Skip to content
Licensed Unlicensed Requires Authentication Published by De Gruyter January 18, 2017

CASZ1 loss-of-function mutation contributes to familial dilated cardiomyopathy

  • Xing-Biao Qiu , Xin-Kai Qu , Ruo-Gu Li , Hua Liu , Ying-Jia Xu , Min Zhang , Hong-Yu Shi , Xu-Min Hou , Xu Liu , Fang Yuan , Yu-Min Sun , Jun Wang , Ri-Tai Huang , Song Xue and Yi-Qing Yang EMAIL logo

Abstract

Background:

The zinc finger transcription factor CASZ1 plays a key role in cardiac development and postnatal adaptation, and in mice, deletion of the CASZ1 gene leads to dilated cardiomyopathy (DCM). However, in humans whether genetically defective CASZ1 contributes to DCM remains unclear.

Methods:

The coding exons and splicing junction sites of the CASZ1 gene were sequenced in 138 unrelated patients with idiopathic DCM. The available family members of the index patient harboring an identified CASZ1 mutation and 200 unrelated, ethnically matched healthy individuals used as controls were genotyped for CASZ1. The functional characteristics of the mutant CASZ1 were analyzed in contrast to its wild-type counterpart using a luciferase reporter assay system.

Results:

A novel heterozygous CASZ1 mutation, p.K351X, was identified in an index patient with DCM. Genetic analysis of the mutation carrier’s family showed that the mutation co-segregated with DCM, which was transmitted in an autosomal dominant pattern with complete penetrance. The nonsense mutation, which was absent in 400 referential chromosomes, altered the amino acid that was highly conserved evolutionarily. Biological investigations revealed that the mutant CASZ1 had no transcriptional activity.

Conclusions:

The current study reveals CASZ1 as a new gene responsible for human DCM, which provides novel mechanistic insight and potential therapeutic target for CASZ1-associated DCM, implying potential implications in improved prophylactic and therapeutic strategies for DCM, the most common type of primary myocardial disease.


Corresponding author: Dr. Yi-Qing Yang, Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, 241 West Huaihai Road, 200030 Shanghai, P.R. China, Phone: +86-21-62821990, Fax: +86-21-62821105

Acknowledgments

We really thank the study participants for their participation in this investigation.

  1. Author contributions: All the authors have accepted responsibility for the entire content of this submitted manuscript and approved submission.

  2. Research funding: This work was financially supported by grants from the National Natural Science Fund of China (81470372 and 81270161), the Key Program for Basic Research of Shanghai, China (14JC1405500), the Natural Science Fund of Shanghai, China (14ZR1438000 and 15ZR1438100), and the Key Project of Shanghai Chest Hospital, China (2014YZDH10102).

  3. Employment or leadership: None declared.

  4. Honorarium: None declared.

  5. Competing interests: The funding organization(s) played no role in the study design; in the collection, analysis, and interpretation of data; in the writing of the report; or in the decision to submit the report for publication.

References

1. Elliott P, Andersson B, Arbustini E, Bilinska Z, Cecchi F, Charron P, et al. Classification of the cardiomyopathies: a position statement from the European Society of Cardiology Working Group on Myocardial and Pericardial Diseases. Eur Heart J 2008;29:270–6.10.1093/eurheartj/ehm342Search in Google Scholar PubMed

2. Dhandapany PS, Razzaque MA, Muthusami U, Kunnoth S, Edwards JJ, Mulero-Navarro S, et al. RAF1 mutations in childhood-onset dilated cardiomyopathy. Nat Genet 2014;46:635–9.10.1038/ng.2963Search in Google Scholar PubMed PubMed Central

3. Garcia-Pavia P, Cobo-Marcos M, Guzzo-Merello G, Gomez-Bueno M, Bornstein B, Lara-Pezzi E, et al. Genetics in dilated cardiomyopathy. Biomark Med 2013;7:517–33.10.2217/bmm.13.77Search in Google Scholar PubMed

4. Hershberger RE, Hedges DJ, Morales A. Dilated cardiomyopathy: the complexity of a diverse genetic architecture. Nat Rev Cardiol 2013;10:531–47.10.1038/nrcardio.2013.105Search in Google Scholar PubMed

5. McNally EM, Golbus JR, Puckelwartz MJ. Genetic mutations and mechanisms in dilated cardiomyopathy. J Clin Invest 2013;123:19–26.10.1172/JCI62862Search in Google Scholar PubMed PubMed Central

6. Arndt AK, Schafer S, Drenckhahn JD, Sabeh MK, Plovie ER, Caliebe A, et al. Fine mapping of the 1p36 deletion syndrome identifies mutation of PRDM16 as a cause of cardiomyopathy. Am J Hum Genet 2013;93:67–77.10.1016/j.ajhg.2013.05.015Search in Google Scholar PubMed PubMed Central

7. van Spaendonck-Zwarts KY, Posafalvi A, van den Berg MP, Hilfiker-Kleiner D, Bollen IA, Sliwa K, et al. Titin gene mutations are common in families with both peripartum cardiomyopathy anddilated cardiomyopathy. Eur Heart J 2014;35:2165–73.10.1093/eurheartj/ehu050Search in Google Scholar PubMed

8. Agrawal PB, Pierson CR, Joshi M, Liu X, Ravenscroft G, Moghadaszadeh B, et al. SPEG interacts with myotubularin, and its deficiency causes centronuclear myopathy with dilated cardiomyopathy. Am J Hum Genet 2014;95:218–26.10.1016/j.ajhg.2014.07.004Search in Google Scholar PubMed PubMed Central

9. Roh JI, Cheong C, Sung YH, Lee J, Oh J, Lee BS, et al. Perturbation of NCOA6 leads todilated cardiomyopathy. Cell Rep 2014;8:991–8.10.1016/j.celrep.2014.07.027Search in Google Scholar PubMed PubMed Central

10. Haas J, Frese KS, Peil B, Kloos W, Keller A, Nietsch R, et al. Atlas of the clinical genetics of humandilated cardiomyopathy. Eur Heart J 2015;36:1123–35a.10.1093/eurheartj/ehu301Search in Google Scholar PubMed

11. Reinstein E, Orvin K, Tayeb-Fligelman E, Stiebel-Kalish H, Tzur S, Pimienta AL, et al. Mutations in TAX1BP3 cause dilated cardiomyopathy with septo-optic dysplasia. Hum Mutat 2015;36:439–42.10.1002/humu.22759Search in Google Scholar PubMed

12. Liaquat A, Shauket U, Ahmad W, Javed Q. The tumor necrosis factor-α -238G/A and IL-6 -572G/C gene polymorphisms and the risk of idiopathic dilated cardiomyopathy: a meta-analysis of 25 studies including 9493 cases and 13,971 controls. Clin Chem Lab Med 2015;53:307–18.10.1515/cclm-2014-0502Search in Google Scholar PubMed

13. Qu XK, Yuan F, Li RG, Xu L, Jing WF, Liu H, et al. Prevalence and spectrum of LRRC10 mutations associated with idiopathicdilated cardiomyopathy. Mol Med Rep 2015;12:3718–24.10.3892/mmr.2015.3843Search in Google Scholar PubMed

14. Akinrinade O, Ollila L, Vattulainen S, Tallila J, Gentile M, Salmenperä P, et al. Genetics and genotype-phenotype correlations in Finnish patients withdilated cardiomyopathy. Eur Heart J 2015;36:2327–37.10.1093/eurheartj/ehv253Search in Google Scholar PubMed PubMed Central

15. Zhao Y, Feng Y, Zhang YM, Ding XX, Song YZ, Zhang AM, et al. Targeted next-generation sequencing of candidate genes reveals novel mutations in patients with dilated cardiomyopathy. Int J Mol Med 2015;36:1479–86.10.3892/ijmm.2015.2361Search in Google Scholar PubMed PubMed Central

16. Cuenca S, Ruiz-Cano MJ, Gimeno-Blanes JR, Jurado A, Salas C, Gomez-Diaz I, et al. Inherited Cardiac Diseases Program of the Spanish Cardiovascular Research Network (Red Investigación Cardiovascular). Genetic basis of familial dilated cardiomyopathy patients undergoing heart transplantation. J Heart Lung Transplant 2016;35:625–35.10.1016/j.healun.2015.12.014Search in Google Scholar PubMed

17. Al-Yacoub N, Shaheen R, Awad SM, Kunhi M, Dzimiri N, Nguyen HC, et al. FBXO32, encoding a member of the SCF complex, is mutated indilated cardiomyopathy. Genome Biol 2016;17:2.10.1186/s13059-015-0861-4Search in Google Scholar PubMed PubMed Central

18. Akazawa H, Komuro I. Cardiac transcription factor Csx/Nkx2-5: its role in cardiac development and diseases. Pharmacol Ther 2005;107:252–68.10.1016/j.pharmthera.2005.03.005Search in Google Scholar PubMed

19. Pikkarainen S, Tokola H, Kerkelä R, Ruskoaho H. GATA transcription factors in the developing and adult heart. Cardiovasc Res 2004;63:196–207.10.1016/j.cardiores.2004.03.025Search in Google Scholar PubMed

20. Greulich F, Rudat C, Kispert A. Mechanisms of T-box gene function in the developing heart. Cardiovasc Res 2011;91:212–22.10.1093/cvr/cvr112Search in Google Scholar PubMed

21. Srivastava D. HAND proteins: molecular mediators of cardiac development and congenital heart disease. Trends Cardiovasc Med 1999;9:11–8.10.1016/S1050-1738(98)00033-4Search in Google Scholar

22. Oka T, Xu J, Molkentin JD. Re-employment of developmental transcription factors in adult heart disease. Semin Cell Dev Biol 2007;18:117–31.10.1016/j.semcdb.2006.11.012Search in Google Scholar PubMed PubMed Central

23. McCulley DJ, Black BL. Transcription factor pathways and congenital heart disease. Curr Top Dev Biol 2012;100:253–77.10.1016/B978-0-12-387786-4.00008-7Search in Google Scholar PubMed PubMed Central

24. Costa MW, Guo G, Wolstein O, Vale M, Castro ML, Wang L, et al. Functional characterization of a novel mutation in NKX2-5 associated with congenital heart disease and adult-onset cardiomyopathy. Circ Cardiovasc Genet 2013;6:238–47.10.1161/CIRCGENETICS.113.000057Search in Google Scholar PubMed PubMed Central

25. Yuan F, Qiu XB, Li RG, Qu XK, Wang J, Xu YJ, et al. A novel NKX2-5 loss-of-function mutation predisposes to familial dilated cardiomyopathy and arrhythmias. Int J Mol Med 2015;35:478–86.10.3892/ijmm.2014.2029Search in Google Scholar PubMed

26. Li RG, Li L, Qiu XB, Yuan F, Xu L, Li X, et al. GATA4 loss-of-function mutation underlies familial dilated cardiomyopathy. Biochem Biophys Res Commun 2013;439:591–6.10.1016/j.bbrc.2013.09.023Search in Google Scholar PubMed

27. Li J, Liu WD, Yang ZL, Yuan F, Xu L, Li RG, et al. Prevalence and spectrum of GATA4 mutations associated with sporadic dilated cardiomyopathy. Gene 2014;548:174–81.10.1016/j.gene.2014.07.022Search in Google Scholar PubMed

28. Zhang XL, Dai N, Tang K, Chen YQ, Chen W, Wang J, et al. GATA5 loss-of-function mutation in familial dilated cardiomyopathy. Int J Mol Med 2015;35:763–70.10.3892/ijmm.2014.2050Search in Google Scholar PubMed

29. Xu L, Zhao L, Yuan F, Jiang WF, Liu H, Li RG, et al. GATA6 loss-of-function mutations contribute to familial dilated cardiomyopathy. Int J Mol Med 2014;34:1315–22.10.3892/ijmm.2014.1896Search in Google Scholar PubMed

30. Zhang XL, Qiu XB, Yuan F, Wang J, Zhao CM, Li RG, et al. TBX5 loss-of-function mutation contributes to familial dilated cardiomyopathy. Biochem Biophys Res Commun 2015;459:166–71.10.1016/j.bbrc.2015.02.094Search in Google Scholar PubMed

31. Zhou W, Zhao L, Jiang JQ, Jiang WF, Yang YQ, Qiu XB. A novel TBX5 loss-of-function mutation associated with sporadic dilated cardiomyopathy. Int J Mol Med 2015;36:282–8.10.3892/ijmm.2015.2206Search in Google Scholar PubMed

32. Kirk EP, Sunde M, Costa MW, Rankin SA, Wolstein O, Castro ML, et al. Mutations in cardiac T-box factor gene TBX20 are associated with diverse cardiac pathologies, including defects of septation and valvulogenesis and cardiomyopathy. Am J Hum Genet 2007;81:280–91.10.1086/519530Search in Google Scholar PubMed PubMed Central

33. Zhao CM, Sun B, Song HM, Wang J, Xu WJ, Jiang JF, et al. TBX20 loss-of-function mutation associated with familial dilated cardiomyopathy. Clin Chem Lab Med 2016;54:325–32.10.1515/cclm-2015-0328Search in Google Scholar PubMed

34. Zhou YM, Dai XY, Qiu XB, Yuan F, Li RG, Xu YJ, et al. HAND1 loss-of-function mutation associated with familial dilated cardiomyopathy. Clin Chem Lab Med 2016;54:1161–7.10.1515/cclm-2015-0766Search in Google Scholar PubMed

35. Liu Z, Li W, Ma X, Ding N, Spallotta F, Southon E, et al. Essential role of the zinc finger transcription factor Casz1 for mammalian cardiac morphogenesis and development. J Biol Chem 2014;289:29801–16.10.1074/jbc.M114.570416Search in Google Scholar PubMed PubMed Central

36. Dorr KM, Amin NM, Kuchenbrod LM, Labiner H, Charpentier MS, Pevny LH, et al. Casz1 is required for cardiomyocyte G1-to-S phase progression during mammalian cardiac development. Development 2015;142:2037–47.10.1242/dev.119107Search in Google Scholar PubMed PubMed Central

37. Jordan VK, Zaveri HP, Scott DA. 1p36 deletion syndrome: an update. Appl Clin Genet 2015;8:189–200.10.2147/TACG.S65698Search in Google Scholar PubMed PubMed Central

38. Huang RT, Xue S, Wang J, Gu JY, Xu JH, Li YJ, et al. CASZ1 loss-of-function mutation associated with congenital heart disease. Gene 2016;595:62–68.10.1016/j.gene.2016.09.044Search in Google Scholar PubMed

39. Elliott P, O’Mahony C, Syrris P, Evans A, Rivera Sorensen C, Sheppard MN, et al. Prevalence of desmosomal protein gene mutations in patients with dilated cardiomyopathy. Circ Cardiovasc Genet 2010;3:314–22.10.1161/CIRCGENETICS.110.937805Search in Google Scholar PubMed

40. Wei D, Bao H, Zhou N, Zheng GF, Liu XY, Yang YQ. GATA5 loss-of-function mutation responsible for the congenital ventriculoseptal defect. Pediatr Cardiol 2013;34:504–11.10.1007/s00246-012-0482-6Search in Google Scholar PubMed

41. Warner LR, Babbitt CC, Primus AE, Severson TF, Haygood R, Wray GA. Functional consequences of genetic variation in primates on tyrosine hydroxylase (TH) expression in vitro. Brain Res 2009;1288:1–8.10.1016/j.brainres.2009.06.086Search in Google Scholar PubMed

42. Liu Z, Lam N, Thiele CJ. Zinc finger transcription factor CASZ1 interacts with histones, DNA repair proteins and recruits NuRD complex to regulate gene transcription. Oncotarget 2015;6:27628–40.10.18632/oncotarget.4733Search in Google Scholar PubMed PubMed Central

43. Virden RA, Thiele CJ, Liu Z. Characterization of critical domains within the tumor suppressor CASZ1 required for transcriptional regulation and growth suppression. Mol Cell Biol 2012;32:1518–28.10.1128/MCB.06039-11Search in Google Scholar PubMed PubMed Central

44. Lykke-Andersen S, Jensen TH. Nonsense-mediated mRNA decay: an intricate machinery that shapes transcriptomes. Nat Rev Mol Cell Biol 2015;16:665–77.10.1038/nrm4063Search in Google Scholar PubMed

45. Inácio A, Silva AL, Pinto J, Ji X, Morgado A, Almeida F, et al. Nonsense mutations in close proximity to the initiation codon fail to trigger full nonsense-mediated mRNA decay. J Biol Chem 2004;279:32170–80.10.1074/jbc.M405024200Search in Google Scholar PubMed

46. Amin NM, Gibbs D, Conlon FL. Differential regulation of CASZ1 protein expression during cardiac and skeletal muscle development. Dev Dyn 2014;243:948–56.10.1002/dvdy.24126Search in Google Scholar PubMed PubMed Central

47. Christine KS, Conlon FL. Vertebrate CASTOR is required for differentiation of cardiac precursor cells at the ventral midline. Dev Cell 2008;14:616–23.10.1016/j.devcel.2008.01.009Search in Google Scholar PubMed PubMed Central

48. Liu Z, Yang X, Tan F, Cullion K, Thiele CJ. Molecular cloning and characterization of human Castor, a novel human gene upregulated during cell differentiation. Biochem Biophys Res Commun 2006;344:834–44.10.1016/j.bbrc.2006.03.207Search in Google Scholar PubMed

49. Zaidi S, Choi M, Wakimoto H, Ma L, Jiang J, Overton JD, et al. De novo mutations in histone-modifying genes in congenital heart disease. Nature 2013;498:220–3.10.1038/nature12141Search in Google Scholar PubMed PubMed Central

50. Wang C, Liu Z, Woo CW, Li Z, Wang L, Wei JS, et al. EZH2 mediates epigenetic silencing of neuroblastoma suppressor genes CASZ1, CLU, RUNX3, and NGFR. Cancer Res 2012;72:315–24.10.1158/0008-5472.CAN-11-0961Search in Google Scholar PubMed PubMed Central

Received: 2016-7-10
Accepted: 2016-12-9
Published Online: 2017-1-18
Published in Print: 2017-8-28

©2017 Walter de Gruyter GmbH, Berlin/Boston

Downloaded on 27.5.2024 from https://www.degruyter.com/document/doi/10.1515/cclm-2016-0612/html
Scroll to top button