Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

Identification of candidate gene FAM183A and novel pathogenic variants in known genes: High genetic heterogeneity for autosomal recessive intellectual disability

  • Megan McSherry ,

    Contributed equally to this work with: Megan McSherry, Katherine E. Masih

    Roles Conceptualization, Data curation, Formal analysis, Investigation, Visualization, Writing – original draft, Writing – review & editing

    Affiliation John P. Hussmann Institute for Human Genomics, Miller School of Medicine, University of Miami, Miami, FL, United States of America

  • Katherine E. Masih ,

    Contributed equally to this work with: Megan McSherry, Katherine E. Masih

    Roles Conceptualization, Data curation, Formal analysis, Investigation, Visualization, Writing – original draft, Writing – review & editing

    Affiliation John P. Hussmann Institute for Human Genomics, Miller School of Medicine, University of Miami, Miami, FL, United States of America

  • Nursel H. Elcioglu,

    Roles Conceptualization, Data curation, Investigation, Resources, Writing – review & editing

    Affiliations Department of Pediatric Genetics, Marmara University Medical School, Istanbul, Turkey, Eastern Mediterranean University Medical School, Cyprus, Mersin 10, Turkey

  • Pelin Celik,

    Roles Data curation, Investigation, Writing – review & editing

    Affiliation Division of Developmental Pediatrics, Department of Pediatrics, Ankara University School of Medicine, Ankara, Turkey

  • Ozge Balci,

    Roles Data curation, Investigation, Writing – review & editing

    Affiliation Division of Developmental Pediatrics, Department of Pediatrics, Ankara University School of Medicine, Ankara, Turkey

  • Filiz Basak Cengiz,

    Roles Methodology, Software, Validation

    Affiliation John P. Hussmann Institute for Human Genomics, Miller School of Medicine, University of Miami, Miami, FL, United States of America

  • Daniella Nunez,

    Roles Data curation, Methodology

    Affiliation John P. Hussmann Institute for Human Genomics, Miller School of Medicine, University of Miami, Miami, FL, United States of America

  • Claire J. Sineni,

    Roles Data curation, Writing – review & editing

    Affiliation John P. Hussmann Institute for Human Genomics, Miller School of Medicine, University of Miami, Miami, FL, United States of America

  • Serhat Seyhan,

    Roles Data curation, Investigation, Writing – review & editing

    Affiliation John P. Hussmann Institute for Human Genomics, Miller School of Medicine, University of Miami, Miami, FL, United States of America

  • Defne Kocaoglu,

    Roles Investigation, Writing – review & editing

    Affiliation Department of Pediatric Genetics, Marmara University Medical School, Istanbul, Turkey

  • Shengru Guo,

    Roles Formal analysis, Software

    Affiliation John P. Hussmann Institute for Human Genomics, Miller School of Medicine, University of Miami, Miami, FL, United States of America

  • Duygu Duman,

    Roles Investigation, Resources

    Affiliation Division of Genetics, Department of Pediatrics, Ankara University School of Medicine, Ankara, Turkey

  • Guney Bademci,

    Roles Data curation, Formal analysis, Investigation, Software, Supervision, Visualization, Writing – review & editing

    Affiliation John P. Hussmann Institute for Human Genomics, Miller School of Medicine, University of Miami, Miami, FL, United States of America

  • Mustafa Tekin

    Roles Conceptualization, Data curation, Formal analysis, Funding acquisition, Investigation, Methodology, Project administration, Resources, Supervision, Writing – review & editing

    mtekin@med.miami.edu

    Affiliations John P. Hussmann Institute for Human Genomics, Miller School of Medicine, University of Miami, Miami, FL, United States of America, Dr. John T. Macdonald Foundation Department of Human Genetics, Miller School of Medicine, University of Miami, Miami, FL, United States of America

Abstract

The etiology of intellectual disability (ID) is heterogeneous including a variety of genetic and environmental causes. Historically, most research has not focused on autosomal recessive ID (ARID), which is a significant cause of ID, particularly in areas where parental consanguinity is common. Identification of genetic causes allows for precision diagnosis and improved genetic counseling. We performed whole exome sequencing to 21 Turkish families, seven multiplex and 14 simplex, with nonsyndromic ID. Based on the presence of multiple affected siblings born to unaffected parents and/or shared ancestry, we consider all families as ARID. We revealed the underlying causative variants in seven families in MCPH1 (c.427dupA, p.T143Nfs*5), WDR62 (c.3406C>T, p.R1136*), ASPM (c.5219_5225delGAGGATA, p.R1740Tfs*7), RARS (c.1588A>G, p.T530A), CC2D1A (c.811delG, p.A271Pfs*30), TUSC3 (c.793C>T, p.Q265*) and ZNF335 (c.808C>T, p.R270C and c.3715C>A, p.Q1239K) previously linked with ARID. Besides ARID genes, in one family, affected male siblings were hemizygous for PQBP1 (c.459_462delAGAG, p.R153Sfs*41) and in one family the proband was female and heterozygous for X-chromosomal SLC9A6 (c.1631+1G>A) variant. Each of these variants, except for those in MCPH1 and PQBP1, have not been previously published. Additionally in one family, two affected children were homozygous for the c.377G>A (p.W126*) variant in the FAM183A, a gene not previously associated with ARID. No causative variants were found in the remaining 11 families. A wide variety of variants explain half of families with ARID. FAM183A is a promising novel candidate gene for ARID.

Introduction

Intellectual disability (ID) is an early-onset neurodevelopmental disorder affecting 1% of the general population [1]. ID is characterized by a significant impairment in cognitive ability and adaptive behavior affecting memory, language, problem solving, and visual comprehension, which can lead to impairments in activities of daily living such as self-care and interpersonal communication.

Environmental factors, such as teratogens, infections, malnutrition, and neurological trauma as well as genetic conditions can cause ID. The literature supports a strong genetic etiology for ID, with a varying proportion of cases (ranging from 17% to 50%) being attributable to genetic causes [25]. Variants in more than 1,000 genes have been connected to ID [6]. Support for rare de novo variants as a major cause of ID in simplex cases have been reported [7]. Microarray and exome sequencing have demonstrated the importance of de novo copy number variations (CNVs) and single‐nucleotide variations (SNVs) in ID. While research elucidating chromosomal aberrations, CNVs, autosomal dominant, and X-linked variants as causes for ID has been well established, it was not until recently that studies focusing on autosomal recessive forms of ID (ARID) have gained attention [6,8]. ARID either presents as the sole clinical feature (nonsyndromic) or with additional features (syndromic), and it is extremely heterogeneous [9,10]. To date, there are fifty-five genes in the phenotypic series [PS249500] for non-syndromic ARID in the Online Mendelian Inheritance in Man (OMIM) database [6]. ARID appears to be a common form of ID, an unsolved healthcare problem creating an enormous socioeconomic burden on society, especially in the underdeveloped countries where there is a high rate of consanguinity [8,11]. To reveal the causative variants in ARID, we performed Whole Exome Sequencing (WES) in 21 families affected by non-syndromic ID; these families either had multiple affected family members or pedigrees suggestive of consanguinity.

Methods

Ethics statement

This study was approved by the University of Miami Institutional Review (USA), the Marmara University Medical School Ethics Committee (Turkey), and the Ankara University Medical School Ethics Committee (Turkey). A signed informed consent form was obtained from each participant or, in the case of a minor, from the parents.

Subjects

We included families with ID and a pedigree structure suggestive of autosomal recessive inheritance. These were a mix of multiplex families with parental consanguinity (7 families), simplex families with reported parental consanguinity (5 families), or parental origin from a small town (9 families). All families were from Turkey and evaluated at Marmara University Medical School or Ankara University Medical School. All affected children received a thorough physical examination and were evaluated by a pediatrician, geneticist, and neurologist, when available.

Patients with major anomalies, syndrome specific phenotypic features, and specific neurological signs were excluded from the study. Patients with non-specific minor anomalies (e.g., clinodactyly) and neurological signs (e.g., seizures) were not excluded from the classification of non-syndromic ID, as most patients with ID have such findings. DNA was extracted from patient blood samples for genomic analysis.

Whole exome sequencing and bioinformatics analysis

In 6 families, we performed WES in two affected siblings, and in 15 families, only the probands were sequenced. Agilent SureSelect Human All Exon 50 Mb was used for the capture and HiSeq 2000 was used for the sequencing per our previously published protocol [12]. We filtered variants based on minor allele frequency of <0.005 for recessive and <0.0005 for dominant using ExAC (http://exac.broadinstitute.org/; accessed 08/15/2018) and GnomAD (http://gnomad.broadinstitute.org/; accessed 08/15/2018), a genotype quality (GQ) score >35 for the variant quality, and minimum read depth of ≥ 8. Combined Annotation Dependent Depletion (CADD: http://cadd.gs.washington.edu/info), Sorting Intolerant from Tolerant (SIFT: http://sift.jcvi.org/) and Mutationtaster (http://www.mutationtaster.org/) were used for the in silico prediction. ClinVar (https://www.ncbi.nlm.nih.gov/clinvar/) and Human Gene Mutation Database (HGMD: http://www.hgmd.cf.ac.uk/ac/index.php) were used for the mutation databases. Genomic evolutionary rate profiling (GERP: http://mendel.stanford.edu/SidowLab/downloads/gerp/index.html) score was used to determine variant conservation. While autosomal recessive inheritance with both homozygous and compound heterozygous models were chosen during the initial analysis, all inheritance modes were subsequently investigated. Missense, nonsense, splice site, in-frame INDEL and frame-shift INDELs were selected. We searched for variants in genes already implicated in ID. These genes were retrieved from OMIM with query words including “intellectual disability” or “mental retardation” or “microcephaly” or “cognitive impairment” in Clinical Synopsis (S1 Table). We used CoNIFER (Copy Number Inference From Exome Reads) [13] and XHMM (eXome-Hidden Markov Model) [14] to detect Copy Number Variants (CNVs) [15]. For the CNV analysis, we evaluated genes in our list. We filtered out common CNVs using our WES controls that consists of >500 individuals and Database of Genomic Variants (DGV; http://dgv.tcag.ca/dgv/app/home). ACMG 2015 Guidelines were used for the variant interpretation [16]. Sanger sequencing was used for variant confirmation and co-segregation (dx.doi.org/10.17504/protocols.io.tzxep7n) (S2 Table).

For those families without causative variants in known ID genes, we obtained runs of homozygosity (>2 MB) shared by affected members of each family (S3 Table). Only homozygous INDELs, single nucleotide variants, and CNVs mapping to runs of homozygosity were included in the analysis.

Results

On average, 99%, 94% and 89% of the captured regions were covered by 1X, 5X, and 10X reads, respectively. Average read depth was 59X in our cohort.

Known variants associated with intellectual disability

We identified two variants that have previously been reported to cause ID in two genes (Table 1). In family S223, one male proband (II:1) was homozygous for c.427dupA (p.T143Nfs*5) in MCPH1. In family S25, both affected brothers (II:1 and II:2) were found to be hemizygous for c.459_462delAGAG (p.R153Sfs*41) in PQBP1.

thumbnail
Table 1. Summary of the identified variants in this study.

https://doi.org/10.1371/journal.pone.0208324.t001

Novel variants in known genes associated with intellectual disability

Whole exome sequencing of the probands resulted in identification of seven novel variants in seven genes known to cause ID (Table 1 and S4 Table). These variants were associated with varying phenotypes (Table 2). Variant segregation was confirmed with Sanger sequencing (Fig 1). Homozygous variants in CC2D1A and ASPM led to frameshift mutations. Additionally, homozygous nonsense variants in TUSC3 and WDR62 and a heterozygous variant in SLC9A6 occurring at a splice site were identified. Lastly, a homozygous substitution in RARS and two heterozygous variants in ZNF335 were found. These results are described in more detail in Table 1.

thumbnail
Fig 1. The electropherograms of the identified variants and pedigrees of the families in this study.

WT: Wildtype, MUT: Mutant.

https://doi.org/10.1371/journal.pone.0208324.g001

thumbnail
Table 2. Phenotypic features of probands with causative variants.

https://doi.org/10.1371/journal.pone.0208324.t002

Mutated FAM183A as a causative candidate for intellectual disability

Whole exome sequencing revealed a nonsense variant (c.377G>A; p.W126*) in FAM183A, a novel candidate gene for ARID. Two siblings, MR25-II:1 and MR25-II:2, were homozygous for the variant, and in the family, the variant segregated in an autosomal recessive fashion (Fig 1).

Discussion

We have identified causative variants in 10 out of 21 Turkish families with nonsyndromic ARID.

11 families remained unsolved in our cohort (S1 Fig and S5 Table).

Known variants associated with intellectual disability

The two families with previously reported causative variants for ID are phenotypically consistent with the previously described findings. Both brothers (II:1 and II:2) in family S25 show findings indicative of Renpenning syndrome [MIM 309500], an X-linked condition [17]. Zhang et al (2017) discusses the molecular pathogenesis of mutations in PQBP1, pointing toward the promotion of ubiquitin-mediated degradation of fragile X mental retardation protein (FMRP) resulting in synaptic dysfunction [19]. WES, elucidating the presence of a known causative variant, accompanied with phenotypic data confirmed a diagnosis of Microcephaly 1 in proband S223-II:1 [18].

Novel variants in known genes associated with intellectual disability

CC2D1A variants are associated with ARID (MRT3) [MIM 608443]. This disorder is characterized by significantly below average general intellectual functioning associated with impairments in adaptive behavior which manifest during the developmental period. Analysis in Drosophila links CC2D1A, a member of the mammal lethal giant discs (lgd) protein family, to endosomal trafficking of Notch proteins, well known transmembrane receptors that regulate cell fate during development [20]. Basel-Vanagaite et al. (2006) reported homozygosity for a protein truncating mutation in CC2D1A in affected members of 9 consanguineous Israeli-Arab families with nonsyndromic ID [21]. The homozygous deletion at c.811delG (p.A271Pfs*30) of CC2D1A found in family AU10 is the fifth variant associated with ID reported in this gene.

TUSC3 variants are known to cause Mental Retardation, Autosomal Recessive 7 (MRT7) [MIM 611093]. Including the finding reported here, four independent autosomal recessive variants in TUSC3 are known to cause ARID [2224]. The exact molecular pathogenesis is unknown. One study suggests the involvement of N-glycosylation in higher brain functions [24], while another postulates disturbed magnesium levels due to TUSC3 impairment may play a role in the pathogenesis of intellectual disability [22]. This novel nonsense variant at c.793C>T (p.Q265*) in TUSC3 in family MR32 further demonstrates the significance of this gene’s association with ARID.

Seventeen variants in numerous domains of WDR62 have been reported to be associated with Microcephaly 2, primary, autosomal recessive, with or without cortical malformations (MCPH2) [MIM 604317] [2528]. Cellular studies indicate that WDR62 is a crucial protein in enabling spindle poles to position the cytokinetic furrow and prolong neural precursor generation, a process that is uniquely vital to the proper growth of the human cerebral cortex [25]. The attributes of the proband, S228-II:1, described in Table 2, are consistent with this phenotype. This novel homozygous nonsense variant in WDR62, c.3406C>T (p.R1136*) along with the phenotype of the proband further implicate this gene’s involvement in ARID.

Variants in SLC9A6 are associated with Mental retardation, X-linked syndromic (MRXSCH), Christianson type [MIM 300243]. SLC9A6 encodes a monovalent sodium-selective sodium/hydrogen exchanger (NHE) found in the membranes of intracellular organelles such as mitochondria and endosomes; NHEs participate in a wide array of essential cellular processes [29]. MRXSCH is characterized by profound ID, epilepsy, ataxia, and microcephaly. The phenotype of the female proband (S234-II:1), described in Table 2, is consistent with the presentation of MRXSCH. Linkage analysis and DNA sequencing of families with MRXSCH have identified multiple variants in the SLC9A6, the majority of which are truncating and/or splice mutations [3034]. The novel heterozygous splice-site variant, c.1631+1G>A in SLC9A6, adds to the body of literature on pathogenic splice-site variants reported in this gene.

ZNF335 mutations are associated with Microcephaly 10, primary, autosomal recessive (MCPH10) [MIM 615095]. One homozygous variant in ZNF335 (S236-II:1), causing both a missense change and a splice site defect, is linked to this syndrome [35]. This same study demonstrated that ZNF335 deficiency causes disrupted neuronal proliferation and differentiation in vitro and in vivo mouse models [35]. Both discovered variants in our study are predicted to be disease causing and are likely to result in a hypomorphic variant of ZNF335. This, in combination with the gene’s association with MCPH10 and altered neuronal growth in vitro and in vivo, make it likely that these mutations are the cause of this family’s ID.

Variants in ASPM are associated with Microcephaly 5, primary, autosomal recessive (MCPH5) [MIM 608716]. ASPM has been implicated in the determination of human cerebral cortical size via maintenance of a cleavage plane orientation allowing for symmetric, proliferative division of neuroepithelial cells during brain development [36]. Several aberrations in ASPM have been reported in the literature with a clear majority resulting in premature termination [3741]. Similarly to previously reported variants, the novel homozygous deletion c.5219_5225delGAGGATA (p.R1740Tfs*7) found in ASPM (S243-II:1) leads to a premature stop codon, suggesting that this variant is involved in ARID.

Lastly, variants in RARS are associated with Leukodystrophy, hypomyelinating, 9 (HLD9) [MIM 616140]. RARS encodes the cytoplasmic arginyl‐tRNA synthetase, an enzyme essential for RNA translation and a key player in myelination, among the subunits of the multisynthase complex [42]. Thus, mutations in RARS cause a hypomylenating disorder of the central nervous system. Previously identified variants in RARS causing the HLD9 phenotype are compound heterozygous mutations [42]. In contrast to previously identified variants in RARS, the novel variant found in family S244 is a homozygous missense mutation. After exome sequencing and segregation analysis, these mutations are the only variants that segregate with the family’s phenotype. Therefore, we propose that this homozygous missense variant has a role in ARID and is the most likely cause of ID this family.

Mutated FAM183A as a causative candidate for intellectual disability

In family, MR 25, there were two siblings with ID, low set ears, and microcephaly along with other correlating phenotypic features (Table 2, Fig 1). The two affected children are homozygous for a FAM183A variant (c.377G>A; p.W126*). This variant, found in MR25-II:1 and MR25-II:2, is located in the longest shared homozygous genomic region in these siblings (S6 Table). FAM183A is expressed in human brain, including the hippocampus, caudate nucleus, and medulla oblongata [43]. Five heterozygous large deletions including FAM183A are reported in the DECIPHER database, (https://decipher.sanger.ac.uk/; accessed 07/08/2018). Three out of five variants are found in patients with intellectual disability, developmental delay, or microcephaly; however, the variants’ pathogenicities are not known [44].

The pathogenicity models, expression in brain tissue, and validated autosomal recessive segregation of this variant with the phenotype in family MR 25 support a role for this homozygous nonsense variant as contributing to the inheritance of ID. More research, including functional studies, should be done to further support the role of FAM183A in the pathogenesis of ID.

In 10 out of 21 families investigated (47.6%) we identified likely disease-causing DNA variants. Although CNVs are important cause of ID, we did not detect any CNV in our cohort. As expected for affected offspring of healthy consanguineous parents, the clear majority of these were autosomal recessive defects. This yield is comparable to other recent investigations in highly consanguineous populations [7,4550].

Supporting information

S1 Table. List of the genes used for the analysis in our cohort.

The gene list is created by using the command ((((mental retardation[Clinical Synopsis]) OR microcephaly[Clinical Synopsis]) OR intellectual disability[Clinical Synopsis]) OR cognitive impairment[Clinical Synopsis]) AND “prefix pound”[Properties] within the web site https://www.ncbi.nlm.nih.gov/omim (accessed 10/02/2018). After curation of the gene list, we analyzed single nucleotide variants, INDELs and copy number variants in the gene list. Due to limitation of the next generation sequencing method that we used, we did not analyze the repeat expansions.

https://doi.org/10.1371/journal.pone.0208324.s001

(XLSX)

S2 Table. Sequences of the primers used for the Sanger sequencing.

https://doi.org/10.1371/journal.pone.0208324.s002

(XLSX)

S3 Table. Parameters used for detecting homozygous runs by using Enlis software (https://www.enlis.com/).

https://doi.org/10.1371/journal.pone.0208324.s003

(XLSX)

S4 Table. Chromosomal position of the identified variants (hg19).

https://doi.org/10.1371/journal.pone.0208324.s004

(XLSX)

S5 Table. Phenotypic features of the unsolved probands.

https://doi.org/10.1371/journal.pone.0208324.s005

(XLSX)

S6 Table. Autozygous regions shared by MR25- II:1 and II:2 (hg19).

https://doi.org/10.1371/journal.pone.0208324.s006

(XLSX)

S1 Fig. Pedigrees of the unsolved families.

https://doi.org/10.1371/journal.pone.0208324.s007

(TIF)

Acknowledgments

The authors are grateful to the patients and families for their participation.

References

  1. 1. Dhandha MM, Seiffert-Sinha K, Sinha AA. Specific immunoglobulin isotypes correlate with disease activity, morphology, duration and HLA association in Pemphigus vulgaris. Autoimmunity. 2012;45: 516–526. pmid:22779708
  2. 2. Moeschler JB, Shevell M, American Academy of Pediatrics Committee on Genetics. Clinical genetic evaluation of the child with mental retardation or developmental delays. Pediatrics. 2006;117: 2304–16. pmid:16740881
  3. 3. Kaufman L, Ayub M, Vincent JB. The genetic basis of non-syndromic intellectual disability: a review. J Neurodev Disord. 2010;2: 182–209. pmid:21124998
  4. 4. Hunter AG. Outcome of the routine assessment of patients with mental retardation in a genetics clinic. Am J Med Genet. 2000;90: 60–8. pmid:10602119
  5. 5. Shashi V, McConkie-Rosell A, Rosell B, Schoch K, Vellore K, McDonald M, et al. The utility of the traditional medical genetics diagnostic evaluation in the context of next-generation sequencing for undiagnosed genetic disorders. Genet Med. 2014;16: 176–82. pmid:23928913
  6. 6. Online Mendelian Inheritance in Man: An Online Catalog of Human Genes and Genetic Disorders 2018 [Internet]. [cited 16 Aug 2018]. Available: https://www.omim.org/
  7. 7. de Ligt J, Willemsen MH, van Bon BWM, Kleefstra T, Yntema HG, Kroes T, et al. Diagnostic exome sequencing in persons with severe intellectual disability. N Engl J Med. 2012;367: 1921–9. pmid:23033978
  8. 8. Musante L, Ropers HH. Genetics of recessive cognitive disorders. Trends Genet. 2014;30: 32–9. pmid:24176302
  9. 9. Kochinke K, Zweier C, Nijhof B, Fenckova M, Cizek P, Honti F, et al. Systematic Phenomics Analysis Deconvolutes Genes Mutated in Intellectual Disability into Biologically Coherent Modules. Am J Hum Genet. 2016;98: 149–64. pmid:26748517
  10. 10. Vissers LELM, Gilissen C, Veltman JA. Genetic studies in intellectual disability and related disorders. Nat Rev Genet. 2016;17: 9–18. pmid:26503795
  11. 11. Iqbal Z, van Bokhoven H. Identifying Genes Responsible for Intellectual Disability in Consanguineous Families. Hum Hered. 2014;77: 150–160. pmid:25060278
  12. 12. Bademci G, Foster J, Mahdieh N, Bonyadi M, Duman D, Cengiz FB, et al. Comprehensive analysis via exome sequencing uncovers genetic etiology in autosomal recessive nonsyndromic deafness in a large multiethnic cohort. Genet Med. 2016;18: 364–371. pmid:26226137
  13. 13. Krumm N, Sudmant PH, Ko A, O’Roak BJ, Malig M, Coe BP, et al. Copy number variation detection and genotyping from exome sequence data. Genome Res. 2012;22: 1525–32. pmid:22585873
  14. 14. Fromer M, Moran JL, Chambert K, Banks E, Bergen SE, Ruderfer DM, et al. Discovery and statistical genotyping of copy-number variation from whole-exome sequencing depth. Am J Hum Genet. 2012;91: 597–607. pmid:23040492
  15. 15. Bademci G, Diaz-Horta O, Guo S, Duman D, Van Booven D, Foster J, et al. Identification of copy number variants through whole-exome sequencing in autosomal recessive nonsyndromic hearing loss. Genet Test Mol Biomarkers. 2014;18: 658–61. pmid:25062256
  16. 16. 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. pmid:25741868
  17. 17. Kalscheuer VM, Freude K, Musante L, Jensen LR, Yntema HG, Gécz J, et al. Mutations in the polyglutamine binding protein 1 gene cause X-linked mental retardation. Nat Genet. 2003;35: 313–5. pmid:14634649
  18. 18. Trimborn M, Bell SM, Felix C, Rashid Y, Jafri H, Griffiths PD, et al. Mutations in microcephalin cause aberrant regulation of chromosome condensation. Am J Hum Genet. 2004;75: 261–6. pmid:15199523
  19. 19. Zhang X-Y, Qi J, Shen Y-Q, Liu X, Liu A, Zhou Z, et al. Mutations of PQBP1 in Renpenning syndrome promote ubiquitin-mediated degradation of FMRP and cause synaptic dysfunction. Hum Mol Genet. 2017;26: 955–968. pmid:28073926
  20. 20. Jaekel R, Klein T. The Drosophila Notch inhibitor and tumor suppressor gene lethal (2) giant discs encodes a conserved regulator of endosomal trafficking. Dev Cell. 2006;11: 655–69. pmid:17084358
  21. 21. Basel-Vanagaite L, Attia R, Yahav M, Ferland RJ, Anteki L, Walsh CA, et al. The CC2D1A, a member of a new gene family with C2 domains, is involved in autosomal recessive non-syndromic mental retardation. J Med Genet. 2006;43: 203–10. pmid:16033914
  22. 22. Garshasbi M, Hadavi V, Habibi H, Kahrizi K, Kariminejad R, Behjati F, et al. A defect in the TUSC3 gene is associated with autosomal recessive mental retardation. Am J Hum Genet. 2008;82: 1158–64. pmid:18452889
  23. 23. Garshasbi M, Kahrizi K, Hosseini M, Nouri Vahid L, Falah M, Hemmati S, et al. A novel nonsense mutation in TUSC3 is responsible for non-syndromic autosomal recessive mental retardation in a consanguineous Iranian family. Am J Med Genet A. 2011;155A: 1976–80. pmid:21739581
  24. 24. Molinari F, Foulquier F, Tarpey PS, Morelle W, Boissel S, Teague J, et al. Oligosaccharyltransferase-subunit mutations in nonsyndromic mental retardation. Am J Hum Genet. 2008;82: 1150–7. pmid:18455129
  25. 25. Bhat V, Girimaji SC, Mohan G, Arvinda HR, Singhmar P, Duvvari MR, et al. Mutations in WDR62, encoding a centrosomal and nuclear protein, in Indian primary microcephaly families with cortical malformations. Clin Genet. 2011;80: 532–40. pmid:21496009
  26. 26. Bilgüvar K, Oztürk AK, Louvi A, Kwan KY, Choi M, Tatli B, et al. Whole-exome sequencing identifies recessive WDR62 mutations in severe brain malformations. Nature. 2010;467: 207–10. pmid:20729831
  27. 27. Nicholas AK, Khurshid M, Désir J, Carvalho OP, Cox JJ, Thornton G, et al. WDR62 is associated with the spindle pole and is mutated in human microcephaly. Nat Genet. 2010;42: 1010–4. pmid:20890279
  28. 28. Yu TW, Mochida GH, Tischfield DJ, Sgaier SK, Flores-Sarnat L, Sergi CM, et al. Mutations in WDR62, encoding a centrosome-associated protein, cause microcephaly with simplified gyri and abnormal cortical architecture. Nat Genet. 2010;42: 1015–20. pmid:20890278
  29. 29. Numata M, Petrecca K, Lake N, Orlowski J. Identification of a mitochondrial Na+/H+ exchanger. J Biol Chem. 1998;273: 6951–9. pmid:9507001
  30. 30. Garbern JY, Neumann M, Trojanowski JQ, Lee VM-Y, Feldman G, Norris JW, et al. A mutation affecting the sodium/proton exchanger, SLC9A6, causes mental retardation with tau deposition. Brain. 2010;133: 1391–402. pmid:20395263
  31. 31. Gilfillan GD, Selmer KK, Roxrud I, Smith R, Kyllerman M, Eiklid K, et al. SLC9A6 mutations cause X-linked mental retardation, microcephaly, epilepsy, and ataxia, a phenotype mimicking Angelman syndrome. Am J Hum Genet. 2008;82: 1003–10. pmid:18342287
  32. 32. Tarpey PS, Smith R, Pleasance E, Whibley A, Edkins S, Hardy C, et al. A systematic, large-scale resequencing screen of X-chromosome coding exons in mental retardation. Nat Genet. 2009;41: 535–43. pmid:19377476
  33. 33. Pescosolido MF, Stein DM, Schmidt M, El Achkar CM, Sabbagh M, Rogg JM, et al. Genetic and phenotypic diversity of NHE6 mutations in Christianson syndrome. Ann Neurol. 2014;76: 581–93. pmid:25044251
  34. 34. Fichou Y, Bahi-Buisson N, Nectoux J, Chelly J, Héron D, Cuisset L, et al. Mutation in the SLC9A6 gene is not a frequent cause of sporadic Angelman-like syndrome. Eur J Hum Genet. 2009;17: 1378–80. pmid:19471312
  35. 35. Yang YJ, Baltus AE, Mathew RS, Murphy EA, Evrony GD, Gonzalez DM, et al. Microcephaly gene links trithorax and REST/NRSF to control neural stem cell proliferation and differentiation. Cell. 2012;151: 1097–112. pmid:23178126
  36. 36. Fish JL, Kosodo Y, Enard W, Pääbo S, Huttner WB. Aspm specifically maintains symmetric proliferative divisions of neuroepithelial cells. Proc Natl Acad Sci U S A. 2006;103: 10438–10443. pmid:16798874
  37. 37. Nicholas AK, Swanson EA, Cox JJ, Karbani G, Malik S, Springell K, et al. The molecular landscape of ASPM mutations in primary microcephaly. J Med Genet. 2009;46: 249–53. pmid:19028728
  38. 38. Darvish H, Esmaeeli-Nieh S, Monajemi GB, Mohseni M, Ghasemi-Firouzabadi S, Abedini SS, et al. A clinical and molecular genetic study of 112 Iranian families with primary microcephaly. J Med Genet. 2010;47: 823–8. pmid:20978018
  39. 39. Passemard S, Verloes A, Billette de Villemeur T, Boespflug-Tanguy O, Hernandez K, Laurent M, et al. Abnormal spindle-like microcephaly-associated (ASPM) mutations strongly disrupt neocortical structure but spare the hippocampus and long-term memory. Cortex. 2016;74: 158–76. pmid:26691732
  40. 40. Bond J, Roberts E, Mochida GH, Hampshire DJ, Scott S, Askham JM, et al. ASPM is a major determinant of cerebral cortical size. Nat Genet. 2002;32: 316–20. pmid:12355089
  41. 41. Bond J, Scott S, Hampshire DJ, Springell K, Corry P, Abramowicz MJ, et al. Protein-truncating mutations in ASPM cause variable reduction in brain size. Am J Hum Genet. 2003;73: 1170–7. pmid:14574646
  42. 42. Wolf NI, Salomons GS, Rodenburg RJ, Pouwels PJW, Schieving JH, Derks TGJ, et al. Mutations in RARS cause hypomyelination. Ann Neurol. 2014;76: 134–9. pmid:24777941
  43. 43. Abugessaisa I, Shimoji H, Sahin S, Kondo A, Harshbarger J, Lizio M, et al. FANTOM5 transcriptome catalog of cellular states based on Semantic MediaWiki. Database (Oxford). 2016;2016: baw105. pmid:27402679
  44. 44. DatabaseE of genomiC varIation and Phenotype in Humans using Ensembl Resources 2018 [Internet]. Available: https://decipher.sanger.ac.uk
  45. 45. Hu H, Kahrizi K, Musante L, Fattahi Z, Herwig R, Hosseini M, et al. Genetics of intellectual disability in consanguineous families. Mol Psychiatry. 2018; pmid:29302074
  46. 46. Alazami AM, Patel N, Shamseldin HE, Anazi S, Al-Dosari MS, Alzahrani F, et al. Accelerating novel candidate gene discovery in neurogenetic disorders via whole-exome sequencing of prescreened multiplex consanguineous families. Cell Rep. 2015;10: 148–61. pmid:25558065
  47. 47. Karaca E, Harel T, Pehlivan D, Jhangiani SN, Gambin T, Coban Akdemir Z, et al. Genes that Affect Brain Structure and Function Identified by Rare Variant Analyses of Mendelian Neurologic Disease. Neuron. 2015;88: 499–513. pmid:26539891
  48. 48. Najmabadi H, Hu H, Garshasbi M, Zemojtel T, Abedini SS, Chen W, et al. Deep sequencing reveals 50 novel genes for recessive cognitive disorders. Nature. 2011;478: 57–63. pmid:21937992
  49. 49. Riazuddin S, Hussain M, Razzaq A, Iqbal Z, Shahzad M, Polla DL, et al. Exome sequencing of Pakistani consanguineous families identifies 30 novel candidate genes for recessive intellectual disability. Mol Psychiatry. 2017;22: 1604–1614. pmid:27457812
  50. 50. Reuter MS, Tawamie H, Buchert R, Hosny Gebril O, Froukh T, Thiel C, et al. Diagnostic Yield and Novel Candidate Genes by Exome Sequencing in 152 Consanguineous Families With Neurodevelopmental Disorders. JAMA psychiatry. 2017;74: 293–299. pmid:28097321