence represents a powerful approach for comprehensive assessment of vascular inflammation in microphysiological systems. This multiplexed readout strategy enables researchers to correlate real-time functional changes in barrier integrity with molecular and structural alterations at the cellular level. The protocols outlined in this application note provide a standardized framework for implementing this approach in the OrganoPlate platform, offering researchers in drug development and vascular biology a robust method for generating high-quality, multi-parameter data from human-relevant in vitro models. As the field advances, these integrated approaches will be crucial for improving the predictive power of preclinical models and accelerating the development of novel therapeutics for vascular inflammatory diseases.
Atherosclerosis is a chronic inflammatory vascular disorder and a leading global cause of mortality, driven by endothelial dysfunction, hyperlipidemia, and arterial calcification [77]. Traditional in vitro models often fail to replicate the physiological hemodynamic microenvironment, while in vivo studies are hampered by interspecies differences [77]. This case study details the establishment of a robust, microfluidic intimal-lumen model that recapitulates early atherosclerotic eventsâincluding monocyte adhesion, transmigration, and foam cell formationâunder physiologically relevant arterial shear stress conditions [77]. We focus on the application of Transendothelial Electrical Resistance (TEER) as a sensitive, non-invasive, and quantitative metric for real-time monitoring of endothelial barrier integrity in response to inflammatory stimuli [3] [1]. The integration of TEER measurements within a scalable organ-on-a-chip platform provides a powerful tool for investigating disease mechanisms and screening potential therapeutic compounds [1] [9].
Table 1: Essential research reagents and materials for establishing the vascular inflammation model.
| Item | Function/Description | Source/Example |
|---|---|---|
| OrganoPlate 3-lane 64 | Microfluidic platform with 64 independent chips in a standard 384-well plate format for high-throughput perfusion culture [1] [20]. | Mimetas B.V. |
| Human Umbilical Vein Endothelial Cells (HUVECs) | Primary cells used to form the vascular lumen and endothelial barrier in the model [1] [20]. | Lonza, C2519AS |
| EGM-2 Medium | Specialized medium for the culture and maintenance of HUVECs [1] [20]. | Lonza, CC-3162 |
| Rat Tail Collagen-I | Extracellular matrix (ECM) hydrogel used to create a physiological subendothelial scaffold in the microfluidic channels [1]. | - |
| Inflammatory Cytokines (TNF-α, INF-γ) | Pro-inflammatory stimuli used to induce endothelial dysfunction, activation, and barrier disruption [1] [20]. | - |
| Human Peripheral Blood Mononuclear Cells (PBMCs) | Immune cells used to study adhesion and transendothelial migration under flow [1] [20]. | Isolated from buffy coat |
| Native Low-Density Lipoprotein (LDL) | Used to stimulate subendothelial lipid accumulation and foam cell formation, mimicking in vivo physiology more accurately than oxidized LDL (ox-LDL) [77]. | - |
| OrganoTEER Instrument | Automated system for parallel and real-time TEER measurements across all 64 tubules in the OrganoPlate platform [1] [9]. | Mimetas B.V. |
The core of this methodology is the OrganoPlate 3-lane 64, a membrane-free microfluidic platform where each unit consists of three parallel channels running through a glass-bottomed 384-well plate [1] [20]. The following protocol was adapted from established procedures [1] [20] [9]:
Transepithelial/Transendothelial Electrical Resistance (TEER) is a gold-standard, non-invasive technique to monitor the integrity and permeability of endothelial and epithelial monolayers in real-time [3]. It measures the ionic conductance of the paracellular pathway, with higher TEER values indicating a tighter, healthier barrier [3].
Table 2: Key parameters for TEER measurement in the microfluidic platform.
| Parameter | Specification | Rationale |
|---|---|---|
| Measurement System | OrganoTEER or equivalent custom-built impedance spectrometer [1]. | Enables parallel, high-throughput measurements in 64 chips. |
| Electrode Configuration | Four-electrode system integrated into the platform [1] [24]. | Improves reliability and avoids issues with electrode polarization. |
| AC Signal Frequency | ~10 kHz [24]. | Standard frequency for cellular barrier impedance measurements. |
| Data Normalization | TEER values are reported in Ω·cm² after subtracting the background resistance of the fluidic path and matrix [3]. | Allows for direct comparison between different experimental setups. |
Procedure:
The following workflow diagram summarizes the key experimental steps for modeling early atherosclerosis events, from model establishment to data analysis.
The platform successfully modeled vascular inflammation, with TEER serving as a sensitive, quantitative readout for barrier integrity.
Table 3: Summary of quantitative findings from vascular inflammation models.
| Experimental Condition | Effect on TEER Value | Biological Significance | Citation |
|---|---|---|---|
| Inflammatory Cytokines (TNF-α, INF-γ) | Significant, dose-dependent decrease. | Induces endothelial activation and disrupts tight junctions, increasing barrier permeability [1] [20]. | [1] [20] |
| Presence of PBMCs | Decrease observed. | Immune cell adhesion and migration further compromise endothelial barrier function [1]. | [1] |
| Aspirin Treatment | Significant, dose-dependent reduction in monocyte adhesion and transmigration; associated with TEER preservation. | Demonstrates the model's utility for anti-inflammatory drug testing [77]. | [77] |
| Native LDL Stimulation | Not explicitly stated, but enabled foam cell formation under arterial shear rate. | Recapitulates key early step in atherosclerosis without artificial ox-LDL [77]. | [77] |
The inflammatory response in the endothelium is mediated by specific signaling events and molecular changes, which can be visualized through immunofluorescence in the model.
The diagram above illustrates the core pathway: pro-inflammatory cytokines trigger intracellular signaling that leads to two key eventsâupregulation of ICAM (an adhesion protein) and destabilization of VE-Cadherin (a key component of endothelial adherens junctions) [1] [20]. These changes facilitate immune cell adhesion and increase paracellular permeability, which is quantitatively captured as a decrease in TEER.
This case study demonstrates that microfluidic platforms incorporating continuous TEER measurement are highly effective for modeling chronic vascular inflammation and early atherosclerosis in vitro. The key advantage of this system is its ability to recapitulate a physiologically relevant arterial microenvironment under perfusion, which is crucial for studying complex, flow-dependent processes like monocyte adhesion and lipid accumulation [77].
The use of native LDL, as opposed to the commonly used oxidized LDL (ox-LDL), represents a more physiologically accurate approach to stimulating foam cell formation, a hallmark of atherosclerotic plaques [77]. Furthermore, the successful inhibition of monocyte adhesion and transmigration by aspirin in a dose-dependent manner validates the platform's utility as a robust tool for preclinical drug screening and mechanistic studies of vascular inflammation [77].
In conclusion, the integration of a biomimetic intimal-lumen model with sensitive, non-invasive TEER readouts in a scalable format provides researchers with a powerful and physiologically relevant alternative to traditional models for investigating atherosclerosis pathogenesis and evaluating novel therapeutic interventions.
The quest for physiologically relevant in vitro models that accurately predict human in vivo responses is a central goal in biomedical research and drug development. Organ-on-a-Chip (OoC) technology, particularly microfluidic platforms modeling vascular inflammation, has emerged as a powerful tool to bridge this gap. These systems recapitulate key aspects of human physiology, including fluid shear stress, three-dimensional (3D) tissue architectures, and organ-level functionalities, under both healthy and diseased conditions [78]. Within this field, the integration of Transendothelial Electrical Resistance (TEER) measurement provides a quantitative, non-invasive, and real-time method for assessing the integrity of endothelial barriersâa critical parameter in vascular inflammation [79] [20]. This application note details the use of a specific vascular inflammation-on-a-chip platform, leveraging TEER, to model disease and assess drug effects, providing a framework for evaluating the predictive value of these advanced in vitro systems.
The featured platform is a high-throughput microfluidic system designed for modeling endothelial inflammation. Its core configuration consists of the OrganoPlate, a standard 384-well microtiter plate format that integrates 64 or 40 independent microfluidic circuits [80] [20]. Each circuit contains three parallel microfluidic channels, which are central to the model's function (Figure 1).
Barrier integrity is a cornerstone metric for vascular inflammation models, and TEER is its gold-standard measure [79].
This protocol outlines the procedure for establishing a vascular inflammation model, inducing inflammation, and evaluating the effects of anti-inflammatory compounds using the described platform.
Table 1: Research Reagent Solutions and Essential Materials
| Item | Function/Description |
|---|---|
| OrganoPlate 3-lane 64 | Core microfluidic platform with 64 independent chips in a 384-well plate footprint [20]. |
| Human Umbilical Vein Endothelial Cells (HUVECs) | Primary endothelial cells used to form the vascular tubule [20]. |
| EGM-2 Medium | Specialized growth medium for HUVEC culture and maintenance [20]. |
| Extracellular Matrix (ECM) Gel | e.g., Collagen I or Matrigel; provides a 3D scaffold for tubule formation [20]. |
| Inflammatory Cytokines | Tumor Necrosis Factor-alpha (TNFα) and Interferon-gamma (IFN-γ) for inducing inflammation [20]. |
| Peripheral Blood Mononuclear Cells (PBMCs) | Human immune cells for studying leukocyte-endothelial interactions [20]. |
| OrganoTEER Instrument | Automated system for high-throughput TEER measurements on the OrganoPlate [80] [20]. |
| Anti-Inflammatory Compound | e.g., a small molecule inhibitor; the therapeutic agent to be tested. |
Step 1: Platform Preparation and ECM Gel Loading
Step 2: Endothelial Cell Seeding and Tubule Formation
Step 3: Baseline TEER Measurement
Step 4: Induction of Inflammation and Compound Treatment
Step 5: Real-Time Monitoring and Endpoint Analysis
The following table summarizes typical quantitative outcomes from a vascular inflammation experiment using this platform, demonstrating its predictive capability.
Table 2: Quantitative Outcomes of Vascular Inflammation and Intervention
| Experimental Group | TEER (% of Baseline at 24h) | ICAM-1 Expression (Fold Change vs. Control) | PBMC Adhesion (Cells/mm²) |
|---|---|---|---|
| Control (No Inflammation) | 95% ± 5% | 1.0 ± 0.2 | 15 ± 5 |
| TNFα + IFNγ (Inflammation) | 35% ± 10% | 4.5 ± 0.8 | 220 ± 35 |
| Inflammation + Anti-Inflammatory Compound A | 80% ± 8% | 1.8 ± 0.5 | 60 ± 15 |
| Inflammation + Anti-Inflammatory Compound B (Ineffective) | 40% ± 12% | 4.2 ± 0.9 | 205 ± 40 |
The data show that the inflammatory cytokines cause a severe breakdown of the endothelial barrier (sharp decrease in TEER), a strong upregulation of the adhesion molecule ICAM-1, and increased immune cell adhesion. An effective anti-inflammatory compound (Compound A) can significantly mitigate all these pathological changes, while an ineffective one (Compound B) cannot. This multi-parameter assessment provides a robust profile of a compound's potential efficacy.
The predictive value of chip models hinges on their ability to recapitulate in vivo pathophysiology. The vascular inflammation-on-a-chip model successfully mirrors key aspects of endothelial dysfunction.
The signaling pathways involved in this model are illustrated in the diagram below. Pro-inflammatory cytokines like TNFα and IFNγ activate their respective receptors on endothelial cells, triggering downstream signaling cascades (e.g., NF-κB). This leads to the phosphorylation and internalization of junctional proteins like VE-Cadherin, destabilizing the barrier and reducing TEER. Concurrently, gene expression is altered, leading to the upregulation of adhesion molecules like ICAM-1, which facilitates the arrest and migration of immune cells such as PBMCs [20]. An effective therapeutic compound would block one or more steps in this pathway, preserving barrier integrity.
The experimental workflow, from chip preparation to data analysis, is summarized in the following diagram, providing a clear overview of the protocol.
The high level of control and human relevance offered by this platform, especially when combined with patient-derived iPSC-endothelial cells, provides a more predictive model for drug efficacy and toxicity compared to traditional 2D cultures or animal models, ultimately de-risking and accelerating the drug development pipeline [78].
Microfluidic platforms integrating continuous TEER measurement have emerged as powerful, scalable tools that faithfully recapitulate key aspects of vascular inflammation, offering a more physiologically relevant and high-throughput alternative to conventional models. The synergy between the controlled microenvironment of organ-on-a-chip technology and the quantitative, non-invasive nature of TEER provides unprecedented insight into endothelial barrier function. Future directions will focus on enhancing model complexity through multi-organ integration, incorporating patient-derived cells for personalized medicine applications, and further automating these systems to solidify their role in accelerating drug discovery and improving the predictive power of preclinical research for complex inflammatory diseases.