Conventional approaches to the therapy of hereditary myopathies

Cover Page

Cite item

Abstract

The aim of the work was to analyze the available therapeutic options for the conventional therapy of hereditary myopathies.

Materials and methods. When searching for the material for writing a review article, such abstract databases as PubMed and Google Scholar were used. The search was carried out on the publications during the period from 1980 to September 2022. The following words and their combinations were selected as parameters for the literature selection: “myopathy”, “Duchenne”, “myodystrophy”, “metabolic”, “mitochondrial”, “congenital”, “symptoms”, “replacement”, “recombinant”, “corticosteroids”, “vitamins”, “tirasemtiv”, “therapy”, “treatment”, “evidence”, “clinical trials”, “patients”, “dichloracetate”.

Results. Congenital myopathies are a heterogeneous group of pathologies that are caused by atrophy and degeneration of muscle fibers due to mutations in genes. Based on a number of clinical and pathogenetic features, hereditary myopathies are divided into: 1) congenital myopathies; 2) muscular dystrophy; 3) mitochondrial and 4) metabolic myopathies. At the same time, treatment approaches vary significantly depending on the type of myopathy and can be based on 1) substitution of the mutant protein; 2) an increase in its expression; 3) stimulation of the internal compensatory pathways expression; 4) restoration of the compounds balance associated with the mutant protein function (for enzymes); 5) impact on the mitochondrial function (with metabolic and mitochondrial myopathies); 6) reduction of inflammation and fibrosis (with muscular dystrophies); as well as 7) an increase in muscle mass and strength. The current review presents current data on each of the listed approaches, as well as specific pharmacological agents with a description of their action mechanisms.

Conclusion. Currently, the following pharmacological groups are used or undergoing clinical trials for the treatment of various myopathies types: inotropic, anti-inflammatory and antifibrotic drugs, antimyostatin therapy and the drugs that promote translation through stop codons (applicable for nonsense mutations). In addition, metabolic drugs, metabolic enzyme cofactors, mitochondrial biogenesis stimulators, and antioxidants can be used to treat myopathies. Finally, the recombinant drugs alglucosidase and avalglucosidase have been clinically approved for the replacement therapy of metabolic myopathies (Pompe’s disease).

Full Text

Abbreviations: ETC – electronic transport chain; mRNA – matrix ribonucleic acid; tRNA – transport ribonucleic acid; siRNAs – small interfering ribonucleic acids; NAD, nicotineamide-adenine dinucleotide; FAD – flavin adenine dinucleotide; NADP – nicotinamide adenine dinucleotide phosphate; ATP – adenosine triphosphate; ADP – adenosine diphosphate; CTGF – connective tissue growth factor; TGFβ – transforming growth factor-beta; NSAIDs – non-steroidal anti-inflammatory drugs; XLMTM – X-linked myotubular myopathy; TCA – tricarboxylic acid cycle; TNF-α – tumor necrosis factor-alpha; CTGF/CCN2 – connective tissue growth factor.

INTRODUCTION

Hereditary myopathies are a clinically, histologically and genetically heterogeneous group of muscle pathologies that are caused by atrophy and degeneration of striated muscles due to mutations in genes whose role is closely related to the functioning of myocytes. Most often, the proteins encoded by these genes are involved in the formation or maintenance of the structural integrity of the cytoskeleton and plasma membrane. At the same time, myopathies associated with the pathology of cytoskeletal proteins are characterized by a progressive course (muscular dystrophies), and myopathies caused by а function loss of membrane proteins are fully manifested from birth (congenital myopathies). In addition, hereditary myopathies can be caused by mutations in the genes associated with the work of mitochondria (mitochondrial myopathies), or the genes encoding enzymes of intracellular metabolism (metabolic myopathies) [1].

Initially, classifications of hereditary myopathies were based on the clinical presentation or typical histological features found in muscle biopsy specimens. However, according to the current recommendations, the diagnosis of myopathy should be accompanied by the data from molecular genetic studies. In addition to precision diagnostics, this approach leads to an expansion list of the nosological group genetic correlates [2].

THE AIM of the work was to analyze the available therapeutic options for the conventional therapy of hereditary myopathies.

MATERIALS AND METHODS

When searching for the material for writing a review article, such abstract databases as PubMed and Google Scholar were used. The search was carried out on the publications during the period from 1980 to September 2022. The following words and their combinations were selected as parameters for the literature selection: “myopathy”, “Duchenne”, “myodystrophy”, “metabolic”, “mitochondrial”, “congenital”, “symptoms”, “replacement”, “recombinant”, “corticosteroids”, “vitamins”, “tirasemtiv”, “therapy”, “treatment”, “evidence”, “clinical trials”, “patients”, “dichloracetate”.

RESULTS AND DISCUSSION

1. General characteristics of hereditary myopathies

The most typical symptoms of myopathies are muscle weakness, myalgia, myopenia, and exercise intolerance. The clinical picture of myopathies can vary from asymptomatic forms with an increase in serum creatine kinase values and an increased tendency to hyperthermia to severe forms leading to skeletal deformities, as well as respiratory and heart failures. The groups of affected muscles can differ significantly – from an isolated lesion of the oculomotor muscles [3] to a systemic muscle atrophy involving myocardium and diaphragm. The variability of clinical signs is associated both with the diversity of causative genes and with the degree of their function loss. For example, a severe muscle phenotype in Duchenne’s muscle dystrophy is associated with neuropsychiatric disorders [4], and muscle symptoms in the phosphofructokinase deficiency (Tarui disease) are accompanied by hemolytic anemia and hyperuricemia [5]. Mitochondrial myopathies are characterized by especially high clinical heterogeneity [6]. Since multisystem disorders, nervous, digestive, urinary, cardiovascular, endocrine and reproductive systems, as well as the organs of vision and hearing often accompany a mitochondrial dysfunction, they can be involved in the pathological process (Table 1).

 

Table 1 – General clinical characteristics of hereditary myopathies

Group of congenital myopathies

Examples of diseases

Clinical manifestations

Proteins with impaired function

Pathogenesis

Muscular dystrophies

Miyoshi myopathy

Distal skeletal muscle weakness

Elevated levels of creatine kinase in the blood

First symptoms occur during adolescence [28, 29]

Dystrophin [30]

Dysferlin is a membrane-associated linker protein; its function is to mediate calcium-dependent regeneration of mechanical damage to sarcolemma.

With mutations that disrupt the dysferlin function; the accumulation of damage to sarcolemma occurs, which leads to progressive dystrophy of skeletal muscles [31].

Limb-girdle muscular dystrophy type 2B (LGMD2B)

Proximal skeletal muscle weakness

Elevated levels of creatine blood kinase

Manifestation at the age from 10 to 30 years [32, 33]

Duchenne’s myodystrophy

Muscular hypotension

Heart failure

Respiratory distress

Debut in early postnatal or postnatal age

Death before age of 20 [34, 35]

Dystrophin [36]

Dystrophin is involved in the mechanical stabilization of sarcolemma. In case of loss of the protein product dysferlin due to large deletions or a shift in the reading frame, sarcolemma becomes vulnerable to mechanical deformations that occur during muscle contraction or stretching [37].

Since dystrophin plays an important role in the processes of mitotic division, Duchenne’s disease disrupts cell polarity and myogenic differentiation of stem cells. Stem cells lacking functional dystrophin undergo aberrant asymmetric division with centrosome amplification, spindle orientation errors, and an extended cell cycle [38, 39].

Congenital myopathies

Myosin storage myopathy

Muscular hypotension

Hypertrophic or dilated cardiomyopathy

Manifestation in the neonatal or postnatal period

MYH7 (heavy chain of slow/β-cardiac myosin)

MYH7 is the main myosin isoform in slow oxidative type 1 muscle fibers of skeletal muscles and myocardium. Numerous missense mutations in the MYH7 globular head lead to disruption of the structural protein function and the formation of large inclusions consisting of myosin chains.

Bethlem muscular dystrophy

Weakness of proximal muscles

Joints contracture

Hypotension progresses slowly, and more than two-thirds of patients older than 50, continue to move independently.

Possible damage to the respiratory muscles [40].

Type VI collagen

Collagen VI is an extracellular matrix protein that forms a microfibrillar network. The protein consists of three different α-chains encoded by separate genes named COL6A1, COL6A2 and COL6A3 in humans.

Potential effects on muscles include progressive dystrophic changes, fibrosis, and signs of increased apoptosis [41].

Metabolic myopathies

Pompe’s disease

Muscular hypotension

Hepatomegaly

Heart failure

Neurological disorders

Debut at any age (an early debut correlates with a more severe course) [42]

Acid maltase [43]

After entering the lysosomes, acid maltase mediates the catalytic breakdown of glycogen by interacting with the mannose-6-phosphate receptor [44]. More than 500 mutations including insertions, deletions, splicing site mutations, nonsense and missense mutations, have been found. They disrupt the functional activity of acid maltase, leading to glycogenosis and energy deficiency of the muscle tissue [45, 46].

Tarui disease

Muscle weakness

Muscle cramps

Encephalopathy

Hemolytic anemia

Rhabdomyolysis risk

Debut at any age [47]

Phosphofructokinase [48]

Phosphofructokinase catalyzes the transfer of a phosphate group from ATP to fructose-6-phosphate, which is one of the key elements of glycolysis. In humans, three isozymes named M (muscle), L (liver), and P (platelets), have been identified,

Mutations in phosphofructokinase-M lead to muscle weakness due to an energy deficiency in working muscles [49].

Mitochondrial myopathies

Myoclonus epilepsy with myopathy and sensory ataxia (MEMSA)

Proximal and/or distal myopathy

Muscular hypotension

Myoclonic epilepsy

Encephalopathy

Sensory ataxia

Debut at any age [50]

Polymerase gamma (POLG) [51]

Gamma polymerase is a key enzyme of a mitochondrial DNA replication Mutations in the POLG gene lead to the energy deficiency due to the accumulation of defective mitochondria and a decrease in the number of mtDNA copies (mtDNA depletion), especially in muscle, brain, or liver cells [52].

 

1.1. Muscular dystrophies

More than 30 muscular dystrophies have been identified, the most common of which are as follows: Duchenne’s muscle dystrophy, facioscapulohumeral muscular dystrophy, Becker muscular dystrophy, limb-girdle and myotonic kinds of muscular dystrophy. Etiologically, these diseases are very heterogeneous. For example, Duchenne’s muscle dystrophy and Becker dystrophy are caused by dystrophin mutations, while lumbar-limb muscular dystrophies can be caused by an impaired function of calpain, dysferlin, sarcoglycan, lamin, anoctamine, etc. [7]. In all cases, early signs of degeneration and then regeneration of some muscle fibers are usually found. The fibers that regenerate become larger than usual, and eventually the muscle is almost completely replaced by a fibrous scar tissue and fat.

The most classic type of such muscle disorders is Duchenne’s dystrophy. It is caused by frameshift mutations in the MDD gene encoding the dystrophin protein, which is a plasma membrane-associated protein that plays a critical role in sarcolemma stabilizing in mechanical shifts during the muscle contraction or stretching [8, 9]. The dystrophin absence leads to a decrease in the resistance of sarcolemma and the subsequent necrosis of the muscle fibers [10]. The muscle fibers destruction is exacerbated by a mechanical stress and improves while the muscle immobilization [11, 12]. Thus, the accumulation of the damaged muscle fibers is the cause of the progressive course of Duchenne’s myodystrophy. At the same time, the exact molecular mechanisms by which dystrophin plays the role of a mechanical stabilizer, are still unclear [13].

1.2. Congenital myopathies

Unlike muscular dystrophies, congenital myopathies are already manifested in the neonatal period [14]. This is due to the fact that the function of defective proteins is not associated with maintaining the integrity of already differentiated myocytes, but with the structural organization of the muscle tissue even at the stage of histogenesis. Basically, these are the proteins involved in the formation of the cytoskeleton or intercellular substance. At the same time, these can be such multifunctional proteins as myotubularin, which is involved in the transfer of endosomes, coupling of excitation and contraction, the organization of intermediate filaments, and apoptosis.

Although the exact epidemiology of congenital myopathies is unknown, researchers estimate their incidence to be around 1:25 000 [15]. The classification of congenital myopathy is constantly being revised as more genes that are associated with its various phenotypic and histological manifestations, are identified. At the moment, it continues to be based mainly on the features observed in muscle biopsy [16]. Accordingly, congenital myopathy can be divided into the following five forms: rod myopathy; cardiac myopathy; centronuclear myopathy; congenital myopathy of a fiber type imbalance; myosin storage myopathy.

Clinically, congenital myopathies are manifested by muscle hypotonia and weakness, present at birth or appearing in infancy and not progressive during life. Depending on the causative gene and the nature of the mutation, the clinical spectrum varies from severe neonatal forms with congenital arthrogryposis to mild forms with isolated hyposthenia [14, 16]. In the neonatal period, symptoms tend to be more pronounced and may include reduced fetal movement and a subsequent development of arthrogryposis and clubfoot. Severe muscular hypotonia is often present at birth and in the first months of life (a sign of a lethargic baby) along with a frog-like posture, difficulty sucking, and a respiratory failure [17].

1.3. Metabolic myopathies

Metabolic myopathies are associated with mutations in the genes encoding energy metabolism enzymes. Biochemical disorders include disorders of fatty acids, glucose, or glycogen oxidation. As a result, the functional reserves of the muscle tissue are reduced, which is manifested by hypotension, increased fatigue, myalgia, convulsions, episodes of rhabdomyolysis, etc. [18]. At the same time, fatty acids utilization defects are characterized by a low tolerance to long-term endurance exercises, while disorders of glucose and glycogen metabolism are manifested by an intolerance to fast high-intensity exercises [19]. A separate feature of myopathies also associated with mutations of glycogenolysis enzymes, is the accumulation of intracellular glycogen inclusions [20].

1.4. Mitochondrial myopathies

The pathogenetic basis of mitochondrial myopathies is a violation of energy metabolism processes due to defects in the oxidative phosphorylation. In this regard, some authors consider mitochondrial myopathies as a subtype of metabolic ones. Nevertheless, a number of features of inheritance and pathogenesis, as well as some clinical characteristics, make it possible to distinguish them into a separate group. Thus, mitochondrial myopathies are always associated with impaired functioning of the electronic transport chain (ETC), most often with defects in complex 1 [21–23]. In addition, mitochondrial myopathies can be caused by mutations in both nuclear and mitochondrial genes. In case of mitochondrial DNA mutations, inheritance occurs almost exclusively maternally [24]. The severity of symptoms is determined not only by the pathogenicity of the mutation, but also by the number of the mutant mitochondrial DNA copies that the body has inherited from the mother [25]. The fact is that the mitochondrial genome is heterogeneous (the phenomenon of heteroplasmy) and, along with mutant ones, healthy mitochondria are always present in the cell. Thus, the proportion of defective mitochondria is determined randomly with a random distribution of mitochondria between the daughter cells, which is called the “bottleneck” phenomenon [26].

In general terms, mitochondrial myopathies are mitochondrial diseases, in the spectrum of clinical manifestations of which there are pronounced symptoms from the muscle tissue. Mitochondrial myopathies are characterized by a progressive course and a wide range of associated symptoms, including epilepsy, neuropathy, sensory impairments, etc. [27].

2. Treatment of hereditary myopathy

Treatment of hereditary myopathies varies widely depending on the type and specific disease. A significant proportion of therapeutic interventions in the treatment of myopathies patients are the approaches based on diet, exercise therapy and massage. For example, in metabolic myopathies associated with impaired glucose utilization, the most important therapeutic approach is a low-carbohydrate ketogenic diet [53]. When correcting congenital myopathies, patients are recommended a controlled physical activity, as well as the use of special corsets to prevent the development of bone deformities.

Pharmacological approaches occupy an important place in symptomatic, supportive, and pathogenetic-oriented kinds of therapy. In addition, high rates development of antisense and gene therapy have recently made it possible to focus on etiotropic approaches in the treatment of myopathies patients.

As with most monogenic diseases associated with the gene function loss, conventional specific therapy can be aimed at: 1) replacement of the mutant protein; 2) increase in its expression; 3) stimulation of the expression of internal compensatory pathways; 4) restoration of the balance of compounds associated with the function of the mutant protein (for enzymes).

2.1. Mutant protein substitution

Currently, a number of recombinant enzymes have been approved for the specific myopathies therapy. One of the approved approaches is the enzyme replacement therapy for type II glycogenosis (Pompe’s disease) with recombinant human alglucosidase alfa (rhGAA; Myozyme© (ex-US) and Lumizyme© (USA), which has been available since 2006, or with avalglucosidase alfa (NEXVIAZYME™; avalglucosidase alfa-ngpt) available since 2021 [54, 55].

Obviously, replacement therapy with recombinant forms of proteins is not the main strategy, since most exogenous proteins cannot penetrate intracellularly to carry out their functions. However, the approaches to directly modify proteins and peptides to enhance cytosolic translocation, continue to be a promising method for improving the delivery efficiency and extending the viability of intracellular protein therapeutics. Among the proposed approaches to improve the cytosolic delivery of exogenous proteins, there have been such as chemical recharging or the inclusion of intracellular internalization motifs [56]. For example, the enzyme replacement therapy with a modified recombinant protein has been proposed for the treatment of X-linked myotubular myopathy. In the preclinical study on Mtm1 δ4 mice with a myotubularin gene knockout, the replacement therapy with recombinant 3E10Fv-MTM1 protein (0.1 mg/kg) into the tibialis anterior muscle twice a week significantly improved the muscle function [57].

2.2. Increase in expression

Some nucleotide substitutions called nonsense mutations, lead to the formation of a stop codon in the coding gene region, resulting in the premature termination of the desired protein synthesis. In addition, mRNA resulting from nonsense mutations is destabilized by a nonsense-mediated decay [58]. Similar mutations are often the cause of hereditary myopathies. Such mutations are found in approximately 10% of Duchenne’s patients [59] and in 20% of individuals with X-linked myotubular myopathy (XLMTM) [60].

To restore the expression of the full amino acid sequence, the drugs that force the reading of termination codons were proposed [61]. For example, these were aminoglycosides containing a 2-deoxystreptamine ring bind to the small ribosomal RNA subunit, reducing the accuracy of translation [62]. This property made it possible to propose the use of aminoglycosides for the treatment of Duchenne’s myodystrophy [63] and a number of other monogenic diseases caused by premature stop codon mutations [64, 65].

However, serious side effects of aminoglycosides, such as nephrotoxicity and ototoxicity, limit their long-term use. In this regard, alternative agents have been proposed, including suppressor tRNAs and small interfering RNAs (siRNAS) [66] and ataluren [67]. However, only ataluren is currently approved for a clinical use [68].

Theoretically, the termination forcing approach could help treat all hereditary myopathies associated with premature stop codons. Restoration of translation does not always lead to the formation of a functional protein, which, apparently, is associated with impaired intracellular traffic and post-translational modifications of the product [69]. To date, the termination-forcing strategy has been approved for only use with nonsense mutations that cause Duchenne’s muscular dystrophy. In addition, despite a high proportion of nonsense mutations in myopathies, their heterogeneity and a low prevalence of each specific disease in the general population make it difficult to conduct full-fledged clinical studies [57].

2.3. Stimulation of internal compensatory pathways expression

In some cases, a decrease or absence of protein expression can be partially compensated for hyperactivation of internal pathways that can functionally mitigate the defect. For example, the severity of muscle pathology in dystrophin defects can be reduced due to the myogenic stimulation, which leads to an increase in the expression of myocyte structural proteins. Non-clinical studies demonstrate that inhibitors of histone deacetylases have a pronounced therapeutic effect in some myopathies. Apparently, due to the regulatory activity in relation to epigenetic modifications, such compounds increase the activity of myogenic differentiation of myocyte precursors. In vitro studies have found out that inhibitors of histone deacetylases enhance myogenesis and the formation of enlarged skeletal myotubes [70, 71]. When administered to the dystrophy mice, the drugs had similar beneficial effects. In the mdx mice, the inhibitors increased the cross-sectional area of myofibrils, reducing the histological signs of inflammation and remodeling [72]. Interestingly, among the compounds with an inhibitory activity against histone deacetylases, such well-known drugs as trichostatin A and valproic acid can be distinguished. Herewith, computational biology methods have shown that trichostatin A has the ability to weaken the posttranscriptional repression of utrophin, which has a significant similarity in sequence and functional motifs with dystrophin, including the ability to bind the same dystrophin-associated glycoprotein complex [73, 74]. Utrophin is expressed in fetal tissues at high levels and is inhibited during its development in adults. It was found out that in mice, a decrease in the level of utrophin programmed in embryogenesis corresponds to the onset of muscle necrosis [75]. At the same time, the gene therapy approaches aimed at the delivery of utrophin, significantly improve the condition of Duchenne’s myodystrophy mice [76]. Trichostatin A is currently undergoing clinical trials for the treatment of Duchenne’s muscular dystrophy. At the same time, the specific utrophin modulator ezutromid/SMT C1100 demonstrated unsatisfactory results in phase II clinical trials and was withdrawn [77]. At present, the search for an optimal candidate for increasing utrophin expression continues [78].

2.4. Restoring Balance of Compounds Associated with Mutant Protein Function

In some cases, in addition to repairing the deficiency of the protein itself, a strategy for the delivery of compounds related to its catalytic function can be used (Fig. 1). Obviously, such an approach can be implemented in only metabolic and mitochondrial myopathies, where the cause of the disease is the metabolic enzyme deprivation function, and not the one of structural protein or kinase. The main principle of this approach is based on the fact that the use of an exogenous metabolite compensates for its endogenous deficiency, restoring the efficiency of the entire biochemical chain. For example, it has been known since the 1960s that intravenous glucose improves the exercise tolerance in patients with McArdle’s disease, which is associated with a defect in the conversion of glycogen to glucose [79]. Glucose therapy is also effective in some other diseases associated with mutations in the proximal enzymes of glycogen catabolism [80–82]. Another example is the use of triheptanoin, a synthetic medium-length triglyceride that restores the energy efficiency of long-chain fatty acid oxidation in the presence of mutations in proximal catabolism enzymes [83]. Triheptanoin has demonstrated a significant improvement in cardiac and muscle symptoms in VLCAD syndrome patients and in patients with carnitine palmitoyltransferase 2 deficiency [84, 85]. In some cases, an effective strategy to increase the concentration of compounds that serve as substrates for the bypass or an alternative biochemical cascade is also used. Thus, for example, in case of a defect in the formation of ATP along the pathway of fatty acid oxidation, an increase in glucose concentration can compensate for the total energy deficiency due to glycolysis [86]. A similar effect can be achieved with the use of creatine. Creatine is a skeletal muscle amino acid that serves as a substrate for the formation of creatine phosphate, a phosphate group donor for the conversion of ADP to ATP by the enzyme creatine kinase. In a number of studies, the administration of exogenous creatine has shown a therapeutic effect on the muscle symptoms in metabolic myopathies [62, 63].

 

Figure 1 – Classical pharmacological methods to compensate for inadequate functioning of mitochondria

 

2.5. Mitotropic drugs

Various cofactors, including riboflavin, coenzyme Q10, vitamins B6 and B3, can be used to partially compensate for the disorders caused by a dysfunction of one of the metabolic pathways (Fig. 1). These drugs can partially increase the energy efficiency of cells due to a positive effect on oxidative phosphorylation in mitochondria [88]. It is known that vitamin B3 (nicotinic acid) serves as a substrate for the formation of NAD and NADP, thereby facilitating the transfer of hydrogen from the tricarboxylic acid cycle to complex 1. Coenzyme Q10 (ubiquinone), in turn, is directly involved in the transfer of electrons from the NADH dehydrogenase complex (complex I) and succinate dehydrogenase complex (II) to complex III.

The use of cofactors is one of the main therapeutic options for mitochondrial myopathies. However, due to the lack of full-fledged clinical studies, it is impossible to judge the effectiveness of this approach in terms of evidence-based medicine. Moreover, the vast majority of these compounds are registered as food additives [89]. Obviously, the approaches based on the use of cofactors do not have a dramatic clinical effect due to e thweak mitochondrial transport, nonselectivity of the action, and a weak overlap with the pathogenetic mechanisms of the disease [90, 91]. The use of vitamin and cofactor cocktails is more justified when the number of factors considered is reduced due to their deficiency or transport defect now when this approach can be considered as replacement therapy [42, 68, 69].

In general, there are still not so many effective methods for restoring a mitochondrial function in mitochondrial mutations from the point of view of evidence-based medicine. In addition to cofactors, mitotropic compounds are represented by antioxidants, mitoprotectors, incl. dichloroacetate, arginine, coenzyme Q10, idebenone, etc. [70, 71]. Pharmacological approaches aimed at improving the function of mitochondria are based on the use of a very wide range of drugs [89, 90, 94]. Some of the most requested connections are shown in Fig. 1.

Classical pharmacological methods of compensating for inadequate functioning of mitochondria are based on increasing the activity of mitochondrial metabolic cascades and reducing the content of toxic agents such as lactate and reactive oxygen species (ROS). E.g., bezafibrate has been shown to stimulate mitochondrial biogenesis by activating the PGC-1a/PPAR pathway. In addition, acipimox, nicotinamide and riboside restore the content of NAD+, increasing the efficiency of electron transfer to the ETC.

Thiamine, lipoic acid and dichloroacetate activate pyruvate dehydrogenase, which leads to a decrease in the lactate accumulation due to the conversion of pyruvate to another metabolite, acetyl-CoA. Succinate, riboflavin, and CoQ10 promote the ETC electron transfer or restore the function of complexes I and II. Some compounds, such as idebenone, N-acetylcysteine, and lipoic acid, have the ability to reduce or inactivate the ROS production. Elamepritide stabilizes mitochondrial membrane lipids, preventing a mitochondrial destruction.

In case of a deficiency of certain lipid or carbohydrate metabolism enzymes, the deficiency replenishing strategy of compounds in the biochemical chain after the reaction catalyzed by the mutant enzyme, has a therapeutic efficacy. E.g., with a defect in the utilization of long-chain fatty acids, the use of heptanoin, a more proximal component included in the tricarboxylic acid (TCA) cycle, is justified. Similarly, in case of glycogen cleavage defects, the therapeutic potential is the use of exogenous glucose. Finally, the defects in the oxidative phosphorylation and mitochondrial function can be partially compensated for by the use of creatine, which acts in muscles as an alternative carrier of the high-energy phosphate bond in the formation of creatine phosphate.

2.6. Anti-inflammatory therapy

Anti-inflammatory therapy is one of the key approaches to the treatment of muscular dystrophies [96]. Inflammatory changes may accompany other types of myopathies, but this is extremely rare [97].

Currently, the only approved approach aimed at suppressing the inflammatory process in myodystrophy is corticosteroid therapy. However, it is important to emphasize that, despite the progressive death of muscle fibers, anti-inflammatory therapy is not necessary for all muscular dystrophies. E.g., treatment with deflazacort in dysferlinopathies patients neither improved nor showed any trend towards a decrease in muscle strength [98].

Corticosteroids have been shown in clinical trials to improve muscle strength and function without clinically serious side effects [99, 100]. Moreover, glucocorticoids have been shown to increase the utrophin expression [101].

In view of the serious side effects developed during a long-term use of corticosteroids, the search for other anti-inflammatory therapy strategies continues. For example, among the strategies tested in myodystrophy, inhibitors of cyclooxygenase (COX), tumor necrosis factor-alpha (TNF-α) and its receptor, as well as TRPV2 channels, can be distinguished.

Non-steroidal anti-inflammatory drugs (NSAIDs) have shown a relatively modest efficacy in a mouse model of Duchenne’s dystrophy. Despite the fact that the use of aspirin and ibuprofen improved the morphological picture of muscles and reduced the inflammatory infiltration and necrosis, the percentage of regenerating myofibrils and isometric tension did not change significantly [102].

The spectrum of the pharmacological activity of the antiallergic drug tranilast includes blockade of TRPV2 [103], therefor its use led to a decrease in fibrosis in skeletal muscles and an increase in the exercise tolerance [104, 105].

Inhibitors of TNF-α have shown some potential in the treatment of myodystrophy. The use of etanercept or an anti-TNF-α antibody slowed down the course of the disease and also reduced the inflammation and destruction of dystrophic muscles in mdx mice without the development of pronounced side effects [106, 107].

2.7. Antifibrotic therapy

An extracellular matrix is an important component of skeletal muscles. It provides a scaffold structure that holds the myofibrils and vessels. In addition, it plays a major role in the processes of biomechanical contraction, as well as in maintaining the integrity and repair of muscle fibers. An excessive accumulation of extracellular matrix components, especially collagen, is defined as fibrosis. An excess formation of the connective tissue as a result of death and defect in muscle cells proliferation is the most important distinguishing feature of muscular dystrophies. Since the dynamics of fibrotic replacement in myodystrophy strongly correlates with the development of muscle symptoms, antifibrotic therapy is one of the main approaches to treating such patients [108].

Tamoxifen is a prodrug, and some of its metabolites interact with the nuclear estrogen receptor, mediating antifibrotic and myoprotective effects. A multicenter, prospective study in 13 outpatient boys aged 6–14 years with genetically confirmed Duchenne’s muscular dystrophy demonstrated that patients treated with tamoxifen 20 mg/daily maintained a motor and respiratory functions, compared with a significant deterioration in the patients of the same history of age who had been administrated with only corticosteroids [109].

A similar approach has also shown off its efficacy in a mouse model of dystroglycanopathy. In the studies by Wu B. et al. it has been demonstrated that tamoxifen and raloxifene significantly alleviate a disease progression in the animals with the c.1343C>T mutation of the FKRP gene, demonstrating a pronounced phenotype of a limb-girdle muscular dystrophy [110].

A primary profibrotic signal in skeletal muscles, as in other tissues, is a transforming growth factor-beta (TGFβ) [111]. A high expression of TGFβ is a characteristic feature of dystrophic muscles [112] and is considered one of the main therapeutic targets for reducing fibrosis. It has been shown that Wnt-TGFβ2 is one of the key factors mediating the differentiation of dystrophin-deficient muscle cell precursors in the fibrogenic direction. Antibodies stabilizing LTBP4, which is a TGFβ binding factor, demonstrated a high efficiency. Anti-LTBP4 treatment also reduced muscle fibrosis and increased muscle strength, including the ones in the diaphragm muscles [113].

The renin-angiotensin system plays an important role in the transmission of profibrotic signals. In particular, the activation of the angiotensin 1 receptor stimulates fibrosis. At the same time, it has been shown that the antihypertensive drug with an inhibitory activity against TGFβ2 losartan led to an increase in the level of myogenic factors with a reduced expression of fibrogenic genes in mdx mice (Duchenne’s myodystrophy model) [112].

Interestingly, another drug that blocks the renin-angiotensin-aldosterone axis, enalapril, also exhibits inhibitory effects on the connective tissue growth factor (CTGF/CCN2) [114] and is another regulator of profibrotic signaling [115, 116]. Pharmacological blockade of CTGF has been shown to slow the progression of fibrosis and improve a muscle function in mdx mice [114]. Moreover, anti-muscle CTGF therapy is currently undergoing clinical trials for the treatment of Duchenne’s muscular dystrophy [117].

2.8. Means with a positive effect on muscle strength

A decreased muscle strength is the main symptom of myopathies. In this regard, in addition to other approaches, strategies have been developed to increase the effectiveness of the muscle contraction or the prevention of myopenia.

For example, tirasemtiv, fast skeletal troponin activator acting on thin filaments, has been shown to be effective as an agent that increases muscle strength, and can be used to compensate for hypotension in the muscle dysfunction. In the studies on genetically modified mice and cells from a patient with rod myopathy carrying an actin mutation (ACTA1H40Y), treatment with tirasemtiv increased inotropic parameters to those comparable to healthy controls [118].

One of the most popular targets for regulating a muscle mass is myostatin. A decrease in signaling of this myokine leads to a sharp increase in the muscle mass due to the intensification of the muscle fiber growth [119]. The first similar drug, domagruzumab (PF-06252616, Pfizer), which is a recombinant humanized antibody to myostatin, was withdrawn during the second phase of clinical trials, despite the fact that in the first phase, a 6.1% increase in the muscle mass after treatment was shown compared with a placebo group [120]. Another antimyostatin drug, BMS-986089, has demonstrated its high efficacy in preclinical test systems in mice and cynomolgus monkeys, and is currently undergoing clinical trials. However, in general, despite the theoretical promise of the approach and positive initial results, the recent clinical data demonstrate that antimyostatin therapy is less effective than expected. In addition, the long-term effects of antimyostatin therapy require a particularly close study, due to the possible negative impact on the pool of myosatellite cells [121].

CONCLUSION

Hereditary myopathies are a group of incurable diseases with a wide range of symptoms and a high variability in the clinical course. Currently, a large number of therapeutic approaches have been developed and approved for the use in various types of myopathies (Fig. 2). The most developed are the methods for the correction of muscular dystrophies, which, due to the progressive nature of the course, have the largest number of pathogenetic pathways that can be targets for therapy. At the same time, the smallest number of therapeutic options is available for the treatment of congenital myopathies, where the hereditary defect is permanently manifested throughout life, and there are no secondary alteration factors, such as inflammation and fibrosis. In addition, with the exception of some nosologies, there are no effective approaches to correct metabolic and mitochondrial myopathies.

 

Figure 2 – Varies of existing pharmacological methods for hereditary myopathies correction depending on disease type

 

In the treatment of all myopathies, an important role is played by symptomatic and supportive therapy aimed at treating pain and symptoms from other organs and systems. Osteoporosis [122] and pneumonia are regular consequences arising from hypodynamia myopathies, which are treated according to standard schemes.

It should be notified that in recent years, gene therapy approaches that correct or compensate for a defect at the gene level have become increasingly important. These approaches were not been covered in the work, the aim of which was to analyze the existing conventional strategies. However, to date, it is gene and cell therapy that constitute the most growing and promising layer of pharmacological agents for the treatment of hereditary myopathies.

In congenital myopathies, tirasemtiv, rapid skeletal troponin activator acting on thin filaments, has been shown to be effective. Theoretically, this approach can be effective in other types of myopathies. For the treatment of muscular dystrophies, anti-inflammatory and antifibrotic drugs, as well as antimyostatin therapy and a strategy aimed at translation through stop codons (applicable for nonsense mutations), can be used. In addition, metabolic drugs, metabolic enzyme cofactors, mitochondrial biogenesis stimulants, and antioxidants can be used to treat mitochondrial and metabolic myopathies. Finally, the recombinant drugs alglucosidase and avalglucosidase have been clinically approved for the replacement therapy of metabolic myopathies (Pompe’s disease).

FUNDING

The study was financially supported by the Ministry of Education and Science of the Russian Federation, Agreement No. 075-15-2021-1346.

CONFLICT OF INTEREST

The authors declare no conflict of interest.

AUTHORS’ CONTRIBUTION

MVP – idea creating, article concept planning, advising on writing of individual manuscript sections; MVK – idea development, article writing; AMK –literature analysis, article writing; NSZ – literature analysis, article writing; KNL – article writing, graphic material preparing; MOS – literature analysis, article writing; EAK– literature analysis, article writing; OSG – literature analysis, article writing; ISK – literature analysis, article writing; AVD – article concept planning, article writing.

×

About the authors

Mikhail V. Pokrovsky

Belgorod State National Research University

Author for correspondence.
Email: mpokrovsky@yandex.ru
ORCID iD: 0000-0002-2761-6249

Doctor of Sciences (Medicine), Professor of the Department of Pharmacology and Clinical Pharmacology, Head of the Research Institute of Pharmacology of Living Systems

Russian Federation, 85, Pobeda Str., Belgorod, 308015

Mikhail V. Korokin

Belgorod State National Research University

Email: mkorokin@mail.ru
ORCID iD: 0000-0001-5402-0697

Doctor of Medical Sciences, Associate Professor, Professor of the Department of Pharmacology and Clinical Pharmacology

Russian Federation, 85, Pobeda Str., Belgorod, 308015

Anastasia M. Krayushkina

Belgorod State National Research University

Email: annkrayushkina98@gmail.com
ORCID iD: 0000-0002-6830-3820

Assistant of the Department of Pharmacology and Clinical Pharmacology

Russian Federation, 85, Pobeda Str., Belgorod, 308015

Nikita S. Zhunusov

Belgorod State National Research University

Email: nzhunu@mail.ru
ORCID iD: 0000-0002-1969-3615

Assistant of the Department of Pharmacology and Clinical Pharmacology

Russian Federation, 85, Pobeda Str., Belgorod, 308015

Konstantin N. Lapin

V.A. Negovsky Research Institute of General Reanimatology, Federal Scientific and Clinical Center for Resuscitation and Rehabilitology

Email: k.n.lapin@gmail.com
ORCID iD: 0000-0002-7760-3526

Researcher, Laboratory of Experimental Research

Russian Federation, Bld. 2, 25, Petrovka Str., Moscow, 107031

Maria O. Soldatova

Kursk State Medical University

Email: mar.sold46@gmail.com
ORCID iD: 0000-0001-6637-1654

laboratory researcher at the Research Institute of Population Genetics and Epidemiology

Russian Federation, 3, Karl Marx Str., Kursk, 305041

Egor A. Kuzmin

Sechenov First Moscow State Medical University (Sechenov University)

Email: eg.ku@yandex.ru
ORCID iD: 0000-0003-4098-1125

laboratory assistant of the Department of Histology

Russian Federation, Bld. 2, 8, Trubetskaya Str., Moscow, 119991

Oleg S. Gudyrev

Belgorod State National Research University

Email: gudyrev@mail.ru
ORCID iD: 0000-0003-0097-000X

Candidate of Sciences (Medicine), Associate Professor, Associate Professor of the Department of Pharmacology and Clinical Pharmacology

Russian Federation, 85, Pobeda Str., Belgorod, 308015

Indira S. Kochkarova

Belgorod State National Research University

Email: kochkarova@bsu.edu.ru
ORCID iD: 0000-0001-6202-9923

Junior Researcher, Research Institute of Pharmacology of Living Systems

Russian Federation, 85, Pobeda Str., Belgorod, 308015

Aleksey V. Deikin

Belgorod State National Research University

Email: deykin@bsu.edu.ru
ORCID iD: 0000-0002-8151-6337

Candidate of Sciences (Biology), Associate Professor of the Department of Pharmacology and Clinical Pharmacology

Russian Federation, 85, Pobeda Str., Belgorod, 308015

References

  1. Cardamone M, Darras BT, Ryan MM. Inherited myopathies and muscular dystrophies. Semin Neurol. 2008 Apr;28(2):250–9. doi: 10.1055/s-2008-1062269
  2. Butterfield RJ. Congenital Muscular Dystrophy and Congenital Myopathy. Continuum (Minneap Minn). 2019 Dec;25(6):1640–61. doi: 10.1212/CON.0000000000000792
  3. Yu Wai Man CY, Smith T, Chinnery PF, Turnbull DM, Griffiths PG. Assessment of visual function in chronic progressive external ophthalmoplegia. Eye (Lond). 2006 May;20(5):564–8. doi: 10.1038/sj.eye.6701924
  4. Naidoo M, Anthony K. Dystrophin Dp71 and the Neuropathophysiology of Duchenne Muscular Dystrophy. Mol Neurobiol. 2020 Mar;57(3):1748–67. doi: 10.1007/s12035-019-01845-w
  5. Toscano A, Musumeci O. Tarui disease and distal glycogenoses: clinical and genetic update. Acta Myol. 2007 Oct;26(2):105–7.
  6. Pfeffer G, Chinnery PF. Diagnosis and treatment of mitochondrial myopathies. Ann Med. 2013 Feb;45(1):4–16. doi: 10.3109/07853890.2011.605389
  7. Isabekova PS, Alekseeva TM. [The hereditary progressive limb-girdle muscular dystrophy type 2l (anoctominopathy)]. Modern problems of science and education. 2020;(4):62. Russian
  8. Ervasti JM, Campbell KP. A role for the dystrophin-glycoprotein complex as a transmembrane linker between laminin and actin. J Cell Biol. 1993 Aug;122(4):809–23. doi: 10.1083/jcb.122.4.809
  9. Vilquin JT, Brussee V, Asselin I, Kinoshita I, Gingras M, Tremblay JP. Evidence of mdx mouse skeletal muscle fragility in vivo by eccentric running exercise. Muscle Nerve. 1998 May;21(5):567–76. doi: 10.1002/(sici)1097-4598(199805)21:5<567::aid-mus2>3.0.co;2-6
  10. Weller B, Karpati G, Carpenter S. Dystrophin-deficient mdx muscle fibers are preferentially vulnerable to necrosis induced by experimental lengthening contractions. J Neurol Sci. 1990 Dec;100(1–2):9–13. doi: 10.1016/0022-510x(90)90005-8
  11. Mizuno Y. Prevention of myonecrosis in mdx mice: Effect of immobilization by the local tetanus method. Brain and Development. 1992;14 (Issue 5):319–22. doi: 10.1016/S0387-7604(12)80151-3
  12. Mokhtarian A, Lefaucheur JP, Even PC, Sebille A. Hindlimb immobilization applied to 21-day-old mdx mice prevents the occurrence of muscle degeneration. J Appl Physiol (1985). 1999 Mar;86(3):924–31. doi: 10.1152/jappl.1999.86.3.924
  13. Le S, Yu M, Hovan L, Zhao Z, Ervasti J, Yan J. Dystrophin As a Molecular Shock Absorber. ACS Nano. 2018 Dec 26;12(12):12140–8. doi: 10.1021/acsnano.8b05721
  14. North KN, Wang CH, Clarke N, Jungbluth H, Vainzof M, Dowling JJ, Amburgey K, Quijano-Roy S, Beggs AH, Sewry C, Laing NG, Bönnemann CG; International Standard of Care Committee for Congenital Myopathies. Approach to the diagnosis of congenital myopathies. Neuromuscul Disord. 2014 Feb;24(2):97–116. doi: 10.1016/j.nmd.2013.11.003
  15. Tubridy N, Fontaine B, Eymard B. Congenital myopathies and congenital muscular dystrophies. Curr Opin Neurol. 2001 Oct;14(5):575–82. doi: 10.1097/00019052-200110000-00005
  16. Jungbluth H, Voermans NC. Congenital myopathies: not only a paediatric topic. Curr Opin Neurol. 2016 Oct;29(5): 642–50. doi: 10.1097/WCO.0000000000000372
  17. Cassandrini D, Trovato R, Rubegni A, Lenzi S, Fiorillo C, Baldacci J, Minetti C, Astrea G, Bruno C, Santorelli FM; Italian Network on Congenital Myopathies. Congenital myopathies: clinical phenotypes and new diagnostic tools. Ital J Pediatr. 2017 Nov 15;43(1):101. doi: 10.1186/s13052-017-0419-z
  18. Olpin SE, Murphy E, Kirk RJ, Taylor RW, Quinlivan R. The investigation and management of metabolic myopathies. J Clin Pathol. 2015 Jun;68(6):410–7. doi: 10.1136/jclinpath-2014-202808
  19. Tein I. Metabolic myopathies. Semin Pediatr Neurol. 1996 Jun;3(2):59–98. doi: 10.1016/s1071-9091(96)80038-6
  20. Tarnopolsky MA. Metabolic Myopathies. Continuum (Minneap Minn). 2016 Dec;22(6, Muscle and Neuromuscular Junction Disorders):1829–51. doi: 10.1212/CON.0000000000000403
  21. Kirby DM, Crawford M, Cleary MA, Dahl HH, Dennett X, Thorburn DR. Respiratory chain complex I deficiency: an underdiagnosed energy generation disorder. Neurology. 1999 Apr 12;52(6):1255–64. doi: 10.1212/wnl.52.6.1255
  22. Fassone E, Rahman S. Complex I deficiency: clinical features, biochemistry and molecular genetics. J Med Genet. 2012 Sep;49(9):578–90. doi: 10.1136/jmedgenet-2012-101159. Erratum in: J Med Genet. 2012 Oct;49(10):668.
  23. Abramov AY, Angelova PR. Cellular mechanisms of complex I-associated pathology. Biochem Soc Trans. 2019 Dec 20;47(6):1963–9. doi: 10.1042/BST20191042
  24. Chiaratti MR, Macabelli CH, Augusto Neto JD, Grejo MP, Pandey AK, Perecin F, Collado MD. Maternal transmission of mitochondrial diseases. Genet Mol Biol. 2020 Mar 2;43(suppl.1):e20190095. doi: 10.1590/1678-4685-GMB-2019-0095
  25. van den Ameele J, Li AYZ, Ma H, Chinnery PF. Mitochondrial heteroplasmy beyond the oocyte bottleneck. Semin Cell Dev Biol. 2020 Jan;97:156–66. doi: 10.1016/j.semcdb.2019.10.001
  26. Floros VI, Pyle A, Dietmann S, Wei W, Tang WCW, Irie N, Payne B, Capalbo A, Noli L, Coxhead J, Hudson G, Crosier M, Strahl H, Khalaf Y, Saitou M, Ilic D, Surani MA, Chinnery PF. Segregation of mitochondrial DNA heteroplasmy through a developmental genetic bottleneck in human embryos. Nat Cell Biol. 2018 Feb;20(2):144–51. doi: 10.1038/s41556-017-0017-8
  27. Ahmed ST, Craven L, Russell OM, Turnbull DM, Vincent AE. Diagnosis and Treatment of Mitochondrial Myopathies. Neurotherapeutics. 2018 Oct;15(4):943–53. doi: 10.1007/s13311-018-00674-4
  28. Miyoshi K, Kawai H, Iwasa M, Kusaka K, Nishino H. Autosomal recessive distal muscular dystrophy as a new type of progressive muscular dystrophy. Seventeen cases in eight families including an autopsied case. Brain. 1986 Feb;109 (Pt 1):31–54. doi: 10.1093/brain/109.1.31
  29. Bushby K, Straub V. One gene, one or many diseases? Simplifying dysferlinopathy. Neurology. 2010 Jul 27;75(4):298–9. doi: 10.1212/WNL.0b013e3181ea1649
  30. Nguyen K, Bassez G, Bernard R, Krahn M, Labelle V, Figarella-Branger D, Pouget J, Hammouda el H, Béroud C, Urtizberea A, Eymard B, Leturcq F, Lévy N. Dysferlin mutations in LGMD2B, Miyoshi myopathy, and atypical dysferlinopathies. Hum Mutat. 2005 Aug;26(2):165. doi: 10.1002/humu.9355
  31. Le Rumeur E, Winder SJ, Hubert JF. Dystrophin: more than just the sum of its parts. Biochim Biophys Acta. 2010 Sep;1804(9):1713–22. doi: 10.1016/j.bbapap.2010.05.001
  32. Liu J, Aoki M, Illa I, Wu C, Fardeau M, Angelini C, Serrano C, Urtizberea JA, Hentati F, Hamida MB, Bohlega S, Culper EJ, Amato AA, Bossie K, Oeltjen J, Bejaoui K, McKenna-Yasek D, Hosler BA, Schurr E, Arahata K, de Jong PJ, Brown RH Jr. Dysferlin, a novel skeletal muscle gene, is mutated in Miyoshi myopathy and limb girdle muscular dystrophy. Nat Genet. 1998 Sep;20(1):31–6. doi: 10.1038/1682
  33. Harris E, Bladen CL, Mayhew A, James M, Bettinson K, Moore U, Smith FE, Rufibach L, Cnaan A, Bharucha-Goebel DX, Blamire AM, Bravver E, Carlier PG, Day JW, Díaz-Manera J, Eagle M, Grieben U, Harms M, Jones KJ, Lochmüller H, Mendell JR, Mori-Yoshimura M, Paradas C, Pegoraro E, Pestronk A, Salort-Campana E, Schreiber-Katz O, Semplicini C, Spuler S, Stojkovic T, Straub V, Takeda S, Rocha CT, Walter MC, Bushby K; Jain COS Consortium. The Clinical Outcome Study for dysferlinopathy: An international multicenter study. Neurol Genet. 2016 Aug 4;2(4):e89. doi: 10.1212/NXG.0000000000000089
  34. Yiu EM, Kornberg AJ. Duchenne muscular dystrophy. Neurol India. 2008;56:236–47. doi: 10.4103/0028-3886.43441
  35. Yiu EM, Kornberg AJ. Duchenne muscular dystrophy. J Paediatr Child Health. 2015 Aug;51(8):759–64. doi: 10.1111/jpc.12868
  36. Flanigan KM. Duchenne and Becker muscular dystrophies. Neurol Clin. 2014 Aug;32(3):671–88, viii. doi: 10.1016/j.ncl.2014.05.002
  37. Muntoni F, Torelli S, Ferlini A. Dystrophin and mutations: one gene, several proteins, multiple phenotypes. Lancet Neurol. 2003 Dec;2(12):731–40. doi: 10.1016/s1474-4422(03)00585-4
  38. Chang NC, Chevalier FP, Rudnicki MA. Satellite Cells in Muscular Dystrophy – Lost in Polarity. Trends Mol Med. 2016 Jun;22(6):479–96. doi: 10.1016/j.molmed.2016.04.002
  39. Dumont NA, Wang YX, von Maltzahn J, Pasut A, Bentzinger CF, Brun CE, Rudnicki MA. Dystrophin expression in muscle stem cells regulates their polarity and asymmetric division. Nat Med. 2015 Dec;21(12):1455–63. doi: 10.1038/nm.3990
  40. Bönnemann CG. The collagen VI-related myopathies Ullrich congenital muscular dystrophy and Bethlem myopathy. Handb Clin Neurol. 2011;101:81–96. doi: 10.1016/B978-0-08-045031-5.00005-0
  41. Bönnemann CG. The collagen VI-related myopathies: muscle meets its matrix. Nat Rev Neurol. 2011 Jun 21;7(7):379–90. doi: 10.1038/nrneurol.2011.81
  42. Katzin LW, Amato AA. Pompe disease: a review of the current diagnosis and treatment recommendations in the era of enzyme replacement therapy. J Clin Neuromuscul Dis. 2008 Jun;9(4):421–31. doi: 10.1097/CND.0b013e318176dbe4
  43. Taverna S, Cammarata G, Colomba P, Sciarrino S, Zizzo C, Francofonte D, Zora M, Scalia S, Brando C, Curto AL, Marsana EM, Olivieri R, Vitale S, Duro G. Pompe disease: pathogenesis, molecular genetics and diagnosis. Aging (Albany NY). 2020 Aug 3;12(15):15856–74. doi: 10.18632/aging.103794
  44. Ghosh P, Dahms NM, Kornfeld S. Mannose 6-phosphate receptors: new twists in the tale. Nat Rev Mol Cell Biol. 2003 Mar;4(3):202–12. doi: 10.1038/nrm1050
  45. Kohler L, Puertollano R, Raben N. Pompe Disease: From Basic Science to Therapy. Neurotherapeutics. 2018 Oct;15(4):928–42. doi: 10.1007/s13311-018-0655-y
  46. Peruzzo P, Pavan E, Dardis A. Molecular genetics of Pompe disease: a comprehensive overview. Ann Transl Med. 2019 Jul;7(13):278. doi: 10.21037/atm.2019.04.13
  47. Tarlow MJ, Ellis DA, Pearce GW, Anderson M. Muscle phosphofructokinase deficiency (Tarui’s disease). Proc Nutr Soc. 1979 Dec;38(3):110A.
  48. Vorgerd M, Zange J, Kley R, Grehl T, Hüsing A, Jäger M, Müller K, Schröder R, Mortier W, Fabian K, Malin JP, Luttmann A. Effect of high-dose creatine therapy on symptoms of exercise intolerance in McArdle disease: double-blind, placebo-controlled crossover study. Arch Neurol. 2002 Jan;59(1):97–101. doi: 10.1001/archneur.59.1.97
  49. Yamasaki T, Nakajima H. [Phosphofructokinase (PFK)]. Nihon Rinsho. 2004 Dec;62 Suppl 12:835-9. Japanese
  50. Wong LJ, Naviaux RK, Brunetti-Pierri N, Zhang Q, Schmitt ES, Truong C, Milone M, Cohen BH, Wical B, Ganesh J, Basinger AA, Burton BK, Swoboda K, Gilbert DL, Vanderver A, Saneto RP, Maranda B, Arnold G, Abdenur JE, Waters PJ, Copeland WC. Molecular and clinical genetics of mitochondrial diseases due to POLG mutations. Hum Mutat. 2008 Sep;29(9):E150–72. doi: 10.1002/humu.20824
  51. Cohen BH, Chinnery PF, Copeland WC. POLG-Related Disorders. 2010 Mar 16 [Updated 2018 Mar 1]. In: Adam MP, Everman DB, Mirzaa GM, et al., editors. GeneReviews® [Internet]. Seattle (WA): University of Washington, Seattle; 1993–2022. Available from: https://www.ncbi.nlm.nih.gov/books/NBK26471/.
  52. Rajakulendran S, Pitceathly RD, Taanman JW, Costello H, Sweeney MG, Woodward CE, Jaunmuktane Z, Holton JL, Jacques TS, Harding BN, Fratter C, Hanna MG, Rahman S. A Clinical, Neuropathological and Genetic Study of Homozygous A467T POLG-Related Mitochondrial Disease. PLoS One. 2016 Jan 6;11(1):e0145500. doi: 10.1371/journal.pone.0145500
  53. Adler M, Shieh PB. Metabolic Myopathies. Semin Neurol. 2015 Aug;35(4):385–97. doi: 10.1055/s-0035-1558973
  54. Meena NK, Raben N. Pompe Disease: New Developments in an Old Lysosomal Storage Disorder. Biomolecules. 2020 Sep 18;10(9):1339. doi: 10.3390/biom10091339
  55. Dhillon S. Avalglucosidase alfa: First Approval. Drugs. 2021 Oct;81(15):1803–9. doi: 10.1007/s40265-021-01600-3
  56. Horn JM, Obermeyer AC. Genetic and Covalent Protein Modification Strategies to Facilitate Intracellular Delivery. Biomacromolecules. 2021;22:4883–904. doi: 10.1021/acs.biomac.1c00745
  57. Lawlor MW, Armstrong D, Viola MG, Widrick JJ, Meng H, Grange RW, Childers MK, Hsu CP, O’Callaghan M, Pierson CR, Buj-Bello A, Beggs AH. Enzyme replacement therapy rescues weakness and improves muscle pathology in mice with X-linked myotubular myopathy. Hum Mol Genet. 2013 Apr 15;22(8):1525–38. doi: 10.1093/hmg/ddt003
  58. Wu RP, Youngblood DS, Hassinger JN, Lovejoy CE, Nelson MH, Iversen PL, Moulton HM. Cell-penetrating peptides as transporters for morpholino oligomers: effects of amino acid composition on intracellular delivery and cytotoxicity. Nucleic Acids Res. 2007;35(15):5182–91. doi: 10.1093/nar/gkm478
  59. Bladen CL, Salgado D, Monges S, Foncuberta ME, Kekou K, Kosma K, Dawkins H, Lamont L, Roy AJ, Chamova T, Guergueltcheva V, Chan S, Korngut L, Campbell C, Dai Y, Wang J, Barišić N, Brabec P, Lahdetie J, Walter MC, Schreiber-Katz O, Karcagi V, Garami M, Viswanathan V, Bayat F, Buccella F, Kimura E, Koeks Z, van den Bergen JC, Rodrigues M, Roxburgh R, Lusakowska A, Kostera- Pruszczyk A, Zimowski J, Santos R, Neagu E, Artemieva S, Rasic VM, Vojinovic D, Posada M, Bloetzer C, Jeannet PY, Joncourt F, Díaz-Manera J, Gallardo E, Karaduman AA, Topaloğlu H, El Sherif R, Stringer A, Shatillo AV, Martin AS, Peay HL, Bellgard MI, Kirschner J, Flanigan KM, Straub V, Bushby K, Verschuuren J, Aartsma-Rus A, Béroud C, Lochmüller H. The TREAT-NMD DMD Global Database: analysis of more than 7,000 Duchenne muscular dystrophy mutations. Hum Mutat. 2015 Apr;36(4):395–402. doi: 10.1002/humu.22758
  60. Laporte J, Biancalana V, Tanner SM, Kress W, Schneider V, Wallgren-Pettersson C, Herger F, Buj-Bello A, Blondeau F, Liechti-Gallati S, Mandel JL. MTM1 mutations in X-linked myotubular myopathy. Hum Mutat. 2000;15(5):393–409. doi: 10.1002/(SICI)1098-1004(200005)15:5<393::AID-HUMU1>3.0.CO;2-R
  61. Diop D, Chauvin C, Jean-Jean O. Aminoglycosides and other factors promoting stop codon readthrough in human cells. C R Biol. 2007 Jan;330(1):71–9. doi: 10.1016/j.crvi.2006.09.001
  62. Schroeder R, Waldsich C, Wank H. Modulation of RNA function by aminoglycoside antibiotics. EMBO J. 2000 Jan 4;19(1):1–9. doi: 10.1093/emboj/19.1.1
  63. Barton-Davis ER, Cordier L, Shoturma DI, Leland SE, Sweeney HL. Aminoglycoside antibiotics restore dystrophin function to skeletal muscles of mdx mice. J Clin Invest. 1999 Aug;104(4):375–81. doi: 10.1172/JCI7866
  64. Clancy JP, Bebök Z, Ruiz F, King C, Jones J, Walker L, Greer H, Hong J, Wing L, Macaluso M, Lyrene R, Sorscher EJ, Bedwell DM. Evidence that systemic gentamicin suppresses premature stop mutations in patients with cystic fibrosis. Am J Respir Crit Care Med. 2001 Jun;163(7):1683–92. doi: 10.1164/ajrccm.163.7.2004001
  65. Howard M, Frizzell RA, Bedwell DM. Aminoglycoside antibiotics restore CFTR function by overcoming premature stop mutations. Nat Med. 1996 Apr;2(4): 467–9. doi: 10.1038/nm0496-467
  66. Carnes J, Jacobson M, Leinwand L, Yarus M. Stop codon suppression via inhibition of eRF1 expression. RNA. 2003 Jun;9(6):648–53. doi: 10.1261/rna.5280103
  67. Welch EM, Barton ER, Zhuo J, Tomizawa Y, Friesen WJ, Trifillis P, Paushkin S, Patel M, Trotta CR, Hwang S, Wilde RG, Karp G, Takasugi J, Chen G, Jones S, Ren H, Moon YC, Corson D, Turpoff AA, Campbell JA, Conn MM, Khan A, Almstead NG, Hedrick J, Mollin A, Risher N, Weetall M, Yeh S, Branstrom AA, Colacino JM, Babiak J, Ju WD, Hirawat S, Northcutt VJ, Miller LL, Spatrick P, He F, Kawana M, Feng H, Jacobson A, Peltz SW, Sweeney HL. PTC124 targets genetic disorders caused by nonsense mutations. Nature. 2007 May 3;447(7140):87–91. doi: 10.1038/nature05756
  68. Berger J, Li M, Berger S, Meilak M, Rientjes J, Currie PD. Effect of Ataluren on dystrophin mutations. J Cell Mol Med. 2020 Jun;24(12):6680–9. doi: 10.1111/jcmm.15319
  69. Allamand V, Bidou L, Arakawa M, Floquet C, Shiozuka M, Paturneau-Jouas M, Gartioux C, Butler-Browne GS, Mouly V, Rousset JP, Matsuda R, Ikeda D, Guicheney P. Drug-induced readthrough of premature stop codons leads to the stabilization of laminin alpha2 chain mRNA in CMD myotubes. J Gene Med. 2008 Feb;10(2):217–24. doi: 10.1002/jgm.1140
  70. Iezzi S, Cossu G, Nervi C, Sartorelli V, Puri PL. Stage-specific modulation of skeletal myogenesis by inhibitors of nuclear deacetylases. Proc Natl Acad Sci USA. 2002 May 28;99(11):7757–62. doi: 10.1073/pnas.112218599
  71. Iezzi S, Di Padova M, Serra C, Caretti G, Simone C, Maklan E, Minetti G, Zhao P, Hoffman EP, Puri PL, Sartorelli V. Deacetylase inhibitors increase muscle cell size by promoting myoblast recruitment and fusion through induction of follistatin. Dev Cell. 2004 May;6(5):673–84. doi: 10.1016/s1534-5807(04)00107-8
  72. Minetti GC, Colussi C, Adami R, Serra C, Mozzetta C, Parente V, Fortuni S, Straino S, Sampaolesi M, Di Padova M, Illi B, Gallinari P, Steinkühler C, Capogrossi MC, Sartorelli V, Bottinelli R, Gaetano C, Puri PL. Functional and morphological recovery of dystrophic muscles in mice treated with deacetylase inhibitors. Nat Med. 2006 Oct;12(10):1147–50. doi: 10.1038/nm1479
  73. Love DR, Hill DF, Dickson G, Spurr NK, Byth BC, Marsden RF, Walsh FS, Edwards YH, Davies KE. An autosomal transcript in skeletal muscle with homology to dystrophin. Nature. 1989 May 4;339(6219):55–8. doi: 10.1038/339055a0
  74. Khurana TS, Hoffman EP, Kunkel LM. Identification of a chromosome 6-encoded dystrophin-related protein. J Biol Chem. 1990 Oct 5;265(28):16717–20.
  75. Khurana TS, Watkins SC, Chafey P, Chelly J, Tomé FM, Fardeau M, Kaplan JC, Kunkel LM. Immunolocalization and developmental expression of dystrophin related protein in skeletal muscle. Neuromuscul Disord. 1991;1(3):185–94. doi: 10.1016/0960-8966(91)90023-l
  76. Starikova AV, Skopenkova VV, Polikarpova AV, Reshetov DA, Vassilieva SG, Velyaev OA, Shmidt AA, Savchenko IM, Soldatov VO, Egorova TV, Bardina MV. Therapeutic potential of highly functional codon-optimized microutrophin for muscle-specific expression. Sci Rep. 2022 Jan 17;12(1):848. doi: 10.1038/s41598-022-04892-x
  77. Vuorinen A, Wilkinson IVL, Chatzopoulou M, Edwards B, Squire SE, Fairclough RJ, Bazan NA, Milner JA, Conole D, Donald JR, Shah N, Willis NJ, Martínez RF, Wilson FX, Wynne GM, Davies SG, Davies KE, Russell AJ. Discovery and mechanism of action studies of 4,6-diphenylpyrimidine-2-carbohydrazides as utrophin modulators for the treatment of Duchenne muscular dystrophy. Eur J Med Chem. 2021 Aug 5;220:113431. doi: 10.1016/j.ejmech.2021.113431
  78. Chatzopoulou M, Conole D, Emer E, Rowleya JA, Willisa NJ, Squireb SE, Gillc B, Broughc S, Wilsond FX, Wynnea GM, Daviesa SG, Daviesb KE, Russellae AJ. Structure-activity relationships of 2-pyrimidinecarbohydrazides as utrophin modulators for the potential treatment of Duchenne muscular dystrophy. Bioorg Med Chem. 2022;69:116812. doi: 10.1016/j.bmc.2022.116812
  79. Pearson CM, Rimer DG, Mommaerts WF. A metabolic myopathy due to absence of muscle phosphorylase. Am J Med. 1961 Apr;30:502–17. doi: 10.1016/0002-9343(61)90075-4
  80. Preisler N, Pradel A, Husu E, Madsen KL, Becquemin MH, Mollet A, Labrune P, Petit F, Hogrel JY, Jardel C, Maillot F, Vissing J, Laforêt P. Exercise intolerance in Glycogen Storage Disease Type III: weakness or energy deficiency? Mol Genet Metab. 2013 May;109(1):14–20. doi: 10.1016/j.ymgme.2013.02.008
  81. Preisler N, Laforêt P, Echaniz-Laguna A, Ørngreen MC, Lonsdorfer-Wolf E, Doutreleau S, Geny B, Stojkovic T, Piraud M, Petit FM, Vissing J. Fat and carbohydrate metabolism during exercise in phosphoglucomutase type 1 deficiency. J Clin Endocrinol Metab. 2013 Jul;98(7):E1235–40. doi: 10.1210/jc.2013-1651
  82. Stojkovic T, Vissing J, Petit F, Piraud M, Orngreen MC, Andersen G, Claeys KG, Wary C, Hogrel JY, Laforêt P. Muscle glycogenosis due to phosphoglucomutase 1 deficiency. N Engl J Med. 2009 Jul 23;361(4):425–7. doi: 10.1056/NEJMc0901158
  83. Roe CR, Mochel F. Anaplerotic diet therapy in inherited metabolic disease: therapeutic potential. J Inherit Metab Dis. 2006 Apr-Jun;29(2–3):332–40. doi: 10.1007/s10545-006-0290-3
  84. Roe CR, Sweetman L, Roe DS, David F, Brunengraber H. Treatment of cardiomyopathy and rhabdomyolysis in long-chain fat oxidation disorders using an anaplerotic odd-chain triglyceride. J Clin Invest. 2002 Jul;110(2): 259–69. doi: 10.1172/JCI15311
  85. Roe CR, Yang BZ, Brunengraber H, Roe DS, Wallace M, Garritson BK. Carnitine palmitoyltransferase II deficiency: successful anaplerotic diet therapy. Neurology. 2008 Jul 22;71(4):260–4. doi: 10.1212/01.wnl.0000318283.42961.e9
  86. Laforêt P, Ørngreen M, Preisler N, Andersen G, Vissing J. Blocked muscle fat oxidation during exercise in neutral lipid storage disease. Arch Neurol. 2012 Apr;69(4):530–3. doi: 10.1001/archneurol.2011.631
  87. Farshidfar F, Pinder MA, Myrie SB. Creatine Supplementation and Skeletal Muscle Metabolism for Building Muscle Mass-Review of the Potential Mechanisms of Action. Curr Protein Pept Sci. 2017;18(12):1273–87. doi: 10.2174/1389203718666170606105108
  88. Marriage B, Clandinin MT, Glerum DM. Nutritional cofactor treatment in mitochondrial disorders. J Am Diet Assoc. 2003 Aug;103(8):1029–38. doi: 10.1016/s0002-8223(03)00476-0
  89. Avula S, Parikh S, Demarest S, Kurz J, Gropman A. Treatment of mitochondrial disorders. Curr Treat Options Neurol. 2014 Jun;16(6):292. doi: 10.1007/s11940-014-0292-7
  90. Tinker RJ, Lim AZ, Stefanetti RJ, McFarland R. Current and Emerging Clinical Treatment in Mitochondrial Disease. Mol Diagn Ther. 2021 Mar;25(2):181–206. doi: 10.1007/s40291-020-00510-6
  91. Viscomi C, Bottani E, Zeviani M. Emerging concepts in the therapy of mitochondrial disease. Biochim Biophys Acta. 2015 Jun-Jul;1847(6-7):544–57. doi: 10.1016/j.bbabio.2015.03.001
  92. Barshop BA, Naviaux RK, McGowan KA, Levine F, Nyhan WL, Loupis-Geller A, Haas RH. Chronic treatment of mitochondrial disease patients with dichloroacetate. Mol Genet Metab. 2004 Sep-Oct;83(1–2):138–49. doi: 10.1016/j.ymgme.2004.06.009
  93. Prietsch V, Lindner M, Zschocke J, Nyhan WL, Hoffmann GF. Emergency management of inherited metabolic diseases. J Inherit Metab Dis. 2002 Nov;25(7):531–46. doi: 10.1023/a:1022040422590
  94. Parikh S, Saneto R, Falk MJ, Anselm I, Cohen BH, Haas R, Medicine Society TM. A modern approach to the treatment of mitochondrial disease. Curr Treat Options Neurol. 2009 Nov;11(6):414–30. doi: 10.1007/s11940-009-0046-0
  95. Angelova PR, Esteras N, Abramov AY. Mitochondria and lipid peroxidation in the mechanism of neurodegeneration: Finding ways for prevention. Med Res Rev. 2021 Mar;41(2):770–84. doi: 10.1002/med.21712
  96. Birnkrant DJ, Bushby K, Bann CM, Apkon SD, Blackwell A, Brumbaugh D, Case LE, Clemens PR, Hadjiyannakis S, Pandya S, Street N, Tomezsko J, Wagner KR, Ward LM, Weber DR; DMD Care Considerations Working Group. Diagnosis and management of Duchenne muscular dystrophy, part 1: diagnosis, and neuromuscular, rehabilitation, endocrine, and gastrointestinal and nutritional management. Lancet Neurol. 2018 Mar;17(3):251–67. doi: 10.1016/S1474-4422(18)30024-3. Epub 2018 Feb 3. Erratum in: Lancet Neurol. 2018.
  97. McNeil SM, Woulfe J, Ross C, Tarnopolsky MA. Congenital inflammatory myopathy: a demonstrative case and proposed diagnostic classification. Muscle Nerve. 2002 Feb;25(2):259–64. doi: 10.1002/mus.10043
  98. Walter MC, Reilich P, Thiele S, Schessl J, Schreiber H, Reiners K, Kress W, Müller-Reible C, Vorgerd M, Urban P, Schrank B, Deschauer M, Schlotter-Weigel B, Kohnen R, Lochmüller H. Treatment of dysferlinopathy with deflazacort: a double-blind, placebo-controlled clinical trial. Orphanet J Rare Dis. 2013 Feb 14;8:26. doi: 10.1186/1750-1172-8-26
  99. Bonifati MD, Ruzza G, Bonometto P, Berardinelli A, Gorni K, Orcesi S, Lanzi G, Angelini C. A multicenter, double-blind, randomized trial of deflazacort versus prednisone in Duchenne muscular dystrophy. Muscle Nerve. 2000 Sep;23(9):1344–7. doi: 10.1002/1097-4598(200009)23:9<1344::aid-mus4>3.0.co;2-f
  100. Escolar DM, Hache LP, Clemens PR, Cnaan A, McDonald CM, Viswanathan V, Kornberg AJ, Bertorini TE, Nevo Y, Lotze T, Pestronk A, Ryan MM, Monasterio E, Day JW, Zimmerman A, Arrieta A, Henricson E, Mayhew J, Florence J, Hu F, Connolly AM. Randomized, blinded trial of weekend vs daily prednisone in Duchenne muscular dystrophy. Neurology. 2011 Aug 2;77(5):444–52. doi: 10.1212/WNL.0b013e318227b164
  101. Pasquini F, Guerin C, Blake D, Davies K, Karpati G, Holland P. The effect of glucocorticoids on the accumulation of utrophin by cultured normal and dystrophic human skeletal muscle satellite cells. Neuromuscul Disord. 1995 Mar;5(2):105–14. doi: 10.1016/0960-8966(94)00042-8
  102. Serra F, Quarta M, Canato M, Toniolo L, De Arcangelis V, Trotta A, Spath L, Monaco L, Reggiani C, Naro F. Inflammation in muscular dystrophy and the beneficial effects of non-steroidal anti-inflammatory drugs. Muscle Nerve. 2012 Nov;46(5):773–84. doi: 10.1002/mus.23432
  103. Aoyagi K, Ohara-Imaizumi M, Nishiwaki C, Nakamichi Y, Nagamatsu S. Insulin/phosphoinositide 3-kinase pathway accelerates the glucose-induced first-phase insulin secretion through TrpV2 recruitment in pancreatic β-cells. Biochem J. 2010 Dec 1;432(2):375–86. doi: 10.1042/BJ20100864
  104. Iwata Y, Katanosaka Y, Shijun Z, Kobayashi Y, Hanada H, Shigekawa M, Wakabayashi S. Protective effects of Ca2+ handling drugs against abnormal Ca2+ homeostasis and cell damage in myopathic skeletal muscle cells. Biochem Pharmacol. 2005 Sep 1;70(5):740–51. doi: 10.1016/j.bcp.2005.05.034
  105. Swiderski K, Todorov M, Gehrig SM, Naim T, Chee A, Stapleton DI, Koopman R, Lynch GS. Tranilast administration reduces fibrosis and improves fatigue resistance in muscles of mdx dystrophic mice. Fibrogenesis Tissue Repair. 2014 Jan 30;7(1):1. doi: 10.1186/1755-1536-7-1
  106. Hodgetts S, Radley H, Davies M, Grounds MD. Reduced necrosis of dystrophic muscle by depletion of host neutrophils, or blocking TNFalpha function with Etanercept in mdx mice. Neuromuscul Disord. 2006 Oct;16(9–10):591–602. doi: 10.1016/j.nmd.2006.06.011
  107. Piers AT, Lavin T, Radley-Crabb HG, Bakker AJ, Grounds M, Pinniger GJ. Blockade of TNF in vivo using cV1q antibody reduces contractile dysfunction of skeletal muscle in response to eccentric exercise in dystrophic mdx and normal mice. Neuromuscul Disord. 2011;21(Issue 2): 132–41. doi: 10.1016/j.nmd.2010.09.013
  108. Mahdy MAA. Skeletal muscle fibrosis: an overview. Cell Tissue Res. 2019 Mar;375(3):575–88. doi: 10.1007/s00441-018-2955-2
  109. Tsabari R, Simchovitz E, Lavi E, Eliav O, Avrahami R, Ben-Sasson S, Do T. Safety and clinical outcome of tamoxifen in Duchenne muscular dystrophy. Neuromuscul Disord. 2021;31:803–13. doi: 10.1016/j.nmd.2021.05.005
  110. Wu B, Shah SN, Lu P, Bollinger LE, Blaeser A, Sparks S, Harper AD, Lu QL. Long-Term Treatment of Tamoxifen and Raloxifene Alleviates Dystrophic Phenotype and Enhances Muscle Functions of FKRP Dystroglycanopathy. Am J Pathol. 2018 Apr;188(4):1069–80. doi: 10.1016/j.ajpath.2017.12.011
  111. Ceco E, McNally EM. Modifying muscular dystrophy through transforming growth factor-β. FEBS J. 2013 Sep;280(17):4198–209. doi: 10.1111/febs.12266
  112. Biressi S, Miyabara EH, Gopinath SD, Carlig PM, Rando TA. A Wnt-TGFβ2 axis induces a fibrogenic program in muscle stem cells from dystrophic mice. Sci Transl Med. 2014 Dec 17;6(267):267ra176. doi: 10.1126/scitranslmed.3008411
  113. Demonbreun AR, Fallon KS, Oosterbaan CC, Vaught LA, Reiser NL, Bogdanovic E, Velez MP, Salamone IM, Page PGT, Hadhazy M, Quattrocelli M, Barefield DY, Wood LD, Gonzalez JP, Morris C, McNally EM. Anti-latent TGFβ binding protein 4 antibody improves muscle function and reduces muscle fibrosis in muscular dystrophy. Sci Transl Med. 2021 Sep 8;13(610):eabf0376. doi: 10.1126/scitranslmed.abf0376
  114. Morales MG, Cabrera D, Céspedes C, Vio CP, Vazquez Y, Brandan E, Cabello-Verrugio C. Inhibition of the angiotensin-converting enzyme decreases skeletal muscle fibrosis in dystrophic mice by a diminution in the expression and activity of connective tissue growth factor (CTGF/CCN-2). Cell Tissue Res. 2013 Jul;353(1):173–87. doi: 10.1007/s00441-013-1642-6
  115. Sun G, Haginoya K, Wu Y, Chiba Y, Nakanishi T, Onuma A, Sato Y, Takigawa M, Iinuma K, Tsuchiya S. Connective tissue growth factor is overexpressed in muscles of human muscular dystrophy. J Neurol Sci. 2008 Apr 15;267(1-2): 48–56. doi: 10.1016/j.jns.2007.09.043
  116. Frazier K, Williams S, Kothapalli D, Klapper H, Grotendorst GR. Stimulation of fibroblast cell growth, matrix production, and granulation tissue formation by connective tissue growth factor. J Invest Dermatol. 1996 Sep;107(3):404–11. doi: 10.1111/1523-1747.ep12363389
  117. Smith LR, Barton ER. Regulation of fibrosis in muscular dystrophy. Matrix Biol. 2018 Aug;68–69:602–615. doi: 10.1016/j.matbio.2018.01.014
  118. de Winter JM, Gineste C, Minardi E, Brocca L, Rossi M, Borsboom T, Beggs AH, Bernard M, Bendahan D, Hwee DT, Malik FI, Pellegrino MA, Bottinelli R, Gondin J, Ottenheijm CAC. Acute and chronic tirasemtiv treatment improves in vivo and in vitro muscle performance in actin-based nemaline myopathy mice. Hum Mol Genet. 2021 Jun 26;30(14):1305–20. doi: 10.1093/hmg/ddab112
  119. McPherron AC, Lawler AM, Lee SJ. Regulation of skeletal muscle mass in mice by a new TGF-beta superfamily member. Nature. 1997 May 1;387(6628):83–90. doi: 10.1038/387083a0
  120. Guiraud S, Davies KE. Pharmacological advances for treatment in Duchenne muscular dystrophy. Curr Opin Pharmacol. 2017 Jun;34:36–48. doi: 10.1016/j.coph.2017.04.002
  121. Rybalka E, Timpani CA, Debruin DA, Bagaric RM, Campelj DG, Hayes A. The Failed Clinical Story of Myostatin Inhibitors against Duchenne Muscular Dystrophy: Exploring the Biology behind the Battle. Cells. 2020 Dec 10;9(12):2657. doi: 10.3390/cells9122657
  122. Korokin MV, Soldatov VO, Gudyrev OS, Koklin IS, Taran EI, Mishenin MO, Korokina LV, Kochkarov AA, Pokrovskii MV, Varaksin MV, Chupakhin ON. [The role of cortisol metabolism in the realization of pathogenetic links in the development of osteoporosis – the rationale for the search for new pharmacotherapeutic targets (review)]. Research Results in Biomedicine. 2022;8(4):457–473. doi: 10.18413/2658-6533-2022-8-4-0-5. Russian

Supplementary files

Supplementary Files
Action
1. JATS XML
2. Figure 1 – Classical pharmacological methods to compensate for inadequate functioning of mitochondria

Download (277KB)
3. Figure 2 – Varies of existing pharmacological methods for hereditary myopathies correction depending on disease type

Download (378KB)

Copyright (c) 2023 Pokrovsky M.V., Korokin M.V., Krayushkina A.M., Zhunusov N.S., Lapin K.N., Soldatova M.O., Kuzmin E.A., Gudyrev O.S., Kochkarova I.S., Deikin A.V.

Creative Commons License
This work is licensed under a Creative Commons Attribution 4.0 International License.

СМИ зарегистрировано Федеральной службой по надзору в сфере связи, информационных технологий и массовых коммуникаций (Роскомнадзор).
Регистрационный номер и дата принятия решения о регистрации СМИ: ПИ № ФС 77 - 67428 от 13.10.2016. 

This website uses cookies

You consent to our cookies if you continue to use our website.

About Cookies