US20160305927A1 - Patterned cardiomyocyte culture on microelectrode array - Google Patents

Patterned cardiomyocyte culture on microelectrode array Download PDF

Info

Publication number
US20160305927A1
US20160305927A1 US15/190,958 US201615190958A US2016305927A1 US 20160305927 A1 US20160305927 A1 US 20160305927A1 US 201615190958 A US201615190958 A US 201615190958A US 2016305927 A1 US2016305927 A1 US 2016305927A1
Authority
US
United States
Prior art keywords
culture
cardiomyocytes
cardiac
patterned
multielectrode array
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US15/190,958
Inventor
Peter Molnar
Anupama Natarajan
Maria Stancescu
James Hickman
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
University of Central Florida Research Foundation Inc UCFRF
Original Assignee
University of Central Florida Research Foundation Inc UCFRF
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by University of Central Florida Research Foundation Inc UCFRF filed Critical University of Central Florida Research Foundation Inc UCFRF
Priority to US15/190,958 priority Critical patent/US20160305927A1/en
Assigned to UNIVERSITY OF CENTRAL FLORIDA RESEARCH FOUNDATION, INC. reassignment UNIVERSITY OF CENTRAL FLORIDA RESEARCH FOUNDATION, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: STANCESCU, MARIA, NATARAJAN, ANUPAMA, HICKMAN, JAMES, MOLNAR, PETER
Publication of US20160305927A1 publication Critical patent/US20160305927A1/en
Assigned to NATIONAL INSTITUTES OF HEALTH (NIH), U.S. DEPT. OF HEALTH AND HUMAN SERVICES (DHHS), U.S. GOVERNMENT reassignment NATIONAL INSTITUTES OF HEALTH (NIH), U.S. DEPT. OF HEALTH AND HUMAN SERVICES (DHHS), U.S. GOVERNMENT CONFIRMATORY LICENSE (SEE DOCUMENT FOR DETAILS). Assignors: UNIVERSITY OF CENTRAL FLORIDA
Abandoned legal-status Critical Current

Links

Images

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/483Physical analysis of biological material
    • G01N33/4833Physical analysis of biological material of solid biological material, e.g. tissue samples, cell cultures
    • G01N33/4836Physical analysis of biological material of solid biological material, e.g. tissue samples, cell cultures using multielectrode arrays
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0652Cells of skeletal and connective tissues; Mesenchyme
    • C12N5/0657Cardiomyocytes; Heart cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/02Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving viable microorganisms
    • C12Q1/04Determining presence or kind of microorganism; Use of selective media for testing antibiotics or bacteriocides; Compositions containing a chemical indicator therefor
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • G01N33/5044Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics involving specific cell types
    • G01N33/5061Muscle cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2533/00Supports or coatings for cell culture, characterised by material
    • C12N2533/30Synthetic polymers
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2535/00Supports or coatings for cell culture characterised by topography
    • C12N2535/10Patterned coating
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/02Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving viable microorganisms
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/483Physical analysis of biological material
    • G01N33/487Physical analysis of biological material of liquid biological material
    • G01N33/48707Physical analysis of biological material of liquid biological material by electrical means
    • G01N33/48728Investigating individual cells, e.g. by patch clamp, voltage clamp

Definitions

  • the present invention relates to the field of new drugs and, more particularly, to a patterned culture of cardiomyocytes formed on a multielectrode array and useful for in vitro testing for possible cardiac side effects by a new drug.
  • VF ventricular fibrillation
  • Arrhythmia is a known side effect of commercial drugs.
  • One of the mechanisms by which drugs can cause a potentially fatal form of ventricular tachy arrhythmia, called Torsades de pointes (Tdp) is through the prolongation of the QT interval (in an ECG the length of the ventricular action potential). It has been reported that approximately 2-3% of all prescribed drugs can cause long QT syndrome[6, 7].
  • a broad range of cardiovascular drugs and antibiotics also have the potential risk of causing drug induced Tdp[8, 9].
  • prolongation of the QT interval does not necessarily lead to Tdp; lengthening of the QT interval could even be antiarrhythmogenic, as it is considered a mechanism of action of the class III anti-arrhythmics[8, 9].
  • a relatively high-throughput method to identify cardiac side effects and differentiate between arrhythmic and anti-arrhythmic effects at an early stage of drug development would have a significant impact on the field.
  • Gap junctions play an important role in the propagation of excitation in cardiac tissue. Changes in gap junction function affect major cardiac parameters, such as conduction velocity (CV). It has been observed in several cardiovascular diseases that the expression of connexins (protein molecules that form gap junction channels) is decreased or their distribution is changed, leading to a malfunction in gap junction coupling[10]. Understanding the pharmacological modulation of cardiac gap junction channels would further aid the drug development enterprise.
  • CV conduction velocity
  • in vitro testing would have a significant impact on drug development as it could also reduce the cost, time and the number of drugs failing in clinical trials[11].
  • in vitro testing would also reduce the need for animal testing and could be used to study drug effects with a functional assay, but at the cellular level.
  • Other in vitro methods such as whole heart experiments (Langendorff heart model) or the Purkinje fiber preparation are difficult and time consuming[11].
  • Traditional methods used to study QT interval prolongation at the cellular level include patch-clamp experiments. However, these experiments are time intensive, require a skilled operator and cannot be used to study action potential (AP) propagation or parameters such as CV and re-entry.
  • AP action potential
  • Cardiac myocytes cultured on microelectrode arrays have several benefits compared to either traditional patch clamp electrophysiology or isolated organ methods.
  • the use of MEAs in the investigation of cardiac side effects would provide information in a relatively high-throughput and low cost manner compared to standard patch-clamp electrophysiology.
  • Cardiac myocytes on MEAs have been used in a number of studies to investigate the effect of toxins, such as pesticides[12] and cardioactive drugs[13] on cardiac field potentials.
  • a commercial system has also been introduced to measure QT intervals in a relatively high-throughput fashion[14], but, to date, it has only limited applications.
  • cardiac myocytes can now be maintained over longer periods of time[15], thus chronic experiments, such as the monitoring of network remodeling for specific diseases, is now feasible.
  • serum-free formulations for cardiac culture have also been introduced, which would increase the reproducibility of such a system[16].
  • One category of this technique is based on direct placement of cells or extracellular matrix molecules on desired locations and includes patterning through microfluidic channels[21-23], microcontact printing[24, 25] and inkjet printing[26]. Cardiac myocytes have previously been patterned on glass using photoresist[27] as well as other techniques[15, 17, 19, 20, 25]. Another method utilized photolithography following surface modification with self-assembled monolayers (SAMs) for neurons[28-30] as well as myocytes[15, 31]. The benefit of this method is the compatibility of the technique with cheap automated silicon manufacturing steps and the ability of the cells to self-assemble after random plating.
  • SAMs self-assembled monolayers
  • SAMs are one molecule thick monolayers attached to a surface composed of organic molecules, which have been extensively used for surface patterning[31-33]. Surface modification with SAMs is also compatible with advanced photolithography methods[30, 34]. Studies have also shown that cells survive on these surfaces for extended periods of time[35, 36], do not migrate off the patterned areas[34] and exhibit the typical morphology and physiology of the specific cell type[16, 37].
  • the goal of this study was the development of patterned, rat, cardiomyocyte cultures on MEAs in a serum-free medium for the study of cardiac physiology and pharmacology utilizing a high-throughput technique, but with high information content.
  • An adsorbed fibronectin layer was used as the foreground because it supported cardiac myocyte attachment and growth and a 2-[Methoxy(Polyethyleneoxy) Propyl]TrimethoxySilane (SiPEG) SAM was used as the cell repellent background because of its excellent protein adsorption resistant properties[38].
  • the measurement of CV with the patterned cardiac myocyte monolayers and the feasibility to apply different stimulation protocols to the MEA/cardiac system was demonstrated.
  • the action of 1-Heptanol and Sparfloxacin was also assessed. This method could easily be adapted for use with human cardiac myocytes to eliminate interspecies differences in drug side effect screening and could become an alternative to the existing more complex, expensive and time consuming methods.
  • the present invention advantageously provides a culture of patterned cardiomyocytes.
  • the culture comprises a support substrate bearing a multielectrode array and a negative surface resistant to cell attachment and deposited on the support substrate covering the multi-electrode array.
  • the negative surface bears a pattern ablated thereon, preferably by laser photolithography.
  • a positive surface promoting cell attachment is deposited on the pattern ablated on the negative surface and cardiomyocytes adherent to the positive surface and growing are aligned along the pattern.
  • the negative surface comprises a material selected from the group consisting of polyethylene glycol, polyacrylic acid and polyacrylamide. Most preferably, the material selected is polyethylene glycol and it is deposited essentially as a monolayer.
  • the pattern on the negative surface is preferably ablated by laser lithography through a mask, as further explained below.
  • the positive surface preferably comprises a material selected from the group consisting of fibronectin, trimethoxysilylpropyldiethylenetriamine and pure glass.
  • the cardiomyocytes are preferably of mammalian origin, and may be of neonatal rat. Additionally, however, the cardiomyocytes may be derived from human embryonic stem cells.
  • the invention further includes a method of making the described culture of patterned myocardiocytes.
  • the method comprises preparing a support substrate bearing a multi-electrode array, overlaying on the support substrate a negative surface resistant to cell adherence thereon, comprising polyethylene glycol and covering the multi-electrode array, ablating a pattern on the negative surface, depositing on the ablated pattern a positive surface promoting cell adherence thereon and including fibronectin, adhering cardiomyocytes on the positive surface, and culturing the cardiomyocytes so that they grow on the positive patterned surface and aligned therewith.
  • the invention also includes an in vitro method of testing the effect of a compound on cardiac function.
  • This method comprises providing patterned spontaneously beating cardiomyocytes according to the described culture. Recording a first electrical output from the cardiomyocytes. Contacting the cardiomyocytes with the compound being tested, Recording a second electrical output from the cardiomyocytes after being contacting with the compound. Finally, determining a change between the first and second electrical outputs as an indicator of altered cardiac function.
  • FIG. 1 Patterned neonatal cardiomyocytes.
  • A Pattern design and dimensions.
  • B, C Phase contrast pictures on day 12 of different regions of the pattern indicated in A. Field potentials measured at individual electrodes are marked in red in B. Electrode distance: 200 ⁇ m, D: Immunostaining with Rhodamine phalloidin for F-actin indicating cardiac myofibrils (note the visible striations). Nuclei are stained with DAPI (blue).
  • FIG. 2 Action potential (AP) conduction in patterned cardiac myocyte monolayers.
  • A phase contrast picture of cardiac myocyte patterns on the top of the substrate embedded electrodes (electrode distance is 200 ⁇ m) with a sketch of the conduction pathway on the completed pattern and with the recorded extracellular signals on the given electrodes.
  • B Overlay of the recorded traces showing the temporal relationship of the signals.
  • a spontaneous AP was generated in the cardiac myocyte monolayer in the right fork of the pattern, close to electrode 74 . This AP spread through the pattern reaching electrode 74 first, electrode 72 second and with a much longer delay (due to the longer path) electrode 52 last.
  • Conduction velocity (CV) was determined based on the time delay in the signal between electrode 74 and 52 and the known pattern geometry. The calculated CV in this experiment was 0.190.
  • FIG. 3 Measurement of conduction velocity in patterned cardiac myocyte monolayers with electrical stimulation.
  • FIG. 4 Measurement of refractory period after cardiac action potential. Electrodes 63 and 85 (see pattern geometries on the left) were stimulated alternatively according to the following protocol: E 63 , 100 ms delay, E 85 , 400 ms delay, E 63 again. Right panel shows recording at electrode 87 . Stimulating E 85 100 ms after the stimulation of E 63 failed to evoke an AP because this delay was too short, cells were stimulated in their refractory period. By chance, cell electrode coupling on electrode E 85 was especially strong in this experiment, thus the extracellular recording approximately replicated the shape of the intracellular AP illustrating the relationship of the AP and the stimulation. When the stimulation delay between E 63 and E 85 was 250 ms, both stimulations evoked an action potential at electrode 87 .
  • FIG. 5 Effect of the gap junction blocker 1-Heptanol.
  • A Propagation of excitation between the two end points in the pattern (Electrodes 84 and 52 ) before addition of the drug.
  • the CV was measured to be 0.197 m/sec
  • B Propagation of excitation after the drug.
  • the CV was calculated as 0.0066 m/sec.
  • the drug effect on the CV measured over 4 MEAs.
  • FIG. 6 Effect of the HERG channel antagonist Sparfloxacin.
  • A Field potential recordings before addition of Sparfloxacin indicated stable beating frequencies and synchronous activity
  • B Field potential recording after the addition of 2 ⁇ M Sparfloxacin showed burst like activity with higher intra-burst frequencies and no synchrony between electrodes.
  • any temperature, weight, volume, time interval, pH, salinity, molarity or molality, range, concentration and any other measurements, quantities or numerical expressions given herein are intended to be approximate and not exact or critical figures unless expressly stated to the contrary. Accordingly, where appropriate to the invention and as understood by those of skill in the art, it is proper to describe the various aspects of the invention using approximate or relative terms and terms of degree commonly employed in the art, for example, so dimensioned, about, approximately, substantially, essentially, consisting essentially of, comprising, and effective amount.
  • the arrays were then rinsed once in toluene, twice in ethanol, twice in water and sonicated in water for 2 minutes to remove the non-covalently linked material[39].
  • the arrays were air dried with nitrogen and stored in a dessicator overnight.
  • the MEAs were patterned by laser ablation photolithography using a deep UV (193 nm) excimer laser (Lambda Physik) at a pulse power of 230 mW and a frequency of 10 Hz for 45 seconds through a quartz photomask (Bandwidth foundry, Eveleigh, Australia).
  • the arrays were sterilized using 70% isopropanol and then incubated with 5 ⁇ g/ml of fibronectin in a Phosphate buffered solution (Invitrogen) for 20 minutes at room temperature. The solution was removed and the surface was first rinsed with PBS, followed by the plating medium, and then dried before the cells were plated.
  • the neonatal rat cardiomyocyte culture was prepared using the cardiac isolation kit from Worthington[40]. All animal work was approved by the UCF IACUC and followed NIH guidelines. Briefly, two day-old rat pups were euthanized in a precharged CO 2 chamber. Hearts were dissected and minced in ice cold Hanks balanced salt solution (HBSS). Cardiac myocytes were dissociated by incubation of the hearts in trypsin (100 ⁇ g/ml in HBSS) for 16 hours at 2-8° C. The hearts in the trypsin solution were briefly warmed with a trypsin inhibitor before adding collegenase (300 units/ml in L-15 medium) for 45 minutes in a water bath at 37° C.
  • HBSS Hanks balanced salt solution
  • the cell solution was filtered to remove any remaining tissue and centrifuged at 50 g for 5 minutes at 22° C.
  • the cells were resuspended in high glucose Dulbecco's modified eagle medium (DMEM, Gibco/Invitrogen) supplemented with 10 ml Fetal Bovine Serum (Gibco/Invitrogen) and 1 ml penicillin streptomycin (Gibco/Invitrogen), preplated in Petri dishes and incubated at 37° C. and in 5% CO 2 for 45 minutes. This was necessary to eliminate the fibroblasts. The supernatant from the Petri dishes was centrifuged at 50 g for 5 min at 22° C.
  • DMEM high glucose Dulbecco's modified eagle medium
  • penicillin streptomycin Gibco/Invitrogen
  • the cells were then resuspended in the plating medium.
  • the serum-free plating medium consisted of: 100 ml Ultraculture medium (Bio Whittaker Cambrex) supplemented with 10 ml 827, 1 ml L-glutamine (Gibco/Invitrogen), 1 ml Penicillin Streptomycin, 0.375 g dextrose (Fisher Scientific) in 800 ⁇ l water, 1 ml non-essential amino acids and 1 ml of Hepes buffer (Gibco/Invitrogen)[41]. Additional growth factors were also added to improve cell survival in the serum-free conditions.
  • Patterned Cardiomyocytes were immunostained for F-Actin with Rhodamine Phalloidin (Invitrogen, R415), using a protocol provided by the company. Briefly, the cells were washed with PBS and fixed using 3% Formaldehyde. The coverslips were extracted with 0.1 ml Triton X®. The staining solution (with 1% Bovine Serum Albumin to prevent background staining) was added at a dilution of 1:40 in PBS and coverslips were incubated for 30 minutes. Imaging was done using confocal microscopy.
  • the cardiac myocytes were cultured on patterned metal MEAs (Planar 10 ⁇ m electrodes, 200 ⁇ m separation, Multichannel-systems).
  • MEA1040 Multichannel-systems
  • STG 1002 Multichannel systems
  • the cells were stimulated utilizing 500 mV, 1 ms wide bipolar pulses at 2 Hz.
  • the recording medium was the same as the plating medium with the pH adjusted to 7.3 using HEPES buffer. After a 30 minute incubation period, APs were detected and recorded using built in functions of the Multichannel System software.
  • a cell resistant background surface was needed which was also resistant to protein adsorption as this would allow fibronectin incubation on the foreground and prevent its adherence to the background, enabling cardiac myocyte attachment.
  • poly(ethylene glycol) (PEG) was chosen for the background because of its excellent protein adsorption resistance.
  • XPS X-ray Photoelectron Spectroscopy
  • contact angle measurements were used to analyze the results of the surface modifications and throughout the entire study for quality assurance purposes.
  • the growth and activity of neonatal rat cardiomyocytes on the patterned MEAs were assessed for over 2 weeks.
  • the cells attached to the fibronectin, but not to the PEG background and showed clearly delineated regions ( FIG. 1 ).
  • the cells formed monolayers by day 2 and spontaneous contraction and beating activity began on day 4 and was consistent throughout the pattern.
  • Cell survival and activity improved markedly with the addition of L-thyroxine, Epidermal growth factor and Hydrocortisone to the culture medium as indicated in the Methods.
  • L-thyroxine L-thyroxine
  • Epidermal growth factor Epidermal growth factor
  • Hydrocortisone Hydrocortisone
  • the electrical activity of the spontaneously contracting cells patterned on the microelectrodes was extracellularly recorded using the MEA system.
  • Cardiac myocytes formed a morphologically homogenous, aligned, integrated network of cells that communicated through gap junctions and displayed normal tissue electrophysiology.
  • the recorded field potential (FP) signals correlated with the contraction cycle.
  • FIG. 2 shows a typical recording and the method for the determination of propagation direction.
  • FIG. 213 emphasizes the delay between FPs at different electrodes on the pattern, which enabled the determination of CV with high temporal resolution.
  • the signal propagation was also visualized using an Imaging software routine written in Matlab 6.5[47].
  • the program converted recordings from the electrodes into a video indicating the field potential movement across an 8 ⁇ 8 grid. Snapshots from the video are shown in the supplementary data. This program also verified the origin of the excitation wave and confirmed our CV results obtained using the earlier method ( FIG. 2 .).
  • the conduction velocity was calculated to be 0.190 ⁇ 0.025 m/s for spontaneous firing of the patterned cardiomyocytes over eight different MEAs. Previous research has shown that in vitro conduction velocities ranged from 0.12 m/s[48] to 0.242 m/s[49]. In a computer simulation of AP propagation in cardiac fibers in realistic conditions (taking into account extracellular solutions and spaces) the conduction velocity was 0.504 m/s[50]. In the human heart the AP generated by the Sino-atrial node spreads through the atria at a conduction velocity of 0.5 m/sec[51].
  • the conduction velocity on patterned cardiac monoloyers was also measured with stimulation.
  • the cardiac pattern was stimulated at the two ends of the pattern, over a distance of 0.77 mm with 1 ms wide bipolar pulses with an amplitude of 500 mV at a frequency of 2 to 4 Hz ( FIG. 3 ).
  • Previous research has indicated that rapid electrical stimulation in cultured, neonatal rat cardiomyocytes had an effect on the expression of Connexin [43] leading to changes in conduction properties[52], including an increase in conduction velocity.
  • the stimulation protocol was optimized by determining a minimal threshold which reliably generated contraction from the cells.
  • the cardiomyocytes were stimulated with 2 Hz, 500 mV, 1 ms wide bidirectional pulses at different electrodes. As seen in FIG. 3 , the time-delay between the stimulation artifact and the FP at the different electrodes is in good correlation with the distance between the stimulation and recording electrodes.
  • the excitation wave spread evenly on branched patterns as evidenced by the zero time difference at the end of the branches.
  • paired pulse stimulation protocols could also be used with the defined cardiac patterns.
  • the absolute/relative refractory period after AP generation could be mapped ( FIG. 4 ). If the stimulation delay between the two electrodes was too short, the second stimulus failed to evoke a second propagating AP.
  • This experiment suggests new possibilities to study phenomena difficult to measure in random (not constrained) monolayers, such as re-entry, the refractory period after an AP and arrhythmia.
  • Patterned spontaneously beating cardiomyocytes were exposed to 50 ⁇ M 1-Heptanol in the recording medium and the FPs were recorded at 15 minute intervals. A control recording was taken before the toxin application.
  • 1-Heptanol had a marked effect on the time for propagation of the excitation wave; it significantly increased the value measured before treatment.
  • the increase in the time delay indicates a block and uncoupling of the gap junctions and delays the FP between the two recording electrodes.
  • Sparfloxacin is a fluoroquinone antibiotic and HERG channel antagonist that is known to cause QT prolongation, Tdp and ventricular fibrillation[13, 53].
  • Application of 2 ⁇ M of Sparfloxacin to the recording medium caused a significant change in the spontaneous beating frequency within 25 minutes, with an increased FP length, non-synchronous contractions similar to fibrillation and burst-like beating activity as shown in FIG. 5 .
  • the beating frequency changed from a stable value of 0.6 Hz-1 Hz to intra-bursts with a higher frequency range of 1 Hz-1.2 Hz.
  • the loss of synchrony between the electrodes measured also indicated a propagation block in the monolayer.
  • the surface of commercial multielectrode arrays could be functionalized with SiPEG self-assembled monolayers.
  • the PEG layer may be ablated and subsequently patterned using photolithographical techniques and that incubation of the patterns with 5 ⁇ g/ml fibronectin for 20 minutes did not affect the cell resistive properties of PEG, but significantly improved the attachment of cardiac cells to the glass surface.
  • Surface analysis of the MEAs after modification indicated that both PEG-Silane modification and fibronectin adsorption on the ablated areas of the MEA surface was successful.
  • the patterned commercial electrodes were reusable 6-10 times without observable decline in the quality of electrophysiological recordings. Rat neonatal cardiac myocytes demonstrated normal physiology and formed beating monolayers on the areas designated by the patterns.
  • the patterned cells formed a confluent, aligned and interconnected network of spontaneously contracting cells communicating via gap junctions.
  • the activity of the cells could be measured using the MEA.
  • the extracellular electrodes allowed not only recording, but also stimulation of the patterned cardiac myocyte monolayers.
  • three different methods were used: 1) traditional video-monitoring of the excitation wave in the monolayer, 2) measurement of the speed of the spontaneous excitation waves on the patterns via the MEA and 3) measurement of the propagation of a stimulation-evoked extracellular field potential. All of these methods produced consistent results which were in reasonable agreement with published in vivo data. Conduction velocity increased with rapid electrical stimulation indicating that cardiac cell physiology can be studied using this method.
  • 1-Heptanol Two pharmacological agents, 1-Heptanol and Sparfloxacin, were used to demonstrate the effectiveness of this method for drug screening. Both of these compounds are known to have cardiac side effects.
  • 1-Heptanol is a gap junction blocker
  • Sparfloxacin is an antibiotic that was taken off the market due to cardiac side effects.
  • Our results were in agreement with the literature as 1-Heptanol drastically decreased the CV in the cardiac myocyte monolayers without completely blocking the conduction, whereas Sparfloxacin caused fibrillations which initiated a complete conduction block. These results indicated that specific drug actions can be studied using this system.
  • 1-Heptanol affected the conduction velocity properties, while maintaining the synchronicity of the beating cells.
  • a simple and reliable method for patterning cardiac myocytes on multielectrode arrays has been disclosed. This method is compatible with standard silicon manufacturing steps. Patterning of the cardiac myocytes increased the information content of the traditional multielectrode array recordings by enabling measurement of CV in a fast and simple way. Moreover, using paired stimulation, the measurement of the refractory period after the action potentials became possible. Measurement of the effect of drugs with known cardiac effects, was in agreement with the literature. Further development of this method could result in cheaper, faster, pharmaceutical side-effect screening with higher predictive value.

Abstract

The invention provides a culture of patterned cardiomyocytes, the culture including a support substrate bearing a multielectrode array, a negative surface resistant to cell attachment and deposited on the support substrate covering the multi-electrode array, a pattern ablated on the negative surface, a positive surface promoting cell attachment deposited on the pattern ablated on the negative surface; and cardiomyocytes adherent to the positive surface and growing aligned along the pattern ablated on the negative surface. The invention also includes a method of making the culture of patterned cardiomyocytes and a method of in vitro method of testing the effect of a compound on cardiac function by measuring electrical output from the patterned cardiomyocytes.

Description

    RELATED APPLICATION
  • This application claims priority from co-pending provisional application Ser. No. 61/257,504, filed on 3 Nov. 2009, which is incorporated herein by reference in its entirety.
  • STATEMENT OF GOVERNMENT RIGHTS
  • The invention claimed herein was made with at least partial support from the U.S. Government. Accordingly, the government may have certain rights in the invention, as specified by law.
  • FIELD OF THE INVENTION
  • The present invention relates to the field of new drugs and, more particularly, to a patterned culture of cardiomyocytes formed on a multielectrode array and useful for in vitro testing for possible cardiac side effects by a new drug.
  • BACKGROUND OF THE INVENTION
  • The development of a high-throughput, high-information content device to study and understand cardiac electrophysiology would be important for the fields of cardiac physiology, tissue engineering and drug research. More than 850,000 people are hospitalized for arrhythmias each year and ventricular fibrillation (VF) is a leading cause of cardiac death[1]. Despite the intensive research in this area, the mechanism of VF is still poorly understood[2-5].
  • Arrhythmia is a known side effect of commercial drugs. One of the mechanisms by which drugs can cause a potentially fatal form of ventricular tachy arrhythmia, called Torsades de pointes (Tdp), is through the prolongation of the QT interval (in an ECG the length of the ventricular action potential). It has been reported that approximately 2-3% of all prescribed drugs can cause long QT syndrome[6, 7]. A broad range of cardiovascular drugs and antibiotics also have the potential risk of causing drug induced Tdp[8, 9]. At the same time, prolongation of the QT interval does not necessarily lead to Tdp; lengthening of the QT interval could even be antiarrhythmogenic, as it is considered a mechanism of action of the class III anti-arrhythmics[8, 9]. Thus, a relatively high-throughput method to identify cardiac side effects and differentiate between arrhythmic and anti-arrhythmic effects at an early stage of drug development would have a significant impact on the field.
  • Gap junctions play an important role in the propagation of excitation in cardiac tissue. Changes in gap junction function affect major cardiac parameters, such as conduction velocity (CV). It has been observed in several cardiovascular diseases that the expression of connexins (protein molecules that form gap junction channels) is decreased or their distribution is changed, leading to a malfunction in gap junction coupling[10]. Understanding the pharmacological modulation of cardiac gap junction channels would further aid the drug development enterprise.
  • Introduction of an in vitro method for cardiac side effect testing, which has high predictive value, would have a significant impact on drug development as it could also reduce the cost, time and the number of drugs failing in clinical trials[11]. in vitro testing would also reduce the need for animal testing and could be used to study drug effects with a functional assay, but at the cellular level. Other in vitro methods, such as whole heart experiments (Langendorff heart model) or the Purkinje fiber preparation are difficult and time consuming[11]. Traditional methods used to study QT interval prolongation at the cellular level include patch-clamp experiments. However, these experiments are time intensive, require a skilled operator and cannot be used to study action potential (AP) propagation or parameters such as CV and re-entry. Moreover, evidence suggests that prolongation of QT intervals is not the best predictor of Torsades de pointes. The measurement of the length, or the variability in the length, of the refractory period after a cardiac action potential may have more relevance for predicting arrhythmic behavior[9].
  • Cardiac myocytes cultured on microelectrode arrays (MEA) have several benefits compared to either traditional patch clamp electrophysiology or isolated organ methods. The use of MEAs in the investigation of cardiac side effects would provide information in a relatively high-throughput and low cost manner compared to standard patch-clamp electrophysiology. However, at this time, it is still a low information content method and this has limited its use. Cardiac myocytes on MEAs have been used in a number of studies to investigate the effect of toxins, such as pesticides[12] and cardioactive drugs[13] on cardiac field potentials. A commercial system has also been introduced to measure QT intervals in a relatively high-throughput fashion[14], but, to date, it has only limited applications. However, cardiac myocytes can now be maintained over longer periods of time[15], thus chronic experiments, such as the monitoring of network remodeling for specific diseases, is now feasible. In addition, serum-free formulations for cardiac culture have also been introduced, which would increase the reproducibility of such a system[16].
  • All of the above mentioned studies utilized unorganized monolayers of cardiomyocytes on the MEAs. Development of a patterned cardiac myocyte layer that is aligned with the electrodes of a MEA could solve several problems associated with the random spread of excitation in a cardiac monolayer, which makes evaluation of the obtained data, such as CV, difficult. It would also enable the development of specific open-loop or closed-loop stimulation protocols to measure critical parameters, such as the length of the refractory period after the action potential. It could also be used to create a high-throughput, low-cost functional reentry model.
  • There are several lines of evidence indicating that not only contact interaction with the surface but the shape of the attachment area determines the physiology of cardiac myocytes[17]. Pattern geometries determine the extent of the alignment of the long axis of cardiac myocytes, alignment determines CV[18] and other physiological and pharmacological properties of cardiac tissues[19, 20].
  • Several different methods have been developed for cell patterning. One category of this technique is based on direct placement of cells or extracellular matrix molecules on desired locations and includes patterning through microfluidic channels[21-23], microcontact printing[24, 25] and inkjet printing[26]. Cardiac myocytes have previously been patterned on glass using photoresist[27] as well as other techniques[15, 17, 19, 20, 25]. Another method utilized photolithography following surface modification with self-assembled monolayers (SAMs) for neurons[28-30] as well as myocytes[15, 31]. The benefit of this method is the compatibility of the technique with cheap automated silicon manufacturing steps and the ability of the cells to self-assemble after random plating.
  • SAMs are one molecule thick monolayers attached to a surface composed of organic molecules, which have been extensively used for surface patterning[31-33]. Surface modification with SAMs is also compatible with advanced photolithography methods[30, 34]. Studies have also shown that cells survive on these surfaces for extended periods of time[35, 36], do not migrate off the patterned areas[34] and exhibit the typical morphology and physiology of the specific cell type[16, 37].
  • The goal of this study was the development of patterned, rat, cardiomyocyte cultures on MEAs in a serum-free medium for the study of cardiac physiology and pharmacology utilizing a high-throughput technique, but with high information content. An adsorbed fibronectin layer was used as the foreground because it supported cardiac myocyte attachment and growth and a 2-[Methoxy(Polyethyleneoxy) Propyl]TrimethoxySilane (SiPEG) SAM was used as the cell repellent background because of its excellent protein adsorption resistant properties[38]. The measurement of CV with the patterned cardiac myocyte monolayers and the feasibility to apply different stimulation protocols to the MEA/cardiac system was demonstrated. The action of 1-Heptanol and Sparfloxacin was also assessed. This method could easily be adapted for use with human cardiac myocytes to eliminate interspecies differences in drug side effect screening and could become an alternative to the existing more complex, expensive and time consuming methods.
  • SUMMARY OF THE INVENTION
  • With the foregoing in mind, the present invention advantageously provides a culture of patterned cardiomyocytes. The culture comprises a support substrate bearing a multielectrode array and a negative surface resistant to cell attachment and deposited on the support substrate covering the multi-electrode array. The negative surface bears a pattern ablated thereon, preferably by laser photolithography. A positive surface promoting cell attachment is deposited on the pattern ablated on the negative surface and cardiomyocytes adherent to the positive surface and growing are aligned along the pattern.
  • Preferably, in the culture of patterned myocardiocytes the negative surface comprises a material selected from the group consisting of polyethylene glycol, polyacrylic acid and polyacrylamide. Most preferably, the material selected is polyethylene glycol and it is deposited essentially as a monolayer. The pattern on the negative surface is preferably ablated by laser lithography through a mask, as further explained below.
  • The positive surface preferably comprises a material selected from the group consisting of fibronectin, trimethoxysilylpropyldiethylenetriamine and pure glass.
  • In the culture described the cardiomyocytes are preferably of mammalian origin, and may be of neonatal rat. Additionally, however, the cardiomyocytes may be derived from human embryonic stem cells.
  • The invention further includes a method of making the described culture of patterned myocardiocytes. The method comprises preparing a support substrate bearing a multi-electrode array, overlaying on the support substrate a negative surface resistant to cell adherence thereon, comprising polyethylene glycol and covering the multi-electrode array, ablating a pattern on the negative surface, depositing on the ablated pattern a positive surface promoting cell adherence thereon and including fibronectin, adhering cardiomyocytes on the positive surface, and culturing the cardiomyocytes so that they grow on the positive patterned surface and aligned therewith.
  • Yet additionally, the invention also includes an in vitro method of testing the effect of a compound on cardiac function. This method comprises providing patterned spontaneously beating cardiomyocytes according to the described culture. Recording a first electrical output from the cardiomyocytes. Contacting the cardiomyocytes with the compound being tested, Recording a second electrical output from the cardiomyocytes after being contacting with the compound. Finally, determining a change between the first and second electrical outputs as an indicator of altered cardiac function.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • Some of the features, advantages, and benefits of the present invention having been stated, others will become apparent as the description proceeds when taken in conjunction with the accompanying drawings. The drawings are presented solely for exemplary purposes and not with intent to limit the invention thereto. The following descriptions apply.
  • FIG. 1: Patterned neonatal cardiomyocytes. A: Pattern design and dimensions. B, C: Phase contrast pictures on day 12 of different regions of the pattern indicated in A. Field potentials measured at individual electrodes are marked in red in B. Electrode distance: 200 μm, D: Immunostaining with Rhodamine phalloidin for F-actin indicating cardiac myofibrils (note the visible striations). Nuclei are stained with DAPI (blue).
  • FIG. 2: Action potential (AP) conduction in patterned cardiac myocyte monolayers. A: phase contrast picture of cardiac myocyte patterns on the top of the substrate embedded electrodes (electrode distance is 200 μm) with a sketch of the conduction pathway on the completed pattern and with the recorded extracellular signals on the given electrodes. B: Overlay of the recorded traces showing the temporal relationship of the signals. A spontaneous AP was generated in the cardiac myocyte monolayer in the right fork of the pattern, close to electrode 74. This AP spread through the pattern reaching electrode 74 first, electrode 72 second and with a much longer delay (due to the longer path) electrode 52 last. Conduction velocity (CV) was determined based on the time delay in the signal between electrode 74 and 52 and the known pattern geometry. The calculated CV in this experiment was 0.190.
  • FIG. 3: Measurement of conduction velocity in patterned cardiac myocyte monolayers with electrical stimulation. A: Electrodes 52 and 84 were stimulated alternatively with a time delay of 300 ms and the response was recorded at electrode 72. B: E72 exhibited both long and short time delays in FP generation based on the distance the AP had to travel from the stimulation site.
  • FIG. 4: Measurement of refractory period after cardiac action potential. Electrodes 63 and 85 (see pattern geometries on the left) were stimulated alternatively according to the following protocol: E63, 100 ms delay, E85, 400 ms delay, E63 again. Right panel shows recording at electrode 87. Stimulating E85 100 ms after the stimulation of E63 failed to evoke an AP because this delay was too short, cells were stimulated in their refractory period. By chance, cell electrode coupling on electrode E85 was especially strong in this experiment, thus the extracellular recording approximately replicated the shape of the intracellular AP illustrating the relationship of the AP and the stimulation. When the stimulation delay between E63 and E85 was 250 ms, both stimulations evoked an action potential at electrode 87.
  • FIG. 5: Effect of the gap junction blocker 1-Heptanol. A: Propagation of excitation between the two end points in the pattern (Electrodes 84 and 52) before addition of the drug. The CV was measured to be 0.197 m/sec B: Propagation of excitation after the drug. The CV was calculated as 0.0066 m/sec. The drug effect on the CV measured over 4 MEAs.
  • FIG. 6: Effect of the HERG channel antagonist Sparfloxacin. A: Field potential recordings before addition of Sparfloxacin indicated stable beating frequencies and synchronous activity B: Field potential recording after the addition of 2 μM Sparfloxacin showed burst like activity with higher intra-burst frequencies and no synchrony between electrodes.
  • DETAILED DESCRIPTION OF THE PREFERRED EMBODIMENT
  • The present invention will now be described more fully hereinafter with reference to the accompanying drawings, in which preferred embodiments of the invention are shown.
  • Unless otherwise defined, all technical and scientific terms used herein are intended to have the same meaning as commonly understood in the art to which this invention pertains and at the time of its filing. Although various methods and materials similar or equivalent to those described herein can be used in the practice or testing of the present invention, suitable methods and materials are described below. However, the skilled should understand that the methods and materials used and described are examples and may not the only ones suitable for use in the invention.
  • Moreover, it should also be understood that any temperature, weight, volume, time interval, pH, salinity, molarity or molality, range, concentration and any other measurements, quantities or numerical expressions given herein are intended to be approximate and not exact or critical figures unless expressly stated to the contrary. Accordingly, where appropriate to the invention and as understood by those of skill in the art, it is proper to describe the various aspects of the invention using approximate or relative terms and terms of degree commonly employed in the art, for example, so dimensioned, about, approximately, substantially, essentially, consisting essentially of, comprising, and effective amount.
  • Further, any publications, patent applications, patents, and other references mentioned herein are incorporated by reference in their entirety as if they were part of this specification. However, in case of conflict, the present specification, including any definitions, will control. In addition, the materials, methods and examples given are illustrative in nature only and not intended to be limiting.
  • Accordingly, this invention may be embodied in many different forms and should not be construed as limited to the illustrated embodiments set forth herein. Rather, these illustrated embodiments are provided so that this disclosure will be thorough, complete, and will fully convey the scope of the invention to those skilled in the art. Other features, and advantages of the invention will be apparent from the following detailed description, and from the claims.
  • Materials and Methods
  • Surface Modification of Microelectrode Arrays with PEG Silanes
  • MEAs containing 60, 10 μm diameter electrodes (Multichannel Systems, Germany) were cleaned by soaking the arrays in a detergent solution for 2 hours followed by sonication for 10 minutes. The arrays were then oxygen plasma cleaned for 20 minutes. Surface modification was completed by incubation of the MEAs in a 3 mM PEG silane, 2-[Methoxypoly(ethyleneoxy)propyl]trimethoxysilane (MW=460-590, Gelest), solution in toluene, with 37% concentrated HCL added to achieve a final value of 0.08% (0.8 ml HCL/L), for 45 minutes at room temperature. The arrays were then rinsed once in toluene, twice in ethanol, twice in water and sonicated in water for 2 minutes to remove the non-covalently linked material[39]. The arrays were air dried with nitrogen and stored in a dessicator overnight.
  • Laser Ablation and Patterning of the Microelectrode Arrays
  • The MEAs were patterned by laser ablation photolithography using a deep UV (193 nm) excimer laser (Lambda Physik) at a pulse power of 230 mW and a frequency of 10 Hz for 45 seconds through a quartz photomask (Bandwidth foundry, Eveleigh, Australia). The arrays were sterilized using 70% isopropanol and then incubated with 5 μg/ml of fibronectin in a Phosphate buffered solution (Invitrogen) for 20 minutes at room temperature. The solution was removed and the surface was first rinsed with PBS, followed by the plating medium, and then dried before the cells were plated.
  • Neonatal Rat Cardiomyocyte Culture
  • The neonatal rat cardiomyocyte culture was prepared using the cardiac isolation kit from Worthington[40]. All animal work was approved by the UCF IACUC and followed NIH guidelines. Briefly, two day-old rat pups were euthanized in a precharged CO2 chamber. Hearts were dissected and minced in ice cold Hanks balanced salt solution (HBSS). Cardiac myocytes were dissociated by incubation of the hearts in trypsin (100 μg/ml in HBSS) for 16 hours at 2-8° C. The hearts in the trypsin solution were briefly warmed with a trypsin inhibitor before adding collegenase (300 units/ml in L-15 medium) for 45 minutes in a water bath at 37° C. followed by mechanical trituration. The cell solution was filtered to remove any remaining tissue and centrifuged at 50 g for 5 minutes at 22° C. The cells were resuspended in high glucose Dulbecco's modified eagle medium (DMEM, Gibco/Invitrogen) supplemented with 10 ml Fetal Bovine Serum (Gibco/Invitrogen) and 1 ml penicillin streptomycin (Gibco/Invitrogen), preplated in Petri dishes and incubated at 37° C. and in 5% CO2 for 45 minutes. This was necessary to eliminate the fibroblasts. The supernatant from the Petri dishes was centrifuged at 50 g for 5 min at 22° C. The cells were then resuspended in the plating medium. The serum-free plating medium consisted of: 100 ml Ultraculture medium (Bio Whittaker Cambrex) supplemented with 10 ml 827, 1 ml L-glutamine (Gibco/Invitrogen), 1 ml Penicillin Streptomycin, 0.375 g dextrose (Fisher Scientific) in 800 μl water, 1 ml non-essential amino acids and 1 ml of Hepes buffer (Gibco/Invitrogen)[41]. Additional growth factors were also added to improve cell survival in the serum-free conditions. They included 0.1 μg/ml of L-thyroxine, 10 ng/ml of Epidermal growth factor (Sigma-aldrich) and 0.5 μg/ml of Hydrocortisone (BD biosciences). Cells were plated at a density of 1000 cells/mm2 on the MEAs. The medium was changed 24 hours after plating. Subsequent changing of the medium was performed every third day.
  • Immunostaining
  • Patterned Cardiomyocytes were immunostained for F-Actin with Rhodamine Phalloidin (Invitrogen, R415), using a protocol provided by the company. Briefly, the cells were washed with PBS and fixed using 3% Formaldehyde. The coverslips were extracted with 0.1 ml Triton X®. The staining solution (with 1% Bovine Serum Albumin to prevent background staining) was added at a dilution of 1:40 in PBS and coverslips were incubated for 30 minutes. Imaging was done using confocal microscopy.
  • Multielectrode Extracellular Recordings
  • The cardiac myocytes were cultured on patterned metal MEAs (Planar 10 μm electrodes, 200 μm separation, Multichannel-systems). A 60 channel amplifier (MEA1040, Multichannel-systems) was used to record electrical activity from the spontaneously beating cardiac cells. The same electrodes were also used for stimulation utilizing a stimulus generator (STG 1002, Multichannel systems). The cells were stimulated utilizing 500 mV, 1 ms wide bipolar pulses at 2 Hz. The recording medium was the same as the plating medium with the pH adjusted to 7.3 using HEPES buffer. After a 30 minute incubation period, APs were detected and recorded using built in functions of the Multichannel System software. For drug experiments, 50 μM 1-Heptanol (Gibco/Invitrogen) was added to the bathing medium and recordings were performed before and 15 minutes after drug administration with additional recordings done at 15 minute intervals. For Sparfloxacin (Sigma-Aldrich), 2 μM of the drug was added to the recording medium and recordings were taken in 15 minute intervals before and after drug administration. The data was further analyzed using software written using Matlab and Clampfit (Axon Instruments).
  • Results Surface Modification of the Microelectrode Arrays
  • Our laboratory routinely uses SAMs to modify and pattern glass coverslips for cell culture applications[15, 16, 30-32, 42]. However, patterning cardiac myocytes on MEAs presented a challenge for three primary reasons: 1) The complex composition of the MEA surface made the verification of the surface modification step difficult, 2) MEAs are expensive, thus methods needed to be developed that enabled them to be cleaned and refunctionalized for repeated use and 3) patterned cardiac myocytes do not grow optimally on the standard trimethoxysilylpropyldiethylenetriamine (DETA) surfaces[15] nor on clean glass. Thus, a cell resistant background surface was needed which was also resistant to protein adsorption as this would allow fibronectin incubation on the foreground and prevent its adherence to the background, enabling cardiac myocyte attachment. Thus, poly(ethylene glycol) (PEG) was chosen for the background because of its excellent protein adsorption resistance. X-ray Photoelectron Spectroscopy (XPS) and contact angle measurements were used to analyze the results of the surface modifications and throughout the entire study for quality assurance purposes.
  • Cultured Neonatal Rat Cardiomyocytes on Patterned Surfaces in Serum-Free Medium
  • The growth and activity of neonatal rat cardiomyocytes on the patterned MEAs were assessed for over 2 weeks. The cells attached to the fibronectin, but not to the PEG background and showed clearly delineated regions (FIG. 1). This allowed the beating cells to grow exclusively over specific electrodes. The cells formed monolayers by day 2 and spontaneous contraction and beating activity began on day 4 and was consistent throughout the pattern. Cell survival and activity improved markedly with the addition of L-thyroxine, Epidermal growth factor and Hydrocortisone to the culture medium as indicated in the Methods. In a previous publication cells were shown to maintain spontaneous beating actively for up to 2 months in vitro [15]. Additionally, and by way of prophetic example, we believe the cardiomyocytes used in the invention could be derived from human embryonic stem cells (HESCs).
  • Extracellular Recordings from Patterned Cardiomyocytes on the MEAs
  • The electrical activity of the spontaneously contracting cells patterned on the microelectrodes was extracellularly recorded using the MEA system. Cardiac myocytes formed a morphologically homogenous, aligned, integrated network of cells that communicated through gap junctions and displayed normal tissue electrophysiology. The recorded field potential (FP) signals correlated with the contraction cycle.
  • In random cultures the direction and speed of the propagation of excitation (action potentials) have been shown to depend on a number of variables, including intercellular resistance across the gap junctions and the depolarizing sodium current[43-45]. For extracellular MEA recordings, previous research had indicated that the excitation wave could be followed through the monolayer as the increasing delay in field potential peak times and could be used to determine the pathway of the excitation as it moved away from the initiation site[46]. However, a direct determination of the CV has been difficult in unpatterned monolayers, even though the timing of FP generation could be estimated. With patterning of the cells on the electrode array, the exact path of a spontaneous excitation wave can be determined and then, using the path length, conduction velocity can be calculated with a high degree of accuracy. Moreover, because of the pre-determined pathway, there is no need to image the wave propagation, just the measurement of the start and end times of the waves on the patterns as recorded by the microelectrodes in the array. Thus, in later high-throughput applications significantly fewer electrodes would be necessary for these measurements and imaging would not be a necessary requirement for analysis. FIG. 2 shows a typical recording and the method for the determination of propagation direction. FIG. 213 emphasizes the delay between FPs at different electrodes on the pattern, which enabled the determination of CV with high temporal resolution. The signal propagation was also visualized using an Imaging software routine written in Matlab 6.5[47]. The program converted recordings from the electrodes into a video indicating the field potential movement across an 8×8 grid. Snapshots from the video are shown in the supplementary data. This program also verified the origin of the excitation wave and confirmed our CV results obtained using the earlier method (FIG. 2.).
  • The conduction velocity was calculated to be 0.190±0.025 m/s for spontaneous firing of the patterned cardiomyocytes over eight different MEAs. Previous research has shown that in vitro conduction velocities ranged from 0.12 m/s[48] to 0.242 m/s[49]. In a computer simulation of AP propagation in cardiac fibers in realistic conditions (taking into account extracellular solutions and spaces) the conduction velocity was 0.504 m/s[50]. In the human heart the AP generated by the Sino-atrial node spreads through the atria at a conduction velocity of 0.5 m/sec[51].
  • The conduction velocity on patterned cardiac monoloyers was also measured with stimulation. In these experiments, the cardiac pattern was stimulated at the two ends of the pattern, over a distance of 0.77 mm with 1 ms wide bipolar pulses with an amplitude of 500 mV at a frequency of 2 to 4 Hz (FIG. 3). The conduction velocity at 2 Hz stimulation was calculated to be 0.315±0.011 m/s (n=8). Previous research has indicated that rapid electrical stimulation in cultured, neonatal rat cardiomyocytes had an effect on the expression of Connexin [43] leading to changes in conduction properties[52], including an increase in conduction velocity.
  • This study showed that rapid electrical stimulation caused an increase in conduction velocity compared to the non-stimulated or spontaneous case. These results also indicated that accurate estimation of conduction velocity and propagation path could be determined in both the stimulated and in the spontaneous case. The measurement of the frequency dependence of conduction velocity could help in the understanding of the electrophysiological properties of cultured, neonatal rat cardiac myocytes, and ultimately, the physiology and pharmacology of the heart. The stimulated conduction velocity was also closer to the modeled values in the literature as well as to those generated by the SA node in the heart in viva
  • Electrical Stimulation Optimization Experiments and Measurement of Refractory Period after the AP
  • The stimulation protocol was optimized by determining a minimal threshold which reliably generated contraction from the cells. The cardiomyocytes were stimulated with 2 Hz, 500 mV, 1 ms wide bidirectional pulses at different electrodes. As seen in FIG. 3, the time-delay between the stimulation artifact and the FP at the different electrodes is in good correlation with the distance between the stimulation and recording electrodes. The excitation wave spread evenly on branched patterns as evidenced by the zero time difference at the end of the branches.
  • As shown in FIG. 4, paired pulse stimulation protocols could also be used with the defined cardiac patterns. By varying the time-delay between stimulations the absolute/relative refractory period after AP generation could be mapped (FIG. 4). If the stimulation delay between the two electrodes was too short, the second stimulus failed to evoke a second propagating AP. This experiment suggests new possibilities to study phenomena difficult to measure in random (not constrained) monolayers, such as re-entry, the refractory period after an AP and arrhythmia.
  • Pharmacology and Toxin Studies
  • One of the most important applications of this system could be the measurement of the effect of drugs and toxins on cardiac function. Two pharmacological agents, 1-Heptanol and Sparfloxacin, were tested to demonstrate the usability of this system for pharmacological and toxicology studies.
  • Patterned spontaneously beating cardiomyocytes were exposed to 50 μM 1-Heptanol in the recording medium and the FPs were recorded at 15 minute intervals. A control recording was taken before the toxin application. As shown in FIG. 5, 1-Heptanol had a marked effect on the time for propagation of the excitation wave; it significantly increased the value measured before treatment. The increase in the time delay indicates a block and uncoupling of the gap junctions and delays the FP between the two recording electrodes. The CV was calculated to be 0.0066±0.0004 (n=4), a significant decrease is from 0.197±0.014 m/s (n=4), and this result clearly shows that the effect of a gap junction blocker can be analyzed using this system.
  • Effect of Sparfloxacin
  • Sparfloxacin is a fluoroquinone antibiotic and HERG channel antagonist that is known to cause QT prolongation, Tdp and ventricular fibrillation[13, 53]. Application of 2 μM of Sparfloxacin to the recording medium caused a significant change in the spontaneous beating frequency within 25 minutes, with an increased FP length, non-synchronous contractions similar to fibrillation and burst-like beating activity as shown in FIG. 5. The beating frequency changed from a stable value of 0.6 Hz-1 Hz to intra-bursts with a higher frequency range of 1 Hz-1.2 Hz. The loss of synchrony between the electrodes measured also indicated a propagation block in the monolayer.
  • Discussion Surface Modification, Photolithographic Patterning and Patterned Cardiac Myocyte Cultures on Microelectrode Arrays
  • In this study, we have shown that the surface of commercial multielectrode arrays could be functionalized with SiPEG self-assembled monolayers. The PEG layer may be ablated and subsequently patterned using photolithographical techniques and that incubation of the patterns with 5 μg/ml fibronectin for 20 minutes did not affect the cell resistive properties of PEG, but significantly improved the attachment of cardiac cells to the glass surface. Surface analysis of the MEAs after modification indicated that both PEG-Silane modification and fibronectin adsorption on the ablated areas of the MEA surface was successful. The patterned commercial electrodes were reusable 6-10 times without observable decline in the quality of electrophysiological recordings. Rat neonatal cardiac myocytes demonstrated normal physiology and formed beating monolayers on the areas designated by the patterns.
  • Manipulating cells, determining their attachment, growth and differentiation in cultures has became important in applications such as tissue engineering, toxin detection, drug screening and robotics. Unfortunately, there is no generally applicable method to pattern all cell types. The method developed here using a protein adsorption resistant background, SiPEG, followed by protein adsorption (fibronectin) to the ablated area is a relatively simple, fast and inexpensive solution for this problem and could be generalized for ‘difficult to pattern’ cells.
  • Electrophysiology, Toxin and Drug Effects
  • The patterned cells formed a confluent, aligned and interconnected network of spontaneously contracting cells communicating via gap junctions. The activity of the cells could be measured using the MEA. The extracellular electrodes allowed not only recording, but also stimulation of the patterned cardiac myocyte monolayers. For the measurement of conduction velocity three different methods were used: 1) traditional video-monitoring of the excitation wave in the monolayer, 2) measurement of the speed of the spontaneous excitation waves on the patterns via the MEA and 3) measurement of the propagation of a stimulation-evoked extracellular field potential. All of these methods produced consistent results which were in reasonable agreement with published in vivo data. Conduction velocity increased with rapid electrical stimulation indicating that cardiac cell physiology can be studied using this method. Also, it was seen that the variability of the CV measurements decreased from about 15% as seen with the recordings from spontaneous APs to about 5% with stimulated APs. Patterning of the monolayers made CV measurements simpler, faster and more reliable. In future commercial applications this may be a more cost effective and a higher throughput technique for screening the cardiac side effects of drug candidates as it also eliminates the need for imaging of the propagation waves in the system. Patterning would also drastically reduce the number of required electrodes.
  • We have also demonstrated that alternating stimulation at different electrodes on the patterns could be used to measure the refractory period after APs, an important parameter for determining potentially fatal pharmacological side effects. This technique could also be a valuable tool for the study of cardiac defects, such as reentrant arrhythmia. Further development of this technology, especially for use with human cells, could significantly affect pharmaceutical drug development enabling high information content cardiac side effect screening.
  • Two pharmacological agents, 1-Heptanol and Sparfloxacin, were used to demonstrate the effectiveness of this method for drug screening. Both of these compounds are known to have cardiac side effects. 1-Heptanol is a gap junction blocker, whereas Sparfloxacin is an antibiotic that was taken off the market due to cardiac side effects. Our results were in agreement with the literature as 1-Heptanol drastically decreased the CV in the cardiac myocyte monolayers without completely blocking the conduction, whereas Sparfloxacin caused fibrillations which initiated a complete conduction block. These results indicated that specific drug actions can be studied using this system. 1-Heptanol affected the conduction velocity properties, while maintaining the synchronicity of the beating cells. The addition of Sparfloxacin to the cells caused a loss in rhythmicity resulting in a burst-like activity similar to fibrillations that are shown to have occurred in animal and human trials for the drug. These promising initial results support the need for further development of this system as a high-throughput, high-information content functional drug screening platform.
  • CONCLUSION
  • A simple and reliable method for patterning cardiac myocytes on multielectrode arrays has been disclosed. This method is compatible with standard silicon manufacturing steps. Patterning of the cardiac myocytes increased the information content of the traditional multielectrode array recordings by enabling measurement of CV in a fast and simple way. Moreover, using paired stimulation, the measurement of the refractory period after the action potentials became possible. Measurement of the effect of drugs with known cardiac effects, was in agreement with the literature. Further development of this method could result in cheaper, faster, pharmaceutical side-effect screening with higher predictive value.
  • Accordingly, in the drawings and specification there have been disclosed typical preferred embodiments of the invention and although specific terms may have been employed, the terms are used in a descriptive sense only and not for purposes of limitation. The invention has been described in considerable detail with specific reference to these illustrated embodiments. It will be apparent, however, that various modifications and changes can be made within the spirit and scope of the invention as described in the foregoing specification and as defined in the appended claims.
  • REFERENCES
    • 1. Jackson J Ht, Frech F, Ronen R, Mullany L, Lennert B, Jhaveri V. Assessment of drug therapy management and the prevalence of heart failure in a managed care population with hypertension. J Manag Care Pharm 2004; 10(6):513-520.
    • 2. Lin J W, Garber L, Qi Y R, Chang M G, Cysyk J, Tung L. Region [corrected] of slowed conduction acts as core for spiral wave reentry in cardiac cell monolayers. Am J Physiol Heart Circ Physiol 2008; 94(1):H58-65.
    • 3. Moe G K. On the multiple wavelet hypothesis of atrial fibrillation. Arch Int Pharmacodyn Ther 1962; (140):183-188.
    • 4. Nash M P, Mourad A, Clayton R H, Sutton P M, Bradley C P, Hayward M, et al. Evidence for multiple mechanisms in human ventricular fibrillation. Circulation 2006; 114(6):536-542.
    • 5, Pijnappels D A, van Tuyn J, de Vries A A, Grauss R W, van der Laarse A, Ypey D L, et al. Resynchronization of separated rat cardiomyocyte fields with genetically modified human ventricular scar fibroblasts. Circulation 2007; 116(18):2018-2028.
    • 6. Recanatini M, Poluzzi E, Masetti M, Cavalli A, De Ponti F. QT prolongation through hERG K(+) channel blockade: current knowledge and strategies for the early prediction during drug development. Med Res Rev 2005; 25(2):133-166.
    • 7. Sala M, Lazzaretti M, De Vidovich G, Caverzasi E, Barale F, d'Allio G, et al. Electrophysiological changes of cardiac function during antidepressant treatment. Ther Adv Cardiovasc Dis 2009; 3(1):29-43.
    • 8. Campbell T J, Williams K M. Therapeutic drug monitoring: antiarrhythmic drugs. Br J Clin Pharmacol 2001; 52 Suppl 1:21 S-34S.
    • 9. Hondeghem L M. Relative contributions of TRIaD and QT to proarrhythmia. J Cardiovasc Electrophysiol 2007; 18(6):655-657.
    • 10. van Rijen H V, van Veen T A, Gros D, Wilders R, de Bakker J M. Connexins and cardiac arrhythmias. Adv Cardiol 2006; 42:150-160.
    • 11. Suter W. Predictive value of in vitro safety studies. Curr Opin Chem Biol 2006; 10(4):362-366.
    • 12. Natarajan A, Molnar P, Sieverdes K, Jamshidi A, Hickman J J. Microelectrode array recordings of cardiac action potentials as a high throughput method to evaluate pesticide toxicity. Toxicol in vitro 2006; 20(3):375-381.
    • 13. Reppel M, Igelmund P, Egert U, Juchelka F, Hescheler J, Drobinskaya I. Effect of cardioactive drugs on action potential generation and propagation in embryonic stem cell-derived cardiomyocytes. Cell Physiol Biochem 2007; 19(5-6):213-224.
    • 14. Meyer T, Boven K H, Gunther E, Fejtl M. Micro-electrode arrays in cardiac safety pharmacology: a novel tool to study QT interval prolongation. Drug Saf 2004; 27(11):763-772.
    • 15. Dhir V, Natarajan A, Stancescu M, Chunder A, Bhargava N, Das M, et al. Patterning of diverse mammalian cell types in serum free medium with photoablation. Biotechnol Prog 2009; 25(2):594-60.3.
    • 16. Das M, Molnar P, Gregory C, Riedel L, Hickman J J. Long-term Culture Of Embyonic Rat Cardiomyocytes on an Organosilane Surface in a Serum Free Medium. Biomaterials 2004; 25(25):5643-5647.
    • 17. Parker K K, Tan J, Chen C S, Tung L. Myofibrillar architecture in engineered cardiac myocytes. Circ Res 2008; 103(4):340-342.
    • 18. Bourgeois E B, Fast V G, Collins R L, Gladden J D, Rogers J M. Change in Conduction Velocity due to Fiber Curvature in Cultured Neonatal Rat Ventricular Myocytes. Ieee Trans Biomed Eng 2009; 56(3):855-861.
    • 19. Badie N, Satterwhite L, Bursac N. A Method to Replicate the Microstructure of Heart Tissue in vitro Using DTMRI-Based Cell Micropatterning. Ann Biomed Eng 2009; 37(12):2510-2521.
    • 20. Kaji H, Takii Y, Nishizawa M, Matsue T. Pharmacological characterization of micropatterned cardiac myocytes. Biomaterials 2003; 24(23):4239-4244.
    • 21. Andersson H, van den Berg A. Microfabrication and microfluidics for tissue engineering: state of the art and future opportunities. Lab Chip 2004; 4(2):98-103.
    • 22. Khademhosseini A, Bettinger C, Karp J M, Yeh J, Ling Y, Borenstein J, et al. Interplay of biomaterials and micro-scale technologies for advancing biomedical applications. J Biomater Sci Polym Ed 2006; 17(11):1221-1240.
    • 23. Morin F, Nishimura N, Griscom L, Lepioufle B, Fujita H, Takamura Y, et al. Constraining the connectivity of neuronal networks cultured on microelectrode arrays with microfluidic techniques: a step towards neuron-based functional chips. Biosens Bioelectron 2006; 21(7):1093-1100.
    • 24. Kane R S, Takayama S, Ostuni E, Ingber D E, Whitesides G M. Patterning proteins and cells using soft lithography. Biomaterials 1999; 20(23-24):2363-2376.
    • 25. McDevitt T C, Angello J C, Whitney M L, Reinecke H, Hauschka S D, Murry C E, et al. in vitro generation of differentiated cardiac myofibers on micropatterned laminin surfaces. J Biomed Mater Res 2002; 60(3):472-479.
    • 26. Xu T, Gregory C A, Molnar P, Gui X, Jalota S, Bhaduri S B, et al. Viability and electrophysiology of neural cell structures generated by the inkjet printing method. Biomaterials 2006; 27(19):3580-3588.
    • 27. Rohr S, Scholly D M, Kleber A G. Patterned growth of neonatal rat heart cells in culture. Morphological and electrophysiological characterization. Circ Res 1991; 68:114-130.
    • 28. Hickman J J, Bhatia, S. K., Quong, J. N., Shoen, P., Stenger, D. A., Pike, C. J. and Cotman, C. W. Rational pattern design for in vitro cellular networks using surface photochemistry. J Vac Sci Technol A 1994; 12:607-616.
    • 29. Kleinfeld D, Kahler K H, Hockberger P E. Controlled outgrowth of dissociated neurons on patterned substrates. J Neurosci 1988; 8(11):4098-4120.
    • 30. Ravenscroft M S, Bateman K E, Shaffer K M, Schessler H M, Jung D R, Schneider T W, et al. Developmental neurobiology implications from fabrication and analysis of hippocampal neuronal networks on patterned silane-modified surfaces. J Am Chem Soc 1998; 120(47):12169-12177.
    • 31. Molnar P, Wang W, Natarajan A, Rumsey J W, Hickman J J. Photolithographic patterning of C2C12 myotubes using vitronectin as growth substrate in serum-free medium. Biotechnol Prog 2007; 23(1):265-268.
    • 32. Rumsey J W, Das M, Bhalkikar A, Stancescu M, Hickman J J. Tissue engineering the mechanosensory circuit of the stretch reflex arc: sensory neuron innervation of intrafusal muscle fibers. Biomaterials 2010; 31:8218-8227.
    • 33. Stenger D A, Georger J H, Dulcey C S, Hickman J J, Rudolph A S, Nielsen T B, et al. Coplanar Molecular Assemblies of Aminoalkylsilane and Perfluorinated Alkylsilane—Characterization and Geometric Definition of Mammalian-Cell Adhesion and Growth. J Am Chem Soc 1992; 114(22):8435-8442.
    • 34. Corey J M, Wheeler B C, Brewer G J. Compliance of hippocampal neurons to patterned substrate networks. J Neurosci Res 1991; 30(2):300-307.
    • 35. Das M, Bhargava N, Bhalkikar A, Kang J-F, Hickman J J. Temporal neurotransmitter conditioning restores the functional activity of adult spinal cord neurons in long-term culture. Exp Neurol 2008; 209:171-180
    • 36. Das M, Rumsey J W, Bhargava N, Stancescu M, Hickman J J. A defined long-term in vitro tissue engineered model of neuromuscular junctions. Biomaterials 2010; 31:4880-4888.
    • 37. Lochter A, Taylor J, Braunewell K H, Holm J, Schachner M. Control of neuronal morphology in vitro: interplay between adhesive substrate forces and molecular instruction. J Neurosci Res 1995; 42(2):145-158.
    • 38. Popat K C, Sharma S, Desai T A. Quantitative XPS Analysis of PEG-Modified Silicon Surfaces. J Phys Chem B 2004; 108(17):5185-5188.
    • 39. Popat K C, Mor G, Grimes C A, Desai T A. Surface modification of nanoporous alumina surfaces with poly(ethylene glycol). Langmuir 2004; 20(19):8035-8041.
    • 40. Natarajan A R, Rong Q, Katchman A N, Ebert S N. Intrinsic cardiac catecholamines help maintain beating activity in neonatal rat cardiomyocyte cultures. Pediatr Res 2004; 56(3):411-417.
    • 41. Sathaye A, Bursac N, Sheehy S, Tung L. Electrical pacing counteracts intrinsic shortening of action potential duration of neonatal rat ventricular cells in culture. J Mal Cell Cardiol 2006t; 41(4):633-641.
    • 42. Molnar P, Kang J F, Bhargava N, Das M, Hickman J J. Synaptic connectivity in engineered neuronal networks. Methods Mol Biol 2007; 403:165-173.
    • 43. Egert U, Banach K, Meyer T. Analysis of cardiac myocyte activity dynamics with microelectrode arrays. In: Taketani M B M, editor. Advances in network electrophysiology using multi electrode arrays: Springer, 2006. p. 274-290.
    • 44. Spach M S. The role of cell-to-cell coupling in cardiac conduction disturbances. Adv Exp Med Biol 1983; 161:61-77.
    • 45. Spach M S, Heidlage J F. The stochastic nature of cardiac propagation at a microscopic level. Electrical description of myocardial architecture and its application to conduction. Circ Res 1995; 76(3):366-380.
    • 46. Halbach M, Egert U, Hescheler J, Banach K. Estimation of action potential changes from field potential recordings in multicellular mouse cardiac myocyte cultures. Cell Physiol Biochem 2003; 13(5):271-284.
    • 47. Yeung C K, Sommerhage F, Wrobel G, Offenhausser A, Chan M, Ingebrandt S. Drug profiling using planar microelectrode arrays. Anal Bioanal Chem 2007; 387(8):2673-2680.
    • 48. Iravanian S, Nabutovsky Y, Kong C R, Saha S, Bursac N, Tung L. Functional reentry in cultured monolayers of neonatal rat cardiac cells. Am J Physiol Heart Circ Physiol 2003; 285(1):H449-456.
    • 49. Entcheva E K, Y. Tchernev, E. Tung, L. Fluorescence imaging of electrical activity in cardiac cells using an all-solid-state system IEEE Trans Biomed Eng 2004; 51(2):331-341.
    • 50. Stinstra J, Roberts S, Pormann J, Macleod R, Henriquez C. A Model of 3D Propagation in Discrete Cardiac Tissue, Comput Cardiol 2006; 33:41-44.
    • 51. Klabunde R E. Cardiovascular physiology concepts. illustrated ed: Lippincott Williams & Wilkins, 2004.
    • 52. Inoue N, Ohkusa T, Nao T, Lee J K, Matsumoto T, Hisamatsu Y, et al. Rapid electrical stimulation of contraction modulates gap junction protein in neonatal rat cultured cardiomyocytes: Involvement of Mitogen-Activated Protein Kinases and Effects of Angiotensin II-Receptor Antagonist. J Am Coll Cardiol 2004; 44; 914-922,
    • 53. Lu H R, Vlaminckx E, Van de Water A, Rohrbacher J, Hermans A, Gallacher D J. In-vitro experimental models for the risk assessment of, antibiotic-induced Q T prolongation. Eur J Pharmacol 2006; 553(1-3):229-239.

Claims (14)

1-10. (canceled)
11. A culture of patterned cardiomyocytes, said culture comprising:
i) a support substrate bearing a multielectrode array;
ii) a first surface resistant to cell attachment and deposited on said support substrate wherein the first surface covers the multi-electrode array;
iii) a pattern ablated on said first surface;
iv) a second surface that promotes cell attachment deposited on the pattern ablated on the first surface; and
v) cardiomyocytes adherent to said second surface and growing aligned along said pattern to provide said culture of patterned spontaneously beating cardiomyocytes.
12. The culture of claim 11, wherein the first surface comprises at least one material selected from the group consisting of polyethylene glycol, polyacrylic acid and polyacrylamide.
13. The culture of claim 12, wherein the material selected is polyethylene glycol deposited essentially as a monolayer.
14. The culture of claim 10, wherein the second surface comprises at least one material selected from the group consisting of fibronectin, trimethoxysilylpropyldiethylenetriamine and pure glass.
15. The culture of claim 10, wherein the pattern is ablated by photolithography.
16. The culture of claim 10, wherein the ablated pattern comprises a path extending over two or more electrodes of the multielectrode array.
17. The culture of claim 16, wherein the support substrate comprises both an multielectrode array-bearing portion and a portion devoid of a multielectrode array, the first surface is deposited on both the multielectrode array-bearing portion and the portion devoid of a multielectrode array, and the path extends over both the multielectrode array-bearing portion and the portion devoid of a multielectrode array.
18. The culture of claim 16, wherein the path comprises a first end, a middle region, and a second end, wherein the second end comprises a branched region.
19. The culture of claim 18, wherein the first end of the path contacts at least two electrodes of the multielectrode array, the middle region of the path extends over the portion of the support substrate devoid of the multielectrode array, and the branched region extends over at least two electrodes of the multielectrode array.
20. The culture of claim 19, wherein the branched region comprises a first branch and a second branch in a Y-shaped configuration, and each of the first branch and the second branch extend over at least two electrodes of the multielectrode array.
21. The culture of claim 10, wherein the cardiomyocytes are of mammalian origin.
22. The culture of claim 10, wherein the cardiomyocytes are of rat origin.
23. The culture of claim 10, wherein the cardiomyocytes are derived from human embryonic stem cells.
US15/190,958 2009-11-03 2016-06-23 Patterned cardiomyocyte culture on microelectrode array Abandoned US20160305927A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US15/190,958 US20160305927A1 (en) 2009-11-03 2016-06-23 Patterned cardiomyocyte culture on microelectrode array

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US25750409P 2009-11-03 2009-11-03
US12/938,701 US9404140B1 (en) 2009-11-03 2010-11-03 Patterned cardiomyocyte culture on microelectrode array
US15/190,958 US20160305927A1 (en) 2009-11-03 2016-06-23 Patterned cardiomyocyte culture on microelectrode array

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US12/938,701 Division US9404140B1 (en) 2009-11-03 2010-11-03 Patterned cardiomyocyte culture on microelectrode array

Publications (1)

Publication Number Publication Date
US20160305927A1 true US20160305927A1 (en) 2016-10-20

Family

ID=56507197

Family Applications (2)

Application Number Title Priority Date Filing Date
US12/938,701 Active 2033-06-02 US9404140B1 (en) 2009-11-03 2010-11-03 Patterned cardiomyocyte culture on microelectrode array
US15/190,958 Abandoned US20160305927A1 (en) 2009-11-03 2016-06-23 Patterned cardiomyocyte culture on microelectrode array

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US12/938,701 Active 2033-06-02 US9404140B1 (en) 2009-11-03 2010-11-03 Patterned cardiomyocyte culture on microelectrode array

Country Status (1)

Country Link
US (2) US9404140B1 (en)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11022605B2 (en) 2016-08-26 2021-06-01 University Of Central Florida Research Foundation, Inc. Multi-component in vitro system to deduce cell signaling pathways by electronic stimulation patterns
US11614437B2 (en) 2013-01-30 2023-03-28 University Of Central Florida Research Foundation, Inc. Devices, systems, and methods for evaluating cardiac parameters
WO2023183371A3 (en) * 2022-03-22 2023-10-26 The Trustees Of Columbia University In The City Of New York Engineered heart tissue model of restrictive cardiomyopathy for drug discovery

Families Citing this family (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA3048884A1 (en) 2016-12-30 2018-07-05 University Of Central Florida Research Foundation, Inc. Pumpless microfluidic organ-on-a-chip system including a functional immune system
US10767164B2 (en) 2017-03-30 2020-09-08 The Research Foundation For The State University Of New York Microenvironments for self-assembly of islet organoids from stem cells differentiation
US20220196636A1 (en) * 2019-03-29 2022-06-23 The Johns Hopkins University Grid of responses indicating drug sensitivity
CN113621514B (en) * 2021-07-16 2023-04-07 武汉大学 Cell atrial fibrillation model stimulation culture system

Family Cites Families (48)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5682899A (en) 1991-05-16 1997-11-04 Ami-Med Corporation Apparatus and method for continuous cardiac output monitoring
US5422510A (en) 1992-10-30 1995-06-06 Analog Devices, Incorporated MOS transistor with non-uniform channel dopant profile
US20090239940A1 (en) * 1997-07-22 2009-09-24 Del Monte Federica Treating heart failure and ventricular arrhythmias
US7923015B2 (en) 1997-08-14 2011-04-12 Institut Pasteur Methods for direct visualization of active synapses
US5948621A (en) 1997-09-30 1999-09-07 The United States Of America As Represented By The Secretary Of The Navy Direct molecular patterning using a micro-stamp gel
US7266457B1 (en) 1999-05-21 2007-09-04 Hesperos, Llc High throughput functional genomics
EP1226239A4 (en) 1999-10-15 2003-02-12 Advanced Cell Tech Inc Methods of producing differentiated progenitor cells and lineage-defective embryonic stem cells
JP2003518258A (en) 1999-12-23 2003-06-03 ファルマシア・アンド・アップジョン・カンパニー Assay methods and diagnostic and therapeutic methods based on the use of sodium channels as targets of amyloid β or aggregates thereof
EP1395330A1 (en) 2001-04-20 2004-03-10 The Board of Regents for the University of Oklahoma Cardiac neuromodulation and methods of using same
ES2287351T3 (en) 2001-04-25 2007-12-16 Cornell Research Foundation, Inc. DEVICES AND METHODS FOR CELL SYSTEMS BASED ON PHARMACOCINETICS.
US6844028B2 (en) 2001-06-26 2005-01-18 Accelr8 Technology Corporation Functional surface coating
AU2002365121A1 (en) 2001-08-06 2003-07-09 Vanderbilt University System and methods for measuring at least one metabolic rate of a plurality of cells
KR20040030142A (en) 2001-08-17 2004-04-08 애버리 데니슨 코포레이션 Topcoat compositions, substrates containing a topcoat derived therefrom, and methods of preparing the same
US6916541B2 (en) 2001-09-07 2005-07-12 Penn State Research Foundation Modified substrates for the attachment of biomolecules
AU2002336504A1 (en) 2001-09-12 2003-03-24 The State Of Oregon, Acting By And Through The State Board Of Higher Education On Behalf Of Oregon S Method and system for classifying a scenario
US8153424B2 (en) 2001-10-03 2012-04-10 Wisconsin Alumni Research Foundation Method of in vitro differentiation of neural stem cells, motor neurons and dopamine neurons from primate embryonic stem cells
US20030144823A1 (en) 2001-11-01 2003-07-31 Fox Jeffrey J. Scale-free network inference methods
WO2003081204A2 (en) 2002-03-20 2003-10-02 Purdue Research Foundation Microscale sensor element and related device and method of manufacture
US20040076961A1 (en) 2002-10-21 2004-04-22 Lewis Mark A. Biomolecule retaining material and methods for attaching biomolecules to a surface
US7341841B2 (en) 2003-07-12 2008-03-11 Accelr8 Technology Corporation Rapid microbial detection and antimicrobial susceptibility testing
EP1649008A2 (en) * 2003-07-16 2006-04-26 Boston Scientific Limited Aligned scaffolds for improved myocardial regeneration
US7198855B2 (en) 2003-09-12 2007-04-03 Becton, Dickinson And Company Methods of surface modification of a flexible substrate to enhance cell adhesion
WO2005043126A2 (en) 2003-10-27 2005-05-12 Drexel University Piezoelectric cantilever sensors
WO2005108598A1 (en) * 2004-05-11 2005-11-17 Axiogenesis Ag Assay for drug discovery based on in vitro differentiated cells
US7807462B2 (en) 2004-11-16 2010-10-05 University Of Medicine And Dentistry Of New Jersey Method for producing a functional neuron
RU2434636C2 (en) 2004-11-17 2011-11-27 Ньюралстем, Инк. Transplantation of nerve cells for treatment of neurodegenerative states
US7718846B2 (en) 2005-05-16 2010-05-18 Gideon Koren Animal models of long QT syndrome and uses thereof
EP1913393A4 (en) 2005-06-16 2010-03-03 Univ California Large scale parallel immuno-based allergy test and device for evanescent field excitation of fluorescence
NZ565683A (en) 2005-07-08 2011-04-29 Braincells Inc Methods for identifying agents and conditions that modulate neurogenesis in human neural cells
US20070129447A1 (en) 2005-12-02 2007-06-07 Sra Jasbir S Method of drug therapy in treatment of cardiac arrhythmias
GB2447191B (en) * 2005-12-22 2011-08-03 Es Cell Int Pte Ltd Direct differentiation of cardiomyocytes from human embryonic stem cells
WO2007098065A2 (en) * 2006-02-17 2007-08-30 The United States Of America As Represented By The Department Of Veterans Affairs Human sodium channel isoforms
US7927671B2 (en) 2006-09-21 2011-04-19 Chisso Corporation Trifunctional compound, composition and polymer thereof
US8193225B2 (en) * 2006-10-13 2012-06-05 The Board Of Regents Of The University Of Texas System Isoxazole amides, derivatives and methods of chemical induction of neurogenesis
US20090029463A1 (en) 2007-07-25 2009-01-29 Bioe, Inc. Differentiation of Multi-Lineage Progenitor Cells to Chondrocytes
WO2009036573A1 (en) 2007-09-21 2009-03-26 Institut De Recherches Cliniques De Montreal Apparatus and method for monitoring cell behaviour
WO2009114133A1 (en) 2008-03-10 2009-09-17 The J. David Gladstone Institutes Cells and assays for use in detecting long qt syndrome
US8835168B2 (en) 2009-04-23 2014-09-16 University Of Central Florida Research Foundation, Inc. Synthetic mammalian neuromuscular junction and method of making
US9012172B2 (en) 2009-05-01 2015-04-21 President And Fellows Of Harvard College Devices comprising muscle thin films and uses thereof in high throughput assays for determining contractile function
WO2010138782A1 (en) 2009-05-28 2010-12-02 University Of Central Florida Research In vitro production of oligodendrocytes from human umbilical cord stem cells
US8828721B1 (en) 2009-05-28 2014-09-09 University Of Central Florida Research Foundation, Inc. Method of myelinating isolated motoneurons
EP2435585A4 (en) 2009-05-28 2013-02-20 Univ Central Florida Res Found Method of screening drugs for reversal of amyloid beta neurotoxicity
EP2531910B1 (en) 2010-02-05 2015-04-08 University of Central Florida Research Foundation, Inc. Methods for identifying points of action in electrically active cells
WO2012158923A1 (en) 2011-05-17 2012-11-22 University Of Central Florida Research Foundation, Inc. Stable electrically active neurons from adult tissue
KR101966208B1 (en) 2011-07-21 2019-04-09 더 보드 어브 트러스티스 어브 더 리랜드 스탠포드 주니어 유니버시티 Cardiomyocytes from induced pluripotent stem cells from patients and methods of use
US20150219622A1 (en) 2012-08-17 2015-08-06 University Of Central Florida Research Foundation, Inc. Methods, systems and compositions for functional in vitro cellular models of mammalian systems
CA2899445C (en) 2013-01-30 2023-04-04 University Of Central Florida Research Foundation, Inc. Devices and systems for mimicking heart function
US20140274796A1 (en) 2013-03-15 2014-09-18 University Of Central Florida Research Foundation, Inc. Methods, Systems, and Compositions for In Vitro Concentric Cell Culture Analog Systems

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11614437B2 (en) 2013-01-30 2023-03-28 University Of Central Florida Research Foundation, Inc. Devices, systems, and methods for evaluating cardiac parameters
US11022605B2 (en) 2016-08-26 2021-06-01 University Of Central Florida Research Foundation, Inc. Multi-component in vitro system to deduce cell signaling pathways by electronic stimulation patterns
WO2023183371A3 (en) * 2022-03-22 2023-10-26 The Trustees Of Columbia University In The City Of New York Engineered heart tissue model of restrictive cardiomyopathy for drug discovery

Also Published As

Publication number Publication date
US9404140B1 (en) 2016-08-02

Similar Documents

Publication Publication Date Title
Natarajan et al. Patterned cardiomyocytes on microelectrode arrays as a functional, high information content drug screening platform
US20160305927A1 (en) Patterned cardiomyocyte culture on microelectrode array
Banach et al. Development of electrical activity in cardiac myocyte aggregates derived from mouse embryonic stem cells
Jo et al. Midbrain-like organoids from human pluripotent stem cells contain functional dopaminergic and neuromelanin-producing neurons
Braam et al. Prediction of drug-induced cardiotoxicity using human embryonic stem cell-derived cardiomyocytes
Ivashchenko et al. Human-induced pluripotent stem cell-derived cardiomyocytes exhibit temporal changes in phenotype
Nguemo et al. In vitro model for assessing arrhythmogenic properties of drugs based on high-resolution impedance measurements
Ma et al. High purity human-induced pluripotent stem cell-derived cardiomyocytes: electrophysiological properties of action potentials and ionic currents
Guo et al. The electrophysiological effects of cardiac glycosides in human iPSC-derived cardiomyocytes and in guinea pig isolated hearts
Takeuchi et al. Device for co-culture of sympathetic neurons and cardiomyocytes using microfabrication
Schultz et al. The protective effect of ursodeoxycholic acid in an in vitro model of the human fetal heart occurs via targeting cardiac fibroblasts
Hu et al. Synchronized electromechanical integration recording of cardiomyocytes
Riedel et al. Functional and pharmacological analysis of cardiomyocytes differentiated from human peripheral blood mononuclear-derived pluripotent stem cells
Cantley et al. Functional and sustainable 3D human neural network models from pluripotent stem cells
EP2951281B1 (en) System and method for mimicking heart function
Belinsky et al. Physiological properties of neurons derived from human embryonic stem cells using a dibutyryl cyclic AMP-based protocol
EP2723853B1 (en) In vitro cardiovascular model
Daus et al. A spheroid-based biosensor for the label-free detection of drug-induced field potential alterations
Kofron et al. Gq-activated fibroblasts induce cardiomyocyte action potential prolongation and automaticity in a three-dimensional microtissue environment
Jimenez-Vazquez et al. SNTA1 gene rescues ion channel function and is antiarrhythmic in cardiomyocytes derived from induced pluripotent stem cells from muscular dystrophy patients
Zhang et al. Characterization and regulation of T-type Ca2+ channels in embryonic stem cell-derived cardiomyocytes
Sokal et al. Multi-neuronal recordings reveal a differential effect of thapsigargin on bicuculline-or gabazine-induced epileptiform excitability in rat hippocampal neuronal networks
Kadir et al. Embryonic stem cell‐derived cardiomyocytes as a model to study fetal arrhythmia related to maternal disease
KR20160099704A (en) Method for producing sinoatrial node cells (pacemaker cells) from stem cells, and use of the produced sinoatrial node cells
Takahashi et al. Contraction control of aligned myofiber sheet tissue by parallel oriented induced pluripotent stem cell-derived neurons

Legal Events

Date Code Title Description
AS Assignment

Owner name: UNIVERSITY OF CENTRAL FLORIDA RESEARCH FOUNDATION,

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:MOLNAR, PETER;NATARAJAN, ANUPAMA;STANCESCU, MARIA;AND OTHERS;SIGNING DATES FROM 20160630 TO 20160810;REEL/FRAME:039899/0072

AS Assignment

Owner name: NATIONAL INSTITUTES OF HEALTH (NIH), U.S. DEPT. OF

Free format text: CONFIRMATORY LICENSE;ASSIGNOR:UNIVERSITY OF CENTRAL FLORIDA;REEL/FRAME:043569/0014

Effective date: 20170808

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: ADVISORY ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION