Introduction

Ryanodine receptors (RyRs) encode intracellular Ca2+ release channels located on the endo/sarcoplasmic reticulum (ER/SR)1. In mammals, there are three isoforms. RyR1 and RyR2 are involved in skeletal and cardiac muscle excitation–contraction coupling, respectively2, and are also found in non-muscle tissues3, while RyR3, originally identified in the brain4, is expressed in many tissues5. RyRs are >2 million dalton homotetramers and are the largest known ion channels. RyR1 and RyR2 are stabilized by two forms of FKBP-encoded proteins: calstabin (calcium channel stabilizing binding protein, referring to its role in calcium homeostasis) 1 and calstabin2 (FKBP12 and FKBP12.6, respectively)6. The function of RyRs is further modulated by kinases7, phosphatases8, phosphodiesterases7,9 and calmodulin7. RyR oxidation, phosphorylation and nitrosylation can lead to dissociation of calstabin from the channel, leading to Ca2+ leaking from the ER/SR7. Sustained leak can excessively elevate cytosolic and mitochondrial Ca2+, inducing increased ROS production, mitochondrial dysfunction and cell damage10. This has been linked to pathological conditions such as heart failure

Fig. 4: Proteomic analysis showing the global map** of RyR1 Ca2+ leak on muscle adaptations to S-SIT.
figure 4

ac Proteomic analysis of protein groups related to Gene Ontology Biological Processes (GoBP), Molecular Function (GoMF) and Cellular Component (GoCC) that are significantly decreased S-SIT after 10 µM S107 treatment for 72 h (the treatment was applied immediately after stimulation). Protein groups exceeding 400 proteins were excluded. The median values of S-SIT − S-SIT S107 difference were calculated, and a score affected to the amplitude of the difference. The positive scores display the pathways significantly inhibited by S107 treatment. n = 5 per group. Benjamini–Hochberg corrected t test. Source data are provided as a Source Data file.

Inhibition of RyR1 Ca2+ leak may also decrease SERCA-mediated Ca2+ uptake and ATP utilization in the cytosol, and that could be a confounding factor for the observed mitochondrial changes in our S-SIT and S-SIT S107 myotubes. Sarcolipin (SLN) is a small molecule that is known to bind to SERCA inhibiting its activity. SERCA uses the energy derived from the hydrolysis of ATP to transport Ca2+ ions across the SR membrane61. SLN binding to SERCA promotes the uncoupling of the SERCA pump and slippage of Ca2+ into the cytoplasm instead of the SR lumen62. Expression of SERCA1 (the main isoform of SERCA expressed in C2C12 myotubes63) and its main regulator in the skeletal muscle, SLN64, was not different between SIT and S-SIT S107 myotubes at 72 h post stimulation (Supplementary Fig. 4b, c). SERCA/SLN co-immunoprecipitation assay showed no significant difference between S-SIT and S-SIT S107 myotubes compared to controls (Supplementary Fig. 4a, d). However, others have shown that SLN decreases SERCA Ca2+ uptake but does not alter ATP hydrolysis65, thus implicating SLN as an uncoupler of SERCA (it continues to hydrolyse ATP but less Ca2+ is transported to the SR lumen)66. We, therefore, prepared microsomes (SR-enriched fractions) (Supplementary Fig. 4e, f) to directly measure SERCA1 ATPase activity. Our results showed increased ATPase activity in S-SIT myotubes compared to control and S-SIT S107 myotubes at 72 h post stimulation (Supplementary Fig. 4g, h), suggesting increased ATP utilization at S-SIT myotube SR. Interestingly, ATP levels in whole-cell lysates showed a similar increase in S-SIT and S-SIT S107 myotubes compared to controls (Supplementary Fig. 4i). The high levels of ATP in S-SIT myotubes, despite the increased ATP utilization at the SR, suggest a higher metabolic state in S-SIT myotubes compared to control and S-SIT S107 myotubes.

S-SIT increases mitochondrial Ca2+ content

Ca2+ is a well-known second messenger involved in many physiological processes in numerous tissues including the skeletal muscle67,68. Ca2+ released through the SR can be taken up by mitochondria through the outer mitochondrial membrane via the voltage-dependent anion channel69,70 to cross the inner mitochondrial membrane via the MCU71. Such mitochondrial Ca2+ flux has been linked to improved mitochondrial bioenergetics72. We, therefore, investigated whether the Ca2+ leak through RyR1 in response to S-SIT induced mitochondrial Ca2+ uptake to trigger positive mitochondrial adaptations. We used the Rhod-2/AM probe and time-lapse confocal live imaging to investigate mitochondrial Ca2+ uptake14 in response to a single bout of S-SIT or S-MICT. Control myotubes loaded with Rhod-2/AM probe did not show any mitochondrial Ca2+ increase in response to light exposure (Fig. 5a), while mitochondrial Ca2+ significantly increased in both S-MICT and S-SIT myotubes (Fig. 5b, c, f). Mitochondrial Ca2+ during S-MICT returned rapidly towards basal levels, while it remained elevated after S-SIT (Fig. 5b, c, g), suggesting Ca2+ accumulation in the mitochondria after S-SIT. S-SIT myotubes pre-treated with 10 μM S107 did not show mitochondrial Ca2+ accumulation (Fig. 5d), indicating that Ca2+ leak through RyR1 is the main source of the observed mitochondrial Ca2+ uptake after S-SIT. A similar result was obtained when S-SIT myotubes were pre-treated with 20 μM of the MCU inhibitor mitoxantrone (MTX)73 (Fig. 5e, g). These results suggest that part of the Ca2+ leaking through RyR1 is directed to the mitochondria through MCU, which supports the decreased SR Ca2+ observed immediately after a SIT session (Fig. 2g–j).

Fig. 5: (S-)SIT-induced leaky RyR1 increases mitochondrial Ca2+ uptake and decreases PDH phosphorylation levels in muscle cells.
figure 5

ae Normalized Rhod-2 fluorescence imaging in C2C12 myotubes. A 300 s time-lapse confocal recording of a bout of slightly modified S-SIT and S-MICT protocols: b S-MICT and c S-SIT. d, e Mitochondrial Ca2+ uptake in S-SIT myotubes pre-treated with 10 µM S107 for 1 h (d) or 20 µM mitoxantrone (MTX) (e). Horizontal black lines indicate the periods of stimulation; vertical red arrows indicate the amplitude of mitochondrial Ca2+ at the end of the recordings. fg Maximal amplitude of normalized Rhod-2 fluorescence during stimulations (f) and 80 s after the end of S-MICT and S-SIT stimulations (g) in (ae); n = 5 (CTRL, S-SIT and S-SIT S107) and 3 (S-MICT, S-SIT MTX) independent biological experiments. One-way ANOVA followed by Tukey’s multiple comparisons test. hj Normalized Rhod-2 fluorescence imaging in mouse FDB intact single muscle fibres. Same protocols as described for (ac), except that the voltage was 40 V. kl Maximal amplitude of normalized Rhod-2 fluorescence in FDB muscle fibres in (hj) during (k) and 80 s after the end (l) of S-MICT and S-SIT stimulations; n = 3 mice per group. One-way ANOVA followed by Tukey’s multiple comparisons test. mn Representative immunoblots (m) and quantification (n) of phosphorylated PDH E1α at serine 293 related to total PDH E1α in myotubes 1 h after stimulation; 10 µM S107 or 20 µM MTX were applied after the stimulation for 1 h when indicated; n = 7 (CTRL, S-MICT, S-SIT and S-SIT S107) and 3 (S-SIT MTX) independent biological experiments. One-way ANOVA followed by Sidak’s multiple comparisons test. o, p Representative immunoblots (o) and quantification (p) of phosphorylated PDH E1α at serine 293 related to total PDH E1α in human muscles; n = 8 participants per group. Two-way ANOVA followed by Sidak’s multiple comparisons test. q Proposed schematic of RyR1 Ca2+ leak-activated mitochondrial PDH E1α dephosphorylation in response to S-SIT. Data are mean ± SD. *p ≤ 0.05, **p ≤ 0.01 and ***p ≤ 0.001. Source data are provided as a Source Data file.

To confirm S-SIT-induced mitochondrial Ca2+ accumulation in mature skeletal muscle, we performed mitochondrial Ca2+ uptake measurements in dissociated mouse FDB muscle fibres. We applied S-MICT and S-SIT to FDB muscle fibres and followed mitochondrial Ca2+ accumulation after the end of the stimulation as in the C2C12 myotubes. Control muscle fibres showed no increase in mitochondrial Ca2+ with light exposure (Fig. 5h). Like in C2C12 myotubes, S-MICT and S-SIT resulted in altered mitochondrial Ca2+ transients in FDB muscle fibres (Fig. 5i, j, k). Again, the amount of Ca2+ taken up by the mitochondria at the end of the S-SIT session was higher than at the end of the S-MICT session (Fig. 5l).

We then examined whether RyR1 Ca2+ leak elicits enough Ca2+ domains to trigger mitochondrial Ca2+ uptake through the MCU in our in vitro models. Mitochondrial Ca2+ uptake is known to occur in specific conditions, which require local microdomains of elevated Ca2+ between the SR and the mitochondria74. Despite the consensus that MCU needs a Ca2+ concentration in the micromolar range, there is evidence that mitochondria also can take up Ca2+ at nanomolar concentrations75,76. We hypothesized that such Ca2+ domains are formed in response to leaky RyR1 and can trigger mitochondrial Ca2+ uptake. We first checked the pattern of RyR1 opening-induced mitochondrial Ca2+ accumulation in our myotubes by treating them with 2.5 mM caffeine while monitoring mitochondrial Ca2+ with the Rhod-2/AM probe. Caffeine induced a rapid, large mitochondrial Ca2+ increase, which progressively decreased, upon which the addition of 10 μM of rapamycin (which dissociates calstabin1 from RyR114) induced a small additional mitochondrial Ca2+ uptake (Supplementary Fig. 4j). When the myotubes were first treated with 10 μM rapamycin, they also showed a rapid mitochondrial Ca2+ increase, but with a lower amplitude than that of caffeine treatment (Supplementary Fig. 4k). This mitochondrial Ca2+ uptake showed a progressive increase that reached a plateau, and the addition of caffeine elicited no further increase in mitochondrial Ca2+ (Supplementary Fig. 4k). These results suggest that rapamycin-induced leaky RyR1 is sufficient to elicit a progressive and sustained mitochondrial Ca2+ uptake. Overall, our observations in C2C12 myotubes and mouse FDB fibres indicate that, in response to S-SIT, a leaky RyR1 channel leads to Ca2+ uptake by the mitochondria. While electrical stimulation is known to elicit mitochondrial Ca2+ uptake72, we here show a specific mitochondrial Ca2+ accumulation following S-SIT, after the electrical stimulation ended, which suggests a role of RyR1 Ca2+ leak rather than Ca2+ release in response to electrical stimulation.

Mitochondrial Ca2+ uptake induced by RyR1 Ca2+ leak dephosphorylates PDH and underpins OXPHOS complex I increase in response to S-SIT

Several mitochondrial enzymes are Ca2+ sensitive. Among them, PDH is a gateway enzyme for carbohydrate-derived pyruvate entry into the TCA cycle for complete oxidation77. PDH catalyses pyruvate decarboxylation to acetyl-CoA, and the reaction leads to a reduction of NAD+ to NADH. PDH activity is covalently regulated by phosphorylation on four residues of the PDH E1 subunit: Ser293, Ser295, Ser300 and Ser232. PDH kinases inactivate PDH by phosphorylation, whereas PDH phosphatases activate PDH by dephosphorylation78. PDH activity is regulated in human muscle in response to exercise79. The following arguments prompted us to investigate PDH phosphorylation in response to SIT: (i) skeletal muscle PDH activity is dependent on exercise intensity80 and MCU−/− mice showed marked impairment in their ability to perform strenuous work81; (ii) MCU knockout induced an increase in PDH phosphorylation similarly to PDH phosphatases knockout linking mitochondrial Ca2+ uptake to PDH phosphorylation levels82.

We, therefore, measured PDH phosphorylation levels as a readout of the increased mitochondrial Ca2+ uptake in S-SIT myotubes. PDH phosphorylation level on Ser293 (P PDH E1α Ser293) was significantly decreased in S-SIT myotubes at 1 h post stimulation, but not in S-MICT myotubes (Fig. 5m, n), reflecting SIT-induced mitochondrial Ca2+ uptake and PDH activation. PDH dephosphorylation was also observed in our human muscle biopsies collected post SIT—but not MICT (Fig. 5o, p), indicating again that our S-SIT model mirrors the effects of a single SIT session in human muscle. To investigate the potential role of RyR1 Ca2+ leak in the process, we treated S-SIT myotubes with 10 μM S107 immediately after the stimulation and for 1 h before measurement of PDH phosphorylation levels. The decreased PDH phosphorylation was blunted (Fig. 5m, n), pointing to a causal role of RyR1 Ca2+ leak in PDH dephosphorylation in response to S-SIT. S-SIT myotube treatment with 20 μM MTX immediately after the stimulation and for 1 h also restored PDH phosphorylation levels, confirming the role of mitochondrial Ca2+ uptake in the process (Fig. 5m, n). It follows that SIT induces RyR1 PTMs and calstabin1 dissociation from the RyR1, which leads to a leaky RyR1, mitochondrial Ca2+ uptake and PDH dephosphorylation (Fig. 5q).

We finally investigated whether RyR1 Ca2+ leak-induced mitochondrial Ca2+ uptake plays a role in the late (72 h post) mitochondrial adaptations in response to S-SIT. For this purpose, we transfected S-SIT myotubes with small interfering RNAs (siRNAs) directed against MCU immediately after the stimulation and measured OXPHOS proteins 72 h later. We first confirmed that the si-MCU was effective to decrease MCU expression and mitochondrial Ca2+ uptake (Fig. 6a, b) in differentiated myotubes without altering the expression of the main proteins involved in SR Ca2+ transients (RyR1, SERCA, and calstabin1) (Fig. 6a). S-SIT myotubes transfected with the si-MCU immediately after the stimulation showed decreased MCU protein levels 72 h after the stimulation as compared to scrambled siRNA transfection (si-CTRL and S-SIT si-CTRL) (Fig. 6d). S-SIT si-CTRL myotubes showed significantly increased OXPHOS CI and IV proteins, whereas S-SIT si-MCU specifically prevented OXPHOS CI modifications (Fig. 6c, e–i). Since we downregulated MCU after S-SIT, our results suggest that mitochondrial Ca2+ uptake after completion of S-SIT (which we linked to the leaky RyR1) is responsible for the increase in mitochondrial OXPHOS CI expression. These results are in agreement with recent work showing an association between decreased MCU levels and a specifically decreased OXPHOS CI protein in cardiac tissues obtained from Barth syndrome patients83. Altogether, these observations point to a new role of MCU and mitochondrial Ca2+ in the regulation of OXPHOS CI that opens an area for further investigations.

Fig. 6: Mitochondrial Ca2+ uptake contributes to mitochondrial adaptations to S-SIT.
figure 6

a Immunoblots of MCU, RyR1, SERCA1, GAPDH and calstabin1 in si-MCU (siRNAs directed against MCU) compared to si-CTRL (negative control siRNAs) myotubes. b Original recordings of normalized Rhod-2 fluorescence imaging in si-MCU compared to si-CTRL myotubes at 72 h post transfection. c Representative immunoblots of mitochondrial OXPHOS and MCU proteins expression in S-SIT si-MCU myotubes at 72 h post stimulation (cells were transfected immediately after the stimulation with the siRNAs against MCU) compared to S-SIT si-CTRL myotubes (cells were transfected immediately after the stimulation with the negative control siRNAs) and si-CTRL (non-stimulated myotubes transfected with the negative control siRNAs). All the cropped parts of OXPHOS proteins are part of the same blot that is shown in Supplementary Fig. 6. di Quantifications of the immunoblots in (c) related to total protein and expressed as % of si-CTRL; n = 6 independent biological experiments per group. One-way ANOVA followed by Tukey’s multiple comparisons test (d, e, h). Data are mean ± SD. *p ≤ 0.05, **p ≤ 0.01 and ***p ≤ 0.001. Source data are provided as a Source Data file.