Summary

The Fabrication and Operation of a Continuous Flow, Micro-Electroporation System with Permeabilization Detection

Published: January 07, 2022
doi:

Summary

This protocol describes the microfabrication techniques required to build a lab-on-a-chip, microfluidic electroporation device. The experimental setup performs controlled, single-cell-level transfections in a continuous flow and can be extended to higher throughputs with population-based control. An analysis is provided showcasing the ability to electrically monitor the degree of cell membrane permeabilization in real-time.

Abstract

Current therapeutic innovations, such as CAR-T cell therapy, are heavily reliant on viral-mediated gene delivery. Although efficient, this technique is accompanied by high manufacturing costs, which has brought about an interest in using alternative methods for gene delivery. Electroporation is an electro-physical, non-viral approach for the intracellular delivery of genes and other exogenous materials. Upon the application of an electric field, the cell membrane temporarily allows molecular delivery into the cell. Typically, electroporation is performed on the macroscale to process large numbers of cells. However, this approach requires extensive empirical protocol development, which is costly when working with primary and difficult-to-transfect cell types. Lengthy protocol development, coupled with the requirement of large voltages to achieve sufficient electric-field strengths to permeabilize the cells, has led to the development of micro-scale electroporation devices. These micro-electroporation devices are manufactured using common microfabrication techniques and allow for greater experimental control with the potential to maintain high throughput capabilities. This work builds off a microfluidic-electroporation technology capable of detecting the level of cell membrane permeabilization at a single-cell level under continuous flow. However, this technology was limited to 4 cells processed per second, and thus a new approach for increasing the system throughput is proposed and presented here. This new technique, denoted as cell-population-based feedback control, considers the cell permeabilization response to a variety of electroporation pulsing conditions and determines the best-suited electroporation pulse conditions for the cell type under test. A higher-throughput mode is then used, where this ‘optimal’ pulse is applied to the cell suspension in transit. The steps for fabricating the device, setting up and running the microfluidic experiments, and analyzing the results are presented in detail. Finally, this micro-electroporation technology is demonstrated by delivering a DNA plasmid encoding for green fluorescent protein (GFP) into HEK293 cells.

Introduction

Current therapeutic innovations in biomedical research, such as CAR-T (Chimeric Antigen Receptor Engineered T cell) cell therapy and genetic editing using CRISPR (clustered regularly interspaced short palindromic repeat DNA sequences)/Cas9, heavily rely on the ability to deliver exogenous material both successfully and efficiently into the intracellular space1. In CAR-T therapy, the gold standard to perform the gene delivery step in cell therapy manufacturing is using viral vectors2. Though viral-mediated gene delivery is an efficient delivery modality, it also has several drawbacks. These include manufacturing costs, cytotoxicity, immunogenicity, mutagenesis/tumorigenesis potential, and size limitations on the gene(s) to be delivered3. These limitations have led to the research and development of alternative, non-viral delivery technologies.

Electroporation, an alternative to viral-mediated gene delivery, relies on the application of an optimal electrical pulse waveform to perform DNA, RNA, and protein transfections of cells. Following the application of an external electric field, the cell membrane is briefly compromised, making the cell susceptible to the intracellular delivery of otherwise impermeable exogenous materials4. Compared to viral-mediated delivery, electroporation is advantageous as it is generally safe, easy to operate, and has low operating costs. Electroporation can deliver both small and large molecular cargo and can be efficient in transfecting cells regardless of lineage5. To achieve desirable outcomes following electroporation, i.e., good viability and good electro-transfection efficiency, a variety of experimental parameters need to be co-optimized. These include cell type6, cell density, molecule concentration7, electroporation buffer properties (e.g., molecular composition, conductivity, and osmolarity)8, electrode size/geometry9, and electrical pulse waveform (shape, polarity, number of pulses)10 (refer to Figure 1 for an illustration). Although each of these parameters can have a significant effect on the outcomes of electroporation experiments, pulse waveform has been especially studied in great detail, as the electrical energy of the applied pulse(s) is the root of the intrinsic trade-off between the resulting cell viability and electro-transfection efficiency8.

Typically, electroporation experiments are performed on the macro-scale, where cells are suspended in 100s of microliters of buffer between a set of large, parallel-plate electrodes within an electroporation cuvette. The electrodes are commonly manufactured out of aluminum with an electrode distance of 1-4 mm. Once the cells are manually loaded via pipette, the cuvette is electrically connected to a bulky, electrical pulse generator where the user can set and apply the pulse waveform parameters to electroporate the cell suspension. Although macro-scale or bulk electroporation can process cell densities >106 cells/mL, this feature can be wasteful when optimizing the electrical pulse waveform settings. This is particularly of concern when electroporating primary cell types where the cell population numbers can be limited. Additionally, due to the large distance between the electrodes, the pulse generator must be able to supply large voltages to achieve electric field strengths >1kV/cm11. These high voltages cause resistive power dissipation through the electrolyte buffer resulting in Joule heating, which can be detrimental to the resulting cell viability12. Lastly, performing electroporation on a dense suspension of cells will consistently be burdened with an innate variability in the resulting electro-transfection efficiency and cell viability. Each cell in suspension could experience a different electric field strength due to the surrounding cells. Depending on whether the experienced electric field strength is either increased or decreased, the resulting cell viability or electro-transfection efficiency may each be negatively impacted11. These downsides to macro-scale electroporation have led to the pursuit and development of alternative technologies that operate on the micro-scale and allow for better control at the single-cell level.

The field of BioMEMS, or biomedical micro-electro-mechanical systems, stems from the technological advancements made in the microelectronics industry. Specifically, utilizing microfabrication processes to develop micro-devices for the advancement of biomedical research. These advancements include the development of micro-electrode arrays for in vivo electrical monitoring13, capacitive micro-electrodes for in situ electroporation14, miniaturized organ-on-a-chip devices15, microfluidic point-of-care diagnostics16, biosensors17, and drug delivery systems18, including nano- and micro-electroporation devices19,20,21. Due to the ability to design and manufacture devices at the same size scale as biological cells, nano- and micro-electroporation technologies are advantageous when compared to their macro-scale counterpart22,23. These electroporation devices eliminate the requirement of high voltage pulse applications, as electrode sets with spacings of 10s to 100s of micrometers are typically integrated. This feature drastically reduces the current through the electrolyte, which in turn reduces the accumulation of toxic electrolysis products and the effects of Joule heating in these systems. The micro-scale channels also ensure that a much more uniform electric field is reliably applied to the cells during pulse application, resulting in more consistent outcomes24. In addition, it is also commonplace for micro-electroporation devices to be integrated into a microfluidic platform which lends itself for future integration into a fully automated technology, a highly desirable capability in cell therapy manufacturing25. Lastly, micro-scale electroporation allows for the electrical interrogation of electroporation events. For example, the degree of cell membrane permeabilization can be monitored in real-time at a single cell level26,27. The purpose of this method is to describe the microfabrication, system operation, and analysis of a microfluidic, single-cell micro-electroporation device capable of measuring the degree of cell membrane permeabilization for optimizing electroporation protocols, yet increasing throughput over the previous state-of-the-art.

Performing single-cell level electroporation is no longer a novel technique, as it was first demonstrated by Rubinsky et al. in 2001 with the development of a static cell electroporation technology28. Their micro-device was innovative as they were the first to demonstrate the ability to electrically monitor the event of electroporation. This has further led to the development of static, single-cell electroporation technologies capable of electrically detecting the degree of cell membrane permeabilization in a parallelized manner to increase the throughputs of the devices. However, even with parallelization and batch processing, these devices severely lack the total number of cells they can process per unit time29,30. This limitation has led to the development of flow-through devices capable of performing single-cell level micro-electroporation at much greater throughputs31. This device transition, from static to flow-through environment, limits the capability of electrically monitoring the degree of cell membrane permeabilization following the application of the electroporation pulse. The method described in this work bridges the gap between these two technologies, a micro-electroporation technology capable of electrically detecting, pulsing, and monitoring the degree of cell membrane permeabilization of individual cells, in a continuous-flow, serial fashion.

This technology was recently described in Zheng et al. In that work, the capabilities of this technology were introduced with the completion of a parametric study, where both the amplitude and duration of the electroporation pulse were varied, and the ensuing electrical signal, indicative of cell membrane permeabilization, was explored32. The results showed that an increase in the intensity of the electroporation pulse (i.e., increase in applied electric field or increase in pulse duration) caused an increase in the measured cell membrane permeabilization. To further validate the system, a common fluorescent indicator of successful electroporation, propidium iodide33, was added to the cell suspension, and a fluorescence image was captured immediately following the application of the electrical pulse. The optical signal, i.e., the fluorescence intensity of propidium iodide inside the cell, was strongly correlated with the electrical measurement of the degree of cell membrane permeabilization, verifying the reliability of this electrical measurement. However, this work only considered the delivery of the small molecule propidium iodide, which has little to no translatable significance.

In this work, a new application of this technology is introduced to improve upon the throughput of the system while delivering a biologically active plasmid DNA (pDNA) vector and assessing the electro-transfection efficiency of cells replated and cultured following electroporation. Though the previous work outperforms existing micro-electroporation technologies that are capable of electrically measuring the event of electroporation, the current state of the device still requires long cell transit times between the electrode set (~250 ms) to perform the cell detection, pulse application, and the cell membrane permeabilization measurement. With a single channel, this limits the throughput to 4 cells/s. To combat this limitation, a new concept of cell-population-based feedback-controlled electroporation is introduced to perform pDNA electro-transfection. By using a hypo-physiologic conductivity electroporation buffer, this system allows for the electrical interrogation of single cells across a multitude of electroporation pulse applications. Based on the electrical response, an 'optimal' electroporation pulse is then determined. A 'high-throughput' mode is then implemented where the cell membrane permeabilization determination is nullified, the flow rate is increased, and the electroporation pulse duty cycle is matched to the cell transit time to ensure one pulse per cell in transit between the electrodes. This work will provide extensive details into the microfabrication steps for the manufacturing of the micro-device, the material/equipment and their setup required to perform the experimentation, and the operation/analysis of the device and its electro-transfection efficiency (eTE).

Figure 1
Figure 1: Experimental factors affecting electroporation outcomes. (Left) Cell Suspension-Important factors to consider prior to the onset of electroporation include: Payload (in this case, pDNA), concentration, cell density, and electroporation buffer properties. Electroporation buffer properties to consider are conductivity, osmolarity, and the exact molecular composition contributing to these values. (Middle) Pulse Application-The exact pulse-type (square wave vs. exponential decay) and pulse waveform (single pulse vs. pulse train) must be optimized to maximize both the resulting cell viability and electro-transfection efficiency. Common pulse trains implemented in electroporation processes are typically composed of a series of High Voltage (HV) pulses or series of pulses rotating between HV and Low Voltage (LV) pulse magnitudes. (Right) Cell Recovery-Down-stream processing steps, in particular, the recovery cell culture media that cells are transferred to, should be optimized. Not featured (Far Left), additional upstream cell processing steps can be implemented for overall electroporation process optimization. Please click here to view a larger version of this figure.

Protocol

NOTE: Users should review all MSDS for the materials and supplies used in this protocol. Appropriate PPE should be worn at each step and sterile technique used during experimentation. Sections 1-7 discuss the device fabrication. 1. Device fabrication- Mask design NOTE: Refer to Figure 2 for an illustration of the microfabrication process. The microfabrication steps are to be carried out in a cleanroom environment. Additio…

Representative Results

Figure 4 highlights the operating principles behind the single-cell-level membrane permeabilization detection for a single pulse amplitude. Following the initiation of the electroporation experiment, the cell detection algorithm determines an optimal threshold for cell detection via a point-by-point, slope-based detection method. The system then continuously monitors (1) for a significant negative change in the measured electrical current, which is indicative of the entry of a cell. This is …

Discussion

The methodology presented within this protocol primarily focuses on the microfabrication of a microfluidic device that is then integrated into a specialized electroporation experimental setup. The term 'recipe', which is often used when describing the specifics of the microfabrication process, hints at the importance of following/optimizing each step to successfully fabricate a functioning device. However, certain critical steps within the process, when not optimized, such as UV exposure time/energy, PVD sputteri…

Divulgations

The authors have nothing to disclose.

Acknowledgements

The authors would like to acknowledge financial support by the National Science Foundation (NSF CBET 0967598, DBI IDBR 1353918) and the U.S. Department of Education's Graduate Training in Emerging Areas of Precision and Personalized Medicine (P200A150131) for funding graduate student J.J.S. on fellowship.

Materials

150-mm diameter petri dishes VWR 25384-326 step 6.1.1 to secure wafer
24-well tissue culture plates VWR 10062-896 step 10.3.6 to plate electroporated cells
33220A Waveform/Function generator Agilent step 9.2.3 electroporation pulse generator
4'' Si-wafers University Wafer subsection 2.1 for microfluidic channel fabrication
6-well tissue culture plates VWR 10062-892 step 8.1.8 to plate cells
Acetone Fisher Scientific A18-4 step 2.1.2 for cleaning and  step 5.1 photoresist lift-off
Allegra X-22R Centrifuge Beckman Coulter steps 8.1.4 , 8.3.2. and 8.3.3. to spin down cells
AutoCAD 2018 Autodesk subsection 1.1. to design transparency masks
Buffered oxide etchant 10:1 VWR 901621-1L subsection 3.1 for HF etching
CCD Monochrome microscope camera Hamamatsu Orca 285 C4742-96-12G04 step 11.2.3. for imaging
CMOS camera- Sensicam QE 1.4MP PCO subsection 9.3 part of the experimental setup
Conductive Epoxy CircuitWorks CW2400 subsection 7.6. for wire attachement
Conical Centrifuge Tubes, 15 mL Fisher Scientific 14-959-70C step 8.1.4. for cell centrifuging
Dektak 3ST Surface Profilometer Veeco (Sloan/Dektak) step 2.1.15 and 5.4 for surface profilometry
Disposable biopsy punch, 0.75 mm Robbins Instruments RBP075 step 6.2.3 for inlet access
Disposable biopsy punch, 3 mm Robbins Instruments RBP30P step 6.2.3 for outlet access
DRAQ5 abcam ab108410 step 11.2.2. for live cell staining
Dulbecco’s Modified Eagle’s Medium ThermoFisher Scientific 11885084 step 8.1.2. part of media composition
E3631A Bipolar Triple DC power supply Agilent step 9.2.1.-9.2.2.part of the experimental setup
Eclipse TE2000-U Inverted  Microscope Nikon  subsection 9.3. part of the experimental setup
EVG620 UV Lithography System EVG  step 2.1.9. and 2.2.7. for UV Exposure
Fetal Bovine Serum Neuromics FBS001 step 8.1.2. part of media composition
FS20 Ultrasonic Cleaner Fisher Scientific subsection 5.1. for photoresist lift-off
Glass Media Bottle with Cap, 100mL Fisher Scientific FB800100 step 8.2.1. for buffer storage
Glass Media Bottle with Cap, 500mL Fisher Scientific FB800500 step 8.1.2.for media storage
HEK-293 cell line ATCC CRL-1573 subsection 8.1 for cell culturing
HEPES buffer solution Sigma Aldrich 83264-100ML-F step 8.2.1 part of electroporation buffer composition
Hexamethyldisilazane Sigma Aldrich 379212-25ML step 2.2.3 adhesion promoter
HF2LI Lock-in Amplifier Zurich Instruments subsection 9.2 part of the experimental setup
HF2TA Current amplifier Zurich Instruments subsection 9.2 part of the experimental setup
Isopropyl Alcohol Fisher Scientific A459-1 step 2.1.2 for cleaning, step 2.1.14 for rinsing wafer following SU-8 development, and step 6.3.1 for cleaning PDMS
IX81 fluorescence microscope Olympus step 11.2.3 for imaging
L-Glutamine Solution Sigma Aldrich G7513-20ML step 8.1.2. part of media composition
M16878/1BFA 22 gauge wire AWC B22-1 subsection 7.5 for device fabrication
Magnesium chloride Sigma Aldrich 208337-100G step 8.1.2 part of electroporation buffer composition
MF 319 Developer Kayaku Advanced Materials 10018042 step 2.2.9. photoresist developer
Microposit S1818 photoresist Kayaku Advanced Materials 1136925 step 2.2.4 positive photoresist for electrode patterning
Microscope slides, 75 x 25 mm VWR 16004-422 step 2.2.1 electrode soda lime glass substrate
Model 2350 High voltage amplifier TEGAM 2350 step 9.2.5. part of the experimental setup
National Instruments LabVIEW National Instruments data acquisition
Needle, 30G x 1 in BD Scientific 305128 step 10.1.1. part of the system priming
PA90 IC OPAMP Power circuit Digi-key 598-1330-ND Part of the custom circuit
Penicillin-Streptomycin Sigma Aldrich P4458-20ML step 8.1.2. part of media composition
Plasmid pMAX-GFP Lonza VCA-1003 step 8.3.4. for intracellular delivery
Plastic tubing, 0.010'' x 0.030" VWR 89404-300 step 10.1.2. for system priming
Platinum targets Kurt J. Lesker subsection 4.2. for physical vapor deposition
Potassium chloride Sigma Aldrich P9333-500G step 8.2.1. part of electroporation buffer composition
Pump 11 PicoPlus microfluidic syringe pump Harvard Apparatus MA1 70-2213 step 10.1.4. for system priming
PVD75 Physical vapor deposition system Kurt J. Lesker subsection 4.1. for physical vapor deposition
PWM32 Spinner System Headway Research steps 2.1.6 and 2.2.2. for substrate coating with photoresist
PX-250 Plasma treatment system March Instruments subsection 7.2 for PDMS and glass substrate bonding
SDG1025 Function/Waveform generator Siglent step 9.2.2. part of the experimental setup
Sodium hydroxide Sigma Aldrich S8045-500G step 8.2.1. part of electroporation buffer composition
SU-8 2010 negative photoresist Kayaku Advanced Materials Y111053 step 2.1.7. for microfluidic channel patterning
SU-8 developer Microchem Y010200 step 2.1.12. for photoresist developing
Sucrose Sigma Aldrich S7903-1KG step 8.2.1. part of electroporation buffer composition
Sylgard 184 elastomer kit Dow Corning 3097358-1004 step 6.2.1  10 : 1 mixture of PDMS polymer and hardening agent
Syringe, 1 ml BD Scientific 309628 step 8.3.4. part of system priming
SZ61 Stereomicroscope System Olympus subsection 7.3. for channel and electrode alignment
Tissue Culture Treated T25 Flasks Falcon 353108 step 8.1.2 for cell culturing
Titanium targets Kurt J. Lesker subsection 4.2. for physical vapor deposition
Transparency masks CAD/ART Services steps 2.1.9. and 2.2.7. for photolithography
Trichloro(1H,1H,2H,2H-perfluorooctyl)silane Sigma Aldrich 448931-10G step 6.1.2. for wafer silanization
Trypsin-EDTA solution Sigma Aldrich T4049-100ML steps 8.1.3. and 8.3.1. for cell harvesting

References

  1. Gao, Q. Q., et al. Therapeutic potential of CRISPR/Cas9 gene editing in engineered T-cell therapy. Cancer Medicine. 8 (9), 4254-4264 (2019).
  2. Aijaz, A., et al. Biomanufacturing for clinically advanced cell therapies. Nature Biomedical Engineering. 2 (6), 362-376 (2018).
  3. Milone, M. C., O’Doherty, U. Clinical use of lentiviral vectors. Leukemia. 32 (7), 1529-1541 (2018).
  4. Weaver, J. C., Chizmadzhev, Y. A. Theory of electroporation: A review. Bioelectrochemistry and Bioenergetics. 41 (2), 135-160 (1996).
  5. Kotnik, T., Rems, L., Tarek, M., Miklavcic, D. Membrane electroporation and electropermeabilization: mechanisms and models. Annual Review of Biophysics. 48, 63-91 (2019).
  6. Rosazza, C., Meglic, S. H., Zumbusch, A., Rols, M. P., Miklavcic, D. Gene electrotransfer: A mechanistic perspective. Current Gene Therapy. 16 (2), 98-129 (2016).
  7. Clauss, J., et al. Efficient non-viral T-cell engineering by sleeping beauty minicircles diminishing DNA toxicity and miRNAs silencing the endogenous T-cell receptors. Human Gene Therapy. 29 (5), 569-584 (2018).
  8. Sherba, J. J., et al. The effects of electroporation buffer composition on cell viability and electro-transfection efficiency. Scientific Reports. 10 (1), 3053 (2020).
  9. Lu, H., Schmidt, M. A., Jensen, K. F. A microfluidic electroporation device for cell lysis. Lab on a Chip. 5 (1), 23-29 (2005).
  10. Kar, S., et al. Single-cell electroporation: current trends, applications and future prospects. Journal of Micromechanics and Microengineering. 28 (12), (2018).
  11. Shi, J. F., et al. A review on electroporation-based intracellular delivery. Molecules. 23 (11), (2018).
  12. Wang, S. N., Zhang, X. L., Wang, W. X., Lee, L. J. Semicontinuous flow electroporation chip for high-throughput transfection on mammalian cells. Analytical Chemistry. 81 (11), 4414-4421 (2009).
  13. Wei, W. J., et al. An implantable microelectrode array for simultaneous L-glutamate and electrophysiological recordings in vivo. Microsystems & Nanoengineering. 1, (2015).
  14. Maschietto, M., Dal Maschio, M., Girardi, S., Vassanelli, S. In situ electroporation of mammalian cells through SiO2 thin film capacitive microelectrodes. Scientific Reports. 11 (1), (2021).
  15. Wu, Q. R., et al. Organ-on-a-chip: recent breakthroughs and future prospects. Biomedical Engineering Online. 19 (1), (2020).
  16. Pandey, C. M., et al. Microfluidics Based Point-of-Care Diagnostics. Biotechnology Journal. 13 (1), (2018).
  17. Vigneshvar, S., Sudhakumari, C. C., Senthilkumaran, B., Prakash, H. Recent advances in biosensor technology for potential applications – An overview. Frontiers in Bioengineering and Biotechnology. 4, (2016).
  18. Nuxoll, E. BioMEMS in drug delivery. Advanced Drug Delivery Reviews. 65 (11-12), 1611-1625 (2013).
  19. Kang, S., Kim, K. H., Kim, Y. C. A novel electroporation system for efficient molecular delivery into Chlamydomonas reinhardtii with a 3-dimensional microelectrode. Scientific Reports. 5, (2015).
  20. Zheng, M. D., Shan, J. W., Lin, H., Shreiber, D. I., Zahn, J. D. Hydrodynamically controlled cell rotation in an electroporation microchip to circumferentially deliver molecules into single cells. Microfluidics and Nanofluidics. 20 (1), (2016).
  21. Santra, T. S., Kar, S., Chang, H. Y., Tseng, F. G. Nano-localized single-cell nano-electroporation. Lab on a Chip. 20 (22), 4194-4204 (2020).
  22. Lee, W. G., Demirci, U., Khademhosseini, A. Microscale electroporation: challenges and perspectives for clinical applications. Integrative Biology. 1 (3), 242-251 (2009).
  23. Santra, T. S., Chang, H. Y., Wang, P. C., Tseng, F. G. Impact of pulse duration on localized single-cell nano-electroporation. Analyst. 139 (23), 6249-6258 (2014).
  24. Geng, T., Lu, C. Microfluidic electroporation for cellular analysis and delivery. Lab on a Chip. 13 (19), 3803-3821 (2013).
  25. Hsi, P., et al. Acoustophoretic rapid media exchange and continuous-flow electrotransfection of primary human T cells for applications in automated cellular therapy manufacturing. Lab on a Chip. 19 (18), 2978-2992 (2019).
  26. Khine, M., Ionescu-Zanetti, C., Blatz, A., Wang, L. P., Lee, L. P. Single-cell electroporation arrays with real-time monitoring and feedback control. Lab on a Chip. 7 (4), 457-462 (2007).
  27. Ye, Y. F., et al. Single-cell electroporation and real-time electrical monitoring on a microfluidic chip. 2020 33rd Ieee International Conference on Micro Electro Mechanical Systems (Mems 2020). , 1040-1043 (2020).
  28. Huang, Y., Rubinsky, B. Microfabricated electroporation chip for single cell membrane permeabilization. Sensors and Actuators a-Physical. 89 (3), 242-249 (2001).
  29. Guo, X. L., Zhu, R. Controllable in-situ cell electroporation with cell positioning and impedance monitoring using micro electrode array. Scientific Reports. 6, (2016).
  30. Punjiya, M., Nejad, H. R., Mathews, J., Levin, M., Sonkusale, S. A flow through device for simultaneous dielectrophoretic cell trapping and AC electroporation. Scientific Reports. 9, (2019).
  31. Wang, H. Y., Lu, C. Microfluidic electroporation for delivery of small molecules and genes into cells using a common DC power supply. Biotechnology and Bioengineering. 100 (3), 579-586 (2008).
  32. Zheng, M. D., et al. Continuous-flow, electrically-triggered, single cell-level electroporation. Technology. 5 (1), 31-41 (2017).
  33. Batista Napotnik, T., Miklavcic, D. In vitro electroporation detection methods – An overview. Bioelectrochemistry. 120, 166-182 (2018).
  34. MICROPOSIT™ S1800® G2 Series Photoresists. KAYAKU Available from: https://kayakuam.com/wp-content/uploads/2019/09/S1800-G2.pdf (2021)
  35. SU-8 2000 Permanent Negative Epoxy Photoresist. KAYAKU Available from: https://kayakuam.com/wp-content/uploads/2020/08/KAM-SU-8-2000-2000.5-2015-Datasheet-8.13.20-final.pdf (2001)
  36. Substrate Preparation. MicroChemicals Available from: https://www.microchemicals.com/technical_information/subtrate_cleaning_adhesion_photoresist.pdf (2021)
  37. Lisinenkova, M., Hahn, L., Schulz, J. . 4M 2006 – Second International Conference on Multi-Material Micro Manufacture. , 91-94 (2006).
  38. Beh, C. W., Zhou, W. Z., Wang, T. H. PDMS-glass bonding using grafted polymeric adhesive – alternative process flow for compatibility with patterned biological molecules. Lab on a Chip. 12 (20), 4120-4127 (2012).
  39. PA90 High Voltage Power Operational Amplifiers. APEX Available from: https://www.apexanalog.com/resources/products/pa90u.pdf (2021)
  40. Lissandrello, C. A., et al. High-throughput continuous-flow microfluidic electroporation of mRNA into primary human T cells for applications in cellular therapy manufacturing. Scientific Reports. 10 (1), 18045 (2020).

Play Video

Citer Cet Article
Sherba, J. J., Atzampou, M., Lin, H., Shan, J. W., Shreiber, D. I., Zahn, J. D. The Fabrication and Operation of a Continuous Flow, Micro-Electroporation System with Permeabilization Detection. J. Vis. Exp. (179), e63103, doi:10.3791/63103 (2022).

View Video