Congenital muscular dystrophy due to laminin α2 (merosin) deficiency (MDC1A) in an ethnic Malay girl

Similar documents
Congenital Muscular Dystrophy: Hospital Based Study in Egyptian Pediatric Patients

Muscular Dystrophies. Pinki Munot Consultant Paediatric Neurologist Great Ormond Street Hospital Practical Neurology Study days April 2018

A rare case of muscular dystrophy with POMT2 and FKRP gene mutation. Present by : Ghasem Khazaei Supervisor :Dr Mina Mohammadi Sarband

Merosin-Deficient Congenital Muscular Dystrophy with Polymicrogyria and Subcortical Heterotopia: A Case Report

The Floppy Baby. Clare Betteridge

Immunohistochemical Study of Dystrophin Associated Glycoproteins in Limb-girdle Muscular Dystrophies

Exercise induced cramps and myoglobinuria in dystrophinopathy a report of three Malaysian patients

ORIGINAL CONTRIBUTION. New Mutations, Prenatal Diagnosis, and Founder Effect

Association of motor milestones and SMN2 copy and outcome in spinal muscular. atrophy types 0 4

Iowa Wellstone Center Muscle Tissue and Cell Culture Repository

Genetic diagnosis of limb girdle muscular dystrophy type 2A, A Case Report

Early-Onset LMNA-Associated Muscular Dystrophy with Later Involvement of Contracture

Next-generation sequencing-based molecular diagnosis of neonatal hypotonia in Chinese

A novel POMT2 mutation causes mild congenital muscular dystrophy with normal brain MRI

Muscular Dystrophy. Biol 405 Molecular Medicine

Tel: , Fax: ,

A CASE OF GIANT AXONAL NEUROPATHY HEMANANTH T SECOND YEAR POST GRADUATE IN PAEDIATRICS INSTITUTE OF SOCIAL PAEDIATRICS GOVERNMENT STANLEY HOSPITAL

Statutory Approvals Committee minutes

Case 1: History of J.H. Outside Evaluation. Outside Labs. Question #1

Genetic test for Bilateral frontoparietal polymicrogyria

Profile, types, duration and severity of muscular dystrophy: a clinical study at a tertiary care hospital

III./10.4. Diagnosis. Introduction. A.) Laboratory tests. Laboratory tests, electrophysiology, muscle biopsy, genetic testing, imaging techniques

Hypotonia Care Pathway Update: Diagnosis Garey Noritz, MD...

Introduction. Overview

Anesthesia recommendations for patients suffering from Welander distal myopathy

READ ORPHA.NET WEBSITE ABOUT BETA-SARCOGLYOCANOPATHY LIMB-GIRDLE MUSCULAR DYSTROPHIES

Clinical and pathologic aspects of congenital myopathies

Congenital muscular dystrophies: New aspects of an expanding group of disorders

Corporate Medical Policy

The Brain Pathology in Fukuyama Type Congenital Muscular Dystrophy -CT and Autopsy Findings-

Diseases of Muscle and Neuromuscular Junction

Learn the steps to identify pediatric muscle weakness and signs of neuromuscular disease.

Limb girdle muscular dystrophy type 2B masquerading as inflammatory myopathy: case report

Nemaline (rod) myopathies

18 (2), DOI: /bjmg

Neonatal Hypotonia Guideline Prepared by Dan Birnbaum MD August 27, 2012

The congenital muscular dystrophies in 2004: a century of exciting progress

Peripheral neuropathy in merosin-negative congenital muscular dystrophy

Breathing problems: and how to get on top of them

Ullrich congenital muscular dystrophy: Report of nine cases from India

Muscle Pathology Surgical Pathology Unknown Conference. November, 2008 Philip Boyer, M.D., Ph.D.

Evaluation of the Hypotonic Infant and Child

Neuromuscular in the Pediatric Clinic: Recognition and Referral

Myotubular (centronuclear) myopathy

Proposal form for the evaluation of a genetic test for NHS Service Gene Dossier

Genetics of Inclusion Body Myositis

Prevalence of congenital muscular dystrophy in Italy A population study

Dysferlinopathies. LGMD2B, Miyoshi & Others. 2B Empowered Conference

Neonatal Hypotonia. Encephalopathy acute No encephalopathy. Neurology Chapter of IAP

Diagnosis, management and new treatments for Spinal Muscular Atrophy Special Focus: SMA Type 1

Case : A Newborn Boy with Hypotonia

Hutterite brothers both affected with two forms of limb girdle muscular dystrophy: LGMD2H and LGMD2I

Limb-girdle Muscular Dystrophy with New Mutation in Sarcoglycan Beta Gene: A Case Report

muscle biopsy How to do it

Systematizing in vivo modeling of pediatric disorders

DSS-1. No financial disclosures

Abnormalities of dystrophin, the sarcoglycans, and laminin a2 in the muscular dystrophies

Below are photos from a 6 month old child with Werdnig-Hoffman s disease.

Neuroradiological, clinical and genetic characterization of new forms of hereditary leukoencephalopathies

Corporate Medical Policy

The spectrum of elderly myopathies in an Asian population

Clinical features, particularly those of the central nervous system, of patients with Becker s muscular dystrophy, including autopsied cases

Cigna Drug and Biologic Coverage Policy

CLASSIFICATION OF PERIPHERAL NERVE DISEASES

Case 1 A 65 year old college professor came to the neurology clinic referred by her family physician because of frequent falling. She had a history of

Should Universal Carrier Screening be Universal?

Dubowitz Neuromuscular Centre, Institute of Child Health and Great Ormond Street Hospital for Children, UCL, London, UK. 2

Cover Page. The handle holds various files of this Leiden University dissertation.

SMA IS A SEVERE NEUROLOGICAL DISORDER [1]

What s New in Newborn Screening?

CARDIAC muscle is commonly affected in muscular

Red flags for clinical practice - guidance on indicators that your patient may have a genetic condition

Proposal form for the evaluation of a genetic test for NHS Service Gene Dossier

Three Muscular Dystrophies: Loss of Cytoskeleton-Extracellular Matrix Linkage

variant led to a premature stop codon p.k316* which resulted in nonsense-mediated mrna decay. Although the exact function of the C19L1 is still

Fukutin Gene Mutations Cause Dilated Cardiomyopathy with Minimal Muscle Weakness

Corporate Medical Policy

An Unusual Triad in Pediatric Neurology: A Case Report on Cerebral Palsy, Epilepsy, and Duchenne Muscular Dystrophy

Cerebral Malformation gene panel

Spinal Muscular Atrophy: Case Study. Spinal muscular atrophy (SMA) is a fairly common genetic disorder, affecting

1/28/2019. OSF HealthCare INI Care Center Team. Neuromuscular Disease: Muscular Dystrophy. OSF HealthCare INI Care Center Team: Who are we?

Case Report GNE Myopathy in Turkish Sisters with a Novel Homozygous Mutation

Genetics of Hereditary Spastic Paraplegia Dr. Arianna Tucci

Clinical Policy: Nusinersen (Spinraza) Reference Number: CP.PHAR.327

EDX in Myopathies Limitations. EDX in Myopathies Utility Causes of Myopathy. Myopathy: Issues for Electromyographers

Scholars Journal of Medical Case Reports

SURVEY OF DUCHENNE TYPE AND CONGENITAL TYPE OF MUSCULAR DYSTROPHY IN SHIMANE, JAPAN 1

TREAT-NMD Conference 2013

Approach to the Child with Developmental Delay

Electron microscopy in the investigation and diagnosis of muscle disease

Whole Exome Sequencing Reveals DYSF, FKTN, and ISPD Mutations in Congenital Muscular Dystrophy Without Brain or Eye Involvement

Pediatric Residency Child Neurology Elective

Title. CitationInternal Medicine, 46(8): Issue Date Doc URL. Type. File Information

See Important Reminder at the end of this policy for important regulatory and legal information.

Diagnosis of McArdle's disease in an elderly patient: Case report and review of literature

Disorders of Muscle. Disorders of Muscle. Muscle Groups Involved in Myopathy. Needle Examination of EMG. History. Muscle Biopsy

Diffusion-Weighted and Conventional MR Imaging Findings of Neuroaxonal Dystrophy

Pediatric Aspects of EDX

Proposal form for the evaluation of a genetic test for NHS Service Gene Dossier

Gene therapy and genome editing technologies for the study and potential treatment of :

Transcription:

Neurology Asia 2017; 22(2) : 155 159 Congenital muscular dystrophy due to laminin α2 (merosin) deficiency (MDC1A) in an ethnic Malay girl 1 MK Thong, 3 Sofiah Ali, 4 YE Park, 5 DS Kim, 6 KJ Goh, 2 KT Wong 1 Departments of Paediatrics, 2 Pathology and 6 Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia; 3 Sime Darby Medical Centre, Subang Jaya, Selangor, Malaysia; 4 Department of Neurology and Biomedical Research Institute, Pusan National University Hospital, Busan, Korea; 5 Department of Neurology, Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, Korea Abstract We report the first known ethnic Malay patient with laminin alpha-2 (merosin) deficiency (MDC1A), a subtype of congenital muscular dystrophy (CMD)as a result of novel LAMA2 gene mutations. The 21-month-old female presented with hypotonia at birth and gross motor delay of her distal lower limbs. Physical examination showed generalised hypotonia, hyporeflexia and myopathic facies but good cognitive functions. Serum creatine kinase was elevated and white matter changes were detected in the brain MRI. Muscle biopsy showed dystrophic changes with complete laminin α2 deficiency by immunohistochemistry. Mutation analysis of LAMA2 showed compound heterozygote at exon 21, c.2888delg(p.gly963alafs*111) and exon 34, c.4886dupc(p.pro1629profs*40) leading to premature stop codon for each of the frameshift mutations. Patient review at seven years of age showed satisfactory cognitive functions despite having contractures and weakness. Genetic testing of LAMA2 related muscular dystrophy facilitated the earlier diagnosis of MDC1A and genetic counselling for this family. Keywords: laminin alpha-2 deficiency; merosin deficiency. LAMA2, Malaysia, congenital muscular dystrophy, MDC1A INTRODUCTION Congenital muscular dystrophy (CMD) is a group of inherited muscle diseases affecting 1:10,000 to 1:50,000 of the population. The common presentation is muscle weakness beginning at birth or early infancy, severe hypotonia, poor spontaneous movements with contractures of the large joints and scoliosis, feeding difficulties with failure to thrive, recurrent aspiration pneumonia and respiratory failure. 1 Serum creatine kinase is elevated and muscle biopsy shows dystrophy. 2 However, there is wide clinical and genetic heterogeneity and the diagnosis relies on clinical findings, brain and muscle imaging, muscle biopsy immunohistochemistry (IHC) and genetic testing. Currently recognised CMD subtypes include laminin alpha-2 (merosin) deficiency (MDC1A), collagen VI-deficient CMD (Ullrich disease), the alpha dystroglycanopathies (caused by POMT1, POMT2, FKTN, FKRP, LARGE, and POMGNT1 mutations), SEPN1-related CMD and LMNArelated CMD (L-CMD). 3 MDC1A (OMIM number 605788) is caused by mutations in the laminin alpha-2 (LAMA2)gene. Loss-of-function mutations had been reported in severely-affected neonates, but only missense mutations had been found in milder cases with partial LAMA2 deficiency. 4 Over 50% of MDC1A are due to complete LAMA2 deficiency. 5,6 Brain MRI white matter abnormalities without major cognitive involvement are associated with the MDC1A subtype. We present a Malay child with MDC1A associated with novel LAMA2stop-codon gene mutations. There is limited information on MDC1A in the South-East Asian population, and this report extends the genotype-phenotype correlation of this condition CASE REPORT The patient, a one-year and 9-month old ethnic Malay girl was hypotonic since birth with gross motor delay involving the lower more than the upper limbs. There was no significant family history and she was the only child. Her parents were not related. Physical examination showed she has normal stature with generalised hypotonia, Address correspondence to: Wong Kum-Thong, Department of Pathology, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia. E-mail: wongkt@ummc.edu.my 155

Neurology Asia June 2017 hyporeflexia and good cognitive functions with normal developmental milestones. The distal part of the lower limbs was more affected than the proximal part. There were no dysmorphic features. She had myopathic facies but no abnormal eye movements, cranial nerve palsies or myoclonus. Visual acuity and funduscopy were normal. There were no joint contractures, calf hypertrophy or positive Gower s sign. Cardiorespiratory examination was normal. The serum creatine kinase was 1858 IU/L. Brain MRI showed hyperintense white matter changes of the frontal, temporal, parietal and occipital lobes bilaterally with sparing of the occipital sub-cortical and cerebellar white matter. (Figure 1). The electromyogram showed evidence of myopathy. The left biceps muscle biopsy showed moderate variation in fibre size with randomly distributed atrophic but there were no hypertrophic fibres. Many fibres showed evidence of degeneration, regeneration and focal necrosis with surrounding inflammation and increased acid phosphatase activity. Moderate increase of endomysial and perimysial fibrous and adipose tissues in the fascicles were observed. There was no fibre type grouping and there were no abnormal vacuoles, cores and inclusions or ragged red, whorled and split fibres. There was only mild intermyofibrillar network disruption and lobulated fibres were absent. Immunohistochemistry (IHC) for merosin was negative. The dystrophin and sarcoglycan IHC generally showed faint and patchy staining, while α-dystroglycan and collagen 6 were normal. These findings were in keeping with the diagnosis of MDC1A. (Figure 2) At seven years of age, she attended school and had appropriate cognitive function. Physical examination showed a thin child with myopathic facies, hyporeflexia and hypotonia of both upper and lower limbs. The hypotonia was more obvious over the lower rather than the upper limbs. She had generalised contractures predominantly of the Figure 1 (A-D): Axial T2 MR images show hyperintense white matter of the frontal, temporal, parietal and occipital lobes bilaterally. The sub-cortical white matter of the occipital lobes (B) and cerebellar white matter are spared. 156

Figure 2: Muscle biopsy of the left biceps showed dystrophic muscle with fibre atrophy, hypertrophy, degeneration and regeneration, interstitial fibrosis and fat infiltration. (H&E, Magnification x20 objective) elbow and knee joints. There were limited joint movements and motor power of MRC grade 3. She was unable to walk without support but she did not require any respiratory support. Mutation analysis of LAMA2 showed compound heterozygote at exon 21, c.2888delg (p.gly963alafs*111) and exon 34, c.4886dupc (p.pro1629profs*40) leading to premature stop codon for each of the frameshift mutations. (Figure 3 and 4). Since there was no similar mutations reported in the LAMA2 mutation database and the findings were likely to be novel. 15 Parental mutation studies were not available. DISCUSSION The clinical findings of CMD with elevated serum creatine kinase, white matter changes in the brain MRI and the IHC finding of complete laminin α2 deficiency in muscle fibres are considered diagnostic of MDC1A. 7 While the prevalence of CMDs has been estimated between 0.7/100,000 and 2.5/100,000, the accurate prevalence of early-onset MDC1A is unknown. 8 MDC1A is being considered as the most common type of CMD all around the world, which accounts for about 30% of CMDs in European countries. The only exception would be Japan where it had been reported to account for only 6% of CMDs Figure 3: Sequencing results of LAMA2 gene (exon 21) which showed c.2888delg (p.gly963alafs*111) 157

Neurology Asia June 2017 Figure 4: Sequencing results of LAMA2 gene (exon 34) which showed c.4886dupc (p.pro1629profs*40) following Fukuyama type CMD. 9 To date, there are anecdotal case report of confirmed MDC1A in East Asia, mainly from Korea and Japan (by IHC analysis) 10,11 or confirmed by genetic and IHC analyses in two patients from China and one patient each from India and Samoa, respectively. 12-14 To the best of our knowledge, this is the first report of a patient with confirmed MDC1A based on IHC study and presence of deleterious LAMA2 mutations in South East Asia, as well as the first Malay patient with the condition. At birth, there is an increased in immune response leading to increased infiltration by T and B cells. Muscle biopsy done during infancy may be misinterpreted as infantile polymyositis. Girgenrath found that treatment of Lama2-null mice with either minocycline or doxycycline, which inhibit apoptosis, showed clinical and pathologic improvement. 15 Muscles derived from the treated mice showed decreased inflammation, and decreased markers of apoptosis, such as Bax and caspase-3. The findings indicated that increased apoptosis is a major pathogenic mechanism in LAMA2 deficiency. The persistence of laminin α4 and α5 leads to abnormal basal lamina causing poor myofibre regeneration in patients beyond one year of age. This leads to dystrophic picture and subsequently end-stage myopathic change. These findings were found in our patient s muscle biopsy which showed inflammatory changes and absent merosin stain on IHC. Compared to the residual merosin group, patients with absent merosin had an earlier presentation (<7days), were more likely to lack independent ambulation, or require enteral feeding and ventilatory support. 16 The T 2 -weighted brain MRI in our patient showed hyperintense white matter of the frontal, temporal, parietal and occipital lobes. These abnormalities were likely to be due to leaky basal laminar connections and increased water content and did not represent areas of demyelination and are detected in some children as early as age six months 17 and were seen in both complete and partial laminin α2 deficiency. 16 Cognitive impairment is found only in 7% of affected individuals and do not correlate with cranial MRI abnormalities. 18 Patients with intellectual disability and epilepsy were associated with bilateral occipital pachygyria 19 or dysplastic cortical changes affecting predominantly the occipital and temporal regions. 16 In view of the lack of common mutations and limited information on genotype-phenotype correlations in the LAMA2-related muscular dystrophy at the moment, there is still a need for IHC study for MDC1A in all patients with CMD. 19,20 The clinical utility of the mutation analysis was the earlier diagnosis of MDC1A as this facilitated timely avoidance of certain anaesthetic agents such as succinylcholine during induction of anaesthesia because of risk of hyperkalaemia and cardiac conduction abnormalities. 21 Prenatal diagnosis of a foetus with CMD may be made by immunostaining of chorionic villus sample. With the availability of mutation analysis, prenatal diagnosis can be an option for some families and preimplantation genetic diagnosis can be made for families where termination of pregnancy is not an option. 22 With more information on the phenotype-genotype correlation, a more accurate genetic counselling can be offered for affected families with an autosomal recessive disorder. Mutation studies may also be useful as a confirmatory test in patients with atypical presentation, for example during early infancy where a muscle biopsy showed an inflammatory picture or cranial MRI showed 158

leukodystrophic findings or partial merosin deficiency. 23 These is a need to increase the awareness of medical practitioners on MDC1A in this region, to facilitate early diagnosis, genetic counselling and avoidance of certain anaesthetic agents for these patients. ACKNOWLEDGEMENTS Professor K.J. Goh received funding from the High Impact Research grant (HIR H20001-00-E00031). This research was also supported by Basic Science Research Program through the National Research Foundation of Korea (NRF) funded by Ministry of Education (2013R1A1A2005521). REFERENCES 1. Bönnemann CG, Rutkowski A, Mercuri E, Muntoni F. CMD Outcomes Consortium.; 173rd ENMC International Workshop: congenital muscular dystrophy outcome measures 5-7 March 2010, Naarden, The Netherlands. Neuromuscul Disord 2011; 21:513-22. 2. Wang CH, Bönnemann CG, Rutkowski A, et al. International Standard of Care Committee for Congenital Muscular Dystrophy: Consensus statement on standard of care for congenital muscular dystrophies. J Child Neurol 2010: 1559-81 3. Muntoni F, Voit T. The congenital muscular dystrophies in 2004: a century of exciting progress. Neuromuscul Disord 2004; 14:635-49. 4. Tezak Z, Prandini P, Boscaro M, et al. Clinical and molecular study in congenital muscular dystrophy with partial laminin alpha-2 (LAMA2) deficiency. Hum Mutat 2003; 21:103-11. 5. Tome FMS, Evangelista T, Leclerc A, et al. Congenital muscular dystrophy with merosin deficiency. Comp Rend Acad Sci (Paris) 1994; 317: 351-7. 6. Helbling-Leclerc A, Zhang X, Topaloglu H, et al. Mutations in the laminin alpha-2-chain gene (LAMA2) cause merosin-deficient congenital muscular dystrophy. Nature Genet 1995; 11:216-8. 7. Sunada Y, Edgar TS, Lotz BP, Rust RS, Campbell KP. Merosin-negative congenital muscular dystrophy associated with extensive brain abnormalities. Neurology 1995; 45:2084-9. 8. Darin N, Tulinius M. Neuromuscular disorders in childhood: a descriptive epidemiological study from western Sweden. Neuromuscul Disord 2000; 10:1-9. 9. Allamand V, Guicheney P. Merosin-deficient congenital muscular dystrophy, autosomal recessive (MDC1A, MIM#156225, LAMA2 gene coding for alpha2 chain of laminin). Eur J Hum Genet 2002; 10:91-4. 10. Fujii Y, Sugiura C, Fukuda C, Maegaki Y, Ohno K. Sequential neuroradiological and neurophysiological studies in a Japanese girl with merosin-deficient congenital muscular dystrophy. Brain Dev 2011; 33(2):140-4. 11. Chae JH, Lee JS, Hwang H, et al. Merosin-deficient congenital muscular dystrophy in Korea. Brain Dev 2009; 31(5):341-6. 12. Yuan J, Takashima H, Higuchi I, et al. Genetically confirmed patients with merosin-deficient congenital muscular dystrophy in China. Neuropediatrics 2008; 39(5):264-7. 13. Turner C, Mein R, Sharpe C, Love D. Merosindeficient congenital muscular dystrophy: A novel homozygous mutation in the laminin-2 gene. J Clin Neurosci 2015; 22(12):1983-5. 14. Bhowmik AD, Dalal AB, Matta D, Sundaram C, Aggarwal S. Targeted next generation sequencing identifies a novel deletion in LAMA2 gene in a merosindeficient congenital muscular dystrophy patient. Indian J Pediatr 2016; 83(4):354-5. 15. Girgenrath M, Beermann ML, Vishnudas VK, Homma S, Miller JB. Pathology is alleviated by doxycycline in a laminin-alpha2-null model of congenital muscular dystrophy. Ann Neurol 2009; 65:47-56. 16. Geranmayeh F, Clement E, Feng LH, et al. Genotypephenotype correlation in a large population of muscular dystrophy patients withlama2 mutations Neuromusc Disord 2010; 20:241-50. 17. Alkan A, Sigirci A, Kutlu R, Aslan M, Doganay S, Yakinci C. Merosin-negative congenital muscular dystrophy: diffusion-weighted imaging findings of brain. J Child Neurol 2007; 22:655-9. 18. Messina S, Bruno C, Moroni I, et al. Congenital muscular dystrophies with cognitive impairment. A population study. Neurology 2010; 75:898-903. 19. Jones KJ, Morgan G, Johnston H, et al. The expanding phenotype of laminin alpha-2 chain (merosin) abnormalities: case series and review. J Med Genet 2001; 38: 649-57. 20. Di Blasi C, Piga D, Brioschi P, et al. LAMA2 gene analysis in congenital muscular dystrophy: new mutations, prenatal diagnosis, and founder effect. Arch Neurol 2005; 62:1582-6. 21. Scrivener TA, Ross SM, Street NE, Webster RI, De Lima JC. A case series of general anesthesia in children with laminin alpha2 (merosin)- deficient congenital muscular dystrophy. Paediatr Anaesth 2014; 24(4):464-5. 22. Naom I, D Alessandro M, Sewry C, et al. The role of immunocytochemistry and linkage analysis in the prenatal diagnosis of merosin-deficient congenital muscular dystrophy. Hum Genet 1997; 99:535-40. 23. LAMA2 Mutation Database. Leiden Open Variation Database (LOVD) (http://www.dmd.nl/lama2_home. html) (Last accessed April 20 th 2015) 159