Introduction

Muscle LIM protein (MLP), encoded by CSRP3, is primarily expressed in cardiomyocytes and skeletal muscle cells and consists of 194 amino acids with two LIM domains1. These LIM domains mediate interactions with a variety of proteins in different subcellular regions, including the cytoplasm and the nucleus. In the cytoplasm of cardiomyocytes, MLP has been found in the sarcomeres, intercalated disks, and costameres of striated myocytes. In addition, MLP can directly affect F-actin cytoskeleton dynamics by enhancing CFL2-dependent F-actin depolymerization2 and stabilizing actin filaments into bundles3. These studies suggest that MLP plays an integral role in sarcomeric structure. Previous studies have shown that mutations (missense, small insertions, and deletions) in CSRP3 are associated with hypertrophic cardiomyopathy (HCM)4,5 and dilated cardiomyopathy (DCM)6 in humans. In addition, MLP has been shown to be significantly downregulated in chronic human heart failure (HF)7. In mouse models, MLP-deficient mice develop dilated DCM with hypertrophy after birth, and are the first transgenic animal model of HF1. However, the underlying molecular mechanisms of CSRP3 mutation-induced cardiomyopathy are not well understood.

Despite the existence of genetically modified mouse models for MLP deficiency1,8, phenotypic results have shown significant discrepancies between identical CSRP3 mutations in animals and humans. For example, MLPW4R/+ transgenic mice are observed to develop features of HCM and HF8, whereas MLPW4R/+ is a known to be a benign polymorphism in humans carrying the mutation4,9. Efforts to obtain primary human cardiac tissue to resolve these discrepancies have not been successful due to difficulties in obtaining and culturing human cardiac tissue in vitro. Advances in disease modeling using human pluripotent stem cells (PSCs) in recent years have provided a novel pathophysiological approach to study the mechanics of disease and discover new targets for pharmacological intervention10.

We thus created a MLP-deficient cardiomyocyte model using from CSRP3−/− hESC-H9 cells, to elucidate the mechanism of cardiomyopathy and HF caused by MLP defects. We report here that MLP-deficient human embryonic stem cell-derived cardiomyocytes (hESC-CMs) recapitulate the phenotypes of HCM and HF. Mechanistically, impaired calcium handling is a major mechanism underlying the pathogenesis of MLP-associated HCM and HF, which can be prevented by pharmaceutical blockade of calcium entry into the cell.

Results

Generation of homozygous CSRP3 −/− hESCs

To generate the human PSC model of MLP deficiency, we designed a single-guide RNA targeting exon 3 of CSRP3 using the epiCRISPR/Cas9 gene editing system (Fig. 1a). H9 hESCs were then electroporated with a plasmid containing sgRNA and epiCRISPR/Cas9

Fig. 2: Phenoty** hypertrophic cardiomyopathy in MLP-deficient hESC-CMs.
figure 2

a Immunostaining of sarcomeric α-actinin (green) and cTnT (red) demonstrates sarcomeric disarray in MLP KO CMs at day 30. Scale bar, 50 μm. b Compared with WT hESC-CMs (n = 189), a significant higher percentage of MLP KO hESC-CMS (n = 191) showed disorganized sarcomeric α-actinin staining pattern in greater than one fourth of the total cellular area. c Images of α-actinin/DAPI-immunostained hESC-CMs and d quantification of mono-, bi-, and multi-nucleation in WT (n = 267) and MLP KO (n = 259) hESC-CMs. Scale bar, 50 μm. e, f Calibration of forward scatter (FSC; 10,000 cells/sample, n = 3) showing an increased cellular size beginning 22 days post cardiac differentiation in MLP KO CMS. f Results are presented as means ± S.E.M. of three independent experiments. *P < 0.05; **P < 0.01; ns, not significant, unpaired two-sided Student’s t test

MLP deficiency leads to impaired calcium handling

Calcium handling plays a fundamental role in regulation of excitation–contraction in both skeletal and cardiac myocytes, dysfunction of which is commonly involved in the development of HCM and HF10,23. To examine the impact of MLP deficiency on calcium handling, we applied nickase Cas9 to introduce the green fluorescent calcium-modulated protein 6 fast type (GCaMP6f) calcium sensor into the AAVS1 locus of the WT and MLP KO H9 hESC lines as previously reported24 (Fig. 3a). Recording of calcium transients at days 15, 22, and 30 (Fig. 3b, c) show that compared with WT hESC-CMs, MLP KO hESC-CMs have slower beating rate (Figs. 3c and S3A). At day 15, the calcium release amplitude of MLP KO CMs was significantly higher than WT CMs, but gradually decreased (Fig. 3c, d). At day 30, compared with WT hESC-CMs, MLP KO hESC-CMs exhibited lower calcium release amplitude and longer time to peak (Fig. 3c–e). Beginning at day 22, MLP KO hESC-CMs started to demonstrate longer transient durations (Fig. 3c, f) and prolonged decay times (Fig. S3B) compared with WT hESC-CMs. Adjustment to heart rate variation did not impact prolonged transients of MLP KO hESC-CMs as compared with WT hESC-CMs (Fig. S3C). Interestingly, increased calcium release amplitude was observed to occur in MLP KO CMs before cellular hypertrophy (Figs. 2f and 3d), suggesting that abnormal Ca2+ handling may be a causal factor for the induction of HCM phenotype in MLP-deficient hESC-CMs.

Fig. 3: MLP-deficient hESC-CMs exhibit abnormal Ca2+ handling properties.
figure 3

a Schematic demonstrating the GCaMP-expression cassette that was integrated into AAVS1 of WT and MLP KO hESCs via nickase CRISPR/Cas9 editing. b Space-averaged calcium transients showing parameters measured for analysis of calcium handling. c Representative line-scan images in WT-GCaMP and MLP KO-GCaMP hESC-CMs at days 15, 22, and 30. df Quantification of peak, time to peak, and calcium transient duration in WT-GCaMP and MLP KO-GCaMP hESC-CMs (n = 15 cells per group). g Representative Ca2+ transients induced with 10 mM caffeine in Ca2+-free conditions between WT-GCaMP and MLP KO-GCaMP hESC-CMs at days 15, 22, and 30 (n = 5 cells per group). hj Peak amplitude, transient duration, and decay time in caffeine-evoked Ca2+ transients WT-GCaMP and MLP KO-GCaMP hESC-CMs. Results are presented as means ± S.E.M. of three independent experiments. *P < 0.05; **P < 0.01; ***P < 0.001; ****P < 0.0001; ns, not significant, unpaired two-sided Student’s t test

To further profile calcium handling properties, we also measured caffeine-induced calcium release from the SR of WT and MLP KO hESC-CMs at days 15, 22, and 30 (Figs. 3g and S3D). Beginning at day 22, MLP KO hESC-CMs exhibited relatively smaller amplitudes, longer duration, and prolonged decay time compared with WT hESC-CMs (Figs. 3g, j and S3D), suggesting that MLP KO hESC-CMs have reduced SR calcium storage and impaired function of Ca2+ pum** and Ca2+ release channels in the SR and plasma membranes. Consistently, increased CACNA1C and RYR2 expression (Fig. S3E, F) and decreased SERCA 2a levels were observed in MLP KO hESC-CMs by qPCR (Fig. S3G) and western blot (Fig. S3H, I). These results indicate that MLP deficiency decreases the rate of calcium uptake and release in hESC-CMs as demonstrated in other models of HCM25,26 and HF27.

MLP-deficient cardiomyocytes exhibit elevated activity of hypertrophic signaling pathways

To understand how MLP deficiency leads to cardiomyopathy, we next accessed the expression of a panel of cardiomyopathy related genes at day 30 by qRT-PCR. Compared with WT hESC-CMs, MLP KO hESC-CMs displayed increased expression of genes involved in fetal development, calcium handling, hypertrophic signaling pathways, and autophagy (Fig. 4a–c). From this transcriptional data, we found significantly elevated activation of hypertrophic signaling in MLP KO hESC-CMs. To further examine signaling pathways involved28,29, we performed western blot for Ca2+-calcineurin–NFAT and Ca2+-Calmodulin-CaMKII. As compared with WT hESC-CMs, MLP KO hESC-CMs exhibited higher levels of P-CaMKII, which is a major mediator of hypertrophic signaling (Fig. 4d, e, g). Furthermore, MLP KO hESC-CMs exhibited higher level of MYH7, cTnT, and α-actinin, consistent with increased sarcomeric gene expression, which is a known feature of HCM (Fig. 4d, h–j)19.

Fig. 4: Increased gene expression of hypertrophic signaling pathways in MLP-deficient hESC-CMs.
figure 4

a Heatmap showing changes in the expression of genes involved in HCM signaling pathways, calcium handling, fibrosis, and autophagy in WT and MLP KO hESC-CMs at day 30. b qRT-PCR analysis of HCM related genes in WT and MLP KO CMs at day 30. c Heatmap showing the expression of HCM associated genes at days 15, 22, and 30 of cardiac differentiation. d Immunoblot analysis of HCM signaling (calcineurin A, CaMKIIδ, and phosphorylated CaMKII) and sarcomere proteins (MYH7, α-actinin, and cTnT) in WT and MLP KO hESC-CMs at day 30. ej Quantification of calcineurin A, CaMKIIδ, P-CaMKII, MYH7, α-actinin, and cTnT normalized by GAPDH in WT and MLP KO hESC-CMs. Results are presented as means ± S.E.M. of three independent experiments. *P < 0.05; **P < 0.01; ***P < 0.001; ****P < 0.0001; ns, not significant, unpaired two-sided Student’s t test

Because clinical symptoms of HCM usually take decades to manifest in affected individuals30, we also examined temporal gene expression in MLP KO hESC-CMs as compared with WT hESC-CMs by qRT-PCR. We found that from day 15 to day 30, the relative expression of hypertrophy related genes gradually increased in MLP KO hESC-CMs (Figs. 4c and S4A–G), and began to pick up in earnest around day 22 for genes such as MYH6, MYH7, MYL2, MYL7, and TNNT2, while NPPA and NPPB were significantly increased by day 30 (Figs. 4c and S4A–G).

MLP-deficient CMs develop mitochondrial dysfunction associated with HF

Previous studies have shown that cardiomyopathies and HF are commonly associated with pathological events such as mitochondrial dysfunction, energy depletion, increased generation of ROS, and cardiac dysfunction31,32. MLP-deficient mice have also been shown to exhibit regional absence of mitochondria and energy depletion33. In addition, increased intracellular basal [Ca2+] caused by MLP deficiency has been shown to damage mitochondria34. Thus, we next quantified mitochondrial content in MLP KO and WT hESC-CMs by comparing the amount of the mitochondrial-specific ND1 and ND2 genes to the housekee** ACTB (encoding β-actin) at days 15, 22, and 3035. Beginning at day 30 post differentiation, MLP KO hESC-CMs exhibited a significant decrease in mtDNA/nDNA ratio compared with WT hESC-CMs (Fig. 5a). To further investigate the impacts of MLP deficiency on the metabolism of hESC-CMs, we next assessed mitochondrial content, total cellular ROS, and mitochondrial-specific ROS by flow cytometry using Mitotracker, Cell ROS green, and MitoSOX, respectively, at day 30. Compared with WT hESC-CMs, the mitochondrial content of MLP KO CMs was significantly reduced (Fig. 5b–d). Moreover, the staining pattern of Mitotracker in MLP KO hESC-CMs demonstrated that many mitochondria were punctate and fragmented (Fig. S5A) as compared with normal linear morphology in WT hESC-CMs, indicating that MLP KO hESC-CMs had hallmarks of mitochondrial damage. Flow cytometry assessment of cell ROS (Fig. 5e–g) and MitoSOX (Fig. 5h–j) staining demonstrated that both total cellular and mitochondrial-specific ROS are significantly elevated in MLP KO hESC-CMs, which is consistent with features of mitochondrial injury32.

Fig. 5: Mitochondrial damage and increased ROX in MLP-deficient CMs.
figure 5

a qPCR analysis of mitochondrial DNA (ND1 and ND2) to nuclear DNA (β-actin) ratio at days 15, 22, and 30 of cardiac differentiation (n = 9). b Mitotracker images and c, d quantification of Mitotracker green intensity obtained by flow cytometry demonstrates a significantly reduced fluorescence intensity in MLP KO hESC-CMs at day 30 as compared with WT hESC-CMs (n = 3). Scale bar, 50 μm. eg Cell ROS green intensity and hj MitoSox Red intensity suggests increase in ROS for MLP KO hESC-CMs as compared with WT hESC-CMs (n = 3). Scale bar, 50 μm. Results are presented as means ± S.E.M. of three independent experiments. **P < 0.01; ***P < 0.001; ****P < 0.0001; ns, not significant, unpaired two-sided Student’s t test

Since damaged mitochondria can lead to decreased ATP production and increased AMP/ATP ratio, we next determined the functional change of mitochondria by AMP-activated protein kinase (AMPK) activity, a “metabolic sensor”, that can be activated by ATP depletion and increased AMP/ATP ratio36. MLP KO hESC-CMs had a significantly higher P-AMPK levels compared with WT CMs at day 30 (Fig. S5B, C). These findings suggest that MLP deficiency may lead to HF phenotypes due to mitochondrial damage, increased ROS, and energy depletion.

MLP-deficient cardiomyocytes exhibit early failure after isoproterenol treatments

Chronic administration of β-adrenergic agonists, such as isoproterenol37, have been shown to aggravate HCM and induce HF in HCM models of disease38. Deletion of the β2-adrenergic receptor39 and overexpression of a β-adrenergic receptor kinase 1 inhibitor40 has been shown to prevent the development of cardiomyopathy in MLP-deficient mice. We thus examined whether treatment with ISO could accelerate the development of HCM disease phenotype in MLP KO hESC-CMs. One week of 10 μM ISO treatment from the day 15 of cardiac differentiation resulted in markedly increased calcium release amplitudes of WT hESC-CMs and significantly decreased calcium release amplitudes in MLP KO hESC-CMs (Fig. 6a–c). ISO treatment did not result in significant changes of time to peak in WT and MLP KO hESC-CMs (Fig. 6a, b, d). Although ISO treatment can reduce the duration of calcium transients both in WT and MLP KO hESC-CMs, the duration of calcium transients in MLP KO hESC-CMs was still maintained at higher levels than in WT hESC-CMs (Fig. 6a, b, e). Gene expression of cardiac hypertrophy related genes such as MYH6, TNNT2, NPPA, and NPPB was modestly induced in ISO-treated WT hESC-CMs, but was found to be markedly elevated in MLP KO hESC-CMs (Figs. 6f and S6A–G). These results suggest that β-adrenergic stimulation can exacerbate HCM disease phenotypes in MLP-deficient hESC-CMs.

Fig. 6: Exacerbation of HF in response to β-adrenergic stimulation in MLP-deficient hESC-CMs.
figure 6

a, b Representative line-scan images from WT and MLP KO hESC-CMs after 1 week of 10 μM ISO treatment beginning at day 15 of cardiac differentiation. ce Quantification of peak, time to peak, and calcium transient duration in WT, MLP KO, WT/ISO, and MLP KO/ISO hESC-CMs (WT n = 13, WT/ISO n = 15, MLP KO n = 13, and MLP KO/ISO n = 12). e Heatmap representations of hypertrophy related gene expression in WT, MLP KO, WT/ISO, and MLP KO/ISO hESC-CMs (n = 3). Results are presented as means ± S.E.M. of three independent experiments. *P < 0.05; **P < 0.01; ***P < 0.001; ****P < 0.0001; ns, not significant, unpaired two-sided Student’s t test

Treatment of Ca2+ dysregulation prevents development of the HCM phenotype in MLP-deficient cardiomyocytes

As Ca2+ dysregulation was observed to occur before other hallmarks of HCM disease phenotypes, we next evaluated whether pharmaceutical inhibition of calcium entry with the L-type Ca2+ channel blocker verapamil could prevent the development of HCM phenotype in MLP KO hESC-CMs. Continuous addition of verapamil using therapeutic dosages (100 nM) for 10 days beginning at day 20 of cardiac differentiation was found to significantly improve impaired calcium handling function in MLP KO hESC-CMs, including increased rate of calcium uptake and release (Fig. 7a–d). Consistent with higher calcium amplitudes (Fig. 7b), increased SR Ca2+ storage capacity was also observed in verapamil-treated MLP KO hESC-CMs as compared with nontreated cells (Fig. S7A–C). Assessment of cell size and mitochondrial content by flow cytometry showed that administration of verapamil ameliorated cell enlargement (Fig. 7e, f) and mitochondrial damage (Fig. 7g, h) in MLP KO hESC-CMs. Treatment with verapamil was also found to downregulate the expression of genes related to cardiac hypertrophy (Figs. 7i and S7D) and reduce activation of the hypertrophic signaling pathways (Fig. 7j, k). These results suggest early intervention to treat calcium handling deficiencies can help prevent the development of HCM disease phenotypes in MLP-deficient hESC-CMs.

Fig. 7: Verapamil rescued HCM phenotype in MLP-deficient CMs.
figure 7

a Representative line-scan images in WT (blue), MLP KO (red), and MLP KO hESC-CMs treated with 100 nM verapamil (green) for 10 continuous days beginning at day 20 of cardiac differentiation (n = 13). bd Quantification of peak, time to peak, and calcium transient duration in WT, MLP KO, and verapamil-treated MLP KO hESC-CMs (n = 12). e, f Forward scatter (FSC) and g, h Mitotracker green intensity obtained by flow cytometry in WT, MLP KO, and verapamil-treated MLP KO hESC-CMs. i Heatmap representations of hypertrophy related gene expression in WT, MLP KO, and verapamil-treated MLP KO hESC-CMs. j, k Immunoblot analysis of P-CaMKII and quantification of P-CAMKII normalized by GAPDH in WT, MLP KO, and verapamil-treated MLP KO hESC-CMs (n = 3). Results are presented as means ± S.E.M. of three independent experiments. *P < 0.05; **P < 0.01; ***P < 0.001; ****P < 0.0001; ns, not significant, unpaired two-sided Student’s t test