Age-associated systemic inflammation and MuSCs epigenetic remodelling
To better understand the intricate relationship between systemic aging and MuSCs, we conducted a comprehensive analysis of the molecular and cellular characteristics of the murine aging circulatory system and compared them with both aged MuSC intrinsic changes and aged MuSC niche extrinsic changes (Figure 1A). First, we used whole blood counts followed by plasma collection to analyze the molecular and cellular blood profiles of young and aged mice. To ensure an unbiased approach, we first screened over 100 cytokines in young and aged mice using a proteome profiler array, followed by secondary validation of the identified candidates using quantitative multiplexing immunoassays. As expected, the plasma from aged mice displayed elevated levels of circulatory cytokines (TNFa, IL1a, IL1b IL-4, IL-6, CCL2, CCL7, CCL11, and CCL12) and myeloid cells, and a decline in lymphoid cells (Figure 1B, 1C, and Table S1). Together, these data support the widely recognized age-associated chronic low-grade inflammation, often referred to as "inflammaging," and myeloid bias resulting from the aging hematopoietic lineage. Secondly, when we interrogated the aged skeletal muscle transcriptome using our previously published dataset16, we found that the top three enriched activated pathway (Z-score > 2) were “Chemokine signaling”, “Cytokine-Cytokine Interaction” and “Complement & Coagulation Cascades” (Table S1). This led us to wonder whether increased circulatory cytokine levels could affect aged skeletal muscle. Using a suite of computational analyses, we integrated our proteomics data with transcriptomic results to generate a ligand-receptor predictive model and the subsequent signaling activity17. We identified five signaling predicted to be activated by our ligand-receptor model, including “Chemokine Signaling”, “IL-7 signaling” and “TLR-signaling” (Figure 1D and E and Table S1). Interaction analysis predicted CCR2 to have the highest activation score, significantly contributing to all but one of the signaling pathways (Figure 1E).
We previously established that CCR2 signaling activation is a hallmark of skeletal muscle aging1. The levels of circulatory CCR2-ligands have also been thoroughly characterized as a measure of biological age, frailty, and age-associated systemic inflammation in both humans and mice11,18-20. Building on this foundation, our first objective was to elucidate the connection between systemic inflammation and the intrinsic changes observed in skeletal muscles and MuSCs with advancing age. We first simulated a directed CCR2-mediated inflammatory response by systemically injecting recombinant CCL2, CCL7, and CCL8 into young animals as previously described1. Following injection, we recorded a transient inflammatory response, as evidenced by the increase in both the number of monocytes and the levels of pro-inflammatory cytokines in the blood of the treated mice (Figure S1E-J). We also assessed how this acute inflammatory response affects MuSCs, using CCR2-null mice as a negative control1. Although we observed no change in the raw number of MuSCs (Pax7+), stem cells from mice injected with chemokines displayed enhanced activation (Ki67+), cell cycling (EdU+), and accelerated myogenesis (MyoD/MyoG+) (Figure S1A-D). These have been previously described as features of the Galert state21,22. Interestingly, these enhanced characteristics remained long after chemokine levels returned to baseline (Figure S1K-M), whereby we observed accelerated regeneration (Figure S1N-P), in line with previous reports regarding Galert. MuSCs that have experienced prior injury have a long-term enhanced regenerative capacity23, yet the mechanisms underlying this phenomenon are not well understood. These long-term functional changes prompted us to believe that MuSCs can adapt to acute inflammatory signals, which are indicative of epigenetic reprogramming24,25.
We hypothesized that age-associated systemic inflammation might affect the epigenome and transcriptome of MuSCs. Following our results, we performed an in-depth examination of the aged MuSC transcriptome and found that one of the most significantly enriched pathways was “Chromatin organization” (Figure 1F), in with our initial hypothesis and previous reports7,26. Of particular interest was the lysine methyltransferase Kmt5a, because of its crucial role in maintaining H4K20 methylation. Kmt5a-mediated catalysis of H4K20 monomethylation is required for subsequent di- and trimethylation, which is necessary for establishing constitutive heterochromatin. The dynamic regulation of both Kmt5a and H4K20me1 is also indispensable for proper cell cycle progression, which is intriguing given that MuSCs predominantly exist in a state of quiescence and aged MuSCs often die upon activation. We confirmed our RNA-seq results to show that both the Kmt5a gene and protein decreased in aged MuSCs (Figure 1I and J). Loss of Kmt5a was functionally reflected by decreased methylation of its main substrate, H4K20me1 (Figure G-M). Single-cell analysis revealed a shift in H4K20me1 intensity in the aged MuSC population.
Next, we assessed whether CCR2 activity is linked to Kmt5a repression. In response to the systemic delivery of CCR2-ligands, Kmt5a, but no other epigenetic genes, was significantly repressed in MuSCs (Figure S1Q and R). It was also accompanied by long-term transcriptional changes in both myogenic and cell cycle genes (Figure S1S and S1T). Notably, we observed the upregulation of activation genes (Myf5 and Dek) and downregulation of quiescence genes (Hes1 and Hey1). We found that MuSCs displayed long-term erosion of H4K20me1 and a sustained increase in MyoD+ cells lasting for at least six-weeks post-injection (Figure S1U-X). Single-cell analysis also pointed that CCR2-ligands treatment promoting a shift in H4K20me1 intensity, with the population of MuSCs derived from treated animals displaying lower H4K20me1 levels (Figure S1W and S1X). These results suggest that CCR2-mediated inflammation may trigger long-term epigenetic remodelling in MuSCs, which is potentially mediated by Kmt5a and H4K20 methylation. Thus, we further investigated the role of Kmt5a in MuSCs to better understand its effect on muscle aging.
Kmt5a is required for quiescence maintenance and MuSC survival after activation.
To assess the role of Kmt5a in quiescent MuSCs, we generated inducible Kmt5a MuSC-specific knockout mice (Pax7CreERT2/+; Kmt5afl/fl [Kmt5aKO]) (Figure S2A). Immediately following the deletion of Kmt5a, we did not observe a change in MuSC number in vivo, despite the vast majority of Kmt5aKO MuSCs lacking Kmt5a (>95% efficiency; orange arrows) (Figure S2B-D). Next, we cultured both wildtype (Pax7+/+; Kmt5afl/fl) and Kmt5aKO mice to assess myogenic potential, cell proliferation, and survival. Freshly isolated MuSCs (Pax7+) were stained with myogenic markers to assess myogenesis progression toward commitment (MyoD) and terminal differentiation (MyoG) over time1. Ki67 and EdU were used to assess cell cycle re-entry (activation) and active cell proliferation, respectively. When we challenged the cells outside their niche, cultured Kmt5a-null cells displayed enhanced activation (Pax7+Ki67+), which directly correlated with the loss of H4K20me1 as early as 24hours after plating (Figure S2E-I). However, once MuSCs started actively dividing (EdU+) after 72h of culture, we observed a near-complete loss of H4K20me1, which is in line with the cell cycle regulation of H4K20me1. This decrease was accompanied by impaired myogenic terminal differentiation and cell expansion (Figure S2E-K). Further analyses revealed that Kmt5aKO MuSCs displayed aberrant morphological phenotypes, such as blebbing, pycnotic, and multiple nuclei, and a molecular signature suggestive of genomic instability (Figure S2L, M; red arrows). Molecular analysis of 72h cultured MuSCs supported this premise, as mutant cells displayed elevated y-H2AX and serine 15 phosphorylation of P53.
When challenged by acute sterile injury, the contribution of Kmt5aKO MuSCs to regeneration was disrupted, resulting in the apparent absence of muscle regeneration, as we did not observe centrally nucleated fibers (CNF) among the injured mutants (Figure 2A). We also noted a macroscopic decrease in injured muscle size 21-day post-injury (dpi) (Figure S3A, B). To better index regeneration, we used embryonic myosin heavy chain (eMHC), a marker of immature (regenerating) skeletal muscle fibers, throughout the regeneration process (Figure S4A). As expected, wild-type regenerating fibers were nearly all positive for eMHC at early regeneration time points (4- and 7dpi), followed by the generation of large centrally nucleated fibers (CNF), a hallmark of regeneration. In contrast, we did not observe any regenerating fibers in Kmt5aKO mice past 7dpi (Figure S4B). By 60dpi, most of the injured area appeared to be reduced to remnants of the ECM and mononucleated cells. Additionally, virtually no surviving MuSCs were observed after regeneration (Figure 2B and S4C). When we investigated the fate of these MuSCs and their derived progenitors, we found a sharp decline in the number of myoblasts (MyoD+) in the mutant at 4dpi (Figure S4D-G). The number of terminally committed progenitors (MyoG+) briefly increased at 4dpi before virtually disappearing at 7dpi (Figure S4E-F). Next, we aimed to further understand the fate of Kmt5aKO MuSCs in response to direct and indirect environmental pressures. Since MuSCs and derived progenitors were lost between 4- and 7dpi, we assessed proliferative and myogenic capacity at 4dpi in both injured and uninjured contralateral (CL) limbs, where MuSCs are expected to enter the Galert state (Figure S4A)21. While MuSC numbers in Kmt5aKO mice rapidly declined in both injured and CL muscles (Figure S4C, S4I, and S4K), MuSCs derived from the injured limb displayed primed features reminiscent of Galert, including swifter entry into the cell cycle (Figure S4J and S4L), accelerated myogenic progression (Figure S4E-G), and high p-S6 levels compared to wild-type MuSCs (Figure S4M-O)22.
Previous reports have shown that H4K20me1 levels are regulated in a cell cycle-dependent manner in some somatic cells27. Based on our observations, H4K20me1 levels in quiescent MuSCs were relatively stable and fell mostly after the first cell division, which occurs approximately 60h post-activation21 (Figure S2E and S2O, P). We hypothesized that long-term loss of Kmt5a would eventually disrupt H4K20me1 maintenance by preventing de novo deposition of H4K20me1 and potentially altering the capacity of MuSCs to remain quiescent. To test this theory, we deleted Kmt5a and waited for 1, 3, or 6 weeks before assessing MuSC number and function in vivo (Figure 2C-O). Over time, the MuSC number progressively declined in a stochastic manner (Figure 2D). While MuSCs were nearly undetectable after six weeks, the few remaining MuSCs still displayed detectable levels of H4K20me1 (Figure S2O, P). Taken together, these results support our theory that the loss of H4K20me1 over time following Ktmt5a deletion does not occur immediately and simultaneously in every cell. Notably, from three weeks and onward, Kmt5aKO MuSCs displayed features of quiescence exit, depicted by elevated levels of pS6 (Figure 2E) and a higher number of Ki67-positive cells (Figure 2F,G), but a low number of actively cycling cells (EdU+) (Figure 2F, H). Next, we investigated the fate of the Kmt5aKO MuSCs in vivo. A previous report has demonstrated that tempering with H4K20me2 leads to spontaneous and precocious differentiation of MuSCs during homeostasis9. However, we did not find MyoG+ cells in our mutants in homeostasis (Figure 1I). Since we observed a decline in survival and genomic instability in cultured Kmt5aKO MuSCs, which correlated with the loss of H4K20me1 (Figure S2E-N), we tested the possibility that Kmt5aKO MuSCs exited quiescence and entered cell death. We found a significant portion of mutant cells showing signs of cell death (~28% TUNEL+) (Figure 1J), together with an increased DNA damage response depicted by increased yH2AX and p53 signaling (Figure 1J, K).
Taken together, these results suggest that H4K20me1 regulation is an important factor in fine-tuning MuSC quiescence and, consequently, ensuring MuSC survival in response to extrinsic cues.
Kmt5a maintains MuSC quiescence by epigenetically regulating Notch signaling through promoter-proximal pausing.
To identify the mechanisms by which Kmt5a affects MuSC quiescence maintenance, we conducted a suite of transcriptomic assays. Because the Kmt5aKO MuSC number declined stochastically over time (Figure 2D), we used single-cell RNA sequencing to assess the dynamic transcriptional changes in Kmt5a-null MuSC fate over time. Unbiased clustering and UMAP reduction showed that wild-type cells were significantly different from the Kmt5aKO MuSCs. As expected, the Kmt5aKO MuSC population 2-weeks post deletion displayed a different transcriptomic profile than the 6-weeks post deletion MuSCs, which surprisingly resembled 2-years knockout MuSCs (Figure 3A). Pseudo-time analysis show that a small subset of MuSCs exist only 2-weeks after Kmt5a deletion and present upregulation of myogenic differentiation genes Myog and Mymk, along with the cell cycle arrest gene Cdkn1c (Figure 3B, and Figure S5C-E). In every mutant sample, we observed a drift in the Kmt5aKO MuSC population, with large changes in quiescence regulatory genes, such as Notch Signaling28, Mitophagy22, and ECM-receptor interactions29 (Figure S5A). Within this population, pseudo-time analysis showed that several key Notch-related genes and downstream targets (Notch, Jag1, Numb, and Rbpj) were downregulated over time, along with other myogenic markers (Pax7, Sdc4, and Spry1), consistent with the loss of quiescence (Figure S5C and E). Next, we combined Bulk RNA-seq with PRO-seq to compare wild-type and Kmt5aKO MuSCs. RNA-seq was used as a measure of total mRNA, whereas PRO-seq was used to determine dynamic transcriptional changes at base-pair resolution30. Most of the significantly altered genes within the Notch signaling pathway were regulated through promoter-proximal pausing (Figure 3C). The pausing index significantly increased for several key Notch effectors (e.g., Rbpj and Dll1) and downstream targets (e.g., HeyL, Hes6, Dtx4, and Snw1), which correlated with significant gene repression. We also found the opposite to be true, as a decrease in the pausing index at Dll4 and Jag2 promoters, both Notch1 ligands that are usually repressed, correlated with increased expression31. Overall, Kmt5a deletion accounted for >80% of the altered Notch pathway gene expression, highlighting Kmt5a as a potential master transcriptional regulator of Notch signaling in quiescent MuSCs. This translated into lower levels of Rbp-jκ protein, a critical regulator of MuSC quiescence32 (Figure 3D). Genetic deletion of Kmt5a in MuSCs did not immediately result in the repression of Notch genes (Figure S6A) despite the loss of detectable Kmt5a binding at the Rbpj TSS (Figure S6B). Instead, Notch target genes repression was observed at the earliest timepoint in our scRNA analysis and onward (Figure S5D and S5E). This was in line with the progressive loss of MuSC quiescence observed in the mutants during homeostasis (Figure 2D). Therefore, we wondered whether Kmt5a-mediated regulation of Notch genes was catalytically dependent. Since H4K20me1 was previously shown to promote transcription through RNA Polymerase II release during promoter-proximal pausing, we tested whether loss of H4K20me1 might be a necessary step for Notch genes repression33. We used a well-characterized selective Kmt5a inhibitor34 that resulted in rapid H4K20me1 decline without affecting Kmt5a levels (Figure 3E, F). Upon catalytic inhibition of Kmt5a in MuSCs, the transcript levels of Hes1, Hey1, Notch1, and Rbpj were significantly decreased without affecting myogenesis genes (Figure 3G). We concluded that Kmt5a is an epigenetic regulator of Notch genes transcription.
Based on these results, we hypothesized that restoring Notch target gene expression could rescue Kmt5a-mediated loss of quiescence. To test this hypothesis, we crossed MuSC-specific Kmt5aKO mice with the ROSANICD mouse line35. Following tamoxifen-mediated induction, MuSCs are both null for Kmt5a and overexpress the Notch intracellular domain (NICD), a DNA-binding domain that promotes the expression of Notch target genes. NICD overexpression rescued the expression of key Notch target genes such as Rbpj and Hes1 without altering Kmt5a expression (Figure S6C). Restoring Notch signaling in Kmt5aKO MuSCs prevented their loss and decreased the number of Ki67+ cells, suggesting that it preserved quiescence (Figure 3H-J). However, skeletal muscle regeneration was still severely impaired compared to that in wild-type mice, analogous to that observed in Kmt5aKO mice, and we did not detect surviving MuSC at 21dpi (Figure S6D, E). To further assess the fate of MuSCs derived from double-mutant mice, we cultured freshly isolated cells and assessed their survival. Similar to the single mutant Kmt5aKO cells, the double mutant cells displayed a decline in cell survival after 72 h of culture (Figure S6F). These results indicate that, while Kmt5a epigenetically controls MuSC quiescence maintenance through Notch signaling, subsequent cell survival after quiescence exit involves a distinct mechanism.
Kmt5a safeguards MuSCs from ferroptosis.
We took advantage of our transcriptomic data to further assess the cell fate and determine the fate of the subpopulation of Kmt5aKO MuSCs that exit quiescence. Curated enrichment analysis for significantly modified genes highlighted ferroptosis in both the RNA- and PRO-seq datasets, whereas the pausing index was enriched for Notch signaling and several cancer-related pathways, consistent with the observations in Figure 3 (Figure S7A-E). Ferroptosis is a unique form of programmed cell death regulated by iron-mediated increases in reactive oxygen species that lead to lipid peroxidation, ultimately resulting in membrane rupture, cell death, and release of pro-inflammatory factors36. Mechanistically, this process is not completely understood and is the focus of extensive investigations for its potential contribution to cancer, aging, and frailty37,38. Owing to recent advances in this field, ferroptosis is associated with several molecular, morphological, and biochemical hallmarks of ferroptosis39. The transcriptional signature in Kmt5a-null MuSCs suggested a pro-ferroptotic fate with a pathway activation z-score of 4.15 and an enrichment score of 44.06 (Adj. p value = 0.02650) (Figure S7A). We did not find a strong correlation between ferroptosis markers and the pausing index (Figure S7F), suggesting that ferroptosis might not be regulated through promoter-proximal pausing. Interestingly, most of the genes silenced in Kmt5aKO MuSCs were anti-ferroptotic and either involved in iron processing and export (Ftl1 and Pcbp2) or inhibition of lipid peroxidation and cellular damage (Gclc, Gss, and Gpx4). In contrast, the upregulated genes were either markers of ferroptosis (Ptgs2, Cybb, and Hmox1) or genes that promoted intracellular iron import (Tfrc) (Figure S7F).
Based on these results, we hypothesized that Kmt5a-deficient MuSCs would exhibit impaired iron metabolism, aberrant accumulation of intracellular iron, and increased levels of lipid peroxidation, resulting in cell death by ferroptosis (Figure S7G). To test this hypothesis more directly, we first characterized MuSC sensitivity to ferroptosis using gold standard compounds to induce ferroptosis (erastin and RSL3), along with Ferrostatin-1 (Fer1), which traps lipid radicals to rescue the effects of erastin and RSL3 (Figure S8)36. Both erastin and RSL3 induced a canonical ferroptotic response, with decreased cell viability, increased ROS production, and enhanced lipid peroxidation, which were rescued by Fer1 treatment (Figure S8). MuSCs were more sensitive to RSL3 exposure, perhaps because they directly target GPX4, which plays a critical role in preventing lipid peroxidation, and thus provides a major safeguard against ferroptosis. Interestingly, Gpx4 was among the most repressed genes identified in our scRNA-seq analysis (Figure S5B), particularly in the population of Kmt5a-deficient MuSCs that exhibited a cell cycle re-entry molecular signature. In vivo, we found that GPX4 was expressed in tibialis anterior muscle fibers with a small cross-sectional area, implying a potential role for GPX4 in type 2A fibers40 (Figure 4A). Interestingly, Kmt5aKO MuSCs were virtually devoid of GPX4 and exhibited iron-rich pockets in the vicinity of the MuSC niche (Figure 4B and 4C). Less than 2% of MuSCs from wild-type mice exhibited detectable levels of intracellular iron (Fe2+), whereas over 80% of Kmt5aKO MuSCs exhibited accumulation of iron foci (Figure 4C). Electron microscopy further suggested that Kmt5aKO MuSCs displayed hallmarks of ferroptosis, including cell swelling, plasma membrane blebbing and rupture (blue arrows), increased mitochondrial content, and intracellular iron accumulation (black arrows) (Figure 4D). ICP-MS was used to measure the total iron content in freshly sorted MuSCs. Kmt5aKO MuSCs had ~3.4x10-2ng of iron per cell, which was nearly 70-fold higher than that measured in wild-type MuSCs (Figure 4E). Consistent with these findings, the Kmt5aKO MuSCs exhibited higher levels of lipid peroxidation (Figure 4F), which progressively increased over time (Figure 4G). Likewise, genetic Kmt5a deletion resulted in the progressive rise of a pro-ferroptotic transcriptomic signature in Kmt5aKO MuSCs, including repression of Gpx4 and Rgs4 and higher expression of Ptgs2 41,42 (Figure 4H).
Next, we wondered if Kmt5a catalytic inhibition could promote ferroptosis. Kmt5a inhibition was accompanied by signs of ferroptosis at both the cellular and molecular levels. Dosage under the inhibitor IC50 ([C] = 4uM < IC50 = 7.3uM), while still significantly decreasing H4K20me1, was seemingly lethal in under 24h (Figure 4I, 4J, S8J and S8K). Loss of H4K20me1 following treatment was accompanied by decreased GPX4 protein and mRNA levels, as well as repression of Rgs4 and increased Ptgs2 and Hmox1 levels, mirroring the effects observed in RSL3 treated cells (Figure 4I-K and S8H-L). Kmt5a inhibition also significantly increased lipid peroxidation, which was reduced when co-treated with Fer-1 (Figure 4L and 4M), further supporting that cell death promoted by Kmt5a inhibition is ferroptosis.
Taken together, these results suggest that Kmt5a activity prevents premature ferroptotic MuSC death by epigenetically regulating key genes involved in iron metabolism and antioxidative activity upon cell cycle re-entry.
Epigenetic erosion of H4K20 drives MuSC population drift toward ferroptosis during aging.
Although not previously molecularly linked, increased pro-inflammatory features, loss of MuSC quiescence, and premature death are the hallmarks of skeletal muscle aging. While ferroptosis is an incompletely understood form of programmed cell death, especially in stem cell homeostasis, recent evidence indicates that ferroptosis is implicated in the pathogenesis of various age-dependent disorders38. Ferroptosis shares most, if not all, of the hallmarks of aging. Here, we aimed to confirm whether the key findings observed in Kmt5aKO MuSCs are physiologically relevant to aging. Specifically, we assessed the contribution of Kmt5a and H4K20me1 to the age-dependent decline in MuSC number and function. Since we found that aged MuSCs displayed lower levels of both H4K20me1 and Kmt5a than MuSCs from young mice (Figure 1G-M), we used flow cytometry to assess the activation status of aged MuSCs upon loss of H4K20me1 at the single-cell level. Flow cytometry analyses revealed that while virtually all quiescent MuSCs from young mice exhibited high levels of H4K20me1 (Ki67- H4K20me1high), an age-dependent reduction in this population of MuSCs suggests H4K20me1 erosion (Figure 5a), consistent with the information theory of aging4. Furthermore, more than 95% of MuSCs positive for the cell cycle entry marker Ki67 showed lower levels of H4K20me1, consistent with decreased H4K20 methylation being necessary for quiescence exit, and thus a hallmark of MuSC early activation (Pax7+ Ki67+ H4K20me1low) (Figure S9A).
To further understand how Kmt5a epigenetically controls MuSC fate, we mapped H4K20me1 in MuSCs from young and aged mice using CUT&Tag (Figure S9B). Consistent with previous studies using somatic cells, we found that H4K20me1 is a broad epigenetic mark with narrow peaks mostly localized near the TSS of genes and broader peaks spanning over the body of expressed genes (Figure 5A, B, and Figure S9C). To gain further insights into H4K20me1 regulation, we integrated H4K20me1 maps with our RNA-seq data and found that H4K20me1 was associated with transcriptional status regardless of age (Figure S9D). Term analysis revealed that genes with a loss of H4K20me1 at their TSS were associated with the cell cycle and chromatin organization, while genes that gained H4K20me1 signal were notably enriched for the activation of cell death (Figure S9E). Further, GSEA identified key processes that were also identified in Kmt5aKO MuSCs, including Notch signaling, P53 signaling, and myogenesis (FDR<0.1) (Figure S9F-H). Most notably, we confirmed that Kmt5a acts as a master epigenetic regulator of Notch Signaling, as over 78% of Notch genes were repressed in aged MuSCs in an H4K20me1-dependent manner with a Pearson correlation p-value of 0.0023 (Figure 5C; bold). Although Notch relies on signaling-based gene regulation, ferroptosis is primarily a metabolic process. Therefore, the role of H4K20me1 in the transcriptional regulation of ferroptotic genes was unclear, except for a few key genes, notably Gpx4, which exhibited the most significant reduction in transcript levels (Figure 5D, bold). Instead, these data highlight that aged MuSCs display signs of a pro-ferroptotic transcriptional signature with repressed Glutathione Metabolism genes such as Gclm and Gclc, enhanced iron import, and handling with elevated iron-responsible element genes such as Tfrc, Fth1, and Ftl1, as well as canonical markers of ferroptosis, such as high Ncoa4, Hmox1, and Ptgs2.
To further characterize the possible presence of a pro-ferroptosis MuSC population during aging, we performed high-depth scRNA-seq on MuSCs from adult and aged mice (Figure 6A-C). Enrichment analysis for the most altered genes highlighted Ribosome as the top pathway, indicative of the high transcriptional and translational turnover associated with MuSC activation. We identified Ferroptosis and Glutathione Metabolism as among the pathways most enriched in MuSCs from aged mice (Figure 6D). Pseudotime analysis identified a novel subpopulation of aged MuSCs (Cluster 3) displaying hallmarks of ferroptosis such as low levels of Gpx4, Slc7a11, and Pcbp2, and high Ftl1 and Fth1 (Figure 6C-E). Both aging clusters (2 and 3) displayed similar aging signatures, such as decreased quiescence gene expression (Pax7, Spry1, and Rbpj). However, unlike the canonical aging cluster 243,44, the ferroptotic cluster (cluster 3) differentiated itself by displaying no signs of premature commitment (MyoD and MyoG) or premature senescence (Cdkn2a and Trp53). Interestingly, Kmt5a was also significantly downregulated along with Gpx4, mostly in cluster 3, suggesting that the loss of Kmt5a could directly contribute to enhanced ferrosensitivity (Figure 5E and Figure S9I, J).
To better understand the extent to which MuSC ferroptosis contributes to skeletal muscle aging, we performed flow cytometry on adult and aged MuSCs, and assessed their viability, cell fate, and different forms of programmed cell death12,43. As previously shown, only a small percentage of aged MuSCs entered senescence prematurely (as opposed to geriatric MuSCs)43 (Figure 5F; SPiDER+)11. The remaining and the majority of dying aged MuSCs were skewed toward ferroptosis (~42%; AnnexinV-; Lipid PeroxidationHigh), with fewer cells undergoing apoptosis (~27%; AnnexinV+), indicating that ferroptosis is the dominant mode of regulated cell death in aged MuSCs (Figure 6F). Upon sorting, ferroptotic aged MuSCs expressed very low levels of Gpx4 and Kmt5a, but high levels of Hmox1 and Ptgs2, consistent with a canonical pro-ferroptotic fate response and similar to cluster 3 (Figure 6E and G). Upon assessing ferroptosis in vivo, we found that <10% of aged MuSCs displayed faint levels of cytoplasmic GPX4 in concert with an abundance of intracellular iron (Figure 6H and Figure S9K), which was not observed in MuSCs from young mice. This suggests that while aged MuSCs are defective in processing iron and fail to resist ferroptosis, some aged MuSCs retain the capacity to express GPX4, thus suggesting a reversible process. Similar to Kmt5aKO MuSCs, a significant proportion of aged MuSCs displayed high intracellular iron levels (>31%), with a concentration far exceeding that measured in MuSCs from young mice (Figure 6H, I). Interestingly, labile iron accumulation has also been observed in aged hematopoietic stem cells45. While circulating iron is largely depleted with age (anemia), labile iron is often enriched in the aging tissues. Iron enrichment in aged skeletal muscles is believed to contribute to both functional and regeneration impairments as well as forms of muscle wasting, such as sarcopenia37. Consistent with reports showing that increased intramuscular iron is associated with increased lipid peroxidation46, we found that freshly isolated aged MuSCs displayed a 1.5-fold increase in lipid peroxidation during homeostasis (Figure 6J). To avoid stress response bias, we used MuSCs from RosaCreERT2;Nrf2f/f;Gclcf/f mice47,48 treated with tamoxifen (50 mg/kg i.p. for five days), which produces Nrf2-Gclc double-KO cells, as a control, since a lack of glutathione in these cells naturally increases ROS production and lipid peroxidation49. Notably, Glutathione Metabolism was also enriched in aged MuSCs (Figure 6D), consistent with altered glutathione metabolism, which is a hallmark of stem cell ageing50. Upon plating, the rate of lipid peroxidation dramatically increased (>4.5-fold increase) in aged MuSCs compared to that in younger cells, highlighting the enhanced sensitivity of aged MuSCs to oxidative and replicative stress outside their niche (Figure 6K). Although RSL3 treatment increased lipid peroxidation to comparable levels in MuSCs from both young and aged mice, Fer1 co-treatment rescued these effects. Importantly, Fer1 treatment alone prevented the increase in basal levels of lipid peroxidation observed in aged MuSCs (Figure 6K) and significantly enhanced both their viability and myogenic potential (Figure 6L-O). Taken together, these results demonstrate that ferroptosis is a dominant contributor to the decline in MuSC numbers with age, where activation of the ferroptotic program is triggered by the loss of Kmt5a.
Lastly, because human hematopoietic stem cells were recently shown to be selectively vulnerable to ferroptosis51 and labile iron accumulation was observed in aged hematopoietic stem cells45, we wondered whether age-associated death by ferroptosis is a shared fate across other adult stem cells. As previously described, we found that both short- and long-term HSCs were enriched in the aged bone marrow (Figure S10 A, B), with long-term HSCs displaying myeloid-bias CD41, a hallmark of HSC aging (Figure S10C). Upon sorting long-term HSCs from young and aged mice, we found that only aged LT-HSCs displayed enhanced lipid peroxidation (Figure S10D).
Taken together, these data indicate that lipid peroxidation and cell death by ferroptosis are important mediators of stem cell aging, and warrant further exploration.
Prevention of systemic inflammation averts muscle stem cell aging.
Finally, because we found that Kmt5a levels decreased in MuSCs in response to both distal injury and ectopic induction of acute systemic inflammation, we assessed whether long-term inhibition of these pro-inflammatory molecules could restore Kmt5a levels in MuSCs, prevent Kmt5a-mediated loss of quiescence and ferroptotic cell death, and improve skeletal muscle homeostasis and regeneration during aging (Figure 7). First, we confirmed that aged mice treated with an anti-inflammatory drug specifically targeting our pathway of interest (Bindarit 1/wk IP 30 mg/kg [12to24-30mo]) exhibited significantly diminished cellular and molecular inflammatory profiles similar to those observed in young mice (Figure S11A-K). As expected, Bindarit treatment restored Kmt5a levels in MuSCs from aged mice back to young levels (Figure S11L). In vivo, the number of MuSCs was significantly improved both at homeostasis and after regeneration compared to vehicle-treated aged mice (Figure 7A-D), complemented by enhanced muscle repair and restoration of muscle strength after injury (Figure 7D-G). Finally, both intracellular iron and lipid peroxidation significantly decreased in MuSCs derived from Bindarit-treated aged mice (Figure 7H and I). Unexpectedly, we observed that Bindarit-treated mice were leaner (Figure S11M) and had an extended lifespan (Figure S11N). Improvement in muscle regeneration was not observed in adult mice, despite the presence of fewer circulatory monocytes (Figure S12A-F). It was also not detrimental, possibly benefiting from circumstances with chronic inflammation only, as previously reported52-54.