Introduction

Skeletal muscle forms through the fusion of mononucleated muscle progenitor cells during development1,2. Mature, multinucleated myofibers are composed of different contractile and metabolic machinery that result in independent muscle fiber types (IIb, IIx, IIa, and I)3. In addition to possessing fiber type diversity, skeletal muscle also contains a stem cell population (satellite cells) that can be activated upon a stimulus and ultimately fuse with myofibers or each other, thereby allowing regeneration4. The requirement of multinucleation for skeletal muscle function lacks experimental validation, but explanations include that each nucleus is capable of controlling a finite volume of cytoplasm and that regions of the myofiber perform specialized functions necessitating localized transcripts.

Compelling recent studies have enhanced our understanding of the transcriptional diversity of skeletal muscle. Single-cell RNA-sequencing and mass cytometry studies have outlined the major mononuclear cell types present in skeletal muscle, ranging from 10 to 15 cell types depending on cluster assignments and the granularity of subty**5,6,7. The major cell types always include the following broad categories: fibroadipogenic progenitors (FAPs), tenocytes, endothelial cells, smooth muscle cells, immune cells (B cells, T cells, macrophages, neutrophils), neural/glial cells, and satellite cells. Notably, mature myofibers are a small minority of captures in single-cell studies despite their dominance within the tissue, owing to the fact that multinucleated cells are not easily isolated in single-cell dissociation approaches5,6,7.

Myofibers are known to exhibit functional specialization at postsynaptic endplates (neuromuscular junction: NMJ), where a small cluster of myonuclei are responsible for formation and maintenance of the synaptic apparatus8,9,10,

Fig. 2: Temporal myonuclear heterogeneity revealed in develo** muscle.
figure 2

a Unbiased clustering of nuclei from postnatal (P) day 21 presented in a UMAP revealed all major populations in addition to unique myonuclei outside the canonical Type IIb and Type IIx myonuclei. These nuclei populations were marked by Nos1 and Enah, respectively. b Subclustering of myonuclear populations from P21 muscle revealed sarcomere assembly myonuclear states. c Feature plots assessing expression of mature muscle markers (Ttn, Myh1, Myh4, and Ckm) and differentiation markers (Myod1, Myog, Mymk) from the myonuclear populations in b. d Violin plots showing the sarcomere assembly myonuclei are enriched for a transcriptional profile associated with early myofibrillogenesis (expression of Nrap, Enah, Flnc, Myh9, Myh10, Fhod3).

One interpretation of P21 sarcomere assembly myonuclei is that they are the most recently fused nuclei and have not yet established their fiber type. In this scenario, these sarcomere assembly myonuclei could be in a more immature state and suggest that they progress through a postfusion maturation program to establish their adult identity. Another possibility is that these myonuclear states are entrained after the majority of myonuclear accretion has occurred (around P21). To distinguish these possibilities, we generated 8611 nuclear transcriptomes from the tibialis anterior at P10 when fusion is ongoing and recently fused nuclei would be present. Here we were unable to detect robust independent clustering of Enah+ myonuclear populations and instead detected myonuclear populations similar to adult muscle (Fig. 3a, Supplementary Fig. 12a). Interestingly, we did detect a cluster of cells at P10, which were close to satellite cells, but were not present at P21 or 5 months of age. We hypothesized that this population represented myocytes (activated satellite cells) since fusion is ongoing at P10. We subclustered the satellite cell and myocyte populations and detected the presence of differentiation and fusion genes including Myog and Mymk in the myocyte population, while Pax7 was enriched in the satellite cell population (Supplementary Fig. 12b). The presence of myocytes demonstrated ongoing fusion at P10 and the absence of this population at P21 confirmed that the majority of fusion was terminated in these samples. The lack of sarcomere assembly populations at P10 indicates that they do not represent a maturational phase that myonuclei transition through proximal to fusion, but instead more nuclei adopt this state in response to a physiological stimulus during the postfusion growth of skeletal muscle. We validated the transient expansion of Enah+ myonuclei at P21 by performing smRNA-FISH on tibialis anterior muscles from P10, P21, and 5-month-old mice (Fig. 3b). Flnc and Enah expression was present at all ages, but the percentage of Flnc+/Enah+ nuclei was significantly increased in P21 compared to P10 and 5 months (Fig. 3b). This result validated the P21 snRNA-seq data while also confirming our initial observation of low-level heterogeneity and Enah+ myonuclei in adult muscle (Supplementary Fig. 5c).

Fig. 3: Myonuclear transcriptional states exhibit temporal specificity in development.
figure 3

a UMAP representing snRNA-seq data from a postnatal (P) day 10 tibialis anterior shows the presence of an activated muscle progenitor population (myocytes) but the absence of defined sarcomere myonuclear states. b smRNA-FISH for Flnc and Enah shows an expansion of sarcomere assembly state myonuclei in P21 tibialis anterior compared to P10 and adult muscle. Flnc+ Enah+ nuclei were quantified at each timepoint (n = 3). Data are presented as mean ± standard deviation. A one-way ANOVA with Tukey post-hoc comparison was used to determine statistical significance, **P < 0.01. Scale bar: 20 μm. Source data are provided as a Source Data file.

In addition to functional diversity found in normal development, transcriptional heterogeneity can arise from pathological or compensatory processes associated with aging34,35,36. Skeletal muscle undergoes dramatic age-related changes with respect to functional deterioration and regenerative decline37,38,39,40, and so we next asked if the aging process impacted myonuclear transcription. We profiled 18,087 nuclei from 24- and 30-month-old tibialis anterior muscles (Fig. 4a, b, Supplementary Figs. 13 and 14). In 30-month muscle only, we observed emerging myonuclear populations that were not present in 5 or 24 months, including clusters expressing Nr4a3, Ampd3, and Enah, respectively (Fig. 4b). We integrated myonuclei from 5, 24, and 30 months and found a consistent aging-related gene-expression signature including upregulation of Nr4a3 and Smad3 at both aged timepoints (Fig. 4c, Supplementary Data 3), indicating that this may be a progressive feature of aging muscle. Smad3 has been implicated during the muscle aging process41,42, while Nr4a3 has a role for metabolic adaptations to exercise but has not been studied during aging43,44. smRNA-FISH for Nr4a3 in the gastrocnemius muscle confirmed elevated expression in myonuclei in aged muscle, following a heterogeneous pattern (Fig. 4d). The Ampd3+ population were identified as myonuclei based on the presence of the myogenic regulatory factor Myf6, the pro-atrophy gene Fbxo32, and neuromuscular markers Musk, Chrnb1, and Hdac4 (Supplementary Fig. 15). The cluster was also enriched for genes associated with the immune response, apoptosis, and proteasomal degradation (Tnfrsf23, Traf3, and Psma5) (Supplementary Fig. 15). Taken together, these data suggest that the Ampd3+ population could represent a denervated state that is dysfunctional (Supplementary Fig. 15). Surprisingly, the Enah+ myonuclear cluster was found to be highly similar to the Enah+ sarcomere assembly state from P21 muscle, with numerous marker genes in common including Atf3, Flnc, and Nrap (Fig. 4e). The presence of an Enah+ cluster at 30 months presents further evidence for the appearance of this sarcomere assembly population throughout the lifespan, appearing most prominently in development and aging.

Fig. 4: Uniform gene expression between myonuclei is disrupted in aged muscle.
figure 4

a UMAP of snRNA-seq data from the tibialis anterior of a 24-month-old mouse displaying similar clusters as at 5 months of age. b UMAP of snRNA-seq data from 30-month-old muscle displaying the presence of unique clusters of myonuclei marked by Ampd3, Enah, and Nr4a3. c Heatmap of integrated myonuclei from 5, 24, and 30-month tibialis anterior muscle showing a consistent transcriptional signature of aging muscle. Columns are individual nuclei belonging to each to timepoint. d Representative image of smRNA-FISH for Nr4a3 validated its upregulation in myonuclei in 30-month skeletal muscle (n = 3 for each group). Scale bar: 10 μm. e Feature plots of key marker genes of the Enah+ population within 30-month myonuclei.

In order to more systematically investigate the presence of sarcomere assembly myonuclei over time, as well as to determine patterns of nuclear composition across the lifespan, we integrated our snRNA-seq data from the tibialis anterior at P10, P21, 5 months, 24 months, and 30 months (Fig. 5a). Notably, integration confirmed the independent clustering of Enah+ and Ampd3+ populations (Fig. 5a). Enah+ myonuclei were present in all timepoints but increased in P21 muscle (Fig. 5b), consistent with our smRNA-FISH findings and analysis of individual datasets. Intriguingly, we also identified a population of nuclei (Ckm+ Col1a1+) expressing a number of skeletal muscle-specific genes (e.g., Tnni2, Tnnt3, Ckm) as well as marker genes involved in extracellular matrix remodeling (e.g., Col1a1, Col3a1, Col5a3, Col6a1, and Dcn) (Fig. 5a, Supplementary Data 1). This population was overrepresented in development, comprising 4.7% of nuclei at P10 compared to 0.9% of nuclei at 5 months (Fig. 5b). This cluster could represent an intermediate population that shares characteristics of myonuclei as well as cells of fibroblastic or tendon origin. We confirmed that our combined datasets are likely to represent a faithful trajectory of development and aging, as an independent approach for combined analysis without batch correction found overlap** cells in all clusters, indicative of temporally related cell states (Supplementary Fig. 16).

Fig. 5: Integration of snRNA-seq across the lifespan reveals myonuclear population dynamics.
figure 5

a Integrated UMAP of snRNA-seq data from postnatal day (P) 10, P21, 5-month, 24-month, and 30-month tibialis anterior muscle. b Proportional composition of nuclear types across the lifespan. Major categories (FAPs, endothelial cells, smooth muscle) include closely-related subpopulations. Myonuclei are combined (top graph) and subsetted (bottom graph) to display myonuclear composition dynamics.

In addition to revealing the emergence of shared transcriptional responses at distinct stages of the life cycle, our data also establish an atlas of myonuclear transcriptomics and reveal transcripts associated with rare myonuclear populations such as the NMJ. Previous studies have elegantly uncovered unique gene expression in NMJ-associated nuclei, which allowed for identification of fundamental molecular components of postsynaptic function45,46,47. However, our data reveal numerous previously unreported genes associated with the NMJ, likely because our approach was able to resolve global transcription at the nuclear level. This represents a unique opportunity to further reveal mechanisms of postsynaptic development and function. To test the validity of genes not previously associated with the NMJ, we confirmed localization of 4 of the genes not previously associated with the NMJ (Ufsp1, Lrfn5, Ano4, and Vav3) by smRNA-FISH and co-labeling of the NMJ marker acetylcholine receptor (AchR) (Fig. 6a). Moreover, we found that multiple top NMJ genes identified here showed significant changes in mRNA levels following hindlimb denervation, an expression pattern associated with postsynaptic regulation (Fig. 6b)48. While the expression of the majority of those genes increased after denervation, similar to the canonical NMJ factor Musk49, Ufsp1 and D430041D05Rik exhibited a decrease in expression, while Pdzrn4 did not change (Fig. 6b). To assess the functional relevance of these factors for NMJ formation and maintenance, we used an in vitro culture system in which C2C12 myoblasts, when plated on laminin, form aneural NMJ-like clusters that are marked by AChR (Fig. 6c)50,51,52. We treated C2C12 cells with a control siRNA or siRNAs targeting Musk, Ufsp1, Vav3, B4galnt3, or Gramd1b, then assayed for AChR clustering. Reduced expression of the target genes was observed after treatment with the appropriate siRNA (Fig. 6d). Musk reduction results in a blockade of AChR clustering (Fig. 6e), showing fidelity of the system50. Obvious AChR clustering abnormalities were not observed for cultures where Vav3 or B4galnt3 was reduced, but Ufsp1 and Gramd1b were identified as regulators of the NMJ (Fig. 6e). Loss of Gramd1b resulted in reduced AChR clusters, whereas reduction of Ufsp1 elicited an increase (Fig. 6f). These data indicate that Ufsp1 is a negative regulator of AChR clustering and NMJ formation, consistent with its downregulation following denervation. Reduction of Ufsp1 or Gramd1b did not impact overall myogenesis or transcript levels of canonical NMJ genes (Fig. 6g), suggesting they specifically regulate the NMJ in a transcriptionally independent manner. These data highlight the utility of nuclear-level resolution for the discovery of factors that regulate the biology of skeletal muscle.

Fig. 6: Discovery and functional characterization of previously unknown NMJ marker genes.
figure 6

a Representative images from smRNA-FISH for Ufsp1, Lfrn5, Vav3, and Ano4 on skeletal muscle sections showing co-localization with the canonical NMJ protein, acetylcholine receptor (AChR) (n = 3). AChR was visualized through α-bungarotoxin labeling. b Quantitative real-time PCR (qPCR) analysis for the indicated genes not previously associated with the NMJ from normal (n = 4) and denervated (n = 5) muscle. c Schematic for a siRNA screen in C2C12 myoblasts designed to test the function of candidate NMJ genes. siRNA was transfected two days after differentiation and three days later α-bungarotoxin was used to analyze AChR clustering as a surrogate for NMJ formation. d qPCR analysis for the genes targeted with siRNA. A scrambled siRNA was used as a control. e Representative images of C2C12 myotube cultures after treatment with various siRNAs and staining with α-bungarotoxin. Cells were also stained with phalloidin and DAPI. f Quantification of AChR clusters per field of view from e. g qPCR analysis for genes associated with myogenesis and NMJ formation. Scale bars: a 50 μm (top left panel), 10 μm (top right and bottom panels), e 50 μm. Data in d, f, and g are from three independent experiments. All data are represented as mean ± standard deviation. An unpaired two-sided t-test was used to determine statistical significance, **P < 0.01, ***P < 0.001, ****P < 0.0001. Source data are provided as a Source Data file.