Skip to main content
Top
Published in: Orphanet Journal of Rare Diseases 1/2020

Open Access 01-12-2020 | Intellectual Disability | Research

Five new cases of syndromic intellectual disability due to KAT6A mutations: widening the molecular and clinical spectrum

Authors: Roser Urreizti, Estrella Lopez-Martin, Antonio Martinez-Monseny, Montse Pujadas, Laura Castilla-Vallmanya, Luis Alberto Pérez-Jurado, Mercedes Serrano, Daniel Natera-de Benito, Beatriz Martínez-Delgado, Manuel Posada-de-la-Paz, Javier Alonso, Purificación Marin-Reina, Mar O’Callaghan, Daniel Grinberg, Eva Bermejo-Sánchez, Susanna Balcells

Published in: Orphanet Journal of Rare Diseases | Issue 1/2020

Login to get access

Abstract

Background

Pathogenic variants of the lysine acetyltransferase 6A or KAT6A gene are associated with a newly identified neurodevelopmental disorder characterized mainly by intellectual disability of variable severity and speech delay, hypotonia, and heart and eye malformations. Although loss of function (LoF) mutations were initially reported as causing this disorder, missense mutations, to date always involving serine residues, have recently been associated with a form of the disorder without cardiac involvement.

Results

In this study we present five new patients, four with truncating mutations and one with a missense change and the only one not presenting with cardiac anomalies. The missense change [p.(Gly359Ser)], also predicted to affect splicing by in silico tools, was functionally tested in the patient’s lymphocyte RNA revealing a splicing effect for this allele that would lead to a frameshift and premature truncation.

Conclusions

An extensive revision of the clinical features of these five patients revealed high concordance with the 80 cases previously reported, including developmental delay with speech delay, feeding difficulties, hypotonia, a high bulbous nose, and recurrent infections. Other features present in some of these five patients, such as cryptorchidism in males, syndactyly, and trigonocephaly, expand the clinical spectrum of this syndrome.
Appendix
Available only for authorised users
Literature
1.
go back to reference Rozman M, Camos M, Colomer D, Villamor N, Esteve J, Costa D, et al. Type I MOZ/CBP (MYST3/CREBBP) is the most common chimeric transcript in acute myeloid leukemia with t(8;16)(p11;p13) translocation. Genes Chromosomes Cancer. 2004;40(2):140–5.PubMedCrossRef Rozman M, Camos M, Colomer D, Villamor N, Esteve J, Costa D, et al. Type I MOZ/CBP (MYST3/CREBBP) is the most common chimeric transcript in acute myeloid leukemia with t(8;16)(p11;p13) translocation. Genes Chromosomes Cancer. 2004;40(2):140–5.PubMedCrossRef
2.
go back to reference Rokudai S, Laptenko O, Arnal SM, Taya Y, Kitabayashi I, Prives C. MOZ increases p53 acetylation and premature senescence through its complex formation with PML. PNAS. 2013;110(10):3895–900.PubMedCrossRef Rokudai S, Laptenko O, Arnal SM, Taya Y, Kitabayashi I, Prives C. MOZ increases p53 acetylation and premature senescence through its complex formation with PML. PNAS. 2013;110(10):3895–900.PubMedCrossRef
3.
go back to reference Pelletier N, Champagne N, Stifani S, Yang XJ. MOZ and MORF histone acetyltransferases interact with the runt-domain transcription factor Runx2. Oncogene. 2002;21(17):2729–40.PubMedCrossRef Pelletier N, Champagne N, Stifani S, Yang XJ. MOZ and MORF histone acetyltransferases interact with the runt-domain transcription factor Runx2. Oncogene. 2002;21(17):2729–40.PubMedCrossRef
4.
go back to reference Tham E, Lindstrand A, Santani A, Malmgren H, Nesbitt A, Dubbs HA, et al. Dominant mutations in KAT6A cause intellectual disability with recognizable syndromic features. Am J Hum Genet. 2015;96(3):507–13.PubMedPubMedCentralCrossRef Tham E, Lindstrand A, Santani A, Malmgren H, Nesbitt A, Dubbs HA, et al. Dominant mutations in KAT6A cause intellectual disability with recognizable syndromic features. Am J Hum Genet. 2015;96(3):507–13.PubMedPubMedCentralCrossRef
5.
go back to reference Arboleda VA, Lee H, Dorrani N, Zadeh N, Willis M, Macmurdo CF, et al. De novo nonsense mutations in KAT6A, a lysine acetyl-transferase gene, cause a syndrome including microcephaly and global developmental delay. Am J Hum Genet. 2015;96(3):498–506.PubMedPubMedCentralCrossRef Arboleda VA, Lee H, Dorrani N, Zadeh N, Willis M, Macmurdo CF, et al. De novo nonsense mutations in KAT6A, a lysine acetyl-transferase gene, cause a syndrome including microcephaly and global developmental delay. Am J Hum Genet. 2015;96(3):498–506.PubMedPubMedCentralCrossRef
6.
go back to reference Millan F, Cho MT, Retterer K, Monaghan KG, Bai R, Vitazka P, et al. Whole exome sequencing reveals de novo pathogenic variants in KAT6A as a cause of a neurodevelopmental disorder. Am J Med Genet A. 2016;170(7):1791–8.PubMedCrossRef Millan F, Cho MT, Retterer K, Monaghan KG, Bai R, Vitazka P, et al. Whole exome sequencing reveals de novo pathogenic variants in KAT6A as a cause of a neurodevelopmental disorder. Am J Med Genet A. 2016;170(7):1791–8.PubMedCrossRef
7.
go back to reference Murray CR, Abel SN, McClure MB, Foster J 2nd, Walke MI, Jayakar P, et al. Novel causative variants in DYRK1A, KARS, and KAT6A associated with intellectual disability and additional phenotypic features. J Pediatr Genet. 2017;6(2):77–83.PubMedPubMedCentralCrossRef Murray CR, Abel SN, McClure MB, Foster J 2nd, Walke MI, Jayakar P, et al. Novel causative variants in DYRK1A, KARS, and KAT6A associated with intellectual disability and additional phenotypic features. J Pediatr Genet. 2017;6(2):77–83.PubMedPubMedCentralCrossRef
9.
go back to reference Gauthier-Vasserot A, Thauvin-Robinet C, Bruel AL, Duffourd Y, St-Onge J, Jouan T, et al. Application of whole-exome sequencing to unravel the molecular basis of undiagnosed syndromic congenital neutropenia with intellectual disability. Am J Med Genet A. 2017;173(1):62–71.PubMedCrossRef Gauthier-Vasserot A, Thauvin-Robinet C, Bruel AL, Duffourd Y, St-Onge J, Jouan T, et al. Application of whole-exome sequencing to unravel the molecular basis of undiagnosed syndromic congenital neutropenia with intellectual disability. Am J Med Genet A. 2017;173(1):62–71.PubMedCrossRef
10.
go back to reference Satoh C, Maekawa R, Kinoshita A, Mishima H, Doi M, Miyazaki M, et al. Three brothers with a nonsense mutation in KAT6A caused by parental germline mosaicism. Hum Gen Var. 2017;4:17045.CrossRef Satoh C, Maekawa R, Kinoshita A, Mishima H, Doi M, Miyazaki M, et al. Three brothers with a nonsense mutation in KAT6A caused by parental germline mosaicism. Hum Gen Var. 2017;4:17045.CrossRef
11.
go back to reference Zwaveling-Soonawala N, Maas SM, Alders M, Majoie CB, Fliers E, van Trotsenburg ASP, et al. Variants in KAT6A and pituitary anomalies. Am J Med Genet A. 2017;173(9):2562–5.PubMedCrossRef Zwaveling-Soonawala N, Maas SM, Alders M, Majoie CB, Fliers E, van Trotsenburg ASP, et al. Variants in KAT6A and pituitary anomalies. Am J Med Genet A. 2017;173(9):2562–5.PubMedCrossRef
12.
go back to reference Trinh J, Huning I, Yuksel Z, Baalmann N, Imhoff S, Klein C, et al. A KAT6A variant in a family with autosomal dominantly inherited microcephaly and developmental delay. J Hum Genet. 2018;63(9):997–1001.PubMedCrossRef Trinh J, Huning I, Yuksel Z, Baalmann N, Imhoff S, Klein C, et al. A KAT6A variant in a family with autosomal dominantly inherited microcephaly and developmental delay. J Hum Genet. 2018;63(9):997–1001.PubMedCrossRef
13.
go back to reference Kennedy J, Goudie D, Blair E, Chandler K, Joss S, McKay V, et al. KAT6A syndrome: genotype-phenotype correlation in 76 patients with pathogenic KAT6A variants. Genet Med. 2019;21(4):850–60.PubMedCrossRef Kennedy J, Goudie D, Blair E, Chandler K, Joss S, McKay V, et al. KAT6A syndrome: genotype-phenotype correlation in 76 patients with pathogenic KAT6A variants. Genet Med. 2019;21(4):850–60.PubMedCrossRef
14.
go back to reference Alkhateeb A, Alazaizeh W. A novel de novo frameshift mutation in KAT6A identified by whole exome sequencing. J Pediatr Genet. 2019;8(1):10–4.PubMedCrossRef Alkhateeb A, Alazaizeh W. A novel de novo frameshift mutation in KAT6A identified by whole exome sequencing. J Pediatr Genet. 2019;8(1):10–4.PubMedCrossRef
15.
go back to reference Efthymiou, S, Salpietro V, Bettencourt B and Houlden H. Paroxysmal movement disorder and epilepsy caused by a de novo truncating mutation in KAT6A. J Pediatr Genet. 2018;7(3):114–116.CrossRef Efthymiou, S, Salpietro V, Bettencourt B and Houlden H. Paroxysmal movement disorder and epilepsy caused by a de novo truncating mutation in KAT6A. J Pediatr Genet. 2018;7(3):114–116.CrossRef
16.
go back to reference Good-Jacobson KL, Chen Y, Voss AK, Smyth GK, Thomas T, Tarlinton D. Regulation of germinal center responses and B-cell memory by the chromatin modifier MOZ. PNAS. 2014;111(26):9585–90.PubMedCrossRef Good-Jacobson KL, Chen Y, Voss AK, Smyth GK, Thomas T, Tarlinton D. Regulation of germinal center responses and B-cell memory by the chromatin modifier MOZ. PNAS. 2014;111(26):9585–90.PubMedCrossRef
17.
go back to reference Katsumoto T, Aikawa Y, Iwama A, Ueda S, Ichikawa H, Ochiya T, et al. MOZ is essential for maintenance of hematopoietic stem cells. Genes Dev. 2006;20(10):1321–30.PubMedPubMedCentralCrossRef Katsumoto T, Aikawa Y, Iwama A, Ueda S, Ichikawa H, Ochiya T, et al. MOZ is essential for maintenance of hematopoietic stem cells. Genes Dev. 2006;20(10):1321–30.PubMedPubMedCentralCrossRef
18.
go back to reference Sanz-Pamplona R, Lopez-Doriga A, Pare-Brunet L, Lazaro K, Bellido F, Alonso MH, et al. Exome sequencing reveals AMER1 as a frequently mutated gene in colorectal cancer. Clin Cancer Res. 2015;21(20):4709–18.PubMedPubMedCentralCrossRef Sanz-Pamplona R, Lopez-Doriga A, Pare-Brunet L, Lazaro K, Bellido F, Alonso MH, et al. Exome sequencing reveals AMER1 as a frequently mutated gene in colorectal cancer. Clin Cancer Res. 2015;21(20):4709–18.PubMedPubMedCentralCrossRef
19.
go back to reference Desvignes JP, Bartoli M, Delague V, Krahn M, Miltgen M, Beroud C, et al. VarAFT: a variant annotation and filtration system for human next generation sequencing data. Nucleic Acids Res. 2018;46(W1):W545–53.PubMedPubMedCentralCrossRef Desvignes JP, Bartoli M, Delague V, Krahn M, Miltgen M, Beroud C, et al. VarAFT: a variant annotation and filtration system for human next generation sequencing data. Nucleic Acids Res. 2018;46(W1):W545–53.PubMedPubMedCentralCrossRef
20.
go back to reference Urreizti R, Cueto-Gonzalez AM, Franco-Valls H, Mort-Farre S, Roca-Ayats N, Ponomarenko J, et al. A de novo nonsense mutation in MAGEL2 in a patient initially diagnosed as Opitz-C: similarities between Schaaf-Yang and Opitz-C syndromes. Sci Rep. 2017;7:44138.PubMedPubMedCentralCrossRef Urreizti R, Cueto-Gonzalez AM, Franco-Valls H, Mort-Farre S, Roca-Ayats N, Ponomarenko J, et al. A de novo nonsense mutation in MAGEL2 in a patient initially diagnosed as Opitz-C: similarities between Schaaf-Yang and Opitz-C syndromes. Sci Rep. 2017;7:44138.PubMedPubMedCentralCrossRef
21.
go back to reference Firth HV, Richards SM, Bevan AP, Clayton S, Corpas M, Rajan D, et al. DECIPHER: database of chromosomal imbalance and phenotype in humans using ensembl resources. Am J Hum Genet. 2009;84(4):524–33.PubMedPubMedCentralCrossRef Firth HV, Richards SM, Bevan AP, Clayton S, Corpas M, Rajan D, et al. DECIPHER: database of chromosomal imbalance and phenotype in humans using ensembl resources. Am J Hum Genet. 2009;84(4):524–33.PubMedPubMedCentralCrossRef
22.
go back to reference Samocha KE, Robinson EB, Sanders SJ, Stevens C, Sabo A, McGrath LM, et al. A framework for the interpretation of de novo mutation in human disease. Nat Genet. 2014;46(9):944–50.PubMedPubMedCentralCrossRef Samocha KE, Robinson EB, Sanders SJ, Stevens C, Sabo A, McGrath LM, et al. A framework for the interpretation of de novo mutation in human disease. Nat Genet. 2014;46(9):944–50.PubMedPubMedCentralCrossRef
23.
go back to reference Lopez-Martin E, Martinez-Delgado B, Bermejo-Sanchez E, Alonso J, SpainUDP network and Posada M. SpainUDP: The Spanish Undiagnosed Rare Diseases Program. Int J Environ Res Public Health. 2018;15(8).PubMedCentralCrossRef Lopez-Martin E, Martinez-Delgado B, Bermejo-Sanchez E, Alonso J, SpainUDP network and Posada M. SpainUDP: The Spanish Undiagnosed Rare Diseases Program. Int J Environ Res Public Health. 2018;15(8).PubMedCentralCrossRef
24.
go back to reference DePristo MA, Banks E, Poplin R, Garimella KV, Maguire JR, Hartl C, et al. A framework for variation discovery and genotyping using next-generation DNA sequencing data. Nat Genet. 2011;43(5):491–8.PubMedPubMedCentralCrossRef DePristo MA, Banks E, Poplin R, Garimella KV, Maguire JR, Hartl C, et al. A framework for variation discovery and genotyping using next-generation DNA sequencing data. Nat Genet. 2011;43(5):491–8.PubMedPubMedCentralCrossRef
25.
go back to reference Lochmuller H, Badowska DM, Thompson R, Knoers NV, Aartsma-Rus A, Gut I, et al. RD-connect, NeurOmics and EURenOmics: collaborative European initiative for rare diseases. Eur J Hum Genet. 2018;26(6):778–85.PubMedPubMedCentralCrossRef Lochmuller H, Badowska DM, Thompson R, Knoers NV, Aartsma-Rus A, Gut I, et al. RD-connect, NeurOmics and EURenOmics: collaborative European initiative for rare diseases. Eur J Hum Genet. 2018;26(6):778–85.PubMedPubMedCentralCrossRef
26.
go back to reference Thompson R, Johnston L, Taruscio D, Monaco L, Beroud C, Gut IG, et al. RD-connect: an integrated platform connecting databases, registries, biobanks and clinical bioinformatics for rare disease research. J Gen Intern Med. 2014;29(Suppl 3):S780–7.PubMedCrossRef Thompson R, Johnston L, Taruscio D, Monaco L, Beroud C, Gut IG, et al. RD-connect: an integrated platform connecting databases, registries, biobanks and clinical bioinformatics for rare disease research. J Gen Intern Med. 2014;29(Suppl 3):S780–7.PubMedCrossRef
27.
go back to reference Kohler S, Vasilevsky NA, Engelstad M, Foster E, McMurry J, Ayme S, et al. The human phenotype ontology in 2017. Nucleic Acids Res. 2017;45(D1):D865–76.PubMedCrossRef Kohler S, Vasilevsky NA, Engelstad M, Foster E, McMurry J, Ayme S, et al. The human phenotype ontology in 2017. Nucleic Acids Res. 2017;45(D1):D865–76.PubMedCrossRef
28.
go back to reference Girdea M, Dumitriu S, Fiume M, Bowdin S, Boycott KM, Chenier S, et al. PhenoTips: patient phenotyping software for clinical and research use. Hum Mutat. 2013;34(8):1057–65.PubMedCrossRef Girdea M, Dumitriu S, Fiume M, Bowdin S, Boycott KM, Chenier S, et al. PhenoTips: patient phenotyping software for clinical and research use. Hum Mutat. 2013;34(8):1057–65.PubMedCrossRef
Metadata
Title
Five new cases of syndromic intellectual disability due to KAT6A mutations: widening the molecular and clinical spectrum
Authors
Roser Urreizti
Estrella Lopez-Martin
Antonio Martinez-Monseny
Montse Pujadas
Laura Castilla-Vallmanya
Luis Alberto Pérez-Jurado
Mercedes Serrano
Daniel Natera-de Benito
Beatriz Martínez-Delgado
Manuel Posada-de-la-Paz
Javier Alonso
Purificación Marin-Reina
Mar O’Callaghan
Daniel Grinberg
Eva Bermejo-Sánchez
Susanna Balcells
Publication date
01-12-2020
Publisher
BioMed Central
Published in
Orphanet Journal of Rare Diseases / Issue 1/2020
Electronic ISSN: 1750-1172
DOI
https://doi.org/10.1186/s13023-020-1317-9

Other articles of this Issue 1/2020

Orphanet Journal of Rare Diseases 1/2020 Go to the issue