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

Open Access 01.12.2023 | Research

Nexus between genome-wide copy number variations and autism spectrum disorder in Northeast Han Chinese population

verfasst von: Shuang Qiu, Yingjia Qiu, Yong Li, Xiaojuan Zhu, Yunkai Liu, Yichun Qiao, Yi Cheng, Yawen Liu

Erschienen in: BMC Psychiatry | Ausgabe 1/2023

Abstract

Background

Autism spectrum disorder (ASD) is a common neurodevelopmental disorder, with an increasing prevalence worldwide. Copy number variation (CNV), as one of genetic factors, is involved in ASD etiology. However, there exist substantial differences in terms of location and frequency of some CNVs in the general Asian population. Whole-genome studies of CNVs in Northeast Han Chinese samples are still lacking, necessitating our ongoing work to investigate the characteristics of CNVs in a Northeast Han Chinese population with clinically diagnosed ASD.

Methods

We performed a genome-wide CNVs screening in Northeast Han Chinese individuals with ASD using array-based comparative genomic hybridization.

Results

We found that 22 kinds of CNVs (6 deletions and 16 duplications) were potentially pathogenic. These CNVs were distributed in chromosome 1p36.33, 1p36.31, 1q42.13, 2p23.1-p22.3, 5p15.33, 5p15.33-p15.2, 7p22.3, 7p22.3-p22.2, 7q22.1-q22.2, 10q23.2-q23.31, 10q26.2-q26.3, 11p15.5, 11q25, 12p12.1-p11.23, 14q11.2, 15q13.3, 16p13.3, 16q21, 22q13.31-q13.33, and Xq12-q13.1. Additionally, we found 20 potential pathogenic genes of ASD in our population, including eight protein coding genes (six duplications [DRD4, HRAS, OPHN1, SHANK3, SLC6A3, and TSC2] and two deletions [CHRNA7 and PTEN]) and 12 microRNAs-coding genes (ten duplications [MIR202, MIR210, MIR3178, MIR339, MIR4516, MIR4717, MIR483, MIR675, MIR6821, and MIR940] and two deletions [MIR107 and MIR558]).

Conclusion

We identified CNVs and genes implicated in ASD risks, conferring perception to further reveal ASD etiology.
Hinweise

Supplementary Information

The online version contains supplementary material available at https://​doi.​org/​10.​1186/​s12888-023-04565-7.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
ASD
Autism spectrum disorder
CNV
Copy number variation
Kb
Kilobase
aCGH
Array-based comparative genomic hybridization
miRNAs
MicroRNAs
GO
Gene Ontology
BP
Biological processes
CC
Cellular component
MF
Molecular function

Background

Autism spectrum disorder (ASD) is a common neurodevelopmental disorder with an increasing prevalence worldwide [1, 2]. ASD manifests the wide range of symptoms and severity in perceivability and socialization with others, such as limited and repetitive patterns of behavior. Both genetic and environmental factors are involved in ASD pathogenesis. Environmental factors, including viral infections, medications during pregnancy, and air pollutants, may contribute to ASD risks [3]. Compared with environmental factors, genetic factors appear to be a prerequisite for ASD development: genetic changes (mutations) may increase ASD risks; additionally, genes, such as CHD8 [4], CNTNAP2 [5], DCC [6], neurexin genes [7], SHANK1 [8], SHANK2 [9], SHANK3 [10], and WNT2 [11] may affect brain development or brain-cells communication. ASD heritability has been estimated to be 50%, reflecting that genetic factors afford main components in ASD etiology [12].
ASD begins in early childhood. Children with ASD usually show symptoms of autism within the first year, and regress during a period between one and two years of age. Although there is no specific medication for ASD patients [13], early treatment can confer the lives of children with ASD beneficially. Gene-based test provides an impressive opportunity to identify potential infants with ASD [8].
Accumulating whole-genome, association, and linkage studies have strongly documented the roles of genes in ASD [1416]. Copy-number variants (CNVs) are defined as deletions and duplications of DNA segments in the genome greater than one kilobase (Kb) [17, 18]. De novo CNV events have been found to be implicated in the etiology of depression, schizophrenia, bipolar disorder, attention deficit hyperactivity disorder, and ASD [1922]. Array-based comparative genomic hybridization (aCGH) technology has proven to be a rapid method to detect the association between CNVs and ASD risks [23, 24]. Large simplex ASD cohort studies show that the rate of rare de novo CNVs is significantly higher in affected siblings (5.8–7.9%) than that in unaffected siblings (1.7–1.9%) [25, 26]. CNVs at 1q21.1, 2p16.3, 3q29, 7q11.23, 15q11.2–13.1, 16p11.2, 17p11.2, 17q12, and 22q11.2 are associated with ASD risks [24, 27]. Moreover, CNVs in NRXN1, SETD5, HDAC9, and PARK2 are found to be associated with ASD risks [2830]. However, there exist substantial differences in terms of location and frequency of some CNVs in the general Asian population [31]. In this paper, we investigated in CNVs in Northeast Han Chinese individuals with ASD.

Methods

Study subjects

We enrolled 16 individuals with ASD aged 2 to 7 years from the Chunguang Rehabilitation hospital in Jilin Province, after cases with fragile X syndrome, Rett syndrome, chromosomal abnormalities, or any neurological or psychiatric disorders were excluded. The individuals with ASD were diagnosed by Pediatric Neurology and Neurorehabilitation doctors using the Diagnostic and Statistical Manual of Mental Disorders (5th edition) [32]. All the individuals with ASD were northeast Han Chinese.

DNA extraction and Detection of CNVs

Genomic DNA was extracted from peripheral blood samples using DNA extraction kits, according to the manufacturer’s instructions (DP319 TIANamp Blood DNA Kit, TIANGEN BiotechCo. Ltd, Beijing, China) [33]. We used Nano Drop (Cat#ND-1000, ThermoFisher, Waltham, MA, US) and 1% agarose gel electrophoresis to check the quantity and quality of the isolated DNA. We used aCGH for genome-wide CNVs screening (Agilent SurePrint G3 Human CGH 60 K). Male and female DNA samples were hybridized with male and female reference DNA samples (G1471, G1521, Promega), respectively.

aCGH data analysis

We converted the raw data using FEATURE EXTRACTION software 10.7 and analyzed CNVs using Agilent CytoGenomics software 4.0.3.12 (Agilent technologies, Santa Clara, CA, US). The human genome assembly NCBI36/hg18 was used as a reference. The analysis settings for CNVs calling were Aberration Detection Method 2 algorithm, centralization threshold 6, bin size 10, and minimum number of adjacent probes 3. Thresholds were set via log2-ratio (log2 R) (for detecting duplications, log2 R ≥ 0.25; for detecting deletions, log2 R ≤ -0.25).

Identification of potential pathogenic CNVs of ASD

We calculated the frequency of each overlapping or non-overlapping CNV in DNA samples from our subjects. CNVs with same overlapping sequence were defined as one kind of CNV, and a non-overlapping CNV was also sorted as one kind of CNV. The circular plot of CNVs distribution in chromosome was visualized using circlize package in R3.6.2 software [34]. We converted our bed file from exon coordinates for human build NCBI36 (hg18) into GRCh37 (hg19) using UCSC LiftOver tool (http://​genome.​ucsc.​edu/​cgi-bin/​hgLiftOver). The classification of CNVs was based on Database of Genomic Variants (DGV, http://​dgv.​tcag.​ca/​dgv/​app/​home), Database of Genomic Structural Variation (dbVar, https://​www.​ncbi.​nlm.​nih.​gov/​dbvar), Clinical Genome Resources (ClinGen, https://​clinicalgenome.​org/​), and Online Mendelian Inheritance in Man (OMIM, https://​www.​ncbi.​nlm.​nih.​gov/​omim). CNVs were classified as benign, likely benign, a variant of unknown significance (VOUS), likely pathogenic, and pathogenic using AnnotSV program (https://​lbgi.​fr/​AnnotSV/​) according to American College of Medical Genetics guideline [35].
CNVs were considered of strong putative interest when they reached the following criteria: (1) they were classified as likely pathogenic or pathogenic; (2) they were of large size (> 100 kb); (3) they had been found in the knowledgebases for the genetic evidence of ASD (Simons Foundation Autism Research Initiative [SFARI, https://​www.​sfari.​org/​resource/​sfari-gene/​], or AutismKB [http://​www.​autismkb.​com]); (4) they had been found in the Database of genomic variation and phenotype in Humans using Ensembl Resources (DECIPHER, https://​decipher.​sanger.​ac.​uk/​about#overview); and (5) they contained previously reported ASD-relative genes.

Identification of potential pathogenic genes of ASD

We selected potential pathogenic genes within potential pathogenic CNVs on the basis of the following criteria: (1) genes enriched in ASD-related pathways; and (2) same genes shared with 363 genes in SFARI classified as high-confidence or strong-candidate, or with 228 genes in AutismKB classified as high-confidence.

Identification of potential pathogenic microRNAs of ASD

MicroRNAs (miRNAs) are involved in the pathogenesis of ASD [30, 36]. Because genes implicated in CNVs that we found encode miRNAs, we further selected potential-pathogenic-CNVs-encoded miRNAs by retrieving PubMed according to experimental evidence documenting nervous system dysfunction.

Bioinformatic analysis

The Gene Ontology (GO) and KEGG pathway analyses of the genes from potential pathogenic CNVs were performed using clusterProfiler package in R3.6.2 software [37, 38]. P-value < 0.05 was considered statistically significant. miRWalk 2.0 database, which contained 12 miRNA-target-prediction database, was used to predict target genes of CNVs-encoded miRNAs [39]. We selected the target genes according to the criteria—target genes existed in at least seven of the 12 databases. Moreover, interactive relationship between CNVs-encoded miRNAs and target genes was presented using Cytoscape 3.8.0 (http://​www.​cytoscape.​org/​).

Results

Identification of CNVs

To detect CNVs, aCGH was performed in all DNA samples from the 16 subjects with ASD (13 males and 3 females). We identified 364 CNVs (153 deletions and 211 duplications) with an average genomic size of 211.982 kb (114.091 kb for deletions and 258.705 kb for duplications). The mean number of CNVs per subject was 22.750 (9.563 for deletions and 13.188 for duplications). The mean number of deletions in male (10.462) was greater than that in females (5.667) (Table 1).
Table 1
The Characteristics of genome-wide CNVs among our subjects
Characters
Number (proportion %) of CNVs
Median CNV Size (kb)
Mean Number of CNVs per Subject
Total
364 (100.0)
211.982 (78.813, 705.031)
22.750
 Male
 299 (82.1)
 213.241 (81.797, 864.947)
23.000
 Female
 65 (17.9)
 132.360 (73.256, 305.926)
21.667
Duplication
211 (58.0)
258.705 (86.903, 708.853)
13.188
 Male
 163 (77.3)
 305.926 (86.903, 1092.277)
12.538
 Female
 48 (22.7)
 133.712 (87.668, 377.979)
16.000
Deletion
153 (42.0)
114.091 (73.256, 656.149)
9.563
 Male
 136 (88.9)
 114.518 (73.256, 693.563)
10.462
 Female
 17 (11.1)
 73.256 (60.021, 135.433)
5.667
A total of 13 males and 3 females

Identification potential pathogenic CNVs of ASD

A total of 20 CNVs from 364 CNVs failed to be converted to GRCh37 (hg19); thus, we obtained 72 benign, 65 likely benign, 9 VOUS, 167 likely pathogenic, and 31 pathogenic CNVs (Table 2). We found that more than half CNVs were likely pathogenic or pathogenic.
Table 2
The Classification of CNVs based on ACMG
Classification
Total (%)
Duplication (%)
Deletion (%)
Benign
72 (19.8)
35 (16.6)
37 (24.2)
Likely Benign
65 (17.9)
59 (28.0)
6 (3.9)
VOUS
9 (2.5)
9 (4.3)
0 (0.0)
Likely Pathogenic
167 (45.9)
68 (32.2)
99 (64.7)
Pathogenic
31 (8.5)
28 (13.3)
3 (2.0)
VOUS variant of unknown significance; A total of 20 CNVs failed to be converted to GRCh37 (hg19), thus, the total proportion was not equal to 100%. ACMG American College of Medical Genetics guideline
After we calculated the frequency of each overlapping or non-overlapping CNV in DNA samples from our subjects, 344 CNVs were converted into 115 kinds of CNVs (45 deletions and 70 duplications). All the 115 kinds of CNVs were further classified (benign: 13 kinds; likely benign: 18 kinds; VOUS: two kinds; likely pathogenic: 60 kinds; and pathogenic: 13 kinds) (Supplementary Table 1). The distribution of the 115 kinds of CNVs in chromosome is visualized by circular plot (Fig. 1).
We investigated SFARI, AutismKB, and DECIPHER database to identify potential pathogenic CNVs from the 115 kinds of CNVs, revealing that 22 kinds of CNVs (6 deletions and 16 duplications) were potentially pathogenic. The 22 kinds of CNVs were distributed in chromosome 1, 2, 5, 7, 10, 11, 12, 14, 15, 16, 22, and X. Among them, 19 kinds of CNVs were rare (Table 3).
Table 3
Summary of candidate CNVs of ASD
M/F No
Coordinates, hg18
Cytoband
Size (Kb)
CNV Type
Classification
Number of genes
Gene Name
1/0
chr1:1,179,223–2,271,500
1p36.33
1092.277
Duplication
LP
42
DVL1, TMEM52
1/0
chr1:5,998,727–6,334,157
1p36.31
335.430
Duplication
LP
9
CHD5
4/0
chr1:225,876,894–226,738,916
1q42.13
862.022
Duplication
LP
26
PRSS38
1/0
chr2:31,412,158–32,712,484
2p23.1-p22.3
1300.327
Deletion
P
12
BIRC6, SPAST, SRD5A2
1/0
chr5:360,041–873,365
5p15.33
513.324
Duplication
LP
11
AHRR, EXOC3, PDCD6
1/0
chr5:1,115,468–8,452,427
5p15.33-p15.2
7336.959
Duplication
P
49
ADCY2, SLC6A3, TERT
2/0
chr7:524,935–1,037,461
7p22.3
512.526
Duplication
LP
13
ADAP1, PRKAR1B
1/0
chr7:1,037,461–2,536,804
7p22.3-p22.2
1499.343
Duplication
LP
26
INTS1
1/0
chr7:103,622,888–104,803,388
7q22.1-q22.2
1180.501
Deletion
LP
8
KMT2E, LHFPL3
13/0
chr10:89,540,133–91,524,263
10q23.2-q23.31
1984.131
Deletion
LP
31
PTEN
1/0
chr10:127,658,856–135,254,513
10q26.2-q26.3
7595.658
Duplication
P
58
DOCK1, EBF3, GLRX3
5/2
chr11:498,019–2,179,368
11p15.5
1681.349
Duplication
P
84
BRSK2, CD151, CTSD, DEAF1, DRD4, HRAS, IGF2, PHRF1, TALDO1
3/0
chr11:132,773,688–134,043,707
11q25
1270.019
Duplication
LP
16
IGSF9B
6/0
chr12:25,156,062–27,414,420
12p12.1-p11.23
2258.359
Deletion
LP
17
KRAS, MED21
1/0
chr14:22,086,438–22,354,007
14q11.2
267.569
Deletion
LP
4
SLC7A7
1/0
chr15:29,809,025–30,298,155
15q13.3
489.131
Deletion
P
1
CHRNA7
0/2
chr16:2,021,433–2,484,806
16p13.3
463.373
Duplication
LP
32
PGP, PKD1, RNPS1, SLC9A3R2, TRAF7, TSC2
0/3
chr16:2,484,806–2,747,528
16p13.3
262.722
Duplication
LP
13
SRRM2
2/0
chr16:61,464,644–64,965,235
16q21
3500.591
Duplication
LP
4
CDH11
4/0
chr22:46,395,224–49,412,774
22q13.31-q13.33
3017.550
Duplication
P
44
CHKB, MAPK12, MAPK8IP2, PANX2, PPP6R2, SBF1, TRABD
2/0
chr22:49,412,774–49,525,130
22q13.31-q13.33
112.356
Duplication
P
3
SHANK3
1/0
chrX:67,331,017–68,768,438
Xq12-q13.1
1437.422
Duplication
P
8
OPHN1
M Male, F Female. M/F No. means the number of CNV among male/female. P Pathogenic, LP Likely Pathogenic. The genes were reported to be related with ASD

Identification of potential pathogenic genes with CNVs of ASD

A total of 511 genes from the 22 potential pathogenic CNVs were functionally annotated by GO. The annotated genes were classified into three GO domains (biological processes [BP], cellular component [CC], and molecular function [MF]). For BP, some gene sets were enriched in synaptic-related functions, including modulation of chemical synaptic transmission (GO: 0050804), regulation of trans-synaptic signaling (GO: 0099177), positive regulation of excitatory postsynaptic potential (GO: 2000463), positive regulation of synaptic transmission (GO: 0050806), chemical postsynaptic transmission, (GO: 0099565), modulation of excitatory postsynaptic potential (GO: 0098815), and regulation of postsynaptic membrane potential (GO: 0060078), and in central nervous system related functions (positive regulation of neurological system process [GO: 0031646]). For CC, the top five CC terms included keratin filament (GO: 0045095), myelin sheath (GO: 0043209), Golgi lumen (GO: 0005796), glutamatergic synapse (GO: 0098978), and neuron to neuron synapse (GO: 0098984). For MF, the top five MF terms encompassed catecholamine binding (GO: 1901338), dopamine binding (GO: 0035240), magnesium ion binding (GO: 0000287), insulin receptor binding (GO: 0005158), and lipase activity (GO: 0016298). The top 20 GO functions are presented in Fig. 2 and Supplementary Tables 2, 3, and 4.
KEGG pathway enrichment analysis showed enriched key pathways, such as dopaminergic synapse (hsa04728), mTOR signaling pathway (hsa04150), insulin signaling pathway (hsa04910), and cholinergic synapse (hsa04725). The top 20 pathways are presented in Fig. 2 and Supplementary Table 5.
We constructed intersections among 511 genes that we found, 363 high-confidence or strong-candidate risk genes of ASD reported in SFARI database, and 228 high-confidence risk genes related to ASD reported in AutismKB database (Fig. 3). After investigating genes in the intersections, we found that cholinergic receptor nicotinic alpha 7 subunit gene (CHRNA7) was involved in the regulation of excitatory postsynaptic potential and cholinergic synapse; dopamine receptor D4 gene (DRD4) was involved in the regulation of synaptic transmission, dopamine binding, and glutamatergic synapse; HRas proto-oncogene (HRAS) played roles in the regulation of excitatory postsynaptic potential, glutamatergic synapse, and mTOR signal pathway; oligophrenin 1 gene (OPHN1) correlated with regulated synaptic signal, ionic glutamate receptor binding, and glutamatergic synapse; phosphatase and tensin homolog (PTEN) was implicated in the regulation of synaptic signal, neuron differentiation of central nervous system, ionic glutamate receptor binding, sphingolipid signaling, and mTOR signaling; SH3 and multiple ankyrin repeat domains 3 gene (SHANK3) was involved in the regulation of synaptic signal, ionic glutamate receptor binding, neuronal synapse, postsynaptic density, and asymmetric synapse; solute carrier family 6 member 3 gene (SLC6A3) played roles in dopamine binding, neurotransmitter: sodium cotransporter activity, and neurotransmitter transport activity; and TSC complex subunit 2 gene (TSC2) was involved in synapses, postsynaptic density, asymmetric synapses, and mTOR signaling pathways. Scores of all these genes (CHRNA7, DRD4, HRAS, OPHN1, PTEN, SHANK3, SLC6A3, and TSC2) in AustismKB and corresponding ranks in SFARI are listed in Table 4. DRD4, HRAS, OPHN1, SHANK3, SLC6A3, and TSC2 were in the regions of CNVs duplication. CHRNA7 and PTEN were in the regions of CNVs deletion.
Table 4
Summary of candidate genes of ASD
Gene Name
CNV Type
M/F No
Category of gene in SFARI
Score of gene in AutismKB
CHRNA7
Deletion
1/0
2
#
DRD4
Duplication
5/2
30
HRAS
Duplication
5/2
1
20
OPHN1
Duplication
1/0
2
#
PTEN
Deletion
13/0
1
78
SHANK3
Duplication
2/0
1
62
SLC6A3
Duplication
1/0
2
30
TSC2
Duplication
0/2
1
46
M Male, F Female. M/F No. means the number of CNV among male/female. —: Not reported as high-confidence or strong candidate autism risk genes in SFARI. #: Not reported as high-confidence autism risk genes in AutismKB

Identification and analysis of potential pathogenic CNVs-encoded miRNAs of ASD

We found 50 potential-pathogenic-CNVs-encoded miRNAs (45 encoded by duplication regions and 5 encoded by deletion regions). According to experimental evidence documenting nervous system dysfunction, we retrieved PubMed, identifying that 12 CNVs-encoded miRNAs were previously reported to be associated with brain or nervous system dysfunction (Table 5).
Table 5
miRNAs with function related to brain or nervous system in CNVs
miRNA ID
CNV Type
Functional relevance
Reference (PMID)
miR-202
Duplication
Depression, Glioma, Neuroblastoma
32425535; 28714009; 21654684; 24337320
miR-210
Duplication
Alzheimer's disease, Epilepsy, Glioblastoma, Glioma, Head and neck paragangliomas, Neuroblastoma, Neuroprotective effects
31092279; 23108914; 21655185; 22977270; 23902947; 25279461; 24729345; 24382515; 25481483; 24930954; 25756397; 29126304; 29362886; 31146085; 32194691; 31896490; 29226333; 30947960; 30746749; 27471387
miR-3178
Duplication
Neuropsychiatric diseases
30766477
miR-339
Duplication
Alzheimer's disease, Glioblastoma, Neuroendocrine neoplasias
32176627; 29983867; 30564636; 24352696; 30176243
miR-4516
Duplication
Glioblastoma
30559405
miR-4717
Duplication
Guillain–Barre Syndrome
27836180
miR-483
Duplication
Alzheimer's disease, Glioma, Neuroblastoma
31938135; 24577456; 22465663
miR-675
Duplication
Glioma
31468534; 28187439; 24466011
miR-6821
Duplication
Alzheimer's disease
27050411
miR-940
Duplication
Glioblastoma, Glioma
31497204; 30906627; 31934283; 29296221; 30431124
miR-107
Deletion
Alzheimer's disease, Amnestic mild cognitive impairment, Bipolar disorder, Brain disorders, Frontotemporal dementia, Glioblastomas, Glioma, Major depression, Neuroblastoma, Neurogenesis, Schizophrenia
31556571; 29258209; 28847283; 26084601; 30543171; 31250578; 27343180; 21625387; 20489155; 28578378; 25662174; 22811466; 20413881; 31778666; 31787850; 29885309; 29671226; 30056425; 30480816; 18234899; 29136645; 23811124; 27143098; 29073742; 25596705; 31605836; 31420923; 23220650; 22594617; 26223576; 23572380; 27501295; 27878295; 32124921; 23962497; 29286086; 21179570; 21111402
miR-558
Deletion
Neuroblastoma
25616966; 27276678
We intersected CNVs-encoded-miRNAs-targeted genes predicted using miRWalk 2.0 database with the union between SFARI and AutismKB (Supplementary Fig. 1). A total of 219 target genes were chosen for further study. We presented the interaction networks between CNVs-encoded miRNAs and 219 target genes (Figs. 4 and 5). The CNVs-encoded miRNAs and target genes are presented in Supplementary Tables 6 and 7.
We further investigated potential functions of the 219 target genes using GO analysis. For BP, some gene sets were enriched in synaptic-related functions, including synapse organization (GO: 0050808), modulation of chemical synaptic transmission (GO: 0050804), regulation of trans-synaptic signaling (GO: 0099177), synaptic transmission, glutamatergic (GO: 0035249), postsynaptic density organization (GO: 0097106), postsynaptic specialization organization (GO: 0099084) and regulation of glutamatergic synaptic transmission (GO: 0051966), and in central nervous system related functions, including learning or memory (GO: 0007611), cognition (GO: 0050890), and neurotransmitter transport (GO: 0006836). For CC, some gene sets were enriched in synaptic-related cellular components, including synapse membrane (GO: 0097060), postsynaptic specialization (GO: 0099572), and neuron to neuron synapse (GO: 0098984). For MF, some gene sets were enriched in ion-gated channel activity (GO: 0022839), gated channel activity (GO: 0022836), ion channel activity (GO: 0,005,216), ionotropic glutamate receptor activity (GO: 0004970), and transmitter gated channel activity (GO: 0022835). The top 20 GO functions are presented in Fig. 6 and Supplementary Tables 8, 9, and 10.
KEGG pathway enrichment analysis showed enriched key pathways, such as glutamatergic synapse (hsa04724), dopaminergic synapse (hsa04728), and Wnt signaling pathway (hsa04310). The top 20 pathways are presented in Fig. 6 and Supplementary Table 11.

Discussion

In the present study, we identified that 22 kinds of CNVs (six deletions and 16 duplications), eight protein-coding genes, and 12 miRNAs-coding genes are associated with ASD risks in northeast Chinese Han from Jilin province, China.
CNVs have repeatedly been found to correlate with ASD risks [40, 41]. In our study, we filtered 22 potential pathogenic CNVs. Individuals with deletions and duplications of 15q13.3 have been found to manifest neuropsychiatric disease and cognitive deficits [42]. In line with the discoveries of Bitar et al. [43], Bremer et al. [44], Celestino-Soper et al. [45], Chen et al. [23], Chen et al. [46], Pinto et al. [28], and Rosenfeld et al. [47], we further documented that CNVs at 5p15.33, 5p15.33-p15.2, 7p22.3, 7p22.3-p22.2, 7q22.1-q22.2, 10q26.2-q26.3, 11q25, 12p12.1-p11.23, 15q13.3, 16p13.3, 22q13.31-q13.33, and Xq12-q13.1 were associated with ASD risks. Autism-related phenotypes are common in patients with deletion or duplication at 22q13.3 [4851]. Most of the defects are due to haploinsufficiency of SHANK3 [49]. Chen et al. found a deletion at 22q13.3 in two male children with ASD and a duplication at 22q13.31-q13.33 in one male child with ASD from Taiwan, China [46]. In our study, we found a duplication at 22q13.31-q13.33 that overlaps SHANK3 from two male children with ASD, indicating that the duplication at 22q13.31-q13.33 may play a key role in ASD etiology in our population. CNVs at 15q13.3 have been found to be involved in a variety of neuropsychiatric diseases, including intellectual disability/developmental delay, epilepsy, schizophrenia, and ASD [42, 5254]. The relation between CHRNA7 at 15q13.3 and neuropsychiatric disorder phenotype has been validated intensively [53]. In accordance with the discovery of Pinto et al. [28], we also found that a deletion of CHRNA7 was associated with ASD risks.
Except CHRNA7 and SHANK3, we found CNVs-duplications (DRD4, HRAS, OPHN1, SLC6A3, and TSC2) and CNVs-deletions (PTEN). For DRD4 and HARS, we found seven children with ASD had duplications at 11p15.5, which overlaps DRD4 and HARS. Mutations in DRD4 are associated with ASD risks [5557]. The mRNA expression levels of DRD4 in peripheral blood lymphocytes are higher in people with ASD than those in healthy controls [58, 59]. Herault et al. also found positive association between HRAS and autism in French-Caucasian [60, 61]. For OPHN1 at Xq12-q13.1, Celestino-Soper et al. found a deletion of exons 7–15 of OPHN1 at Xq12 in a male child with ASD [45]. In contrast, we found a male child with ASD had a duplication at Xq12-q13.1. For SLC6A3 at 5p15.33-p15.2, Bowton et al. found SLC6A3 coding variant Ala559Val is related to ASD [62]. We further found a child with ASD had a duplication at 5p15.33-p15.2. For TSC2 at 16p13.3 and PTEN at 10q23.2-q23.31, Bourgeron et al. found that mutations in TSC2 and PTEN activate the mTOR/PI3K pathway, associating with ASD risks [63]. We found duplications at 16p13.3 in two female children with ASD. PTEN loss involved in white matter pathology in human with ASD is consistent with that in mouse models [64]. We revealed that deletions at 10q23.2-q23.31 overlapping PTEN in 13 male children with ASD, rather than 3 female children with ASD. Thus, these eight genes may be implicated in ASD etiology.
MiRNAs encoded within CNVs are important functional variants, providing a new dimension to recognize the association between genotype and phenotype [65]. MiRNAs play vital roles in governing essential aspects of inhibitory transmission and interneuron development in nervous system [66]. Deletion or duplication of a chromosomal loci changes the levels of miRNAs which further impact on neuronal function and communication [36]. In our study, 12 candidate-susceptible miRNAs-coding genes of ASD were identified (ten duplications [MIR202, MIR210, MIR3178, MIR339, MIR4516, MIR4717, MIR483, MIR675, MIR6821, and MIR940] and two deletions [MIR107 and MIR558]). BDNF, a brain-derived neurotrophic factor and a member of the neurotrophic factor family, is a target gene of miR-202 [67]. Moreover, we further predicted that miR-4717-5p, miR-483-3p, and miR-940 also targeted BNDF. Skogstrand et al. found that lower BDNF levels in serum correlate with ASD risks [68, 69]. miR-339-5p has been found to be a drug target for Alzheimer's disease, and is low expressed in mature neurons and related to axon guidance [70, 71]. In our study, we found that miR-339-5p targets 42 genes associated with ASD risks. Among these genes, the association of DIP2A and ASD risks has been validated by our team [72]; moreover, Dip2a knockout mice exhibit autism-like behaviors, including excessive repetitive behavior and social novelty defects [73]. Notably, autism-like behaviors and germline transmission in MECP2 transgenic monkeys corroborate association between miR-339-5p and MECP2 [74]. In addition, miR-202-5p, miR-483-3p, and miR-940 also targets MECP2. For these reasons, miRNAs encoded within CNVs may be implicated in ASD etiology.
For enrichment analysis, we found that genes were enriched in synapse, synapse-related signal regulation, neurotransmitter activity, neurotransmitter transport, and neurotransmitter binding. Mutations in synapse-related or neurotransmitter-related genes are associated with ASD risks in multiple unbiased, targeted sequencing, and neuropathological studies, evidencing that dysregulation in synaptogenesis and neurotransmission is implicated in the pathogenesis of ASD [7578]. We corroborated that ASD pathogenesis was related to dopaminergic synapse, mTOR signaling pathway, insulin signaling pathway, and cholinergic synapse [7982]. Dopamine affects ASD-related-brain regions (basal ganglia, cortex, and amygdala) via dopaminergic synapse [79]. mTOR is involved in integrating signaling from ASD synaptic and regulatory proteins, such as SHANK3, FMRP and the glutamate receptors mGluR1/5 [63, 83]. Dysfunction in mTOR signaling affords one of mechanisms of ASD — an imbalance between excitatory and inhibitory currents [80]. Insulin signaling pathway is feasible for development of autism [81]. Neurochemical abnormalities in the cholinergic system are involved in ASD pathogenesis, highlighting the potential for intervention-targeted cholinergic synapses [82].
Functional network analysis of the 219 CNVs-encoded-miRNAs-targeted genes elicited that a novel regulating mechanism of these CNVs-encoded miRNAs consisted of synapse-related functions (glutamatergic synapse, dopaminergic synapse, serotonergic synapse, and GABAergic synapse), axon guidance, ion channel (ion-gated channel and cation channel complex), and Wnt signaling pathway. Synaptic function and Wnt signaling pathway are affected by mutations in diverse ASD-related genes, and altered Wnt pathway signaling may confer an involvement in ASD pathogenesis [78]. Interestingly, dysfunction of axon-guidance signaling is integral to the microstructural abnormalities of the brain in people with ASD [84]. Notably, the involvement of ion channel-related genes has been found in ASD etiology [85]. Mutations in ion channel genes contribute to low-to-moderate susceptibility of ASD [85].
Both GO and Pathway enrichment analyses showed that CNVs-relating genes and CNVs-encoded-miRNAs-targeted genes mapped synapse-related functions. Additionally, CNVs-relating genes also enriched in mTOR signaling pathway and insulin signaling pathway. In contrast, CNVs-encoded-miRNAs-targeted genes enriched in axon guidance, ion channel, and Wnt signaling pathway. These results documented the high complexity and heterogeneity of ASD, suggesting that different genomic alteration on same chromosomal location may confer distinct but complementary effects on the brain of people with ASD.
Our study had some limitations: (1) the sample size in our study may confer limited statistical power to discover significant findings; (2) genetic and environmental factors contribute to ASD risk; however, environmental factors were not available for us; and (3) de novo or inherited of the CNVs were not be classified because of the lack of data from parents.
Despite these limitations above, our study also had some strength. Firstly, we found eight de novo CNVs (duplications at 1p36.31, 1p36.33, 1q42.13, 11p15.5, and 16q21; deletions at 2p23.1-p22.3, 10q23.2-q23.31, and 14q11.2) and 12 validated CNVs (duplication at 5p15.33, 5p15.33-p15.2, 7p22.3, 7p22.3-p22.2, 10q26.2-q26.3, 11q25, 16p13.3, 22q13.31-q13.33, and Xq12-q13.1; deletion at 7q22.1-q22.2, 12p12.1-p11.23, and 15q13.3), further documenting that ASD is of high genetic heterogeneity after comparing our results and previous findings (Supplementary Table 12). Secondly, we identified 20 genes (eight protein-coding genes supported by SFARI and AutismKB and 12 microRNAs-coding genes that refine understanding of involving approach of ASD-susceptible-genes in etiology) are implicated in ASD risks. Thirdly, we performed GO and KEGG pathway analyses of CNVs-relating genes and CNVs-encoded-miRNAs-targeted genes, providing a new dimension to revealing ASD etiology.

Conclusions

In summary, we identified that 22 kinds of CNVs (six deletions and 16 duplications), eight protein-coding genes, and 12 miRNAs-coding genes are implicated in ASD risks, conferring perception to further reveal ASD etiology.

Acknowledgements

The authors would like to thank the subjects in this study.

Declarations

All the methods were performed in accordance with Declaration of Helsinki. This study was approved by the ethics committee of Jilin University. The parents or guardians of each individual with ASD signed the written informed consent forms.
Not applicable.

Competing interests

The authors declare no 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
Literatur
1.
Zurück zum Zitat WHO. Autism spectrum disorders and other developmental disorders: From raising awareness to building capacity. Geneva: World Health Organization; 2013. WHO. Autism spectrum disorders and other developmental disorders: From raising awareness to building capacity. Geneva: World Health Organization; 2013.
2.
Zurück zum Zitat Sahin M, Sur M. Genes, circuits, and precision therapies for autism and related neurodevelopmental disorders. Science. 2015;350:6263. Sahin M, Sur M. Genes, circuits, and precision therapies for autism and related neurodevelopmental disorders. Science. 2015;350:6263.
3.
Zurück zum Zitat Kim JY, Son MJ, Son CY, Radua J, Eisenhut M, Gressier F, et al. Environmental risk factors and biomarkers for autism spectrum disorder: an umbrella review of the evidence. Lancet Psychiatry. 2019;6:590–600.CrossRef Kim JY, Son MJ, Son CY, Radua J, Eisenhut M, Gressier F, et al. Environmental risk factors and biomarkers for autism spectrum disorder: an umbrella review of the evidence. Lancet Psychiatry. 2019;6:590–600.CrossRef
4.
Zurück zum Zitat Xu Q, Liu YY, Wang X, Tan GH, Li HP, Hulbert SW, et al. Autism-associated CHD8 deficiency impairs axon development and migration of cortical neurons. Mol Autism. 2018;9:65.CrossRef Xu Q, Liu YY, Wang X, Tan GH, Li HP, Hulbert SW, et al. Autism-associated CHD8 deficiency impairs axon development and migration of cortical neurons. Mol Autism. 2018;9:65.CrossRef
5.
Zurück zum Zitat Peñagarikano O, Abrahams BS, Herman EI, Winden KD, Gdalyahu A, Dong H, et al. Absence of CNTNAP2 leads to epilepsy, neuronal migration abnormalities, and core autism-related deficits. Cell. 2011;147:235–46.CrossRef Peñagarikano O, Abrahams BS, Herman EI, Winden KD, Gdalyahu A, Dong H, et al. Absence of CNTNAP2 leads to epilepsy, neuronal migration abnormalities, and core autism-related deficits. Cell. 2011;147:235–46.CrossRef
6.
Zurück zum Zitat Li Y, Qiu S, Zhong W, Li Y, Liu Y, Cheng Y, et al. Association Between DCC Polymorphisms and Susceptibility to Autism Spectrum Disorder. J Autism Dev Disord. 2020;50(10):3800–9.CrossRef Li Y, Qiu S, Zhong W, Li Y, Liu Y, Cheng Y, et al. Association Between DCC Polymorphisms and Susceptibility to Autism Spectrum Disorder. J Autism Dev Disord. 2020;50(10):3800–9.CrossRef
7.
Zurück zum Zitat da Silva Montenegro EM, Costa CS, Campos G, Scliar M, de Almeida TF, Zachi EC, et al. Meta-analyses support previous and novel autism candidate genes: outcomes of an unexplored Brazilian cohort. Autism Res. 2020;13:199–206.CrossRef da Silva Montenegro EM, Costa CS, Campos G, Scliar M, de Almeida TF, Zachi EC, et al. Meta-analyses support previous and novel autism candidate genes: outcomes of an unexplored Brazilian cohort. Autism Res. 2020;13:199–206.CrossRef
8.
Zurück zum Zitat Qiu S, Li Y, Bai Y, Shi J, Cui H, Gu Y, et al. SHANK1 polymorphisms and SNP-SNP interactions among SHANK family: a possible cue for recognition to autism spectrum disorder in infant age. Autism Res. 2019;12:375–83.CrossRef Qiu S, Li Y, Bai Y, Shi J, Cui H, Gu Y, et al. SHANK1 polymorphisms and SNP-SNP interactions among SHANK family: a possible cue for recognition to autism spectrum disorder in infant age. Autism Res. 2019;12:375–83.CrossRef
9.
Zurück zum Zitat Bai Y, Qiu S, Li Y, Li Y, Zhong W, Shi M, et al. Genetic association between SHANK2 polymorphisms and susceptibility to autism spectrum disorder. IUBMB Life. 2018;70:763–76.CrossRef Bai Y, Qiu S, Li Y, Li Y, Zhong W, Shi M, et al. Genetic association between SHANK2 polymorphisms and susceptibility to autism spectrum disorder. IUBMB Life. 2018;70:763–76.CrossRef
10.
Zurück zum Zitat Wang J, Gong J, Li L, Chen Y, Liu L, Gu H, et al. Neurexin gene family variants as risk factors for autism spectrum disorder. Autism Res. 2018;11:37–43.CrossRef Wang J, Gong J, Li L, Chen Y, Liu L, Gu H, et al. Neurexin gene family variants as risk factors for autism spectrum disorder. Autism Res. 2018;11:37–43.CrossRef
11.
Zurück zum Zitat Marui T, Funatogawa I, Koishi S, Yamamoto K, Matsumoto H, Hashimoto O, et al. Association between autism and variants in the wingless-type MMTV integration site family member 2 ( WNT2) gene. Int J Neuropsychopharmacol. 2010;13:443–9.CrossRef Marui T, Funatogawa I, Koishi S, Yamamoto K, Matsumoto H, Hashimoto O, et al. Association between autism and variants in the wingless-type MMTV integration site family member 2 ( WNT2) gene. Int J Neuropsychopharmacol. 2010;13:443–9.CrossRef
12.
Zurück zum Zitat Sandin S, Lichtenstein P, Kuja-Halkola R, Larsson H, Hultman CM, Reichenberg A. The familial risk of autism. JAMA. 2014;311:1770–7.CrossRef Sandin S, Lichtenstein P, Kuja-Halkola R, Larsson H, Hultman CM, Reichenberg A. The familial risk of autism. JAMA. 2014;311:1770–7.CrossRef
13.
Zurück zum Zitat Ratto AB, Mesibov GB. Autism spectrum disorders in adolescence and adulthood: long-term outcomes and relevant issues for treatment and research. Sci China Life Sci. 2015;58:1010–5.CrossRef Ratto AB, Mesibov GB. Autism spectrum disorders in adolescence and adulthood: long-term outcomes and relevant issues for treatment and research. Sci China Life Sci. 2015;58:1010–5.CrossRef
14.
Zurück zum Zitat C Yuen RK, Merico D, Bookman M, L Howe J, Thiruvahindrapuram B, et al. Whole genome sequencing resource identifies 18 new candidate genes for autism spectrum disorder. Nat Neurosci. 2017;20:602–11.CrossRef C Yuen RK, Merico D, Bookman M, L Howe J, Thiruvahindrapuram B, et al. Whole genome sequencing resource identifies 18 new candidate genes for autism spectrum disorder. Nat Neurosci. 2017;20:602–11.CrossRef
15.
Zurück zum Zitat Safari MR, Omrani MD, Noroozi R, Sayad A, Sarrafzadeh S, Komaki A, et al. Synaptosome-associated protein 25 (SNAP25) gene association analysis revealed risk variants for ASD, in Iranian population. J Mol Neurosci. 2017;61:305–11.CrossRef Safari MR, Omrani MD, Noroozi R, Sayad A, Sarrafzadeh S, Komaki A, et al. Synaptosome-associated protein 25 (SNAP25) gene association analysis revealed risk variants for ASD, in Iranian population. J Mol Neurosci. 2017;61:305–11.CrossRef
16.
Zurück zum Zitat Murdoch JD, State MW. Recent developments in the genetics of autism spectrum disorders. Curr Opin Genet Dev. 2013;23:310–5.CrossRef Murdoch JD, State MW. Recent developments in the genetics of autism spectrum disorders. Curr Opin Genet Dev. 2013;23:310–5.CrossRef
17.
Zurück zum Zitat Iafrate AJ, Feuk L, Rivera MN, Listewnik ML, Donahoe PK, Qi Y, et al. Detection of large-scale variation in the human genome. Nat Genet. 2004;36:949–51.CrossRef Iafrate AJ, Feuk L, Rivera MN, Listewnik ML, Donahoe PK, Qi Y, et al. Detection of large-scale variation in the human genome. Nat Genet. 2004;36:949–51.CrossRef
18.
Zurück zum Zitat Sebat J, Lakshmi B, Troge J, Alexander J, Young J, Lundin P, et al. Large-scale copy number polymorphism in the human genome. Science. 2004;305:525–8.CrossRef Sebat J, Lakshmi B, Troge J, Alexander J, Young J, Lundin P, et al. Large-scale copy number polymorphism in the human genome. Science. 2004;305:525–8.CrossRef
19.
Zurück zum Zitat Xu B, Roos JL, Levy S, van Rensburg EJ, Gogos JA, Karayiorgou M. Strong association of de novo copy number mutations with sporadic schizophrenia. Nat Genet. 2008;40:880–5.CrossRef Xu B, Roos JL, Levy S, van Rensburg EJ, Gogos JA, Karayiorgou M. Strong association of de novo copy number mutations with sporadic schizophrenia. Nat Genet. 2008;40:880–5.CrossRef
20.
Zurück zum Zitat Sebat J, Lakshmi B, Malhotra D, Troge J, Lese-Martin C, Walsh T, et al. Strong association of de novo copy number mutations with autism. Science. 2007;316:445–9.CrossRef Sebat J, Lakshmi B, Malhotra D, Troge J, Lese-Martin C, Walsh T, et al. Strong association of de novo copy number mutations with autism. Science. 2007;316:445–9.CrossRef
21.
Zurück zum Zitat Glessner JT, Li J, Wang D, March M, Lima L, Desai A, et al. Copy number variation meta-analysis reveals a novel duplication at 9p24 associated with multiple neurodevelopmental disorders. Genome Med. 2017;9:106.CrossRef Glessner JT, Li J, Wang D, March M, Lima L, Desai A, et al. Copy number variation meta-analysis reveals a novel duplication at 9p24 associated with multiple neurodevelopmental disorders. Genome Med. 2017;9:106.CrossRef
22.
Zurück zum Zitat Erikson GA, Deshpande N, Kesavan BG, Torkamani A. SG-ADVISER CNV: copy-number variant annotation and interpretation. Genet Med. 2015;17:714–8.CrossRef Erikson GA, Deshpande N, Kesavan BG, Torkamani A. SG-ADVISER CNV: copy-number variant annotation and interpretation. Genet Med. 2015;17:714–8.CrossRef
23.
Zurück zum Zitat Chen CH, Chen HI, Chien WH, Li LH, Wu YY, Chiu YN, et al. High resolution analysis of rare copy number variants in patients with autism spectrum disorder from Taiwan. Sci Rep. 2017;7:11919.CrossRef Chen CH, Chen HI, Chien WH, Li LH, Wu YY, Chiu YN, et al. High resolution analysis of rare copy number variants in patients with autism spectrum disorder from Taiwan. Sci Rep. 2017;7:11919.CrossRef
24.
Zurück zum Zitat Rosti RO, Sadek AA, Vaux KK, Gleeson JG. The genetic landscape of autism spectrum disorders. Dev Med Child Neurol. 2014;56:12–8.CrossRef Rosti RO, Sadek AA, Vaux KK, Gleeson JG. The genetic landscape of autism spectrum disorders. Dev Med Child Neurol. 2014;56:12–8.CrossRef
25.
Zurück zum Zitat Levy D, Ronemus M, Yamrom B, Lee YH, Leotta A, Kendall J, et al. Rare de novo and transmitted copy-number variation in autistic spectrum disorders. Neuron. 2011;70:886–97.CrossRef Levy D, Ronemus M, Yamrom B, Lee YH, Leotta A, Kendall J, et al. Rare de novo and transmitted copy-number variation in autistic spectrum disorders. Neuron. 2011;70:886–97.CrossRef
26.
Zurück zum Zitat Sanders SJ, Ercan-Sencicek AG, Hus V, Luo R, Murtha MT, Moreno-De-Luca D, et al. Multiple recurrent de novo CNVs, including duplications of the 7q11.23 Williams syndrome region, are strongly associated with autism. Neuron. 2011;70:863–85.CrossRef Sanders SJ, Ercan-Sencicek AG, Hus V, Luo R, Murtha MT, Moreno-De-Luca D, et al. Multiple recurrent de novo CNVs, including duplications of the 7q11.23 Williams syndrome region, are strongly associated with autism. Neuron. 2011;70:863–85.CrossRef
27.
Zurück zum Zitat Liu X, Takumi T. Genomic and genetic aspects of autism spectrum disorder. Biochem Biophys Res Commun. 2014;452:244–53.CrossRef Liu X, Takumi T. Genomic and genetic aspects of autism spectrum disorder. Biochem Biophys Res Commun. 2014;452:244–53.CrossRef
28.
Zurück zum Zitat Pinto D, Pagnamenta AT, Klei L, Anney R, Merico D, Regan R, et al. Functional impact of global rare copy number variation in autism spectrum disorders. Nature. 2010;466:368–72.CrossRef Pinto D, Pagnamenta AT, Klei L, Anney R, Merico D, Regan R, et al. Functional impact of global rare copy number variation in autism spectrum disorders. Nature. 2010;466:368–72.CrossRef
29.
Zurück zum Zitat Yin CL, Chen HI, Li LH, Chien YL, Liao HM, Chou MC, et al. Genome-wide analysis of copy number variations identifies PARK2 as a candidate gene for autism spectrum disorder. Mol Autism. 2016;7:23.CrossRef Yin CL, Chen HI, Li LH, Chien YL, Liao HM, Chou MC, et al. Genome-wide analysis of copy number variations identifies PARK2 as a candidate gene for autism spectrum disorder. Mol Autism. 2016;7:23.CrossRef
30.
Zurück zum Zitat Marrale M, Albanese NN, Cali F, Romano V. Assessing the impact of copy number variants on miRNA genes in autism by Monte Carlo simulation. PLoS ONE. 2014;9:e90947.CrossRef Marrale M, Albanese NN, Cali F, Romano V. Assessing the impact of copy number variants on miRNA genes in autism by Monte Carlo simulation. PLoS ONE. 2014;9:e90947.CrossRef
31.
Zurück zum Zitat Park H, Kim JI, Ju YS, Gokcumen O, Mills RE, Kim S, et al. Discovery of common Asian copy number variants using integrated high-resolution array CGH and massively parallel DNA sequencing. Nat Genet. 2010;42:400–5.CrossRef Park H, Kim JI, Ju YS, Gokcumen O, Mills RE, Kim S, et al. Discovery of common Asian copy number variants using integrated high-resolution array CGH and massively parallel DNA sequencing. Nat Genet. 2010;42:400–5.CrossRef
32.
Zurück zum Zitat American Psychiatric Association. Diagnostic and Statistical Manual of Mental Disorders. 5th ed. Washington DC: American Psychiatric Publishing Inc; 2013.CrossRef American Psychiatric Association. Diagnostic and Statistical Manual of Mental Disorders. 5th ed. Washington DC: American Psychiatric Publishing Inc; 2013.CrossRef
33.
Zurück zum Zitat Qiu S, Li Y, Li Y, Zhong W, Shi M, Zhao Q, et al. Association between SHANK3 polymorphisms and susceptibility to autism spectrum disorder. Gene. 2018;651:100–5.CrossRef Qiu S, Li Y, Li Y, Zhong W, Shi M, Zhao Q, et al. Association between SHANK3 polymorphisms and susceptibility to autism spectrum disorder. Gene. 2018;651:100–5.CrossRef
34.
Zurück zum Zitat Gu Z, Gu L, Eils R, Schlesner M, Brors B. circlize Implements and enhances circular visualization in R. Bioinformatics. 2014;30:2811–2.CrossRef Gu Z, Gu L, Eils R, Schlesner M, Brors B. circlize Implements and enhances circular visualization in R. Bioinformatics. 2014;30:2811–2.CrossRef
35.
Zurück zum Zitat Kearney HM, Thorland EC, Brown KK, Quintero-Rivera F, South ST. American College of Medical Genetics standards and guidelines for interpretation and reporting of postnatal constitutional copy number variants. Genet Med. 2011;13:680–5.CrossRef Kearney HM, Thorland EC, Brown KK, Quintero-Rivera F, South ST. American College of Medical Genetics standards and guidelines for interpretation and reporting of postnatal constitutional copy number variants. Genet Med. 2011;13:680–5.CrossRef
36.
Zurück zum Zitat Vaishnavi V, Manikandan M, Tiwary BK, Munirajan AK. Insights on the functional impact of microRNAs present in autism-associated copy number variants. PLoS ONE. 2013;8:e56781.CrossRef Vaishnavi V, Manikandan M, Tiwary BK, Munirajan AK. Insights on the functional impact of microRNAs present in autism-associated copy number variants. PLoS ONE. 2013;8:e56781.CrossRef
37.
Zurück zum Zitat Yu G, Wang LG, Han Y, He QY. clusterProfiler: an R package for comparing biological themes among gene clusters. OMICS. 2012;16:284–7.CrossRef Yu G, Wang LG, Han Y, He QY. clusterProfiler: an R package for comparing biological themes among gene clusters. OMICS. 2012;16:284–7.CrossRef
38.
Zurück zum Zitat Kanehisa M, Goto S. KEGG: kyoto encyclopedia of genes and genomes. Nucleic Acids Res. 2000;28:27–30.CrossRef Kanehisa M, Goto S. KEGG: kyoto encyclopedia of genes and genomes. Nucleic Acids Res. 2000;28:27–30.CrossRef
39.
Zurück zum Zitat Dweep H, Gretz N. miRWalk2.0: a comprehensive atlas of microRNA-target interactions. Nat Methods. 2015;12:697.CrossRef Dweep H, Gretz N. miRWalk2.0: a comprehensive atlas of microRNA-target interactions. Nat Methods. 2015;12:697.CrossRef
40.
Zurück zum Zitat Abrahams BS, Geschwind DH. Advances in autism genetics: on the threshold of a new neurobiology. Nat Rev Genet. 2008;9:341–55.CrossRef Abrahams BS, Geschwind DH. Advances in autism genetics: on the threshold of a new neurobiology. Nat Rev Genet. 2008;9:341–55.CrossRef
41.
Zurück zum Zitat Kushima I, Aleksic B, Nakatochi M, Shimamura T, Okada T, Uno Y, et al. Comparative analyses of copy-number variation in autism spectrum disorder and schizophrenia reveal etiological overlap and biological insights. Cell Rep. 2018;24:2838–56.CrossRef Kushima I, Aleksic B, Nakatochi M, Shimamura T, Okada T, Uno Y, et al. Comparative analyses of copy-number variation in autism spectrum disorder and schizophrenia reveal etiological overlap and biological insights. Cell Rep. 2018;24:2838–56.CrossRef
42.
Zurück zum Zitat Gillentine MA, Yin J, Bajic A, Zhang P, Cummock S, Kim JJ, et al. Functional consequences of CHRNA7 copy-number alterations in induced pluripotent stem cells and neural progenitor cells. Am J Hum Genet. 2017;101:874–87.CrossRef Gillentine MA, Yin J, Bajic A, Zhang P, Cummock S, Kim JJ, et al. Functional consequences of CHRNA7 copy-number alterations in induced pluripotent stem cells and neural progenitor cells. Am J Hum Genet. 2017;101:874–87.CrossRef
43.
Zurück zum Zitat Bitar T, Hleihel W, Marouillat S, Vonwill S, Vuillaume ML, Soufia M, et al. Identification of rare copy number variations reveals PJA2, APCS, SYNPO, and TAC1 as novel candidate genes in Autism Spectrum Disorders. Mol Genet Genomic Med. 2019;7:e786.CrossRef Bitar T, Hleihel W, Marouillat S, Vonwill S, Vuillaume ML, Soufia M, et al. Identification of rare copy number variations reveals PJA2, APCS, SYNPO, and TAC1 as novel candidate genes in Autism Spectrum Disorders. Mol Genet Genomic Med. 2019;7:e786.CrossRef
44.
Zurück zum Zitat Bremer A, Giacobini M, Eriksson M, Gustavsson P, Nordin V, Fernell E, et al. Copy number variation characteristics in subpopulations of patients with autism spectrum disorders. Am J Med Genet B Neuropsychiatr Genet. 2011;156:115–24.CrossRef Bremer A, Giacobini M, Eriksson M, Gustavsson P, Nordin V, Fernell E, et al. Copy number variation characteristics in subpopulations of patients with autism spectrum disorders. Am J Med Genet B Neuropsychiatr Genet. 2011;156:115–24.CrossRef
45.
Zurück zum Zitat Celestino-Soper PB, Shaw CA, Sanders SJ, Li J, Murtha MT, Ercan-Sencicek AG, et al. Use of array CGH to detect exonic copy number variants throughout the genome in autism families detects a novel deletion in TMLHE. Hum Mol Genet. 2011;20:4360–70.CrossRef Celestino-Soper PB, Shaw CA, Sanders SJ, Li J, Murtha MT, Ercan-Sencicek AG, et al. Use of array CGH to detect exonic copy number variants throughout the genome in autism families detects a novel deletion in TMLHE. Hum Mol Genet. 2011;20:4360–70.CrossRef
46.
Zurück zum Zitat Chen CH, Chen HI, Liao HM, Chen YJ, Fang JS, Lee KF, et al. Clinical and molecular characterization of three genomic rearrangements at chromosome 22q13.3 associated with autism spectrum disorder. Psychiatr Genet. 2017;27:23–33.CrossRef Chen CH, Chen HI, Liao HM, Chen YJ, Fang JS, Lee KF, et al. Clinical and molecular characterization of three genomic rearrangements at chromosome 22q13.3 associated with autism spectrum disorder. Psychiatr Genet. 2017;27:23–33.CrossRef
47.
Zurück zum Zitat Rosenfeld JA, Ballif BC, Torchia BS, Sahoo T, Ravnan JB, Schultz R, et al. Copy number variations associated with autism spectrum disorders contribute to a spectrum of neurodevelopmental disorders. Genet Med. 2010;12:694–702.CrossRef Rosenfeld JA, Ballif BC, Torchia BS, Sahoo T, Ravnan JB, Schultz R, et al. Copy number variations associated with autism spectrum disorders contribute to a spectrum of neurodevelopmental disorders. Genet Med. 2010;12:694–702.CrossRef
48.
Zurück zum Zitat Phelan MC. Deletion 22q13.3 syndrome. Orphanet J Rare Dis. 2008;3:14.CrossRef Phelan MC. Deletion 22q13.3 syndrome. Orphanet J Rare Dis. 2008;3:14.CrossRef
49.
Zurück zum Zitat Phelan K, McDermid HE. The 22q13.3 deletion syndrome (Phelan-McDermid Syndrome). Mol Syndromol. 2012;2:186–201.CrossRef Phelan K, McDermid HE. The 22q13.3 deletion syndrome (Phelan-McDermid Syndrome). Mol Syndromol. 2012;2:186–201.CrossRef
50.
Zurück zum Zitat Soorya L, Kolevzon A, Zweifach J, Lim T, Dobry Y, Schwartz L, et al. Prospective investigation of autism and genotype-phenotype correlations in 22q13 deletion syndrome and SHANK3 deficiency. Mol Autism. 2013;4:18.CrossRef Soorya L, Kolevzon A, Zweifach J, Lim T, Dobry Y, Schwartz L, et al. Prospective investigation of autism and genotype-phenotype correlations in 22q13 deletion syndrome and SHANK3 deficiency. Mol Autism. 2013;4:18.CrossRef
51.
Zurück zum Zitat Sarasua SM, Boccuto L, Sharp JL, Dwivedi A, Chen CF, Rollins JD, et al. Clinical and genomic evaluation of 201 patients with Phelan-McDermid syndrome. Hum Genet. 2014;133:847–59.CrossRef Sarasua SM, Boccuto L, Sharp JL, Dwivedi A, Chen CF, Rollins JD, et al. Clinical and genomic evaluation of 201 patients with Phelan-McDermid syndrome. Hum Genet. 2014;133:847–59.CrossRef
52.
Zurück zum Zitat Lowther C, Costain G, Stavropoulos DJ, Melvin R, Silversides CK, Andrade DM, et al. Delineating the 15q13.3 microdeletion phenotype: a case series and comprehensive review of the literature. Genet Med. 2015;17:149–57.CrossRef Lowther C, Costain G, Stavropoulos DJ, Melvin R, Silversides CK, Andrade DM, et al. Delineating the 15q13.3 microdeletion phenotype: a case series and comprehensive review of the literature. Genet Med. 2015;17:149–57.CrossRef
53.
Zurück zum Zitat Gillentine MA, Schaaf CP. The human clinical phenotypes of altered CHRNA7 copy number. Biochem Pharmacol. 2015;97:352–62.CrossRef Gillentine MA, Schaaf CP. The human clinical phenotypes of altered CHRNA7 copy number. Biochem Pharmacol. 2015;97:352–62.CrossRef
54.
Zurück zum Zitat Stefansson H, Rujescu D, Cichon S, Pietilainen OP, Ingason A, Steinberg S, et al. Large recurrent microdeletions associated with schizophrenia. Nature. 2008;455:232–6.CrossRef Stefansson H, Rujescu D, Cichon S, Pietilainen OP, Ingason A, Steinberg S, et al. Large recurrent microdeletions associated with schizophrenia. Nature. 2008;455:232–6.CrossRef
55.
Zurück zum Zitat Klein M, van Donkelaar M, Verhoef E, Franke B. Imaging genetics in neurodevelopmental psychopathology. Am J Med Genet B Neuropsychiatr Genet. 2017;174:485–537.CrossRef Klein M, van Donkelaar M, Verhoef E, Franke B. Imaging genetics in neurodevelopmental psychopathology. Am J Med Genet B Neuropsychiatr Genet. 2017;174:485–537.CrossRef
56.
Zurück zum Zitat Eisenegger C, Knoch D, Ebstein RP, Gianotti LR, Sandor PS, Fehr E. Dopamine receptor D4 polymorphism predicts the effect of L-DOPA on gambling behavior. Biol Psychiatry. 2010;67:702–6.CrossRef Eisenegger C, Knoch D, Ebstein RP, Gianotti LR, Sandor PS, Fehr E. Dopamine receptor D4 polymorphism predicts the effect of L-DOPA on gambling behavior. Biol Psychiatry. 2010;67:702–6.CrossRef
57.
Zurück zum Zitat Gadow KD, Devincent CJ, Olvet DM, Pisarevskaya V, Hatchwell E. Association of DRD4 polymorphism with severity of oppositional defiant disorder, separation anxiety disorder and repetitive behaviors in children with autism spectrum disorder. Eur J Neurosci. 2010;32:1058–65.CrossRef Gadow KD, Devincent CJ, Olvet DM, Pisarevskaya V, Hatchwell E. Association of DRD4 polymorphism with severity of oppositional defiant disorder, separation anxiety disorder and repetitive behaviors in children with autism spectrum disorder. Eur J Neurosci. 2010;32:1058–65.CrossRef
58.
Zurück zum Zitat Sener EF, Taheri S, Sahin MC, Bayramov KK, Marasli MK, Zararsiz G, et al. Altered global mRNA expressions of pain and aggression related genes in the blood of children with autism spectrum disorders. J Mol Neurosci. 2019;67:89–96.CrossRef Sener EF, Taheri S, Sahin MC, Bayramov KK, Marasli MK, Zararsiz G, et al. Altered global mRNA expressions of pain and aggression related genes in the blood of children with autism spectrum disorders. J Mol Neurosci. 2019;67:89–96.CrossRef
59.
Zurück zum Zitat Emanuele E, Boso M, Cassola F, Broglia D, Bonoldi I, Mancini L, et al. Increased dopamine DRD4 receptor mRNA expression in lymphocytes of musicians and autistic individuals: bridging the music-autism connection. Neuro Endocrinol Lett. 2010;31:122–5. Emanuele E, Boso M, Cassola F, Broglia D, Bonoldi I, Mancini L, et al. Increased dopamine DRD4 receptor mRNA expression in lymphocytes of musicians and autistic individuals: bridging the music-autism connection. Neuro Endocrinol Lett. 2010;31:122–5.
60.
Zurück zum Zitat Herault J, Perrot A, Barthelemy C, Buchler M, Cherpi C, Leboyer M, et al. Possible association of c-Harvey-Ras-1 (HRAS-1) marker with autism. Psychiatry Res. 1993;46:261–7.CrossRef Herault J, Perrot A, Barthelemy C, Buchler M, Cherpi C, Leboyer M, et al. Possible association of c-Harvey-Ras-1 (HRAS-1) marker with autism. Psychiatry Res. 1993;46:261–7.CrossRef
61.
Zurück zum Zitat Herault J, Petit E, Martineau J, Perrot A, Lenoir P, Cherpi C, et al. Autism and genetics: clinical approach and association study with two markers of HRAS gene. Am J Med Genet. 1995;60:276–81.CrossRef Herault J, Petit E, Martineau J, Perrot A, Lenoir P, Cherpi C, et al. Autism and genetics: clinical approach and association study with two markers of HRAS gene. Am J Med Genet. 1995;60:276–81.CrossRef
62.
Zurück zum Zitat Bowton E, Saunders C, Reddy IA, Campbell NG, Hamilton PJ, Henry LK, et al. SLC6A3 coding variant Ala559Val found in two autism probands alters dopamine transporter function and trafficking. Transl Psychiatry. 2014;4:e464.CrossRef Bowton E, Saunders C, Reddy IA, Campbell NG, Hamilton PJ, Henry LK, et al. SLC6A3 coding variant Ala559Val found in two autism probands alters dopamine transporter function and trafficking. Transl Psychiatry. 2014;4:e464.CrossRef
63.
Zurück zum Zitat Bourgeron T. A synaptic trek to autism. Curr Opin Neurobiol. 2009;19:231–4.CrossRef Bourgeron T. A synaptic trek to autism. Curr Opin Neurobiol. 2009;19:231–4.CrossRef
64.
Zurück zum Zitat Frazier TW. Autism Spectrum Disorder Associated with Germline Heterozygous PTEN Mutations. Cold Spring Harb Perspect Med. 2019;9:a037002.CrossRef Frazier TW. Autism Spectrum Disorder Associated with Germline Heterozygous PTEN Mutations. Cold Spring Harb Perspect Med. 2019;9:a037002.CrossRef
65.
Zurück zum Zitat Marcinkowska M, Szymanski M, Krzyzosiak WJ, Kozlowski P. Copy number variation of microRNA genes in the human genome. BMC Genomics. 2011;12:183.CrossRef Marcinkowska M, Szymanski M, Krzyzosiak WJ, Kozlowski P. Copy number variation of microRNA genes in the human genome. BMC Genomics. 2011;12:183.CrossRef
66.
Zurück zum Zitat Hsu R, Schofield CM, Dela Cruz CG, Jones-Davis DM, Blelloch R, Ullian EM. Loss of microRNAs in pyramidal neurons leads to specific changes in inhibitory synaptic transmission in the prefrontal cortex. Mol Cell Neurosci. 2012;50:283–92.CrossRef Hsu R, Schofield CM, Dela Cruz CG, Jones-Davis DM, Blelloch R, Ullian EM. Loss of microRNAs in pyramidal neurons leads to specific changes in inhibitory synaptic transmission in the prefrontal cortex. Mol Cell Neurosci. 2012;50:283–92.CrossRef
67.
Zurück zum Zitat Xin C, Xia J, Liu Y, Zhang Y. MicroRNA-202-3p Targets Brain-Derived Neurotrophic Factor and Is Involved in Depression-Like Behaviors. Neuropsychiatr Dis Treat. 2020;16:1073–83. Xin C, Xia J, Liu Y, Zhang Y. MicroRNA-202-3p Targets Brain-Derived Neurotrophic Factor and Is Involved in Depression-Like Behaviors. Neuropsychiatr Dis Treat. 2020;16:1073–83.
68.
Zurück zum Zitat Skogstrand K, Hagen CM, Borbye-Lorenzen N, Christiansen M, Bybjerg-Grauholm J, Bækvad-Hansen M, et al. Reduced neonatal brain-derived neurotrophic factor is associated with autism spectrum disorders. Transl Psychiatry. 2019;9:252.CrossRef Skogstrand K, Hagen CM, Borbye-Lorenzen N, Christiansen M, Bybjerg-Grauholm J, Bækvad-Hansen M, et al. Reduced neonatal brain-derived neurotrophic factor is associated with autism spectrum disorders. Transl Psychiatry. 2019;9:252.CrossRef
69.
Zurück zum Zitat Katoh-Semba R, Wakako R, Komori T, Shigemi H, Miyazaki N, Ito H, et al. Age-related changes in BDNF protein levels in human serum: differences between autism cases and normal controls. Int J Dev Neurosci. 2007;25:367–72.CrossRef Katoh-Semba R, Wakako R, Komori T, Shigemi H, Miyazaki N, Ito H, et al. Age-related changes in BDNF protein levels in human serum: differences between autism cases and normal controls. Int J Dev Neurosci. 2007;25:367–72.CrossRef
70.
Zurück zum Zitat Long JM, Ray B, Lahiri DK. MicroRNA-339-5p down-regulates protein expression of β-site amyloid precursor protein-cleaving enzyme 1 (BACE1) in human primary brain cultures and is reduced in brain tissue specimens of Alzheimer disease subjects. J Biol Chem. 2014;289:5184–98.CrossRef Long JM, Ray B, Lahiri DK. MicroRNA-339-5p down-regulates protein expression of β-site amyloid precursor protein-cleaving enzyme 1 (BACE1) in human primary brain cultures and is reduced in brain tissue specimens of Alzheimer disease subjects. J Biol Chem. 2014;289:5184–98.CrossRef
71.
Zurück zum Zitat Liu DZ, Ander BP, Tian Y, Stamova B, Jickling GC, Davis RR, et al. Integrated analysis of mRNA and microRNA expression in mature neurons, neural progenitor cells and neuroblastoma cells. Gene. 2012;495:120–7.CrossRef Liu DZ, Ander BP, Tian Y, Stamova B, Jickling GC, Davis RR, et al. Integrated analysis of mRNA and microRNA expression in mature neurons, neural progenitor cells and neuroblastoma cells. Gene. 2012;495:120–7.CrossRef
72.
Zurück zum Zitat Li Y, Qiu S, Zhong W, Shi J, Cui H, Li Y, et al. rs1007893 and rs2070435 in DIP2A are associated with visual-reaction-behavior phenotype in Chinese Han population with autism spectrum disorder. Research in Autism Spectrum Disorders. 2020;70:101475.CrossRef Li Y, Qiu S, Zhong W, Shi J, Cui H, Li Y, et al. rs1007893 and rs2070435 in DIP2A are associated with visual-reaction-behavior phenotype in Chinese Han population with autism spectrum disorder. Research in Autism Spectrum Disorders. 2020;70:101475.CrossRef
73.
Zurück zum Zitat Ma J, Zhang LQ, He ZX, He XX, Wang YJ, Jian YL, et al. Autism candidate gene DIP2A regulates spine morphogenesis via acetylation of cortactin. PLoS Biol. 2019;17:e3000461.CrossRef Ma J, Zhang LQ, He ZX, He XX, Wang YJ, Jian YL, et al. Autism candidate gene DIP2A regulates spine morphogenesis via acetylation of cortactin. PLoS Biol. 2019;17:e3000461.CrossRef
74.
Zurück zum Zitat Liu Z, Li X, Zhang JT, Cai YJ, Cheng TL, Cheng C, et al. Autism-like behaviours and germline transmission in transgenic monkeys overexpressing MeCP2. Nature. 2016;530:98–102.CrossRef Liu Z, Li X, Zhang JT, Cai YJ, Cheng TL, Cheng C, et al. Autism-like behaviours and germline transmission in transgenic monkeys overexpressing MeCP2. Nature. 2016;530:98–102.CrossRef
75.
Zurück zum Zitat de la Torre-Ubieta L, Won H, Stein JL, Geschwind DH. Advancing the understanding of autism disease mechanisms through genetics. Nat Med. 2016;22:345–61.CrossRef de la Torre-Ubieta L, Won H, Stein JL, Geschwind DH. Advancing the understanding of autism disease mechanisms through genetics. Nat Med. 2016;22:345–61.CrossRef
76.
Zurück zum Zitat Bourgeron T. From the genetic architecture to synaptic plasticity in autism spectrum disorder. Nat Rev Neurosci. 2015;16:551–63.CrossRef Bourgeron T. From the genetic architecture to synaptic plasticity in autism spectrum disorder. Nat Rev Neurosci. 2015;16:551–63.CrossRef
77.
Zurück zum Zitat Pinto D, Delaby E, Merico D, Barbosa M, Merikangas A, Klei L, et al. Convergence of genes and cellular pathways dysregulated in autism spectrum disorders. Am J Hum Genet. 2014;94:677–94.CrossRef Pinto D, Delaby E, Merico D, Barbosa M, Merikangas A, Klei L, et al. Convergence of genes and cellular pathways dysregulated in autism spectrum disorders. Am J Hum Genet. 2014;94:677–94.CrossRef
78.
Zurück zum Zitat Quesnel-Vallieres M, Weatheritt RJ, Cordes SP, Blencowe BJ. Autism spectrum disorder: insights into convergent mechanisms from transcriptomics. Nat Rev Genet. 2019;20:51–63.CrossRef Quesnel-Vallieres M, Weatheritt RJ, Cordes SP, Blencowe BJ. Autism spectrum disorder: insights into convergent mechanisms from transcriptomics. Nat Rev Genet. 2019;20:51–63.CrossRef
79.
Zurück zum Zitat Kosillo P, Bateup HS. Dopaminergic dysregulation in syndromic autism spectrum disorders: insights from genetic mouse models. Front Neural Circuits. 2021;15:700968.CrossRef Kosillo P, Bateup HS. Dopaminergic dysregulation in syndromic autism spectrum disorders: insights from genetic mouse models. Front Neural Circuits. 2021;15:700968.CrossRef
80.
Zurück zum Zitat Tang G, Gudsnuk K, Kuo SH, Cotrina ML, Rosoklija G, Sosunov A, et al. Loss of mTOR-dependent macroautophagy causes autistic-like synaptic pruning deficits. Neuron. 2014;83:1131–43.CrossRef Tang G, Gudsnuk K, Kuo SH, Cotrina ML, Rosoklija G, Sosunov A, et al. Loss of mTOR-dependent macroautophagy causes autistic-like synaptic pruning deficits. Neuron. 2014;83:1131–43.CrossRef
81.
Zurück zum Zitat Stern M. Insulin signaling and autism. Front Endocrinol (Lausanne). 2011;2:54.CrossRef Stern M. Insulin signaling and autism. Front Endocrinol (Lausanne). 2011;2:54.CrossRef
82.
Zurück zum Zitat Perry EK, Lee ML, Martin-Ruiz CM, Court JA, Volsen SG, Merrit J, et al. Cholinergic activity in autism: abnormalities in the cerebral cortex and basal forebrain. Am J Psychiatry. 2001;158:1058–66.CrossRef Perry EK, Lee ML, Martin-Ruiz CM, Court JA, Volsen SG, Merrit J, et al. Cholinergic activity in autism: abnormalities in the cerebral cortex and basal forebrain. Am J Psychiatry. 2001;158:1058–66.CrossRef
83.
Zurück zum Zitat Peça J, Feng G. Cellular and synaptic network defects in autism. Curr Opin Neurobiol. 2012;22:866–72.CrossRef Peça J, Feng G. Cellular and synaptic network defects in autism. Curr Opin Neurobiol. 2012;22:866–72.CrossRef
84.
Zurück zum Zitat Suda S, Iwata K, Shimmura C, Kameno Y, Anitha A, Thanseem I, et al. Decreased expression of axon-guidance receptors in the anterior cingulate cortex in autism. Mol Autism. 2011;2:14.CrossRef Suda S, Iwata K, Shimmura C, Kameno Y, Anitha A, Thanseem I, et al. Decreased expression of axon-guidance receptors in the anterior cingulate cortex in autism. Mol Autism. 2011;2:14.CrossRef
85.
Zurück zum Zitat Lee J, Ha S, Ahn J, Lee ST, Choi JR, Cheon KA. The role of ion channel-related genes in autism spectrum disorder: a study using next-generation sequencing. Front Genet. 2021;12:595934.CrossRef Lee J, Ha S, Ahn J, Lee ST, Choi JR, Cheon KA. The role of ion channel-related genes in autism spectrum disorder: a study using next-generation sequencing. Front Genet. 2021;12:595934.CrossRef
Metadaten
Titel
Nexus between genome-wide copy number variations and autism spectrum disorder in Northeast Han Chinese population
verfasst von
Shuang Qiu
Yingjia Qiu
Yong Li
Xiaojuan Zhu
Yunkai Liu
Yichun Qiao
Yi Cheng
Yawen Liu
Publikationsdatum
01.12.2023
Verlag
BioMed Central
Erschienen in
BMC Psychiatry / Ausgabe 1/2023
Elektronische ISSN: 1471-244X
DOI
https://doi.org/10.1186/s12888-023-04565-7

Weitere Artikel der Ausgabe 1/2023

BMC Psychiatry 1/2023 Zur Ausgabe

„Übersichtlicher Wegweiser“: Lauterbachs umstrittener Klinik-Atlas ist online

17.05.2024 Klinik aktuell Nachrichten

Sie sei „ethisch geboten“, meint Gesundheitsminister Karl Lauterbach: mehr Transparenz über die Qualität von Klinikbehandlungen. Um sie abzubilden, lässt er gegen den Widerstand vieler Länder einen virtuellen Klinik-Atlas freischalten.

ADHS-Medikation erhöht das kardiovaskuläre Risiko

16.05.2024 Herzinsuffizienz Nachrichten

Erwachsene, die Medikamente gegen das Aufmerksamkeitsdefizit-Hyperaktivitätssyndrom einnehmen, laufen offenbar erhöhte Gefahr, an Herzschwäche zu erkranken oder einen Schlaganfall zu erleiden. Es scheint eine Dosis-Wirkungs-Beziehung zu bestehen.

Klinikreform soll zehntausende Menschenleben retten

15.05.2024 Klinik aktuell Nachrichten

Gesundheitsminister Lauterbach hat die vom Bundeskabinett beschlossene Klinikreform verteidigt. Kritik an den Plänen kommt vom Marburger Bund. Und in den Ländern wird über den Gang zum Vermittlungsausschuss spekuliert.

Typ-2-Diabetes und Depression folgen oft aufeinander

14.05.2024 Typ-2-Diabetes Nachrichten

Menschen mit Typ-2-Diabetes sind überdurchschnittlich gefährdet, in den nächsten Jahren auch noch eine Depression zu entwickeln – und umgekehrt. Besonders ausgeprägt ist die Wechselbeziehung laut GKV-Daten bei jüngeren Erwachsenen.