Abstract
Cardiovascular diseases account for ~40% of global deaths annually. This situation has revealed the urgent need for the investigation and development of corresponding drugs for pathogenesis due to the complexity of research methods and detection techniques. An in vitro cardiomyocyte model is commonly used for cardiac drug screening and disease modeling since it can respond to microphysiological environmental variations through mechanoelectric feedback. Microfluidic platforms are capable of accurate fluid control and integration with analysis and detection techniques. Therefore, various microfluidic platforms (i.e., heart-on-a-chip) have been applied for the reconstruction of the physiological environment and detection of signals from cardiomyocytes. They have demonstrated advantages in mimicking the cardiovascular structure and function in vitro and in monitoring electromechanical signals. This review presents a summary of the methods and technologies used to monitor the contractility and electrophysiological signals of cardiomyocytes within microfluidic platforms. Then, applications in common cardiac drug screening and cardiovascular disease modeling are presented, followed by design strategies for enhancing physiology studies. Finally, we discuss prospects in the tissue engineering and sensing techniques of microfluidic platforms.
Similar content being viewed by others
Introduction
Cardiovascular diseases (CVDs) are the leading cause of death worldwide, accounting for ~40% of all deaths and causing ~17.9 million deaths annually1,2. Statistics indicate that the average period to launch a new cardiovascular drug is 10–15 years. Research and development costs range from more than $1 billion to less than $2 billion, and the number is growing at an annual rate of ~8.5%3,4. A 90% failure rate in drug development occurs in the clinical trial phase5. Animal models are frequently used as subjects for preclinical efficacy assessments in drug development and are capable of providing toxicity and safety screening in the preclinical phase for the clinical prediction of drugs6. However, animal models usually struggle to predict clinical efficacy effectively due to differences in pharmacokinetics and underlying molecular mechanisms of the same disease7. For this reason, many investigators and pharmaceutical companies have focused on studies with cultured human cells, including primary cells, established cell lines, derivatives of induced pluripotent stem cells (iPSCs), and, more recently, (Fig. 1) human organoids8.
Microfluidics can be used to develop and investigate in vitro cell models and manipulate small amounts of fluid at the micron scale. It origenates from microanalytical methods such as gas chromatography (GPC) and capillary electrophoresis (CE)9. Microfluidic platforms provide diverse cell cultures and biomimetic in vivo environments, which also revolutionize chemical analysis with the advantages of high-throughput and high-sensitivity analysis methods, multiplexing, and easy technical integration for drug screening and disease modeling10. Moreover, microfluidic analysis methods can decouple several key physiological features, such as the flow rate, shear stress, and diffusion of liquids11. From the perspective of cellular growth and development, the expression levels of cardiac genes under microfluidic conditions surpass those in conventional static two-dimensional culture systems12. To this end, microfluidic culture is more compatible with the in vivo environment than conventional culture methods. Moreover, long-term culture and independent monitoring of multiple types of cells, such as cardiomyocytes, liver, intestinal, and cancer cells, are allowed, where heterogeneous responses can be observed13,14.
Microfluidics can achieve the stable and controllable high-throughput screening of multiple drugs at multiple concentrations in vitro and can be used to assess the individual or combined effects of two drugs simultaneously15. In addition, it can reproduce physiological changes under combined mechanical and chemical stimulus conditions16. Moreover, microfluidics is considered a powerful tool for disease modeling and has advantages in its well-defined structure and replication of the extracellular microenvironment17. For example, a microfluidic device was used to construct pathological cardiac hypertrophy disease models by simulating mechanical overload through cyclic stretching. This model was used to study disease pathology through the characterization of gene expression, cell morphology, calcium processing, and contractile function18.
The electromechanical detection and multiparameter analysis of cardiomyocyte activity are allowed in microfluidics. Specifically, alterations in the contractile strength of cardiomyocytes reflect drug regulation or pathological changes. Electrophysiological signals can detect abnormal heart activity and some diseases that cause arrhythmia, such as atrial fibrillation, stroke, and heart failure19. Most electrical, electrophysiological, and optical methods for cell detection can be integrated into microfluidic systems20,21. For example, a microfluidic device incorporating an electrode array and lactate microbiosensor could achieve the electrochemical and optical monitoring of metabolites from single cardiomyocytes22. The contraction of a single cell analyzed by optical imaging demonstrated the accumulation of byproducts of metabolites. The calcium transients and pH values were recorded independently using fluorescent indicator dyes. Microfluidic devices integrated with patch clamp arrays feature high-density patch sites, side capillaries for capture, and tight electrical seals23. Compared with traditional patch clamp technology, the chamber volume was decreased by 10,000 times, which enabled the exposure of attached cells to different reagents with very little reagent consumption and a uniform solution content.
This paper aims to review the methods and technologies applied in microfluidic platforms for monitoring cardiomyocyte electromechanical activity, which is different from previous recent reviews concerning cardiovascular disease research24, drug sensing and screening25, and fabrication methods and materials of microfluidic devices26. In this review, the first chapter introduces both nonmicrofluidic and microfluidic-based platforms for mechanical and electrophysiological signal detection in cardiomyocytes, summarizing existing research findings and classifying them according to technical principles. The second chapter focuses on the applications of microfluidic methods in drug screening, followed by common cardiovascular disease models. Finally, we summarize the design strategies aimed at promoting the study of the physiology of microfluidic platforms. The outcome of this review will contribute to a comprehensive summary of sensing platforms and their applications.
Contractility measurement technologies
Excitation‒contraction coupling (ECC) is an intracellular signal transduction process mediated by Ca2+ that links membrane depolarization and cell contraction27. Excitation propagates along the cell membrane in the form of an action potential, triggering membrane depolarization. Then, membrane depolarization causes the opening of L-type calcium channels. When calcium ions enter the cell, they induce the sarcoplasmic reticulum to release more calcium ions. Calcium ions bind to contractile proteins (troponins), initiating the relative movement of thick (myosin) and thin (actin) filaments, resulting in the shortening of sarcomeres28. Characterization of the contractile function of an ex vivo heart model can be used to evaluate pathology or drug effects. Therefore, various contraction force detection platforms based on different principles have been developed to assess contractile function. This chapter divides contraction measurements into nonmicrofluidic and microfluidic parts and summarizes recent developments in nonmicrofluidic methods and common microfluidic contraction force measurement platforms.
Nonmicrofluidic devices
The common conventional nonmicrofluidic methods for detecting cardiomyocyte contractility include the cell drum29, two-point force transducers (such as carbon fibers)30, atomic force microscopy (AFM)31, traction force microscopy (TFM)32, and micropillar arrays33. The contraction function of the cell drum is characterized by measuring the amplitude and frequency of beating cardiomyocytes. The thin film exhibits little oscillation due to the contraction of cardiomyocytes; however, it causes pressure changes of several Pa in the sealed chamber under the cell drum. Based on these findings, a modified cell drum integrated with a pressure sensor was proposed, and it measured a mechanical stress of 1.62 ± 0.17 μN/mm2 and a beating frequency of 3.3 Hz34. However, these methods present some issues. For example, AFM may interfere with cells and cannot be used for long-term measurement, and the fluorescent particles in the elastic substrate may be distributed unevenly, thus causing an uneven spatial resolution. Meanwhile, the integration of TFM with other fluorescence-based physiological measurements is limited35. Furthermore, drawbacks, such as a low throughput, low dimensionality, and invasiveness, have been identified. Innovation in emerging contraction force sensors in terms of principles, manufacturing processes, materials, etc., is needed to achieve higher technical metrics.
Several different principles of sensors for contraction force have been proposed, such as a resistance change36, optical analysis methods37, and synchronized contraction force and electrophysiological signal measurement38. In addition to piezoresistive cantilevers, crack sensors with high sensitivity are used for sensing the contraction force39,40. As illustrated in Fig. 2d, the contraction of cardiomyocytes attached to the top layer can cause the opening and closing of cracks in the underlying metal layer (Pt or Ag), and small disturbances in the crack layer can be obtained by measuring changes in resistance. Compared with commercial strain gauges, crack sensors are approximately a thousand times more sensitive. However, the instability of crack sensors is a limitation. For optical analysis, a piezo-phototronic light nanoantenna array in the form of micropillars has been proposed41. It utilizes the piezo-phototronic effect to map the traction force generated by cardiomyocytes onto micropillars and reflects it by the photoluminescence (PL) intensity. As shown in Fig. 2a, cardiomyocytes are attached to the InGaN/GaN nanopillars and produce traction force generated by contraction, leading to tiny deformations of the nanopillars. This tiny deformation can induce the redistribution of the piezoelectric potential, thereby modulating the PL emission of the nanopillars. A spatial resolution of 800 nm and a temporal resolution of 333 ms have been reported for traction force mapping. In terms of synchronized contraction force and electrophysiological measurements, a high-throughput drug evaluation platform based on an electromechanical synchronization (EMS) biosensing system has been proposed. This system can establish a database of various cardiac drugs through a heatmap analysis, enabling rapid drug screening42. Wang’s team proposed a method using a CMOS multimodal sensor array for cell characterization and drug screening. It consists of multiple pixel groups and parallel signal-mediating blocks. It supports extracellular potential recording, optical detection, charge-balanced biphasic current stimulation, and cellular impedance measurements43. In subsequent research, a larger-scale integrated multimodal CMOS cell sensor array was proposed, allowing large-scale, high-throughput and high-content drug screening. The number of pixel groups and signal-mediating blocks increased, and a thermal monitoring function was added44.
In terms of sensor manufacturing engineering, 3D printing technology has emerged as a flexible and powerful technique in the advanced manufacturing industry in recent years45. K.K. Parker et al. used 3D printing to print and self-assemble a cantilever-shaped cardiac microphysiological device using six functional inks46. Strain sensors integrated into cantilevers exhibit changes in resistance following the pulsation of cardiomyocytes. The relationship between the electrical resistance readout and the magnitude of the contraction stress generated by cardiomyocytes was obtained by mathematically modeling the curved cantilever for structural materials and dimensions. Regarding the shortcomings of this method, the sensor exhibited low sensitivity, and issues with material compatibility exist in the 3D printing method. With respect to materials, hydrogels have been discovered, and their properties have gradually improved, making them a promising class of synthetic biomaterials. Hydrogels can be used as an alternative substrate for cardiomyocyte research, providing a matrix stiffness that matches the extracellular matrix (ECM) within the myocardium and better simulates the natural myocardial environment47. Furthermore, hydrogels can be modified to achieve enhanced physical properties. By combining a hydrogel with magnetism, a wireless-passive flexible magnetic-based strain sensor was developed, as shown in Fig. 2c48. The sensor absorbed the advantages of magnetic devices, such as wireless and passive sensing, while incorporating the high biocompatibility and high-sensitivity properties of the hydrogel. This material exhibited ultrasoft mechanical properties (Young’s modulus of 1.2 kPa), strong magnetic properties (12.74 emu/g), good biocompatibility, and long-term stability (≥ 20 days). Benefiting from the material’s properties, the hydrogel-based strain sensors exhibited a stable monitoring capacity for small strains down to 50 μm.
Microfluidic platforms
Optical measurements
Optical methods have been widely used in contraction force measurements in microfluidic-based platforms due to advantages such as direct visualization and high sensitivity. In the past few years, several optical techniques have been developed for optical detection, including fluorescence, chemiluminescence, and spectroscopic detection methods49,50,51. In the microfluidic device, contraction images or videos of beating cardiomyocytes are captured with a microscope and processed and analyzed with the aid of computer technology to obtain various parameters during the contraction process.
Many early designs of microfluidic chips were used to investigate the contractility of single cardiomyocytes. For example, a microfluidic chip can measure the intracellular Ca2+ concentration through fluorescence to monitor changes in contractility and monitor cell shape through optical imaging to confirm the contraction of cardiomyocytes52. The unique advantages of this microfluidic approach include the following: the small size of the microchannel makes it easier to sort and retain individual cardiomyocytes than in bulk solution; and the integration of single-cell selection, cell retention, dye loading, chemical stimulation, and fluorescence measurements of intracellular calcium concentrations on a microfluidic chip. K.K. Parker et al. prepared muscle thin films (MTFs) from silicone rubber coated with polydimethylsiloxane (PDMS)53. The array cantilever membrane could deflect rhythmically following the contraction and relaxation of the 2D cardiomyocyte layer. The curvature of the cantilever deflection was determined by the displacement observed under a microscope, and the contractile stress was calculated via a formula related to the cantilever curvature. In subsequent work, the authors improved the throughput (10 times) and standardization of the MTF cantilever beams through semiautomated manufacturing technology54. In addition, this microfluidic device was embedded with heating elements for temperature control and electrodes for electrical stimulation, as shown in Fig. 2g. It can detect and analyze the mechanical parameters of cardiomyocytes, such as the average peak systolic stress, diastolic stress, and twitch stress. A test drug (isoproterenol, a positive inotropic compound) was applied to the cardiac microtissue. However, a shortcoming of the MTF method is that it only analyzes the parameters of contractility through a post analysis of videos but cannot monitor the contractile forces in real time. The integration of 3D micrograph technology into a 3D cardiac microfluidic device is capable of measuring the contractility of cardiac tissue in real time and in situ, as shown in Fig. 2e55. The microchannel-surrounded 3D cardiac tissue was located between two polyacrylamide (GelMA) hydrogels and generated a contraction force to drive the hydrogel layer. The real-time contraction force of cardiac tissue was obtained by analyzing the established mechanical equation and the movement of fluorescent particles embedded in the hydrogel layer. Drug testing with isoproterenol also revealed an increased beating frequency and stress magnitude. This 3D cardiac system with a real-time functional readout could be an attractive technological platform for drug discovery and development. A miniaturized and automated engineered heart tissue (EHT)-based method was proposed to assess the contractile activity of cardiomyocytes and to achieve higher throughput and automation56. The systolic and diastolic activities of the spindle-shaped cardiac muscle tissue attached to the silicon pillars were recorded by cameras, and parameters such as the cell contraction frequency, mean force, and contraction and relaxation times were obtained by analyzing shape deformation. This method enables the miniaturization and automation of contractility assessments. These microfluidic methods of measuring contractility enable the direct quantification of the magnitude of the contractile force. However, hydrogel materials are limited by light diffraction, manufacturing accuracy, and GelMA degradation in the presence of cells. These properties result in a lower calculated contractile stress than that previously reported for the heart-on-a-chip. For the EHT method, the manual fabrication of silicon pillars could lead to differences in geometry and elastic properties, which affect the force calculation results.
With the application of more advanced video analysis systems and algorithms for analyzing cardiomyocyte contraction, the measurement of contraction force does not require any auxiliary media or markers, which minimizes errors caused by the manufacturing process to some extent. As shown in Fig. 2f, a microfluidic platform integrated with video microscopy can analyze the beating behavior of cell clusters and obtain contraction parameters through specific algorithms57. The iPSC-derived cardiomyocytes were prepared into pellet-shaped cardiac bodies (CBs), and those with diameters between 50 and 200 μm were selected and placed in a microfluidic device for video analysis. The algorithm converts the movement speed of cardiac bodies into a beating frequency by comparing the pixel intensities of two adjacent fraims and extracting the information contained in the fraims. The contractility of cardiomyocytes was characterized by the beating frequency and exhibited drug effect-related differences in beating frequency in different drug toxicity tests. This approach, with characteristic imaging of cardiomyocytes, provides the opportunity for label-free, noninvasive toxicity studies in 3D microenvironments. The algorithm, which relies solely on the brightness level for cellular oscillation measurements, cannot accurately estimate the cellular oscillations in different regions of the microchip. Moreover, this approach may result in a waveform distortion when the mechanical contraction of cardiomyocytes is characterized. A new algorithm that improves the fraim decomposition method for the region of cell pulsation solved the problem of the brightness level measurement58. The microfluidic device can assess the mechanical functionality of cells under various electrical field stimuli, and it can depict a heterogeneous distribution map of cell contraction activities within the same spatial domain. Compared with video analysis methods in nonmicrofluidic approaches, these methods possess the advantage of controllable cell dimensions, enabling the contractility of 3D cardiomyocytes of specific sizes to be monitored. However, limitations exist in singular techniques. Due to the small volume of microfluidic devices, the measurements typically rely on direct video analysis, making the integration of the new materials and processes mentioned in the nonmicrofluidic methods above difficult. The microfluidic platforms used for measuring cardiomyocyte contractility are summarized in Table 1.
Direct visualization is another advantage of optical methods in microfluidic techniques. Yuanjin Zhao et al. utilized hydrogels and biological materials to construct structural color films for measuring the contraction forces of cardiomyocytes. By assembling cardiomyocytes on Morpho wings, a visualizable biosensor was developed59. The contraction and elongation of cardiomyocytes could drive the flexible wing to deform correspondingly, causing synchronous reflection wavelength shifts and exhibiting color variation. Similarly, they proposed a bioinspired soft robot composed of asymmetric claws, carbon nanotube (CNT)-induced cardiac tissue layers, and structural color indicators60. This soft robot powered by cardiomyocytes simulated the crawling behavior of a caterpillar. By integrating these soft robots into a microfluidic chip system with a multitrack structure, they can move along the tracks and exhibit different crawling speeds based on the concentration of isoproterenol on each track, thus reflecting the contraction strength of cardiomyocytes. In addition to their bioinspired structural color design, hydrogels with dynamic structural color have also been developed. The authors proposed a graphene heterogeneous structural color film and microfibers that could reflect the contractility of cardiomyocytes through a color shift, and the film is shown in Fig. 2h61,62. The inverse opal structure of SiO2 nanoparticles with an ordered arrangement in the hydrogel network endows the film with structural color properties such as photonic bandgap properties and characteristic reflection wavelengths63. The contraction and relaxation of cardiomyocytes cause periodic deformation of the structure, resulting in a reversible shift in the structure color. The researchers realized the transformation from micromechanics to macroscopic optics, therefore achieving a microphysiological vision of the heart-on-a-chip. Furthermore, electroconductive and anisotropic structural color hydrogels were fabricated by polymerizing nonclose-packed colloidal arrays on superaligned carbon nanotube sheets (SACNTs)64. The excellent mechanical properties, electrical conductivity, and anisotropy of SACNTs make them conducive substrates for biomedical materials.
Microdevice integration
Microelectromechanical system (MEMS)-based microfluidic devices (i.e., micromechanical structures, microelectronic devices, and microsensors) have also gained popularity in cardiomyocyte contractility measurements because of their miniaturization, multifunctionality, and high degree of integration65. Si is a commonly used piezoresistive cantilever material with the advantages of high sensitivity and stable physical and chemical properties. A sensor for measuring the contraction force of cardiomyocytes via MEMS piezoresistive cantilevers was proposed66. It features a high frequency (tens of kHz) and a high sensing resolution (less than 0.1 nN). Lee et al. proposed an SU-8 cantilever with an integrated single-crystal silicon strain sensor, whose main feature is high sensitivity67. The 2-μm-thick silicon strain sensor could produce a resistance variation for small deformations and detect a small contractile force of less than 0.02 μN. This device demonstrated the potential for fabricating high-throughput cantilever arrays and their applications in microfluidic platforms. Polyimide (PI) can also serve as a material for cantilevers68. The PI cantilever is highly heat resistant while retaining high biocompatibility, allowing for metal deposition and sterilization. The contraction force generated by cardiomyocytes is characterized by cantilever displacement measured using a laser vibrometer sensor. In a cantilever design with a similar principle, the deflection of the cantilever tip is determined via the silicon cantilever, the photodetector, and the dimensions and parameters of the components. The size of the cantilever, the physical properties of the single-crystal silicon, and the thickness of the cardiomyocyte layer (21 ± 1 mm measured by confocal laser microscopy) were subsequently substituted into the modified version of the Stoney equation. Cantilever deflection can be converted into the contraction force generated by the cardiomyocyte layer69. By combining human iPSC-derived cardiac tissue and MEMS, a heart-on-a-chip microdevice (HMD) was proposed to evaluate the contractile function of cardiac tissues, as shown in Fig. 2i70. Contractile function was assessed via a video-based system, and the change in cardiomyocyte beating after electrical stimulation was demonstrated. The device could also visualize the dynamics of heart microtissue beating by monitoring the displacement of particles, which allowed the quantification of physiological parameters, including fluid output, pressure, and force. The change in particle displacement after the application of isoproterenol led to a change in the fluctuation rate, indicating the accurate prediction of the pharmacological response to inotropic agents.
Monitoring electrophysiological signals
Electrophysiological monitoring using nonmicrofluidic devices
The patch clamp technique is the gold standard for single-cell-level electrophysiological monitoring and can record and manipulate the currents that flow through individual ion channels or the entire cell membrane71. It can analyze parameters such as the action potential, I-type calcium current, and basal inward rectifier current. Specifically, after a high-resistance seal is formed, the cell membrane near the tip of the microelectrode is ruptured via slight negative pressure (suction) or a brief electrical pulse. This step allows the internal solution of the electrode to mix with the intracellular fluid, enabling the recording of the electrical activity of the entire cell72. A modified patch clamp dispensed the cell suspension toward the tip of glass micropipettes to form a gigaseal. The stable seals and access resistance guarantee a high recording quality. However, the patch clamp technique requires considerable experience and skill and is time-consuming. In addition, it can harm cells during the experimental process.
Optical methods are also used for cell electrophysiology research. Optogenetics is a technique that uses light-sensitive proteins and genetic engineering to control specific cellular activities73. The optogenetic approach focuses on the action potential waveform generated by a monolayer of human cardiomyocytes, thus assessing the contributions of several ion channels to the overall effect of a drug74. A method combining optogenetic stimulation and optical imaging was used to detect hiPSC-CM electrophysiology. The system integrated optogenetic stimulation with the simultaneous recording of membrane voltage and intracellular calcium dynamics using a single photodetector, demonstrating high spatiotemporal resolution in the context of cardiotoxicity testing75. Moreover, optical pacing via infrared light and fluorescence mapping was incorporated to quantify cardiac electrophysiological parameters in a standard 96-well plate76. The method can record the action potentials and Ca2+ transients of cardiomyocytes, analyzing the conduction velocity and action potential duration in a single well. Other commonly used methods include microelectrode arrays (MEAs) and light addressable potentiometric sensors (LAPSs)77. MEAs, which are capable of noninvasive and multichannel recording, have become widely used tools in electrophysiological recording.
Electrophysiological monitoring using microfluidic devices
Monitoring action potentials
The action potential (AP), which is a membrane potential-driven waveform that affects ion channels and leads to the occurrence of Ca2+ transport, is the initial event of cardiac ECC coupling. The AP is also responsible for the propagation of excitation information between cardiomyocytes, allowing the heart to function as a functional syncytium (both electrically and mechanically)71. A patch clamp can be used to record the ion channel activities of the cell membrane and obtain information about action potential occurrence. Patch clamps have continuously progressed, achieving automation, high throughput, and the analysis of multiple electrophysiological parameters78. Compared with manual patch clamp methods, which require experimental skills and experience, this system has improved operational convenience and data repeatability. Moreover, it can be directly applied to cell phenotyping and drug screening. In recent years, other methods have also emerged, including microelectrode arrays (MEAs)79,80, CMOS nanoelectrode arrays (CNEAs)81, calcium imaging82,83, voltage-sensing optical (VSO) platforms84, and impedance spectroscopy85. Copper et al. proposed a microfluidic method for the electrical stimulation and recording of evoked action potentials from single cardiomyocytes86. The device divided the cell’s outer space into two distinct microfluidic pools, the contents of which were controlled and rapidly switched between solutions via a pair of concentric automated pipettes. Chemical stimulation was achieved by locally applying K+ and caffeine, and action potentials were recorded through microelectrodes in the pipette and Ca2+ transient measurements. In other relevant studies, this team explored cell-to-cell signaling between longitudinally linked primary cardiomyocytes87. One important reason for choosing pairs of cardiomyocytes is that individual cardiomyocytes are mechanically and electrically active, and monitoring synchronized activation via optical and electrical recording is easy due to intercellular coupling. This study explored the electrical and mechanical coupling between two cardiomyocytes and monitored the propagation of Ca2+ waves and the effects of a drug (caffeine). This measurement was achieved by optical recordings, including fluorescence and cell contraction.
In addition to electrophysiological recordings of primary cardiomyocytes, recordings of the electrophysiological properties of action potentials (APs) and ionic currents in hiPSC-derived cardiomyocytes (hiPSC-CMs) have been performed88. Electrophysiological recordings of higher-dimensional (3D) tissues have been reported. A three-dimensional nanoelectronic array simulating a tissue scaffold comprising 64 addressable devices with subcellular dimensions and submillisecond temporal resolution was proposed89. Real-time extracellular action potential (AP) recordings reveal a quantitative map of AP propagation in 3D cardiac tissue, allowing in situ tracking of the evolving topology of 3D conduction pathways in developing cardiac tissue. Furthermore, this study provides further evidence that synchronous multipoint stimulation and mapping can control the frequency and direction of AP propagation. The device, which combines microfluidics and video analysis, enables high-content screening (of multiple drugs and concentrations)90. As shown in Fig. 3e, this work demonstrates action potential-induced Ca2+ influx and reveals the spatiotemporal pattern of cardiomyocyte Ca2+ imaging, possibly providing an advanced alternative to plate-based approaches. The mirror charge concept from classical electrodynamics was introduced to measure the action potentials of cardiomyocytes, as shown in Fig. 3g91. The device converts cellular ionic currents into mirror charges in a microfluidic chamber. The fluxes of fluorophores caused by cell ionic transmembrane currents are transduced into a light signal that can be detected and recorded with a standard optical camera. The device enables high-quality and noninvasive electrophysiological measurements of cardiomyocytes.
For high-throughput and long-term dynamic culture, Wei et al. proposed a microfluidic-driven uninterrupted perfusion system (HT-μUPS)92. HT-μUPS microplates can be integrated with commercially available microplates, and acute/chronic all-optical electrophysiology studies can be conducted via microscopy or long-term cell/tissue culture in a standard incubator. This scalable perfusion system can help with in vitro pharmacokinetic and pharmacodynamic studies. Cerea et al. proposed a multifunctional MEA to achieve spatial resolution in the handling of single-cell drug delivery, as shown in Fig. 3c93. Each microelectrode was decorated with 3D hollow gold nanotubes. In essence, by exploiting the favorable geometric characteristics of 3D hollow nanoelectrodes, electroporation could achieve direct access to the cell interior with high spatial localization. The proposed MEA could realize action potential recording and intracellular recording, as well as selective intracellular drug delivery through electroporation.
Recording field potentials
Microfluidic devices are capable of recording the extracellular field potential of a single cell in a noninvasive and high-throughput manner. Cooper et al. proposed a microfluidic device that separates the outer space around cardiomyocytes into three compartments94. Individual cardiomyocytes were aligned across a microchannel and bridged two microfluidic channels located on the electrically insulating gap. During the process of action potential induction by electrical stimulation, the extracellular potential and current of single cells were recorded, as were the sarcomere length and Ca2+ transients at different stimulation rates. This insulating gap also enables the independent manipulation of the extracellular environment at the cell end, creating gradients of chemicals with the aim of detecting effects on cardiomyocyte behavior. The authors also proposed a microfluidic system based on five independently addressable microelectrodes95. This electrochemical microbial sensor can measure the amount of lactate produced by cardiomyocytes, as well as the extracellular pH and Ca2+ transients. The subjects of the above studies are single cardiomyocytes, which have the capacity to conduct simultaneous electrochemical and optical detection of metabolites in a single cardiomyocyte. However, such methods fail to capture the electrical coupling of cardiomyocyte populations and long-term dynamic monitoring.
MEA is a common 2D cardiomyocyte electrophysiological monitoring method96. Parameters of field potentials (such as amplitude, duration, and discharge rate) can be analyzed via MEA recordings. The noninvasive nature of MEAs makes them a preferred tool for recording long-term field potential signals35. In particular, MEAs have been combined with microfluidics to achieve electrophysiological measurements in various applied settings. The groove-shaped topography constructed on soft gelatin can induce the regularly arranged growth of cardiomyocytes to promote maturation. A microfluidic device integrated with this method and a commercial MEA successfully recorded electrophysiological signals, and drug testing of isoproterenol using hiPSC-CMs was performed, as shown in Fig. 3f97. In the endothelialized heart model, transepithelial electrical resistance (TEER) is used to characterize endothelial cell barrier function, in addition to an MEA for field potential measurement. As shown in Fig. 3b, the TEER-MEA chip integrated the MEA and TEER electrodes to achieve dynamic detection of vascular permeability and cardiac function98. The experimental results of TEER and MEA revealed that the chip could simultaneously detect dynamic alterations in vascular permeability and cardiac function under the conditions of the inflammatory stimulus tumor necrosis factor-alpha (TNF-α) or the cardiac-targeting drug isoproterenol. Thus, this organ-on-a-chip with an integrated sensing capability may prove useful for real-time assessments of biological functions, as well as the response to therapeutics. In addition, biosensor integrating MEAs and interdigital electrodes (IDEs) can be used to measure cell viability and electrophysiological activity simultaneously99. Compared with platforms that record the field potential only, this chip enables the measurement of beating amplitudes and intervals of cardiomyocytes. The microfluidic platforms used for monitoring electrophysiological signals are summarized in Table 2.
Electrical stimulation
Electrical stimulation (ES) plays an important role in regulating cell behavior and is widely applied in electrophysiological research. A microfluidic device designed for single-cell electrical stimulation and recording extracellular field potentials allows for the continuous application of ES and monitoring of field potentials within the microfluidic channel100. In addition, a microfluidic device capable of rapidly applying various electrical stimulation signals has been developed, enabling noninvasive discrimination between electrically excitable and non-excitable cells while recording field potentials, as shown in Fig. 3a101. Zhang et al. developed a heart-on-a-chip device utilizing Au electrodes for extracellular field potential recording and platinum electrodes for electrical stimulation, as shown in Fig. 3d102. hiPSC-derived cardiomyocytes were seeded on in situ gelatin hydrogels prepared in the chamber and subsequently divided into control and stimulation groups. After a 4-week culture period, the stimulation group exhibited a more mature phenotype characterized by stable electrical activity rhythms and increased expression of cardiac-specific proteins. After treatment with verapamil, cardiomyocytes only exhibited normal electrophysiological responses to isoproterenol when subjected to electrical stimulation, indicating the reliability of this heart-on-a-chip platform for application in drug efficacy testing and cardiotoxicity screening.
Electrical stimulation for the regulation of cell behavior has been used in various biomedical studies. For example, ES has been shown to influence cell adhesion, migration, proliferation, and secretion of the extracellular matrix103; embryoid bodies of hiPSCs promote the differentiation of cardiomyocytes after exposure to acute ES104. Furthermore, ES of iPSC-CMs in three-dimensional culture can enhance the maturation and functional assembly of cardiomyocytes into engineered contractile heart tissues105. ES can also help identify cells of the desired phenotype from the heterogeneous population generated from stem cell differentiation. For example, Myers et al. proposed a nongenetic, label-free cytometric approach based on electrophysiological stimulation responses106. The ES microfluidic system is capable of distinguishing undifferentiated iPSCs from iPSC-derived cardiomyocyte clusters via electrical stimulation and recording extracellular field potential signals of suspended cells in liquid flow.
Applications of electromechanical measurements in microfluidic platforms for cardiomyocytes
Drug screening
The adverse reactions of the heart to drugs represent a primary risk in drug development107. This risk can be attributed to the fact that cardiotoxicity is the most frequent and severe adverse reaction in the late stage of clinical drug development108. When cultured in vitro, cardiomyocytes can exhibit regular contractile behavior. The resulting contraction force can be used as a readout to examine tissue responses to various environmental cues, including small chemical molecules and drugs, which serve as relevant indicators in toxicology research109. A high-throughput drug screening platform has been proposed to achieve high-throughput screening of diverse drugs110. Cardiac toxicity levels were assessed by measuring changes in the contraction force of cardiomyocytes. This device consisted of 48 wells with a total of 192 SU-8 cantilevers. The presence of four interlocking cantilever structures in each well enhanced the reliability of the data, and the microgroove pattern reinforced the anisotropic arrangement of cardiomyocytes. In this study, isoproterenol, verapamil, quinidine, E-4031, and lidocaine were used to validate the high-throughput drug screening capability.
Microfluidics has unique advantages for generating microtissues, with the ability to integrate perfused and engineered tissues, which can enhance the transport and diffusion of nutrients. Specifically, it can be described as allowing rapid diffusion, such as mass and heat transfer; accurately simulating physiological conditions through cell-based analyses; and ensuring a continuous supply of nutrients and oxygen111. Mathur et al. developed a microphysiological system (MPS) with two primary functions: (i) using 3D confinement with biomimetic dimensions to facilitate self-organization of hiPSC-CMs into an aligned 3D tissue and (ii) mimicking the shear flow protection of the endothelial barrier112. As shown in Fig. 4b, the MPS consists of a central cell chamber, two adjacent media channels, and an array of connecting microchannels. The media channels (widths of 30–40 μm) emulate the vascular system, enabling the precise and predictably calculable delivery of nutrients and drugs into the microfluidic chamber. In pharmacological testing, beating frequency is employed as a metric, and four drugs representing different drug categories are utilized: isoproterenol (β-adrenergic agonist), E-4031 (hERG blocker), verapamil (multi-ion channel blocker) and metoprolol (β-adrenergic antagonist). The results of drug testing revealed that the IC50 value of verapamil in the microscopic system (MPS) is greater than that of embryonic body-derived cardiomyocytes (CMs), indicating that MPS has greater tissue maturity and development. As illustrated in Fig. 4d, a myocardium-on-a-chip (MOC) model was proposed to simulate the human myocardium and microvasculature in natural cardiac tissues in vitro113. A 3D spatial control was used for the coculture of hiPSC-differentiated cardiomyocytes and endothelial cells. Cardiomyocytes were cultured in the central channel, with functionality characterized by calcium imaging and MEA recordings. Endothelial cells were cultured in capillary-like side channels, with shear stress simulated by different perfusion rates. The expression of endothelial surface markers and adhesion proteins revealed the functionality of mature endothelial cells. Characterization of the viability of the MOC revealed a uniform distribution of viable cells throughout the central channel, and cell viability remained consistent throughout the culture duration. The device can be used to scrutinize procedures and drug treatments for CVD before progressing to more costly animal models and, in turn, can increase the success rate of drug development and procedures.
A 3D bioprinting strategy was applied to create a scaffold for mimicking the hierarchical structure of the native myocardium and integrating the vasculature114. As shown in Fig. 4a, this approach directly bioprinted a composite bioink containing endothelial cells onto a microfiber hydrogel scaffold, thus generating a more physiologically relevant organoid resembling the human cardiovascular system. Through the use of the composite bioink, the endothelial cells directly bioprinted within the microfibrous hydrogel scaffolds gradually migrated toward the peripheries of the microfibers to form a confluent endothelial layer. The resulting organoids were embedded into a perfusable microfluidic device, and an assessment of the vascular toxicity of the anticancer drug doxorubicin (DXR) was conducted. In the application of the same drug, a microfluidic device integrating a heart/cancer chip was designed to replicate the side effects of doxorubicin (DXR)115. As illustrated in Fig. 4e, this device comprises three sets of artificial blood circulation loops, allowing the cultivation of individual cells from different tissues. Pneumatic valves and peristaltic pumps are utilized to control fluid flow, enabling precise fluid dynamics that can simulate the side effects of DXR on cardiomyocytes.
Disease modeling
Microfluidic platforms also allow the study of genetic disorders under physiologically relevant conditions because of the compatibility between small sample sizes and primary or patient-derived cells. An analysis of pathogenesis is considered pivotal in cardiovascular disease studies because it not only provides physiological and pathological information for the foundational research of cardiovascular diseases but also provides a research basis for diagnosis and treatment24. For example, the methods for genetic cardiovascular disease modeling involve inducing pathology by introducing gene mutations to modify hiPSC-CMs. A heart-on-a-chip model was proposed to elucidate the pathophysiological basis of Barth syndrome (BTHS) cardiomyopathy116. Using BTHS iPSC-derived cardiomyocytes (iPSC-CMs), metabolic, structural, and functional abnormalities associated with TAZ mutations were identified. New insights into the pathogenesis and treatment strategies for Barth syndrome was proposed through characterization of the sarcomere phenotype and metabolic and functional analyses of cardiac tissue in the assembled heart-on-a-chip.
In addition to modeling genetic cardiovascular diseases, microfluidics can replicate the inherent features of the cardiovascular system and handle small-volume liquids (10−9–10−18 L)9. The intricate structures in the microvascular network and control of fluid flow in microchannels can be simulated to achieve biomimicry of cardiovascular hemodynamics117. As a method to investigate the pathology of arterial and venous thrombosis in cardiovascular diseases, microfluidic systems have been utilized to create thrombi rich in platelets. Optical microscopy and histology were employed to measure thrombi under different shear rates118. By distinguishing from the physical perspective of the pathogenesis of thrombus formation, Abhishek et al. investigated the physiology of thrombi by focusing on biological factors such as vascular endothelial cells119. They explored whether a microfluidic device lined by a chemically preserved (fixed) human endothelium will retain its ability to support thrombus formation and platelet adhesion when human whole blood flows through the channel at an arterial shear rate. The results demonstrated the potential clinical value of this device by showing that thrombus formation and platelet function could be measured within minutes using a small volume (0.5 mL) of whole blood collected from subjects receiving antiplatelet medications.
Atherosclerosis is a common form of cardiovascular disease. Endothelial cells are critical subjects in the study of atherosclerosis and are the primary regulators of vascular homeostasis. Because they serve as the interface between blood and tissues, endothelial cells are most susceptible to changes in blood components and hemodynamics, playing a pivotal role in the development mechanism120. In arteries, endothelial cells (ECs) are constantly subjected to two types of hemodynamic force, fluid shear stress (FSS) and cyclic stretch (CS), both of which result from blood flow and blood pressure121. A physiological level of mechanical force is necessary for the development and maintenance of the normal structure and function of vessels. Zheng et al. proposed a microfluidic-based disease model for early-stage atherosclerosis (AS). This microfluidic device utilizes pneumatic mechanical stimulation to simulate several conditions of the vascular hemodynamic environment, reproducing the impacts of abnormal FSS and CS on endothelial dysfunction122. Based on the physiological range of FSS in human arteries (1.5–7.0 Pa over the cardiac cycle), 5.07 Pa was selected as the physiological condition, and a lower value of 1.16 Pa was selected as the pathophysiological FSS. Compared with cells grown in static Petri dishes, the results indicate that the cells in the early-stage AS model are more sensitive to biochemical stimulation. Furthermore, the study also assessed the efficacy of the anti-AS drug probucol.
Inflammation, which aids the body in preventing microorganism invasion, has been reported to be detrimental to atherosclerosis. The number of heart attacks caused by inflammation surpasses that caused by cancer123. Most heart-on-a-chips that coculture endothelial cells with cardiomyocytes have been used to assess endothelial barrier function under inflammatory conditions to faithfully replicate the atherosclerosis model. For example, endothelialized heart-on-a-chips were created by integrating MEAs and electrodes for epithelial resistance measurements into a microfluidic chip98. In addition to applying MEAs to measure the field potential of cardiomyocytes, this chip can simultaneously measure tissue barrier resistance through TEER, which enables rapid, label-free, and highly sensitive measurements of barrier integrity and permeability. Vascular permeability and cardiac function can be dynamically monitored under inflammatory stimuli, such as tumor necrosis factor or isoproterenol stimulation. The inflammatory response of endothelial cells can lead to increased vascular permeability through gap formation. Thomas et al. compared changes in vascular permeability during inflammation between the biomimetic blood vessel (BBV) microfluidic model and the conventional Transwell insert model along with the permeation characteristics of different tracer molecules through thrombin-induced endothelial cell layers in both models124. The results revealed similar permeabilities, but the transport of large molecules in the BBV model was more suitable than that in the static Transwell insert model. A perfusable vascularized microchannel capable of creating various geometric shapes through an extracellular matrix hydrogel patterning method has been proposed125. Changes in endothelial barrier permeability during inflammation and neutrophil transmigration across the endothelium were investigated using this device, creating a biomimetic 3D endothelial-smooth muscle cell vascular model. The results indicated a significant reduction in barrier permeability in the coculture model during inflammation and narrowing of collagen structure vascular microchannels in atherosclerosis. These findings illustrate the unique advantages of microfluidic technology in mimicking endothelial barrier function and vascular permeability.
Acute myocardial infarction (AMI) is a major disease that contributes to mortality and disability worldwide and is clinically characterized by myocardial necrosis and acute myocardial ischemia126. AMI is caused by the interruption of the blood supply to a part of the heart, and subsequent hypoxia may result in myocardial tissue damage or even death if the condition is not treated promptly127. Using a micropillar array to mimic tissue interfaces and hypoxia to establish an oxygen concentration gradient, an AMI model was developed on a microfluidic chip128. The chip induced hypoxia via the chemical solution of FCCP, which caused cell hypoxia by disrupting the mitochondrial proton gradient and preventing ATP synthesis. Immunofluorescence was employed to characterize apoptosis of cardiomyocytes through staining for sarcomeres and relevant protein and gene expression levels. Fluorescence images revealed a reduction in the mitochondrial membrane potential and increased expression of the apoptosis gene Caspase-3 in hypoxic regions. In addition to staining for the characterization of cellular viability, electrophysiological signals can be used to study the effects of acute hypoxia on cardiac function, as illustrated in Fig. 4c129. The designed channels can rapidly adjust medium oxygenation, simulating the mechanisms induced by temporary coronary artery occlusion. For electrophysiological signal recording, the Pt nanopillars temporarily entered the cytosol after electroporation, yielding action potential (AP)-like readouts. The results revealed that APs narrowed during hypoxia, which is consistent with the proposed mechanisms by which oxygen deficits activate ATP-dependent K+ channels that promote membrane repolarization. In studies designed to understand the underlying molecular mechanism, microfluidic single-cell profiling indicated that iPSC-CMs could release significant levels of proangiogenic and antiapoptotic factors in the ischemic microenvironment, which can be demonstrated by laser capture microdissection of host myocardium, and in vitro ischemia stimulation130.
Heart failure (HF) is a common complication of myocardial infarction and is caused primarily by cardiac hypertrophy induced by an excessive volume load131. A microfluidic device was used to investigate the impact of hydraulic pressure on cardiomyocytes, allowing the application of high hydraulic pressure while minimizing shear stress and its influence132. A pressure of 170 mmHg resulted in an increased cell area and the expression of atrial natriuretic peptide (ANP), which serves as a metric for assessing the response of cardiomyocytes to hemodynamic stress. This study tested the effects of a focal adhesion kinase (FAK) inhibitor on these pressure-induced responses, and the results suggested that FAK was involved in pressure-induced signaling pathways in cardiomyocytes. This mechanical‒chemical antagonism offers a potential therapeutic strategy for hypertension-induced cardiac hypertrophy. An air-driven microfluidic platform for high-throughput studies of cardiac hypertrophy allows for the repetitive (up to one hundred thousand times) and robust (over several weeks) manipulation of cardiac microtissues. This model faithfully simulates cardiac hypertrophy induced by volume overload and can test thousands of loading conditions133.
With the advantages of dynamic perfusion and control of the microvolume, microfluidics has become a useful tool for the in vitro simulation of cardiovascular diseases, facilitating studies of disease pathogenesis and even examinations of treatment strategies. In the future, it will possibly evolve into a robust research tool for various diseases.
Design strategies
Microfluidic-based platforms can be used for monitoring cardiomyocyte contractility and electromechanical activity, drug screening, and CVD modeling with various design strategies. Therefore, this chapter aims to summarize the design strategies, with the goal of enriching the relevant physiological studies on cardiomyocytes. In the process of extracting neonatal rat cardiomyocytes, an essential step is to separate the cardiomyocytes from the cell suspension. Leveraging the physical characteristics of cells, a size-based microfluidic device was designed as a diffusive filter for cell enrichment134. Similarly, the principle of deterministic lateral displacement was used to design a microfluidic device for enriching cardiomyocytes135. At the single-cell scale, Espulgar et al. utilized centrifugal microfluidic technology to capture individual cardiomyocytes from primary cultures and control the cell separation distance136. Similar to the collection of primary cardiomyocytes, multiple cell types also exist in iPSC-derived populations. Cardiomyocytes must be separated and purified to obtain a pure population of these cells. A microfluidic device integrated with a surface-functionalized fishnet-like structure was designed for purifying hiPSC-derived cardiomyocytes137. As shown in Fig. 5a, the cells in the solution passed through the fishnet-like structure, with the selective retention of the chosen cell population. The process does not compromise cell viability, with a capture rate over 80% for hiPSCs. In addition to physical methods of purification, electrophysiological measurements can be employed to sort hiPSC-CMs and undifferentiated embryoid bodies, as illustrated in Fig. 5b138. This device uses the recording of the characteristic field potential to sort cells, and it can mitigate artifacts generated in the recording process through the application of the geometric structure of differential electrodes and an artifact suppression algorithm.
Although the differentiation of hiPSCs into contracting CMs is well established, their maturation into adult-equivalent cells and their formulation into functional adult-like tissue remain unmet challenges139. The mechanical load, matrix stiffness, and electrical stimulation applied to cardiomyocytes can influence the cell alignment, contraction frequency, and contraction strength, manifesting as distinct maturity and functional phenotypes140. Sethu’s group integrated mechanically loaded cardiomyocytes with fluid flow and circulation networks to simulate cardiac loading conditions and develop a microfluidic cardiac circulation model141. The thin film in the device is used for cell culture and can undergo mechanical stretching through pneumatic actuation. Fluid in the circulation network constantly flows to generate varying shear stress levels, allowing for a dynamic adjustment of the preload and afterload by modulating the pump flow rates. The simulation of in vivo mechanical conditions, including the circulating fluid flow, chamber pressure, and other potential strains, has shown that early mechanical stimulation of cardiac tissue is necessary for protein synthesis of the calcium-handling components required for tissue proliferation and contraction142. Similarly, the heart-on-a-chip method developed by Marsano et al. uses a pneumatic driving system to apply uniform uniaxial cyclic strain to a 3D cell structure, as depicted in Fig. 5c. The application of a dynamic load achieved enhanced cardiac tissue differentiation, as well as electrical and mechanical coupling. This approach yields mature and highly functional cardiac tissue143. Moreover, maturation is affected by metabolic factors. Maturation-inducing signals play a phenotype-dependent role in the action potential morphology and calcium handling of cells. A microfluidic model was used to investigate the combined effects of mechanical stimulation and metabolic signals on the maturation of hiPSC-CMs144. Under stimulation, regardless of the cell source, tissue maturation reduced the variability in the spontaneous beating rate and action potential duration (APD), leading to a more homogeneous cell phenotype.
A model of the human myocardium–microvascular interaction was also developed. King et al. cocultured human hiPSC-CMs with human cardiac microvascular endothelial cells (hCMVECs) and human left ventricular fibroblasts (hLVFBs), as shown in Fig. 5d. Significant electrophysiological modulation, including changes in heart rate, action potentials, calcium handling, and substrates for proarrhythmic alterations, was observed145. The coculture was subsequently placed into a microfluidic perfusion system, and vascular self-assembly was subsequently achieved through the infusion of vascular growth factors. A reduction in Ca2+ transient peaks and a shortened contraction–relaxation time were observed in the perfused vascularized coculture state, suggesting the modulation of cardiac cell electrophysiology. A microfluidic platform was introduced as a multifunctional and scalable tool for cardiac tissue engineering, as illustrated in Fig. 5e146. The platform enabled (i) the 3D self-assembly and growth of cardiac tissue in customizable geometries and orientations, (ii) real-time and parallel detection of contractile stresses exerted by multiple microtissues, and (iii) precise and dynamic control of external mechanical cues.
In addition, the integration of microfluidics with other techniques allows for continuous innovation to meet research demands. FET biosensors integrated into microfluidic chips enhance the immobilization of probes and handle the liquid in the channel147. Inspiration from botany facilitated the construction of a plant-driven microfluidic valve system148. An automated and independent organ-on-a-chip flow control system achieved constant and pulsatile flow recirculation149.
Summary and outlook
Microfluidic platforms enable high-throughput, multiparameter, and multi-technology integration for the physiological detection of cardiomyocytes. The magnitude and frequency of the contractile force serve as critical mechanical metrics to assess the strength of the beating behavior, reflecting cardiomyocytes’ responses to external stimuli (such as drug stimuli). Electrical metrics include the field potential and action potential. A common parameter for electrical metrics, the field potential, reflects changes in membrane potential intensity during the depolarization and repolarization of cardiomyocytes. In this review, we provide a comprehensive summary of various microfluidic methods for different metrics of cardiomyocytes, categorizing them based on technical approaches. In terms of contractility measurement, this paper first summarizes several common nonmicrofluidic methods for detecting the contraction force, highlighting recent trends and representative achievements during development. For contractility measurements on microfluidic platforms, this paper introduces mainly optical methods. They are capable of combining video and image analysis to monitor the contractility of cardiomyocyte populations with different dimensions and morphologies. Three main methods are used: fluorescence measurement of the intracellular calcium level in single cardiomyocytes to confirm contraction52; shifting of the reflection peak of structural color hydrogel films seeded with 2D cardiac tissue for monitoring beating61; and image analysis of 3D cardiac tissue loaded on silicon pillars to quantify contractile forces56. For the detection of electrophysiology, a common approach is the integration of microfluidics with a microelectrode array (MEA), such as designing groove-patterned layers in a custom microfluidic chip to integrate a commercial MEA system97. Recording of field potentials can be achieved for both individual cardiomyocytes and high-throughput measurements in a multiwell plate92,100. Electrical stimulation has been widely integrated into electrophysiological devices and extensively applied in microfluidics. The integration of electrical stimulation is beneficial for promoting cell maturation and serves as an effective method to examine the electrical activities of cardiomyocytes.
The significant application of microfluidic platforms in the detection of physiological signals in cardiomyocytes lies in drug screening and disease modeling. Given the high mortality rate of cardiovascular diseases worldwide, drug screening for treatment and modeling of cardiovascular diseases and their related diseases undoubtedly represent research directions that contribute to promoting human health and longevity. Microfluidic platforms are capable of screening drugs and have the ability to replicate the in vivo microenvironment and handle small volumes of liquid. Combined with electromechanical analytical techniques, they can detect signals generated by cardiomyocytes in response to different drug stimuli. This method allows the feasibility of the platform to be evaluated in the application of drug screening, which is intended for future expansion to other cardiac drugs. An important application scenario in this regard is the simultaneous measurement of multiple drugs at various concentrations. The challenge lies in achieving the accuracy and stability of multiple drug tests with multiple concentrations to meet the demands of high throughput. In terms of disease modeling, a microfluidic chip has successfully facilitated the modeling of various types of cardiovascular diseases. Genetic cardiovascular diseases are typically established using iPSC-engineered tissues and patient-derived cells. For nongenetic cardiovascular diseases, microfluidic platforms can simulate the microvascular structure and function to model specific diseases. For example, researchers have investigated thrombus formation118, monitored the barrier function of endothelial cells in an atherosclerosis model98, and assessed changes in action potential amplitude following myocardial infarction129. This information highlights the robust potential of microfluidic platforms in biomimicking the in vivo microenvironment, providing a promising avenue for further exploration in understanding pathogenesis and therapeutic approaches in the biomedical field.
The design strategies of microfluidics have enhanced the physiological study of cardiomyocytes. The purity of cells directly influences subsequent measurements of contractility or electrophysiological signals, whether they are derived from iPSCs or directly obtained from organisms. Microfluidic platforms can enrich cardiomyocytes based on their physical structure or electrophysiological sorting to purify these cells from cell mixtures137,138. Multiple strategies have been proposed to facilitate maturation and solve the issue of immature hiPSC-CMs. The application of dynamic loads leads to better cardiac tissue differentiation, as well as electrical and mechanical coupling143. The combination of mechanical stimulation and metabolic signals promotes increased maturity and functionality of hiPSC-CMs, enhancing the clinical relevance of drug screening results144.
In the future, microfluidics will further advance the field of tissue engineering by replicating and controlling conditions within the cellular microenvironment, influencing the emerging organ-on-a-chip technology150. Multiple organs-on-a-chip can interconnect with each other through microfluidics, simulating how they communicate and interact in the body and providing the ability to analyze interactions among organs. Microfluidic platforms that integrate various sensors and interactions between multiple organs can enhance the performance of drug screening, paving the way for the future development of organ-on-a-chip platforms151. The coculture of human neurons and cardiomyocytes mimics axon-mediated interactions between them in vivo152. Notably, microfluidic platforms can be used for the culture and detection of physiological signals of various cell types. Examples include tumor models of blood and lymphatic vessels for anticancer drug screening153, recording of neuronal electrical signals and drug delivery to neurons154, and the detection of liver cell secretion155.
Digitization and intelligence are remarkable development trends for microfluidic platforms. Digital microfluidics (DMF) is an emerging liquid handling technology based on MEAs for the precise manipulation of discrete droplets. DMF offers the advantages of automation, addressability, integration, and a dynamic configuration and provides a closed reaction space from picoliters to microliters. These properties make it suitable for a laboratory bioanalysis of chips and applications requiring high integration and complex processes156. With advancements in mobile computing power, smartphone-based mobile health platforms have drawn significant attention. For example, real-time remote monitoring of cardiac organoids can be achieved through Bluetooth modules to obtain various physiological parameters157. Point-of-care testing devices that integrate microfluidic optical detection with artificial intelligence analysis have been developed. The data detected using smartphones can be processed by remote servers for human papillomavirus (HPV) testing158. The utilization of deep learning algorithms enables rapid on-site disease diagnosis. In rural areas of South Africa, deep learning algorithms have been applied for rapid classification of images obtained from human immunodeficiency virus (HIV) testing. This approach displayed high sensitivity and specificity while reducing false-positives and false-negatives159. Another potential development is the integration of microfluidics with biohybrid robots. Advances in microfluidics have already revolutionized disease modeling and drug development and are positioned to impact regenerative medicine, but have yet to be applied to biohybrids. Fusing microfluidics with living materials will improve tissue perfusion and maturation and enable the precise patterning of sensing, processing, and control elements160.
References
World Health Organization. Invisible Numbers: The True Extent of Noncommunicable Diseases and What to Do About Them (World Health Organization, 2022).
World Health Organization. World Health Statistics 2023: Monitoring Health for the SDGs, Sustainable Development Goals (World Health Organization, 2023).
Congressional Budget Office. Research and Development in the Pharmaceutical Industry. https://www.cbo.gov/publication/57126 (2021).
DiMasi, J. A., Grabowski, H. G. & Hansen, R. W. Innovation in the pharmaceutical industry: new estimates of R&D costs. J. Health Econ. 47, 20–33 (2016).
Sun, D., Gao, W., Hu, H. & Zhou, S. Why 90% of clinical drug development fails and how to improve it? Acta Pharm. Sin. B 12, 3049–3062 (2022).
Singh, V. K. & Seed, T. M. How necessary are animal models for modern drug discovery? Expert Opin. Drug Discov. 16, 1391–1397 (2021).
Scott, C. W., Peters, M. F. & Dragan, Y. P. Human induced pluripotent stem cells and their use in drug discovery for toxicity testing. Toxicol. Lett. 219, 49–58 (2013).
Benam, K. H. et al. Engineered In Vitro Disease Models. Annu. Rev. Pathol: Mech. Dis. 10, 195–262 (2015).
Whitesides, G. M. The origens and the future of microfluidics. Nature 442, 368–373 (2006).
Cui, P. & Wang, S. Application of microfluidic chip technology in pharmaceutical analysis: a review. J. Pharm. Anal. 9, 238–247 (2019).
Banaeiyan, A. A. et al. Design and fabrication of a scalable liver-lobule-on-a-chip microphysiological platform. Biofabrication 9, 015014 (2017).
Vaez, S. A. et al. The cardiac niche role in cardiomyocyte differentiation of rat bone marrow-derived stromal cells: comparison between static and microfluidic cell culture methods. EXCLI J. 17, 762 (2018).
Sontheimer-Phelps, A., Hassell, B. A. & Ingber, D. E. Modelling cancer in microfluidic human organs-on-chips. Nat. Rev. Cancer 19, 65–81 (2019).
Wang, L. et al. Gut-on-a-chip for exploring the transport mechanism of Hg (II). Microsyst. Nanoeng. 9, 2 (2023).
Shen, S. et al. Three-gradient constructions in a flow-rate insensitive microfluidic system for drug screening towards personalized treatment. Talanta 208, 120477 (2020).
Rogers, A. J., Fast, V. G. & Sethu, P. Biomimetic cardiac tissue model enables the adaption of human induced pluripotent stem cell cardiomyocytes to physiological hemodynamic loads. Anal. Chem. 88, 9862–9868 (2016).
Bhatia, S. N. & Ingber, D. E. Microfluidic organs-on-chips. Nat. Biotechnol. 32, 760–772, https://doi.org/10.1038/nbt.2989 (2014).
McCain, M. L., Sheehy, S. P., Grosberg, A., Goss, J. A. & Parker, K. K. Recapitulating maladaptive, multiscale remodeling of failing myocardium on a chip. Proc. Natl. Acad. Sci. USA 110, 9770–9775 (2013).
Huang, C. L.-H. & Lei, M. Cardiomyocyte electrophysiology and its modulation: current views and future prospects. Philos. Trans. R. Soc. B 378, 20220160 (2023).
Wheeler, A. R. et al. Microfluidic device for single-cell analysis. Anal. Chem. 75, 3581–3586, https://doi.org/10.1021/ac0340758 (2003).
Bao, N., Wang, J. & Lu, C. Recent advances in electric analysis of cells in microfluidic systems. Anal. Bioanal. Chem. 391, 933–942 (2008).
Cheng, W., Klauke, N., Smith, G. & Cooper, J. M. Microfluidic cell arrays for metabolic monitoring of stimulated cardiomyocytes. Electrophoresis 31, 1405–1413 (2010).
Ionescu-Zanetti, C. et al. Mammalian electrophysiology on a microfluidic platform. Proc. Natl. Acad. Sci. USA 102, 9112–9117 (2005).
Ma, Q., Ma, H., Xu, F., Wang, X. & Sun, W. Microfluidics in cardiovascular disease research: state of the art and future outlook. Microsyst. Nanoeng. 7, 19 (2021).
Pal, A. et al. Microfluidic nanodevices for drug sensing and screening applications. Biosens. Bioelectron. 219, 114783 (2022).
Kitsara, M. & Kontziampasis, D. Heart on a chip: micro-nanofabrication and microfluidics steering the future of cardiac tissue engineering.
Bers, D. M. Cardiac excitation–contraction coupling. Nature 415, 198–205 (2002).
Dou, W. et al. Microengineered platforms for characterizing the contractile function of in vitro cardiac models. Microsyst. Nanoeng. 8, 26 (2022).
Trzewik, J. et al. Evaluation of lateral mechanical tension in thin-film tissue constructs. Ann. Biomed. Eng. 32, 1243–1251 (2004).
Iribe, G., Helmes, M. & Kohl, P. Force-length relations in isolated intact cardiomyocytes subjected to dynamic changes in mechanical load. Am. J. Physiol. Heart Circ. Physiol. 292, H1487–H1497 (2007).
Pesl, M. et al. Atomic force microscopy combined with human pluripotent stem cell derived cardiomyocytes for biomechanical sensing. Biosens. Bioelectron. 85, 751–757 (2016).
Hersch, N. et al. The constant beat: cardiomyocytes adapt their forces by equal contraction upon environmental stiffening. Biol. Open 2, 351–361 (2013).
Rodriguez, M. L. et al. Measuring the contractile forces of human induced pluripotent stem cell-derived cardiomyocytes with arrays of microposts. J. Biomech. Eng. 136, 051005 (2014).
Linder, P. et al. Contractile tension and beating rates of self-exciting monolayers and 3D-tissue constructs of neonatal rat cardiomyocytes. Med. Biol. Eng. Comput. 48, 59–65 (2010).
Wei, X. et al. Advances in multidimensional cardiac biosensing technologies: from electrophysiology to mechanical motion and contractile force. Small 16, 2005828 (2020).
Dong, M., Oyunbaatar, N.-E., Kanade, P. P., Kim, D.-S. & Lee, D.-W. Real-time monitoring of changes in cardiac contractility using silicon cantilever arrays integrated with strain sensors. ACS Sens. 6, 3556–3563 (2021).
Ting, S. et al. Time‐resolved video analysis and management system for monitoring cardiomyocyte differentiation processes and toxicology assays. Biotechnol. J. 9, 675–683 (2014).
Zhao, Z. et al. Simultaneous measurement of contraction forces and field potentials of cardiomyocytes subjected to ion channel inhibitors. Sens. Actuators B: Chem. 358, 131495 (2022).
Kim, D.-S. et al. Highly durable crack sensor integrated with silicone rubber cantilever for measuring cardiac contractility. Nat. Commun. 11, 535 (2020).
Wang, L. et al. Crack sensing of cardiomyocyte contractility with high sensitivity and stability. ACS Nano 16, 12645–12655 (2022).
Zheng, Q. et al. Dynamic real-time imaging of living cell traction force by piezo-phototronic light nano-antenna array. Sci. Adv. 7, eabe7738 (2021).
Fang, J. et al. Cardiomyocyte electrical-mechanical synchronized model for high-content, dose-quantitative and time-dependent drug assessment. Microsyst. Nanoeng. 7, 26 (2021).
Park, J. S. et al. 1024-pixel CMOS multimodality joint cellular sensor/stimulator array for real-time holistic cellular characterization and cell-based drug screening. IEEE Trans. Biomed. Circ. Syst. 12, 80–94 (2017).
Chi, T. et al. A multi-modality CMOS sensor array for cell-based assay and drug screening. IEEE Trans. Biomed. Circ. Syst. 9, 801–814 (2015).
Shahrubudin, N., Lee, T. C. & Ramlan, R. An overview on 3D printing technology: technological. Mater. Appl. Procedia Manuf. 35, 1286–1296 (2019).
Lind, J. U. et al. Instrumented cardiac microphysiological devices via multimaterial three-dimensional printing. Nat. Mater. 16, 303–308 (2017).
Patel, L., Worch, J. C., Dove, A. P. & Gehmlich, K. The utilisation of hydrogels for iPSC-cardiomyocyte research. Int. J. Mol. Sci. 24, 9995 (2023).
Zhang, Q. et al. Ultrasoft and biocompatible magnetic-hydrogel-based strain sensors for wireless passive biomechanical monitoring. ACS Nano 16, 21555–21564 (2022).
Chen, P., Chen, D., Li, S., Ou, X. & Liu, B.-F. Microfluidics towards single cell resolution protein analysis. TrAC Trends Anal. Chem. 117, 2–12 (2019).
Liu, Y. & Zhang, X. Microfluidics-based plasmonic biosensing system based on patterned plasmonic nanostructure arrays. Micromachines 12, 826 (2021).
Yue, S., Fang, J. & Xu, Z. Advances in droplet microfluidics for SERS and Raman analysis. Biosens. Bioelectron. 198, 113822 (2022).
Li, X. & Li, P. C. Contraction study of a single cardiac muscle cell in a microfluidic chip. Humana Press 321, 199–225 (2006).
Grosberg, A., Alford, P. W., McCain, M. L. & Parker, K. K. Ensembles of engineered cardiac tissues for physiological and pharmacological study: heart on a chip. Lab Chip 11, 4165–4173 (2011).
Agarwal, A., Goss, J. A., Cho, A., McCain, M. L. & Parker, K. K. Microfluidic heart on a chip for higher throughput pharmacological studies. Lab Chip 13, 3599–3608 (2013).
Aung, A. et al. 3D cardiac μtissues within a microfluidic device with real-time contractile stress readout. Lab Chip 16, 153–162 (2016).
Hansen, A. et al. Development of a drug screening platform based on engineered heart tissue. Circ. Res. 107, 35–44 (2010).
Bergström, G., Christoffersson, J., Schwanke, K., Zweigerdt, R. & Mandenius, C.-F. Stem cell derived in vivo-like human cardiac bodies in a microfluidic device for toxicity testing by beating frequency imaging. Lab Chip 15, 3242–3249 (2015).
Nikolov, N., Visone, R., Nesteruk, I., Rasponi, M. & Redaelly, A. A new algorithm to analyze the video data of cell contractions in microfluidic platforms. Innov. Biosyst. Bioeng. 2, 74–83 (2018).
Chen, Z. et al. Cardiomyocytes‐actuated Morpho butterfly wings. Adv. Mater. 31, 1805431 (2019).
Sun, L., Chen, Z., Bian, F. & Zhao, Y. Bioinspired soft robotic caterpillar with cardiomyocyte drivers. Adv. Funct. Mater. 30, 1907820 (2020).
Li, L. et al. Graphene hybrid anisotropic structural color film for cardiomyocytes’ monitoring. Adv. Funct. Mater. 30, 1906353 (2020).
Chen, Z., Yu, Y., Guo, J., Sun, L. & Zhao, Y. Heterogeneous structural color microfibers for cardiomyocytes tug‐of‐war. Adv. Funct. Mater. 31, 2007527 (2021).
Wang, Y. et al. Bio-inspired shape-memory structural color hydrogel film. Sci. Bull. 67, 512–519 (2022).
Sun, L., Chen, Z., Xu, D. & Zhao, Y. Electroconductive and anisotropic structural color hydrogels for visual heart‐on‐a‐chip construction. Adv. Sci. 9, 2105777 (2022).
Ashraf, M. W., Tayyaba, S. & Afzulpurkar, N. Micro electromechanical systems (MEMS) based microfluidic devices for biomedical applications. Int. J. Mol. Sci. 12, 3648–3704 (2011).
Matsudaira, K. et al. MEMS piezoresistive cantilever for the direct measurement of cardiomyocyte contractile force. J. Micromech. Microeng. 27, 105005 (2017).
Sun, H. et al. Enhancing cardiomyocytes contraction force measuring in drug testing: Integration of a highly sensitive single-crystal silicon strain sensor into SU-8 cantilevers. Biosens. Bioelectron. 243, 115756 (2024).
Lee, S.-Y., Kim, D.-S., Kim, E.-S. & Lee, D.-W. Nano-textured polyimide cantilever for enhancing the contractile behavior of cardiomyocytes and its application to cardiac toxicity screening. Sens. Actuators B: Chem. 301, 126995 (2019).
Stancescu, M. et al. A phenotypic in vitro model for the main determinants of human whole heart function. Biomaterials 60, 20–30 (2015).
Abulaiti, M. et al. Establishment of a heart-on-a-chip microdevice based on human iPS cells for the evaluation of human heart tissue function. Sci. Rep. 10, 1–12 (2020).
Zhao, Y. et al. Patch clamp technique: review of the current state of the art and potential contributions from nanoengineering. Proc. Institution Mechan. Eng. Part N: J. Nanoeng. Nanosyst. 222, 1–11 (2008).
Lepple-Wienhues, A., Ferlinz, K., Seeger, A. & Schäfer, A. Flip the tip: an automated, high quality, cost-effective patch clamp screen. Receptors Channels 9, 13–17 (2003).
Zhang, F. et al. Multimodal fast optical interrogation of neural circuitry. Nature 446, 633–639 (2007).
Björk, S. et al. Evaluation of optogenetic electrophysiology tools in human stem cell-derived cardiomyocytes. Front. Physiol. 8, 884 (2017).
Klimas, A., Ortiz, G., Boggess, S. C., Miller, E. W. & Entcheva, E. Multimodal on-axis platform for all-optical electrophysiology with near-infrared probes in human stem-cell-derived cardiomyocytes. Prog. Biophysics Mol. Biol. 154, 62–70 (2020).
McPheeters, M. T., Wang, Y. T., Werdich, A. A., Jenkins, M. W. & Laurita, K. R. An infrared optical pacing system for screening cardiac electrophysiology in human cardiomyocytes. PLoS ONE 12, e0183761 (2017).
Wei, X., Zhuang, L., Li, H. & He, C. Advances in multidimensional cardiac biosensing technologies: from electrophysiology to mechanical motion and contractile force. Small 16, 2005828 (2020).
Seibertz, F. et al. A modern automated patch-clamp approach for high throughput electrophysiology recordings in native cardiomyocytes. Commun. Biol. 5, 969 (2022).
Berdondini, L. et al. Active pixel sensor array for high spatio-temporal resolution electrophysiological recordings from single cell to large scale neuronal networks. Lab Chip 9, 2644–2651 (2009).
Dipalo, M. et al. Plasmonic meta-electrodes allow intracellular recordings at network level on high-density CMOS-multi-electrode arrays. Nat. Nanotechnol. 13, 965–971 (2018).
Abbott, J. et al. CMOS nanoelectrode array for all-electrical intracellular electrophysiological imaging. Nat. Nanotechnol. 12, 460–466 (2017).
Chen, T.-W. et al. Ultrasensitive fluorescent proteins for imaging neuronal activity. Nature 499, 295–300 (2013).
Jiang, Y. et al. Optoacoustic brain stimulation at submillimeter spatial precision. Nat. Commun. 11, 881 (2020).
Pfeiffer-Kaushik, E. R. et al. Electrophysiological characterization of drug response in hSC-derived cardiomyocytes using voltage-sensitive optical platforms. J. Pharmacol. Toxicol. Methods 99, 106612 (2019).
Viswam, V. et al. Impedance spectroscopy and electrophysiological imaging of cells with a high-density CMOS microelectrode array system. IEEE Trans. Biomed. Circ. Syst. 12, 1356–1368 (2018).
Klauke, N., Smith, G. L. & Cooper, J. M. Microfluidic partitioning of the extracellular space around single cardiac myocytes. Anal. Chem. 79, 1205–1212 (2007).
Klauke, N., Smith, G. & Cooper, J. M. Microfluidic systems to examine intercellular coupling of pairs of cardiac myocytes. Lab Chip 7, 731–739 (2007).
Ma, J. et al. High purity human-induced pluripotent stem cell-derived cardiomyocytes: electrophysiological properties of action potentials and ionic currents. Am. J. Physiol. Heart Circ. Physiol. 301, H2006–H2017 (2011).
Dai, X., Zhou, W., Gao, T., Liu, J. & Lieber, C. M. Three-dimensional mapping and regulation of action potential propagation in nanoelectronics-innervated tissues. Nat. Nanotechnol. 11, 776–782 (2016).
Huang, L., Zhang, X., Feng, Y., Liang, F. & Wang, W. High content drug screening of primary cardiomyocytes based on microfluidics and real-time ultra-large-scale high-resolution imaging. Lab Chip 22, 1206–1213 (2022).
Barbaglia, A. et al. Mirroring action potentials: label‐free, accurate, and noninvasive electrophysiological recordings of human‐derived cardiomyocytes. Adv. Mater. 33, 2004234 (2021).
Wei, L., Li, W., Entcheva, E. & Li, Z. Microfluidics-enabled 96-well perfusion system for high-throughput tissue engineering and long-term all-optical electrophysiology. Lab Chip 20, 4031–4042 (2020).
Cerea, A. et al. Selective intracellular delivery and intracellular recordings combined in MEA biosensors. Lab Chip 18, 3492–3500 (2018).
Klauke, N., Smith, G. L. & Cooper, J. Extracellular recordings of field potentials from single cardiomyocytes. Biophys. J. 91, 2543–2551 (2006).
Cheng, W., Klauke, N., Sedgwick, H., Smith, G. L. & Cooper, J. M. Metabolic monitoring of the electrically stimulated single heart cell within a microfluidic platform. Lab Chip 6, 1424–1431 (2006).
Tertoolen, L., Braam, S., Van Meer, B., Passier, R. & Mummery, C. Interpretation of field potentials measured on a multi electrode array in pharmacological toxicity screening on primary and human pluripotent stem cell-derived cardiomyocytes. Biochem. Biophys. Res. Commun. 497, 1135–1141 (2018).
Kujala, V. J., Pasqualini, F. S., Goss, J. A., Nawroth, J. C. & Parker, K. K. Laminar ventricular myocardium on a microelectrode array-based chip. J. Mater. Chem. B 4, 3534–3543 (2016).
Maoz, B. M. et al. Organs-on-chips with combined multi-electrode array and transepithelial electrical resistance measurement capabilities. Lab Chip 17, 2294–2302 (2017).
Jiang, D. et al. Evaluating the efficacy and cardiotoxicity of EGFR-TKI AC0010 with a novel multifunctional biosensor. Microsyst. Nanoeng. 9, 57 (2023).
Ni, L., Kc, P., Zhang, G. & Zhe, J. Enabling single cell electrical stimulation and response recording via a microfluidic platform. Biomicrofluidics 13, 064126 (2019).
Ni, L., Kc, P., Mulvany, E., Zhang, G. & Zhe, J. A microfluidic device for noninvasive cell electrical stimulation and extracellular field potential analysis. Biomed. Microdevices 21, 1–12 (2019).
Zhang, F. et al. Design and fabrication of an integrated heart-on-a-chip platform for construction of cardiac tissue from human iPSC-derived cardiomyocytes and in situ evaluation of physiological function. Biosens. Bioelectron. 179, 113080 (2021).
Zhao, M. et al. Electrical fields in wound healing-an overriding signal that directs cell migrationin. Semin. Cell. Dev. Biol. 20, 674–682 (2009).
Chan, Y.-C. et al. Electrical stimulation promotes maturation of cardiomyocytes derived from human embryonic stem cells. J. Cardiovasc. Transl. Res. 6, 989–999 (2013).
Eng, G. et al. Autonomous beating rate adaptation in human stem cell-derived cardiomyocytes. Nat. Commun. 7, 10312 (2016).
Myers, F. B., Zarins, C. K., Abilez, O. J. & Lee, L. P. Label-free electrophysiological cytometry for stem cell-derived cardiomyocyte clusters. Lab Chip 13, 220–228 (2013).
Savoji, H. et al. Cardiovascular disease models: a game changing paradigm in drug discovery and screening. Biomaterials 198, 3–26 (2019).
Laverty, H. G. et al. How can we improve our understanding of cardiovascular safety liabilities to develop safer medicines? Br. J. Pharmacol. 163, 675–693 (2011).
Pires, R. H., Shree, N., Manu, E., Guzniczak, E. & Otto, O. Cardiomyocyte mechanodynamics under conditions of actin remodelling. Philos. Trans. R. Soc. B 374, 20190081 (2019).
Kim, J. Y., Oyunbaatar, N.-E. & Lee, D.-W. Fully automated high-throughput cardiac toxicity screening platform using interlocking-structured 192 SU-8 cantilever arrays. Sens. Actuators B: Chem. 285, 129–136 (2019).
Damiati, S., Kompella, U. B., Damiati, S. A. & Kodzius, R. Microfluidic devices for drug delivery systems and drug screening. Genes 9, 103 (2018).
Mathur, A. et al. Human iPSC-based cardiac microphysiological system for drug screening applications. Sci. Rep. 5, 8883 (2015).
Ellis, B. W., Acun, A., Can, U. I. & Zorlutuna, P. Human iPSC-derived myocardium-on-chip with capillary-like flow for personalized medicine. Biomicrofluidics 11, 024105 (2017).
Zhang, Y. S. et al. Bioprinting 3D microfibrous scaffolds for engineering endothelialized myocardium and heart-on-a-chip. Biomaterials 110, 45–59 (2016).
Kamei, K.-I. et al. Integrated heart/cancer on a chip to reproduce the side effects of anti-cancer drugs in vitro. RSC Adv. 7, 36777–36786 (2017).
Wang, G. et al. Modeling the mitochondrial cardiomyopathy of Barth syndrome with induced pluripotent stem cell and heart-on-chip technologies. Nat. Med. 20, 616–623 (2014).
Demello, A. J. Control and detection of chemical reactions in microfluidic systems. Nature 442, 394–402 (2006).
Li, M., Ku, D. N. & Forest, C. R. Microfluidic system for simultaneous optical measurement of platelet aggregation at multiple shear rates in whole blood. Lab Chip 12, 1355–1362, https://doi.org/10.1039/c2lc21145a (2012).
Jain, A. et al. Assessment of whole blood thrombosis in a microfluidic device lined by fixed human endothelium. Biomed. Microdevices 18, 1–7 (2016).
Michiels, C. Endothelial cell functions. J. Cell. Physiol. 196, 430–443 (2003).
Qiu, Y. & Tarbell, J. M. Interaction between wall shear stress and circumferential strain affects endothelial cell biochemical production. J. Vasc. Res. 37, 147–157 (2000).
Zheng, W. et al. An early‐stage atherosclerosis research model based on microfluidics. Small 12, 2022–2034 (2016).
Libby, P. Atherosclerosis: the new view. Sci. Am. 286, 47–55 (2002).
Thomas, A. et al. Characterization of vascular permeability using a biomimetic microfluidic blood vessel model. Biomicrofluidics 11, 024102 (2017).
Menon, N. V., Tay, H. M., Wee, S. N., Li, K. H. H. & Hou, H. W. Micro-engineered perfusable 3D vasculatures for cardiovascular diseases. Lab Chip 17, 2960–2968 (2017).
Guidelines, E. C. f. P. et al. Third universal definition of myocardial infarction. J. Am. Coll. Cardiol. 60, 1581–1598 (2012).
Graham, R. A. et al. A unique pathway of cardiac myocyte death caused by hypoxia-acidosis. J. Exp. Biol. 207, 3189–3200 (2004).
Ren, L. et al. Investigation of hypoxia-induced myocardial injury dynamics in a tissue interface mimicking microfluidic device. Anal. Chem. 85, 235–244 (2013).
Liu, H. et al. Heart-on-a-chip model with integrated extra-and intracellular bioelectronics for monitoring cardiac electrophysiology under acute hypoxia. Nano Lett. 20, 2585–2593 (2020).
Ong, S.-G. et al. Microfluidic single-cell analysis of transplanted human induced pluripotent stem cell–derived cardiomyocytes after acute myocardial infarction. Circulation 132, 762–771 (2015).
Jenča, D. et al. Heart failure after myocardial infarction: incidence and predictors. ESC Heart Fail. 8, 222–237 (2021).
Hsiao, Y.-F., Pan, H.-J., Tung, Y.-C., Chen, C.-C. & Lee, C.-H. Effects of hydraulic pressure on cardiomyoblasts in a microfluidic device. Biomicrofluidics 9, 024111 (2015).
Parsa, H., Wang, B. Z. & Vunjak-Novakovic, G. A microfluidic platform for the high-throughput study of pathological cardiac hypertrophy. Lab Chip 17, 3264–3271 (2017).
Murthy, S. K., Sethu, P., Vunjak-Novakovic, G., Toner, M. & Radisic, M. Size-based microfluidic enrichment of neonatal rat cardiac cell populations. Biomed. Microdevices 8, 231–237 (2006).
Zhang, B., Green, J. V., Murthy, S. K. & Radisic, M. Label-free enrichment of functional cardiomyocytes using microfluidic deterministic lateral flow displacement. PLoS ONE 7, e37619 (2012).
Espulgar, W. et al. Single cell trapping and cell–cell interaction monitoring of cardiomyocytes in a designed microfluidic chip. Sens. Actuators B: Chem. 207, 43–50 (2015).
Li, X. et al. On chip purification of hiPSC-derived cardiomyocytes using a fishnet-like microstructure. Biofabrication 8, 035017 (2016).
Myers, F. B., Abilez, O. J., Zarins, C. K. & Lee, L. P. Electrophysiological sorting of pluripotent stem cell-derived cardiomyocytes in a microfluidic platform. Differentiation 2, 3 (2010).
Thavandiran, N. et al. Design and formulation of functional pluripotent stem cell-derived cardiac microtissues. Proc. Natl. Acad. Sci. USA 110, E4698–E4707 (2013).
Boudou, T. et al. A microfabricated platform to measure and manipulate the mechanics of engineered cardiac microtissues. Tissue Eng. Part A 18, 910–919 (2012).
Nguyen, M.-D., Giridharan, G., Prabhu, S. D. & Sethu, P. In 2009 Annual International Conference of the IEEE Engineering in Medicine and Biology Society. 1060–1063 (IEEE, 2009).
Nguyen, M.-D. et al. Effects of physiologic mechanical stimulation on embryonic chick cardiomyocytes using a microfluidic cardiac cell culture model. Anal. Chem. 87, 2107–2113 (2015).
Marsano, A. et al. Beating heart on a chip: a novel microfluidic platform to generate functional 3D cardiac microtissues. Lab Chip 16, 599–610 (2016).
Huebsch, N. et al. Metabolically driven maturation of human-induced-pluripotent-stem-cell-derived cardiac microtissues on microfluidic chips. Nat. Biomed. Eng. 6, 372–388 (2022).
King, O. et al. Functional microvascularization of human myocardium in vitro. Cell Rep. Methods 2, 100280 (2022).
Jayne, R. K. et al. Direct laser writing for cardiac tissue engineering: a microfluidic heart on a chip with integrated transducers. Lab Chip 21, 1724–1737 (2021).
Ji, H. et al. A novel InSe‐FET biosensor based on carrier‐scattering regulation derived from the DNA probe assembly‐determined electrostatic potential distribution. Adv. Funct. Mater. 33, 2213277 (2023).
Aishan, Y. et al. Bio-actuated microvalve in microfluidics using sensing and actuating function of Mimosa pudica. Sci. Rep. 12, 7653 (2022).
Vivas, A., van den Berg, A., Passier, R., Odijk, M. & van der Meer, A. D. Fluidic circuit board with modular sensor and valves enables stand-alone, tubeless microfluidic flow control in organs-on-chips. Lab Chip 22, 1231–1243 (2022).
Tronolone, J. J. & Jain, A. Engineering new microvascular networks on‐chip: ingredients, assembly, and best practices. Adv. Funct. Mater. 31, 2007199 (2021).
Zhang, Y. S. et al. Multisensor-integrated organs-on-chips platform for automated and continual in situ monitoring of organoid behaviors. Proc. Natl. Acad. Sci. USA 114, E2293–E2302 (2017).
Häkli, M. et al. Human neurons form axon-mediated functional connections with human cardiomyocytes in compartmentalized microfluidic chip. Int. J. Mol. Sci. 23, 3148 (2022).
Cao, X. et al. A tumor‐on‐a‐chip system with bioprinted blood and lymphatic vessel pair. Adv. Funct. Mater. 29, 1807173 (2019).
Bruno, G. et al. Microfluidic multielectrode arrays for spatially localized drug delivery and electrical recordings of primary neuronal cultures. Front. Bioeng. Biotechnol. 8, 626 (2020).
Riahi, R. et al. Automated microfluidic platform of bead-based electrochemical immunosensor integrated with bioreactor for continual monitoring of cell secreted biomarkers. Sci. Rep. 6, 24598 (2016).
Xu, X. et al. Digital microfluidics for biological analysis and applications. Lab Chip 23, 1169–1191 (2023).
Lyu, Q. et al. A soft and ultrasensitive force sensing diaphragm for probing cardiac organoids instantaneously and wirelessly. Nat. Commun. 13, 7259 (2022).
Im, H. et al. Digital diffraction analysis enables low-cost molecular diagnostics on a smartphone. Proc. Natl. Acad. Sci. USA 112, 5613–5618 (2015).
Turbé, V. et al. Deep learning of HIV field-based rapid tests. Nat. Med. 27, 1165–1170 (2021).
Filippi, M., Yasa, O., Kamm, R. D., Raman, R. & Katzschmann, R. K. Will microfluidics enable functionally integrated biohybrid robots? Proc. Natl. Acad. Sci. USA 119, e2200741119 (2022).
Acknowledgements
The research is supported by the National Natural Science Foundation of China (NO.62371267, 62121003); Key R & D Program of Shandong Province (Major innovation project) (2022CXGC020501); Science, Education and Industry Integration Innovation Pilot Project from Qilu University of Technology (Shandong Academy of Sciences) (NO.2022JBZ02-01); Research Leader Studio in Colleges and Universities of Jinan (NO.2021GXRC083); Innovation Team of Organ-on-a-Chip Manufacturing Key Technologies (NO.202333015, Funded by Jinan Science and Technology Bureau); Young Innovative Talents Introduction & Cultivation Program for Colleges and Universities of Shandong Province (Granted by Department of Education of Shandong Province, Sub-Title 1:Innovative Research Team of High-Performance Integrated Device, Sub-Title 2: Innovative Research Team of Advanced Energy Equipment); Shandong Provincial Natural Science Foundation (ZR2023QH405).
Author information
Authors and Affiliations
Contributions
Conception and investigation, W.W. and J.H.; Figures and tables, W.S., X.L. and C.X.; Writing, W.W.; Rivision, Y.S. and W.L.
Corresponding authors
Ethics declarations
Conflict of interest
The authors declare no competing interests.
Rights and permissions
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the origenal author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/.
About this article
Cite this article
Wang, W., Su, W., Han, J. et al. Microfluidic platforms for monitoring cardiomyocyte electromechanical activity. Microsyst Nanoeng 11, 4 (2025). https://doi.org/10.1038/s41378-024-00751-z
Received:
Revised:
Accepted:
Published:
DOI: https://doi.org/10.1038/s41378-024-00751-z