Skip to main content
Erschienen in: BMC Medical Genetics 1/2019

Open Access 01.12.2019 | Research article

Feeding difficulty is the dominant feature in 12 Chinese newborns with CHD7 pathogenic variants

verfasst von: Xiang Chen, Kai Yan, Yanyan Gao, Huijun Wang, Guoqiang Chen, Bingbing Wu, Qian Qin, Lin Yang, Wenhao Zhou

Erschienen in: BMC Medical Genetics | Ausgabe 1/2019

Abstract

Background

CHARGE syndrome is characterized by coloboma, heart defects, choanal atresia, growth retardation, genitourinary malformation and ear abnormalities. The chromodomain helicase DNA-binding protein 7 (CHD7) gene is the major cause of CHARGE syndrome and is inherited in an autosomal dominant manner. Currently, the phenotype spectrum of CHARGE syndrome in neonatal population remain elusive. We aimed to investigate the phenotype spectrum of neonatal patients suspected to have CHARGE syndrome with pathogenic or likely pathogenic variants in the CHD7 gene.

Methods

We pooled next-generation sequencing data from the Neonatal Birth Defects Cohort (NBDC, ClinicalTrials.​gov Identifier: NCT02551081) in Children’s Hospital of Fudan University. The pathogenicity of novel variants was analyzed by bioinformatic and genetic analyses. Clinical information collection, Sanger sequencing and follow-up interviews were performed when possible. Cranial MRI of these patients was performed, the volumes of different regions of the brain were analyzed.

Results

A total of 12 unrelated patients in our cohort were found with CHD7 variants. Eight patients received a firm clinical diagnosis of CHARGE syndrome (Bergmann criteria, Blake criteria, Verloes criteria and Hale criteria). Three patients did not match any diagnostic criteria, and no patients matched the Verloes criteria. Phenotype spectrum analysis found that feeding difficulty was the dominant feature among this neonatal cohort. Six novel variants in the CHD7 gene (Glu2408*, Lys651*, c.5607 + 1G > T, Leu373Val, Lys2005Asnfs*37 and Gln1991*) were identified, expanding the variant database of the CHD7 gene. Cranial MRI analysis revealed significant volume loss in cingulate gyrus, occipital lobe, and cerebellum and volume gain in the left medial and inferior temporal gyri anterior white matter parts.

Conclusions

Based on a relatively unbiased neonatal cohort, we concluded that CHARGE syndrome and CHD7 gene variants should be suspected in newborns who have feeding difficulty, and one or more malformations.

Trial registration

Neonatal Birth Defects Cohort (NBDC, ClinicalTrials.gov identifier: NCT02551081).
Hinweise

Electronic supplementary material

The online version of this article (https://​doi.​org/​10.​1186/​s12881-019-0813-z) contains supplementary material, which is available to authorized users.
Xiang Chen and Kai Yan are Co-first author

Background

CHARGE syndrome ([MIM:214800]) is a rare disease with a prevalence between 1:15,000 and 1:17,000. This syndrome is mainly characterized by coloboma, heart defects, choanal atresia, growth retardation, genitourinary malformation and ear abnormalities [1]. Minor manifestations include a distinctive face, esophagus malformation, hypothalamo-hypophyseal dysfunction and mental retardation [2, 3]. Clinical diagnostic criteria were proposed by Blake in 1998 [2] and revised by Verloes in 2005 [3]. The CHD7 gene is the only known gene [4] responsible for 90%~ 95% of typical CHARGE syndrome [1]. CHD is an acronym for chromodomian helicase DNA-binding proteins. CHD7 belongs to the CHD protein subfamily III, along with CHD5, CHD6, CHD8, CHD9 [5, 6]. CHD7 acts as an ATP-dependent chromatin remodeler and transcriptional regulator preferring short linker DNA [7]. In mouse models, CHD7 participates in embryonic stem cell differentiation and regulates the transcription of tissue-specific genes [8]. The broad spectrum of CHARGE syndrome symptoms is related to the regulatory function of CHD7 in the multipotent migratory neural crest in the embryonic period [9]. Bergmann et al. reported that a large number of patients with a CHD7 variant do not fulfill the clinical criteria of CHARGE syndrome. Mildly affected patients may be overlooked easily. So, in 2011, they emphasized that CHD7 analysis is helpful in the CHARGE syndrome diagnosis process and proposed a guideline for CHD7 analysis [10]. In 2016, new CHARGE syndrome diagnostic criteria was proposed by Hale with four major criteria and seven minor criteria [11]. Major standards include pathogenic variants in the CHD7 gene, coloboma, choanal atresia or cleft palate and ear (external, middle or inner) abnormalities. The inclusion criteria of CHARGE syndrome are two major criteria and any number of minor criteria. So genetic testing is increasingly important in CHARGE syndrome diagnostic process. In this study, we describe the phenotype spectrum of 12 neonatal patients carrying CHD7 variants. This is the largest sample size with a focus on CHARGE syndrome in the Chinses Han population.

Methods

Inclusion criteria based on clinical features and genetic analysis

We pooled data from the Neonatal Birth Defects Cohort (NBDC, ClinicalTrials.​gov Identifier: NCT02551081) in Children’s Hospital of Fudan University during 2016–01 to 2018–11. This data included 11,572 whole exome sequencing, 13,636 clinical exome sequencing and 1284 whole genome copy number microarray analyses. In this retrospective study, enrolled newborns met one of the following four criteria: 1) a previously established heterozygous pathogenic variant in the CHD7 gene, with data obtained from both the public database (HGMD and ClinVar) and internal database; 2) the same amino acid change as a previously established pathogenic variant; however, different nucleotide changes were accepted; 3) a novel (both public database and internal database) heterozygous null variant (nonsense, frameshift, canonical +/− 1 or 2 splice sites, initiation codon, single or multiexon deletion) in CHD7 gene; and 4) a novel and de novo heterozygous variant with negative family history or inherited from the affected parents. All variants were classified according to ACMG guideline [12]. Patients were excluded if pathogenic copy number variants were identified using array-based comparative genomic hybridization (arry-CGH). Patients’ information was obtained from clinical records. A clinical diagnosis of CHARGE syndrome was made based on the Bergmann criteria [10], Blake criteria [2], Verloes criteria [3] and Hale criteria [11].

Next generation sequencing and sanger confirmation

The criteria for genetic testing were approved by ethics committees of Children’s Hospital, Fudan University (2014–107). Pretest counseling was performed by physicians. Informed consent was obtained from the patient’s parents. High-throughput sequencing was performed according to standard protocols in Clinical Laboratory Improvement Amendments (CLIA: 99D2064856) compliant sequencing laboratory in Wuxi NEXTCODE (China). Sequences were generated using the Agilent ClearSeq Inherited Disease Kit, Illumina Cluster and SBS Kit. Next generation sequencing was performed on an Illumina HiSeq 2000/2500 platform. The detected variants were confirmed using PCR and PCR-amplified DNA products were subjected to direct automated sequencing (3500XL Genetic Analyzer, Applied Biosystems) according to the manufacturer’s specifications. De novo variants were detected by parental evaluating via Sanger sequencing.

Cranial MRI analysis

Among the 12 affected neonates, 8 had T1- and T2-weighted MRI head scans from the picture archiving and communication system (PACS) in Children’s hospital of Fudan University. We matched these affected neonates with 92 control neonates of the same corrected gestational age and excluded 4 patients because of image registration failure caused by poor image quality. All digital imaging and communications in medicine (DICOMs) were concatenated into a NIfTi volume format by dcm2niix software [13]. Then, the brain images of the neonates were extracted through FSL BET based on the T2-weighted modality followed by N4 bias correction [14]. The skull-striped T2 images were normalized to the neonate-specific T2 weighted image template at 44 weeks corrected gestational age [15] using the SyN registration method in ANTS [16]. The T2 brain template included a parcellation atlas with 87 regions of interest (ROI). The volumes of each ROI in the brain were measured by the sum of the log relative Jacobian determinant from the nonlinear deformable field of the registration. The total brain volume was used as a nuisance variable for regression, and the t statistic map was constructed from the linear regression as follows: ROI_volume ~α variants+ β corrected_age + θ gender+ 1.

Results

Clinical diagnosis

A total of 12 pathogenic/likely pathogenic variants in the CHD7 gene were identified by next generation sequencing in 12 unrelated newborns. All enrolled patients were from nonconsanguineous couples from the Chinese Han population. All patients’ family histories were negative. The clinical manifestations of each patient are shown in Table 1. The Bergmann criteria focuses on patients with suspected features of CHARGE syndrome with CHD7 analysis. According to Bergmann’s criteria, Patient 2, 6, 7, 8, 9, 10, 11, and 12 matched the guidelines and needed further CHD7 analysis. Patients 7, 8 and 11 matched two cardinal and one supportive criteria and need CHD7 analysis including MLPA. Patient 2, 6, 9 and 10 matched one cardinal criteria and one or more than one supportive criteria. According to Bergmann’s criteria, the four patients needed a temporal bone CT first to detect typical semicircular canal abnormalities. The Blake criteria has four major criteria (coloboma, choanal atresia/stenosis, characteristic ear anomalies and cranial nerve dysfunction) and seven minor criteria. Definitive CHARGE syndrome should match 4 major or 3 major features and 3 minor features. Probable or possible CHARGE is defined as 1 or 2 major features and several minor features. Based on the Blake criteria, 8 patients (2, 3, 6, 7, 8, 10, 11 and 12) were diagnosed with probable or possible CHARGE. The major criteria of Hale proposed in 2016 were coloboma, choanal atresia or cleft palate, ear abnormalities and pathogenic CHD7 variants. Patients matching two major criteria and any number of minor criteria are diagnosed with CHARGE syndrome. In this study, four patients (2, 3, 7 and 11) met Hale’s criteria for CHARGE syndrome. Major Verlos criteria include coloboma, choanal atresia/stenosis and hypoplasia/aplasia of the semicircular canals. Patient matching as least two major criteria can be considered to have CHARGE syndrome. As no patient in our cohort was reported with choanal atresia/stenosis and abnormalities of the semicircular canals, no patient could be diagnosed with CHARGE syndrome based on the Verloes criteria. In this population, some typical features of CHARGE syndrome were observed, including aplasia/dysplasia of the semicircular canals, cleft lip/palate and choanal atresia.
Table 1
Phenotype spectrum of 12 neonatal patients
Item
Positive observed
Bergman 2011 [10]
1
2
3
4
5
6
7
8
9
10
11
12
Sex
75% (9/12 male)
/
M
M
M
F
M
M
M
F
M
M
F
M
GA (week)
37.54 (average)
/
41 + 2
36 + 3
NA
38 + 3
37 + 2
35 + 6
39
40
36 + 2
34 + 3
38
39 + 4
Weight (Kg)
2.77 (average)
/
3.2
3.45
NA
2.2
2.76
2.2
3.75
2.8
2.4
1.85
2.65
3.2
Age of onset
1.83 (average)
/
IAB
9d
10d
IAB
IAB
3d
IAB
IAB
IAB
IAB
IAB
IAB
Head and Neck
83% (10/12)
/
            
Abnormality of the external ear
50% (6/12)
97.0% (224/231)
N
yes
yes
N
N
yes, L
yes
N
N
N
yes, R
yes, B
Abnormal location of ears
17% (2/12)
/
N
N
N
N
N
N
N
N
yes
N
N
yes
Colobomas
17% (2/12)
80.8% (189/234)
N
N
N
N
N
N
N
yes
N
yes
N
N
Hypertelorism
8% (1/12)
/
N
N
N
N
N
N
yes
N
N
N
N
N
Asymmetric crying face
33% (4/12)
/
N
N
yes
N
N
N
yes
yes
N
N
yes
N
Flat nasal bridge
8% (1/12)
/
N
N
N
N
N
N
yes
N
N
N
N
N
Micrognathia
17% (2/12)
/
N
N
N
N
N
N
N
N
N
N
yes
yes
Short neck
8% (1/12)
/
N
N
N
N
N
N
N
N
N
yes
N
N
Head circumference
33.45 (average)
/
33
36
NA
33
32
NA
35
35
33
30.5
33
34
SD (standard deviation)
/
/
(−1.37 SD)
(−0.60 SD)
(−1.22 SD)
(−1.71 SD)
(−0.40 SD)
(−1.56 SD)
(−1.27 SD)
(−2.47 SD)
(−1.22 SD)
(−1.59 SD)
Cardiovascular system
58% (7/12)
75.8% (191/252)
            
PDA
100% (12/12)
/
yes
yes
yes
yes
yes
yes
yes
yes
yes
yes
yes
yes
Atrial septal defect
33% (4/12)
/
N
yes
N
N
N
N
N
yes
N
yes
yes
N
Ventricular septal defect
8% (1/12)
/
N
N
N
N
N
N
N
N
yes
N
N
N
Pulmonary valve stenosis
8% (1/12)
/
N
N
N
N
N
N
N
N
yes
N
N
N
Right aortic arch
17% (2/12)
/
yes
N
N
N
N
yes
N
N
N
N
N
N
Coarctation of aorta
17% (2/12)
/
N
N
N
N
N
N
N
yes
N
yes
N
N
Respiratory system
17% (2/12)
             
Tracheal stenosis
8% (1/12)
/
N
N
N
N
N
N
N
yes
N
N
N
N
Laryngeal obstruction
8% (1/12)
/
N
N
N
N
N
N
N
N
N
N
yes (II)
N
Hypoplasia of Laryngeal cartilage
8% (1/12)
/
N
N
N
N
N
N
N
yes
N
N
N
N
Digestive system
67% (8/12)
/
            
Tracheo-oesophageal malformation
35% (3/12)
28.8% (42/146)
yes
N
N
yes
N
N
yes
N
N
N
N
N
Hiatal hernia
8% (1/12)
/
N
N
N
N
N
N
N
N
N
N
N
yes
Feeding difficulty
58% (7/12)
/
yes
yes
N
yes
N
yes
yes
yes
N
N
yes
N
Urogenital system
67% (6/9)
81.4% (118/145)
            
Cryptorchidism
44% (4/9)
/
N
N
N
/
N
yes, R
yes
/
yes, B
yes, B
/
N
Micropenis
33% (3/9)
/
N
N
N
/
N
N
yes
/
N
yes
/
yes
Hypospadias
11% (1/9)
/
N
N
N
/
N
N
yes
/
N
N
/
N
Small scrotum
11% (1/9)
/
N
N
N
/
yes, B
N
N
/
N
N
/
N
Limbs
17% (2/12)
/
            
Polydactyly
8% (1/12)
/
N
yes, LH
N
N
N
N
N
N
N
N
N
N
Syndactyly
8% (1/12)
/
N
N
N
N
N
N
N
yes, LF, 4–5
N
N
N
N
Immudeficiency
8% (1/12)
/
NA
NA
NA
NA
NA
NA
NA
NA
NA
NA
NA
yes
Diagnosis
75% (9/12)
/
            
Bergmann
67% (8/12)
/
N
yes
N
N
N
yes
yes
yes
yes
yes
yes
yes
Blake
67% (8/12)
/
N
yes
yes
N
N
yes
yes
yes
N
yes
yes
yes
Verlos
0
/
N
N
N
N
N
N
N
N
N
N
N
N
Hale
42% (5/12)
/
N
yes
yes
N
N
N
yes
yes
N
N
yes
N
Death
33% (4/12)
/
N
N
yes
N
N
N
yes
yes
N
yes
N
N
B bilateral, Blank patient without this feature, F female, GA gestational age, IAB immediately after birth, L left, LF left food, LH left hand, M male, N do not have this feature, NA the missing value, PDA patent ductus arteriosus, R right, T term baby

Variants of CHD7

Twelve pathogenic/likely pathogenic variants in the CHD7 gene were identified. We detected 6 reported pathogenic variants and 6 novel variants, including 4 frameshifts, 4 stop-gain, 2 splice-donor region, 1 intron variant and 1 missense variant (Table 2). Stop-gain and frameshift variants accounted for 67% of variants in this study. None of these variants are included in the gnomeAD database (http:// gnomad.​broadinstitute.​org/), the 1000 gnommeAD database (http://​gnomad-old.​broadinstitute.​org/​) or our internal database (1833 probands and 6893 families). Among all six novel variants, Glu2408* and Lys651* (NM_017780) were identified as de novo variants by Sanger sequencing. We mapped the 12 variants into the structure of the CHD7 protein (http://​www.​ebi.​ac.​uk/​interpro/​). Two variants were located in the chromodomain, two were located in the SANT domain, one in the ATP-binding domain belongs to the Helicase superfamily (Helicase N domain) and one was in the BRK domain. We did not detect CHD7 copy number variants in our study.
Table 2
Genotype of Patients
NO.
Region
Nucleotide Change
Amino Acid Change
Type of Variants
Familial Targeted Variants Study
gnomAD/1000gene
Score of S/PP/MT
S/PP/MT
References
Classification
1
exon12
c.3082A > G
p.Ile1028Val
missense
/
0/0
0/0.894/1
D/PD/D
PMID 15300250
P
2
exon31
c.6165_6166del
p.Tyr2056Profs*3
frameshift
/
0/0
/
/
PMID 22461308
P
3
intron 25
c.5405-17G > A
/
intron
De novo
0/0
/
/
PMID 16155193
P
4
exon8
c.2504_2508del
p.Tyr835Serfs*14
frameshift
/
0/0
/
/
PMID 16155193
P
5
intron 27
c.5607 + 1G > T
/
splice_donor
/
0/0
/
/
/
LP
6
exon34
c.7222G > T
p.Glu2408*
stop-gain
De novo
0/0
0.19/NA/1
T/NA/D
/
P
7
intron 11
c.2957 + 5G > A
/
splice_donor
/
0/0
/
/
PMID 22461308
P
8
exon3
c.1951_1952delinsT
p.Lys651*
stop-gain
De novo
0/0
/
/
/
P
9
exon37
c.8062dupA
p.Ile2688Asnfs*3
frameshift
/
0/0
/
/
/
LP
10
exon30
c.6015_6018delAAAA
p.Lys2005Asnfs*37
frameshift
/
0/0
/
/
/
LP
11
exon8
c.2572C > T
p.Arg858*
stop-gain
De novo
0/0
0.1/N/1
T/N/D
PMID 16155193
P
12
exon30
c.5971C > T
p.Gln1991*
stop-gain
/
0/0
0.11/N/1
T/N/D
/
LP
D deleterious, MT MutationTaster, N normal, NA none, LP likely pathogenic, P pathogenic, PD probably damaging, PMID PubMed ID number, PP Polyphen2, S SIFT, T tolerant

Cranial MRI analysis results

MRI analysis revealed significant volume loss in the cingulate gyrus, occipital lobe, and cerebellum and volume gain in the left medial and inferior temporal gyri anterior white matter parts among neonates with the CHD7 variant (Fig. 1). In addition, different subregions showed different levels of volume loss among these three regions (Table 3).
Table 3
Volume loss regions of CHARGE syndrome patients
Region of Brain
Volume loss
p-value
atlas50_17_Cerebellum left
−0.20947782
0.027094616
atlas50_18_Cerebellum right
− 0.229208438
0.016167437
atlas50_22_Occipital lobe right
−0.341721802
0.000452598
atlas50_23_Occipital lobe left
−0.210090999
0.037293534
atlas50_34_Cingulate gyrus, posterior part right
−0.219671229
0.028380689
atlas50_35_Cingulate gyrus, posterior part left
−0.220531078
0.029805553
atlas7_6_Cerebellum + Brainstem
−0.20603902
0.026648778
atlas87_22_Occipital lobe right GM
−0.370674616
0.000147048
atlas87_23_Occipital lobe left GM
−0.230148083
0.022420291
atlas87_27_Lateral occipitotemporal gyrus, gyrus fusiformis posterior part left GM
−0.192364134
0.0431107
atlas87_34_Cingulate gyrus, posterior part right GM
−0.213154535
0.034225732
atlas87_59_Medial and inferior temporal gyri anterior part left WM
0.214177329
0.03865777
atlas87_65_Occipital lobe right WM
−0.307703804
0.001591836
atlas87_77_Cingulate gyrus, posterior part right WM
−0.217167864
0.030227307
atlas87_78_Cingulate gyrus, posterior part left WM
−0.243852741
0.015847061
atlas87_84_Extra-cranial background
−0.228192206
0.0277017

Discussion

The NGS test largely reduced the turnaround time of high-sequencing genetic tests; therefore, genetic diseases can be rapidly diagnosed in neonatal patients. To the best of our knowledge, this is the first study to investigate the phenotype spectrum of neonatal patients with likely pathogenic or pathogenic CHD7 variants. According to the four reported diagnostic criteria of CHARGE syndrome, 0–67% of newborns received a clinical diagnosis. This may be because some typical features of CHARGE syndrome (aplasia/dysplasia of the semicircular canals, cleft lip/palate and choanal atresia) are not observed in this population, and some features present later in childhood (growth retardation and mental retardation) cannot be diagnosed in neonatal period. Proposed in 2016, pathogenic CHD7 variant status is now a major criterion in CHARGE syndrome diagnoses [11]. Criteria focusing on typical clinical phenotypes may exclude patients with a mild phenotype in early life.
Digestive and respiratory problems are the primary causes of postneonatal demise in CHARGE syndrome [17]. Complex digestive anomalies in patients with CHARGE syndrome are often highly prevalent at birth and require long-term management [18, 19]. In this study, 67% (8/12) of patients had digestive system anomalies, including feeding difficulty, tracheo-oesophageal malformation and hiatal hernia. Tracey Allen et al. [20] reported that tracheo-oesophageal malformation was regarded as a conditional finding in neonates and a major factor influencing morbidity. Tracheoesophageal fistula was previously reported as an uncommon feature among CHARGE syndrome patients with a prevalence between 8%~ 18% [21]. However, Conny van Ravenswaaij-Arts [22] reported a higher prevalence (28.8%, 42/146). In our study, 3 of 12 (25%) patients had tracheo-oesophageal malformation. Our results showed that feeding difficulty was the main feature among neonatal patients. Therefore, the possibility of CHARGE syndrome should be kept in mind when newborns suffer from feeding difficulty for a period.
Heart defects are predominant features in CHARGE syndrome patients and patients with pathogenic CHD7 variants [23]. Patent ductus arteriosus is more commonly seen in patients with CHD7 pathogenic variants than nonsyndromic heart malformations [24]. In our study, all patients had patent ductus arteriosus. As the patent ductus arteriosus can be closed within 3 months of age in most babies, the proportion of heart defects, including patent ductus arteriosus, may be overestimated in the neonatal population.
In our study, 4 neonates with CHARGE syndrome had an asymmetric crying face. To the best of our knowledge, this study first reported the asymmetric crying face in CHARGE syndrome. An asymmetric crying face is recognized in neonates as lower lip asymmetry present only with crying. The etiology includes facial nerve compression and faulty facial muscle/nerve development. An asymmetric crying face is often seen in 22q11.2 deletion syndrome patients; however, it has not been reported in CHARGE syndrome patients [25, 26]. This could be because the importance of this symptom has not received enough attention.
Respiratory distress during feeding, regurgitation and persistent frothy salivation indicate a high risk of esophageal atresia or tracheoesophageal fistulae [27]. Treatment of feeding difficulty caused by esophageal atresia needs a multidisciplinary team to manage additional specific medical or physiological problems [28]. Although the survival of these patients is as high as 90%, long-term complications are common, continuous and challenging [29]. Additionally, surviving patients diagnosed with CHARGE syndrome most likely suffer from development delays, especially in motor and language development [30].
For CHARGE syndrome patients with a CHD7 pathogenic variant, the relationship between genotype and phenotype is unclear [10]. In this study, stop-gain, frameshift and splice variants were the main variants, which is in agreement with a previous study [31]. The patients in this study with the known pathogenic variant presented different phenotypes from the reported patients. All this indicates that CHARGE syndrome is a highly heterogeneous disease and CHD7 gene analysis is important for comprehensive assessment.
MRI was used to detect cochlear abnormalities in CHARGE syndrome patients. Recent studies showed that CHD7 affects neurogenesis by activating neuron stem cells and progenitors [32]. Conditional genetic deficiency of Chd7 in mice led to abnormalities in corpus callosum and cerebellum [33, 34]. Christa M. de Geus et al. published a cranial imaging evaluation checklist for CHARGE patients based on a literature review [35]. Cerebellum dysplasia was included as a cranial abnormality, but abnormalities of the cingulate gyrus and occipital lobe were not mentioned. Therefore, our study provides evidence to expand the abnormalities presented in the cranial imaging of CHARGE patients. According to previously studies, the cingulate gyrus processed and modulated gastrointestinal sensory signals [36], and the cerebellum involved in the regulation of feeding behavior [37]. These findings may explain why feeding difficulty is the dominant feature of neonatal CHARGE patients. The relationship between the occipital lobe, temporal lobe and feeding difficulty remains unknown. As only a few cranial MRI data of neonatal patients can be used for region division, only four patients were analyzed in this study. High-quality data collection from neonatal patients will be helpful for further study.
In this study, we reported 12 neonatal patients with CHD7 pathogenic/likely pathogenic variants. Six novel variants in the CHD7 gene were identified, expanding the variant database of the CHD7 gene. Diagnostic criteria of CHARGE syndrome focusing on only typical clinical features may underestimate its neonatal incidence. CHARGE syndrome and CHD7 pathogenic variants should be suspected in newborns who have feeding difficulty and one or more malformations.

Conclusions

Our study found that diagnostic criteria of CHARGE syndrome focusing on only typical clinical features may underestimate neonatal incidence. Based on a relatively unbiased neonatal cohort, we concluded that CHARGE syndrome and CHD7 gene variants should be suspected in newborns who have feeding difficulty, and one or more malformations.

Acknowledgements

We are very grateful to the patient families and thanks for their trust for our lab. Thanks the bioinformatic team members in our lab for data analysis. This work was funded by the National Natural Science Foundation of China (81720108018), the National Key Research and Development Program of China (2016YFC0905100), the Shanghai Municipal Commission of Health and Family Planning (GDEK201701), the Shanghai Shen Kang Hospital Development Center (SHDC12017110).

Funding

This work was supported by grants from the National Natural Science Foundation of China (81720108018), the National Key Research and Development Program of China (2016YFC0905100), the Shanghai Municipal Commission of Health and Family Planning (GDEK201701), the Shanghai Shen Kang Hospital Development Center (SHDC12017110). They provided support for data collection and analysis.

Availability of data and materials

In this study, filtered variants’ information of 12 patients were provided as Additional file 1: Table S1. Please contact corresponding author (Lin Yang) for more data.
This study was approved by the ethics committees of Children’s Hospital, Fudan University. The criteria for genetic testing were approved by ethics committees of Children’s Hospital, Fudan University (2014–107). The informed consent was in the written from and was obtained from all participants.
Written informed consents were obtained from the patients for publication of this study.

Competing interests

The authors declare that they have no competing interests.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. 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.
Literatur
1.
Zurück zum Zitat Hsu P, Ma A, Wilson M, Williams G, Curotta J, Munns CF, Mehr S. CHARGE syndrome: a review. J Paediatr Child Health. 2014;50:504–11.CrossRef Hsu P, Ma A, Wilson M, Williams G, Curotta J, Munns CF, Mehr S. CHARGE syndrome: a review. J Paediatr Child Health. 2014;50:504–11.CrossRef
2.
Zurück zum Zitat Blake KD, Davenport SL, Hall BD, Hefner MA, Pagon RA, Williams MS, et al. CHARGE association: an update and review for the primary pediatrician. Clin Pediatr (Phila). 1998;37:159–73.CrossRef Blake KD, Davenport SL, Hall BD, Hefner MA, Pagon RA, Williams MS, et al. CHARGE association: an update and review for the primary pediatrician. Clin Pediatr (Phila). 1998;37:159–73.CrossRef
3.
Zurück zum Zitat Verloes A. Updated diagnostic criteria for CHARGE syndrome: a proposal. Am J Med Genet A. 2005;133A:306–8.CrossRef Verloes A. Updated diagnostic criteria for CHARGE syndrome: a proposal. Am J Med Genet A. 2005;133A:306–8.CrossRef
4.
Zurück zum Zitat Vissers LE, van Ravenswaaij CM, Admiraal R, Hurst JA, de Vries BB, Janssen IM, et al. Mutations in a new member of the chromodomain gene family cause CHARGE syndrome. Nat Genet. 2004;36:955–7.CrossRef Vissers LE, van Ravenswaaij CM, Admiraal R, Hurst JA, de Vries BB, Janssen IM, et al. Mutations in a new member of the chromodomain gene family cause CHARGE syndrome. Nat Genet. 2004;36:955–7.CrossRef
5.
Zurück zum Zitat Hall JA, Georgel PT. CHD proteins: a diverse family with strong ties. Biochem Cell Biol. 2007;85:463–76.CrossRef Hall JA, Georgel PT. CHD proteins: a diverse family with strong ties. Biochem Cell Biol. 2007;85:463–76.CrossRef
6.
Zurück zum Zitat Marfella CG, Imbalzano AN. The Chd family of chromatin remodelers. Mutat Res. 2007;618:30–40.CrossRef Marfella CG, Imbalzano AN. The Chd family of chromatin remodelers. Mutat Res. 2007;618:30–40.CrossRef
7.
Zurück zum Zitat Manning BJ, Yusufzai T. The ATP-dependent chromatin remodeling enzymes CHD6, CHD7, and CHD8 exhibit distinct nucleosome binding and remodeling activities. J Biol Chem. 2017;292:11927–36.CrossRef Manning BJ, Yusufzai T. The ATP-dependent chromatin remodeling enzymes CHD6, CHD7, and CHD8 exhibit distinct nucleosome binding and remodeling activities. J Biol Chem. 2017;292:11927–36.CrossRef
8.
Zurück zum Zitat Schnetz MP, Bartels CF, Shastri K, Balasubramanian D, Zentner GE, Balaji R, et al. Genomic distribution of CHD7 on chromatin tracks H3K4 methylation patterns. Genome Res. 2009;19:590–601.CrossRef Schnetz MP, Bartels CF, Shastri K, Balasubramanian D, Zentner GE, Balaji R, et al. Genomic distribution of CHD7 on chromatin tracks H3K4 methylation patterns. Genome Res. 2009;19:590–601.CrossRef
9.
Zurück zum Zitat Bajpai R, Chen DA, Rada-Iglesias A, Zhang J, Xiong Y, Helms J, et al. CHD7 cooperates with PBAF to control multipotent neural crest formation. Nature. 2010;463:958–62.CrossRef Bajpai R, Chen DA, Rada-Iglesias A, Zhang J, Xiong Y, Helms J, et al. CHD7 cooperates with PBAF to control multipotent neural crest formation. Nature. 2010;463:958–62.CrossRef
10.
Zurück zum Zitat Bergman JE, Janssen N, Hoefsloot LH, Jongmans MC, Hofstra RM, van Ravenswaaij-Arts CM. CHD7 mutations and CHARGE syndrome: the clinical implications of an expanding phenotype. J Med Genet. 2011;48:334–42.CrossRef Bergman JE, Janssen N, Hoefsloot LH, Jongmans MC, Hofstra RM, van Ravenswaaij-Arts CM. CHD7 mutations and CHARGE syndrome: the clinical implications of an expanding phenotype. J Med Genet. 2011;48:334–42.CrossRef
11.
Zurück zum Zitat Hale CL, Niederriter AN, Green GE, Martin DM. Atypical phenotypes associated with pathogenic CHD7 variants and a proposal for broadening CHARGE syndrome clinical diagnostic criteria. Am J Med Genet A. 2016;170A:344–54.CrossRef Hale CL, Niederriter AN, Green GE, Martin DM. Atypical phenotypes associated with pathogenic CHD7 variants and a proposal for broadening CHARGE syndrome clinical diagnostic criteria. Am J Med Genet A. 2016;170A:344–54.CrossRef
12.
Zurück zum Zitat 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;17:405–24.CrossRef 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;17:405–24.CrossRef
13.
Zurück zum Zitat Li X, Morgan PS, Ashburner J, Smith J, Rorden C. The first step for neuroimaging data analysis: DICOM to NIfTI conversion. J Neurosci Methods. 2016;264:47–56.CrossRef Li X, Morgan PS, Ashburner J, Smith J, Rorden C. The first step for neuroimaging data analysis: DICOM to NIfTI conversion. J Neurosci Methods. 2016;264:47–56.CrossRef
14.
Zurück zum Zitat Tustison NJ, Avants BB, Cook PA, Zheng Y, Egan A, Yushkevich PA, Gee JC. N4ITK: improved N3 bias correction. IEEE Trans Med Imaging. 2010;29:1310–20.CrossRef Tustison NJ, Avants BB, Cook PA, Zheng Y, Egan A, Yushkevich PA, Gee JC. N4ITK: improved N3 bias correction. IEEE Trans Med Imaging. 2010;29:1310–20.CrossRef
15.
Zurück zum Zitat Oishi K, Mori S, Donohue PK, Ernst T, Anderson L, Buchthal S, et al. Multi-contrast human neonatal brain atlas: application to normal neonate development analysis. Neuroimage. 2011;56:8–20.CrossRef Oishi K, Mori S, Donohue PK, Ernst T, Anderson L, Buchthal S, et al. Multi-contrast human neonatal brain atlas: application to normal neonate development analysis. Neuroimage. 2011;56:8–20.CrossRef
16.
Zurück zum Zitat Avants BB, Epstein CL, Grossman M, Gee JC. Symmetric diffeomorphic image registration with cross-correlation: evaluating automated labeling of elderly and neurodegenerative brain. Med Image Anal. 2008;12:26–41.CrossRef Avants BB, Epstein CL, Grossman M, Gee JC. Symmetric diffeomorphic image registration with cross-correlation: evaluating automated labeling of elderly and neurodegenerative brain. Med Image Anal. 2008;12:26–41.CrossRef
17.
Zurück zum Zitat Bergman JE, Blake KD, Bakker MK, du Marchie SG, Free RH, Van Ravenswaaij-Arts CM. Death in CHARGE syndrome after the neonatal period. Clin Genet. 2010;77:232–40.CrossRef Bergman JE, Blake KD, Bakker MK, du Marchie SG, Free RH, Van Ravenswaaij-Arts CM. Death in CHARGE syndrome after the neonatal period. Clin Genet. 2010;77:232–40.CrossRef
18.
Zurück zum Zitat Hudson A, Macdonald M, Friedman JN, Blake K. CHARGE syndrome gastrointestinal involvement: from mouth to anus. Clin Genet. 2016. Hudson A, Macdonald M, Friedman JN, Blake K. CHARGE syndrome gastrointestinal involvement: from mouth to anus. Clin Genet. 2016.
19.
Zurück zum Zitat Dobbelsteyn C, Peacocke SD, Blake K, Crist W, Rashid M. Feeding difficulties in children with CHARGE syndrome: prevalence, risk factors, and prognosis. Dysphagia. 2008;23:127–35.CrossRef Dobbelsteyn C, Peacocke SD, Blake K, Crist W, Rashid M. Feeding difficulties in children with CHARGE syndrome: prevalence, risk factors, and prognosis. Dysphagia. 2008;23:127–35.CrossRef
20.
Zurück zum Zitat Allen T. CHARGE syndrome: diagnosis and clinical management in the NICU. Adv Neonatal Care 2012,12:336–342, 343–344.CrossRef Allen T. CHARGE syndrome: diagnosis and clinical management in the NICU. Adv Neonatal Care 2012,12:336–342, 343–344.CrossRef
21.
Zurück zum Zitat Lalani SR, Safiullah AM, Fernbach SD, Harutyunyan KG, Thaller C, Peterson LE, et al. Spectrum of CHD7 mutations in 110 individuals with CHARGE syndrome and genotype-phenotype correlation. Am J Hum Genet. 2006;78:303–14.CrossRef Lalani SR, Safiullah AM, Fernbach SD, Harutyunyan KG, Thaller C, Peterson LE, et al. Spectrum of CHD7 mutations in 110 individuals with CHARGE syndrome and genotype-phenotype correlation. Am J Hum Genet. 2006;78:303–14.CrossRef
22.
Zurück zum Zitat van Ravenswaaij-Arts C, Martin DM. New insights and advances in CHARGE syndrome: diagnosis, etiologies, treatments, and research discoveries. Am J Med Genet C Semin Med Genet. 2017;175:397–406.CrossRef van Ravenswaaij-Arts C, Martin DM. New insights and advances in CHARGE syndrome: diagnosis, etiologies, treatments, and research discoveries. Am J Med Genet C Semin Med Genet. 2017;175:397–406.CrossRef
23.
Zurück zum Zitat Corsten-Janssen N, Scambler PJ. Clinical and molecular effects of CHD7 in the heart. Am J Med Genet C Semin Med Genet. 2017;175:487–95.CrossRef Corsten-Janssen N, Scambler PJ. Clinical and molecular effects of CHD7 in the heart. Am J Med Genet C Semin Med Genet. 2017;175:487–95.CrossRef
24.
Zurück zum Zitat Corsten-Janssen N, Kerstjens-Frederikse WS, du Marchie SG, Baardman ME, Bakker MK, Bergman JE, et al. The cardiac phenotype in patients with a CHD7 mutation. Circ Cardiovasc Genet. 2013;6:248–54.CrossRef Corsten-Janssen N, Kerstjens-Frederikse WS, du Marchie SG, Baardman ME, Bakker MK, Bergman JE, et al. The cardiac phenotype in patients with a CHD7 mutation. Circ Cardiovasc Genet. 2013;6:248–54.CrossRef
25.
Zurück zum Zitat Pasick C, McDonald-McGinn DM, Simbolon C, Low D, Zackai E, Jackson O. Asymmetric crying facies in the 22q11.2 deletion syndrome: implications for future screening. Clin Pediatr (Phila). 2013;52:1144–8.CrossRef Pasick C, McDonald-McGinn DM, Simbolon C, Low D, Zackai E, Jackson O. Asymmetric crying facies in the 22q11.2 deletion syndrome: implications for future screening. Clin Pediatr (Phila). 2013;52:1144–8.CrossRef
26.
Zurück zum Zitat Liang X, He B. Congenital asymmetric crying facies syndrome: a case report. Medicine (Baltimore). 2018;97:e11403.CrossRef Liang X, He B. Congenital asymmetric crying facies syndrome: a case report. Medicine (Baltimore). 2018;97:e11403.CrossRef
27.
Zurück zum Zitat Aminde LN, Ebenye VN, Arrey WT, Takah NF, Awungafac G. Oesophageal atresia with tracheo-oesophageal fistula in a preterm neonate in Limbe, Cameroon: case report & brief literature review. BMC Res Notes. 2014;7:692.CrossRef Aminde LN, Ebenye VN, Arrey WT, Takah NF, Awungafac G. Oesophageal atresia with tracheo-oesophageal fistula in a preterm neonate in Limbe, Cameroon: case report & brief literature review. BMC Res Notes. 2014;7:692.CrossRef
28.
Zurück zum Zitat Ramsay M, Birnbaum R. Feeding difficulties in children with esophageal atresia: treatment by a multidisciplinary team. Dis Esophagus. 2013;26:410–2.CrossRef Ramsay M, Birnbaum R. Feeding difficulties in children with esophageal atresia: treatment by a multidisciplinary team. Dis Esophagus. 2013;26:410–2.CrossRef
29.
Zurück zum Zitat Smith N. Oesophageal atresia and tracheo-oesophageal fistula. Early Hum Dev. 2014;90:947–50.CrossRef Smith N. Oesophageal atresia and tracheo-oesophageal fistula. Early Hum Dev. 2014;90:947–50.CrossRef
30.
Zurück zum Zitat Lalani SR, Hefner MA, Belmont JW, Davenport S. CHARGE Syndrome; 1993. Lalani SR, Hefner MA, Belmont JW, Davenport S. CHARGE Syndrome; 1993.
31.
Zurück zum Zitat Janssen N, Bergman JE, Swertz MA, Tranebjaerg L, Lodahl M, Schoots J, et al. Mutation update on the CHD7 gene involved in CHARGE syndrome. Hum Mutat. 2012;33:1149–60.CrossRef Janssen N, Bergman JE, Swertz MA, Tranebjaerg L, Lodahl M, Schoots J, et al. Mutation update on the CHD7 gene involved in CHARGE syndrome. Hum Mutat. 2012;33:1149–60.CrossRef
32.
Zurück zum Zitat Feng W, Khan MA, Bellvis P, Zhu Z, Bernhardt O, Herold-Mende C, Liu HK. The chromatin remodeler CHD7 regulates adult neurogenesis via activation of SoxC transcription factors. Cell Stem Cell. 2013;13:62–72.CrossRef Feng W, Khan MA, Bellvis P, Zhu Z, Bernhardt O, Herold-Mende C, Liu HK. The chromatin remodeler CHD7 regulates adult neurogenesis via activation of SoxC transcription factors. Cell Stem Cell. 2013;13:62–72.CrossRef
33.
Zurück zum Zitat He D, Marie C, Zhao C, Kim B, Wang J, Deng Y, et al. Chd7 cooperates with Sox10 and regulates the onset of CNS myelination and remyelination. Nat Neurosci. 2016;19:678–89.CrossRef He D, Marie C, Zhao C, Kim B, Wang J, Deng Y, et al. Chd7 cooperates with Sox10 and regulates the onset of CNS myelination and remyelination. Nat Neurosci. 2016;19:678–89.CrossRef
34.
Zurück zum Zitat Feng W, Kawauchi D, Korkel-Qu H, Deng H, Serger E, Sieber L, et al. Chd7 is indispensable for mammalian brain development through activation of a neuronal differentiation programme. Nat Commun. 2017;8:14758.CrossRef Feng W, Kawauchi D, Korkel-Qu H, Deng H, Serger E, Sieber L, et al. Chd7 is indispensable for mammalian brain development through activation of a neuronal differentiation programme. Nat Commun. 2017;8:14758.CrossRef
35.
Zurück zum Zitat de Geus CM, Free RH, Verbist BM, Sival DA, Blake KD, Meiners LC, van Ravenswaaij-Arts C. Guidelines in CHARGE syndrome and the missing link: cranial imaging. Am J Med Genet C Semin Med Genet. 2017;175:450–64.CrossRef de Geus CM, Free RH, Verbist BM, Sival DA, Blake KD, Meiners LC, van Ravenswaaij-Arts C. Guidelines in CHARGE syndrome and the missing link: cranial imaging. Am J Med Genet C Semin Med Genet. 2017;175:450–64.CrossRef
36.
Zurück zum Zitat Lawal A, Kern M, Sanjeevi A, Antonik S, Mepani R, Rittmann T, et al. Neurocognitive processing of esophageal central sensitization in the insula and cingulate gyrus. Am J Physiol Gastrointest Liver Physiol. 2008;294:G787–94.CrossRef Lawal A, Kern M, Sanjeevi A, Antonik S, Mepani R, Rittmann T, et al. Neurocognitive processing of esophageal central sensitization in the insula and cingulate gyrus. Am J Physiol Gastrointest Liver Physiol. 2008;294:G787–94.CrossRef
37.
Zurück zum Zitat Zhu JN, Wang JJ. The cerebellum in feeding control: possible function and mechanism. Cell Mol Neurobiol. 2008;28:469–78.CrossRef Zhu JN, Wang JJ. The cerebellum in feeding control: possible function and mechanism. Cell Mol Neurobiol. 2008;28:469–78.CrossRef
Metadaten
Titel
Feeding difficulty is the dominant feature in 12 Chinese newborns with CHD7 pathogenic variants
verfasst von
Xiang Chen
Kai Yan
Yanyan Gao
Huijun Wang
Guoqiang Chen
Bingbing Wu
Qian Qin
Lin Yang
Wenhao Zhou
Publikationsdatum
01.12.2019
Verlag
BioMed Central
Erschienen in
BMC Medical Genetics / Ausgabe 1/2019
Elektronische ISSN: 1471-2350
DOI
https://doi.org/10.1186/s12881-019-0813-z

Weitere Artikel der Ausgabe 1/2019

BMC Medical Genetics 1/2019 Zur Ausgabe