Skip to main content

CaMKIIβ deregulation contributes to neuromuscular junction destabilization in Myotonic Dystrophy type I

Abstract

Background

Myotonic Dystrophy type I (DM1) is the most common muscular dystrophy in adults. Previous reports have highlighted that neuromuscular junctions (NMJs) deteriorate in skeletal muscle from DM1 patients and mouse models thereof. However, the underlying pathomechanisms and their contribution to muscle dysfunction remain unknown.

Methods

We compared changes in NMJs and activity-dependent signalling pathways in HSALR and Mbnl1ΔE3/ΔE3 mice, two established mouse models of DM1.

Results

Muscle from DM1 mouse models showed major deregulation of calcium/calmodulin-dependent protein kinases II (CaMKIIs), which are key activity sensors regulating synaptic gene expression and acetylcholine receptor (AChR) recycling at the NMJ. Both mouse models exhibited increased fragmentation of the endplate, which preceded muscle degeneration. Endplate fragmentation was not accompanied by changes in AChR turnover at the NMJ. However, the expression of synaptic genes was up-regulated in mutant innervated muscle, together with an abnormal accumulation of histone deacetylase 4 (HDAC4), a known target of CaMKII. Interestingly, denervation-induced increase in synaptic gene expression and AChR turnover was hampered in DM1 muscle. Importantly, CaMKIIβ/βM overexpression normalized endplate fragmentation and synaptic gene expression in innervated Mbnl1ΔE3/ΔE3 muscle, but it did not restore denervation-induced synaptic gene up-regulation.

Conclusions

Our results indicate that CaMKIIβ-dependent and -independent mechanisms perturb synaptic gene regulation and muscle response to denervation in DM1 mouse models. Changes in these signalling pathways may contribute to NMJ destabilization and muscle dysfunction in DM1 patients.

Background

Myotonic Dystrophy type I (DM1) is a multisystemic disorder caused by expanded CTG triplet repeats in the 3’UTR of the DMPK (Dystrophia Myotonica Protein Kinase) gene that leads to muscle wasting, weakness and inability to relax (myotonia). Accumulation of toxic transcripts containing the expanded CUG repeats leads to the nuclear sequestration of splicing factors. The consecutive mis-splicing of specific genes is determinant in the pathogenesis of DM1-associated muscle alterations [1, 2]. For example, mis-splicing of the CLCN1 gene, which encodes the chloride channel ClC-1, is implicated in myotonia development. Mis-splicing of several genes encoding proteins of Ca2+-associated signalling pathways has also been shown to contribute to muscle dysfunction. In particular, previous reports revealed that mis-splicing of CAMK2 genes, encoding Ca2+/calmodulin-dependent protein kinases II (CaMKIIs), is a hallmark of DM1 [3,4,5]. However, the functional consequences of CAMKII deregulation in DM1 muscle and its contribution to DM1 pathogenesis have not been explored.

CaMKIIs are important for the maintenance of neuromuscular junctions (NMJs), which are the synapses connecting motor neurons to muscle fibres. Especially, CaMKIIs enhance the recycling of acetylcholine receptors (AChRs) upon their internalization in the sub-synaptic region of muscle fibres (i.e., the endplate) [6]. Moreover, CaMKIIs indirectly repress the expression of synaptic genes in non-synaptic regions of innervated muscle, by inhibiting the myogenic transcription factor myogenin and the histone deacetylase 4 (HDAC4) [7,8,9]. Hence, deregulation of the CaMKII signalling pathway may affect NMJ maintenance, by perturbing the expression and the dynamics of synaptic proteins.

Early studies on DM1 muscle biopsies pointed to NMJ-associated abnormalities, such as a reduced number of presynaptic vesicles, enlarged endplates or angular muscle fibres [10,11,12,13]. NMJ alterations have also been reported in DMSXL mice and in mice deficient for Muscleblind-like 1/2 (Mbnl1/2), two mouse models of DM1, as well as in C. elegans DM1 mutants [14, 15]. The lack of denervation markers (e.g., non-junctional AChR clusters) and the absence of massive motor neuron loss rejected the hypothesis that spontaneous denervation is part of DM1 pathogenesis [16, 17]. However, the contribution of neuronal deregulations to NMJ defects has recently been proposed from human motoneuron / muscle cells co-culture [18]. Nuclear foci, characteristic of DM1-associated accumulation of toxic RNA, were detected at the endplate and may alter the expression of synaptic genes [19]. Consistently, mis-splicing of some synaptic genes has been reported in muscle cells from DM1 patients [20]. Hence, NMJ deterioration in DM1 is likely to contribute to muscle dysfunction, but the underlying pathomechanisms remain unknown.

Here, we analysed changes in activity-dependent signalling pathways in HSALR and Mbnl1ΔE3/ΔE3 mice, two established DM1 mouse models. Both mouse models displayed increased fragmentation of the endplate, which preceded muscle alterations. HSALR and Mbnl1ΔE3/ΔE3 mice also showed major deregulation of CaMKIIβ signalling in muscle, which contributed to increase synaptic gene expression and endplate fragmentation in innervated conditions. Interestingly, the muscle response to denervation was hampered in Mbnl1ΔE3/ΔE3 mice, independently from CaMKIIβ/βM deficiency. Hence, perturbations in activity-dependent signalling pathways may contribute to NMJ destabilization and muscle dysfunction in DM1 patients.

Methods

Mice

Homozygous mice of the mouse line LR20b carrying about 250 (CTG) repeats within the HSA transgene (HSALR) were obtained from Thornton and colleagues (University of Rochester Medical Centre, Rochester, New York, USA) [21]. Mice of the corresponding background strain (FVB/N) were used as controls. Mice were genotyped for the HSALR transgene by quantifying human ACTA1 levels normalized to endogenous actin (mouse Acta1) in genomic DNA. Mice from the Mbnl1ΔE3 line were obtained from Swanson and colleagues (College of Medicine, University of Florida, Gainesville, Florida, USA) [22]. Mbnl1+/+ littermates from the corresponding background strain (C57BL/6) were used as controls. Mice from the Mbnl1ΔE3 line were genotyped for exon 3 depletion at the Mbnl1 locus. Mice were maintained in a conventional specific-pathogen-free facility with a fixed light cycle (23 °C, 12 h dark-light cycle). For the AAV study, anterior hindlimb compartments of control and mutant mice were injected with adeno-associated virus serotype 9 (AAV9; GeneCopoeia) carrying either the transgene for CaMKIIβ (AA09-Mm33884-AV01-A00-GS), CaMKIIβM (AA09-Mm44818-AV01-A00-GS) or GFP (AA09-NEG-AV07-A00) at a dose of 8 × 1010 viral particles per compartment. Sciatic nerve cut was conducted as described previously [23]. All animal studies were performed in accordance with the European Union guidelines for animal care and approved by the Veterinary Office of the Cantons of Basel city (application number 2601) and Geneva (application number GE220/GE227).

Muscle force and relaxation

In vitro force measurement of EDL muscle and late relaxation time evaluation were conducted as previously described [24, 25].

Western blotting

Muscles powdered in liquid nitrogen were lysed in cold RIPA + buffer (50 mM Tris-HCl pH 8, 150 mM NaCl, 1% NP-40, 0.5% sodium deoxycholate, 0.1% SDS, 1% Triton X, 10% glycerol, phosphatase and protease inhibitors). Subcellular fractionation was done according to Dimauro et al. (2012) [26]. Following dosage (BCA Protein Assay, Pierce), proteins were separated on SDS-polyacrylamide gels and transferred to nitrocellulose membrane. Blots were blocked in TBS, 3% BSA, 0.1% Tween-20, and incubated overnight at 4 °C with primary antibodies, then for 2 h with HRP-labelled secondary antibodies. Immunoreactivity was detected using the Western blot chemiluminescent substrate LumiGLO (Seracare) and exposed to Super RX-N films (Fujifilm) or revealed with iBright™ Imaging System (ThermoFisher). Protein expression was normalized to α-actinin or GAPDH, or to total protein levels of the corresponding phosphorylated form. The list of antibodies used is provided in Supplementary Materials.

Polymerase chain reaction

Total RNAs were extracted with the RNeasy Mini Kit (Qiagen), reverse transcribed with the SuperScript III First-Strand Synthesis System (Invitrogen), and amplified with the Power SYBR Green Master Mix (Applied Biosystems) or the Hot FirePol EvaGreen qPCR Mix (Solis BioDyne). Expression of specific spliced or pan transcripts was analysed by end-point PCR and electrophoresis, or by quantitative PCR with the Step-One software and normalization to Tbp expression. The list of primers used is provided in Table S1.

Histology and immunofluorescence

Muscles were frozen in liquid nitrogen-cooled isopentane. Eight-micrometre muscle sections were stained with H&E and observed with an upright microscope (Olympus). For immunostaining, sections were fixed with 4% paraformaldehyde (PFA) or kept unfixed, then blocked in PBS, 3% BSA, incubated sequentially with primary and secondary fluorescent antibodies (Invitrogen, Jackson ImmunoResearch), mounted with Vectashield medium (Vector), and observed with Leica or Zeiss fluorescent microscopes. Quantification of fibre type and size was done as previously reported [27].

Muscle bundle staining

To analyse NMJ organization, muscles were bathed ex vivo with α-bungarotoxin(Btx)-Alexa555 (2 µg/ml - Invitrogen) for 30 min, before being washed and fixed with 4% PFA. Muscle bundles were cut, permeabilized in PBS, 1% Triton-X100, and blocked in PBS, 1% BSA, 0.1% Triton-X100. Bundles were then successively incubated with primary antibodies against neurofilament and synaptophysin (to stain pre-synaptic compartment), and the corresponding secondary antibodies (Invitrogen, Jackson ImmunoResearch). Images were recorded using Leica and Zeiss confocal microscopes. 3D reconstructed images of endplates, based on Btx staining, were obtained with Imaris 10.1.1 software. The number of AChR fragments per endplate were counted manually in a double-blind manner. Volume and projected area of Btx-stained AChRs per endplate were measured with ImageJ/Fiji software.

AChR turnover

AChR turnover was assessed by injecting Btx-Alexa647 and -Alexa555 (25 pmoles - Invitrogen) into TA/EDL muscles at days 1 and 10, respectively (5 and 14 days after nerve cut when combined with denervation), as previously described [23]. For turnover quantification, images were recorded using Leica or Zeiss confocal microscopes. Pixel dominance (old vs. new receptors) was calculated using Fiji and MATLAB software. The assays could be applied to TA and EDL muscles, but not to the gastrocnemius muscle because of its size and heterogeneity.

Statistics

Quantitative data are displayed as mean ± SEM of independent samples, with n (number of individual experiments) ≥ 3. The statistical analysis of values was performed using unpaired Student’s t test or two-way ANOVA test with Tukey’s multiple comparisons test correction, with prior log transformation of the data that were not normally distributed (RNA and protein levels, relaxation time). A 0.05 level of confidence was accepted for statistical significance.

Results

DM1-associated muscle alterations are similar in Mbnl1 ΔE3/ΔE3 and HSA LR mice

To evaluate changes in NMJs and activity-dependent pathways in DM1, we selected the Mbnl1ΔE3/ΔE3 and HSALR mouse lines, which are two well-established DM1 mouse models. In Mbnl1ΔE3/ΔE3 mice, deletion of Mbnl1 exon 3 leads to body-wide depletion of the splicing factor MBNL1 [22]. In contrast, HSALR mice express the HSA transcript (Human Skeletal Actin) with long (CTG) repeats only in skeletal muscle, which allowed to unveil cell-autonomous defects [21]. First, we compared the muscle phenotype between the two mouse models. There was no dystrophic sign in muscles from 3-month-old mutant mice (Fig. 1A, B), as previously reported [21, 22]. The dystrophic phenotype remained moderate in 9- and 12-month-old Mbnl1ΔE3/ΔE3 and HSALR mice, respectively (Fig. 1A, B). In particular, fibre size variation, increased proportion of fibres with internalized myonuclei, as well as intracellular vacuoles were present in Mbnl1ΔE3/ΔE3tibialis anterior (TA) and gastrocnemius muscles (Fig. 1A), as well as in gastrocnemius muscle from HSALR mice (Fig. 1B). In contrast, these alterations were rare in TA muscle from HSALR mice (Fig. 1B). This was consistent with previous reports describing differential muscle severity in this mouse model [28], which may pertain to the HSA promoter. This heterogeneity between muscles was not observed in Mbnl1ΔE3/ΔE3 mice. Both Mbnl1ΔE3/ΔE3 and HSALR mice exhibited a myotonic phenotype, as shown by the increased late relaxation time of extensor digitorum longus (EDL) muscle after ex vivo stimulation at 3 months (Fig. 1C and Fig. S1A) and 9/12 months (Fig. S1B) of age. In contrast, specific tetanic muscle force (sP0) was unaffected in 3-month-old mutant mice (Fig. 1D) and only slightly reduced in 9-month-old Mbnl1ΔE3/ΔE3 mice (Fig. S1C).

To evaluate DM1-associated mis-splicing, we next quantified the inclusion of exons 7a and 22 of the Clcn1 and Atp2a1 genes, which encode ClC-1 channel and SERCA1 (Sarco/Endoplasmic Reticulum Ca2+-ATPase), respectively. The inclusion of Clcn1 exon 7a was strongly increased in Mbnl1ΔE3/ΔE3 TA muscle (Fig. 1E) and was accompanied by major reduction in total Clcn1 transcript levels (Fig. 1F). Clcn1 mis-splicing and down-regulation were similar in gastrocnemius muscle from HSALR mice (Fig. 1E, F). In contrast, TA muscle from HSALR mice showed milder changes in Clcn1 splicing and no reduction in total Clcn1 transcript levels (Fig. 1E, F). Similarly, the inclusion of Atp2a1 exon 22 was abrogated in Mbnl1ΔE3/ΔE3 TA muscle and in gastrocnemius muscle from HSALR mice, while it was reduced by only half in HSALR TA muscle (Fig. 1G). These results confirm that both mouse models have mild muscle alterations, with similar DM1-associated phenotype observed in Mbnl1ΔE3/ΔE3 muscles and HSALRgastrocnemius muscle, and milder changes detected in TA muscle from HSALR mice.

Fig. 1
figure 1

Muscle phenotype in Mbnl1ΔE3/ΔE3 and HSALR mice. A, B H&E coloration reveals moderate myopathic alterations in TA and gastrocnemius (GA) muscles from 3- and 9-month(M)-old Mbnl1ΔE3/ΔE3 (A) and 3- and 12-month(M)-old HSALR mice (B). Asterisks and arrows point to internalized nuclei and vacuoles, respectively. Scale bar, 100 μm. C Late relaxation time upon stimulation is increased in EDL muscle from 3-month-old Mbnl1ΔE3/ΔE3 and HSALR mice, as compared to control mice. n = 4 Ctrl / 3 Mbnl1ΔE3/ΔE3; 9 Ctrl / 30 HSALR. D Specific tetanic force (sP0) of EDL muscle is unchanged in 3-month-old Mbnl1ΔE3/ΔE3 (n = 4 Ctrl / 3 KO) and HSALR (n = 9 Ctrl / 30 HSALR) mice. E-G qPCR analysis of Clcn1 exon7a inclusion (E), Clcn1 total mRNA levels (F) and Atp2a1 exon22 inclusion (G) in TA muscle from Mbnl1ΔE3/ΔE3 mice and gastrocnemius (GA) or TA muscle from HSALR mice. Expression is normalised on total mRNA expression for splice variants (E, G) or on Tbp expression (F). Levels are relative to control and expressed as log2(Fold Change). n = 4 Ctrl / 3 Mbnl1ΔE3/ΔE3 (E, F); 6 Ctrl / 5 Mbnl1ΔE3/ΔE3 (G); 4 Ctrl / 4 HSALR. All data represent mean ± SEM. **p < 0.01; ****p < 0.0001; unpaired two-tailed Student’s t test

CaMKIIs are strongly deregulated in muscle from DM1 patients and mouse models

Previous reports pointed to mis-splicing of CAMK2 genes in DM1 tissues [3,4,5]. However, it remains unclear which CaMKII isoforms are affected in DM1 muscle, and what the consequences of their deregulation are. CaMKIIβ/γ/δ isoforms are encoded by the three genes CAMK2B, 2G and 2D, and are expressed in skeletal muscle. Each gene is expressed as different splice variants. In particular, CaMKIIβ splice variants include CaMKIIβ/βe/β’/βe’ and the muscle-specific variant CaMKIIβM, which arise from the alternative splicing of CAMK2b exons 13, 16 and 18–20 (Fig. S2A) [29,30,31]. Protein levels of CaMKIIβM, which can be distinguished from other CaMKII isoforms based on its size, were strongly reduced in both Mbnl1ΔE3/ΔE3 (Fig. 2A) and HSALR (Fig. 2B) muscles. Auto-phosphorylation of CaMKIIβM (phospho-Thr287) decreased as well (Fig. 2C), suggesting reduced CaMKIIβM activity. Notably, additional bands around the size of the other isoforms of CaMKIIβ and of CaMKIIγ/δ suggest the expression of alternative CaMKII isoforms in mutant muscles (Fig. 2A, B).

To characterize splicing events in the Camk2b, 2d and 2g genes in DM1 muscle, we used RNA-seq data that we obtained from HSALRgastrocnemius muscle. We detected significant changes in Camk2b transcript, with a major exclusion of exons 13 and 18 to 20 in HSALR muscle, compared to control (Fig. S2B, C). There was no change in the splicing of Camk2b exon 16 in HSALR muscle (Fig. S2B, C). We confirmed the expression of Camk2b transcripts lacking exon 13 (corresponding to CaMKIIβe/e’) in HSALR muscle by RT-PCR (Fig. S2D). Moreover, the amplicon including exons 18 to 20, which encode the variable inserts of CaMKIIβM, was barely detected in HSALR muscle (Fig. S2D). By quantitative RT-PCR, total levels of Camk2b transcripts increased in Mbnl1ΔE3/ΔE3 TA muscle, as well as in HSALRgastrocnemius, TA and EDL muscles (Fig. 2D). Using primers spanning Camk2b exons 12–14, we confirmed that the expression of Camk2b transcripts without exon 13 was more than four times higher in mutant muscles than in controls (Fig. 2E). Conversely, levels of Camk2b transcripts with exons 18 to 20 were reduced by half in Mbnl1ΔE3/ΔE3 TA muscle and in gastrocnemius muscle from HSALR mice (Fig. 2F) compared to controls. Notably, TA and EDL muscles from HSALR mice showed similar extent of Camk2b mis-splicing compared to gastrocnemius muscle (Fig. S2E, F). Interestingly, the mis-splicing of exon 13 and exons 18–20 is also confirmed by RNA-seq data of TA muscle from DM1 patients (Fig. S3) [32]. These results indicate that skeletal muscles from DM1 patients and mouse models shift to the expression of CaMKIIβe isoform (arising from exons 13/18–20 exclusion, with exon 16 inclusion), while CaMKIIβ (+ ex13 + ex16Δex18-20) and CaMKIIβM (+ ex13 + ex16 + ex18-20) are the predominant isoforms expressed in control muscles.

Transcripts encoding CaMKIIγ were also mis-spliced in HSALR muscle. These included increased inclusion of exons 13, 15 and 19 in Camk2g transcripts in HSALR muscle, as seen in RNA-seq reads (Fig. S4A, B) and by RT-PCR (Fig. S4C). Similar Camk2g mis-splicing was observed in Mbnl1ΔE3/ΔE3 TA muscle (Fig. S4D). Increased inclusion of CAMK2G exon 19 was also observed in TA biopsies from DM1 patients (Fig. S4E) [32]. In contrast, there was no major splicing change detected for Camk2d in HSALR muscle (Fig. S5A) and CAMK2D in DM1 muscle (Fig. S5B) [32]. Taken together, these results show that mis-splicing of some Camk2 transcripts alters the expression pattern of CaMKIIs in DM1 muscle.

Fig. 2
figure 2

CaMKIIβ deregulation in Mbnl1ΔE3/ΔE3 and HSALR muscles. A-C Western blot analysis of CaMKII isoforms and quantification of CaMKIIβM levels in TA muscle from 3-month-old Mbnl1ΔE3/ΔE3 mice (A) and in gastrocnemius muscle from 3-month-old HSALR mice (B). Quantification of CaMKIIβM phosphorylated form in Mbnl1ΔE3/ΔE3 and HSALR muscles is shown in C. Protein levels are normalized on α-actinin (A, B) or on total CaMKIIβM (C), relative to control and expressed as log2(Fold Change). n = 3 (Ctrl/Mbnl1ΔE3/ΔE3) and 4 (Ctrl/HSALR) per group. D-F Quantitative RT-PCR analysis of total Camk2b mRNA levels (D), Camk2 exon 13 exclusion (E), and Camk2b exons 18–20 inclusion (F) in TA muscle from Mbnl1ΔE3/ΔE3 mice and in gastrocnemius (GA), TA and EDL muscles from HSALR mice. Data are normalized on Tbp levels (D) or on total Camk2b transcripts (E, F), relative to control and expressed as log2(Fold Change). n = 4 Ctrl / 3 Mbnl1ΔE3/ΔE3; 4 Ctrl / 4 HSALR (D, E); 3 Ctrl / 3 HSALR (F). All data are mean ± SEM; * p < 0.05; ** p < 0.01; ***p < 0.001; **** p < 0.0001; two-tailed unpaired Student’s t-test

Endplate fragmentation is not caused by abnormal AChR turnover in Mbnl1 ΔE3/ΔE3 and HSA LR mice

As CaMKIIs are key sensors of neural activity involved in NMJ maintenance, we analysed NMJ structures in EDL, TA and gastrocnemius muscles from 3-month-old and 9- or 12-month-old Mbnl1ΔE3/ΔE3 and HSALR mice. Pre- and post-synaptic compartments were stained in whole-mount muscle bundles with antibodies against neurofilament/synaptophysin and with α-bungarotoxin (Btx), which binds specifically to AChRs, respectively. The overall organization of the NMJs was preserved in mutant mice (Fig. 3A, B). In particular, we did not observe signs of denervation or abnormal axonal termination in mutant muscles. Acetylcholinesterase staining was also similar in control and mutant mice (Fig. S6A, B). However, the number of AChR fragments per endplate increased in muscles from both 3- and 9-month-old Mbnl1ΔE3/ΔE3 mice, compared to age-matched controls (Fig. 3C and Fig. S6C). A similar increase in endplate fragmentation was observed in gastrocnemius, TA and EDL muscles from HSALR mice (Fig. 3D and Fig. S6D). Of note, the volume occupied by AChRs was unchanged in Mbnl1ΔE3/ΔE3 and HSALR mice (Fig. S6E). There was a slight increase in the projected endplate area in HSALR EDL muscle, which may arise from endplate fragmentation (Fig. S6F). As the HSALR transgene is specifically expressed in skeletal muscle, these results indicate that post-synaptic perturbations contribute to endplate fragmentation in HSALR mice. Moreover, these NMJ alterations were detected as soon as 3 months of age, i.e., before changes in muscle histology, suggesting that they are primary deficit in DM1 mouse models and not a consequence of muscle degeneration/regeneration.

Fig. 3
figure 3

Altered NMJ maintenance in Mbnl1ΔE3/ΔE3 and HSALR mice. A, B Fluorescent images of NMJ regions stained with α-bungarotoxin (Btx, red), antibodies against neurofilament/synaptophysin (green), and dapi (blue) in EDL muscles from 3- and 9-month-old Mbnl1ΔE3/ΔE3 mice (A) and 3-and 12-month-old HSALR mice (B). Scale bar, 50 μm. C, D Quantification of the number of fragments per endplate in EDL, TA and gastrocnemius (GA) muscles from 3- and 9/12-month-old Mbnl1ΔE3/ΔE3 (C) and HSALR (D) mice. n = 5/8 (EDL 3 M), 3/4 (TA 3 M), 4/4 (GA 3 M), 5/3 (all muscles 9 M) Ctrl/ Mbnl1ΔE3/ΔE3 (C); 11/12 (EDL 3 M), 7/8 (TA 3 M), 7/8 (GA 3 M), 4/3 (EDL 12 M), 4/4 (TA and GA 12 M) Ctrl/HSALR (D), with more than 50 fibres per muscle. Data represent mean ± SEM. **p < 0.01, ***p < 0.001, ****p < 0.0001, unpaired two-tailed Student’s t test

As CaMKIIs have been shown to regulate AChR recycling at the endplate [6], we next evaluated AChR turnover, by labelling “old” and newly formed receptors by two sequential injections of differently labelled Btx (Fig. 4A, B) [23, 33]. AChR turnover was low in all muscles, as described before for innervated conditions, with no change detected in Mbnl1ΔE3/ΔE3 muscles compared to controls (Fig. 4C). Similarly, there was no significant change in AChR turnover in TA and EDL muscles from HSALR mice (Fig. 4D). These results indicate that CaMKII deregulation, and more specifically the loss of CaMKIIβM, do not alter AChR dynamics in innervated DM1 muscle.

Fig. 4
figure 4

AChR dynamics is not altered in Mbnl1ΔE3/ΔE3 and HSALR muscles. A Timeline of injections of α-bungarotoxin (Btx) for AChR turnover assay. d, days. Created with BioRender.com. B Turnover assay in EDL and TA muscles from Mbnl1ΔE3/ΔE3 and HSALR mice. Fluorescent images show “old” (green) and “new” (red) AChRs in Mbnl1ΔE3/ΔE3 and HSALR muscles. Scale bar, 50 μm. C, D AChR turnover in EDL and TA muscles from Mbnl1ΔE3/ΔE3 (C) and HSALR (D) mice. n = 10/11 (EDL) and 5/5 (TA) Ctrl/Mbnl1ΔE3/ΔE3 (C); 7/8 (EDL) and 3/3 (TA) Ctrl/HSALR (D), with more than 22 fibres per muscle. Data are mean ± SEM.

Synaptic gene expression and muscle fibre type composition are altered in DM1 mouse models

CaMKIIs are well known to mediate activity-dependent regulations (e.g., of synaptic gene expression) in adult skeletal muscle [34]. To assess the consequences of CaMKII deregulation in DM1 muscle, we first measured mRNA levels of Myog, which encodes myogenin, and of the synaptic genes Musk, Chrna1, Chrne and Chrng genes, which encode MUSK and the α, ε and γ subunits of AChR, respectively. During muscle development, expression of Musk and Chrna1 becomes restricted to sub-synaptic myonuclei upon muscle innervation [35]. Simultaneously, Chrng transcripts are downregulated, while Chrne starts to be expressed in sub-synaptic nuclei (AChRε subunits replace AChRγ subunits). In non-synaptic regions of muscle fibres, synaptic gene repression is mediated by CaMKIIs and dependent on HDAC4/5 and myogenin inhibition (Fig. 5A) [8, 36]. This adult gene expression pattern depends on neural activity, as denervation reverts it back to a developmental pattern. Transcript levels of Myog tended to be higher in Mbnl1ΔE3/ΔE3 TA and HSALRgastrocnemius innervated muscles, compared to control muscles (Fig. 5B, C). In parallel, transcript levels of Musk, Chrna1, Chrne and Chrng were strongly increased in both mutant muscles (Fig. 5B, C). Their levels were less or not changed in EDL and TA muscles from HSALR mice (Fig. S7A, B).

Expression of Myh genes, which encode myosin heavy chains (MHC), is also dependent on neural activity and Ca2+-associated signalling [37]. In particular, by inhibiting the activity of myogenin and HDAC4, CaMKIIs may perturb the expression of Myh2 and Myh4, encoding MHCIIA and MHCIIB, respectively (Fig. 5A). Indeed, HDAC4 was shown to regulate the switch to type IIA fibres in TA muscle after nerve injury, by indirectly promoting the expression of Myh2 and directly repressing Myh4 [8]. Expression of Myh2 was strongly increased in Mbnl1ΔE3/ΔE3 innervated muscle, as compared to control (Fig. 5D). In contrast, Myh4 transcript levels were reduced in mutant muscle (Fig. 5D). Consistently, innervated TA muscle from Mbnl1ΔE3/ΔE3 mice displayed major accumulation of type IIA fibres and a loss of type IIB fibres compared to controls (Fig. 5E, F). Similarly, we previously described a mild switch towards type IIA fibres in TA muscle from HSALR mice [25]. There was also a tendency towards increased Myh2 transcript levels and reduced expression of Myh4 in HSALR TA muscle, as compared to controls (Fig. 5G). These results were in line with recent data describing myotonia-dependent switch towards oxidative muscle [38]. These results highlight that activity-dependent regulations underlying synaptic gene expression and fibre type composition, are perturbed in muscles from DM1 mouse models, which may involve CaMKII deregulation.

Fig. 5
figure 5

DM1 model mice display deregulation of activity-dependent signalling pathways. A Regulation of activity-dependent pathways by CaMKII. B, C Quantitative RT-PCR analysis of Myog, Musk, Chrna1, Chrne and Chrng in TA muscle from 3-month-old Mbnl1ΔE3/ΔE3 mice (B) and in gastrocnemius muscle from 3-month-old HSALR mice (C). n = 6/5 Ctrl/ Mbnl1ΔE3/ΔE3 (except for Myog, n = 4/3) and 5/7 Ctrl/HSALR (except for Myog and Chrna1, n = 4). D mRNA levels of Myh2 and Myh4, encoding MHC2A and MHC2B, in Mbnl1ΔE3/ΔE3 TA muscle. n = 6/5 Ctrl/Mbnl1ΔE3/ΔE3. E, F Fluorescent images of control and Mbnl1ΔE3/ΔE3 muscles, stained with antibodies against MHC2A (red) and laminin (green), and quantification of the proportion of type IIA, IIX and IIB fibres in control and mutant muscles (F). Scale bar, 500 μm. G mRNA levels of Myh2 and Myh4 in HSALR TA muscle. n = 3/4 Ctrl/ HSALR. All transcript levels (B, C, D, G) are normalized to Tbp, relative to control and expressed as log2(Fold Change). All data are mean ± SEM; * p < 0.05; ** p < 0.01; *** p < 0.001; ****p < 0.0001; two-tailed unpaired Student’s t-test

HDAC4 accumulates in Mbnl1 ΔE3/ΔE3 and HSA LR muscles

CaMKIIs inhibit the nuclear import, and thereby the activity of HDACs, such as HDAC4 [39]. To determine whether changes in HDAC4 pathway mediate the effect of CaMKII deregulation on activity-dependent regulations in DM1 mouse models, we analysed the expression of HDAC4 and of its target genes in Mbnl1ΔE3/ΔE3 and HSALR muscles. While transcript levels of Hdac4 were unchanged in Mbnl1ΔE3/ΔE3 and HSALR muscles (Fig. S8A), HDAC4 protein levels were higher in mutant muscles, as compared to controls (Fig. 6A, B). CaMKII-dependent phosphorylation of HDAC4 (Ser632) remained unchanged in Mbnl1ΔE3/ΔE3 muscle, but decreased in HSALR muscle (Fig. 6A and Fig. S8B). Notably, HDAC4 accumulated in nuclear fractions of Mbnl1ΔE3/ΔE3 and HSALR muscles (Fig. 6C, D). In contrast, HDAC4 protein levels were unchanged in cytosolic fractions of mutant muscles (Fig. 6E, F). This suggests that HDAC4 nuclear import is increased in Mbnl1ΔE3/ΔE3 and HSALR muscles. HDAC4 remained, however, barely detectable in mutant muscles by immunostaining (Fig. S8C). To assess whether HDAC4 accumulation translates into higher nuclear activity, we next evaluated the expression of target genes directly repressed by HDAC4. We focused on Dach2 and Mitr [36, 40], as well as on Ramp2 (Receptor Activity Modifying Protein 2), Actc1 (Actin alpha cardiac muscle 1), Cdh1 (Cadherin 1) and Dhrs7c (Dehydrogenase/Reductase 7 C), which were identified in public RNA-seq data as HDAC4 targets repressed after denervation [41]. Consistent with higher HDAC4 activity, transcript levels of Dach2, Actc1, Ramp1, and to a lesser extent of Dhrs7c, were reduced in Mbnl1ΔE3/ΔE3 and/or HSALR muscles (Fig. 6G), as observed for Myh4 (Fig. 5D, G). However, the expression of other genes was unchanged (Cdh1) or increased (Mitr) in mutant muscles compared to controls (Fig. 6G), suggesting incomplete or mild increase in HDAC4 activity. Hence, nuclear HDAC4 accumulation in skeletal muscle from DM1 mouse models may contribute to transcriptional changes of some activity-dependent genes downstream of CaMKII deregulation in innervated conditions.

Fig. 6
figure 6

Changes in HDAC4 signalling pathway in Mbnl1ΔE3/ΔE3 and HSALR muscles. A, B Western blot analysis of HDAC4 and its phosphorylated form (Ser632) in total protein lysate of Mbnl1ΔE3/ΔE3 and HSALR muscles. Quantification of total levels is given in B. Quantification of phosphorylated levels is given in Supplementary Material, Fig. S8B. Total levels are normalized to α-actinin. n = 3 per group. C, D Western blot analysis of HDAC4 in nuclear (C, D) and cytosolic (E, F) fractions of gastrocnemius muscle from Mbnl1ΔE3/ΔE3 mice and HSALR mice. Quantifications of HDAC4 levels are given in D and F. Levels are normalized to lamin-B (D) and tubulin (F). n = 5/4 Ctrl/Mbnl1ΔE3/ΔE3 and 3/4 Ctrl/HSALR. G mRNA levels of Dach2, Actc1, Ramp1, Dhrs7c, Cdh1 and Mitr in TA muscle from Mbnl1ΔE3/ΔE3 mice and in gastrocnemius muscle from HSALR mice. Transcript levels are normalized to Tbp. n = 4/3 Ctrl/Mbnl1ΔE3/ΔE3 (except for Dhrs7c and Cdh1, n = 6/5) and 4/4 (Dach2/Mitr), 6/6 (Actc1/Cdh1), 6/5 (Ramp1/Dhrs7c) Ctrl/HSALR. All protein (B, D, F) and RNA (G) levels are relative to control and expressed as log2(Fold Change). All data are mean ± SEM; * p < 0.05; ** p < 0.01; ****p < 0.0001; two-tailed unpaired Student’s t-test

Mbnl1 ΔE3/ΔE3 mice show resistance to denervation-induced muscle atrophy

Perturbations observed in innervated muscle from DM1 mouse models are reminiscent of the changes induced after denervation (i.e., synaptic gene up-regulation, HDAC4 accumulation, fibre type switch). CaMKII deregulation in mutant mice may interfere with the effect of neural activity in skeletal muscle. In turn, it may impair the muscle response to neural inactivity and reduce adaptive changes in activity-dependent processes. This reduced muscle plasticity may contribute to progressive NMJ deterioration observed in DM1 mouse models. To test this hypothesis, we challenged activity-dependent signalling pathways in DM1 mouse models with nerve injury. To this end, we cut the sciatic nerve of 3-month-old Mbnl1ΔE3/ΔE3 and HSALR mice to obtain complete denervation of hind limb muscles. Unexpectedly, we observed that HSALR mice lose the expression of the HSA transgene after 3 days of denervation (Fig. S9A). Consequently, ribonuclear foci accumulation and Clcn1 mis-splicing were reduced in denervated HSALR muscle, as compared to innervated muscle (Fig. S9B, C). Therefore, we limited the analysis to the Mbnl1ΔE3/ΔE3 mouse line.

After denervation, the loss of muscle mass was significantly less in Mbnl1ΔE3/ΔE3 mice, as compared to control mice (Fig. 7A). Nerve injury did not aggravate muscle degeneration in Mbnl1ΔE3/ΔE3 mice (Fig. S10A). To get insights into the mechanisms of this atrophy resistance, we first evaluated changes in pathways known to contribute to muscle atrophy after denervation. Transcript levels of the atrogenes Fbxo32 and Trim63, which are induced by HDAC4 and FoxO pathways after nerve injury [42, 43], were similar in Mbnl1ΔE3/ΔE3 and control muscles 3 days after denervation (Fig. S10B). We and others have reported that the anabolic pathway Akt/mTORC1 (mammalian Target Of Rapamycin Complex 1) is deregulated in DM1 muscle [25], and that its activation contributes to muscle atrophy upon denervation [23, 43]. Thus, we assessed whether Akt/mTORC1 activity is perturbed in DM1 denervated muscle. At 3 days of denervation, there was no change in the levels of the phosphorylated, active form of Akt (AktP473) in Mbnl1ΔE3/ΔE3 and control muscles, and levels of the active form of S6 (S6P235) increased similarly in both denervated muscles (Fig. S10C). Moreover, levels of the autophagic marker LC3II remained largely unchanged in Mbnl1ΔE3/ΔE3 and control muscles, suggesting that autophagy is not strongly affected (Fig. S10C). These results suggest that atrophy resistance does not arise from perturbations in atrogene expression or autophagy after denervation in DM1 muscle. We next evaluated muscle fibre type and size after 3 weeks of denervation. Control TA muscle shifted to type IIA fibres upon denervation, approaching the fibre type composition observed in Mbnl1ΔE3/ΔE3 innervated muscle (Fig. 7B). Fibre type proportion remained unchanged in Mbnl1ΔE3/ΔE3 denervated muscle (Fig. 7B). The minimum ferret of type IIA fibres was increased in Mbnl1ΔE3/ΔE3 innervated muscle as compared to control innervated muscle, but it decreased upon denervation in mutant mice (Fig. 7C, D). In parallel, the size of type IIX and IIB fibres was similar in Mbnl1ΔE3/ΔE3 and control innervated muscles, and strongly decreased upon denervation in both mutant and control mice (Fig. 7C, D). Hence, the predominance of type IIA fibres in mutant mice and their relative resistance to denervation-induced atrophy as compared to type IIX/B fibres (Fig. 7D) may explain why Mbnl1ΔE3/ΔE3 mice show limited muscle mass loss upon nerve injury.

Fig. 7
figure 7

Mbnl1ΔE3/ΔE3 muscle shows resistance to atrophy upon denervation. A Mass variation after 3 weeks of denervation in control and Mbnl1ΔE3/ΔE3 mice, for TA and gastrocnemius (GA) muscles. n = 4 per group. B Fluorescent images of denervated muscles from control and Mbnl1ΔE3/ΔE3 mice, stained with antibodies against type IIA myosin heavy chain (MHC2A - red) and laminin (green) and quantification of fibre type proportion in control and mutant denervated muscles. Scale bar, 500 μm. n = 4 mice per group. C Quantification of fibre minimum ferret in innervated and 3-week-denervated muscles from control and Mbnl1ΔE3/ΔE3 mice. n = 4 mice per group. D Fibre size distribution for type IIA, IIX and IIB fibres in innervated and 3-week-denervated muscles from Mbnl1ΔE3/ΔE3 and control mice. n = 4/4 Ctrl/Mbnl1ΔE3/ΔE3. All data are mean ± SEM; $p < 0.05, $$p < 0.01, $$$p < 0.001, $$$$p < 0.0001 between Inn/Den; * p < 0.05, ** p < 0.01, **** p < 0.0001 between genotypes; two-way ANOVA with a Tukey’s post-hoc analysis (Inn/Den and Ctrl/ Mbnl1ΔE3/ΔE3; C) or two-tailed unpaired Student’s t-test (A, B)

Endplate remodelling is impaired in Mbnl1 ΔE3/ΔE3 muscle upon denervation

Synaptic remodelling after denervation includes a strong increase in AChR turnover in the sub-synaptic region, together with synaptic gene up-regulation throughout the fibre [44]. The release of synaptic gene repression in non-synaptic muscle regions depends on HDAC4 induction, which may rely on CaMKII inhibition [8, 36]. To further characterize the muscle response to denervation in Mbnl1ΔE3/ΔE3 mice, we first evaluated the expression of synaptic genes 3 days after nerve injury. Importantly, the up-regulation of Myog, Musk, Chrna1 and Chrng was hampered in mutant TA muscle, as compared to controls (Fig. 8A). Inversely, the expression of Chrne remained abnormally high in Mbnl1ΔE3/ΔE3 muscle upon denervation (Fig. 8B). This defect occurred despite an efficient induction of HDAC4 in mutant mice. Indeed, there was a strong increase in the transcript (Fig. S11A) and protein (Fig. S11B) levels of HDAC4 in denervated muscle from both mutant and control mice. Consistently, HDAC4 accumulated in myonuclei of Mbnl1ΔE3/ΔE3 and control denervated muscles (Fig. S11C). Of note, changes in CaMKII expression pattern persisted upon denervation in Mbnl1ΔE3/ΔE3 muscle (Fig. S11B). Expression of the direct targets of HDAC4, Dach2 and Mitr, was similarly repressed in Mbnl1ΔE3/ΔE3 and control denervated muscles (Fig. S11D).

To assess the consequences of the defective up-regulation of synaptic genes in mutant muscle, we evaluated changes at the endplate after 2 weeks of denervation. At this stage, endplate fragmentation increased and remained higher in EDL and TA muscles from Mbnl1ΔE3/ΔE3 mice, compared to controls (Fig. 8C, D). We next quantified AChR turnover by labelling receptors 5 days after nerve injury and assessing their turnover 10 days later. In control muscle, AChR turnover increased drastically after denervation (Fig. 8E, F), as previously reported [45]. Interestingly, AChR turnover was reduced in TA and EDL denervated muscles from Mbnl1ΔE3/ΔE3 mice, as compared to control denervated muscle (Fig. 8E, F). Reduced AChR turnover in mutant muscle may arise from the defective expression of synaptic genes and reduced insertion of AChRs. Together, these results point to an impaired response to denervation and reduced plasticity to neural (in)activity of Mbnl1ΔE3/ΔE3 muscle.

Fig. 8
figure 8

Denervation-induced changes in the expression and dynamics of synaptic proteins are impaired in Mbnl1ΔE3/ΔE3 mice. A, B mRNA levels of Myog, Musk, Chrna1 and Chrng (A) and Chrne (B) in TA muscle from control and Mbnl1ΔE3/ΔE3 mice 3 days post-denervation. Levels are normalized to Tbp, relative to control innervated muscle and expressed as log2(Fold Change). n = 6/5 Ctrl/ Mbnl1ΔE3/ΔE3 (except for Myog, n = 4/3). C, D Fluorescent images of endplates stained with α-bungarotoxin in EDL muscle from control and Mbnl1ΔE3/ΔE3 mice 2 weeks post-denervation. Quantification of endplate fragmentation in EDL and TA muscles is given in D. n = 3/4 Ctrl/KO. Scale bar, 50 μm. E, F AChR turnover in denervated muscle from Mbnl1ΔE3/ΔE3 and control mice. Fluorescent images of “old” and “new” receptors are shown in (E). Scale bar, 50 μm. Quantification of AChR turnover is given in (F). n = 10/11 (EDL) and 5/5 (TA) Ctrl/Mbnl1ΔE3/ΔE3. Data are mean ± SEM; *p < 0.05; ** p < 0.01; *** p < 0.001; ****p < 0.0001 between genotypes; two-way ANOVA with a Tukey’s post-hoc analysis (Inn/Den and Ctrl/ Mbnl1ΔE3/ΔE3)

CaMKIIβ/βM overexpression normalizes endplate fragmentation and synaptic gene expression in innervated Mbnl1 ΔE3/ΔE3 muscle

To determine the contribution of CaMKII deregulation in endplate fragmentation and changes in synaptic gene expression observed in Mbnl1ΔE3/ΔE3 mice, we assessed the consequences of CaMKII modulation in mutant muscle. As DM1 mouse models shifted from CaMKIIβ/βM to CaMKIIβe expression, we injected isotype 9 adeno-associated virus (AAV9) in the anterior compartment of control and Mbnl1ΔE3/ΔE3 mice to overexpress either CaMKIIβ or CaMKIIβM. Three weeks later, we unilaterally denervated the mice and harvested innervated and denervated muscles 3 days later (Fig. 9A). A single AAV injection was sufficient to strongly increase the expression of CaMKIIβ/βM in TA innervated and denervated muscles, as compared to muscle injected with AAV-GFP (Fig. 9B). CaMKIIβ/βM overexpression did not modify TA muscle mass normalized to body weight, as compared to GFP-overexpressing muscle (Fig. S12A). Moreover, it did not alter muscle histology in control and mutant mice (Fig. S12B). In parallel, the specific tetanic force (Fig. 9C) and the late relaxation time (Fig. 9D) of EDL innervated control and mutant muscles were unchanged upon CaMKIIβ/βM overexpression, as compared to GFP-overexpressing muscle.

Importantly, overexpression of CaMKIIβ or CaMKIIβM was sufficient to normalize endplate fragmentation in Mbnl1ΔE3/ΔE3 EDL innervated muscle, as compared to control (Fig. 9E and Fig. S12C). CaMKIIβ/βM overexpression did not perturb synaptic gene expression in control innervated muscle (Fig. 9F-H). Conversely, CaMKIIβ overexpression normalized Myog expression in mutant innervated muscle (Fig. 9F) and tended to reduce Chrna1 and Chrng expression (Fig. 9G, H), as compared to GFP-overexpressing mutant muscle. CaMKIIβM overexpression showed the same trend but was less efficient (Fig. 9F-H). In control muscle, CaMKIIβ/βM overexpression was sufficient to reduce the up-regulation of Myog, Chrna1 and Chrng upon denervation (Fig. 9I-K). In contrast, it did not further reduce synaptic gene expression in Mbnl1ΔE3/ΔE3 denervated muscle (Fig. 9I-K). Together, these results indicate that CaMKIIβ/βM deficiency contributes to synaptic gene deregulation and endplate fragmentation in innervated Mbnl1ΔE3/ΔE3 muscles, but not to their defective response to denervation. To determine whether the effect of CaMKIIβ/βM overexpression involves changes in HDAC4 pathway, we evaluated the expression of HDAC4 target genes. CaMKIIβ/βM overexpression normalized the expression of Dach2 and Ramp1 in Mbnl1ΔE3/ΔE3 innervated muscle, as compared to control muscle (Fig. S13A). Moreover, it restricted the repression of Dach2 and Mitr after denervation in control mice (Fig. S13A). Strikingly, denervation-induced HDAC4 accumulation was abrogated by CaMKIIβ/βM overexpression in control and mutant mice (Fig. S13B), which was consistent with the defective repression of HDAC4 target genes. These changes suggest that HDAC4 inhibition mediates some of the transcriptional changes observed in control and mutant mice after CaMKIIβ/βM overexpression.

Fig. 9
figure 9

CaMKIIβ/βM overexpression normalizes endplate fragmentation in Mbnl1ΔE3/ΔE3 innervated muscle. A AAV-based overexpression strategy to evaluate the role of CaMKIIβ/βM deregulation in Mbnl1ΔE3/ΔE mice. Created with Biorender.com. B mRNA levels of Camk2b in control and Mbnl1ΔE3/ΔE3 TA muscles infected with AAV-GFP, -CaMKIIβ, or -CaMKIIβM. Levels are normalized to Tbp, relative to control innervated muscle and expressed as log2(Fold Change). n = 4 Ctrl and 3 Mbnl1ΔE3/ΔE3 (except in Den, n = 4 IIβM). C, D Specific tetanic force (sP0; C) and late relaxation time (D) upon stimulation of control and Mbnl1ΔE3/ΔE3 EDL muscles infected with AAV-GFP, -CaMKIIβ, or -CaMKIIβM. n = 4/5/6 Ctrl and 3 Mbnl1ΔE3/ΔE3 per group. E Number of fragments per endplate in control and Mbnl1ΔE3/ΔE3 EDL innervated muscles injected with AAV-GFP, -CaMKIIβ, or -CaMKIIβM. n = 5/5/7 Ctrl and 3 Mbnl1ΔE3/ΔE3 per group. F-K mRNA levels of Myog (F, I), Chrna1 (G, J) and Chrng (H, K) in control and Mbnl1ΔE3/ΔE3 TA innervated (F-H) and denervated (3 days; I-K) muscles injected with AAV-GFP, -CaMKIIβ, or -CaMKIIβM. Levels are normalized to Tbp, relative to control innervated muscle and expressed as log2(Fold Change). n = 4 Ctrl and 3 Mbnl1ΔE3/ΔE3 (except for Den IIβM, n = 4). Data are mean ± SEM; *p < 0.05; ** p < 0.01; *** p < 0.001; ****p < 0.0001; two-way ANOVA with Tukey’s post-hoc correction

Discussion

Although mis-splicing events are at the basis of DM1 pathogenesis, how the consecutive perturbations lead to muscle alterations is unclear. NMJ deteriorations have been described in muscle biopsies from DM1 patients and in muscle from DM1 mouse models [10,11,12,13,14, 46]. However, the mechanisms underlying these perturbations and whether these defects arise from DM1-related changes in the muscle or in the nerve remain unknown. Here, we showed that NMJs are affected in Mbnl1ΔE3/ΔE3 and HSALR mice, two well-characterized mouse models of DM1. We established that CaMKIIs and activity-dependent signalling pathways are disrupted in Mbnl1ΔE3/ΔE3 and HSALR muscles, which may contribute to endplate destabilization and to the abnormal response of mutant muscles to denervation.

Signs of NMJ deterioration, without denervation of muscle fibres, have been reported in muscle biopsies from DM1 patients, as well as in DMSXL and Mbnl1/2-deficient mice [10,11,12,13,14, 46]. As motor neurons are also affected in these mouse models, it is unclear whether the defects arise from pre- or post-synaptic perturbations. Here, we found endplate fragmentation in the muscles of Mbnl1ΔE3/ΔE3 and HSALR mice at different ages. As HSALR mice express the transgene carrying the CTG repeats only in muscle, perturbations in the post-synaptic compartment, i.e. the muscle, are likely responsible for NMJ deterioration in DM1. Importantly, endplate fragmentation was similar between TA, EDL and gastrocnemius muscles, which are affected differentially in HSALR mice. In particular, TA muscle showed only few signs of muscle degeneration, suggesting that endplate destabilization is a primary defect in DM1. NMJ deterioration may in fact contribute to and precede muscle atrophy, weakness and fatigue observed in DM1 muscle.

To determine the pathomechanisms that compromise NMJ integrity in DM1, we examined the potential role of CaMKIIs. Mis-splicing in Camk2b, 2d and 2 g has been reported in DM1 patients, as well as in mouse models [3,4,5]. In particular, exclusion of Camk2b exon 13 appeared as one of the most important splicing changes detected in DM1 tissues. Although the pathophysiological consequences have been investigated in the brain, the consequences of CaMKII deregulation in skeletal muscle have not yet been analysed. Here, we report that, together with exon 13 exclusion, the three exons specifically included in the muscle-specific isoform of CaMKIIβ are excluded in HSALR and Mbnl1ΔE3/ΔE3 muscles. This indicates that Camk2b splicing is MBNL1-dependent. Of note, in HSALR mice, Camk2b mis-splicing was detected in gastrocnemius, TA and EDL muscles. Together with exons 13 and 16, exons 18–20 are part of the highly variable region of the Camk2b gene, which allows the expression of tissue-specific variants. Especially, CaMKIIβM was shown to be the only isoform accumulating at the NMJ [6]. Consistent with the abnormal splicing of Camk2b in DM1 muscle, CaMKIIβM was not detected in Mbnl1ΔE3/ΔE3 and HSALR muscles. CaMKIIs have been shown to contribute to synaptic gene repression in non-synaptic regions of muscle fibres, by inhibiting myogenin activity and HDAC4 signalling pathway [7, 36]. We reveal that HDAC4 accumulates in myonuclei from Mbnl1ΔE3/ΔE3 and HSALR muscles, which was not caused by spontaneous denervation of mutant muscles. CaMKII deficiency and the consecutive increase in HDAC4 activity may hence contribute to the up-regulation of synaptic genes and to the fibre type switch observed in mutant muscles. As some HDAC4 targets remained unchanged in mutant muscles, changes in synaptic genes detected in DM1 muscle may occur predominantly at the endplate, as their regulation there is independent from HDAC4/myogenin in innervated muscle [35]. Alternatively, abnormal accumulation of other CaMKII isoforms, such as CaMKIIβe, may inhibit the repressor MITR/HDAC9 and thereby mediate some of the effects observed on activity-dependent genes. Importantly, we show that overexpression of CaMKIIβ, and to a lesser extent of CaMKIIβM, normalizes synaptic gene expression in innervated mutant muscle, consistent with a primary role of CaMKII deficiency in activity-dependent gene deregulation. Simultaneously, CaMKIIβ/βM overexpression reverses endplate fragmentation in mutant muscle. As synaptic gene up-regulation was detected in gastrocnemius muscle, but not in TA muscle from HSALR mice, it is unlikely that it underlies endplate fragmentation that was observed in all muscles. Despite the known role of CaMKIIs in regulating AChR recycling, we did not find perturbation in AChR dynamics at the endplate in mutant muscle. CaMKII deregulation may alternatively increase endplate fragmentation by perturbing AChR clustering at the endplate, as suggested by previous studies [6, 47]. Of note, the fibre type switch previously reported in HSALR muscle [25] was exacerbated in Mbnl1ΔE3/ΔE3 muscle. This suggested that perturbations both in the muscle and in non-muscle tissues (e.g., the motor neuron) contribute to these changes.

To obtain further insights into the capacity of DM1 muscle to regulate activity-dependent signalling and to maintain endplates, we challenged Mbnl1ΔE3/ΔE3 and HSALR mice with nerve injury. As transgene expression driven by the HSA promoter was lost in HSALR muscle upon denervation, we needed to limit the analysis to Mbnl1ΔE3/ΔE3 mice. In this model, denervation-induced increase in AChR turnover and synaptic gene up-regulation were hampered, suggesting that incorporation of new receptors at the endplate is limited in denervated mutant muscle. Synaptic gene induction upon denervation relies on the release of Myog expression, through the repression of Mitr/Hdac9 and Dach2 by HDAC4 [8, 48, 49]. Expression of these two repressors was efficiently reduced upon denervation in DM1 muscle, which was consistent with the major accumulation of HDAC4 in mutant denervated muscle. In addition, CaMKIIβ/βM overexpression did not restore synaptic gene expression in denervated mutant muscles, but rather decreased synaptic gene up-regulation in control denervated muscle. Hence, the incomplete induction of synaptic genes in Mbnl1ΔE3/ΔE3 denervated muscle is unlikely to occur via HDAC4 signalling deregulation or CaMKIIβ/βM deficiency. However, we cannot rule out contribution of other CaMKII isoforms to the defective muscle response to denervation of Mbnl1ΔE3/ΔE3 mice. Changes in the activity of other repressors of Myog, such as Msy3/Ybx3 [50] or CtBP1 [51], may also be involved in the defects observed. Finally, ClC-1 deregulation may hinder the response to denervation in DM1 muscle, as primary changes triggered by nerve injury at the endplate have been associated with chloride influx [52]. Consistently, another myotonic mouse model deficient for ClC-1 also displayed impaired muscle response to denervation [53].

Conclusions

In conclusion, our study identified NMJ deterioration as an integral part of muscle dysfunction in DM1, which likely involves muscle perturbations in activity-dependent pathways. Especially, deregulation of CaMKII isoforms may be a key event in endplate destabilization, as well as in DM1 pathogenesis in muscle and non-muscle tissues.

Data availability

Data generated during the study are freely accessible on the Yareta repository database: https://doi.org/10.26037/yareta:qx7k7d2vcva2fo7jr5t3bn2bly. RNAseq data are available under the GEO accession number: GSE226676.

References

  1. Lin X, Miller JW, Mankodi A, Kanadia RN, Yuan Y, Moxley RT, et al. Failure of MBNL1-dependent post-natal splicing transitions in myotonic dystrophy. Hum Mol Genet. 2006;15(13):2087–97.

    Article  CAS  PubMed  Google Scholar 

  2. Miller JW, Urbinati CR, Teng-Umnuay P, Stenberg MG, Byrne BJ, Thornton CA, et al. Recruitment of human muscleblind proteins to (CUG)(n) expansions associated with myotonic dystrophy. EMBO J. 2000;19(17):4439–48.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Nakamori M, Sobczak K, Puwanant A, Welle S, Eichinger K, Pandya S, et al. Splicing biomarkers of disease severity in myotonic dystrophy. Ann Neurol. 2013;74(6):862–72.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Suenaga K, Lee KY, Nakamori M, Tatsumi Y, Takahashi MP, Fujimura H, et al. Muscleblind-like 1 knockout mice reveal novel splicing defects in the myotonic dystrophy brain. PLoS ONE. 2012;7(3):e33218.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Sobczak K, Wheeler TM, Wang W, Thornton CA. RNA interference targeting CUG repeats in a mouse model of myotonic dystrophy. Mol Ther. 2013;21(2):380–7.

    Article  CAS  PubMed  Google Scholar 

  6. Martinez-Pena y Valenzuela I, Mouslim C, Akaaboune M. Calcium/calmodulin kinase II-dependent acetylcholine receptor cycling at the mammalian neuromuscular junction in vivo. J Neurosci. 2010;30(37):12455–65.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Tang H, Macpherson P, Argetsinger LS, Cieslak D, Suhr ST, Carter-Su C, et al. CaM kinase II-dependent phosphorylation of myogenin contributes to activity-dependent suppression of nAChR gene expression in developing rat myotubes. Cell Signal. 2004;16(5):551–63.

    Article  CAS  PubMed  Google Scholar 

  8. Tang H, Macpherson P, Marvin M, Meadows E, Klein WH, Yang XJ, et al. A histone deacetylase 4/myogenin positive feedback loop coordinates denervation-dependent gene induction and suppression. Mol Biol Cell. 2009;20(4):1120–31.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Macpherson P, Kostrominova T, Tang H, Goldman D. Protein kinase C and calcium/calmodulin-activated protein kinase II (CaMK II) suppress nicotinic acetylcholine receptor gene expression in mammalian muscle. A specific role for CaMK II in activity-dependent gene expression. J Biol Chem. 2002;277(18):15638–46.

    Article  CAS  PubMed  Google Scholar 

  10. Macdermot V. The histology of the neuromuscular junction in dystrophia myotonica. Brain. 1961;84:75–84.

    Article  CAS  PubMed  Google Scholar 

  11. Allen DE, Johnson AG, Woolf AL. The intramuscular nerve endings in dystrophia myotonica–a biopsy study by vital staining and electron microscopy. J Anat. 1969;105(Pt 1):1–26.

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Coers C, Telerman-Toppet N, Gerard JM. Terminal innervation ratio in neuromuscular disease. II. Disorders of lower motor neuron, peripheral nerve, and muscle. Arch Neurol. 1973;29(4):215–22.

    Article  CAS  PubMed  Google Scholar 

  13. Engel A, Jerusalem F, Tsujihata M, Gomez M. The neuromuscular junction in myopathies: a quantitative ultrastructural study. In: Recent Advances in Myology: Proceedings of the Third International Congress on Muscle Diseases, International Congress Series no 360 Edited by Bradley W, Gardner-Medwin D, Walton J. New York: Elsevier; 1975: 132–143.

  14. Panaite PA, Kuntzer T, Gourdon G, Lobrinus JA, Barakat-Walter I. Functional and histopathological identification of the respiratory failure in a DMSXL transgenic mouse model of myotonic dystrophy. Dis Model Mech. 2013;6(3):622–31.

    CAS  PubMed  Google Scholar 

  15. Spilker KA, Wang GJ, Tugizova MS, Shen K. Caenorhabditis elegans Muscleblind homolog mbl-1 functions in neurons to regulate synapse formation. Neural Dev. 2012;7:7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Drachman DB, Fambrough DM. Are muscle fibers denervated in myotonic dystrophy? Arch Neurol. 1976;33(7):485–8.

    Article  CAS  PubMed  Google Scholar 

  17. Walton JN, Irving D, Tomlinson BE. Spinal cord limb motor neurons in dystrophia myotonica. J Neurol Sci. 1977;34(2):199–211.

    Article  CAS  PubMed  Google Scholar 

  18. Tahraoui-Bories J, Merien A, Gonzalez-Barriga A, Laine J, Leteur C, Polveche H et al. MBNL-dependent impaired development within the neuromuscular system in myotonic dystrophy type 1. 2023; 49(1):e12876.

  19. Wheeler TM, Krym MC, Thornton CA. Ribonuclear foci at the neuromuscular junction in myotonic dystrophy type 1. Neuromuscul Disord. 2007;17(3):242–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Klinck R, Fourrier A, Thibault P, Toutant J, Durand M, Lapointe E, et al. RBFOX1 cooperates with MBNL1 to control splicing in muscle, including events altered in myotonic dystrophy type 1. PLoS ONE. 2014;9(9):e107324.

    Article  PubMed  PubMed Central  Google Scholar 

  21. Mankodi A, Logigian E, Callahan L, McClain C, White R, Henderson D, et al. Myotonic dystrophy in transgenic mice expressing an expanded CUG repeat. Science. 2000;289(5485):1769–73.

    Article  CAS  PubMed  Google Scholar 

  22. Kanadia RN, Johnstone KA, Mankodi A, Lungu C, Thornton CA, Esson D, et al. A muscleblind knockout model for myotonic dystrophy. Science. 2003;302(5652):1978–80.

    Article  CAS  PubMed  Google Scholar 

  23. Castets P, Rion N, Theodore M, Falcetta D, Lin S, Reischl M, et al. mTORC1 and PKB/Akt control the muscle response to denervation by regulating autophagy and HDAC4. Nat Commun. 2019;10(1):3187.

    Article  PubMed  PubMed Central  Google Scholar 

  24. Castets P, Lin S, Rion N, Di Fulvio S, Romanino K, Guridi M, et al. Sustained activation of mTORC1 in skeletal muscle inhibits constitutive and starvation-induced autophagy and causes a severe, late-onset myopathy. Cell Metab. 2013;17(5):731–44.

    Article  CAS  PubMed  Google Scholar 

  25. Brockhoff M, Rion N, Chojnowska K, Wiktorowicz T, Eickhorst C, Erne B, et al. Targeting deregulated AMPK/mTORC1 pathways improves muscle function in myotonic dystrophy type I. J Clin Invest. 2017;127(2):549–63.

    Article  PubMed  PubMed Central  Google Scholar 

  26. Dimauro I, Pearson T, Caporossi D, Jackson MJ. A simple protocol for the subcellular fractionation of skeletal muscle cells and tissue. BMC Res Notes. 2012;5:513.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Ham DJ, Borsch A, Lin S, Thurkauf M, Weihrauch M, Reinhard JR, et al. The neuromuscular junction is a focal point of mTORC1 signaling in Sarcopenia. Nat Commun. 2020;11(1):4510.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Moyer M, Berger DS, Ladd AN, Van Lunteren E. Differential susceptibility of muscles to myotonia and force impairment in a mouse model of myotonic dystrophy. Muscle Nerve. 2011;43(6):818–27.

    Article  PubMed  Google Scholar 

  29. Bayer KU, De Koninck P, Schulman H. Alternative splicing modulates the frequency-dependent response of CaMKII to ca(2+) oscillations. EMBO J. 2002;21(14):3590–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Brocke L, Srinivasan M, Schulman H. Developmental and regional expression of multifunctional Ca2+/calmodulin-dependent protein kinase isoforms in rat brain. 1995; 15(10):6797–808.

  31. Tombes RM, Faison MO, Turbeville JM. Organization and evolution of multifunctional ca(2+)/CaM-dependent protein kinase genes. Gene. 2003;322:17–31.

    Article  CAS  PubMed  Google Scholar 

  32. Wang ET, Treacy D, Eichinger K, Struck A, Estabrook J, Olafson H, et al. Transcriptome alterations in myotonic dystrophy skeletal muscle and heart. Hum Mol Genet. 2019;28(8):1312–21.

    Article  CAS  PubMed  Google Scholar 

  33. Strack S, Khan MM, Wild F, Rall A, Rudolf R. Turnover of acetylcholine receptors at the endplate revisited: novel insights into nerve-dependent behavior. J Muscle Res. 2015;36(6):517–24.

    CAS  Google Scholar 

  34. Chin ER. Role of Ca2+/calmodulin-dependent kinases in skeletal muscle plasticity. J Appl Physiol. 2005;99(2):414–23.

    Article  CAS  PubMed  Google Scholar 

  35. Tintignac LA, Brenner HR, Ruegg MA. Mechanisms regulating neuromuscular Junction development and function and causes of muscle wasting. Physiol Rev. 2015;95(3):809–52.

    Article  CAS  PubMed  Google Scholar 

  36. Cohen TJ, Waddell DS, Barrientos T, Lu Z, Feng G, Cox GA, et al. The histone deacetylase HDAC4 connects neural activity to muscle transcriptional reprogramming. J Biol Chem. 2007;282(46):33752–9.

    Article  CAS  PubMed  Google Scholar 

  37. Blaauw B, Schiaffino S, Reggiani C. Mechanisms modulating skeletal muscle phenotype. Compr Physiol. 2013;3(4):1645–87.

    Article  PubMed  Google Scholar 

  38. Hu N, Kim E, Antoury L, Wheeler TM. Correction of Clcn1 alternative splicing reverses muscle fiber type transition in mice with myotonic dystrophy. 2023; 14(1):1956.

  39. Wang AH, Yang XJ. Histone deacetylase 4 possesses intrinsic nuclear import and export signals. Mol Cell Biol. 2001;21(17):5992–6005.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Tang H, Goldman D. Activity-dependent gene regulation in skeletal muscle is mediated by a histone deacetylase (HDAC)-Dach2-myogenin signal transduction cascade. Proc Natl Acad Sci U S A. 2006;103(45):16977–82.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Ma W, Cai Y, Shen Y, Chen X, Zhang L, Ji Y, et al. HDAC4 Knockdown alleviates Denervation-Induced muscle atrophy by inhibiting myogenin-dependent atrogene activation. Front Cell Neurosci. 2021;15:663384.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Moresi V, Williams AH, Meadows E, Flynn JM, Potthoff MJ, McAnally J, et al. Myogenin and class II HDACs control neurogenic muscle atrophy by inducing E3 ubiquitin ligases. Cell. 2010;143(1):35–45.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Tang H, Inoki K, Lee M, Wright E, Khuong A, Sugiarto S, et al. mTORC1 promotes denervation-induced muscle atrophy through a mechanism involving the activation of FoxO and E3 ubiquitin ligases. Sci Signal. 2014;7(314):ra18.

    Article  PubMed  Google Scholar 

  44. Akaaboune M, Culican SM, Turney SG, Lichtman JW. Rapid and reversible effects of activity on acetylcholine receptor density at the neuromuscular junction in vivo. Science. 1999;286(5439):503–7.

    Article  CAS  PubMed  Google Scholar 

  45. Strack S, Petersen Y, Wagner A, Roder IV, Albrizio M, Reischl M, et al. A novel labeling Approach identifies three Stability Levels of Acetylcholine Receptors in the mouse Neuromuscular Junction in vivo. PLoS ONE. 2011;6(6):e20524.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Lee KY, Li M, Manchanda M, Batra R, Charizanis K, Mohan A, et al. Compound loss of muscleblind-like function in myotonic dystrophy. EMBO Mol Med. 2013;5(12):1887–900.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Koh YH, Popova E, Thomas U, Griffith LC, Budnik V. Regulation of DLG localization at synapses by CaMKII-dependent phosphorylation. Cell. 1999;98(3):353–63.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Cohen TJ, Barrientos T, Hartman ZC, Garvey SM, Cox GA, Yao TP. The deacetylase HDAC4 controls myocyte enhancing factor-2-dependent structural gene expression in response to neural activity. FASEB J. 2009;23(1):99–106.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Mejat A, Ramond F, Bassel-Duby R, Khochbin S, Olson EN, Schaeffer L. Histone deacetylase 9 couples neuronal activity to muscle chromatin acetylation and gene expression. Nat Neurosci. 2005;8(3):313–21.

    Article  CAS  PubMed  Google Scholar 

  50. De Angelis L, Balasubramanian S, Berghella L. Akt-mediated phosphorylation controls the activity of the Y-box protein MSY3 in skeletal muscle. Skelet Muscle. 2015;5:18.

    Article  PubMed  PubMed Central  Google Scholar 

  51. Thomas JL, Moncollin V, Ravel-Chapuis A, Valente C, Corda D, Mejat A, et al. PAK1 and CtBP1 regulate the coupling of neuronal activity to muscle chromatin and Gene Expression. Mol Cell Biol. 2015;35(24):4110–20.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Vyskocil F. Early postdenervation depolarization is controlled by acetylcholine and glutamate via nitric oxide regulation of the chloride transporter. Neurochem Res. 2003;28(3–4):575–85.

    Article  CAS  PubMed  Google Scholar 

  53. Klocke R, Steinmeyer K, Jentsch TJ, Jockusch H. Role of innervation, excitability, and myogenic factors in the expression of the muscular chloride channel ClC-1. A study on normal and myotonic muscle. J Biol Chem. 1994;269(44):27635–9.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank Prof. C.A. Thornton and Prof. M.S. Swanson for the generous gift of HSALR and Mbnl1ΔE3/ΔE3mice, as well as Dr. D. Ham, M. Reischl, and N. Liaudet for their help with the macros for fibre typing, AChR turnover and 3D NMJ representation. MHC antibodies, developed by H.M. Blau and S. Schiaffino, were obtained from the Developmental Studies Hybridoma Bank (University of Iowa, Iowa City, Iowa, USA).

Funding

This work was supported by the Swiss Foundation for Research on Muscle Diseases and the Swiss National Science Foundation.

Open access funding provided by University of Geneva

Author information

Authors and Affiliations

Authors

Contributions

D.F., S.Q., I.C., F.C., M.T., A.S., S.L., performed the experiments and acquired the data. D.F. and P.C. analysed the data. R.I. and J.K. analysed RNAseq data. D.F., L.T., J.K., M.A.R., M.S., and P.C. interpreted the data. D.F. and P.C. wrote the manuscript. All authors were involved in revising the manuscript.

Corresponding author

Correspondence to Perrine Castets.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Electronic supplementary material

Below is the link to the electronic supplementary material.

Supplementary Material 1

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Falcetta, D., Quirim, S., Cocchiararo, I. et al. CaMKIIβ deregulation contributes to neuromuscular junction destabilization in Myotonic Dystrophy type I. Skeletal Muscle 14, 11 (2024). https://doi.org/10.1186/s13395-024-00345-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13395-024-00345-3

Keywords