746
Views
17
CrossRef citations to date
0
Altmetric
Editorial

What insights can we gain from studying early-onset schizophrenia? The neurodevelopmental pathway and beyond

, &
Pages 1243-1247 | Published online: 09 Jan 2014

Schizophrenia is a complex and debilitating brain disorder that commonly presents clinically in late adolescents and early adulthood. However, several converging lines of evidence point to its pathogenesis being rooted in early development Citation[1,2]. The current theory posits that schizophrenia is neurodevelopmental in origin Citation[3], with the interaction between genes and environment resulting in the overt expression of clinical symptomatology. Recently, there has been an upsurge of research interest in studying children and adolescents with schizophrenia since they provide an opportunity to understand the underlying neurodevelopment pathway to disease expression.

Early-onset schizophrenia (EOS; manifestation of psychotic symptoms before 18 years of age) occurs in 4% of all schizophrenia cases, representing a rare, usually more severe and familial variant of the adult-onset counterpart Citation[4–6]. EOS lies at one end of the continuum of disease severity and genetic liability Citation[7]. Early-onset cases show clinical, cognitive, genetic and neurobiologic continuity with adult-onset schizophrenia Citation[7–9], albeit with enduring clinical morbidity and psychosocial disability Citation[10]. Thus, the study of this enriched population is likely to provide important pointers to the neurodevelopmental origins and pathophysiological mechanisms of schizophrenia.

Early-onset schizophrenia runs a chronic course, characterized by severe premorbid neurodevelopmental abnormalities, worse long-term outcome and delays in reaching crucial developmental milestones in motor coordination, language, speech and social functioning before psychosis onset Citation[10–16]. Young-onset cases are associated with a greater number of hospitalizations Citation[17], developmental deviance and higher rate of re-admissions Citation[18] compared with adult-onset schizophrenia.

Many studies suggest that cognitive deficits are an extension of the phenotype of schizophrenia, but few studies have investigated cognition in EOS. Overall, EOS shows generalized cognitive deficits across a broad range of ability domains, with the largest effect sizes being reported in general intellectual functioning, verbal learning and memory, and executive function Citation[19–24]. Similar cognitive impairments have been reported in adult-onset cases, although there are some discrepancies in specific domains of cognition. Some studies have reported that visual information processing and sustained attention are selective strengths in EOS Citation[25–27], but this has not been reported in other studies Citation[20,22]. A plausible explanation for these discrepancies may be disease heterogeneity, the effects of medication and varying IQ. In a group of medication-naive EOS patients, Brickman and colleagues reported a large deficit (<3 standard deviations below normative means) in attention function, as measured using the digit span subtest from the Wechsler Intelligence Scale for Children Citation[21]. By contrast, Ueland et al. confirmed the findings documented in earlier reports showing least impairment in sustained attention and early visual information processing Citation[25].

The issue of whether cognitive functioning changes over time in EOS has been a focus of intense interest. Longitudinal studies on adult-onset patients have shown relative stability or minor improvements over a 2- to 20-year period Citation[28–30]. In a review, Kurtz investigated the trajectory of cognitive deficits across the lifespan in community-dwelling outpatients between the ages of 20 and 85 years, and showed that impairments across a majority of cognitive measures remained stable after illness onset Citation[30]. At 13-year follow-up, Gochman et al. prospectively examined a childhood-onset schizophrenia cohort at 2-year intervals on general intellectual functioning and reported that cognitive decline occurred 2 years prior to disease onset and 2 years after the onset of psychotic symptoms, which was subsequently followed by cognitive stability Citation[31]. Therefore, there is no evidence of a progressive loss of cognitive functioning after the initial decline at illness onset.

During normal neurodevelopment, synaptic density increases until 2 years, followed by gradual decline during childhood and a steep fall-off during late childhood and early adolescence Citation[32]. During adolescence, decreases in synaptic density, axon retraction and neuronal fallout in the prefrontal cortex coincide with an increased ability to solve abstract reasoning, response inhibition, processing speed and complex high-order cognitive tasks. The neurodevelopmental hypothesis proposes that patients with schizophrenia may have too many, too few or unnecessary synaptic connections that are eliminated during adolescence; abnormality in pruning would lead to a reduction in synaptic connectivity that goes beyond the liability threshold, ultimately leading to the manifestation of psychotic symptoms Citation[33].

There are numerous studies of brain structural abnormalities in schizophrenia Citation[34]. Neuroimaging studies of adult-onset schizophrenia have consistently reported increased lateral ventricular volume Citation[35], reduced gray and white matter volumes Citation[36–38], particularly in the prefrontal region Citation[39], and a reduction in thalamic size Citation[40]. Similarly, imaging studies on early-onset cases have reported enlarged ventricular size, lower regional gray matter volumes (e.g., prefrontal cortex and cerebellum), white matter abnormalities Citation[41], increased basal ganglia volume Citation[14,17,42] and reduced volume of the thalamus Citation[9,43], cerebellum Citation[42] and anterior cingulated gyrus Citation[44]. Adult-onset studies have also reported hippocampal volume reduction Citation[45]. While four independent studies reported no hippocampal volume reduction in young-onset cases Citation[46–49], this may have been secondary to a lack of statistical power and heterogeneity with respect to clinical characteristics and pharmacotherapy Citation[49]. Indeed, modifications in methodology and the study of a larger sample revealed reduced hippocampal volume in the National Institute of Mental Health (NIMH) EOS cohort Citation[50–52] which, moreover, was shown to be associated with specific abnormalities in memory function Citation[53]. Longitudinal data available from the NIMH EOS cohort (treatment-refractory group of EOS patients) suggest a back-to-front wave of cortical gray matter volume reduction, beginning in the parietal lobes, progressing rostrally to the superior frontal and temporal cortices Citation[54], together with bilateral dorsal-to-ventral frontal gray matter reductions across the medial hemispheric surfaces separated from cingulate and limbic areas Citation[55]. The progressive loss of the cortical gray matter developmental trajectory normalizes in parietal regions and becomes restricted to the prefrontal and superior temporal cortices as EOS subjects move into adulthood Citation[56].

Few studies have used positron emission tomography to investigate neurobiological and neurochemical mechanisms underlying EOS Citation[14], possibly because of ionizing radiation exposure and the need for ready access to a cyclotron (for a review, see Citation[57]).

Schizophrenia has a heritability of 80% Citation[58]. Twin, family and genetic studies support a strong genetic basis Citation[6,59–62]. EOS patients provide a unique opportunity to understand the pathophysiology of schizophrenia at a neurobiological and genetic level as they provide evidence for the development of symptom expression and are associated with higher genetic loading and familiality. The NIMH group have identified four putative schizophrenia-susceptibility genes in childhood-onset schizophrenia implicated in adult-onset schizophrenia, namely DAOA or G72 (previously known as G72/G30) at 13q33.2 Citation[63], GAD1 (2q31.1) Citation[64], NRG1 (8p12) Citation[65] and DTNBP1 (6p22.3) Citation[66]. GAD1 is associated with cortical gray matter loss Citation[64]; NRG1 is associated with reduction in gray and white matter volume (and lateral ventricular enlargement) Citation[65]; while DTNBP1 expression is influenced by cis-acting variation at the gene locus, which may mediate association between DTNBP1 and schizophrenia, and by trans-acting polymorphism on chromosome 8p Citation[67,68].

Compared with adults with schizophrenia, a higher rate of cytogenetic abnormalities has been reported in individuals in the NIMH EOS cohort, implicating a strong association with very early-onset age. These include an inherited balanced 1:7 translocation, mosaic trisomy X (50% XXX), Turner’s syndrome (XO) and 22q11 deletion (velocardiofacial) syndrome Citation[69]. Atypical sex chromosomal anomalies have also been identified at a higher rate in EOS than adult-onset cases Citation[70]. Technological advances in high-density microarrays have provided us with important insights into difficult to detect deleterious mutations that could affect neurodevelopment. Structural genomic variations, or copy number variations (CNVs), are large duplications and deletions involved in disease etiology. Although some CNVs may be benign, and less likely to be fully penetrant owing to the genetic complexity and clinical heterogeneity of schizophrenia, emerging evidence has reported overabundance of rare CNVs on known genes in EOS Citation[71]. These include the genetic loci 22q11.21, 2p25.3 (MYT1L), 16p11.2 (neurexin 1, NRXN1) and 3p25.3 (SRGAP3), some of which have been associated with early neurodevelopmental difficulties (NRXN1) and neuronal development (SRGAP3). Recent studies have shown that rare de novo copy-number mutations exist in familial and nonfamilial (sporadic) forms in childhood disorders Citation[72]. Investigating the association between specific susceptibility genes and cognitive functioning in schizophrenia has been an area of interest. The Maudsley EOS study showed an association of the serotonin receptor 2A gene (5HTR2A) with attention and executive functioning in EOS probands and their first-degree relatives [Vyas NS, Ternouth A, Powell JF, Puri BK. Association of a 5HTR2A gene polymorphism with executive and attentional functions in patients with early-onset schizophrenia and their first-degree relatives. Manuscript Submitted], which supports an earlier study in adult-onset patients Citation[73]. Further similar investigations using a family-based design Citation[74] may help us to better understand the mechanisms underlying the transition to psychosis in apparently healthy siblings of schizophrenia patients who have not passed the period of risk for developing the illness.

In conclusion, EOS represents a rare, chronic and possibly more severe variant of the adult form of schizophrenia, associated with premorbid abnormalities, cognitive deficits, poor psychosocial outcome, higher genetic loading and greater loss of cortical gray matter volume compared with adult-onset cases. Future research could investigate gene–environment interactions to understand the developmental trajectories and disease progression at a young age in schizophrenia. Advances in current methodologies will provide a blueprint for unraveling the impact of rare structural genetic mutations on the neurodevelopmental pathway in EOS. It is anticipated that investigation into the clinical, genetic, cognitive and neuroimaging aspects of EOS will provide a window of opportunity for creating a novel model that systematically explains the neurobiological pathway underlying the occurrence of events leading to the clinical syndrome.

Financial & competing interests disclosure

Sanjiv Kumra is consultant to Bristol-Myers Squibb, AstraZeneca and Schering Plough. He has also conducted studies that were industry sponsored for AstraZeneca and Bristol-Myers Squibb. The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed.

No writing assistance was utilized in the production of this manuscript.

References

  • Cannon M, Caspi A, Moffitt TE et al. Evidence for early-childhood, pan-developmental impairment specific to schizophreniform disorder: results from a longitudinal birth cohort. Arch. Gen. Psychiatry59, 449–456 (2002).
  • Fatemi SH, Folsom TD. The neurodevelopmental hypothesis of schizophrenia, revisited. Schizophr. Bull.35(3), 528–548 (2009).
  • Weinberger DR. Implications of normal brain development for the pathogenesis of schizophrenia. Arch. Gen. Psychiatry44(7), 660–669 (1987).
  • Cannon M, Jones P, Huttunen MO et al. School performance in Finnish children and later development of schizophrenia: a population-based longitudinal study. Arch. Gen. Psychiatry56(5), 457–463 (1999).
  • Häfner H, Maurer K, Loffler W, Riecher-Rossler A. The influence of age and sex on the onset and early course of schizophrenia. Br. J. Psychiatry162, 80–86 (1993).
  • Asarnow RF, Nuechterlein KH, Fogelson D et al. Schizophrenia and schizophrenia-spectrum personality disorders in the first-degree relatives of children with schizophrenia: the UCLA family study. Arch. Gen. Psychiatry58(6), 581–588 (2001).
  • Hollis C. Adult outcomes of child- and adolescent-onset schizophrenia: diagnostic stability and predictive validity. Am. J. Psychiatry157(10), 1652–1659 (2000).
  • Asarnow RF, Asamen J, Granholm E, Sherman T, Watkins JM, Williams ME. Cognitive/neuropsychological studies of children with a schizophrenic disorder. Schizophr. Bull.20(4), 647–669 (1994).
  • Frazier JA, Giedd JN, Hamburger SD et al. Brain anatomic magnetic resonance imaging in childhood-onset schizophrenia. Arch. Gen. Psychiatry53(7), 617–624 (1996).
  • Vyas NS, Hadjulis M, Vourdas A, Byrne P, Frangou S. The Maudsley Early Onset Schizophrenia Study. Predictors of psychosocial outcome at 4-year follow-up. Eur. Child Adolesc. Psychiatry16(7), 465–470 (2007).
  • Walker EF, Savoie T, Davis D. Neuromotor precursors of schizophrenia. Schizophr. Bull.20(3), 441–451 (1994).
  • Alaghband-Rad J, McKenna K, Gordon CT et al. Childhood-onset schizophrenia: the severity of premorbid course. J. Am. Acad. Child Adolesc. Psychiatry34(10), 1273–1283 (1995).
  • Asarnow RF, Brown N, Strandburg R. Children with a schizophrenic disorder: neurobehavioural studies. Eur. Arch. Psychiatry Clin. Neurosci.245(2), 70–79 (1995).
  • Jacobsen LK, Rapoport JL. Research update: childhood-onset schizophrenia: implications of clinical and neurobiological research. J. Child Psychol. Psychiatry39(1), 101–113 (1998).
  • Nicolson R, Lenane M, Singaracharlu S et al. Premorbid speech and language impairments in childhood-onset schizophrenia: association with risk factors. Am. J. Psychiatry157(5), 794–800 (2000).
  • McClellan J, Breiger D, McCurry C, Hlastala SA. Premorbid functioning in early-onset psychotic disorders. J. Am. Acad. Child Adolesc. Psychiatry42(6), 666–672 (2003).
  • Frazier JA, Alaghband-Rad J, Jacobsen L et al. Pubertal development and onset of psychosis in childhood onset schizophrenia. Psychiatry Res.70(1), 1–7 (1997).
  • Rabinowitz J, Lewine SZ, Hafner H. A population based elaboration of the role of age of onset on the course of schizophrenia. Schizophr. Res.88(1–3), 96–101 (2006).
  • Kenny JT, Friedman L, Findling RL et al. Cognitive impairment in adolescents with schizophrenia. Am. J. Psychiatry154(11), 1613–1615 (1997).
  • Kumra S, Wiggs E, Bedwell J et al. Neuropsychological deficits in pediatric patients with childhood-onset schizophrenia and psychotic disorder not otherwise specified. Schizophr. Res.42(2), 135–144 (2000).
  • Brickman AM, Buchsbaum MS, Bloom R et al. Neuropsychological functioning in first-break, never-medicated adolescents with psychosis. J. Nerv. Ment. Dis.192(9), 615–622 (2004).
  • Rhinewine JP, Lencz T, Thaden EP et al. Neurocognitive profile in adolescents with early-onset schizophrenia: clinical correlates. Biol. Psychiatry58(9), 705–712 (2005).
  • Cervellione KL, Burdick KE, Cottone JG, Rhinewine, JP, Kumra S. Neurocognitive deficits in adolescents with schizophrenia: longitudinal stability and predictive utility for short-term functional outcome. J. Am. Acad. Child Adolesc. Psychiatry46(7), 867–878 (2007).
  • Sharma T, Harvey PD. The Early Course of Schizophrenia. Oxford University Press, Oxford, UK (2006).
  • Ueland T, Øie MN, Inge Landrø N, Rund BR. Cognitive functioning in adolescents with schizophrenia spectrum disorders. Psychiatry Res.126(3), 229–239 (2004).
  • Øie M, Rund BR. Neuropsychological deficits in adolescent-onset schizophrenia compared with attention deficit hyperactivity disorder. Am. J. Psychiatry156(8), 1216–1222 (1999).
  • Kravariti E, Morris RG, Rabe-Hesketh S, Murray RM, Frangou S. The Maudsley early onset schizophrenia study: cognitive function in adolescents with recent onset schizophrenia. Schizophr. Res.61(2–3), 137–148 (2003).
  • Kondel TK, Mortimer AM, Leeson VC, Laws KR, Hirsch SR. Intellectual differences between schizophrenic patients and normal controls across the adult lifespan. J. Clin. Exp. Neuropsychol.25(8), 1045–1056 (2003).
  • Hoff AL, Svetina C, Shields G, Stewart J, DeLisi LE. Ten year longitudinal study of neuropsychological functioning subsequent to a first episode of schizophrenia. Schizophr. Res.78(1), 27–34 (2005).
  • Kurtz MM. Neurocognitive impairment across the lifespan in schizophrenia: an update. Schizophr. Res.74(1), 15–26 (2005).
  • Gochman PA, Greenstein D, Sporn A et al. IQ stabilization in childhood-onset schizophrenia. Schizophr. Res.77(2–3), 271–277 (2005).
  • Huttenlocher PR. Synaptic density in human frontal cortex – developmental changes and effects of aging. Brain Res.163(2), 195–205 (1979).
  • McGlashan TH, Hoffman RE. Schizophrenia as a disorder of developmentally reduced synaptic connectivity. Arch. Gen. Psychiatry57(7), 637–648 (2000).
  • Puri BK. Progressive structural brain changes in schizophrenia. Expert Rev. Neurother.10(1), 33–42 (2010).
  • Wright IC, Rabe-Hesketh S, Woodruff PW, David AS, Murray RM, Woodruff ET. Meta-analysis of regional brain volume in schizophrenia. Am. J. Psychiatry157, 16–25 (2000).
  • Lawrie SM, Abukmeil SS. Brain abnormality in schizophrenia: a systematic and quantitative review of volumetric magnetic resonance imaging studies. Br. J. Psychiatry172, 110–120 (1998).
  • Wright IC, Rabe-Hesketh S, Woodruff PW, David AD, Murray RM, Bullmore ET. Meta-analysis of regional brain volumes in schizophrenia. Am. J. Psychiatry157(1), 16–25 (2000).
  • Gur RE, Cowell PE, Latshaw A et al. Reduced dorsal and orbital prefrontal gray matter volumes in schizophrenia. Arch. Gen. Psychiatry57(8), 761–768 (2000).
  • Gur RE, Turetsky BI, Cowell PE et al. Temporolimbic volume reductions in schizophrenia. Arch. Gen. Psychiatry57(8), 769–775 (2000).
  • Konick LC, Friedman L. Meta-analysis of thalamic size in schizophrenia. Biol. Psychiatry49(1), 28–38 (2001).
  • Pagsberg AK, Baare WF, Raabjerg Christensen AM et al. Structural brain abnormalities in early onset first-episode psychosis. J. Neural Transm.114(4), 489–498 (2007).
  • Jacobsen LK, Giedd JN, Berquin PC et al. Quantitative morphology of the cerebellum and fourth ventricle in childhood-onset schizophrenia. Am. J. Psychiatry154(12), 1663–1669 (1997).
  • Dasari M, Friedman L, Jesberger J et al. A magnetic resonance imaging study of thalamic area in adolescent patients with either schizophrenia or bipolar disorder as compared to healthy controls. Psychiatry Res.91(3), 155–162 (1999).
  • Marquardt RK, Levitt JG, Blanton RE et al. Abnormal development of the anterior cingulated in childhood-onset schizophrenia: a preliminary quantitative MRI study. Psychiatry Res.138(3), 221–233 (2005).
  • Nelson MD, Saykin AJ, Flashman LA, Riordan HJ. Hippocampal volume reduction in schizophrenia as assessed by magnetic resonance imaging: a meta-analytic study. Arch. Gen. Psychiatry55(5), 433–440 (1998).
  • Matsumoto H, Simmons A, Williams S et al. Superior temporal gyrus abnormalities in early-onset schizophrenia: similarities and differences with adult-onset schizophrenia. Am. J. Psychiatry158(8), 1299–1304 (2001).
  • Jacobsen LK, Giedd N, Vaituzis AC et al. Temporal lobe morphology in childhood-onset schizophrenia. Am. J. Psychiatry153(3), 355–361 (1996).
  • Findling RL, Friedman L, Kenny JT, Swales TP, Cola DM, Schulz SC. Adolescent schizophrenia: a methodologic review of the current neuroimaging and neuropsychologic literature. J. Autism Dev. Disord.25(6), 627–639 (1995).
  • Levitt JG, Blanton RE, Caplan R et al. Medial temporal lobe in childhood-onset schizophrenia. Psychiatry Res.108(1), 17–27 (2001).
  • Giedd JN, Jeffries NO, Blumenthal J et al. Childhood-onset schizophrenia: progressive brain changes during adolescence. Biol. Psychiatry46(7), 892–898 (1999).
  • Jacobsen LK, Giedd JN, Castellanos FX et al. Progressive reduction of temporal lobe structures in childhood-onset schizophrenia. Am. J. Psychiatry155(5), 678–685 (1998).
  • Nugent TF III, Herman DH, Ordonez A et al. Dynamic mapping of hippocampal development in childhood onset schizophrenia. Schizophr. Res.90(1–3), 62–70 (2006).
  • Roofeh D, Cottone J, Burdick KE et al. Deficits in memory strategy use are related to verbal memory impairments in adolescents with schizophrenia-spectrum disorders. Schizophr. Res.85(1–3), 201–212 (2006).
  • Thompson PM, Vidal C, Giedd JN et al. Mapping adolescent brain change reveals dynamic wave of accelerated gray matter loss in very early-onset schizophrenia. Proc. Natl Acad. Sci. USA98(20), 11650–11655 (2001).
  • Vidal CN, Rapoport JL, Hayashi KM et al. Dynamically spreading frontal and cingulate deficits mapped in adolescents with schizophrenia. Arch. Gen. Psychiatry63(1), 25–34 (2006).
  • Greenstein D, Lerch J, Shaw P et al. Childhood-onset schizophrenia: cortical brain abnormalities as young adults. J. Child Adolesc. Psychiatry47(10), 1003–1012 (2006).
  • Patel NH, Vyas NS, Puri BK, Nijran KS, Al-Nahhas A. Positron emission tomography in schizophrenia: a new perspective. J. Nucl. Med.51(4), 511–520 (2010).
  • Sullivan PF, Kendler KS, Neale MC. Schizophrenia as a complex traits: evidence from a meta-analysis of twin studies. Arch. Gen. Psychiatry60(12), 1187–1192 (2003).
  • Kallmann FJ, Roth B. Genetic aspects of preadolescent schizophrenia. Am. J. Psychiatry112(8), 599–606 (1956).
  • Kolvin I, Ounsted C, Humphrey M, McNay A. Studies in the childhood psychoses. II. The phenomenology of childhood psychoses. Br. J. Psychiatry118(545), 385–395 (1971).
  • Murray RM, Jones PB, Susser E, van Os J, Cannon M. The Epidemiology of Schizophrenia. Cambridge University Press, Cambridge, UK (2003).
  • Nicolson R, Brookner FB, Lenane M et al. Parental schizophrenia spectrum disorders in childhood-onset and adult-onset schizophrenia. Am. J. Psychiatry160(3), 490–495 (2003).
  • Addington AM, Gornick M, Sporn AL et al. Polymorphisms in the 13q33.2 gene G72/G30 are associated with childhood-onset schizophrenia and psychosis not otherwise specified. Biol. Psychiatry55, 876–880 (2004).
  • Addington AM, Gornick M, Duckworth J et al. GAD (2q31.1), which encodes glutamic acid decarboxylase (GAD67) is associated with childhood onset schizophrenia and cortical gray matter volume loss. Mol. Psychiatry10(6), 581–588 (2005).
  • Addington AM, Gornick MC, Shaw P et al. Neuregulin 1 (8p12) and childhood-onset schizophrenia: susceptibility haplotypes for diagnosis and brain developmental trajectories. Mol. Psychiatry12(2), 195–205 (2007).
  • Gornick MC, Addington AM, Sporn A et al. Dysbindin (DTNBP1, 6p22.3) is associated with childhood-onset psychosis and endophenotypes measured by the Premorbid Adjustment Scale (PAS). J. Autism Dev. Disord.35(6), 831–838 (2005).
  • Bray NJ, Holmans PA, van den Bree MB et al. Cis- and trans loci influence expression of the schizophrenia susceptibility gene DTNBP1. Hum. Mol. Genet.17(8), 1169–1174 (2008).
  • Bray NJ, Preece A, Williams NM et al. Haplotypes at the dystrobrevin binding protein 1 (DTNBP1) gene locus mediate risk for schizophrenia through reduced DTNBP1 expression. Hum. Mol. Genet.14(14), 1947–1954 (2005).
  • Kumra S, Wiggs E, Krasnewich D et al. Brief Report: association of sex chromosome anomalies with childhood-onset psychotic disorders. J. Am. Acad. Child Adolesc. Psychiatry37(3), 292–296 (1998).
  • Eckstrand K, Addington AM, Stromberg T et al. Sex chromosome anomalies in childhood onset schizophrenia. Mol. Psychiatry13(10), 910–911 (2008).
  • Stefansson H, Rujescu D, Cichon S et al. Large recurrent microdeletions associated with schizophrenia. Nature455(7210), 232–236 (2008).
  • Sebat J, Lakshmi B, Malhotra D et al. Strong association of de novo copy number mutations with autism. Science316(5823), 445–449 (2007).
  • Üçok A, Alpsan H, Cakir S, Saruhan-Direskeneli G. Association of a serotonin receptor 2A gene polymorphism with cognitive functions in patients with schizophrenia. Am. J. Med. Genet. B Neuropsychiatr. Genet.144B(5), 704–707 (2007).
  • Groom MJ, Jackson GM, Calton TG et al. Cognitive deficits in early-onset schizophrenia spectrum patients and their non-psychotic siblings: a comparison with ADHD. Schizophr. Res.99(1–3), 85–95 (2008).

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.