The American Psychiatric Association (APA) has updated its Privacy Policy and Terms of Use, including with new information specifically addressed to individuals in the European Economic Area. As described in the Privacy Policy and Terms of Use, this website utilizes cookies, including for the purpose of offering an optimal online experience and services tailored to your preferences.

Please read the entire Privacy Policy and Terms of Use. By closing this message, browsing this website, continuing the navigation, or otherwise continuing to use the APA's websites, you confirm that you understand and accept the terms of the Privacy Policy and Terms of Use, including the utilization of cookies.

×

What is the likely probability that a parent with schizophrenia in whom the 16p11.2 microduplication syndrome is found will pass along that CNV to a particular offspring?

  1. 1%, the population base rate of schizophrenia

  2. 10%, the likelihood of parent to child transmission of schizophrenia

  3. 50%, the likelihood of inheriting one of the parent's two chromosomes in meiosis

  4. Depends upon whether an older offspring already has it

“Mr. A” is a 22-year-old man with a slim appearance who resides with his parents and works in their convenience store. For the past 2 years, he has been in a romantic relationship with a woman who lives independently nearby. Mr. A experienced his first psychotic episode at the age of 17, during which he was hospitalized for 6 weeks and treated with 5 mg/day of haloperidol, leading to a significant reduction in his psychotic symptoms. Mr. A was discharged with a diagnosis of psychotic disorder not otherwise specified, on maintenance treatment with haloperidol, and he began outpatient visits with a psychiatrist. During the first year after his discharge from the hospital, Mr. A’s medication regimen was transitioned to 4 mg/day of risperidone because of extrapyramidal side effects on haloperidol, and his diagnosis was revised to schizophrenia. Since then, Mr. A has experienced several periods of increased psychotic symptoms. However, these were well controlled by transient increases of his risperidone dosage.

During outpatient family meetings with Mr. A and his parents, it became apparent that Mr. A’s mother suffered from intermittent nonaffective psychotic episodes, which had never been formally evaluated or treated. The family agreed that Mr. A bore a close resemblance to his mother, both in psychiatric symptoms and in physical appearance. Mr. A’s mother also reported having a younger sister whom she described as “a little odd.” Throughout elementary school, Mr. A’s performance was below average, and he was late in reaching developmental milestones. His elementary school teacher suggested to his parents that he may suffer from attention deficit hyperactivity disorder, but this was never formally evaluated. In secondary school, Mr. A continued to struggle academically and decided to stop attending school beyond the legal requirement at age 16. During the course of his outpatient treatment, neuropsychological testing revealed that Mr. A had substantial impairments in both attention and working memory. His full-scale IQ was determined to be 70, with a verbal IQ of 68 and a performance IQ of 72. Coincidentally, the family read an article in their local newspaper about genetic testing in patients with autism spectrum disorder, which prompted Mr. A to discuss genetic testing with his psychiatrist, who referred him for clinical genetic counseling.

During the physical examination by the clinical geneticist, Mr. A was noted to have dysmorphic facial features, including elongated face with low-set ears, elongated fingers and toes, and a flat nasal bridge. Oral examination revealed a high-arched palate. Genotyping of Mr. A and his parents revealed a maternally inherited 16p11.2 duplication. Brain MRI was notable for cavum septum pellucidum and enlargement of the third and fourth ventricles.

The psychiatric symptoms and dysmorphic features of Mr. A and his mother are consistent with the 16p11.2 microduplication syndrome (OMIM #614671). The genetic counselor discussed with Mr. A and his parents the findings and the implications for future offspring. Both Mr. A and his parents reported that they were comforted to learn that there was an identifiable cause of the familial psychiatric illness. However, Mr. A’s mother also described experiencing some ambivalence about the outcome of the genetic testing, with feelings of guilt about her responsibility in “passing on” the pathogenic copy number variant to her son. His mother’s concerns were discussed over several sessions with a clinical psychologist who is a member of the medical genetic counseling team. Mr. A is now aware that there is a 50% chance for each of his children to inherit the 16p11.2 microduplication. Mr. A and his partner have expressed a desire to have children and were specifically counseled about family planning. Additional family members potentially at risk for carrying the 16p11.2 microduplication by Mendelian inheritance were informed by Mr. A and his mother about the option to contact their clinical genetic counselor to discuss the possibility of having a genetic evaluation for themselves.

Despite extensive efforts and recent major successes (1, 2), the genetic architecture of psychiatric disorders has remained insufficiently understood to recommend genetic testing as part of the routine diagnostic evaluation in clinical psychiatry (3). With recent developments in advanced molecular genetic techniques and a considerable reduction in costs, there has been a dramatic improvement in our understanding of the genetic underpinnings of severe psychiatric disorders (1, 2, 46). The strong heritability of severe psychiatric disorders was initially suggested by high rates of concordance in studies of monozygotic twins as well as in adoption studies, especially in those involving monozygotic twins (7). Additionally, in large epidemiological studies, it has been repeatedly demonstrated that the risk of psychiatric illness is inversely proportional to the genetic distance from an affected relative (8).

Large international consortia have undertaken highly successful efforts to unravel the genetic underpinnings for several of the major psychiatric disorders. For schizophrenia, a genome-wide association study (GWAS) was performed using >36,000 patients and >110,000 controls, which allowed for the identification of 108 loci reaching genome-wide significance (1). Using a polygenic risk score, these loci accounted for an estimated 3.4% of the variance in schizophrenia liability, which is generally considered insufficient for implementation as a routine clinical diagnostic measure (9). The largest effort to date for identifying rare coding variants was performed using roughly 2,500 cases and an equal number of controls, yielding no variants reaching genome-wide significance (4; but see also 10). Multiple genes have been reported to exhibit an increased burden of rare de novo mutations among patients with schizophrenia, which disproportionately involve genes coding for proteins that govern synaptic function (1113). For bipolar disorder, the largest GWAS reported included a total of approximately 12,000 patients and 52,000 controls, in which genome-wide significant associations were observed for CACNA1C and ODZ4 (14). For major depressive disorder, no genome-wide significant loci were identified in a 2013 GWAS mega-analysis comprising 9,200 cases and 9,500 controls in the discovery phase and 6,800 cases and 50,000 controls in the replication phase (15). However, consistent with the widely held assumption that the genomic architecture of many polygenic disorders will yield to analyses involving increasingly large cohort sizes, a recent GWAS of major depression involving a discovery cohort of 75,000 cases and 231,000 controls, and a replication phase with 45,000 cases and 106,000 controls, identified 15 genome-wide significant loci (16).

Genetic studies of autism spectrum disorder (ASD) have yielded notably strong associations with rare monogenic syndromes, including fragile X syndrome, tuberous sclerosis complex, and Angelman syndrome (17, 18). Investigations of idiopathic autism spectrum disorder have converged on de novo mutations as a frequent genetic determinant (19). The most recent large-scale study (2), involving 2,500 pedigrees, found that de novo coding mutations and copy number variants (CNVs) together explain approximately 30% of simplex ASD cases. The CNV results confirmed multiple previously identified susceptibility loci for ASD, including 1q21.1, 3q29, 7q11.23, 16p11.2, 15q11.2–13, and 22q11.2 (20). Notably, de novo point mutations and CNVs were also recently demonstrated as the major cause of severe intellectual disability (21). Moreover, neurodevelopmental abnormalities, congenital heart disease, and extracardiac congenital anomalies appear to cosegregate among patients with de novo mutations (22), together providing further evidence of pathogenic rare genetic variation in syndromic forms of psychiatric illness that involve comorbidity between axis I psychiatric disorders, intellectual disability, congenital abnormalities, and dysmorphic features.

Copy Number Variants

CNVs are a common source of structural genomic variation involving gains (duplications or insertions), losses (deletions), or complex rearrangements of genomic sequence resulting in deviations from the diploid state (23, 24) (Figure 1). On average, each individual is estimated to have >1,000 CNVs, with similar rates of gains and losses, together involving 10 Mb of genomic sequence (25). CNVs can include one or more genes, leading to disruptions of coding regions or alterations of gene dosage, although many CNVs involve exclusively intergenic sequence. A wide variety of CNVs have been shown to be important sources of pathogenic mutations, particularly those of large size (>100 kb), low frequency (<1% in the general population), and containing genes (5, 21, 2527). With the advent of microarray technology, reliable detection of CNVs has been widely implemented for both research and clinical diagnostic use in a standardized and relatively low-cost workflow (25).

FIGURE 1.

FIGURE 1. Schematic of Major CNV Typesa

a Segments of DNA can be deleted, duplicated, or inserted. The CNV duplication shown here is in an adjacent tandem alignment, but duplications can also be located on more distant parts of the same chromosome or on other chromosomes, thereby potentially disrupting other genes.

Rare CNVs have been well demonstrated as enriched in patients with a variety of severe psychiatric disorders (5, 2830). The CNV and Schizophrenia Working Groups of the Psychiatric Genomics Consortium and the Psychosis Endophenotypes International Consortium recently published a collaborative study (5) involving a large cohort of patients with schizophrenia (N=21,094) and unaffected controls (N=20,227). Overall, patients with schizophrenia were found to have a significantly increased burden of CNVs (>10 kb in size with a population frequency <1%). Eight loci reached genome-wide significance for their pathogenicity: deletions at 1q21.1, 2p16.3 (NRXN1), 3q29, 15q13.3, 16p11.2 (distal), and 22q11.2, as well as duplications at 7q11.23 and 16p11.2 (proximal). Remarkably, CNV deletions at the 22q11.2 locus were observed in 64 of 21,094 cases, compared to one of 20,227 controls (p=5.7×10−18; odds ratio=67.7, 95% CI=9.3–492.8). Carriers of these rare pathogenic CNVs have a significantly higher risk of developing psychosis and a range of other neuropsychiatric disorders, including mood and anxiety disorders, attention deficit hyperactivity disorder (ADHD) (31), ASD (32), and Parkinson’s disease (33), as well as a variety of congenital malformations (31, 34, 35).

The eight CNVs found to be significantly associated with schizophrenia are collectively present in 1.4% of patients and together explain 0.85% of the variance of schizophrenia liability (5), while the 108 genome-wide significant loci of the latest schizophrenia GWAS explain 3.4% of the variance (1). Although the relatively low prevalence of highly penetrant CNVs among patients with nonsyndromic schizophrenia currently appears to limit the cost-effectiveness of CNV testing in the standard psychiatric diagnostic workup, with increasingly large data sets and continued reductions in cost, this remains a distinct possibility in the future.

Compared to patients with nonsyndromic forms of psychiatric illness, those with syndromic features have a significantly higher frequency of CNVs. In one of the first studies to examine CNVs in patients with syndromic psychiatric illness, deletion of CNTNAP2 was found to be associated with comorbid schizophrenia and epilepsy (36). A subsequent study described patients with comorbid schizophrenia and epilepsy, in which 4% of the cases had a 15q11-q13 duplication while none were found in controls (37). Similarly, in a study investigating the relative frequency of CNVs in schizophrenia patients with or without intellectual disability, an excess of large (>1Mb) 15q11.2 duplications or deletions was found in patients with schizophrenia and comorbid intellectual disability (38). In an exemplary case, the 15q11.2 duplication carried by a proband was found to cosegregate in his family with schizophrenia and comorbid intellectual disability, hearing impairment, and ophthalmological problems.

The Clinical Genetic Examination and Consultation

Clinical geneticists provide a diagnostic service and counseling for individuals or families with, or at risk for, conditions that may have a discernible genetic etiology. Ideally, a clinical genetics consultation for patients with severe psychiatric disorders is an opportunity to assess etiology, prognosis, and risk for offspring through probability estimates based on empirical genetic findings (8, 39, 40). In certain instances, clinical, molecular, or metabolic diagnostics can provide insight into the genetic cause of the disorder or syndrome (19, 22, 41). Client-centered psychiatric genetic counseling for patients and their families has already been implemented successfully in a few specialized centers, with demonstrable enhancement of patient empowerment and self-efficacy (39). Moreover, prenatal genetic counseling has also been demonstrated to be an important opportunity for discussing psychiatric illness risk, facilitating insight, and increasing etiological understanding in a therapeutic and collaborative manner (40).

When a patient is referred for genetic consultation, the proband, parents, and/or spouse are typically interviewed to learn about the motivation for their visit. In addition to the anamnesis, a detailed family history is obtained with a particular focus on pregnancy, delivery, and early developmental milestones. The clinical geneticist performs a physical examination of the proband, including dysmorphology assessment, in order to evaluate for the presence of syndromic features and/or minor physical anomalies (MPAs) that might be suggestive of genetic disease. MPAs are congenital anatomical defects (e.g., deformities of the head, eyes, ears, mouth, palate, hands, and feet) thought to be indicative of abnormal ectodermal development during the first and/or second trimester (42). Since the CNS is of ectodermal origin, MPAs are generally viewed as potential indicators of abnormal CNS development (43). A large number of studies have documented an increased prevalence of MPAs in patients with psychiatric disorders of a presumed neurodevelopmental etiology, such as schizophrenia and ASD (44).

In the present study, we specifically focused our investigation on patients with a syndromic form of psychiatric illness, defined as an axis I disorder in combination with multiple congenital abnormalities and/or dysmorphic features. A total of 50 patients with syndromic psychiatric illness, comprising two independent prospectively recruited consecutive case series, were screened for CNVs. In total, we identified pathogenic or likely pathogenic CNVs in 24.0% of patients (95% CI=12.2–35.8), with analogous findings in each independent cohort.

Method

Cohorts

Pilgrim Psychiatric Center cohort.

Medical ethical approval was obtained from the institutional review board of Pilgrim Psychiatric Center in Brentwood, N.Y., and the Mt. Sinai School of Medicine in New York City. Patients, none of whom had previously undergone any genetic testing, were evaluated between February 2000 and June 2001. Informed consent was obtained from all patients. Patients with schizophrenia or schizoaffective disorder were diagnosed according to DSM-IV-TR criteria using the Comprehensive Assessment of Symptoms and History (45). Intelligence testing was performed using the Wechsler Adult Intelligence Scale, 3rd ed. (WAIS-III) with the relevant classification terminology proposed by Groth-Marnat (46): average (full-scale IQ, 90–109), low average (full-scale IQ, 80–89), well below average (full-scale IQ, 70–79), and lower extreme (full-scale IQ, ≤69). Medical chart review, family history, dysmorphology assessment, and physical examination were performed by a qualified psychiatrist (J.I.F., S.M.).

Erasmus University Medical Center cohort.

Medical ethical approval was obtained by the institutional review board of the Erasmus University Medical Center in Rotterdam, the Netherlands. Patients were referred by their treating psychiatrist to the Department of Clinical Genetics for diagnostic evaluations between January 2012 and July 2013. All patients were diagnosed according to DSM-IV-TR using the Structured Clinical Interview for DSM-IV Axis I Disorders. Medical chart review, family history, dysmorphology assessment, and physical examination were performed by a qualified medical geneticist (A.J.A.K.). The parents of the patients were invited to provide DNA samples, but participation was declined in all but one case.

Presentation and History

For the purposes of this study, we operationally defined a “syndromic” presentation as a DSM-IV-TR axis I psychiatric disorder in combination with at least two dysmorphic features and/or congenital abnormalities involving the head, hair, face, chin, eyes, ears, nose, mouth, lips, teeth, neck, thorax, abdomen, genitalia, skin, extremities, stature, or spine.

Family history of psychiatric illness was obtained by constructing a three-generation pedigree for each proband and systematically interviewing the proband and available family members regarding the psychiatric history of each person represented on the pedigree.

Genotyping

DNA was extracted from venous whole blood. Genotyping of the Pilgrim Psychiatric Center cohort was performed using the Affymetrix GeneChip Human Mapping 250K Nsp Assay, with the Affymetrix GeneChip Command Console (Affymetrix, Santa Clara, Calif.) for quality control analysis and variant calls. Genotyping of the Erasmus University Medical Center cohort was performed using the Illumina HumanCytoSNP-12v2.1 microarray, with the Illumina iScan Control and GenomeStudio software program, version 2.1, as well as the Nexus Copy Number Discovery software program, version 5.0 (BioDiscovery, Hawthorne, Calif.). Deletions were considered if they were supported by more than five sequential probes and larger than 150 kb. Duplications were considered if they were supported by more than seven sequential probes and larger than 200 kb. The results were filtered to remove common CNVs (population frequency >1%) present in the UCSC Genome Browser (hg18 build), the Database of Genomic Variants, and in-house databases. Copy-neutral regions of homozygosity were considered if they were larger than 5 Mb. Fragile X Syndrome testing was performed in the Erasmus University Medical Center cohort by Southern blot analysis of the FMR1 trinucleotide repeat length.

Pathogenicity Classification

CNV pathogenicity classification was implemented according to the guidelines of the American College of Medical Genetics (47) through consensus between a molecular geneticist and clinical psychologist (C.G.B.) and a medical geneticist (A.J.A.K). In rare instances of discordance regarding variant classification, a consensus decision was made through a collaborative discussion together with a qualified psychiatrist (S.A.K.). Variants were classified as pathogenic if there were at least two published articles in the literature describing the variant as being associated with the proband’s phenotype. All other CNVs were classified as variants of uncertain significance (VUSs), with the specifier “likely pathogenic” assigned when there were at least two published fundamental neurobiological, functional genomic, and/or human genetic studies involving genes contained within a given CNV for which there was evidence for a causal influence on disease-relevant neurodevelopment, neural circuit function, or behavior.

Metabolic Studies

Erasmus University Medical Center patients were screened for metabolic abnormalities in blood plasma and urine. Blood plasma fraction was isolated according to standardized clinical protocols from venous whole blood collected using lithium heparin–coated blood tubes. Metabolic diagnostics included the following: acylcarnitines, amino acids, bile acids, creatine, guanidinoacetate, homocysteine, homogentisic acid, imidazole compounds, methylmalonic acid, mucopolysaccharides, oligosaccharides, organic acids, orotic acids, phenylalanine, tyrosine, phytanic and pristanic acid, purines and pyrimidines, sialic acid, sialotransferrins, sugars and sugar alcohols, sulfatides, tetraglucoside, and very long chain fatty acids.

Results

Pilgrim Psychiatric Center

The basic demographic and clinical characteristics of the sample are summarized in Table 1. Axis I diagnoses were schizophrenia (78.9%) and schizoaffective disorder (21.1%). Nearly all patients (94.4%) had below average IQ, with a majority (77.8%) classified as lower extreme (full-scale IQ ≤69). Pathogenic or likely pathogenic CNVs were found in five of 19 patients (26.3%), and another five patients had VUSs (see the data supplement that accompanies the online edition of this article). The genomic coordinates of the identified CNVs are provided in Table 2, and detailed characteristics of these 10 patients regarding congenital abnormalities, dysmorphic features, intelligence testing, and psychiatric diagnoses are reported in Table 3.

TABLE 1. Characteristics of Patient Cohorts Referred for Genetic Testing and Counseling

Center and Characteristic
Pilgrim Psychiatric Center (N=19)MeanSD
Age (years)38.410.0
N%
Male947.4
Primary psychiatric diagnosis
 Schizophrenia1578.9
 Schizoaffective disorder421.1
WAIS IQ classification (N=18)
 Average (90–109)15.6
 Low average (80–89)00.0
 Well below average (70–79)316.7
 Lower extreme (≤69)1477.8
Erasmus Medical Center (N=31)MeanSD
Age (years)34.611.7
N%
Male2064.5
WAIS IQ classification (N=29)
 Average (90–109)00.0
 Low average (80–89)931.0
 Well below average (70–79)413.8
 Lower extreme (≤69)1655.2
Primary psychiatric diagnosis
 Autism spectrum disorder1032.3
 Schizophrenia619.4
 Psychotic disorder not otherwise specified516.1
 Schizoaffective disorder412.9
 Major depressive disorder13.2
 Dysthymic disorder13.2
 Bipolar disorder13.2
 Attention deficit hyperactivity disorder13.2
 Behavioral disorder not otherwise specified13.2
 Impulse control disorder13.2
Family history of psychiatric illness2374.2

TABLE 1. Characteristics of Patient Cohorts Referred for Genetic Testing and Counseling

Enlarge table

TABLE 2. Genomic Coordinates (hg18) of the Identified CNVs

Center and PatientVariantGenomic Coordinates (hg18)
Pilgrim Psychiatric Center
PPC–2DupChr22:48,114,921–48,458,127
PPC–3DelChr7:145,582,575–145,741,967
PPC–3DupChr8:133,398,572–133,821,537
PPC–3DupChr22:16,615,108–18,594,783
PPC–7DupChr15:58,705,395–58,764,487
PPC–9DupChr6:124,846,876–125,239,117
PPC–9DupChr7:149,198,031–149,883,752
PPC–9DupChr9:132,351,166–132,715,588
PPC–9DupChr10:128,282,537–129,138,470
PPC–10DupChr1:212,247,344–212,557,152
PPC–10DelChr22:17,275,227–19,790,008
PPC–11DupChr16:18,028,700–20,139,336
PPC–12DelChr7:146,360,344–147,928,119
PPC–15DupChr4:4,511,826–4,748,685
PPC–16DupChr6:162,807,280–163,306,684
PPC–18DupChr2:45,303,454–45,813,464
Erasmus University Medical Center
EMC–1DelChr5:37,423,970–37,662,323
EMC–4DupChr7:0–481,295
EMC–9DelChr3:113,618,774–116,995,580
EMC–10DelChr1:144,959,767–146,292,125
EMC–11DupChr11:107,153,898–107,489,611
EMC–11DupChrX:87,540,740–87,757,911
EMC–12DupChrY:0–2,736,035
EMC–12DupChrY:154,587,409–154,913,754
EMC–12DupChrY:0–27,198,031
EMC–14DelChr16:29,528,999–30,171,562
EMC–16DupChr3:836,618–1,438,346
EMC–16DupChr11:50,080,786–50,675,483
EMC–21DupChr5:99,785,428–100,021,429
EMC–30DelChr13:106,234,020–107,960,598
EMC–31DelChr3:2,226,296–2,292,563
EMC–31DupChr3:2,341,695–2,571,325
EMC–31DelChr8:82,798,696–83,515,397

TABLE 2. Genomic Coordinates (hg18) of the Identified CNVs

Enlarge table

TABLE 3. Characteristics of Patients in the Pilgrim Psychiatric Center Sample Who Had Positive Genetic Findings

PatientSexAge (years)Psychiatric DisorderFull-Scale IQPerformance IQVerbal IQEducation LevelSomatic SymptomsFacial and Body CharacteristicsMicroarray ResultsDiagnostic ClassificationaRegions of Homozygosity
PPC-2Male32Schizophrenia75797511th gradeMRI findings, psychogenic polydipsiaElongated face, cleft palate, pectus excavatumdup22q13.33 (343 kb)VUSNone
PPC-3Male52Schizophrenia776879High school graduateHyperlipidemia, sideroblastic anemia, late-onset seizure disorder (at age 59)Elongated face, narrow orbital fissures, hypertelorism, flat nasal bridge, malar flatness, retrognathiadel7q35 (159 kb), dup8q24.22 (423 kb), dup22q11.21 (2 Mb)PathogenicNone
PPC-7Female35Schizoaffective disorder545164Special education (12 years)Seizure disorder, mitral valve prolapse and tricuspid insufficiency, strabismus, migraine headachesNarrow orbital fissures, square nasal root with hypoplastic alae nasi, retrognathiadup15q22 (580 kb)VUS, likely pathogenicNone
PPC-9Female21Schizoaffective disorder595865UnknownPilonidal cystShort staturedup6q22.31 (392 kb), dup7q36.1 (686 kb), dup9q34.11–34.12 (364 kb), dup10q26.2 (856 kb)VUS, likely pathogenicNone
PPC-10Female27Schizophrenia605966Completed secondary school; special educationCleft palate, seizure disorder, velopharyngeal insufficiency, chronic middle ear infections, recurrent episodes of pneumonia, esotropia, aortic insufficiency and tricuspid regurgitationNarrow orbital figures, telecanthus, square nasal root, retrognathia, prominent/bulbous nose, short staturedup1q41 (310 kb), del22q11.2 (2.5 Mb)PathogenicNone
PPC-11Male40Schizophrenia646568Special educationNoneSloping forehead, protruding supraorbital ridges, square nasal root, wide nasal tip, short philtrum, retrognathia, malar flatness, short and broad sternum, cleft palatedup16p12.3 (2.11 Mb)VUSNone
PPC-12Female50Schizophrenia636566Dropped out of secondary schoolSeizure disorder, unilateral exotropia, strabismusNonedel7q35–36.1 (1.5 Mb)PathogenicNone
PPC-15Female42Schizophrenia6169619th gradePsychogenic polydipsia, hyponatremia, seizures secondary to hyponatremia, MRI findingsHigh nasal root protrusion, kyphosisdup4p16.3–16.2 (237 kb)VUSNone
PPC-16Male48SchizophreniaN/AbHigh school graduateMitral valve prolapse with mitral regurgitation, seizures after clozapine treatmentNarrow orbital fissures, retrognathia, protruding supraorbital ridges, pectus excavatumdup6q26 (499 kb)VUSNone
PPC-18Male36Schizoaffective disorder908392High school graduateHypothyroidism, psychogenic polydipsia, hyponatremia, chronic neutropenia, hiatal hernia, obesityHigh nasal root protrusion, flat alae nasi, diminished vermilion of the upper lip, clefted nasal tip, mild midline cleft lower lip, hypoplastic teeth, hypoplastic earlobes, slender tapered fingers, high-arched V-shaped steepled cleft palatedup2p21 (510 kb)VUSNone

aVUS=variant of uncertain significance.

bIQ testing could not be performed.

TABLE 3. Characteristics of Patients in the Pilgrim Psychiatric Center Sample Who Had Positive Genetic Findings

Enlarge table

Case Descriptions of Pilgrim Psychiatric Center Patients With Pathogenic CNVs

Patient PPC-3 was a 52-year-old man diagnosed with schizophrenia with an onset age of 14 years. His medical history was notable for hyperlipidemia and sideroblastic anemia. The patient exhibited dysmorphic features, including an elongated face with narrow orbital fissures, hypertelorism, flat nasal bridge, malar flatness, and retrognathia. He completed secondary school but was never employed and never married. His illness course was chronic and unremitting, with increasingly severe cognitive impairment and regressive behavior. WAIS-III testing at age 52 revealed a verbal IQ of 79, a performance IQ of 68, and a full-scale IQ of 77. CNV screening revealed a 2-Mb duplication of the long arm of chromosome 22 (22q11.21). 22q11.2 microduplications have been associated with both schizophrenia and ASD (OMIM #608363) (48). This variant was classified as a pathogenic variant. The patient was also noted to have a deletion involving CNTNAP2. However, it concerns a 159-kb intronic deletion in CNTNAP2 that does not involve coding sequence and was therefore classified as a VUS. Lastly, we identified a 423-kb duplication on the long arm of chromosome 8 (8q24.22) containing KCNQ3, LRRC6, and TMEM71. KCNQ3 encodes the voltage-gated potassium channel, subfamily Q, member 3. Heterozygous missense mutations in this gene have been associated with seizures of the benign neonatal subtype, type 2 (4951). The protein is mainly expressed in the brain. LRRC6 is involved in autosomal recessive primary ciliary dyskinesia (52, 53). TMEM71 has not been implicated with this phenotype. Based on the gene content, this variant was classified as a VUS, likely pathogenic, especially in relation to the seizure disorder. On the basis of the risk associated with this genetic diagnosis, the patient was evaluated for the possibility of a previously unrecognized and/or late-onset seizure disorder, which was identified and successfully treated.

Patient PPC-10 was a 27-year-old woman diagnosed with a severe, deteriorating course of schizophrenia (onset age unknown). She received special education and completed secondary school, and was never married and never employed. Her medical history included the surgical repair of a congenital cleft palate, velopharyngeal insufficiency with hypernasal voice, chronic middle ear infections, recurrent episodes of pneumonia, short stature, esotropia, aortic insufficiency, and tricuspid regurgitation. The patient had a seizure disorder with an abnormal background EEG showing bifrontal and bitemporal slowing. She exhibited prominent dysmorphic features, including narrow orbital fissures, telecanthus, square nasal root, bulbous/prominent nose, and retrognathia. WAIS-III testing revealed a verbal IQ of 66, a performance IQ of 59, and a full-scale IQ of 60. Microarray screening revealed a 2.6-Mb deletion on the long arm of chromosome 22 (22q11.21), a well-established CNV associated with the 22q11.2 deletion syndrome (OMIM #188400). 22q11.2 microdeletions are currently the most well established genetic risk factor for schizophrenia (5, 30). This variant was classified as pathogenic. In addition, we identified a 310-kb duplication on the long arm of chromosome 1 (1q41) containing PROX1 and SMYD2. These genes have not been previously associated with the phenotype. This variant was classified as a VUS. On the basis of the risk associated with this genetic diagnosis, the patient was screened for the possibility of hypocalcemia, which was identified and successfully treated, resulting in a significant reduction in seizure frequency.

Patient PPC-12 was a 50-year-old woman diagnosed with schizophrenia, the symptoms of which first manifested at age 18. She attended high school until the 10th grade, and was never married and never employed. Her medical history was notable for seizure disorder, unilateral exotropia, and strabismus. WAIS-III testing revealed a verbal IQ of 66, a performance IQ of 65, and a full-scale IQ of 63. She was assessed by the Wide-Range Achievement Test–Revised reading subtest to estimate a premorbid verbal IQ and was found to have a premorbid full-scale IQ of 94, suggesting a trajectory of cognitive decline following the onset of schizophrenia. Her mother and both of her mother’s siblings suffered from severe psychiatric illnesses that also required long-term hospitalization. At age 34, she experienced her only documented seizure, characterized as generalized tonic-clonic. EEG demonstrated slow dysrhythmia in theta with occasional delta waves, predominantly localized to the frontal lobes. Brain MRI demonstrated mild cerebral atrophy. As previously described, microarray screening identified a heterozygous 1.57-Mb deletion at the 7q357q36.1 locus, which includes CUL1, EZH2, PDIA4, and CNTNAP2 (36). CNTNAP2 encodes contactin-associated protein-like 2, which regulates neuronal-glial interactions and neural cell migration (54, 55). Both CNV deletions and exonic mutations of CNTNAP2 have been previously associated with schizophrenia and ASD (OMIM #604569) (56, 57). This variant was classified as pathogenic. Knowledge of the genetic diagnosis promoted increased awareness of clinically significant prognostic risks, including late-onset language regression and cognitive deterioration.

Erasmus University Medical Center

The basic demographic and clinical characteristics of the sample are summarized in Table 1. Axis I disorders were predominantly ASD (32.3%) or a psychotic disorder (48.4%), and a majority of the sample (74.2%) had a positive family history for psychiatric illness. All patients who could be assessed had below-average IQ, and a substantial proportion (55.2%) had a lower-extreme IQ. Pathogenic or likely pathogenic CNVs were found in seven of the 31 patients (22.6%). Another four patients had VUSs (see the online data supplement). The genomic coordinates of the identified CNVs are listed in Table 2. Detailed characteristics of these 11 patients regarding congenital abnormalities, dysmorphic features, intelligence testing, psychiatric diagnoses, and family history are provided in Table 4. Results of metabolic screening tests of blood plasma and urine, as well as fragile X testing, were normal for all patients.

TABLE 4. Characteristics of Patients in the Erasmus University Medical Center Sample Who Had Positive Genetic Findings

PatientSexAge (years)Psychiatric DisorderaIQEducational LevelFamily HistoryaSomatic SymptomsFacial CharacteristicsBody CharacteristicsSNP ArrayDiagnostic ClassificationRegions of HomozygosityMetabolic Screen, Blood PlasmaMetabolic Screen, UrineFragile X Testing
EMC-1Male34Bipolar II disorder76Lower-level secondary educationMother with bipolar disorderDiabetes, hypothyroidism, hypercholesterolemia, cryptorchidism, amblyopia, retractile testesUpslanted palpebral fissures, epicanthus, downturned mouth corners, asymmetric prominent jaw, dental caries, flat philtrum, high skullEdema in the lower legs, panniculusdel5p13.2 (238 kb)VUSNoneNormalNormalNormal
EMC-4Male37PDD-NOS, psychotic disorder NOS, PTSD69Special educationMother and siblings have learning disabilitiesNoneReceding hairline, hypertelorism, broad palpebral fissures, bushy eyebrows with synophrys, prominent nose tip, narrow upper lip, edentate upper jaw, dysplastic ear helicesIncreased lordosis and kyphosis, mild pectus excavatum, ugly scar formation, striae on the upper legs, Beighton score 2/9, normal tonus, slim posture, hypotrophic muscular systemdup7p22.3 (481 kb)VUSNoneNormalNormalNormal
EMC-9Male53Dysthymic disorder55Special educationAlcohol abuse in fatherDiabetes, hearing impairment (AD>AS), varicesElongated face, flattened center of face, ptosis, narrow palpebral fissures, prominent pear-shaped noseHeight 1 SD below the mean, weight 2 SD above the mean, skull circumference 1 SD above the mean, central adipositydel3q13.2q13.31 (3,377 kb)PathogenicNoneNormalSlightly elevated lactate levelsNormal
EMC-10Male22PDD-NOS, intermittent explosive disorder69Special educationA sibling with intellectual disabilityEar infections, back problemsHypoplastic alae nasiNonedel1q21.1 (1,332 kb)PathogenicNoneNormalNormalNormal
EMC-11Male27PDD-NOS, dysthymic disorder85Special educationNo positive family historyNoneReceding hairlineClinodactyly in fifth finger, hemangioma, striae on arms and abdomendup 11q22.3 (336 kb) and dupXq21.31 (217 kb)VUS, likely pathogenicNoneNormalNormalNormal
EMC-12Male23PDD-NOS, ADHD, ADD, substance abuseN/AbSpecial educationBipolar disorder and ASDInguinal hernia requiring surgeryHigh receding hairline, turned-up nose tip, deeply grooved philtrumHyperpigmentation on left shoulderXYYPathogenicNoneNormalNormalNormal
EMC-14Male18PDD-NOS, ADHDN/AbSpecial educationBrother has ADHDChronic obstructive pulmonary disease, migraine, speech and language disorderBroad and prominent eyebrows with synophrys, broad palpebral fissures, full pouching lips, high and flat philtrumHeight 2.5 SDs below the mean, obese, tapering fingers, hyperextension of the proximal interphalangeal jointdel16p11.2 (643 kb)PathogenicNoneNormalNormalNormal
EMC-16Male43Psychotic disorder NOS80cPrimary education onlyNone; parents are consanguineousHypermetropia, syphilisFlattened face, attached earlobes, hairy ears, prominent eyebrowsIncreased thoracic kyphosis, hyperpigmentation, axial hypotonia, height 2.5 SDs below the mean, weight 2.5 SDs above the meandup3p26.3 (602 kb), dup11p11.12 (595 kb)VUS, likely pathogenicMultiple, parents consanguineousHigh alanine and glutamineNormalNormal
EMC-21Male55Autistic disorder, depressive disorder NOS, OCD82Lower professional educationFather has schizophrenia, sibling has psychotic disorderObesityObesity, small palpebral fissures, hypoplastic alae nasi, high palate, long earsWeight 2 SDs above the mean, hernia umbilicalis, panniculus, thoracic kyphosis, long hands and feetdup5q21.1 (236 kb)VUSNoneNormalNormalNormal
EMC-30Female50Psychotic disorder NOS64Special educationOne brother with schizophrenia and one brother with intellectual disabilityNoneLong face, lipoma on the head, exophthalmos, full nose tip, hypotonic in the faceHeight and skull circumference 0.5 SD below the mean, hallux valgus (right>left), increased thoracic kyphosisdel13q33.3 (1.7 Mb)VUSNoneNot performedNormalNormal
EMC-31Female36Psychotic disorder, major depressive disorderdSpecial educationOne brother with intellectual disabilityNoneNoneNonedel3p26.3 (66 kb) and dup3p26.3 (230 kb), del8q21.13 (717 kb)VUS, likely pathogenicNoneNot performedNormalNormal

aADD=attention deficit disorder; ADHD=attention deficit hyperactivity disorder; ASD=autism spectrum disorder; NOS=not otherwise specified; OCD=obsessive-compulsive disorder; PDD=pervasive developmental disorder; PTSD=posttraumatic stress disorder; VUS=variant of uncertain significance.

bIQ testing could not be performed.

cEstimated IQ.

dIQ estimated as lower extreme (≤69).

TABLE 4. Characteristics of Patients in the Erasmus University Medical Center Sample Who Had Positive Genetic Findings

Enlarge table

Case Descriptions of Erasmus University Medical Center Patients With Pathogenic CNVs

Patient EMC-9 was a 53-year-old man with a diagnosis of dysthymic disorder and intellectual disability and a lower-extreme IQ of 55. His social-emotional development was estimated to be that of a 6-year-old. He exhibited an elongated face, ptosis, narrow palpebral fissures, a large, pear-shaped nose, and central obesity. We found a 3.38-Mb deletion on the long arm of chromosome 3 (3q13.2-q13.31) that included 25 genes. Molin et al. (58) and Lowther et al. (59) described several patients with 3q13.31 microdeletion syndrome. The 3q13.31 microdeletion syndrome is associated with developmental delay, muscular hypotonia, hypoplastic male genitalia, characteristic facial features, and obesity (OMIM #615433). This variant was classified as pathogenic. On the basis of the obesity risk associated with this genetic diagnosis, increased attention was given to avoiding medications associated with weight gain and metabolic side effects.

Patient EMC-10 was a 22-year-old man with a diagnosis of pervasive developmental disorder not otherwise specified, intermittent explosive disorder, and intellectual disability (full-scale IQ of 69). His social-emotional development level was estimated as that of a 3-year-old. We identified a 1.33-Mb deletion on the long arm of chromosome 1 containing the critical region of the 1q21.1 deletion syndrome (OMIM #612474), a susceptibility locus associated with developmental delay, hypotonia, microcephaly, cardiac anomalies, hypermobility, seizures, and dysmorphology (60). This variant was classified as pathogenic. On the basis of the risks associated with this genetic diagnosis, a cardiac screening examination was performed and medications known to lower seizure threshold were avoided.

Patient EMC-12 was a 23-year-old man with a diagnosis of pervasive developmental disorder not otherwise specified, ADHD, oppositional defiant disorder, and substance abuse. He exhibited a high receding hairline, a region of hyperpigmentation on the left shoulder, an upturned nose, and a deeply grooved philtrum. Microarray screening revealed a gain for the entire Y chromosome, which was supported by gains in the pseudoautosomal regions located on X and Y. Karyotyping confirmed a single additional Y chromosome (47,XYY). Studies of patients with 47,XYY syndrome have identified an increased risk of ASD and ADHD, as well as a higher burden of externalizing symptoms (61). This variant was classified as pathogenic. Discussion of the genetic diagnosis helped to strengthen the therapeutic alliance with the mental health care team.

Patient EMC-14 was an 18-year-old man with a diagnosis of pervasive developmental disorder not otherwise specified and ADHD, and he was a convicted sex offender. He reported speech and language problems during childhood requiring special education. He exhibited broad and prominent eyebrows with synophrys, long palpebral fissures, full pouching lips, a high and flat philtrum, obesity, tapering fingers, and hyperextension of the proximal interphalangeal joint. His height was 2.5 standard deviations below the mean. Genotyping revealed a 643-kb deletion on the short arm of chromosome 16 (16p11.2) overlapping with the critical region of the 16p11.2 microdeletion syndrome (OMIM #611913) (31, 60, 62). Microdeletions of 16p11.2 confer high susceptibility to developmental delay, craniofacial dysmorphology, and ASD, as well as severe early-onset obesity, congenital cardiac abnormalities, seizures, and intellectual disability. This variant was classified as pathogenic. On the basis of the risks associated with this genetic diagnosis, a cardiac screening examination was performed and medications known to lower seizure threshold, induce weight gain, or promote metabolic syndrome were strictly avoided.

Discussion

We performed comprehensive genetic analyses in two cohorts of patients with a syndromic presentation of psychiatric illness. In 12 patients (24%), we identified a pathogenic or likely pathogenic genetic variant. We found a similar percentage of pathogenic or likely pathogenic CNVs in each independent cohort (Pilgrim Psychiatric Center cohort: 5/19, or 26.3%; Erasmus University Medical Center cohort: 7/31, or 22.6%). Among the identified CNVs, many have been previously established as known genetic risk factors for psychiatric disorders (22q11.2 microdeletion, 16p11.2 microdeletion, XYY syndrome) and developmental delay (3q13.31 microdeletion, 1q21.1 microdeletion). Furthermore, we observed likely pathogenic CNVs involving only a single gene that have been strongly linked to psychiatric illness (CNTN4, CNTN6).

Pathogenic CNVs associated with psychiatric phenotypes have a broad range of penetrance, varying from 2%−33% for bipolar disorder, schizophrenia, or ASD, while the exposed attributable risk for pathogenic de novo CNVs varies between 79% and 87% (63). In particular, among individuals carrying a microdeletion at 22q11.2, the lifetime prevalence of psychotic disorders is 30% (64). Increasingly precise penetrance estimates will greatly facilitate discussions between patients, their families, and their health care providers regarding diagnosis, treatment, and prognosis (25).

Our findings are consistent with a recent study by Stobbe et al. (65), who reported clinical diagnostic findings in 24 consecutively evaluated adult patients with syndromic ASD, of whom 20.8% were found to have pathogenic or likely pathogenic CNVs. Furthermore, our observed rate of pathogenic CNVs (7/50, or 14.0%) is significantly higher than in the general-population schizophrenia cohort reported by Rees et al. (28) (171/6882, or 2.48%) (Fisher’s exact test, p=2.6×10−4). Therefore, clinical genetic testing for CNVs may be particularly relevant in patients with syndromic forms of psychiatric illness.

Clinical genetic testing should be considered in the presence or absence of a significant family history. Although a positive family history suggests the presence of an inherited genetic variant, a negative family history can be indicative of a de novo variant. Notably, however, the absence of an identifiable inherited or de novo genetic variant does not rule out more complex genetic events due to somatic mosaicism that might not be detectable in DNA isolated from peripheral blood (66, 67). The considerable diagnostic importance of de novo mutations has been firmly established for intellectual disability (19, 21, 68). Accordingly, this might also be an important genetic mechanism underlying syndromic forms of psychiatric illness that should be evaluated in future studies. Moreover, future investigations with larger cohorts should be conducted to determine the relative contribution of the severity of intellectual disability, distinct congenital abnormalities, craniofacial dysmorphologies, and seizure disorders to the prior probability of CNVs in patients with psychiatric disorders.

Important clinical benefits have been shown to result from a genetic diagnosis. Multiple studies have reported a preference on the part of patients and their families to obtain an etiological genetic diagnosis, the benefits of which include improved knowledge of their disease and a feeling of empowerment to better advocate for themselves (39, 69). Patients who receive a genetic diagnosis experience a strengthening of the therapeutic alliance with their psychiatrist and mental health care providers (70). In addition, having an etiological genetic diagnosis facilitates access to medical benefits, educational opportunities, and social services for patients and their families (71, 72). Accordingly, formal genetic counseling is a critically important opportunity for patients and their families to understand the risks and opportunities for reproductive planning (39, 40).

Confirmation of a known genetic syndrome can also improve diagnosis, prognosis, treatment, and prevention. The knowledge of a genetic diagnosis provides a unique opportunity to consider future clinical course and prognosis on the basis of published information regarding patients with comparable genetic diagnoses. For example, CNVs involving CNTNAP2 have been reported to increase the risk for adult-onset mutism (73) and epilepsy (36). Furthermore, pathogenic CNVs associated with neuropsychiatric symptoms frequently involve medical comorbidities for which diagnostic screening and preventive treatment are often available and of significant clinical consequence. For example, patients with 22q11.2 microdeletion exhibit high rates of immune deficiency, congenital heart disease, hypocalcemia, seizures (often provoked by episodes of hypocalcemia), scoliosis, obesity, hearing loss (sensorineural and/or conductive), thrombocytopenia, thyroid dysfunction, and renal anomalies (74). Additional examples include 16p11.2 microdeletion, which is associated with obesity, gastrointestinal symptoms (e.g., reflux, constipation, diarrhea), seizures, immune deficiency, scoliosis, and congenital heart disease (75, 76). 1q21.1 microduplication is associated with cardiac abnormalities (in particular tetralogy of Fallot), seizures, and macrocephaly (77).

Regarding treatment, there are important examples of CNV-associated risks of adverse effects or complications of psychopharmacological treatment, as well as emerging therapeutic opportunities. For example, patients with schizophrenia and 22q11.2 microdeletion appear to be at increased risk of adverse events from clozapine, including seizures, movement-related side effects (including myoclonus, tremor, unsteady gait, rigidity, and slurred speech), neutropenia, and myocarditis (78, 79). Clinicians should also be aware of the potential to exacerbate the predilection toward hypocalcemia in patients with 22q11.2 microdeletion through medication (e.g., anticonvulsants) or alcohol consumption, and the corresponding importance of vitamin D and calcium supplementation. Moreover, 16p11.2 microdeletion results in a strong predisposition to obesity, for which there should be concern regarding medications associated with weight gain and metabolic syndrome (75, 80). With the increasing success of psychiatric genetics, neuroscientists are gaining knowledge about the underlying neurobiological mechanisms of CNV-associated neurogenetic syndromes for which clinicians, patients, and their families should be aware of emerging clinical research studies. Notable recent examples of translational drug development studies include vigabatrin for 22q11.2 microdeletion syndrome (81) and oxytocin for CNTNAP2 deletion (82).

Craniofacial abnormalities are known to be associated with alterations in brain development, consistent with the shared neurodevelopmental origin of the brain and the facial skeleton (83). However, the issue is complex, given that there is a generally broad phenotypic spectrum associated with carriership of pathogenic CNVs (8486). In an Icelandic population-based study of more than 100,000 people without a history of schizophrenia or autism, 1% were identified as carriers of pathogenic CNVs. Notably, although none of the people carrying these CNVs met traditional clinical diagnostic criteria for a neurodevelopmental or neuropsychiatric disorder, the majority exhibited a range of functionally significant cognitive and neuropsychological deficits (87).

For both schizophrenia (5) and autism (20, 88), case-control studies have shown a clear excess of CNVs. In contrast, the genome-wide burden of CNVs does not appear to be increased for bipolar disorder or recurrent major depression (8991) (however, there is strong evidence for an association of bipolar disorder and recurrent depression with duplications at 16p11.2 [48, 89]). Notably, large (≥100 kb) CNVs—which we have focused on in the present study—generally appear to have larger effect sizes than small CNVs, but they are also significantly more rare. Accordingly, the contribution of smaller CNVs to phenotypic diversity in the general population remains less well studied, because of the requirement for increasingly large population-based cohort studies with detailed phenotypic data and higher-density genotyping (25). Future studies using high-resolution variant detection by whole genome sequencing are expected to greatly facilitate the identification of smaller pathogenic CNVs and indels.

The majority of the patients in our cohorts had intellectual disability in addition to axis I psychiatric disorders, for which the nature of the causality of the genetic effects remains an open question. Specifically, it remains unclear to what extent a given CNV might exert its effects directly through endogenous neurobiological mechanisms and indirectly through impairments in cognitive reserve (92) or social or emotional functioning. In this regard, it is notable that genome-wide CNV burden appears to be elevated in patients who have a psychiatric disorder and comorbid intellectual disability (2, 93, 94). This finding might therefore suggest that CNVs can exert their influence on psychiatric disease risk indirectly through reduced intellectual capacity (95). Conversely, the pattern of allelic pleiotropy observed for distinct CNVs and rare single-nucleotide variants suggests that the associated risk of psychiatric illness is likely to be independent of intellectual disability (48, 96, 97).

With regard to the classification of CNV pathogenicity, it is important to be aware that there remains a considerable degree of subjectivity when implementing the consensus guidelines (47). Classifications of pathogenic and benign are relatively higher confidence, given the stronger available evidence base required for these specifiers, while likely pathogenic variants and VUSs are dynamically evolving with the increasing availability of population-based genotyping data (e.g., the ExAC browser [98]) and clinical genetic information (e.g., ClinVar [99]), as well as the standardized implementation of genome-wide functional genomic, neuroimaging, and cognitive analyses (e.g., the ENIGMA consortium [100]).

Despite the substantial benefits for patients and their clinicians, genetic testing is not without clinically significant risks. Given that genetic testing currently leads to an etiological diagnosis in only a minority of cases, patients’ expectations should be appropriately tempered. In the absence of identifiable pathogenic CNVs, there remains a distinct possibility that CNVs classified as VUSs and/or other types of genetic variants, such as rare protein-coding mutations or common polygenic risk, may be etiologically significant factors. Moreover, for patients who receive an inconclusive diagnosis (e.g., VUSs), the resulting uncertainty might increase their level of concern (69). Conversely, a confirmed pathogenic finding may induce anxiety regarding future prognosis and later-onset symptoms (70). Importantly, the decision of whether to perform genetic testing for diagnostic or predictive purposes should be made by the patient (or by a legal guardian, for those who are incapable of providing informed consent), and only after a comprehensive discussion with their psychiatrist and genetic counselor to weigh the risks and benefits of testing. Moreover, the outcome of genetic testing for a patient can have significant consequences for the entire family, for whom genetic counseling is strongly recommended to allow them a forum, either individually or as a group, regarding the implications for themselves and their children.

In summary, on the basis of the best available evidence, we propose that CNV screening should be considered for implementation within routine clinical practice for patients with syndromic forms of psychiatric illness. Although formal cost-effectiveness analyses are not yet complete, the current price of comparative genomic hybridization microarray testing is approximately $500–$1500 (101, 102), with a number needed to test of 4.35 based on the combination of our results and those of Stobbe et al. (65) (17 cases with pathogenic or likely pathogenic variants of a total of 74 screened). With the availability of exponentially larger data sets, an increasing proportion of the variants now considered likely pathogenic or VUSs on the basis of insufficient information are likely to be reclassified in the near future with even further improvements in the diagnostic yield for genetic testing of patients with syndromic forms of psychiatric illness.

Summary and Conclusions

Pathogenic or likely pathogenic CNVs are present in a substantial fraction of patients with syndromic forms of psychiatric illness. Currently, the diagnostic yield appears to be highest when genetic testing is implemented in patients with axis I psychiatric disorders in combination with multiple congenital abnormalities and/or dysmorphic features. Based on our case series and the published literature, intellectual disability is also a frequent comorbid symptom. Given that pathogenic variants associated with syndromic disorders are often de novo, genetic testing should be considered even if the family history is negative.

Below we summarize some important implications for clinical management following a genetic diagnosis in patients with syndromic forms of mental illness.

Therapeutic alliance: Psychiatric genetic diagnoses generally lead to a strengthening of the therapeutic alliance, with patients and their families often reporting a resulting feeling of empowerment.

Resources: Having an etiological genetic diagnosis facilitates access to medical benefits, educational opportunities, and social services for patients and their families. When available, genetic diagnosis–focused patient organizations are a valuable source of information, advocacy, and support.

Genetic counseling: Formal genetic counseling is critically important for patients and their families to understand the risks and opportunities for reproductive planning.

Prognosis: Genetic diagnostics provides the opportunity to consider a patient’s future clinical course on the basis of published information regarding patients with comparable CNVs.

Medical comorbidity: Pathogenic CNVs associated with neuropsychiatric symptoms frequently involve medical comorbidities for which diagnostic screening and preventive treatment might be available and of significant clinical consequence.

Standard-of-care treatment: Clinicians should be aware of the CNV-associated risks of pharmacological treatment. Particularly notable are the risks of exacerbating underlying predilections toward obesity, metabolic syndrome, immune dysfunction, endocrine abnormalities, intellectual disability, and seizures.

Emerging therapies: Clinical research and treatment studies of established genetic diagnoses are increasing in frequency and scope. Clinical trial registries, mostly notably ClinicalTrials.gov and EudraCT, as well as genetic diagnosis–focused patient organizations, are a valuable resource for identifying active studies.

C. If the CNV is on one of a parent's two chromosomes, then the likelihood that the CNV chromosome will be placed by meiosis into the sperm or egg that becomes an offspring is 50%. It is 50% for each of the offspring in that family, because each is the independent outcome of a different meiosis. The CNV may or may not lead to a psychiatric illness in a particular individual, and therefore whether or not a particular sibling will become ill is unknown.

Want more? A CME course is available in the APA Learning Center at education.psychiatry.org

From the Department of Psychiatry and the Department of Clinical Genetics, Erasmus University Medical Center (Erasmus MC), Rotterdam, the Netherlands; the Department of Psychiatry and the Center for Precision Neuropsychiatry, Columbia University, New York; New York State Psychiatric Institute, New York; the Departments of Psychiatry, Neuroscience, and Human Genetics, Mount Sinai School of Medicine, New York; the Clinical Neuroscience Center, Pilgrim Psychiatric Center, West Brentwood, N.Y.; the Department of Genetics, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht, the Netherlands; Delta Psychiatric Center, Portugaal, the Netherlands; the Department of Human Genetics, Donders Center for Neuroscience, Radboud University Nijmegen Medical Center, Nijmegen, the Netherlands; and the Department of Clinical Genetics, School for Oncology and Developmental Biology, Maastricht University Medical Center, Maastricht, the Netherlands.
Address correspondence to Dr. Kushner ().

The first four authors contributed equally to this article.

Dr. Bonifati has received research grants from the Erasmus MC, Rotterdam, the Stichting ParkinsonFonds (Netherlands), and ZonMw under the aegis of the EU Joint Programme–Neurodegenerative Disease Research; he has received compensation for serving as Section Editor of Current Neurology and Neuroscience Reports and Associate Editor of Parkinsonism and Related Disorders; he has received honoraria from the International Parkinson and Movement Disorder Society, Elsevier, Springer Science + Business Media, and LSP Life Sciences Fund. Dr. Kushner has received funding from ZonMw Vidi (grant 017.106.384) from the Netherlands Organization for Scientific Research (NWO), the Dutch Technology Foundation (the applied science division of NWO), and the Technology Program of the Ministry of Economic Affairs (Project 12197), as well as by the NeuroBasic PharmaPhenomics consortium and the Erasmus University Medical Center. The other authors report no financial relationships with commercial interests.

References

1 Ripke S, Neale BM, Corvin A, et al.: Biological insights from 108 schizophrenia-associated genetic loci. Nature 2014; 511:421–427Crossref, MedlineGoogle Scholar

2 Iossifov I, O’Roak BJ, Sanders SJ, et al.: The contribution of de novo coding mutations to autism spectrum disorder. Nature 2014; 515:216–221Crossref, MedlineGoogle Scholar

3 International Society of Psychiatric Genetics, Genetics Testing Taskforce: Genetic Testing Statement [Internet]. 2014. http://ispg.net/genetic-testing-statement/Google Scholar

4 Purcell SM, Moran JL, Fromer M, et al.: A polygenic burden of rare disruptive mutations in schizophrenia. Nature 2014; 506:185–190Crossref, MedlineGoogle Scholar

5 Marshall CR, Howrigan DP, Merico D, et al.: Contribution of copy number variants to schizophrenia from a genome-wide study of 41,321 subjects. Nat Genet 2017; 49:27–35Crossref, MedlineGoogle Scholar

6 Smoller JW, Craddock N, Kendler K, et al.: Identification of risk loci with shared effects on five major psychiatric disorders: a genome-wide analysis. Lancet 2013; 381:1371–1379Crossref, MedlineGoogle Scholar

7 Shields J: Monozygotic Twins Brought Up Apart and Brought Together. London, Oxford University Press, 1962Google Scholar

8 Austin JC, Peay HL: Applications and limitations of empiric data in provision of recurrence risks for schizophrenia: a practical review for healthcare professionals providing clinical psychiatric genetics consultations. Clin Genet 2006; 70:177–187Crossref, MedlineGoogle Scholar

9 Dudbridge F: Power and predictive accuracy of polygenic risk scores. PLoS Genet 2013; 9:e1003348Crossref, MedlineGoogle Scholar

10 Kim MJ, Biag J, Fass DM, et al.: Functional analysis of rare variants found in schizophrenia implicates a critical role for GIT1-PAK3 signaling in neuroplasticity. Mol Psychiatry 2017; 22:417–429Crossref, MedlineGoogle Scholar

11 Takata A, Xu B, Ionita-Laza I, et al.: Loss-of-function variants in schizophrenia risk and SETD1A as a candidate susceptibility gene. Neuron 2014; 82:773–780Crossref, MedlineGoogle Scholar

12 Xu B, Ionita-Laza I, Roos JL, et al.: De novo gene mutations highlight patterns of genetic and neural complexity in schizophrenia. Nat Genet 2012; 44:1365–1369Crossref, MedlineGoogle Scholar

13 Fromer M, Pocklington AJ, Kavanagh DH, et al.: De novo mutations in schizophrenia implicate synaptic networks. Nature 2014; 506:179–184Crossref, MedlineGoogle Scholar

14 Psychiatric GWAS Consortium Bipolar Disorder Working Group: Large-scale genome-wide association analysis of bipolar disorder identifies a new susceptibility locus near ODZ4. Nat Genet 2011; 43:977–983Crossref, MedlineGoogle Scholar

15 Ripke S, Wray NR, Lewis CM, et al.: A mega-analysis of genome-wide association studies for major depressive disorder. Mol Psychiatry 2013; 18:497–511Crossref, MedlineGoogle Scholar

16 Hyde CL, Nagle MW, Tian C, et al.: Identification of 15 genetic loci associated with risk of major depression in individuals of European descent. Nat Genet 2016; 48:1031–1036Crossref, MedlineGoogle Scholar

17 Geschwind DH, State MW: Gene hunting in autism spectrum disorder: on the path to precision medicine. Lancet Neurol 2015; 14:1109–1120Crossref, MedlineGoogle Scholar

18 Miles JH: Autism spectrum disorders: a genetics review. Genet Med 2011; 13:278–294Crossref, MedlineGoogle Scholar

19 Veltman JA, Brunner HG: De novo mutations in human genetic disease. Nat Rev Genet 2012; 13:565–575Crossref, MedlineGoogle Scholar

20 Sanders SJ, He X, Willsey AJ, et al.: Insights into autism spectrum disorder genomic architecture and biology from 71 risk loci. Neuron 2015; 87:1215–1233Crossref, MedlineGoogle Scholar

21 Gilissen C, Hehir-Kwa J, Thung D, et al.: Genome sequencing identifies major causes of severe intellectual disability. Nature 2014; 511:344–347Crossref, MedlineGoogle Scholar

22 Homsy J, Zaidi S, Shen Y, et al.: De novo mutations in congenital heart disease with neurodevelopmental and other congenital anomalies. Science 2015; 350:1262–1266Crossref, MedlineGoogle Scholar

23 Iafrate AJ, Feuk L, Rivera MN, et al.: Detection of large-scale variation in the human genome. Nat Genet 2004; 36:949–951Crossref, MedlineGoogle Scholar

24 Sebat J, Lakshmi B, Troge J, et al.: Large-scale copy number polymorphism in the human genome. Science 2004; 305:525–528Crossref, MedlineGoogle Scholar

25 Zarrei M, MacDonald JR, Merico D, et al.: A copy number variation map of the human genome. Nat Rev Genet 2015; 16:172–183Crossref, MedlineGoogle Scholar

26 Vulto-van Silfhout AT, Hehir-Kwa JY, van Bon BWM, et al.: Clinical significance of de novo and inherited copy-number variation. Hum Mutat 2013; 34:1679–1687Crossref, MedlineGoogle Scholar

27 Zhang F, Gu W, Hurles ME, et al.: Copy number variation in human health, disease, and evolution. Annu Rev Genomics Hum Genet 2009; 10:451–481Crossref, MedlineGoogle Scholar

28 Rees E, Walters JTR, Georgieva L, et al.: Analysis of copy number variations at 15 schizophrenia-associated loci. Br J Psychiatry 2014; 204:108–114Crossref, MedlineGoogle Scholar

29 Kirov G, Rees E, Walters JTR, et al.: The penetrance of copy number variations for schizophrenia and developmental delay. Biol Psychiatry 2014; 75:378–385Crossref, MedlineGoogle Scholar

30 Sullivan PF, Daly MJ, O’Donovan M: Genetic architectures of psychiatric disorders: the emerging picture and its implications. Nat Rev Genet 2012; 13:537–551Crossref, MedlineGoogle Scholar

31 Shinawi M, Liu P, Kang S-HL, et al.: Recurrent reciprocal 16p11.2 rearrangements associated with global developmental delay, behavioural problems, dysmorphism, epilepsy, and abnormal head size. J Med Genet 2010; 47:332–341Crossref, MedlineGoogle Scholar

32 Schneider M, Debbané M, Bassett AS, et al.: Psychiatric disorders from childhood to adulthood in 22q11.2 deletion syndrome: results from the International Consortium on Brain and Behavior in 22q11.2 Deletion Syndrome. Am J Psychiatry 2014; 171:627–639LinkGoogle Scholar

33 Mok KY, Sheerin U, Simón-Sánchez J, et al.: Deletions at 22q11.2 in idiopathic Parkinson’s disease: a combined analysis of genome-wide association data. Lancet Neurol 2016; 15:585–596Crossref, MedlineGoogle Scholar

34 Mefford HC, Sharp AJ, Baker C, et al.: Recurrent rearrangements of chromosome 1q21.1 and variable pediatric phenotypes. N Engl J Med 2008; 359:1685–1699Crossref, MedlineGoogle Scholar

35 Karayiorgou M, Simon TJ, Gogos JA: 22q11.2 microdeletions: linking DNA structural variation to brain dysfunction and schizophrenia. Nat Rev Neurosci 2010; 11:402–416Crossref, MedlineGoogle Scholar

36 Friedman JI, Vrijenhoek T, Markx S, et al.: CNTNAP2 gene dosage variation is associated with schizophrenia and epilepsy. Mol Psychiatry 2008; 13:261–266Crossref, MedlineGoogle Scholar

37 Stewart LR, Hall AL, Kang SH, et al.: High frequency of known copy number abnormalities and maternal duplication 15q11-q13 in patients with combined schizophrenia and epilepsy. BMC Med Genet 2011; 12:154Crossref, MedlineGoogle Scholar

38 Derks EM, Ayub M, Chambert K, et al.: A genome wide survey supports the involvement of large copy number variants in schizophrenia with and without intellectual disability. Am J Med Genet B Neuropsychiatr Genet 2013; 162:847–854CrossrefGoogle Scholar

39 Inglis A, Koehn D, McGillivray B, et al.: Evaluating a unique, specialist psychiatric genetic counseling clinic: uptake and impact. Clin Genet 2015; 87:218–224Crossref, MedlineGoogle Scholar

40 Inglis A, Morris E, Austin J: Prenatal genetic counselling for psychiatric disorders. Prenat Diagn 2016; 37:6–13Crossref, MedlineGoogle Scholar

41 Novarino G, El-Fishawy P, Kayserili H, et al.: Mutations in BCKD-kinase lead to a potentially treatable form of autism with epilepsy. Science 2012; 338:394–397Crossref, MedlineGoogle Scholar

42 Murphy KC, Owen MJ: Minor physical anomalies and their relationship to the aetiology of schizophrenia. Br J Psychiatry 1996; 168:139–142Crossref, MedlineGoogle Scholar

43 O’Callaghan E, Larkin C, Kinsella A, et al.: Familial, obstetric, and other clinical correlates of minor physical anomalies in schizophrenia. Am J Psychiatry 1991; 148:479–483LinkGoogle Scholar

44 Schiffman J, Ekstrom M, LaBrie J, et al.: Minor physical anomalies and schizophrenia spectrum disorders: a prospective investigation. Am J Psychiatry 2002; 159:238–243LinkGoogle Scholar

45 Andreasen NC, Flaum M, Arndt S: The Comprehensive Assessment of Symptoms and History (CASH): an instrument for assessing diagnosis and psychopathology. Arch Gen Psychiatry 1992; 49:615–623Crossref, MedlineGoogle Scholar

46 Groth-Marnat G: Handbook of Psychological Assessment, 5th ed. Hoboken, NJ, Wiley, 2009Google Scholar

47 Kearney HM, Thorland EC, Brown KK, et al.: American College of Medical Genetics standards and guidelines for interpretation and reporting of postnatal constitutional copy number variants. Genet Med 2011; 13:680–685Crossref, MedlineGoogle Scholar

48 Malhotra D, Sebat J: CNVs: harbingers of a rare variant revolution in psychiatric genetics. Cell 2012; 148:1223–1241Crossref, MedlineGoogle Scholar

49 Fister P, Soltirovska-Salamon A, Debeljak M, et al.: Benign familial neonatal convulsions caused by mutation in KCNQ3, exon 6: a European case. Eur J Paediatr Neurol 2013; 17:308–310Crossref, MedlineGoogle Scholar

50 Li H, Li N, Shen L, et al.: A novel mutation of KCNQ3 gene in a Chinese family with benign familial neonatal convulsions. Epilepsy Res 2008; 79:1–5Crossref, MedlineGoogle Scholar

51 Hirose S, Zenri F, Akiyoshi H, et al.: A novel mutation of KCNQ3 (c.925T-->C) in a Japanese family with benign familial neonatal convulsions. Ann Neurol 2000; 47:822–826Crossref, MedlineGoogle Scholar

52 Zariwala MA, Gee HY, Kurkowiak M, et al.: ZMYND10 is mutated in primary ciliary dyskinesia and interacts with LRRC6. Am J Hum Genet 2013; 93:336–345Crossref, MedlineGoogle Scholar

53 Kott E, Duquesnoy P, Copin B, et al.: Loss-of-function mutations in LRRC6, a gene essential for proper axonemal assembly of inner and outer dynein arms, cause primary ciliary dyskinesia. Am J Hum Genet 2012; 91:958–964Crossref, MedlineGoogle Scholar

54 Strauss KA, Puffenberger EG, Huentelman MJ, et al.: Recessive symptomatic focal epilepsy and mutant contactin-associated protein-like 2. N Engl J Med 2006; 354:1370–1377Crossref, MedlineGoogle Scholar

55 Peñagarikano O, Abrahams BS, Herman EI, et al.: Absence of CNTNAP2 leads to epilepsy, neuronal migration abnormalities, and core autism-related deficits. Cell 2011; 147:235–246Crossref, MedlineGoogle Scholar

56 Rodenas-Cuadrado P, Pietrafusa N, Francavilla T, et al.: Characterisation of CASPR2 deficiency disorder: a syndrome involving autism, epilepsy, and language impairment. BMC Med Genet 2016; 17:8Crossref, MedlineGoogle Scholar

57 Rodenas-Cuadrado P, Ho J, Vernes SC: Shining a light on CNTNAP2: complex functions to complex disorders. Eur J Hum Genet 2014; 22:171–178Crossref, MedlineGoogle Scholar

58 Molin A-M, Andrieux J, Koolen DA, et al.: A novel microdeletion syndrome at 3q13.31 characterised by developmental delay, postnatal overgrowth, hypoplastic male genitals, and characteristic facial features. J Med Genet 2012; 49:104–109Crossref, MedlineGoogle Scholar

59 Lowther C, Costain G, Melvin R, et al.: Adult expression of a 3q13.31 microdeletion. Mol Cytogenet 2014; 7:23Crossref, MedlineGoogle Scholar

60 Kaminsky EB, Kaul V, Paschall J, et al.: An evidence-based approach to establish the functional and clinical significance of copy number variants in intellectual and developmental disabilities. Genet Med 2011; 13:777–784Crossref, MedlineGoogle Scholar

61 Ross JL, Tartaglia N, Merry DE, et al.: Behavioral phenotypes in males with XYY and possible role of increased NLGN4Y expression in autism features. Genes Brain Behav 2015; 14:137–144Crossref, MedlineGoogle Scholar

62 Weiss LA, Shen Y, Korn JM, et al.: Association between microdeletion and microduplication at 16p11.2 and autism. N Engl J Med 2008; 358:667–675Crossref, MedlineGoogle Scholar

63 Gershon ES, Alliey-Rodriguez N: New ethical issues for genetic counseling in common mental disorders. Am J Psychiatry 2013; 170:968–976LinkGoogle Scholar

64 Green T, Gothelf D, Glaser B, et al.: Psychiatric disorders and intellectual functioning throughout development in velocardiofacial (22q11.2 deletion) syndrome. J Am Acad Child Adolesc Psychiatry 2009; 48:1060–1068Crossref, MedlineGoogle Scholar

65 Stobbe G, Liu Y, Wu R, et al.: Diagnostic yield of array comparative genomic hybridization in adults with autism spectrum disorders. Genet Med 2014; 16:70–77Crossref, MedlineGoogle Scholar

66 Cohen ASA, Wilson SL, Trinh J, et al.: Detecting somatic mosaicism: considerations and clinical implications. Clin Genet 2015; 87:554–562Crossref, MedlineGoogle Scholar

67 de Kock L, Wang YC, Revil T, et al.: High-sensitivity sequencing reveals multi-organ somatic mosaicism causing DICER1 syndrome. J Med Genet 2016; 53:43–52Crossref, MedlineGoogle Scholar

68 de Ligt J, Willemsen MH, van Bon BWM, et al.: Diagnostic exome sequencing in persons with severe intellectual disability. N Engl J Med 2012; 367:1921–1929Crossref, MedlineGoogle Scholar

69 Lawrence RE, Appelbaum PS: Genetic testing in psychiatry: a review of attitudes and beliefs. Psychiatry 2011; 74:315–331Crossref, MedlineGoogle Scholar

70 Costain G, Chow EWC, Ray PN, et al.: Caregiver and adult patient perspectives on the importance of a diagnosis of 22q11.2 deletion syndrome. J Intellect Disabil Res 2012; 56:641–651Crossref, MedlineGoogle Scholar

71 Coulter ME, Miller DT, Harris DJ, et al.: Chromosomal microarray testing influences medical management. Genet Med 2011; 13:770–776Crossref, MedlineGoogle Scholar

72 Riggs ER, Wain KE, Riethmaier D, et al.: Chromosomal microarray impacts clinical management. Clin Genet 2014; 85:147–153Crossref, MedlineGoogle Scholar

73 Stein MB, Yang BZ, Chavira DA, et al.: A common genetic variant in the neurexin superfamily member CNTNAP2 is associated with increased risk for selective mutism and social anxiety-related traits. Biol Psychiatry 2011; 69:825–831Crossref, MedlineGoogle Scholar

74 Fung WLA, Butcher NJ, Costain G, et al.: Practical guidelines for managing adults with 22q11.2 deletion syndrome. Genet Med 2015; 17:599–609Crossref, MedlineGoogle Scholar

75 Zufferey F, Sherr EH, Beckmann ND, et al.: A 600 kb deletion syndrome at 16p11.2 leads to energy imbalance and neuropsychiatric disorders. J Med Genet 2012; 49:660–668Crossref, MedlineGoogle Scholar

76 Miller DT, Chung W, Nasir R, et al.: 16p11.2 recurrent microdeletion. GeneReviews [Internet]), Sept 22, 2009, pp 1–16Google Scholar

77 Dolcetti A, Silversides CK, Marshall CR, et al.: 1q21.1 Microduplication expression in adults. Genet Med 2013; 15:282–289Crossref, MedlineGoogle Scholar

78 Butcher NJ, Fung WLA, Fitzpatrick L, et al.: Response to clozapine in a clinically identifiable subtype of schizophrenia. Br J Psychiatry 2015; 206:484–491Crossref, MedlineGoogle Scholar

79 Boot E, Butcher NJ, van Amelsvoort TAMJ, et al.: Movement disorders and other motor abnormalities in adults with 22q11.2 deletion syndrome. Am J Med Genet A 2015; 167:639–645CrossrefGoogle Scholar

80 Walters RG, Jacquemont S, Valsesia A, et al.: A new highly penetrant form of obesity due to deletions on chromosome 16p11.2. Nature 2010; 463:671–675Crossref, MedlineGoogle Scholar

81 Crabtree GW, Park AJ, Gordon JA, et al.: Cytosolic accumulation of L-proline disrupts GABA-ergic transmission through GAD blockade. Cell Reports 2016; 17:570–582Crossref, MedlineGoogle Scholar

82 Peñagarikano O, Lázaro MT, Lu X-H, et al.: Exogenous and evoked oxytocin restores social behavior in the Cntnap2 mouse model of autism. Sci Transl Med 2015; 7:271ra8Crossref, MedlineGoogle Scholar

83 Marcucio R, Hallgrimsson B, Young NM: Facial morphogenesis: physical and molecular interactions between the brain and the face. Curr Top Dev Biol 2015; 115:299–320Crossref, MedlineGoogle Scholar

84 Girirajan S, Eichler EE: Phenotypic variability and genetic susceptibility to genomic disorders. Hum Mol Genet 2010; 19:176–187Crossref, MedlineGoogle Scholar

85 Rosenfeld JA, Coe BP, Eichler EE, et al.: Estimates of penetrance for recurrent pathogenic copy-number variations. Genet Med 2013; 15:478–481Crossref, MedlineGoogle Scholar

86 Torres F, Barbosa M, Maciel P: Recurrent copy number variations as risk factors for neurodevelopmental disorders: critical overview and analysis of clinical implications. J Med Genet 2015; 53:73–90Crossref, MedlineGoogle Scholar

87 Stefansson H, Meyer-Lindenberg A, Steinberg S, et al.: CNVs conferring risk of autism or schizophrenia affect cognition in controls. Nature 2013; 505:361–366Crossref, MedlineGoogle Scholar

88 Leppa VM, Kravitz SN, Martin CL, et al.: Rare inherited and de novo CNVs reveal complex contributions to ASD risk in multiplex families. Am J Hum Genet 2016; 99:540–554Crossref, MedlineGoogle Scholar

89 Green EK, Rees E, Walters JTR, et al.: Copy number variation in bipolar disorder. Mol Psychiatry 2016; 21:89–93Crossref, MedlineGoogle Scholar

90 Rucker JJH, Tansey KE, Rivera M, et al.: Phenotypic association analyses with copy number variation in recurrent depressive disorder. Biol Psychiatry 2016; 79:329–336Crossref, MedlineGoogle Scholar

91 Grozeva D, Kirov G, Ivanov D, et al.: Rare copy number variants: a point of rarity in genetic risk for bipolar disorder and schizophrenia. Arch Gen Psychiatry 2010; 67:318–327Crossref, MedlineGoogle Scholar

92 Koenen KC, Moffitt TE, Roberts AL, et al.: Childhood IQ and adult mental disorders: a test of the cognitive reserve hypothesis. Am J Psychiatry 2009; 166:50–57LinkGoogle Scholar

93 Wolfe K, Strydom A, Morrogh D, et al.: Chromosomal microarray testing in adults with intellectual disability presenting with comorbid psychiatric disorders. Eur J Hum Genet 2017; 25:66–72CrossrefGoogle Scholar

94 Singh T, Walters JTR, Johnstone M, et al.: The contribution of rare variants to risk of schizophrenia in individuals with and without intellectual disability. Nat Genet 2017; 49:1167–1173Crossref, MedlineGoogle Scholar

95 Pescosolido MF, Gamsiz ED, Nagpal S, et al.: Distribution of disease-associated copy number variants across distinct disorders of cognitive development. J Am Acad Child Adolesc Psychiatry 2013; 52:414–430.e14Crossref, MedlineGoogle Scholar

96 O’Donovan MC, Owen MJ: The implications of the shared genetics of psychiatric disorders. Nat Med 2016; 22:1214–1219Crossref, MedlineGoogle Scholar

97 Moreno-De-Luca D, Moreno-De-Luca A, Cubells JF, et al.: Cross-disorder comparison of four neuropsychiatric CNV loci. Curr Genet Med Rep 2014; 2:1–11CrossrefGoogle Scholar

98 Lek M, Karczewski KJ, Minikel EV, et al.: Analysis of protein-coding genetic variation in 60,706 humans. Nature 2016; 536:285–291Crossref, MedlineGoogle Scholar

99 Landrum MJ, Lee JM, Benson M, et al.: ClinVar: public archive of interpretations of clinically relevant variants. Nucleic Acids Res 2016; 44:D862–D868Crossref, MedlineGoogle Scholar

100 Thompson PM, Stein JL, Medland SE, et al.: The ENIGMA Consortium: large-scale collaborative analyses of neuroimaging and genetic data. Brain Imaging Behav 2014; 8:153–182Crossref, MedlineGoogle Scholar

101 Sagoo GS, Mohammed S, Barton G, et al.: Cost effectiveness of using array-CGH for diagnosing learning disability. Appl Health Econ Health Policy 2015; 13:421–432Crossref, MedlineGoogle Scholar

102 Martin CL, Ledbetter DH: Chromosomal microarray testing for children with unexplained neurodevelopmental disorders. JAMA 2017; 317:2545–2546Crossref, MedlineGoogle Scholar