electrical propagation of three-dimensional engineered hearts using decellularized extracellular...

1
fluorescent proteins to a voltage sensitive membrane protein (1) . However, one factor that has limited the utility of these indicators is poor voltage sensitivity, making it a challenge to express the indicators in neurons and still observe detectable responses (2) . In addition, none of the genetically-encoded voltage indicators reported can be combined with channelrhodopsin for optical readout of electrical activity in an all-optical electrophysiology setup. To address these limitations, we set up a directed evolution strategy to screen for both voltage indicator brightness and voltage sensitivity. First, bacterial expression enables us to screen thousands of clones in colonies of E. coli to improve the brightness of the indicator. Second, a medium throughput mammalian electric field stim- ulation system allows us to test for function of the voltage indicators. This screening has resulted in a family of red voltage indicators (VSDRs). They consist of the voltage-sensing domain of Ciona intestinalis voltage-sensitive phosphatase linked to red fluorescent protein mApple. The fluorescence inten- sity of the VSDRs increases in response to depolarization. We show that the combination of signal size and response speed of VSDRs allows the reliable detection of spontaneous action potentials in cultured mammalian neurons in single trials with widefield fluorescent light microscopy. Critically, we also demonstrate that VSDRs faithfully reports signals from all-optical electrophys- iology experiments using channelrhodopsin to depolarize mammalian cells. 1. B. J. Baker et al., J. Neurosci. Methods. 2007, 161, 32-38. 2. W. Akemann et al., Nat Methods. 7, 2010, 643-649. 3184-Plat Optogenetic Stimulation of Channelrhodopsin-2 Expressing Neurons Flips Cortical Networks from Low to High Activity State Parijat Sengupta, Lindsay Fague. ISP-Applied Sciences Laboratory, Washington State University, Spokane, WA, USA. Optogenetics, the single-component strategy for neuronal stimulation using light, allows for non-invasive and temporally precise stimulation of neurons. In our lab, we have recently developed an investigative tool that combines op- togenetic stimulation of channelrhodopsin-2 expressing neuronal networks (Figure-1: left panel; red: neuronal nucleus marker; violet: glial fibrillary acidic protein; green: channelrhodopsin-2-YFP expressing neurons) with long-term electrophysiological measurement from multiple locations of the network using a multi-electrode array (MEA) dish. This combinatorial approach can be used to induce, detect and track short- and long-term changes in spontaneous and evoked neuronal network activity in response to optical stimuli. Using this technique we have found that even a single 20s-long excitation light pulse can flip a dissociated cortical network from a low-activity state to a high- activity state (Figure-1: right, top panel). Interestingly, a single pulse does not seem to affect the levels of network activity if the network is already in high-activity state (Figure-1: right, bottom panel). A more comprehensive study of this phenomenon and its relationship with network plasticity is currently underway. Acknowledgement: Channelrhodopsin-2- YFP construct was a gift from Karl Deisser- oth lab at Stanford University. Platform: Cardiac, Smooth, and Skeletal Muscle Electrophysiology 3185-Plat Myocyte Stretch Slows Cardiac Conduction by a Caveolae-Dependent In- crease in Sarcolemmal Capacitance Emily Pfeiffer, Adam Wright, Andrew Edwards, Jennifer Stowe, Katie McNall, Justin Tan, Hemal Patel, Jeffrey Omens, Andrew McCulloch. UCSD, San Diego, CA, USA. Alterations in cardiac electrophysiology by mechanical perturbations have been long observed, however the cellular basis for these changes remains poorly understood. Most studies have focused on pro-arrhythmic consequences of stretch-activated currents (SAC). However, alterations in cardiac conduction that can create an arrhythmogenic substrate have also been observed even in the presence of SAC blockade. We tested the hypotheses that stretch slows conduc- tion by increasing cell membrane capacitance via recruitment and sarcolemmal integration of caveolae and found that: (1) Conduction slowing occurred with stretch in both (a) pressure-loaded mouse hearts and (b) biaxially-stretched mi- cropatterned murine cardiomyocyte monolayers, (2) Inhibition of stretch- activated currents had no effect on this slowing in both models, (3) Slowing was abrogated by caveolae depletion in both models, using cav3-knockout mice or incubation with 1 mM MbCD, (4) Stretch significantly increased cell membrane capacitance and time constant in single wild-type, but not caveolae-depleted cells, and (5) These effects on cell membrane capacitance and conduction slowing concide with recruitment and integration of caveolae by stretch. Together, these data suggest that myocyte stretch promotes recruit- ment and integration of caveolae and causes slowing of cardiac conduction by increasing cell membrane capacitance. 3186-Plat Electrical Propagation of Three-Dimensional Engineered Hearts using De- cellularized Extracellular Matrix Haruyo Yasui 1 , Jong-Kook Lee 2 , Akira Yoshida 1 , Teruki Yokoyama 1 , Junichi Nakai 3 , Issei Komuro 4 . 1 Cardiovascular Medicine, Osaka University Graduate School of Medicine, Suita, Japan, 2 Cardiovascular Regenerative Medicine, Osaka University Graduate School of Medicine, Suita, Japan, 3 Brain Science Institute, Saitama University, Saitama, Japan, 4 Cardiovascular Medicine, Tokyo University Graduate School of Medicine, Tokyo, Japan. [Background] Construction of three-dimensional cardiac tissues using decellu- larized extracellular matrix could be a new technique to create an ‘‘organ-like’’ structure of the heart. To engineer functional artificial hearts comparable for or- thotopic hearts, however, much remain to be solved including electrical stabil- ity for efficient contraction. To elucidate the points, we examined electrophysiological properties of recellularized heart tissues. [Methods] Entire hearts of adult Wistar rats were decellularized using 0.5% SDS and 1% triton-X, and then recellularized with enzymatically-dispersed neonatal rat cardiac cells (1x10t8 cells) through antegrade coronary circulation. Three and seven days later after cell seeding, we observed excitation of spontaneous and pacing-induced beatings of recellularized heart tissues expressing Ca 2þ - indicating protein (GCaMP2) using high resolution cameras. We also conduct- ed immunofluorescence staining to examine morphological aspects of engi- neered tissues. [Results] Live tissue fluorescence imaging revealed that GFP- labeled-isolated cardiac cells were dispersed into interstitial spaces through extravasation from coronary arteries. Engineered hearts seeded with GCaMP2-expressing cardiac cells started showing spontaneous beating, and were subjected to further electrophysiological experiments using optical imag- ing system. Both in spontaneous and pacing-induced beating hearts, we observed well-organized conduction of stable excitation in substantial areas of the engineered heart tissues, whereas we also recorded disorganized propa- gation of asynchronous excitation with multiple origins. Conduction velocity was markedly decreased (~5 cm/sec) during early stages (up to nine days) after recellularization. Immunofluorescence study revealed randomly-mixed align- ment of cardiomyocytes, endothelial cells and smooth muscle cells, stained with alpha-actinin, CD31 and sm-actin, respectively. Recellularized heart tis- sues also showed disarray of cardiomyocytes and markedly decreased expres- sion of connexin43. [Conclusion] Three-dimensional engineered hearts showed electrical heterogeneity and proarrhythmic propensity during early stages of culture. These findings may be due to, at least in part, disorganized alignment of microstructure and immature formation of gap junction. 3187-Plat Spatiotemporal Transitions in Cardiac Neuronal Co-Cultures Rebecca A.B. Burton, Guy Stephens, Amy Sharkey, Sam Bilton, Hege Larsen, Holger Kramer, Carla Schmidt, Claudia Molina, Dan Li, Gary Mirams, Carol Robinson, David Paterson, Gil Bub. University of Oxford, Oxford, United Kingdom. A variety of arrhythmias have been attributed to formation of large scale pat- terns of excitation such as formation and break-up of spiral waves. We inves- tigate the relationship between neuronal activity and evolution of macroscopic spatiotemporal dynamics. We also aim to understand the role of target-derived trophic nerve growth factors (NGF) on the formation and function of synapses between sympathetic neurons and heart tissue. Hearts were isolated from neonatal rat pups, ventricular myocytes were iso- lated, cultured and after 24h, stellate ganglia were isolated from litter mates enzymatically digested, and neurons were plated on top on myocyte dishes. We used dye-free imaging techniques using to record wave pattern and beat- rate (in the presence/absence of NGF). For the first time, we report wave speeds in cardiac neuron co-cultures. Our re- sults show that (i) basal speed of co-cultures is significantly faster than myocyte only cultures (p<0.001); (ii) nicotine causes an altered behaviour in wave pat- terns; (iii) application of nicotine to co-culture causes a transient increase in contraction rate; (iv) the spatial organization of cells in myocyte monocultures is significantly altered by the addition of NGF. Our observations may be relevant to understanding how neuronal activity mod- ulates macroscopic cardiac function. The finding that NGF modulates the devel- opment and patterning of myocytes raises the question of how NGF and their receptors interact to lead to long term changes in cardiac and synaptic function. 630a Wednesday, February 19, 2014

Upload: issei

Post on 30-Dec-2016

217 views

Category:

Documents


3 download

TRANSCRIPT

Page 1: Electrical Propagation of Three-Dimensional Engineered Hearts using Decellularized Extracellular Matrix

630a Wednesday, February 19, 2014

fluorescent proteins to a voltage sensitive membrane protein (1). However, onefactor that has limited the utility of these indicators is poor voltage sensitivity,making it a challenge to express the indicators in neurons and still observedetectable responses (2). In addition, none of the genetically-encoded voltageindicators reported can be combined with channelrhodopsin for optical readoutof electrical activity in an all-optical electrophysiology setup. To address theselimitations, we set up a directed evolution strategy to screen for both voltageindicator brightness and voltage sensitivity. First, bacterial expression enablesus to screen thousands of clones in colonies of E. coli to improve the brightnessof the indicator. Second, a medium throughput mammalian electric field stim-ulation system allows us to test for function of the voltage indicators. Thisscreening has resulted in a family of red voltage indicators (VSDRs). Theyconsist of the voltage-sensing domain of Ciona intestinalis voltage-sensitivephosphatase linked to red fluorescent protein mApple. The fluorescence inten-sity of the VSDRs increases in response to depolarization. We show that thecombination of signal size and response speed of VSDRs allows the reliabledetection of spontaneous action potentials in cultured mammalian neurons insingle trials with widefield fluorescent light microscopy. Critically, we alsodemonstrate that VSDRs faithfully reports signals from all-optical electrophys-iology experiments using channelrhodopsin to depolarize mammalian cells.1. B. J. Baker et al., J. Neurosci. Methods. 2007, 161, 32-38.2. W. Akemann et al., Nat Methods. 7, 2010, 643-649.

3184-PlatOptogenetic Stimulation of Channelrhodopsin-2 Expressing Neurons FlipsCortical Networks from Low to High Activity StateParijat Sengupta, Lindsay Fague.ISP-Applied Sciences Laboratory, Washington State University, Spokane,WA, USA.Optogenetics, the single-component strategy for neuronal stimulation usinglight, allows for non-invasive and temporally precise stimulation of neurons.In our lab, we have recently developed an investigative tool that combines op-togenetic stimulation of channelrhodopsin-2 expressing neuronal networks(Figure-1: left panel; red: neuronal nucleus marker; violet: glial fibrillary acidicprotein; green: channelrhodopsin-2-YFP expressing neurons) with long-termelectrophysiological measurement frommultiple locations of the network usinga multi-electrode array (MEA) dish. This combinatorial approach can be usedto induce, detect and track short- and long-term changes in spontaneous andevoked neuronal network activity in response to optical stimuli. Using thistechnique we have found that even a single 20s-long excitation light pulsecan flip a dissociated cortical network from a low-activity state to a high-activity state (Figure-1: right, top panel). Interestingly, a single pulse doesnot seem to affect the levels of network activity if the network is already in

high-activity state (Figure-1: right, bottompanel). A more comprehensive study ofthis phenomenon and its relationship withnetwork plasticity is currently underway.Acknowledgement: Channelrhodopsin-2-YFP construct was a gift from Karl Deisser-oth lab at Stanford University.

Platform: Cardiac, Smooth, and Skeletal MuscleElectrophysiology

3185-PlatMyocyte Stretch Slows Cardiac Conduction by a Caveolae-Dependent In-crease in Sarcolemmal CapacitanceEmily Pfeiffer, Adam Wright, Andrew Edwards, Jennifer Stowe,Katie McNall, Justin Tan, Hemal Patel, Jeffrey Omens, Andrew McCulloch.UCSD, San Diego, CA, USA.Alterations in cardiac electrophysiology by mechanical perturbations havebeen long observed, however the cellular basis for these changes remainspoorly understood. Most studies have focused on pro-arrhythmic consequencesof stretch-activated currents (SAC). However, alterations in cardiac conductionthat can create an arrhythmogenic substrate have also been observed even in thepresence of SAC blockade. We tested the hypotheses that stretch slows conduc-tion by increasing cell membrane capacitance via recruitment and sarcolemmalintegration of caveolae and found that: (1) Conduction slowing occurred withstretch in both (a) pressure-loaded mouse hearts and (b) biaxially-stretched mi-cropatterned murine cardiomyocyte monolayers, (2) Inhibition of stretch-activated currents had no effect on this slowing in both models, (3) Slowingwas abrogated by caveolae depletion in both models, using cav3-knockoutmice or incubation with 1 mM MbCD, (4) Stretch significantly increased cellmembrane capacitance and time constant in single wild-type, but not

caveolae-depleted cells, and (5) These effects on cell membrane capacitanceand conduction slowing concide with recruitment and integration of caveolaeby stretch. Together, these data suggest that myocyte stretch promotes recruit-ment and integration of caveolae and causes slowing of cardiac conduction byincreasing cell membrane capacitance.

3186-PlatElectrical Propagation of Three-Dimensional Engineered Hearts using De-cellularized Extracellular MatrixHaruyo Yasui1, Jong-Kook Lee2, Akira Yoshida1, Teruki Yokoyama1,Junichi Nakai3, Issei Komuro4.1Cardiovascular Medicine, Osaka University Graduate School of Medicine,Suita, Japan, 2Cardiovascular Regenerative Medicine, Osaka UniversityGraduate School of Medicine, Suita, Japan, 3Brain Science Institute, SaitamaUniversity, Saitama, Japan, 4Cardiovascular Medicine, Tokyo UniversityGraduate School of Medicine, Tokyo, Japan.[Background] Construction of three-dimensional cardiac tissues using decellu-larized extracellular matrix could be a new technique to create an ‘‘organ-like’’structure of the heart. To engineer functional artificial hearts comparable for or-thotopic hearts, however, much remain to be solved including electrical stabil-ity for efficient contraction. To elucidate the points, we examinedelectrophysiological properties of recellularized heart tissues. [Methods]Entire hearts of adult Wistar rats were decellularized using 0.5% SDS and1% triton-X, and then recellularized with enzymatically-dispersed neonatalrat cardiac cells (1x10t8 cells) through antegrade coronary circulation. Threeand seven days later after cell seeding, we observed excitation of spontaneousand pacing-induced beatings of recellularized heart tissues expressing Ca2þ-indicating protein (GCaMP2) using high resolution cameras. We also conduct-ed immunofluorescence staining to examine morphological aspects of engi-neered tissues. [Results] Live tissue fluorescence imaging revealed that GFP-labeled-isolated cardiac cells were dispersed into interstitial spaces throughextravasation from coronary arteries. Engineered hearts seeded withGCaMP2-expressing cardiac cells started showing spontaneous beating, andwere subjected to further electrophysiological experiments using optical imag-ing system. Both in spontaneous and pacing-induced beating hearts, weobserved well-organized conduction of stable excitation in substantial areasof the engineered heart tissues, whereas we also recorded disorganized propa-gation of asynchronous excitation with multiple origins. Conduction velocitywas markedly decreased (~5 cm/sec) during early stages (up to nine days) afterrecellularization. Immunofluorescence study revealed randomly-mixed align-ment of cardiomyocytes, endothelial cells and smooth muscle cells, stainedwith alpha-actinin, CD31 and sm-actin, respectively. Recellularized heart tis-sues also showed disarray of cardiomyocytes and markedly decreased expres-sion of connexin43. [Conclusion] Three-dimensional engineered heartsshowed electrical heterogeneity and proarrhythmic propensity during earlystages of culture. These findings may be due to, at least in part, disorganizedalignment of microstructure and immature formation of gap junction.

3187-PlatSpatiotemporal Transitions in Cardiac Neuronal Co-CulturesRebecca A.B. Burton, Guy Stephens, Amy Sharkey, Sam Bilton,Hege Larsen, Holger Kramer, Carla Schmidt, Claudia Molina, Dan Li,Gary Mirams, Carol Robinson, David Paterson, Gil Bub.University of Oxford, Oxford, United Kingdom.A variety of arrhythmias have been attributed to formation of large scale pat-terns of excitation such as formation and break-up of spiral waves. We inves-tigate the relationship between neuronal activity and evolution of macroscopicspatiotemporal dynamics. We also aim to understand the role of target-derivedtrophic nerve growth factors (NGF) on the formation and function of synapsesbetween sympathetic neurons and heart tissue.Hearts were isolated from neonatal rat pups, ventricular myocytes were iso-lated, cultured and after 24h, stellate ganglia were isolated from litter matesenzymatically digested, and neurons were plated on top on myocyte dishes.We used dye-free imaging techniques using to record wave pattern and beat-rate (in the presence/absence of NGF).For the first time, we report wave speeds in cardiac neuron co-cultures. Our re-sults show that (i) basal speed of co-cultures is significantly faster than myocyteonly cultures (p<0.001); (ii) nicotine causes an altered behaviour in wave pat-terns; (iii) application of nicotine to co-culture causes a transient increase incontraction rate; (iv) the spatial organization of cells in myocyte monoculturesis significantly altered by the addition of NGF.Our observations may be relevant to understanding how neuronal activity mod-ulates macroscopic cardiac function. The finding that NGFmodulates the devel-opment and patterning of myocytes raises the question of how NGF and theirreceptors interact to lead to long term changes in cardiac and synapticfunction.