Skip to main content
Erschienen in: BMC Pediatrics 1/2023

Open Access 01.12.2023 | Case report

White-Sutton syndrome and congenital heart disease: case report and literature review

verfasst von: Jing Duan, Yuanzhen Ye, Jianxiang Liao, Li Chen, Xia Zhao, Chao Liu, Jialun Wen

Erschienen in: BMC Pediatrics | Ausgabe 1/2023

Abstract

Background

White-Sutton syndrome is an autosomal dominant neurodevelopmental disorder caused by heterozygous mutation in POGZ (Pogo Transposable Element Derived with ZNF Domain). This syndrome is characterized by delayed psychomotor development apparent in infancy and abnormal facial features. To date, 80 cases have been reported in the literature; however, the phenotypic characterizations remain incomplete.

Case presentation

We herein describe a 2-year-old girl harboring a novel frameshift de novo POGZ variant: c.2746del (p.Thr916ProfsTer12). This patient presented with multisystem abnormalities affecting the digestive tract and neurological functioning, as well as congenital heart disease, which involved an atrial septal defect (18 × 23 × 22 mm) with pulmonary arterial hypertension (42 mmHg). The relationship between congenital heart disease and White-Sutton syndrome as described in both the GeneReview and OMIM databases (#616,364) remains unclear. A review of the current literature revealed 18 cases of White-Sutton syndrome with POGZ variants and congenital heart disease, and we summarize their clinical features in this study.

Conclusions

Our findings based on the present case and those in the literature indicate a relationship between POGZ mutation and congenital heart disease.
Begleitmaterial
Hinweise

Supplementary Information

The online version contains supplementary material available at https://​doi.​org/​10.​1186/​s12887-023-03972-9.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
WES
Whole-exome sequencing

Background

POGZ encodes a zinc finger protein that is mainly found in the nucleus [1] and known to be involved in neuronal proliferation, chromatin remodeling, cell cycle progression and gene transcription regulation [2, 3]. Previous research has shown that POGZ is enriched in cerebrocortical and hippocampal neurons of early mouse embryos and regulates cortical neuronal development by promoting neuronal differentiation [4]. De novo disruptive mutations of POGZ are associated with White-Sutton syndrome, a syndromic neurodevelopmental disorder characterized by developmental delay, cerebral malformation, hearing loss, facial dimorphisms, and seizures [5, 6]. To date, 80 cases of White-Sutton syndrome have been reported [519]. However, the phenotypic characterizations of this syndrome remain incomplete.
Herein, we present a case of White-Sutton syndrome with a novel POGZ frameshift mutation. The patient presented multisystem manifestations, including developmental delay, hypokalemia, congenital heart disease, incomplete intestinal obstruction, and dystonia. A review of the existing literature returned 18 additional cases of White-Sutton syndrome with de novo POGZ variants that presented with congenital heart disease. The findings of the present case and literature analysis provide insight for further establishing the phenotypic spectrum of White-Sutton syndrome.

Case presentation

We report a case of White-Sutton syndrome in a 2-year-old girl. She was the second child of healthy and unrelated Chinese parents. She was born at 39 weeks of gestation by cesarean section, with a birth weight of 2840 g. The mother had gestational diabetes mellitus. The patient had a 20-year-old brother who was healthy, and her family history was negative for heart disease, epilepsy, and other neurological disorders. Nineteen hours after delivery, the girl was admitted to the neonatal unit due to repeated vomiting and diagnosed with digestive tract bleeding, which was managed with fasting and thrombin. The passage of meconium was not delayed, but abdominal distension was observed from 4 days after birth and persisted. Abdominal ultrasound showed a dilated bowel and bowel gas. Abdominal distension recurred several times over the next 2 years, culminating in mechanical ileus (Fig. 1) at the age of 1 year. Mechanical ileus was improved by fasting, gastrointestinal decompression, and glycerin enema.
At the age of 5 months, the patient was diagnosed with an atrial septal defect (18 × 23 × 22 mm), and pulmonary arterial hypertension (42 mmHg) was recorded for the first time. Atrial septal defect closure surgery was performed when the patient was 5 months of age, and the patient was treated with digoxin (0.1 mg/kg.d), spironolactone (2.4 mg/kg.d), and hydrochlorothiazide (0.8 mg/kg.d) after surgery. The patient had recurrent pneumonia after surgery and was admitted to the ICU 6 months after surgery for heart failure. Her left ventricular ejection fraction dropped to 22% at the lowest recording. At the 1-year follow-up after ICU discharge, the patient’s left ventricular ejection fraction ranged from 47 to 55%.
At the age of 9 months, she presented with epileptic spasm with hypsarrhythmia several times a day. She was successively treated with courses of topiramate (TPM; maximum dosage of 5 mg/kg.d), valproate (VPA; maximum dosage of 24 mg/kg.d), and cocktail therapy. No obvious seizure attack was observed between the ages of 13 and 19 months after combined treatment with TPM, VPA and cocktail therapy, which was a combination nutraceutical therapy consisting of vitamin B1 50 mg/d, vitamin B2 100 mg/d, vitamin C 200 mg/d, vitamin E 100 mg/d, L-carnitine 1000 mg/d, and coenzyme Q10 100 mg/d. At 19 months old, seizure returned, occurring several times a day, and did not improve with successively administered courses of levetiracetam (LEV; maximum dosage of 20 mg/kg.d), vigabatrin (VGB: the maximum dosage of 160 mg/kg.d), nitrazepam (NZP: the maximum dosage of 0.07 mg/kg.d) and clobazam (CLB; maximum dosage of 0.27 mg/kg.d). Adrenocorticotropic hormone (ATCH, 1.5 IU/kg) was added to the combined levetiracetam, vigabatrin and cocktail therapy for 2 weeks when the patient was 1 year and 10 months old. Thereafter, prednisone was continued before being gradually reduced and withdrawn over 1 month. The frequency of seizures decreased to two times per week.
From 1 year of age, the patient suffered from sleep disturbance, which mainly manifested as light sleep, often crying in sleep, and being difficult to soothe. The patient had several dysmorphic features, including a high-arched palate, frontal bossing, a congenital preauricular fistula, a tented mouth, a broad nasal root, a flat nasal bridge, and tongue protrusion. Brain magnetic resonance imaging performed at 1 year showed cerebral atrophy associated with enlargement of the supratentorial ventricles, thinned corpus callosum, and delayed myelination. She did not pass the newborn hearing screening conducted with otoacoustic emissions testing, and hearing loss was confirmed by otoacoustic emissions testing at the age of 1 year.
Peripheral venous blood samples were collected from the proband and her parents with their informed consent. Chromosomal microarray analysis for the proband was performed using Affymetrix Cytoscan 750 K. The results of the chromosomal microarray analysis and mitochondrial genetic testing for the proband were normal. The results of prenatal karyotype analysis on a cord blood sample also were normal. Trio-based WES revealed that the POGZ gene had a de novo heterozygous frameshift mutation [NM_015100.4:c.2746delA (p.Thr916ProfsTer12)], which was not found in current population databases (dbSNP, GnomAD, and ExAC). Most previously reported mutations in the POGZ gene are null variant [5, 7, 20]. According to the guidelines of the American College of Medical Genetics and Genomics (ACMG) and the Association of Molecular Pathology (AMP), the variant identified in the present case is considered pathogenic.
At the last follow-up at 2 years of age, the patient was experiencing a seizure every 3–5 days. Her parents had stopped all anti-seizure medications against medical advice, and she was receiving traditional Chinese massage. Developmentally, she could turn over, sit without support, make eye contact, and laugh, but could not stand or speak.

Discussion and conclusions

The clinical spectrum of White-Sutton Syndrome is relatively wide, with known manifestations including autism spectrum disorder, developmental delays, and intellectual disability [5, 7, 17, 20, 21]. Additional commonly reported features include feeding and gastrointestinal problems, seizures, sleep problems, hearing loss, vision problems and genitourinary abnormalities. However, the association of congenital heart disease with POGZ haploinsufficiency has not been well characterized in the previous literature. As a result, the relationship between heart disease and White-Sutton syndrome as described in both the GeneReview [21] and OMIM databases (#616,364) remains unclear. The present case report describes a new patient with a pathogenic variant of the POGZ gene who presented with congenital heart disease. This case was then compared to all cases of patients with POGZ mutations and heart disease that were found in the literature.
Peer-reviewed articles were identified by searching PubMed with the search terms: “POGZ” and “White-Sutton syndrome.” A total of 141 cases of White-Sutton syndrome caused by mutation of POGZ were identified [519, 2236] (Supplementary Table 1, Fig. 2). The types of mutations in these cases included frameshift mutation (61/141, 43.3%), nonsense mutation (49/141, 34.8%), splicing mutation (9/141, 5.6%), large deletion (3/141, 2.5%), missense mutation (17/141, 12.1%), intronic mutation (1/141, 0.7%) and in-frame deletion (1/141, 0.7%). Overall, 80.1% of the reported mutations were null variants, which suggests that loss of function is the main mechanism of pathogenicity. A previous function study revealed that de novo mutations Q1042R and R1008X in POGZ disrupt its DNA-binding activity, and a de novo missense mutation (Q1042R) is associated with an approximately 60% reduction in the DNA-binding activity of POGZ [37], which further proves that loss of function is the pathogenic mechanism. The mutation identified in the present case is a novel frameshift mutation, which is a common type of loss-of-function mutation.
The clinical descriptions of the 141 cases included varying phenotypic details, and a relatively detailed phenotype information was provided for 125 cases. Among those 125 cases, 16 cases (16/125, 12.5%) had previously received a diagnosis of congenital heart disease [5, 11, 14, 17, 19, 22, 24, 25, 27, 29, 31, 36]. In addition, we found in the Decipher database (https://​decipher.​sanger.​ac.​uk/​) two cases (Patients: 333,151 and 284,226) with heart disease and a pathogenic mutation in the POGZ gene to which the patients’ whole phenotype was attributed. Therefore, we found a total of 19 cases (including the present case) with congenital heart disease (Tables 1 and 2). Among these 19 cases with a cardiovascular defect, clinical exome sequencing was performed for 6 cases, Trio-WES for 4 cases, both Trio-WGS and microarray analysis for 1 case, and both Trio-WES and microarray analysis for 4 cases. As such, the patients’ genetic test results were relatively comprehensive. However, no other suspected pathogenic mutations were reported in these cases. In particular, four of the cases were reported in studies on congenital heart disease [11, 19, 22], and no other disease-causing mutations were found in genes associated with congenital heart disease. In conclusion, we believe the likelihood of other another underlying genetic etiology causing congenital heart disease in these patients with White-Sutton syndrome is low. In addition, according to the cases we reviewed, the incidence of congenital heart disease in patients with POGZ mutation was 12.5%, compared with only 0.8 ~ 1% in all newborns [38, 39]. This finding suggests that the incidence of congenital heart disease is significantly higher in patients with POGZ mutation than in the general population and supports the hypothesis that congenital heart disease is a relatively uncommon feature in White-Sutton syndrome.
Table 1
Characteristics of White-Sutton cases with congenital heart disease
Individual
Our case
157
Reuter et al. (2020) [11]
1-00961
Homsy et al. (2015) [22]
1-02312a
Homsy et al. (2015) [22]
1-07689
Jin et al. (2017) [19]
PT23
Assia Batzir et al. (2020)[5]
3 cases
Assia Batzir et al. (2020)
Patient:333,151 Decipher database
Patient:284,226
Decipher database
Gender
F
NA
NA
NA
NA
M
NA
NA
NA
Age at onset
Birth
NA
NA
NA
NA
2 years
NA
NA
NA
Mutation(s)
c.2746del
p.Thr916Profs*12
c.3403del
p.Glu1135Argfs*3
c.284-1G > T
c.1838 A > G
p.His613Arg
c.1558_1559delinsT
p.Asp520Phefs*7
c.1669G > T
p.Glu557*
NA
c.1837​del
p.His613Metfs*13
c.2935 C​>T
p.(Arg979*)
Genetic testing
Trio-WES, prenatal karyotype, microarry
Trio-WGS, prenatal microarray, FISH 22q
Trio-WES
Trio-WES
Trio-WES
Clinical exome sequencing
Clinical exome sequencing
  
Inheritance
De novo
De novo
De novo
De novo
De novo
De novo
NA
De novo
De novo
Cardiovascular defect
Atrial septal defect (18 × 23 × 22 mm)
Mitral atresia, aortic atresia
Hypoplastic right ventricle; pulmonary atresia congenital; pulmonary atresia, intact ventricular septum
Aortic arch hypoplasia; atrial septal defect, secundum; hypoplastic aortic annulus; hypoplastic left ventricle; mitral stenosis, valvar; ventricular septal defect, single
Congenital coronary anomaly; DORV, ventricular defect committed to aorta; left aortic arch with normal branching pattern; SDD; subaortic conus; ventricular septal defect, malalignment
Mitral valve prolapse
Atrial septal defect/ patent foramen ovale (2 patients), and aortic root dilatation (1 patient)
Abnormality of the cardiovascular system
Dextrocardia
Developmental delay
YES
YES
YES
NO
NA
YES
NA
NA
NA
Seizure
YES
NA
NA
NO
NA
NO
NA
NA
NA
ASD
NA
NA
NO
NO
NA
NO
NA
NA
NA
Microcephaly
YES
NA
NA
NO
NA
NO
NA
NA
NA
Gastrointestinal issues
YES
YES
NA
NO
NA
NA
NA
NA
NA
Hearing impairment
YES
NA
NA
NO
NA
NA
NA
NA
NA
Abnormal brain imaging
YES
NA
NA
NO
NA
NO
NA
NA
NA
Other
Recurrent respiratory infection
Borderline short stature
Laryngo bronchio tracheomalacia, subglottic cyst and learning disability
NA
NA
Myopia, astigmatism, pectus excavatum arachnodactyly
NA
Nervous system abnormality
Aganglionic megacolon
aPathogenicity of the variant is dubious
Table 2
Characteristics of White-Sutton cases with congenital heart disease
Individual
Individual 10
Murch et al. (2022) [25]
L01
Nagy,Dóra et al. (2022) [24]
Patient 2
White et al. (2016) [17]
Patient 1
Dentici et al. (2017) [14]
Patient
Pascolini et al. (2020) [36]
Patient
Trimarchi et al. (2021) [31]
Patient 8
Garde et al. (2021) [27]
Patient
Dal et al. (2021) [29]
Gender
male
male
Female
Female
Male
Female
Male
Male
Age at onset
Birth
NA
NA
Birth
NA
Birth
NA
NA
Mutation
c2933_2934dupTT
p.Arg979Phefs*3
c.2873_2874delCA; p.Ala958Valfs*6
c.2763dupC p.Thr922Hisfs*22
c.2820dupG
p.Asn941Glufs*3
c.3631 C > T
p.Arg1211*
c,2546-1G > A
c.2545 + 1delG
c.3631 C > T
p.Arg1211*
Genetic testing
Trio-WES, karyotype, array
NA
diagnostic WES
WES, chromosomal microarray
Family-based WES
Trio-WES, array-CGH
NA
Exome sequencing
Inheritance
De novo
De novo
De novo
De novo
De novo
De novo
De novo
De novo
Cardiovascular defect
Atrial septal defect
Atrial septal defect
Atrial septal defect, Patent ductus arteriosus, patent foramen ovale
Atrial septal defect
Aortic bicuspid valve with mild ascending aorta dilatation
Congenital heart disease
Atrial septal defect
Dextrocardia
Developmental Delay
YES
YES
YES
YES
YES
YES
YES
YES
Seizure
NO
YES
NO
YES
NO
NO
NO
NA
ASD
NA
NA
NO
YES
YES
NA
NA
NA
Microcephaly
YES
YES
YES
YES
NO
YES
NA
NA
Gastrointestinal issues
YES
YES
YES
YES
Yes
YES
NA
NA
Hearing impairment
Yes
NO
Yes
Yes
Yes
YES
Yes
NA
Abnormal brain imaging
NA
NA
YES
YES
YES
YES
YES
NA
Other
Prominent right eye with strabismus, cystic hygroma, pyloric stenosis, bilateral cryptorchidism
Pendle nystagmus and choroidal atrophy
Congenital diaphragmatic hernia; duplicated renal collecting system, cortical blindness
Vitiligo
Overweight
Dystonia
Common mesentary, micropenis, cryptorchidism, vision impairment
Overweight
All of the variants in cases with congenital heart disease were truncation variants (i.e., frameshift, nonsense, splicing and large deletion mutation) except for c.1838 A > G (p.His613Arg). The only missense mutation, c.1838 A > G reported by Homsy et al. [19], was identified de novo in a case without neurodevelopmental disabilities. According to the Sequence Variant Interpretation Working Group (SVI WG) general recommendations for using ACMG/AMP criteria (https://​clinicalgenome.​org/​working-groups/​sequence-variant-interpretation/​), c.1838 A > G was reclassified as a variant of uncertain significance, and this patient lacked other pathogenomic features (specifically neurodevelopmental disabilities) of White-Sutton syndrome. Thus, we believe that the pathogenicity of c.1838 A > G is dubious, and more evidence is needed to support it. Therefore, we only discuss the remaining 18 cases when considering the relationship between congenital heart disease and White-Sutton syndrome. The variants in these cases were scattered across genes and not concentrated in specific domains (Fig. 2). Moreover, two mutations, c.2545 + 1del and c.1180_1181del, have been reported in patients with and without congenital heart disease. Therefore, no significant difference was found in the type or distribution of variants between patients with and without congenital heart disease. In terms of the type of cardiac abnormalities, two of these 18 cases had no detailed phenotype of congenital heart disease. The cardiac abnormalities in the remaining 16 cases varied widely and included many types of congenital heart disease (Table 1). It is worth noting that atrial septal defects were presented in 8 cases (8/16, 50%) (including the present case), making this the most common defect type.
Animal models are an important tool for understanding the relationship between genes and disease. A mouse model with a heterozygous or homozygous nervous system-specific deletion of the Pogz gene mimicked several of the human symptoms, showing microcephaly, growth impairment, increased sociability, and learning and motor deficits [40]. Mice heterozygous for the Q1038R mutation exhibited decreased brain size, decreased cortical thickness, and ASD-related behavioral abnormalities [4]. Significantly, Complete knockout of Pogz [41] or homozygosity for the Q1038R mutation in mice [4] both cause early embryonic lethality. Micro computed tomography (CT) scanning of Q1038R homozygous mouse embryos (E15.5) showed a ventricular septal defect, which was suspected to result in embryonic lethality. This finding in a mouse model further supports the relationship between congenital heart disease and POGZ mutation.
In summary, we herein described a new White-Sutton syndrome patient with a novel frameshift de novo POGZ variant, c.2746delA (p.Thr916ProfsTer12). Furthermore, we reviewed all previously reported cases of White-Sutton syndrome with POGZ mutation and focused on patients with congenital heart disease. Our findings suggest that the White-Sutton syndrome phenotype may align with congenital heart disease. More cases showing a similar presentation would support our findings. In addition, the role of POGZ in cardiac development has not been functionally verified, and such analysis may be needed in the future.

Acknowledgements

We thank the patient and her parents for their participation and cooperation.

Declarations

The studies involving human participants were reviewed and approved by ethics committee of Shenzhen Children’s Hospital. Written informed consent was obtained from the parents of the patient.
Written informed consent was obtained from the parents of the patient for publication of this case report.

Competing interests

CL was employed by the Berry Genomics Co. Ltd. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.
Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Anhänge

Supplementary Information

Literatur
1.
Zurück zum Zitat Ibaraki K, Hamada N, Iwamoto I, Ito H, Kawamura N, Morishita R, et al. Expression analyses of POGZ, a responsible gene for Neurodevelopmental Disorders, during mouse Brain Development. Dev Neurosci. 2019;41(1–2):139–48.CrossRefPubMed Ibaraki K, Hamada N, Iwamoto I, Ito H, Kawamura N, Morishita R, et al. Expression analyses of POGZ, a responsible gene for Neurodevelopmental Disorders, during mouse Brain Development. Dev Neurosci. 2019;41(1–2):139–48.CrossRefPubMed
2.
Zurück zum Zitat Zhao W, Quan Y, Wu H, Han L, Bai T, Ma L, et al. POGZ de novo missense variants in neuropsychiatric disorders. Mol Genet Genomic Med. 2019;7(9):e900.CrossRefPubMedPubMedCentral Zhao W, Quan Y, Wu H, Han L, Bai T, Ma L, et al. POGZ de novo missense variants in neuropsychiatric disorders. Mol Genet Genomic Med. 2019;7(9):e900.CrossRefPubMedPubMedCentral
3.
Zurück zum Zitat Zhao W, Tan J, Zhu T, Ou J, Li Y, Shen L, et al. Rare inherited missense variants of POGZ associate with autism risk and disrupt neuronal development. J Genet Genomics. 2019;46(5):247–57.CrossRefPubMed Zhao W, Tan J, Zhu T, Ou J, Li Y, Shen L, et al. Rare inherited missense variants of POGZ associate with autism risk and disrupt neuronal development. J Genet Genomics. 2019;46(5):247–57.CrossRefPubMed
4.
Zurück zum Zitat Matsumura K, Seiriki K, Okada S, Nagase M, Ayabe S, Yamada I, et al. Pathogenic POGZ mutation causes impaired cortical development and reversible autism-like phenotypes. Nat Commun. 2020;11(1):859.CrossRefPubMedPubMedCentral Matsumura K, Seiriki K, Okada S, Nagase M, Ayabe S, Yamada I, et al. Pathogenic POGZ mutation causes impaired cortical development and reversible autism-like phenotypes. Nat Commun. 2020;11(1):859.CrossRefPubMedPubMedCentral
5.
Zurück zum Zitat Assia Batzir N, Posey JE, Song X, Akdemir ZC, Rosenfeld JA, Brown CW, et al. Phenotypic expansion of POGZ-related intellectual disability syndrome (White-Sutton syndrome). Am J Med Genet A. 2020;182(1):38–52.CrossRefPubMed Assia Batzir N, Posey JE, Song X, Akdemir ZC, Rosenfeld JA, Brown CW, et al. Phenotypic expansion of POGZ-related intellectual disability syndrome (White-Sutton syndrome). Am J Med Genet A. 2020;182(1):38–52.CrossRefPubMed
6.
Zurück zum Zitat Ferretti A, Barresi S, Trivisano M, Ciolfi A, Dentici ML, Radio FC, et al. POGZ-related epilepsy: case report and review of the literature. Am J Med Genet A. 2019;179(8):1631–6.PubMed Ferretti A, Barresi S, Trivisano M, Ciolfi A, Dentici ML, Radio FC, et al. POGZ-related epilepsy: case report and review of the literature. Am J Med Genet A. 2019;179(8):1631–6.PubMed
7.
Zurück zum Zitat Stessman HAF, Willemsen MH, Fenckova M, Penn O, Hoischen A, Xiong B, et al. Disruption of POGZ is Associated with Intellectual Disability and Autism Spectrum Disorders. Am J Hum Genet. 2016;98(3):541–52.CrossRefPubMedPubMedCentral Stessman HAF, Willemsen MH, Fenckova M, Penn O, Hoischen A, Xiong B, et al. Disruption of POGZ is Associated with Intellectual Disability and Autism Spectrum Disorders. Am J Hum Genet. 2016;98(3):541–52.CrossRefPubMedPubMedCentral
8.
Zurück zum Zitat Iossifov I, O’Roak BJ, Sanders SJ, Ronemus M, Krumm N, Levy D, et al. The contribution of de novo coding mutations to autism spectrum disorder. Nature. 2014;515(7526):216–21.CrossRefPubMedPubMedCentral Iossifov I, O’Roak BJ, Sanders SJ, Ronemus M, Krumm N, Levy D, et al. The contribution of de novo coding mutations to autism spectrum disorder. Nature. 2014;515(7526):216–21.CrossRefPubMedPubMedCentral
9.
Zurück zum Zitat Deciphering Developmental Disorders S. Large-scale discovery of novel genetic causes of developmental disorders. Nature. 2015;519(7542):223–8.CrossRef Deciphering Developmental Disorders S. Large-scale discovery of novel genetic causes of developmental disorders. Nature. 2015;519(7542):223–8.CrossRef
10.
Zurück zum Zitat Fukai R, Hiraki Y, Yofune H, Tsurusaki Y, Nakashima M, Saitsu H, et al. A case of autism spectrum disorder arising from a de novo missense mutation in POGZ. J Hum Genet. 2015;60(5):277–9.CrossRefPubMed Fukai R, Hiraki Y, Yofune H, Tsurusaki Y, Nakashima M, Saitsu H, et al. A case of autism spectrum disorder arising from a de novo missense mutation in POGZ. J Hum Genet. 2015;60(5):277–9.CrossRefPubMed
11.
Zurück zum Zitat Reuter MS, Chaturvedi RR, Liston E, Manshaei R, Aul RB, Bowdin S, et al. The Cardiac Genome Clinic: implementing genome sequencing in pediatric heart disease. Genet Med. 2020;22(6):1015–24.CrossRefPubMedPubMedCentral Reuter MS, Chaturvedi RR, Liston E, Manshaei R, Aul RB, Bowdin S, et al. The Cardiac Genome Clinic: implementing genome sequencing in pediatric heart disease. Genet Med. 2020;22(6):1015–24.CrossRefPubMedPubMedCentral
12.
Zurück zum Zitat Samanta D, Ramakrishnaiah R, Schaefer B. The neurological aspects related to POGZ mutation: case report and review of CNS malformations and epilepsy. Acta Neurol Belg. 2020;120(2):447–50.CrossRefPubMed Samanta D, Ramakrishnaiah R, Schaefer B. The neurological aspects related to POGZ mutation: case report and review of CNS malformations and epilepsy. Acta Neurol Belg. 2020;120(2):447–50.CrossRefPubMed
13.
Zurück zum Zitat Du X, Gao X, Liu X, Shen L, Wang K, Fan Y, et al. Genetic diagnostic evaluation of Trio-Based whole exome sequencing among children with diagnosed or suspected Autism Spectrum Disorder. Front Genet. 2018;9:594.CrossRefPubMedPubMedCentral Du X, Gao X, Liu X, Shen L, Wang K, Fan Y, et al. Genetic diagnostic evaluation of Trio-Based whole exome sequencing among children with diagnosed or suspected Autism Spectrum Disorder. Front Genet. 2018;9:594.CrossRefPubMedPubMedCentral
14.
Zurück zum Zitat Dentici ML, Niceta M, Pantaleoni F, Barresi S, Bencivenga P, Dallapiccola B, et al. Expanding the phenotypic spectrum of truncating POGZ mutations: Association with CNS malformations, skeletal abnormalities, and distinctive facial dysmorphism. Am J Med Genet A. 2017;173(7):1965–9.CrossRefPubMed Dentici ML, Niceta M, Pantaleoni F, Barresi S, Bencivenga P, Dallapiccola B, et al. Expanding the phenotypic spectrum of truncating POGZ mutations: Association with CNS malformations, skeletal abnormalities, and distinctive facial dysmorphism. Am J Med Genet A. 2017;173(7):1965–9.CrossRefPubMed
15.
Zurück zum Zitat Wang T, Guo H, Xiong B, Stessman HA, Wu H, Coe BP, et al. De novo genic mutations among a chinese autism spectrum disorder cohort. Nat Commun. 2016;7:13316.CrossRefPubMedPubMedCentral Wang T, Guo H, Xiong B, Stessman HA, Wu H, Coe BP, et al. De novo genic mutations among a chinese autism spectrum disorder cohort. Nat Commun. 2016;7:13316.CrossRefPubMedPubMedCentral
16.
Zurück zum Zitat Tan B, Zou Y, Zhang Y, Zhang R, Ou J, Shen Y, et al. A novel de novo POGZ mutation in a patient with intellectual disability. J Hum Genet. 2016;61(4):357–9.CrossRefPubMed Tan B, Zou Y, Zhang Y, Zhang R, Ou J, Shen Y, et al. A novel de novo POGZ mutation in a patient with intellectual disability. J Hum Genet. 2016;61(4):357–9.CrossRefPubMed
17.
Zurück zum Zitat White J, Beck CR, Harel T, Posey JE, Jhangiani SN, Tang S, et al. POGZ truncating alleles cause syndromic intellectual disability. Genome Med. 2016;8(1):3.CrossRefPubMedPubMedCentral White J, Beck CR, Harel T, Posey JE, Jhangiani SN, Tang S, et al. POGZ truncating alleles cause syndromic intellectual disability. Genome Med. 2016;8(1):3.CrossRefPubMedPubMedCentral
18.
Zurück zum Zitat Hildebrand MS, Jackson VE, Scerri TS, Van Reyk O, Coleman M, Braden RO, et al. Severe childhood speech disorder: gene discovery highlights transcriptional dysregulation. Neurology. 2020;94(20):e2148–e67.CrossRefPubMed Hildebrand MS, Jackson VE, Scerri TS, Van Reyk O, Coleman M, Braden RO, et al. Severe childhood speech disorder: gene discovery highlights transcriptional dysregulation. Neurology. 2020;94(20):e2148–e67.CrossRefPubMed
19.
Zurück zum Zitat Jin SC, Homsy J, Zaidi S, Lu Q, Morton S, DePalma SR, et al. Contribution of rare inherited and de novo variants in 2,871 congenital heart disease probands. Nat Genet. 2017;49(11):1593–601.CrossRefPubMedPubMedCentral Jin SC, Homsy J, Zaidi S, Lu Q, Morton S, DePalma SR, et al. Contribution of rare inherited and de novo variants in 2,871 congenital heart disease probands. Nat Genet. 2017;49(11):1593–601.CrossRefPubMedPubMedCentral
20.
Zurück zum Zitat Ye Y, Cho MT, Retterer K, Alexander N, Ben-Omran T, Al-Mureikhi M, et al. De novo POGZ mutations are associated with neurodevelopmental disorders and microcephaly. Cold Spring Harb Mol Case Stud. 2015;1(1):a000455.CrossRefPubMedPubMedCentral Ye Y, Cho MT, Retterer K, Alexander N, Ben-Omran T, Al-Mureikhi M, et al. De novo POGZ mutations are associated with neurodevelopmental disorders and microcephaly. Cold Spring Harb Mol Case Stud. 2015;1(1):a000455.CrossRefPubMedPubMedCentral
21.
Zurück zum Zitat Assia Batzir N, White J, Sutton VR. White-Sutton Syndrome. 2021 Sep 16. In: Adam MP, Mirzaa GM, Pagon RA, Wallace SE, Bean LJH, Gripp KW, Amemiya A, editors. GeneReviews® [Internet]. Seattle: University of Washington; 1993–2023. Assia Batzir N, White J, Sutton VR. White-Sutton Syndrome. 2021 Sep 16. In: Adam MP, Mirzaa GM, Pagon RA, Wallace SE, Bean LJH, Gripp KW, Amemiya A, editors. GeneReviews® [Internet]. Seattle: University of Washington; 1993–2023.
22.
Zurück zum Zitat Homsy J, Zaidi S, Shen Y, Ware JS, Samocha KE, Karczewski KJ, et al. De novo mutations in congenital heart disease with neurodevelopmental and other congenital anomalies. Science. 2015;350(6265):1262–6.CrossRefPubMedPubMedCentral Homsy J, Zaidi S, Shen Y, Ware JS, Samocha KE, Karczewski KJ, et al. De novo mutations in congenital heart disease with neurodevelopmental and other congenital anomalies. Science. 2015;350(6265):1262–6.CrossRefPubMedPubMedCentral
23.
Zurück zum Zitat Longoni M, High FA, Qi H, Joy MP, Hila R, Coletti CM, et al. Genome-wide enrichment of damaging de novo variants in patients with isolated and complex congenital diaphragmatic hernia. Hum Genet. 2017;136(6):679–91.CrossRefPubMedPubMedCentral Longoni M, High FA, Qi H, Joy MP, Hila R, Coletti CM, et al. Genome-wide enrichment of damaging de novo variants in patients with isolated and complex congenital diaphragmatic hernia. Hum Genet. 2017;136(6):679–91.CrossRefPubMedPubMedCentral
24.
Zurück zum Zitat Nagy D, Verheyen S, Wigby KM, et al. Genotype-phenotype comparison in POGZ-related neurodevelopmental disorders by using clinical scoring. Genes (Basel).2022;13(1):154. Nagy D, Verheyen S, Wigby KM, et al. Genotype-phenotype comparison in POGZ-related neurodevelopmental disorders by using clinical scoring. Genes (Basel).2022;13(1):154.
25.
Zurück zum Zitat Murch O, Jain V, Benneche A, Metcalfe K, Hobson E, Prescott K, et al. Further delineation of the clinical spectrum of White-Sutton syndrome: 12 new individuals and a review of the literature. Eur J Hum Genet. 2022;30(1):95–100.CrossRefPubMed Murch O, Jain V, Benneche A, Metcalfe K, Hobson E, Prescott K, et al. Further delineation of the clinical spectrum of White-Sutton syndrome: 12 new individuals and a review of the literature. Eur J Hum Genet. 2022;30(1):95–100.CrossRefPubMed
26.
Zurück zum Zitat Villalba MF, Chang TC. Congenital corneal opacities as a new feature in an unusual case of White-Sutton syndrome. J AAPOS. 2022;26(5):265–268. Villalba MF, Chang TC. Congenital corneal opacities as a new feature in an unusual case of White-Sutton syndrome. J AAPOS. 2022;26(5):265–268.
27.
Zurück zum Zitat Garde A, Cornaton J, Sorlin A, Moutton S, Nicolas C, Juif C, et al. Neuropsychological study in 19 french patients with White-Sutton syndrome and POGZ mutations. Clin Genet. 2021;99(3):407–17.CrossRefPubMed Garde A, Cornaton J, Sorlin A, Moutton S, Nicolas C, Juif C, et al. Neuropsychological study in 19 french patients with White-Sutton syndrome and POGZ mutations. Clin Genet. 2021;99(3):407–17.CrossRefPubMed
28.
Zurück zum Zitat Mahjani B, De Rubeis S, Gustavsson Mahjani C, Mulhern M, Xu X, Klei L, et al. Prevalence and phenotypic impact of rare potentially damaging variants in autism spectrum disorder. Mol Autism. 2021;12(1):65.CrossRefPubMedPubMedCentral Mahjani B, De Rubeis S, Gustavsson Mahjani C, Mulhern M, Xu X, Klei L, et al. Prevalence and phenotypic impact of rare potentially damaging variants in autism spectrum disorder. Mol Autism. 2021;12(1):65.CrossRefPubMedPubMedCentral
29.
Zurück zum Zitat Dal S, Hopper B, du Chattel MVR, Goel H. A case of White-Sutton syndrome with previously described loss-of-function variant in DDE domain of POGZ (p.Arg1211*) and Kartagener syndrome. Am J Med Genet A. 2021;185(3):1006–7.CrossRefPubMed Dal S, Hopper B, du Chattel MVR, Goel H. A case of White-Sutton syndrome with previously described loss-of-function variant in DDE domain of POGZ (p.Arg1211*) and Kartagener syndrome. Am J Med Genet A. 2021;185(3):1006–7.CrossRefPubMed
30.
Zurück zum Zitat Giraldo-Ocampo S, Pacheco-Orozco RA, Pachajoa H. A novel POGZ variant in a patient with intellectual disability and obesity. Appl Clin Genet. 2022;15:63–8.CrossRefPubMedPubMedCentral Giraldo-Ocampo S, Pacheco-Orozco RA, Pachajoa H. A novel POGZ variant in a patient with intellectual disability and obesity. Appl Clin Genet. 2022;15:63–8.CrossRefPubMedPubMedCentral
31.
Zurück zum Zitat Trimarchi G, Caraffi SG, Radio FC, et al. Adducted thumb and peripheral polyneuropathy: diagnostic supports in suspecting white-sutton syndrome:case report and review of the literature. Genes (Basel). 2021;12(7):950. Trimarchi G, Caraffi SG, Radio FC, et al. Adducted thumb and peripheral polyneuropathy: diagnostic supports in suspecting white-sutton syndrome:case report and review of the literature. Genes (Basel). 2021;12(7):950.
32.
Zurück zum Zitat Wright CM, Guter SJ, Cook EH. Case Report: Association of Comorbid Psychiatric Disorders and Sigmoid Prolapse with de novo POGZ mutation. J Autism Dev Disord. 2022;52(3):1408–11.CrossRefPubMed Wright CM, Guter SJ, Cook EH. Case Report: Association of Comorbid Psychiatric Disorders and Sigmoid Prolapse with de novo POGZ mutation. J Autism Dev Disord. 2022;52(3):1408–11.CrossRefPubMed
33.
Zurück zum Zitat Donnarumma B, Riccio MP, Terrone G, Palma M, Strisciuglio P, Scala I. Expanding the neurological and behavioral phenotype of White-Sutton syndrome: a case report. Ital J Pediatr. 2021;47(1):148.CrossRefPubMedPubMedCentral Donnarumma B, Riccio MP, Terrone G, Palma M, Strisciuglio P, Scala I. Expanding the neurological and behavioral phenotype of White-Sutton syndrome: a case report. Ital J Pediatr. 2021;47(1):148.CrossRefPubMedPubMedCentral
34.
Zurück zum Zitat Bruno LP, Doddato G, Valentino F, et al. New Candidates for Autism/Intellectual Disability Identified by Whole-Exome Sequencing. Int J Mol Sci. 2021;22(24):13439. Bruno LP, Doddato G, Valentino F, et al. New Candidates for Autism/Intellectual Disability Identified by Whole-Exome Sequencing. Int J Mol Sci. 2021;22(24):13439. 
35.
Zurück zum Zitat Merriweather A, Murdock DR, Rosenfeld JA, Dai H, Ketkar S, Emrick L, et al. A novel, de novo intronic variant in POGZ causes White-Sutton syndrome. Am J Med Genet A. 2022;188(7):2198–203.CrossRefPubMed Merriweather A, Murdock DR, Rosenfeld JA, Dai H, Ketkar S, Emrick L, et al. A novel, de novo intronic variant in POGZ causes White-Sutton syndrome. Am J Med Genet A. 2022;188(7):2198–203.CrossRefPubMed
36.
Zurück zum Zitat Pascolini G, Agolini E, Fleischer N, Gulotta E, Cesario C, D’Elia G, et al. A novel patient with White-Sutton syndrome refines the mutational and clinical repertoire of the POGZ-related phenotype and suggests further observations. Am J Med Genet A. 2020;182(7):1791–5.CrossRefPubMed Pascolini G, Agolini E, Fleischer N, Gulotta E, Cesario C, D’Elia G, et al. A novel patient with White-Sutton syndrome refines the mutational and clinical repertoire of the POGZ-related phenotype and suggests further observations. Am J Med Genet A. 2020;182(7):1791–5.CrossRefPubMed
37.
Zurück zum Zitat Matsumura K, Nakazawa T, Nagayasu K, Gotoda-Nishimura N, Kasai A, Hayata-Takano A, et al. De novo POGZ mutations in sporadic autism disrupt the DNA-binding activity of POGZ. J Mol Psychiatry. 2016;4:1.CrossRefPubMedPubMedCentral Matsumura K, Nakazawa T, Nagayasu K, Gotoda-Nishimura N, Kasai A, Hayata-Takano A, et al. De novo POGZ mutations in sporadic autism disrupt the DNA-binding activity of POGZ. J Mol Psychiatry. 2016;4:1.CrossRefPubMedPubMedCentral
38.
Zurück zum Zitat Williams K, Carson J, Lo C. Genetics of Congenital Heart Disease. Biomolecules. 2019;9(12):879. Williams K, Carson J, Lo C. Genetics of Congenital Heart Disease. Biomolecules. 2019;9(12):879.
39.
Zurück zum Zitat van der Bom T, Zomer AC, Zwinderman AH, Meijboom FJ, Bouma BJ, Mulder BJ. The changing epidemiology of congenital heart disease. Nat Rev Cardiol. 2011;8(1):50–60.CrossRefPubMed van der Bom T, Zomer AC, Zwinderman AH, Meijboom FJ, Bouma BJ, Mulder BJ. The changing epidemiology of congenital heart disease. Nat Rev Cardiol. 2011;8(1):50–60.CrossRefPubMed
40.
Zurück zum Zitat Suliman-Lavie R, Title B, Cohen Y, Hamada N, Tal M, Tal N, et al. Pogz deficiency leads to transcription dysregulation and impaired cerebellar activity underlying autism-like behavior in mice. Nat Commun. 2020;11(1):5836.CrossRefPubMedPubMedCentral Suliman-Lavie R, Title B, Cohen Y, Hamada N, Tal M, Tal N, et al. Pogz deficiency leads to transcription dysregulation and impaired cerebellar activity underlying autism-like behavior in mice. Nat Commun. 2020;11(1):5836.CrossRefPubMedPubMedCentral
41.
Zurück zum Zitat Gudmundsdottir B, Gudmundsson KO, Klarmann KD, Singh SK, Sun L, Singh S, et al. POGZ is required for silencing mouse embryonic beta-like hemoglobin and human fetal hemoglobin expression. Cell Rep. 2018;23(11):3236–48.CrossRefPubMedPubMedCentral Gudmundsdottir B, Gudmundsson KO, Klarmann KD, Singh SK, Sun L, Singh S, et al. POGZ is required for silencing mouse embryonic beta-like hemoglobin and human fetal hemoglobin expression. Cell Rep. 2018;23(11):3236–48.CrossRefPubMedPubMedCentral
Metadaten
Titel
White-Sutton syndrome and congenital heart disease: case report and literature review
verfasst von
Jing Duan
Yuanzhen Ye
Jianxiang Liao
Li Chen
Xia Zhao
Chao Liu
Jialun Wen
Publikationsdatum
01.12.2023
Verlag
BioMed Central
Erschienen in
BMC Pediatrics / Ausgabe 1/2023
Elektronische ISSN: 1471-2431
DOI
https://doi.org/10.1186/s12887-023-03972-9

Weitere Artikel der Ausgabe 1/2023

BMC Pediatrics 1/2023 Zur Ausgabe

Ähnliche Überlebensraten nach Reanimation während des Transports bzw. vor Ort

29.05.2024 Reanimation im Kindesalter Nachrichten

Laut einer Studie aus den USA und Kanada scheint es bei der Reanimation von Kindern außerhalb einer Klinik keinen Unterschied für das Überleben zu machen, ob die Wiederbelebungsmaßnahmen während des Transports in die Klinik stattfinden oder vor Ort ausgeführt werden. Jedoch gibt es dabei einige Einschränkungen und eine wichtige Ausnahme.

Alter der Mutter beeinflusst Risiko für kongenitale Anomalie

28.05.2024 Kinder- und Jugendgynäkologie Nachrichten

Welchen Einfluss das Alter ihrer Mutter auf das Risiko hat, dass Kinder mit nicht chromosomal bedingter Malformation zur Welt kommen, hat eine ungarische Studie untersucht. Sie zeigt: Nicht nur fortgeschrittenes Alter ist riskant.

Begünstigt Bettruhe der Mutter doch das fetale Wachstum?

Ob ungeborene Kinder, die kleiner als die meisten Gleichaltrigen sind, schneller wachsen, wenn die Mutter sich mehr ausruht, wird diskutiert. Die Ergebnisse einer US-Studie sprechen dafür.

Bei Amblyopie früher abkleben als bisher empfohlen?

22.05.2024 Fehlsichtigkeit Nachrichten

Bei Amblyopie ist das frühzeitige Abkleben des kontralateralen Auges in den meisten Fällen wohl effektiver als der Therapiestandard mit zunächst mehrmonatigem Brilletragen.

Update Pädiatrie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.