Keywords: extracellular matrix (ECM), fibro-adipogenic progenitors (FAPs), fibrosis, skeletal muscle, stem cells
Abstract
Fibro-adipogenic progenitors (FAPs) are key regulators of skeletal muscle regeneration and homeostasis. However, dysregulation of these cells leads to fibro-fatty infiltration across various muscle diseases. FAPs are the key source of extracellular matrix (ECM) deposition in muscle, and disruption to this process leads to a pathological accumulation of ECM, known as fibrosis. The replacement of contractile tissue with fibrotic ECM functionally impairs the muscle and increases muscle stiffness. FAPs and fibrotic muscle form a progressively degenerative feedback loop where, as a muscle becomes fibrotic, it induces a fibrotic FAP phenotype leading to further development of fibrosis. In this review, we summarize FAPs’ role in fibrosis in terms of their activation, heterogeneity, contributions to fibrotic degeneration, and role across musculoskeletal diseases. We also discuss current research on potential therapeutic avenues to attenuate fibrosis by targeting FAPs.
INTRODUCTION
Fibrosis is the pathological accumulation of extracellular matrix (ECM) components, predominantly fibrillar collagens, which impair tissue function (1, 2). Fibrosis is a consequence of a multitude of diseases across tissues where the regenerative processes are impaired (3–7). In skeletal muscle, the excessive accumulation of ECM in fibrosis takes the place of functional, contractile tissue, weakening the muscle and increasing muscle passive stiffness (8, 9). The weakened muscle becomes more prone to injury leading to further degeneration and fibrosis as the muscle becomes chronically injured.
The main cell driver of fibrosis in skeletal muscle is fibro-adipogenic progenitors (FAPs). FAPs are muscle-resident nonmyogenic cells that reside in the interstitial space (10). FAPs play an essential role in supporting regeneration. In uninjured muscle, FAPs exist at relatively low levels but rapidly proliferate after injury (11, 12). After rapid expansion in response to injury, FAPs deposit ECM components to replace the damaged matrix and release promyogenic factors to induce muscle differentiation (13, 14). FAPs then rapidly undergo apoptosis, returning to preinjury levels, which halts ECM deposition and allows for full regeneration of the muscle (12, 15). FAPs become resistant to apoptosis in fibrotic conditions, remaining at chronically high levels and continuously depositing ECM resulting in fibrosis (11, 12, 16). The development of fibrosis triggers FAPs to develop a fibrotic phenotype leading to a progressively degenerative feedback loop. As FAPs become activated into a fibrotic phenotype, they contribute to fibrotic ECM deposition and aberrant cell signaling, leading to further fibrotic activation and degeneration. Research on FAPs has rapidly expanded in the past couple of years, dramatically increasing our understanding. However, the mechanosensitivity of FAPs has only recently been defined and requires further research to understand the interplay between FAPs and the fibrotic ECM. The goal of this review is to highlight the positive feedback loop of FAPs and fibrosis; what contributes to this cyclic activation; and methods to disrupt the feedback loop to stop the progression of fibrosis.
ACTIVATION AND PERSISTENCE OF FIBROTIC PHENOTYPE
FAPs were first defined by their ability to spontaneously differentiate into adipocytes and activate into myofibroblasts (10). Notably, there is considerable overlap between FAPs and what may also be referred to as a muscle-resident fibroblast (17). Activation of myofibroblasts is often considered a marker of fibrosis with alpha-smooth muscle action (αSMA), a myofibroblast marker, significantly increasing in fibrotic conditions and strongly correlating with increased ECM deposition (18–23). A series of soluble factors and mechanical signaling from the ECM trigger FAP activation in myofibroblasts. These activation signals create a progressively degenerative feedback loop; as FAPs become activated into myofibroblasts, ECM deposition and release of profibrotic signals increase leading to additional induction of myofibroblast activation and fibrosis (Fig. 1).
Figure 1.
The profibrotic positive feedback loop between fibro-adipogenic progenitors (FAPs) and fibrotic muscle. As FAPs are activated into myofibroblasts, they produce fibrotic levels of extracellular matrix (ECM) leading to increased stiffness, which in turn activates FAPs further into myofibroblasts. Transforming growth factor-beta (TGF-β) contributes to myofibroblast activation and ECM production, furthering the fibrotic loop. Graph axis are stress (σ) and strain (ε) indicating the stiffness of a material. Pink line indicates the stiffness of healthy ECM, and red line indicates the stiffness of fibrotic ECM. Created with BioRender.com.
Soluble Signaling to FAPs
Various soluble factors induce FAP activation in myofibroblasts. The most established one being transforming growth factor-beta (TGF-β). In skeletal muscle, TGF-β is released by both FAPs and macrophages, which play a key role in modulating FAP behavior through soluble factors (24–26). TGF-β is widely regarded as a strong profibrotic signal, with its expression increased across fibrotic diseases (27). Exposure to TGF-β induces myofibroblast activation and is strongly correlated with fibrotic ECM deposition (12, 16, 27, 28). Other soluble factors, downstream of TGF-β, increase myofibroblast activation, further implicating TGF-β has a strong profibrotic factor. Connective tissue growth factor, also known as cellular communication network factor-2 (CTGF/CCN-2), downstream targets of TGF-β, increases αSMA when overexpressed in wildtype (wt) mice (29). In a rat model for an overuse fibrotic injury, TGF-β, αSMA, and CCN2 expression were all increased (30). Myostatin, which signals through the same SMAD pathway as TGF-β, increases FAP proliferation and αSMA expression (23). FAPs produce bone morphogenetic protein 1 (BMP1) and matrix metalloproteinase 14 (MMP14) that activate latent TGF-β into an active form (24). This leads to a feedforward loop as activated FAPs lead to the activation of TGF-β and therefore, more FAP activation.
TGF-β is involved in activating the Hedgehog (Hh) signaling pathway. This signaling pathway is known to block FAP differentiation into adipocytes after injury and enhance regeneration (31). However, ectopic activation of the Hh pathway leads to increased activation of FAPs into myofibroblasts, increased collagen deposition, and impaired regeneration (32). As with TGF-β, Hh signaling plays a key role in promoting regeneration in muscle, however, excessive and sustained expression leads to overactivation of FAPs and initiates fibrotic degeneration.
Platelet-derived growth factor receptor A (PDGFRα) is a tyrosine kinase receptor that is used as the canonical marker for FAP isolation (13, 21, 33). PDGFRα expression is strongly correlated with collagen expression in FAPs suggesting that the surface marker plays a fibrogenic role. Exposure to PDGF-AA in vitro significantly inhibited FAP adipogenic potential while retaining fibrogenic potential and collagen expression (34, 35). Constitutively active PDGFRα expression in an injured mouse model resulted in increased collagen deposition and fibrosis (35, 36). Exposure to TGF-β decreases PDGFRα expression while increasing αSMA expression, suggesting FAPs lose their canonical marker as they commit to a fibrotic phenotype (19). PDGFRα modulates TGF-β-induced myofibroblast activation, with blockage of PDGFRα decreasing myofibroblast activation and fibrosis, highlighting the cross talk between these two play a key role in regulating FAPs’ activation and fate (19, 37). The persistence of FAPs in fibrosis leads to chronically high expression of PDGFRα that activates FAPs into a fibrotic phenotype, creating a positive feedback loop of fibrosis and fibrotic activation.
Excessive reactive oxygen species (ROS), particularly NADPH oxidase 4 (NOX4), have been implicated in myofibroblast activation in FAPs and other fibroblast cell types across tissues (38, 39). TGF-β induces NOX4 expression across different cell types, indicating it as a downstream target of the profibrotic pathway in FAPs (38–40). In an mdx mouse, a model for skeletal muscle fibrosis, NOX4 expression is significantly higher than in wt mice. In a NOX4-KO mdx mouse model, the number of myofibroblast is decreased suggesting NOX4 plays a role in FAP activation (39). ROS levels are elevated in fibrotic skeletal muscle, therefore leading to cyclic activation of FAPs in an already fibrotic condition (41, 42).
The persistence of a fibrotic phenotype is not only driven by FAP activation into myofibroblasts but also their resistance to apoptosis, resulting in the maintenance of chronically high levels of FAPs. Clearance of FAPs is an essential part of the regeneration and removes them from the profibrotic feedback loop. FAPs undergo apoptosis, mediated by tumor necrosis factor-alpha (TNFα) after rapidly proliferating and depositing ECM postinjury (12). This prevents excessive ECM deposition and keeps FAPs at relatively low levels in nonfibrotic and uninjured (11, 12, 43). TGF-β blocks TNFα-induced apoptosis, resulting in chronically high levels of FAPs and furthered fibrosis (12, 16).
Mechanical Signaling to FAPs
The mechanical properties and organization of the ECM are altered in fibrosis. Excessive ECM deposition leads to increased stiffness, crosslinking, and altered alignment (44–46). Similar to mesenchymal stem cells (MSCs), FAPs are responsive to substrate stiffness (47, 48). Increasing substrate stiffness dramatically increases αSMA expression in FAPs. Stiffness that is physiologically representative of fibrotic ECM has significantly more myofibroblast activation compared with stiffness representative of healthy ECM (48). In aged skeletal muscle, the increase in muscle stiffening leads to increased YAP/TAZ expression in muscle fibroblasts (49). YAP/TAZ are known to mediate TGF-β signaling to promote myofibroblast activation and collagen deposition (3, 4). Therefore, the increase in ECM stiffness due to fibrosis induces FAP activation into myofibroblasts through the YAP/TAZ pathway. As a muscle becomes fibrotic it leads to further myofibroblast activation in FAPs, contributing to the progressively degenerative positive feedback loop.
When cultured in standard conditions, FAPs largely spontaneously activate into myofibroblasts as measured by αSMA stress fibers (10, 13, 19). Given the mechanosensitivity of FAPs, this could be due to the excessive-high stiffness of tissue-cultured plastic. Therefore, culture conditions that are more physiologically representative of the ECM stiffness in vivo better represent FAP activation and behavior. A physiological substrate would thus include an elastic stiffness on the order of ∼10 kPa of healthy muscle (50). However, mimicking the mechanical environment would also dictate transitioning from standard two-dimensional (2-D) culture systems to three-dimensional (3-D) culture that can influence the response to substrate stiffness (51). Furthermore, MSCs respond not only to elastic stiffness but also viscoelasticity (52). The specific impact of these more complex mechanical features on FAPs is still currently unknown.
Apoptosis provides a mechanism for FAPs to exit the profibrotic feedback loop and restore homeostatic conditions in skeletal muscle. However, profibrotic signaling, such as TGF-β and increased stiffness, block this exit pathway leading to a cyclic process of profibrotic activation, further fibrotic signaling and activation, and fibrotic development. This profibrotic feedback loop leads to continuous fibrotic activation of FAPs and the progressively degenerative nature of fibrosis.
HETEROGENEITY WITHIN FAPs
FAPs overall are one of the main cell drivers of fibrosis. Recent studies have identified key subpopulations in FAPs that affect their fibrotic and regenerative capacities through single-cell RNA sequencing (scRNAseq). Studies have divided these subpopulations by different markers, but overall, there is significant heterogeneity within FAPs that is altered in injury and disease.
Malecova et al. identified distinct subpopulations of FAPs that changed in injury and fibrosis. These subpopulations were defined by the relative expression of two surface markers, vascular cell adhesion molecule 1 (Vcam1) and tyrosine kinase receptor (Tie2), that have unique transcriptional profiles. Vcam1 expression was significantly increased after acute injury and in mdx mice, but relatively low in wt mice. Tie2 expression levels were the opposite, most FAPs expressed some level of Tie2 in healthy muscle and those levels significantly decreased after injury and in fibrosis (11). High expression of Vcam1+ FAPs was observed in other fibrotic conditions. In oculopharyngeal muscular dystrophy (OPMD), a genetic disorder affecting the muscles in the eyelids and pharynx, there was a higher abundance of Vcam1+ FAPs in the fibrotic muscle compared with nonfibrotic muscle (43). Human FAPs isolated from nonfibrotic muscles clustered differently from those isolated from fibrotic muscles, suggesting changes in gene expression in injury and disease (43). Vcam1 levels are upregulated in other tissue-type diseases and associated with increased proliferation including in idiopathic pulmonary fibrosis and lung cancer (5, 53). In lung fibroblasts, TGFβ significantly increased Vcam1 expression and fibroblast proliferation (5). If TGFβ triggers Vcam1 expression in FAPs as well, it further implicates this subpopulation as the more fibrotic phenotype.
Type 2 diabetes (T2D) is a disease that is associated with fibro-fatty infiltration of skeletal muscle, with contractile tissue being replaced by both excessive ECM deposition and adipocytes (34). Farup et al. analyzed human FAPs isolated from patients with T2D clustered into four distinct subpopulations. A population high in αSMA expression, also differentially expressed collagens and other ECM proteins, indicating that the myofibroblast population of FAPs is primarily responsible for ECM deposition (6, 34). FAPs from patients with T2D clustered based on CD90+/− expression, with genes upregulated in CD90+ FAPs were also upregulated in T2D muscle. CD90+ FAPs had higher proliferative capacity and clonal formation, suggesting a progenitor phenotype while CD90− FAPs expressed a more adipogenic phenotype. CD90 positive and negative FAPs had distinct transcriptomes. CD90− FAPs were enriched for angiogenesis and regulation of endothelial proliferation whereas CD90+ subset was enriched for ECM proteins, collagen synthesis, and TGFβ response, suggesting this subset is a more fibrotic phenotype. Patients with T2D had a significantly higher population of CD90+ FAPs, exclusively, implying this population is primarily responsible for the fibrotic degeneration seen in T2D (34).
Davies et al. (54) divided murine FAPs into subpopulations based on their expression of uncoupling protein 1 (UCP1), a precursor marker for beige adipose. UCP1+ FAPs released promyogenic exosomes whereas UCP1− FAPs were defined by profibrotic exosomes. FAPs isolated from humans with rotator cuff tears, an injury associated with fatty infiltration and fibrosis, clustered into six distinct subpopulations. These distinct subpopulations had differential expression of fibrogenic and adipogenic genes including COL1A1, PDGRFA, ADIPOQ, and UCP1 (54).
In cells isolated from patients with limb-girdle muscular dystrophy (LGMD), Depuydt et al. divided FAPS based on lumican (LUM+) and proteoglycan 4 (PRG4+) expression. LUM+ expression positively correlated with disease severity (55). In nondiseased human muscle, Rubenstein et al. also found FAPs to cluster based on LUM+ and PRG4+ markers. After using mouse samples to validate the human sequencing, however, FAPs were divided into two subpopulations based on lumican (LUM+) and fibrillin 1 (FBN1+) expression, highlighting the heterogeneity between species. Both of these subpopulations had high expression of fibrillar collagens I and III, the predominant proteins in fibrosis, but LUM+ FAPs differentially expressed types IV and XV collagens whereas FBN1+ FAPs had heightened expression of collage XIV.
Several studies have looked at FAP heterogeneity in the context of regeneration. Although these do not give direct insights into FAP subpopulations in fibrosis, the subpopulations of FAPs responsible for ECM deposition and TGF-β signaling in regeneration likely share characteristics with FAPs that persist in fibrosis (56, 57). Scott et al. (58) classified FAPs into two subpopulations that differentially expressed genes for ECM components and cell signaling pathways. The temporal expression of these genes after injury with notexin (NTX) suggests a subset of the FAP population is primarily responsible for ECM deposition after injury. This same subpopulation, defined by ECM expression, may become persistent in chronic injury and disease leading to fibrosis. Oprescu et al. identified subpopulations of murine FAPs that are dynamically altered in homeostasis and cardiotoxin (CTX)-induced injury. After muscle damage, FAPs diverged into subpopulations, the markers of which are time-dependent. At 21 days postinjury (DPI), FAPs could be divided into Osr1+ FAPs and fibroblasts enriched with collagen-I, which might represent the more persistent, fibrotic subset of FAPs (57). Osr1+ is associated with pro-regenerative signaling in FAPs and is upregulated in FAPs after acute injury (14, 15). Leinroth et al. described six distinct subpopulations of FAPs with varying adipogenic potential and ability to respond to BaCl2-induced injury. Osr1+ defined one subpopulation that expressed genes related to stemness and development, indicating a precursor population (15, 59). Two clusters defined by Adam12 and Gap43 had enhanced enrichment of fibrotic genes including TGFβ and Fibronectin 1 (Fn1) and downregulation of Ltbp4, a negative regulator of TGF-β (59).
FAPs are the primary contributors of fibrotic ECM deposition and play essential roles in regeneration and homeostasis. Although ablating FAPs reduces excessive ECM deposition, it results in muscle atrophy and muscle satellite cell (MuSC) depletion, even under homeostatic conditions (60). Recent illumination of FAP subpopulations implies an opportunity for more targeted approaches to reduce fibrosis. Inhibiting a profibrotic subpopulation of FAPs while leaving a pro-regenerative subpopulation intact offers an avenue to maintain the supportive role of FAPs while attenuating fibrosis. However, there seems to be little consensus about how to define these subpopulations in both the context of regeneration and fibrosis. This is likely due to biological and technical variability that exists across samples, disease models, and analysis techniques. Despite different markers, these subpopulations share similarities across studies. For example, in healthy mice, Rubenstein et al. (61) defined a subpopulation by FBN1+ expression, notably, the FBN1+ subset coexpressed TEK, the gene for Tie2, the defining marker for a subpopulation in the Malecova et al. (11) study. Although αSMA is the canonical marker of myofibroblasts it does not have a distinguishing factor in profibrotic FAPs at the transcriptional level across studies in mice and humans (Table 1). Although the identifying markers can shift, a consistent feature of profibrotic FAPs is increased expression of fibrillar collagens I and III as well as collagen crosslinking enzymes.
Table 1.
Summary of FAP heterogeneity and potential markers for profibrotic populations, determined by high fibrillar collagen expression and relative expression in fibrotic conditions
Profibrotic Subpopulation(s) | Other Subpopulation(s) | Sample | Condition | Ref. |
---|---|---|---|---|
Vcam1+ | Tie2+ | Mouse | Fibrosis (mdx) | Malecova et al. (11) |
UCP1− | UCP1+ | Mouse | Rotator cuff tear | Davies et al. (54) |
CD90+ | CD90− | Human | T2D | Farup et al. (34) |
Lumican+ | Proteoglycan 4+ | Human | LGMD | Depuydt et al. (55) |
Lumican+Fibrillin 1+ | Mouse, human | Healthy, resting | Rubenstein et al. (61) | |
Dlk1+ | Dpp4+ | Mouse | CTX-injury | Oprescu et al. (57) |
Fibroblasts | Cxcl14+ | |||
Ors1+ | ||||
Wisp1+ | ||||
FAP1 | FAP2 | Mouse | NTX-injury | Scott et al. (58) |
Gli1 + | Mouse | NTX-injury | Yao et al. (56) | |
Gli1− | ||||
Adam12+ | Osr1+ | Mouse | BaCl2 injury | Leinroth et al. (59) |
Gap43+ | Gli1+ | |||
Hsd11b1+ | ||||
Clu+ | ||||
Osr1+ | Mouse | Freeze injury, glycerol injury | Stumm et al. (15) | |
Osr1− | ||||
MME+ | MME− | Human | HOA | Fitzgerald et al. (62) |
CTX, cardiotoxin; FAP, fibro-adipogenic progenitor; LGMD, limb-girdle muscular dystrophy; NTX, notexin; T2D, type 2 diabetes.
CONTRIBUTIONS TO FIBROSIS
In acute injury, FAPs play a key role in supporting healthy regeneration by both depositing ECM to replace the damaged matrix and signaling to MuSCs through the release of promyogenic signals. In a fibrotic state, these pathways get disrupted leading to excessive ECM deposition, fatty infiltration, and aberrant cell signaling. The fibrotic contributions of FAPs lead to further profibrotic activation, continually disrupting the proregenerative pathways and leading to progressively degenerative fibrosis.
ECM Deposition
The defining characteristic of fibrosis is a pathological excess of ECM deposition, which stiffens the muscle and impairs contractile function (2, 7, 43–45, 63). FAPs account for the majority of collagen expression in skeletal muscle both in regenerating injuries and fibrotic conditions (10, 12, 13, 19–21, 31, 64). The number of FAPs strongly correlates with muscle collagen levels (22, 23). This is supported by the increase in the expression of ECM proteins, particularly fibrillar collagens I and III, from FAPs derived from fibrotic conditions (58, 65). Increased synthesis of various proteoglycans, including decorin and biglycan, found within the ECM is seen in both patients with Duchenne muscular dystrophy (DMD) and mdx mice (65, 66). Lumican and fibrillin, proteoglycans upregulated in FAPs are known to accumulate with fibrillar collagens, promote thicker collagen fibril formation, and induce myofibroblast activation through the TGF-β pathway in other fibrotic diseases (67–70). The increase in ECM deposition stiffens the muscle, leading to further myofibroblast activation in FAPs and fibrotic development (45, 48).
The skeletal muscle ECM not only increases in content throughout fibrosis but also the organization of the ECM and collagen fibrils is altered. Collagen crosslinking and alignment are increased in fibrotic muscle, indicating a change in how FAPs deposit and organize the ECM (44, 71). FAPs express high levels of not only ECM genes but also regulatory genes that control ECM degradation and organization. FAPs have increased expression of lysyl oxidases (LOXs), which promote collagen crosslinking (65, 72, 73). This makes the ECM stiffer and resistant to degradation, contributing to the continuous buildup of ECM and stiffening of the muscle (44, 45, 71). Fibrotic FAPs express CD147, an inducer of matrix metalloproteinases (MMPs), at higher levels than nonfibrotic FAPs (43, 74, 75). MMPs are a large class of proteinases, many of which degrade ECM and are considered antifibrotic. However, MMPs affect a large range of biological processes, and some have been identified as having profibrotic properties. Particularly mdx FAPs had increased expression of MMP2 and MMP14, both of which are upregulated in the presence of TGF-β and have been implicated in the furthering of fibrosis (76–78). Although it may be counterintuitive that MMPs that breakdown ECM are increased in fibrosis, the expression of tissue inhibitors of MMPs (TIMPs) are also overexpressed in fibrotic conditions that shift the balance toward ECM accumulation (79).
FAPs contribution to the fibrotic ECM perpetuates the positive feedback loop of FAPs and fibrosis. FAPs are sensitive to both the ECM stiffness and architecture, inducing myofibroblast activation (48). Fibrotic FAPs have altered the expression of regulatory ECM genes affecting ECM organization and remodeling. As FAPs produce a more fibrotic ECM in terms of stiffness, content, and architecture, it induces further myofibroblast activation in the FAPs leading to more fibrotic ECM deposition. This cycle continues leading to continuing stiffer and more disorganized ECM as fibrosis progresses.
Fatty Infiltration
Fibrosis is, by definition, a pathological accumulation of ECM, which is a result of FAPs profibrotic activation into myofibroblasts. However, the dysregulation of FAPs that occurs in fibrotic diseases, often results in fatty infiltration of the muscle too, as FAPs both differentiate into adipocytes and activate into myofibroblasts, highlighting the multipotency of these cells. The persistently high levels of FAPs due to their apoptotic resistance during fibrosis can lead to high levels of both fibroblastic and adipogenic activation.
Fibro-fatty infiltration is seen in various chronic injuries including rotator cuff tears and DMD (80–82). MRI assessment of skeletal muscles reveals a high-fat fraction in DMD, predominantly in the hip and thigh muscles (83, 84). The amount of fatty infiltration negatively correlates with muscle function and strength (85). Therefore, FAPs impairment of muscle function in fibrotic diseases is in part through the replacement of contractile tissue with not only ECM but also fatty connective tissue.
FAP activation into myofibroblasts and differentiation into adipocytes are often viewed as opposing sides of a coin with profibrotic factors, such as TGF-β, inhibiting adipogenesis whereas promoting fibrogenesis and proadipogenic factors having the opposite effect (86, 87). However, the existence of fibro-fatty infiltration across various skeletal muscle diseases suggests a dual capacity of FAPs to activate in both directions under the same conditions. This is attributed in part to the heterogeneity of FAPs, with certain subsets of FAPs accounting for almost all adipogenic differentiation in muscle (54, 62). The dysregulation of signaling in fibrotic conditions may induce subsets of FAPs to undertake a fibrotic or adipogenic phenotype.
Aberrant Cell Signaling
The altered ECM provides a nondirect way in which FAPs signal to surrounding muscle cells. MuSCs are sensitive to their mechanical and architectural environment and therefore, are affected by the FAP-produced ECM (88). The increased collagen cross-linking and stiffness, seen in fibrosis, impair MuSC differentiation (50, 88, 89). Since the fibrotic response and the regenerative response are opposing pathways to dealing with damage, the inhibition of myogenesis propagates the progressive nature of fibrosis.
FAPs also signal to MuSCs through the release of soluble factors. In a regenerative state, FAPs release promyogenic exosomes that promote MuSC migration to the injury site and differentiation into muscle (54, 56). In fibrosis, these signaling pathways are disrupted, with FAPs impairing rather than promoting myogenesis (90, 91). MuSCs cultured in conditioned media obtained from fibrotic myofibroblasts in skeletal muscle had declined proliferation and increased apoptosis, resulting in poor wound healing (37). Coculturing of FAPs isolated from fibrotic conditions with MuSCs had a negative effect on fusion index compared with coculture of nonfibrotic FAPs, both in direct coculture and in indirect transwell experiments, indicating at least a portion of the negative effect comes from FAP soluble factor release rather than direct cell contact (14, 43).
Fibrotic degeneration is a complex process involving various cell types, but FAPs play a main role in promoting deterioration through multiple pathways. The proregenerative nature of FAPs is switched to a degenerative phenotype of fibro-fatty infiltration and impaired myogenesis advancing fibrotic development across skeletal muscle diseases. FAPs also receive signaling from MuSCs, differentiated muscle fibers play a role in determining FAPs’ adipogenic or fibrogenic fate (92, 93). As FAPs’ signaling to MuSCs becomes dysregulated in fibrosis so does MuSCs signaling to FAPs as a result. Therefore, the cyclic nature of fibrosis continues leading to fibrotic activation of FAPs and aberrant cell signaling.
FAPs IN FIBROTIC CONDITIONS
Fibrosis is a consequence of nearly all skeletal muscle diseases and chronic injuries. Although the causes of the disease may differ, a common feature is the persistent presence of FAPs and their contributions to fibrosis.
Muscular Dystrophies
One of the more commonly studied groups of fibrotic diseases in skeletal muscles is muscular dystrophies, a group of genetic diseases that progressively weaken muscles. The most studied one being DMD. DMD is an x-linked recessive disorder resulting from a mutation in the dystrophin gene, a structural protein that provides mechanical stability and strength to the muscle. The muscles in DMD are more prone to injury without a fully functioning dystrophin protein and are significantly weaker leading to chronic damage and fibrosis. FAPs in DMD muscle are more highly activated into myofibroblasts and exist in larger numbers than in nondiseased muscle driving the progressively degenerative fibrosis (94). Profibrotic markers are enhanced in DMD FAPs including TGF-β, αSMA, and fibrillar collagens (11, 34). Profibrotic miRNAs are elevated in FAPs isolated from DMD versus control muscle. These profibrotic miRNAs strongly correlate with TGF-β and ECM protein expression levels (94).
Limb-girdle muscular dystrophy (LGMD) is a disease caused by a mutation in the dysferlin gene, impairing muscle repair and leading to chronic injury. FAPs in LGMD, specifically the subpopulation expressing lumican (LUM), progressively increase with disease severity (55, 95). LGMD muscle had upregulation of genes related to fibro-fatty infiltration whereas genes related to protein synthesis and regeneration were downregulated (55). Particularly, FAPs contribute significantly to adipogenic replacement of contractile tissue in LGMD, impairing muscle function and furthering disease progression (95). FAPs isolated from patients with OPMD have the significant increased proliferative capacity and were more motile compared with control FAPs. OPMD FAPs have increased the expression of ECM proteins and altered the expression of genes related to cell signaling, which resulted in an impaired fusion index of MuSCs (43).
Skeletal Muscle Diseases and Chronic Injuries
Skeletal muscle fibrosis is most commonly studied in the context of muscular dystrophies, however, almost any chronic skeletal muscle injury or disease results in some level of fibrosis. Across these diseases, FAP levels are elevated and highly activated leading to increased ECM deposition and fibrosis. In cerebral palsy, a group of motor disorders, collagen expression is elevated in FAPs compared with typically developing controls (22, 46, 96). In a mouse model for amyotrophic lateral sclerosis (ALS), a motor neuron disease, αSMA expression is increased over wild-type controls, indicating enhanced FAP activation into myofibroblasts (20). Chronic kidney disease (CKD) and kidney dysfunction are strongly correlated with a progressively fibrotic muscle phenotype. This is purportedly due to decreased TNF-α expression leading to a persistently expanding pool of FAPs resulting in the number of FAPs nearly doubling in CKD muscle (8), or increased myostatin in CKD stimulating the differentiation of FAPs to myofibroblasts (23). In a mouse model for CKD, αSMA and ECM protein expressions are elevated (23). T2D is associated with fibro-fatty degeneration of skeletal muscle and increased PDGFRα expression. Genes related to ECM turnover and remodeling are differently expressed in FAPs, including increased collagen-I deposition (34).
FAPs are responsible for the high levels of fatty infiltration and fibrotic deposition observed in rotator cuff tears (82, 97). FAP levels are elevated and the apoptotic index is decreased in a mouse model for rotator cuff tears (81). TGF-β levels are increased after rotator cuff injury and increase FAP survival (81, 97). Rotator cuff injuries develop fatty infiltration at a high rate and concentration than other muscles across diseases. This may in part be due to the heterogeneity of FAPs discussed earlier not only within a muscle but across anatomical regions. FAPs from rotator cuff muscles reside at a higher concentration and demonstrate greater adipogenic potential than lower limb muscle FAPs (98).
Skeletal muscle diseases and chronic injuries vary in their cause, symptoms, and development. However, a common factor across diseases is the role FAPs play in the progressively degenerative nature of fibrosis. Therefore, targeting FAPs through various antifibrotic therapies offers an avenue to attenuate a broad range of fibrotic conditions (Fig. 2).
Figure 2.
Signaling pathways involved in fibro-adipogenic progenitor (FAP) fibrotic activation and potential therapeutic targets. Therapeutics to block fibrotic pathways are marked by the red boxes. ECM, extracellular matrix; TGF-β, transforming growth factor-beta; TNFα, tumor necrosis factor alpha Created with BioRender.com.
ATTENUATION OF FIBROSIS THROUGH TARGETING FAPs
The positive feedback loop of FAPs and fibrosis causes muscles to progressively degenerate. To attenuate fibrosis and improve muscle function, this cycle needs to be disrupted otherwise FAPs will continually activate and contribute to fibrosis. Methods to break this cycle include blocking the soluble and mechanical signaling within the cycle as well as promoting apoptosis, to remove FAPs from the fibrotic feedback loop.
Blockage of TGF-β Signaling
TGF-β plays a significant role in driving FAPs’ fibrotic behavior by inducing proliferation, myofibroblast activation, and blocking apoptosis. Methods that inhibit TGF-β signaling to FAPs help to promote apoptosis, restoring a key aspect of the regenerative cycle, and limiting myofibroblast activation and ECM deposition. Small molecule inhibitors of TGF-β reduce FAP number and fibrotic signaling in mice (81, 99). In a mouse model of a rotator cuff injury, SB431542, a small molecule TGF-β inhibitor reduced αSMA expression, collagen deposition, the number of FAP cells, and increased FAPs’ apoptotic index (81). ITD-1, which targets TGF-β receptors, reduced TGF-β activity, FAP accumulation, and fibrotic area in mdx mice (99). Nilotinib, a tyrosine kinase inhibitor, blocks TGF-β signaling, restoring FAP apoptosis and as a result, reducing collagen expression (12, 100).
The benefits of blocking TGF-β to attenuate fibrosis have been widely studied and proven successful in promoting FAP apoptosis and reduction of ECM deposition. However, these techniques need to be applied with caution as total inhibition can have adverse side effects. TGF-β plays a role in many different biological signaling pathways including myogenesis (101). Therefore, complete inhibition could impair regeneration, rendering the reduction in fibrosis functionally inconsequential. Targeting downstream targets of TGF-β specifically involved in myofibroblast activation provides a more specific method to attenuating fibrosis while leaving most broad signaling pathways intact. TGF-β acts on many different signaling pathways, with most fibrotic upregulation occurring through the SMAD2/3 pathway (4, 38, 43). Therefore, blocking downstream targets of TGF-β related to this pathway allows for the inhibition of fibrotic signaling while leaving other important signaling pathways intact. Blocking CTGF/CCN-2, a downstream target of TGF-β/SMAD pathway, reduces αSMA expression and fibrosis in a rat overuse injury model (30). ATA 842, a drug that inhibits the myostatin/SMAD pathway, prevents fibrosis progression, promotes apoptosis, and reduces myofibroblast activation in fibrotic FAPs (23, 102).
Blocking Mechanosensing
The mechanical environment of the muscle is altered in fibrosis with stiffness increasing as fibrosis progresses. FAPs are sensitive to these increases in stiffness, inducing higher myofibroblast activation on increased stiffness (48). Blocking FAPs’ ability to sense fibrotic stiffness provides an avenue to reduce myofibroblast activation in fibrotic muscle by stopping the profibrotic feedback loop between myofibroblast activation, ECM deposition, and stiffness. YAP/TAZ are proteins involved in mechanosensing, translocating to the nucleus on stiffer substrates, and are linked to myofibroblast activation in FAPs and other fibroblast-like cells (3, 48, 103). Inhibiting YAP/TAZ translocation to the nucleus prevents myofibroblast activation on stiffer substrates and therefore, could be applied to fibrotic conditions where the muscles have elevated stiffness (4, 48, 104). Drugs that block YAP translocation to the nucleus have shown preclinical efficacy as antifibrotic drugs including verteporfin (105) and a dopamine receptor agonist (106). However, to our knowledge, YAP inhibitors have not been applied in the context of skeletal muscle tissue.
A stiff mechanical environment, also, inhibits apoptosis of profibrotic myofibroblasts. Despite the presence of apoptotic signals being present, stiffness induces the expression of antiapoptotic protein BCL-XL (107). To combat this pathway and reduce the number of myofibroblasts, strategies to block BCL-XL can be applied. The drug ABT-263 inhibits BCL-XL and has demonstrated antifibrotic activity in the lung (108), liver (109), and heart (110), but similarly has not been applied to skeletal muscle.
Restoring Cell Signaling Pathways
In fibrosis, FAP signaling to MuSCs is disrupted with FAPs releasing inhibitory exosomes rather than promyogenic exosomes. Histone deacetylase inhibitors (HDACis) have emerged as a therapeutic method to tune FAP signaling to a more pro-regenerative state. Extracellular vesicles (EVs) released from both human DMD FAPs and mouse mdx FAPs had altered expressions of miRNAs. HDACis increased the expression of pro-regenerative miR-206 from FAPs which in turn increased differentiation in MuSCs and decreased the amount of collagen in the muscle (90, 111). HDACis also inhibit FAPs adipogenic potential and increase FAP expression of follistatin, a glycoprotein known to inhibit myostatin, therefore blocking myofibroblast activation (112, 113). Givinostat, an HDACi, has been used in clinical trials for DMD with successful reduction of fibrotic and fatty areas (114, 115).
However, experiments in mdx mice showed HDACis were only effective in restoring FAPs’ pro-regenerative signaling in young mice, whereas old mdx FAPs were resistant to HDACi treatment (112). Similarly, antibody treatments against myostatin increased muscle size and specific force, only when administered to young mdx mice, and had no effect on adult mdx mice (102). Fibrosis is largely considered irreversible, although there is some evidence to the contrary (106, 116). Treatments that prevent the development of fibrosis rather than reversing already existing fibrosis tend to be more efficacious. Therefore, targeting FAPs to prevent profibrotic activation before excessive fibrotic development provides the best method for improving muscle function.
PERSPECTIVES AND FUTURE STUDIES
Fibrosis is a progressively degenerative consequence of nearly all skeletal muscle diseases as a result of the profibrotic feedback loop that, not only activates FAPs into a profibrotic state but keeps them there increasing fibrotic contributions (7, 80, 117). Soluble and mechanical signaling contribute to the activation of FAPs and block their exit from the profibrotic feedback loop (12, 16, 48, 118). Investigations into methods to attenuate fibrosis must address ways to disrupt this loop otherwise the cyclic nature of FAPs and fibrosis will continue.
Two interesting avenues to break the profibrotic feedback loop are the relatively new discoveries of FAPs’ mechanosensitivity and heterogeneity. FAPs’ mechanosensitivity has only recently been defined and adds insight into the cyclic nature of fibrotic tissue stiffening and the activation of myofibroblasts (48). However, the signaling pathways that induce myofibroblast activation of FAPs on stiff substrates are not fully defined and further investigation could provide more insight and therapeutic targets for disrupting FAP activation and fibrosis. Methods to block YAP/TAZ or other mechanosensing pathways in vivo have proved effective in other fibrotic tissue (104, 119). Another prospective strategy to break the fibrotic feedback loop would be to target the ECM structure directly. Direct injection of collagenase to digest fibrotic collagen is performed in Dupuytren’s contracture (120) and has been proposed as a method of reducing muscle stiffness in muscle contractures of children with cerebral palsy (121, 122). Blocking collagen crosslinking is a potential target as well to increase the turnover by endogenous collagenase and decrease the stiffness of the collagen network (123). A range of collagen crosslinking inhibitors have been explored in other tissues (124) although a common collagen crosslinking inhibitor was found to be ineffective in blocking crosslinking in skeletal muscle. Methods to disrupt the profibrotic cycle through mechanosensing or altering the mechanics should be further developed in the skeletal muscle environment to determine their efficacy in treating skeletal muscle fibrosis.
The heterogeneity of FAPs in the context of regeneration and fibrosis offers various avenues for future studies to parse out the individual contributions of each subpopulation. Experimentation isolating out individual subpopulations and defining their matrisome and secretome would better define the profibrotic and proregenerative nature of the subpopulations. This could be explored through depletion experiments to ablate a specific subpopulation of FAPs and assess changes in skeletal muscle function and fibrosis in vivo.
CONCLUSIONS
FAPs play an essential role in the regeneration and homeostasis of skeletal muscle through their contributions to the ECM and myogenic signaling. However, in disease, these pathways get disrupted and FAPs quickly take on a pathogenic phenotype. The transient nature of FAPs after injury is essential for maintaining their pro-regenerative role. The persistence of FAPs and resistance to apoptosis leads to the chronic cycle of ECM deposition, fibrosis, and fibrotic activation of FAPs. Recent progress in RNAseq and transcriptomics has allowed insights into the vast heterogeneity of FAPs in healthy and diseased muscle. The importance of these subpopulations is not fully understood but may provide insights into the multidirectional nature of FAPs and more specific targets for antifibrotic therapies.
FAPs and fibrosis contribute to a progressively degenerative positive feedback loop. The development of fibrosis disrupts cell signaling leading to fibrogenic activation of FAPs, furthering fibrosis and, as a result, FAP activation and contributions to fibrosis. Disrupting this feedforward pathway is necessary to attenuate fibrosis and restore muscle function. Targeting the various signaling pathways that induce myofibroblast activation and inhibit apoptosis in FAPs will disrupt the fibrotic feedback loop.
GRANTS
This work was supported by grants from the National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS) R01AR079545 (to L.R. Smith) and 1F31AR082700 (to T. Loomis) and Congressionally Directed Medical Research Programs Grant MD210110 (to L.R. Smith).
DISCLOSURES
No conflicts of interest, financial or otherwise, are declared by the authors.
AUTHOR CONTRIBUTIONS
T.L. prepared figures; T.L. drafted manuscript; T.L. and L.R.S. edited and revised manuscript; T.L. and L.R.S. approved final version of manuscript.
ACKNOWLEDGMENTS
We acknowledge the members of the MyoMatrix Lab whose discussions helped to frame and motivate this review.
REFERENCES
- 1. Seynnes O, Schiaffino S, Blottner D, Csapo R, Gumpenberger M, Wessner B. Skeletal muscle extracellular matrix—what do we know about its composition, regulation, and physiological roles? A narrative review. Front Physiol 11: 253, 2020. doi: 10.3389/fphys.2020.00253. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2. Smith LR, Barton ER, States U. Regulation of fibrosis in muscular dystrophy. Matrix Biol 68–69: 602–615, 2018. doi: 10.1016/j.matbio.2018.01.014.Regulation. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3. He X, Tolosa MF, Zhang T, Goru SK, Severino LU, Misra PS, McEvoy CM, Caldwell L, Szeto SG, Gao F, Chen X, Atin C, Ki V, Vukosa N, Hu C, Zhang J, Yip C, Krizova A, Wrana JL, Yuen DA. Myofibroblast YAP/TAZ activation is a key step in organ fibrogenesis. JCI Insight 7: 22–24, 2022. doi: 10.1172/jci.insight.146243. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4. Szeto SG, Narimatsu M, Lu M, He X, Sidiqi AM, Tolosa MF, Chan L, De Freitas K, Bialik JF, Majumder S, Boo S, Hinz B, Dan Q, Advani A, John R, Wrana JL, Kapus A, Yuen DA. YAP/TAZ are mechanoregulators of TGF-β-Smad signaling and renal fibrogenesis. J Am Soc Nephrol 27: 3117–3128, 2016. doi: 10.1681/ASN.2015050499. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5. Agassandian M, Tedrow JR, Sembrat J, Kass DJ, Zhang Y, Goncharova EA, Kaminski N, Mallampalli RK, Vuga LJ. VCAM-1 is a TGF-β1 inducible gene upregulated in idiopathic pulmonary fibrosis. Cell Signal 27: 2467–2473, 2015. doi: 10.1016/j.cellsig.2015.09.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6. Schuster R, Younesi F, Ezzo M, Hinz B. The role of myofibroblasts in physiological and pathological tissue repair. Cold Spring Harb Perspect Biol 15: a041231, 2023. doi: 10.1101/cshperspect.a041231. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7. Lieber RL, Ward SR. Cellular mechanisms of tissue fibrosis. IV. Structural and functional consequences of skeletal muscle fibrosis. Am J Physiol Cell Physiol 305: C241–C252, 2013. doi: 10.1152/ajpcell.00173.2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8. Abramowitz MK, Paredes W, Zhang K, Brightwell CR, Newsom JN, Kwon HJ, Custodio M, Buttar RS, Farooq H, Zaidi B, Pai R, Pessin JE, Hawkins M, Fry CS. Skeletal muscle fibrosis is associated with decreased muscle inflammation and weakness in patients with chronic kidney disease. Am J Physiol Renal Physiol 315: F1658–F1669, 2018. doi: 10.1152/ajprenal.00314.2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9. Smith LR, Lee KS, Ward SR, Chambers HG, Lieber RL. Hamstring contractures in children with spastic cerebral palsy result from a stiffer extracellular matrix and increased in vivo sarcomere length. J Physiol 589: 2625–2639, 2011. doi: 10.1113/jphysiol.2010.203364. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10. Uezumi A, Ito T, Morikawa D, Shimizu N, Yoneda T, Segawa M, Yamaguchi M, Ogawa R, Matev MM, Miyagoe-Suzuki Y, Takeda S, Tsujikawa K, Tsuchida K, Yamamoto H, Fukada SI. Fibrosis and adipogenesis originate from a common mesenchymal progenitor in skeletal muscle. J Cell Sci 124: 3654–3664, 2011. doi: 10.1242/jcs.086629. [DOI] [PubMed] [Google Scholar]
- 11. Malecova B, Gatto S, Etxaniz U, Passafaro M, Cortez A, Nicoletti C, Giordani L, Torcinaro A, De Bardi M, Bicciato S, De Santa F, Madaro L, Puri PL. Dynamics of cellular states of fibro-adipogenic progenitors during myogenesis and muscular dystrophy. Nat Commun 9: 3670, 2018. doi: 10.1038/s41467-018-06068-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12. Lemos DR, Babaeijandaghi F, Low M, Chang CK, Lee ST, Fiore D, Zhang RH, Natarajan A, Nedospasov SA, Rossi FMV. Nilotinib reduces muscle fibrosis in chronic muscle injury by promoting TNF-mediated apoptosis of fibro/adipogenic progenitors. Nat Med 21: 786–794, 2015. doi: 10.1038/nm.3869. [DOI] [PubMed] [Google Scholar]
- 13. Joe AWB, Yi L, Natarajan A, Le Grand F, So L, Wang J, Rudnicki MA, Rossi FMV. Muscle injury activates resident fibro/adipogenic progenitors that facilitate myogenesis. Nat Cell Biol 12: 153–163, 2010. doi: 10.1038/ncb2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14. Kotsaris G, Qazi TH, Bucher CH, Zahid H, Pöhle-Kronawitter S, Ugorets V, Jarassier W, Börno S, Timmermann B, Giesecke-Thiel C, Economides AN, Le Grand F, Vallecillo-García P, Knaus P, Geissler S, Stricker S. Odd skipped-related 1 controls the pro-regenerative response of fibro-adipogenic progenitors. NPJ Regen Med 8: 19, 2023. doi: 10.1038/s41536-023-00291-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15. Stumm J, Vallecillo-García P, Vom Hofe-Schneider S, Ollitrault D, Schrewe H, Economides AN, Marazzi G, Sassoon DA, Stricker S. Odd skipped-related 1 (Osr1) identifies muscle-interstitial fibro-adipogenic progenitors (FAPs) activated by acute injury. Stem Cell Res 32: 8–16, 2018. doi: 10.1016/j.scr.2018.08.010. [DOI] [PubMed] [Google Scholar]
- 16. Hinz B, Lagares D. Evasion of apoptosis by myofibroblasts: a hallmark of fibrotic diseases. Nat Rev Rheumatol 16: 11–31, 2020. doi: 10.1038/s41584-019-0324-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17. Contreras O, Rossi FM, Brandan E. Adherent muscle connective tissue fibroblasts are phenotypically and biochemically equivalent to stromal fibro/adipogenic progenitors. Matrix Biol Plus 2: 100006, 2019. doi: 10.1016/j.mbplus.2019.04.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18. Contreras O, Rebolledo DL, Oyarzún JE, Olguín HC, Brandan E. Connective tissue cells expressing fibro/adipogenic progenitor markers increase under chronic damage: relevance in fibroblast-myofibroblast differentiation and skeletal muscle fibrosis. Cell Tissue Res 364: 647–660, 2016. doi: 10.1007/s00441-015-2343-0. [DOI] [PubMed] [Google Scholar]
- 19. Contreras O, Cruz-Soca M, Theret M, Soliman H, Tung LW, Groppa E, Rossi FM, Brandan E. Cross-talk between TGF-β and PDGFRα signaling pathways regulates the fate of stromal fibro-adipogenic progenitors. J Cell Sci 132: jcs232157, 2019. doi: 10.1242/jcs.232157. [DOI] [PubMed] [Google Scholar]
- 20. Gonzalez D, Contreras O, Rebolledo DL, Espinoza JP, Van Zundert B, Brandan E. ALS skeletal muscle shows enhanced TGF-β signaling, fibrosis and induction of fibro/adipogenic progenitor markers. PLoS One 12: e0177649, 2017. doi: 10.1371/journal.pone.0177649. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21. Uezumi A, Fukada SI, Yamamoto N, Takeda S, Tsuchida K. Mesenchymal progenitors distinct from satellite cells contribute to ectopic fat cell formation in skeletal muscle. Nat Cell Biol 12: 143–152, 2010. doi: 10.1038/ncb2014. [DOI] [PubMed] [Google Scholar]
- 22. Chapman MA, Mukund K, Subramaniam S, Brenner D, Lieber RL. Three distinct cell populations express extracellular matrix proteins and increase in number during skeletal muscle fibrosis. Am J Physiol Cell Physiol 312: C131–C143, 2017. doi: 10.1152/ajpcell.00226.2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23. Dong J, Dong Y, Chen Z, Mitch WE, Zhang L. The pathway to muscle fibrosis depends on myostatin stimulating the differentiation of fibro/adipogenic progenitor cells in chronic kidney disease. Kidney Int 91: 119–128, 2017. doi: 10.1016/j.kint.2016.07.029. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24. Juban G, Saclier M, Yacoub-Youssef H, Kernou A, Arnold L, Boisson C, Ben Larbi S, Magnan M, Cuvellier S, Théret M, Petrof BJ, Desguerre I, Gondin J, Mounier R, Chazaud B. AMPK activation regulates LTBP4-dependent TGF-β1 secretion by pro-inflammatory macrophages and controls fibrosis in Duchenne muscular dystrophy. Cell Rep 25: 2163–2176.e6, 2018. doi: 10.1016/j.celrep.2018.10.077. [DOI] [PubMed] [Google Scholar]
- 25. Muñoz-Cánoves P, Serrano AL. Macrophages decide between regeneration and fibrosis in muscle. Trends Endocrinol Metab 26: 449–450, 2015. doi: 10.1016/j.tem.2015.07.005. [DOI] [PubMed] [Google Scholar]
- 26. Stepien DM, Hwang C, Marini S, Pagani CA, Sorkin M, Visser ND, Huber AK, Edwards NJ, Loder SJ, Vasquez K, Aguilar CA, Kumar R, Mascharak S, Longaker MT, Li J, Levi B. Tuning macrophage phenotype to mitigate skeletal muscle fibrosis. J Immunol 204: 2203–2215, 2020. doi: 10.4049/jimmunol.1900814. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27. Kim J, Lee J. Role of transforming growth factor-β in muscle damage and regeneration: focused on eccentric muscle contraction. J Exerc Rehabil 13: 621–626, 2017. doi: 10.12965/jer.1735072.536. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28. Mann CJ, Perdiguero E, Kharraz Y, Aguilar S, Pessina P, Serrano AL, Muñoz-Cánoves P. Aberrant repair and fibrosis development in skeletal muscle. Skelet Muscle 1: 21–20, 2011. doi: 10.1186/2044-5040-1-21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29. Morales MG, Cabello-Verrugio C, Santander C, Cabrera D, Goldschmeding R, Brandan E. CTGF/CCN-2 over-expression can directly induce features of skeletal muscle dystrophy. J Pathol 225: 490–501, 2011. doi: 10.1002/path.2952. [DOI] [PubMed] [Google Scholar]
- 30. Barbe MF, Hilliard BA, Amin M, Harris MY, Hobson LJ, Cruz GE, Popoff SN. Blocking CTGF/CCN2 reduces established skeletal muscle fibrosis in a rat model of overuse injury. FASEB J 34: 6554–6569, 2020. doi: 10.1096/fj.202000240RR. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31. Kopinke D, Roberson EC, Reiter JF. Ciliary Hedgehog signaling restricts injury-induced adipogenesis. Cell 170: 340–351.e12, 2017. doi: 10.1016/j.cell.2017.06.035. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32. Norris AM, Appu AB, Johnson CD, Zhou LY, McKellar DW, Renault MA, Hammers D, Cosgrove BD, Kopinke D. Hedgehog signaling via its ligand DHH acts as cell fate determinant during skeletal muscle regeneration. Nat Commun 14: 3766, 2023. doi: 10.1038/s41467-023-39506-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33. Molina T, Fabre P, Dumont NA. Fibro-adipogenic progenitors in skeletal muscle homeostasis, regeneration and diseases. Open Biol 11: 210110, 2021. doi: 10.1098/rsob.210110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34. Farup J, Just J, de Paoli F, Lin L, Jensen JB, Billeskov T, Roman IS, Cömert C, Møller AB, Madaro L, Groppa E, Fred RG, Kampmann U, Gormsen LC, Pedersen SB, Bross P, Stevnsner T, Eldrup N, Pers TH, Rossi FMV, Puri PL, Jessen N. Human skeletal muscle CD90+ fibro-adipogenic progenitors are associated with muscle degeneration in type 2 diabetic patients. Cell Metab 33: 2201–2214.e11, 2021. doi: 10.1016/j.cmet.2021.10.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. Fernández-Simón E, Suárez-Calvet X, Carrasco-Rozas A, Piñol-Jurado P, López-Fernández S, Pons G, Bech Serra JJ, de la Torre C, de Luna N, Gallardo E, Díaz-Manera J. RhoA/ROCK2 signalling is enhanced by PDGF-AA in fibro-adipogenic progenitor cells: implications for Duchenne muscular dystrophy. J Cachexia Sarcopenia Muscle 13: 1373–1384, 2022. doi: 10.1002/jcsm.12923. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36. Ieronimakis N, Hays A, Prasad A, Janebodin K, Duffield JS, Reyes M. PDGFRα signalling promotes fibrogenic responses in collagen-producing cells in Duchenne muscular dystrophy. J Pathol 240: 410–424, 2016. doi: 10.1002/path.4801. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37. Thooyamani AS, Mukhopadhyay A. PDGFRα mediated survival of myofibroblasts inhibit satellite cell proliferation during aberrant regeneration of lacerated skeletal muscle. Sci Rep 11: 63–15, 2021. doi: 10.1038/s41598-020-79771-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38. Barnes JL, Gorin Y. Myofibroblast differentiation during fibrosis: role of NAD(P)H oxidases. Kidney Int 79: 944–956, 2011. doi: 10.1038/ki.2010.516. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39. Hammers DW, Hammers DW. NOX4 inhibition promotes the remodeling of dystrophic muscle NOX4 inhibition promotes the remodeling of dystrophic muscle. JCI Insight 7: e158316, 2022. doi: 10.1172/jci.insight.158316. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40. Carmona-Cuenca I, Roncero C, Sancho P, Caja L, Fausto N, Fernández M, Fabregat I. Upregulation of the NADPH oxidase NOX4 by TGF-β in hepatocytes is required for its pro-apoptotic activity. J Hepatol 49: 965–976, 2008. doi: 10.1016/j.jhep.2008.07.021. [DOI] [PubMed] [Google Scholar]
- 41. Cabello-Verrugio C, Acuña MJ, Morales MG, Becerra A, Simon F, Brandan E. Fibrotic response induced by angiotensin-II requires NAD(P)H oxidase-induced reactive oxygen species (ROS) in skeletal muscle cells. Biochem Biophys Res Commun 410: 665–670, 2011. doi: 10.1016/j.bbrc.2011.06.051. [DOI] [PubMed] [Google Scholar]
- 42. Kozakowska M, Pietraszek-Gremplewicz K, Jozkowicz A, Dulak J. The role of oxidative stress in skeletal muscle injury and regeneration: focus on antioxidant enzymes. J Muscle Res Cell Motil 36: 377–393, 2015. doi: 10.1007/s10974-015-9438-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43. Bensalah M, Muraine L, Boulinguiez A, Giordani L, Albert V, Ythier V, Dhiab J, Oliver A, Hanique V, Gidaro T, Perié S, Lacau St-Guily J, Corneau A, Butler-Browne G, Bigot A, Mouly V, Negroni E, Trollet C. A negative feedback loop between fibroadipogenic progenitors and muscle fibres involving endothelin promotes human muscle fibrosis. J Cachexia Sarcopenia Muscle 13: 1771–1784, 2022. doi: 10.1002/jcsm.12974. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44. Brashear SE, Wohlgemuth RP, Gonzalez G, Smith LR. Passive stiffness of fibrotic skeletal muscle in mdx mice relates to collagen architecture. J Physiol 599: 943–962, 2021. doi: 10.1113/JP280656. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45. Wohlgemuth RP, Feitzinger RM, Henricson KE, Dinh DT, Brashear SE, Smith LR. The extracellular matrix of dystrophic mouse diaphragm accounts for the majority of its passive stiffness and is resistant to collagenase digestion. Matrix Biol Plus 18: 100131, 2023. doi: 10.1016/j.mbplus.2023.100131. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46. Smith LR, Pichika R, Meza RC, Gillies AR, Baliki MN, Chambers HG, Lieber RL. Contribution of extracellular matrix components to the stiffness of skeletal muscle contractures in patients with cerebral palsy. Connect Tissue Res 62: 287–298, 2021. doi: 10.1080/03008207.2019.1694011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47. Engler AJ, Sen S, Sweeney HL, Discher DE. Matrix elasticity directs stem cell lineage specification. Cell 126: 677–689, 2006. doi: 10.1016/j.cell.2006.06.044. [DOI] [PubMed] [Google Scholar]
- 48. Loomis T, Hu LY, Wohlgemuth RP, Chellakudam RR, Muralidharan PD, Smith LR. Matrix stiffness and architecture drive fibro-adipogenic progenitors’ activation into myofibroblasts. Sci Rep 12: 13582, 2022. doi: 10.1038/s41598-022-17852-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49. Stearns-Reider KM, D'Amore A, Beezhold K, Rothrauff B, Cavalli L, Wagner WR, Vorp DA, Tsamis A, Shinde S, Zhang C, Barchowsky A, Rando TA, Tuan RS, Ambrosio F. Aging of the skeletal muscle extracellular matrix drives a stem cell fibrogenic conversion. Aging Cell 16: 518–528, 2017. doi: 10.1111/acel.12578. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50. Engler AJ, Griffin MA, Sen S, Bönnemann CG, Sweeney HL, Discher DE. Myotubes differentiate optimally on substrates with tissue-like stiffness: pathological implications for soft or stiff microenvironments. J Cell Biol 166: 877–887, 2004. doi: 10.1083/jcb.200405004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51. Baker BM, Chen CS. Deconstructing the third dimension-how 3D culture microenvironments alter cellular cues. J Cell Sci 125: 3015–3024, 2012. doi: 10.1242/jcs.079509. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52. Chaudhuri O, Cooper-White J, Janmey PA, Mooney DJ, Shenoy VB. Effects of extracellular matrix viscoelasticity on cellular behaviour. Nature 584: 535–546, 2020. doi: 10.1038/s41586-020-2612-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53. Zhou Z, Zhou Q, Wu X, Xu S, Hu X, Tao X, Li B, Peng J, Li D, Shen L, Cao Y, Yang L. VCAM-1 secreted from cancer-associated fibroblasts enhances the growth and invasion of lung cancer cells through AKT and MAPK signaling. Cancer Lett 473: 62–73, 2020. doi: 10.1016/j.canlet.2019.12.039. [DOI] [PubMed] [Google Scholar]
- 54. Davies MR, Garcia S, Liu M, Chi H, Kim HT, Raffai RL, Liu X, Feeley BT. Muscle-derived beige adipose precursors secrete promyogenic exosomes that treat rotator cuff muscle degeneration in mice and are identified in humans by single-cell RNA sequencing. Am J Sports Med 50: 2247–2257, 2022. [Erratum in Am J Sports Med 50: NP38, 2022]. doi: 10.1177/03635465221095568. [DOI] [PubMed] [Google Scholar]
- 55. Depuydt CE, Goosens V, Janky R, D’hondt A, De Bleecker JL, Noppe N, Derveaux S, Thal DR, Claeys KG. Unraveling the molecular basis of the dystrophic process in limb-girdle muscular dystrophy LGMD-R12 by differential gene expression profiles in diseased and healthy muscles. Cells 11: 1508, 2022. doi: 10.3390/cells11091508. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56. Yao L, Tichy ED, Zhong L, Mohanty S, Wang L, Ai E, Yang S, Mourkioti F, Qin L. Gli1 defines a subset of fibro-adipogenic progenitors that promote skeletal muscle regeneration with less fat accumulation. J Bone Miner Res 36: 1159–1173, 2021. doi: 10.1002/jbmr.4265. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57. Oprescu SN, Yue F, Qiu J, Brito LF, Kuang S. Temporal dynamics and heterogeneity of cell populations during skeletal muscle regeneration. iScience 23: 100993, 2020. doi: 10.1016/j.isci.2020.100993. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58. Scott RW, Arostegui M, Schweitzer R, Rossi FMV, Underhill TM. Hic1 defines quiescent mesenchymal progenitor subpopulations with distinct functions and fates in skeletal muscle regeneration. Cell Stem Cell 25: 797–813.e9, 2019. doi: 10.1016/j.stem.2019.11.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59. Leinroth AP, Mirando AJ, Rouse D, Long JT, Chakkalakal JV, Hilton MJ, Leinroth AP, Mirando AJ, Rouse D, Kobayahsi Y, Tata PR. Identification of distinct non-myogenic skeletal- muscle-resident mesenchymal cell populations. Cell Rep 39: 110785, 2022. doi: 10.1016/j.celrep.2022.110785. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60. Wosczyna MN, Konishi CT, Perez Carbajal EE, Wang TT, Walsh RA, Gan Q, Wagner MW, Rando TA. Mesenchymal stromal cells are required for regeneration and homeostatic maintenance of skeletal muscle. Cell Rep 27: 2029–2035.e5, 2019. doi: 10.1016/j.celrep.2019.04.074. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61. Rubenstein AB, Smith GR, Raue U, Begue G, Minchev K, Ruf-Zamojski F, Nair VD, Wang X, Zhou L, Zaslavsky E, Trappe TA, Trappe S, Sealfon SC. Single-cell transcriptional profiles in human skeletal muscle. Sci Rep 10: 229–215, 2020. doi: 10.1038/s41598-019-57110-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62. Fitzgerald G, Turiel G, Gorski T, Soro-Arnaiz I, Zhang J, Casartelli NC, Masschelein E, Maffiuletti NA, Sutter R, Leunig M, Farup J, De Bock K. MME+ fibro-adipogenic progenitors are the dominant adipogenic population during fatty infiltration in human skeletal muscle. Commun Biol 6: 111, 2023. doi: 10.1038/s42003-023-04504-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63. Stantzou A, Relizani K, Morales-Gonzalez S, Gallen C, Grassin A, Ferry A, Schuelke M, Amthor H. Extracellular matrix remodelling is associated with muscle force increase in overloaded mouse plantaris muscle. Neuropathol Appl Neurobiol 47: 218–235, 2021. doi: 10.1111/nan.12655. [DOI] [PubMed] [Google Scholar]
- 64. Gutpell KM, Hrinivich WT, Hoffman LM. Skeletal muscle fibrosis in the mdx/utrn+/− mouse validates its suitability as a murine model of Duchenne muscular dystrophy. PLoS One 10: e0117306, 2015. doi: 10.1371/journal.pone.0117306. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65. Wang X, Chen J, Homma ST, Wang Y, Smith GR, Ruf-Zamojski F, Sealfon SC, Zhou L. Diverse effector and regulatory functions of fibro/adipogenic progenitors during skeletal muscle fibrosis in muscular dystrophy. iScience 26: 105775, 2023. doi: 10.1016/j.isci.2022.105775. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66. Fadic R, Mezzano V, Alvarez K, Cabrera D, Holmgren J, Brandan E. Increase in decorin and biglycan in Duchenne muscular dystrophy: role of fibroblasts as cell source of these proteoglycans in the disease. J Cell Mol Med 10: 758–769, 2006. doi: 10.1111/j.1582-4934.2006.tb00435.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67. Rixon C, Andreassen K, Shen X, Erusappan PM, Almaas VM, Palmero S, Dahl CP, Ueland T, Sjaastad I, Louch WE, Stokke MK, Tønnessen T, Christensen G, Lunde IG. Lumican accumulates with fibrillar collagen in fibrosis in hypertrophic cardiomyopathy. ESC Heart Fail 10: 858–871, 2023. doi: 10.1002/ehf2.14234. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68. Xiao D, Liang T, Zhuang Z, He R, Ren J, Jiang S, Zhu L, Wang K, Shi D. Lumican promotes joint fibrosis through TGF-β signaling. FEBS Open Bio 10: 2478–2488, 2020. doi: 10.1002/2211-5463.12974. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69. Kissin EY, Lemaire R, Korn JH, Lafyatis R. Transforming growth factor β induces fibroblast fibrillin-1 matrix formation. Arthritis Rheum 46: 3000–3009, 2002. doi: 10.1002/art.10621. [DOI] [PubMed] [Google Scholar]
- 70. Bouzeghrane F, Reinhardt DP, Reudelhuber TL, Thibault G. Enhanced expression of fibrillin-1, a constituent of the myocardial extracellular matrix in fibrosis. Am J Physiol Heart Circ Physiol 289: H982–H991, 2005. doi: 10.1152/ajpheart.00151.2005. [DOI] [PubMed] [Google Scholar]
- 71. Smith LR, Hammers DW, Sweeney HL, Barton ER. Increased collagen cross-linking is a signature of dystrophin-deficient muscle. Muscle Nerve 54: 71–78, 2016. doi: 10.1002/mus.24998. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72. Wu Y, Wu Y, Yang Y, Yu J, Wu J, Liao Z, Guo A, Sun Y, Zhao Y, Chen J, Xiao Q. Lysyl oxidase-like 2 inhibitor rescues d-galactose-induced skeletal muscle fibrosis. Aging Cell 21: e13659, 2022. doi: 10.1111/acel.13659. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73. Mitra D, Yasui OW, Harvestine JN, Link JM, Hu JC, Athanasiou KA, Leach JK. Exogenous lysyl oxidase-like 2 and perfusion culture induce collagen crosslink formation in osteogenic grafts. Biotechnol J 14: e1700763, 2019. doi: 10.1002/biot.201700763. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74. Asgari R, Vaisi-Raygani A, Aleagha MSE, Mohammadi P, Bakhtiari M, Arghiani N. CD147 and MMPs as key factors in physiological and pathological processes. Biomed Pharmacother 157: 113983, 2023. doi: 10.1016/j.biopha.2022.113983. [DOI] [PubMed] [Google Scholar]
- 75. Chuliá-Peris L, Carreres-Rey C, Gabasa M, Alcaraz J, Carretero J, Pereda J. Matrix metalloproteinases and their inhibitors in pulmonary fibrosis: EMMPRIN/CD147 comes into play. Int J Mol Sci 23: 6894, 2022. doi: 10.3390/ijms23136894. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76. Eldred JA, Hodgkinson LM, Dawes LJ, Reddan JR, Edwards DR, Wormstone IM. MMP2 activity is critical for TGFβ2-induced matrix contraction-implications for fibrosis. Invest Ophthalmol Vis Sci 53: 4085–4098, 2012. doi: 10.1167/iovs.12-9457. [DOI] [PubMed] [Google Scholar]
- 77. Taylor SH, Yeung CYC, Kalson NS, Lu Y, Zigrino P, Starborg T, Warwood S, Holmes DF, Canty-Laird EG, Mauch C, Kadler KE. Matrix metalloproteinase 14 is required for fibrous tissue expansion. eLife 4: e09345, 2015. doi: 10.7554/eLife.09345. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78. Giannandrea M, Parks WC. Diverse functions of matrix metallopreinases during fibrosis. Dis Model Mech 7: 193–203, 2014. doi: 10.1242/dmm.012062. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79. Alameddine HS, Morgan JE. Matrix metalloproteinases and tissue inhibitor of metalloproteinases in inflammation and fibrosis of skeletal muscles. J Neuromuscul Dis 3: 455–473, 2016. doi: 10.3233/JND-160183. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80. Kaur G, Davies MR, Liu X, Feeley BT. The role of fibro-adipogenic progenitors in musculoskeletal disease. Muscles Ligaments Tendons J 11: 201–214, 2021. doi: 10.32098/mltj.02.2021.01. [DOI] [Google Scholar]
- 81. Davies MR, Liu X, Lee L, Laron D, Ning AY, Kim HT, Feeley BT. TGF-β small molecule inhibitor sb431542 reduces rotator cuff muscle fibrosis and fatty infiltration by promoting fibro/adipogenic progenitor apoptosis. PLoS One 11: e0155486, 2016. doi: 10.1371/journal.pone.0155486. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82. Liu X, Ning AY, Chang NC, Kim H, Nissenson R, Wang L, Feeley BT. Investigating the cellular origin of rotator cuff muscle fatty infiltration and fibrosis after injury. Muscles Ligaments Tendons J 6: 6–15, 2016. doi: 10.11138/mltj/2016.6.1.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83. Kim HK, Merrow AC, Shiraj S, Wong BL, Horn PS, Laor T. Analysis of fatty infiltration and inflammation of the pelvic and thigh muscles in boys with Duchenne muscular dystrophy (DMD): grading of disease involvement on MR imaging and correlation with clinical assessments. Pediatr Radiol 43: 1327–1335, 2013. doi: 10.1007/s00247-013-2696-z. [DOI] [PubMed] [Google Scholar]
- 84. Wokke BH, van den Bergen JC, Versluis MJ, Niks EH, Milles J, Webb AG, van Zwet EW, Aartsma-Rus A, Verschuuren JJ, Kan HE. Quantitative MRI and strength measurements in the assessment of muscle quality in Duchenne muscular dystrophy. Neuromuscul Disord 24: 409–416, 2014. doi: 10.1016/j.nmd.2014.01.015. [DOI] [PubMed] [Google Scholar]
- 85. Polavarapu K, Manjunath M, Preethish-Kumar V, Sekar D, Vengalil S, Thomas PT, Sathyaprabha TN, Bharath RD, Nalini A. Muscle MRI in Duchenne muscular dystrophy: evidence of a distinctive pattern. Neuromuscul Disord 26: 768–774, 2016. doi: 10.1016/j.nmd.2016.09.002. [DOI] [PubMed] [Google Scholar]
- 86. Vumbaca S, Giuliani G, Fiorentini V, Tortolici F, Perpetuini AC, Riccio F, Sennato S, Gargioli C, Fuoco C, Castagnoli L, Cesareni G. Characterization of the skeletal muscle secretome reveals a role for extracellular vesicles and il1α/il1β in restricting fibro/adipogenic progenitor adipogenesis. Biomolecules 11: 1171, 2021. doi: 10.3390/biom11081171. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87. Giuliani G, Rosina M, Reggio A. Signaling pathways regulating the fate of fibro/adipogenic progenitors (FAPs) in skeletal muscle regeneration and disease. FEBS J 289: 6484–6517, 2022. doi: 10.1111/febs.16080. [DOI] [PubMed] [Google Scholar]
- 88. Hu L-Y, Mileti CJ, Loomis T, Brashear SE, Ahmad S, Chellakudam RR, Wohlgemuth RP, Gionet-Gonzales MA, Leach JK, Smith LR. Skeletal muscle progenitors are sensitive to collagen architectural features of fibril size and cross linking. Am J Physiol Cell Physiol 321: C330–C342, 2021. doi: 10.1152/ajpcell.00065.2021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89. Trensz F, Lucien F, Couture V, Söllrald T, Drouin G, Rouleau AJ, Grandbois M, Lacraz G, Grenier G. Increased microenvironment stiffness in damaged myofibers promotes myogenic progenitor cell proliferation. Skelet Muscle 5: 5–16, 2015. [Erratum in Skelet Muscle 5:31, 2015]. doi: 10.1186/s13395-015-0030-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90. Mozzetta C, Consalvi S, Saccone V, Tierney M, Diamantini A, Mitchell KJ, Marazzi G, Borsellino G, Battistini L, Sassoon D, Sacco A, Puri PL, Sandonà M, Consalvi S, Tucciarone L, De Bardi M, Scimeca M, Angelini DF, Buffa V, D’Amico A, Bertini ES, Cazzaniga S, Bettica P, Bouché M, Bongiovanni A, Puri PL, Saccone V. HDAC inhibitors tune miRNAs in extracellular vesicles of dystrophic muscle-resident mesenchymal cells. EMBO Rep 21: e50863, 2020. doi: 10.15252/embr.202050863. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91. Lukjanenko L, Karaz S, Stuelsatz P, Gurriaran-Rodriguez U, Michaud J, Dammone G, Sizzano F, Mashinchian O, Ancel S, Migliavacca E, Liot S, Jacot G, Metairon S, Raymond F, Descombes P, Palini A, Chazaud B, Rudnicki MA, Bentzinger CF, Feige JN. Aging disrupts muscle stem cell function by impairing matricellular WISP1 secretion from fibro-adipogenic progenitors. Cell Stem Cell 24: 433–446.e7, 2019. doi: 10.1016/j.stem.2018.12.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92. Moratal C, Arrighi N, Dechesne CA, Dani C. Control of muscle fibro-adipogenic progenitors by myogenic lineage is altered in aging and Duchenne muscular dystrophy. Cell Physiol Biochem 53: 1029–1045, 2019. doi: 10.33594/000000196. [DOI] [PubMed] [Google Scholar]
- 93. Biferali B, Proietti D, Mozzetta C, Madaro L. Fibro–adipogenic progenitors cross-talk in skeletal muscle: the social network. Front Physiol 10: 1074–10, 2019. doi: 10.3389/fphys.2019.01074. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94. Arrighi N, Moratal C, Savary G, Fassy J, Nottet N, Pons N, Clément N, Fellah S, Larrue R, Magnone V, Lebrigand K, Pottier N, Dechesne C, Vassaux G, Dani C, Peraldi P, Mari B. The fibromiR miR-214-3p is upregulated in Duchenne muscular dystrophy and promotes differentiation of human fibro-adipogenic muscle progenitors. Cells 10: 1832, 2021. doi: 10.3390/cells10071832. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95. Hogarth MW, Defour A, Lazarski C, Gallardo E, Manera JD, Partridge TA, Nagaraju K, Jaiswal JK. Fibroadipogenic progenitors are responsible for muscle loss in limb girdle muscular dystrophy 2B. Nat Commun 10: 2430–13, 2019. doi: 10.1038/s41467-019-10438-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96. Booth CM, Cortina-Borja MJF, Theologis TN. Collagen accumulation in muscles of children with cerebral palsy and correlation with severity of spasticity. Dev Med Child Neurol 43: 314–320, 2001. doi: 10.1017/s0012162201000597. [DOI] [PubMed] [Google Scholar]
- 97. Jensen AR, Kelley BV, Mosich GM, Ariniello A, Eliasberg CD, Vu B, Shah P, Devana SK, Murray IR, Péault B, Dar A, Petrigliano FA. Platelet-derived growth factor receptor α co-expression typifies a subset of platelet-derived growth factor receptor β-positive progenitor cells that contribute to fatty degeneration and fibrosis of the murine rotator cuff. J Shoulder Elbow Surg 27: 1149–1161, 2018. doi: 10.1016/j.jse.2018.02.040. [DOI] [PubMed] [Google Scholar]
- 98. Lee C, Agha O, Liu M, Davies M, Bertoy L, Kim HT, Liu X, Feeley BT. Rotator cuff fibro-adipogenic progenitors demonstrate highest concentration, proliferative capacity, and adipogenic potential across muscle groups. J Orthop Res 38: 1113–1121, 2020. doi: 10.1002/jor.24550. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99. Mázala DAG, Novak JS, Hogarth MW, Nearing M, Adusumalli P, Tully CB, Habib NF, Gordish-Dressman H, Chen YW, Jaiswal JK, Partridge TA. TGF-β-driven muscle degeneration and failed regeneration underlie disease onset in a DMD mouse model. JCI Insight 5: e135703, 2020. doi: 10.1172/jci.insight.135703. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100. Fiore D, Judson RN, Low M, Lee S, Zhang E, Hopkins C, Xu P, Lenzi A, Rossi FMV, Lemos DR. 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, 2016. doi: 10.1016/j.scr.2016.06.007. [DOI] [PubMed] [Google Scholar]
- 101. Miyake T, Alli NS, McDermott JC. Nuclear function of Smad7 promotes myogenesis. Mol Cell Biol 30: 722–735, 2010. doi: 10.1128/mcb.01005-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102. Murphy KT, Ryall JG, Snell SM, Nair L, Koopman R, Krasney PA, Ibebunjo C, Holden KS, Loria PM, Salatto CT, Lynch GS. Antibody-directed myostatin inhibition improves diaphragm pathology in young but not adult dystrophic mdx mice. Am J Pathol 176: 2425–2434, 2010. doi: 10.2353/ajpath.2010.090932. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103. Yu H, Xia Yao Y, Bu F, Tian Chen Y, Wu Y, Ting Yang Y, Chen X, Zhu Y, Wang Q, Pan X, Yin Meng X, Ming Huang C, Li J. Blockade of YAP alleviates hepatic fibrosis through accelerating apoptosis and reversion of activated hepatic stellate cells. Mol Immunol 107: 29–40, 2019. doi: 10.1016/j.molimm.2019.01.004. [DOI] [PubMed] [Google Scholar]
- 104. Du K, Hyun J, Premont RT, Choi SS, Michelotti GA, Swiderska-Syn M, Dalton GD, Thelen E, Rizi BS, Jung Y, Diehl AM. Hedgehog-YAP signaling pathway regulates glutaminolysis to control activation of hepatic stellate cells. Gastroenterology 154: 1465–1479.e13, 2018. doi: 10.1053/j.gastro.2017.12.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105. Mannaerts I, Leite SB, Verhulst S, Claerhout S, Eysackers N, Thoen LFR, Hoorens A, Reynaert H, Halder G, Van Grunsven LA. The Hippo pathway effector YAP controls mouse hepatic stellate cell activation. J Hepatol 63: 679–688, 2015. doi: 10.1016/j.jhep.2015.04.011. [DOI] [PubMed] [Google Scholar]
- 106. Lagares D, Santos A, Grasberger PE, Liu F, Probst CK, Rahimi RA, Sakai N, Kuehl T, Ryan J, Bhola P, Montero J, Kapoor M, Baron M, Varelas X, Tschumperlin DJ, Letai A, Tager AM. Targeted apoptosis of myofibroblasts with the BH3 mimetic ABT-263 reverses established fibrosis. Sci Transl Med 9: eaal3765, 2017. doi: 10.1126/scitranslmed.aal3765. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107. Haak AJ, Kostallari E, Sicard D, Ligresti G, Choi KM, Caporarello N, Jones DL, Tan Q, Meridew J, Espinosa AMD, Aravamudhan A, Maiers JL, Britt RD, Roden AC, Pabelick CM, Prakash YS, Nouraie SM, Li X, Zhang Y, Kass DJ, Lagares D, Tager AM, Varelas X, Shah VH, Tschumperlin DJ. Selective YAP/TAZ inhibition in fibroblasts via dopamine receptor D1 agonism reverses fibrosis. Sci Transl Med 11: eaau6296, 2019. doi: 10.1126/scitranslmed.aau6296. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108. Pan J, Li D, Xu Y, Zhang J, Wang Y, Chen M, Lin S, Huang L, Chung EJ, Citrin DE, Wang Y, Hauer-Jensen M, Zhou D, Meng A. Inhibition of Bcl-2/xl With ABT-263 selectively kills senescent type II pneumocytes and reverses persistent pulmonary fibrosis induced by ionizing radiation in mice. Int J Radiat Oncol Biol Phys 99: 353–361, 2017. doi: 10.1016/j.ijrobp.2017.02.216. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109. Moncsek A, Al-Suraih MS, Trussoni CE, O'Hara SP, Splinter PL, Zuber C, Patsenker E, Valli PV, Fingas CD, Weber A, Zhu Y, Tchkonia T, Kirkland JL, Gores GJ, Müllhaupt B, LaRusso NF, Mertens JC. Targeting senescent cholangiocytes and activated fibroblasts with Bcl-xL inhibitors ameliorates fibrosis in Mdr2−/− mice. Hepatology 67: 247–259, 2018. doi: 10.1002/hep.29464. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110. Walaszczyk A, Dookun E, Redgrave R, Tual-Chalot S, Victorelli S, Spyridopoulos I, Owens A, Arthur HM, Passos JF, Richardson GD. Pharmacological clearance of senescent cells improves survival and recovery in aged mice following acute myocardial infarction. Aging Cell 18: e12945, 2019. doi: 10.1111/acel.12945. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111. Saccone V, Consalvi S, Giordani L, Mozzetta C, Barozzi I, Sandoná M, Ryan T, Rojas-Muñoz A, Madaro L, Fasanaro P, Borsellino G, De Bardi M, Frigè G, Termanini A, Sun X, Rossant J, Bruneau BG, Mercola M, Minucci S, Puri PL. HDAC-regulated myomiRs control BAF60 variant exchange and direct the functional phenotype of fibro-adipogenic progenitors in dystrophic muscles. Genes Dev 28: 841–857, 2014. doi: 10.1101/gad.234468.113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112. Mozzetta C, Consalvi S, Saccone V, Tierney M, Diamantini A, Mitchell KJ, Marazzi G, Borsellino G, Battistini L, Sassoon D, Sacco A, Puri PL. 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, 2013. doi: 10.1002/emmm.201202096. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113. Rodino-Klapac LR, Haidet AM, Kota J, Handy C, Kaspar BK, Mendell JR. Inhibition of myostatin with emphasis on follistatin as a therapy for muscle disease. Muscle Nerve 39: 283–296, 2009. doi: 10.1002/mus.21244. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114. Bettica P, Petrini S, D'Oria V, D'Amico A, Catteruccia M, Pane M, Sivo S, Magri F, Brajkovic S, Messina S, Vita GL, Gatti B, Moggio M, Puri PL, Rocchetti M, De Nicolao G, Vita G, Comi GP, Bertini E, Mercuri E. Histological effects of givinostat in boys with Duchenne muscular dystrophy. Neuromuscul Disord 26: 643–649, 2016. doi: 10.1016/j.nmd.2016.07.002. [DOI] [PubMed] [Google Scholar]
- 115. Consalvi S, Mozzetta C, Bettica P, Germani M, Fiorentini F, Del Bene F, Rocchetti M, Leoni F, Monzani V, Mascagni P, Puri PL, Saccone V. Preclinical studies in the mdx mouse model of duchenne muscular dystrophy with the histone deacetylase inhibitor givinostat. Mol Med 19: 79–87, 2013. doi: 10.2119/molmed.2013.00011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116. Herrera J, Henke CA, Bitterman PB. Extracellular matrix as a driver of progressive fibrosis. J Clin Invest 128: 45–53, 2018. doi: 10.1172/JCI93557. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117. Chapman MA, Meza R, Lieber RL. Skeletal muscle fibroblasts in health and disease. Differentiation 92: 108–115, 2016. doi: 10.1016/j.diff.2016.05.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118. Ismaeel A, Kim J-SS, Kirk JSJS, Smith RS, Bohannon WT, Koutakis P. Role of transforming growth factor-β in skeletal muscle fibrosis: a review. Int J Mol Sci 20: 2446, 2019. doi: 10.3390/ijms20102446. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119. Martin K, Pritchett J, Llewellyn J, Mullan AF, Athwal VS, Dobie R, Harvey E, Zeef L, Farrow S, Streuli C, Henderson NC, Friedman SL, Hanley NA, Hanley KP. PAK proteins and YAP-1 signalling downstream of integrin β-1 in myofibroblasts promote liver fibrosis. Nat Commun 7: 12502, 2016. doi: 10.1038/ncomms12502. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120. Yeo JH, Kim JY. Minimally invasive treatments of Dupuytren disease: an overview. J Hand Surg Am 26: 131–141, 2021. doi: 10.1142/S2424835521400026. [DOI] [PubMed] [Google Scholar]
- 121. Howard JJ, Huntley JS, Graham HK, Herzog WL. Intramuscular injection of collagenase clostridium histolyticum may decrease spastic muscle contracture for children with cerebral palsy. Med Hypotheses 122: 126–128, 2019. doi: 10.1016/j.mehy.2018.11.002. [DOI] [PubMed] [Google Scholar]
- 122. Howard JJ, Joumaa V, Robinson KG, Lee SK, Akins RE, Syed F, Shrader MW, Huntley JS, Graham HK, Leonard T, Herzog W. Collagenase treatment decreases muscle stiffness in cerebral palsy: a preclinical ex vivo biomechanical analysis of hip adductor muscle fiber bundles. Dev Med Child Neurol, 2023. doi: 10.1111/dmcn.15637. [DOI] [PubMed] [Google Scholar]
- 123. Chen W, Yang A, Jia J, Popov YV, Schuppan D, You H. Lysyl oxidase (LOX) family members: rationale and their potential as therapeutic targets for liver fibrosis. Hepatology 72: 729–741, 2020. doi: 10.1002/hep.31236. [DOI] [PubMed] [Google Scholar]
- 124. Brashear SE, Wohlgemuth RP, Hu LY, Jbeily EH, Christiansen BA, Smith LR. Collagen cross-links scale with passive stiffness in dystrophic mouse muscles, but are not altered with administration of a lysyl oxidase inhibitor. PLoS One 17: e0271776, 2022. doi: 10.1371/journal.pone.0271776. [DOI] [PMC free article] [PubMed] [Google Scholar]