Skip to main content
JACC: Basic to Translational Science logoLink to JACC: Basic to Translational Science
. 2022 Mar 9;7(6):608–625. doi: 10.1016/j.jacbts.2021.11.004

Cardiovascular Disease in Duchenne Muscular Dystrophy

Overview and Insight Into Novel Therapeutic Targets

Taylor I Schultz a,, Frank J Raucci Jr a,b,, Fadi N Salloum a,c,
PMCID: PMC9270569  PMID: 35818510

Highlights

  • Cardiomyopathy is the leading cause of death in patients with DMD.

  • DMD has no cure, and there is no current consensus for treatment of DMD cardiomyopathy.

  • This review discusses therapeutic strategies to potentially reduce or prevent cardiac dysfunction in DMD patients.

  • Additional studies are needed to firmly establish optimal treatment modalities for DMD cardiomyopathy.

Key Words: arrhythmias, cardiomyopathy, Duchenne muscular dystrophy, inflammatory modulators, myocardial fibrosis

Abbreviations and Acronyms: ACE, angiotensin-converting enzyme; ApN, adiponectin; ARB, angiotensin receptor blocker; BB, beta-blocker; BDNF, brain-derived neurotrophic factor; CMR, cardiac magnetic resonance imaging; Cx, connexin; DMD, Duchenne muscular dystrophy; DPC, dystrophin-associated protein complex; FFA, free fatty acid; HF, heart failure; LNP, lipid nanoparticle; LV, left ventricular; LVEF, left ventricular ejection fraction; miR, microRNA; NIV, noninvasive ventilation; Nrf2, nuclear factor erythroid 2-related factor 2; PKA, protein kinase A; PTX3, pentraxin 3; Px, pannexin; RNP, ribonucleoprotein complexes; RT-qPCR, reverse transcription-quantitative polymerase chain reaction; RyR2, ryanodine receptor isoform 2; sgRNA, single guide RNA; SR, sarcoplasmic reticulum; TrkB, tyrosine kinase B; TRPV2, transient receptor potential cation channel, subfamily V, member 2

Summary

Duchenne muscular dystrophy (DMD) is a devastating disease affecting approximately 1 in every 3,500 male births worldwide. Multiple mutations in the dystrophin gene have been implicated as underlying causes of DMD. However, there remains no cure for patients with DMD, and cardiomyopathy has become the most common cause of death in the affected population. Extensive research is under way investigating molecular mechanisms that highlight potential therapeutic targets for the development of pharmacotherapy for DMD cardiomyopathy. In this paper, the authors perform a literature review reporting on recent ongoing efforts to identify novel therapeutic strategies to reduce, prevent, or reverse progression of cardiac dysfunction in DMD.

Central Illustration

graphic file with name fx1.jpg


Duchenne muscular dystrophy (DMD) is a progressive myopathic disorder caused by a recessive mutation in the dystrophin gene on the X chromosome. DMD affects roughly 1.3 to 2.1 per 10,000 live male births.1 This devastating disease results in severe clinical symptoms. Progressive muscle weakness typically begins to affect young boys around the age of 2 or 3 years, eventually becoming wheelchair bound by the age of 12. Orthopedic fractures commonly occur mainly due to frequent falls. Scoliosis often manifests in patients with DMD, increasing the possibility of respiratory failure.2,3 Severity of symptoms primarily correlate with the quantity of dystrophin in the muscle ranging from absent/minimal dystrophin to higher levels; the less dystrophin, the more severe the phenotype.4

Dystrophin, part of a large glycoprotein complex, is located on the cytoplasmic side of the plasma membrane of muscle fibers.1 As part of this complex, known as the dystrophin-associated protein complex (DPC), dystrophin functions as mechanical reinforcement to the sarcolemma and to withstand contraction-induced injury, while also stabilizing the DPC, preventing its degradation. In the absence of dystrophin, the DPC degrades, resulting in several downstream detrimental effects including weakening of the sacrolemmal membrane, loss of membrane proteins, disrupted calcium homeostasis, up-regulation of inflammatory factors, and mitochondrial dysfunction, ultimately leading to degeneration of muscle fibers, necrosis, and cardiac fibrosis1 (Figure 1).

Figure 1.

Figure 1

Dystrophin Acts as a Molecular Scaffold and Influences Mechanisms of Calcium Handling in Cardiomyocytes

(A) Dystrophin present: Cytosolic Ca2+ is regulated primarily by LTCC, SACs, and NCX. In normal excitation–contraction (E-C) coupling, small influx of Ca2+ through LTCCs stimulates Ca2+ release from the SR through RYR2. Ca2+ activates nNOS within the dystrophin complex in a calmodulin-dependent manner. NO subsequently further activates SR Ca2+ turnover through s-nitrosylation of RYR2, IP3, and SERCA2. NO also augments E-C coupling through production of cGMP, which also reduces cardiac afterload by stimulating vasodilation. Normal physiological stretch activates NOX-2–dependent ROS production, which increases Ca2+ influx through SACs. Phospholamban (PLN) negatively regulates SERCA2 and β-adrenergic activation leads to PLN phosphorylation and dissociation from SERCA2, with a resultant increase in SR Ca2+ reuptake. Dystrophin helps to stabilize the sarcolemmal membrane during repeated stretch–relaxation cycling. (B) Dystrophin absent: Sarcolemmal influx of Ca2+ increases through disruption of the normal function of LTCCs, NCX, SACs, and microtears in the membrane. Mislocalization of nNOS disrupts NO signaling, which reduces s-nitrosylation of the SR channels and contributes to SR Ca2+ leak. Increased cytosolic Ca2+ also actives CAMKII, PKC, and the purinergic signaling cascade, leading to further increase in intracellular Ca2+. Lower NO levels also reduce mitochondrial ATP production leading to increased ROS generation. Increased intracellular ROS, combined with mitochondrial energetics dysregulation and the high intracellular Ca2+, induce inflammatory, apoptotic, and necrotic pathway activation. The CamKII = calcium/calmodulin-dependent protein kinase II; cyt c = cytochrome c; IP3 = inositol triphosphate receptor; LTCC = L-type calcium channel; MCU = mitochondrial Ca2+ uniporter; mNCX = mitochondrial Na+-Ca2+ exchanger; NCX = Na+-Ca2+ exchanger; nNOS = neuronal nitric oxide synthase; NF-kB = nuclear factor kappa-light-chain-enhancer of activated B cells; NOX-2 = NADPH oxidase 2; P2X7 = P2X7 purinergic receptor; PKC = protein kinase C; PLC = phospholipase C; PLN = phospholamban; Px = pannexin channels; ROS = reactive oxygen species; RYR2 = ryanodine receptor type 2; SAC = stretch-activated channels; SERCA2 = sarco/endoplasmic reticulum Ca2+-ATPase 2; SR = sarcoplasmic reticulum; VGCC = voltage-gated Ca2+ channels.

With advances in respiratory and other therapies, the leading cause of death in the current era for DMD patients is cardiovascular disease (Central Illustration).2,5,6 DMD is associated with dilated cardiomyopathy and rhythm abnormalities, mainly supraventricular arrhythmias. The dilated cardiomyopathy seen in these patients is characterized by widespread fibrosis of the left ventricular (LV) free wall.7 Heart failure (HF) and arrhythmias will eventually develop as the disease progresses. Therefore, early diagnosis and management of cardiovascular disease is critical for the survival and/or improved quality of life for these patients. In addition, accumulating evidence over the last 2 decades has indicated that female carriers of DMD mutations are also at increased risk of developing cardiac disease.8, 9, 10

Central Illustration.

Central Illustration

Cardiomyopathy in Duchenne Muscular Dystrophy: Potential Therapeutic Targets

Duchenne muscular dystrophy (DMD) has several systemic effects, including cardiomyopathy. DMD cardiomyopathy is characterized by cardiac fibrosis, arrhythmias, and heart failure. Inflammatory modulation and mitochondrial regulation could reduce cardiac fibrosis associated with DMD. In addition, gap junction regulation and therapy with antiarrhythmic agents could reduce incidence of arrhythmia in DMD. Furthermore, gene therapy and neurohormonal modulation could be beneficial in reducing heart failure in DMD. BDNF = brain-derived neurotrophic factor; Cas9 = CRISPR associated protein 9; CRISPR = clustered regularly interspaced short palindromic repeats; DHA = docosahexaenoic acid; EPA = eicosapentaenoic acid; Lox = lysyl oxidase; NLRP3 = NOD-LRR-and pyrin domain-containing protein3; Nox4 = NADPH oxidase 4; Nrf2 = nuclear factor-erythroid factor 2-related factor; PARKIN = E3 ubiquitin ligase; PINK1 = PTEN-induced kinase; PTX3 = pentraxin 3; TrkB = tropomyosin receptor kinase B.

DMD is a devastating disease with early fatality. Current treatment options for cardiac management are aimed primarily at delaying development and progression of HF. Despite extensive research, there remains no cure. In this report, we review emerging potential therapeutic targets to reduce or prevent cardiac dysfunction in DMD (Table 1).

Table 1.

Current Experimental Investigations for DMD Cardiomyopathy

Drug/Class Target/Mechanism Stage of Development Advantages Disadvantages
Exon skipping gene therapy
 Casimersen DMD exon 45 FDA approved under accelerated review Specifically targets the causative defect, produces at least somewhat functional dystrophins Require regular infusions, only available for specific mutations, cardiac benefits unclear
 Eteplirsen DMD exon 51 FDA approved
 Golodirsen DMD exon 53 FDA approved under accelerated review
 Vitolarsen DMD exon 53 FDA approved
Nonsense mutation suppression
 Ataluren Release factor inhibition Approved in EU, orphan status with FDA Specifically targets the causative defect, produces at least somewhat functional dystrophin Only effective for patients with a relatively small subset of DMD mutation types (nonsense)
 G418 sulfate Binds 80s ribosome, increased near-cognate tRNA mispairing Preclinical for DMD
 Direct CRISP/Cas9 editing Specific mutation correction Preclinical for DMD Potentially curative May result in permanent side effects
 KT5720 Selective PKA inhibition Preclinical for DMD
 Tranilast TRPV2 inhibition Phase 1 and 2 clinical investigation Side-effect profile known through its use as antiallergic medication
 Gap19 Cx43 hemichannel-specific inhibition Preclinical for DMD
 Gap26 Cx43 gap junction channel-specific inhibition Preclinical for DMD
 Probenecid Px channel inhibition, TRPV2 agonist, inhibits renal tubular urate resorption FDA approved for gout, Phase 2 investigation for HF Well-established safety profile Several potential mechanisms, may be less effective than more specific agents
 Aldosterone inhibitors Aldosterone inhibition, Px channel inhibition (?) Phase 3 clinical investigation, spironolactone FDA approved for HF Evidence of improvement in subclinical HF in DMD population Mild diuretic effect, risk of hyperkalemia and gynecomastia
 ACEI inhibitors Inhibit Ang II formation and bradykinin metabolism FDA approved for HF Risk of angioedema and chronic cough, hypotension and hyperkalemia
 Angiotensin receptor blockers Competitive inhibition of Ang II binding to the angiotensin 1 receptor FDA approved for HF Less angioedema and cough than ACE inhibitors Potentially increased risk of hypotension and hyperkalemia compared to ACE inhibitors
 β-blockers Nonselective or selective inhibition of β adrenergic receptors FDA approved for HF and arrhythmia Risk of hypotension and bradycardia
 Sacubitril Neprilysin inhibition FDA approved for HF May be superior to ACE inhibitors in reducing risk hospitalization and death in symptomatic HF Incidence of hypotension and hyperkalemia may be more common than with ACE inhibitors
 Eicosapentaenoic acid, docosahexaenoic acid Inflammatory pathway inhibition FDA approved for risk reduction in major cardiovascular events Minimal side effect profile, may improve lipid profile in patients with concomitant dyslipidemia
 Zidovudine Reverse transcriptase inhibition, P2X7 receptor antagonism FDA approved for HIV, preclinical for DMD/HF Reports of cardiomyopathy and myopathy (particularly at higher doses), class 2B carcinogenic risk
 Ivabradine If inhibition FDA approved for HF Improves outcomes in symptomatic HF with reduced LVEF and persistent heart rate ≥70 beats/min, HR reduction with low risk of hypotension Risk of bradycardia and/or atrial fibrillation
 Sulforaphane Nrf2-mediated TGF-β/Smad signaling, NLRP3 inhibition (?) Preclinical for DMD, Phase 1 for other indications

ACE = angiotensin-converting enzyme; DMD = Duchenne muscular dystrophy; EU = European Union; FDA = Food and Drug Administration; HF = heart failure; LVEF = left ventricular ejection fraction.

Noncardiac Therapeutic Strategies

Over the past several decades, several therapeutic advances have led to improved survival and quality of life for DMD patients. Long-term corticosteroids have been the mainstay of medical therapy over the past several decades. There is clear benefit to skeletal muscle function with early glucocorticoid use, depending on the type and frequency of dosing, evidenced by delay of loss of ambulation by an average of 1 to 3 years.11 The cardiac benefits of chronic steroid use are less well-established. Acute glucocorticoid administration activates endothelial nitric oxide synthase and has potent anti-inflammatory and vasodilatory effects.12,13 Chronic use in DMD patients, particularly with deflazacort, has been associated with longer preservation of LV systolic function.14 However, chronic steroid use also can increase the risk of obesity, dyslipidemia, hypertension, adrenal insufficiency, and osteopenia.15,16 There is also some evidence that pulsed corticosteroid regimens maintain many of the benefits of chronic use with less effects on bone density or adrenal function.17

Nonpharmacologic therapies have also improved morbidity and mortality for DMD patients. Impaired airway clearance increases risk for pneumonia and other respiratory infections that may accelerate the need for invasive ventilatory support. Cough assist devices, usually initiated when peak cough expiratory flow is <160 to 240 L/min, help to improve airway secretion clearance and delay initiation of invasive ventilation.18 Nocturnal noninvasive ventilatory (NIV) strategies, such as bilevel positive airway pressure ventilation, have had a tremendous effect on the care of this population. NIV improves survival compared with hypoventilatory DMD patients (RR: 0.62, 95% CI: 0.42 to 0.91; P = 0.01) and may delay need for chronic invasive ventilatory support.19 There may also be cardioprotective effects of NIV, with 1 study of DMD patients on long-term NIV (5 years), showing similar rates of cardiac events (acute HF, cardiac arrhythmia, and ischemic stroke) to patients on invasive ventilation and no difference between those with or without angiotensin-converting enzyme (ACE) inhibitor treatment.20 Scoliosis occurs in up to 90% of untreated DMD boys as they become nonambulatory due to muscle weakness and pelvic imbalance.21 As scoliosis progresses, geometric changes in the thoracic cage can compromise cardiopulmonary function, including reducing lung capacity, increased risk of pulmonary infection, and direct cardiac impingement.21 Early spinal stabilization can reduce the incidence of surgical complications; however, the long-term benefits on cardiopulmonary function have been mixed.22,23

Cardiac Characteristics of Mouse Models of DMD

Many of the preclinical studies discussed in this review have been performed in murine models of DMD. Although several mouse models have been developed, none of the current models has been able to completely imitate the natural history of disease progression seen in humans (Table 2). Cardiac phenotypes can vary substantially, and care should be taken when selecting a model or interpreting results depending on the variables being assessed in the study. The original murine DMD model, C57BL/10ScSn-Dmdmdx/J (henceforth mdx), does not accurately recapitulate the cardiac phenotype seen in human DMD patients with fibrosis and dysfunction occurring very late, if at all.24 The inbred D2.B10-Dmdmdx/J (D2mdx) strain has an accelerated cardiac phenotype with early fibrosis and mild-to-moderate dysfunction by echocardiography typically peaking at approximately 6 months.25,26 However, there is evidence that there is some recovery that occurs in older animals as well as an increase in fibrosis seen in older wild-type (DBA/2J) control animals,27 making it potentially less reliable for longer-term studies of cardiac function. Dystrophin/telomerase RNA double deficient (mdx/mTR) models have a severe cardiac phenotype including fibrosis, mitochondrial fragmentation, and development of dilated cardiomyopathy.28,29 Unlike most other murine models, and similar to human DMD patients, early death from HF is common.

Table 2.

Cardiac and Noncardiac Phenotypes of Established DMD Mouse Models

First Author, Year, Ref. # Model Mutation Background Strain Lifespan Histopathologic Changes (Onset) Cardiac Dysfunction (Onset) Cardiac Phenotype Comments Other Comments
Dystrophin-deficient models
Bulfield et al, 198424  Mdx Exon 23 point mutation C57BL/10 2 y Mild (≥10 mo) Mild/none (≥10 mo) Frequent ECG abnormalities, DCM in females and HCM in males Most widely used model, available through Jackson Labs (C57BL/10ScSn-Dmdmdx/J, stock #001801)
Krivov et al, 2009130  Albino Mdx Exon 23 point mutation Albino 2 y Mild (≥10 mo) Mild/none (≥10 mo) Same as mdx
Schmidt el al, 2011131  Mdx/BALB/c Exon 23 point mutation BALB/c
Duan et al, unpublished data  Mdx/BL6 Exon 23 point mutation C57BL/6
Schmidt el al, 2011131  Mdx/C3H Exon 23 point mutation C3H
Fukada et al, 201025  Mdx/DBA2 Exon 23 point mutation DBA2 1.5-2 y Severe (≥8 wk) Mild/moderate (≥10 wk) Frequent ECG abnormalities, normalization of fractional shortening reported at 1 year More severe dystrophic phenotype (polymorphism in LTBP4 gene), increased fibrosis and fat accumulation, calcifications seen in both dystrophic and wild type strains, available through Jackson Labs (D2.B10-Dmdmdx/J, stock #013141)
Wasala et al, 2015132  Mdx/FVB Exon 23 point mutation FVB
Chapman et al, 1989133  Mdx2cv Intron 42 point mutation C57BL/6 2 y Mild None Chemically induced mutation, fewer revertant fibers, available through Jackson Labs (B6Ros.Cg-Dmdmdx-2Cv/J, stock #002388)
Chapman et al, 1989133  Mdx3cv Intron 65 point mutation C57BL/6 2 y Chemically induced mutation, full-length dystrophin expressed at ∼5% wild-type levels with all other isoforms eliminated, available through Jackson Labs (B6Ros.Cg-Dmdmdx-3Cv/J)
Chapman et al, 1989133  Mdx4cv Exon 53 point mutation C57BL/6 2 y Chemically induced mutation, fewer revertant fibers, available through Jackson Labs (B6Ros.Cg-Dmdmdx-4Cv/J, stock #002378)
Chapman et al, 1989134  Mdx5cv Exon 10 point mutation C57BL/6 2 y None None No significant cardiac phenotype observed Chemically induced mutation, more severe skeletal muscle disease, available through Jackson Labs (B6Ros.Cg-Dmdmdx-5Cv/J, stock #002379)
Araki et al, 1997134  Mdx52 Exon 52 deletion C57BL/6 2 y Targeted inactivation of hotspot (between exons 45-55), fewer revertant fibers
Wertz and Füchtbauer, 1998136  Mdx βgeo Insertion of gene trap vector (ROSAβgeo) in exon 63 along with LacZ reporter C57BL/10 2 y Mild (≥10 mo) Mild/none (≥10 mo) All dystrophin isoforms affected, LacZ reporter replaces CR and CT domains
Kudoh et al, 2005137  DMD-null Cre-loxP mediated deletion of entire DMD gene 2 y None None No significant cardiac phenotype observed No revertant fibers
Double knockout models
Guo et al, 2006137  mdx/α7−/− α7-Integrin/dystrophin double deficient mdx ≤4 wk Mild (≥20 days) None Ultrastructural changes seen by electron microscopy including necrosis and cardiomyocyte and mitochondrial disarray
Deconinck et al, 1997138  Mdx/Utr−/− (Deconinck strain) Utrophin/dystrophin double deficient mdx 20 wk Moderate (≥8 wk) Moderate (≥8 wk) Cardiomyocyte fragility and necrosis early then fibrosis, LV dilation, and reduced functional parameters late, frequent ECG abnormalities Largest utrophin isoform is inactivated by targeted mutation at utrophin exon 7 (other isoforms are active), severe dystrophic phenotype, available through Jackson Labs (Utrntm1Ked/Dmdmdx/J, stock #014563)
Grady et al, 1997139  Mdx/Utr−/− (Grady strain) Utrophin/dystrophin double deficient mdx 20 wk Moderate (≥8 wk) Moderate (≥8 wk) Cardiomyocyte fragility and necrosis early then fibrosis, LV dilation, and reduced functional parameters late, frequent ECG abnormalities All utrophin isoforms are inactivated by targeted mutation at utrophin CR domain, severe dystrophic phenotype, available through Jackson Labs (stock #016622)
Megeney et al, 1996140  Mdx/Myod1 MyoD/dystrophin double deficient 12 mo Severe (≥5 mo) Mild/moderate (≥6 mo) DCM occurs after 5-6 mo, fibrosis occurs by 10 mo, epicardial involvement of LV similar to human DMD cardiomyopathy Severe dystrophic phenotype, MyoD only expressed in skeletal muscle
Chandrasekharan et al, 2010141  Mdx/Cmah Cmah/dystrophin double deficient 11 mo Moderate/severe (≥3 mo) None Cardiomyocyte necrosis early, no overt DCM Humanized model of cytidine monophosphate-N-acetylneuraminic acid hydroxylase-like protein deletion, severe dystrophic phenotype, available from the Jackson Laboratory (stock #017929)
Sacco et al, 201028  Mdx/mTRG2 Telomerase RNA/dystrophin double deficient mdx/BL6 4-18 mo Severe (≥32 wk) Severe (≥32 wk) DCM occurs by 8 mo Severe dystrophic phenotype, available through Jackson Labs (stock #018915).
Sacco et al, 201028, Mourkioti et al, 201329  Mdx4cv/mTRG2 Telomerase RNA/dystrophin double deficient mdx4cv 4-18 mo Severe (≥32 wk) Severe (≥32 wk) DCM occurs by 8 mo Severe dystrophic phenotype, available through Jackson Labs (stock #023535)
Grady et al, 1999142  Mdx/Dtna−/− α-Dystrobrevin/dystrophin double deficient 8-10 mo Moderate/severe (≥4 wk) none No overt dilation/hypertrophy but increased nuclear cell infiltration and necrosis, increased susceptibility to stress-induced injury Pronounced skeletal muscle phenotype but less severe than mdx/Utr−/−, available through Jackson Labs (B6.Cg-Terctm1Rdp Dmdmdx-4Cv/BlauJ, stock #023535)
Li et al, 2009143  Mdx/Sgcd−/− δ-Sarcoglycan/dystrophin double deficient mdx/BL6 10-14 mo Moderate/severe (≥8 wk) Frequent ECG abnormalities, DCM ≥8 wk, increased risk of spontaneous death at 6 mo Severe phenotype, knockdown-targeted replacement of Sgcd exon 2 leading to loss of whole sarcoglycan complex and sarcospan

DCM = dilated cardiomyopathy; ECG = electrocardiogram; HCM = hypertrophic cardiomyopathy; LV = left ventricular.

Gene Therapy

An ideal, potentially curative therapy for DMD in patients would be to correct the genetic defect in the affected cells, leading to functional dystrophin protein and preventing or greatly mitigating symptomatic involvement. However, there are currently several hurdles to achieving this goal. First, dystrophin is the largest known human gene, with 79 exons and roughly 2,200 kb, which makes packaging the gene into traditional viral vectors challenging. Additionally, the size of the gene also leads to hundreds of documented mutations of various types, including deletions, duplications, and missense mutations.30,31 DMD typically results from a deletion of more than 1 exon causing a premature stop codon, which in turn leads to a lack of dystrophin.32 Because cardiac dysfunction associated with DMD is quite variable, studies have investigated whether the underlying genetic mutations that cause DMD could predict the severity of cardiac dysfunction in DMD patients. Studies have suggested that dystrophin gene mutations in exons 48-49 have been associated with earlier onset of cardiomyopathy whereas mutations in exons 51-52 could be cardioprotective.33,34 However, the genotype/phenotype correlation is also complicated by emerging evidence that polymorphic variation in other genes, such as β2-adrenergic receptors,35 secreted phosphoprotein 1,36 and brain-derived neurotrophic factor (BDNF),37 may affect functional, respiratory, and cardiac outcomes. Timing of initiation of therapy is also an important consideration, because the primary benefit is in preventing functional decline and not with recovery of lost functional parameters, which may limit the utility in older patients. Despite these hurdles, there are several promising gene therapy strategies that have received approval for use in patients.

Exon skipping

Exon skipping involves using specific synthetic antisense oligonucleotides vector-targeted to skip out-of-frame mutations at the level of the messenger RNA. This results in re-establishment of the correct reading frame and the generation of functional micro- or mini-dystrophins. An estimated 80% of all DMD mutations are potentially amenable to exon skipping. It is important to note that these truncated dystrophins are still abnormal proteins, and functional dystrophin expression in treated patients is estimated at approximately 0.3% of normal levels, and as such, it is not necessarily a curative therapy.38, 39, 40 The most common regions in the DMD gene for mutations to occur are between exons 45-55, with the deletion of exon 50 being most frequent.41 Clinically, there are currently 4 exon skipping therapies approved for human use: eteplirsen (exon 51), casimersen (exon 45), golodirsen (exon 53), and viltolarsen (exon 53). Approximately 30% of DMD patients have mutations that would be amenable to treatment with 1 of these drugs. Although initial data regarding skeletal muscle function are encouraging for these therapies, the cardiac benefits require ongoing evaluation. To this end, it is not a foregone conclusion that skeletal muscle benefit will extend to cardiac muscle. Preclinical studies in DMD mouse models have had mixed results in terms of expression of microdystrophins in cardiac tissue.42,43

Nonsense mutation suppression

Up to 25% of DMD patients have mutations resulting in premature stop codons. DMD mutations that result in premature stop codons may be targeted using small molecule nonsense suppressors, also known as translational read-through–inducing drugs, which attempt to bypass the premature stop codon and produce functional dystrophin. The 2 most well-characterized translational read-through–inducing drugs are ataluren (currently approved for use in DMD patients in Europe) and the aminoglycoside G418. Ataluren acts by inhibiting release factor activity and thus preventing the termination of translation when a stop codon is encountered.44 By contrast, G418 binds tightly to the ribosome and increases functional near-cognate tRNA (single nucleotide) mispairing with the premature stop codon, resulting in continuation of translation past the stop codon.44 The long-term cardiovascular effects of these medications remain unexplored, however, and further data will be necessary as their use becomes more widespread.

CRISPR/Cas9

Clustered regularly interspaced short palindromic repeats/CRISPR-associated 9 (CRISPR/Cas9) has emerged as a powerful genome editing tool that relies on delivery of ribonucleoprotein complexes (RNPs). There have been reported delivery options into cells such as DNA nanoclews,45 cationic lipid nanoparticles (LNPs),46, 47, 48 lipoplexes, gold-based nanoparticles,49, 50, 51 and zeolitic imidazole frameworks.52 Despite the various approaches there are still numerous problems that present themselves such as controlling the size, uniformity, and stability of the formulations, limiting in vivo application of CRISPR-Cas9 technology. One study was completed to further investigate a more applicable delivery system of CRISPR-Cas9.53 The investigators successfully developed an approach designed to preserve RNP integrity through inclusion of a permanently cationic lipid in ionized LNP formulation. This was applicable to different classes of LNPs such as dendrimer LNPs, stable nucleic acid lipid particles, and lipid-like nanoparticles. Furthermore, they successfully demonstrated that modified dendrimer LNPs delivered RNPs to restore dystrophin expression in DMD mice with a deletion of exon 44. In this study, mice were injected with LNPs into the tibialis anterior muscles weekly (3 injections, 1 mg/kg single guide RNA [sgRNA]) and detected the expression of dystrophin protein in tibialis anterior muscles 3 weeks after the last injection. This was determined through immunofluorescence and Western blot analysis.53

The use of CRISPR/Cas9 has the promise to potentially correct DMD mutations, thereby serving as a potential for curative therapeutic strategies. One preclinical study generated a mouse model using CRISPR/Cas9 that resulted in a deletion of exon 50.41 These mice displayed severe muscle dysfunction including a depletion of dystrophin in the cardiac muscle tissue. Through the use of CRISPR/Cas9, the authors used a sgRNA that created reading-frame mutations that allowed for skipping of exon 51. The results from this study showed a significant recovery of dystrophin in the cardiac tissue, establishing a new mouse model of DMD.41 Additionally, findings from this study demonstrate that systemic delivery of AAV9 encoding a sgRNA directed toward exon 51 recovered the expression of dystrophin thereby preventing the onset of DMD.41 Recently, a 2-virus system with separate AAV9 constructs for the sgRNA and Cas9 (associated with a creatine kinase cassette to target muscle cells) was successful in repairing the exon 50 deletion in the deltaE50-MD canine model.54 The promise of gene editing with a CRISPR/Cas9 approach is that the result would be permanent and would not require periodic treatments as with the other approaches mentioned in the preceding text. However, this permanency is also a reason for caution, because it cannot easily be reversed should adverse side effects become apparent in the long term. In addition, toxicity has been observed with systems such as AAV9-mediated gene delivery. An article recently published discusses the severe toxicity observed in nonhuman primates and piglets that received high-dose intravenous administration of adeno-associated virus vectors.55 Examples include systemic and sensory neuron toxicity, with hepatic toxicity potentially manifesting as disseminated intravascular coagulation and liver necrosis.

Inflammatory Modulators

Cardiac dysfunction in DMD has been associated with up-regulation of inflammatory pathways,56 which in turn likely contribute to increased cardiac fibrosis.57 Numerous studies have therefore examined the roles of inflammatory modulators in cardiac dysfunction in DMD to help identify potential therapeutic targets.

Pentraxin 3

Given the inflammatory processes occurring in DMD, numerous studies have further examined the roles of inflammatory modulators as potential targets for therapies. The protein pentraxin 3 (PTX3) is an inflammatory mediator that plays a critical role in the inflammatory process by contributing to activation of the complement pathway and coordinating macrophage function.58 PTX3 is induced by myeloid differentiation primary response protein 88 (MYD88) in cardiac tissue, leading to downstream effects on critical inflammatory pathways involving Toll-like receptors and interleukins.59 PTX3 has been found to be present in high concentrations in DMD animal models60 and overexpressed in cardiac tissue of mdx mice in an age-dependent manner.61 Results from the latter study demonstrated that dystrophic expression of PTX3 was correlated with inflammatory and fibrotic pathways in cardiac tissue in mdx mice (C57BL6/10ScSn-DMDmdx/J).61 Expression of PTX3 was slightly up-regulated in 9 month old mice and significantly increased as the mice approached 18 months of age. Results were determined through reverse transcription-quantitative polymerase chain reaction (RT-qPCR), immunohistochemistry, and Western blot analysis. These findings suggest that PTX3 could serve as potentially a prognostic marker, as well as a target for cardiac therapies in DMD patients.

Polyunsaturated fatty acids

Eicosapentaenoic acid and docosahexaenoic acid are polyunsaturated fatty acids that have anti-inflammatory effects and have shown benefit in improving inflammatory processes in cardiac disease and rheumatoid arthritis.62 One study further investigated the involvement of free fatty acid (FFA) receptors in the anti-inflammatory role of eicosapentaenoic and docosahexaenoic acid in dystrophic muscles.63 Mdx mice (C57BL/10-Dmdmdx/PasUnib) were treated with fish oil capsules alone or with FFA1 or FFA4 blockers. Following 1 month of treatment, results showed that in the group receiving the blockers of FFA1 and FFA4, the anti-inflammatory effects of fish oil were inhibited due to a significant increase in the proinflammatory markers tumor necrosis factor-α (TNF-α) and interleukin-1β (IL-1β), which was determined through Western blot analysis. These findings indicate that the anti-inflammatory effects of fish oil in the dystrophic heart are mediated through FFA1 and FFA4, offering a mechanistic view of omega-3 in DMD. Furthermore, these results suggest that modulation of key inflammatory markers such as TNF-α and IL-1β could be potential therapeutic targets to improve cardiac function in DMD.

Statins

Statins are well known to be beneficial in cardiovascular disease such as lowering cholesterol, improved endothelial function, increase stability of atherosclerotic plaques, and reducing inflammation. However, data in DMD models thus far have been mixed. One study examined whether simvastatin could halt or rescue cardiac function in male dystrophin-deficient (mdx) mouse model. Results from this study determined that simvastatin administration significantly improves LV diastolic and systolic function and prevents cardiac fibrosis based on echocardiography, electrocardiography, and Western blot analysis.64 In this study, simvastatin was administered at 40 mg/kg or 80 mg/kg for short-term (8 weeks) or long-term (12 months) study intervals. Another study was completed to further investigate the effect of rosuvastatin and its potential effects in both male and female mdx mice (C57BL/10-DMDmdx/PasUnib).65 Rosuvastatin was administered for 30 days beginning at the time of weaning, before muscle degeneration had occurred. Results of histopathological and morphometric analysis, immunofluorescence, and Western blot analysis showed no changes in serum creatine kinase or in markers of inflammation such as TNF-α, NF-kB, or 4-HNE in mdx cardiac tissue. In addition, there was no significant reduction in cardiac fibrosis in mdx mice treated with rosuvastatin.65 Taken together, these results suggest that rosuvastatin may not have the beneficial impact on dystrophy seen in mdx mice. Additional studies are necessary to better delineate the specific role of statins on cardiac function in DMD. Interestingly, statins also have a common adverse side effect of drug-induced myopathy.66 The risk of myopathy or rhabdomyolysis is particularly concerning in patients with underlying muscle disease such as DMD, and thus statin use in this population is not routine.

Adiponectin

Adiponectin (ApN), a hormone physiologically secreted by adipocytes, is an inflammatory regulator in a variety of muscle tissues, playing a role in the NLRP3 inflammasome pathway.67,68 NLRP3 is a highly characterized inflammasome pathway, and its specific involvement in the pathogenesis and progression of DMD continues to be investigated.69,70 Results from 1 study using NLRP3-KO, mdx, and mdx/NLRP3-KO mouse models determined that NLRP3 was expressed in skeletal muscle, and that ApN down-regulates NLRP3 expression through the inflammatory mediator, microRNA (miR)-711.71 These findings confirm that the NLRP3 inflammasome plays a key role in DMD, and ApN-mediated down-regulation of the NLRP3 inflammasome could serve as a potential therapeutic target for inhibiting the inflammatory process occurring in DMD.

Ghrelin

Ghrelin, a hormone produced by the stomach, plays a key role in regulating appetite and growth hormone release. Ghrelin also exerts anti-inflammatory effects in numerous inflammatory states including cardiovascular disease; however, its effects in DMD are not fully understood.72, 73, 74 One study investigated the effect and potential mechanism of ghrelin on muscle morphology and function in male mdx mice (C57BL/10ScSn-Dmdmdx/NJU).75 Mdx mice were injected with ghrelin (100 μg/kg) or saline for 4 weeks. Results determined that ghrelin significantly improved motor performance and decreased inflammatory cell infiltration. Additionally, ghrelin administration inhibited NLRP3 inflammasome activation, partly through suppression of JAK2-STAT3 and p38 mitogen-activated protein kinase (MAPK) pathway. Results were determined through histologic analysis, immunohistochemistry, immunofluorescence, grip strength test, and RT-qPCR. These findings suggest the ghrelin could serve as a potential therapy to decrease muscle inflammation occurring in DMD, slowing disease progression and decreasing symptoms.

Purinergic receptors

Purinergic receptors have been implicated in inflammation and fibroproliferation in different tissues, including the heart.76,77 In pressure overload–induced fibrosis in murine models, the G-protein coupled P2Y6 receptor activation by extracellular purine stimulation leads to cardiac fibrosis.78,79 In mdx mouse models of DMD, increased purine metabolism has been noted with involvement of G-protein coupled (P2Y2) and non–G-protein coupled (P2X7) purinergic receptors.80 P2X7 contributes to up-regulation of IL-1β and NLRP3 inflammasome activation.81 Activation of P2X7 pathways in DMD result in muscle cell damage and death along with increase inflammation.82,83 One study was completed in male mdx mice to determine the outcome of suppressing P2X7 through a known allosteric inhibitor, zidovudine.81 Results indicated that administration of 4 weeks of zidovudine led to a significant decrease in CD68 macrophage markers, as well as a significant decrease in TNF-α and inducible nitric oxide synthase (iNOS) transcript levels in murine cardiac tissue. In addition, although not statistically significant, it was also observed that the myocardial area affected by increased inflammation was reduced with zidovudine administration. Results were determined through histochemical analysis, immunofluorescence, Western blot analysis, and RT-qPCR. These findings suggest that P2X7 could be a potential therapeutic target for DMD cardiomyopathy. Interestingly, purinergic receptor activation may be tied to ATP release through pannexin (Px) channels, which are found in close proximity within the plasma membrane and may represent an important mechanism for crosstalk between cardiomyocytes and inflammatory cells.76

Proteoglycans

Proteomic studies have shown the correlation of the deficiency in dystrophin and the secondary changes that occur in cardiac tissue, including a recent study that was the first to successfully identify members of the cardiac dystrophin-glycoprotein complex through whole-tissue proteomics in mdx-4c mice.84 Results from this study identified changes in various proteins such as the extracellular matrix protein laminin, the Ca2+ binding protein sarcalumenin, the matricellular protein periostin, the proteoglycans asporin and lumican, the cardiac-specific myosin light chain kinase, heat shock proteins, and a large number of mitochondrial and glycolytic enzymes. Fibrotic changes were observed through the presence of the matricellular protein, periostin, and other extracellular matrix proteins, such as the proteoglycans lumican and asporin, which are markers of reactive myofibrosis. These results could potentially contribute to the development of future therapeutic strategies targeted toward the progressive fibrosis that occurs in DMD.

Redox/Mitochondrial Targets

Mitochondrial dysfunction has been identified in mouse models of DMD specifically in cardiac, diaphragmatic, and skeletal muscle, although the specific mechanisms remain unclear.85, 86, 87, 88, 89 It is believed that microtubules regulate mitochondrial bioenergetics through binding of the outer mitochondrial membrane voltage-dependent anion channel and influencing ADP/ATP cycling. The mechanism of action in cardiac dysfunction in DMD remains under investigation. One study examined mitochondrial bioenergetics involved in cardiac function focusing on the ability of creatine to facilitate mitochondrial-cytoplasmic phosphate shuttling and LV mitochondrial responsiveness to ADP in the D2-mdx/2J mouse model of DMD.85 At 4 weeks of age, impairments in ADP-stimulated respiration and ADP attenuation of H2O2 emission were noted. The ability of creatine to increase ADP’s control of mitochondrial bioenergetics was also decreased, whereas mitochondrial H2O2 emission was elevated.85 By using echocardiography, Western blot analysis, mitochondrial respiration, H2O2 emission, and calcium retention capacity of mitochondria, findings showed that selective mitochondria dysfunction precedes the onset of cardiomyopathy in DMD mice. These findings indicate mitochondrial bioenergetic modulators, including ADP and creatine phosphate shuttle, could be potential therapeutic targets to improve cardiac function in DMD.

Dystrophic cardiomyopathy is associated with increased oxidative stress and defective intracellular Ca2+ signaling.90, 91, 92 One study aimed to determine whether mitochondrial damage or loss of function were due to impaired autophagic/mitophagic mechanisms in a mouse model of DMD (C57BL/10ScSn-Dmdmdx/J).93 Mitochondrial structure and function were assessed along with autophagy and mitophagy through techniques such as Western blot analysis, RT-PCR, and confocal and electron microscopy. Structural damage of mitochondria, a significant decrease in ATP production, and increased autophagy were observed in mdx mice, but interestingly, mitophagy was suppressed.93 Down-regulation of several proteins in the PINK1/PARKIN pathway of mitophagy were identified, suggesting that reduced mitophagy could be contributing to the worsened mitochondrial impact in DMD. Targeting improved mitophagic mechanisms by modulation of the PINK1/PARKIN pathway could be a potential therapeutic strategy to improve cardiac function in DMD.

Leucine zipper

Nuclear factor erythroid 2-related factor 2 (Nrf2) is a basic leucine zipper that regulates antioxidant pathways to reduce negative effects of inflammation.94 Sulforaphane (SNF) is an activator of Nrf2, protecting against fibrosis in the liver and lung;95 however, its effects on dystrophic cardiac muscle are unknown. Results from 1 study in a mouse model of DMD (C57BL/10ScSn/Dmdmdx/NJU) showed that SNF administration from 3 to 6 months of age significantly decreased cardiac muscle fibrosis through Nrf2-mediated inhibition of TGF-β/Smad signaling, as determined through RT-qPCR, histologic and morphometric analysis, immunohistochemistry, and immunofluorescence.96 Notably, Nrf2 is also known to inhibit activation of the NLRP3 inflammasome in cardiac and other tissue types.97,98 These findings suggest that modulation of Nrf2 is a potential target to promote antifibrotic effects in DMD.

In addition to Nrf2, other genes implicated in the fibrotic process have been studied in DMD, namely Nox4 and Lox. A study in a mouse model of DMD (C57BL/10ScSn-Dmdmdx/J) used echocardiography, histomorphometry, and gene expression analysis to identify overexpression of the profibrotic genes Nox4 and Lox in LV tissue associated with significant LV thickening.99 Interestingly, Lox expression was increased from the 3-month time point, and Nox4 was increased in cardiac tissue, but not skeletal tissue. These findings suggest that Nox4 and Lox may be contributing to the development of cardiac fibrosis and may represent cardiac-specific targets in DMD.

Calcium Channel Targets

Dystrophin is not only important in force transduction, but also serves as an essential scaffolding protein in muscle cells for several signaling proteins near the cell membrane, including sarcoglycans and matrix metalloproteins, among others. As a result of the lack of dystrophin in DMD, the mechanisms of normal calcium handling are disrupted including at the sarcoplasmic reticulum (SR). Similar to other forms of HF, the calcium sensitivity of the ryanodine receptor isoform 2 (RyR2) in the SR is increased in dystrophic cardiomyocytes. However, unlike in other forms of HF, there is a hypersensitivity to excitation–contraction coupling in dystrophic cardiomyocytes instead of a decrease. This creates the possibility that targeting RYR2 may offer a unique benefit in DMD cardiomyopathy.

One such possible target in DMD cardiomyopathy may be protein kinase A (PKA). Genetic or pharmacologic (using the selective PKA inhibitor KT5720) inhibition of PKA-mediated phosphorylation of RyR2 prevents dystrophic cardiomyopathy by reducing SR Ca2+ leak.100

Another calcium channel target is the stretch-sensitive transient receptor potential cation channel, subfamily V, member 2 (TRPV2). Overexpression of TRPV2 has been observed in the sarcolemmal membrane of DMD patients, as well as in the cytoplasmic membrane of mdx mouse skeletal and cardiac cells.101 A recent study examined a small group of DMD patients with advanced HF and treated them with the antiallergy medication tranilast, which has anti-TRPV2 activity in addition to inhibitory effects on cytokine release and NLRP3 inflammasome activation. Tranilast led to reduction in HF biomarkers such as B-type natriuretic peptide, as well as a reduction in TRPV2 cytoplasmic membrane of mononuclear cells. Tranilast also attenuated the increase in circulating levels of miR-208a-5p, a known regulator of cardiac hypertrophy, and miR-223-3p, a marker of heart disease.102

Gap Junction Protein Targets

Gap junctions are collections of intracellular proteins that connect adjacent cells and allow for direct cell-to-cell transfer of ions and small molecules. The family of structurally similar proteins includes connexins (Cxs) and Pxs. Recently, these proteins have been implicated as important disease modulators in animal models of DMD and represent potential therapeutic targets for DMD patients.

Connexins

Cxs are 6-subunit hemichannels that form functional channels after docking end-to-end at the plasma membranes between adjacent cells. The primary isoforms expressed in cardiac tissue are Cx43 (ventricle) and Cx40 (atria), and they have been shown to be stretch-activated.103 Recent studies in dystrophic mice and humans have demonstrated a role for Cx43 in DMD cardiomyopathy.104,105 In these models, Cx43 is up-regulated and becomes lateralized beyond the borders of the cardiomyocyte gap junctions, thus creating the potential for ionic “leak” and triggered arrhythmias. High-dose isoproterenol–induced severe ventricular arrhythmias in mdx and mdx:utr mouse models of DMD were prevented by treatment with Cx43 mimetic peptides targeted to inhibit hemichannel (Gap19) and gap junction channel (Gap26) opening.104 Additionally, genetic mdx models ablating Cx43 (mdxCx−/−)105 or altering the phosphorylation of Cx43 by replacing the serine triplet with phospho-mimicking glutamic acids (mdxS3E)106 resulted in improvement in calcium handling, reduced cardiac fibrosis, less inducible arrhythmia, and overall improved survival. There is also a link between cellular oxidation stress and Cx activity. Treatment of mdx mice with the NADPH oxidase inhibitor apocynin (40 mg/kg/day over 1 month) resulted in decreased lateralization of Cx43, reduction in hyper-S-nitrosylation of Cx43, and an overall reduction in apoptosis in the dystrophic heart.107

Pannexins

Unlike Cxs, Pxs are not isolated to gap junctions and have larger conductance (300-500 pS).108,109 They have been shown to be important in apoptosis signaling through release of ATP through their large pore region. Like Cxs, they are also stimulated by stretch but are additionally activated by elevated intracellular calcium and directly by ATP. Recent studies have shown increased Px expression in mdx mouse skeletal110 and cardiac111 muscle. In isolated mdx hearts, infusion with ATP stimulates ventricular ectopy, which is suppressed by coinfusion with carbenoxolone at concentrations selective for Px channels.111 Recently, the aldosterone inhibitor spironolactone was found to have Px inhibitory properties in endothelial cells,112 allowing for a possible dual action of this pharmacologic class.

Neurohormonal Modulators

Neurohormonal modulators have also been implicated in DMD cardiomyopathy. Specifically, BDNF, aldosterone inhibitors, and sacubitril/valsartan have been studied in the context of DMD cardiomyopathy.

BDNF/tyrosine kinase B

BDNF is a neuronal growth and survival factor, and plasma levels have been shown to correlate with cardiovascular risk in general113 and specifically LV ejection fraction (LVEF) in DMD patients.114,115 In DMD animal models, polymorphisms within the BDNF gene lead to reduced BDNF metabolism into the active form, alteration in cardiac-specific gene expression, and reduced cardiomyocyte contractility.37 BDNF acts primarily by binding to the tyrosine kinase B (TrkB) receptor, leading to autophosphorylation of the receptor and downstream activation of several signaling pathways, including MAPK, the phospholipase C-gamma (PLC-γ), and the phosphatidylinositol 3-kinase (PI3K). Dystrophic mice with genetic ablation of neutral sphingomyelinase 2/sphingomyelin phosphodiesterase 3, important enzymes in the signaling cascades of inflammation and apoptosis, showed recovery of BDNF levels.116 Recently, it has also been demonstrated that acute inhibition of TrkB in wild-type mice leads to reduced heart rate and LVEF,37 further reinforcing the potential therapeutic benefit of this target in DMD cardiomyopathy.

Aldosterone inhibitors

Early mineralocorticoid receptor antagonist therapy with spironolactone in combination with ACE inhibitors has been shown to preserve normal circumferential strain and prevent cardiomyocyte damage in a utrophin haploinsufficient mdx mouse model of DMD,117 although concurrent prednisolone therapy may attenuate these effects.118 Low-dose eplerenone stabilizes left ventricular strain in DMD boys with detectible myocardial damage but preserved ejection fraction.119 Spironolactone appears to be equally effective as eplerenone in DMD patients.120 Concurrent use of aldosterone inhibitors with traditional modulators of cardiac remodeling, including ACE inhibitors/angiotensin receptor blockers (ARBs) or beta-blockade,121 is currently recommended to be started by age 12 in asymptomatic DMD boys or sooner if evidence of late gadolinium enhancement is seen on cardiac magnetic resonance imaging (CMR).

Sacubitril/valsartan

Sacubitril is a prodrug that is metabolized to sacubitrilat, a potent inhibitor of the enzyme neprilysin, which is responsible for the degradation of atrial and brain natriuretic peptides. Combined with an ARB (valsartan), sacubitril has been recommended for patients with acquired symptomatic HF with reduced ejection fraction and has demonstrated partial recovery of LVEF in these patients.122 The PARADIGM-HF (Prospective Comparison of ARNI with ACEI to Determine Impact on Global Mortality and Morbidity in Heart Failure) trial demonstrated a 20% reduction in mortality and a reduction in HF admission with sacubitril/valsartan compared with standard treatment with an ACE inhibitor.123 More recently, clinical experience has been increasing with the use of sacubitril/valsartan in DMD patients. Initial reports suggest that sacubitril/valsartan may improve cardiac remodeling and LVEF in DMD patients compared with ACE inhibitor/ARB alone.124

Antiarrhythmic Agents

There has been growing evidence that antiarrhythmic agents, particularly beta-blockers (BBs), may have a beneficial role in improving cardiac function in DMD. One study examined the effects of adrenergic agonists and BB treatments in DMD patient-specific human induced pluripotent stem cell–derived cardiomyocytes.125 Results from this study determined that BB therapy in vitro and in vivo decreased the incidence of arrhythmogenesis and rescued lethality in mdx mice after beta-adrenergic stimulation.

DMD patients treated with the BB carvedilol over 5 years demonstrated significantly higher survival rate free from primary endpoints, which included all-cause death, advanced HF, and severe arrhythmia, compared with the non-BB group.126 No significant differences in LVEF were identified between the BB group and non-BB group. These results suggest that BBs, specifically carvedilol, may be a safe and effective approach to reduce cardiac events in DMD patients. Despite the benefit seen with carvedilol, not all BBs have similar efficacy in DMD. One study in male mdx mice was completed, testing the effects of the BB metoprolol (2.5 mg/kg/day) on ventricular function and myocardial calcium influx.127 Results from this study concluded that administration of metoprolol at an early stage of cardiomyopathy lead to worsening right ventricular ejection fraction though with no effect on myocardial calcium influx. These results were determined through CMR and in vivo myocardial calcium influx with manganese enhanced CMR. Given the conflicting impact of different BBs on left or right ventricular function, taken together, these findings suggest that more studies are needed to better elucidate the specific effect of BBs on cardiomyopathy in DMD.

Recently, the sinoatrial pacemaker or “funny” current inhibitor, ivabradine, has also been investigated as a possible treatment for end-stage DMD cardiomyopathy. In a study of 20 teenage DMD patients with LVEF <40% and on long-term ACE inhibitor therapy, patients who underwent a heart rate reduction therapy with BB alone or with ivabradine had significantly lower major acute cardiac events compared with those who did not have aggressive rate control.128 Additionally, it has been suggested that ivabradine has an independent effect on cardiac remodeling and thus may have a role in DMD cardiomyopathy treatment in combination with standard therapy.129

Conclusions

DMD is a devastating disease with an ultimately poor prognosis and no cure. In recent years, cardiovascular disease has become the leading cause of death in patients with DMD. Research has identified therapeutic strategies that could help delay the progression of cardiac disease in patients with DMD including genetic manipulation, antiarrhythmic therapy, mitochondrial regulation, and modulation of inflammatory and neurohormonal factors. Gene therapy approaches such as CRISPR/Cas9 technology and exon skipping offer cautious optimism for the development of novel pharmacologic therapies for DMD. Although genetic strategies to correct the dystrophin mutation have shown promising results for skeletal muscles in DMD, their impact on cardiac dysfunction in DMD remains unclear. Furthermore, improved skeletal function in DMD may even cause decline in cardiac function in these patients due to earlier unmasking of underlying cardiac dysfunction facilitated by improved exertional capacity. As gene therapy becomes more widespread, some of these questions will begin to be answered; however, further research is necessary to firmly establish specific therapeutic approaches in DMD cardiomyopathy and to establish guidelines for early implementation of cardioprotective therapies and medications in these patients.

Preclinical evaluation of potential treatments for DMD cardiomyopathy has been limited by the phenotypic variation of currently available animal models. As has been discussed earlier, there is no 1 murine model that ideally mimics the cardiac and skeletal muscle DMD phenotype seen in humans. Experiments in larger animals, such as canine or porcine models, may more faithfully recapitulate the DMD cardiac phenotype; however, their utility is limited by the inability to easily control for genetic background variation, the length of time to maturity, and the sample sizes needed to power studies depending on the effect size of the outcome variables. In addition, ethical and cost concerns make widespread use of these models prohibitive in all but the most select studies. Advances in human pluripotent stem cell technology has made studying cellular mechanisms in human cells more reliable. These systems have the advantage of being able to have human background genetic variations and, through the use of CRISPR/Cas9 systems, the ability to create isogenic control cell lines. Initial assessment of cellular toxicities can potentially be addressed in these systems as well, making it easier to move forward with the most promising therapies in humans. However, such in vitro studies cannot yet address complex interactions beyond monolayers or differential cell–cell interfaces.

There are several impediments to performing cardiac-specific randomized controlled trials in DMD patients. The relative rarity of DMD and the prevalence of established therapies such as glucocorticoids make study design more complicated, particularly for recruiting proper control patients. Additionally, many treatments are justifiably focused on mitigating the skeletal muscle effects of dystrophinopathies; however, until relatively recently, cardiac outcomes were either not evaluated or only evaluated as secondary outcomes. This has led to several multi-institutional efforts to pool outcomes and natural history data that can used as control data and also for increasing the power of studies investigating newer therapies. Consortiums such as the Advanced Cardiac Therapies Improving Outcomes Network (ACTION) and Parent Project Muscular Dystrophy (PPMD) have developed patient registries that collect extensive data on DMD patients, including advance imaging parameters (echocardiographic and CMR), biomarkers, and medication adherence. As they continue to grow, these extensive data networks will allow for more streamlined and consistent evaluation of newer cardiac therapies in this population.

Based on the studies presented here, of particular promise are treatments that modulate the calcium regulation and the ongoing activation of the inflammatory cascade. Across several pharmaceutical classes, preclinical experiments that normalize intracellular calcium have a significant phenotype-mitigating effect. Additionally, better understanding of the mechanisms behind the efficacy of aldosterone inhibitors and combination sacubitril/valsartan in DMD may lead to more targeted therapies. More human trials are needed, however, in order to establish long-term cardiovascular efficacy. Even as newer therapies become more standard for the treatment of skeletal muscle disease, it is likely that cardiac care will continue to be an important factor in monitoring DMD patients. It would not be surprising if cardiac-specific medications are still necessary even as skeletal muscles involvement improves and earlier initiation of cardioprotective therapies may soon become standard of care in this setting.

Funding Support and Author Disclosures

This work was supported by the National Heart, Lung, and Blood Institute of the National Institutes of Health under grants R35HL155651 (Dr Salloum) and K08HL155852 (Dr Raucci). The authors have reported that they have no relationships relevant to the contents of this paper to disclose.

Footnotes

The authors attest they are in compliance with human studies committees and animal welfare regulations of the authors’ institutions and Food and Drug Administration guidelines, including patient consent where appropriate. For more information, visit the Author Center.

References

  • 1.Romitti P.A., Zhu Y., Puzhankara S., et al. Prevalence of Duchenne and Becker muscular dystrophies in the United States. Pediatrics. 2015;135:513–521. doi: 10.1542/peds.2014-2044. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Gartz M., Lin C.W., Sussman M.A., Lawlor M.W., Strande J.L. Duchenne muscular dystrophy (DMD) cardiomyocyte-secreted exosomes promote the pathogenesis of DMD-associated cardiomyopathy. Dis Model Mech. 2020;13(11) doi: 10.1242/dmm.045559. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.MacKintosh E.W., Chen M.L., Benditt J.O. Lifetime care of Duchenne muscular dystrophy. Sleep Med Clin. 2020;15:485–495. doi: 10.1016/j.jsmc.2020.08.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Eiholzer U., Boltshauser E., Frey D., Molinari L., Zachmann M. Short stature: a common feature in Duchenne muscular dystrophy. Eur J Pediatr. 1988;147:602–605. doi: 10.1007/BF00442472. [DOI] [PubMed] [Google Scholar]
  • 5.Cheeran D., Khan S., Khera R., et al. Predictors of death in adults with Duchenne muscular dystrophy-associated cardiomyopathy. J Am Heart Assoc. 2017;6(10) doi: 10.1161/JAHA.117.006340. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Van Ruiten H.J., Marini Bettolo C., Cheetham T., et al. Why are some patients with Duchenne muscular dystrophy dying young: an analysis of causes of death in North East England. Eur J Paediatr Neurol. 2016;20:904–909. doi: 10.1016/j.ejpn.2016.07.020. [DOI] [PubMed] [Google Scholar]
  • 7.Sanyal S.K., Johnson W.W., Thapar M.K., Pitner S.E. An ultrastructural basis for electrocardiographic alterations associated with Duchenne's progressive muscular dystrophy. Circulation. 1978;57:1122–1129. doi: 10.1161/01.cir.57.6.1122. [DOI] [PubMed] [Google Scholar]
  • 8.Schade van Westrum S.M., Hoogerwaard E.M., Dekker L., et al. Cardiac abnormalities in a follow-up study on carriers of Duchenne and Becker muscular dystrophy. Neurology. 2011;77:62–66. doi: 10.1212/WNL.0b013e318221ad14. [DOI] [PubMed] [Google Scholar]
  • 9.Florian A., Rösch S., Bietenbeck M., et al. Cardiac involvement in female Duchenne and Becker muscular dystrophy carriers in comparison to their first-degree male relatives: a comparative cardiovascular magnetic resonance study. Eur Heart J Cardiovasc Imaging. 2016;17:326–333. doi: 10.1093/ehjci/jev161. [DOI] [PubMed] [Google Scholar]
  • 10.Adachi K., Hashiguchi S., Saito M., et al. Detection and management of cardiomyopathy in female dystrophinopathy carriers. J Neurol Sci. 2018;386:74–80. doi: 10.1016/j.jns.2017.12.024. [DOI] [PubMed] [Google Scholar]
  • 11.Escolar D.M., Hache L.P., Clemens P.R., et al. Randomized, blinded trial of weekend vs daily prednisone in Duchenne muscular dystrophy. Neurology. 2011;77:444–452. doi: 10.1212/WNL.0b013e318227b164. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Hafezi-Moghadam A., Simoncini T., Yang Z., et al. Acute cardiovascular protective effects of corticosteroids are mediated by non-transcriptional activation of endothelial nitric oxide synthase. Nat Med. 2002;8:473–479. doi: 10.1038/nm0502-473. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Markham L.W., Spicer R.L., Khoury P.R., Wong B.L., Mathews K.D., Cripe L.H. Steroid therapy and cardiac function in Duchenne muscular dystrophy. Pediatr Cardiol. 2005;26:768–771. doi: 10.1007/s00246-005-0909-4. [DOI] [PubMed] [Google Scholar]
  • 14.Silversides C.K., Webb G.D., Harris V.A., Biggar D.W. Effects of deflazacort on left ventricular function in patients with Duchenne muscular dystrophy. Am J Cardiol. 2003;91:769–772. doi: 10.1016/s0002-9149(02)03429-x. [DOI] [PubMed] [Google Scholar]
  • 15.Bylo M., Farewell R., Coppenrath V.A., Yogaratnam D. A review of deflazacort for patients with Duchenne muscular dystrophy. Ann Pharmacother. 2020;54:788–794. doi: 10.1177/1060028019900500. [DOI] [PubMed] [Google Scholar]
  • 16.Liu D., Ahmet A., Ward L., et al. A practical guide to the monitoring and management of the complications of systemic corticosteroid therapy. Allergy Asthma Clin Immunol. 2013;9:30. doi: 10.1186/1710-1492-9-30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Quattrocelli M., Zelikovich A.S., Jiang Z., et al. Pulsed glucocorticoids enhance dystrophic muscle performance through epigenetic-metabolic reprogramming. JCI Insight. 2019;4(24) doi: 10.1172/jci.insight.132402. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Boitano L.J. Management of airway clearance in neuromuscular disease. RespirCare. 2006;51:913. [PubMed] [Google Scholar]
  • 19.Annane D., Orlikowski D., Chevret S. Nocturnal mechanical ventilation for chronic hypoventilation in patients with neuromuscular and chest wall disorders. Cochrane Database Syst Rev. 2014;2014:Cd001941. doi: 10.1002/14651858.CD001941.pub3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Fayssoil A., Ogna A., Chaffaut C., et al. Natural history of cardiac function in Duchenne and Becker muscular dystrophies on home mechanical ventilation. Medicine (Baltimore) 2018;97 doi: 10.1097/MD.0000000000011381. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Hsu J.D., Quinlivan R. Scoliosis in Duchenne muscular dystrophy (DMD) Neuromuscul Disord. 2013;23(8):611–617. doi: 10.1016/j.nmd.2013.05.003. [DOI] [PubMed] [Google Scholar]
  • 22.Takaso M., Nakazawa T., Imura T., et al. Surgical management of severe scoliosis with high-risk pulmonary dysfunction in Duchenne muscular dystrophy. Int Orthop. 2010;34:401–406. doi: 10.1007/s00264-009-0764-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Van Opstal N., Verlinden C., Myncke J., Goemans N., Moens P. The effect of Luque-Galveston fusion on curve, respiratory function and quality of life in Duchenne muscular dystrophy. Acta Orthop Belg. 2011;77:659–665. [PubMed] [Google Scholar]
  • 24.Bulfield G., Siller W.G., Wight P.A., Moore K.J. X chromosome-linked muscular dystrophy (mdx) in the mouse. Proc Natl Acad Sci U S A. 1984;81:1189–1192. doi: 10.1073/pnas.81.4.1189. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Fukada S., Morikawa D., Yamamoto Y., et al. Genetic background affects properties of satellite cells and mdx phenotypes. Am J Pathol. 2010;176:2414–2424. doi: 10.2353/ajpath.2010.090887. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Coley W.D., Bogdanik L., Vila M.C., et al. Effect of genetic background on the dystrophic phenotype in mdx mice. Hum Mol Genet. 2016;25:130–145. doi: 10.1093/hmg/ddv460. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Hakim C.H., Wasala N.B., Pan X., et al. A five-repeat micro-dystrophin gene ameliorated dystrophic phenotype in the severe DBA/2J-mdx model of Duchenne muscular dystrophy. Mol Ther Methods Clin Dev. 2017;6:216–230. doi: 10.1016/j.omtm.2017.06.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Sacco A., Mourkioti F., Tran R., et al. Short telomeres and stem cell exhaustion model Duchenne muscular dystrophy in mdx/mTR mice. Cell. 2010;143:1059–1071. doi: 10.1016/j.cell.2010.11.039. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Mourkioti F., Kustan J., Kraft P., et al. Role of telomere dysfunction in cardiac failure in Duchenne muscular dystrophy. Nat Cell Biol. 2013;15:895–904. doi: 10.1038/ncb2790. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Neri M., Rossi R., Trabanelli C., et al. The genetic landscape of dystrophin mutations in Italy: a nationwide study. Front Genet. 2020;11:131. doi: 10.3389/fgene.2020.00131. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Flanigan K.M., Dunn D.M., von Niederhausern A., et al. Mutational spectrum of DMD mutations in dystrophinopathy patients: application of modern diagnostic techniques to a large cohort. Hum Mutat. 2009;30:1657–1666. doi: 10.1002/humu.21114. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Ashwath M.L., Jacobs I.B., Crowe C.A., Ashwath R.C., Super D.M., Bahler R.C. Left ventricular dysfunction in duchenne muscular dystrophy and genotype. Am J Cardiol. 2014;114:284–289. doi: 10.1016/j.amjcard.2014.04.038. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Eagle M., Baudouin S.V., Chandler C., Giddings D.R., Bullock R., Bushby K. Survival in Duchenne muscular dystrophy: improvements in life expectancy since 1967 and the impact of home nocturnal ventilation. Neuromuscul Disord. 2002;12:926–929. doi: 10.1016/s0960-8966(02)00140-2. [DOI] [PubMed] [Google Scholar]
  • 34.Kirchmann C., Kececioglu D., Korinthenberg R., Dittrich S. Echocardiographic and electrocardiographic findings of cardiomyopathy in Duchenne and Becker-Kiener muscular dystrophies. Pediatr Cardiol. 2005;26:66–72. doi: 10.1007/s00246-004-0689-2. [DOI] [PubMed] [Google Scholar]
  • 35.Kelley E.F., Cross T.J., Snyder E.M., McDonald C.M., Hoffman E.P., Bello L. Influence of β(2) adrenergic receptor genotype on risk of nocturnal ventilation in patients with Duchenne muscular dystrophy. Respir Res. 2019;20:221. doi: 10.1186/s12931-019-1200-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Pegoraro E., Hoffman E.P., Piva L., et al. SPP1 genotype is a determinant of disease severity in Duchenne muscular dystrophy. Neurology. 2011;76:219–226. doi: 10.1212/WNL.0b013e318207afeb. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Raucci F.J., Jr., Singh A.P., Soslow J., et al. The BDNF rs6265 polymorphism is a modifier of cardiomyocyte contractility and dilated cardiomyopathy. Int J Mol Sci. 2020;21(20):7466. doi: 10.3390/ijms21207466. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Ramos J., Chamberlain J.S. Gene therapy for Duchenne muscular dystrophy. Expert Opin Orphan Drugs. 2015;3:1255–1266. doi: 10.1517/21678707.2015.1088780. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Shin J.H., Pan X., Hakim C.H., et al. Microdystrophin ameliorates muscular dystrophy in the canine model of duchenne muscular dystrophy. Mol Ther. 2013;21:750–757. doi: 10.1038/mt.2012.283. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Yue Y., Pan X., Hakim C.H., et al. Safe and bodywide muscle transduction in young adult Duchenne muscular dystrophy dogs with adeno-associated virus. Hum Mol Genet. 2015;24:5880–5890. doi: 10.1093/hmg/ddv310. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Amoasii L., Long C., Li H., et al. Single-cut genome editing restores dystrophin expression in a new mouse model of muscular dystrophy. Sci Transl Med. 2017;9(418) doi: 10.1126/scitranslmed.aan8081. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Malerba A., Boldrin L., Dickson G. Long-term systemic administration of unconjugated morpholino oligomers for therapeutic expression of dystrophin by exon skipping in skeletal muscle: implications for cardiac muscle integrity. Nucleic Acid Ther. 2011;21:293–298. doi: 10.1089/nat.2011.0306. [DOI] [PubMed] [Google Scholar]
  • 43.Blain A.M., Greally E., McClorey G., et al. Peptide-conjugated phosphodiamidate oligomer-mediated exon skipping has benefits for cardiac function in mdx and Cmah-/-mdx mouse models of Duchenne muscular dystrophy. PloS One. 2018;13 doi: 10.1371/journal.pone.0198897. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Ng M.Y., Li H., Ghelfi M.D., Goldman Y.E., Cooperman B.S. Ataluren and aminoglycosides stimulate read-through of nonsense codons by orthogonal mechanisms. Proc Natl Acad Sci U S A. 2021;118(2) doi: 10.1073/pnas.2020599118. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Sun W., Ji W., Hall J.M., et al. Self-assembled DNA nanoclews for the efficient delivery of CRISPR-Cas9 for genome editing. Angew Chem Int Ed Engl. 2015;54:12029–12033. doi: 10.1002/anie.201506030. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Gao X., Tao Y., Lamas V., et al. Treatment of autosomal dominant hearing loss by in vivo delivery of genome editing agents. Nature. 2018;553:217–221. doi: 10.1038/nature25164. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Wang M., Zuris J.A., Meng F., et al. Efficient delivery of genome-editing proteins using bioreducible lipid nanoparticles. Proc Natl Acad Sci U S A. 2016;113:2868–2873. doi: 10.1073/pnas.1520244113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Zuris J.A., Thompson D.B., Shu Y., et al. Cationic lipid-mediated delivery of proteins enables efficient protein-based genome editing in vitro and in vivo. Nat Biotechnol. 2015;33:73–80. doi: 10.1038/nbt.3081. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Wang P., Zhang L., Xie Y., et al. Genome editing for cancer therapy: delivery of Cas9 protein/sgRNA plasmid via a gold nanocluster/lipid core-shell nanocarrier. Adv Sci (Weinh) 2017;4:1700175. doi: 10.1002/advs.201700175. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Mout R., Ray M., Yesilbag Tonga G., et al. Direct cytosolic delivery of CRISPR/Cas9-ribonucleoprotein for efficient gene editing. ACS Nano. 2017;11:2452–2458. doi: 10.1021/acsnano.6b07600. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Lee K., Conboy M., Park H.M., et al. Nanoparticle delivery of Cas9 ribonucleoprotein and donor DNA in vivo induces homology-directed DNA repair. Nat Biomed Eng. 2017;1:889–901. doi: 10.1038/s41551-017-0137-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Alsaiari S.K., Patil S., Alyami M., et al. Endosomal escape and delivery of CRISPR/Cas9 genome editing machinery enabled by nanoscale zeolitic imidazolate framework. J Am Chem Soc. 2018;140:143–146. doi: 10.1021/jacs.7b11754. [DOI] [PubMed] [Google Scholar]
  • 53.Wei T., Cheng Q., Min Y.L., Olson E.N., Siegwart D.J. Systemic nanoparticle delivery of CRISPR-Cas9 ribonucleoproteins for effective tissue specific genome editing. Nat Commun. 2020;11:3232. doi: 10.1038/s41467-020-17029-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Amoasii L., Hildyard J.C.W., Li H., et al. Gene editing restores dystrophin expression in a canine model of Duchenne muscular dystrophy. Science. 2018;362:86–91. doi: 10.1126/science.aau1549. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Hinderer C., Katz N., Buza E.L., et al. Severe toxicity in nonhuman primates and piglets following high-dose intravenous administration of an adeno-associated virus vector expressing human SMN. Hum Gene Ther. 2018;29:285–298. doi: 10.1089/hum.2018.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Noviello M., Tedesco F.S., Bondanza A., et al. Inflammation converts human mesoangioblasts into targets of alloreactive immune responses: implications for allogeneic cell therapy of DMD. Mol Ther. 2014;22:1342–1352. doi: 10.1038/mt.2014.62. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Silva M.C., Magalhaes T.A., Meira Z.M., et al. Myocardial fibrosis progression in Duchenne and Becker muscular dystrophy: a randomized clinical trial. JAMA Cardiol. 2017;2:190–199. doi: 10.1001/jamacardio.2016.4801. [DOI] [PubMed] [Google Scholar]
  • 58.Garlanda C., Bottazzi B., Bastone A., Mantovani A. Pentraxins at the crossroads between innate immunity, inflammation, matrix deposition, and female fertility. Annu Rev Immunol. 2005;23:337–366. doi: 10.1146/annurev.immunol.23.021704.115756. [DOI] [PubMed] [Google Scholar]
  • 59.Kunes P., Holubcova Z., Kolackova M., Krejsek J. Pentraxin 3 (PTX 3): an endogenous modulator of the inflammatory response. Mediators Inflamm. 2012;2012:920517. doi: 10.1155/2012/920517. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Frohlich T., Kemter E., Flenkenthaler F., et al. Progressive muscle proteome changes in a clinically relevant pig model of Duchenne muscular dystrophy. Sci Rep. 2016;6:33362. doi: 10.1038/srep33362. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Farini A., Villa C., Di Silvestre D., et al. PTX3 predicts myocardial damage and fibrosis in Duchenne muscular dystrophy. Front Physiol. 2020;11:403. doi: 10.3389/fphys.2020.00403. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Simonetto M., Infante M., Sacco R.L., Rundek T., Della-Morte D. A novel anti-inflammatory role of omega-3 PUFAs in prevention and treatment of atherosclerosis and vascular cognitive impairment and dementia. Nutrients. 2019;11 doi: 10.3390/nu11102279. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Maciel Junior M., Camacari de Carvalho S., Saenz Suarez P.A., Santo Neto H., Marques M.J. Fish oil attenuated dystrophic muscle markers of inflammation via FFA1 and FFA4 in the mdx mouse model of DMD. Anat Rec (Hoboken) 2020;304(6):1305–1312. doi: 10.1002/ar.24563. [DOI] [PubMed] [Google Scholar]
  • 64.Kim M.J., Bible K.L., Regnier M., Adams M.E., Froehner S.C., Whitehead N.P. Simvastatin provides long-term improvement of left ventricular function and prevents cardiac fibrosis in muscular dystrophy. Physiol Rep. 2019;7 doi: 10.14814/phy2.14018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Finkler J.M.G., de Carvalho S.C., Santo Neto H., Marques M.J. Cardiac and skeletal muscle changes associated with rosuvastatin therapy in dystrophic mdx mice. Anat Rec (Hoboken) 2020;303:2202–2212. doi: 10.1002/ar.24341. [DOI] [PubMed] [Google Scholar]
  • 66.Allenbach Y., Drouot L., Rigolet A., et al. Anti-HMGCR autoantibodies in European patients with autoimmune necrotizing myopathies: inconstant exposure to statin. Medicine (Baltimore) 2014;93:150–157. doi: 10.1097/MD.0000000000000028. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Luo Y., Liu M. Adiponectin: a versatile player of innate immunity. J Mol Cell Biol. 2016;8:120–128. doi: 10.1093/jmcb/mjw012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Ouchi N., Walsh K. Adiponectin as an anti-inflammatory factor. Clin Chim Acta. 2007;380:24–30. doi: 10.1016/j.cca.2007.01.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Benetti E., Chiazza F., Patel N.S., Collino M. The NLRP3 Inflammasome as a novel player of the intercellular crosstalk in metabolic disorders. Mediators Inflamm. 2013;2013:678627. doi: 10.1155/2013/678627. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Rawat R., Cohen T.V., Ampong B., et al. Inflammasome up-regulation and activation in dysferlin-deficient skeletal muscle. Am J Pathol. 2010;176:2891–2900. doi: 10.2353/ajpath.2010.090058. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Boursereau R., Abou-Samra M., Lecompte S., Noel L., Brichard S.M. Downregulation of the NLRP3 inflammasome by adiponectin rescues Duchenne muscular dystrophy. BMC Biol. 2018;16:33. doi: 10.1186/s12915-018-0501-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Baatar D., Patel K., Taub D.D. The effects of ghrelin on inflammation and the immune system. Mol Cell Endocrinol. 2011;340:44–58. doi: 10.1016/j.mce.2011.04.019. [DOI] [PubMed] [Google Scholar]
  • 73.Huang C.X., Yuan M.J., Huang H., et al. Ghrelin inhibits post-infarct myocardial remodeling and improves cardiac function through anti-inflammation effect. Peptides. 2009;30:2286–2291. doi: 10.1016/j.peptides.2009.09.004. [DOI] [PubMed] [Google Scholar]
  • 74.Sun G.X., Ding R., Li M., et al. Ghrelin attenuates renal fibrosis and inflammation of obstructive nephropathy. J Urol. 2015;193:2107–2115. doi: 10.1016/j.juro.2014.11.098. [DOI] [PubMed] [Google Scholar]
  • 75.Chang L., Niu F., Chen J., et al. Ghrelin improves muscle function in dystrophin-deficient mdx mice by inhibiting NLRP3 inflammasome activation. Life Sci. 2019;232:116654. doi: 10.1016/j.lfs.2019.116654. [DOI] [PubMed] [Google Scholar]
  • 76.Zhou J., Zhou Z., Liu X., Yin H.Y., Tang Y., Cao X. P2X7 receptor-mediated inflammation in cardiovascular disease. Front Pharmacol. 2021;12:654425. doi: 10.3389/fphar.2021.654425. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Tian G., Zhou J., Quan Y., Kong Q., Wu W., Liu X. P2Y1 receptor agonist attenuates cardiac fibroblasts activation triggered by TGF-β1. Front Pharmacol. 2021;12:627773. doi: 10.3389/fphar.2021.627773. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Shimoda K., Nishimura A., Sunggip C., et al. Modulation of P2Y(6)R expression exacerbates pressure overload-induced cardiac remodeling in mice. Sci Rep. 2020;10:13926. doi: 10.1038/s41598-020-70956-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Nishida M., Sato Y., Uemura A., et al. P2Y6 receptor-Galpha12/13 signalling in cardiomyocytes triggers pressure overload-induced cardiac fibrosis. EMBO J. 2008;27:3104–3115. doi: 10.1038/emboj.2008.237. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Róg J., Oksiejuk A., Gosselin M.R.F., et al. Dystrophic mdx mouse myoblasts exhibit elevated ATP/UTP-evoked metabotropic purinergic responses and alterations in calcium signalling. Biochim Biophys Acta Mol Basis Dis. 2019;1865:1138–1151. doi: 10.1016/j.bbadis.2019.01.002. [DOI] [PubMed] [Google Scholar]
  • 81.Al-Khalidi R., Panicucci C., Cox P., et al. Zidovudine ameliorates pathology in the mouse model of Duchenne muscular dystrophy via P2RX7 purinoceptor antagonism. Acta Neuropathol Commun. 2018;6:27. doi: 10.1186/s40478-018-0530-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Di Virgilio F. Purinergic signalling in the immune system. A brief update. Purinergic Signal. 2007;3:1–3. doi: 10.1007/s11302-006-9048-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Di Virgilio F., Dal Ben D., Sarti A.C., Giuliani A.L., Falzoni S. The P2X7 receptor in infection and inflammation. Immunity. 2017;47:15–31. doi: 10.1016/j.immuni.2017.06.020. [DOI] [PubMed] [Google Scholar]
  • 84.Murphy S., Dowling P., Zweyer M., et al. Proteomic analysis of dystrophin deficiency and associated changes in the aged mdx-4cv heart model of dystrophinopathy-related cardiomyopathy. J Proteomics. 2016;145:24–36. doi: 10.1016/j.jprot.2016.03.011. [DOI] [PubMed] [Google Scholar]
  • 85.Hughes M.C., Ramos S.V., Turnbull P.C., et al. Impairments in left ventricular mitochondrial bioenergetics precede overt cardiac dysfunction and remodelling in Duchenne muscular dystrophy. J Physiol. 2020;598:1377–1392. doi: 10.1113/JP277306. [DOI] [PubMed] [Google Scholar]
  • 86.Whitehead N.P., Yeung E.W., Allen D.G. Muscle damage in mdx (dystrophic) mice: role of calcium and reactive oxygen species. Clin Exp Pharmacol Physiol. 2006;33:657–662. doi: 10.1111/j.1440-1681.2006.04394.x. [DOI] [PubMed] [Google Scholar]
  • 87.Hughes M.C., Ramos S.V., Turnbull P.C., et al. Early myopathy in Duchenne muscular dystrophy is associated with elevated mitochondrial H2 O2 emission during impaired oxidative phosphorylation. J Cachexia Sarcopenia Muscle. 2019;10:643–661. doi: 10.1002/jcsm.12405. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Godin R., Daussin F., Matecki S., Li T., Petrof B.J., Burelle Y. Peroxisome proliferator-activated receptor gamma coactivator1- gene alpha transfer restores mitochondrial biomass and improves mitochondrial calcium handling in post-necrotic mdx mouse skeletal muscle. J Physiol. 2012;590:5487–5502. doi: 10.1113/jphysiol.2012.240390. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Timpani C.A., Hayes A., Rybalka E. Revisiting the dystrophin-ATP connection: how half a century of research still implicates mitochondrial dysfunction in Duchenne Muscular Dystrophy aetiology. Med Hypotheses. 2015;85:1021–1033. doi: 10.1016/j.mehy.2015.08.015. [DOI] [PubMed] [Google Scholar]
  • 90.Jung D.W., Baysal K., Brierley G.P. The sodium-calcium antiport of heart mitochondria is not electroneutral. J Biol Chem. 1995;270:672–678. doi: 10.1074/jbc.270.2.672. [DOI] [PubMed] [Google Scholar]
  • 91.Williams I.A., Allen D.G. Intracellular calcium handling in ventricular myocytes from mdx mice. Am J Physiol Heart Circ Physiol. 2007;292:H846–H855. doi: 10.1152/ajpheart.00688.2006. [DOI] [PubMed] [Google Scholar]
  • 92.Yasuda S., Townsend D., Michele D.E., Favre E.G., Day S.M., Metzger J.M. Dystrophic heart failure blocked by membrane sealant poloxamer. Nature. 2005;436:1025–1029. doi: 10.1038/nature03844. [DOI] [PubMed] [Google Scholar]
  • 93.Kang C., Badr M.A., Kyrychenko V., Eskelinen E.L., Shirokova N. Deficit in PINK1/PARKIN-mediated mitochondrial autophagy at late stages of dystrophic cardiomyopathy. Cardiovasc Res. 2018;114:90–102. doi: 10.1093/cvr/cvx201. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Gold R., Kappos L., Arnold D.L., et al. Placebo-controlled phase 3 study of oral BG-12 for relapsing multiple sclerosis. N Engl J Med. 2012;367:1098–1107. doi: 10.1056/NEJMoa1114287. [DOI] [PubMed] [Google Scholar]
  • 95.Juge N., Mithen R.F., Traka M. Molecular basis for chemoprevention by sulforaphane: a comprehensive review. Cell Mol Life Sci. 2007;64:1105–1127. doi: 10.1007/s00018-007-6484-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Sun C., Li S., Li D. Sulforaphane mitigates muscle fibrosis in mdx mice via Nrf2-mediated inhibition of TGF-beta/Smad signaling. J Appl Physiol (1985) 2016;120:377–390. doi: 10.1152/japplphysiol.00721.2015. [DOI] [PubMed] [Google Scholar]
  • 97.Yarmohammadi F., Hayes A.W., Karimi G. Protective effects of curcumin on chemical and drug-induced cardiotoxicity: a review. Naunyn Schmiedebergs Arch Pharmacol. 2021;394(7):1341–1353. doi: 10.1007/s00210-021-02072-8. [DOI] [PubMed] [Google Scholar]
  • 98.Su Y., Wang Y., Liu M., Chen H. Hydrogen sulfide attenuates renal I/R-induced activation of the inflammatory response and apoptosis via regulating Nrf2-mediated NLRP3 signaling pathway inhibition. Mol Med Rep. 2021;24(1):518. doi: 10.3892/mmr.2021.12157. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Spurney C.F., Knoblach S., Pistilli E.E., Nagaraju K., Martin G.R., Hoffman E.P. Dystrophin-deficient cardiomyopathy in mouse: expression of Nox4 and Lox are associated with fibrosis and altered functional parameters in the heart. Neuromuscul Disord. 2008;18:371–381. doi: 10.1016/j.nmd.2008.03.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Sarma S., Li N., van Oort R.J., Reynolds C., Skapura D.G., Wehrens X.H. Genetic inhibition of PKA phosphorylation of RyR2 prevents dystrophic cardiomyopathy. Proc Natl Acad Sci U S A. 2010;107:13165–13170. doi: 10.1073/pnas.1004509107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Iwata Y., Ohtake H., Suzuki O., Matsuda J., Komamura K., Wakabayashi S. Blockade of sarcolemmal TRPV2 accumulation inhibits progression of dilated cardiomyopathy. Cardiovasc Res. 2013;99:760–768. doi: 10.1093/cvr/cvt163. [DOI] [PubMed] [Google Scholar]
  • 102.Matsumura T., Matsui M., Iwata Y., et al. A pilot study of tranilast for cardiomyopathy of muscular dystrophy. Intern Med. 2018;57:311–318. doi: 10.2169/internalmedicine.8651-16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Yamada K., Green K.G., Samarel A.M., Saffitz J.E. Distinct pathways regulate expression of cardiac electrical and mechanical junction proteins in response to stretch. Circ Res. 2005;97:346–353. doi: 10.1161/01.RES.0000178788.76568.8a. [DOI] [PubMed] [Google Scholar]
  • 104.Gonzalez J.P., Ramachandran J., Xie L.H., Contreras J.E., Fraidenraich D. Selective connexin43 inhibition prevents isoproterenol-induced arrhythmias and lethality in muscular dystrophy mice. Sci Rep. 2015;5:13490. doi: 10.1038/srep13490. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Gonzalez J.P., Ramachandran J., Himelman E., et al. Normalization of connexin 43 protein levels prevents cellular and functional signs of dystrophic cardiomyopathy in mice. Neuromusc Disord. 2018;28:361–372. doi: 10.1016/j.nmd.2018.01.012. [DOI] [PubMed] [Google Scholar]
  • 106.Himelman E., Lillo M.A., Nouet J., et al. Prevention of connexin-43 remodeling protects against Duchenne muscular dystrophy cardiomyopathy. J Clin Invest. 2020;130:1713–1727. doi: 10.1172/JCI128190. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Vielma A.Z., Boric M.P., Gonzalez D.R. Apocynin treatment prevents cardiac connexin 43 hemichannels hyperactivity by reducing nitroso-redox stress in mdx mice. Int J Mol Sci. 2020;21(15):5415. doi: 10.3390/ijms21155415. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Kienitz M.C., Bender K., Dermietzel R., Pott L., Zoidl G. Pannexin 1 constitutes the large conductance cation channel of cardiac myocytes. J Biol Chem. 2011;286:290–298. doi: 10.1074/jbc.M110.163477. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Chiu Y.H., Schappe M.S., Desai B.N., Bayliss D.A. Revisiting multimodal activation and channel properties of Pannexin 1. J Gen Physiol. 2018;150:19–39. doi: 10.1085/jgp.201711888. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Valladares D., Almarza G., Contreras A., et al. Electrical stimuli are anti-apoptotic in skeletal muscle via extracellular ATP. Alteration of this signal in Mdx mice is a likely cause of dystrophy. PloS One. 2013;8 doi: 10.1371/journal.pone.0075340. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Raucci F.J., Jr., Kim K., Huke S., Knollmann B.C. Increased pannexin 1 expression and activity in ventricle of mdx dystrophic hearts. Biophys J. 2018;114(3 suppl 1):486A–487A. [Google Scholar]
  • 112.Good M.E., Chiu Y.H., Poon I.K.H., et al. Pannexin 1 channels as an unexpected new target of the anti-hypertensive drug spironolactone. Circ Res. 2018;122:606–615. doi: 10.1161/CIRCRESAHA.117.312380. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Fulgenzi G., Tomassoni-Ardori F., Babini L., et al. BDNF modulates heart contraction force and long-term homeostasis through truncated TrkB.T1 receptor activation. J Cell Biol. 2015;210:1003–1012. doi: 10.1083/jcb.201502100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Galindo C.L., Soslow J.H., Brinkmeyer-Langford C.L., et al. Translating golden retriever muscular dystrophy microarray findings to novel biomarkers for cardiac/skeletal muscle function in Duchenne muscular dystrophy. Pediatr Res. 2016;79:629–636. doi: 10.1038/pr.2015.257. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Comim C.M., Mathia G.B., Hoepers A., et al. Neurotrophins, cytokines, oxidative parameters and funcionality in progressive muscular dystrophies. An Acad Bras Cienc. 2015;87:1809–1818. doi: 10.1590/0001-3765201520140508. [DOI] [PubMed] [Google Scholar]
  • 116.Matsuzaka Y., Tanihata J., Ooshima Y., et al. The nSMase2/Smpd3 gene modulates the severity of muscular dystrophy and the emotional stress response in mdx mice. BMC Med. 2020;18:343. doi: 10.1186/s12916-020-01805-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Rafael-Fortney J.A., Chimanji N.S., Schill K.E., et al. Early treatment with lisinopril and spironolactone preserves cardiac and skeletal muscle in Duchenne muscular dystrophy mice. Circulation. 2011;124:582–588. doi: 10.1161/CIRCULATIONAHA.111.031716. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Janssen P.M., Murray J.D., Schill K.E., et al. Prednisolone attenuates improvement of cardiac and skeletal contractile function and histopathology by lisinopril and spironolactone in the mdx mouse model of Duchenne muscular dystrophy. PloS One. 2014;9 doi: 10.1371/journal.pone.0088360. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Raman S.V., Hor K.N., Mazur W., et al. Eplerenone for early cardiomyopathy in Duchenne muscular dystrophy: a randomised, double-blind, placebo-controlled trial. Lancet Neurol. 2015;14:153–161. doi: 10.1016/S1474-4422(14)70318-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Raman S.V., Hor K.N., Mazur W., et al. Stabilization of early Duchenne cardiomyopathy with aldosterone inhibition: results of the multicenter AIDMD trial. J Am Heart Assoc. 2019;8 doi: 10.1161/JAHA.119.013501. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Feingold B., Mahle W.T., Auerbach S., et al. Management of cardiac involvement associated with neuromuscular diseases: a scientific statement from the American Heart Association. Circulation. 2017;136:e200–e231. doi: 10.1161/CIR.0000000000000526. [DOI] [PubMed] [Google Scholar]
  • 122.Sauer A.J., Cole R., Jensen B.C., et al. Practical guidance on the use of sacubitril/valsartan for heart failure. Heart Fail Rev. 2019;24:167–176. doi: 10.1007/s10741-018-9757-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.McMurray J.J., Packer M., Desai A.S., et al. Angiotensin-neprilysin inhibition versus enalapril in heart failure. N Engl J Med. 2014;371:993–1004. doi: 10.1056/NEJMoa1409077. [DOI] [PubMed] [Google Scholar]
  • 124.Lamendola P., Lanza G.A., Melita V., et al. Duchenne muscular dystrophy: preliminary experience with sacubitril-valsartan in patients with asymptomatic left ventricular dysfunction. Eur Rev Med Pharmacol Sci. 2020;24:9112–9115. doi: 10.26355/eurrev_202009_22857. [DOI] [PubMed] [Google Scholar]
  • 125.Kamdar F., Das S., Gong W., et al. Stem cell-derived cardiomyocytes and beta-adrenergic receptor blockade in Duchenne muscular dystrophy cardiomyopathy. J Am Coll Cardiol. 2020;75:1159–1174. doi: 10.1016/j.jacc.2019.12.066. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Matsumura T., Tamura T., Kuru S., Kikuchi Y., Kawai M. Carvedilol can prevent cardiac events in Duchenne muscular dystrophy. Intern Med. 2010;49:1357–1363. doi: 10.2169/internalmedicine.49.3259. [DOI] [PubMed] [Google Scholar]
  • 127.Blain A., Greally E., Laval S., Blamire A., Straub V., MacGowan G.A. Beta-blockers, left and right ventricular function, and in-vivo calcium influx in muscular dystrophy cardiomyopathy. PLoS One. 2013;8 doi: 10.1371/journal.pone.0057260. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Adorisio R., Calvieri C., Cantarutti N., et al. Heart rate reduction strategy using ivabradine in end-stage Duchenne cardiomyopathy. Int J Cardiol. 2019;280:99–103. doi: 10.1016/j.ijcard.2019.01.052. [DOI] [PubMed] [Google Scholar]
  • 129.De Benedittis G., Della Rosa G., D'Ettorre E., Piscitelli P., Distante A., de Gregorio C. Effect of ivabradine in dilated cardiomyopathy from Duchenne muscular dystrophy: a chance for slowing progression of heart failure? Int J Cardiol. 2016;223:286–288. doi: 10.1016/j.ijcard.2016.08.135. [DOI] [PubMed] [Google Scholar]
  • 130.Krivov L.I., Stenina M.A., Yarygin V.N., et al. A new genetic variant of mdx mice: study of the phenotype. Bull Exp Biol Med. 2009;147(5):625–629. doi: 10.1007/s10517-009-0564-5. [DOI] [PubMed] [Google Scholar]
  • 131.Schmidt W.M., Uddin M.H., Dysek S., et al. DNA damage, somatic aneuploidy, and malignant sarcoma susceptibility in muscular dystrophies. PLoS Genet. 2011;7(4) doi: 10.1371/journal.pgen.1002042. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Wasala N.B., Zhang K., Wasala L.P., Hakim C.H., Duan D. The FVB background does not dramatically alter the dystrophic phenotype of Mdx mice. PLoS Curr. 2015;7 doi: 10.1371/currents.md.28266819ca0ec5fefcac767ea9a3461c. 28266819ca0ec5fefcac767ea9a3461c. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Chapman V.M., Miller D.R., Armstrong D., Caskey C.T. Recovery of induced mutations for X chromosome-linked muscular dystrophy in mice. Proc Natl Acad Sci U S A. 1989;86(4):1292–1296. doi: 10.1073/pnas.86.4.1292. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Araki E., Nakamura K., Nakao K., et al. Targeted disruption of exon 52 in the mouse dystrophin gene induced muscle degeneration similar to that observed in Duchenne muscular dystrophy. Biochem Biophys Res Commun. 1997;238(2):492–497. doi: 10.1006/bbrc.1997.7328. [DOI] [PubMed] [Google Scholar]
  • 135.Wertz K., Füchtbauer E.M. Dmd(mdx-beta geo): a new allele for the mouse dystrophin gene. Dev Dyn. 1998;212(2):229–241. doi: 10.1002/(SICI)1097-0177(199806)212:2<229::AID-AJA7>3.0.CO;2-J. [DOI] [PubMed] [Google Scholar]
  • 136.Kudoh H., Ikeda H., Kakitani M., et al. A new model mouse for Duchenne muscular dystrophy produced by 2.4 Mb deletion of dystrophin gene using Cre-loxP recombination system. Biochem Biophys Res Commun. 2005;328(2):507–516. doi: 10.1016/j.bbrc.2004.12.191. [DOI] [PubMed] [Google Scholar]
  • 137.Guo C., Willem M., Werner A., et al. Absence of alpha 7 integrin in dystrophin-deficient mice causes a myopathy similar to Duchenne muscular dystrophy. Hum Mol Genet. 2006;15(6):989–998. doi: 10.1093/hmg/ddl018. [DOI] [PubMed] [Google Scholar]
  • 138.Deconinck A.E., Rafael J.A., et al. Utrophin-dystrophin-deficient mice as a model for Duchenne muscular dystrophy. Cell. 1997;90(4):717–727. doi: 10.1016/s0092-8674(00)80532-2. [DOI] [PubMed] [Google Scholar]
  • 139.Grady R.M., Teng H., Nichol M.C., Cunningham J.C., Wilkinson R.S., Sanes J.R. Skeletal and cardiac myopathies in mice lacking utrophin and dystrophin: a model for Duchenne muscular dystrophy. Cell. 1997;90(4):729–738. doi: 10.1016/s0092-8674(00)80533-4. [DOI] [PubMed] [Google Scholar]
  • 140.Megeney L.A., Kablar B., Garrett K., Anderson J.E., Rudnicki M.A. MyoD is required for myogenic stem cell function in adult skeletal muscle. Genes Dev. 1996;10(10):1173–1183. doi: 10.1101/gad.10.10.1173. [DOI] [PubMed] [Google Scholar]
  • 141.Chandrasekharan K., Yoon J.H., Xu Y., et al. A human-specific deletion in mouse Cmah increases disease severity in the mdx model of Duchenne muscular dystrophy. Sci Transl Med. 2010;2(42):42ra54. doi: 10.1126/scitranslmed.3000692. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Grady R.M., Grange R.W., Lau K.S., et al. Role for alpha-dystrobrevin in the pathogenesis of dystrophin-dependent muscular dystrophies. Nat Cell Biol. 1999;1(4):215–220. doi: 10.1038/12034. [DOI] [PubMed] [Google Scholar]
  • 143.Li D., Long C., Yue Y., Duan D. Sub-physiological sarcoglycan expression contributes to compensatory muscle protection in mdx mice. Hum Mol Genet. 2009;18(7):1209–1220. doi: 10.1093/hmg/ddp015. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from JACC: Basic to Translational Science are provided here courtesy of Elsevier

RESOURCES