Identification of LAMA2 compound heterozygous variants: a case report
Case Report

Identification of LAMA2 compound heterozygous variants: a case report

Yingchao Ying1 ORCID logo, Jia Ye1, Jue Shen2, Guojuan Chen3, Kewen Jiang1 ORCID logo

1Department of Child Psychology, The Children’s Hospital Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China; 2Department of Neurology, The Children’s Hospital Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China; 3Department of Child Health, Shaoxing Shangyu Maternal and Child Health Hospital, Shaoxing, China

Contributions: (I) Conception and design: K Jiang; (II) Administrative support: K Jiang; (III) Provision of study materials or patients: J Shen; (IV) Collection and assembly of data: Y Ying; (V) Data analysis and interpretation: Y Ying, J Ye, G Chen; (VI) Manuscript writing: All authors; (VII) Final approval of manuscript: All authors.

Correspondence to: Kewen Jiang, MD, PhD. Department of Child Psychology, The Children’s Hospital Zhejiang University School of Medicine, National Clinical Research Center for Child Health, 3333 Binsheng Road, Binjiang District, Hangzhou 310051, China. Email: jiangkw_zju@zju.edu.cn.

Background: Laminin-α2 (LAMA2) chain-deficient muscular dystrophy (LAMA2-MD) is the most common congenital muscular dystrophy (CMD) in the world. Its main manifestations are muscle weakness and hypotonia that occur after birth or at early infancy.

Case Description: We reported a case of a 3-year-old and 6-month-old boy presented with delayed motor development, elevated creatine kinase (CK) levels, and abnormal white matter in the brain. Whole exome sequencing (WES) showed compound heterozygous variants of the LAMA2 gene. This case reports for the first time the compound heterozygous LAMA2 variants c.5476C>T (p.R1826*) (paternal inheritance) with c.2749 + 2dup (maternal inheritance), as both variants are interpreted as pathogenic/potentially pathogenic variants.

Conclusions: This study reports a novel heterozygous variant, including two pathogenic variants in the LAMA2 gene, and highlights the effectiveness of highly efficient exome sequencing applying in patients with undefined CMDs.

Keywords: Laminin-α2 (LAMA2); merosin-deficient congenital muscular dystrophy (merosin-deficient CMD); limb-girdle muscular dystrophy; whole exome sequencing (WES); case report


Submitted Feb 28, 2024. Accepted for publication May 29, 2024. Published online Jun 14, 2024.

doi: 10.21037/tp-24-62


Highlight box

Key findings

• Through exome sequencing, this study reports a novel heterozygous variant in the laminin-α2 (LAMA2) gene in a boy manifesting as congenital muscular dystrophy (CMD).

What is known and what is new?

LAMA2 chain-deficient muscular dystrophy is an autosomal recessive genetic disorder caused by variants in the LAMA2 gene leading to the decreased or absent production of protein LAMA2.

• This case reports for the first time the compound heterozygous LAMA2 variants c.5476C>T (p.R1826*) (paternal inheritance) with c.2749 + 2dup (maternal inheritance), as both variants are interpreted as pathogenic/likely pathogenic variants.

What is the implication, and what should change now?

• Whole exome sequencing is an effective method for diagnosing CMD, a helpful tool for genetic counseling and prenatal testing. In the future, it is necessary to evaluate the impact of identified variants on LAMA2 function through experiments to promote understanding of the CMD pathogenesis and personalized gene therapy for patients.


Introduction

Congenital muscular dystrophies (CMDs) are phenotypic heterogeneous diseases. Its main manifestations are muscle weakness and hypotonia that occur after birth or at early infancy (1). CMDs are not common, with an incidence of 0.82/100,000 in live births (1). According to previous reports, 37 genes have been identified to be associated with CMDs (2,3). Laminin-α2 (LAMA2) chain-deficient muscular dystrophy (LAMA2-MD) is the most common CMD in the world, which affects about 36.4% in China (4).

LAMA2-MD is an autosomal recessive genetic disorder caused by variants in the LAMA2 gene leading to the decreased or absent production of protein LAMA2, which leads to a deficiency of laminin-211 and/or laminin-221, resulting in a decrease in the strength and stability of skeletal muscle. Based on the degree of LAMA2 deficiency, LAMA2-MD varies from severe merosin-deficient CMD type 1A (MDC1A, OMIM # 607855) to milder late-onset limb-girdle MD-23 (LGMDR23, OMIM # 618138). Blood biochemical indicators such as elevated serum creatine kinase (CK) levels, immunohistochemistry results mainly from skin or muscle biopsies, and brain magnetic resonance imaging (MRI) findings such as white matter abnormalities can be used to diagnose MDC1A (5,6). It is currently recommended to use next-generation sequencing (NGS) for genetic testing in LAMA2-MD (7,8).

There are over 800 unique variants that have been reported in the LAMA2. Among them, a total of 666 pathogenic variants in the LAMA2 gene have been reported in the Human Gene Mutation Database (HGMD) Professional release 2023.4 (http://www.hgmd.cf.ac.uk/ac/gene.php?gene=LAMA2) (Table 1). Furthermore, according to the Leiden Open Variation Database (LOVD, v3.0), 2.64% cases with more than two variants and 30.53% individuals with two variants in the LAMA2 gene.

Table 1

Pathogenic mutations in the LAMA2 gene in HGMD

Mutation type Number of mutations Proportion (%)
Missense/nonsense 310 46.55
Splicing 106 15.92
Regulatory 0 0
Small deletions 131 19.67
Small insertions 49 7.36
Small indels 1 0.15
Gross deletions 50 7.51
Gross insertions/duplications 15 2.25
Complex rearrangements 4 0.60
Repeat variations 0 0
HGMD Professional 2023.4 total 666 100

LAMA2, laminin-α2; HGMD, Human Gene Mutation Database.

Here, we report a boy presenting a delayed motor development, and elevated CK levels, with compound heterozygous variants of the LAMA2 gene. We present this case in accordance with the CARE reporting checklist (available at https://tp.amegroups.com/article/view/10.21037/tp-24-62/rc).


Case presentation

The proband was a boy aged 3 years and 6 months who had been seeking medical attention for high CK levels. This boy can only walk when he was 15 months old. At the age of 3, there was a noticeable lack of running, limb weakness, inability to perform leg jumps, and poor stair walking, with good language development. There was no history of birth rescue, allergies, nor major illnesses. Screening for major genetic metabolic diseases in newborns was shown negative. He was the first and only child of the family and there was no family history of hereditary diseases. No obvious positive signs were found in the physical examination. Neurological examination showed cranial nerve negative, limb muscle strength grade IV, hypotonia, normal tendon reflex, abdominal wall reflex led out, negative Babinski sign, and mild hypertrophy of gastrocnemius muscle. Laboratory tests found that his serum CK level was as high as 987 U/L. Thyroid function test showed no abnormalities. TORCH test (panel for four serious infectious conditions in infants: toxoplasmosis, rubella, cytomegalovirus and herpes simplex virus) showed an increase of 105 U/mL (normal range, <14 U/mL) in cytomegalovirus antibody immunoglobulin G (IgG). The electromyogram (EMG) showed increased multiphase waves and shortened duration of motor units of limb muscles, indicating myogenic lesions. Brain MRI results showed symmetrical abnormal signals in bilateral deep white matter of the brain (Figure 1). Patients with white matter abnormalities may have a risk of epilepsy (9,10), but no prominent epileptiform activity was observed in the electroencephalogram (EEG) of this patient. No specific changes were found in the genetic metabolism map. A trio-based whole exome sequencing (WES) on genomic DNA was performed (Department of Screening Laboratory). The results showed compound heterozygous LAMA2 variants, c.5476C>T (p.R1826*) that was paternally inherited, and c.2749 + 2dup that was maternally inherited (Figure 2). Both variants were interpreted as pathogenic/likely pathogenic variants, suggesting a diagnosis of LAMA2-MD.

Figure 1 Brain MRI. Abnormal signal symmetry in the deep white matter of both sides of the brain with slight hypointensity on T1 (A), slight hyperintensity on T2 (B) and FLAIR (C). MRI, magnetic resonance imaging; FLAIR, fluid attenuated inversion recovery.
Figure 2 Information of LAMA2 mutation in the family of the proband. (A) Family map of gene mutation. (B) Next-generation sequencing of mutation. LAMA2, laminin-α2.

This study was reviewed and approved by the Ethics Committee of The Children’s Hospital Zhejiang University School of Medicine (Ref. 2024-IRB-0038). All procedures performed in this study were in accordance with the ethical standards of the institutional and/or national research committee(s) and with the Helsinki Declaration (as revised in 2013). Written informed consent was obtained from the participant’s legal guardian for the publication of this case report and accompanying images. A copy of the written consent is available for review by the editorial office of this journal.


Discussion

In addition to the delayed motor development of this case, other main diagnostic evidences were: elevated CK level, EMG-suggested myogenic lesions, and brain MRI-shown symmetrical abnormal signals in bilateral deep white matter. Importantly, the proband of this case inherited chromosomes from parents who were with different pathogenic variants in LAMA2 gene: c.5476C>T(p.R1826*) and c.2749 + 2dup (Figure 2), conforming the diagnosis of LAMA2-MD.

This is a relatively mild case of MDC1A, with the following phenotypic characteristics: (I) delayed diagnose until he came to the department of neurology at 3 years and 6 months old. He received routine follow-up at the local health care clinic and was not promptly referred to a higher-level hospital, despite suffering from early onset of hypotonia and inability to walk and exhibiting an obvious delayed motor development at the age of 3. (II) CK level is relatively low, as in severe cases of MDC1A, the serum CK level is usually between several thousand to ten thousand. This relatively low CK level indicates that the muscle damage is not very serious, so the symptoms displayed are also mild, which can easily lead to negligence. (III) No CMD-family history and neither parent shows the phenotype.

For genotype characteristics, the compound variants occurred in this case with them originating from the parents separately. In exon 38 of the LAMA2 gene, a nonsense variant c.5476C>T(p.R1826*) is inherited from the father. The identified rare LAMA2 c.5476C>T was pathogenic, classified as PSV1, PM3, PP3, and PP5 according to the American College of Medical Genetics and Genomics (ACMG) guidelines classification (11). And the variant is expected to lose function due to premature protein truncation or nonsense-mediated mRNA decay. Based on data from the Genome Aggregation Database (gnomAD), the LAMA2 c.5476C>T variant is observed in <0.0001 (7/251,122) of total alleles studied, and the frequency of the East Asian subgroup is 0.0001 (2/18,386). This variant was mainly detected in the compound heterozygous state (12-14). The c.2749 + 2dup is a splicing variant inherited from the mother and has been reported in previous studies (15,16). The splice donor site duplication (c.2749 + 2dup) may lead to splicing abnormalities. The identified rare LAMA2 c.2749 + 2dup was likely pathogenic classified as PM3, PP1, PP3, PP4 and PP5 according to the ACMG guidelines classification (11). GnomAD shows that the overall frequency of this variant in the population is 1/250,932 (with a frequency of 0/18,390 in the East Asian population), and it is not supported as a polymorphic locus. Results from a study by using spliceAI analysis suggest this variant may affect splicing (15), but it has not been experimentally investigated (17). Although this variant achieved a high spliceAI score of 0.99 for splice altering (15), it is also possible that the in frame/out of frame exon skips or the splice donor site changes to another location could lead to mild phenotypes in patients like this one (17). This variant is not related to complete loss-of-function of the gene (16) and is consistent with the observed in our case. Besides, a previous study reported that c.2749 + 2dup might be related to white matter abnormalities (16), as observed in this case of brain MRI. Due to the expression of LAMA2 in cerebral blood vessels, it may be important for the selective filtering ability of the blood-brain barrier. Dysfunction of LAMA2 may lead to impaired selective filtering, resulting in plasma component leakage and central nervous system damage (18).

In addition, according to ClinVar Database, there are currently five reported cases of LAMA2: c.5476C>T, and two reported cases of LAMA2: c.2749 + 2dup (Table 2) (12-16). Currently (by January 2024), the LAMA2-LOVD (v3.0), an open-source DNA variation database system, contains a total of 2,628 public reported LAMA2-related variants (871 unique) identified in a total of 1,703 individuals (https://databases.lovd.nl/shared/genes/LAMA2). There were 45 cases with more than two variants, and 520 individuals with two variants. This case reports for the first time the compound heterozygous LAMA2 variants c.5476C>T (p.R1826*) (paternal inheritance) with c.2749 + 2dup (maternal inheritance), as both variants are interpreted as pathogenic/likely pathogenic variants.

Table 2

Reported cases of the same variants in the LAMA2 gene with our case

Patient CK EMG Muscle biopsy MRI NGS Inheritance
P1 (13) N/A N/A + N/A c.8038delG N/A
c.5476C>T N/A
P2 (14) + + + + c.2958G>A Paternal
c.5476C>T Maternal
P3 (14) + + + c.7987delG Paternal
c.5476C>T Maternal
P4/P5 (12) + + + + c.5525C>T Paternal
c.43511-1G>C Maternal
P6 (15) + + N/A + c.2749 + 2dup N/A
c.8689C>T N/A
P7 (16) + N/A N/A + c.2749 + 2dup Paternal
Maternal
c.1338_1339del
P8 (our case) + + N/A + c.5476C>T Paternal
c.2749 + 2dup Maternal

LAMA2, laminin-α2; CK, creatine kinase; EMG, electromyogram; MRI, magnetic resonance imaging; NGS, next-generation sequencing; N/A, not available.

Furthermore, diagnosis requires muscle biopsy, immunohistochemical staining, or genetic testing. It was reported that there was no merosin staining in MDC1A, but low levels of residual merosin can be detected in LGMDR23 (15,19). Unfortunately, muscle biopsy and immunohistochemistry staining had not been performed in this case. Thyroid function test is to exclude hypothyroid myopathy. TORCH testing is to exclude CK elevation caused by infectious myositis. The positive cytomegalovirus IgG antibody indicated that the patient had previously been infected with cytomegalovirus and not related to the current increase in CK.

Altogether, WES is an effective method for diagnosing CMD, a helpful tool for genetic counseling and prenatal testing. In the future, it is necessary to evaluate the impact of identified variants on LAMA2 function through experiments to promote understanding of the CMD pathogenesis and personalized gene therapy for patients.


Conclusions

The dual pathogenic variants in the LAMA2 gene produce a series of CMDs (LAMA2-MDs). By using WES, we identified two variants in the LAMA2 gene in a boy. The variant c.5476C>T (p.R1826*) is expected to lose function due to premature protein truncation or nonsense-mediated mRNA decay and is classified as pathogenic. The splicing variant c.2749 + 2dup is also interpreted as pathogenic variants. The predominant clinical manifestations observed in this patient included delayed motor development, elevated CK levels, and abnormal white matter in the brain. This report emphasizes the effectiveness of WES application in undefined CMD patients.


Acknowledgments

We thank the patient and his parents for their cooperation.

Funding: This study was supported by the National Natural Science Foundation of China (Nos. 81871012 and 81571263, to K.J.), and by the Science and Technology Plan of Zhejiang Provincial Department of Health (No. 2022519065, to G.C.).


Footnote

Reporting Checklist: The authors have completed the CARE reporting checklist. Available at https://tp.amegroups.com/article/view/10.21037/tp-24-62/rc

Peer Review File: Available at https://tp.amegroups.com/article/view/10.21037/tp-24-62/prf

Conflicts of Interest: All authors have completed the ICMJE uniform disclosure form (available at https://tp.amegroups.com/article/view/10.21037/tp-24-62/coif). The authors have no conflicts of interest to declare.

Ethical Statement: The authors are accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved. This study was reviewed and approved by the Ethics Committee of The Children’s Hospital Zhejiang University School of Medicine (Ref. 2024-IRB-0038). All procedures performed in this study were in accordance with the ethical standards of the institutional and/or national research committee(s) and with the Helsinki Declaration (as revised in 2013). Written informed consent was obtained from the participant’s legal guardian for the publication of this case report and accompanying images. A copy of the written consent is available for review by the editorial office of this journal.

Open Access Statement: This is an Open Access article distributed in accordance with the Creative Commons Attribution-NonCommercial-NoDerivs 4.0 International License (CC BY-NC-ND 4.0), which permits the non-commercial replication and distribution of the article with the strict proviso that no changes or edits are made and the original work is properly cited (including links to both the formal publication through the relevant DOI and the license). See: https://creativecommons.org/licenses/by-nc-nd/4.0/.


References

  1. Pasrija D, Tadi P. Congenital Muscular Dystrophy. In: StatPearls. Treasure Island (FL): StatPearls Publishing; July 3, 2023.
  2. Zambon AA, Muntoni F. Congenital muscular dystrophies: What is new? Neuromuscul Disord 2021;31:931-42. [Crossref] [PubMed]
  3. Tran VK, Nguyen NL, Tran LNT, et al. Merosin-deficient congenital muscular dystrophy type 1a: detection of LAMA2 variants in Vietnamese patients. Front Genet 2023;14:1183663. [Crossref] [PubMed]
  4. Ge L, Zhang C, Wang Z, et al. Congenital muscular dystrophies in China. Clin Genet 2019;96:207-15. [Crossref] [PubMed]
  5. Previtali SC, Zambon AA. LAMA2 Neuropathies: Human Findings and Pathomechanisms From Mouse Models. Front Mol Neurosci 2020;13:60. [Crossref] [PubMed]
  6. Sarkozy A, Foley AR, Zambon AA, et al. LAMA2-Related Dystrophies: Clinical Phenotypes, Disease Biomarkers, and Clinical Trial Readiness. Front Mol Neurosci 2020;13:123. [Crossref] [PubMed]
  7. Saredi S, Gibertini S, Matalonga L, et al. Exome sequencing detects compound heterozygous nonsense LAMA2 mutations in two siblings with atypical phenotype and nearly normal brain MRI. Neuromuscul Disord 2019;29:376-80. [Crossref] [PubMed]
  8. El Kadiri Y, Ratbi I, Laarabi FZ, et al. Identification of a novel LAMA2 c.2217G > A, p.(Trp739*) mutation in a Moroccan patient with congenital muscular dystrophy: a case report. BMC Med Genomics 2021;14:113. [Crossref] [PubMed]
  9. Urquia-Osorio H, Pimentel-Silva LR, Rezende TJR, et al. Superficial and deep white matter diffusion abnormalities in focal epilepsies. Epilepsia 2022;63:2312-24. [Crossref] [PubMed]
  10. Mito R, Pedersen M, Pardoe H, et al. Exploring individual fixel-based white matter abnormalities in epilepsy. Brain Commun 2024;6:fcad352. [Crossref] [PubMed]
  11. 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. [Crossref] [PubMed]
  12. Naom I, D'Alessandro M, Sewry CA, et al. Laminin alpha 2-chain gene mutations in two siblings presenting with limb-girdle muscular dystrophy. Neuromuscul Disord 1998;8:495-501. [Crossref] [PubMed]
  13. Cauley ES, Pittman A, Mummidivarpu S, et al. Novel mutation identification and copy number variant detection via exome sequencing in congenital muscular dystrophy. Mol Genet Genomic Med 2020;8:e1387. [Crossref] [PubMed]
  14. Xiong H, Tan D, Wang S, et al. Genotype/phenotype analysis in Chinese laminin-α2 deficient congenital muscular dystrophy patients. Clin Genet 2015;87:233-43. [Crossref] [PubMed]
  15. Wang DZ, Li BH, Ma Q, et al. Novel compound heterozygous mutations of LAMA2-limb-girdle muscular dystrophy: A case report and literature review. Front Neurol 2023;14:1078151. [Crossref] [PubMed]
  16. Yamamoto-Shimojima K, Ono H, Imaizumi T, et al. Novel LAMA2 variants identified in a patient with white matter abnormalities. Hum Genome Var 2020;7:16. [Crossref] [PubMed]
  17. Jaganathan K, Kyriazopoulou Panagiotopoulou S, McRae JF, et al. Predicting Splicing from Primary Sequence with Deep Learning. Cell 2019;176:535-548.e24. [Crossref] [PubMed]
  18. Miyagoe-Suzuki Y, Nakagawa M, Takeda S. Merosin and congenital muscular dystrophy. Microsc Res Tech 2000;48:181-91. [Crossref] [PubMed]
  19. Løkken N, Born AP, Duno M, et al. LAMA2-related myopathy: Frequency among congenital and limb-girdle muscular dystrophies. Muscle Nerve 2015;52:547-53. [Crossref] [PubMed]
Cite this article as: Ying Y, Ye J, Shen J, Chen G, Jiang K. Identification of LAMA2 compound heterozygous variants: a case report. Transl Pediatr 2024;13(6):1001-1006. doi: 10.21037/tp-24-62

Download Citation