Introduction

Throughout an organism’s lifetime, cell mechanosensation (i.e., the ability to perceive and respond to mechanical stimuli in the form of shear stress, tension, or compression) is essential in a myriad of developmental, physiological, and pathophysiological processes, including embryogenesis, homeostasis, metastasis, and wound healing1. How these processes incorporate active feedback via force sensing at the cellular level is an area of active study, and a wide range of tools have been developed to interrogate cell mechanics2,3.

Stretch-activated ion channels, including the Piezo proteins, can respond to various external mechanical stimuli4,5. Most vertebrates have two Piezo genes, Piezo1 and Piezo24, and functional homologs have been identified both in plants6 and invertebrates7. While Piezo2 function is mainly restricted to the peripheral nervous system, Piezo1 is expressed in a wide range of tissues and has been shown to contribute to mechanotransduction in various organs5. Mutations in human Piezo1 have been implicated in diseases, such as dehydrated hereditary stomatocytosis8,9 and general lymphatic dysplasia10,11, and global knockout of Piezo1 in mice causes embryonic lethality12,13, highlighting the importance of this channel for development and homeostasis5. However, how cells and tissues integrate Piezo1 activity has been mainly examined by indirect outputs, such as morphological changes, protein expression, electrophysiological signaling, cytosolic calcium (Ca2+) imaging, and transcriptional activity in response to mechanical stimuli14.

Here, we rationally engineer and validate a genetically-encoded fluorescent reporter of Piezo1-dependent activity, named GenEPi, which has broad applicability across biological scales. We demonstrate that GenEPi has a high spatiotemporal resolution to report Piezo1-dependent activity from the single-cell level to that of the entire organism. We use Total Internal Reflection Fluorescence Microscopy (TIRFM) to showcase that GenEPi reveals transient, local mechanical stimuli in the plasma membrane of single cells. We demonstrate that GenEPi resolves repetitive contraction-triggered mechanical stimulation of beating cardiomyocytes within cardiac microtissues derived from mouse embryonic stem cells (ESCs). Finally, we validate GenEPi in vivo by generating a GenEPi zebrafish transgenic line which allows for robust and reliable monitoring of Piezo1-dependent activity using non-invasive optical imaging. Our work establishes GenEPi as a versatile and powerful tool for studying mechanosensitive processes, providing invaluable insights into Piezo1 dynamics in diverse biological contexts.

Results

Reporter engineering

In order to develop a non-invasive, genetically-encoded, mechanosensitive fluorescent reporter that is applicable to a wide variety of cells and types of mechanical stimuli, we set out to design a reporter of Piezo1 activity. It has been shown that the C-terminus of Piezo1 resides in the cytosol and contains the ion-permeating channel15,16, which has a preference for divalent cations, such as Ca2+ 15,16. Upon opening, Ca2+ concentration near the channel, referred to as the Ca2+ microdomain, is typically several-fold higher than resting levels17. We, therefore, hypothesized that by targeting a Genetically-Encoded Ca2+ Indicator (GECI) to the ion-permeating channel of Piezo1, we can obtain an optical readout for its activity.

We reasoned that a fluorescent reporter of channel activation would require a GECI with low Ca2+ affinity and a wide dynamic range to reliably monitor the considerable Ca2+ increase in the microdomains, while displaying a low response to cytosolic Ca2+, which serves as an important secondary messenger in many other cellular processes17. To meet these requirements, we decided to evaluate GCaMPs, a class of GECIs18, as fluorescent reporters of the Piezo1 function. In contrast to FRET-based GECIs, GCaMPs occupy a narrower spectral range, allowing for the simultaneous imaging of multiple fluorescent markers. Progressive protein engineering efforts have yielded GCaMP variants that display a wide dynamic range of response with high signal-to-noise ratios (SNR)19.

In a systematic screen, we generated a library of reporters by fusing five different low-affinity GCaMPs20,21 (here denoted as GCaMP-G1 to GCaMP-G5) (with Kd-s in the 0.6 to 6 µM range) to the C-terminus of human Piezo1 (Fig. 1a). Given the influence of linker length on the sensing mechanism22, we employed flexible linker peptides with varying lengths to attach GCaMPs to Piezo1 (Fig. 1b). The generated variants were evaluated based on their response to both mechanical stimuli and cytosolic Ca2+ fluctuations that were independent of Piezo1 activity. To test their responses to mechanical stimuli, variants were exposed to physiological levels of fluid shear stress23 (see Methods) (Fig. 1c), which causes a Piezo1-dependent Ca2+ increase in HEK293T cells12. To determine the sensitivity of the variants to intracellular Ca2+ levels independent of Piezo1 function, we recorded their response to the Ca2+ ionophore ionomycin (Fig. 1c)24. Among the candidates tested, we identified one Piezo1-GCaMP fusion variant that satisfied our requirements (here: robust response to shear stress) (Fig. 1d), Piezo1-1xGSGG-GCaMP-G4 (containing the GCaMP6s RS1 EF4 variant21, hereby referred to as GenEPi (Fig. 1c).

Fig. 1: Design and systematic screen to identify an optical reporter of Piezo1 activity.
figure 1

a Schematic of the reporter working principle. A low-affinity GCaMP is targeted near the C-terminal site of the Piezo1 channel. When mechanical stimuli induce channel opening, incoming Ca2+ (in yellow) binds to GCaMP, causing an increase in green fluorescence. b Design of the reporter screen. Five different GCaMPs with low affinity for Ca2+ (GCaMP-G1 to -G5) were fused to the C-terminus of human Piezo1 without any linker, with the addition of a short linker (i.e., 1xGly-Ser-Gly-Gly (1xGSGG) linker sequence) and long linker (i.e., 2xGSGG linker sequence). c Results of the reporter screen. The response of the resulting 20 variants (including the co-transfected cytosolic GCaMPs and human Piezo1) to shear stress (10 dyne/cm2, blue) and ionomycin (1 µM, grey) and identified Piezo1-1xGSGG-GCaMP-G4 (GenEPi) as a specific reporter for mechanical stimuli. eGFP fused to human Piezo1 serves as a control for the noise acquired during imaging under either stimulus. Data are presented as means ± SEM. Data from three independent experiments. For detailed statistical information for the screen, see Supplementary Table 3. d Summary of systematic screening results that illustrate which tested variant fulfilled our key requirements (here: higher response to shear stress and lower response to ionomycin administration compared to the cytosolic response). The only variant which fulfilled both initial requirements (statistically significant) was the fusion Piezo1-1XGSGG-GCaMP-G4 which is labeled with green shading. Source data are provided as a Source Data file.

Reporter characterization

The systemic expression of GenEPi required to confer mechanical sensitivity in HEK293T cells25 (Supplementary Fig. 1) did not affect their viability (Supplementary Fig. 2). The localization of GenEPi in plasma membrane and endoplasmic reticulum (ER) reflected that of wild-type Piezo1 while loss-of-function GenEPi-S217L mutant showed ER-retention and affected functionality as previously described in ref. 26 (Supplementary Note 1 and Supplementary Fig. 3).

The optical response of GenEPi (Supplementary Note 1) to fluid shear stress (Fig. 2a–c) was considerably higher (1.61 ± 0.09, mean ± SEM, n = 12 cells) than that of cytosolic GCaMP-G4 (1.36 ± 0.02, n = 13 cells) (Fig. 2c), indicating that channel tethering of GCaMP-G4 in this particular configuration provides optimal access to high Ca2+ levels upon Piezo1 channel opening. As GenEPi retained the low affinity for Ca2+ (Supplementary Fig. 4), it had a low level of response to cytosolic Ca2+ induced by ionomycin (1.16 ± 0.05, n = 15 cells), indistinguishable from the response levels of the control fusion protein, Piezo1-eGFP (1.12 ± 0.02, n = 19 cells) (Fig. 2c). In contrast, cytosolic GCaMP-G4 could not distinguish between shear stress and ionomycin and responded to both stimuli (Fig. 2c). Changing the level (1–30 dyne/cm2) or duration (10–120 s) of fluid shear stress did not result in any significant difference in GenEPi response (Supplementary Fig. 5), which confirms the highly cooperative nature of GCaMP-G4 to Ca2+ binding21. Notably, the tethering of all investigated GCaMP variants to the Piezo1 channel consistently reduced their response to cytosolic Ca2+ evoked by ionomycin (Fig. 1c) which suggests that genetically-encoded Ca2+ indicators placed near the channel are protected from cytosolic Ca2+ fluctuations, supporting the Ca2+ microdomain hypothesis17.

Fig. 2: GenEPi specifically reports Piezo1-dependent activity without affecting the functionality of the channel.
figure 2

a Representative example of GenEPi activation and b F/F0 signal intensity profile (black) in response to 10 dyne/cm2 shear stress (green) in HEK293T cells. Time stamps in the images correspond to the stimulation and response profile in the graph. Scale bar, 10 μm. c Response of HEK293T cells expressing Piezo1 and GCaMP-G4 (n = 13, shear stress; n = 18, ionomycin), GenEPi (n = 12, shear stress; n = 15, ionomycin), or Piezo1-eGFP (n = 16, shear stress, n = 19, ionomycin) to shear stress and ionomycin. Two-tailed Mann–Whitney test, ****p < 0.0001; *p < 0.05; n.s. = p > 0.05, data from three independent experiments. Data were presented as means ± SEM. d Response of GenEPi-expressing HEK293T cells to 10 μM Yoda1 (n = 14) or DMSO (n = 17). Two-tailed Mann–Whitney test, ****p < 0.0001, data from three independent experiments. Data were presented as means ± SEM. e Response of GenEPi and jRCaMP1a expressing HEK293T cells (n = 19) to intracellular Ca2+ triggered by 30 μM ATP. Two-tailed unpaired t-test, ****p < 0.0001, data from six independent experiments. Data were presented as means ± SEM. f Representative images of AFM cantilever stimulation of GenEPi-expressing HEK293T cells stimulated by the compressing AFM cantilever. Brightfield image of cantilever position before (top left) and during stimulation (top right) and corresponding fluorescent images of the stimulated cell before and after stimulation. Scale bar, 10 μm. g The mechanical stimulation procedure of compressive forces ranging from 100–400 nN (purple) along with the brightfield (gray) and fluorescent (green) traces from the cell depicted in f. h Amplitude of Ca2+ responses from GenEPi (n = 21), and Piezo-eGFP (n = 45) expressing cells. Data were presented as means ± SEM. Two-tailed Mann–Whitney test, ****p < 0.0001. i Amplitude of Ca2+ responses of GenEPi in cells transfected with GenEPi before (n = 13) and after addition of 3 μM GsMTx-4 (n = 11). Data were presented as means ± SEM. Two-tailed Mann–Whitney test, ****p < 0.0001. j Threshold forces and pressures for cells co-transfected with human Piezo1 and cytosolic GCaMP-G4 (n = 21), n.s. = p > 0.05, Data were presented as means ± SEM. Two-tailed unpaired t-test. k Duration of Ca2+ responses from cells co-transfected with cytosolic GCaMP-G4 and human Piezo1 (n = 27) and GenEPi (n = 16). Two-tailed Mann–Whitney test, ****p < 0.0001. Data were presented as means ± SEM. Data from three independent experiments. l Representative traces of the currents from wild-type Piezo1 (black) and GenEPi (green) evoked by the negative pressure of 0, −20, −40, −60, −80, and −100 mmHg at −80 mV with the cell-attached recording configuration. The current evoked by individual negative pressure was normalized to the maximum evoked by the negative pressure of −100 mmHg. m Both wild-type Piezo1 (n = 8 cells; 8 experiments) (black) and GenEPi (n = 6 cells; 6 experiments) (green) show a similar pressure-dependent response. Data were presented as means ± SEM. Two-tailed unpaired t-test, p = 0.11, p = 0.06, p = 0.20, and p = 0.07 for pressure sensitivities to −20, −40, −60, and −80 mmHg between Piezo1 and GenEPi. n Representative traces of wild-type Piezo1 (black) or GenEPi (green) currents evoked by −60 mmHg pressure at different voltages of +30, −30, −50, and −80 mV. o The conductance value for wild-type Piezo1 (black) is 24.9 ± 0.8 pS (n = 6 cells from six experiments, mean ± SEM) and the conductance value for GenEPi (green) is 25.5 ± 0.6 pS (n = 6 cells from six experiments, mean ± SEM). Both channels show the typical 25 pS conductance of the Piezo1 channel, indicating that the ion selectivity of Piezo1 is preserved within GenEPi. p Example traces of wild-type Piezo1 channels (black) or GenEPi (green) activated by 0.1, 0.3, 1, 3, 10 µM Yoda1 in the absence of mechanical stimulation. After the channel activities were evoked by 10 µM Yoda1, 10 µM ruthenium red (RR) was used to inhibit the currents (blue). Source data are provided as a Source Data file.

After we confirmed the functional specificity of GenEPi to the Piezo1-specific agonist Yoda127 (Fig. 2d and Supplementary Fig. 6) and the dependence of its response to extracellular calcium (Supplementary Fig. 7), we determined GenEPi response to physiological Ca2+ signaling in the cell upon addition of ATP (see Methods). We detected an ATP-dependent cytosolic Ca2+ increase using the Ca2+ indicator jRCaMP1a28 (Fig. 2e and Supplementary Fig. 8), yet these elevated Ca2+ levels were not detected by GenEPi (Supplementary Fig. 8). These results indicate that GenEPi is indeed responding specifically to Piezo1-dependent activity and does not sense physiological fluctuations of cytosolic Ca2+, whereas cytosolic Ca2+ indicators respond to both Piezo1-dependent and Piezo1-independent stimuli (Fig. 2c, 2e and Supplementary Fig. 8). The specificity of GenEPi response was further corroborated by the observation that membrane localization of GCaMP-G4 was not sufficient to confer functional specificity (Supplementary Note 2).

As Piezo1 is known to respond to other forms of mechanical stimuli, such as compression, we characterized the force sensitivity and temporal kinetics of GenEPi under this stimulus. We turned to a previously described Atomic Force Microscopy (AFM)-based setup29 that allows probing Piezo1 sensitivity to mechanical stimuli while simultaneously recording the optical response of GenEPi (Fig. 2f). We applied precisely timed compressive forces ranging from 100 to 400 nN in 50 nN increments on single HEK293T cells expressing GenEPi using a 5 µm bead attached to an AFM cantilever (Fig. 2g). These compressive forces are related to pressures ranging from 2.6 to 10.2 kPa or 19.1 to 76.5 mmHg (Supplementary Note 3). GenEPi responded to short (250 ms) compressive forces with fast kinetics, but on average with comparable signal amplitude to shear stress (1.65 ± 0.12, n = 21 cells) (Fig. 2h). GenEPi signals in response to compressive forces were abolished in the presence of the calcium channel inhibitor GsMTx-430 (Fig. 2i). In contrast, the Piezo1-eGFP fusion did not show any optical response (Fig. 2h).

To characterize the force sensitivity and duration of Piezo1-induced fluorescent signals reported by GenEPi and cytosolic GCaMP-G4, we applied timed compression onto GenEPi-transfected cells and control cells co-transfected with both human Piezo1 and cytosolic GCaMP-G4. Measured threshold forces were comparable for GenEPi and cytosolic GcaMP4 (243.50 ± 13.68 nN and 241.20 ± 13.87 nN, each n = 21 cells, respectively) (Fig. 2j), demonstrating that the mechanical sensitivity of the channel is not affected by the protein fusion. Notably, GenEPi response to the cantilever-triggered compression lasted on average 7.56 ± 1.09 s (n = 16 cells), which was much shorter than that of the cytosolic indicator (18.39 ± 1.84 s, n = 27 cells) (Fig. 2k).

To confirm that Piezo1 within GenEPi remains functional, we conducted a series of patch-clamp electrophysiology experiments. The pressure-dependent responses evoked by negative pressure applied in a cell-attached recording configuration were similar for both GenEPi and wild-type Piezo1 (Fig. 2l–m). Single channel measurements in cell-attached patches (Fig. 2n-o) showed that GenEPi has similar conductance values as wild-type Piezo1 (GenEPi, 25.5 ± 0.6 pS and Piezo1, 24.9 ± 0.8 pS), confirming that the conductivity of Piezo1 is not affected. Likewise, the channel kinetics of Piezo1 within GenEPi were preserved in response to repetitive mechanical stimulation (Supplementary Fig. 9), the agonist Yoda1 and the generic ion channel inhibitor Ruthenium Red (RR) (Fig. 2p and Supplementary Fig. 10). Furthermore, the electrochemical inactivation kinetic of GenEPi in response to mechanical stimuli was comparable to wild-type Piezo1 and shorter than that of the Piezo1 delayed inactivation mutant R2456H31 (Supplementary Fig. 11).

In conclusion, GenEPi provides high spatiotemporal resolution and functional specificity of Piezo1-dependent activity compared to cytosolic Ca2+ indicators without compromising the nature and functionality of the channel.

Analysis of transient, local mechanical stimuli on the membrane of cells

After the characterization under controlled experimental conditions (Fig. 2), we tested whether GenEPi can visualize Piezo1-dependent activity during autonomous cellular processes, such as local cellular traction forces, using total internal reflection fluorescence microscopy (TIRFM)32,33,34.

First, we tested the ability of GenEPi to resolve the localization dynamics of Piezo1 on the cell membrane of different cell types that transiently expressed GenEPi using the dox-inducible XLGenEPi plasmid (Supplementary Fig. 12) (see Methods, Molecular cloning, cell culture, and transfections). After doxycycline induction to temporally control the expression of the sensor, GenEPi was evenly distributed over the ventral surface of HEK293T, HFF, and HeLa cells (Fig. 3a), reflecting the previously reported wild-type Piezo1 membrane localization in cells32,33,34 (Supplementary Fig. 3). The number of GenEPi clusters per cell (Fig. 3b) was similar among the different cell types. Likewise, the area (Fig. 3c) and perimeter (Fig. 3e) of individual GenEPi clusters and their size distributions (Fig. 3d, f) did not differ between the cell lines, demonstrating that transient expression of GenEPi provides consistent localization dynamics in different cellular contexts.

Fig. 3: GenEPi captures Piezo1-dependent activity to transient, local mechanical stimuli on the cell membrane.
figure 3

a Representative TIRFM images from the cell-substrate interface of live HEK293T, HFF, and HeLa cells expressing GenEPi. Scale bar, 10 μm. b Number of GenEPi clusters per cell in HEK293T, HFF, and HeLa cells expressing GenEPi. n = 13 cells, error bar: SEM. Ordinary one-way ANOVA test. cf Quantification of GenEPi cluster area and perimeter as violin plots in HEK293T, HFF, and HeLa cells expressing GenEPi. HEK293T cells (n = 279), HFF cells (n = 503), HeLa cells (n = 669); n = 1 represent a single GenEPi (Piezo1) cluster. Kruskal–Wallis test. g Tracking of GenEPi (Piezo1) clusters imaged at 10 frames per second in live HEK293T cells reveals the motility of the channel clusters. The background image shows the fluorescence of GenEPi clusters captured during a single imaging frame (rectangular region (G) in image i, two-pixel median filter applied in FiJi/ImageJ (see Methods, Image processing and analysis). White lines depict the tracks of these clusters over several successive frames. Scale bar, 1 μm. h Mean-squared displacement calculated from 5332 GenEPi tracks plotted as a function of time. Data fit to a straight line with a slope corresponding to a two-dimensional (2D) diffusion coefficient of 0.0044 µm2/s. R2 for linear fit to data is 0.9969. i Representative time-lapse images of GenEPi (Piezo1) cluster dynamics in a single HEK293T cell expressing GenEPi. i1i3 (i1) Region without GenEPi cluster adjacent to a GenEPi cluster. (i2) Region with a GenEPi cluster which shows dynamic behavior and changes in fluorescence. (i3) Region with a GenEPi cluster which does not show dynamic behavior. Scale bars, 10 and 1 μm, respectively. j Raw values of fluorescence intensity profiles of the ROIs i1–i3 (Trace i1, Trace i2, and Trace i3) which show different GenEPi activity dynamics. k Fluorescence intensity profiles (F/F0) of the ROIs i1–i3 which illustrate the different GenEPi activity dynamics. Source data are provided as a Source Data file.

As wild-type Piezo1 has been previously shown to readily diffuse in the cell membrane32,33,34, we tested whether GenEPi shows similar dynamics. Time-lapse TIRFM imaging revealed that individual GenEPi clusters were mobile in the plasma membrane (Fig. 3g–i and Supplementary Movie 1). To obtain apparent diffusion coefficients, we tracked GenEPi in the plasma membrane to build trajectories of individual clusters (Fig. 3g) and plotted the Mean-Squared Displacement (MSD) of 5332 GenEPi tracks (Fig. 3h). The slope of the MSD yielded an apparent two-dimensional (2D) diffusion coefficient of ~0.005 μm2/s, which is in the range of previously reported values for wild-type Piezo132,33,34.

Piezo1 function has been reported to be related to autonomous cell processes in the absence of externally applied mechanical forces, such as local cellular traction forces32. Indeed, we observed distinguishable GenEPi responses over the ventral surface of the cells (Supplementary Fig. 13). Notably, some of the GenEPi responses lasted for several seconds (Fig. 3i and trace i2 in 3j, k). These responses are comparable in duration to GenEPi activation in response to local mechanical forces applied by an AFM cantilever (Fig. 2k) and could be the source of previously reported calcium flickers32,35. Taken together, GenEPi robustly reports diffusion dynamics comparable to wild-type Piezo1 channels and captures Piezo1-dependent activity to transient, local mechanical stimuli on the membrane of the cells.

Analysis of contraction-triggered mechanical stimulation in microtissues

In order to test the functional specificity and performance of GenEPi in an in vitro three-dimensional (3D), multicellular environment, we tested its response to homeostatic cell motions, such as cardiomyocyte contraction where Piezo1 has been shown to act as a key mechanotransducer36. To this end, we generated doxycycline-inducible GenEPi mouse embryonic stem cells (mESCs)37 (Supplementary Fig. 14) and differentiated these cells into cardiomyocytes38. We confirmed GenEPi activity in undifferentiated mESC by monitoring its specific response to Yoda1 (Supplementary Fig. 14). After 10 days of differentiation (Supplementary Fig. 15), regular beating patches of cells could be identified in microtissues (Supplementary Movie 2) consisting predominantly of cardiomyocytes and other mesodermal lineage cells, such as endothelial cells (Supplementary Fig. 15). After transient induction of GenEPi expression, we observed cells that displayed noticeable GenEPi responses due to contraction-triggered mechanical stimulation within beating patches. Differentiated microtissues with multiple beating patches caused a robust systemic activation of GenEPi in response to the autonomous regular contraction of cardiomyocytes (Fig. 4a). Notably, in dissected microtissue, where single cells were attached to autonomously beating cardiomyocytes, GenEPi responses were restricted to places of local membrane displacement (Fig. 4b and Supplementary Fig. 16). In both conditions, the response amplitude range (F/F0 = 1.15 to 2.94) was comparable to that of shear stress and compressive forces, while the sub-second responses were qualitatively coupled to the autonomous regular beating of the cardiomyocytes (Fig. 4b, Supplementary Fig. 17, and Supplementary Movie 3).

Fig. 4: GenEPi reports cardiomyocyte contraction-triggered mechanical stimulation with high spatiotemporal resolution.
figure 4

a Multicellular and systemic activation of GenEPi in response to autonomous contraction of cardiomyocytes in differentiated tissue within multiple beating patches. Representative time-lapse fluorescence intensity (F/F0) images and profile of multiple responding cells embedded in the differentiated tissue of beating cardiomyocyte patches. Scale bar, 10 μm. b Single cell local responses of GenEPi in a dissected beating patch in response to the autonomous beating of cardiomyocytes. b1, b2 Representative time-lapse fluorescence intensity (F/F0) images and profiles of a responding (b1) and a non-responding (b2) ROI in a single cell attached to the beating cardiomyocyte patch. Scale bar, 5 μm. c Representative fluorescence intensity (F/F0) profile from an ROI with systemic activation of GenEPi in response to cardiomyocyte contraction before (control) and after the addition of 100 μM blebbistatin. d Frequency of peaks, e amplitude of fluorescence intensity (F/F0) changes and f median ± SEM values of d and e before (control) and after the addition of norepinephrine (range of concentrations, 10 nM–10 μM). n = 6 ROIs, error bar: SEM. Ordinary one-way ANOVA and Kruskal–Wallis test, respectively. Data from three independent experiments. g Representative fluorescence intensity (F/F0) profiles from an ROI with GenEPi activation in response to cardiomyocyte contraction after the addition of norepinephrine (range of concentrations, 10 nm–10 μM). h Representative fluorescence intensity (F/F0) profile from an ROI with systemic activation of GenEPi in response to cardiomyocyte contraction before (control) and after the addition of 100 nM nifedipine. i Beating rate of cardiac microtissues before and after the addition of 100 nM nifedipine which abolishes the contraction of the beating cardiomyocytes. n = 17 measurements from three independent biological replicates, error bar: SEM. Two-tailed Wilcoxon rank-sum test. j Amplitude of fluorescence intensity (F/F0) changes before (control) and after the addition of 100 nM nifedipine. n = 101 and n = 20 measurements for control and nifedipine treatments, respectively, from three individual biological replicates, error bar: SEM. Two-tailed Mann–Whitney test. k Baseline fluorescence (F0) before and after the addition of 100 nM nifedipine. n = 24 ROIs containing multiple cells for each condition from 3 individual biological replicates, error bars: SEM. Two-tailed Mann–Whitney test. l Median ± SEM values of (i) and (j) before (control) and after the addition of 100 nM nifedipine. Source data are provided as a Source Data file.

In order to confirm that the source of GenEPi responses were indeed cardiomyocyte contractions, we applied blebbistatin, a myosin inhibitor, which blocks the contractions and uncouples mechanical stimuli-induced Ca2+ influx from Ca2+ processes accompanying spontaneous cell contractions39. Fast GenEPi responses decreased when contractions stopped in response to blebbistatin (Fig. 4c), as demonstrated by the significant decrease in the amplitude and frequency of the GenEPi response (Supplementary Fig. 18), confirming cardiomyocyte contractions as the source of GenEPi signals.

To complement our analysis, we tested the ability of GenEPi to respond and resolve higher-frequency cardiomyocyte contractions. To this end, we employed norepinephrine, a ligand which activates adrenoreceptor signals and increases the contraction rate in cardiomyocytes40. We indeed observed a significant increase in the frequency and the amplitude of GenEPi responses in proportion to the norepinephrine regime (10 nΜ to 10 μM) (Fig. 4d–g), validating the ability of GenEPi to temporally resolve cardiomyocyte contractions. Additionally, to further demonstrate that other channels, such as L-type channels, do not contribute to the response of GenEPi, we treated differentiated beating cardiac microtissues with nifedipine which is known to block most L-type channels and has a strong negative ionotropic effect41,42,Full size image

As zebrafish are potent models for drug and toxicity screening74,75 and only a few modulators of Piezo1 activity are known27,76, we assessed the activity of GenEPi upon pharmacological intervention in vivo. As a proof-of-principle, we used the agonist Yoda1 and the generic ion channel inhibitor RR to probe Piezo1 function following a similar approach to our in vitro analyses and electrophysiology experiments (Fig. 2d, p). Heat-shocked Tg(hsp70:GenEPi) zebrafish embryos in egg medium (E3 medium) were sequentially incubated in 10 μM Yoda1, 10 μM Yoda1 and 10 μM RR, and 10 μM RR followed by washing out in E3 medium (see Methods, Zebrafish transgenesis and in vivo pharmacological intervention experiments). To enable rapid interpretation of GenEPi response over a large field of view, we used a widefield fluorescence microscope. We focused on the develo** eye, a large organ in direct contact with the egg medium easily accessible for imaging and pharmacological interventions. Heat-shocked Tg(hsp70:GenEPi) fish showed instantaneous and significant fluorescence increase in response to 10 μM Yoda1, whereas transfer to a solution containing 10 μM Yoda1 and 10 μM of RR inhibited GenEPi activation (Fig. 5g, h), consistent with our electrophysiology results (Fig. 2p). Zebrafish transferred to 10 μM RR demonstrated further decrease in fluorescence intensity (Fig. 5g, h). Notably, washing out of the zebrafish in the original E3 medium restored the fluorescence of GenEPi to pre-treatment activity levels (Fig. 5g, h), demonstrating that GenEPi retains the pharmacological properties of Piezo1.

We previously showed (Fig. 4) that GenEPi can report cardiomyocyte contraction-triggered mechanical stimulation with high spatiotemporal resolution within 3D cardiac microtissues. To confirm that GenEPi can also monitor Piezo1-dependent activity during heart beating in vivo, we imaged heat-shocked Tg(hsp70:GenEPi) zebrafish embryo hearts at 3dpf (Supplementary Movie 4). GenEPi is strongly expressed in both cell layers of the develo** zebrafish heart (Fig. 5i) once fluorescence reaches a steady state. Performing dynamic time-lapse widefield imaging, we observed a mechanical stimuli-dependent activation of GenEPi in the atrioventricular canal (AVC) of the heart (Fig. 5j–l and Supplementary Fig. 22). The GenEPi activation was abolished when the zebrafish heart was stopped with 2,3-Butanedione 2-monoxime (BDM) treatment (Fig. 5j–l), confirming the mechanical nature of GenEPi activation.

Overall, we confirm that the ability of GenEPi to resolve Piezo1-dependent activity is preserved in vivo and demonstrate a robust and reliable in vivo GenEPi system that is able to generate information-rich and physiologically relevant readouts.