Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2014 Nov 20.
Published in final edited form as: Mol Psychiatry. 2014 Feb;19(2):141–143. doi: 10.1038/mp.2013.160

DISC1 as a genetic risk factor for schizophrenia and related major mental illness: response to Sullivan

David J Porteous 1, Pippa Thomson 1, J Kirsty Millar 1, Kathryn L Evans 1, William Hennah 2, Dinesh Soares 1, Shane McCarthy 3, Richard McCombie 3, Stephen Clapcote 4, Carsten Korth 5, Nicholas Brandon 6, Akira Sawa 7, Atsushi Kamiya 8, John Roder 9, Stephen M Lawrie 10, Andrew McIntosh 10, David St Clair 11, Douglas Blackwood 11
PMCID: PMC4238281  NIHMSID: NIHMS638739  PMID: 24457522

In a recent guest editorial1, Patrick Sullivan questioned several aspects of the original Scottish t(1;11) family through which DISC1 was discovered24. He challenged the wider significance of the original genetic finding and questioned the relevance of DISC1 biology to psychiatry, casting doubt on the validity of the ‘integrative’ approach. Sullivan prefaced his strong views on DISC1 by welcoming the ‘uncompromising statistical rigour and replication’ that is now being applied to his area of expertise in genome wide association studies (GWAS). In his opinion, the evidence in support of DISC1 falls short of the mark, but the methods he champions are based on the assumption of the ‘common disease; common variant’ hypothesis and are not valid tests of the ‘common disease; rare variant’ hypothesis of which DISC1 is a prime example. If we were to apply his criteria universally, we would, for example, be obliged to ignore everything learnt about Alzheimer’s Disease through APP, PSEN1 and PSEN2, none of which show up in GWAS, yet these clear-cut, family-based rare variant discoveries have provided profound insights into the disorder.5 We would likewise have to ignore much of the insights that have been gained from copy number variant analysis in autism6, 7 and, indeed, schizophrenia8, 9 Denying an important role for rare variants makes no sense and contradicts theoretical considerations10: insights from exceptional cases and families have repeatedly served medical research well across the full spectrum of conditions generally considered ‘common and complex’.

So why does Sullivan pick out DISC1 for special criticism? This is not the time or place to reiterate the wealth of evidence in favour of DISC1 and of the DISC1 pathway, but readers might find it useful to consult recent reviews1114 in addition to those he cites1. Here, we summarise his Table 1, answer his main questions and expand upon these in the main text.

Table 1.

Answers to questions from Sullivan about DISC1

The pedigree
The classical karyotyping methods developed in Edinburgh by Jacobs et al42 are robust for tracking the t(1;11) translocation2, were used effectively again in Blackwood et al, 2001, but for convenience, efficiency and utility, have since been supplemented with fluorescence in situ hybridisation and now PCR methods: all methods give entirely congruous results. There is no evidence from follow up studies for any significant change from the original report that the t(1;11) tracks susceptibility to a spectrum of schizophrenia, bipolar disorder and major depressive disorder with genome wide significance. A comprehensive follow up on the t(1;11) family, including brain imaging, has recently been completed and will be submitted for publication in due course.
The phenotypes
These are not ‘worrying’. The presence of major depressive disorder along with bipolar disorder and schizophrenia is supported by GWAS-based co-heritability analysis. The absence of mental retardation is unsurprising: mental retardation is a common co-morbid feature of many deletion syndromes, with or without other psychiatric features, but not of balanced translocations.
The focus on DISC1
The focus on DISC1 derives from the remarkable properties attributable to the protein – interaction and modulation of multiple proteins that have been independently linked genetically to psychiatric and neurodevelopmental disorders and biologically to fundamental neurological processes of signalling, trafficking, synaptic function and neurogenesis. Other molecular genetic consequences of the t(1;11) have been considered and are published. None challenge the primacy of the DISC1 disruption.
Genetic results
The examples cited as the ‘most rigorously conducted genomic studies’ are selective and biased towards meta-analyses of linkage and association, blunt tools to test for rare variants. Positive published findings with genome wide significance have been ignored.

Sullivan queries the Robertsonian t(13q;14q) translocation reported in a small branch of the Scottish t(1;11) pedigree, and the chromosome 1 constriction that is more-prevalent but predominantly in branches of the family that do not carry the t(1;11). The former was in fact discussed in the Lancet study2, and neither materially alters the conclusion that the t(1;11) robustly tracks the high risk of psychiatric illness in this family. The LOD score for schizophrenia alone meets stringent genome wide significance; for schizophrenia plus bipolar disorder and major depressive disorder it substantially exceeds genome wide significance (MLOD=7.1). Regarding the alleged disparity between the pedigree in the original 1970 report15 and the follow up report in 20013, there is nothing suspicious here. Any clinician who has followed up families in their care over such a long period of time will fully understand that not all of the original members will still be willing or able to participate and that new members and new cases may become available. That the follow up study confirmed and substantiated the original study should give comfort not concern. For the avoidance of any further doubt, we can confirm that the pedigree presented in the 2001 study focuses only upon branches of the family carrying the t(1;11), that the t(1;11) shows unequivocal linkage to the high burden of major mental illness in this family and that the t(1;11) has been scored both by classical chromosome banding and by fluorescence in situ hybridisation16. The molecular cloning of the t(1;11) breakpoint4 has more recently allowed us to develop a PCR-based assay spanning the breakpoint with sequence confirmation: we routinely apply this test to validate samples in contemporary studies (Thomson, unpublished). Finally, regarding the Scottish t(1;11) pedigree, a major third wave of follow up has recently been completed with brain imaging added to the clinical phenotype: this will be submitted shortly for publication.

Sullivan finds the spectrum of psychiatric diagnoses seen in the t(1;11) family ‘worrying’, but what the t(1;11) family showed par example is evidence for genetic and biological overlap between schizophrenia, bipolar disorder and major depressive disorder. The epidemiological evidence is now clear on this17, 18 as too is the accumulating GWAS evidence for shared genetic liability across DSM-defined diagnoses19, 20. The presence of both schizophrenia and major depressive disorder in the t(1;11) family is entirely consistent with recent GWAS-derived estimates of co-heritability: schizophrenia and bipolar disorder (0.68); bipolar disorder and major depressive disorder (0.47); schizophrenia and major depressive disorder (0.43)21. The variability in presentation in the Scottish t(1;11) family may be accounted for by genetic (independently segregating modifiers) or non-genetic (experiences and exposures) factors: these are under active investigation. In a similar vein, there is nothing ‘worrying’ about the absence of mental retardation: mental retardation is seen frequently in chromosome deletion conditions without any other psychiatric features22. Finally, it is indeed the case that the original propositus was a male with adolescent conduct disorder: schizophrenia is known to increase in prevalence with an increasing number of conduct problems in childhood23.

With regards to the focus on DISC1, other possible explanations – a functional effect of the disrupted chromosome 11 locus, an independent or co-dependent effect of the long non-coding RNA antiparallel to DISC1, named DISC2, and/or a dominant negative effect of DISC1 fusion proteins - have not been ignored. We and others have considered and actively investigated all of these alternative hypotheses2426, and will continue to do so as the necessary tools, reagents and assays are developed. As part of on-going work, we are carrying out whole-genome sequencing of t(1;11) family members and will report the findings in due course.

With regards to independent genetic results, the failure of meta-analyses of genome wide linkage and association to replicate DISC1 is misleading: it is well understood that the presence and impact of rare or recent genetic events go undetected by these methods5. GWAS depends upon genotyped alleles tagging risk loci that are then implicated indirectly by a significant difference in allele frequency between cases and controls. The statistical tests may be highly significant, but the population level odds ratios are typically less than 1.2. Given the reliance on strong linkage disequilibrium at a whole-sample level between the genotyped common allele and the risk allele, this method is neither designed nor suited to the detection of rare risk alleles: this is true even for those of high penetrance and impact, for example structural or coding variants, for which the odds ratios are typically greater than 210. Family-based linkage studies in relatively isolated populations have the potential to bridge the gap between ‘common’ and ‘rare’ variant detection, and this is true for DISC1. In a population based study of 221 Finnish families with schizophrenia, D1S2709, an intragenic marker for DISC1, gave a Z(max) of 2.71 in the combined cohort increasing to 3.21 when an internal isolate was excluded. Linkage to DISC1 was replicated in an independent set of 70 Finnish families with schizophrenia27. Analysis of the combined Finnish samples exceeds genome-wide significant linkage for DISC1 and schizophrenia (LOD=3.6)28. When conditioned on the DISC1 risk haplotype, the NDE1 locus was identified as a strong linkage peak (LOD = 3.17), a finding that was replicated by association (P<0.011 after correction for multiple testing)28. Although NDE1 does not appear as a top GWAS ‘hit’, deletions and duplications spanning NDE1 are among the most common copy number variants in schizophrenia29. NDE1 is critical for multiple neurodevelopmental processes: through protein-protein interaction, DISC1 regulates NDE1 function30. Thus, consistent with current neurodevelopmental concepts in schizophrenia31, the genetic and biological evidence for DISC1 and NDE1 provide evidence for a shared ‘risk’ pathway. NDE1 is just one of a large number of proteins for which there is firm evidence for direct interaction with DISC1: several likewise have biological functions that have been independently linked to psychiatric illness1114. None independently meet PGC criteria for genome wide significance, but some meet gene-wide significance. Of note, the DISC1 interactor PDE4B encodes a regulator of cAMP signalling that is targeted by the antidepressant/antipsychotic rolipram and the PDE4B gene is directly disrupted by a t(1;16) translocation in two cousins diagnosed with schizophrenia or a psychotic disorder13, 30, 32.

What have we learnt so far from sequencing DISC1? Perhaps more than Sullivan suggests1. An early sequencing study of DISC1 identified six ultra-rare non-synonymous amino-acid substitutions in 288 schizophrenia cases but not 288 matched controls33. Moens et al34 reported an increased burden of rare missense mutations in a Swedish cohort. Green et al35 reported an excess of exon 11 rare variants in schizotypy. Crowley et al36 found nothing of statistical significance on sequencing 2.7kb of DISC1 in 727 schizophrenia cases and 733 controls. In a recently published study, we sequenced the full 528kb of the genomic DISC1 locus in 1,562 cases and controls37. We found no significant increase in the burden of non-synonymous coding variants in cases, but did find evidence for gene-wide enrichment of conserved, regulatory variants in major depressive disorder. We also reported the ultra-rare variant R37W in a case of major depressive disorder, transmitted to two affected offspring, one of the ultra-rare variants first reported by Song et al33 in a case of schizophrenia. We now know that R37W alters the cellular distribution of DISC1, the modulatory role of DISC1 in cellular stress38 and also mitochondrial trafficking39.

Sullivan’s dictum that ‘biology does not have a role in establishing a genetic association (but only later in understanding its role)’ is not one that we or a large section of the genetics community would agree with, but ironically it does apply to the case for the Scottish t(1;11) family: only once the breakpoint was cloned and DISC1 identified could the biology begin4. It is worth remembering that in 2000, DISC1 was, and still is, a singleton protein with no close homology to any other and thus no neat and meaningful descriptor. Naming the protein in the context of its discovery (genome wide significance for schizophrenia in the t(1;11) family) made sense then and no change is warranted now.

There is no harm and much to gain from reasoned and vigorous debate over alternative approaches to cracking a scientific problem as pressing and challenging as understanding the genetic and biological basis of major mental illness. Indeed, we should take as clear evidence of progress that the relative merits of case, family and population approaches can be debated on the basis of evidence. The value of a biological pathway based approach is very well established in other fields: in our view, shared by others31, 40, 41, it is critical to the advancement of psychiatric research. DISC1 has already made a valuable contribution to this endeavour. We believe strongly that DISC1 and the DISC1 pathway paradigm have served the field well, will continue to do so and that the foundational evidence is sound. We will continue to take this systems approach as the one most likely to provide actionable biological insight.

References

  • 1.Sullivan PF. Questions about DISC1 as a genetic risk factor for schizophrenia. Mol Psychiatry. 2013;18(10):1050–1052. doi: 10.1038/mp.2012.182. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.St Clair D, Blackwood D, Muir W, Carothers A, Walker M, Spowart G, et al. Association within a Family of a Balanced Autosomal Translocation with Major Mental-Illness. Lancet. 1990;336(8706):13–16. doi: 10.1016/0140-6736(90)91520-k. [DOI] [PubMed] [Google Scholar]
  • 3.Blackwood DH, Fordyce A, Walker MT, St Clair DM, Porteous DJ, Muir WJ. Schizophrenia and affective disorders--cosegregation with a translocation at chromosome 1q42 that directly disrupts brain-expressed genes: clinical and P300 findings in a family. Am J Hum Genet. 2001;69 (2):428–433. doi: 10.1086/321969. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Millar JK, Wilson-Annan JC, Anderson S, Christie S, Taylor MS, Semple CA, et al. Disruption of two novel genes by a translocation co-segregating with schizophrenia. Hum Mol Genet. 2000;9(9):1415–1423. doi: 10.1093/hmg/9.9.1415. [DOI] [PubMed] [Google Scholar]
  • 5.Bertram L, Lill CM, Tanzi RE. The genetics of Alzheimer disease: back to the future. Neuron. 2010;68(2):270–281. doi: 10.1016/j.neuron.2010.10.013. [DOI] [PubMed] [Google Scholar]
  • 6.Sebat J, Lakshmi B, Malhotra D, Troge J, Lese-Martin C, Walsh T, et al. Strong association of de novo copy number mutations with autism. Science. 2007;316(5823):445–449. doi: 10.1126/science.1138659. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Gilman SR, Iossifov I, Levy D, Ronemus M, Wigler M, Vitkup D. Rare de novo variants associated with autism implicate a large functional network of genes involved in formation and function of synapses. Neuron. 2011;70(5):898–907. doi: 10.1016/j.neuron.2011.05.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Stefansson H, Rujescu D, Cichon S, Pietilainen OP, Ingason A, Steinberg S, et al. Large recurrent microdeletions associated with schizophrenia. Nature. 2008;455(7210):232–236. doi: 10.1038/nature07229. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Kirov G, Pocklington AJ, Holmans P, Ivanov D, Ikeda M, Ruderfer D, et al. De novo CNV analysis implicates specific abnormalities of postsynaptic signalling complexes in the pathogenesis of schizophrenia. Mol Psychiatry. 2012;17(2):142–153. doi: 10.1038/mp.2011.154. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Bodmer W, Bonilla C. Common and rare variants in multifactorial susceptibility to common diseases. Nat Genet. 2008;40(6):695–701. doi: 10.1038/ng.f.136. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Bradshaw NJ, Porteous DJ. DISC1-binding proteins in neural development, signalling and schizophrenia. Neuropharmacology. 2012;62(3):1230–1241. doi: 10.1016/j.neuropharm.2010.12.027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Brandon NJ, Sawa A. Linking neurodevelopmental and synaptic theories of mental illness through DISC1. Nat Rev Neurosci. 2011;12(12):707–722. doi: 10.1038/nrn3120. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Soares DC, Carlyle BC, Bradshaw NJ, Porteous DJ. DISC1: Structure, Function, and Therapeutic Potential for Major Mental Illness. ACS Chem Neurosci. 2011;2(11):609–632. doi: 10.1021/cn200062k. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Thomson PA, Malavasi EL, Grunewald E, Soares DC, Borkowska M, Millar JK. DISC1 genetics, biology and psychiatric illness. Front Biol (Beijing) 2013;8(1):1–31. doi: 10.1007/s11515-012-1254-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Jacobs PA, Brunton M, Frackiewicz A, Newton M. Studies on a family with three cytogenetic markers. Ann Hum Genet. 1970;33:325–336. [Google Scholar]
  • 16.Millar JK, Christie S, Anderson S, Lawson D, Hsiao-Wei Loh D, Devon RS, et al. Genomic structure and localisation within a linkage hotspot of Disrupted In Schizophrenia 1, a gene disrupted by a translocation segregating with schizophrenia. Mol Psychiatry. 2001;6(2):173–178. doi: 10.1038/sj.mp.4000784. [DOI] [PubMed] [Google Scholar]
  • 17.Lichtenstein P, Yip BH, Björk C, Pawitan Y, Cannon TD, Sullivan PF, et al. Common genetic determinants of schizophrenia and bipolar disorder in Swedish families: a population-based study. The Lancet. 2009;373(9659):234–239. doi: 10.1016/S0140-6736(09)60072-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Gottesman II, Laursen TM, Bertelsen A, Mortensen PB. Severe mental disorders in offspring with 2 psychiatrically ill parents. Arch Gen Psychiatry. 2010;67(3):252–257. doi: 10.1001/archgenpsychiatry.2010.1. [DOI] [PubMed] [Google Scholar]
  • 19.Purcell SM, Wray NR, Stone JL, Visscher PM, O’Donovan MC, Sullivan PF, et al. Common polygenic variation contributes to risk of schizophrenia and bipolar disorder. Nature. 2009;460(7256):748–752. doi: 10.1038/nature08185. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Smoller JW, Craddock N, Kendler K, Lee PH, Neale BM, Nurnberger JI, et al. Identification of risk loci with shared effects on five major psychiatric disorders: a genome-wide analysis. Lancet. 2013;381(9875):1371–1379. doi: 10.1016/S0140-6736(12)62129-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Lee SH, Ripke S, Neale BM, Faraone SV, Purcell SM, Perlis RH, et al. Genetic relationship between five psychiatric disorders estimated from genome-wide SNPs. Nat Genet. 2013;45 (9):984–994. doi: 10.1038/ng.2711. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Swaminathan GJ, Bragin E, Chatzimichali EA, Corpas M, Bevan AP, Wright CF, et al. DECIPHER: web-based, community resource for clinical interpretation of rare variants in developmental disorders. Hum Mol Genet. 2012;21(R1):R37–44. doi: 10.1093/hmg/dds362. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Robins LN, Price RK. Adult disorders predicted by childhood conduct problems: results from the NIMH Epidemiologic Catchment Area project. Psychiatry. 1991;54(2):116–132. doi: 10.1080/00332747.1991.11024540. [DOI] [PubMed] [Google Scholar]
  • 24.Taylor MS, Devon RS, Millar JK, Porteous DJ. Evolutionary constraints on the disrupted in schizophrenia locus. Genomics. 2003;81(1):67–77. doi: 10.1016/s0888-7543(02)00026-5. [DOI] [PubMed] [Google Scholar]
  • 25.Zhou X, Chen Q, Schaukowitch K, Kelsoe JR, Geyer MA. Insoluble DISC1-Boymaw fusion proteins generated by DISC1 translocation. Mol Psychiatry. 2010;15(7):669–672. doi: 10.1038/mp.2009.127. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Eykelenboom JE, Briggs GJ, Bradshaw NJ, Soares DC, Ogawa F, Christie S, et al. A t(1;11) translocation linked to schizophrenia and affective disorders gives rise to aberrant chimeric DISC1 transcripts that encode structurally altered, deleterious mitochondrial proteins. Hum Mol Genet. 2012;21(15):3374–3386. doi: 10.1093/hmg/dds169. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Ekelund J, Hennah W, Hiekkalinna T, Parker A, Meyer J, Lonnqvist J, et al. Replication of 1q42 linkage in Finnish schizophrenia pedigrees. Mol Psychiatry. 2004;9(11):1037–1041. doi: 10.1038/sj.mp.4001536. [DOI] [PubMed] [Google Scholar]
  • 28.Hennah W, Tomppo L, Hiekkalinna T, Palo OM, Kilpinen H, Ekelund J, et al. Families with the risk allele of DISC1 reveal a link between schizophrenia and another component of the same molecular pathway, NDE1. Hum Mol Genet. 2007;16(5):453–462. doi: 10.1093/hmg/ddl462. [DOI] [PubMed] [Google Scholar]
  • 29.Ingason A, Rujescu D, Cichon S, Sigurdsson E, Sigmundsson T, Pietilainen OP, et al. Copy number variations of chromosome 16p13. 1 region associated with schizophrenia. Mol Psychiatry. 2011;16(1):17–25. doi: 10.1038/mp.2009.101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Bradshaw NJ, Soares DC, Carlyle BC, Ogawa F, Davidson-Smith H, Christie S, et al. PKA Phosphorylation of NDE1 Is DISC1/PDE4 Dependent and Modulates Its Interaction with LIS1 and NDEL1. J Neurosci. 2011;31(24):9043–9054. doi: 10.1523/JNEUROSCI.5410-10.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Insel TR. Rethinking schizophrenia. Nature. 2010;468(7321):187–193. doi: 10.1038/nature09552. [DOI] [PubMed] [Google Scholar]
  • 32.Millar JK, Pickard BS, Mackie S, James R, Christie S, Buchanan SR, et al. DISC1 and PDE4B are interacting genetic factors in schizophrenia that regulate cAMP signaling. Science. 2005;310(5751):1187–1191. doi: 10.1126/science.1112915. [DOI] [PubMed] [Google Scholar]
  • 33.Song W, Li W, Feng J, Heston LL, Scaringe WA, Sommer SS. Identification of high risk DISC1 structural variants with a 2% attributable risk for schizophrenia. Biochemical and Biophysical Research Communications. 2008;367(3):700–706. doi: 10.1016/j.bbrc.2007.12.117. [DOI] [PubMed] [Google Scholar]
  • 34.Moens LN, De Rijk P, Reumers J, Van den Bossche MJ, Glassee W, De Zutter S, et al. Sequencing of DISC1 pathway genes reveals increased burden of rare missense variants in schizophrenia patients from a northern Swedish population. PLoS ONE. 2011;6(8):e23450. doi: 10.1371/journal.pone.0023450. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Green EK, Grozeva D, Sims R, Raybould R, Forty L, Gordon-Smith K, et al. DISC1 exon 11 rare variants found more commonly in schizoaffective spectrum cases than controls. Am J Med Genet B Neuropsychiatr Genet. 2011;156B(4):490–492. doi: 10.1002/ajmg.b.31187. [DOI] [PubMed] [Google Scholar]
  • 36.Crowley JJ, Hilliard CE, Kim Y, Morgan MB, Lewis LR, Muzny DM, et al. Deep resequencing and association analysis of schizophrenia candidate genes. Mol Psychiatry. 2012 doi: 10.1038/mp.2012.28. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Thomson PA, Parla JS, McRae AF, Kramer M, Ramakrishnan K, Yao J, et al. 708 Common and 2010 rare DISC1 locus variants identified in 1542 subjects: analysis for association with psychiatric disorder and cognitive traits. Mol Psychiatry. 2013 doi: 10.1038/mp.2013.68. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Malavasi EL, Ogawa F, Porteous DJ, Millar JK. DISC1 variants 37W and 607F disrupt its nuclear targeting and regulatory role in ATF4-mediated transcription. Hum Mol Genet. 2012;21 (12):2779–2792. doi: 10.1093/hmg/dds106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Ogawa F, Malavasi EL, Crummie DK, Eykelenboom JE, Soares DC, Mackie S, et al. DISC1 complexes with TRAK1 and Miro1 to modulate anterograde axonal mitochondrial trafficking. Hum Mol Genet. 2013 doi: 10.1093/hmg/ddt485. [epub] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Hyman SE. A glimmer of light for neuropsychiatric disorders. Nature. 2008;455(7215):890–893. doi: 10.1038/nature07454. [DOI] [PubMed] [Google Scholar]
  • 41.Konopka G, Bomar JM, Winden K, Coppola G, Jonsson ZO, Gao F, et al. Human-specific transcriptional regulation of CNS development genes by FOXP2. Nature. 2009;462(7270):213–217. doi: 10.1038/nature08549. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Jacobs PA, Aitken J, Frackiewicz A, Law P, Newton MS, Smith PG. The inheritance of translocations in man: data from families ascertained through a balanced heterozygote. Ann Hum Genet. 1970;34(2):119–136. doi: 10.1111/j.1469-1809.1970.tb00226.x. [DOI] [PubMed] [Google Scholar]

RESOURCES