Dysferlin Enables Tubular Membrane Proliferation in Cardiac Hypertrophy
Abstract
BACKGROUND:
Cardiac hypertrophy compensates for increased biomechanical stress of the heart induced by prevalent cardiovascular pathologies but can result in heart failure if left untreated. Here, we hypothesized that the membrane fusion and repair protein dysferlin is critical for the integrity of the transverse-axial tubule (TAT) network inside cardiomyocytes and contributes to the proliferation of TAT endomembranes during pressure overload–induced cardiac hypertrophy.
METHODS:
Stimulated emission depletion and electron microscopy were used to localize dysferlin in mouse and human cardiomyocytes. Data-independent acquisition mass spectrometry revealed the cardiac dysferlin interactome and proteomic changes of the heart in dysferlin-knockout mice. After transverse aortic constriction, we compared the hypertrophic response of wild-type versus dysferlin-knockout hearts and studied TAT network remodeling mechanisms inside cardiomyocytes by live-cell membrane imaging.
RESULTS:
We localized dysferlin in a vesicular compartment in nanometric proximity to contact sites of the TAT network with the sarcoplasmic reticulum, a.k.a. junctional complexes for Ca2+-induced Ca2+ release. Interactome analyses demonstrated a novel protein interaction of dysferlin with the membrane-tethering sarcoplasmic reticulum protein juncophilin-2, a putative interactor of L-type Ca2+ channels and ryanodine receptor Ca2+ release channels in junctional complexes. Although the dysferlin-knockout caused a mild progressive phenotype of dilated cardiomyopathy, global proteome analysis revealed changes preceding systolic failure. Following transverse aortic constriction, dysferlin protein expression was significantly increased in hypertrophied wild-type myocardium, while dysferlin-knockout animals presented markedly reduced left-ventricular hypertrophy. Live-cell membrane imaging showed a profound reorganization of the TAT network in wild-type left-ventricular myocytes after transverse aortic constriction with robust proliferation of axial tubules, which critically depended on the increased expression of dysferlin within newly emerging tubule components.
CONCLUSIONS:
Dysferlin represents a new molecular target in cardiac disease that protects the integrity of tubule-sarcoplasmic reticulum junctional complexes for regulated excitation-contraction coupling and controls TAT network reorganization and tubular membrane proliferation in cardiomyocyte hypertrophy induced by pressure overload.
Graphical Abstract
In This Issue, see p 551
Meet the First Author, see p 552
Dysferlin, a 238-kDa transmembrane protein with multiple Ca2+-binding C2-domains, mediates Ca2+-dependent membrane fusion and repair events in striated muscle cells,1 as previously reviewed.2–5 Biallelic loss-of-function mutations in the dysferlin gene cause skeletal muscle diseases, referred to as dysferlinopathies, such as Miyoshi muscular dystrophy,6 limb-girdle muscular dystrophy type 2B,7 and distal myopathy with anterior tibial onset.8 In addition to the skeletal myopathies, mild forms of dilated cardiomyopathy can occur,9–11 attributed to a to date hypothetical protective role of dysferlin in cardiomyocytes. Dysferlin-mediated membrane repair was suggested to maintain the integrity of postmitotic cardiomyocytes in the context of recurring sarcolemmal lesions during heart contractions and particularly under conditions of increased biomechanical stress, aiding in the prevention of cardiac failure.12,13
Recently, dysferlin was proposed to be localized to the ventricular myocyte (VM) cell surface sarcolemma and the transverse-axial tubule (TAT) network,14 which is composed of regular tubular invaginations at the lateral surface membrane called transverse tubules (TTs), interconnected by axial tubule (AT) components.15 TAT membranes form thousands of junctional membrane complexes with the sarcoplasmic reticulum (SR), a.k.a. cardiac dyads that facilitate the regulated Ca2+-induced Ca2+ release in cardiomyocytes,16 but undergo disruption in the failing heart.17 While heterologous expression of dysferlin was suggested to mediate the biogenesis of a TAT-like endomembrane system in nonmuscle cells,18 little is known about the exact role of dysferlin with regard to TAT network maintenance and its stress-induced reorganization in terminally differentiated cardiomyocytes. However, TAT network reorganization has emerged as a hallmark of subcellular cardiomyocyte remodeling, particularly in the case of cardiac hypertrophy.15,19,20
Cardiac hypertrophy is considered a cardiomyocyte-specific response to increased biomechanical stress induced by prevalent stressors like arterial hypertension, valvular heart disease, or familial hypertrophic cardiomyopathy and results in progressive cardiomyopathy if left untreated. Global reorganization of the TAT network starts early in compensated hypertrophy of VMs, preceding the transition to left-ventricular (LV) failure.19 Hence, cardiomyocyte hypertrophy may depend on and can potentially be controlled by molecular mechanisms driving the remodeling of the TAT network. In this study, we addressed the role of dysferlin in the proliferation and remodeling of the TAT network during cardiac hypertrophy.
We applied stimulated emission depletion (STED) nanoscopy in combination with immunoelectron microscopy and electron tomography (ET) to analyze the subcellular expression of dysferlin in cardiomyocytes. Notably, we identified the specific localization of dysferlin in a new vesicular compartment in the vicinity of tubule-SR junctions inside VMs and atrial myocytes (AMs). Next, we used proteomic techniques to characterize the cardiac dysferlin interactome, demonstrating a previously unknown protein-protein interaction with the SR membrane and channel-tethering protein JP2 (junctophilin-2). While we confirmed a mild progressive phenotype of dilated cardiomyopathy in dysferlin knockout mice at higher ages, quantitative mass spectrometry identified proteomic changes preceding LV failure in dysferlin deficiency. Interestingly, following transverse aortic constriction (TAC) in mice, a model of cardiac hypertrophy induced by LV pressure overload, dysferlin deficiency diminished the hypertrophic response of cardiomyocytes. At consecutive intervals post-TAC, live-membrane imaging and ET of wild-type cardiomyocytes revealed a fundamental reorganization and proliferation of the TAT network with dilated AT membranes and increased AT-SR junctions, whose biogenesis critically depended on the high local expression of dysferlin.
METHODS
Data Availability
Proteomic data were deposited to the ProteomeXchange Consortium via the PRIDE21 partner repository with the dataset identifiers PXD043108, PXD053051, PXD045738, and PXD053037.
For more detailed methods description and the Major Resources Table please refer to the Supplemental Material.
Mouse Experiments
The generation of constitutive dysferlin-KO and cardiomyocyte-specific JP2 overexpression mice was described previously.1,22 Sex-mixed mouse cohorts in the C57BL/6J background aged 8 to 16 weeks and 30 to 32 weeks were used for experiments. Mouse cardiomyocytes were isolated using collagenase type II (Worthington) and modified Langendorff-perfusion.20,23 Mice were anesthetized with 2% isoflurane before cervical dislocation. TAC was induced with a 27 G spacer in 9- to 15-week-old mice after IP anesthesia. Blinded transthoracic echocardiographic measurements were performed in parasternal views with ECG monitoring under isoflurane anesthesia. Animal procedures were approved by the veterinarian state authority (LAVES, Oldenburg, Germany; animal protocols no. T11.2, 33.9-42502-04-18/2975, 33.9-42502-04-16/2102, and 33.9-42502-04-21/3785).
Human Endomyocardial LV Biopsies
LV biopsies were obtained from patients suffering from severe symptomatic aortic valve stenosis (AS) who underwent transfemoral transcatheter aortic valve replacement according to current guidelines. Myocardial biopsies from nonfailing (NF) donor hearts rejected for transplantation served as controls. The institutional ethics committee approved the study (no. 10/5/16), and written informed consent was obtained from all patients.
STED Nanoscopy
For live-cell membrane STED imaging, VMs were incubated with 5 µmol/L of the custom-made fluorescent cholesterol analog Chol-PEG-KK114,24 or 40 µmol/L of di-8-ANEPPS (Molecular Probes) diluted in nominally Ca2+-free perfusion buffer. Refer to Table S1 for information on physiological buffer compositions. For multicolor immunofluorescence STED imaging, isolated cardiomyocytes were fixed with 4% PFA on laminin-coated coverslips and permeabilized with 0.2% Triton in PBS. Human myocardial biopsies were fixed in 4% PFA, paraffin-embedded, and cut into 4-µm thick sections. Following deparaffinization and rehydration, epitopes were unmasked in sodium citrate buffer, and samples permeabilized with 0.1% Triton. For information on primary antibodies, see Table S2 and the Major Resources Table. Primary antibody specificity was verified by experiments including knockout mice (dysferlin), overexpression and knockdown mice (JP2), and knockin mice (ryanodine receptor type 2 [RyR2]). In addition, secondary antibody-only controls were used to distinguish genuine target staining from background. STED images were acquired with a Leica TCS SP8 laser-scanning microscope with an HC-PL-APO-C2S 100×/1.40 oil objective. Images were processed in ImageJ/Fiji.
Electron Tomography
VMs were high-pressure frozen in perfusion buffer plus 8% BSA using a Leica HPM100 system as described.20 Freeze-substitution was performed in an EM-AFS2 (Leica Microsystems) according to Wong et al.25 For ET, semi-thick (300 nm) sections were prepared and imaged using 300 kV Tecnai TF30 (Thermo Fisher Scientific). Tilt series were aligned, reconstructed, and combined using IMOD as described.26
Label-Free Quantitative Mass Spectrometry
LV tissue and isolated left-VMs from wild-type (WT) versus dysferlin-KO mice at an age of 16 and 32 weeks, and WT sham- versus TAC-operated animals were subjected to data-independent acquisition mass spectrometry (DIA-MS) and quantitative analysis using a hybrid ion mobility/time-of-flight mass spectrometer hyphenated to nanoflow chromatography.27 Using a custom prefractionated MS/MS spectral library, precursors were extracted at 1% FDR, which were then combined to peptide and protein levels. Protein profiles were log-transformed and quantile-normalized before proteome analyses. The raw data have been deposited to the ProteomeXchange Consortium via the PRIDE21 partner repository.
Protein Analysis
Quantitative SDS-PAGE was performed as described.20 Twenty micrograms of protein per lane were resolved by SDS-PAGE on 4% to 20% Tris-HCl protein gradient gels. Full immunoblot scans are provided in the supplemental figures. For coimmunoprecipitation experiments, we used a solubilization buffer containing 0.15% CHAPS and incubated 500 µg protein with 4 µg anti-dysferlin antibody (ab124684) or unspecific rabbit IgG antibody. Dynabeads protein G (Thermo Fisher Scientific) was used to precipitate dysferlin. For cell surface protein crosslinking, isolated VMs were incubated for 30 minutes with 10 mmol/L of the membrane-impermeable crosslinker bis-sulfosuccinimidyl suberate (BS3, Thermo Fisher Scientific).
Complexome Profiling
Membrane pellets (100 000 g) from isolated mouse VMs were solubilized, and 100 µg of protein per lane were separated by blue native gel electrophoresis as described.28 Gel lanes were cut into 60 equally sized slices and subjected to in-gel tryptic digestion. Peptides were analyzed by mass spectrometry (LC-MS/MS) in replicates and processed using MaxQuant software.
Statistics
Data are presented as mean±SEM. P<0.05 was accepted to indicate statistical difference. Statistical analyses were performed with GraphPad Prism not older than 9.4.0, SPSS Statistics 28.0.1.1, and R versions 4.2.2 and 4.3.1. Details regarding experimental design, sample sizes, normalization procedures, tests establishing normality, named statistical tests, post hoc correction for multiple comparisons, and precise P values are provided in the Supplemental Table: statistical details and the corresponding figure legends. Experiment-wide multiple test correction was not applied. Representative images were selected to visualize the average in each group.
RESULTS
Dysferlin Is Localized in Nanometric Proximity to Ca2+ Release Units in VMs
Among the group of 6 ferlin proteins, only dysferlin is substantially expressed in isolated mouse VMs on the RNA transcript level, with myoferlin transcripts at a 9 times lower level (Figure 1A). All other ferlin isoforms including otoferlin, which is essential for hearing as a mediator of the Ca2+-triggered exocytosis in hair cells,29 are not expressed at detectable levels in mouse VMs (Figure 1A). Quantitative analysis of immunoblots from mouse myocardial tissue lysates revealed the highest expression of dysferlin protein in the ventricles, compared with 59% relative dysferlin expression in the atria (Figure 1B; Figure S1). Atrial and ventricular tissue lysates from heterozygous dysferlin knockout mice showed ≈25% dysferlin protein expression in comparison to WT myocardium; homozygous dysferlin knockout (hereafter referred to as KO) lysates confirmed the antibody specificity (Figure 1B). Of note, only full-length dysferlin and no cleavage products were detected in mouse myocardium (Figure 1B; Figure S1).30
To localize dysferlin in isolated VMs in situ, we established coimmunostaining protocols for dysferlin and the membrane/tubule marker caveolin-3 for STED nanoscopy (see Supplemental Material for detailed information). Relatively small dysferlin immunofluorescent spots in groups of up to 10 signals accumulated near caveolin-3-positive Z-line striations, apparently associated with endomembrane TAT network components and the lateral surface membrane, where dysferlin signals alternated with caveolin-3-positive regions and did not directly overlap with caveolin-3 at the level of STED resolution (Figure 1C, left). Dysferlin-KO VMs were used to exclude unspecific antibody signals (Figure 1C, right), and Figure S2A documents the superior resolution of STED nanoscopy versus confocal microscopy using the same optical setup. While the majority of dysferlin signals were localized to the TAT network or the lateral surface membrane in VMs, we noted additional dysferlin signals with a high local density at the terminal intercalated disc (ICD) membrane folds (Figure S2A). To further estimate the quantitative distribution of dysferlin at specific membrane domains in the whole cell volume, we acquired confocal Z-stacks of dysferlin-immunostained VMs and applied image segmentation for quantitative immunofluorescence analyses (Figure S2B and S2C). Notably, compared with the predominant signal intensity-weighted dysferlin area fraction associated with the TAT network, only 16% and 7% were attributed to the lateral surface membrane or ICD membrane folds, respectively (Figure S2C). Immunohistological stainings and STED imaging of intact mouse ventricular myocardium confirmed a similar dysferlin and caveolin-3 signal structure and distribution at TAT membranes and ICD membrane folds (Figure S3A and S3B) as shown in isolated VMs. Confocal imaging and quantitative immunofluorescence analyses, however, demonstrated a signal intensity-weighted dysferlin area fraction of 15% at the ICD membrane folds compared with TAT-associated signals, twice as high as seen in isolated VMs (Figure S3C and S3D). This result is explained by 2 neighboring myocytes at the lateral ICD cell poles, whose dysferlin signals cannot be analyzed separately due to highly interdigitated ICD membrane folds.
In AMs, which express a TAT network primarily composed of AT membranes interconnected by sparse TT components,20,24 STED imaging also identified punctate dysferlin signals accumulating close to the TAT network and alternating with caveolin-3 positive membrane regions (Figure 1D, left). Of note, the lower expression level of dysferlin in atrial myocardium correlated with a significantly smaller TAT network density in AMs versus VMs.20 While dysferlin-KO AMs confirmed the specificity of the immunolabeling approach (Figure 1D, right), immunostainings of VMs and AMs from dysferlin-KO hearts did not show obvious differences in the organization of the caveolin-3 signals (Figure 1C and 1D, right).
At regular intervals, TAT membranes are known to form junctional membrane complexes with the SR, a.k.a. cardiac dyads, nanodomains regulating local intracellular Ca2+ release in cardiomyocytes.16 Hence, we wondered if the dysferlin signals border on SR Ca2+ release sites, and thus co-immunostained dysferlin and the cardiac RyR2 Ca2+ release channel in VMs. STED imaging in fact confirmed plenty of dysferlin signals in nanometric proximity to RyR2 channel clusters (Figure 1E, left). Accordingly, ET of mouse VMs identified numerous small membrane vesicles in the vicinity of tubule-SR junctions with an average diameter of 76.9±3.6 nm (Figure S4A and S4B). Moreover, immunoelectron microscopy confirmed dysferlin signals in vesicular membranes next to tubule-SR junctions (Figure S4C), suggestive of a nearby dysferlin-positive vesicular compartment.
To further characterize dysferlin and RyR2 signals spatially, immunofluorescence in both STED channels was subjected to image segmentation protocols31 before quantitative signal analysis (Figure 1E, right). Intracellular dysferlin immunofluorescent spots were 1 order of magnitude smaller than RyR2 clusters (average cluster size 0.006 versus 0.05 µm²) but exhibited a significantly higher cluster density (Figure 1F and 1G). Importantly, the dysferlin cluster frequency increased exponentially with smaller distances to RyR2 (Figure 1H), while the average nearest neighbor distance of dysferlin to RyR2 clusters was 0.11 µm small (Figure 1I). Accordingly, co-immunostaining of dysferlin and the L-type Ca2+ channel (LTCC) CaV1.2, which controls voltage-dependent Ca2+ influx through membrane tubules into cardiac dyads, confirmed a similar arrangement of dysferlin signals immediately next to CaV1.2 clusters (Figure S5A). However, our attempts to resolve the specific membrane compartment of dysferlin signals by co-immunostaining with vesicle markers have, to date, failed. For example, dysferlin colocalized neither with the vesicular coating protein clathrin (Figure S5B) nor with the synaptic vesicle protein synaptotagmin-1 (Figure S5C). Together, these data establish dysferlin as the predominant ferlin expressed in VMs that localizes in close proximity to Ca2+ release units, where it exists in a previously unknown vesicular compartment.
Dysferlin Knockout Mice Develop a Progressive Phenotype and Proteotype of Dilated Cardiomyopathy at Increasing Age
As dysferlin is the only ferlin expressed at substantial protein levels in VMs, we aimed to reassess the cardiac phenotype in 10 to 16 versus 30 to 32 weeks old dysferlin-KO mice. In total, we observed 202 animals per genotype and did not identify an increased mortality in the overall cohort (Figure 2A). However, more detailed analyses revealed that dysferlin-KO male mice showed a significantly higher mortality of ≈10% in the 32-week observation period (Figure 2B) in contrast to unimpaired dysferlin-KO female mice (Figure S6A). Importantly, none of the observed KO animals presented obvious symptoms related to muscular dystrophy in routine animal observation. In addition, body weight and heart weight were not changed between WT and KO animals at any time point (Figure S6B). However, investigator-blinded transthoracic echocardiography showed that the LV end-diastolic diameter increased significantly, whereas LV anterior wall thickness and LV ejection fraction decreased at higher ages in dysferlin-KO animals (Figure S6B; Figure 2C; Tables S3 and S4). The echocardiographic changes indicated a progressive but mild form of dilated cardiomyopathy in 30-week-old dysferlin-KO mice, which may contribute to the slightly increased mortality in male mice. Interestingly, the heart rate during echocardiography was reduced in young and older dysferlin-KO mice (Figure 2C, right), implicating a role of dysferlin for cardiac pacemaking and conduction propagation, which may be the subject of future studies.
On a single-cell basis, isolated dysferlin-KO VMs exhibited an increased cell area and surface perimeter, mainly reflecting an enlarged cell length compared with WT cells (Figure 2D), contributing to the mild dilated cardiomyopathy phenotype at 30 weeks. We also sought to analyze the TAT network in 30-week-old dysferlin-KO mice using the bright, photostable fluorescent cholesterol analog Chol-PEG-KK11424 for live confocal membrane imaging. Surprisingly, we could not identify changes in TAT network density or individual TAT component orientations at weeks 30 to 32 (Figure 2E) in contrast to a previous report for 50- to 60-week-old mice.14
To investigate proteomic changes at higher ages, we used ventricular tissue from WT versus dysferlin-KO hearts at 16 versus 32 weeks of age for DIA-MS analyses after tissue lysis by pressure-cycling technology.32 Based on 2 technical replicate injections from each n=5 WT versus KO hearts of both ages, DIA-MS reproducibly quantified 3746 proteins across all samples, of which 1265 proteins showed statistically significant abundance changes. Unsupervised hierarchical clustering clearly segregated WT from dysferlin-KO and 16 from 32 weeks old ventricular tissue samples as presented in the heat map in Figure 2F. Most interestingly, the pairwise comparison of 16-week-old mouse hearts commonly revealed 34 downregulated versus 28 upregulated proteins, while 69 downregulated versus 107 upregulated proteins were identified by the 32-week pairwise comparison (Figure 2G). Differentially abundant proteins from the 32-week pairwise comparison were subjected to gene ontology term enrichment analysis. Figure 2H highlights the top-10 most regulated gene ontology term biological pathways, 7 of which are directly linked to the regulation of lipid or phospholipid metabolism, and 3 to changes in respiratory electron chain transport, corroborating a progressive phenotype of dilated cardiomyopathy before echocardiographic changes.33 Additional information on gene ontology term enrichment and gene-concept-networks from the proteomic readout can be found in Figures S7 and S8. To characterize cardiomyocyte-specific proteomic changes in the dysferlin-KO, we additionally studied isolated VMs from each n=5 WT versus KO hearts at 16 weeks of age (Figure S9). Altogether, DIA-MS quantified 3178 proteins across all samples. The principal component analysis of the DIA-MS readout and the unsupervised hierarchical clustering of the differentially abundant proteins segregated WT from KO samples, thereby confirming a myocyte-specific dysferlin-KO proteotype (Figure S9A and S9B). The pairwise comparison of KO and WT VMs revealed 412 upregulated versus 957 downregulated proteins (Figure S9C), while the top 3 enriched gene ontology terms for biological pathways highlighted proteomic changes in Golgi vesicle transport, protein folding and establishment of protein localization to organelle, indicative of a disturbed vesicular transport in KO left-VMs (Figure S9D and S9E). Taken together, dysferlin-KO mice show a progressive phenotype of dilated cardiomyopathy based on and reflected by proteomic changes occurring before full expression of dilated cardiomyopathy in ventricular tissue and isolated VMs.
Interactomic Analyses Identify Dysferlin in Functional High Molecular Weight Complexes With Proteins of the Cardiac Ca2+ Release Unit
STED nanoscopy of coimmunostained VMs and immunofluorescence intensity line profiling underlined the close spatial relationship of dysferlin and RyR2 clusters (Figure 3A). Accordingly, co-immunostaining with JP2, a RyR2 and LTCC CaV1.2 binding protein,16,22,34 confirmed dysferlin signals in close proximity to JP2 clusters (Figure 3B). Indeed, image segmentation and detailed cluster signal analysis established similar characteristics for dysferlin/JP2 compared with dysferlin/RyR2 co-immunostainings with regard to cluster size and density (Figure S10, compare with Figure 1F and 1G). Hence, we wondered if dysferlin may interact with RyR2 and JP2 in the heart, although quantitative analysis of immunoblots from ventricular WT versus dysferlin-KO tissue lysates did not reveal changes in protein expression of RyR2, JP2 or caveolin-3 (Figures S11 and S12).
First, to study proteins in their functional high molecular weight complexes, we employed mass spectrometry-based complexome profiling, a global interaction analysis approach based on nondenaturating blue native gel electrophoresis separation followed by LC-MS/MS analysis (Figure S13A).28 Interestingly, the blue native gel electrophoresis migration pattern of dysferlin presented 3 local maxima: the first and highest maximum presumably indicates dysferlin monomers/dimers in smaller protein complexes, whereas the third maximum at ≈5 MDa represents the functional high molecular weight protein complex including dysferlin and RyR2 (Figure 3C). Interestingly, the JP2 migration profile showed a total of seven local maxima, 3 of which overlapped clearly with the 3 dysferlin maxima (Figure 3D). In contrast, the caveolin-3 pattern did not show any high molecular weight maximum overlapping with dysferlin and neither did we detect protein subunits of the LTCC CaV1.2 (CACNA1C, CACNB2, and CACNA2D1) in the high molecular weight range (Figure S13B). However, lower molecular weight maxima for caveolin-3 and CACNA1C overlapped with the low molecular weight complex for dysferlin.
Second, to further explore the proteomic interactome of dysferlin in the heart, we used recombinant expression of N-terminally Twin-Strep II-tagged full-length human dysferlin to establish an in vitro binding assay. Specifically, isolated mouse heart membrane fractions were incubated with dysferlin-coupled beads under 4 different detergent conditions. Dysferlin pull-down eluates were analyzed by DIA-MS, revealing altogether 412 potential protein interaction partners (disregarding dysferlin) as highlighted in Figure 3E, among which JP2 was identified. To provide a subcellular synopsis, the percentage of dysferlin interactors was mapped on VM organelles and compartments in a cartoon presentation (Figure 3F). Top dysferlin interactor protein functions included the endosomal and secretory pathway as well as (tubular) membrane repair and biogenesis (Figure 3G; Supplemental Data S2).
Finally, as dysferlin is thought to function as a Ca2+-binding protein,35 activated upon local plasma membrane rupture, we wondered if the above described protein interactions depend on increased Ca2+ concentrations. Coimmunoprecipitation experiments of ventricular tissue membrane fractions in absence and presence of Ca2+ (1 mmol/L) showed that the protein-protein interactions of dysferlin with RyR2 and JP2 are independent of the Ca2+ concentration (Figure 3H; Figure S14), including dysferlin-KO and IgG controls to rule out unspecific protein binding. In summary, 3 independent biochemical approaches demonstrated protein-protein interactions of dysferlin with the cardiac SR proteins RyR2 and JP2, which may be crucial to stabilize the functionally important junctional membrane complexes during VM contractions and increased biomechanical stress.
Polyadic Tubule-SR Junctions in VMs Overexpressing Junctophilin-2 Increase the Density of Local Dysferlin Clusters
Previous studies have shown that JP2 overexpression induces the formation of complex polyadic tubule-SR junctions in VMs and AMs.31,36 Hence, in light of the interaction of dysferlin with proteins of the cardiac dyad, we sought to study dysferlin clustering in VMs isolated from cardiomyocyte-restricted transgenic JP2 overexpressing mice.21 Live-cell membrane STED imaging using Chol-PEG-KK114 confirmed stacked tubular superstructures (Figure 4A) and parallelized ATs stacked on top of each other in JP2 overexpressing VMs (Figure S15A and S15B), providing a profoundly increased tubular membrane surface for gain-of-function polyadic tubule-SR junctions as demonstrated by ET and Ca2+ imaging previously.31 Stacked tubular superstructures in JP2 overexpressing VMs showed an average diameter of 600 nm, which is approximately thrice as large as TTs and ATs in WT and JP2 overexpressing VMs (Figure 4B).
Coimmunostaining confirmed the stacked tubular membrane structures as caveolin-3-positive membrane signals (Figure 4C; Figure S15C). Most interestingly, tubular superstructures were accompanied by a markedly increased local density of dysferlin clusters (Figure 4C, red). To quantify the changes of dysferlin expression in JP2 overexpression, cluster analysis was performed based on segmented STED images. Although the histogram distribution showed no obvious changes in the dysferlin cluster size in WT versus JP2 overexpressing VMs (Figure 4D left; Figure S16A), a mild but significant increase of the average dysferlin cluster size was documented (Figure 4D, middle). This increase in dysferlin cluster size may have resulted from overlapping but distinct dysferlin signals, which could be observed in context of an increased dysferlin cluster density (Figure 4D, right) and area fraction in JP2 overexpressing VMs (Figure S16B). Although the dysferlin cluster density grew, the nearest distance of dysferlin to adjacent RyR2 clusters remained unchanged, indicating a specific localization of dysferlin at Ca2+ release units (Figure S16C).
Coimmunostaining of dysferlin and JP2 confirmed large JP2 clusters in JP2 overexpressing VMs in vicinity of numerous dysferlin clusters (Figure 4E). Of note, the average JP2 cluster size and area fraction was increased by 27% in JP2 overexpression, whereas the JP2 cluster density did not change significantly (Figure 4F; Figure S16D and S16E). In addition, JP2 overexpression exhibited apparently larger RyR2 clusters compared with WT VMs, which were surrounded by a high local density of dysferlin clusters (Figure 4G). This was confirmed by the histogram distribution of the RyR2 cluster size suggesting a small shift toward larger RyR2 clusters in JP2 overexpression (Figure 4H; Figure S16F and S16G). In summary, polyadic tubule-SR junctions induced by JP2 overexpression resulted in an increased local density of dysferlin clusters in VMs.
Dysferlin Deficiency Ameliorates Hypertrophic Remodeling in Left-VMs
Dysferlin was previously shown to enable tubular membrane proliferation in heterologous cell systems.18 Hence, we hypothesized that dysferlin may impact cellular hypertrophic remodeling induced by LV pressure overload through the biogenesis of TAT network components. Therefore, we applied TAC (Figure 5A) in 9- to 15-week-old WT and dysferlin-KO mice to assess potential differences in LV hypertrophy 4 weeks post-surgery (4pTAC; Figure 5B). The TAC-induced transaortic pressure gradients were equally elevated in the first 3 days post-surgery in WT and KO animals (Figure S17A). Unexpectedly, despite a mild tendency toward dilated cardiomyopathy per se (Figure 2), dysferlin-KO mice did not show a significant higher mortality 4pTAC (Figure 5C). However, the heart weights of dysferlin-KO mice were considerably lower compared with WT mice 4 weeks post-TAC (−25%; Figure 5D), while the body weight was unchanged (Figure S17B). Investigator-blinded echocardiographic measurements revealed that the LV end-diastolic diameter had not changed, while the LV anterior and posterior wall thickness, both indicators of hypertrophy, increased to a lesser extent in KO versus WT hearts 4pTAC (Figure 5E; Figure S17C; Table S5). To understand the reduced LV hypertrophy development in KO hearts, we studied isolated left-VMs. Dysferlin-KO left-VMs were significantly smaller in regard to cell area and width 4pTAC compared with WT cells (Figure 5F; Figure S17D). These results were further underlined by 3D volumetry of isolated left-VMs, confirming a decrease in volume of −29% in dysferlin-KO compared with WT cells 4 weeks post-TAC (Figure 5G; Figure S17E). Hence, we conclude that dysferlin is necessary for the hypertrophic growth of left-VMs following TAC.
Interestingly, immunoblots of LV tissue lysates identified an elevation of dysferlin expression up to 143% 1pTAC and 192% 4pTAC, while JP2 expression was slightly reduced (−24%, Figures S12 and S17F; Figure 5H), further highlighting the role of dysferlin in post-TAC cardiac hypertrophy. To differentiate between dysferlin expression on the cell surface versus intracellular membrane vesicles, we employed VM surface protein crosslinking using the membrane-impermeable crosslinker BS3 (Figure S17G). BS3 protein crosslinking requires extracellular lysines, of which surface-expressed dysferlin possesses 2 in the extracellular domain. Under baseline conditions, 30-minute BS3 treatment of VMs resulted in at least 4 additional crosslinked dysferlin bands as shown by immunoblotting (Figure 5I). In the crosslinking experiments, the Na+-K+-ATPase (NaK) served as positive and the intracellular membrane protein caveolin-3 as negative control. Quantitative analysis of the BS3 crosslinking revealed 22% of dysferlin versus 84% of NaK protein crosslinking as normalized to the total protein (Figure 5I). In VMs 4pTAC, crosslinked dysferlin products were significantly elevated up to 47% (Figure 5J), implying an active role of dysferlin for membrane stabilization and remodeling 4pTAC.
Proteomic Profiling of Isolated Myocytes Identifies Early Versus Late Proteomic Changes in Pressure Overload–Induced LV Hypertrophy
To assess early versus late cardiomyocyte-specific proteomic changes in LV hypertrophy, we isolated left-VMs from WT hearts 1 and 4 weeks post-TAC for DIA-MS analysis (Figure S18). Based on 1814 proteins reproducibly quantified across all samples, 515 proteins showed significant abundance changes between the sham- versus TAC-operated experimental groups. The principal component analysis of the DIA-MS readout and the unsupervised hierarchical clustering of differentially abundant proteins clearly segregated left-VM proteotypes depending on intervention and time points (Figure S18A and S18B). Most interestingly, the 1pTAC proteotype showed the greatest Euclidean distance from sham controls based on proteomic differences observed in early left-VM remodeling post-TAC. More specifically, the pairwise comparison of 1pTAC and 1pSham samples identified 487 upregulated versus 96 downregulated proteins (Figure S18C), of which dysferlin was confirmed as a significantly upregulated protein 1pTAC in line with our immunoblot quantifications. In the 4pTAC versus sham comparison we detected 314 up- versus 83 downregulated proteins, highlighting persistent proteomic differences in late left-VM hypertrophic remodeling (Figure S18D). Finally, Figure S18E directly compares 1pTAC and 4pTAC samples to point out 274 versus 45 proteins in early and late left-VM remodeling, respectively. The top-10 most regulated proteins in the pairwise comparisons are listed in Figure S18F. In addition, differentially abundant proteins from individual group comparisons were subjected to gene ontology term enrichment analysis. Notably, most of the top-10 overrepresented gene ontology terms molecular function in the pairwise comparisons can be linked to regulation of protein translation, protein folding, and actin binding (Figure S19). Figure S20 highlights the most significantly enriched gene ontology terms related to cardiac development and hypertrophy, visualizing an increase in the myosin-7/myosin-6 ratio among numerous other changes on the protein level in isolated left-VMs post-TAC.
Additionally, we analyzed the transcriptome of WT versus dysferlin-KO LV tissue from mouse hearts 4 weeks after sham versus TAC surgery (Figure S21; Supplemental Data S1). While we confirmed genotype-specific differences in all group comparisons, most of the transcriptomic changes were identified in WT sham versus TAC samples (1476 significantly regulated genes, Figure S21D). Some of the differentially regulated genes in the WT sham versus TAC comparison can be linked to the enriched gene ontology terms cardiac cell/muscle development (Figure S21D and S21F). In contrast, none of these genes, nor previously established transcription factors involved in cardiac development or reactivation of the fetal gene program during hypertrophy (like Gata4, Mef2a, Nkx2-5, Tbx, etc)37,38 were regulated in WT versus KO TAC samples (Figure S21E). Likewise, no significant changes in the myosin heavy chain isoforms Myh6 and Myh7 or the natriuretic peptides Nppa and Nppb were detected in WT versus KO TAC samples on the mRNA level (Figure S21E). Overall, we did not observe any significantly enriched gene ontology terms for biological pathways in the WT versus KO TAC comparison. Altogether, the attenuated hypertrophic remodeling observed in the dysferlin-KO mice was not associated with prominent changes in cardiac gene regulation compared with WT controls post-TAC.
AT Proliferation in Cardiac Hypertrophy Is Attenuated by Dysferlin Deficiency
To study the consequences of VM hypertrophy on the TAT network architecture, we applied live-cell membrane STED imaging on isolated left-VMs post-TAC, uncovering dramatic changes in the network composition after signal skeletonization (Figure 6A). Most prominently, the TAT network density increased 1 and 4pTAC (Figure 6B), mainly driven by the proliferation of AT membranes along the longitudinal direction of the cell axis (Figure 6C; Figure S22A). Consequently, the AT density 1 and 4pTAC was increased (Figure 6D), contributing to an elevated number of network junctions (Figure S22B). In sharp contrast, left-VMs from dysferlin-KO hearts lacked a comparable proliferation of AT membranes 4pTAC, while the TT components were preserved (Figure 6E and 6G).
ET of isolated left-VMs did not only confirm the proliferation of AT components in cross-sectional views but also identified a markedly increased AT cross-sectional area 4pTAC (Figure 6H and 6I), together with enlarged tubule-SR contact sites as visualized by 3D reconstruction of an AT and its surrounding SR in Figure 6H. Additional 3D ET reconstructions of longitudinal left-VM sections demonstrated changes in tubule geometry with membrane extensions and protrusions not seen in sham left-VMs (Figure 6J). These tubular extensions may indicate active membrane remodeling and branching mechanisms necessary for the TAT network proliferation. Next to the TAT system, ET allowed the detection of membrane vesicles 4pTAC in a high local density that could not be found in sham controls and may additionally contribute to TAT component proliferation (Figure 6J; Figure S23). In conclusion, AT membrane proliferation together with AT dilation seem to be hallmarks of subcellular membrane remodeling in left-VM hypertrophy induced by pressure overload but are significantly attenuated in the dysferlin-KO heart.
Newly Shaped AT Membranes Are Highly Decorated by Dysferlin Clusters
To further explore the contribution of dysferlin to post-TAC tubule proliferation, we studied co-immunostainings of dysferlin and caveolin-3 in left-VMs 4pTAC. The confocal overviews in Figure 7A highlight the increased expression of dysferlin as visualized by the red signal intensity in hypertrophied left-VMs 4pTAC, which was particularly prominent in the center of the cells between the 2 nuclei. As dysferlin clusters are associated tightly with TAT network structures, confocal images were used for skeletonization before network analysis to identify the contribution of dysferlin to specific TAT network changes (Figure 7A). Of note, the dysferlin skeleton density increased by 28% in favor of axial network components (Figure 7B). Consequently, the frequency of axial skeleton components was elevated in left-VMs 4pTAC (Figure 7B), consistent with a role of dysferlin in AT membrane proliferation (Figure 6).
Parallel analysis of the caveolin-3 signal showed an unchanged caveolin-3 TAT network density 4pTAC, and a minor change toward more axial network components but markedly reduced TT elements (Figure 7C). These differences in dysferlin versus caveolin-3 relative to the TAT network composition were substantiated by frequency subtraction histograms (Figure 7D), showing a pronounced axial component proliferation in the dysferlin versus prominent transverse component reduction in the caveolin-3 network signal. In line with a tentatively reduced caveolin-3 protein expression 4pTAC facing an elevated TAT network density (Figure 5H), caveolin-3 expression on tubular membranes was generally limited in left-VMs 4pTAC but mainly reduced on TT elements.
STED nanoscopy of dysferlin and caveolin-3 coimmunostained left-VMs confirmed the presence of dysferlin clusters at regular intervals at the TAT network in sham cells (Figure 7E). In left-VMs 4pTAC, however, more numerous dysferlin clusters highly decorated newly shaped AT membranes in string-of-pearls arrangements (Figure 7E, right). In light of an increased dysferlin surface crosslinking 4pTAC, these data suggest a direct role for dysferlin in the proliferation of AT membranes during VM hypertrophy.
Dysferlin Clusters Are Strongly Expressed on AT Membranes in Left-VMs From Human Hearts With Severe Aortic Valve Stenosis
LV biopsies from human hearts were used to extend our imaging data to a highly prevalent human disease context.32 In healthy nonfailing LV sections fluorescently stained for dysferlin and the continuous membrane marker wheat germ agglutinin,24 punctate dysferlin signals were localized in close association to TT membranes of left-VMs, further highlighted by the line profiles of the wheat germ agglutinin and dysferlin signals (Figure 8A). Dysferlin signals were also found in high local density at ICD membrane folds (Figure 8B), thereby fully validating our mouse VM imaging data in human LV biopsy samples.
To study dysferlin in human LV pressure overload in situ, we obtained LV biopsies from patients with 2 different hemodynamic subtypes of severe aortic valve stenosis, who underwent transfemoral aortic valve replacement: classic normal ejection fraction and high gradient aortic stenosis with compensated LV hypertrophy versus low ejection fraction and low gradient aortic stenosis representing decompensated LV hypertrophy that progressed into systolic heart failure (Figure 8C).32 Please refer to Supplemental Material for detailed patient information. While confocal overviews of the dysferlin and wheat germ agglutinin signals in coimmunostained biopsy sections confirmed substantially hypertrophic left-VMs in both aortic stenosis subtypes in contrast to nonfailing myocardium, TAT network composition and dysferlin clustering showed clear differences. In normal ejection fraction and high gradient aortic stenosis, left-VMs contained AT membrane structures clearly associated with many dysferlin clusters, as highlighted by STED magnifications (Figure 8C, middle). Left-VMs from low ejection fraction and low gradient aortic stenosis biopsies, in contrast, hardly contained any AT structures and only rare residual TT elements (Figure 8C, right). Interestingly, dysferlin clusters in low ejection fraction and low gradient aortic stenosis mainly decorated the lateral surface membrane and the ICD membrane folds but could rarely be found on TAT structures (Figure 8C, STED magnification). Thus, dysferlin clusters are located at different subcellular membrane compartments in left-VMs of distinct aortic stenosis subtypes and may be regulated differentially in the context of compensated versus decompensated LV hypertrophy and acute murine versus chronic human LV pressure overload.
DISCUSSION
Combining the strengths of super-resolution light and electron microscopy, we identified that dysferlin, the predominant ferlin expressed in VMs, is mainly localized in a vesicular compartment in close proximity to tubule-SR junctions. In JP2 overexpression VMs, junctional membrane complexes composed of polyadic tubule-SR junctions provoked a local accumulation of additional dysferlin clusters. Notably, our interactomic approaches discovered >400 protein-protein interaction partners of dysferlin in VMs by complementary biochemical/proteomic techniques including complexome profiling, pull-down, and coimmunoprecipitation, demonstrating the interaction of dysferlin with the SR proteins RyR2 and JP2 (Figure 3). We confirmed a mild and progressive phenotype of dilated cardiomyopathy in dysferlin-KO mice and revealed significant protein abundance changes preceding LV systolic failure in dysferlin deficiency by DIA-MS proteome analysis of LV myocardium and isolated left-VMs. Strikingly, in TAC-induced LV pressure overload, WT mice expressed 92% more dysferlin, while dysferlin-KO animals presented reduced LV hypertrophy. On the subcellular level, VMs post-TAC showed a substantial proliferation of the TAT network based on new and dilated AT membranes that were highly decorated by dysferlin clusters (Figures 6 and 7). Together with elevated dysferlin surface crosslinking post-TAC, we suggest an active role of dysferlin in recruiting a previously unknown vesicular compartment for the proliferation of TAT membranes to enable VM growth during pressure overload. STED imaging data from human VMs confirmed increased dysferlin labeling of AT membranes in normal ejection fraction and high gradient aortic stenosis patients.
Previous studies reported a dysfunctional TAT network in skeletal myocytes from dysferlin-KO mice39 and demonstrated a protein interaction of dysferlin with the LTCC CaV1.1.40 In VMs, which primarily express CaV1.2, confocal images from Hofhuis et al14 suggested a colocalization of dysferlin with CaV1.2. However, our super-resolution STED imaging data identified dysferlin clustering in vicinity to and not directly overlapping with CaV1.2, RyR2, and JP2 clusters (Figure 1), thereby extending the previous model of subcellular dysferlin organization in VMs. As immunoelectron microscopy showed dysferlin in distinct membrane vesicles next to tubule-SR junctions (Figure S4), we propose a dysferlin-containing vesicular membrane compartment next to tubule-SR structures, which can be recruited to stabilize and seal the cardiac dyads upon tubular membrane lesions. Notably, SR membranes are tethered to the tubular surface through direct binding of JP2 to the CaV1.2 C-terminus.16,34 Cardiac dyads face an increased biomechanical stress in the context of constantly contracting cardiomyocytes, requiring a rapidly activated membrane repair strategy during stress exercise of the heart.12 Previously, JP2 was shown to be a key regulator of the architecture of the cardiac dyads.22 In addition, JP2 may organize a pool of vesicles in close proximity to the dyadic cleft based on a protein interaction with dysferlin (Figures 3 and 4). The role of dysferlin in stabilization of tubule-SR junctions is consistent with previously published data, showing decreased LTCC current versus increased spontaneous SR Ca2+ release events in dysferlin-KO VMs.14 Hence, on top of JP2’s proposed roles as a membrane tether of the cardiac Ca2+ release unit and as a regulator of gene transcription upon Ca2+-dependent calpain cleavage,16,41 JP2 may protect the Ca2+ release unit by direct recruitment of dysferlin-containing vesicles as a vital membrane reservoir that facilitates rapid Ca2+-mediated membrane repair.
Previously, in vitro assays revealed membrane modulatory properties of dysferlin, myoferlin, and otoferlin.42 However, in COS-7 and HeLa cells, exogenous dysferlin but not myoferlin or otoferlin were suggested to enable membrane tubulation.18 To our best knowledge, we are the first to show a direct role of dysferlin in the progressive remodeling of the TAT network through proliferation of AT membranes in primary VMs, specifically under pressure overload–induced LV hypertrophy in transition to heart failure (Figures 6 and 7). In dysferlin-deficient VMs, however, AT membrane proliferation was absent, thereby limiting VM hypertrophy post-TAC (Figure 5). While AT membrane proliferation in VMs has been associated with different stages of heart failure before,15,17,43 our data propose that the dysferlin-mediated proliferation of the TAT network may be a prerequisite for the cellular hypertrophy of VMs, and mark the transition to heart failure.
Among potential study limitations, we note that the correlation of STED imaging and immunoelectron microscopy can only partly reveal the localization of dysferlin on vesicular versus tubular/surface membranes, particularly in the absence of a continuous membrane marker like wheat germ agglutinin in mouse VM (immuno-)labeling.24 Therefore, BS3 protein crosslinking was used to semi-quantitatively detect surface versus intracellularly expressed dysferlin (Figure 5). The exact vesicular compartment, in which dysferlin is organized in VMs, may be explored in future studies guided by our interactomic readouts. We decided to use isolated VMs for most of our experiments in view of superior immunofluorescence signal quality compared with myocardial tissue slices (compare Figures S2 and S3). Furthermore, the cell axis of isolated VMs can easily be aligned for the extensive TAT network analyses presented in this study in contrast to myocardial tissue slices, strengthening the reliability of the TAT network readouts. The cardiomyocyte isolation protocol was carefully optimized to preserve membrane nanodomains like the endomembrane TAT network or the ICD membrane folds as demonstrated in our previously published work.20,23,24,31,32,44 Although we identified dysferlin as an important mediator of TAT membrane proliferation in differentiated VMs post-TAC, molecular mechanisms from dysferlin activation to membrane recruitment and vesicle exocytosis should be elaborated to therapeutically target membrane repair and proliferation in the heart. While the vast majority of the dysferlin protein in VMs is associated with TAT membranes as estimated in Figure S2, ICD membrane folds also show a high local density of dysferlin clustering consistent with cardiac muscle data,13 and may be addressed in the context of dysferlinopathy in the future. Potentially, dysferlin may be recruited to different sites of action through varying protein-protein interactions in cardiomyocytes.
In conclusion, our current data suggest dysferlin as a potential therapeutic target (1) to stabilize tubule-SR junctions for regulated excitation-contraction coupling in VMs, (2) to promote TAT membrane and junctional membrane contact site regeneration in heart failure, and (3) to control TAT membrane remodeling and cardiomyocyte growth in pressure overload–induced cardiac hypertrophy. To translate our findings into medical interventions, future studies are necessary to identify strategies for the regulation of local dysferlin expression and activation of vesicular membrane recruitment in cardiomyocytes.
ARTICLE INFORMATION
Supplemental Material
Supplemental Methods
Figures S1–S23
Tables S1–S5
Data S1 and S2
Supplemental Table: Statistical details
Major Resources Table
References 45–62
Acknowledgments
The authors gratefully thank Brigitte Korff, Birgit Schumann, Timo Schulte, Lisa Neuenroth, Sina Langer, Dörte Hesse, Marina Uecker, and the European Molecular Biology Laboratory (EMBL) Heidelberg electron microscopy core facility for excellent technical support. The authors extend their gratitude to Gabriela Salinas and Maren Sitte from the Next-Generation Sequencing (NGS)-Integrative Genomics Core Unit at the Institute of Pathology, University Medical Center Göttingen for their excellent support in conducting RNA-sequencing and data analysis of the transcriptomic data. The graphical abstract was created with www.BioRender.com.
Footnote
Nonstandard Abbreviations and Acronyms
- AM
- atrial myocyte
- AT
- axial tubule
- BS3
- bissulfosuccinimidyl suberate
- DIA-MS
- data-independent acquisition mass spectrometry
- ET
- electron tomography
- ICD
- intercalated disc
- JP2
- junctophilin-2
- LTCC
- L-type Ca2+ channel
- LV
- left-ventricular
- NaK
- Na+-K+-ATPase
- RyR2
- ryanodine receptor type 2
- SR
- sarcoplasmic reticulum
- STED
- stimulated emission depletion
- TAC
- transverse aortic constriction
- TAT
- transverse-axial tubule
- TT
- transverse tubule
- VM
- ventricular myocyte
- WT
- wild-type
Supplemental Material
File (circres_circres-2024-324588d_supp1.pdf)
- Download
- 21.26 KB
File (circres_circres-2024-324588d_supp2.xlsx)
- Download
- 4.01 MB
File (circres_circres-2024-324588d_supp3.xlsx)
- Download
- 48.98 KB
File (circres_circres-2024-324588d_supp6.pdf)
- Download
- 568.35 KB
File (supplement_material_paulke_circres_2024.pdf)
- Download
- 6.78 MB
REFERENCES
1.
Bansal D, Miyake K, Vogel SS, Groh S, Chen CC, Williamson R, McNeil PL, Campbell KP. Defective membrane repair in dysferlin-deficient muscular dystrophy. Nature. 2003;423:168–172. doi: 10.1038/nature01573
2.
Bulankina AV, Thoms S. Functions of vertebrate ferlins. Cells. 2020;9:534. doi: 10.3390/cells9030534
3.
Lek A, Evesson FJ, Sutton RB, North KN, Cooper ST. Ferlins: regulators of vesicle fusion for auditory neurotransmission, receptor trafficking and membrane repair. Traffic. 2012;13:185–194. doi: 10.1111/j.1600-0854.2011.01267.x
4.
Han R, Campbell KP. Dysferlin and muscle membrane repair. Curr Opin Cell Biol. 2007;19:409–416. doi: 10.1016/j.ceb.2007.07.001
5.
Pangrsic T, Reisinger E, Moser T. Otoferlin: a multi-C2 domain protein essential for hearing. Trends Neurosci. 2012;35:671–680. doi: 10.1016/j.tins.2012.08.002
6.
Miyoshi K, Kawai H, Iwasa M, Kusaka K, Nishino H. Autosomal recessive distal muscular dystrophy as a new type of progressive muscular dystrophy. Seventeen cases in eight families including an autopsied case. Brain. 1986;109 (Pt 1):31–54. doi: 10.1093/brain/109.1.31
7.
Liu J, Aoki M, Illa I, Wu C, Fardeau M, Angelini C, Serrano C, Urtizberea JA, Hentati F, Hamida MB, et al. Dysferlin, a novel skeletal muscle gene, is mutated in Miyoshi myopathy and limb girdle muscular dystrophy. Nat Genet. 1998;20:31–36. doi: 10.1038/1682
8.
Illa I, Serrano-Munuera C, Gallardo E, Lasa A, Rojas-Garcia R, Palmer J, Gallano P, Baiget M, Matsuda C, Brown RH. Distal anterior compartment myopathy: a dysferlin mutation causing a new muscular dystrophy phenotype. Ann Neurol. 2001;49:130–134. doi: 10.1002/1531-8249(200101)49:1<130::AID-ANA22>3.0.CO;2-0
9.
Nishikawa A, Mori-Yoshimura M, Segawa K, Hayashi YK, Takahashi T, Saito Y, Nonaka I, Krahn M, Levy N, Shimizu J, et al. Respiratory and cardiac function in japanese patients with dysferlinopathy. Muscle Nerve. 2016;53:394–401. doi: 10.1002/mus.24741
10.
Wenzel K, Geier C, Qadri F, Hubner N, Schulz H, Erdmann B, Gross V, Bauer D, Dechend R, Dietz R, et al. Dysfunction of dysferlin-deficient hearts. J Mol Med (Berl). 2007;85:1203–1214. doi: 10.1007/s00109-007-0253-7
11.
Choi ER, Park SJ, Choe YH, Ryu DR, Chang SA, Choi JO, Lee SC, Park SW, Kim BJ, Kim DK, et al. Early detection of cardiac involvement in Miyoshi myopathy: 2D strain echocardiography and late gadolinium enhancement cardiovascular magnetic resonance. J Cardiovasc Magn Reson. 2010;12:31. doi: 10.1186/1532-429X-12-31
12.
Han R, Bansal D, Miyake K, Muniz VP, Weiss RM, McNeil PL, Campbell KP. Dysferlin-mediated membrane repair protects the heart from stress-induced left ventricular injury. J Clin Invest. 2007;117:1805–1813. doi: 10.1172/JCI30848
13.
Chase TH, Cox GA, Burzenski L, Foreman O, Shultz LD. Dysferlin deficiency and the development of cardiomyopathy in a mouse model of limb-girdle muscular dystrophy 2B. Am J Pathol. 2009;175:2299–2308. doi: 10.2353/ajpath.2009.080930
14.
Hofhuis J, Bersch K, Wagner S, Molina C, Fakuade FE, Iyer LM, Streckfuss-Bomeke K, Toischer K, Zelarayan LC, Voigt N, et al. Dysferlin links excitation-contraction coupling to structure and maintenance of the cardiac transverse-axial tubule system. Europace. 2020;22:1119–1131. doi: 10.1093/europace/euaa093
15.
Wagner E, Lauterbach MA, Kohl T, Westphal V, Williams GS, Steinbrecher JH, Streich JH, Korff B, Tuan HT, Hagen B, et al. Stimulated emission depletion live-cell super-resolution imaging shows proliferative remodeling of T-tubule membrane structures after myocardial infarction. Circ Res. 2012;111:402–414. doi: 10.1161/CIRCRESAHA.112.274530
16.
Lehnart SE, Wehrens XHT. The role of junctophilin proteins in cellular function. Physiol Rev. 2022;102:1211–1261. doi: 10.1152/physrev.00024.2021
17.
Song LS, Sobie EA, McCulle S, Lederer WJ, Balke CW, Cheng H. Orphaned ryanodine receptors in the failing heart. Proc Natl Acad Sci U S A. 2006;103:4305–4310. doi: 10.1073/pnas.0509324103
18.
Hofhuis J, Bersch K, Bussenschutt R, Drzymalski M, Liebetanz D, Nikolaev VO, Wagner S, Maier LS, Gartner J, Klinge L, et al. Dysferlin mediates membrane tubulation and links T-tubule biogenesis to muscular dystrophy. J Cell Sci. 2017;130:841–852. doi: 10.1242/jcs.198861
19.
Wei S, Guo A, Chen B, Kutschke W, Xie YP, Zimmerman K, Weiss RM, Anderson ME, Cheng H, Song LS. T-tubule remodeling during transition from hypertrophy to heart failure. Circ Res. 2010;107:520–531. doi: 10.1161/CIRCRESAHA.109.212324
20.
Brandenburg S, Kohl T, Williams GS, Gusev K, Wagner E, Rog-Zielinska EA, Hebisch E, Dura M, Didie M, Gotthardt M, et al. Axial tubule junctions control rapid calcium signaling in atria. J Clin Invest. 2016;126:3999–4015. doi: 10.1172/JCI88241
21.
Perez-Riverol Y, Bai J, Bandla C, Garcia-Seisdedos D, Hewapathirana S, Kamatchinathan S, Kundu DJ, Prakash A, Frericks-Zipper A, Eisenacher M, et al. The PRIDE database resources in 2022: a hub for mass spectrometry-based proteomics evidences. Nucleic Acids Res. 2022;50:D543–D552. doi: 10.1093/nar/gkab1038
22.
van Oort RJ, Garbino A, Wang W, Dixit SS, Landstrom AP, Gaur N, De Almeida AC, Skapura DG, Rudy Y, Burns AR, et al. Disrupted junctional membrane complexes and hyperactive ryanodine receptors after acute junctophilin knockdown in mice. Circulation. 2011;123:979–988. doi: 10.1161/CIRCULATIONAHA.110.006437
23.
Wagner E, Brandenburg S, Kohl T, Lehnart SE. Analysis of tubular membrane networks in cardiac myocytes from atria and ventricles. J Vis Exp. 2014;(92):e51823. doi: 10.3791/51823
24.
Brandenburg S, Pawlowitz J, Fakuade FE, Kownatzki-Danger D, Kohl T, Mitronova GY, Scardigli M, Neef J, Schmidt C, Wiedmann F, et al. Axial tubule junctions activate atrial Ca(2+) release across species. Front Physiol. 2018;9:1227. doi: 10.3389/fphys.2018.01227
25.
Wong AB, Rutherford MA, Gabrielaitis M, Pangrsic T, Gottfert F, Frank T, Michanski S, Hell S, Wolf F, Wichmann C, et al. Developmental refinement of hair cell synapses tightens the coupling of Ca2+ influx to exocytosis. EMBO J. 2014;33:247–264. doi: 10.1002/embj.201387110
26.
Rog-Zielinska EA, Scardigli M, Peyronnet R, Zgierski-Johnston CM, Greiner J, Madl J, O’Toole ET, Morphew M, Hoenger A, Sacconi L, et al. Beat-by-beat cardiomyocyte T-tubule deformation drives tubular content exchange. Circ Res. 2021;128:203–215. doi: 10.1161/CIRCRESAHA.120.317266
27.
Meier F, Brunner AD, Frank M, Ha A, Bludau I, Voytik E, Kaspar-Schoenefeld S, Lubeck M, Raether O, Bache N, et al. diaPASEF: parallel accumulation-serial fragmentation combined with data-independent acquisition. Nat Methods. 2020;17:1229–1236. doi: 10.1038/s41592-020-00998-0
28.
Alsina KM, Hulsurkar M, Brandenburg S, Kownatzki-Danger D, Lenz C, Urlaub H, Abu-Taha I, Kamler M, Chiang DY, Lahiri SK, et al. Loss of protein phosphatase 1 regulatory subunit PPP1R3A promotes atrial fibrillation. Circulation. 2019;140:681–693. doi: 10.1161/CIRCULATIONAHA.119.039642
29.
Roux I, Safieddine S, Nouvian R, Grati M, Simmler MC, Bahloul A, Perfettini I, Le Gall M, Rostaing P, Hamard G, et al. Otoferlin, defective in a human deafness form, is essential for exocytosis at the auditory ribbon synapse. Cell. 2006;127:277–289. doi: 10.1016/j.cell.2006.08.040
30.
Lek A, Evesson FJ, Lemckert FA, Redpath GM, Lueders AK, Turnbull L, Whitchurch CB, North KN, Cooper ST. Calpains, cleaved mini-dysferlinC72, and L-type channels underpin calcium-dependent muscle membrane repair. J Neurosci. 2013;33:5085–5094. doi: 10.1523/JNEUROSCI.3560-12.2013
31.
Brandenburg S, Pawlowitz J, Eikenbusch B, Peper J, Kohl T, Mitronova GY, Sossalla S, Hasenfuss G, Wehrens XH, Kohl P, et al. Junctophilin-2 expression rescues atrial dysfunction through polyadic junctional membrane complex biogenesis. JCI Insight. 2019;4:e127116. doi: 10.1172/jci.insight.127116
32.
Brandenburg S, Drews L, Schonberger HL, Jacob CF, Paulke NJ, Beuthner BE, Topci R, Kohl T, Neuenroth L, Kutschka I, et al. Direct proteomic and high-resolution microscopy biopsy analysis identifies distinct ventricular fates in severe aortic stenosis. J Mol Cell Cardiol. 2022;173:1–15. doi: 10.1016/j.yjmcc.2022.08.363
33.
Da Dalt L, Cabodevilla AG, Goldberg IJ, Norata GD. Cardiac lipid metabolism, mitochondrial function and heart failure. Cardiovasc Res. 2023;119:1905. doi: 10.1093/cvr/cvad100
34.
Yang ZF, Panwar P, McFarlane CR, Tuinte WE, Campiglio M, Van Petegem F. Structures of the junctophilin/voltage-gated calcium channel interface reveal hot spot for cardiomyopathy mutations. Proc Natl Acad Sci U S A. 2022;119:e2120416119. doi: 10.1073/pnas.2120416119
35.
Wang Y, Tadayon R, Santamaria L, Mercier P, Forristal CJ, Shaw GS. Calcium binds and rigidifies the dysferlin C2A domain in a tightly coupled manner. Biochem J. 2021;478:197–215. doi: 10.1042/BCJ20200773
36.
Guo A, Zhang X, Iyer VR, Chen B, Zhang C, Kutschke WJ, Weiss RM, Franzini-Armstrong C, Song LS. Overexpression of junctophilin-2 does not enhance baseline function but attenuates heart failure development after cardiac stress. Proc Natl Acad Sci U S A. 2014;111:12240–12245. doi: 10.1073/pnas.1412729111
37.
Olson EN. Gene regulatory networks in the evolution and development of the heart. Science. 2006;313:1922–1927. doi: 10.1126/science.1132292
38.
Dirkx E, da Costa Martins PA, De Windt LJ, da Costa Martins PA, De Windt LJ. Regulation of fetal gene expression in heart failure. Biochim Biophys Acta. 2013;1832:2414–2424. doi: 10.1016/j.bbadis.2013.07.023
39.
Demonbreun AR, Rossi AE, Alvarez MG, Swanson KE, Deveaux HK, Earley JU, Hadhazy M, Vohra R, Walter GA, Pytel P, et al. Dysferlin and myoferlin regulate transverse tubule formation and glycerol sensitivity. Am J Pathol. 2014;184:248–259. doi: 10.1016/j.ajpath.2013.09.009
40.
Kerr JP, Ziman AP, Mueller AL, Muriel JM, Kleinhans-Welte E, Gumerson JD, Vogel SS, Ward CW, Roche JA, Bloch RJ. Dysferlin stabilizes stress-induced Ca2+ signaling in the transverse tubule membrane. Proc Natl Acad Sci U S A. 2013;110:20831–20836. doi: 10.1073/pnas.1307960110
41.
Guo A, Wang Y, Chen B, Wang Y, Yuan J, Zhang L, Hall D, Wu J, Shi Y, Zhu Q, et al. E-C coupling structural protein junctophilin-2 encodes a stress-adaptive transcription regulator. Science. 2018;362:eaan3303. doi: 10.1126/science.aan3303
42.
Marty NJ, Holman CL, Abdullah N, Johnson CP. The C2 domains of otoferlin, dysferlin, and myoferlin alter the packing of lipid bilayers. Biochemistry. 2013;52:5585–5592. doi: 10.1021/bi400432f
43.
Sachse FB, Torres NS, Savio-Galimberti E, Aiba T, Kass DA, Tomaselli GF, Bridge JH. Subcellular structures and function of myocytes impaired during heart failure are restored by cardiac resynchronization therapy. Circ Res. 2012;110:588–597. doi: 10.1161/CIRCRESAHA.111.257428
44.
Brandenburg S, Pawlowitz J, Steckmeister V, Subramanian H, Uhlenkamp D, Scardigli M, Mushtaq M, Amlaz SI, Kohl T, Wegener JW, et al. A junctional cAMP compartment regulates rapid Ca(2+) signaling in atrial myocytes. J Mol Cell Cardiol. 2022;165:141–157. doi: 10.1016/j.yjmcc.2022.01.003
45.
Toischer K, Rokita AG, Unsold B, Zhu W, Kararigas G, Sossalla S, Reuter SP, Becker A, Teucher N, Seidler T, et al. Differential cardiac remodeling in preload versus afterload. Circulation. 2010;122:993–1003. doi: 10.1161/CIRCULATIONAHA.110.943431
46.
Vahanian A, Beyersdorf F, Praz F, Milojevic M, Baldus S, Bauersachs J, Capodanno D, Conradi L, De Bonis M, De Paulis R, et al; ESC/EACTS Scientific Document Group. 2021 ESC/EACTS Guidelines for the management of valvular heart disease. Eur Heart J. 2022;43:561–632. doi: 10.1093/eurheartj/ehab395
47.
Puls M, Beuthner BE, Topci R, Vogelgesang A, Bleckmann A, Sitte M, Lange T, Backhaus SJ, Schuster A, Seidler T, et al. Impact of myocardial fibrosis on left ventricular remodelling, recovery, and outcome after transcatheter aortic valve implantation in different haemodynamic subtypes of severe aortic stenosis. Eur Heart J. 2020;41:1903–1914. doi: 10.1093/eurheartj/ehaa033
48.
Weil MT, Ruhwedel T, Meschkat M, Sadowski B, Mobius W. Transmission electron microscopy of oligodendrocytes and myelin. Methods Mol Biol. 2019;1936:343–375. doi: 10.1007/978-1-4939-9072-6_20
49.
Dobin A, Davis CA, Schlesinger F, Drenkow J, Zaleski C, Jha S, Batut P, Chaisson M, Gingeras TR. STAR: ultrafast universal RNA-seq aligner. Bioinformatics. 2013;29:15–21. doi: 10.1093/bioinformatics/bts635
50.
Liao Y, Smyth GK, Shi W. featureCounts: an efficient general purpose program for assigning sequence reads to genomic features. Bioinformatics. 2014;30:923–930. doi: 10.1093/bioinformatics/btt656
51.
Love MI, Huber W, Anders S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 2014;15:550. doi: 10.1186/s13059-014-0550-8
52.
Gu Z, Eils R, Schlesner M. Complex heatmaps reveal patterns and correlations in multidimensional genomic data. Bioinformatics. 2016;32:2847–2849. doi: 10.1093/bioinformatics/btw313
53.
Cai X, Xue Z, Wu C, Sun R, Qian L, Yue L, Ge W, Yi X, Liu W, Chen C, et al. High-throughput proteomic sample preparation using pressure cycling technology. Nat Protoc. 2022;17:2307–2325. doi: 10.1038/s41596-022-00727-1
54.
Atanassov I, Urlaub H. Increased proteome coverage by combining PAGE and peptide isoelectric focusing: comparative study of gel-based separation approaches. Proteomics. 2013;13:2947–2955. doi: 10.1002/pmic.201300035
55.
Meier F, Brunner AD, Koch S, Koch H, Lubeck M, Krause M, Goedecke N, Decker J, Kosinski T, Park MA, et al. Online Parallel Accumulation-Serial Fragmentation (PASEF) with a novel trapped ion mobility mass spectrometer. Mol Cell Proteomics. 2018;17:2534–2545. doi: 10.1074/mcp.TIR118.000900
56.
Skowronek P, Thielert M, Voytik E, Tanzer MC, Hansen FM, Willems S, Karayel O, Brunner AD, Meier F, Mann M. Rapid and in-depth coverage of the (phospho-)proteome with deep libraries and optimal window design for dia-PASEF. Mol Cell Proteomics. 2022;21:100279. doi: 10.1016/j.mcpro.2022.100279
57.
Wu T, Hu E, Xu S, Chen M, Guo P, Dai Z, Feng T, Zhou L, Tang W, Zhan L, et al. clusterProfiler 4.0: a universal enrichment tool for interpreting omics data. Innovation (Camb). 2021;2:100141. doi: 10.1016/j.xinn.2021.100141
58.
Yu G, Wang LG, Han Y, He QY. clusterProfiler: an R package for comparing biological themes among gene clusters. OMICS. 2012;16:284–287. doi: 10.1089/omi.2011.0118
59.
Fitzgerald DJ, Schaffitzel C, Berger P, Wellinger R, Bieniossek C, Richmond TJ, Berger I. Multiprotein expression strategy for structural biology of eukaryotic complexes. Structure. 2007;15:275–279. doi: 10.1016/j.str.2007.01.016
60.
Bieniossek C, Nie Y, Frey D, Olieric N, Schaffitzel C, Collinson I, Romier C, Berger P, Richmond TJ, Steinmetz MO, et al. Automated unrestricted multigene recombineering for multiprotein complex production. Nat Methods. 2009;6:447–450. doi: 10.1038/nmeth.1326
61.
Jahn O, Siems SB, Kusch K, Hesse D, Jung RB, Liepold T, Uecker M, Sun T, Werner HB. The CNS myelin proteome: deep profile and persistence after post-mortem delay. Front Cell Neurosci. 2020;14:239. doi: 10.3389/fncel.2020.00239
62.
Sondermann JR, Barry AM, Jahn O, Michel N, Abdelaziz R, Kugler S, Gomez-Varela D, Schmidt M. Vti1b promotes TRPV1 sensitization during inflammatory pain. Pain. 2019;160:508–527. doi: 10.1097/j.pain.0000000000001418
Information & Authors
Information
Published In
Copyright
© 2024 American Heart Association, Inc.
Versions
You are viewing the most recent version of this article.
History
Received: 11 March 2024
Revision received: 5 July 2024
Accepted: 8 July 2024
Published online: 16 July 2024
Published in print: 16 August 2024
Keywords
Subjects
Authors
Disclosures
None.
Funding Information
Deutsche Forschungsgemeinschaft (DE)501100001659
Deutsche Forschungsgemeinschaft (DE)501100001659: EXC 2067/1- 390729940
Deutsche Forschungsgemeinschaft (DE)501100001659: CRC1190 project P03 and Z02
Deutsche Forschungsgemeinschaft (DE)501100001659: CRC1425 project P11
Deutsche Forschungsgemeinschaft (DE)501100001659: 396913060
This article was funded by the Deutsche Forschungsgemeinschaft (DFG, German Research Foundation): collaborative research center SFB1002 (193793266) to S. Brandenburg (A09), K. Toischer (D04), G. Hasenfuß (D01), C. Lenz (A09), and S.E. Lehnart (A09 and S02); Germany’s Excellence Strategy - EXC 2067/1- 390729940 Multiscale Bioimaging to W. Möbius, G. Hasenfuß, T. Moser, J. Preobraschenski, and S.E. Lehnart; and Hertha Sponer College to Y. Zühlke; collaborative research center SFB1190 to S.E. Lehnart (P03) and H. Urlaub (Z02); Heisenberg program (TO 822/1-1) to K. Toischer; collaborative research center SFB1425 to E.A. Rog-Zielinska (P11); Emmy Noether Fellowship to E.A. Rog-Zielinska (396913060); International Research Training Group 1816, doctoral stipend to N.J. Paulke. Y. Zühlke, J.B. Wegener, and L. Liebmann were financially supported through stipends of the Göttingen Promotionskolleg für Medizinstudierende, funded by the Jacob-Henle-Programm/Else-Kröner-Fresenius-Stiftung. C. Fleischhacker and J. Wedemeyer received funding through a doctoral stipend of the German Centre for Cardiovascular Research (DZHK, Deutsches Zentrum für Herz-Kreislauf-Forschung). The German Cardiac Society (DGK, Deutsche Gesellschaft für Kardiologie) funded C. Fleischhacker through a doctoral stipend. C. Lenz and S.E. Lehnart were funded by the Leducq Foundation (CURE-PLaN).
Metrics & Citations
Metrics
Citations
Download Citations
If you have the appropriate software installed, you can download article citation data to the citation manager of your choice. Select your manager software from the list below and click Download.
- PTN secreted by cardiac fibroblasts promotes myocardial fibrosis and inflammation of pressure overload-induced hypertrophic cardiomyopathy through the PTN-SDC4 pathway, Life Sciences, 363, (123356), (2025).https://doi.org/10.1016/j.lfs.2024.123356
- Different effects of cardiomyocyte contractile activity on transverse and axial tubular system luminal content dynamics, Journal of Molecular and Cellular Cardiology, 197, (125-135), (2024).https://doi.org/10.1016/j.yjmcc.2024.10.014
Loading...
View Options
Login options
Check if you have access through your login credentials or your institution to get full access on this article.
Personal login Institutional LoginPurchase Options
Purchase this article to access the full text.
eLetters(0)
eLetters should relate to an article recently published in the journal and are not a forum for providing unpublished data. Comments are reviewed for appropriate use of tone and language. Comments are not peer-reviewed. Acceptable comments are posted to the journal website only. Comments are not published in an issue and are not indexed in PubMed. Comments should be no longer than 500 words and will only be posted online. References are limited to 10. Authors of the article cited in the comment will be invited to reply, as appropriate.
Comments and feedback on AHA/ASA Scientific Statements and Guidelines should be directed to the AHA/ASA Manuscript Oversight Committee via its Correspondence page.