Novel frameshift variant in MYL2 reveals molecular differences between dominant and recessive forms of hypertrophic cardiomyopathy

Sathiya N Manivannan, Sihem Darouich, Aida Masmoudi, David Gordon, Gloria Zender, Zhe Han, Sara Fitzgerald-Butt, Peter White, Kim L McBride, Maher Kharrat, Vidu Garg, Sathiya N Manivannan, Sihem Darouich, Aida Masmoudi, David Gordon, Gloria Zender, Zhe Han, Sara Fitzgerald-Butt, Peter White, Kim L McBride, Maher Kharrat, Vidu Garg

Abstract

Hypertrophic cardiomyopathy (HCM) is characterized by thickening of the ventricular muscle without dilation and is often associated with dominant pathogenic variants in cardiac sarcomeric protein genes. Here, we report a family with two infants diagnosed with infantile-onset HCM and mitral valve dysplasia that led to death before one year of age. Using exome sequencing, we discovered that one of the affected children had a homozygous frameshift variant in Myosin light chain 2 (MYL2:NM_000432.3:c.431_432delCT: p.Pro144Argfs*57;MYL2-fs), which alters the last 20 amino acids of the protein and is predicted to impact the most C-terminal of the three EF-hand domains in MYL2. The parents are unaffected heterozygous carriers of the variant and the variant is absent in control cohorts from gnomAD. The absence of the phenotype in carriers and the infantile presentation of severe HCM is in contrast to HCM associated with dominant MYL2 variants. Immunohistochemical analysis of the ventricular muscle of the deceased patient with the MYL2-fs variant showed a marked reduction of MYL2 expression compared to an unaffected control. In vitro overexpression studies further indicate that the MYL2-fs variant is actively degraded. In contrast, an HCM-associated missense variant (MYL2:p.Gly162Arg) and three other MYL2 stop-gain variants (p.E22*, p.K62*, p.E97*) that result in loss of the EF domains are stably expressed but show impaired localization. The degradation of the MYL2-fs can be rescued by inhibiting the cell's proteasome function supporting a post-translational effect of the variant. In vivo rescue experiments with a Drosophila MYL2-homolog (Mlc2) knockdown model indicate that neither the MYL2-fs nor the MYL2:p.Gly162Arg variant supports normal cardiac function. The tools that we have generated provide a rapid screening platform for functional assessment of variants of unknown significance in MYL2. Our study supports an autosomal recessive model of inheritance for MYL2 loss-of-function variants in infantile HCM and highlights the variant-specific molecular differences found in MYL2-associated cardiomyopathy.

Conflict of interest statement

The authors have declared that no competing interests exist.

Figures

Fig 1. Identification of a novel homozygous…
Fig 1. Identification of a novel homozygous frameshift variant in a proband with infantile hypertrophic cardiomyopathy.
(A) Pedigree of the family with multiple infant deaths due to early-onset cardiac disease. The consanguinity and age of the parents are also shown. The proband (fs/fs) was homozygous for the MYL2-fs allele while the parents were found to be heterozygous (fs/wt) carriers. The status of the MYL2 allele in siblings is unknown (Unk.). Proband is marked by an arrowhead. (B) Table summarizing key clinical findings of the four siblings (IV:1 –IV:4). (C) Dissection of the post-mortem heart from the proband shows severe biatrial dilatation along with hypertrophy of the right ventricle (left). Severe mitral valve dysplasia and hypertrophy of the left ventricle with a small ventricular cavity is also noted (right). RA- right atrium; TV- tricuspid valve; RV- right ventricle; LA- left atrium; MV- mitral valve; LV- left ventricle (D-top) The genomic locus of the MYL2 gene shows the frameshift variant identified in the proband (highlighted in red), which is located in the last exon of the gene. (D-bottom) Sequence chromatograms from Sanger sequencing of control (MYL2 wt), the proband (homozygous fs variant: MYL2:NM_000432.3:c.431_432delCT) and the parents (heterozygous fs).
Fig 2. Analysis of the ventricular myocardium…
Fig 2. Analysis of the ventricular myocardium of the proband shows characteristic features of hypertrophic cardiomyopathy (HCM) and molecular consequences of the variant.
(A) Histochemical analysis of the ventricular myocardium from the proband by H&E staining shows myocyte disarray as compared to control. Masson’s trichrome staining of the proband’s ventricular myocardium shows increased fibrosis (stained with blue). (B) Immunohistochemical analysis of MYL2 expression in the proband’s ventricular myocardium shows a remarkable reduction in the expression of MYL2 as compared to control. The antibody used targets residues at the N-terminal region of the protein (S4 Fig). Cardiac troponin I (TNNI3) was detected at comparable levels in the control and the proband myocardium. The unstained areas correspond to fibrosis. (C) Flow diagram showing the steps used to analyze MYL2 mRNA expression in the ventricular myocardium of the proband. An RT-PCR product of the expected size of MYL2 mRNA is detected in the control and proband tissue, and not in the negative (No-RT) controls. Sanger sequencing of the product from the proband shows the MYL2 mRNA transcript with the dinucleotide deletion.
Fig 3. In vitro analysis of stability…
Fig 3. In vitro analysis of stability and localization of MYL2 variants.
(A) Schematic of MYL2 protein showing N-terminal and C-terminal EF-hand domains along with Serine 15 residue that is target of MLCK phosphorylation, three stop-gain variants (red octagons), the frameshift variant identified in the proband (red block showing extension of C-terminal end) and missense variant associated with HCM (orange oval) in the C-terminal domain. The frameshift mutation changes the canonical MYL2 protein sequence (‘Ref.’, black text) from residue 144 onwards, modifying the last 20 amino acids of the protein in addition to adding 36 non-canonical amino acids to the C-terminus (‘fs’, red text). (B) Western blot image showing overexpression of EGFP-tagged MYL2 wildtype (wt) and variant proteins. The expression of the protein harboring the frameshift variant is significantly reduced while the other stop-gain variants do not show a significant change in expression. Analysis of mCherry signal by western blot does not show any significant changes between MYL2 wt and tested variants. Loading controls: mCherry and Actin. (C) In H9c2 cells, immunofluorescence analysis of EGFP tagged MYL2 wt and tested variants (green) that overexpress mCherry from the same transcript (red) is shown. EGFP-tagged MYL2-wt localizes to the cytoskeleton, while stop-gain variants (E22*, K62*, E97*) do not localize to the cytoskeleton and are observed in a diffuse pattern. EGFP signal from the frameshift variant (fs) is significantly reduced. Localization of the missense (G162R) variant is in a pattern similar to the wt. mCherry signal from the transfected cells does not change between variants. (D) Immunofluorescence images of EGFP-tagged MYL2-wt, MYL2-fs, and MYL2-G162R in MG-132 treated H9c2 cells show the rescue of MYL2-fs variant signal following MG-132 treatment (western blot of the rescue shown in S3B Fig). Scale bar indicates 50 μm length. Quantitation of signal from the western and immunofluorescence experiments is provided in S3B Fig.
Fig 4. In vivo functional analysis of…
Fig 4. In vivo functional analysis of MYL2 variants using Drosophila Mlc2 substitution assay in the heart.
(A) Schematic shows Hand-enhancer driven GAL4 (Hand-GAL4) that was used to knockdown Mlc2 expression by RNAi in the heart and to overexpress human MYL2-wt and variant cDNAs for functional substitution. In parallel, the Hand-GAL4 drives the expression of UAS-CD8.mCherry, which facilitates the visualization of the heart. (B-i) Immunofluorescence image of the Drosophila heart from UAS-CD8.mCherry. Hand-GAL4 animals show expression of mCherry in the heart chambers and aorta. (B-ii) Magnified image of the A7-A8 posterior denticle band region showing expression of mCherry in both cardiomyocytes (arrowheads) as well as pericardial cells (arrows). The expression of pericardin is shown (green, B-iii). Actin expression (blue, B-iv) shows neighboring skeletal muscles (actin is stained with phalloidin). (B-v) Merged image of the three channels. No expression of mCherry was detected in the skeletal muscle. (C) Graph showing percent of progeny that emerge as adults compared to expected numbers following Mlc2 RNAi knockdown using Hand-GAL4, and rescue by overexpression of human MYL2-wildtype (wt) and variants. Partial rescue of the lethality is observed with wt MYL2 cDNA overexpression while no significant difference is observed for the tested variants. Significance is analyzed using Brown-Forsythe and Welch ANOVA across all samples followed by multiple comparisons using Games-Howell multiple comparison test. Significance is indicated using * adj. P-Value < 0.05, ** adj. P-Value < 0.01, **** adj. P-Value < 0.0001. (D) Image showing the rhythmic pattern of cardiac contraction that was observed using UAS-CD8-mCherry reporter driven by Hand-GAL4. Diastolic width is shown by ‘D’, and systolic width by ‘S’. The formula used to calculate fractional shortening is shown. (E) Interleaved scatter plot showing fractional shortening across different genotypes (S1–S5 Movies). Fractional shortening was significantly reduced due to Mlc2 RNAi knockdown which is partially rescued by overexpression of wt human MYL2. The missense and fs variants showed no significant improvement in fractional shortening compared to the wt. Significance was calculated using two-way ANOVA followed by Tukey’s multiple comparison tests. Multiplicity adjusted P-Values of Tukey’s comparison are indicated as * adj. P-Value < 0.05, ** adj. P-Value < 0.01, *** adj. P-Value < 0.001, **** adj.P-Value < 0.0001.

References

    1. Richardson P, McKenna W, Bristow M, Maisch B, Mautner B, O'Connell J, et al. Report of the 1995 World Health Organization/International Society and Federation of Cardiology Task Force on the Definition and Classification of cardiomyopathies. Circulation. 1996;93(5):841–2. Epub 1996/03/01. 10.1161/01.cir.93.5.841 .
    1. Fatkin D, Seidman CE, Seidman JG. Genetics and disease of ventricular muscle. Cold Spring Harb Perspect Med. 2014;4(1):a021063 Epub 2014/01/05. 10.1101/cshperspect.a021063
    1. Regnier M. Mechanistic complexity of contractile dysfunction in hypertrophic cardiomyopathy. J Gen Physiol. 2018;150(8):1051–3. Epub 2018/07/25. 10.1085/jgp.201812091
    1. Elliott P, McKenna WJ. Hypertrophic cardiomyopathy. The Lancet. 2004;363(9424):1881–91. 10.1016/S0140-6736(04)16358-7.
    1. Keren A, Syrris P, McKenna WJ. Hypertrophic cardiomyopathy: the genetic determinants of clinical disease expression. Nat Clin Pract Cardiovasc Med. 2008;5(3):158–68. Epub 2008/01/30. 10.1038/ncpcardio1110 .
    1. Maron BJ, Gardin JM, Flack JM, Gidding SS, Kurosaki TT, Bild DE. Prevalence of hypertrophic cardiomyopathy in a general population of young adults. Echocardiographic analysis of 4111 subjects in the CARDIA Study. Coronary Artery Risk Development in (Young) Adults. Circulation. 1995;92(4):785–9. Epub 1995/08/15. 10.1161/01.cir.92.4.785 .
    1. Seidman JG, Seidman C. The genetic basis for cardiomyopathy: from mutation identification to mechanistic paradigms. Cell. 2001;104(4):557–67. Epub 2001/03/10. 10.1016/s0092-8674(01)00242-2 .
    1. Maron BJ, Doerer JJ, Haas TS, Tierney DM, Mueller FO. Sudden deaths in young competitive athletes: analysis of 1866 deaths in the United States, 1980–2006. Circulation. 2009;119(8):1085–92. Epub 2009/02/18. 10.1161/CIRCULATIONAHA.108.804617 .
    1. Elliott PM, Gimeno JR, Thaman R, Shah J, Ward D, Dickie S, et al. Historical trends in reported survival rates in patients with hypertrophic cardiomyopathy. Heart. 2006;92(6):785–91. Epub 2005/10/12. 10.1136/hrt.2005.068577
    1. Elliott PM, Poloniecki J, Dickie S, Sharma S, Monserrat L, Varnava A, et al. Sudden death in hypertrophic cardiomyopathy: identification of high risk patients. J Am Coll Cardiol. 2000;36(7):2212–8. Epub 2000/12/29. 10.1016/s0735-1097(00)01003-2 .
    1. Fatkin D, Graham RM. Molecular mechanisms of inherited cardiomyopathies. Physiol Rev. 2002;82(4):945–80. Epub 2002/09/25. 10.1152/physrev.00012.2002 .
    1. McKenna WJ, Kleinebenne A, Nihoyannopoulos P, Foale R. Echocardiographic measurement of right ventricular wall thickness in hypertrophic cardiomyopathy: relation to clinical and prognostic features. J Am Coll Cardiol. 1988;11(2):351–8. Epub 1988/02/01. 10.1016/0735-1097(88)90101-5 .
    1. Varnava AM, Elliott PM, Mahon N, Davies MJ, McKenna WJ. Relation between myocyte disarray and outcome in hypertrophic cardiomyopathy. Am J Cardiol. 2001;88(3):275–9. Epub 2001/07/27. 10.1016/s0002-9149(01)01640-x .
    1. Seidman CE, Seidman JG. Identifying sarcomere gene mutations in hypertrophic cardiomyopathy: a personal history. Circ Res. 2011;108(6):743–50. Epub 2011/03/19. 10.1161/CIRCRESAHA.110.223834
    1. Marian AJ, Roberts R. The molecular genetic basis for hypertrophic cardiomyopathy. J Mol Cell Cardiol. 2001;33(4):655–70. Epub 2001/03/29. 10.1006/jmcc.2001.1340
    1. Van Driest SL, Vasile VC, Ommen SR, Will ML, Tajik AJ, Gersh BJ, et al. Myosin binding protein C mutations and compound heterozygosity in hypertrophic cardiomyopathy. J Am Coll Cardiol. 2004;44(9):1903–10. Epub 2004/11/03. 10.1016/j.jacc.2004.07.045 .
    1. Charron P, Komajda M. Molecular genetics in hypertrophic cardiomyopathy: towards individualized management of the disease. Expert Rev Mol Diagn. 2006;6(1):65–78. Epub 2005/12/20. 10.1586/14737159.6.1.65 .
    1. Ho CY, Seidman CE. A contemporary approach to hypertrophic cardiomyopathy. Circulation. 2006;113(24):e858–62. Epub 2006/06/21. 10.1161/CIRCULATIONAHA.105.591982 .
    1. Monasky MM, Ciconte G, Anastasia L, Pappone C. Commentary: Next Generation Sequencing and Linkage Analysis for the Molecular Diagnosis of a Novel Overlapping Syndrome Characterized by Hypertrophic Cardiomyopathy and Typical Electrical Instability of Brugada Syndrome. Front Physiol. 2017;8:1056 Epub 2018/01/10. 10.3389/fphys.2017.01056
    1. Mango R, Luchetti A, Sangiuolo R, Ferradini V, Briglia N, Giardina E, et al. Next Generation Sequencing and Linkage Analysis for the Molecular Diagnosis of a Novel Overlapping Syndrome Characterized by Hypertrophic Cardiomyopathy and Typical Electrical Instability of Brugada Syndrome. Circ J. 2016;80(4):938–49. Epub 2016/03/11. 10.1253/circj.CJ-15-0685 .
    1. Glotov AS, Kazakov SV, Zhukova EA, Alexandrov AV, Glotov OS, Pakin VS, et al. Targeted next-generation sequencing (NGS) of nine candidate genes with custom AmpliSeq in patients and a cardiomyopathy risk group. Clin Chim Acta. 2015;446:132–40. Epub 2015/04/22. 10.1016/j.cca.2015.04.014 .
    1. Millat G, Chanavat V, Rousson R. Evaluation of a new NGS method based on a custom AmpliSeq library and Ion Torrent PGM sequencing for the fast detection of genetic variations in cardiomyopathies. Clin Chim Acta. 2014;433:266–71. Epub 2014/04/12. 10.1016/j.cca.2014.03.032 .
    1. Zhao Y, Feng Y, Ding X, Dong S, Zhang H, Ding J, et al. Identification of a novel hypertrophic cardiomyopathy-associated mutation using targeted next-generation sequencing. Int J Mol Med. 2017;40(1):121–9. Epub 2017/05/13. 10.3892/ijmm.2017.2986
    1. Lopes LR, Zekavati A, Syrris P, Hubank M, Giambartolomei C, Dalageorgou C, et al. Genetic complexity in hypertrophic cardiomyopathy revealed by high-throughput sequencing. J Med Genet. 2013;50(4):228–39. Epub 2013/02/12. 10.1136/jmedgenet-2012-101270
    1. Hershberger RE, Givertz MM, Ho CY, Judge DP, Kantor PF, McBride KL, et al. Genetic evaluation of cardiomyopathy: a clinical practice resource of the American College of Medical Genetics and Genomics (ACMG). Genet Med. 2018;20(9):899–909. Epub 2018/06/16. 10.1038/s41436-018-0039-z .
    1. Ingles J, Goldstein J, Thaxton C, Caleshu C, Corty EW, Crowley SB, et al. Evaluating the Clinical Validity of Hypertrophic Cardiomyopathy Genes. Circ Genom Precis Med. 2019;12(2):e002460 Epub 2019/01/27. 10.1161/CIRCGEN.119.002460
    1. Ramensky V, Bork P, Sunyaev S. Human non-synonymous SNPs: server and survey. Nucleic Acids Res. 2002;30(17):3894–900. Epub 2002/08/31. 10.1093/nar/gkf493
    1. Rentzsch P, Witten D, Cooper GM, Shendure J, Kircher M. CADD: predicting the deleteriousness of variants throughout the human genome. Nucleic Acids Res. 2019;47(D1):D886–D94. Epub 2018/10/30. 10.1093/nar/gky1016
    1. Garber M, Guttman M, Clamp M, Zody MC, Friedman N, Xie X. Identifying novel constrained elements by exploiting biased substitution patterns. Bioinformatics. 2009;25(12):i54–62. Epub 2009/05/30. 10.1093/bioinformatics/btp190
    1. Shihab HA, Gough J, Cooper DN, Stenson PD, Barker GL, Edwards KJ, et al. Predicting the functional, molecular, and phenotypic consequences of amino acid substitutions using hidden Markov models. Hum Mutat. 2013;34(1):57–65. Epub 2012/10/04. 10.1002/humu.22225
    1. Siepel A, Haussler D. Phylogenetic estimation of context-dependent substitution rates by maximum likelihood. Mol Biol Evol. 2004;21(3):468–88. Epub 2003/12/09. 10.1093/molbev/msh039 .
    1. Davydov EV, Goode DL, Sirota M, Cooper GM, Sidow A, Batzoglou S. Identifying a high fraction of the human genome to be under selective constraint using GERP++. PLoS Comput Biol. 2010;6(12):e1001025 Epub 2010/12/15. 10.1371/journal.pcbi.1001025
    1. Richards S, Aziz N, Bale S, Bick D, Das S, Gastier-Foster J, 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(5):405–24. Epub 2015/03/06. 10.1038/gim.2015.30
    1. Strande NT, Brnich SE, Roman TS, Berg JS. Navigating the nuances of clinical sequence variant interpretation in Mendelian disease. Genet Med. 2018;20(9):918–26. Epub 2018/07/11. 10.1038/s41436-018-0100-y .
    1. Claes GR, van Tienen FH, Lindsey P, Krapels IP, Helderman-van den Enden AT, Hoos MB, et al. Hypertrophic remodelling in cardiac regulatory myosin light chain (MYL2) founder mutation carriers. Eur Heart J. 2016;37(23):1815–22. Epub 2015/10/27. 10.1093/eurheartj/ehv522 .
    1. Szczesna D. Regulatory light chains of striated muscle myosin. Structure, function and malfunction. Curr Drug Targets Cardiovasc Haematol Disord. 2003;3(2):187–97. Epub 2003/05/29. 10.2174/1568006033481474 .
    1. Poetter K, Jiang H, Hassanzadeh S, Master SR, Chang A, Dalakas MC, et al. Mutations in either the essential or regulatory light chains of myosin are associated with a rare myopathy in human heart and skeletal muscle. Nat Genet. 1996;13(1):63–9. Epub 1996/05/01. 10.1038/ng0596-63 .
    1. Alvarez-Acosta L, Mazzanti A, Fernández X, Ortí M, Barriales-Villa R, García D, et al. Regulatory light chain (MYL2) mutations in familial hypertrophic cardiomyopathy. J Cardiovasc Dis. 2014;2:82–90.
    1. Olivotto I, Girolami F, Ackerman MJ, Nistri S, Bos JM, Zachara E, et al. Myofilament protein gene mutation screening and outcome of patients with hypertrophic cardiomyopathy. Mayo Clin Proc. 2008;83(6):630–8. Epub 2008/06/06. 10.4065/83.6.630 .
    1. Santos S, Marques V, Pires M, Silveira L, Oliveira H, Lanca V, et al. High resolution melting: improvements in the genetic diagnosis of hypertrophic cardiomyopathy in a Portuguese cohort. BMC Med Genet. 2012;13:17 Epub 2012/03/21. 10.1186/1471-2350-13-17
    1. Andersen PS, Havndrup O, Hougs L, Sorensen KM, Jensen M, Larsen LA, et al. Diagnostic yield, interpretation, and clinical utility of mutation screening of sarcomere encoding genes in Danish hypertrophic cardiomyopathy patients and relatives. Hum Mutat. 2009;30(3):363–70. Epub 2008/11/28. 10.1002/humu.20862 .
    1. Garcia-Pavia P, Vazquez ME, Segovia J, Salas C, Avellana P, Gomez-Bueno M, et al. Genetic basis of end-stage hypertrophic cardiomyopathy. Eur J Heart Fail. 2011;13(11):1193–201. Epub 2011/09/08. 10.1093/eurjhf/hfr110 .
    1. Burns C, Bagnall RD, Lam L, Semsarian C, Ingles J. Multiple Gene Variants in Hypertrophic Cardiomyopathy in the Era of Next-Generation Sequencing. Circ Cardiovasc Genet. 2017;10(4). Epub 2017/08/10. 10.1161/CIRCGENETICS.116.001666 .
    1. Geeves MA. Stretching the lever-arm theory. Nature. 2002;415(6868):129–31. Epub 2002/01/24. 10.1038/415129a .
    1. Sheikh F, Lyon RC, Chen J. Functions of myosin light chain-2 (MYL2) in cardiac muscle and disease. Gene. 2015;569(1):14–20. Epub 2015/06/16. 10.1016/j.gene.2015.06.027
    1. Yu H, Chakravorty S, Song W, Ferenczi MA. Phosphorylation of the regulatory light chain of myosin in striated muscle: methodological perspectives. European Biophysics Journal. 2016;45(8):779–805. 10.1007/s00249-016-1128-z
    1. Sitbon YH, Yadav S, Kazmierczak K, Szczesna‐Cordary D. Insights into myosin regulatory and essential light chains: a focus on their roles in cardiac and skeletal muscle function, development and disease. Journal of Muscle Research and Cell Motility. 2019. 10.1007/s10974-019-09517-x
    1. Grey C, Mery A, Puceat M. Fine-tuning in Ca2+ homeostasis underlies progression of cardiomyopathy in myocytes derived from genetically modified embryonic stem cells. Hum Mol Genet. 2005;14(10):1367–77. Epub 2005/04/15. 10.1093/hmg/ddi146 .
    1. Kampourakis T, Sun YB, Irving M. Myosin light chain phosphorylation enhances contraction of heart muscle via structural changes in both thick and thin filaments. Proc Natl Acad Sci U S A. 2016;113(21):E3039–47. Epub 2016/05/11. 10.1073/pnas.1602776113
    1. Zhou Z, Huang W, Liang J, Szczesna-Cordary D. Molecular and Functional Effects of a Splice Site Mutation in the MYL2 Gene Associated with Cardioskeletal Myopathy and Early Cardiac Death in Infants. Front Physiol. 2016;7:240 Epub 2016/07/06. 10.3389/fphys.2016.00240
    1. Szczesna-Cordary D, Guzman G, Ng SS, Zhao J. Familial hypertrophic cardiomyopathy-linked alterations in Ca2+ binding of human cardiac myosin regulatory light chain affect cardiac muscle contraction. J Biol Chem. 2004;279(5):3535–42. Epub 2003/11/05. 10.1074/jbc.M307092200 .
    1. Yadav S, Kazmierczak K, Liang J, Sitbon YH, Szczesna-Cordary D. Phosphomimetic-mediated in vitro rescue of hypertrophic cardiomyopathy linked to R58Q mutation in myosin regulatory light chain. FEBS J. 2019;286(1):151–68. Epub 2018/11/16. 10.1111/febs.14702
    1. Yuan CC, Kazmierczak K, Liang J, Zhou Z, Yadav S, Gomes AV, et al. Sarcomeric perturbations of myosin motors lead to dilated cardiomyopathy in genetically modified MYL2 mice. Proc Natl Acad Sci U S A. 2018;115(10):E2338–E47. Epub 2018/02/22. 10.1073/pnas.1716925115
    1. Kampourakis T, Ponnam S, Irving M. Hypertrophic cardiomyopathy mutation R58Q in the myosin regulatory light chain perturbs thick filament-based regulation in cardiac muscle. J Mol Cell Cardiol. 2018;117:72–81. Epub 2018/02/17. 10.1016/j.yjmcc.2018.02.009
    1. Szczesna D, Ghosh D, Li Q, Gomes AV, Guzman G, Arana C, et al. Familial hypertrophic cardiomyopathy mutations in the regulatory light chains of myosin affect their structure, Ca2+ binding, and phosphorylation. J Biol Chem. 2001;276(10):7086–92. Epub 2000/12/05. 10.1074/jbc.M009823200 .
    1. Chen J, Kubalak SW, Minamisawa S, Price RL, Becker KD, Hickey R, et al. Selective requirement of myosin light chain 2v in embryonic heart function. J Biol Chem. 1998;273(2):1252–6. Epub 1998/02/14. 10.1074/jbc.273.2.1252 .
    1. Chen Z, Huang W, Dahme T, Rottbauer W, Ackerman MJ, Xu X. Depletion of zebrafish essential and regulatory myosin light chains reduces cardiac function through distinct mechanisms. Cardiovasc Res. 2008;79(1):97–108. Epub 2008/03/18. 10.1093/cvr/cvn073
    1. Sanbe A, Fewell JG, Gulick J, Osinska H, Lorenz J, Hall DG, et al. Abnormal cardiac structure and function in mice expressing nonphosphorylatable cardiac regulatory myosin light chain 2. J Biol Chem. 1999;274(30):21085–94. Epub 1999/07/20. 10.1074/jbc.274.30.21085 .
    1. Tohtong R, Yamashita H, Graham M, Haeberle J, Simcox A, Maughan D. Impairment of muscle function caused by mutations of phosphorylation sites in myosin regulatory light chain. Nature. 1995;374(6523):650–3. Epub 1995/04/13. 10.1038/374650a0 .
    1. Szczesna-Cordary D, Guzman G, Zhao J, Hernandez O, Wei J, Diaz-Perez Z. The E22K mutation of myosin RLC that causes familial hypertrophic cardiomyopathy increases calcium sensitivity of force and ATPase in transgenic mice. J Cell Sci. 2005;118(Pt 16):3675–83. Epub 2005/08/04. 10.1242/jcs.02492 .
    1. Huang W, Liang J, Yuan CC, Kazmierczak K, Zhou Z, Morales A, et al. Novel familial dilated cardiomyopathy mutation in MYL2 affects the structure and function of myosin regulatory light chain. FEBS J. 2015;282(12):2379–93. Epub 2015/04/01. 10.1111/febs.13286
    1. Weterman MA, Barth PG, van Spaendonck-Zwarts KY, Aronica E, Poll-The BT, Brouwer OF, et al. Recessive MYL2 mutations cause infantile type I muscle fibre disease and cardiomyopathy. Brain. 2013;136(Pt 1):282–93. Epub 2013/02/01. 10.1093/brain/aws293 .
    1. Kelly BJ, Fitch JR, Hu Y, Corsmeier DJ, Zhong H, Wetzel AN, et al. Churchill: an ultra-fast, deterministic, highly scalable and balanced parallelization strategy for the discovery of human genetic variation in clinical and population-scale genomics. Genome Biol. 2015;16:6 Epub 2015/01/21. 10.1186/s13059-014-0577-x
    1. Kimes BW, Brandt BL. Properties of a clonal muscle cell line from rat heart. Exp Cell Res. 1976;98(2):367–81. Epub 1976/03/15. 10.1016/0014-4827(76)90447-x .
    1. Thibaudeau TA, Smith DM. A Practical Review of Proteasome Pharmacology. Pharmacol Rev. 2019;71(2):170–97. Epub 2019/03/15. 10.1124/pr.117.015370
    1. Moore JR, Dickinson MH, Vigoreaux JO, Maughan DW. The effect of removing the N-terminal extension of the Drosophila myosin regulatory light chain upon flight ability and the contractile dynamics of indirect flight muscle. Biophys J. 2000;78(3):1431–40. Epub 2000/02/29. 10.1016/S0006-3495(00)76696-3
    1. Marttila M, Win W, Al-Ghamdi F, Abdel-Hamid HZ, Lacomis D, Beggs AH. MYL2-associated congenital fiber-type disproportion and cardiomyopathy with variants in additional neuromuscular disease genes; the dilemma of panel testing. Cold Spring Harb Mol Case Stud. 2019;5(4). Epub 2019/05/28. 10.1101/mcs.a004184 .
    1. MYL2 myosin, light chain 2, regulatory, cardiac, slow Dataset gnomAD v2.1.1 gnomAD SVs [Internet]. 2019 [cited 8/7/2019]. Available from: .
    1. Burghardt TP, Sikkink LA. Regulatory light chain mutants linked to heart disease modify the cardiac myosin lever arm. Biochemistry. 2013;52(7):1249–59. Epub 2013/01/25. 10.1021/bi301500d
    1. Wang Y, Wang Z, Yang Q, Zou Y, Zhang H, Yan C, et al. Autosomal recessive transmission of MYBPC3 mutation results in malignant phenotype of hypertrophic cardiomyopathy. PLoS One. 2013;8(6):e67087 Epub 2013/07/11. 10.1371/journal.pone.0067087
    1. Zaleta-Rivera K, Dainis A, Ribeiro AJS, Sanchez Cordero P, Rubio G, Shang C, et al. Allele-Specific Silencing Ameliorates Restrictive Cardiomyopathy Due to a Human Myosin Regulatory Light Chain Mutation. Circulation. 2019. Epub 2019/07/19. 10.1161/CIRCULATIONAHA.118.036965 .
    1. Cui Y, Zheng Y, Liu X, Yan L, Fan X, Yong J, et al. Single-Cell Transcriptome Analysis Maps the Developmental Track of the Human Heart. Cell Rep. 2019;26(7):1934–50 e5. Epub 2019/02/14. 10.1016/j.celrep.2019.01.079 .
    1. Edelheit O, Hanukoglu A, Hanukoglu I. Simple and efficient site-directed mutagenesis using two single-primer reactions in parallel to generate mutants for protein structure-function studies. BMC Biotechnol. 2009;9:61 Epub 2009/07/02. 10.1186/1472-6750-9-61
    1. Fink M, Callol-Massot C, Chu A, Ruiz-Lozano P, Izpisua Belmonte JC, Giles W, et al. A new method for detection and quantification of heartbeat parameters in Drosophila, zebrafish, and embryonic mouse hearts. Biotechniques. 2009;46(2):101–13. Epub 2009/03/26. 10.2144/000113078
    1. Fischer AH, Jacobson KA, Rose J, Zeller R. Hematoxylin and Eosin Staining of Tissue and Cell Sections. Cold Spring Harbor Protocols. 2008;2008(5):pdb.prot4986. 10.1101/pdb.prot4986
    1. Schipke J, Brandenberger C, Rajces A, Manninger M, Alogna A, Post H, et al. Assessment of cardiac fibrosis: a morphometric method comparison for collagen quantification. J Appl Physiol (1985). 2017;122(4):1019–30. Epub 2017/01/28. 10.1152/japplphysiol.00987.2016 .
    1. Zikova M, Da Ponte JP, Dastugue B, Jagla K. Patterning of the cardiac outflow region in Drosophila. Proc Natl Acad Sci U S A. 2003;100(21):12189–94. Epub 2003/10/02. 10.1073/pnas.2133156100

Source: PubMed

3
订阅