Next Article in Journal
15-Deoxy-Δ12,14-prostaglandin J2 Upregulates VEGF Expression via NRF2 and Heme Oxygenase-1 in Human Breast Cancer Cells
Next Article in Special Issue
Astrocytes in Alzheimer’s Disease: Pathological Significance and Molecular Pathways
Previous Article in Journal
Cellular Energetics of Mast Cell Development and Activation
Previous Article in Special Issue
How Degeneration of Cells Surrounding Motoneurons Contributes to Amyotrophic Lateral Sclerosis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Skeletal Muscle in ALS: An Unappreciated Therapeutic Opportunity?

by
Silvia Scaricamazza
1,2,†,
Illari Salvatori
1,3,†,
Alberto Ferri
1,4,* and
Cristiana Valle
1,4,*
1
Fondazione Santa Lucia IRCCS, c/o CERC, 00143 Rome, Italy
2
Department of Biology, University of Rome Tor Vergata, 00133 Rome, Italy
3
Department of Experimental Medicine, University of Rome “La Sapienza”, 00161 Rome, Italy
4
Institute of Translational Pharmacology, National Research Council, 00133 Rome, Italy
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Cells 2021, 10(3), 525; https://doi.org/10.3390/cells10030525
Submission received: 4 February 2021 / Revised: 23 February 2021 / Accepted: 24 February 2021 / Published: 2 March 2021

Abstract

:
Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disorder characterized by the selective degeneration of upper and lower motor neurons and by the progressive weakness and paralysis of voluntary muscles. Despite intense research efforts and numerous clinical trials, it is still an incurable disease. ALS had long been considered a pure motor neuron disease; however, recent studies have shown that motor neuron protection is not sufficient to prevent the course of the disease since the dismantlement of neuromuscular junctions occurs before motor neuron degeneration. Skeletal muscle alterations have been described in the early stages of the disease, and they seem to be mainly involved in the “dying back” phenomenon of motor neurons and metabolic dysfunctions. In recent years, skeletal muscles have been considered crucial not only for the etiology of ALS but also for its treatment. Here, we review clinical and preclinical studies that targeted skeletal muscles and discuss the different approaches, including pharmacological interventions, supplements or diets, genetic modifications, and training programs.

1. Introduction

Amyotrophic lateral sclerosis (ALS) is a progressive disease characterized by motor neuron degeneration and skeletal muscle atrophy [1]. There are no effective therapies for ALS: riluzole and edaravone are the only FDA-approved drugs; however, they have a rather modest impact on the course of the disease [2].
While the majority of ALS cases are sporadic (sALS), about 10% of the cases are familial (fALS) and characterized by autosomal dominant inheritance. The clinical manifestations of sALS and fALS are indistinguishable, suggesting that different pathways converge, causing the typical neuromuscular degeneration of ALS [3]. However, despite the intense efforts to identify the pathogenetic mechanisms, the etiology of ALS remains elusive.
As ALS has long been considered the prototype of motor neuron diseases, many studies on its pathology have been “neurocentric”. However, since the early 2000s, several papers have started describing the key roles of non-neuronal cell types in triggering or supporting the ALS neuromuscular degenerative processes [4]. Today, ALS is considered a multisystemic and multifactorial disease characterized by the degeneration of motor neurons in the motor cortex and spinal cord and accelerated by physiological alterations of other cell types and organs [5].
Genetic models showed that only the ubiquitous expression of a gain-of-function mutant of the human superoxide dismutase 1 (mSOD1) induced fast and severe paralysis in mice, mimicking ALS progression [6]. The overexpression of mSOD1 in neurons led to a late-onset of the disease with slow progression [7], while restricting the expression of mSOD1 to motor neurons did not trigger the pathology [8]. mSOD1 expression in skeletal muscle elicited muscle atrophy, decreased muscle strength, impaired mitochondrial distribution and the contractile apparatus [9], reduced the spinal cord mass, triggered late motor neuron loss, and shortened the lifespan [10].
The neuromuscular junction (NMJ) connects muscle fibers and motor neurons, allowing their communication; ALS is the classic example of severely compromised communication between muscles and nerves [11]. Motor neuron activity regulates muscle physiology and function; in turn, muscles affect the neuronal activity by sending retrograde signals that preserve NMJ functionality and structure [12]. The so-called “dying back” hypothesis suggests that retrograde signals contribute to the centripetal motor neuron degeneration in ALS [13]. Studies in ALS mouse models have corroborated this hypothesis and described ALS as a distal axonopathy also caused by alterations in skeletal muscle [14].
Different studies have reported that more than 60% of familial and sporadic ALS patients have increased resting and non-resting energy expenditure, an unexpected feature considering the effects of undernutrition on energy balance and the defense mechanisms to lower energy waste [15,16,17]. A study involving a large cohort of ALS patients found that high levels of physical activity were related to an increased risk of ALS [18,19]. It also has been shown that a low premorbid body mass index (BMI) increases the risk of ALS and that weight loss and hypermetabolism correlate with a less favorable prognosis [16,20]. Interestingly, variants of the ACSL5 gene, previously associated with rapid weight loss in humans, have recently been associated with ALS risk and lean body mass reduction in ALS patients [21]. Moreover, ALS incidence is lower among obese individuals, and patients with a high pre-diagnostic body and subcutaneous fat have a lower mortality risk [22,23]. Overall, the studies suggest a key role of energy expenditure and hypermetabolism in ALS pathology.
The cause of defective energy homeostasis in ALS is still unknown; however, since muscle metabolism is the major determinant of the total energy expenditure, we should further investigate the role of skeletal muscle in ALS etiology to understand whether the metabolic disorder contributes to its pathogenesis. Early events before denervation affecting muscle physiology have been described, supporting the “dying back” hypothesis in ALS. Understanding the molecular mechanisms involved in skeletal muscle degeneration may help develop therapeutic strategies that preserve muscle function, slow down the disease progression, and improve ALS patients’ quality of life.
In this review, we have summarized and discussed the therapeutic approaches that have been used to increase the performance of skeletal muscle in ALS animal models and patients.

2. Genetic Interventions

Gene therapy aims to treat a disease by inserting genetic material into cells with viral or non-viral vectors [24]. The technology allowed obtaining remarkable results in patients affected by spinal muscular atrophy (SMA), a childhood neuromuscular disease caused by the deletion or mutation of survival of motor neuron 1 (SMN1). The treatment involved the injection of antisense oligonucleotides (ASOs) [25,26] or viral vectors [27] in restoring SMN1 protein levels [28]. These results have opened doors to new treatments for fALS patients, and gene therapy clinical trials have been proposed.
Genetic interventions in ALS animal models, which targeted not only ALS-linked mutations, have been useful to understand the role of several proteins and pathways in the progression of the disease. Some studies have shown the importance of skeletal muscle in ALS progression, highlighting that motor neuron degeneration is not the only cause of alterations in this tissue. For instance, the modifications of trophic factors, such as glial cell-derived neurotrophic factor (GDNF), vascular endothelial growth factor (VEGF), and insulin-like growth factor 1 (IGF-1), in particular, improved ALS symptoms and survival.
GDNF is a member of the TGF-β family, it was originally isolated in rat glial cells, and it promotes the differentiation and survival of dopaminergic neurons by increasing their dopamine uptake [29,30]. GDNF also has a neurotrophic function in motor neurons, where it enhances survival by preventing apoptosis and degeneration [31,32,33,34,35,36]. Because of this function, GDNF has been proposed as a therapeutic target for ALS [31,36,37]. Its expression in the skeletal muscle of 5–7-day-old SOD1G93A mice slowed down the progression of the disease. The mice were injected with adenoviral vectors (AVR) into the hind limbs and the paraspinal muscles. In treated mice, the disease onset, the reduction of motor performance, and the motor neuron loss were delayed. Moreover, SOD1G93A-GDNF mice survived about two weeks longer than control mice [38]. The reason why the intramuscular injection of GDNF increased the lifespan is not clear, but it could be due to the effect on the NMJ, as GDNF increases the nerve sprouting capacity [38]. Alternatively, GDNF expression in skeletal muscle could promote motor neuron preservation in the spinal cord. Indeed, Acsadi and colleagues detected the protein both in the muscles and the spinal cord, probably because of the retrograde transport of motor neurons [38]. Consistent with these findings, the intramuscular transplantation with human mesenchymal stem cells engineered to secrete GDNF (hMSC-GDNF) protected SOD1G93A rats from motor neuron loss and denervation, significantly delayed motor decline and increased the lifespan by about four weeks [39].
VEGF is another neuroprotective factor that may play a role in ALS. VEGF promotes angiogenesis and neuronal survival [40], as shown by the knockout of VEGF in wild-type mice that led to neurodegeneration and ALS-like symptoms [41]. Azzuoz and colleagues observed that a single injection of VEGF-expressing lentiviral vector (EIAV-VEGF) in different muscles of SOD1G93A mice (gastrocnemius muscle, diaphragm, intercostal, facial, and tongue muscles) had positive effects on ALS symptoms [42]. To replicate potential clinical applications, two groups of mice received the EIAV-VEGF injection at two different time points: one before the onset (21 days of age) and the other at the onset of the disease. Both groups showed longer survival, better motor performances, and delays in motor neuron loss and motor weakness compared to controls [42].
The neurotrophin neuregulin 1 (NRG1) protects motor neurons from degeneration and is involved in the development and maintenance of axons [43,44,45] and NMJs, where it induces the clustering of acetylcholine receptors (AchRs) [46,47,48]. NRG1 exerts its functions by interacting with ErbB receptors; this interaction is impaired in ALS patients and animal models [49,50], which show low levels of expression of ErbB4 mRNA and protein in their skeletal muscle [51]. Similarly, the gastrocnemius muscle of SOD1G93A mice has reduced levels of ErbB4 mRNA, which correlates with denervation [51]. The injection of an adeno-associated viral vector (AAV) expressing NRG1-I into the gastrocnemius muscle of SOD1G93A mice significantly increased the muscle action potential and the collateral sprouting of axons [51]. In line with these results, expressing NRG1 in the skeletal muscles using the human desmin (hDesmin) promoter delayed the onset of ALS and improved the phenotype in SOD1G93A mice. Indeed, NRG1 was shown to activate cell survival pathways in muscles and the spinal cord, protecting against denervation, neuroinflammation, and motor neuron loss. As the NMJs were preserved, treated mice had better neuromuscular and motor functions [52].
In ALS, NMJ degradation is the first event of denervation, and it occurs before motor neuron loss [14,53]. NRG1-ErbBs signaling and, hence, NMJ development and maintenance depend on the activation of muscle-specific receptor tyrosine kinase (MuSK) [54]. MuSK orchestrates the muscle-derived retrograde signal through the interaction with LRP4 and agrin, ensuring NMJ stability and maintenance while preventing disassembly [55,56,57,58]. MuSK overexpression in SOD1G93A double transgenic mice delayed the onset of ALS, improved motor ability, and preserved the integrity of NMJs [59].
DOK-7 (docking protein 7) controls MuSK activation and response to agrin [60]; mutations in the DOK7 gene are responsible for the congenital myasthenic syndrome, which is characterized by impaired NMJ structure and functionality [61]. Since the administration of a recombinant AAV carrying the human DOK7 gene (AAV-D7) improved motor abilities and survival of animal models of DOK-7 myasthenia [62] and Emery–Dreifuss muscular dystrophy [63], Miyoshi and colleagues tried this therapeutic approach in ALS models: intravenous injection of AAV-D7 in SOD1G93A mice at the onset of the disease significantly increased motor abilities, counteracted muscle atrophy, preserved NMJs, and extended the lifespan by more than ten days [64].
The microRNA miR206 and class II histone deacetylase 4 (HDAC4) control the denervation-reinnervation process [65]. The involvement of miR206 and HDAC4 in ALS progression has been proposed since their expression is altered in the skeletal muscles of patients and animal models [65,66,67,68]. HDAC4 is considered as a link between neuronal activity and muscle transcription because of its response to denervation [69]: HDAC4 expression levels in skeletal muscles increase during denervation, activating the muscle atrophy pathway driven by E3 ubiquitin-ligases MuRF1 and atrogin-1 transcription [69,70,71]. HDAC4 mRNA is upregulated in the muscle biopsies of ALS patients and correlates with disease severity, as the expression is higher in patients with a faster progression of the disease [68].
Pharmacological inhibition of class II HDACs in SOD1G93A mice increased skeletal muscle electrical potential and improved motor abilities; however, it had no effect on survival and motor neuron loss [72]. On the other hand, the knockout of HDAC4 in skeletal muscle of SOD1G93A mice worsened the ALS-like phenotype by speeding up the onset, the muscle force decline, and the NMJ loss, demonstrating that HDAC4 specifically has a protective role in ALS [73]. Indeed, HDAC4 induces the reinnervation pathway by activating the transcription of MuSK, miR206, and synaptic AchR through the indirect regulation of myogenin expression [69,70,71].
miR206 could be a prognostic marker for ALS, as high levels in serum correlate with a slower disease progression [74]. SOD1G93A mice genetically deficient in miR206 showed worse ALS symptoms, shorter survival, increased muscle atrophy, and early NMJ loss, corroborating the important role of miR206 in the reinnervation process [65]. Williams and colleagues also observed that miR206 upregulation after denervation was higher in fast-twitch fibers than in slow-twitch fibers, probably because slow-twitch fibers express higher levels of miR206 in physiological conditions [65]. Interestingly, fast-twitch fibers are more vulnerable to denervation in ALS [75,76]; thus, the upregulation of miR206 could be a defense mechanism to protect them [65,77].
The protective role of miR206 in ALS could also be due to its role in satellite cell differentiation: miR206 promotes the differentiation of satellite cells into muscle cells by inhibiting PAX7 expression; PAX7 inhibition is further enhanced by a positive feedback loop in which the muscular differentiation factors MyoD, myogenin, and MEF2 induce miR206 transcription [78,79,80].
In adult muscles, MyoD is predominantly expressed in fast-twitch fibers, while myogenin in slow-twitch fibers [81,82,83], where it stimulates oxidative metabolism [84,85]. Given their differential role and expression patterns, Park and colleagues hypothesized that the postnatal expression of MyoD and myogenin in muscles could affect ALS progression in opposite ways [86]. Indeed, MyoD overexpression in 30-day-old SOD1G93A mice via intramuscular injection with an AVV vector led to a more aggressive phenotype, shorter survival, earlier decline of motor performances, and premature loss of motor neurons and NMJs [86]. On the other hand, myogenin overexpression in skeletal muscle improved motor functions and preserved innervation and motor neurons; however, there were no changes in the lifespan [86].
The results from MyoD and myogenin overexpression and miR206 deletion led to the hypothesis that fibers in ALS switch from the fast to the slow type to preserve integrity and functionality of motor units and skeletal muscle [87]. This switch may reflect the metabolic shift towards oxidative metabolism occurring in ALS patients and animal models [88,89,90]. Since mitochondria regulate metabolism plasticity, they may also play a crucial role in the ALS fiber switch [90]. In ALS mouse models, mitochondrial dysfunctions and energy production impairments occur before the onset of the disease in skeletal muscle and at the onset of the disease in the spinal cord [90,91,92]. In addition, it has been shown that mitochondrial dysfunctions start before muscle differentiation, as they can be detected in the satellite cells of SOD1G93A mice [90].
Studies have shown that genetic interventions that enhance mitochondrial performance can improve muscle functionalities and quality of life. Peroxisome proliferator-activated receptor-gamma coactivator-1a (PGC-1a) is a major regulator of mitochondrial biogenesis and activity [93,94]. In SOD1G37R mice, the overexpression of PGC-1a in skeletal muscle improved muscle performance, locomotor activity, resistance to fatigue, and muscle atrophy; it also increased the mitochondrial area and the oxygen consumption in skeletal muscle [86,95]. Consistent with these results, the overexpression of uncoupling protein 1 (UCP1) (a mitochondrial protein that mediates non-shivering thermogenesis by uncoupling the mitochondrial electron transport chain) shortened the lifespan of SOD1G86R mice and increased disease duration and progression [96]. UCP1 overexpression in skeletal muscle of wild-type mice was sufficient to trigger NMJ dismantlement and distal motor neuron degeneration [96], highlighting the importance of mitochondria for the integrity and functionality of skeletal muscle. Together, these results show that enhancing mitochondrial functions and regulating the energy homeostasis of muscles can delay disease progression and improve the quality of life. However, it should be noted that improving mitochondrial performance in mouse models of ALS does not always produce an increase in survival, as described by some works that through genetic or pharmacological approaches improve mitochondrial proliferation [97,98,99].
IGF-1 regulates skeletal muscle physiology as well as mitochondrial dynamics and turnover [100,101,102]; it prevents inflammation, controls protein synthesis and degradation, and promotes satellite cell proliferation and neuronal survival [101,102,103,104]. The expression of IGF-1 in skeletal muscle of ALS models gave the most remarkable results on disease progression and survival, delaying the death of SOD1G93A mice by about one month [105]. In these mice, the regeneration pathways through calcineurin and CDK5 were induced, while apoptotic and ubiquitin pathways were inhibited, protecting muscles against atrophy and denervation and preserving NMJs and motor neurons [105,106]. Interestingly, high concentrations of IGF-1 in patients’ serum correlate with a better prognosis but not with a lower risk of ALS, suggesting that IGF-1 plays a role in the survival of ALS patients [107].
All preclinical genetic interventions on skeletal muscle highlighted the importance of this tissue in ALS progression as modifying the expression of genes involved in skeletal muscle physiology, metabolism, and functions had a strong impact (either positive or negative) on the disease. Therefore, muscle-directed gene therapy could become a therapeutic approach for ALS.
Table 1 summarizes the genetic interventions on ALS skeletal muscle.

3. Pharmacological and Nutritional Interventions

To date, pharmacological interventions to counteract ALS neuromuscular degeneration target mainly neurons. Drugs targeting skeletal muscle have been tested only recently; they could improve energetic metabolism and allow sprouting and formation of new synapses. Enhancing muscle and, hence, respiratory functions should be a priority in ALS care, also because it can improve the patient’s quality of life.
In this section, we have classified the pharmacological treatments based on their action on pathological events; however, considering their tight interdependence, there is a fine line between the different skeletal muscle alterations.

3.1. Pharmacological Interventions That Target Hypermetabolism

As muscle activity is the major contributor to the whole-body energy metabolism, muscles are likely to play a crucial role in ALS hypermetabolism. Defects in muscular ATP production and altered substrate utilization have been reported in patients and animal models [53,108]. In ALS, fiber transition from fast fatigable to fast intermediate and fast fatigue-resistant occurs before any measurable locomotor defects [88,89,90]. As shown in mSOD1 ALS mice, this transition results in the switch from glycolysis (i.e., the use of glucose as the main energy source) to ß-oxidation (i.e., the use of fat as the main energy source) [88,89,90]. Consistent with these findings, a study found that many ALS patients experience a sharp decrease in BMI and weight due to lipid consumption almost ten years before the onset of the disease and the appearance of motor symptoms [20].
Different metabolic therapies have been tested on animal models and patients to sustain their fatty acid consumption.
Carnitine is a fundamental source of acetyl groups. As it acts by transporting long-chain fatty acids into the mitochondrial matrix, its bioavailability is directly related to the rate of ß-oxidation [109]. 95% of carnitine resides in skeletal muscle since this tissue largely depends on fatty acids as an energy source [110].
Carnitine affects muscle remodeling by preventing atrophy and activating the oxidative stress response [111]. In presymptomatic SOD1G93A mice, oral administration of L-carnitine extended the lifespan while delaying motor impairment and disease onset [112]. Similar results were also obtained with subcutaneous injection at the onset of the disease [112]. The co-administration of acetyl-L-carnitine and riluzole in a small group of ALS patients was well tolerated and resulted in a better ALSFRS score (ALS Functional Rating Scale) as compared to riluzole alone [113] (EudraCT number: 2004-004158-23). Despite these encouraging results, a larger Phase III trial to validate the effectiveness of the treatment is still missing.
A high-fat diet (HFD) in SOD1G93A mice extended the mean survival by about 20% [114], and a high-energy diet based on medium-chain fatty acids and beta-hydroxybutyrate reduced locomotor defects in a Drosophila model of ALS [115]. Interestingly, a study involving more than two hundred patients fed with HFD showed that this diet significantly extended the survival of fast-progressing patients [116].
Creatine has been recently proposed in preclinical and clinical studies to compensate for the progressive depletion of energy reserves in ALS. Creatine is an amino acid endogenously synthesized or found in food, which is mainly absorbed by skeletal muscle. Creatine is phosphorylated by creatine kinase (CK) to phospho-creatine (PK) that is used as a source of energy during rapid and intense muscle contractions [117,118,119,120]. Since it helps improve muscle performance, creatine is often taken by athletes as a dietary supplement [121]. Initially, preclinical studies provided encouraging data as creatin was shown to delay the impairment of locomotor functions and extend lifespan [122,123,124]; however, a later study did not confirm these results [125]. Similarly, randomized controlled human trials evaluating the efficacy of creatine monohydrate, administered alone or in combination with other drugs (NCT00005766, NCT00005674, NCT00355576, NCT00070993, NCT00069186, NCT01257581) [126,127,128], showed that this compound did not improve disease progression or survival in ALS patients [129]. However, high CK levels have been correlated with a slower progression of the disease in ALS patients and mouse models [130], suggesting that providing supplements to the muscles could partially compensate for the catabolic effects of ALS hypermetabolism.
Inhibiting the β-oxidation of fatty acids to induce glycolysis has been another strategy to counter hypermetabolism in ALS. A recent study of SOD1G93A mice showed that starting the chronic administration of Ranolazine, an inhibitor of β-oxidation, at the onset of the disease slowed down the muscle strength loss and improved the motor functions, but not the lifespan. We correlated the administration of Ranolazine with improvements in energy metabolism, as the drug reduced the whole-body energy expenditure of SOD1G93A mice by increasing the levels of ATP in muscles [90].
Consistent with these results, dichloroacetate (DCA), a pyruvate dehydrogenase kinase 4 (PDK4) inhibitor that switches muscle metabolism from β-oxidation to glycolysis, improved muscle strength, maintained NMJ integrity and reduced the expression of denervation markers in SOD1G86R mice [89]. Its administration in presymptomatic SOD1G93A mice delayed the onset of the disease, enhanced motor performance and increased the lifespan by improving the mitochondrial redox status [131]. However, although its use in certain chemotherapies, DCA has severe side effects, hepatotoxicity in particular [132], making it not suitable for long-term treatment of neurodegenerative diseases.
Together, these results indicate that hypermetabolism, and thus skeletal muscle, can be good drug targets for ALS.

3.2. Pharmacological Interventions to Increase Muscle Mass

Several pathological phenomena, including defects in the proliferation and differentiation processes, lead to skeletal muscle mass loss in ALS [53,133]. Different pharmacological approaches have been tried to counter mass loss, such as the treatment with anabolic androgenic steroids (AAS).
AAS are synthetic derivatives of the testosterone hormone that increase muscle mass (and, for this reason, are also often used illegally by athletes to enhance performances). Subcutaneous administration of dihydrotestosterone crystals (an AAS) in early-symptomatic SOD1G93A-induced weight gain, reduced muscle atrophy, increased grip strength, and extended lifespan [134]. Interestingly, dihydrotestosterone treatment increased muscular expression of IGF-1, which protects mitochondria of murine and cellular models of ALS by increasing mitophagy and upregulating the expression of anti-apoptotic proteins [135] (see above). Similarly, the chronic administration of the AAS nandrolone in presymptomatic SOD1G93A mice maintained the mass of the diaphragm muscle, despite mild side effects on muscle fiber innervation [136]. However, an earlier study on the same mouse model showed that the administration of nandrolone significantly increased the expression of TGFβ1a in muscles [137], suggesting that it could even worsen the disease [138].
Myostatin inhibits myogenesis and muscle growth by reducing fiber number and size [139,140,141,142]. Its overexpression led to weight loss, muscle atrophy, and sarcopenia [143], while its downregulation to muscle hypertrophy and a hypermuscular phenotype [144,145]. Because of its impact on muscle mass, several drugs that inhibit the myostatin signaling pathway have been evaluated in preclinical and clinical studies to treat a variety of muscle-wasting diseases [146]. Myostatin levels are significantly higher in ALS patients than in healthy individuals and they are positively correlated with the rate of muscle degeneration [147]. In two rodent models of ALS, neutralizing antibodies against myostatin decreased the weight loss and increased the mass and strength of muscles at the onset of the disease and during the early-stages [142]. However, they did not delay the disease onset nor increased survival [142]. Similar results were obtained in SOD1G93A mice using an Fc chimera of the activin receptor type IIB, an endogenous signaling receptor for myostatin [148].
Although obtained only in preclinical studies, these data suggest that targeting myostatin signaling pathways may have a therapeutic effect in ALS. Supporting this hypothesis, anti-myostatin antibodies improved muscle performance in a mouse model of Duchenne muscular dystrophy and prevented skeletal muscle alterations in a Huntington’s disease mouse model [149]; moreover, the administration of follistatin, a natural antagonist of myostatin, improved the severity of SMA in mice [150].

3.3. Pharmacological Interventions to Preserve NMJs and Reduce Atrophy

NMJ dismantling and muscle atrophy are early events in ALS and precede denervation, supporting the idea that skeletal muscle plays a key role in the disease. The maintenance of NMJs and the inhibition of atrophic processes have been considered as main targets for pharmacological interventions.
ALS patients’ muscles aberrantly express the neurite growth inhibitor Nogo-A, and its levels correlate with the severity of symptoms [151,152]. In ALS, Nogo-A causes retrograde axonal degeneration by destabilizing the NMJs. The overexpression of Nogo-A in murine healthy muscle fibers induced the detachment of NMJs, while its genetic ablation in SOD1G86R mice reduced denervation and increased the lifespan [151]. Despite these encouraging results, the administration of the anti-Nogo-A monoclonal antibody ozanezumab was ineffective in a phase II trial (NCT01753076) [153].
As discussed above, activating MuSK has been another approach to prevent or delay NMJs dismantling. Although ALS patients and animal models do not have alterations in the MuSK pathway, two preclinical studies have stimulated MuSK with an agonist antibody. These studies obtained conflicting results despite using a similar approach (same antibody and same mouse model). In the first study, the administration of the MuSK agonist preserved NMJs, delayed denervation and increased survival in SOD1G93A mice [154]. The second study did not report any improvements in diaphragm functionality or lifespan, despite the retention of NMJs in the diaphragm [155]. Therefore, further investigations are needed to understand the effectiveness of this therapeutic strategy.
Muscle acetylcholine receptors (AchRs) have also been proposed as therapeutic targets in ALS. In a recent clinical study, the administration of palmitoylethanolamide (PEA), an endocannabinoid that reduces the desensitization of AchRs currents following repeated stimulation, improved pulmonary functions and delayed the decrease of forced vital capacity (FVC) (NCT02645461) [156]. The authors also reported that PEA strongly upregulated the expression of the α1 AChR subunit and that it maintained NMJ functionality by reducing the rundown of ε-AChRs currents. Interestingly, another study showed that riluzole blocked muscle AchRs with greater specificity for γ-AChRs than ε-AChRs; however, the resulting biological consequences were not clarified [157].

3.4. Other Pharmacological Interventions

Fast skeletal muscle troponin activators (FSTA), which selectively activate the troponin complex and increase its sensitivity to calcium, have been studied as potential treatments for ALS [158,159]. Troponin is a protein complex that modulates muscle contractility and increases muscular strength and power, slowing down the onset of fatigue, particularly in respiratory muscles. The FSTA tirasemtiv gave good results in both preclinical [159] and early clinical studies (NCT01486849; NCT01089010; NCT02936635; NCT01709149; NCT01378676), maintaining muscle strength and delaying the onset and the level of muscle fatigue. However, in the phase III VITALITY-ALS trial (NCT02496767) involving 81 centers in the United States and Europe, tirasemtiv did not impact the decline of slow vital capacity (SVC), nor secondary outcomes, such as the ALSFRS-R score, the first use of mechanical ventilatory assistance, and death [160]. The fact that many patients did not tolerate the treatment and left the trial may have contributed to its disappointing results [160]. Following these data, reldesemtiv, a next-generation FSTA compound, was synthesized. Its functions are similar to tirasemtiv, but it has different chemical characteristics. A double-blind, randomized, placebo-controlled, variable dosage trial (NCT03160898) tested the safety of reldesemtiv and demonstrated that the drug was well tolerated by patients and that there was a trend towards improvement in primary and secondary outcomes, though it was not statistically significant [161]. The therapeutic potential of reldesemtiv is currently studied for the treatment of other diseases associated with muscle dysfunction and weakness, such as SMA, chronic obstructive pulmonary disease (COPD), and in elderly subjects with reduced mobility (NCT02644668sma- NCT03065959-copd).
The FSTA levosimendan has been recently tested. This compound showed positive effects in a phase II clinical trial [162] (NCT02487407); however, phase III clinical trials did not confirm the data, as the oral administration of levosimendan did not improve the respiratory function nor the general functions of ALS patients (NCT03505021; NCT03948178). In light of these discouraging results, this pharmacological approach has now been abandoned.
Recently, different studies have shown that CTGF/CCN2, a member of the CCN family of extracellular matrix-associated heparin-binding proteins, is upregulated in skeletal muscle and spinal cord of ALS patients [163,164]. CTGF/CCN2 plays a crucial role in tissue fibrosis, as it affects angiogenesis, migration, proliferation, and cell adhesion [165]. Treating SOD1G93A mice with a monoclonal neutralizing antibody against CTGF/CCN2 (FG-3019) improved locomotor performance and reduced muscular fibrosis and atrophy [163]. Preclinical studies using this treatment have provided encouraging results also for other pathologies associated with fibrosis, including skeletal muscle dystrophies [166].
Another therapeutic approach using aminophylline, which is supposed to mainly act on smooth muscle, was tested. Aminophylline is a soluble derivative of theophylline, a compound that relaxes smooth muscles and relieves bronchial spasm. Theophylline functions as a phosphodiesterase inhibitor, an adenosine receptor blocker, and a histone deacetylase activator [167]; it is widely used for the treatment of asthma, bronchospasm, and COPD [168,169,170]. Two studies showed that theophylline improved the strength and endurance of peripheral and respiratory muscles [171,172]. In a double-blind, randomized crossover trial, 25 ALS patients with a disease duration of fewer than five years received intravenous aminophylline; the treatment improved the endurance of respiratory muscles and increased handgrip strength [173]. Despite these promising results, the therapeutic potential of aminophylline in ALS has not been further investigated.
Finally, the activation of P2X7, a purinergic receptor abundantly expressed in skeletal muscle, with the specific agonist 2′(3′)-O-(4-benzoylbenzoyl) adenosine 5′-triphosphate improved muscle metabolism and preserved NMJ morphology in presymptomatic SOD1G93A mice. Interestingly, P2X7 is a key regulator of myofiber differentiation and regeneration [174].
Together, these data highlight the possibility to target skeletal muscle with a wide range of drug classes.
Preclinical and clinical studies are summarized in Table 2 and Table 3, respectively.

4. Physical Exercise as a Therapeutic Approach

Adult skeletal muscle is a highly plastic tissue that adapts in response to external stimuli [175]. For instance, variations in nutrient intake, aerobic, anaerobic conditions, and hormonal responses determine the structure of skeletal muscle. In addition, muscles adapt to physical training with structural and physiological changes, leading to positive health impacts [176].
The benefits of physical activity on motor neuron loss and sarcopenia are widely recognized [177]. As exercise may improve several chronic conditions, it could be considered as a therapeutic intervention to slow down muscle degeneration and preserve NMJ integrity in ALS.
Besides mental and other biological improvements, physical activity preserves specific mechanisms altered in the progression of ALS. For instance, exercise triggers pathways that improve skeletal muscle metabolism, enhance muscle glucose utilization and induce myofiber regeneration by activating satellite cells [178]; moreover, regular physical activity, regardless of the type, strengthens antioxidative defenses [179], and endurance training increases mitochondrial biogenesis in skeletal muscle [180] and neurogenesis [181,182]. However, preclinical studies have so far provided contradictory results, suggesting that the outcomes depend on the type and intensity of physical activity.
The adaptive response to exercise is a hormetic response that follows a biphasic curve, where low levels of stimuli elicit beneficial effects, whereas chronic and/or high levels of the same stimuli lead to negative or even toxic effects [183]. According to the exercise-induced hormesis theory, regular moderate-intensity training counteracts free radicals-induced cell injuries and inflammation processes [184,185,186], improves cardiovascular functions [187,188,189] and protects from different senescence-related processes [190,191], such as mitochondrial alterations in skeletal muscle [192,193,194]. On the other hand, continuous and high-intensity training triggers opposite effects, speeding up aging processes and increasing oxidative stress [195,196].
Models of ALS have shown the effects of exercise-induced hormesis. For instance, the phenotype of SOD1G93A mice that exercised on a motorized treadmill improved only with moderate exercise intensity, whereas high exercise intensity speeded up the decline of motor performance and did not preserve the density of motor neurons in the lumbar spine ventral horn [197]. In line with these observations, SOD1G93A mice exercising on a running wheel at moderate intensity showed a modest improvement in lifespan and locomotor performances [198,199,200]; on the other hand, intense, forced treadmill exercise worsened their phenotype [201].
Kirkinezos et al. and Veldink et al. showed that the effects of exercise on ALS phenotype depended on gender; however, they reported opposite conclusions. In both studies, SOD1G93A mice run daily on a treadmill for 30 and 45 min, respectively, at moderate intensity [202,203]. Kirkinezos et al. concluded that physical activity benefitted only in male mice [202], while Veldink et al. observed a positive neuroprotective effect only in females [203]. The authors proposed a different role of sex hormones to explain the gender-specific responses. However, in both studies, the mice were forced to run using an electric shock; this method has likely introduced biases in the results because of the stress that is induced in the animals. Garbugino et al. described the effects of voluntary exercise in low-copy SOD1G93A mice running on a home-cage running-wheel system [204]. The authors concluded that male mice were worse affected by prolonged and repeated exercise than females, showing shorter survival, increased body weight loss, and poorer prognosis. Their results were in line with the theory of exercise-induced hormesis [204].
The type of exercise may also affect the ALS phenotype. While running exercises have provided controversial results, the benefits of swimming-based exercises have been clear, as Deforges et al. showed that swimming extended the lifespan of SOD1G93A mice by about 25 days [205]. The authors hypothesized that swimming and running involve different motor units: while swimming is characterized by high-frequency and large-amplitude movements mainly recruiting the fast motor units, mostly running triggers the slow motor units [205]. These results were confirmed by other papers describing the beneficial effects of swimming on the phenotype of ALS mouse models [206,207,208,209].
Motor neuron fast-twitch fibers, which are preferentially stimulated when swimming, degenerate first and are more compromised in ALS [210], probably because they use the glycolytic pathway as their main energy source, a pathway heavily impaired in ALS [88,89,90]) (see above). Swimming improved glucose metabolism in SOD1G93A mice more efficiently than running [209]; specifically, it induced the expression of glucose transporter GLUT4 and of glyceraldehyde-3-phosphate dehydrogenase (GAPDH), the key enzyme of the glycolytic pathway, countering the glucose intolerance of SOD1G93A mice [88,90].
Exercise, and swimming, in particular, reduced the deregulation of the BDNF/TrkB pathway in SOD1G93A mice [211]. BDNF, a neurotrophin secreted following muscle contraction, can be either a neuroprotector or a neurotoxin by probably acting in a paracrine way and increasing glutamate excitotoxicity through the activation of TrkB receptors [212,213]. In SOD1G93A mice, the neuronal hyperexcitability and the following muscle contractions induced the over-secretion of BDNF that likely contributed to neurodegeneration by enhancing glutamate toxicity [211]. A recent paper showed that preserving the BDNF/TrkB pathway through a specific swimming-based training improved the phenotype of ALS mice [206]. Consistent with these findings, 70 to 115-day-old SOD1G93A mice trained by swimming (performed in an adjustable-flow swimming pool) and running (performed on a treadmill at moderate intensity) showed a decrease in muscular BDNF concentration; in particular, muscular BDNF reached physiological levels in the mice that underwent the swimming-based training [206].
Overall, preclinical studies have shown that mild-to-moderate aerobic training improves the phenotype of ALS animal models. A recent meta-analysis assessed the impact of exercise on ALS patients by comparing 94 patients that underwent therapeutic exercise with 159 patients treated with conventional therapy [214]. The authors concluded that exercise could positively affect the rate of weakening of physical functions; however, these results should be interpreted with caution due to the limited number of studies and the different protocols used [214].
Although the therapeutic use of physical activity in ALS patients is still debated, a recent randomized controlled study on 22 patients showed that repetitive twitches induced by local magnetic stimulation hampered muscle atrophy, increased local muscular strength, and slowed down the metabolic shift towards ß-oxidation [215]. The molecular analysis of muscular biopsies showed that the magnetic stimulation counteracted muscle atrophy and proteolysis by increasing the efficacy of nicotinic ACh receptors [215].
ALS patients have defects in the energetic metabolism of skeletal muscle and alterations in energy expenditure, which indicate a poor prognosis [16]. The hypothesis that lifetime physical activity is a risk factor for developing ALS is under debate [216] and has raised doubts about the use of physical activity, which increases the body’s energy requirements, as a therapy. These doubts are also justified by clinical evidence showing that the oxidative capacity of skeletal muscle is impaired when ALS patients undergo intense physical exercise [217,218,219]. A study also described a mild mitochondrial dysfunction at the onset of the disease that could be detected only during exercise [218].
The analysis of the oxidative capacity of skeletal muscle in exercised patients has provided heterogeneous results depending on their exercise capacity and clinical profile [218]. Overall, the data have highlighted the need to adapt the type and intensity of physical activity to each patient. For instance, Ferri and colleagues showed that a training program combining moderate-intensity aerobic and strength training improved the patients’ aerobic capacity and physical function when tailored to their individual needs [220].
Consistent with the effectiveness of training programs that combine strength and endurance exercises, Lunetta et al. showed that patients doing strength exercises for the upper and lower limbs and exercises on a cycle ergometer at a moderate intensity improved their ALSFRS-R score; however, the training did not extend their survival [221]. These results were confirmed by a pilot randomized study from Merico et al. that showed how a combined exercise program improved the patients’ functional status measured with the functional independence measure (FIM) [222]. Resistance and strength exercises alone were also well tolerated by ALS patients and generally improved the patient’s quality of life [223]. Indeed, resistance training protocols at moderate intensity improved the scores of the ALSFRS-R test [224,225] and of the 30-second sit-to-stand test [226]. Endurance training performed with moderate aerobic exercises has also been shown to increase the ALSFRS-R score in certain cases [227,228].
In conclusion, physical activity as a therapy option has given interesting results, especially regarding the patient’s quality of life. However, the type of exercise should be tailored to each patient’s needs, and the intensity should always be moderate.

5. Concluding Remarks

Skeletal muscle has been long neglected in ALS, but recent data have highlighted its role in the etiopathogenesis of the disease.
In this review, we have discussed the preclinical and clinical studies that have targeted skeletal muscle to treat ALS. Since all of them emphasized the pivotal role of this tissue in ALS progression, skeletal muscle should be considered an optimal target site for therapeutic intervention.
In our opinion, although the results obtained so far have not introduced substantial innovations in clinical practice, they allow us to draw important conclusions:
Skeletal muscle is the main determinant of the whole-body energy expenditure, and interventions that improve its metabolism bring benefits to the entire organism.
Given that the functions of muscles and motor neurons are tightly intertwined, therapeutic interventions targeting skeletal muscle can counter the dying back process and, ultimately, protect motor neurons.
Both the physiology and the accessibility of muscle tissue make it a good therapeutic target that is worth considering at least to improve the patients’ quality of life.

Author Contributions

Conceptualization, C.V., A.F.; data curation, S.S., I.S.; writing—original draft preparation, S.S., I.S.; writing—review and editing, C.V., A.F. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by AriSLA through the HyperALS project (A.F.); AFM-Telethon project n. 2018; AFM-Telethon project n. 21021 (A.F.); Ministry of Health, Italy–United States of America. 2020—“Whole transcriptome analysis in models of extended healthy lifespan after spermidine treatment”-n.PGR01040 (C.V.).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Acknowledgments

This work is dedicated to the memory of our Mentor and Friend Maria Teresa Carrì.

Conflicts of Interest

The authors declare that they have no conflict of interest. The funding sources had no role in the collection, analysis, and interpretation of the data of this manuscript.

References

  1. Hardiman, O.; Al-chalabi, A.; Chio, A.; Corr, E.M.; Robberecht, W.; Shaw, P.J.; Simmons, Z. Amyotrophic lateral sclerosis. Nat. Rev. Dis. Primers 2017, 3. [Google Scholar] [CrossRef]
  2. Jaiswal, M.K. Riluzole and edaravone: A tale of two amyotrophic lateral sclerosis drugs. Med. Res. Rev. 2019, 733–748. [Google Scholar] [CrossRef]
  3. Van Es, M.A.; Hardiman, O.; Chio, A.; Al-Chalabi, A.; Pasterkamp, R.J.; Veldink, J.H.; van den Berg, L.H. Amyotrophic lateral sclerosis. Lancet 2017, 390, 2084–2098. [Google Scholar] [CrossRef]
  4. Cozzolino, M.; Ferri, A.; Teresa Carrì, M. Amyotrophic lateral sclerosis: From current developments in the laboratory to clinical implications. Antioxid. Redox Signal. 2008, 10, 405–443. [Google Scholar] [CrossRef] [PubMed]
  5. Rossi, S.; Cozzolino, M.; Carrì, M.T. Old versus new mechanisms in the pathogenesis of ALS. In Proceedings of the Brain Pathology; Blackwell Publishing Ltd.: Hoboken, NJ, USA, 2016; Volume 26, pp. 276–286. [Google Scholar]
  6. Gurney, M.E.; Pu, H.; Chiu, A.Y.; Dal Canto, M.C.; Polchow, C.Y.; Alexander, D.D.; Caliendo, J.; Hentati, A.; Kwon, Y.W.; Deng, H.X.; et al. Motor neuron degeneration in mice that express a human Cu, Zn superoxide dismutase mutation. Science 1994, 264, 1772–1775. [Google Scholar] [CrossRef] [PubMed]
  7. Jaarsma, D.; Teuling, E.; Haasdijk, E.D.; De Zeeuw, C.I.; Hoogenraad, C.C. Neuron-specific expression of mutant superoxide dismutase is sufficient to induce amyotrophic lateral sclerosis in transgenic mice. J. Neurosci. 2008, 28, 2075–2088. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  8. Lino, M.M.; Schneider, C.; Caroni, P. Accumulation of SOD1 Mutants in Postnatal Motoneurons Does Not Cause Motoneuron Pathology or Motoneuron Disease. J. Neurosci. 2002, 22, 4825–4832. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  9. Dobrowolny, G.; Aucello, M.; Rizzuto, E.; Beccafico, S.; Mammucari, C.; Boncompagni, S.; Bonconpagni, S.; Belia, S.; Wannenes, F.; Nicoletti, C.; et al. Skeletal muscle is a primary target of SOD1G93A-mediated toxicity. Cell Metab. 2008, 8, 425–436. [Google Scholar] [CrossRef]
  10. Martin, L.J.; Wong, M. Skeletal Muscle-Restricted Expression of Human SOD1 in Transgenic Mice Causes a Fatal ALS-Like Syndrome. Front. Neurol. 2020, 11. [Google Scholar] [CrossRef]
  11. Lepore, E.; Casola, I.; Dobrowolny, G.; Musarò, A. Neuromuscular Junction as an Entity of Nerve-Muscle Communication. Cells 2019, 8, 906. [Google Scholar] [CrossRef] [Green Version]
  12. Heckman, C.J.; Enoka, R.M. Motor unit. Compr. Physiol. 2012, 2, 2629–2682. [Google Scholar] [CrossRef]
  13. Dadon-Nachum, M.; Melamed, E.; Offen, D. The “dying-back” phenomenon of motor neurons in ALS. J. Mol. Neurosci. 2011, 43, 470–477. [Google Scholar] [CrossRef]
  14. Fischer, L.R.; Culver, D.G.; Tennant, P.; Davis, A.A.; Wang, M.; Castellano-sanchez, A.; Khan, J.; Polak, M.A.; Glass, J.D. Amyotrophic lateral sclerosis is a distal axonopathy: Evidence in mice and man. Exp. Neurol. 2004, 185, 232–240. [Google Scholar] [CrossRef]
  15. Fayemendy, P.; Marin, B.; Labrunie, A.; Boirie, Y.; Walrand, S.; Achamrah, N.; Coëffier, M.; Preux, P.M.; Lautrette, G.; Desport, J.C.; et al. Hypermetabolism is a reality in amyotrophic lateral sclerosis compared to healthy subjects. J. Neurol. Sci. 2021, 420. [Google Scholar] [CrossRef]
  16. Steyn, F.J.; Ioannides, Z.A.; Van Eijk, R.P.A.; Heggie, S.; Thorpe, K.A.; Ceslis, A.; Heshmat, S.; Henders, A.K.; Wray, N.R.; Van Den Berg, L.H.; et al. Hypermetabolism in ALS is associated with greater functional decline and shorter survival. J. Neurol. Neurosurg. Psychiatry 2018, 1016–1023. [Google Scholar] [CrossRef] [Green Version]
  17. Ferri, A.; Coccurello, R. Review Article What is “Hyper” in the ALS Hypermetabolism? Mediat. Inflamm. 2017, 2017. [Google Scholar] [CrossRef]
  18. Harwood, C.A.; Westgate, K.; Gunstone, S.; Brage, S.; Wareham, N.J.; McDermott, C.J.; Shaw, P.J. Long-term physical activity: An exogenous risk factor for sporadic amyotrophic lateral sclerosis? Amyotroph. Lateral Scler. Front. Degener. 2016, 17, 377–384. [Google Scholar] [CrossRef] [Green Version]
  19. Pupillo, E.; Bianchi, E.; Vanacore, N.; Montalto, C.; Ricca, G.; Robustelli Della Cuna, F.S.; Fumagalli, F.; Castellani, M.; Poli, F.; Romeo, F.; et al. Increased risk and early onset of ALS in professional players from Italian Soccer Teams. Amyotroph. Lateral Scler. Front. Degener. 2020, 21, 403–409. [Google Scholar] [CrossRef]
  20. Peter, R.S.; Rosenbohm, A.; Dupuis, L.; Brehme, T.; Kassubek, J.; Rothenbacher, D.; Nagel, G.; Ludolph, A.C. Life course body mass index and risk and prognosis of amyotrophic lateral sclerosis: Results from the ALS registry Swabia. Eur. J. Epidemiol. 2017, 32, 901–908. [Google Scholar] [CrossRef]
  21. Iacoangeli, A.; Lin, T.; Al Khleifat, A.; Jones, A.R.; Opie-Martin, S.; Coleman, J.R.I.; Shatunov, A.; Sproviero, W.; Williams, K.L.; Garton, F.; et al. Genome-wide Meta-analysis Finds the ACSL5-ZDHHC6 Locus Is Associated with ALS and Links Weight Loss to the Disease Genetics. Cell Rep. 2020, 33. [Google Scholar] [CrossRef]
  22. Lindauer, E.; Dupuis, L.; Müller, H.-P.; Neumann, H.; Ludolph, A.C.; Kassubek, J. Adipose Tissue Distribution Predicts Survival in Amyotrophic Lateral Sclerosis. PLoS ONE 2013, 8, e67783. [Google Scholar] [CrossRef] [PubMed]
  23. Nakken, O.; Meyer, H.E.; Stigum, H.; Holmøy, T. High BMI is associated with low ALS risk: A population-based study. Neurology 2019, 93, E424–E432. [Google Scholar] [CrossRef]
  24. O’Connor, D.M.; Boulis, N.M. Gene therapy for neurodegenerative diseases. Trends Mol. Med. 2015, 21, 504–512. [Google Scholar] [CrossRef] [PubMed]
  25. Finkel, R.S.; Mercuri, E.; Darras, B.T.; Connolly, A.M.; Kuntz, N.L.; Kirschner, J.; Chiriboga, C.A.; Saito, K.; Servais, L.; Tizzano, E.; et al. Nusinersen versus Sham Control in Infantile-Onset Spinal Muscular Atrophy. N. Engl. J. Med. 2017, 377, 1723–1732. [Google Scholar] [CrossRef] [Green Version]
  26. Mercuri, E.; Darras, B.T.; Chiriboga, C.A.; Day, J.W.; Campbell, C.; Connolly, A.M.; Iannaccone, S.T.; Kirschner, J.; Kuntz, N.L.; Saito, K.; et al. Nusinersen versus Sham Control in Later-Onset Spinal Muscular Atrophy. N. Engl. J. Med. 2018, 378, 625–635. [Google Scholar] [CrossRef]
  27. Mendell, J.R.; Al-Zaidy, S.; Shell, R.; Arnold, W.D.; Rodino-Klapac, L.R.; Prior, T.W.; Lowes, L.; Alfano, L.; Berry, K.; Church, K.; et al. Single-Dose Gene-Replacement Therapy for Spinal Muscular Atrophy. N. Engl. J. Med. 2017, 377, 1713–1722. [Google Scholar] [CrossRef]
  28. Ludolph, A.C.; Wurster, C.D. Therapeutic advances in SMA. Curr. Opin. Neurol. 2019, 32, 777–781. [Google Scholar] [CrossRef]
  29. Lin, L.F.H.; Doherty, D.H.; Lile, J.D.; Bektesh, S.; Collins, F. GDNF: A glial cell line—Derived neurotrophic factor for midbrain dopaminergic neurons. Science 1993, 260, 1130–1132. [Google Scholar] [CrossRef]
  30. Lin, L.H.; Zhang, T.J.; Collins, F.; Armes, L.G. Purification and Initial Characterization of Rat B49 Glial Cell Line-Derived Neurotrophic Factor. J. Neurochem. 1994, 63, 758–768. [Google Scholar] [CrossRef]
  31. Henderson, C.E.; Phillips, H.S.; Pollock, R.A.; Davies, A.M.; Lemeulle, C.; Armanini, M.; Simpson, L.C.; Moffet, B.; Vandlen, R.A.; Koliatsos, V.E.; et al. GDNF: A potent survival factor for motoneurons present in peripheral nerve and muscle. Science 1994, 266, 1062–1064. [Google Scholar] [CrossRef] [PubMed]
  32. Oppenheim, R.W.; Houenou, L.J.; Johnson, J.E.; Lin, L.F.H.; Li, L.; Lo, A.C.; Newsome, A.L.; Prevette, D.M.; Wang, S. Developing motor neurons rescued from programmed and axotomy-induced cell death by gdnf. Nature 1995, 373, 344–346. [Google Scholar] [CrossRef]
  33. Trupp, M.; Rydén, M.; Jörnvall, H.; Funakoshi, H.; Timmusk, T.; Arenas, E.; Ibáñez, C.F. Peripheral expression and biological activities of GDNF, a new neurotrophic factor for avian and mammalian peripheral neurons. J. Cell Biol. 1995, 130, 137–148. [Google Scholar] [CrossRef] [Green Version]
  34. Li, L.; Wu, W.; Lin, L.F.H.; Lei, M.; Oppenheim, R.W.; Houenou, L.J. Rescue of adult mouse motoneurons from injury-induced cell death by glial cell line-derived neurotrophic factor. Proc. Natl. Acad. Sci. USA 1995, 92, 9771–9775. [Google Scholar] [CrossRef] [Green Version]
  35. Ruven, C.; Badea, S.R.; Wong, W.M.; Wu, W. Combination treatment with exogenous GDNF and fetal spinal cord cells results in better motoneuron survival and functional recovery after avulsion injury with delayed root reimplantation. J. Neuropathol. Exp. Neurol. 2018, 77, 325–343. [Google Scholar] [CrossRef] [Green Version]
  36. Cintrón-Colón, A.F.; Almeida-Alves, G.; Boynton, A.M.; Spitsbergen, J.M. GDNF synthesis, signaling, and retrograde transport in motor neurons. Cell Tissue Res. 2020, 382, 47–56. [Google Scholar] [CrossRef]
  37. Alisky, J.M.; Davidson, B.L. Gene Therapy for Amyotrophic Lateral Sclerosis and Other Motor Neuron Diseases. Hum. Gene Ther. 2000, 11, 2315–2329. [Google Scholar] [CrossRef] [PubMed]
  38. Acsadi, G.; Anguelov, R.A.; Yang, H.; Toth, G.; Thomas, R.; Jani, A.; Wang, Y.; Ianakova, E.; Mohammad, S.; Lewis, R.A.; et al. Increased survival and function of SOD1 mice after Glial cell-derived neurotrophic factor gene therapy. Hum. Gene Ther. 2002, 13, 1047–1059. [Google Scholar] [CrossRef] [PubMed]
  39. Suzuki, M.; McHugh, J.; Tork, C.; Shelley, B.; Hayes, A.; Bellantuono, I.; Aebischer, P.; Svendsen, C.N. Direct muscle delivery of GDNF with human mesenchymal stem cells improves motor neuron survival and function in a rat model of familial ALS. Mol. Ther. 2008, 16, 2002–2010. [Google Scholar] [CrossRef] [PubMed]
  40. Shim, J.W.; Madsen, J.R. VEGF signaling in neurological disorders. Int. J. Mol. Sci. 2018, 19, 275. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  41. Oosthuyse, B.; Moons, L.; Storkebaum, E.; Beck, H.; Nuyens, D.; Brusselmans, K.; Van Dorpe, J.; Hellings, P.; Gorselink, M.; Heymans, S.; et al. Deletion of the hypoxia-response element in the vascular endothelial growth factor promoter causes motor neuron degeneration. Nat. Genet. 2001, 28, 131–138. [Google Scholar] [CrossRef] [PubMed]
  42. Azzouz, M.; Ralph, G.S.; Storkebaum, E.; Walmsley, L.E.; Mitrophanous, K.A.; Kingsman, S.M.; Carmellet, P.; Mazarakis, N.D. VEGF delivery with retrogradely transported lentivector prolongs survival in a mouse ALS model. Nature 2004, 429, 413–417. [Google Scholar] [CrossRef] [PubMed]
  43. Loeb, J.A.; Khurana, T.S.; Robbins, J.T.; Yee, A.G.; Fischbach, G.D. Expression patterns of transmembrane and released forms of neuregulin during spinal cord and neuromuscular synapse development. Development 1999, 126, 781–791. [Google Scholar] [PubMed]
  44. Esper, R.M.; Pankonin, M.S.; Loeb, J.A. Neuregulins: Versatile growth and differentiation factors in nervous system development and human disease. Brain Res. Rev. 2006, 51, 161–175. [Google Scholar] [CrossRef]
  45. Fricker, F.R.; Lago, N.; Balarajah, S.; Tsantoulas, C.; Tanna, S.; Zhu, N.; Fageiry, S.K.; Jenkins, M.; Garratt, A.N.; Birchmeier, C.; et al. Axonally derived neuregulin-1 is required for remyelination and regeneration after nerve injury in adulthood. J. Neurosci. 2011, 31, 3225–3233. [Google Scholar] [CrossRef]
  46. Sandrock, A.W.; Dryer, S.E.; Rosen, K.M.; Gozani, S.N.; Kramer, R.; Theill, L.E.; Fischbach, G.D. Maintenance of acetylcholine receptor number by neuregulins at the neuromuscular junction in vivo. Science 1997, 276, 599–603. [Google Scholar] [CrossRef]
  47. Ngo, S.T.; Cole, R.N.; Sunn, N.; Phillips, W.D.; Noakes, P.G. Neuregulin-1 potentiates agrin-induced acetylcholine: Receptor clustering through muscle-specific kinase: Phosphorylation. J. Cell Sci. 2012, 125, 1531–1543. [Google Scholar] [CrossRef] [Green Version]
  48. Wolpowitz, D.; Mason, T.B.A.; Dietrich, P.; Mendelsohn, M.; Talmage, D.A.; Role, L.W. Cysteine-rich domain isoforms of the Neuregulin-1 gene are required for maintenance of peripheral synapses. Neuron 2000, 25, 79–91. [Google Scholar] [CrossRef] [Green Version]
  49. Song, F.; Chiang, P.; Wang, J.; Ravits, J.; Loeb, J.A. Aberrant neuregulin 1 signaling in amyotrophic lateral sclerosis. J. Neuropathol. Exp. Neurol. 2012, 71, 104–115. [Google Scholar] [CrossRef] [Green Version]
  50. Mòdol-Caballero, G.; García-Lareu, B.; Verdés, S.; Ariza, L.; Sánchez-Brualla, I.; Brocard, F.; Bosch, A.; Navarro, X.; Herrando-Grabulosa, M. Therapeutic Role of Neuregulin 1 Type III in SOD1-Linked Amyotrophic Lateral Sclerosis. Neurotherapeutics 2020, 17, 1048–1060. [Google Scholar] [CrossRef]
  51. Mancuso, R.; Martínez-Muriana, A.; Leiva, T.; Gregorio, D.; Ariza, L.; Morell, M.; Esteban-Pérez, J.; García-Redondo, A.; Calvo, A.C.; Atencia-Cibreiro, G.; et al. Neuregulin-1 promotes functional improvement by enhancing collateral sprouting in SOD1G93A ALS mice and after partial muscle denervation. Neurobiol. Dis. 2016, 95, 168–178. [Google Scholar] [CrossRef] [Green Version]
  52. Mòdol-Caballero, G.; Herrando-Grabulosa, M.; García-Lareu, B.; Solanes, N.; Verdés, S.; Osta, R.; Francos-Quijorna, I.; López-Vales, R.; Calvo, A.C.; Bosch, A.; et al. Gene therapy for overexpressing Neuregulin 1 type I in skeletal muscles promotes functional improvement in the SOD1G93A ALS mice. Neurobiol. Dis. 2020, 137, 104793. [Google Scholar] [CrossRef] [PubMed]
  53. Loeffler, J.-P.; Picchiarelli, G.; Dupuis, L.; Gonzalez De Aguilar, J.-L. The Role of Skeletal Muscle in Amyotrophic Lateral Sclerosis. Brain Pathol. 2016, 26, 227–236. [Google Scholar] [CrossRef]
  54. Burden, S.J. Building the vertebrate neuromuscular synapse. J. Neurobiol. 2002, 53, 501–511. [Google Scholar] [CrossRef]
  55. Herbst, R. MuSk function during health and disease. Neurosci. Lett. 2020, 716, 134676. [Google Scholar] [CrossRef]
  56. Rodríguez Cruz, P.M.; Cossins, J.; Beeson, D.; Vincent, A. The Neuromuscular Junction in Health and Disease: Molecular Mechanisms Governing Synaptic Formation and Homeostasis. Front. Mol. Neurosci. 2020, 13, 610964. [Google Scholar] [CrossRef]
  57. Hesser, B.A.; Henschel, O.; Witzemann, V. Synapse disassembly and formation of new synapses in postnatal muscle upon conditional inactivation of MuSK. Mol. Cell. Neurosci. 2006, 31, 470–480. [Google Scholar] [CrossRef]
  58. Kong, X.C.; Barzaghi, P.; Ruegg, M.A. Inhibition of synapse assembly in mammalian muscle in vivo by RNA interference. EMBO Rep. 2004, 5, 183–188. [Google Scholar] [CrossRef] [Green Version]
  59. Pérez-García, M.J.; Burden, S.J. Increasing MuSK Activity Delays Denervation and Improves Motor Function in ALS Mice. Cell Rep. 2012, 2, 497–502. [Google Scholar] [CrossRef] [Green Version]
  60. Inoue, A.; Setoguchi, K.; Matsubara, Y.; Okada, K.; Sato, N.; Iwakura, Y.; Higuchi, O.; Yamanashi, Y. Dok-7 activates the muscle receptor kinase MuSK and shapes synapse formation. Sci. Signal. 2009, 2, ra7. [Google Scholar] [CrossRef]
  61. Beeson, D.; Higuchi, O.; Palace, J.; Cossins, J.; Spearman, H.; Maxwell, S.; Newsom-Davis, J.; Burke, G.; Fawcett, P.; Motomura, M.; et al. Dok-7 mutations underlie a neuromuscular junction synaptopathy. Science 2006, 313, 1975–1978. [Google Scholar] [CrossRef]
  62. Arimura, S.; Okada, T.; Tezuka, T.; Chiyo, T.; Kasahara, Y.; Yoshimura, T.; Motomura, M.; Yoshida, N.; Beeson, D.; Takeda, S.; et al. DOK7 gene therapy benefits mouse models of diseases characterized by defects in the neuromuscular junction. Science 2014, 345, 1505–1508. [Google Scholar] [CrossRef] [PubMed]
  63. Méjat, A.; Decostre, V.; Li, J.; Renou, L.; Kesari, A.; Hantaï, D.; Stewart, C.L.; Xiao, X.; Hoffman, E.; Bonne, G.; et al. Lamin A/C—Mediated neuromuscular junction defects in Emery-Dreifuss muscular dystrophy. J. Cell Biol. 2009, 184, 31–44. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Miyoshi, S.; Tezuka, T.; Arimura, S.; Tomono, T.; Okada, T.; Yamanashi, Y. DOK 7 gene therapy enhances motor activity and life span in ALS model mice. EMBO Mol. Med. 2017, 9, 880–889. [Google Scholar] [CrossRef]
  65. Williams, A.H.; Valdez, G.; Moresi, V.; Qi, X.; McAnally, J.; Elliott, J.L.; Bassel-Duby, R.; Sanes, J.R.; Olson, E.N. MicroRNA-206 delays ALS progression and promotes regeneration of neuromuscular synapses in mice. Science 2009, 326, 1549–1554. [Google Scholar] [CrossRef] [Green Version]
  66. DI Pietro, L.; Baranzini, M.; Berardinelli, M.G.; Lattanzi, W.; Monforte, M.; Tasca, G.; Conte, A.; Logroscino, G.; Michetti, F.; Ricci, E.; et al. Potential therapeutic targets for ALS: MIR206, MIR208b and MIR499 are modulated during disease progression in the skeletal muscle of patients. Sci. Rep. 2017, 7. [Google Scholar] [CrossRef]
  67. Pegoraro, V.; Marozzo, R.; Angelini, C. MicroRNAs and HDAC4 protein expression in the skeletal muscle of ALS patients. Clin. Neuropathol. 2020, 39, 105–114. [Google Scholar] [CrossRef]
  68. Lle Bruneteau, G.; Simonet, T.; Phanie Bauché, S.; Mandjee, N.; Malfatti, E.; Girard, E.; Tanguy, M.-L.; Behin, A.; Dé, F.; Khiami, R.; et al. Muscle histone deacetylase 4 upregulation in amyotrophic lateral sclerosis: Potential role in reinnervation ability and disease progression. Brain 2013, 136, 2359–2368. [Google Scholar] [CrossRef]
  69. Cohen, T.J.; Waddell, D.S.; Barrientos, T.; Lu, Z.; Feng, G.; Cox, G.A.; Bodine, S.C.; Yao, T.P. The histone deacetylase HDAC4 connects neural activity to muscle transcriptional reprogramming. J. Biol. Chem. 2007, 282, 33752–33759. [Google Scholar] [CrossRef] [Green Version]
  70. Moresi, V.; Williams, A.H.; Meadows, E.; Flynn, J.M.; Potthoff, M.J.; McAnally, J.; Shelton, J.M.; Backs, J.; Klein, W.H.; Richardson, J.A.; et al. Myogenin and class II HDACs control neurogenic muscle atrophy by inducing E3 ubiquitin ligases. Cell 2010, 143, 35–45. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  71. Choi, M.C.; Cohen, T.J.; Barrientos, T.; Wang, B.; Li, M.; Simmons, B.J.; Yang, J.S.; Cox, G.A.; Zhao, Y.; Yao, T.P. A Direct HDAC4-MAP Kinase Crosstalk Activates Muscle Atrophy Program. Mol. Cell 2012, 47, 122–132. [Google Scholar] [CrossRef] [Green Version]
  72. Buonvicino, D.; Felici, R.; Ranieri, G.; Caramelli, R.; Lapucci, A.; Cavone, L.; Muzzi, M.; Di Pietro, L.; Bernardini, C.; Zwergel, C.; et al. Effects of Class II-Selective Histone Deacetylase Inhibitor on Neuromuscular Function and Disease Progression in SOD1-ALS Mice. Neuroscience 2018, 379, 228–238. [Google Scholar] [CrossRef] [PubMed]
  73. Pigna, E.; Simonazzi, E.; Sanna, K.; Bernadzki, K.M.; Proszynski, T.; Heil, C.; Palacios, D.; Adamo, S.; Moresi, V. Histone deacetylase 4 protects from denervation and skeletal muscle atrophy in a murine model of amyotrophic lateral sclerosis. EBioMedicine 2019, 40, 717–732. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Dobrowolny, G.; Martone, J.; Lepore, E.; Casola, I.; Petrucci, A.; Inghilleri, M.; Morlando, M.; Colantoni, A.; Scicchitano, B.M.; Calvo, A.; et al. A longitudinal study defined circulating microRNAs as reliable biomarkers for disease prognosis and progression in ALS human patients. Cell Death Discov. 2021, 7. [Google Scholar] [CrossRef]
  75. Hegedus, J.; Putman, C.T.; Tyreman, N.; Gordon, T. Preferential motor unit loss in the SOD1 G93A transgenic mouse model of amyotrophic lateral sclerosis. J. Physiol. 2008, 586, 3337–3351. [Google Scholar] [CrossRef] [PubMed]
  76. Gordon, T.; Putman, C.T.; Hegedus, J. Amyotrophic lateral sclerosis-evidence of early denervation of fast-twitch muscles. Basic Appl. Myol. 2007, 17, 4. [Google Scholar]
  77. Di Pietro, L.; Lattanzi, W.; Bernardini, C. Skeletal muscle microRNAs as key players in the pathogenesis of amyotrophic lateral sclerosis. Int. J. Mol. Sci. 2018, 19, 1534. [Google Scholar] [CrossRef] [Green Version]
  78. Ma, G.; Wang, Y.; Li, Y.; Cui, L.; Zhao, Y.; Zhao, B.; Li, K. MiR-206, a key modulator of skeletal muscle development and disease. Int. J. Biol. Sci. 2015, 11, 345–352. [Google Scholar] [CrossRef] [PubMed]
  79. Chen, J.F.; Tao, Y.; Li, J.; Deng, Z.; Yan, Z.; Xiao, X.; Wang, D.Z. microRNA-1 and microRNA-206 regulate skeletal muscle satellite cell proliferation and differentiation by repressing Pax7. J. Cell Biol. 2010, 190, 867–879. [Google Scholar] [CrossRef] [Green Version]
  80. Dey, B.K.; Gagan, J.; Dutta, A. miR-206 and -486 Induce Myoblast Differentiation by Downregulating Pax7. Mol. Cell. Biol. 2011, 31, 203–214. [Google Scholar] [CrossRef] [Green Version]
  81. Hughes, S.M.; Taylor, J.M.; Tapscott, S.J.; Gurley, C.M.; Carter, W.J.; Peterson, C.A. Selective accumulation of MyoD and myogenin mRNAs in fast and slow adult skeletal muscle is controlled by innervation and hormones. Development 1993, 118, 1137–1147. [Google Scholar]
  82. Maves, L.; Waskiewicz, A.J.; Paul, B.; Cao, Y.; Tyler, A.; Moens, C.B.; Tapscott, S.J. Pbx homeodomain proteins direct Myod activity to promote fast-muscle differentiation. Development 2007, 134, 3371–3382. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Ekmark, M.; Rana, Z.A.; Stewart, G.; Hardie, D.G.; Gundersen, K. De-phosphorylation of MyoD is linking nerve-evoked activity to fast myosin heavy chain expression in rodent adult skeletal muscle. J. Physiol. 2007, 584, 637–650. [Google Scholar] [CrossRef] [PubMed]
  84. Hughes, S.M.; Chi, M.M.Y.; Lowry, O.H.; Gundersen, K. Myogenin induces a shift of enzyme activity from glycolytic to oxidative metabolism in muscles of transgenic mice. J. Cell Biol. 1999, 145, 633–642. [Google Scholar] [CrossRef] [PubMed]
  85. Ekmark, M.; Grønevik, E.; Schjerling, P.; Gundersen, K. Myogenin induces higher oxidative capacity in pre-existing mouse muscle fibres after somatic DNA transfer. J. Physiol. 2003, 548, 259–269. [Google Scholar] [CrossRef]
  86. Park, K.H.J.; Franciosi, S.; Leavitt, B.R. Postnatal muscle modification by myogenic factors modulates neuropathology and survival in an ALS mouse model. Nat. Commun. 2013, 4. [Google Scholar] [CrossRef] [Green Version]
  87. Peggion, C.; Massimino, M.L.; Biancotto, G.; Angeletti, R.; Reggiani, C.; Sorgato, M.C.; Bertoli, A.; Stella, R.; Stella, R. Absolute quantification of myosin heavy chain isoforms by selected reaction monitoring can underscore skeletal muscle changes in a mouse model of amyotrophic lateral sclerosis. Anal. Bioanal. Chem. 2017. [Google Scholar] [CrossRef] [PubMed]
  88. Dobrowolny, G.; Lepore, E.; Martini, M.; Barberi, L.; Nunn, A.; Scicchitano, B.M.; Musarò, A. Metabolic Changes Associated with Muscle Expression of SOD1 G93A. Front. Physiol. 2018, 9, 831. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  89. Palamiuc, L.; Schlagowski, A.; Ngo, S.T.; Vernay, A.; Dirrig-Grosch, S.; Henriques, A.; Boutillier, A.-L.; Zoll, J.; Echaniz-Laguna, A.; Loeffler, J.-P.; et al. A metabolic switch toward lipid use in glycolytic muscle is an early pathologic event in a mouse model of amyotrophic lateral sclerosis. EMBO Mol. Med. 2015, 7, 526–546. [Google Scholar] [CrossRef]
  90. Scaricamazza, S.; Salvatori, I.; Giacovazzo, G.; Loeffler, J.P.; Renè, F.; Rosina, M.; Quessada, C.; Proietti, D.; Heil, C.; Rossi, S.; et al. Skeletal-muscle metabolic reprogramming in ALS-SOD1 mice predates disease onset and is a promising therapeutical target. iScience 2020, 23, 101087. [Google Scholar] [CrossRef]
  91. Mattiazzi, M.; D’Aurelio, M.; Gajewski, C.D.; Martushova, K.; Kiaei, M.; Flint Beal, M.; Manfredi, G. Mutated human SOD1 causes dysfunction of oxidative phosphorylation in mitochondria of transgenic mice. J. Biol. Chem. 2002, 277, 29626–29633. [Google Scholar] [CrossRef] [Green Version]
  92. Cozzolino, M.; Ferri, A.; Ferraro, E.; Rotilio, G.; Cecconi, F.; Carrì, M.T. Apaf1 mediates apoptosis and mitochondrial damage induced by mutant human SOD1s typical of familial amyotrophic lateral sclerosis. Neurobiol. Dis. 2006, 21, 69–79. [Google Scholar] [CrossRef]
  93. Handschin, C. Regulation of skeletal muscle cell plasticity by the peroxisome proliferator-activated receptor γ coactivator 1α. J. Recept. Signal Transduct. 2010, 30, 376–384. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Lin, J.; Handschin, C.; Spiegelman, B.M. Metabolic control through the PGC-1 family of transcription coactivators. Cell Metab. 2005, 1, 361–370. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Da Cruz, S.; Parone, P.A.; Lopes, V.S.; Lillo, C.; McAlonis-Downes, M.; Lee, S.K.; Vetto, A.P.; Petrosyan, S.; Marsala, M.; Murphy, A.N.; et al. Elevated PGC-1α activity sustains mitochondrial biogenesis and muscle function without extending survival in a mouse model of inherited ALS. Cell Metab. 2012, 15, 778–786. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Dupuis, L.; Gonzalez de Aguilar, J.L.; Echaniz-Laguna, A.; Eschbach, J.; Rene, F.; Oudart, H.; Halter, B.; Huze, C.; Schaeffer, L.; Bouillaud, F.; et al. Muscle mitochondrial uncoupling dismantles neuromuscular junction and triggers distal degeneration of motor neurons. PLoS ONE 2009, 4, e5390. [Google Scholar] [CrossRef] [Green Version]
  97. Natale, G.; Lenzi, P.; Lazzeri, G.; Falleni, A.; Biagioni, F.; Ryskalin, L.; Fornai, F. Compartment-dependent mitochondrial alterations in experimental als, the effects of mitophagy and mitochondriogenesis. Front. Cell. Neurosci. 2015, 9, 434. [Google Scholar] [CrossRef] [Green Version]
  98. Parone, P.A.; Da Cruz, S.; Han, J.S.; McAlonis-Downes, M.; Vetto, A.P.; Lee, S.K.; Tseng, E.; Cleveland, D.W. Enhancing mitochondrial calcium buffering capacity reduces aggregation of misfolded SOD1 and motor neuron cell death without extending survival in mouse models of inherited amyotrophic lateral sclerosis. J. Neurosci. 2013, 33, 4657–4671. [Google Scholar] [CrossRef]
  99. Ruffoli, R.; Bartalucci, A.; Frati, A.; Fornai, F. Ultrastructural studies of ALS mitochondria connect altered function and permeability with defects of mitophagy and mitochondriogenesis. Front. Cell. Neurosci. 2015, 9, 341. [Google Scholar] [CrossRef] [Green Version]
  100. Riis, S.; Murray, J.B.; O’Connor, R. IGF-1 Signalling Regulates Mitochondria Dynamics and Turnover through a Conserved GSK-3β–Nrf2–BNIP3 Pathway. Cells 2020, 9, 147. [Google Scholar] [CrossRef] [Green Version]
  101. Ahmad, S.S.; Ahmad, K.; Lee, E.J.; Lee, Y.H.; Choi, I. Implications of Insulin-Like Growth Factor-1 in Skeletal Muscle and Various Diseases. Cells 2020, 9, 1773. [Google Scholar] [CrossRef]
  102. Yoshida, T.; Delafontaine, P. Mechanisms of IGF-1-Mediated Regulation of Skeletal Muscle Hypertrophy and Atrophy. Cells 2020, 9, 1970. [Google Scholar] [CrossRef] [PubMed]
  103. Song, Y.H.; Song, J.L.; Delafontaine, P.; Godard, M.P. The therapeutic potential of IGF-I in skeletal muscle repair. Trends Endocrinol. Metab. 2013, 24, 310–319. [Google Scholar] [CrossRef] [Green Version]
  104. Musaro, A.; Rosenthal, N. The Role of local Insulin-like Growth Factor-1 Isoforms in the Pathophysiology of Skeletal Muscle. Curr. Genom. 2002, 3, 149–162. [Google Scholar] [CrossRef]
  105. Dobrowolny, G.; Giacinti, C.; Pelosi, L.; Nicoletti, C.; Winn, N.; Barberi, L.; Molinaro, M.; Rosenthal, N.; Musarò, A. Muscle expression of a local Igf-1 isoform protects motor neurons in an ALS mouse model. J. Cell Biol. 2005, 168, 193–199. [Google Scholar] [CrossRef] [Green Version]
  106. Dobrowolny, G.; Aucello, M.; Molinaro, M.; Musarò, A. Local expression of mIgf-1 modulates ubiquitin, caspase and CDK5 expression in skeletal muscle of an ALS mouse model. Neurol. Res. 2008, 30, 131–136. [Google Scholar] [CrossRef]
  107. Nagel, G.; Peter, R.S.; Rosenbohm, A.; Koenig, W.; Dupuis, L.; Rothenbacher, D.; Ludolph, A.C. Association of Insulin-like Growth Factor 1 Concentrations with Risk for and Prognosis of Amyotrophic Lateral Sclerosis—Results from the ALS Registry Swabia. Sci. Rep. 2020, 10, 736. [Google Scholar] [CrossRef]
  108. Ngo, S.T.; Steyn, F.J. The interplay between metabolic homeostasis and neurodegeneration: Insights into the neurometabolic nature of amyotrophic lateral sclerosis. Cell Regen. 2015, 4, 4–5. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  109. Sharma, S.; Black, S.M. Carnitine homeostasis, mitochondrial function and cardiovascular disease. Drug Discov. Today Dis. Mech. 2009, 6, e31–e39. [Google Scholar] [CrossRef] [Green Version]
  110. Fielding, R.; Riede, L.; Lugo, J.P.; Bellamine, A. L-carnitine supplementation in recovery after exercise. Nutrients 2018, 10, 349. [Google Scholar] [CrossRef] [Green Version]
  111. Montesano, A.; Senesi, P.; Luzi, L.; Benedini, S.; Terruzzi, I. Potential therapeutic role of L-carnitine in skeletal muscle oxidative stress and atrophy conditions. Oxid. Med. Cell. Longev. 2015, 2015. [Google Scholar] [CrossRef]
  112. Kira, Y.; Nishikawa, M.; Ochi, A.; Sato, E.; Inoue, M. L-Carnitine suppresses the onset of neuromuscular degeneration and increases the life span of mice with familial amyotrophic lateral sclerosis. Brain Res. 2006, 1070, 206–214. [Google Scholar] [CrossRef]
  113. Beghi, E.; Pupillo, E.; Bonito, V.; Buzzi, P.; Caponnetto, C.; Chiò, A.; Corbo, M.; Giannini, F.; Inghilleri, M.; La Bella, V.; et al. Randomized double-blind placebo-controlled trial of acetyl-L-carnitine for ALS. Amyotroph. Lateral Scler. Front. Degener. 2013, 14, 397–405. [Google Scholar] [CrossRef]
  114. Dupuis, L.; Oudart, H.; René, F.; Gonzalez De Aguilar, J.L.; Loeffler, J.P. Evidence for defective energy homeostasis in amyotrophic lateral sclerosis: Benefit of a high-energy diet in a transgenic mouse model. Proc. Natl. Acad. Sci. USA 2004, 101, 11159–11164. [Google Scholar] [CrossRef] [Green Version]
  115. Manzo, E.; O’Conner, A.G.; Barrows, J.M.; Shreiner, D.D.; Birchak, G.J.; Zarnescu, D.C. Medium-chain fatty acids, beta-hydroxybutyric acid and genetic modulation of the carnitine shuttle are protective in a drosophila model of ALS based on TDP-43. Front. Mol. Neurosci. 2018, 11. [Google Scholar] [CrossRef]
  116. Ludolph, A.C.; Dorst, J.; Dreyhaupt, J.; Weishaupt, J.H.; Kassubek, J.; Weiland, U.; Meyer, T.; Petri, S.; Hermann, A.; Emmer, A.; et al. Effect of High-Caloric Nutrition on Survival in Amyotrophic Lateral Sclerosis. Ann. Neurol. 2020, 87, 206–216. [Google Scholar] [CrossRef]
  117. Guimarães-Ferreira, L. Role of the phosphocreatine system on energetic homeostasis in skeletal and cardiac muscles. Einstein 2014, 12, 126–131. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  118. Wallimann, T.; Tokarska-Schlattner, M.; Schlattner, U. The creatine kinase system and pleiotropic effects of creatine. Amino Acids 2011, 40, 1271–1296. [Google Scholar] [CrossRef] [Green Version]
  119. McMahon, S.; Jenkins, D. Factors affecting the rate of phosphocreatine resynthesis following intense exercise. Sport. Med. 2002, 32, 761–784. [Google Scholar] [CrossRef]
  120. Katz, A.; Andersson, D.C.; Yu, J.; Norman, B.; Sandström, M.E.; Wieringa, B.; Westerblad, H. Contraction-mediated glycogenolysis in mouse skeletal muscle lacking creatine kinase: The role of phosphorylase b activation. J. Physiol. 2003, 553, 523–531. [Google Scholar] [CrossRef] [PubMed]
  121. Tarnopolsky, M.A. Caffeine and Creatine Use in Sport. Ann. Nutr. Metab. 2010, 57, 1–8. [Google Scholar] [CrossRef] [PubMed]
  122. Klivenyi, P.; Ferrante, R.J.; Matthews, R.T.; Bogdanov, M.B.; Klein, A.M.; Andreassen, O.A.; Mueller, G.; Wermer, M.; Kaddurah-Daouk, R.; Beal, M.F. Neuroprotective effects of creatine in a transgenic animal model of amyotrophic lateral sclerosis. Nat. Med. 1999, 5, 347–350. [Google Scholar] [CrossRef] [PubMed]
  123. Andreassen, O.A.; Jenkins, B.G.; Dedeoglu, A.; Ferrante, K.L.; Bogdanov, M.B.; Kaddurah-Daouk, R.; Beal, M.F. Increases in cortical glutamate concentrations in transgenic amyotrophic lateral sclerosis mice are attenuated by creatine supplementation. J. Neurochem. 2001, 77, 383–390. [Google Scholar] [CrossRef] [Green Version]
  124. Snow, R.J.; Turnbull, J.; Da Silva, S.; Jiang, F.; Tarnopolsky, M.A. Creatine supplementation and riluzole treatment provide similar beneficial effects in copper, zinc superoxide dismutase (G93A) transgenic mice. Neuroscience 2003, 119, 661–667. [Google Scholar] [CrossRef]
  125. Scott, S.; Kranz, J.E.; Cole, J.; Lincecum, J.M.; Thompson, K.; Kelly, N.; Bostrom, A.; Theodoss, J.; Al-Nakhala, B.M.; Vieira, F.G.; et al. Design, power, and interpretation of studies in the standard murine model of ALS. Amyotroph. Lateral Scler. 2008, 9, 4–15. [Google Scholar] [CrossRef]
  126. Groeneveld, G.J.; Veldink, J.H.; Van der Tweel, I.; Kalmijn, S.; Beijer, C.; De Visser, M.; Wokke, J.H.J.; Franssen, H.; Van den Berg, L.H. A randomized sequential trial of creatine in amyotrophic lateral sclerosis. Ann. Neurol. 2003, 53, 437–445. [Google Scholar] [CrossRef] [PubMed]
  127. Babu, S.; Macklin, E.A.; Jackson, K.E.; Simpson, E.; Mahoney, K.; Yu, H.; Walker, J.; Simmons, Z.; David, W.S.; Barkhaus, P.E.; et al. Selection design phase II trial of high dosages of tamoxifen and creatine in amyotrophic lateral sclerosis. Amyotroph. Lateral Scler. Front. Degener. 2020, 21, 15–23. [Google Scholar] [CrossRef]
  128. Rosenfeld, J.; King, R.M.; Jackson, C.E.; Bedlack, R.S.; Barohn, R.J.; Dick, A.; Phillips, L.H.; Chapin, J.; Gelinas, D.F.; Lou, J.S. Creatine monohydrate in ALS: Effects on strength, fatigue, respiratory status and ALSFRS. Amyotroph. Lateral Scler. 2008, 9, 266–272. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  129. Bender, A.; Klopstock, T. Creatine for neuroprotection in neurodegenerative disease: End of story? Amino Acids 2016, 48, 1929–1940. [Google Scholar] [CrossRef] [PubMed]
  130. Ceccanti, M.; Pozzilli, V.; Cambieri, C.; Libonati, L.; Onesti, E.; Frasca, V.; Fiorini, I.; Petrucci, A.; Garibaldi, M.; Palma, E.; et al. Creatine Kinase and Progression Rate in Amyotrophic Lateral Sclerosis. Cells 2020, 9, 1174. [Google Scholar] [CrossRef]
  131. Miquel, E.; Cassina, A.; Martínez-Palma, L.; Bolatto, C.; Trías, E.; Gandelman, M.; Radi, R.; Barbeito, L.; Cassina, P. Modulation of astrocytic mitochondrial function by dichloroacetate improves survival and motor performance in inherited amyotrophic lateral sclerosis. PLoS ONE 2012, 7, e34776. [Google Scholar] [CrossRef]
  132. DeAngelo, A.B.; George, M.H.; House, D.E. Hepatocarcinogenicity in the male B6C3F1 mouse following a lifetime exposure to dichloroacetic acid in the drinking water: Dose-response determination and modes of action. J. Toxicol. Environ. Heal. Part A 1999, 58, 485–507. [Google Scholar] [CrossRef]
  133. Jensen, L.; Jørgensen, L.H.; Bech, R.D.; Frandsen, U.; Schrøder, H.D. Skeletal Muscle Remodelling as a Function of Disease Progression in Amyotrophic Lateral Sclerosis. Biomed Res. Int. 2016, 2016, 5930621. [Google Scholar] [CrossRef] [Green Version]
  134. Yoo, Y.E.; Ko, C.P. Dihydrotestosterone ameliorates degeneration in muscle, axons and motoneurons and improves motor function in amyotrophic lateral sclerosis model mice. PLoS ONE 2012, 7, e37258. [Google Scholar] [CrossRef] [Green Version]
  135. Wen, D.; Cui, C.; Duan, W.; Wang, W.; Wang, Y.; Liu, Y.; Li, Z.; Li, C. The role of insulin-like growth factor 1 in ALS cell and mouse models: A mitochondrial protector. Brain Res. Bull. 2019, 144, 1–13. [Google Scholar] [CrossRef]
  136. Cappello, V.; Vezzoli, E.; Righi, M.; Fossati, M.; Mariotti, R.; Crespi, A.; Patruno, M.; Bentivoglio, M.; Pietrini, G.; Francolini, M. Analysis of neuromuscular junctions and effects of anabolic steroid administration in the SOD1G93A mouse model of ALS. Mol. Cell. Neurosci. 2012, 51, 12–21. [Google Scholar] [CrossRef]
  137. Galbiati, M.; Onesto, E.; Zito, A.; Crippa, V.; Rusmini, P.; Mariotti, R.; Bentivoglio, M.; Bendotti, C.; Poletti, A. The anabolic/androgenic steroid nandrolone exacerbates gene expression modifications induced by mutant SOD1 in muscles of mice models of amyotrophic lateral sclerosis. Pharmacol. Res. 2012, 65, 221–230. [Google Scholar] [CrossRef] [Green Version]
  138. Peters, S.; Zitzelsperger, E.; Kuespert, S.; Iberl, S.; Heydn, R.; Johannesen, S.; Petri, S.; Aigner, L.; Thal, D.R.; Hermann, A.; et al. The TGF-β system as a potential pathogenic player in disease modulation of amyotrophic lateral sclerosis. Front. Neurol. 2017, 8. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  139. Bonnieu, A.; Carnac, G.; Vernus, B. Myostatin in the Pathophysiology of Skeletal Muscle. Curr. Genom. 2007, 8, 415–422. [Google Scholar] [CrossRef]
  140. Tobin, J.F.; Celeste, A.J. Myostatin, a negative regulator of muscle mass: Implications for muscle degenerative diseases. Curr. Opin. Pharmacol. 2005, 5, 328–332. [Google Scholar] [CrossRef]
  141. Trendelenburg, A.U.; Meyer, A.; Rohner, D.; Boyle, J.; Hatakeyama, S.; Glass, D.J. Myostatin reduces Akt/TORC1/p70S6K signaling, inhibiting myoblast differentiation and myotube size. Am. J. Physiol. Cell Physiol. 2009, 296, 1258–1270. [Google Scholar] [CrossRef] [Green Version]
  142. Holzbaur, E.L.F.; Howland, D.S.; Weber, N.; Wallace, K.; She, Y.; Kwak, S.; Tchistiakova, L.A.; Murphy, E.; Hinson, J.; Karim, R.; et al. Myostatin inhibition slows muscle atrophy in rodent models of amyotrophic lateral sclerosis. Neurobiol. Dis. 2006, 23, 697–707. [Google Scholar] [CrossRef]
  143. Zimmers, T.A.; Davies, M.V.; Koniaris, L.G.; Haynes, P.; Esquela, A.F.; Tomkinson, K.N.; McPherron, A.C.; Wolfman, N.M.; Lee, S.J. Induction of cachexia in mice by systemically administered myostatin. Science 2002, 296, 1486–1488. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. Pirruccello-Straub, M.; Jackson, J.; Wawersik, S.; Webster, M.T.; Salta, L.; Long, K.; McConaughy, W.; Capili, A.; Boston, C.; Carven, G.J.; et al. Blocking extracellular activation of myostatin as a strategy for treating muscle wasting. Sci. Rep. 2018, 8. [Google Scholar] [CrossRef]
  145. Lightfoot, A.P.; Cooper, R.G. The role of myokines in muscle health and disease. Curr. Opin. Rheumatol. 2016, 28, 661–666. [Google Scholar] [CrossRef] [Green Version]
  146. Duan, K.; Gao, X.; Zhu, D. The clinical relevance and mechanism of skeletal muscle wasting. Clin. Nutr. 2021, 40, 27–37. [Google Scholar] [CrossRef] [PubMed]
  147. Tasca, E.; Pegoraro, V.; Merico, A.; Angelini, C. Circulating microRNAs as biomarkers of muscle differentiation and atrophy in ALS. Clin. Neuropathol. 2016, 35, 22–30. [Google Scholar] [CrossRef] [PubMed]
  148. Morrison, B.M.; Lachey, J.L.; Warsing, L.C.; Ting, B.L.; Pullen, A.E.; Underwood, K.W.; Kumar, R.; Sako, D.; Grinberg, A.; Wong, V.; et al. A soluble activin type IIB receptor improves function in a mouse model of amyotrophic lateral sclerosis. Exp. Neurol. 2009, 217, 258–268. [Google Scholar] [CrossRef]
  149. Bondulich, M.K.; Jolinon, N.; Osborne, G.F.; Smith, E.J.; Rattray, I.; Neueder, A.; Sathasivam, K.; Ahmed, M.; Ali, N.; Benjamin, A.C.; et al. Myostatin inhibition prevents skeletal muscle pathophysiology in Huntington’s disease mice. Sci. Rep. 2017, 7, 14275. [Google Scholar] [CrossRef] [Green Version]
  150. Rose, F.F.; Mattis, V.B.; Rindt, H.; Lorson, C.L. Delivery of recombinant follistatin lessens disease severity in a mouse model of spinal muscular atrophy. Hum. Mol. Genet. 2009, 18, 997–1005. [Google Scholar] [CrossRef] [Green Version]
  151. Jokic, N.; Gonzalez De Aguilar, J.-L.; Dimou, L.; Lin, S.; Fergani, A.; Ruegg, M.A.; Schwab, M.E.; Dupuis, L.; Loeffler, J.-P. The neurite outgrowth inhibitor Nogo-A promotes denervation in an amyotrophic lateral sclerosis model. EMBO Rep. 2003. [Google Scholar] [CrossRef] [Green Version]
  152. Pradat, P.F.; Bruneteau, G.; Gonzalez De Aguilar, J.L.; Dupuis, L.; Jokic, N.; Salachas, F.; Le Forestier, N.; Echaniz-Laguna, A.; Dubourg, O.; Hauw, J.J.; et al. Muscle Nogo-A expression is a prognostic marker in lower motor neuron syndromes. Ann. Neurol. 2007, 62, 15–20. [Google Scholar] [CrossRef]
  153. Meininger, V.; Genge, A.; van den Berg, L.H.; Robberecht, W.; Ludolph, A.; Chio, A.; Kim, S.H.; Leigh, P.N.; Kiernan, M.C.; Shefner, J.M.; et al. Safety and efficacy of ozanezumab in patients with amyotrophic lateral sclerosis: A randomised, double-blind, placebo-controlled, phase 2 trial. Lancet Neurol. 2017, 16, 208–216. [Google Scholar] [CrossRef] [Green Version]
  154. Cantor, S.; Zhang, W.; Delestrée, N.; Remédio, L.; Mentis, G.Z.; Burden, S.J. Preserving neuromuscular synapses in ALS by stimulating MuSK with a therapeutic agonist antibody. Elife 2018, 7. [Google Scholar] [CrossRef] [Green Version]
  155. Sengupta-Ghosh, A.; Dominguez, S.L.; Xie, L.; Barck, K.H.; Jiang, Z.; Earr, T.; Imperio, J.; Phu, L.; Budayeva, H.G.; Kirkpatrick, D.S.; et al. Muscle specific kinase (MuSK) activation preserves neuromuscular junctions in the diaphragm but is not sufficient to provide a functional benefit in the SOD1G93A mouse model of ALS. Neurobiol. Dis. 2019, 124, 340–352. [Google Scholar] [CrossRef]
  156. Palma, E.; Reyes-Ruiz, J.M.; Lopergolo, D.; Roseti, C.; Bertollini, C.; Ruffolo, G.; Cifelli, P.; Onesti, E.; Limatola, C.; Miledi, R.; et al. Acetylcholine receptors from human muscle as pharmacological targets for ALS therapy. Proc. Natl. Acad. Sci. USA 2016, 113, 3060–3065. [Google Scholar] [CrossRef] [Green Version]
  157. Deflorio, C.; Palma, E.; Conti, L.; Roseti, C.; Manteca, A.; Giacomelli, E.; Catalano, M.; Limatola, C.; Inghilleri, M.; Grassi, F. Riluzole blocks human muscle acetylcholine receptors. J. Physiol. 2012, 590, 2519–2528. [Google Scholar] [CrossRef] [Green Version]
  158. Collibee, S.E.; Bergnes, G.; Muci, A.; Browne, W.F.; Garard, M.; Hinken, A.C.; Russell, A.J.; Suehiro, I.; Hartman, J.; Kawas, R.; et al. Discovery of Tirasemtiv, the First Direct Fast Skeletal Muscle Troponin Activator. ACS Med. Chem. Lett. 2018, 9, 354–358. [Google Scholar] [CrossRef] [Green Version]
  159. Hwee, D.T.; Kennedy, A.; Ryans, J.; Russell, A.J.; Jia, Z.; Hinken, A.C.; Morgans, D.J.; Malik, F.I.; Jasper, J.R. Fast Skeletal Muscle Troponin Activator tirasemtiv Increases Muscle Function and Performance in the B6SJL-SOD1G93A ALS Mouse Model. PLoS ONE 2014, 9, e96921. [Google Scholar] [CrossRef]
  160. Shefner, J.M.; Cudkowicz, M.E.; Hardiman, O.; Cockroft, B.M.; Lee, J.H.; Malik, F.I.; Meng, L.; Rudnicki, S.A.; Wolff, A.A.; Andrews, J.A. A phase III trial of tirasemtiv as a potential treatment for amyotrophic lateral sclerosis. Amyotroph. Lateral Scler. Front. Degener. 2019, 20, 584–594. [Google Scholar] [CrossRef] [Green Version]
  161. Shefner, J.M.; Andrews, J.A.; Genge, A.; Jackson, C.; Lechtzin, N.; Miller, T.M.; Cockroft, B.M.; Meng, L.; Wei, J.; Wolff, A.A.; et al. A Phase 2, Double-Blind, Randomized, Dose-Ranging Trial Of Reldesemtiv In Patients With ALS. Amyotroph. Lateral Scler. Front. Degener. 2020. [Google Scholar] [CrossRef]
  162. Al-Chalabi, A.; Shaw, P.; Leigh, P.N.; Van Den Berg, L.; Hardiman, O.; Ludolph, A.; Aho, V.V.; Sarapohja, T.; Kuoppamäki, M. Oral levosimendan in amyotrophic lateral sclerosis: A phase II multicentre, randomised, double-blind, placebo-controlled trial. J. Neurol. Neurosurg. Psychiatry 2019, 90. [Google Scholar] [CrossRef] [Green Version]
  163. Gonzalez, D.; Rebolledo, D.L.; Correa, L.M.; Court, F.A.; Cerpa, W.; Lipson, K.E.; Van Zundert, B.; Brandan, E. The inhibition of CTGF/CCN2 activity improves muscle and locomotor function in a murine ALS model. Hum. Mol. Genet. 2018, 27, 2913–2926. [Google Scholar] [CrossRef] [Green Version]
  164. Gonzalez, D.; Brandan, E. CTGF/CCN2 from Skeletal Muscle to Nervous System: Impact on Neurodegenerative Diseases. Mol. Neurobiol. 2019, 56, 5911–5916. [Google Scholar] [CrossRef]
  165. Chen, Z.; Zhang, N.; Chu, H.Y.; Yu, Y.; Zhang, Z.K.; Zhang, G.; Zhang, B.T. Connective Tissue Growth Factor: From Molecular Understandings to Drug Discovery. Front. Cell Dev. Biol. 2020, 8, 1239. [Google Scholar] [CrossRef]
  166. Morales, M.G.; Gutierrez, J.; Cabello-Verrugio, C.; Cabrera, D.; Lipson, K.E.; Goldschmeding, R.; Brandan, E. Reducing CTGF/CCN2 slows down mdx muscle dystrophy and improves cell therapy. Hum. Mol. Genet. 2013, 22, 4938–4951. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  167. Barnes, P.J. Theophylline. Pharmaceuticals 2010, 3, 725–747. [Google Scholar] [CrossRef] [Green Version]
  168. Ramsdell, J. Use of theophylline in the treatment of COPD. Chest 1995, 107, 206S–209S. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  169. Cazzola, M.; Matera, M.G. The effect of doxofylline in asthma and COPD. Respir. Med. 2020, 164, 105904. [Google Scholar] [CrossRef] [PubMed]
  170. Donohue, J.F. Therapeutic responses in asthma and COPD: Bronchodilators. Chest 2004, 126, 125S–137S. [Google Scholar] [CrossRef] [PubMed]
  171. Yu, T.-J.; Liu, Y.-C.; Chu, C.-M.; Hu, H.-C.; Kao, K.-C. Effects of theophylline therapy on respiratory muscle strength in patients with prolonged mechanical ventilation. Medicine 2019, 98, e13982. [Google Scholar] [CrossRef]
  172. Schiffman, P.L.; Belsh, J.M. Effect of Inspiratory Resistance and Theophylline on Respiratory Muscle Strength in Patients with Amyotrophic Lateral Sclerosis. Am. Rev. Respir. Dis. 1989, 139, 1418–1423. [Google Scholar] [CrossRef] [PubMed]
  173. Berto, M.C.; Filha, S.C.; Camelier, A.; Rosa, F.W.; De Souza Bulle Oliveira, A.; Jardim, J.R. Acute action of aminophylline in patients with amyotrophic lateral sclerosis. Acta Neurol. Scand. 2007, 115, 301–305. [Google Scholar] [CrossRef] [PubMed]
  174. Fabbrizio, P.; Apolloni, S.; Bianchi, A.; Salvatori, I.; Valle, C.; Lanzuolo, C.; Bendotti, C.; Nardo, G.; Volonté, C. P2X7 activation enhances skeletal muscle metabolism and regeneration in SOD1G93A mouse model of amyotrophic lateral sclerosis. Brain Pathol. 2020, 30, 272–282. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  175. Ferraro, E.; Giammarioli, A.M.; Chiandotto, S.; Spoletini, I.; Rosano, G. Exercise-induced skeletal muscle remodeling and metabolic adaptation: Redox signaling and role of autophagy. Antioxid. Redox Signal. 2014, 21, 154–176. [Google Scholar] [CrossRef] [PubMed]
  176. Saraulli, D.; Costanzi, M.; Mastrorilli, V.; Farioli-Vecchioli, S. The Long Run: Neuroprotective Effects of Physical Exercise on Adult Neurogenesis from Youth to Old Age. Curr. Neuropharmacol. 2017, 15, 519–533. [Google Scholar] [CrossRef] [Green Version]
  177. Gillon, A.; Nielsen, K.; Steel, C.; Cornwall, J.; Sheard, P. Exercise attenuates age-associated changes in motoneuron number, nucleocytoplasmic transport proteins and neuromuscular health. GeroScience 2018, 40, 177–192. [Google Scholar] [CrossRef]
  178. Rygiel, K.A.; Picard, M.; Turnbull, D.M. The ageing neuromuscular system and sarcopenia: A mitochondrial perspective. J. Physiol. 2016, 594, 4499–4512. [Google Scholar] [CrossRef] [PubMed]
  179. Sailani, M.R.; Halling, J.F.; Møller, H.D.; Lee, H.; Plomgaard, P.; Pilegaard, H.; Snyder, M.P.; Regenberg, B. Lifelong physical activity is associated with promoter hypomethylation of genes involved in metabolism, myogenesis, contractile properties and oxidative stress resistance in aged human skeletal muscle. Sci. Rep. 2019, 9. [Google Scholar] [CrossRef] [Green Version]
  180. Ruegsegger, G.N.; Booth, F.W. Health benefits of exercise. Cold Spring Harb. Perspect. Med. 2018, 8. [Google Scholar] [CrossRef] [Green Version]
  181. Nicolis di Robilant, V.; Scardigli, R.; Strimpakos, G.; Tirone, F.; Middei, S.; Scopa, C.; De Bardi, M.; Battistini, L.; Saraulli, D.; Farioli Vecchioli, S. Running-Activated Neural Stem Cells Enhance Subventricular Neurogenesis and Improve Olfactory Behavior in p21 Knockout Mice. Mol. Neurobiol. 2019, 56, 7534–7556. [Google Scholar] [CrossRef]
  182. Mastrorilli, V.; Scopa, C.; Saraulli, D.; Costanzi, M.; Scardigli, R.; Rouault, J.P.; Farioli-Vecchioli, S.; Tirone, F. Physical exercise rescues defective neural stem cells and neurogenesis in the adult subventricular zone of Btg1 knockout mice. Brain Struct. Funct. 2017, 222, 2855–2876. [Google Scholar] [CrossRef] [PubMed]
  183. Powers, S.K.; Deminice, R.; Ozdemir, M.; Yoshihara, T.; Bomkamp, M.P.; Hyatt, H. Exercise-induced oxidative stress: Friend or foe? J. Sport Heal. Sci. 2020, 9, 415–425. [Google Scholar] [CrossRef]
  184. Monteiro-Junior, R.S.; De Tarso Maciel-Pinheiro, P.; Da Matta Mello Portugal, E.; Da Silva Figueiredo, L.F.; Terra, R.; Carneiro, L.S.F.; Rodrigues, V.D.; Nascimento, O.J.M.; Deslandes, A.C.; Laks, J. Effect of exercise on inflammatory profile of older persons: Systematic review and meta-analyses. J. Phys. Act. Heal. 2018, 15, 64–71. [Google Scholar] [CrossRef]
  185. Webb, R.; Hughes, M.G.; Thomas, A.W.; Morris, K. The ability of exercise-associated oxidative stress to trigger redox-sensitive signalling responses. Antioxidants 2017, 6, 63. [Google Scholar] [CrossRef] [Green Version]
  186. Simioni, C.; Zauli, G.; Martelli, A.M.; Vitale, M.; Sacchetti, G.; Gonelli, A.; Neri, L.M. Oxidative stress: Role of physical exercise and antioxidant nutraceuticals in adulthood and aging. Oncotarget 2018, 9, 17181–17198. [Google Scholar] [CrossRef] [Green Version]
  187. Sayegh, A.L.C.; Degani-Costa, L.H. Effects of exercise training on endothelial and diastolic age-related dysfunctions: A new view of an old problem. J. Physiol. 2017, 595, 4591–4592. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  188. Hotta, K.; Chen, B.; Behnke, B.J.; Ghosh, P.; Stabley, J.N.; Bramy, J.A.; Sepulveda, J.L.; Delp, M.D.; Muller-Delp, J.M. Exercise training reverses age-induced diastolic dysfunction and restores coronary microvascular function. J. Physiol. 2017, 595, 3703–3719. [Google Scholar] [CrossRef] [PubMed]
  189. Shibata, S.; Fujimoto, N.; Hastings, J.L.; Carrick-Ranson, G.; Bhella, P.S.; Hearon, C.M.; Levine, B.D. The effect of lifelong exercise frequency on arterial stiffness. J. Physiol. 2018, 596, 2783–2795. [Google Scholar] [CrossRef] [PubMed]
  190. Rebelo-Marques, A.; Lages, A.D.S.; Andrade, R.; Ribeiro, C.F.; Mota-Pinto, A.; Carrilho, F.; Espregueira-Mendes, J. Aging hallmarks: The benefits of physical exercise. Front. Endocrinol. 2018, 9, 258. [Google Scholar] [CrossRef]
  191. Puterman, E.; Lin, J.; Blackburn, E.; O’Donovan, A.; Adler, N.; Epel, E. The power of exercise: Buffering the effect of chronic stress on telomere length. PLoS ONE 2010, 5. [Google Scholar] [CrossRef] [PubMed]
  192. Wyckelsma, V.L.; Levinger, I.; McKenna, M.J.; Formosa, L.E.; Ryan, M.T.; Petersen, A.C.; Anderson, M.J.; Murphy, R.M. Preservation of skeletal muscle mitochondrial content in older adults: Relationship between mitochondria, fibre type and high-intensity exercise training. J. Physiol. 2017, 595, 3345–3359. [Google Scholar] [CrossRef] [Green Version]
  193. Nyberg, M.; Blackwell, J.R.; Damsgaard, R.; Jones, A.M.; Hellsten, Y.; Mortensen, S.P. Lifelong physical activity prevents an age-related reduction in arterial and skeletal muscle nitric oxide bioavailability in humans. J. Physiol. 2012, 590, 5361–5370. [Google Scholar] [CrossRef] [Green Version]
  194. Joseph, A.M.; Adhihetty, P.J.; Leeuwenburgh, C. Beneficial effects of exercise on age-related mitochondrial dysfunction and oxidative stress in skeletal muscle. J. Physiol. 2016, 594, 5105–5123. [Google Scholar] [CrossRef] [Green Version]
  195. Sahl, R.E.; Andersen, P.R.; Gronbaek, K.; Morville, T.H.; Rosenkilde, M.; Rasmusen, H.K.; Poulsen, S.S.; Prats, C.; Dela, F.; Helge, J.W. Repeated excessive exercise attenuates the anti-inflammatory effects of exercise in older men. Front. Physiol. 2017, 8. [Google Scholar] [CrossRef] [Green Version]
  196. Gomez-Cabrera, M.C.; Viña, J.; Ji, L.L. Interplay of oxidants and antioxidants during exercise: Implications for muscle health. Phys. Sportsmed. 2009, 37, 116–123. [Google Scholar] [CrossRef]
  197. Carreras, I.; Yuruker, S.; Aytan, N.; Hossain, L.; Choi, J.K.; Jenkins, B.G.; Kowall, N.W.; Dedeoglu, A. Moderate exercise delays the motor performance decline in a transgenic model of ALS. Brain Res. 2010, 1313, 192–201. [Google Scholar] [CrossRef] [Green Version]
  198. Liebetanz, D.; Hagemann, K.; Von Lewinski, F.; Kahler, E.; Paulus, W. Extensive exercise is not harmful in amyotrophic lateral sclerosis. Eur. J. Neurosci. 2004, 20, 3115–3120. [Google Scholar] [CrossRef]
  199. Kaspar, B.K.; Frost, L.M.; Christian, L.; Umapathi, P.; Gage, F.H. Synergy of insulin-like growth factor-1 and exercise in amyotrophic lateral sclerosis. Ann. Neurol. 2005, 57, 649–655. [Google Scholar] [CrossRef]
  200. Bennett, E.J.; Mead, R.J.; Azzouz, M.; Shaw, P.J.; Grierson, A.J. Early detection of motor dysfunction in the SOD1G93A mouse model of amyotrophic lateral sclerosis (ALS) using home cage running wheels. PLoS ONE 2014, 9, e107918. [Google Scholar] [CrossRef]
  201. Mahoney, D.J.; Rodriguez, C.; Devries, M.; Yasuda, N.; Tarnopolsky, M.A. Effects of high-intensity endurance exercise training in the G93A mouse model of amyotrophic lateral sclerosis. Muscle Nerve 2004, 29, 656–662. [Google Scholar] [CrossRef]
  202. Kirkinezos, I.G.; Hernandez, D.; Bradley, W.G.; Moraes, C.T. Regular exercise is beneficial to a mouse model of amyotrophic lateral sclerosis. Ann. Neurol. 2003, 53, 804–807. [Google Scholar] [CrossRef] [PubMed]
  203. Veldink, J.H.; Bär, P.R.; Joosten, E.A.J.; Otten, M.; Wokke, J.H.J.; Van Den Berg, L.H. Sexual differences in onset of disease and response to exercise in a transgenic model of ALS. Neuromuscul. Disord. 2003, 13, 737–743. [Google Scholar] [CrossRef]
  204. Garbugino, L.; Golini, E.; Giuliani, A.; Mandillo, S. Prolonged voluntary running negatively affects survival and disease prognosis of male SOD1G93A low-copy transgenic mice. Front. Behav. Neurosci. 2018, 12, 275. [Google Scholar] [CrossRef] [Green Version]
  205. Deforges, S.; Branchu, J.; Biondi, O.; Grondard, C.; Pariset, C.; Lécolle, S.; Lopes, P.; Vidal, P.-P.; Chanoine, C.; Charbonnier, F. Motoneuron survival is promoted by specific exercise in a mouse model of amyotrophic lateral sclerosis. J. Physiol. 2009, 587, 3561–3572. [Google Scholar] [CrossRef] [PubMed]
  206. Just-Borràs, L.; Hurtado, E.; Cilleros-Mañé, V.; Biondi, O.; Charbonnier, F.; Tomàs, M.; Garcia, N.; Tomàs, J.; Lanuza, M.A. Running and swimming prevent the deregulation of the BDNF/TrkB neurotrophic signalling at the neuromuscular junction in mice with amyotrophic lateral sclerosis. Cell. Mol. Life Sci. 2020, 77, 3027–3040. [Google Scholar] [CrossRef] [PubMed]
  207. Flis, D.J.; Dzik, K.; Kaczor, J.J.; Halon-Golabek, M.; Antosiewicz, J.; Wieckowski, M.R.; Ziolkowski, W. Swim training modulates skeletal muscle energy metabolism, oxidative stress, and mitochondrial cholesterol content in amyotrophic lateral sclerosis mice. Oxid. Med. Cell. Longev. 2018, 2018, 5940748. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  208. Flis, D.J.; Dzik, K.; Kaczor, J.J.; Cieminski, K.; Halon-Golabek, M.; Antosiewicz, J.; Wieckowski, M.R.; Ziolkowski, W. Swim training modulates mouse skeletal muscle energy metabolism and ameliorates reduction in grip strength in a mouse model of amyotrophic lateral sclerosis. Int. J. Mol. Sci. 2019, 20, 233. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  209. Desseille, C.; Deforges, S.; Biondi, O.; Houdebine, L.; D’Amico, D.; Lamazière, A.; Caradeuc, C.; Bertho, G.; Bruneteau, G.; Weill, L.; et al. Specific physical exercise improves energetic metabolism in the skeletal muscle of amyotrophic-lateral- sclerosis mice. Front. Mol. Neurosci. 2017, 10, 332. [Google Scholar] [CrossRef] [PubMed]
  210. Nijssen, J.; Comley, L.H.; Hedlund, E. Motor neuron vulnerability and resistance in amyotrophic lateral sclerosis. Acta Neuropathol. 2017, 133, 863–885. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  211. Pradhan, J.; Noakes, P.G.; Bellingham, M.C. The Role of Altered BDNF/TrkB Signaling in Amyotrophic Lateral Sclerosis. Front. Cell. Neurosci. 2019, 13, 368. [Google Scholar] [CrossRef]
  212. Hu, P.; Kalb, R.G. BDNF heightens the sensitivity of motor neurons to excitotoxic insults through activation of TrkB. J. Neurochem. 2003, 84, 1421–1430. [Google Scholar] [CrossRef] [Green Version]
  213. Mojsilovic-Petrovic, J.; Jeong, G.B.; Crocker, A.; Arneja, A.; David, S.; Russell, D.; Kalb, R.G. Protecting motor neurons from toxic insult by antagonism of adenosine A2a and Trk receptors. J. Neurosci. 2006, 26, 9250–9263. [Google Scholar] [CrossRef] [PubMed]
  214. Park, D.; Kwak, S.G.; Park, J.S.; Choo, Y.J.; Chang, M.C. Can Therapeutic Exercise Slow Down Progressive Functional Decline in Patients with Amyotrophic Lateral Sclerosis? A Meta-Analysis. Front. Neurol. 2020, 11, 853. [Google Scholar] [CrossRef] [PubMed]
  215. Musarò, A.; Dobrowolny, G.; Cambieri, C.; Onesti, E.; Ceccanti, M.; Frasca, V.; Pisano, A.; Cerbelli, B.; Lepore, E.; Ruffolo, G.; et al. Neuromuscular magnetic stimulation counteracts muscle decline in ALS patients: Results of a randomized, double-blind, controlled study. Sci. Rep. 2019, 9. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  216. Huisman, M.H.B.; Seelen, M.; de Jong, S.W.; Dorresteijn, K.R.I.S.; van Doormaal, P.T.C.; van der Kooi, A.J.; de Visser, M.; Schelhaas, H.J.; van den Berg, L.H.; Veldink, J.H. Lifetime physical activity and the risk of amyotrophic lateral sclerosis. J. Neurol. Neurosurg. Psychiatry 2013, 84, 976–981. [Google Scholar] [CrossRef] [Green Version]
  217. Siciliano, G.; Pastorini, E.; Pasquali, L.; Manca, M.L.; Iudice, A.; Murri, L. Impaired oxidative metabolism in exercising muscle from ALS patients. J. Neurol. Sci. 2001, 191, 61–65. [Google Scholar] [CrossRef]
  218. Lanfranconi, F.; Ferri, A.; Corna, G.; Bonazzi, R.; Lunetta, C.; Silani, V.; Riva, N.; Rigamonti, A.; Maggiani, A.; Ferrarese, C.; et al. Inefficient skeletal muscle oxidative function flanks impaired motor neuron recruitment in Amyotrophic Lateral Sclerosis during exercise. Sci. Rep. 2017, 7. [Google Scholar] [CrossRef] [Green Version]
  219. Sassani, M.; Alix, J.J.; McDermott, C.J.; Baster, K.; Hoggard, N.; Wild, J.M.; Mortiboys, H.J.; Shaw, P.J.; Wilkinson, I.D.; Jenkins, T.M. Magnetic resonance spectroscopy reveals mitochondrial dysfunction in amyotrophic lateral sclerosis. Brain 2021, 143, 3603–3618. [Google Scholar] [CrossRef]
  220. Ferri, A.; Lanfranconi, F.; Corna, G.; Bonazzi, R.; Marchese, S.; Magnoni, A.; Tremolizzo, L. Tailored Exercise Training Counteracts Muscle Disuse and Attenuates Reductions in Physical Function in Individuals with Amyotrophic Lateral Sclerosis. Front. Physiol. 2019, 10. [Google Scholar] [CrossRef] [Green Version]
  221. Lunetta, C.; Lizio, A.; Sansone, V.A.; Cellotto, N.M.; Maestri, E.; Bettinelli, M.; Gatti, V.; Melazzini, M.G.; Meola, G.; Corbo, M. Strictly monitored exercise programs reduce motor deterioration in ALS: Preliminary results of a randomized controlled trial. J. Neurol. 2016, 263, 52–60. [Google Scholar] [CrossRef]
  222. Merico, A.; Cavinato, M.; Gregorio, C.; Lacatena, A.; Gioia, E.; Piccione, F.; Angelini, C. Effects of combined endurance and resistance training in Amyotrophic Lateral Sclerosis: A pilot, randomized, controlled study. Eur. J. Transl. Myol. 2018, 28, 132–140. [Google Scholar] [CrossRef]
  223. Tsitkanou, S.; Della Gatta, P.; Foletta, V.; Russell, A. The Role of Exercise as a Non-pharmacological Therapeutic Approach for Amyotrophic Lateral Sclerosis: Beneficial or detrimental? Front. Neurol. 2019, 10, 783. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  224. Bello-Haas, V.D.; Florence, J.M.; Kloos, A.D.; Scheirbecker, J.; Lopate, G.; Hayes, S.M.; Pioro, E.P.; Mitsumoto, H. A randomized controlled trial of resistance exercise in individuals with ALS. Neurology 2007, 68, 2003–2007. [Google Scholar] [CrossRef] [PubMed]
  225. Drory, V.E.; Goltsman, E.; Goldman Reznik, J.; Mosek, A.; Korczyn, A.D. The value of muscle exercise in patients with amyotrophic lateral sclerosis. J. Neurol. Sci. 2001, 191, 133–137. [Google Scholar] [CrossRef]
  226. Jensen, L.; Djurtoft, J.B.; Bech, R.D.; Nielsen, J.L.; Jørgensen, L.H.; Schrøder, H.D.; Frandsen, U.; Aagaard, P.; Hvid, L.G. Influence of Resistance Training on Neuromuscular Function and Physical Capacity in ALS Patients. J. Neurodegener. Dis. 2017, 2017, 1436519. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  227. Sanjak, M.; Bravver, E.; Bockenek, W.L.; Norton, H.J.; Brooks, B.R. Supported treadmill ambulation for amyotrophic lateral sclerosis: A pilot study. Arch. Phys. Med. Rehabil. 2010, 91, 1920–1929. [Google Scholar] [CrossRef]
  228. Braga, A.C.M.; Pinto, A.; Pinto, S.; De Carvalho, M. The role of moderate aerobic exercise as determined by cardiopulmonary exercise testing in ALS. Neurol. Res. Int. 2018, 2018, 8218697. [Google Scholar] [CrossRef] [Green Version]
Table 1. Genetic intervenctions.
Table 1. Genetic intervenctions.
GeneFunctionExpression Type in MuscleModelEffectSurvivalReferences
Glial cell-derived neurotrophic factor (GDNF)Trophic effect on motor neuronsExpression by muscle injection (AVR) at not symptomatic stage (age: 5–7days)SOD1G93A mice∧ Motor performance; ∨ motor neuron lossYES[38]
Engineered human mesenchymal stem cells (hMSC-GDNF)SOD1G93A rats∧ Motor performance; ∨ motor neuron loss; ∨ denervationYES[39]
Vascular endothelial growth factor (VEGF)Angiogenesis and neuroprotectionExpression by muscle injection (EIAV) at not symptomatic stage (age: 21 days)SOD1G93A mice∧ Motor performances; ∨ motor weakness; delayed onsetYES[42]
Expression by muscle injection (EIAV) at the onset (age: 90 days)SOD1G93A mice∧ Motor performances; ∨ motor weakness; ∨ motor neuron lossYES[42]
Insulin-like growth factor 1 (IGF-1)Anabolism of muscle and nerve tissues, myogenesis and neuronal survivalTransgenic mice, muscle restricted expressionSOD1G93A mice∧ Muscle regeneration; ∧ preservation NMJ; ∨ Muscle atrophy; ∨ MN loss; ∨ apoptotic and ubiquitin pathwaysYES[105,106]
MicroRNA-206 (miR-206)Myogenesis, NMJ formation, stabilization and repairTransgenic mice, muscle restricted deletionSOD1G93A mice∧ Muscle atrophy; ∧ NMJ loss; ∧ disease progression;
∨ disease duration
NO[65]
Uncoupling protein1 (UCP1)Thermogenesis by uncoupling mitochondrial electron transport from ATP synthesisTransgenic mice, muscle restricted overexpressionSOD1G86R mice∧ Disease progression; ∨ disease durationNO[96]
Muscle-specific kinase (MuSK)Formation and maintenance of NMJTransgenic mice, muscle restricted overexpressionSOD1G93A mice∧ Motor performances; ∨ NMJ denervation; delayed onsetNO[59]
Peroxisome proliferator-activated receptor-gamma coactivator- 1a (PGC-1a)Cellular energy metabolism, mitochondrial
biogenesis and angiogenesis
Transgenic mice, muscle restricted overexpressionSOD1G937R mice∧ Mitochondrial biogenesis; ∧mitochondria area; ∧ resistance to fatigue;
∧ Locomotor activity; ∧ Mitochondrial oxygen consumption in skeletal muscle; ∨ muscle atrophy
NO[95]
SOD1G93A mice∧ Muscle fiber oxidation; ∨ motor functionNO[86]
MyoDMuscle development and differentiationExpression by muscle injection (AV) in adult mice (age: 30 days)SOD1G93A mice∧ Weight loss; ∧ Motor neuron loss; ∨ motor performances;
∨ NMJ innervation; ∨ muscle fiber oxidation
NO[86]
MyogeninMuscle development and differentiation increases oxidative metabolism of muscleExpression by muscle injection (AV) in adult mice (age: 30 days)SOD1G93A mice∧ Motor performances; ∧ NMJ innerve tion; ∧ Muscle fiber oxidation; ∨ motor neuron lossNO[86]
DOK-7Neuromuscular synapsis formation by regulation of Musk activityExpression by intravenous injection (AAV) at the onset (age: 90 days)SOD1G93A mice∧ NMJ innervation (diaphragm); ∧ Motor activity; ∨ muscle atrophyYES[64]
Histone deacetylase 4 (HDAC4)Skeletal muscle response to denervationTransgenic mice, muscle restricted deletionSOD1G93A mice∧ Muscle atrophy; ∧ weight loss; ∨ muscle force; ∨ NMJ innervation; precocious disease onset.NO[72]
Neuregulin 1 (NRG1)Axonal and neuromuscular development and maintenanceOverexpression by intramuscular injection (AAV) at the onset (age: 8 weeks)SOD1G93A mice∧ Axons collateral sprouting and NMJ; ∧ Compound muscle action potentialN/A[51]
Overexpression by intravenous injection (AAV under hDesmin promoter) in adult mice (age: 6 weeks)SOD1G93A mice∧ Neuromuscular functions; ∧ NMJ innervation; ∧ cell survival pathway activation; ∧ locomotor ability; ∨ Motoneuron loss; ∨ neuroinflammation; ∨ oxidative stress in skeletal muscle; delayed onsetN/A[52]
Table 2. Preclinical pharmacological interventions.
Table 2. Preclinical pharmacological interventions.
DrugsFunctionModelEffectsSurvivalReferences
Metabolic modulationL-CarnitineCofactor for the beta-oxidation of long-chain fatty acidsSOD1G93A miceDelayed deterioration of motor activityYES[112]
DichloroacetateImproves glycolysisSOD1G86R mice∧ Maintenance of NMJs; ∧ Muscle strength; ∨ denervation markersYES[89,131]
RanolazineInhibition of beta-oxidationSOD1G93A mice∧ Motor functions; ∧ Muscle ATP;
∧ energy metabolism
NO[90]
Modulation of muscle mass growthAnti-MyostatinEndogenous inhibitor of myogenesisSOD1G93A mice SOD1G93A rats∧ Muscle mass strength; ∨ weight lossNO[142]
ActRIIB.mFcEndogenous signaling receptor for
myostatin
SOD1G93A mice∧ Body weight; ∧ grip strength; ∧ Muscle sizeNO[148]
DihydrotestosteroneActivator of anabolic functionsSOD1G93A mice∧ Weight loss; ∧ grip strengthYES[133]
NandroloneActivator of anabolic functionsSOD1G93A mice∧ Diaphragm muscle massNO[136]
NMJ preservation and atrophy reductionAnti-MuskDevelopment and stability of NMJsSOD1G93A mice∧ Muscle mass; ∧ strength; ∨ muscle; ∨ denervationYES[154]
∧ innervation of the neuromuscular junction; ∨ diaphragm function, motor neuronsNO[155]
Other interventionsTirasemvit (CK-357)Fatigue resistance of the muscleSOD1G93A mice∧ Submaximal isometric force; ∧ forelimb grip strength; ∧ grid hang time; ∧ rotarod performance; ∧ diaphragm forceNO[159]
FG-3019Development and stability of NMJsSOD1G93A mice∧ Locomotor; ∧ performance; ∨ muscular
fibrosis; ∨ atrophy
NO[160]
2′(3′)-O-(4-Benzoylbezoyl) Adenosine5′-triphosphate (BzATP)P2X7 agonistSOD1G93A mice∧ Muscle metabolism; ∧ NMJs morphologyNO[174]
Table 3. Clinical pharmacological interventions.
Table 3. Clinical pharmacological interventions.
DrugsFunctionPhaseClinical TrialReferences
Acetyl L-carnitineCofactor for the beta-oxidation of long-chain fatty acidsIIEudraCT Number: 2004-004158-23[113]
MetabolicmodulationCreatineFacilitates recycling of adenosine triphosphate (ATP), the energy currency of the cell, primarily in muscle and brain tissue.II II II II IIINCT00005766 NCT00005674 NCT00355576 NCT00070993 NCT00069186[126,127,128]
IINCT01257581
NMJ
preservation and atrophy reduction
OzanezumabHumanized monoclonal antibody against Nogo-AIINCT01753076[153]
Otherinterventions IINCT01486849
IINCT01089010
Tirasemvit
(CK-357)
Fast skeletal muscle troponin activators (FSTA)III
II
NCT02936635
NCT01709149
[163]
IINCT01378676
IIINCT02496767
Reldesemtiv (CK-2127107)Protein complex that modulates muscle contractility and increases the strength and power of the muscular systemIINCT03160898[161]
IINCT02487407
LevosimendanIncreases the functionality of the musculoskeletal systemIIINCT03505021[162]
IIINCT03948178
PalmitoylethanolamineAnalgesic and anti-inflammatoryN/ANCT02645461[158]
(PEA)
AminophyllineAdenosine receptor antagonistN/AN/A[173]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Scaricamazza, S.; Salvatori, I.; Ferri, A.; Valle, C. Skeletal Muscle in ALS: An Unappreciated Therapeutic Opportunity? Cells 2021, 10, 525. https://doi.org/10.3390/cells10030525

AMA Style

Scaricamazza S, Salvatori I, Ferri A, Valle C. Skeletal Muscle in ALS: An Unappreciated Therapeutic Opportunity? Cells. 2021; 10(3):525. https://doi.org/10.3390/cells10030525

Chicago/Turabian Style

Scaricamazza, Silvia, Illari Salvatori, Alberto Ferri, and Cristiana Valle. 2021. "Skeletal Muscle in ALS: An Unappreciated Therapeutic Opportunity?" Cells 10, no. 3: 525. https://doi.org/10.3390/cells10030525

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop