Laminin-α2-related congenital muscular dystrophy (LAMA2-CMD) is a neuromuscular disease affecting around 1-9 in 1,000,000 children. LAMA2-CMD is caused by mutations in the LAMA2 gene resulting in the loss of laminin-211/221 heterotrimers in skeletal muscle. LAMA2-CMD patients exhibit severe hypotonia and progressive muscle weakness. Currently, there is no effective treatment for LAMA2-CMD and patients die prematurely. The loss of laminin-α2 results in muscle degeneration, defective muscle repair and dysregulation of multiple signaling pathways. Signaling pathways that regulate muscle metabolism, survival and fibrosis have been shown to be dysregulated in LAMA2-CMD. As vemurafenib is a US Food and Drug Administration (FDA)-approved serine/threonine kinase inhibitor, we investigated whether vemurafenib could restore some of the serine/threonine kinase-related signaling pathways and prevent disease progression in the dyW−/− mouse model of LAMA2-CMD. Our results show that vemurafenib reduced muscle fibrosis, increased myofiber size and reduced the percentage of fibers with centrally located nuclei in dyW−/− mouse hindlimbs. These studies show that treatment with vemurafenib restored the TGF-β/SMAD3 and mTORC1/p70S6K signaling pathways in skeletal muscle. Together, our results indicate that vemurafenib partially improves histopathology but does not improve muscle function in a mouse model of LAMA2-CMD.

Laminin-α2-related congenital muscular dystrophy (LAMA2-CMD), also known as merosin-deficient congenital muscular dystrophy type 1A (MDC1A), is a severe form of congenital muscular dystrophy (CMD) that accounts for ∼30-40% of all CMDs in Europe (Allamand and Guicheney, 2002; Muntoni and Voit, 2004). The prevalence of LAMA2-CMD is not well known, but it is estimated to be about 1-9 per 1,000,000 of the total population (Durbeej, 2015; Graziano et al., 2015; Norwood et al., 2009).

LAMA2-CMD is caused by mutations in the LAMA2 gene inherited in an autosomal recessive fashion that results in a complete loss or truncated expression of the laminin-α2 chain (Geranmayeh et al., 2010; Helbling-Leclerc et al., 1995). Laminin-α2 is essential for the assembly of laminin-211/221 heterotrimers, the major laminin isoforms expressed in adult skeletal muscle (Aumailley et al., 2005; Durbeej, 2010). Laminin-211 is the predominant isoform in the basement membrane surrounding the muscle fibers and axon–Schwann cell units of peripheral nerves, whereas laminin-221 is specifically found in the synaptic cleft of the neuromuscular junctions in adult skeletal muscles (Patton, 2000; Patton et al., 1997).

Laminin-α2 deficiency leads to the disruption of the basement membrane, resulting in muscle degeneration, fibrosis, inflammation (Gawlik and Durbeej, 2011, 2020; Pegoraro et al., 1996), abortive skeletal muscle regeneration (Kuang et al., 1999) and peripheral neuropathy (Mercuri et al., 1996; Shorer et al., 1995). LAMA2-CMD patients rarely achieve independent ambulation and exhibit severe hypotonia with muscle weakness and joint contractures at birth or within the first 6 months of life. Premature death is mainly caused by respiratory insufficiency in LAMA2-CMD patients (Geranmayeh et al., 2010; Jones et al., 2001; Philpot et al., 1995; Xiong et al., 2015; Zambon et al., 2020). Currently, there is no cure or effective treatment for LAMA2-CMD. The clinical practice guidelines recommend the multidisciplinary management of the symptoms. The standard medical care includes, among others, the treatment of joint contractures to promote joint and bone development, the use of orthotics and splinting for facilitation of standing and walking, activities to improve respiratory function, assisted ventilation, and management of pain (Oliveira et al., 2020; Wang et al., 2010). Clinical care only temporarily mitigates the symptoms of the disease.

Transgenic expression of the Lama2 gene has been effective at preventing disease progression in mouse models of LAMA2-CMD (Kuang et al., 1998). However, laminin-α2 is encoded by a ∼10 kb transcript (Miyagoe-Suzuki et al., 2000), and therefore current gene therapy approaches that aim to replace the defective gene and restore laminin-α2 expression remain a challenge. Other gene therapy approaches aiming to stabilize the basement membrane of the muscle fibers have been shown to improve muscle disease in different mouse models of LAMA2-CMD (Aoki et al., 2013; Doe et al., 2011; Gawlik and Durbeej, 2010; Gawlik et al., 2004, 2018; Kemaladewi et al., 2017; McKee et al., 2009; Moll et al., 2001; Reinhard et al., 2017). However, the translation of gene therapies to the clinic remains a challenge at present owing to safety concerns including gene editing accuracy, insertional mutagenesis, limited long-term expression of the corrected gene and host immune response (Tang and Xu, 2020). Therefore, there is an urgent need to develop therapies that can slow disease progression and act on the downstream effects caused by the loss of laminin-α2, such as basement membrane disruption, muscle atrophy, apoptosis, regeneration, inflammation and fibrosis.

Several small molecules and biological treatments have been tested in laminin-α2-deficient mouse models – these include the administration of laminin-111 protein (Barraza-Flores et al., 2020; Rooney, 2012; van Ry et al., 2014) to compensate for the loss of laminin-211/221; IGF-1 (Accorsi et al., 2016; Lynch et al., 2001) and clenbuterol (Hayes and Williams, 1998) to promote muscle growth; omigapil (Erb et al., 2009; Yu et al., 2013), doxycycline (Girgenrath et al., 2009) and losartan (Elbaz et al., 2015) to inhibit apoptosis; anti-fibrotic (Elbaz et al., 2012; Meinen et al., 2012; Nevo et al., 2010, 2011) and anti-inflammatory (Connolly et al., 2002; Dadush et al., 2010) drugs; and several drugs targeting other important signaling pathways (Carmignac et al., 2011a,b; Fontes-Oliveira et al., 2018; Körner et al., 2014; Körner and Durbeej, 2016; Millay et al., 2008; Tomomura et al., 2011).

Studies show that loss of laminin-α2 leads to the dysregulation of several signaling pathways in the skeletal muscle of laminin-α2-deficient mouse models (de Oliveira et al., 2014; Durbeej, 2015; Nguyen et al., 2019; Taniguchi et al., 2006; Mehuron et al., 2014). The Ras-Raf-MEK-ERK signaling pathway, including proteins of the mitogen-activated protein kinase (MAPK) family, is an important signaling pathway that regulates several cellular processes, including metabolism, differentiation, proliferation, survival, apoptosis and inflammation (Peti and Page, 2013; Schultze et al., 2012; Turjanski et al., 2007), and its dysregulation has been associated with several human diseases (Kim and Choi, 2015). Enhanced extracellular signal-regulated kinase (ERK) activation is correlated to muscle wasting in cancer cachexia and Emery–Dreifuss muscular dystrophy, and its inhibition was shown to reduce skeletal muscle loss (Muchir et al., 2013; Penna et al., 2010). Increased ERK phosphorylation has also been reported in a LAMA2-CMD mouse model, and its inhibition after treatment with losartan improved forelimb and hindlimb grip strength and reduced fibrosis (Elbaz et al., 2012).

Vemurafenib, a US Food and Drug Administration (FDA)-approved serine/threonine kinase inhibitor (Zhang et al., 2017), is used for the treatment of melanomas (Czirbesz et al., 2019; Luke and Hodi, 2012) and reported to be effective in the treatment of Langerhans cell histiocytosis (Evseev et al., 2021) and gliomas (Bautista et al., 2014) in pediatric patients through its inhibition of the serine/threonine-protein kinase B-Raf, an upstream activator of ERK (Macdonald et al., 1993; McCubrey et al., 2007). Vemurafenib has also been reported to inhibit JNK signaling (Vin et al., 2013), an important signaling pathway in skeletal muscle development (Xie et al., 2018) and wasting (Mulder et al., 2020), and when combined with other drugs for the treatment of melanomas, it can modulate other signaling pathways (Li et al., 2022). Considering that the loss of laminin-α2 dysregulates several signaling pathways in LAMA2-CMD mouse models (Meinen et al., 2012;de Oliveira et al., 2014; Elbaz et al., 2012), we investigated whether vemurafenib could prevent disease progression by modulating the activity of serine/threonine kinases in the dyW−/− mouse model of LAMA2-CMD, in which Lama2 is disrupted. Our studies show that short-term treatment with vemurafenib partially improves histomorphology, reduces fibrosis and downregulates the TGF-β/SMAD3 and mTORC1 pathways, but not the Ras-Raf-MEK-ERK signaling pathway. Despite the improvements observed, vemurafenib did not improve skeletal muscle function in dyW−/− mice, suggesting that the use of vemurafenib alone might not be a promising treatment option to prevent muscle disease progression in patients with LAMA2-CMD.

Muscle histopathology is partially improved with vemurafenib treatment in dyW−/− mice

The skeletal muscle histopathology in dyW−/− mice is characterized by a significant reduction in muscle area and the number of PAX7-positive cells during fetal development (Nunes et al., 2017). After birth, reduced body weight, muscle cross-sectional area and number of fibers are observed along with increased apoptosis and embryonic myosin heavy chain (eMHC, or MYH3) expression (Mehuron et al., 2014).

In this study, we investigated whether treatment with 5 mg/kg vemurafenib could prevent further growth impairment from 3 weeks to 8 weeks of age in dyW−/− mice. As treatment was started at 3 weeks of age, this study examines the ability of vemurafenib to prevent muscle disease progression after onset. Vemurafenib treatment starting at 3 weeks of age did not show improvements in body weight (Fig. 1A), quadriceps weight (Fig. 1B), tibialis anterior (TA) cross-sectional area (Fig. 1C) and the number of fibers (Fig. 1E) in dyW−/− mice compared to vehicle control-treated dyW−/− mice. We verified that treatment with vemurafenib did not change the percentage of eMHC-positive fibers in the TA (Fig. 1D,F) and was not able to reduce active caspase 3 levels in the gastrocnemius (Fig. 1G) of 8-week-old dyW−/− mice. However, we observed that vemurafenib treatment significantly reduced the percentage of centrally nucleated fibers (CNFs) (Fig. 1D,H, left graph) and increased the percentage of muscle fibers with a minimal Feret's (MinFeret) diameter of 40-50 μm (Fig. 1D and I, left graph) in the TA of dyW−/− mice, compared to the percentages measured for vehicle control-treated mice. We also analyzed the percentage of CNFs and fiber diameter distribution in the triceps of dyW−/− mice, and we observed that the treatment with vemurafenib had no effect in reducing the percentage of CNFs and did not improve fiber diameter in the triceps of dyW−/− mice compared to those measured for vehicle control-treated mice (Fig. 1H,I, right graphs). Therefore, our data show that vemurafenib partially improves muscle histopathology by decreasing the percentage of CNFs and increasing the percentage of bigger fibers in the TA but not in the triceps of dyW−/− mice.

Fig. 1.

Effects of vemurafenib on growth and histopathology of hindlimb muscles in the dyW−/− mouse model of LAMA2-CMD at 8 weeks of age. (A) Body weight measurements of WT, dyW−/− vehicle-treated and dyW−/− vemurafenib-treated mice. (B) Quadriceps muscle weight/body weight (BW) ratio measurements. (C) Tibialis anterior (TA) cross-sectional area measurements. (D) Representative images showing dystrophin (red), embryonic myosin heavy chain (eMHC) (green) and nuclei (blue) staining in TA cryosections from WT mice (left), vehicle-treated dyW−/− mice (middle) and vemurafenib-treated dyW−/− mice (right). Scale bar: 200 µm. (E-I) Quantitative analysis of (E) number of fibers per mm2 of TA muscle, (F) percentage of eMHC-positive fibers in TA muscle, (G) active caspase 3 levels in the protein extract from gastrocnemius muscle, (H) percentage of fibers with centrally located nuclei (CNFs) in TA (left graph) and triceps (right graph), and (I) minimal Feret's (MinFeret) diameter distribution (percentage of the total number of fibers) in TA (left graph) and triceps (right graph) from WT mice, vehicle-treated dyW−/− mice and vemurafenib-treated dyW−/− mice. One-way ANOVA with uncorrected Fisher's LSD test was performed for the data that followed the normal distribution (body weight, muscle weight/body weight, cross-sectional area, fibers/mm2, eMHC-positive fibers, percentage of CNFs). Two-way ANOVA analysis was performed for MinFeret diameter distribution data (*P<0.05 denoting significance between vehicle-treated dyW−/− mice and vemurafenib-treated dyW−/− mice). Kruskal–Wallis test was performed for the data that did not follow the normal distribution (active caspase 3). All data are represented by statistical significance of mean±s.e.m. (n=10 for all groups). ns, not significant; *P<0.05; **P<0.01; ***P<0.001; ****P<0.0001.

Fig. 1.

Effects of vemurafenib on growth and histopathology of hindlimb muscles in the dyW−/− mouse model of LAMA2-CMD at 8 weeks of age. (A) Body weight measurements of WT, dyW−/− vehicle-treated and dyW−/− vemurafenib-treated mice. (B) Quadriceps muscle weight/body weight (BW) ratio measurements. (C) Tibialis anterior (TA) cross-sectional area measurements. (D) Representative images showing dystrophin (red), embryonic myosin heavy chain (eMHC) (green) and nuclei (blue) staining in TA cryosections from WT mice (left), vehicle-treated dyW−/− mice (middle) and vemurafenib-treated dyW−/− mice (right). Scale bar: 200 µm. (E-I) Quantitative analysis of (E) number of fibers per mm2 of TA muscle, (F) percentage of eMHC-positive fibers in TA muscle, (G) active caspase 3 levels in the protein extract from gastrocnemius muscle, (H) percentage of fibers with centrally located nuclei (CNFs) in TA (left graph) and triceps (right graph), and (I) minimal Feret's (MinFeret) diameter distribution (percentage of the total number of fibers) in TA (left graph) and triceps (right graph) from WT mice, vehicle-treated dyW−/− mice and vemurafenib-treated dyW−/− mice. One-way ANOVA with uncorrected Fisher's LSD test was performed for the data that followed the normal distribution (body weight, muscle weight/body weight, cross-sectional area, fibers/mm2, eMHC-positive fibers, percentage of CNFs). Two-way ANOVA analysis was performed for MinFeret diameter distribution data (*P<0.05 denoting significance between vehicle-treated dyW−/− mice and vemurafenib-treated dyW−/− mice). Kruskal–Wallis test was performed for the data that did not follow the normal distribution (active caspase 3). All data are represented by statistical significance of mean±s.e.m. (n=10 for all groups). ns, not significant; *P<0.05; **P<0.01; ***P<0.001; ****P<0.0001.

Short-term treatment with vemurafenib reduces fibrosis and restores TGF-β1 and phosphorylated SMAD3 levels in dyW−/− mice

Interstitial fibrosis is considered an important feature in LAMA2-CMD muscles and it has been discussed to be a critical driver of the pathology (Accorsi et al., 2020; Taniguchi et al., 2006). Considering the importance of the fibrotic process in LAMA2-CMD pathology, we evaluated whether vemurafenib could inhibit the TGF-β/SMAD3 signaling pathway and prevent the progression of fibrosis (Ismaeel et al., 2019) in the dyW−/− mouse model. We verified that treatment with vemurafenib effectively reduced the levels of hydroxyproline, the major component of the collagen protein, in the quadriceps (Fig. 2A) and restored the levels of TGF-β1 (Fig. 2B) and phosphorylated SMAD3 (pS423/S425) (Fig. 2C) in the gastrocnemius of dyW−/− mice compared to those measured for dyW−/− vehicle-treated animals. These results show that vemurafenib is effective in preventing the progression of fibrosis and restores the TGF-β/SMAD3 signaling pathway to wild-type (WT) levels in the analyzed muscles of dyW−/− mice.

Fig. 2.

Vemurafenib reduces fibrosis and restores the TGF-β/SMAD3 signaling pathway in dyW−/− mice. Quantification of (A) hydroxyproline content in quadriceps muscle normalized by the muscle weight, (B) TGF-β1 levels and (C) percentage of SMAD3 (pS423/S425) in protein extracts of gastrocnemius muscle from WT mice, vehicle-treated dyW−/− mice and vemurafenib-treated dyW−/− mice. One-way ANOVA with uncorrected Fisher's LSD test represented by statistical significance of mean±s.e.m. (n=10 for all groups). ns, not significant; *P<0.05; **P<0.01; ***P<0.001; ****P<0.0001.

Fig. 2.

Vemurafenib reduces fibrosis and restores the TGF-β/SMAD3 signaling pathway in dyW−/− mice. Quantification of (A) hydroxyproline content in quadriceps muscle normalized by the muscle weight, (B) TGF-β1 levels and (C) percentage of SMAD3 (pS423/S425) in protein extracts of gastrocnemius muscle from WT mice, vehicle-treated dyW−/− mice and vemurafenib-treated dyW−/− mice. One-way ANOVA with uncorrected Fisher's LSD test represented by statistical significance of mean±s.e.m. (n=10 for all groups). ns, not significant; *P<0.05; **P<0.01; ***P<0.001; ****P<0.0001.

Vemurafenib does not reduce inflammation in dyW−/− mice

Inflammation is a hallmark of early disease progression in LAMA2-CMD patients (Pegoraro et al., 1996). Similarly, increased immune cell infiltration can be detected in dyW−/− mice as early as 1 week of age (Mehuron et al., 2014). In this context, we evaluated the effects of vemurafenib on the inflammatory response in dyW−/− mice. Our results corroborate with literature data demonstrating increased inflammatory cell infiltration in the dyW−/− skeletal muscle. However, no improvements in the inflammatory cell infiltration area were observed in the TA of dyW−/− mice after treatment with vemurafenib (Fig. 3A,B). We also detected increased levels of pro-inflammatory cytokines in the gastrocnemius muscle of 8-week-old dyW−/− mice, such as IP-10 (or CXCL10) (Fig. 3G), MIP-1α (CCL3) (Fig. 3I), IL-9 (Fig. 3J) and KC (CXCL1) (Fig. 3K), and reduced levels of the anti-inflammatory cytokine IL-10 (Fig. 3L). Vemurafenib treatment did not change the levels of these cytokines compared with their levels in dyW−/− vehicle-treated animals. Interestingly, the treatment with vemurafenib significantly restored the levels of eotaxin (CCL11) (Fig. 3C), an eosinophil chemoattractant (Rankin et al., 2000), and reduced the levels of monokine induced by γ interferon (MIG/CXCL9) (Fig. 3D), an important chemokine in inflammatory myopathies expressed by macrophages and T cells (Paparo, 2019). Luminex analysis showed no differences in the levels of IL-1β (Fig. 3E), IL-2 (Fig. 3F), IL-6 (Fig. 3H), MCP-1 (CCL2) (Fig. 3M) and LIF (Fig. 3N) in the three groups analyzed. Our data show that despite reducing the levels of two important chemokines, vemurafenib was not efficient in inhibiting inflammation in dyW−/− mice.

Fig. 3.

Effects of vemurafenib on inflammatory cell infiltration and cytokine profile in hindlimbs from dyW−/− mice at 8 weeks of age. (A) Representative images showing dystrophin (green), CD45-positive cells (red) and nuclei (blue) staining in TA cryosections from WT mice (left), vehicle-treated dyW−/− mice (middle) and vemurafenib-treated dyW−/− mice (right). Scale bar: 200 µm. (B-N) Quantification of (B) percentage of CD45-positive areas in TA muscle, and the levels of (C) eotaxin, (D) MIG, (E) IL-1β, (F) IL-2, (G) IP-10, (H) IL-6, (I) MIP-1α, (J) IL-9, (K) KC, (L) IL-10, (M) MCP-1 and (N) LIF in protein extracts of gastrocnemius muscle from WT mice, vehicle-treated dyW−/− mice and vemurafenib-treated dyW−/− mice. One-way ANOVA with uncorrected Fisher's LSD test was performed for the data that followed the normal distribution (CD45-positive area, MIG, IL-1β, IL-2, IL-6, MIP-1α, KC, MCP-1 and LIF measurements) and the Kruskal–Wallis test was performed for the data that did not follow the normal distribution (eotaxin, IP-10, IL-9 and IL-10 measurements). All data are represented by statistical significance of mean±s.e.m. Samples presenting cytokine levels below the Luminex detection limit were removed from the analysis. ns, not significant; *P<0.05; **P<0.01; ***P<0.001; ****P<0.0001. The individual symbols in the bar graphs represent the individual animals used in each experiment.

Fig. 3.

Effects of vemurafenib on inflammatory cell infiltration and cytokine profile in hindlimbs from dyW−/− mice at 8 weeks of age. (A) Representative images showing dystrophin (green), CD45-positive cells (red) and nuclei (blue) staining in TA cryosections from WT mice (left), vehicle-treated dyW−/− mice (middle) and vemurafenib-treated dyW−/− mice (right). Scale bar: 200 µm. (B-N) Quantification of (B) percentage of CD45-positive areas in TA muscle, and the levels of (C) eotaxin, (D) MIG, (E) IL-1β, (F) IL-2, (G) IP-10, (H) IL-6, (I) MIP-1α, (J) IL-9, (K) KC, (L) IL-10, (M) MCP-1 and (N) LIF in protein extracts of gastrocnemius muscle from WT mice, vehicle-treated dyW−/− mice and vemurafenib-treated dyW−/− mice. One-way ANOVA with uncorrected Fisher's LSD test was performed for the data that followed the normal distribution (CD45-positive area, MIG, IL-1β, IL-2, IL-6, MIP-1α, KC, MCP-1 and LIF measurements) and the Kruskal–Wallis test was performed for the data that did not follow the normal distribution (eotaxin, IP-10, IL-9 and IL-10 measurements). All data are represented by statistical significance of mean±s.e.m. Samples presenting cytokine levels below the Luminex detection limit were removed from the analysis. ns, not significant; *P<0.05; **P<0.01; ***P<0.001; ****P<0.0001. The individual symbols in the bar graphs represent the individual animals used in each experiment.

Vemurafenib treatment reduced mTORC1 signaling pathway activation in dyW−/− mice

Several muscle signaling pathways that regulate metabolism, inflammation and fibrosis have been described to be dysregulated in LAMA2-CMD patients and animal models, including ERK, NFκB and STAT3 (Carmignac et al., 2011a; de Oliveira et al., 2014; Durbeej, 2015; Elbaz et al., 2012, 2015; Nguyen et al., 2019; Nunes et al., 2017; Taniguchi et al., 2006; Mehuron et al., 2014). As vemurafenib is a MEK/ERK inhibitor, we first investigated whether the short-term treatment with vemurafenib would effectively modulate this pathway by inhibiting ERK phosphorylation. We verified that 5 mg/kg of vemurafenib did not inhibit ERK activation in gastrocnemius of 8-week-old dyW−/− mice (Fig. 4A). We next analyzed the effects of vemurafenib on STAT3 and NFκB activation, which have been described to be involved with inflammation, fibrosis, muscle atrophy and muscle wasting, and upregulated due to laminin-α2 deficiency (Chakraborty et al., 2017; Guadagnin et al., 2018; Ma et al., 2017; Sartori et al., 2021). Vemurafenib did not inhibit STAT3 (Fig. 4B) and NFκB p65 (RELA) activation (Fig. 4C) in dyW−/− mice.

Fig. 4.

Vemurafenib inhibited mTOR and p70S6K activation in dyW−/− mice. Levels of (A) phosphorylated ERK1/2 (pT202/Y204), (B) phosphorylated STAT3 (pY705), (C) phosphorylated NFκB p65 (pS536), (D) phosphorylated JNK1/2 (pT183/Y185), (E) phosphorylated mTOR (pS2448) and (F) phosphorylated p70S6K (pT389) in protein extracts of gastrocnemius muscle from WT mice, vehicle-treated dyW−/− mice and vemurafenib-treated dyW−/− mice. One-way ANOVA with uncorrected Fisher's LSD test was performed for the data that followed the normal distribution [ERK1/2 (pT202/Y204), mTOR (pS2448) and NFκB p65 (pS536)]. Kruskal–Wallis test was performed for the data that did not follow the normal distribution [STAT3 (pY705), JNK1/2 (pT183/Y185) and p70S6K (pT389)]. All data are represented by statistical significance of mean±s.e.m. (n=10 for all groups). ns, not significant; *P<0.05; **P<0.01; ***P<0.001; ****P<0.0001.

Fig. 4.

Vemurafenib inhibited mTOR and p70S6K activation in dyW−/− mice. Levels of (A) phosphorylated ERK1/2 (pT202/Y204), (B) phosphorylated STAT3 (pY705), (C) phosphorylated NFκB p65 (pS536), (D) phosphorylated JNK1/2 (pT183/Y185), (E) phosphorylated mTOR (pS2448) and (F) phosphorylated p70S6K (pT389) in protein extracts of gastrocnemius muscle from WT mice, vehicle-treated dyW−/− mice and vemurafenib-treated dyW−/− mice. One-way ANOVA with uncorrected Fisher's LSD test was performed for the data that followed the normal distribution [ERK1/2 (pT202/Y204), mTOR (pS2448) and NFκB p65 (pS536)]. Kruskal–Wallis test was performed for the data that did not follow the normal distribution [STAT3 (pY705), JNK1/2 (pT183/Y185) and p70S6K (pT389)]. All data are represented by statistical significance of mean±s.e.m. (n=10 for all groups). ns, not significant; *P<0.05; **P<0.01; ***P<0.001; ****P<0.0001.

As we observed an improvement in myofiber atrophy after treatment with vemurafenib (Fig. 1D,I), we analyzed the levels of phosphorylated JNK1/2, a MAPK family member, described to be involved in myofiber atrophy (Mulder et al., 2020). No difference was observed in the levels of phosphorylated JNK1/2 (Fig. 4D) in the groups analyzed.

Considering the importance of the serine/threonine kinase mTOR in metabolism regulation (Bodine et al., 2001; Fingar and Blenis, 2004; Giguère, 2018) and the well-described effects of sustained activation of the mTORC1 signaling pathway to promote skeletal muscle atrophy and loss in different pathological processes (Castets et al., 2013; Chiarini et al., 2019; Tang et al., 2014, 2019), we next investigated whether the increased myofiber diameter observed after treatment with vemurafenib was associated with the modulation of the mTORC1 signaling pathway in our animal model of LAMA2-CMD. We verified increased levels of phosphorylated mTOR (pS2448) (Fig. 4E) and phosphorylated p70S6K (RPS6KB1) (pT389), a downstream target of mTOR (Fig. 4F) in gastrocnemius of 8-week-old dyW−/− mice, and treatment with vemurafenib reduced the levels of phosphorylated mTOR (pS2448) (Fig. 4E) and restored the levels of phosphorylated p70S6K (Fig. 4F) to WT levels. Our results show that vemurafenib acted to normalize the mTORC1 signaling pathway in dyW−/− mice.

Ubiquitin-proteasome-related pathways and autophagy are not modulated by vemurafenib in 8-week-old dyW−/− mice

Sustained activation of mTORC1 has been described to promote muscle atrophy by increasing the expression of E3 ubiquitin ligases and impairing autophagy (Castets et al., 2013). To verify whether the ubiquitin-proteasome system and autophagy are impaired and/or being modulated by vemurafenib in dyW−/− mice at 8 weeks of age, we quantified the protein levels of two important ubiquitin-proteasome-related components, atrogin1 (FBXO32) and MuRF1 (TRIM63) (Khalil, 2018), and two proteins involved in autophagy, beclin-1 and p62 (SQSTM1) (Glick et al., 2010). We did not observe significant differences in the protein levels of atrogin1 (Fig. 5A), MuRF1 (Fig. 5B) and beclin-1 (Fig. 5C) in 8-week-old WT, dyW−/− vehicle-treated and dyW−/− vemurafenib-treated mice. However, we detected increased levels of p62 (Fig. 5D) in dyW−/− vehicle-treated mice compared to WT mice, suggesting that autophagy might be impaired in dyW−/− mice at 8 weeks of age. Treatment with vemurafenib was not effective at reducing p62 in dyW−/− mice. Our results suggest that the ubiquitin-proteasome system and autophagy are not modulated by vemurafenib in dyW−/− mice at 8 weeks of age.

Fig. 5.

The E3 ubiquitin ligases atrogin1 and MuRF1 are not upregulated in dyW−/− mice at 8 weeks of age. Protein levels of (A) atrogin1, (B) MuRF1, (C) beclin-1 and (D) p62 in protein extracts of gastrocnemius muscle from WT mice, vehicle-treated dyW−/− mice and vemurafenib-treated dyW−/− mice. One-way ANOVA with uncorrected Fisher's LSD test represented by statistical significance of mean±s.e.m. (n=10 for all groups). ns, nonsignificant; *P<0.05.

Fig. 5.

The E3 ubiquitin ligases atrogin1 and MuRF1 are not upregulated in dyW−/− mice at 8 weeks of age. Protein levels of (A) atrogin1, (B) MuRF1, (C) beclin-1 and (D) p62 in protein extracts of gastrocnemius muscle from WT mice, vehicle-treated dyW−/− mice and vemurafenib-treated dyW−/− mice. One-way ANOVA with uncorrected Fisher's LSD test represented by statistical significance of mean±s.e.m. (n=10 for all groups). ns, nonsignificant; *P<0.05.

Skeletal muscle function is not improved after vemurafenib treatment in dyW−/− mice

To investigate whether the improvements in histopathology and regulation of the mTORC1 signaling pathway were sufficient to improve skeletal muscle function in vemurafenib-treated dyW−/− mice, we performed the ex vivo contractility assay using the extensor digitorum longus (EDL) muscle. Our data show no significant improvements in strength in twitch (Fig. 6A), tetanus (Fig. 6B) and force-frequency measurements (Fig. 6C) in dyW−/− vemurafenib-treated mice compared to dyW−/− vehicle-treated mice. Our results indicate that treatment with 5 mg/kg vemurafenib from 3 to 8 weeks of age does not improve muscle strength in dyW−/− mice.

Fig. 6.

Ex vivo contractility analysis of EDL muscle in dyW−/− mice. Normalized force measurements for (A) twitch, (B) tetanus and (C) force frequency. One-way ANOVA with uncorrected Fisher's LSD test was performed for the twitch and tetanus data and two-way ANOVA analysis was performed for the force-frequency data (****P<0.0001 denoting significance between WT and vehicle- or vemurafenib-treated dyW−/− mice). All data are represented by statistical significance of mean±s.e.m. (n=9 for all groups). ns, not significant; ****P<0.0001.

Fig. 6.

Ex vivo contractility analysis of EDL muscle in dyW−/− mice. Normalized force measurements for (A) twitch, (B) tetanus and (C) force frequency. One-way ANOVA with uncorrected Fisher's LSD test was performed for the twitch and tetanus data and two-way ANOVA analysis was performed for the force-frequency data (****P<0.0001 denoting significance between WT and vehicle- or vemurafenib-treated dyW−/− mice). All data are represented by statistical significance of mean±s.e.m. (n=9 for all groups). ns, not significant; ****P<0.0001.

LAMA2-CMD is a severe form of congenital muscular dystrophy characterized by hypotonia, progressive muscle weakness and respiratory insufficiency (Sarkozy et al., 2020). Only palliative care is available for LAMA2-CMD patients, which aims to reduce disease symptoms and improve quality of life (Wang et al., 2010). Although several treatment approaches have been shown to improve muscle histopathology in mouse models of LAMA2-CMD (Aoki et al., 2013; Durbeej, 2015; Hagiwara et al., 2006; Qiao et al., 2005), only one clinical trial has been performed for this congenital muscular dystrophy. Santhera Pharmaceuticals completed a phase 1 clinical trial with omigapil, an anti-apoptotic drug, which was demonstrated to be safe and well tolerated in children with LAMA2-CMD. Owing to the short duration of the study, data showing disease-related improvements are not available (NCT01805024). The lack of clinical trials reinforces the need for new treatments that could be fast-tracked for LAMA2-CMD. In this context, our group evaluated the efficacy of vemurafenib in preventing disease progression in dyW−/− mice. As an FDA-approved drug, vemurafenib, if proven to be efficacious, could be fast-tracked for the treatment of LAMA2-CMD.

Our data showed that vemurafenib improved skeletal muscle histomorphology by increasing the percentage of muscle fibers with a MinFeret diameter of 40-50 μm and by reducing the percentage of CNFs from 25% in dyW−/− vehicle-treated mice to 18% in the TA of dyW−/− vemurafenib-treated mice. The treatment with vemurafenib did not increase regenerative capacity or reduce apoptosis in skeletal muscle. Therefore, vemurafenib was not able to prevent the loss of muscle from the hindlimbs of dyW−/− mice.

We did not observe a correlation between the percentage of CNFs, the percentage of eMHC-positive fibers and apoptosis in our animal model after treatment with vemurafenib, suggesting that the reduction in CNFs in the TA observed is not due to the reduction in the degeneration-regeneration process. Indeed, the displacement of nuclei in skeletal muscle fibers is not restricted to the regeneration process; the central nucleation can also be caused by skeletal muscle denervation (Carraro and Kern, 2016; Daou et al., 2020). LAMA2-CMD patients and animal models present with impaired motor nerve conduction owing to reduced axon diameter and thickness of the axon myelin sheath, which could cause central nucleation of myofibers (Nakagawa et al., 2001; Patton, 2000; Previtali and Zambon, 2020; Quijano-Roy et al., 2004). In dyW−/− mice, secondary atrophy is observed in the hindlimbs due to paralysis, which is discussed to be associated with peripheral neuropathy (Miyagoe-Suzuki et al., 2000). The forelimbs, such as the triceps, are not affected by paralysis in this animal model of LAMA2-CMD (Meinen et al., 2012). As we only observed the reduction in the percentage of CNFs in the TA and not in the triceps of dyW−/− mice after treatment with vemurafenib, we suggest that the action of vemurafenib to reduce the percentage of CNFs might be related to the reduction of the neuropathy associated with the loss of the laminin-α2 protein in the hindlimbs. Further studies will be required to evaluate whether the impaired nerve conduction might be contributing to the nuclei displacement and whether vemurafenib could be acting to improve LAMA2-CMD neuropathy in the hindlimbs of dyW−/− mice.

Extracellular matrix remodeling resulting in fibrotic tissue deposition in skeletal muscles is a hallmark of muscular dystrophies (Serrano and Muñoz-Cánoves, 2017). In LAMA2-CMD, fibrotic tissue deposition plays a central role in disease progression (Accorsi et al., 2020). In this study, we showed that treatment with vemurafenib significantly reduced fibrosis and restored the TGF-β/SMAD3 signaling pathway to the basal levels.

We also analyzed the effects of vemurafenib on the inflammatory response, another important feature of LAMA2-CMD (Pegoraro et al., 1996). We did not observe an overall improvement in the inflammatory response after the treatment with vemurafenib. The reduced levels of the chemoattractants eotaxin and MIG were not sufficient to decrease the infiltration of immune cells in the muscle of dyW−/− vemurafenib-treated mice. However, as we focused the analysis only on the pan-leukocyte marker CD45, we cannot exclude possible effects of vemurafenib in the differentiation, activation and recruitment of specific leukocyte subpopulations.

We next evaluated the effects of vemurafenib on signaling pathways dysregulated in the context of laminin-α2 deficiency. Vemurafenib was able to restore the mTORC1 signaling pathway but did not inhibit ERK, NFκB and STAT3 activation in dyW−/− mice. It is important to mention that this study demonstrates the overactivation of the mTORC1/p70S6K signaling pathway in the gastrocnemius muscle of dyW−/− mice. The mTOR signaling pathway is well described to be involved in protein synthesis, muscle hypertrophy and growth (Bodine et al., 2001; Schiaffino et al., 2021). However, sustained activation of mTORC1 can lead to pulmonary fibrosis (Gui et al., 2015) and drive neuromuscular junction structural alterations, resulting in myofiber denervation (Ang et al., 2022), muscle atrophy, loss of muscle mass (Tang et al., 2014, 2019) and late-onset myopathy by increasing the expression of the E3 ubiquitin ligases atrogin1 and MuRF1 and impairing autophagy (Castets et al., 2013). Inhibition of mTORC1 by rapamycin improves muscle pathology in the fukutin-deficient mouse model of dystroglycanopathy (Foltz et al., 2016) and the mdx mouse model of Duchenne muscular dystrophy (Eghtesad et al., 2011).

Our results did not show a correlation between the upregulation of the mTORC1/p70S6K signaling pathway with the atrogin1 and MuRF1 protein levels detected in gastrocnemius of dyW−/− mice, suggesting that in our animal model at 8 weeks of age, the overactivation of the mTORC1/p70S6K pathway does not modulate the ubiquitin-proteasome system. Interestingly, studies have shown an increase in global protein ubiquitination and mRNA expression of atrogin1 and MuRF1 in the dy3k/dy3k mouse model of LAMA2-CMD (Carmignac et al., 2011a). dy3k/dy3k mice have a complete deficiency of laminin-α2 (Miyagoe et al., 1997), whereas dyW−/− mice produce a small amount of truncated laminin-α2 protein lacking the LN domain (Guo et al., 2003). Therefore, we suggest that the differences in the ubiquitin-proteasome activation observed could be related to the different levels of laminin-α2 chain expression in the two animal models, which results in different phenotypes, disease progression and lifespan (Gawlik and Durbeej, 2020) and will likely impact laminin-α2-related signaling pathways in the skeletal muscle. However, we did observe an increase in the levels of p62, indicating that autophagy might be impaired in dyW−/− mice. As we did not observe any difference in the levels of beclin-1, another protein involved in autophagy, further studies to evaluate other autophagic markers are essential to confirm whether autophagy is impaired in dyW−/− mice at 8 weeks of age.

Together, our results indicate that vemurafenib partially improves muscle histopathology by reducing the percentage of CNFs, increasing the percentage of muscle fibers with bigger diameter in the TA, reducing fibrosis, and restoring the TGF-β/SMAD3 and mTORC1/p70S6K signaling pathways to WT levels in hindlimbs of dyW−/− mice. Future studies will be necessary to understand the correlation of mTORC1 overactivation with the neuropathy observed in the hindlimbs of the dyW−/− mice, as chronic activation of mTORC1 could drive neuromuscular junction structural alterations and myofiber denervation (Ang et al., 2022).

As a pharmacological therapy that modulates intracellular signaling pathways involved in fibrosis and metabolism, vemurafenib is not capable of restoring the myomatrix and, consequently, does not improve muscle strength in dyW−/− mice. However, vemurafenib was able to partially improve muscle histopathology and restore the TGF-β/SMAD3 and mTORC1/p70S6K signaling pathways to WT levels. Therefore, combinatorial treatment with vemurafenib and therapeutics that restore the myomatrix and improve other pathological features in LAMA2-CMD (Erb et al., 2009; Yu et al., 2013; Barraza-Flores et al., 2020; Rooney, 2012; van Ry et al., 2014) might be a more effective therapeutic approach for LAMA2-CMD and should be investigated in future preclinical studies.

Study design

To evaluate the benefits of vemurafenib as a therapeutic for LAMA2-CMD, we performed a short-term treatment with 5 mg/kg vemurafenib (MedChemExpress, HY-12057) in the dyw−/− mouse model of LAMA2-CMD from 3 to 8 weeks of age. The dyw−/− mouse model is a severe model of the disease presenting low levels of truncated laminin-α2 chain, which represents fairly well a group of patients with LAMA2-CMD, and it is considered a relevant model for LAMA2-CMD preclinical studies. The mice were treated four times a week in the morning via oral gavage with 5 mg/kg vemurafenib diluted in 10% DMSO, 40% PEG300 (MedChemExpress, HY-Y0873), 5% Tween-80 (MedChemExpress, HY-Y1891) and 45% saline, as per the manufacturer's recommendations for in vivo administration. The vehicle-treated mice were treated with an equal volume of vehicle solution (10% DMSO, 40% PEG300, 5% Tween-80 and 45% saline). After 5 weeks of treatment, we evaluated skeletal muscle histology, apoptosis, hydroxyproline content, inflammation, intracellular signaling pathways and skeletal muscle function. Animals were assigned randomly to experimental groups and analyses were performed blinded to treatment allocation. Different doses of vemurafenib were tested and the most efficient dose (5 mg/kg) to reduce hydroxyproline content (measurement of fibrosis) was chosen (Fig. S1). Initial analysis to verify gender effects was made on five males and five females for each group. No clear differences were observed (Fig. S2) and both genders were used for the final analysis. The minimal number of animals in each experiment was determined using power analysis (power=0.8, α=0.05 and r=0.7).

Animals

Heterozygous dyW mice (Kuang et al., 1998) (gift from Eva Engvall via Paul Martin; The Ohio State University, Columbus, OH, USA) were crossed to obtain homozygous dyw−/− mutants. The animals were treated according to the rules and regulations specified in the approved protocol from the University of Nevada Reno Institutional Animal Care and Use Committee. At the end of the study, mice were sacrificed by CO2 asphyxiation followed by cervical dislocation under the American Veterinary Medical Association guidelines for euthanasia. All mice were maintained in a pathogen-free animal care facility with access to food and water ad libitum.

Protein extraction

Gastrocnemius protein was extracted in RIPA buffer (50 mM Tris pH 7.4, 1% NP-40, 0.5% sodium deoxycholate, 0.1% SDS, 150 mM NaCl, 2 mM EDTA, 50 mM NaF) containing protease inhibitor cocktail, sodium fluoride (NaF) and sodium orthovanadate (Na3VO4) phosphatase inhibitors. Extracts were centrifuged at 14,000 g at 4°C for 5 min to obtain the supernatant. Protein quantification was performed using the Pierce BCA protein assay kit (Thermo Fisher Scientific, 23227) according to the manufacturer's recommendations.

Enzyme-linked immunosorbent assay

Enzyme-linked immunosorbent assay (ELISA) kits were used to measure levels of active caspase 3 (MyBioSource, MBS7210856), TGF-β1 (Abcam, ab119557), phosphorylated SMAD3 (pS423/S425) (Abcam, 186038), phosphorylated mTOR (pS2448) (RayBiotech, PEL-mTOR-S2448), phosphorylated p70S6K (pT389) (Abcam, ab176651), phosphorylated ERK1/2 (pT202/Y204) (Abcam, ab176640), phosphorylated STAT3 (pY705) (Invitrogen, KHO0481), phosphorylated NFκB p65 (pS536) (Abcam, ab176647), phosphorylated JNK1/2 (pT183/Y185) (Abcam, ab176645), atrogin1 (LSBio, LS-F35338), MuRF1 (MyBioSource, MBS2502946), beclin-1 (LSBio, LS-F35824) and p62 (MyBioSource, MBS039475) in the gastrocnemius protein homogenate, according to the manufacturer's instructions.

Luminex xMAP immunoassay

Luminex xMAP immunoassay was performed in the University of California, Los Angeles (UCLA) Immune Assessment Core to quantify cytokine levels in the gastrocnemius protein homogenate. Mouse magnetic cytokine/chemokine kits were purchased from EMD Millipore and used per the manufacturer's instructions. Briefly, 25 μl diluted (1:2) samples were mixed with 25 μl magnetic beads and allowed to incubate overnight at 4°C while shaking. After washing the plates twice with wash buffer in a Biotek ELx405 washer, 25 μl of biotinylated detection antibody was added and the samples were incubated for 1 h at room temperature. Then, 25 μl streptavidin-phycoerythrin conjugate was added to the reaction mixture and incubated for another 30 min at room temperature. Following two washes, the beads were resuspended in sheath fluid, and fluorescence was quantified using a Luminex 200TM instrument (Luminex Corporation, TX, USA). Samples presenting cytokine levels below the Luminex detection limit were removed from the analysis.

Immunofluorescence

Freshly collected tibialis anterior (TA) and triceps brachii muscles were rinsed in PBS and placed into a 2:3 (v/v) optimum cutting temperature compound (Tissue-TEK OCT compound, Sakura Finetek, 4583) to 30% sucrose/PBS medium inside a cryomold and frozen in liquid nitrogen-cooled isopentane. Tissues were then cryosectioned at 10 μm thickness using a Leica CM1950 cryostat. Sections were fixed in cold 4% paraformaldehyde in PBS for 10 min and incubated overnight with the following primary antibodies: anti-eMHC [Developmental Studies Hybridoma Bank (DSHB), F1.652-s; 5 µg/ml], anti-dystrophin [DSHB, MANDRA1 (7A10); 5 µg/ml], anti-dystrophin (Abcam, ab15277; 4 µg/ml) and anti-CD45 (Abcam, ab10558; 50 µg/ml). After washing with PBS, muscle sections were incubated with secondary antibodies conjugated with Alexa Fluor 488 (Invitrogen, A11001; 2 µg/ml) and Alexa Fluor 546 (Invitrogen, A11035; 2 µg/ml) at room temperature for 45 min and mounted with Vectashield Antifade Mounting Medium with 4′,6-diamidino-2-phenylindole (DAPI) (Vector Laboratories, H-1200-10). When staining with monoclonal mouse antibodies, the Mouse-On-Mouse (MOM) kit (Vector Laboratories, FMK-2201) was used.

A series of three non-consecutive sections were acquired using the Keyence digital microscope (Keyence Corporation of America, IL USA) and the images were analyzed using Image J software (National Institutes of Health, USA). The measurements of the number of fibers, fiber MinFeret diameter, CNFs, eMHC-positive fibers and the CD45-positive area were performed on three stitched whole sections of the TA muscle and averaged for each animal.

Hydroxyproline assay

Quadricep muscle was minced overnight in 2 ml of 6 M hydrochloric acid at 110°C and the resulting hydrolysate (10 µl) was mixed with 150 µl isopropanol. Hydroxyproline oxidation was then performed for 10 min at room temperature with the addition of 72 µl of 1.4% chloramine-T (Sigma-Aldrich, 402869) in citrate buffer (0.385 M sodium acetate trihydrate, 0.24 M citric acid, 1.2% acetic acid, 0.85 M sodium hydroxide). Ehrlich's reagent [1 ml; 7.5g of 4-(dimethylamino) benzaldehyde, 25 ml ethanol, 1688 μl sulfuric acid] was then added and incubated for 30 min at 55°C. The absorbance measurement was performed at 558 nm, as previously described (Heydemann et al., 2005).

Ex vivo contractility assay

Extensor digitorum longus (EDL) muscles were dissected from deeply anesthetized mice with 2.5% isoflurane and mounted between two platinum electrodes, clamped at one tendon, and attached at the other tendon to a force transducer placed in an oxygenated bath containing a physiologic salt solution (PSS buffer, pH 7.6) at 30°C. Experiments were performed using the isolated muscle test system for mice (Aurora Scientific) as described previously (https://treat-nmd.org/wp-content/uploads/2016/08/cmd-DMD_M.1.2.002.pdf; Sperringer and Grange, 2016). Data were analyzed using DMA software (Aurora Scientific), and the force was normalized by the EDL muscle weight.

Statistical analysis

All data are expressed as mean±standard error mean (s.e.m.). The means of all data that followed the normal distribution were analyzed using one-way ANOVA with uncorrected Fisher's least significant difference (LSD) test, and the data that did not follow the normal distribution were analyzed using the Kruskal–Wallis test. Twitch and tetanus data were analyzed using one-way ANOVA with uncorrected Fisher's LSD test, and force-frequency data were analyzed using two-way ANOVA. Statistical analyses were performed using GraphPad Prism 9 software. P-values, n-values and symbols are described in the figure legends. *P<0.05, **P<0.01, ***P<0.001, ****P<0.0001 were considered statistically significant.

We would like to acknowledge use of the COBRE High Spatial and Temporal Resolution Imaging (HSTRI) Core, which was supported by a grant from the National Institute of General Medical Sciences (P20GM130459).

Author contributions

Conceptualization: A.O.-S., D.J.B.; Methodology: A.O.-S., M.D., J.W., R.S., D.J.B.; Validation: D.J.B.; Formal analysis: A.O.-S.; Investigation: A.O.-S.; Data curation: A.O.-S., J.W., R.S.; Writing - original draft: A.O.-S.; Writing - review & editing: M.D., D.J.B.; Supervision: D.J.B.; Project administration: D.J.B.; Funding acquisition: D.J.B.

Funding

This study was supported by a grant from the Muscular Dystrophy Association (MDA628561) to D.J.B. A.O.-S. was supported by a Raymond H. Berner Graduate School Scholarship. Open Access funding provided by University of Nevada, Reno. Deposited in PMC for immediate release.

Data availability

All data associated with this study are present in the paper or supplementary materials.

Accorsi
,
A.
,
Kumar
,
A.
,
Rhee
,
Y.
,
Miller
,
A.
and
Girgenrath
,
M.
(
2016
).
IGF-1/GH axis enhances losartan treatment in Lama2-related muscular dystrophy
.
Hum. Mol. Genet.
25
,
4624
-
4634
.
Accorsi
,
A.
,
Cramer
,
M. L.
and
Girgenrath
,
M.
(
2020
).
Fibrogenesis in LAMA2-related muscular dystrophy is a central tenet of disease etiology
.
Front. Mol. Neurosci.
13
,
1
-
16
.
Allamand
,
V.
and
Guicheney
,
P.
(
2002
).
Merosin-deficient congenital muscular dystrophy, autosomal recessive (MDC1A, MIM#156225, LAMA2 gene coding for α2 chain of laminin)
.
Eur. J. Hum. Genet.
10
,
91
-
94
.
Ang
,
S.-T. J.
,
Crombie
,
E. M.
,
Dong
,
H.
,
Tan
,
K.-T.
,
Hernando
,
A.
,
Yu
,
D.
,
Adamson
,
S.
,
Kim
,
S.
,
Withers
,
D. J.
,
Huang
,
H.
et al. 
(
2022
).
Muscle 4EBP1 activation modifies the structure and function of the neuromuscular junction in mice
.
Nat. Commun.
13
,
7792
.
Aoki
,
Y.
,
Nagata
,
T.
,
Yokota
,
T.
,
Nakamura
,
A.
,
Wood
,
M. J. A.
,
Partridge
,
T.
and
Takeda
,
S.
(
2013
).
Highly efficient in vivo delivery of PMO into regenerating myotubes and rescue in laminin-α2 chain-null congenital muscular dystrophy mice
.
Hum. Mol. Genet.
22
,
4914
-
4928
.
Aumailley
,
M.
,
Bruckner-Tuderman
,
L.
,
Carter
,
W. G.
,
Deutzmann
,
R.
,
Edgar
,
D.
,
Ekblom
,
P.
,
Engel
,
J.
,
Engvall
,
E.
,
Hohenester
,
E.
,
Jones
,
J. C. R.
et al. 
(
2005
).
A simplified laminin nomenclature
.
Matrix Biol.
24
,
326
-
332
.
Barraza-Flores
,
P.
,
Bukovec
,
K. E.
,
Dagda
,
M.
,
Conner
,
B. W.
,
Oliveira-Santos
,
A.
,
Grange
,
R. W.
and
Burkin
,
D. J.
(
2020
).
Laminin-111 protein therapy after disease onset slows muscle disease in a mouse model of laminin-α2 related congenital muscular dystrophy
.
Hum. Mol. Genet.
29
,
2162
-
2170
.
Bautista
,
F.
,
Paci
,
A.
,
Minard-Colin
,
V.
,
Dufour
,
C.
,
Grill
,
J.
,
Lacroix
,
L.
,
Varlet
,
P.
,
Valteau-Couanet
,
D.
,
Geoerger
,
B.
,
Melchionda
,
F.
et al. 
(
2014
).
Vemurafenib in pediatric patients with BRAFV600E mutated high-grade gliomas
.
Pediatr. Blood Cancer
61
,
1101
-
1103
.
Bodine
,
S. C.
,
Stitt
,
T. N.
,
Gonzalez
,
M.
,
Kline
,
W. O.
,
Stover
,
G. L.
,
Bauerlein
,
R.
,
Zlotchenko
,
E.
,
Scrimgeour
,
A.
,
Lawrence
,
J. C.
,
Glass
,
D. J.
et al. 
(
2001
).
Akt/mTOR pathway is a crucial regulator of skeletal muscle hypertrophy and can prevent muscle atrophy in vivo
.
Nat. Cell Biol.
3
,
1014
-
1019
.
Carmignac
,
V.
,
Quéré
,
R.
and
Durbeej
,
M.
(
2011a
).
Proteasome inhibition improves the muscle of laminin α2 chain-deficient mice
.
Hum. Mol. Genet.
20
,
541
-
552
.
Carmignac
,
V.
,
Svensson
,
M.
,
Körner
,
Z.
,
Elowsson
,
L.
,
Matsumura
,
C.
,
Gawlik
,
K. I.
,
Allamand
,
V.
and
Durbeej
,
M.
(
2011b
).
Autophagy is increased in laminin α2 chain-deficient muscle and its inhibition improves muscle morphology in a mouse model of MDC1A
.
Hum. Mol. Genet.
20
,
4891
-
4902
.
Carraro
,
U.
and
Kern
,
H.
(
2016
).
Severely atrophic human muscle fibers with nuclear misplacement survive many years of permanent denervation
.
Eur. J. Transl. Myol.
26
,
76
-
80
.
Castets
,
P.
,
Lin
,
S.
,
Rion
,
N.
,
di Fulvio
,
S.
,
Romanino
,
K.
,
Guridi
,
M.
,
Frank
,
S.
,
Tintignac
,
L. A.
,
Sinnreich
,
M.
and
Rüegg
,
M. A.
(
2013
).
Sustained activation of mTORC1 in skeletal muscle inhibits constitutive and starvation-induced autophagy and causes a severe, late-onset myopathy
.
Cell Metab.
17
,
731
-
744
.
Chakraborty
,
D.
,
Šumová
,
B.
,
Mallano
,
T.
,
Chen
,
C.-W.
,
Distler
,
A.
,
Bergmann
,
C.
,
Ludolph
,
I.
,
Horch
,
R. E.
,
Gelse
,
K.
,
Ramming
,
A.
et al. 
(
2017
).
Activation of STAT3 integrates common profibrotic pathways to promote fibroblast activation and tissue fibrosis
.
Nat. Commun.
8
,
1130
.
Chiarini
,
F.
,
Evangelisti
,
C.
,
Cenni
,
V.
,
Fazio
,
A.
,
Paganelli
,
F.
,
Martelli
,
A. M.
and
Lattanzi
,
G.
(
2019
).
The cutting edge: The role of mTOR signaling in laminopathies
.
Int. J. Mol. Sci.
20
,
847
.
Connolly
,
A. M.
,
Keeling
,
R. M.
,
Streif
,
E. M.
,
Pestronk
,
A.
and
Mehta
,
S.
(
2002
).
Complement 3 deficiency and oral prednisolone improve strength and prolong survival of laminin α2-deficient mice
.
J. Neuroimmunol.
127
,
80
-
87
.
Czirbesz
,
K.
,
Gorka
,
E.
,
Balatoni
,
T.
,
Pánczél
,
G.
,
Melegh
,
K.
,
Kovács
,
P.
,
Gézsi
,
A.
and
Liszkay
,
G.
(
2019
).
Efficacy of Vemurafenib treatment in 43 metastatic melanoma patients with BRAF mutation. Single-institute retrospective analysis, early real-life survival data
.
Pathol. Oncol. Res.
25
,
45
-
50
.
Dadush
,
O.
,
Aga-Mizrachi
,
S.
,
Ettinger
,
K.
,
Tabakman
,
R.
,
Elbaz
,
M.
,
Fellig
,
Y.
,
Yanay
,
N.
and
Nevo
,
Y.
(
2010
).
Improved muscle strength and mobility in the dy2J/dy2J mouse with merosin deficient congenital muscular dystrophy treated with Glatiramer acetate
.
Neuromuscul. Disord.
20
,
267
-
272
.
Daou
,
N.
,
Hassani
,
M.
,
Matos
,
E.
,
de Castro
,
G. S.
,
Costa
,
R. G. F.
,
Seelaender
,
M.
,
Moresi
,
V.
,
Rocchi
,
M.
,
Adamo
,
S.
,
Li
,
Z.
et al. 
(
2020
).
Displaced myonuclei in cancer cachexia suggest altered innervation
.
Int. J. Mol. Sci.
21
,
1
-
17
.
de Oliveira
,
B. M.
,
Matsumura
,
C. Y.
,
Fontes-Oliveira
,
C. C.
,
Gawlik
,
K. I.
,
Acosta
,
H.
,
Wernhoff
,
P.
and
Durbeej
,
M.
(
2014
).
Quantitative proteomic analysis reveals metabolic alterations, calcium dysregulation, and increased expression of extracellular matrix proteins in laminin α2 chain–deficient muscle
.
Mol. Cell. Proteomics
13
,
3001
-
3013
.
Doe
,
J. A.
,
Wuebbles
,
R. D.
,
Allred
,
E. T.
,
Rooney
,
J. E.
,
Elorza
,
M.
and
Burkin
,
D. J.
(
2011
).
Transgenic overexpression of the α7 integrin reduces muscle pathology and improves viability in the dy(W) mouse model of merosin-deficient congenital muscular dystrophy type 1A
.
J. Cell Sci.
124
,
2287
-
2297
.
Durbeej
,
M.
(
2010
).
Laminins
.
Cell Tissue Res.
339
,
259
-
268
.
Durbeej
,
M.
(
2015
).
Laminin-alpha2 chain-deficient congenital muscular dystrophy: pathophysiology and development of treatment
.
Curr. Top. Membr.
76
,
31
-
60
.
Eghtesad
,
S.
,
Jhunjhunwala
,
S.
,
Little
,
S. R.
and
Clemens
,
P. R.
(
2011
).
Rapamycin ameliorates dystrophic phenotype in mdx mouse skeletal muscle
.
Mol. Med.
17
,
917
-
924
.
Elbaz
,
M.
,
Yanay
,
N.
,
Aga-Mizrachi
,
S.
,
Brunschwig
,
Z.
,
Kassis
,
I.
,
Ettinger
,
K.
,
Barak
,
V.
and
Nevo
,
Y.
(
2012
).
Losartan, a therapeutic candidate in congenital muscular dystrophy: studies in the dy2J/dy2J mouse
.
Ann. Neurol.
71
,
699
-
708
.
Elbaz
,
M.
,
Yanay
,
N.
,
Laban
,
S.
,
Rabie
,
M.
,
Mitrani-Rosenbaum
,
S.
and
Nevo
,
Y.
(
2015
).
Life or death by NFκB, Losartan promotes survival in dy2J/dy2J mouse of MDC1A
.
Cell Death Dis.
6
,
1
-
9
.
Erb
,
M.
,
Meinen
,
S.
,
Barzaghi
,
P.
,
Sumanovski
,
L. T.
,
Courdier-Früh
,
I.
,
Rüegg
,
M. A.
and
Meier
,
T.
(
2009
).
Omigapil ameliorates the pathology of muscle dystrophy caused by Laminin-α2 deficiency
.
J. Pharmacol. Exp. Ther.
331
,
787
-
795
.
Evseev
,
D.
,
Kalinina
,
I.
,
Raykina
,
E.
,
Osipova
,
D.
,
Abashidze
,
Z.
,
Ignatova
,
A.
,
Mitrofanova
,
A.
,
Maschan
,
A.
,
Novichkova
,
G.
and
Maschan
,
M.
(
2021
).
Vemurafenib provides a rapid and robust clinical response in pediatric Langerhans cell histiocytosis with the BRAF V600E mutation but does not eliminate low-level minimal residual disease per ddPCR using cell-free circulating DNA
.
Int. J. Hematol.
114
,
725
-
734
.
Fingar
,
D. C.
and
Blenis
,
J.
(
2004
).
Target of rapamycin (TOR): An integrator of nutrient and growth factor signals and coordinator of cell growth and cell cycle progression
.
Oncogene
23
,
3151
-
3171
.
Foltz
,
S. J.
,
Luan
,
J.
,
Call
,
J. A.
,
Patel
,
A.
,
Peissig
,
K. B.
,
Fortunato
,
M. J.
and
Beedle
,
A. M.
(
2016
).
Four-week rapamycin treatment improves muscular dystrophy in a fukutin-deficient mouse model of dystroglycanopathy
.
Skelet. Muscle
6
,
20
.
Fontes-Oliveira
,
C. C.
,
Soares Oliveira
,
B. M.
,
Körner
,
Z.
,
Harandi
,
V. M.
and
Durbeej
,
M.
(
2018
).
Effects of metformin on congenital muscular dystrophy type 1A disease progression in mice: a gender impact study
.
Sci. Rep.
8
,
16302
.
Gawlik
,
K. I.
and
Durbeej
,
M.
(
2010
).
Transgenic overexpression of laminin α1 chain in laminin α2 chain-deficient mice rescues the disease throughout the lifespan
.
Muscle Nerve
42
,
30
-
37
.
Gawlik
,
K. I.
and
Durbeej
,
M.
(
2011
).
Skeletal muscle laminin and MDC1A: pathogenesis and treatment strategies
.
Skelet. Muscle
1
,
9
.
Gawlik
,
K. I.
and
Durbeej
,
M.
(
2020
).
A family of Laminin α2 chain-deficient mouse mutants: advancing the research on LAMA2-CMD
.
Front. Mol. Neurosci.
13
,
1
-
16
.
Gawlik
,
K.
,
Miyagoe-Suzuki
,
Y.
,
Ekblom
,
P.
,
Takeda
,
S.
and
Durbeej
,
M.
(
2004
).
Laminin α1 chain reduces muscular dystrophy in laminin chain deficient mice
.
Hum. Mol. Genet.
13
,
1775
-
1784
.
Gawlik
,
K. I.
,
Harandi
,
V. M.
,
Cheong
,
R. Y.
,
Petersén
,
Å.
and
Durbeej
,
M.
(
2018
).
Laminin α1 reduces muscular dystrophy in dy2J mice
.
Matrix Biol.
70
,
36
-
49
.
Geranmayeh
,
F.
,
Clement
,
E.
,
Feng
,
L. H.
,
Sewry
,
C.
,
Pagan
,
J.
,
Mein
,
R.
,
Abbs
,
S.
,
Brueton
,
L.
,
Childs
,
A.-M.
,
Jungbluth
,
H.
et al. 
(
2010
).
Genotype-phenotype correlation in a large population of muscular dystrophy patients with LAMA2 mutations
.
Neuromuscul. Disord.
20
,
241
-
250
.
Giguère
,
V.
(
2018
).
Canonical signaling and nuclear activity of mTOR—a teamwork effort to regulate metabolism and cell growth
.
FEBS J.
285
,
1572
-
1588
.
Girgenrath
,
M.
,
Beermann
,
M. L.
,
Vishnudas
,
V. K.
,
Homma
,
S.
and
Miller
,
J. B.
(
2009
).
Pathology is alleviated by doxycycline in a laminin-α2-null model of congenital muscular dystrophy
.
Ann. Neurol.
65
,
47
-
56
.
Glick
,
D.
,
Barth
,
S.
and
Macleod
,
K. F.
(
2010
).
Autophagy: cellular and molecular mechanisms
.
J. Pathol.
221
,
3
-
12
.
Graziano
,
A.
,
Bianco
,
F.
,
D'Amico
,
A.
,
Moroni
,
I.
,
Messina
,
S.
,
Bruno
,
C.
,
Pegoraro
,
E.
,
Mora
,
M.
,
Astrea
,
G.
,
Magri
,
F.
et al. 
(
2015
).
Prevalence of congenital muscular dystrophy in Italy: a population study
.
Neurology
84
,
904
-
911
.
Guadagnin
,
E.
,
Mázala
,
D.
and
Chen
,
Y.-W.
(
2018
).
STAT3 in skeletal muscle function and disorders
.
Int. J. Mol. Sci.
19
,
2265
.
Gui
,
Y.-S.
,
Wang
,
L.
,
Tian
,
X.
,
Li
,
X.
,
Ma
,
A.
,
Zhou
,
W.
,
Zeng
,
N.
,
Zhang
,
J.
,
Cai
,
B.
,
Zhang
,
H.
et al. 
(
2015
).
mTOR overactivation and compromised autophagy in the pathogenesis of pulmonary fibrosis
.
PLoS ONE
10
,
e0138625
.
Guo
,
L. T.
,
Zhang
,
X. U.
,
Kuang
,
W.
,
Xu
,
H.
,
Liu
,
L. A.
,
Vilquin
,
J.-T.
,
Miyagoe-Suzuki
,
Y.
,
Takeda
,
S.
,
Ruegg
,
M. A.
,
Wewer
,
U. M.
et al. 
(
2003
).
Laminin alpha2 deficiency and muscular dystrophy; genotype-phenotype correlation in mutant mice
.
Neuromuscul. Disord.
13
,
207
-
215
.
Hagiwara
,
H.
,
Ohsawa
,
Y.
,
Asakura
,
S.
,
Murakami
,
T.
,
Teshima
,
T.
and
Sunada
,
Y.
(
2006
).
Bone marrow transplantation improves outcome in a mouse model of congenital muscular dystrophy
.
FEBS Lett.
580
,
4463
-
4468
.
Hayes
,
A.
and
Williams
,
D. A.
(
1998
).
Examining potential drug therapies for muscular dystrophy utilising the dy/dy mouse: I. Clenbuterol
.
J. Neurol. Sci.
157
,
122
-
128
.
Helbling-Leclerc
,
A.
,
Zhang
,
X.
,
Topaloglu
,
H.
,
Cruaud
,
C.
,
Tesson
,
F.
,
Weissenbach
,
J.
,
Tomé
,
F. M. S.
,
Schwartz
,
K.
,
Fardeau
,
M.
,
Tryggvason
,
K.
et al. 
(
1995
).
Mutations in the laminin α2-chain gene (LAMA2) cause merosin-deficient congenital muscular dystrophy
.
Nat. Genet.
11
,
216
-
218
.
Heydemann
,
A.
,
Huber
,
J. M.
,
Demonbreun
,
A.
,
Hadhazy
,
M.
and
McNally
,
E. M.
(
2005
).
Genetic background influences muscular dystrophy
.
Neuromuscul. Disord.
15
,
601
-
609
.
Ismaeel
,
A.
,
Kim
,
J.-S.
,
Kirk
,
J. S.
,
Smith
,
R. S.
,
Bohannon
,
W. T.
and
Koutakis
,
P.
(
2019
).
Role of transforming growth factor-β in skeletal muscle fibrosis: a review
.
Int. J. Mol. Sci.
20
,
2446
.
Jones
,
K. J.
,
Morgan
,
G.
,
Johnston
,
H.
,
Tobias
,
V.
,
Ouvrier
,
R. A.
,
Wilkinson
,
I.
and
North
,
K. N.
(
2001
).
The expanding phenotype of laminin α2 chain (merosin) abnormalities: case series and review
.
J. Med. Genet.
38
,
649
-
657
.
Kemaladewi
,
D. U.
,
Maino
,
E.
,
Hyatt
,
E.
,
Hou
,
H.
,
Ding
,
M.
,
Place
,
K. M.
,
Zhu
,
X.
,
Bassi
,
P.
,
Baghestani
,
Z.
,
Deshwar
,
A. G.
et al. 
(
2017
).
Correction of a splicing defect in a mouse model of congenital muscular dystrophy type 1A using a homology-directed-repair-independent mechanism
.
Nat. Med.
23
,
984
-
989
.
Khalil
,
R.
(
2018
).
Ubiquitin-proteasome pathway and muscle atrophy
.
Adv. Exp. Med. Biol.
1088
,
235
-
248
.
Kim
,
E. K.
and
Choi
,
E.-J.
(
2015
).
Compromised MAPK signaling in human diseases: an update
.
Arch. Toxicol.
89
,
867
-
882
.
Körner
,
Z.
and
Durbeej
,
M.
(
2016
).
Bortezomib does not reduce muscular dystrophy in the dy2J/dy2J mouse model of Laminin α2 chain-deficient muscular dystrophy
.
PLoS ONE
11
,
e0146471
.
Körner
,
Z.
,
Fontes-Oliveira
,
C. C.
,
Holmberg
,
J.
,
Carmignac
,
V.
and
Durbeej
,
M.
(
2014
).
Bortezomib partially improves laminin α2 chain deficient muscular dystrophy
.
Am. J. Pathol.
184
,
1518
-
1528
.
Kuang
,
W.
,
Xu
,
H.
,
Vachon
,
P. H.
,
Liu
,
L.
,
Loechel
,
F.
,
Wewer
,
U. M.
and
Engvall
,
E.
(
1998
).
Merosin-deficient congenital muscular dystrophy: partial genetic correction in two mouse models
.
J. Clin. Investig.
102
,
844
-
852
.
Kuang
,
W.
,
Xu
,
H.
,
Vilquin
,
J. T.
and
Engvall
,
E.
(
1999
).
Activation of the lama2 gene in muscle regeneration: Abortive regeneration in laminin α2-deficiency
.
Lab. Investig.
79
,
1601
-
1613
.
Li
,
H.
,
Wang
,
Y.
,
Su
,
R.
,
Jia
,
Y.
,
Lai
,
X.
,
Su
,
H.
,
Fan
,
Y.
,
Wang
,
Y.
,
Xing
,
W.
and
Qin
,
J.
(
2022
).
Dimethyl fumarate combined with vemurafenib enhances anti-melanoma efficacy via inhibiting the Hippo/YAP, NRF2-ARE, and AKT/mTOR/ERK pathways in A375 melanoma cells
.
Front. Oncol.
12
,
1
-
16
.
Luke
,
J. J.
and
Hodi
,
F. S.
(
2012
).
Vemurafenib and BRAF inhibition: a new class of treatment for metastatic melanoma
.
Clin. Cancer Res.
18
,
9
-
14
.
Lynch
,
G. S.
,
Cuffe
,
S. A.
,
Plant
,
D. R.
and
Gregorevic
,
P.
(
2001
).
IGF-I treatment improves the functional properties of fast- and slow-twitch skeletal muscles from dystrophic mice
.
Neuromuscul. Disord.
11
,
260
-
268
.
Ma
,
J. F.
,
Sanchez
,
B. J.
,
Hall
,
D. T.
,
Tremblay
,
A. M. K.
,
di Marco
,
S.
and
Gallouzi
,
I. E.
(
2017
).
STAT3 promotes IFNγ/TNFα–induced muscle wasting in an NF–κB–dependent and IL–6–independent manner
.
EMBO Mol Med
9
,
622
-
637
.
Macdonald
,
S. G.
,
Crews
,
C. M.
,
Wu
,
L.
,
Driller
,
J.
,
Clark
,
R.
,
Erikson
,
R. L.
and
McCormick
,
F.
(
1993
).
Reconstitution of the Raf-1-MEK-ERK signal transduction pathway in vitro
.
Mol. Cell. Biol.
13
,
6615
-
6620
.
McCubrey
,
J. A.
,
Steelman
,
L. S.
,
Chappell
,
W. H.
,
Abrams
,
S. L.
,
Wong
,
E. W. T.
,
Chang
,
F.
,
Lehmann
,
B.
,
Terrian
,
D. M.
,
Milella
,
M.
,
Tafuri
,
A.
et al. 
(
2007
).
Roles of the Raf/MEK/ERK pathway in cell growth, malignant transformation, and drug resistance
.
Biochim. Biophys. Acta
1773
,
1263
-
1283
.
McKee
,
K. K.
,
Capizzi
,
S.
and
Yurchenco
,
P. D.
(
2009
).
Scaffold-forming and adhesive contributions of synthetic laminin-binding proteins to basement membrane assembly
.
J. Biol. Chem.
284
,
8984
-
8994
.
Mehuron
,
T.
,
Kumar
,
A.
,
Duarte
,
L.
,
Yamauchi
,
J.
,
Accorsi
,
A.
and
Girgenrath
,
M.
(
2014
).
Dysregulation of matricellular proteins is an early signature of pathology in laminin-deficientmuscular dystrophy
.
Skelet. Muscle
4
,
14
.
Meinen
,
S.
,
Lin
,
S.
and
Ruegg
,
M. A.
(
2012
).
Angiotensin II type 1 receptor antagonists alleviate muscle pathology in the mouse model for laminin-α2-deficient congenital muscular dystrophy (MDC1A)
.
Skelet. Muscle
2
,
18
.
Mercuri
,
E.
,
Pennock
,
J.
,
Goodwin
,
F.
,
Sewry
,
C.
,
Cowan
,
F.
,
Dubowitz
,
L.
,
Dubowitz
,
V.
and
Muntoni
,
F.
(
1996
).
Sequential study of central and peripheral nervous system involvement in an infant with merosin-deficient congenital muscular dystrophy
.
Neuromuscul. Disroder.
6
,
425
-
429
.
Millay
,
D. P.
,
Sargent
,
M. A.
,
Osinska
,
H.
,
Baines
,
C. P.
,
Barton
,
E. R.
,
Vuagniaux
,
G.
,
Sweeney
,
H. L.
,
Robbins
,
J.
and
Molkentin
,
J. D.
(
2008
).
Genetic and pharmacologic inhibition of mitochondrial-dependent necrosis attenuates muscular dystrophy
.
Nat. Med.
14
,
442
-
447
.
Miyagoe
,
Y.
,
Hanaoka
,
K.
,
Nonaka
,
I.
,
Hayasaka
,
M.
,
Nabeshima
,
Y.
,
Arahata
,
K.
,
Nabeshima
,
Y.-I.
and
Takeda
,
S.
(
1997
).
Laminin α2 chain-null mutant mice by targeted disruption of the Lama2 gene: a new model of merosin (laminin 2)-deficient congenital muscular dystrophy
.
FEBS Lett.
415
,
33
-
39
.
Miyagoe-Suzuki
,
Y.
,
Nakagawa
,
M.
and
Takeda
,
S. I.
(
2000
).
Merosin and congenital muscular dystrophy
.
Microsc. Res. Tech.
48
,
181
-
191
.
Moll
,
J.
,
Barzaghi
,
P.
,
Lin
,
S.
,
Bezakova
,
G.
,
Lochmüller
,
H.
,
Engvall
,
E.
,
Müller
,
U.
and
Ruegg
,
M. A.
(
2001
).
An agrin minigene rescues dystrophic symptoms in a mouse model for congenital muscular dystrophy
.
Nature
413
,
302
-
307
.
Muchir
,
A.
,
Kim
,
Y. J.
,
Reilly
,
S. A.
,
Wu
,
W.
,
Choi
,
J. C.
and
Worman
,
H. J.
(
2013
).
Inhibition of extracellular signal-regulated kinase 1/2 signaling has beneficial effects on skeletal muscle in a mouse model of Emery-Dreifuss muscular dystrophy caused by lamin A/C gene mutation
.
Skelet. Muscle
3
,
17
.
Mulder
,
S. E.
,
Dasgupta
,
A.
,
King
,
R. J.
,
Abrego
,
J.
,
Attri
,
K. S.
,
Murthy
,
D.
,
Shukla
,
S. K.
and
Singh
,
P. K.
(
2020
).
JNK signaling contributes to skeletal muscle wasting and protein turnover in pancreatic cancer cachexia
.
Cancer Lett.
491
,
70
-
77
.
Muntoni
,
F.
and
Voit
,
T.
(
2004
).
The congenital muscular dystrophies in 2004: a century of exciting progress
.
Neuromuscul. Disord.
14
,
635
-
649
.
Nakagawa
,
M.
,
Miyagoe-Suzuki
,
Y.
,
Ikezoe
,
K.
,
Miyata
,
Y.
,
Nonaka
,
I.
,
Harii
,
K.
and
Takeda
,
S. I.
(
2001
).
Schwann cell myelination occurred without basal lamina formation in laminin α2 chain-null mutant (dy3K/dy3K) mice
.
Glia
35
,
101
-
110
.
Nevo
,
Y.
,
Halevy
,
O.
,
Genin
,
O.
,
Moshe
,
I.
,
Turgeman
,
T.
,
Harel
,
M.
,
Biton
,
E.
,
Reif
,
S.
and
Pines
,
M.
(
2010
).
Fibrosis inhibition and muscle histopathology improvement in laminin-α2-deficient mice
.
Muscle Nerve
42
,
218
-
229
.
Nevo
,
Y.
,
Aga-Mizrachi
,
S.
,
Elmakayes
,
E.
,
Yanay
,
N.
,
Ettinger
,
K.
,
Elbaz
,
M.
,
Brunschwig
,
Z.
,
Dadush
,
O.
,
Elad-Sfadia
,
G.
,
Haklai
,
R.
et al. 
(
2011
).
The Ras Antagonist, Farnesylthiosalicylic Acid (FTS), decreases fibrosis and improves muscle strength in dy2J/dy2J mouse model of muscular dystrophy
.
PLoS ONE
6
,
e18049
.
Nguyen
,
Q.
,
Lim
,
K. R. Q.
and
Yokota
,
T.
(
2019
).
Current understanding and treatment of cardiac and skeletal muscle pathology in laminin-α2 chain-deficient congenital muscular dystrophy
.
Appl. Clin. Genet.
12
,
113
-
130
.
Norwood
,
F. L. M.
,
Harling
,
C.
,
Chinnery
,
P. F.
,
Eagle
,
M.
,
Bushby
,
K.
and
Straub
,
V.
(
2009
).
Prevalence of genetic muscle disease in Northern England: In-depth analysis of a muscle clinic population
.
Brain
132
,
3175
-
3186
.
Nunes
,
A. M.
,
Wuebbles
,
R. D.
,
Sarathy
,
A.
,
Fontelonga
,
T. M.
,
Deries
,
M.
,
Burkin
,
D. J.
and
Thorsteinsdóttir
,
S.
(
2017
).
Impaired fetal muscle development and JAK-STAT activation mark disease onset and progression in a mouse model for merosin-deficient congenital muscular dystrophy
.
Hum. Mol. Genet.
26
,
2018
-
2033
.
Oliveira
,
J.
,
Freixo
,
J. P.
,
Santos
,
M.
and
Coelho
,
T.
(
2020
).
LAMA2 muscular dystrophy
. In
GeneReviews® [Internet]
(ed.
M. P.
Adam
,
G. M.
Mirzaa
,
R. A.
Pagon
,
S. E.
Wallace
,
L. J. H.
Bean
,
K. W.
Gripp
and
A.
Amemiya
), pp.
1
-
25
.
Seattle (WA)
:
University of Washington
.
Paparo
,
S. R.
(
2019
).
The MIG chemokine in inflammatory myopathies
.
Clin. Terapeutica
1
,
e55
-
e60
.
Patton
,
B. L.
(
2000
).
Laminins of the neuromuscular system
.
Microsc. Res. Tech
51
,
247
-
261
.
Patton
,
B. L.
,
Miner
,
J. H.
,
Chiu
,
A. Y.
and
Sanes
,
J. R.
(
1997
).
Distribution and function of laminins in the neuromuscular system of developing, adult, and mutant mice
.
J. Cell Biol.
139
,
1507
-
1521
.
Pegoraro
,
E.
,
Mancias
,
P.
,
Swerdlow
,
S. H.
,
Raikow
,
R. B.
,
Garcia
,
C.
,
Marks
,
H.
,
Crawford
,
S. T.
,
Carver
,
V.
,
di Cianno
,
B.
,
Hoffman
,
E. I.
et al. 
(
1996
).
Congenital muscular dystrophy with primary laminin-A2 (merosin) deficiency presenting as inflammatory myopathy
.
Annal. Neurol.
40
,
782
-
791
.
Penna
,
F.
,
Costamagna
,
D.
,
Fanzani
,
A.
,
Bonelli
,
G.
,
Baccino
,
F. M.
and
Costelli
,
P.
(
2010
).
Muscle wasting and impaired myogenesis in tumor bearing mice are prevented by ERK inhibition
.
PLoS ONE
5
,
1
-
11
.
Peti
,
W.
and
Page
,
R.
(
2013
).
Molecular basis of MAP kinase regulation
.
Protein Sci.
22
,
1698
-
1710
.
Philpot
,
J.
,
Sewry
,
C.
,
Pennock
,
J.
and
Dubowitz
,
V.
(
1995
).
Clinical phenotype in congenital muscular dystrophy: correlation with expression of merosin in skeletal muscle
.
Neuromuscul. Disord.
5
,
301
-
305
.
Previtali
,
S. C.
and
Zambon
,
A. A.
(
2020
).
LAMA2 neuropathies: human findings and pathomechanisms from mouse models
.
Front. Mol. Neurosci.
13
,
1
-
10
.
Qiao
,
C.
,
Li
,
J.
,
Zhu
,
T.
,
Draviam
,
R.
,
Watkins
,
S.
,
Ye
,
X.
,
Chen
,
C.
,
Li
,
J.
and
Xiao
,
X.
(
2005
).
Amelioration of laminin-α2-deficient congenital muscular dystrophy by somatic gene transfer of miniagrin
.
Proc. Natl. Acad. Sci. USA
102
,
11999
-
12004
.
Quijano-Roy
,
S.
,
Renault
,
F.
,
Romero
,
N.
,
Guicheney
,
P.
,
Fardeau
,
M.
and
Estournet
,
B.
(
2004
).
EMG and nerve conduction studies in children with congenital muscular dystrophy
.
Muscle Nerve
29
,
292
-
299
.
Rankin
,
S. M.
,
Conroy
,
D. M.
and
Williams
,
T. J.
(
2000
).
Eotaxin and eosinophil recruitment: implications for human disease
.
Mol. Med. Today
6
,
20
-
27
.
Reinhard
,
J. R.
,
Lin
,
S.
,
McKee
,
K. K.
,
Meinen
,
S.
,
Crosson
,
S. C.
,
Sury
,
M.
,
Hobbs
,
S.
,
Maier
,
G.
,
Yurchenco
,
P. D.
,
Rüegg
,
M. A.
et al. 
(
2017
).
Linker proteins restore basement membrane and correct LAMA2-related muscular dystrophy in mice
.
Sci. Transl. Med.
9
,
1
-
28
.
Rooney
,
J. E.
,
Knapp
,
J. R.
,
Hodges
,
B. L.
,
Wuebbles
,
R. D.
and
Burkin
,
D. J.
(
2012
).
Laminin-111 protein therapy reduces muscle pathology and improves viability of a mouse model of merosin-deficient congenital muscular dystrophy
.
Musculoskelet. Pathol.
180
,
1593
-
1602
.
Sarkozy
,
A.
,
Foley
,
A. R.
,
Zambon
,
A. A.
,
Bönnemann
,
C. G.
and
Muntoni
,
F.
(
2020
).
LAMA2-related dystrophies: clinical phenotypes, disease biomarkers, and clinical trial readiness
.
Front. Mol. Neurosci.
13
,
1
-
11
.
Sartori
,
R.
,
Romanello
,
V.
and
Sandri
,
M.
(
2021
).
Mechanisms of muscle atrophy and hypertrophy: implications in health and disease
.
Nat. Commun.
12
,
1
-
12
.
Schiaffino
,
S.
,
Reggiani
,
C.
,
Akimoto
,
T.
and
Blaauw
,
B.
(
2021
).
Molecular mechanisms of skeletal muscle hypertrophy
.
J. Neuromuscul. Dis.
8
,
169
-
183
.
Schultze
,
S. M.
,
Hemmings
,
B. A.
,
Niessen
,
M.
and
Tschopp
,
O.
(
2012
).
PI3K/AKT, MAPK and AMPK signalling: protein kinases in glucose homeostasis
.
Expert Rev. Mol. Med.
14
,
1
-
21
.
Serrano
,
A. L.
and
Muñoz-Cánoves
,
P.
(
2017
).
Fibrosis development in early-onset muscular dystrophies: Mechanisms and translational implications
.
Semin. Cell Dev. Biol.
64
,
181
-
190
.
Shorer
,
Z.
,
Philpot
,
J.
,
Muntoni
,
F.
,
Sewry
,
C.
and
Dubowitz
,
V.
(
1995
).
Demyelinating peripheral neuropathy in merosin-deficient congenital muscular dystrophy
.
J. Child Neurol.
10
,
472
-
475
.
Sperringer
,
J. E.
and
Grange
,
R. W.
(
2016
).
In vitro assays to determine skeletal muscle physiologic function
.
Methods Mol. Biol.
1460
,
271
-
291
.
Tang
,
R.
and
Xu
,
Z.
(
2020
).
Gene therapy: a double-edged sword with great powers
.
Mol. Cell. Biochem.
474
,
73
-
81
.
Tang
,
H.
,
Inoki
,
K.
,
Lee
,
M.
,
Wright
,
E.
,
Khuong
,
A.
,
Khuong
,
A.
,
Sugiarto
,
S.
,
Garner
,
M.
,
Paik
,
J.
,
Depinho
,
R. A.
et al. 
(
2014
).
mTORC1 promotes denervation-induced muscle atrophy through a mechanism involving the activation of FoxO and E3 ubiquitin ligases
.
Muscle Biol.
7
,
1
-
11
.
Tang
,
H.
,
Inoki
,
K.
,
Brooks
,
S. V.
,
Okazawa
,
H.
,
Lee
,
M.
,
Wang
,
J.
,
Kim
,
M.
,
Kennedy
,
C. L.
,
Macpherson
,
C. D.
,
Macpherson
,
P.
et al. 
(
2019
).
mTORC1 underlies age–related muscle fiber damage and loss by inducing oxidative stress and catabolism
.
Aging Cell
18
,
e12943
.
Taniguchi
,
M.
,
Kurahashi
,
H.
,
Noguchi
,
S.
,
Sese
,
J.
,
Okinaga
,
T.
,
Tsukahara
,
T.
,
Guicheney
,
P.
,
Ozono
,
K.
,
Nishino
,
I.
,
Morishita
,
S.
et al. 
(
2006
).
Expression profiling of muscles from Fukuyama-type congenital muscular dystrophy and laminin-α2 deficient congenital muscular dystrophy; is congenital muscular dystrophy a primary fibrotic disease?
Biochem. Biophys. Res. Commun.
342
,
489
-
502
.
Tomomura
,
M.
,
Fujii
,
T.
,
Sakagami
,
H.
and
Tomomura
,
A.
(
2011
).
Serum calcium-decreasing factor, caldecrin, ameliorates muscular dystrophy in dy/dy mice
.
In Vivo (Brooklyn)
25
,
157
-
163
.
Turjanski
,
A. G.
,
Vaqué
,
J. P.
and
Gutkind
,
J. S.
(
2007
).
MAP kinases and the control of nuclear events
.
Oncogene
26
,
3240
-
3253
.
van Ry
,
P. M.
,
Minogue
,
P.
,
Hodges
,
B. L.
and
Burkin
,
D. J.
(
2014
).
Laminin-111 improves muscle repair in a mouse model of merosin-deficient congenital muscular dystrophy
.
Hum. Mol. Genet.
23
,
383
-
396
.
Vin
,
H.
,
Ojeda
,
S. S.
,
Ching
,
G.
,
Leung
,
M. L.
,
Chitsazzadeh
,
V.
,
Dwyer
,
D. W.
,
Adelmann
,
C. H.
,
Restrepo
,
M.
,
Richards
,
K. N.
,
Stewart
,
L. R.
et al. 
(
2013
).
BRAF inhibitors suppress apoptosis through off-target inhibition of JNK signaling
.
eLife
2
,
1
-
25
.
Wang
,
C. H.
,
Bonnemann
,
C. G.
,
Rutkowski
,
A.
,
Sejersen
,
T.
,
Bellini
,
J.
,
Battista
,
V.
,
Florence
,
J. M.
,
Schara
,
U.
,
Schuler
,
P. M.
,
Wahbi
,
K.
et al. 
(
2010
).
Consensus statement on standard of care for congenital muscular dystrophies Ching
.
J. Child Neurol.
25
,
1559
-
1581
.
Xie
,
S.-J.
,
Li
,
J.-H.
,
Chen
,
H.-F.
,
Tan
,
Y.-Y.
,
Liu
,
S.-R.
,
Zhang
,
Y.
,
Xu
,
H.
,
Yang
,
J.-H.
,
Liu
,
S.
,
Zheng
,
L.-L.
et al. 
(
2018
).
Inhibition of the JNK/MAPK signaling pathway by myogenesis-associated miRNAs is required for skeletal muscle development
.
Cell Death Differ.
25
,
1581
.
Xiong
,
H.
,
Tan
,
D.
,
Wang
,
S.
,
Song
,
S.
,
Yang
,
H.
,
Gao
,
K.
,
Liu
,
A.
,
Jiao
,
H.
,
Mao
,
B.
,
Ding
,
J.
et al. 
(
2015
).
Genotype/phenotype analysis in Chinese laminin-α2 deficient congenital muscular dystrophy patients
.
Clin. Genet.
87
,
233
-
243
.
Yu
,
Q.
,
Sali
,
A.
,
van der Meulen
,
J.
,
Creeden
,
B. K.
,
Gordish-Dressman
,
H.
,
Rutkowski
,
A.
,
Rayavarapu
,
S.
,
Uaesoontrachoon
,
K.
,
Huynh
,
T.
,
Nagaraju
,
K.
et al. 
(
2013
).
Omigapil treatment decreases fibrosis and improves respiratory rate in dy2J mouse model of congenital muscular dystrophy
.
PLoS ONE
8
,
e65468
.
Zambon
,
A. A.
,
Ridout
,
D.
,
Main
,
M.
,
Mein
,
R.
,
Phadke
,
R.
,
Muntoni
,
F.
and
Sarkozy
,
A.
(
2020
).
LAMA2-related muscular dystrophy: natural history of a large pediatric cohort
.
Ann. Clin. Transl. Neurol.
7
,
1870
-
1882
.
Zhang
,
W.
,
Heinzmann
,
D.
and
Grippo
,
J. F.
(
2017
).
Clinical Pharmacokinetics of Vemurafenib
.
Clin. Pharmacokinet.
56
,
1033
-
1043
.

Competing interests

The authors declare no competing or financial interests.

This is an Open Access article distributed under the terms of the Creative Commons Attribution License (https://creativecommons.org/licenses/by/4.0), which permits unrestricted use, distribution and reproduction in any medium provided that the original work is properly attributed.

Supplementary information