Clinical and molecular findings in a cohort of ANO5-related myopathy

André M S Silva, Antônio R Coimbra-Neto, Paulo Victor S Souza, Pablo B Winckler, Marcus V M Gonçalves, Eduardo B U Cavalcanti, Alzira A D S Carvalho, Cláudia F D R Sobreira, Clara G Camelo, Rodrigo D H Mendonça, Eduardo D P Estephan, Umbertina C Reed, Marcela C Machado-Costa, Mario E T Dourado-Junior, Vanessa C Pereira, Marcelo M Cruzeiro, Paulo V P Helito, Laís U Aivazoglou, Leonardo V D Camargo, Hudson H Gomes, Amaro J S D Camargo, Wladimir B V D R Pinto, Bruno M L Badia, Luiz H Libardi, Mario T Yanagiura, Acary S B Oliveira, Anamarli Nucci, Jonas A M Saute, Marcondes C França-Junior, Edmar Zanoteli, André M S Silva, Antônio R Coimbra-Neto, Paulo Victor S Souza, Pablo B Winckler, Marcus V M Gonçalves, Eduardo B U Cavalcanti, Alzira A D S Carvalho, Cláudia F D R Sobreira, Clara G Camelo, Rodrigo D H Mendonça, Eduardo D P Estephan, Umbertina C Reed, Marcela C Machado-Costa, Mario E T Dourado-Junior, Vanessa C Pereira, Marcelo M Cruzeiro, Paulo V P Helito, Laís U Aivazoglou, Leonardo V D Camargo, Hudson H Gomes, Amaro J S D Camargo, Wladimir B V D R Pinto, Bruno M L Badia, Luiz H Libardi, Mario T Yanagiura, Acary S B Oliveira, Anamarli Nucci, Jonas A M Saute, Marcondes C França-Junior, Edmar Zanoteli

Abstract

Objective: ANO5-related myopathy is an important cause of limb-girdle muscular dystrophy (LGMD) and hyperCKemia. The main descriptions have emerged from European cohorts, and the burden of the disease worldwide is unclear. We provide a detailed characterization of a large Brazilian cohort of ANO5 patients.

Methods: A national cross-sectional study was conducted to describe clinical, histopathological, radiological, and molecular features of patients carrying recessive variants in ANO5. Correlation of clinical and genetic characteristics with different phenotypes was studied.

Results: Thirty-seven patients from 34 nonrelated families with recessive mutations of ANO5 were identified. The most common phenotype was LGMD, observed in 25 (67.5%) patients, followed by pseudometabolic presentation in 7 (18.9%) patients, isolated asymptomatic hyperCKemia in 4 (10.8%) patients, and distal myopathy in a single patient. Nine patients presented axial involvement, including one patient with isolated axial weakness. The most affected muscles according to MRI were the semimembranosus and gastrocnemius, but paraspinal and abdominal muscles, when studied, were involved in most patients. Fourteen variants in ANO5 were identified, and the c.191dupA was present in 19 (56%) families. Sex, years of disease, and the presence of loss-of-function variants were not associated with specific phenotypes.

Interpretation: We present the largest series of anoctaminopathy outside Europe. The most common European founder mutation c.191dupA was very frequent in our population. Gender, disease duration, and genotype did not determine the phenotype.

Conflict of interest statement

Nothing to declare.

© 2019 The Authors. Annals of Clinical and Translational Neurology published by Wiley Periodicals, Inc on behalf of American Neurological Association.

Figures

Figure 1
Figure 1
Muscle biopsy of patients with anoctaminopathy. Unspecific myopathic findings in patient #1.1 characterized by mild fiber size variability, increase in endomysial connective tissue, and increase in nuclei internalization (A) (H&E). More severe myopathic features (dystrophic aspect) in patient #2 (B) (H&E). Preserved muscle architecture with the presence of grouped necrotic fibers (C) (arrow heads) in patient #4; sparse necrotic fibers (D) in patient #26 (degeneration fiber with macrophagy indicated by the black arrowhead and without macrophagy indicated by the white arrowhead) (H&E). Bar = 50 µm (A, B and C); Bar = 20 µm (D).
Figure 2
Figure 2
The heatmap for muscle involvement in MRI, using the 5‐point scale for each muscle (right and left) of each patient. Muscles and patients were ordered in grade of severity based on the mean score of the muscle and patient. The score of a muscle in a patient is indicated by the color of the square. White squares mean that data are not available. R, right; L, left; LGMD, limb‐girdle muscular dystrophy; MMD3, Miyoshi‐like distal myopathy; NA, not available
Figure 3
Figure 3
Typical MRI findings of ANO5‐related myopathy. (A and B) Axial T1‐weighted images of the thighs reveal striking involvement of the posterior compartment (mainly the semimembranosus and the adductors) in patient #7; black asterisks in A show bilateral fatty degeneration of the adductors, and the white asterisk in A shows asymmetrical involvement of the semitendinosus. White asterisks in B show asymmetrical sparing of the short head of the left biceps femoris. (C and D) Axial T1‐weighted images of the legs show bilateral fatty infiltration of the medial head of the gastrocnemius (white arrows in C) and asymmetrical involvement of the soleus (black asterisk in D) in patient #7. (E) Axial T1‐weighted image of the lumbar spine of patient #9 reveals severe fatty degeneration of the posterior paravertebral muscles (white arrow). (F) Coronal T1‐weighted image of the thighs in patient #15 shows a craniocaudal gradient of involvement (the distal portion of the quadriceps muscles is more affected than the proximal portion). (G and H) Axial T1‐weighted image of the thighs of patient #29.1 shows severe fatty infiltration of the quadriceps and the “undulating fascia” sign (white arrowhead) in G, and areas of hyperintensity indicate muscle edema (white head) in H (axial STIR‐weighted image)

References

    1. Hartzell HC, Yu K, Xiao Q, et al. Anoctamin/TMEM16 family members are Ca2+‐activated Cl– channels. J Physiol 2009;587(Pt 10):2127–39.
    1. Griffin DA, Johnson RW, Whitlock JM, et al. Defective membrane fusion and repair in anoctamin5‐deficient muscular dystrophy. Hum Mol Genet 2016;25:1900–11.
    1. Bolduc V, Marlow G, Boycott KM, et al. Recessive mutations in the putative calcium‐activated chloride channel Anoctamin 5 cause proximal LGMD2L and distal MMD3 muscular dystrophies. Am J Hum Genet 2010;86:213–21.
    1. Hicks D, Sarkozy A, Muelas N, et al. A founder mutation in Anoctamin 5 is a major cause of limb‐girdle muscular dystrophy. Brain 2011;134:171–82.
    1. Schessl J, Kress W, Schoser B. Novel ANO5 mutations causing hyper‐CK‐emia, limb girdle muscular dystrophy and miyoshi type muscular dystrophy. Muscle Nerve 2012;45:740–2.
    1. Papadopoulos C, Laforêt P, Nectoux J, et al. Hyperckemia and myalgia are common presentations of anoctamin‐5‐related myopathy in French patients. Muscle Nerve 2017;56:1096–100.
    1. Tsutsumi S, Kamata N, Vokes TJ, et al. The novel gene encoding a putative transmembrane protein is mutated in gnathodiaphyseal dysplasia (GDD). Am J Hum Genet 2004;74:1255–61.
    1. Magri F, Del Bo R, D'Angelo MG, et al. Frequency and characterisation of anoctamin 5 mutations in a cohort of Italian limb‐girdle muscular dystrophy patients. Neuromuscul Disord 2012;22:934–43.
    1. Penttilä S, Palmio J, et al. E Eight new mutations and the expanding phenotype variability in muscular dystrophy caused by ANO5. Neurology 2012;78:897–903.
    1. Nallamilli BRR, Chakravorty S, Kesari A, et al. Genetic landscape and novel disease mechanisms from a large LGMD cohort of 4656 patients. Ann Clin Transl Neurol 2018;5:1574–87.
    1. Sarkozy A, Hicks D, Hudson J, et al. ANO5 gene analysis in a large cohort of patients with anoctaminopathy: confirmation of male prevalence and high occurrence of the common exon 5 gene mutation. Hum Mutat 2013;34:1111–8.
    1. Kadoya M, Ogata K, Suzuki M, et al. A Japanese male with a novel ANO5 mutation with minimal muscle weakness and muscle pain till his late fifties. Neuromuscul Disord 2017;27:477–80.
    1. Hu B, Xiong L, Zhou Y, et al. First familial limb‐girdle muscular dystrophy 2L in China: Clinical, imaging, pathological, and genetic features. Medicine (Baltimore). 2018;97:e12506.
    1. Bohlega S, Monies DM, Abulaban AA, et al. Clinical and genetic features of anoctaminopathy in Saudi Arabia. Neurosciences (Riyadh). 2015;20:173–7.
    1. Fischer D, Kley RA, Strach K, et al. Distinct muscle imaging patterns in myofibrillar myopathies. Neurology 2008;71:758–65.
    1. Mercuri E, Cini C, Counsell S, et al. Muscle MRI findings in a three‐generation family affected by Bethlem myopathy. Eur J Paediatr Neurol 2002;6:309–14.
    1. Richards S, Aziz N, Bale S, et al. Standards and guidelines for the interpretation of sequence variants: a joint consensus recommendation of the American College of Medical Genetics and Genomics and the Association for Molecular Pathology. Genet Med 2015;17:405–24.
    1. Adzhubei IA, Schmidt S, Peshkin L, et al. A method and server for predicting damaging missense mutations. Nat Methods 2010;7:248–9.
    1. Ng PC, Henikoff S. Predicting deleterious amino acid substitutions. Genome Res 2001;11:863–74.
    1. Rentzsch P, Witten D, Cooper GM, et al. CADD: predicting the deleteriousness of variants throughout the human genome. Nucleic Acids Res 2019;47(D1):D886–D894.
    1. Jagadeesh KA, Wenger AM, Berger MJ, et al. M‐CAP eliminates a majority of variants of uncertain significance in clinical exomes at high sensitivity. Nat Genet 2016;48:1581–6.
    1. Schwarz JM, Cooper DN, Schuelke M, et al. MutationTaster2: mutation prediction for the deep‐sequencing age. Nat Methods 2014;11:361–2.
    1. Cooper GM, Stone EA, Asimenos G, et al. Distribution and intensity of constraint in mammalian genomic sequence. Genome Res 2005;15:901–13.
    1. gnomAD, Genome Aggregation Database, . 2018. (Accessed December 2018).
    1. Browser Genomes, . 2018. (Accessed December 2018).
    1. Guimaraes JB, Zanoteli E, Link TM, et al. Sporadic inclusion body myositis: MRI findings and correlation with clinical and functional parameters. Am J Roentgenol 2017;209:1340–1347.
    1. van der Kooi AJ, Ten Dam L, Frankhuizen WS, et al. ANO5 mutations in the Dutch limb girdle muscular dystrophy population. Neuromuscul Disord 2013;23:456–60.
    1. Liewluck T, Winder TL, Dimberg EL, et al. ANO5‐muscular dystrophy: clinical, pathological and molecular findings. Eur J Neurol 2013;20:1383–9.
    1. Wahbi K, Béhin A, Bécane HM, et al. Dilated cardiomyopathy in patients with mutations in anoctamin 5. Int J Cardiol 2013;168:76–9.
    1. Witting N, Duno M, Petri H, et al. Anoctamin 5 muscular dystrophy in Denmark: prevalence, genotypes, phenotypes, cardiac findings, and muscle protein expression. J Neurol 2013;260:2084–93.
    1. Guerreiro‐Junior G, Bisso‐Machado R, Marrero A, et al. Genetic signatures of parental contribution in black and white populations in Brazil. Genet Mol Biol 2009;32:1–11.
    1. Moura RR, Coelho, et al. Meta‐analysis of Brazilian genetic admixture and comparison with other Latin America Countries. Am J Hum Biol 27:674–80.
    1. Lahoria R, Winder TL, Lui J, et al. Novel ANO5 homozygous microdeletion causing myalgia and unprovoked rhabdomyolysis in an Arabic man. Muscle Nerve 2014;50:610–3.
    1. Little AA, McKeever PE, Gruis KL. Novel mutations in the Anoctamin 5 gene (ANO5) associated with limb‐girdle muscular dystrophy 2L. Muscle Nerve 2013;47:287–91.
    1. Savarese M, Di Fruscio G, Tasca G, et al. Next generation sequencing on patients with LGMD and nonspecific myopathies: findings associated with ANO5 mutations. Neuromuscul Disord 2015;25:533–41.
    1. Sarkozy A, Deschauer M, Carlier RY, et al. Muscle MRI findings in limb girdle muscular dystrophy type 2L. Neuromuscul Disord 2012;22(Suppl 2):S122–9.
    1. de Camargo LV, de Carvalho MS, Shinjo SK, et al. Clinical, histological, and immunohistochemical findings in inclusion body myositis. Biomed Res Int 2018;2018:5069042.

Source: PubMed

Подписаться