Vertebrate skeletal muscle is composed of multinucleate myofibers that are surrounded by muscle connective tissue. Following injury, muscle is able to robustly regenerate because of tissue-resident muscle stem cells, called satellite cells. In addition, efficient and complete regeneration depends on other cells resident in muscle – including fibro-adipogenic progenitors (FAPs). Increasing evidence from single-cell analyses and genetic and transplantation experiments suggests that satellite cells and FAPs are heterogeneous cell populations. Here, we review our current understanding of the heterogeneity of satellite cells, their myogenic derivatives and FAPs in terms of gene expression, anatomical location, age and timing during the regenerative process – each of which have potentially important functional consequences.

Up to 40% of total body mass is made up of skeletal muscle, which is essential for movement, structural support and whole-body metabolism (Baskin et al., 2015). It is composed primarily of contractile multinucleate myofibers. These myofibers are surrounded by muscle connective tissue, which maintains the structural integrity of muscle and functionally transmits muscle contractile force to tendons and bones.

Adult vertebrate skeletal muscle has a remarkable capacity for regeneration, with restoration of complete function by one month, even after severe damage, in both mice and humans (Warren et al., 2007; Laumonier and Menetrey, 2016). Following acute injury, myofibers undergo degeneration and necrosis. As myofibers are post-mitotic, regeneration of skeletal muscle is mediated by tissue-resident stem cells, the satellite cells.

Satellite cells are essential for muscle regeneration (Fig. 1). In response to injury, quiescent satellite cells activate, transition to myoblasts and differentiate into myocytes, which fuse and regenerate multinucleate myofibers (Relaix and Zammit, 2012; Dumont et al., 2015). In addition to regenerating myofibers, some satellite cells self-renew, returning to quiescence and the myofiber-associated niche. Whether satellite cells differentiate or self-renew is tightly regulated to ensure both efficient regeneration of muscle and maintenance of the satellite cell pool (Dumont et al., 2015).

In addition to satellite cells, muscle regeneration requires the coordination of multiple cell types for complete restoration of muscle. These cell types include immune cells (macrophages, eosinophils, neutrophils and regulatory T cells), vascular endothelial cells and pericytes (Wosczyna and Rando, 2018). Of particular note are another population of cells, the fibro-adipogenic progenitors (FAPs; Joe et al., 2010; Uezumi et al., 2010) (Fig. 1). FAPs are a population of fibroblasts embedded in the muscle connective tissue, and during regeneration they play two crucial roles. First, FAPs synthesize the transient connective tissue fibrosis necessary to maintain the structural integrity of regenerating muscle (Goetsch et al., 2003; Serrano and Muñoz-Cánoves, 2010). Second, they provide trophic support for myogenic cells (Murphy et al., 2011; Wosczyna et al., 2019).

Regeneration is a complex cellular process. It requires the temporally and spatially regulated interplay between satellite cells, FAPs and other populations of cells resident in muscle. Furthermore, it has become apparent that satellite cells, their myogenic derivatives and FAPs are heterogeneous populations. The nature and functional consequences of this heterogeneity are long-standing questions. Recently, the introduction of single-cell sequencing and mass cytometry technology has led to the publication of multiple single-cell analyses of skeletal muscle that provide new insights into the heterogeneity of satellite cells, their myogenic derivatives and FAPs. Additional functional insights have been provided by new genetic and transplantation experiments. In this Review, we discuss current advances in our understanding of heterogeneity in satellite cells, their myogenic derivatives and FAPs with regards to gene expression, anatomical location, age, and timing during the regenerative process and functional capacity.

Satellite cells were first identified and named based on their unique ‘satellite’ position between the sarcolemma and basement membrane of myofibers (Fig. 1; Mauro, 1961). They were postulated to be muscle stem cells, but it was not until they were found to specifically express the transcription factor PAX7 (Seale et al., 2000) that their function could be formally demonstrated. The generation of Pax7CreER mice (Table S1) enabled inducible genetic labeling and ablation of satellite cells in the adult and established that satellite cells are both necessary and sufficient for muscle regeneration (Lepper et al., 2011; Murphy et al., 2011; Sambasivan et al., 2011).

Satellite cells are generally thought to uniformly lie in a quiescent G0 state in their niche underneath the basement membrane and adjacent to myofibers (Montarras et al., 2013). In homeostatic muscle, satellite cells constitute less than 10% of myofiber-associated nuclei (Bischoff and Franzini-Armstrong, 2004). Yet, the number of quiescent satellite cells varies stereotypically in different muscles: more satellite cells are associated with slow myofibers (e.g. in the predominantly slow soleus muscle), whereas fewer are associated with fast myofibers [e.g. in the fast extensor digitorum longus (EDL) muscle] (Collins et al., 2005). In addition, culture of C2C12 or MM14 satellite cell lines or primary satellite cells associated with myofibers revealed that subpopulations, termed reserve cells, are more resistant to differentiation (Clegg et al., 1987; Miller, 1990; Yoshida et al., 1998; Olguín and Olwin, 2004; Zammit et al., 2004). These observations suggest that quiescent satellite cells are a heterogeneous population, with differences in functional capacity.

In response to injury or damage, satellite cells activate, leave their niche and re-enter the cell cycle (Fig. 2; reviewed by Relaix and Zammit, 2012). Activated satellite cells increase in size and have a greater RNA content (Chargé and Rudnicki, 2004; Fukada et al., 2007; Rodgers et al., 2014). Upon activation, satellite cells either become committed myoblasts that express sustained levels of MYOD and differentiate to regenerate myofibers (Lahmann et al., 2019) or self-renew to replenish the satellite cell pool. Asymmetric cell division of activated satellite cells is thought to be crucial for establishing these two subpopulations of activated satellite cells (Shinin et al., 2006; Kuang et al., 2007; Feige et al., 2018). An outstanding question is whether these two subpopulations are distinct and exhibit unique molecular signatures.

Satellite cells destined to regenerate myofibers follow a stereotypical series of steps (Fig. 2). Upon activation, myoblasts proliferate and migrate to the site of injury. MYOD+ myoblasts differentiate into myocytes that express myogenin (MYOG) and fuse to regenerate myofibers (Chargé and Rudnicki, 2004). Regenerating myofibers, with their hallmark centralized nuclei, transiently express embryonic myosin heavy chain (MyHCemb) and then mature to express slow or fast myosin (Chargé and Rudnicki, 2004). Thus, the process of myogenesis during regeneration follows a defined trajectory: quiescent PAX7+MYOD− satellite cells activate and become committed PAX7−MyoD+ myoblasts, then differentiate to MYOG+ myocytes and finally fuse into myosin heavy chain+ (MyHC+) myofibers. Although PAX7, MYOD and MYOG have been key transcription factors defining the different myogenic populations, it has been an area of interest whether other previously unrecognized myogenic populations are present and whether cell surface markers [allowing for fluorescence-activated cell sorting (FACS) isolation] identify these populations.

Both regenerated and uninjured myofibers are syncytial, meaning they are composed of hundreds of myonuclei sharing a common cytoplasm and sarcolemma. How homogeneous these hundreds of myonuclei are within one myofiber was largely unknown. The accumulation of AchR (also known as Chrna2) and AchE transcripts adjacent to the neuromuscular junction and Col22a1 transcripts near the myotendinous junction argued that individual myonuclei are molecularly distinct (Jacobson et al., 2001; Koch et al., 2004). The identification of myofibers that express more than one MyHC isoform (MyHCI, MyHCIIA, MyHCIIX, MyHCIIB) also raises the question of whether individual myonuclei are specialized to express one type of MyHC and whether individual myonuclei in a mixed myofiber may express different MyHCs (Dos Santos et al., 2020). Recent single-nucleus RNA-seq experiments, which we discuss below, shed light on this debate.

For many years, researchers have tried to determine the degree of heterogeneity in the quiescent satellite cell population in muscle (reviewed by Biressi and Rando, 2010). One of the first pieces of molecular evidence for heterogeneity came from the identification in cultured isolated myofibers of a rare population of satellite cells that do not express CD34, M-cadherin or MYF5 (Beauchamp et al., 2000). Subsequently, the Rudnicki lab (Kuang et al., 2007) used Myf5cre/+;RosaYFP/+ mice to show that the majority of quiescent satellite cells express yellow fluorescent protein (YFP) (and so are Myf5+ or derived from Myf5+ cells), but 10% are negative for YFP (and so have never expressed Myf5) and are more stem-like.

Distinct subpopulations of quiescent satellite cells have also been identified based on the expression levels of Pax7 and the related transcription factor Pax3 (Rocheteau et al., 2012; Der Vartanian et al., 2019; Scaramozza et al., 2019). Rocheteau and colleagues identified two populations of quiescent satellite cells based on Pax7 expression using transgenic Pax7-nGFP mice (Table S1) (Rocheteau et al., 2012). They showed that Pax7Hi cells expressed higher levels of Cxcr4 and CD34 (markers of stemness; Beauchamp et al., 2000; Sherwood et al., 2004) and lower levels of Myog (a marker of differentiation). Upon activation, Pax7Hi cells took longer to perform the first cell division compared with Pax7Lo cells, and most Pax7Hi cells performed asymmetric DNA segregation during cell division, with the more stem-like daughter retaining template DNA. These results suggest that Pax7Hi cells are a subpopulation of quiescent satellite cells that are more stem-like than Pax7Lo cells. It has also been shown that a subset of Pax7+ satellite cells co-express Pax3 (Montarras et al., 2005; Relaix et al., 2005a). Two independent studies (Der Vartanian et al., 2019; Scaramozza et al., 2019) demonstrated that Pax7+ satellite cells that also express Pax3 are resistant to stress (environmental and irradiation). In normal conditions, Pax3+ satellite cells have limited contribution to muscle regeneration, but under stress they possess extensive regenerative potential (Scaramozza et al., 2019). During muscle homeostasis, Pax3+ satellite cells have also been found to contribute more readily to myofibers during muscle homeostasis (de Morree et al., 2019). Why only subsets of satellite cells express the Pax3 transcript is unclear, but PAX3 protein levels are additionally regulated by alternative polyadenylation (Boutet et al., 2012; de Morree et al., 2019). Overall, these studies identify heterogeneity in quiescent satellite cells present in uninjured muscle, but the relationship between the identified subpopulations has yet to be resolved.

The advent of single-cell RNA-seq and multi-parameter mass cytometry (CyTOF) have enabled the detailed characterization of cell populations within a tissue (Bandura et al., 2009; Macosko et al., 2015; Zheng et al., 2019). Recently, multiple single-cell analyses have been conducted on skeletal muscle (Table S2) and have investigated the composition of quiescent satellite cells (Table S2). Several studies found only a single population of quiescent satellite cells (Cho and Doles, 2017; De Micheli et al., 2020a; Oprescu et al., 2020; Rubenstein et al., 2020), whereas others found two subpopulations – one more quiescent or stem-like and one more primed to activate (Dell'Orso et al., 2019; De Micheli et al., 2020b; Kimmel et al., 2020, 2021; Yartseva et al., 2020) – or even more (Barruet et al., 2020; Perez et al., 2021). However, recent re-analysis and integration of all available single-cell and single-nuclei data calls into question whether subpopulations of quiescent satellite cells can be identified by these technologies (McKellar et al., 2020 preprint). Several studies have revealed that the dissociation of quiescent satellite cells induces stress, and the expression of genes encoding for heat shock proteins, immediate early genes (e.g. Fos and Jun) and other stress-related genes leads to the induction of an artificial cell population that resembles activated satellite cells 4 h post-injury (Machado et al., 2017, 2021; van den Brink et al., 2017). McKellar and colleagues’ re-analysis of all available datasets reveals that subpopulations of quiescent satellite cells are largely found in studies in which these cells were isolated via FACs and/or were cultured and therefore are likely experimentally-induced artifacts (McKellar et al., 2020 preprint). Thus, single-cell and single-nuclei studies do not currently support the existence of distinct subpopulations of quiescent satellite cells. However, it should be noted that these technologies are not well suited for the discovery of rare populations due to the small number and incomplete transcriptome of captured cells (McKellar et al., 2020 preprint).

Another question yet to be resolved is whether upon activation and asymmetric cell division some satellite cells have a unique set of markers that indicate that they will self-renew, whereas others have a different set of markers that indicate that they will differentiate. Several single-cell and single-nuclei studies examining regenerating muscle have only found a single population of activated satellite cells (Table S2; McKellar et al., 2020 preprint; Oprescu et al., 2020; Yartseva et al., 2020). However, two studies that examined multiple time points post injury and conducted pseudo-time analyses distinguished two populations of activated satellite cells (Porpiglia et al., 2017; De Micheli et al., 2020a). A third study of cultured primary satellite cells also identified two clusters of activated satellite cells (Kimmel et al., 2020). The lack of consensus among these studies likely derives from the difficulty in sampling enough activated satellite cells, as they are a relatively small and transitory population. An additional complication is a lack of a definitive marker of activated satellite cells. Although activated satellite cells have canonically been designated as Pax7+ cells which express MYOD protein (Grounds et al., 1992; Yablonka-Reuveni and Rivera, 1994), a recent study has shown that proliferating, activated satellite cells express oscillating levels of MYOD and so MYOD does not definitively mark these cells (Lahmann et al., 2019). A further difficulty is that subpopulations of activated satellite cells may be distinguished by lowly expressed genes or genes subject to post-transcriptional or post-translational modifications (e.g. Bernet et al., 2014). Thus, whether single-cell RNA-seq experiments can discriminate different subpopulations of activated satellite cells is unclear. To help resolve the question of heterogeneity in activated satellite cells, future single-cell studies will need to focus on the early time points post injury [1.5-3 days post injury (dpi)] and explicitly examine cell cycle status.

An area in which single-cell technology has been fruitful is the elucidation of populations of differentiating myogenic cells. Single-cell analyses of regenerating muscle have readily been able to identify proliferating myoblasts and differentiating myocytes as distinct populations (Porpiglia et al., 2017; Dell'Orso et al., 2019; De Micheli et al., 2020a; McKellar et al., 2020 preprint; Oprescu et al., 2020; Yartseva et al., 2020). Having distinguished these populations, these studies have been useful for providing additional markers (see Table S2) and insights into the cell-cycle regulators and the metabolic state of myoblasts and myocytes.

Myofibers are multinucleate, and whether myonuclei in different locations along mature myofibers are molecularly and functionally distinct has been a long-standing question. It has been difficult to assess transcriptional heterogeneity of myonuclei owing to their shared cytoplasm, but with the advent of single-nucleus RNA-seq technology, the transcriptional diversity of myonuclei has now been investigated. Four studies have identified multiple distinct populations of myonuclei (Chemello et al., 2020; Dos Santos et al., 2020; Kim et al., 2020; Petrany et al., 2020). Specifically, myonuclei associated with the myotendinous junctions, neuromuscular junctions, spindle-fibers and perimysium are molecularly distinct and the bulk of myonuclei express only one MyHC isoform. Interestingly, myonuclei populations and their transcriptomes differ during postnatal growth, age and muscular dystrophy (Chemello et al., 2020; Petrany et al., 2020; Perez et al., 2021). Together, these studies demonstrate that myonuclei have distinct transcriptional profiles that are associated with specialized regions and functions of the myofiber. How and when this specialization occurs is not known, but presumably it happens after myocyte fusion, as satellite cells, myoblasts and myocytes do not appear to contain specialized subpopulations correlating to the specialized myonuclear populations.

The anatomical location of satellite cells imparts significant heterogeneity in their gene expression, numbers and behavior. Satellite cells in different anatomical regions are derived from different embryonic regions (Randolph and Pavlath, 2015). Satellite cells in the trunk, limb, diaphragm and tongue arise from the somites. In contrast, satellite cells in the extraocular muscles arise from the prechordal mesoderm and cranial paraxial mesoderm of the first pharyngeal arch, masseter satellite cells arise from the first and second arches, and pharyngeal satellite cells from the third and fourth arches. Reflecting their different embryonic origins, extraocular, head, masseter and limb muscle satellite cells have different gene expression profiles (Harel et al., 2009; Sambasivan et al., 2009; Ono et al., 2010). Recently, two groups have explicitly profiled gene expression of satellite cells derived from the extraocular, masseter and tibialis anterior (TA) muscles. Notably, they found that satellite cells in the TA uniquely express HoxA and HoxC genes, as a result of both cell-intrinsic and -extrinsic (i.e. host environment) determinants (Evano et al., 2020; Yoshioka et al., 2021).

Not only does the number of satellite cells vary between different muscles (Collins et al., 2005; Ono et al., 2010; Keefe et al., 2015), but their behavior differs. During homeostasis, satellite cells continuously contribute to uninjured myofibers, but the degree of contribution varies between slow and fast myofibers and in different anatomical muscles (Keefe et al., 2015), potentially reflecting cell-intrinsic differences among satellite cells in different muscles (de Morree et al., 2019). During regeneration, satellite cells from different muscles are also functionally heterogeneous (see discussion below). Even within individual muscles, heterogeneity in satellite cell behavior exists. In uninjured muscle, satellite cells that reside in closer proximity to endothelial cells have been found to be more quiescent (EdU label-retaining; Verma et al., 2018). This suggests that endothelial cells may be crucial for inducing or maintaining a more stem-like (reserve) satellite cell population in muscle.

Although regeneration of myofibers proceeds in a stereotyped manner, several studies have found that the satellite cell-mediated rate of regeneration can vary. The time to activation is accelerated when a previous or contralateral muscle injury has occurred (Rodgers et al., 2014). In these circumstances, satellite cells primed for activation (termed G-alert) reach their first division sooner than quiescent G0 cells. Similarly, in response to environmental stress, Pax3+ satellite cells exhibit accelerated cell-cycle kinetics and appear to be ‘alerted’ compared with Pax3− cells (Der Vartanian et al., 2019; Kimmel et al., 2020). Differences in activation have also been found in satellite cells derived from different muscles: in myofiber cultures, satellite cells of the soleus muscle (composed mainly of slow MyHCI and IIa myofibers) activate more quickly than cells derived from the EDL muscle (composed of fast MyHCIIb myofibers) (Motohashi et al., 2018). Surprisingly, in vivo satellite cells derived from either the EDL or soleus regenerate myofibers at a faster rate than satellite cells derived from the TA (composed of MyHCIIx and b fibers) when transplanted into the TA muscle (Collins et al., 2005). Therefore, both fiber type and muscle-specific effects regulate the kinetics of associated satellite cells.

Satellite cells and regeneration are also significantly affected by age. With age, muscle mass and function declines (Narici and Maffulli, 2010) and concomitant with this, satellite cell number is generally thought to decline (Collins et al., 2007; Shefer et al., 2010). Aged satellite cells differ from young satellite cells in gene expression (Liu et al., 2013; Bernet et al., 2014; Sousa-Victor et al., 2014) and their epigenetic profile (Liu et al., 2013). In addition, as discussed below, multiple studies have demonstrated that aged satellite cells have compromised regenerative capacity. Finally, two recent studies have demonstrated that age affects satellite cell kinetics: satellite cells derived from older mice are delayed in their regenerative response compared with those derived from younger mice (Kimmel et al., 2020, 2021).

Transplantation studies, pioneered by Mikel Snow, Terence Partridge and Miranda Grounds (Snow, 1977, 1978; Partridge et al., 1978; Watt et al., 1982) have been essential in evaluating the regenerative capacity and potential functional heterogeneity of satellite cell populations in vivo. Such experiments have established that satellite cells from different muscles are functionally different (Collins et al., 2005; Ono et al., 2010). For example, soleus-derived donor satellite cells produce not only a higher number of myofibers, but more satellite cells than TA-derived satellite cells (Motohashi et al., 2018). A comparison of satellite cells derived from extraocular muscles with those derived from limb muscles also revealed that extraocular satellite cells have a significantly higher rate of engraftment when transplanted into the TA (Stuelsatz et al., 2015). Interestingly, the Motohashi study also showed that only donor satellite cells derived from the slow soleus, and not donor cells from the TA, yielded slow MyHCI+ regenerated myofibers when transplanted (Motohashi et al., 2018). Together, these studies indicate that satellite cells derived from different anatomical muscles and associated with different fiber types have different functional capacities.

Transplantation studies have revealed satellite cell subpopulations with distinct functional capabilities. As mentioned earlier, Kuang and colleagues distinguished YFP− and YFP+ satellite cells derived from Myf5cre/+;RosaYFP/+ mice (Kuang et al., 2007). Transplantation experiments showed that YPF− cells generated both YFP− and YFP+ satellite cell populations and produced more myofibers than YFP+ cells, indicating that YFP− cells are more stem-like. Similarly, when satellite cells were isolated based on Pax7 expression (Pax7lo and Pax7Hi), Pax7Hi cells generated both Pax7Hi and Pax7lo, although they did not differ in their fiber engraftment potential following transplantation (Rocheteau et al., 2012).

Functionally distinct satellite cell subpopulations have also been identified based on their cell-cycle behavior. Using H2B-GFP mice, Chakkalakal and colleagues distinguished a subpopulation of rarely-dividing satellite cells, termed label-retaining cells (LRCs) (Chakkalakal et al., 2012). Transplantation of LRCs showed that they give rise to more satellite cells and myofibers than non-LRC satellite cells (Chakkalakal et al., 2012, 2014). Recently, further heterogeneity within the LRC population, based on Mx1 lineage, was demonstrated (Scaramozza et al., 2019). Transplantation of Mx1+ and Mx1− LRC satellite cells showed that Mx1+ LRCs possessed a greater self-renewal potential and myofiber contribution than Mx1− LRCs, suggesting that Mx1+ LRCs are a more potent stem cell than Mx1− LRCs (Scaramozza et al., 2019). Mx1+ LRCs co-express Pax7 and Pax3, and Pax3 is required for their enhanced self-renewal and fiber contribution following transplantation (Scaramozza et al., 2019). Interestingly, under normal regeneration conditions, the majority of satellite cell self-renewal and myofiber contribution is through Mx1−Pax3−Pax7+ satellite cells; only under conditions of elevated stress or multiple injuries is this reserve population of Mx1+Pax3+Pax7+ LRCs recruited.

Transplantation studies have also been crucial for investigating satellite cell functional heterogeneity introduced by aging. Several studies have shown, by comparing heterochronic transplantation of old with isochronic transplantation of young satellite cells into young regenerating muscle, that aged satellite cells are defective in their ability to engraft (Chakkalakal et al., 2012; Bernet et al., 2014; Cosgrove et al., 2014; Sousa-Victor et al., 2014). These studies have identified several molecular changes – an increase in Spry1 expression and p38α/β MAPK activity and de-repression of p16INK4a – that lead to these cell-intrinsic functional defects. Not only do aged satellite cells differ from young ones, but heterogeneity exists within aged satellite cells. Transplantation of small numbers of aged satellite cells revealed that only a small percentage of these cells are functional stem cells (Cosgrove et al., 2014). Chakkalakal and colleagues demonstrated that the slower-dividing, more quiescent LRC aged satellite cells are a subpopulation better able to engraft and differentiate into myonuclei (Chakkalakal et al., 2012).

In summary, transplantation experiments have been crucial for revealing that satellite cells are functionally heterogeneous – differing depending upon the anatomical muscles or myofiber type they reside in, their age, lineage, gene expression and cell cycle behavior.

In uninjured skeletal muscle, fibroblasts are a population of cells that reside in the interstitial space between myofibers and are the major source of extracellular matrix (ECM) surrounding myofibers (Sasse et al., 1981; Kühl et al., 1982). ECM is crucial for muscle structure and function; it provides stability to the myofibers, blood vessels and nerves and transmits the contractile force from myofibers to tendons and bones (Grounds et al., 2005; Passerieux et al., 2007). Studying muscle connective tissue fibroblasts was initially difficult because of the lack of unique molecular markers and efficient methods to isolate these cells. Fibroblasts were originally identified and isolated via their in vitro functional properties; fibroblasts more readily bind to plastic dishes than myogenic cells and so have been isolated by pre-plating (Richler and Yaffe, 1970; Rando and Blau, 1994). Markers such as vimentin and α-smooth muscle actin have been used to identify muscle connective tissue fibroblasts, but these markers are not specific to these cells (e.g. Sato et al., 2003). Subsequently, other fibroblast markers were discovered: the transcription factors TCF4 (TCF7L2; Murphy et al., 2011) and OSR1 (Vallecillo-García et al., 2017; Stumm et al., 2018), fibroblast activation protein α (Roberts et al., 2013) and the receptor PDGFRα (Uezumi et al., 2010). Further lineage studies using tamoxifen-inducible Tcf4CreERT2 (Murphy et al., 2011), Osr1GCE (Stumm et al., 2018) and PdgfrαCreER (Wosczyna et al., 2019) mice demonstrated that these markers label the muscle connective tissue fibroblasts (Table S1). In addition to these studies, two independent groups identified cell surface markers that allow fibroblasts to be isolated via FACS: CD31− CD45− α7 integrin− Sca1+ (Joe et al., 2010) and CD31− CD45− PDGFRα+ (Uezumi et al., 2010). These fibroblasts were named FAPs based on their ability to differentiate into either fibroblasts or adipocytes (Joe et al., 2010). Subsequent experiments showed that these cells are also capable of differentiating into osteocytes in the presence of BMP2 (Wosczyna et al., 2012).

There has been debate in the literature as to the relationship between muscle connective tissue fibroblasts and FAPs (e.g. Wosczyna and Rando, 2018). It is likely that FAPs and fibroblasts identified by TCF4 and OSR1 are largely overlapping (Stumm et al., 2018). A common feature of all these fibroblasts is that they are distinct from myogenic cells and express PDGFRα (Joe et al., 2010; Uezumi et al., 2010, 2014; Murphy et al., 2011; Heredia et al., 2013; Malecova et al., 2018; Stumm et al., 2018; Biferali et al., 2019; Theret et al., 2021). For the sake of simplicity, we collectively refer to all adult muscle connective tissue fibroblasts as FAPs in this Review. Nevertheless, an unresolved question has been whether there are one or more distinct subpopulations of FAPs in uninjured and injured skeletal muscle.

During regeneration, FAPs are responsible for the transient fibrosis that is necessary to maintain the structural integrity of regenerating muscle (Fig. 2). Immediately following injury, FAPs rapidly proliferate and reach peak levels 3-5 dpi and then decline to basal levels by 14 dpi (Joe et al., 2010; Uezumi et al., 2010; Murphy et al., 2011; Lemos et al., 2015; Wosczyna et al., 2019). The rapidly expanding FAPs synthesize the ECM and transient fibrosis that occurs during regeneration. As regeneration concludes, this fibrosis is resolved by decreasing the number of FAPs and dampening their expression of pro-fibrotic genes (Lemos et al., 2015). Unresolved fibrosis impairs muscle function and is characteristic of diseased muscle (Serrano and Munoz-Canoves, 2017). Thus, the number of FAPs and their molecular characteristics are tightly regulated to allow for transient fibrosis, but avoid maladaptive persistent fibrosis.

During regeneration, FAPs can also adopt the alternative fate – becoming adipocytes rather than fibrogenic (Joe et al., 2010; Uezumi et al., 2010, 2011). Transient adipogenesis is a feature of regeneration after muscle injury (Lukjanenko et al., 2013) and this fat deposition is presumably mediated by FAP-derived adipocytes. As the long-term deposition of intramuscular fat by adipocytes is generally considered maladaptive, because it impairs muscle contractile strength and metabolism (Coen et al., 2010; Biltz et al., 2020), FAP differentiation into adipocytes is tightly controlled. Recently, both cell intrinsic (Mueller et al., 2016; Wosczyna et al., 2021) and cell extrinsic (Heredia et al., 2013; Kopinke et al., 2017) mediators of FAP differentiation into adipocytes have been described.

FAPs not only contribute to transient muscle fibrosis and adipogenesis during regeneration, but they also regulate satellite cells. Genetic ablation of FAPs using Tcf4CreERT2;RosaDTA (Murphy et al., 2011) or PDGFRaCreERT2;RosaDTA (Wosczyna et al., 2019) mice demonstrated that in vivo FAPs prevent satellite cells from prematurely differentiating, thereby promoting proliferation of satellite cells during the early phase of regeneration. Some in vitro experiments co-culturing FAPs with satellite cells have corroborated these results and found that FAPs promote satellite cell expansion (Quinn et al., 1990; Fiore et al., 2016; Lukjanenko et al., 2019), whereas others report that FAPs promote satellite cell differentiation (Joe et al., 2010; Mozzetta et al., 2013). Thus, it is possible that different subpopulations of FAPs play different roles in regeneration.

Finally, FAPs have further additional functions in muscle regeneration. FAPs play an important phagocytic role in removing necrotic debris (Heredia et al., 2013). FAPs are also a significant source of secreted signals that regulate satellite cells, macrophages and regulatory T cells (Biferali et al., 2019). The multiple roles of FAPs during regeneration suggest that there may be several subpopulations of FAPs that differ in their properties, functions or fate.

In addition to their functions in regeneration, strikingly FAPs have been found to be essential for maintenance of skeletal muscle. Genetic ablation of fibroblasts using PdgfraCreERT2;RosaDTA mice (Wosczyna et al., 2019; Uezumi et al., 2021) or transgenic FAPα-DTR mice (Roberts et al., 2013) resulted in a dramatic decrease in the cross-sectional area of myofibers and consequent sarcopenic-like loss in muscle mass. Therefore, FAPs are necessary to maintain homeostatic muscle. How FAPs mechanistically regulate muscle homeostasis is just starting to be elucidated; FAPs secrete BMP3B (GDF10), which maintains myofiber size via AKT signaling and neuromuscular junctions via stabilization of Schwann cells (Uezumi et al., 2021). Whether these FAPs in uninjured homeostatic muscle differ molecularly and functionally from activated FAPs during muscle regeneration has yet to be discovered.

Single-cell technologies have allowed us to delve into FAP molecular characteristics and elucidate whether there are different subpopulations of FAPs. Multiple studies have examined the composition of quiescent FAPs in uninjured muscle (Table S2). Several studies have concluded that there is only one population of quiescent FAPs (Giordani et al., 2019; Marinkovic et al., 2019; Pawlikowski et al., 2019 preprint; De Micheli et al., 2020a,b; Petrilli et al., 2020; Kimmel et al., 2021), whereas others have identified two FAP populations (Malecova et al., 2018; Oprescu et al., 2020; Rubenstein et al., 2020; Perez et al., 2021). Another study identified nine populations of quiescent FAPs that are located in different muscle regions (Muhl et al., 2020). Thus, there currently is not a consensus on the number of quiescent FAP populations. Interestingly, the transcriptional repressor HIC1 has emerged as a marker of quiescent FAPs – although it is not exclusively expressed in FAPs (Scott et al., 2019).

A question that has been elucidated by the use of single-cell technology is whether quiescent FAPs differ from FAPs during regeneration (i.e. activated FAPs). Multiple studies have found that quiescent FAPs are molecularly distinct and cluster separately from activated FAPs in injured muscle (Table S2; Malecova et al., 2018; Marinkovic et al., 2019; Pawlikowski et al., 2019 preprint; Scott et al., 2019; Oprescu et al., 2020; Kimmel et al., 2021). Although there is consensus that quiescent and activated FAPs are molecularly distinct, each of these studies stratified activated FAPs via different markers (Table S2). Future studies that integrate these datasets will be required to determine the relationship between identified populations of activated FAPs.

As FAPs can differentiate into fibrogenic or adipogenic cells (Joe et al., 2010; Uezumi et al., 2010, 2011), an outstanding question is whether fibrogenic versus adipogenic FAPs are molecularly distinct. The first group to investigate FAPs during regeneration found one population that was not present in uninjured muscle and unique to differentiation (Malecova et al., 2018). This population was described as fibrogenic based on high expression of known fibrogenic genes. Subsequently, two studies have observed more than one population of FAPs that segregate by fate. Kimmel and colleagues observed pre-adipocytes, adipocytes and myofibroblasts in cultured FAPs (Kimmel et al., 2021). McKellar's integrative analysis of multiple papers found three FAP populations: a stem FAP, a pro-remodeling FAP (which appears to be fibrogenic) and an adipogenic FAP (McKellar et al., 2020 preprint). Newly identified markers of fibrogenic versus adipogenic FAPs will be useful for future studies determining what drives FAPs to these two different fates.

Overall, these analyses from single-cell data indicate that there are multiple subpopulations of FAPs. Quiescent FAPs in uninjured muscle are distinct from FAPs following injury, and fibrogenic and adipogenic FAPs are distinguishable. The relationship between these different subpopulations is currently unclear, but it will be important to resolve and distinguish whether these subpopulations have different functions in muscle homeostasis and regeneration.

An outstanding question is whether FAPs vary in different anatomical regions. Intramuscular connective tissue is organized into three interconnected sheaths: epimysium, perimysium and endomysium (Sanes, 2004). The epimysium enwraps whole muscle, perimysium surrounds fiber bundles or fascicles and endomysium ensheathes individual myofibers. Each of these layers contains distinct proteoglycans and collagens (Jarvinen et al., 2002; Purslow, 2020). This difference in ECM expression in the different layers suggests that FAP populations within each layer may vary. Furthermore, there is some evidence that FAPs derived from different anatomical muscles are different in their growth rates and expression of matrix metalloproteases (Archile-Contreras et al., 2010). Whether FAPs in these different sheaths are molecularly distinct has recently been investigated by single-cell RNA-seq (Muhl et al., 2020). The authors found distinct populations of endomysial (six subclusters) and perimysial (two subclusters) FAPs, as well as FAPs associated with nerves and a newly described FAP population adjacent to the perimysium. Although these populations are distinguished by gene expression and location within the muscle, it has yet to be determined whether they are functionally distinct. Also, it is still unknown whether epimysial FAPs are molecularly and functionally distinct.

Another important question is whether FAPs differ temporally – over the course of regeneration or throughout the lifetime of an individual. During regeneration, the expansion and clearance of FAPs is tightly regulated (Lemos et al., 2015; Contreras et al., 2019; Marinkovic et al., 2019; Contreras et al., 2020; Eisner et al., 2020). A series of studies have shown that FAPs produce and secrete different factors at different times during regeneration to provide an optimal regenerative milieu (Lemos et al., 2012; Mozzetta et al., 2013; Kuswanto et al., 2016; Juban et al., 2018; Biferali et al., 2019; Lukjanenko et al., 2019). For example, FAPs secrete IL33 during the initial phases of regeneration (0-1 dpi; Kuswanto et al., 2016), whereas IL10 and IL6 are secreted at 3 dpi (Lemos et al., 2012). Congruent with these data, single-cell data have also segregated FAP subpopulations based on their progression during the regenerative process (Table S2). Oprescu et al. (2020) observed activated FAPs immediately following injury at 0.5-2 dpi, Wisp1+ FAPs at 3.5-5 dpi, Dlk1+ FAPs at 10 dpi and Osr1+ FAPs at 21 dpi. Wisp1+ FAPs are a fibrogenic population (based on co-expression of ECM genes) and have previously been identified as a distinct FAP population during regeneration (Lukjanenko et al., 2019). Likewise, a progression of FAP populations during regeneration has been shown, with activated FAPs at 2 dpi, remodeling or fibrogenic FAPs at 5 dpi and resolved FAPs at later post-injury time points (De Micheli et al., 2020a).

FAPs also differ over the lifetime of an individual. The quantity and quality of connective tissue changes with age, leading to increased muscle stiffness and reduced muscle function (Haus et al., 2007). Consistent with this, FAPs from aged versus young mice differ in their transcriptional profile (Lukjanenko et al., 2019; Kimmel et al., 2021). Aged FAPs also differ functionally such that, during regeneration, aged FAPs expand and are cleared more slowly and are more fibrogenic and less adipogenic (Lukjanenko et al., 2019).

Transplantation experiments have been an important experimental paradigm to test the fate and function of FAPs. Initial studies of FAPs isolated from uninjured muscle found that they do not engraft when transplanted into uninjured muscle, consistent with a hypothesis that healthy uninjured muscle does not support fibroadipogenic infiltration (Joe et al., 2010). However, it has subsequently been shown that FAPs isolated from uninjured muscle are able to engraft into uninjured muscle in which FAPs have been genetically ablated (Uezumi et al., 2021), into regenerating muscles after a BaCl2 injury (Uezumi et al., 2010; Wosczyna et al., 2019) or into muscles with adipocyte infiltration after a glycerol injury (Joe et al., 2010; Uezumi et al., 2010; Wosczyna et al., 2021). Therefore, it appears that FAPs from uninjured muscle are only able to engraft when transplanted into a disturbed muscle environment. Transplantation experiments have provided strong evidence that the muscle environment determines whether FAPs become either fibrogenic or adipogenic. Multiple studies have found that FAPs transplanted into muscle after a glycerol injury differentiate into adipocytes (Joe et al., 2010; Uezumi et al., 2010; Wosczyna et al., 2021). Uezumi and colleagues explicitly tested whether the local host environment dictates FAP fate by conducting a challenge experiment: FAPs derived from muscle damaged by a myotoxin or glycerol injury were transplanted into muscle damaged by the opposite injury type (Uezumi et al., 2010). FAPs from a glycerol-injured muscle transplanted into a toxin-injured muscle yielded few donor cells, whereas FAPs from a toxin injury transplanted into a glycerol-injured muscle yielded an accumulation of donor-derived adipocytes. These experiments demonstrate that the host environment strongly affects the fate of transplanted FAPs. However, the fate of donor FAPs is not entirely dictated by the host environment. For example, the intrinsic expression of miR-206 in FAPs inhibits FAP differentiation into adipocytes via repression of pro-adipogenic Runx1 – even when transplanted into glycerol-injured muscle (Wosczyna et al., 2021). Thus, although the host environment appears to have a dominant role in determining FAP fate, intrinsic FAP determinants also play a role.

The important function and therapeutic potential of FAPs in regulating muscle has also been elucidated by FAP transplantation experiments. Transplantation of FAPs into regenerating muscle reveals their function in regulating satellite cells and myofiber regeneration. For example, transplantation of young versus aged FAPs into injured muscle showed that young FAPs more strongly supported the expansion of activated satellite cells and demonstrated that the secretion of WISP1 by young FAPs (and not aged FAPs) was a key paracrine signal mediating this function (Lukjanenko et al., 2019). Transplantation of FAPs into uninjured muscle has been crucial for establishing that FAPs are essential for maintaining myofiber size and overall muscle mass (Wosczyna et al., 2019; Uezumi et al., 2021). In contrast, transplantation of pathogenic FAPs (derived from denervated muscle) into uninjured muscles leads to muscle atrophy and fibrosis and so demonstrates the important role of FAPs in regulating muscle pathology. Recently, transplantation experiments have also revealed the therapeutic potential of FAPs. Transplantation of FAPs that have been metabolically rewired by a short-term high-fat diet improved the size of dystrophic myofibers (Reggio et al., 2020) and suggested that FAPs are an important source of signals that can positively affect diseased myofibers.

Overall, transplantation experiments are a powerful tool to dissect the fate and function of FAPs and the role of intrinsic and extrinsic determinants of these processes. To date, these experiments have largely used FAPs isolated from uninjured hindlimb muscle. In the future, such transplantation experiments could be used to test whether different FAP subpopulations – isolated at different times during regeneration, from diverse muscles of different ages or from endomysial, perimysial or epimysial sheaths – are indeed functionally heterogeneous and/or have different fates.

In vertebrate skeletal muscle, satellite cells and FAPs are essential cell populations that contribute to muscle homeostasis and are required for muscle regeneration. However, as a multitude of studies have demonstrated, satellite cells and FAPs are heterogeneous populations. Satellite cells and FAPs vary between different anatomical muscles and even differ within individual muscles. Such anatomical heterogeneity perhaps is a consequence of the structural, functional and metabolic differences of the muscles they reside in. In addition, satellite cells and FAPs from aged compared with young muscles differ in gene expression and their functional properties.

Recent single-cell analyses have provided important insights into satellite cells and their myogenic derivatives (Table S2). Such analyses have been able to distinguish quiescent from activated satellite cells and delineate the series of myoblasts and myocytes that differentiate into myofibers during regeneration (Porpiglia et al., 2017; Dell'Orso et al., 2019; De Micheli et al., 2020a; McKellar et al., 2020 preprint; Oprescu et al., 2020; Yartseva et al., 2020) as well as show that there are several distinct populations of myonuclei (Chemello et al., 2020; Dos Santos et al., 2020; Kim et al., 2020; Petrany et al., 2020). However, current single-cell papers have surprisingly not definitively detected distinct subpopulations of quiescent satellite cells. Based on the identification of reserve cells in satellite cell cultures and My5 lineage, Pax3 and Pax7 studies (Beauchamp et al., 2000; Montarras et al., 2005; Relaix et al., 2005b; Rocheteau et al., 2012; Der Vartanian et al., 2019; Scaramozza et al., 2019), different populations of quiescent satellite cells seem likely. However, single-cell analyses are challenging because of the difficulty of isolating and analyzing truly quiescent satellite cells and the potential rarity of these subpopulations (McKellar et al., 2020 preprint); clearly this is an important issue to resolve. Another unresolved question is whether satellite cells from different anatomical muscles or even within different regions within particular muscles (e.g. near capillaries or associated with slow myofibers; Motohashi et al., 2018; Verma et al., 2018) are transcriptionally distinct and would segregate into distinct subpopulations. Currently, satellite cells from only a small number of muscles (e.g. extraocular, masseter, TA, EDL and soleus) have been explicitly compared and functionally tested. A survey of a wider array of muscles may reveal new populations of satellite cells with advantageous and potentially therapeutically useful properties.

Single-cell analyses have also elucidated heterogeneity in FAPs by identifying several FAP subpopulations. Quiescent FAPs found in uninjured muscle are distinct from FAPs activated during regeneration (Malecova et al., 2018; Marinkovic et al., 2019; Pawlikowski et al., 2019 preprint; Scott et al., 2019; Oprescu et al., 2020; Kimmel et al., 2021) and, within regenerating muscle, FAP subpopulations have been identified that differ in fate (fibrogenic versus adipogenic; Malecova et al., 2018; McKellar et al., 2020 preprint; Kimmel et al., 2021) or appearance in the regenerative process (De Micheli et al., 2020a; Oprescu et al., 2020). Resolving the lineage relationships between these subpopulations and determining their functional significance is an important area for future research. Also yet to be discovered is whether FAPs from different anatomical muscles are transcriptionally and/or functionally distinct. The different developmental origins of FAPs and the important role of these cells in differentially regulating the morphogenesis of anatomical muscles (Sefton and Kardon, 2019) suggests that FAPs residing in different adult anatomical muscles are molecularly distinct. Determining whether FAPs from different muscles differ molecularly and have unique functional capabilities may yield unique insights into their developmental and regenerative functions, as well as their therapeutic potential.

We thank B.D. Cosgrove for helpful discussions of single cell literature.

Funding

Research in G. Kardon's lab is supported by the National Institutes of Health (HD104317 and HD087360) and the Wheeler Foundation. Deposited in PMC for release after 12 months.

Archile-Contreras
,
A. C.
,
Mandell
,
I. B.
and
Purslow
,
P. P.
(
2010
).
Phenotypic differences in matrix metalloproteinase 2 activity between fibroblasts from 3 bovine muscles
.
J. Anim. Sci.
88
,
4006
-
4015
.
Bandura
,
D. R.
,
Baranov
,
V. I.
,
Ornatsky
,
O. I.
,
Antonov
,
A.
,
Kinach
,
R.
,
Lou
,
X.
,
Pavlov
,
S.
,
Vorobiev
,
S.
,
Dick
,
J. E.
and
Tanner
,
S. D.
(
2009
).
Mass cytometry: technique for real time single cell multitarget immunoassay based on inductively coupled plasma time-of-flight mass spectrometry
.
Anal. Chem.
81
,
6813
-
6822
.
Barruet
,
E.
,
Garcia
,
S. M.
,
Striedinger
,
K.
,
Wu
,
J.
,
Lee
,
S.
,
Byrnes
,
L.
,
Wong
,
A.
,
Xuefeng
,
S.
,
Tamaki
,
S.
,
Brack
,
A. S.
et al. 
(
2020
).
Functionally heterogeneous human satellite cells identified by single cell RNA sequencing
.
ELife
9
,
e51576
.
Baskin
,
K. K.
,
Winders
,
B. R.
and
Olson
,
E. N.
(
2015
).
Muscle as a “Mediator” of systemic metabolism
.
Cell Metab.
21
,
237
-
248
.
Beauchamp
,
J. R.
,
Heslop
,
L.
,
Yu
,
D. S. W.
,
Tajbakhsh
,
S.
,
Kelly
,
R. G.
,
Wernig
,
A.
,
Buckingham
,
M. E.
,
Partridge
,
T. A.
and
Zammit
,
P. S.
(
2000
).
Expression of Cd34 and Myf5 defines the majority of quiescent adult skeletal muscle satellite cells
.
J. Cell Biol.
151
,
1221
-
1234
.
Bernet
,
J. D.
,
Doles
,
J. D.
,
Hall
,
J. K.
,
Tanaka
,
K. K.
,
Carter
,
T. A.
and
Olwin
,
B. B.
(
2014
).
p38 MAPK signaling underlies a cell-autonomous loss of stem cell self-renewal in skeletal muscle of aged mice
.
Nat. Med.
20
,
265
-
271
.
Biferali
,
B.
,
Proietti
,
D.
,
Mozzetta
,
C.
and
Madaro
,
L.
(
2019
).
Fibro-adipogenic progenitors cross-talk in skeletal muscle: the social network
.
Front. Physiol.
10
,
1074
.
Biltz
,
N. K.
,
Collins
,
K. H.
,
Shen
,
K. C.
,
Schwartz
,
K.
,
Harris
,
C. A.
and
Meyer
,
G. A.
(
2020
).
Infiltration of intramuscular adipose tissue impairs skeletal muscle contraction
.
J. Physiol.
598
,
2669
-
2683
.
Biressi
,
S.
and
Rando
,
T. A.
(
2010
).
Heterogeneity in the muscle satellite cell population
.
Semin. Cell Dev. Biol.
21
,
845
-
854
.
Bischoff
,
R.
and
Franzini-Armstrong
,
C
. (
2004
).
Satellite and stem cells in muscle regeneration
. In
Myology
(ed.
A. G.
Engel
and
C.
Franzinin-Armstrong
), pp.
66
-
86
.
New York
:
McGraw-Hill
.
Bosnakovski
,
D.
,
Xu
,
Z.
,
Li
,
W.
,
Thet
,
S.
,
Cleaver
,
O.
,
Perlingeiro
,
R. C. R.
and
Kyba
,
M.
(
2008
).
Prospective isolation of skeletal muscle stem cells with a Pax7 reporter
.
Stem Cells
26
,
3194
-
3204
.
Boutet
,
S. C.
,
Cheung
,
T. H.
,
Quach
,
N. L.
,
Liu
,
L.
,
Prescott
,
S. L.
,
Edalati
,
A.
,
Iori
,
K.
and
Rando
,
T. A.
(
2012
).
Alternative polyadenylation mediates microRNA regulation of muscle stem cell function
.
Cell Stem Cell
10
,
327
-
336
.
Chakkalakal
,
J. V.
,
Jones
,
K. M.
,
Basson
,
M. A.
and
Brack
,
A. S.
(
2012
).
The aged niche disrupts muscle stem cell quiescence
.
Nature
490
,
355
-
360
.
Chakkalakal
,
J. V.
,
Christensen
,
J.
,
Xiang
,
W.
,
Tierney
,
M. T.
,
Boscolo
,
F. S.
,
Sacco
,
A.
and
Brack
,
A. S.
(
2014
).
Early forming label-retaining muscle stem cells require p27kip1 for maintenance of the primitive state
.
Development
141
,
1649
-
1659
.
Chargé
,
S. B. P.
and
Rudnicki
,
M. A.
(
2004
).
Cellular and molecular regulation of muscle regeneration
.
Physiol. Rev.
84
,
209
-
238
.
Chemello
,
F.
,
Wang
,
Z.
,
Li
,
H.
,
McAnally
,
J. R.
,
Liu
,
N.
,
Bassel-Duby
,
R.
and
Olson
,
E. N.
(
2020
).
Degenerative and regenerative pathways underlying Duchenne muscular dystrophy revealed by single-nucleus RNA sequencing
.
Proc. Natl. Acad. Sci. USA
117
,
29691
-
29701
.
Cho
,
D. S.
and
Doles
,
J. D.
(
2017
).
Single cell transcriptome analysis of muscle satellite cells reveals widespread transcriptional heterogeneity
.
Gene
636
,
54
-
63
.
Chung
,
M.-I.
,
Bujnis
,
M.
,
Barkauskas
,
C. E.
,
Kobayashi
,
Y.
and
Hogan
,
B. L. M.
(
2018
).
Niche-mediated BMP/SMAD signaling regulates lung alveolar stem cell proliferation and differentiation
.
Development
145
,
dev163014
.
Clegg
,
C. H.
,
Linkhart
,
T. A.
,
Olwin
,
B. B.
and
Hauschka
,
S. D.
(
1987
).
Growth factor control of skeletal muscle differentiation: commitment to terminal differentiation occurs in G1 phase and is repressed by fibroblast growth factor
.
J. Cell Biol.
105
,
949
-
956
.
Coen
,
P. M.
,
Dube
,
J. J.
,
Amati
,
F.
,
Stefanovic-Racic
,
M.
,
Ferrell
,
R. E.
,
Toledo
,
F. G.
and
Goodpaster
,
B. H.
(
2010
).
Insulin resistance is associated with higher intramyocellular triglycerides in type I but not type II myocytes concomitant with higher ceramide content
.
Diabetes
59
,
80
-
88
.
Collins
,
C. A.
,
Olsen
,
I.
,
Zammit
,
P. S.
,
Heslop
,
L.
,
Petrie
,
A.
,
Partridge
,
T. A.
and
Morgan
,
J. E.
(
2005
).
Stem cell function, self-renewal, and behavioral heterogeneity of cells from the adult muscle satellite cell niche
.
Cell
122
,
289
-
301
.
Collins
,
C. A.
,
Zammit
,
P. S.
,
Ruiz
,
A. P.
,
Morgan
,
J. E.
and
Partridge
,
T. A.
(
2007
).
A population of myogenic stem cells that survives skeletal muscle aging
.
Stem Cells
25
,
885
-
894
.
Contreras
,
O.
,
Cruz-Soca
,
M.
,
Theret
,
M.
,
Soliman
,
H.
,
Tung
,
L. W.
,
Groppa
,
E.
,
Rossi
,
F. M.
and
Brandan
,
E.
(
2019
).
Cross-talk between TGF-β and PDGFRα signaling pathways regulates the fate of stromal fibro-adipogenic progenitors
.
J. Cell Sci.
132
,
jcs232157
.
Contreras
,
O.
,
Soliman
,
H.
,
Theret
,
M.
,
Rossi
,
F. M. V.
and
Brandan
,
E.
(
2020
).
TGF-β-driven downregulation of the transcription factor TCF7L2 affects Wnt/β-catenin signaling in PDGFRα +fibroblasts
.
J. Cell Sci.
133
,
jcs242297
.
Cosgrove
,
B. D.
,
Gilbert
,
P. M.
,
Porpiglia
,
E.
,
Mourkioti
,
F.
,
Lee
,
S. P.
,
Corbel
,
S. Y.
,
Llewellyn
,
M. E.
,
Delp
,
S. L.
and
Blau
,
H. M.
(
2014
).
Rejuvenation of the muscle stem cell population restores strength to injured aged muscles
.
Nat. Med.
20
,
255
-
264
.
Dell'Orso
,
S.
,
Juan
,
A. H.
,
Ko
,
K.-D.
,
Naz
,
F.
,
Gutierrez-Cruz
,
G.
,
Feng
,
X.
and
Sartorelli
,
V.
(
2019
).
Single-cell analysis of adult skeletal muscle stem cells in homeostatic and regenerative conditions
.
Development
146
,
dev174177
.
De Micheli
,
A. J.
,
Laurilliard
,
E. J.
,
Heinke
,
C. L.
,
Ravichandran
,
H.
,
Fraczek
,
P.
,
Soueid-Baumgarten
,
S.
,
De Vlaminck
,
I.
,
Elemento
,
O.
and
Cosgrove
,
B. D.
(
2020a
).
Single-cell analysis of the muscle stem cell hierarchy identifies heterotypic communication signals involved in skeletal muscle regeneration
.
Cell Rep
30
,
3583
-
3595.e5
.
De Micheli
,
A. J.
,
Spector
,
J. A.
,
Elemento
,
O.
and
Cosgrove
,
B. D.
(
2020b
).
A reference single-cell transcriptomic atlas of human skeletal muscle tissue reveals bifurcated muscle stem cell populations
.
Skelet Muscle
10
,
19
.
de Morree
,
A.
,
Klein
,
J. D. D.
,
Gan
,
Q.
,
Farup
,
J.
,
Urtasun
,
A.
,
Kanugovi
,
A.
,
Bilen
,
B.
,
van Velthoven
,
C. T. J.
,
Quarta
,
M.
and
Rando
,
T. A.
(
2019
).
Alternative polyadenylation of Pax3 controls muscle stem cell fate and muscle function
.
Science
366
,
734
-
738
.
Der Vartanian
,
A.
,
Quétin
,
M.
,
Michineau
,
S.
,
Auradé
,
F.
,
Hayashi
,
S.
,
Dubois
,
C.
,
Rocancourt
,
D.
,
Drayton-Libotte
,
B.
,
Szegedi
,
A.
,
Buckingham
,
M.
et al. 
(
2019
).
PAX3 confers functional heterogeneity in skeletal muscle stem cell responses to environmental stress
.
Stem Cell
24
,
958
-
973.e9
.
Ding
,
G.
,
Tanaka
,
Y.
,
Hayashi
,
M.
,
Nishikawa
,
S.-I.
and
Kataoka
,
H.
(
2013
).
PDGF Receptor alpha+ mesoderm contributes to endothelial and hematopoietic cells in mice
.
Dev. Dyn.
242
,
254
-
268
.
Dos Santos
,
M.
,
Backer
,
S.
,
Saintpierre
,
B.
,
Izac
,
B.
,
Andrieu
,
M.
,
Letourneur
,
F.
,
Relaix
,
F.
,
Sotiropoulos
,
A.
and
Maire
,
P.
(
2020
).
Single-nucleus RNA-seq and FISH identify coordinated transcriptional activity in mammalian myofibers
.
Nat. Commun.
11
,
5102
.
Dumont
,
N. A.
,
Bentzinger
,
C. F.
,
Sincennes
,
M. C.
and
Rudnicki
,
M. A.
(
2015
).
Satellite cells and skeletal muscle regeneration
.
Compr Physiol
5
,
1027
-
1059
.
Eisner
,
C.
,
Cummings
,
M.
,
Johnston
,
G.
,
Tung
,
L. W.
,
Groppa
,
E.
,
Chang
,
C.
and
Rossi
,
F. M. V.
(
2020
).
Murine tissue-resident PDGFRα+ fibro-adipogenic progenitors spontaneously acquire osteogenic phenotype in an altered inflammatory environment
.
J. Bone Miner. Res.
35
,
1525
-
1534
.
Evano
,
B.
,
Gill
,
D.
,
Hernando-Herraez
,
I.
,
Comai
,
G.
,
Stubbs
,
T. M.
,
Commere
,
P. H.
,
Reik
,
W.
and
Tajbakhsh
,
S.
(
2020
).
Transcriptome and epigenome diversity and plasticity of muscle stem cells following transplantation
.
PLoS Genet.
16
,
e1009022
.
Feige
,
P.
,
Brun
,
C. E.
,
Ritso
,
M.
and
Rudnicki
,
M. A.
(
2018
).
Orienting muscle stem cells for regeneration in homeostasis
.
Aging Disease. Stem Cell
23
,
653
-
664
.
Fiore
,
D.
,
Judson
,
R. N.
,
Low
,
M.
,
Lee
,
S.
,
Zhang
,
E.
,
Hopkins
,
C.
,
Xu
,
P.
,
Lenzi
,
A.
,
Rossi
,
F. M. V.
and
Lemos
,
D. R.
(
2016
).
Pharmacological blockage of fibro/adipogenic progenitor expansion and suppression of regenerative fibrogenesis is associated with impaired skeletal muscle regeneration
.
Stem Cell Res.
17
,
161
-
169
.
Fukada
,
S.-i.
,
Uezumi
,
A.
,
Ikemoto
,
M.
,
Masuda
,
S.
,
Segawa
,
M.
,
Tanimura
,
N.
,
Yamamoto
,
H.
,
Miyagoe-Suzuki
,
Y.
and
Takeda
,
S. i.
(
2007
).
Molecular signature of quiescent satellite cells in adult skeletal muscle
.
Stem Cells
25
,
2448
-
2459
.
Giordani
,
L.
,
He
,
G. J.
,
Negroni
,
E.
,
Sakai
,
H.
,
Law
,
J. Y. C.
,
Siu
,
M. M.
,
Wan
,
R.
,
Corneau
,
A.
,
Tajbakhsh
,
S.
,
Cheung
,
T. H.
et al. 
(
2019
).
High-dimensional single-cell cartography reveals novel skeletal muscle-resident cell populations
.
Mol. Cell
74
,
609
-
621.e6
.
Goetsch
,
S. C.
,
Hawke
,
T. J.
,
Gallardo
,
T. D.
,
Richardson
,
J. A.
and
Garry
,
D. J.
(
2003
).
Transcriptional profiling and regulation of the extracellular matrix during muscle regeneration
.
Physiol. Genomics
14
,
261
-
271
.
Grounds
,
M. D.
,
Garrett
,
K. L.
,
Lai
,
M. C.
,
Wright
,
W. E.
and
Beilharz
,
M. W.
(
1992
).
Identification of skeletal muscle precursor cells in vivo by use of MyoD1 and myogenin probes
.
Cell Tissue Res.
267
,
99
-
104
.
Grounds
,
M. D.
,
Sorokin
,
L.
and
White
,
J.
(
2005
).
Strength at the extracellular matrix-muscle interface
.
Scand. J. Med. Sci. Sports
15
,
381
-
391
.
Hamilton
,
T. G.
,
Klinghoffer
,
R. A.
,
Corrin
,
P. D.
and
Soriano
,
P.
(
2003
).
Evolutionary divergence of platelet-derived growth factor alpha receptor signaling mechanisms
.
Mol. Cell. Biol.
23
,
4013
-
4025
.
Harel
,
I.
,
Nathan
,
E.
,
Tirosh-Finkel
,
L.
,
Zigdon
,
H.
,
Guimaraes-Camboa
,
N.
,
Evans
,
S. M.
and
Tzahor
,
E.
(
2009
).
Distinct origins and genetic programs of head muscle satellite cells
.
Dev. Cell
16
,
822
-
832
.
Haus
,
J. M.
,
Carrithers
,
J. A.
,
Trappe
,
S. W.
and
Trappe
,
T. A.
(
2007
).
Collagen, cross-linking, and advanced glycation end products in aging human skeletal muscle
.
J. Appl. Physiol. (1985)
103
,
2068
-
2076
.
Heredia
,
J. E.
,
Mukundan
,
L.
,
Chen
,
F. M.
,
Mueller
,
A. A.
,
Deo
,
R. C.
,
Locksley
,
R. M.
,
Rando
,
T. A.
and
Chawla
,
A.
(
2013
).
Type 2 innate signals stimulate fibro/adipogenic progenitors to facilitate muscle regeneration
.
Cell
153
,
376
-
388
.
Jacobson
,
C.
,
Cote
,
P. D.
,
Rossi
,
S. G.
,
Rotundo
,
R. L.
and
Carbonetto
,
S.
(
2001
).
The dystroglycan complex is necessary for stabilization of acetylcholine receptor clusters at neuromuscular junctions and formation of the synaptic basement membrane
.
J. Cell Biol.
152
,
435
-
450
.
Jarvinen
,
T. A. H.
,
Jozsa
,
L.
,
Kannus
,
P.
,
Jarvinen
,
T. L. N.
and
Jarvinen
,
M.
(
2002
).
Organization and distribution of intramuscular connective tissue in normal and immobilized skeletal muscles - An immunohistochemical, polarization and scanning electron microscopic study
.
J. Muscle Res. Cell Motil.
23
,
245
-
254
.
Joe
,
A. W. B.
,
Yi
,
L.
,
Natarajan
,
A.
,
Le Grand
,
F.
,
So
,
L.
,
Wang
,
J.
,
Rudnicki
,
M. A.
and
Rossi
,
F. M. V.
(
2010
).
Muscle injury activates resident fibro/adipogenic progenitors that facilitate myogenesis
.
Nat. Cell Biol.
12
,
153
-
163
.
Juban
,
G.
,
Saclier
,
M.
,
Yacoub-Youssef
,
H.
,
Kernou
,
A.
,
Arnold
,
L.
,
Boisson
,
C.
,
Ben Larbi
,
S.
,
Magnan
,
M.
,
Cuvellier
,
S.
,
Theret
,
M.
et al. 
(
2018
).
AMPK activation regulates LTBP4-dependent TGF-beta;1 secretion by pro-inflammatory macrophages and controls fibrosis in duchenne muscular dystrophy
.
CellReports
25
,
2163
-
2176.e6
.
Kang
,
S. H.
,
Fukaya
,
M.
,
Yang
,
J. K.
,
Rothstein
,
J. D.
and
Bergles
,
D. E.
(
2010
).
NG2+ CNS Glial progenitors remain committed to the oligodendrocyte lineage in postnatal life and following neurodegeneration
.
Neuron
68
,
668
-
681
.
Keefe
,
A. C.
,
Lawson
,
J. A.
,
Flygare
,
S. D.
,
Fox
,
Z. D.
,
Colasanto
,
M. P.
,
Mathew
,
S. J.
,
Yandell
,
M.
and
Kardon
,
G.
(
2015
).
Muscle stem cells contribute to myofibres in sedentary adult mice
.
Nat. Commun.
6
,
7087
.
Keller
,
C.
,
Hansen
,
M. S.
,
Coffin
,
C. M.
and
Capecchi
,
M. R.
(
2004
).
Pax3:Fkhr interferes with embryonic Pax3 and Pax7 function: implications for alveolar rhabdomyosarcoma cell of origin
.
Genes Dev.
18
,
2608
-
2613
.
Kim
,
M.
,
Franke
,
V.
,
Brandt
,
B.
,
Lowenstein
,
E. D.
,
Schöwel
,
V.
,
Spuler
,
S.
,
Akalin
,
A.
and
Birchmeier
,
C.
(
2020
).
Single-nucleus transcriptomics reveals functional compartmentalization in syncytial skeletal muscle cells
.
Nat. Commun.
11
,
6375
.
Kimmel
,
J. C.
,
Hwang
,
A. B.
,
Scaramozza
,
A.
,
Marshall
,
W. F.
and
Brack
,
A. S.
(
2020
).
Aging induces aberrant state transition kinetics in murine muscle stem cells
.
Development
147
,
dev183855
.
Kimmel
,
J. C.
,
Yi
,
N.
,
Roy
,
M.
,
Hendrickson
,
D. G.
and
Kelley
,
D. R.
(
2021
).
Differentiation reveals latent features of aging and an energy barrier in murine myogenesis
.
Cell Rep
35
,
109046
.
Koch
,
M.
,
Schulze
,
J.
,
Hansen
,
U.
,
Ashwodt
,
T.
,
Keene
,
D. R.
,
Brunken
,
W. J.
,
Burgeson
,
R. E.
,
Bruckner
,
P.
and
Bruckner-Tuderman
,
L.
(
2004
).
A novel marker of tissue junctions, collagen XXII
.
J. Biol. Chem.
279
,
22514
-
22521
.
Kopinke
,
D.
,
Roberson
,
E. C.
and
Reiter
,
J. F.
(
2017
).
Ciliary hedgehog signaling restricts injury-induced adipogenesis
.
Cell
170
,
340
-
351.e2
.
Kuang
,
S.
,
Kuroda
,
K.
,
Le Grand
,
F.
and
Rudnicki
,
M. A.
(
2007
).
Asymmetric self-renewal and commitment of satellite stem cells in muscle
.
Cell
129
,
999
-
1010
.
Kühl
,
U.
,
Timpl
,
R.
and
von der Mark
,
K.
(
1982
).
Synthesis of type IV collagen and laminin in cultures of skeletal muscle cells and their assembly on the surface of myotubes
.
Dev. Biol.
93
,
344
-
354
.
Kuswanto
,
W.
,
Burzyn
,
D.
,
Panduro
,
M.
,
Wang
,
K. K.
,
Jang
,
Y. C.
,
Wagers
,
A. J.
,
Benoist
,
C.
and
Mathis
,
D.
(
2016
).
Poor repair of skeletal muscle in aging mice reflects a defect in local, interleukin-33–dependent accumulation of regulatory T cells
.
Immuni
44
,
355
-
367
.
Lahmann
,
I.
,
Bröhl
,
D.
,
Zyrianova
,
T.
,
Isomura
,
A.
,
Czajkowski
,
M. T.
,
Kapoor
,
V.
,
Griger
,
J.
,
Ruffault
,
P.-L.
,
Mademtzoglou
,
D.
,
Zammit
,
P. S.
et al. 
(
2019
).
Oscillations of MyoD and Hes1 proteins regulate the maintenance of activated muscle stem cells
.
Genes Dev.
33
,
524
-
535
.
Laumonier
,
T.
and
Menetrey
,
J.
(
2016
).
Muscle injuries and strategies for improving their repair
.
J. Exp. Orthop.
3
,
15
.
Lemos
,
D. R.
,
Paylor
,
B.
,
Chang
,
C.
,
Sampaio
,
A.
,
Underhill
,
T. M.
and
Rossi
,
F. M. V.
(
2012
).
Functionally convergent white adipogenic progenitors of different lineages participate in a diffused system supporting tissue regeneration
.
Stem Cells
30
,
1152
-
1162
.
Lemos
,
D. R.
,
Babaeijandaghi
,
F.
,
Low
,
M.
,
Chang
,
C. K.
,
Lee
,
S. T.
,
Fiore
,
D.
,
Zhang
,
R.-H.
,
Natarajan
,
A.
,
Nedospasov
,
S. A.
and
Rossi
,
F. M. V.
(
2015
).
Nilotinib reduces muscle fibrosis in chronic muscle injury by promoting TNF-mediated apoptosis of fibro/adipogenic progenitors
.
Nat. Med.
21
,
786
-
794
.
Lepper
,
C.
,
Conway
,
S. J.
and
Fan
,
C. M.
(
2009
).
Adult satellite cells and embryonic muscle progenitors have distinct genetic requirements
.
Nature
460
,
627
-
631
.
Lepper
,
C.
,
Partridge
,
T. A.
and
Fan
,
C. M.
(
2011
).
An absolute requirement for Pax7-positive satellite cells in acute injury-induced skeletal muscle regeneration
.
Development
138
,
3639
-
3646
.
Liu
,
L.
,
Cheung
,
T. H.
,
Charville
,
G. W.
,
Hurgo
,
B. M. C.
,
Leavitt
,
T.
,
Shih
,
J.
,
Brunet
,
A.
and
Rando
,
T. A.
(
2013
).
Chromatin modifications as determinants of muscle stem cell quiescence and chronological aging
.
CellReports
4
,
189
-
204
.
Lukjanenko
,
L.
,
Brachat
,
S.
,
Pierrel
,
E.
,
Lach-Trifilieff
,
E.
and
Feige
,
J. N.
(
2013
).
Genomic profiling reveals that transient adipogenic activation is a hallmark of mouse models of skeletal muscle regeneration
.
PLoS ONE
8
,
e71084
.
Lukjanenko
,
L.
,
Karaz
,
S.
,
Stuelsatz
,
P.
,
Gurriaran-Rodriguez
,
U.
,
Michaud
,
J.
,
Dammone
,
G.
,
Sizzano
,
F.
,
Mashinchian
,
O.
,
Ancel
,
S.
,
Migliavacca
,
E.
et al. 
(
2019
).
aging disrupts muscle stem cell function by impairing matricellular WISP1 secretion from fibro-adipogenic progenitors
.
Cell Stem Cell
24
,
433
-
446.e7
.
Machado
,
L.
,
de Lima
,
J. E.
,
Fabre
,
O.
,
Proux
,
C.
,
Legendre
,
R.
,
Szegedi
,
A.
,
Varet
,
H.
,
Ingerslev
,
L. R.
,
Barrès
,
R.
,
Relaix
,
F.
et al. 
(
2017
).
In situ fixation redefines quiescence and early activation of skeletal muscle stem cells
.
CellReports
21
,
1982
-
1993
.
Machado
,
L.
,
Geara
,
P.
,
Camps
,
J.
,
Dos Santos
,
M.
,
Teixeira-Clerc
,
F.
,
Van Herck
,
J.
,
Varet
,
H.
,
Legendre
,
R.
,
Pawlotsky
,
J.-M.
,
Sampaolesi
,
M.
et al. 
(
2021
).
Tissue damage induces a conserved stress response that initiates quiescent muscle stem cell activation
.
Cell Stem Cell
28
,
1125
-
1135.e7
.
Malecova
,
B.
,
Gatto
,
S.
,
Etxaniz
,
U.
,
Passafaro
,
M.
,
Cortez
,
A.
,
Nicoletti
,
C.
,
Giordani
,
L.
,
Torcinaro
,
A.
,
Bardi
,
M.
,
Bicciato
,
S.
et al. 
(
2018
).
Dynamics of cellular states of fibro-adipogenic progenitors during myogenesis and muscular dystrophy
.
Nat. Commun.
9
,
3670
.
Marinkovic
,
M.
,
Fuoco
,
C.
,
Sacco
,
F.
,
Cerquone Perpetuini
,
A.
,
Giuliani
,
G.
,
Micarelli
,
E.
,
Pavlidou
,
T.
,
Petrilli
,
L. L.
,
Reggio
,
A.
,
Riccio
,
F.
et al. 
(
2019
).
Fibro-adipogenic progenitors of dystrophic mice are insensitive to NOTCH regulation of adipogenesis
.
Life Science Alliance
2
,
e201900437
.
Mathew
,
S. J.
,
Hansen
,
J. M.
,
Merrell
,
A. J.
,
Murphy
,
M. M.
,
Lawson
,
J. A.
,
Hutcheson
,
D. A.
,
Hansen
,
M. S.
,
Angus-Hill
,
M.
and
Kardon
,
G.
(
2010
).
Connective tissue fibroblasts and Tcf4 regulate myogenesis
.
Development
138
,
371
-
384
.
Mauro
,
A.
(
1961
).
Satellite cell of skeletal muscle fibers
.
J. Biophys. Biochem. Cytol.
9
,
493
-
495
.
McKellar
,
D. W.
,
Walter
,
L. D.
,
Song
,
L. T.
,
Mantri
,
M.
,
Wang
,
M. F. Z.
,
De Vlaminck
,
I.
and
Cosgrove
,
B. D.
(
2020
).
Strength in numbers: Large-scale integration of single-cell transcriptomic data reveals rare, transient muscle progenitor cell states in muscle regeneration
.
bioRxiv
2020.12.01.407460
.
Miller
,
J. B.
(
1990
).
Myogenic programs of mouse muscle cell lines: expression of myosin heavy chain isoforms, MyoD1, and myogenin
.
J. Cell Biol.
111
,
1149
-
1159
.
Miwa
,
H.
and
Era
,
T.
(
2015
).
Generation and characterization of PDGFRα-GFPCreER t2 knock-in mouse line
.
Genesis
53
,
329
-
336
.
Montarras
,
D.
,
Morgan
,
J.
,
Collins
,
C.
,
Relaix
,
F.
,
Zaffran
,
S. p.
,
Cumano
,
A.
,
Partridge
,
T.
and
Buckingham
,
M.
(
2005
).
Direct isolation of satellite cells for skeletal muscle regeneration
.
Science
309
,
2064
-
2067
.
Montarras
,
D.
,
L'Honoré
,
A.
and
Buckingham
,
M.
(
2013
).
Lying low but ready for action: the quiescent muscle satellite cell
.
FEBS J.
280
,
4036
-
4050
.
Motohashi
,
N.
,
Uezumi
,
A.
,
Asakura
,
A.
,
Ikemoto-Uezumi
,
M.
,
Mori
,
S.
,
Mizunoe
,
Y.
,
Takashima
,
R.
,
Miyagoe-Suzuki
,
Y.
,
Takenda
,
S. i.
and
Shigemoto
,
K.
(
2018
).
Tbx1 regulates inherited metabolic and myogenic abilities of progenitor cells derived from slow- and fast-type muscle
.
Cell Death Differ.
26
,
1024
-
1036
.
Mozzetta
,
C.
,
Consalvi
,
S.
,
Saccone
,
V.
,
Tierney
,
M.
,
Diamantini
,
A.
,
Mitchell
,
K. J.
,
Marazzi
,
G.
,
Borsellino
,
G.
,
Battistini
,
L.
,
Sassoon
,
D.
et al. 
(
2013
).
Fibroadipogenic progenitors mediate the ability of HDAC inhibitors to promote regeneration in dystrophic muscles of young, but not old Mdx mice
.
EMBO Mol. Med.
5
,
626
-
639
.
Mueller
,
A. A.
,
van Velthoven
,
C. T.
,
Fukumoto
,
K. D.
,
Cheung
,
T. H.
and
Rando
,
T. A.
(
2016
).
Intronic polyadenylation of PDGFRα in resident stem cells attenuates muscle fibrosis
.
Nature
540
,
276
-
279
.
Muhl
,
L.
,
Genove
,
G.
,
Leptidis
,
S.
,
Liu
,
J.
,
He
,
L.
,
Mocci
,
G.
,
Sun
,
Y.
,
Gustafsson
,
S.
,
Buyandelger
,
B.
,
Chivukula
,
I. V.
et al. 
(
2020
).
Single-cell analysis uncovers fibroblast heterogeneity and criteria for fibroblast and mural cell identification and discrimination
.
Nat. Commun.
11
,
3953
.
Murphy
,
M. M.
,
Lawson
,
J. A.
,
Mathew
,
S. J.
,
Hutcheson
,
D. A.
and
Kardon
,
G.
(
2011
).
Satellite cells, connective tissue fibroblasts and their interactions are crucial for muscle regeneration
.
Development
138
,
3625
-
3637
.
Narici
,
M. V.
and
Maffulli
,
N.
(
2010
).
Sarcopenia: characteristics, mechanisms and functional significance
.
Br. Med. Bull.
95
,
139
-
159
.
Nishijo
,
K.
,
Hosoyama
,
T.
,
Bjornson
,
C. R. R.
,
Schaffer
,
B. S.
,
Prajapati
,
S. I.
,
Bahadur
,
A. N.
,
Hansen
,
M. S.
,
Blandford
,
M. C.
,
McCleish
,
A. T.
,
Rubin
,
B. P.
et al. 
(
2009
).
Biomarker system for studying muscle, stem cells, and cancer in vivo
.
The FASEB J.
23
,
2681
-
2690
.
Olguín
,
H. C.
and
Olwin
,
B. B.
(
2004
).
Pax-7 up-regulation inhibits myogenesis and cell cycle progression in satellite cells: a potential mechanism for self-renewal
.
Dev. Biol.
275
,
375
-
388
.
Ono
,
Y.
,
Boldrin
,
L.
,
Knopp
,
P.
,
Morgan
,
J. E.
and
Zammit
,
P. S.
(
2010
).
Muscle satellite cells are a functionally heterogeneous population in both somite-derived and branchiomeric muscles
.
Dev. Biol.
337
,
29
-
41
.
Oprescu
,
S. N.
,
Yue
,
F.
,
Qiu
,
J.
,
Brito
,
L. F.
and
Kuang
,
S.
(
2020
).
Temporal Dynamics and Heterogeneity of Cell Populations during Skeletal Muscle Regeneration
.
ISCIENCE
23
,
100993
.
Partridge
,
T. A.
,
Grounds
,
M.
and
Sloper
,
J. C.
(
1978
).
Evidence of fusion between host and donor myoblasts in skeletal muscle grafts
.
Nature Publishing Group
273
,
306
-
308
.
Passerieux
,
E.
,
Rossignol
,
R.
,
Letellier
,
T.
and
Delage
,
J. P.
(
2007
).
Physical continuity of the perimysium from myofibers to tendons: involvement in lateral force transmission in skeletal muscle
.
J. Struct. Biol.
159
,
19
-
28
.
Pawlikowski
,
B.
,
Betta
,
N. D.
,
Elston
,
T.
,
O'Rourke
,
R.
,
Jones
,
K.
and
Olwin
,
B. B.
(
2019
).
A cellular atlas of skeletal muscle regeneration and aging
.
bioRxiv
635805
.
Perez
,
K.
,
McGirr
,
J.
,
Limbad
,
C.
,
Doi
,
R.
,
Nederveen
,
J. P.
,
Nilsson
,
M. I.
,
Tarnopolsky
,
M.
,
Campisi
,
J.
and
Melov
,
S.
(
2021
).
Single nuclei profiling identifies cell specific markers of skeletal muscle aging, sarcopenia and senescence
.
medRxiv
2021
,
2001.2022.21250336
.
Petrany
,
M. J.
,
Swoboda
,
C. O.
,
Sun
,
C.
,
Chetal
,
K.
,
Chen
,
X.
,
Weirauch
,
M. T.
,
Salomonis
,
N.
and
Millay
,
D. P.
(
2020
).
Single-nucleus RNA-seq identifies transcriptional heterogeneity in multinucleated skeletal myofibers
.
Nat. Commun.
11
,
6374
.
Petrilli
,
L. L.
,
Spada
,
F.
,
Palma
,
A.
,
Reggio
,
A.
,
Rosina
,
M.
,
Gargioli
,
C.
,
Castagnoli
,
L.
,
Fuoco
,
C.
and
Cesareni
,
G.
(
2020
).
High-dimensional single-cell quantitative profiling of skeletal muscle cell population dynamics during regeneration
.
Cells
9
,
1723
.
Porpiglia
,
E.
,
Samusik
,
N.
,
Van Ho
,
A. T.
,
Cosgrove
,
B. D.
,
Mai
,
T.
,
Davis
,
K. L.
,
Jager
,
A.
,
Nolan
,
G. P.
,
Bendall
,
S. C.
,
Fantl
,
W. J.
et al. 
(
2017
).
High-resolution myogenic lineage mapping by single-cell mass cytometry
.
Nat. Cell Biol.
19
,
558
-
567
.
Purslow
,
P. P.
(
2020
).
The structure and role of intramuscular connective tissue in muscle function
.
Front. Physiol.
11
,
495
.
Quinn
,
L. S.
,
Ong
,
L. D.
and
Roeder
,
R. A.
(
1990
).
Paracrine control of myoblast proliferation and differentiation by fibroblasts
.
Dev. Biol.
140
,
8
-
19
.
Rando
,
T. A.
and
Blau
,
H. M.
(
1994
).
Primary mouse myoblast purification, characterization, and transplantation for cell-mediated gene therapy
.
J. Cell Biol.
125
,
1275
-
1287
.
Randolph
,
M. E.
and
Pavlath
,
G. K.
(
2015
).
A muscle stem cell for every muscle: variability of satellite cell biology among different muscle groups
.
Front. Aging Neurosci.
7
,
190
.
Reggio
,
A.
,
Rosina
,
M.
,
Krahmer
,
N.
,
Palma
,
A.
,
Petrilli
,
L. L.
,
Maiolatesi
,
G.
,
Massacci
,
G.
,
Salvatori
,
I.
,
Valle
,
C.
,
Testa
,
S.
et al. 
(
2020
).
Metabolic reprogramming of fibro/adipogenic progenitors facilitates muscle regeneration
.
Life Sci. Alliance
3
,
e202000646
.
Relaix
,
F.
and
Zammit
,
P. S.
(
2012
).
Satellite cells are essential for skeletal muscle regeneration: the cell on the edge returns centre stage
.
Development
139
,
2845
-
2856
.
Relaix
,
F.
,
Montarras
,
D.
,
Zaffran
,
S. p.
,
Gayraud-Morel
,
B.
,
Rocancourt
,
D.
,
Tajbakhsh
,
S.
,
Mansouri
,
A.
,
Cumano
,
A.
and
Buckingham
,
M.
(
2005a
).
Pax3 and Pax7 have distinct and overlapping functions in adult muscle progenitor cells
.
J. Cell Biol.
172
,
91
-
102
.
Relaix
,
F.
,
Rocancourt
,
D.
,
Mansouri
,
A.
and
Buckingham
,
M.
(
2005b
).
A Pax3/Pax7-dependent population of skeletal muscle progenitor cells
.
Nature
435
,
948
-
953
.
Richler
,
C.
and
Yaffe
,
D.
(
1970
).
The in vitro cultivation and differentiation capacities of myogenic cell lines
.
Dev. Biol.
23
,
1
-
22
.
Rivers
,
L. E.
,
Young
,
K. M.
,
Rizzi
,
M.
,
Jamen
,
F.
,
Psachoulia
,
K.
,
Wade
,
A.
,
Kessaris
,
N.
and
Richardson
,
W. D.
(
2008
).
PDGFRA/NG2 glia generate myelinating oligodendrocytes and piriform projection neurons in adult mice
.
Nat. Neurosci.
11
,
1392
-
1401
.
Roberts
,
E. W.
,
Deonarine
,
A.
,
Jones
,
J. O.
,
Denton
,
A. E.
,
Feig
,
C.
,
Lyons
,
S. K.
,
Espeli
,
M.
,
Kraman
,
M.
,
McKenna
,
B.
,
Wells
,
R. J.
et al. 
(
2013
).
Depletion of stromal cells expressing fibroblast activation protein-alpha from skeletal muscle and bone marrow results in cachexia and anemia
.
J. Exp. Med.
210
,
1137
-
1151
.
Rocheteau
,
P.
,
Gayraud-Morel
,
B.
,
Siegl-Cachedenier
,
I.
,
Blasco
,
M. A.
and
Tajbakhsh
,
S.
(
2012
).
A subpopulation of adult skeletal muscle stem cells retains all template DNA strands after cell division
.
Cell
148
,
112
-
125
.
Rodgers
,
J. T.
,
King
,
K. Y.
,
Brett
,
J. O.
,
Cromie
,
M. J.
,
Charville
,
G. W.
,
Maguire
,
K. K.
,
Brunson
,
C.
,
Mastey
,
N.
,
Liu
,
L.
,
Tsai
,
C.-R.
et al. 
(
2014
).
mTORC1 controls the adaptive transition of quiescent stem cells from G0 to GAlert
.
Nature
510
,
393
-
396
.
Roesch
,
K.
,
Jadhav
,
A. P.
,
Trimarchi
,
J. M.
,
Stadler
,
M. B.
,
Roska
,
B.
,
Sun
,
B. B.
and
Cepko
,
C. L.
(
2008
).
The transcriptome of retinal Müller glial cells
.
J. Comp. Neurol.
509
,
225
-
238
.
Rubenstein
,
A. B.
,
Smith
,
G. R.
,
Raue
,
U.
,
Begue
,
G.
,
Minchev
,
K.
,
Ruf-Zamojski
,
F.
,
Nair
,
V. D.
,
Wang
,
X.
,
Zhou
,
L.
,
Zaslavsky
,
E.
et al. 
(
2020
).
Single-cell transcriptional profiles in human skeletal muscle
.
Sci. Rep.
10
,
229
.
Sambasivan
,
R.
,
Gayraud-Morel
,
B.
,
Dumas
,
G.
,
Cimper
,
C.
,
Paisant
,
S.
,
Kelly
,
R. G.
and
Tajbakhsh
,
S.
(
2009
).
Distinct regulatory cascades govern extraocular and pharyngeal arch muscle progenitor cell fates
.
Dev. Cell
16
,
810
-
821
.
Sambasivan
,
R.
,
Yao
,
R.
,
Kissenpfennig
,
A.
,
Van Wittenberghe
,
L.
,
Paldi
,
A.
,
Gayraud-Morel
,
B.
,
Guenou
,
H.
,
Malissen
,
B.
,
Tajbakhsh
,
S.
and
Galy
,
A.
(
2011
).
Pax7-expressing satellite cells are indispensable for adult skeletal muscle regeneration
.
Development
138
,
4333
.
Sanes
,
J. R
. (
2004
).
Extracellular matrix
. In
Myology
(ed.
A. G. a. F.-A.
Engel, C.
), pp.
471
-
487
:
New York
:
McGraw -Hill
.
Sasse
,
J.
,
von der Mark
,
H.
,
Kühl
,
U.
,
Dessau
,
W.
and
von der Mark
,
K.
(
1981
).
Origin of collagen types I, III, and V in cultures of avian skeletal muscle
.
Dev. Biol.
83
,
79
-
89
.
Sato
,
K.
,
Li
,
Y.
,
Foster
,
W.
,
Fukushima
,
K.
,
Badlani
,
N.
,
Adachi
,
N.
,
Usas
,
A.
,
Fu
,
F. H.
and
Huard
,
J.
(
2003
).
Improvement of muscle healing through enhancement of muscle regeneration and prevention of fibrosis
.
Muscle Nerve
28
,
365
-
372
.
Scaramozza
,
A.
,
Park
,
D.
,
Kollu
,
S.
,
Beerman
,
I.
,
Sun
,
X.
,
Rossi
,
D. J.
,
Lin
,
C. P.
,
Scadden
,
D. T.
,
Crist
,
C.
and
Brack
,
A. S.
(
2019
).
Lineage tracing reveals a subset of reserve muscle stem cells capable of clonal expansion under stress
.
Cell Stem Cell
24
,
944
-
957.e5
.
Scott
,
R. W.
,
Arostegui
,
M.
,
Schweitzer
,
R.
,
Rossi
,
F. M. V.
and
Underhill
,
T. M.
(
2019
).
Hic1 defines quiescent mesenchymal progenitor subpopulations with distinct functions and fates in skeletal muscle regeneration
.
Cell Stem Cell
25
,
797
-
813.e9
.
Seale
,
P.
,
Sabourin
,
L. A.
,
Girgis-Gabardo
,
A.
,
Mansouri
,
A.
,
Gruss
,
P.
and
Rudnicki
,
M. A.
(
2000
).
Pax7 is required for the specification of myogenic satellite cells
.
Cell
102
,
777
-
786
.
Sefton
,
E. M.
and
Kardon
,
G
. (
2019
).
Connecting muscle development, birth defects, and evolution: An essential role for muscle connective tissue
. In
Organ Development
, pp.
137
-
176
:
Elsevier
.
Serrano
,
A. L.
and
Muñoz-Cánoves
,
P.
(
2010
).
Regulation and dysregulation of fibrosis in skeletal muscle
.
Exp. Cell Res.
316
,
3050
-
3058
.
Serrano
,
A. L.
and
Munoz-Canoves
,
P.
(
2017
).
Fibrosis development in early-onset muscular dystrophies: mechanisms and translational implications
.
Semin. Cell Dev. Biol.
64
,
181
-
190
.
Shefer
,
G.
,
Rauner
,
G.
,
Yablonka-Reuveni
,
Z.
and
Benayahu
,
D.
(
2010
).
Reduced satellite cell numbers and myogenic capacity in aging can be alleviated by endurance exercise
.
PLoS ONE
5
,
e13307
.
Sherwood
,
R. I.
,
Christensen
,
J. L.
,
Weissman
,
I. L.
and
Wagers
,
A. J.
(
2004
).
Determinants of skeletal muscle contributions from circulating cells, bone marrow cells, and hematopoietic stem cells
.
Stem Cells
22
,
1292
-
1304
.
Shinin
,
V.
,
Gayraud-Morel
,
B.
,
Gomès
,
D.
and
Tajbakhsh
,
S.
(
2006
).
Asymmetric division and cosegregation of template DNA strands in adult muscle satellite cells
.
Nature Publishing Group
8
,
677
-
682
.
Snow
,
M. H.
(
1977
).
Myogenic cell formation in regenerating rat skeletal muscle injured by mincing. II. An autoradiographic study
.
Anat. Rec.
188
,
201
-
217
.
Snow
,
M. H.
(
1978
).
An autoradiographic study of satellite cell differentiation into regenerating myotubes following transplantation of muscles in young rats
.
Cell Tissue Res.
186
,
535
-
540
.
Sousa-Victor
,
P.
,
Gutarra
,
S.
,
Garcia-Prat
,
L.
,
Rodriguez-Ubreva
,
J.
,
Ortet
,
L.
,
Ruiz-Bonilla
,
V.
,
Jardi
,
M.
,
Ballestar
,
E.
,
Gonzalez
,
S.
,
Serrano
,
A. L.
et al. 
(
2014
).
Geriatric muscle stem cells switch reversible quiescence into senescence
.
Nature
506
,
316
-
321
.
Stuelsatz
,
P.
,
Shearer
,
A.
,
Li
,
Y.
,
Muir
,
L. A.
,
Ieronimakis
,
N.
,
Shen
,
Q. W.
,
Kirillova
,
I.
and
Yablonka-Reuveni
,
Z.
(
2015
).
Extraocular muscle satellite cells are high performance myo-engines retaining efficient regenerative capacity in dystrophin deficiency
.
Dev. Biol.
397
,
31
-
44
.
Stumm
,
J.
,
Vallecillo-García
,
P.
,
Vom Hofe-Schneider
,
S.
,
Ollitrault
,
D.
,
Schrewe
,
H.
,
Economides
,
A. N.
,
Marazzi
,
G.
,
Sassoon
,
D. A.
and
Stricker
,
S.
(
2018
).
Odd skipped-related 1 (Osr1) identifies muscle-interstitial fibro-adipogenic progenitors (FAPs) activated by acute injury
.
Stem Cell Research
32
,
8
-
16
.
Theret
,
M.
,
Rossi
,
F. M. V.
and
Contreras
,
O.
(
2021
).
Evolving roles of muscle-resident fibro-adipogenic progenitors in health, regeneration, neuromuscular disorders, and aging
.
Front. Physiol.
12
,
673404
.
Uezumi
,
A.
,
Fukada
,
S.-i.
,
Yamamoto
,
N.
,
Takeda
,
S. i.
and
Tsuchida
,
K.
(
2010
).
Mesenchymal progenitors distinct from satellite cells contribute to ectopic fat cell formation in skeletal muscle
.
Nat. Cell Biol.
12
,
143
-
152
.
Uezumi
,
A.
,
Ikemoto-Uezumi
,
M.
and
Tsuchida
,
K.
(
2014
).
Roles of nonmyogenic mesenchymal progenitors in pathogenesis and regeneration of skeletal muscle
.
Front Physiol
5
,
68
.
Uezumi
,
A.
,
Ikemoto-Uezumi
,
M.
,
Zhou
,
H.
,
Kurosawa
,
T.
,
Yoshimoto
,
Y.
,
Nakatani
,
M.
,
Hitachi
,
K.
,
Yamaguchi
,
H.
,
Wakatsuki
,
S.
,
Araki
,
T.
et al. 
(
2021
).
Mesenchymal Bmp3b expression maintains skeletal muscle integrity and decreases in age-related sarcopenia
.
J. Clin. Invest.
131
,
e139617
.
Uezumi
,
A.
,
Ito
,
T.
,
Morikawa
,
D.
,
Shimizu
,
N.
,
Yoneda
,
T.
,
Segawa
,
M.
,
Yamaguchi
,
M.
,
Ogawa
,
R.
,
Matev
,
M. M.
,
Miyagoe-Suzuki
,
Y.
et al. 
(
2011
).
Fibrosis and adipogenesis originate from a common mesenchymal progenitor in skeletal muscle
.
J. Cell Sci.
124
,
3654
-
3664
.
Vallecillo-García
,
P.
,
Orgeur
,
M.
,
Vom Hofe-Schneider
,
S.
,
Stumm
,
J.
,
Kappert
,
V.
,
Ibrahim
,
D. M.
,
Börno
,
S. T.
,
Hayashi
,
S.
,
Relaix
,
F.
,
Hildebrandt
,
K.
et al. 
(
2017
).
Odd skipped-related 1 identifies a population of embryonic fibro-adipogenic progenitors regulating myogenesis during limb development
.
Nat. Commun.
8
,
1218
.
van den Brink
,
S. C.
,
Sage
,
F.
,
Vértesy
,
Á.
,
Spanjaard
,
B.
,
Peterson-Maduro
,
J.
,
Baron
,
C. S.
,
Robin
,
C.
and
van Oudenaarden
,
A.
(
2017
).
Single-cell sequencing reveals dissociation-induced gene expression in tissue subpopulations
.
Nat. Methods
14
,
935
-
936
.
Verma
,
M.
,
Asakura
,
Y.
,
Murakonda
,
B. S. R.
,
Pengo
,
T.
,
Latroche
,
C.
,
Chazaud
,
B.
,
McLoon
,
L. K.
and
Asakura
,
A.
(
2018
).
Muscle satellite cell cross-talk with a vascular niche maintains quiescence via VEGF and notch signaling
.
Cell Stem Cell
23
,
530
-
543.e9
.
Warren
,
G. L.
,
Summan
,
M.
,
Gao
,
X.
,
Chapman
,
R.
,
Hulderman
,
T.
and
Simeonova
,
P. P.
(
2007
).
Mechanisms of skeletal muscle injury and repair revealed by gene expression studies in mouse models
.
J. Physiol.
582
,
825
-
841
.
Watt
,
D. J.
,
Lambert
,
K.
,
Morgan
,
J. E.
,
Partridge
,
T. A.
and
Sloper
,
J. C.
(
1982
).
Incorporation of donor muscle precursor cells into an area of muscle regeneration in the host mouse
.
J. Neurol. Sci.
57
,
319
-
331
.
Wosczyna
,
M. N.
and
Rando
,
T. A.
(
2018
).
A muscle stem cell support group: coordinated cellular responses in muscle regeneration
.
Dev. Cell
46
,
135
-
143
.
Wosczyna
,
M. N.
,
Biswas
,
A. A.
,
Cogswell
,
C. A.
and
Goldhamer
,
D. J.
(
2012
).
Multipotent progenitors resident in the skeletal muscle interstitium exhibit robust BMP-dependent osteogenic activity and mediate heterotopic ossification
.
J. Bone Miner. Res.
27
,
1004
-
1017
.
Wosczyna
,
M. N.
,
Konishi
,
C. T.
,
Carbajal
,
E. E. P.
,
Wang
,
T. T.
,
Walsh
,
R. A.
,
Gan
,
Q.
,
Wagner
,
M. W.
and
Rando
,
T. A.
(
2019
).
Mesenchymal stromal cells are required for regeneration and homeostatic maintenance of skeletal muscle
.
CellReports
27
,
2029
-
2035.e5
.
Wosczyna
,
M. N.
,
Perez Carbajal
,
E. E.
,
Wagner
,
M. W.
,
Paredes
,
S.
,
Konishi
,
C. T.
,
Liu
,
L.
,
Wang
,
T. T.
,
Walsh
,
R. A.
,
Gan
,
Q.
,
Morrissey
,
C. S.
et al. 
(
2021
).
Targeting microRNA-mediated gene repression limits adipogenic conversion of skeletal muscle mesenchymal stromal cells
.
Cell Stem Cell
28
,
1323
-
1334.e8
.
Yablonka-Reuveni
,
Z.
and
Rivera
,
A. J.
(
1994
).
Temporal expression of regulatory and structural muscle proteins during myogenesis of satellite cells on isolated adult rat fibers
.
Dev. Biol.
164
,
588
-
603
.
Yartseva
,
V.
,
Goldstein
,
L. D.
,
Rodman
,
J.
,
Kates
,
L.
,
Chen
,
M. Z.
,
Chen
,
Y.-J. J.
,
Foreman
,
O.
,
Siebel
,
C. W.
,
Modrusan
,
Z.
,
Peterson
,
A. S.
et al. 
(
2020
).
Heterogeneity of satellite cells implicates DELTA1/ NOTCH2 signaling in self-renewal
.
CellReports
30
,
1491
-
1503.e6
.
Yoshida
,
N.
,
Yoshida
,
S.
,
Koishi
,
K.
,
Masuda
,
K.
and
Nabeshima
,
Y.
(
1998
).
Cell heterogeneity upon myogenic differentiation: down-regulation of MyoD and Myf-5 generates 'reserve cells
’.
J. Cell Sci.
111
,
769
-
779
.
Yoshioka
,
K.
,
Nagahisa
,
H.
,
Miura
,
F.
,
Araki
,
H.
,
Kamei
,
Y.
,
Kitajima
,
Y.
,
Seko
,
D.
,
Nogami
,
J.
,
Tsuchiya
,
Y.
,
Okazaki
,
N.
et al. 
(
2020
).
Hoxa10 mediates positional memory to govern stem cell function in adult skeletal muscle
.
Sci. Adv
7
,
eabd7924
.
Zammit
,
P. S.
,
Golding
,
J. P.
,
Nagata
,
Y.
,
Hudon
,
V. r.
,
Partridge
,
T. A.
and
Beauchamp
,
J. R.
(
2004
).
Muscle satellite cells adopt divergent fates: a mechanism for self-renewal?
J. Cell Biol.
166
,
347
-
357
.
Zheng
,
G. X. Y.
,
Terry
,
J. M.
,
Belgrader
,
P.
,
Ryvkin
,
P.
,
Bent
,
Z. W.
,
Wilson
,
R.
,
Ziraldo
,
S. B.
,
Wheeler
,
T. D.
,
McDermott
,
G. P.
,
Zhu
,
J.
et al. 
(
2019
).
Massively parallel digital transcriptional profiling of single cells
.
Nat. Commun.
8
,
14049
.

Competing interests

The authors declare no competing or financial interests.

Supplementary information