Psychiatry and Mental Health-Sci Forschen

Full Text

Review Article
Transcriptional Factor 4 Association with Schizophrenia

  Ting Zhang1      Chaoran Yin1      Xiao Xiao1      Daniel R. Marenda2      Wenhui Hu1,*   

1Center for Metabolic Disease Research, Department of Pathology and Laboratory Medicine, Temple University Lewis Katz School of Medicine, Philadelphia, PA 19140, USA
2Department of Biology, Drexel University, Philadelphia, PA, 19104, USA

*Corresponding author: Wenhui Hu, 3500 N Broad Street, MERB, Room 846, Philadelphia, PA 19140, USA, Tel: 215-707-5164; E-mail: whu@temple.edu


Abstract

Schizophrenia (SCZ) is a highly heritable, severe, common psychotic disorder. Genetic variants play an important role in SCZ pathogenesis and progression. Preclinical and clinical findings, particularly in the genome-wide association studies (GWAS), have identified transcriptional factor 4 (TCF4, E2-2) as a high risk gene for SCZ. Disruption of TCF4 expression and function has been also linked to Pitt-Hopkins syndrome (PTHS). TCF4 is well known to regulate neuronal lineage differentiation during embryonic and adult neurogenesis. Recent studies have identified a novel role of Tcf4 in regulating the neurite branching/pruning and synaptic plasticity of postmitotic differentiated neurons. Further understanding of the cellular and molecular mechanisms underlying the neurodevelopmental disorders and neuropsychological/neuropsychiatric diseases associated with TCF4 deficiency or dysfunction will provide important translational targets for the treatment of SCZ and PTHS.

Keywords

Schizophrenia; TCF4; CRISPR-Cas9; Neurons; Neurogenesis

Introduction

Schizophrenia (SCZ) is a major life-long psychiatric disorder, affecting men and women equally in 1% of the general population. The majority of SCZ patients remain ill after the initial episode, suffering from chronic and severely incapacitating symptoms. Genetic studies identified as high as 80% heritability of this complex and devastating mental disease [1-3]. Genetic variants contribute much to the pathogenesis and progression of SCZ. More and more genome-wide association studies (GWAS) have identified transcription factor 4 (TCF4) as a high risk gene for SCZ [2,4- 6]. A CTG triplet repeat expansion in an intronic region of the TCF4 gene is associated with SCZ [7,8]. Six single nucleotide polymorphisms (SNPs) of TCF4 [rs12966547 (G) P=2.6 × 10-10, rs9960767 (C) P=4.1 × 10-9, rs4309482 (A) P=7.8 × 10-9, rs10401120 (T), rs2958182 (T) and rs17512836 (C) P=2.35 × 10-8] have been identified as risk variants for SCZ [9-12]. In the Han Chinese population, three TCF4 SNPs (rs9320010, rs7235757, and rs1452787) are also significantly related to the risk of SCZ [2]. The findings in recent GWAS from psychiatric GWAS consortium (PGC) with 36,989 cases and 113,075 controls further support TCF4’s role in genetic susceptibility to SCZ [13]. However, no significant association between rare TCF4 sequence variants and SCZ is found [14]. Among the TCF4 variants, rs12966547 and rs8766 are associated with the age at onset (AAO), which is a known prognostic indicator for SCZ [11]. Disruption of TCF4 expression and function has been also linked to several other common and rare diseases such as PittHopkins syndrome (PTHS), Fuchs’ endothelial corneal dystrophy (FECD) [15,16], and primary sclerosing cholangitis (PSC) [6,17]. In particular, PTHS is well known to be caused by TCF4 haploinsufficiency [18-20], and is characterized with intellectual disability, autism, epileptic seizures, absent speech, hyperventilation and gut dysfunction [21-23]. Therefore, targeting TCF4 by modulating its expression/function might be a highly promising therapeutic to treat these already-diagnosed patients with TCF4 mutations or deficiency.

Extensive Function of TCF4, Particularly in Neurogenesis and Neuronal Polarization

TCF4 is also known as E2-2; ITF2; PTHS; SEF2; ITF-2; SEF-2; SEF2- 1; SEF2-1A; SEF2-1B; SEF2-1D; bHLHb19 (http://www.ncbi.nlm.nih. gov/gene/6925). TCF4 should not be confused with the T-cell factor 4 that regulates Wnt signaling and also associates with SCZ [24,25]. TCF4 is a member of class I basic helix-loop-helix (bHLH) transcription factors. The bHLH family, known as E-proteins due to their binding to Ephrussi box (CANNTG) [18,19], play critical roles in a huge array of cellular processes and organogenesis. TCF4 is extensively expressed in many tissues but enriched in neurogenic niche during embryonic, postnatal and adult neurogenesis [6]. Among the bHLH factors, only TCF4 is continuously expressed in the adult nervous system in both animals and humans [6]. Generally, TCF4 heterodimerizes with class II bHLH proteins, particularly the proneuronal factors such as NeuroD, Neurogenin, ATOH1/MATH1, ASCL1/MASH1 to regulate neurogenesis and neuronal lineage differentiation [26]. TCF4 homodimerizes [27] to regulate gene expression [28,29]. TCF4 also heterodimerizes with class V bHLH proteins to inhibit gene expression [30,31]. These findings suggest that TCF4 plays an important role in regulating embryonic and adult neurogenesis and synaptic plasticity. TCF4 is also involved in lymphoid development [32], epithelial-mesenchymal transition [33,34], ocular growth [35], T cell differentiation [36] and dendritic cell development [37,38].

The expression of class I/II bHLH proteins is required to initiate neuronal differentiation of neural stem cell [39]. However, the class II bHLH proteins play a role in regulating the function of terminally differentiated neural cells. For example, in Drosophila, the class II bHLH protein Atonal functions to promote neuronal migration, axon guidance and arborization [40]. In mammals, the Atonal homolog Atoh1/Math1 is essential for migration of postmitotic retrotrapezoid nucleus neurons required for proper respiration [41]. Several class I bHLH proteins are expressed in postmitotic neurons in C. elegans [42], mice [43], humans [43], suggesting that class I bHLH proteins may also play an important role in postmitotic neurons. The daughterless (Da) gene encodes the only type I bHLH protein found in Drosophila [44]. Similar to TCF4, the Da protein is expressed in a large number of tissues and is involved in a diverse number of developmental processes including neurogenesis [28,45], cell proliferation [46,47], muscle development [48], ovarian development [49], and retinal development [47,50]. Over expression of Da in mitotic epithelial cells is sufficient to arrest these cells in G2 phase of the cell cycle [51]. Our recent studies show that Da is expressed in postmitotic neurons of the fly neuromuscular junction, and functions to restrict axonal arborization (bouton), while Tcf4 is also expressed in mouse postmitotic neurons and functions to restrict neurite branching [52]. However, more experimental evidence is warranted to support the importance of Tcf4/ Da in neuronal polarization, in particular the presynaptic (axonal) vs the postsynaptic (dendritic) branching/pruning.

Tcf4 Binding Partners and Downstream Target Genes in Postmitotic Neurons

In addition to binding to the target DNA, TCF4 binds to many proteins, particularly bHLH family of transcription factors. Its homodimerization or heterodimerization with binding partners contributes to its action in regulating the expression of target genes [26,28,29]. Bioinformatics analysis identified thousands of downstream target genes that are regulated by TCF4/Da in mitotically active cells [33,53]. For example, TCF4 binds to proneuronal proteins such as NeuroD, neurogenin, Mash1, Math1 to regulate neurogenesis and neuronal lineage differentiation [54- 56]. Neurexin 1 (Nrxn1) is emerging as susceptibility factor for SCZ and its rare copy number variants are found to contribute to the pathogenesis of SCZ [57,58]. Deletions within the Nrxn1 gene affecting exons confer SCZ risk and are associated with autism and mental retardation [59,60]. TCF4 directly binds to Nrxn1 promoter/enhancer region in mature mammalian neurons to repress its transcription [52]. In Drosophila, Da forms homodimers which mediate synapse restriction via inhibition of Nrxn1 expression [52]. Thus, Nrxn1 may partially mediate TCF4-related phenotypes for SCZ. The variants in miR-137 gene are significantly associated with SCZ. SCZ patients with homozygous miR-137 risk allele display significant decreases in occipital, parietal and temporal lobe gray matter concentration [61]. Dysregulation of miR-137-regulated genes such as TCF4, PTGS2, MAPK1 and MAPK3 may underlie the gray matter loss seen in SCZ patients [61]. TCF4 acts as a regulator of neuronal intrinsic excitability by suppressing two ion channel genes, Kcnq1 and Scn10a [62]. The downstream effectors of TCF4 pathway are essential for understanding the molecular mechanisms underlying TCF4-associated diseases. Altering the expression or function of TCF4 downstream effectors may improve the cognitive in SCZ patients. Potential therapeutic chemical/natural compound(s) could be screened in human cell model or transgenic animal models to control the expression of TCF4 and/or target genes [63]. Recently, HDAC inhibitor treatment in mouse model has been shown to rescue learning and memory deficits caused by loss of TCF4 function [64]. To develop valid therapeutics, controlling specific genes’ expression is pivotal.

The Potential Therapeutic Strategies and CRIPSR-Cas9 Technology

So far, no effective treatment is available for TCF4-associated diseases although different therapeutics has been used to improve the SCZ symptoms, such as antipsychotic prescription and psychosocial intervention [65-67]. It is still a big challenge to develop new effective therapy for SCZ because the etiology and pathogenesis of SCZ remain largely unclear. Many transcripts/isoforms exist in both human TCF4 and mouse Tcf4. Different types of cells (particularly subtypes of neurons) may express different isoforms at various levels. Various types of SNPs may contribute to SCZ development in a different way. Therefore, isoform specific and/or cell-specific manipulation of TCF4 expression and/ or gene mutation correction would provide a novel therapy for TCF4- assocaited diseases. Spatiotemporal control of endogenous non-mutant TCF4 in attempt to restore the “normal” expression/function of TCF4 may be also an interesting therapeutic strategy for patients with TCF4 haploinsufficiency.

The RNA-guided endonuclease CRISPR-Cas9 technology has emerged as a simpler and more versatile technology to target and modify any genomic sequence with high levels of efficacy and specificity [68]. Successful application of Cas9 technology to mammalian system for genome editing was first reported in early 2013 [69,70]. Since then, this novel genome editing system has attracted a huge amount of attention in the biomedical field. In particular, extensive preclinical examples have been seen in the fields of animal models, genetic diseases, cancer biology and infectious diseases [71-75]. Efficient Cas9-mediated transgene knockin has been recently reported in various cell lines [76-81] and different species including mice [82,83], rats [84,85], pigs [86,87], zebrafish [81,88,89], etc. Simultaneously, the use of the catalytically-deficient Cas9 (dCas9) conjugated with a single transcriptional activator or repressor to manipulate cellular gene regulation has been developed [90-93]. This single regulator system has its limitations, such as effectiveness of gene activation or repression and scalability. Thus, recruitment of multiplex transcriptional activators or repressors through guide RNA (gRNA) modification and/or dCas9 fusion has been explored [94-99]. For example, dCas9-based synergistic activation mediator (SAM) system has been developed by engineering the single gRNA (sgRNA) through appending a minimal hairpin aptamer to the tetraloop and stem loop 2 of sgRNA [100]. Such aptamer is capable of binding to the dimerized MS2 bacteriophage coat proteins. Thus, a novel MS2-p65-HSF1 complex guided by targetspecific MS2-mediated sgRNA (msgRNAs) could facilitate the potency of dCas9-mediated gene activation by up to 3,000 fold [100]. This dCas9- SAM technology is capable of activating the provirus in HIV-1 latent cells for the “shock and kill” strategy to cure HIV/AIDS [101]. Similarly, a dCas9-engineered transcriptional repressor (ETR) system that combines several epigenome repressors has been established to achieve long-term suppression of endogenous genes [102]. The dCas9 system relies on target-specific sgRNAs and delivers multiple exogenous transcriptional activators or repressors to the target site. Furthermore, the dCas9 does not keep nuclease activity, and never induce any DNA mutation or chromosome translocations in host cells. Therefore this dCas9 system has high specificity, high efficiency and no/low cytotoxicity.

Most exciting application of Cas9 technology is the genetic correction [103-105] and antiviral treatment in animals [106,107] and likely soon in the clinic. Clinical trials for Cas9-mediated cancer targeting have been initiated [108,109]. The proof of concept to apply the Cas9 and/or dCas9 genome/epigenome editing technology to the manipulation of SCZ risk genes has been proposed in several reviews [110-112]. Experimental evidence to target SCZ-derived induced pluripotent stem (iPS) cells has been reported recently [113,114]. The effective knockout of Tcf4 by Cas9/sgRNA in embryonic neural cells via in-utero electroporation has been shown to alter the intrinsic excitability of prefrontal neurons [62]. However, it is still a long journey to unlock genetics of SCZ diseases using CRISPR-Cas9 technology.

The spatiotemporal expression of the TCF4 gene is extraordinarily intricate and difficult to study because (1) There are many splicing events and variant transcripts of TCF4 in both human and animals, with 30 exons in mouse Tcf4; (2) No good antibody against TCF4 (and particularly its various isoforms) is currently available; (3) Traditional knockin tagging (reporter) modeling is time-consuming and insufficient. Even with a good antibody, the immunohistochemistry study cannot detect the dynamic and quantitative changes in TCF4 protein. TCF4 promoter-driven reporter assay may address the transcriptional regulation of different TCF4 transcripts in human and animals, but this in vitro assay is artificial and does not measure the spatiotemporal expression of endogenous TCF4, rather than various isoforms. Therefore, CRIPSR-Cas9 mediated reporter knockin technology may provide a novel approach to manipulate the spatiotemporal expression of the endogenous TCF4 gene in cultured cells (in vitro) or transgenic animal models (in vivo). Many drugs have been approved by FDA for their safety and pharmacokinetic profiles in patients. In addition, the manufacturing and distribution networks are readily available. Therefore, repurposing existing drugs for novel unanticipated function would be a rapid and effective way to develop potential new therapy for neurodevelopmental disorders and neuropsychological/ neuropsychiatric diseases. The highly sensitive bioluminescence assay in endogenous TCF4 reporter knockin cells may provide an excellent high throughput screening for FDA-approved drugs. By screening, we may identify potential FDA-approved drugs that specifically and efficiently upregulate or downregulate the expression of endogenous TCF4 in human iPS-derived cells or animal models.

Conclusion

In the context of all the TCF4-associated diseases, the critical question is which copy, the insufficient normal TCF4 or the over expressed mutant TCF4, is the de novo cause to the development and progression of these diseases. Therefore, better understanding the expression pattern and different function of the mutant and non-mutant TCF4 in the nervous system, cornea and liver is fundamental to the development of novel therapeutic treatment for these debilitating diseases. The causative effect of the haploinsufficient normal TCF4 and the mutant TCF4 due to SNP variants, triplet repeat or deletion has yet to be characterized. Developing a novel dual reporter disease model by targeting endogenous normal and mutant copies of TCF4 might be an important future direction. We expect to see that manipulation of insufficient normal TCF4 expression may counteract the mutant TCF4 to achieve sufficient normal function.

Acknowledgement

This work was partially supported by a grant from the Pitt Hopkins Research Foundation and the Orphan Disease Center at the Perelman School of Medicine at the University of Pennsylvania (to DRM and WH).

References

  1. Aberg KA, Liu Y, Bukszar J, McClay JL, Khachane AN, et al. (2013) A comprehensive family-based replication study of schizophrenia genes. JAMA Psychiatry 70: 573-581. [Ref.]
  2. Li J, Chen Z, Wang F, Ouyang Y, Zhang N, et al. (2016) Polymorphisms of the TCF4 gene are associated with the risk of schizophrenia in the Han Chinese. Am J Med Genet B Neuropsychiatr Genet 171: 1006-1012. [Ref.]
  3. Nieratschker V, Nöthen MM, Rietschel M (2010) New Genetic Findings in Schizophrenia: Is there Still Room for the Dopamine Hypothesis of Schizophrenia? Front Behav Neurosci 4:23. [Ref.]
  4. Dong J, Wyss A, Yang J, Price T R, Nicolas A, et al. (2016) GenomeWide Association Analysis of the Sense of Smell in U.S. Older Adults: Identification of Novel Risk Loci in African-Americans and EuropeanAmericans. Mol Neurobiol. [Ref.]
  5. Gurung R, Prata DP (2015) What is the impact of genome-wide supported risk variants for schizophrenia and bipolar disorder on brain structure and function? A systematic review. Psychol Med 45: 2461-2480. [Ref.]
  6. Quednow BB, Brzózka MM (2014) Rossner MJ Transcription factor 4 (TCF4) and schizophrenia: integrating the animal and the human perspective. Cell Mol Life Sci 71: 2815-2835. [Ref.]
  7. Fortune MT, Kennedy JL, Vincent JB (2003) Anticipation and CAG*CTG repeat expansion in schizophrenia and bipolar affective disorder. Curr Psychiatry Rep 5: 145-154. [Ref.]
  8. McInnis MG, Swift-Scanlanl T, Mahoney AT, Vincent J, Verheyen G, et al. (2000) Allelic distribution of CTG18.1 in Caucasian populations: association studies in bipolar disorder, schizophrenia, and ataxia. Mol Psychiatry 5: 439-442. [Ref.]
  9. Wirgenes KV, Sønderby IE, Haukvik UK, Mattingsdal M, Tesli M, et al. (2012) TCF4 sequence variants and mRNA levels are associated with neurodevelopmental characteristics in psychotic disorders. Transl Psychiatry 2: e112. [Ref.]
  10. Hui L, Rao WW, Yu Q, Kou C, Wu JQ, et al. (2015) TCF4 gene polymorphism is associated with cognition in patients with schizophrenia and healthy controls. J Psychiatr Res 69: 95-101. [Ref.]
  11. Chow TJ, Tee SF, Yong HS, Tang PY (2016) Genetic Association of TCF4 and AKT1 Gene Variants with the Age at Onset of Schizophrenia. Neuropsychobiology 73: 233-240. [Ref.]
  12. Quednow BB, Brinkmeyer J, Mobascher A, Nothnagel M, Musso F, et al. (2012) Schizophrenia risk polymorphisms in the TCF4 gene interact with smoking in the modulation of auditory sensory gating. Proc Natl Acad Sci U S A 109: 6271-6276. [Ref.]
  13. Schizophrenia Working Group of the Psychiatric Genomics Consortium (2014) Biological insights from 108 schizophrenia-associated genetic loci. Nature 511: 421-427. [Ref.]
  14. Basmanav FB, Forstner AJ, Fier H, Herms S, Meier S, et al. (2015) Investigation of the role of TCF4 rare sequence variants in schizophrenia. Am J Med Genet B Neuropsychiatr Genet 168B: 354-362. [Ref.]
  15. Oldak M, Ruszkowska E, Udziela M, Ozieblo D, Binczyk E, et al. (2015) Fuchs Endothelial Corneal Dystrophy: Strong Association with rs613872 Not Paralleled by Changes in Corneal Endothelial TCF4 mRNA Level. Biomed Res Int 640234. [Ref.]
  16. Wieben ED, Aleff RA, Tosakulwong N, Butz ML, Highsmith WE, (2012) A common trinucleotide repeat expansion within the transcription factor 4 (TCF4, E2-2) gene predicts Fuchs corneal dystrophy. PLoS One 7: e49083. [Ref.]
  17. Forrest MP, Hill MJ, Quantock AJ, Martin-Rendon E, Blake DJ (2014) The emerging roles of TCF4 in disease and development. Trends Mol Med 20: 322-331. [Ref.]
  18. Zweier C, Peippo MM, Hoyer J, Sousa S, Bottani A, et al. (2007) Haploinsufficiency of TCF4 causes syndromal mental retardation with intermittent hyperventilation (Pitt-Hopkins syndrome). Am J Hum Genet 80: 994-1001. [Ref.]
  19. Amiel J, Rio M, de Pontual L, Redon R, Malan V, et al. (2007) Mutations in TCF4, encoding a class I basic helix-loop-helix transcription factor, are responsible for Pitt-Hopkins syndrome, a severe epileptic encephalopathy associated with autonomic dysfunction. Am J Hum Genet 80: 988-993. [Ref.]
  20. Brockschmidt A, Todt U, Ryu S, Hoischen A, Landwehr C, et al. (2007) Severe mental retardation with breathing abnormalities (Pitt-Hopkins syndrome) is caused by haploinsufficiency of the neuronal bHLH transcription factor TCF4. Hum Mol Genet 16:1488-1494.
  21. Grubisic V, Kennedy AJ, Sweatt JD, ParpuraV (2015) Pitt-Hopkins Mouse Model has Altered Particular Gastrointestinal Transits In Vivo. Autism Res 8: 629-633 [Ref.]
  22. Kinsley L, Siddique T (1993) Amyotrophic Lateral Sclerosis Overview. In: Pagon RA, Adam MP, Ardinger HH, Wallace SE, Amemiya A, et al. (eds.), GeneReviews(R), Seattle (WA) USA. [Ref.]
  23. Peippo M, Ignatius J (2012) Pitt-Hopkins Syndrome. Mol Syndromol 2: 171-180.
  24. Alkelai A, Greenbaum L, Lupoli S, Kohn Y, Sarner-Kanyas K, et al. (2012) Association of the type 2 diabetes mellitus susceptibility gene, TCF7L2, with schizophrenia in an Arab-Israeli family sample. PLoS One 7: e29228. [Ref.]
  25. Hansen T, Ingason A, Djurovic S, Melle I, Fenger M, et al. (2011) At-risk variant in TCF7L2 for type II diabetes increases risk of schizophrenia. Biol Psychiatry 70: 59-63. [Ref.]
  26. Powell LM, Jarman AP (2008) Context dependence of proneural bHLH proteins. Curr Opin Genet Dev 18: 411-417. [Ref.]
  27. Cabrer CV, Alonso MC (1991) Transcriptional activation by heterodimers of the achaete-scute and daughterless gene products of Drosophila. EMBO J 10: 2965-2973. [Ref.]
  28. Lim J, Jafar-Nejad H, Hsu YC , Choi KW (2008) Novel function of the class I bHLH protein Daughterless in the negative regulation of proneural gene expression in the Drosophila eye. EMBO Rep 9: 1128-1133. [Ref.]
  29. Tanaka-Matakatsu M, Miller J, Borger D, Tang WJ, Du W (2014) Daughterless homodimer synergizes with Eyeless to induce Atonal expression and retinal neuron differentiation. Dev Biol 392: 256-265. [Ref.]
  30. Quednow BB, Brzozka MM, Rossner MJ (2014) Transcription factor 4 (TCF4) and schizophrenia: integrating the animal and the human perspective. Cell Mol Life Sci 71: 2815-2835. [Ref.]
  31. Massari ME, Murre C (2000) Helix-loop-helix proteins: regulators of transcription in eucaryotic organisms. Mol Cell Biol 20: 429-440. [Ref.]
  32. Quong MW, Romanow W J, Murre C (2002) E protein function in lymphocyte development. Annu Rev Immunol 20: 301-322. [Ref.]
  33. Forrest MP, Waite AJ, Martin-Rendon E, Blake DJ (2013) Knockdown of human TCF4 affects multiple signaling pathways involved in cell survival, epithelial to mesenchymal transition and neuronal differentiation. PLoS One 8: e73169. [Ref.]
  34. Cano A, Portillo F (2010) An emerging role for class I bHLH E2-2 proteins in EMT regulation and tumor progression. Cell Adh Migr 4: 56-60. [Ref.]
  35. Brockschmidt A, Filippi A, Charbel Issa P, Nelles M, Urbach H, (2011) Neurologic and ocular phenotype in Pitt-Hopkins syndrome and a zebrafish model. Hum Genet 130: 645-655. [Ref.]
  36. Bergqvist I, Eriksson M, Saarikettu J, Eriksson B, Corneliussen B, (2000) The basic helix-loop-helix transcription factor E2-2 is involved in T lymphocyte development. Eur J Immunol 30: 2857-2863. [Ref.]
  37. Onai N, Kurabayashi K, Hosoi-Amaike M, Toyama-Sorimachi N, Matsushima K, et al. (2013) A clonogenic progenitor with prominent plasmacytoid dendritic cell developmental potential. Immunity 38: 943-957. [Ref.]
  38. Kneissl S, Abel T, Rasbach A, Brynza J, Schneider-Schaulies J (2012) Measles virus glycoprotein-based lentiviral targeting vectors that avoid neutralizing antibodies. PLoS One 7: e46667. [Ref.]
  39. Guillemot F (2007) Spatial and temporal specification of neural fates by transcription factor codes. Development 134: 3771-3780. [Ref.]
  40. Hassan BA, Bermingham NA, He Y, Sun Y, Jan YN, et al. (2000) atonal regulates neurite arborization but does not act as a proneural gene in the Drosophila brain. Neuron 25: 549-561. [Ref.]
  41. Huang WH, Tupal S, Huang TW, Ward CS, Neul JL, et al. (2012) Atoh1 governs the migration of postmitotic neurons that shape respiratory effectiveness at birth and chemoresponsiveness in adulthood. Neuron 75: 799-809. [Ref.]
  42. Krause M, Park M, Zhang JM, Yuan J, Harfe B, et al. (1997) A C. elegans E/Daughterless bHLH protein marks neuronal but not striated muscle development. Development 124: 2179-2189. [Ref.]
  43. Sepp M, Kannike K, Eesmaa A, Urb M, Timmusk T (2011) Functional diversity of human basic helix-loop-helix transcription factor TCF4 isoforms generated by alternative 5’ exon usage and splicing. PLoS One e22138. [Ref.]
  44. Caudy M, Vässin H, Brand M, Tuma R, Jan LY, et al. (1988) daughterless, a Drosophila gene essential for both neurogenesis and sex determination, has sequence similarities to myc and the achaetescute complex. Cell 55: 1061-1067. [Ref.]
  45. Jafar-Nejad H, Tien AC, Acar M, Bellen HJ (2006) Senseless and Daughterless confer neuronal identity to epithelial cells in the Drosophila wing margin. Development 133: 1683-1692. [Ref.]
  46. Smith JE, Cummings CA, Cronmiller C (2002) Daughterless coordinates somatic cell proliferation, differentiation and germline cyst survival during follicle formation in Drosophila. Development 129: 3255-3267. [Ref.]
  47. Brown NL, Paddock SW, Sattler CA, Cronmiller C, Thomas BJ (1996) daughterless is required for Drosophila photoreceptor cell determination, eye morphogenesis, and cell cycle progression. Dev Biol 179: 65-78. [Ref.]
  48. . Castanon I, Von Stetina S, Kass J, Baylies MK (2001) Dimerization partners determine the activity of the Twist bHLH protein during Drosophila mesoderm development. Development 128: 3145-3159. [Ref.]
  49. Cummings CA, Cronmiller C (1994) The daughterless gene functions together with Notch and Delta in the control of ovarian follicle development in Drosophila. Development 120: 381-394. [Ref.]
  50. Melicharek D, Shah A, DiStefano G, Gangemi AJ, Orapallo A, et al. (2008) Identification of novel regulators of atonal expression in the developing Drosophila retina. Genetics 180: 2095-2110. [Ref.]
  51. Andrade-Zapata I, Baonza A (2014) The bHLH factors extramacrochaetae and daughterless control cell cycle in Drosophila imaginal discs through the transcriptional regulation of the Cdc25 phosphatase string. PLoS genetics e1004233. [Ref.]
  52. D’Rozario M, Zhang T, Waddell EA, Zhang Y, Sahin C, et al. (2016) Type I bHLH Proteins Daughterless and Tcf4 Restrict Neurite Branching and Synapse Formation by Repressing Neurexin in Postmitotic Neurons. Cell Rep 15: 386-397. [Ref.]
  53. MacArthur S, Li XY, Li J, Brown JB, Chu HC, et al. (2009) Developmental roles of 21 Drosophila transcription factors are determined by quantitative differences in binding to an overlapping set of thousands of genomic regions. Genome Biol 10: R80. [Ref.]
  54. Navarrete K, Pedroso I, De Jong S, Stefansson H, Steinberg S, et al (2013) TCF4 (e2-2; ITF2): a schizophrenia-associated gene with pleiotropic effects on human disease.Am J Med Genet B Neuropsychiatr Genet 162B: 1-16. [Ref.]
  55. Fischer B, Azim K, Hurtado-Chong A, Ramelli S, Fernández M, et al. (2014) E-proteins orchestrate the progression of neural stem cell differentiation in the postnatal forebrain. Neural Dev 9: 23. [Ref.]
  56. Brzózka MM, Radyushkin K, Wichert SP, Ehrenreich H, Rossner MJ (2010) Cognitive and sensorimotor gating impairments in transgenic mice overexpressing the schizophrenia susceptibility gene Tcf4 in the brain. Biol Psychiatry 68: 33-40. [Ref.]
  57. Rujescu D, Ingason A, Cichon S, Pietiläinen OP, Barnes MR, et al. (2009) Disruption of the neurexin 1 gene is associated with schizophrenia. Hum Mol Genet 18: 988-996. [Ref.]
  58. Kirov G, Grozeva D, Norton N, Ivanov D, Mantripragada KK, et al. (2009) Support for the involvement of large copy number variants in the pathogenesis of schizophrenia. Hum Mol Genet 18: 1497-1503. [Ref.]
  59. Kirov G, Gumus D, Chen W, Norton N, Georgieva L, et al. (2008) Comparative genome hybridization suggests a role for NRXN1 and APBA2 in schizophrenia. Hum Mol Genet 17: 458-465. [Ref.]
  60. Kim HG, Kishikawa S, Higgins AW, Seong IS, Donovan DJ, et al. (2008) Disruption of neurexin 1 associated with autism spectrum disorder. Am J Hum Genet 82: 199-207. [Ref.]
  61. Wright C, Gupta CN, Chen J, Patel V, Calhoun VD, et al. (2016) Polymorphisms in MIR137HG and microRNA-137-regulated genes influence gray matter structure in schizophrenia. Transl Psychiatry 6: e724. [Ref.]
  62. Rannals MD, Hamersky GR, Page SC, Campbell MN, Briley A, et al. (2016) Psychiatric Risk Gene Transcription Factor 4 Regulates Intrinsic Excitability of Prefrontal Neurons via Repression of SCN10a and KCNQ1. Neuron 90: 43-55. [Ref.]
  63. Sweatt JD (2013) Pitt-Hopkins Syndrome: intellectual disability due to loss of TCF4-regulated gene transcription. Exp Mol Med 45: e21. [Ref.]
  64. Kennedy AJ, Rahn EJ, Paulukaitis BS, Savell KE, Kordasiewicz HB, et al. (2016) Tcf4 Regulates Synaptic Plasticity, DNA Methylation, and Memory Function. Cell Rep 16: 2666-2685. [Ref.]
  65. Khan AY, Salaria S, Ovais M and Ide GD (2016) Depot antipsychotics: Where do we stand? Ann Clin Psychiatry 28: 289-298. [Ref.]
  66. Catts SV and O’Toole BI (2016) The treatment of schizophrenia: Can we raise the standard of care? Aust N Z J Psychiatry 50: 1128-1138. [Ref.]
  67. Gopalakrishna G, Ithman MH and Lauriello J (2016) Update on New and Emerging Treatments for Schizophrenia. Psychiatr Clin North Am 39: 217-238. [Ref.]
  68. Hsu PD, Lander ES and Zhang F (2014) Development and applications of CRISPR-Cas9 for genome engineering. Cell 157: 1262-1278. [Ref.]
  69. Cong L, Ran FA, Cox D, Lin S, Barretto R, et al. (2013) Multiplex genome engineering using CRISPR/Cas systems. Science 339: 819-823. [Ref.]
  70. Mali P, Yang L, Esvelt KM, Aach J, Guell M, et al. (2013) RNA-guided human genome engineering via Cas9. Science 339: 823-826. [Ref.]
  71. Sander JD, Joung JK (2014) CRISPR-Cas systems for editing, regulating and targeting genomes. Nat Biotechnol 32: 347-355. [Ref.]
  72. Vasileva EA, Shuvalov OU, Garabadgiu AV, Melino G and Barlev NA (2015) Genome-editing tools for stem cell biology. Cell death & disease 6: e1831. [Ref.]
  73. Sanchez-Rivera FJ, Jacks T (2015) Applications of the CRISPR-Cas9 system in cancer biology. Nat Rev Cancer 15: 387-395. [Ref.]
  74. Riordan SM, Heruth DP, Zhang LQ, Ye SQ (2015) Application of CRISPR/Cas9 for biomedical discoveries. Cell Biosci 5: 33. [Ref.]
  75. Saayman S, Ali SA, Morris KV, Weinberg MS (2015) The therapeutic application of CRISPR/Cas9 technologies for HIV. Expert Opin Biol Ther 15: 819-830. [Ref.]
  76. Mandai S, Mori T, Sohara E, Rai T, Uchida S (2015) Generation of Hypertension-Associated STK39 Polymorphism Knockin Cell Lines With the Clustered Regularly Interspaced Short Palindromic Repeats/ Cas9 System. Hypertension 66: 1199-1206. [Ref.]
  77. Li S, Xue H, Wu J, Rao MS, Kim DH, et al. (2015) Human induced pluripotent stem cell NEUROG2 dual knockin reporter lines generated by the CRISPR/Cas9 system. Stem Cells Dev 24: 2925-2942. [Ref.]
  78. Li S, Xue H, Long B, Sun L, Truong T et al. (2015) Efficient generation of hiPSC neural lineage specific knockin reporters using the CRISPR/ Cas9 and Cas9 double nickase system. J Vis Exp e52539. [Ref.]
  79. Merkle FT, Neuhausser WM, Santos D, Valen E, Gagnon JA, et al. (2015) Efficient CRISPR-Cas9-mediated generation of knockin human pluripotent stem cells lacking undesired mutations at the targeted locus. Cell Rep 11: 875-883.[Ref.]
  80. Ratz M, Testa I, Hell SW, Jakobs S (2015) CRISPR/Cas9-mediated endogenous protein tagging for RESOLFT super-resolution microscopy of living human cells. Scientific Reports 5: 9592. [Ref.]
  81. Li J, Zhang BB, Ren YG, Gu SY, Xiang YH, et al. (2015) Intron targeting-mediated and endogenous gene integrity-maintaining knockin in zebrafish using the CRISPR/Cas9 system. Cell Res 25: 634-637. [Ref.]
  82. Platt RJ, Chen S, Zhou Y, Yim MJ, Swiech L, et al. (2014) CRISPRCas9 Knockin Mice for Genome Editing and Cancer Modeling. Cell 159: 440-455. [Ref.]
  83. Fujihara Y, Ikawa M (2014) CRISPR/Cas9-based genome editing in mice by single plasmid injection. Methods Enzymol 546: 319-36. [Ref.]
  84. Ma Y, Ma J, Zhang X, Chen W, Yu L, et al. (2014) Generation of eGFP and Cre knockin rats by CRISPR/Cas9. The FEBS journal 281: 3779-3790. [Ref.]
  85. Guan Y, Shao Y, Li D, Liu M (2014) Generation of site-specific mutations in the rat genome via CRISPR/Cas9. Methods Enzymol 546: 297-317. [Ref.]
  86. Wang X, Zhou J, Cao C, Huang J, Hai T, et al. (2015) Efficient CRISPR/ Cas9-mediated biallelic gene disruption and site-specific knockin after rapid selection of highly active sgRNAs in pigs. Sci Rep 5: 13348. [Ref.]
  87. . Ruan J, Li H, Xu K, Wu T, Wei J, et al. (2015) Highly efficient CRISPR/ Cas9-mediated transgene knockin at the H11 locus in pigs. Sci Rep 5: 14253. [Ref.]
  88. Li J, Zhang B, Bu J, Du J (2015) Intron-based genomic editing: a highly efficient method for generating knockin zebrafish. Oncotarget 6: 17891-17894. [Ref.]
  89. Dong Z, Dong X, Jia W, Cao S, Zhao Q (2014) Improving the efficiency for generation of genome-edited zebrafish by labeling primordial germ cells. Int J Biochem Cell Biol 55: 329-334. [Ref.]
  90. Agne M, Blank I, Emhardt AJ, Gäbelein CG, Gawlas F, et al. (2014) Modularized CRISPR/dCas9 effector toolkit for target-specific gene regulation. ACS Synth Biol 3: 986-989. [Ref.]
  91. Maeder ML, Linder SJ, Cascio VM, Fu Y, Ho QH, et al. (2013) CRISPR RNA-guided activation of endogenous human genes. Nature methods 10: 977-979. [Ref.]
  92. Gilbert LA, Larson MH, Morsut L, Liu Z, Brar GA, et al. (2013) CRISPR-mediated modular RNA-guided regulation of transcription in eukaryotes. Cell 154: 442-451. [Ref.]
  93. Cheng AW, Wang H, Yang H, Shi L, Katz Y, et al. (2013) Multiplexed activation of endogenous genes by CRISPR-on, an RNA-guided transcriptional activator system. Cell Res 23: 1163-1171. [Ref.]
  94. Heckl D, Charpentier E (2015) Toward Whole-Transcriptome Editing with CRISPR-Cas9. Mol Cell 58: 560-562. [Ref.]
  95. Zalatan JG, Lee ME, Almeida R, Gilbert LA, Whitehead EH, et al. (2015) Engineering complex synthetic transcriptional programs with CRISPR RNA scaffolds. Cell 160: 339-350. [Ref.]
  96. Tanenbaum ME, Gilbert LA, Qi LS, Weissman JS, Vale RD (2014) A protein-tagging system for signal amplification in gene expression and fluorescence imaging. Cell 159: 635-646. [Ref.]
  97. Liu XS, Wu H, Ji X, Stelzer Y, Wu X, et al. (2016) Editing DNA Methylation in the Mammalian Genome. Cell 167, 233-247 e217. [Ref.]
  98. McDonald JI, Celik H, Rois LE, Fishberger G, Fowler T, et al. (2016) Reprogrammable CRISPR/Cas9-based system for inducing sitespecific DNA methylation. Biol Open 5: 866-874. [Ref.]
  99. Vojta A, Dobrinic P, Tadic V, Bockor L, Korac P, et al. (2016) Repurposing the CRISPR-Cas9 system for targeted DNA methylation. Nucleic Acids Res 44: 5615-5628. [Ref.]
  100. Konermann S, Brigham MD, Trevino AE, Joung J, Abudayyeh OO, et al. (2015) Genome-scale transcriptional activation by an engineered CRISPR-Cas9 complex. Nature 517: 583-588. [Ref.]
  101. . Zhang Y, Yin C, Zhang T, Li F, Yang W, et al. (2015) CRISPR/gRNAdirected synergistic activation mediator (SAM) induces specific, persistent and robust reactivation of the HIV-1 latent reservoirs. Sci Rep 5: 16277. [Ref.]
  102. Amabile A, Migliara A, Capasso P, Biffi M, Cittaro D, et al. (2016) Inheritable Silencing of Endogenous Genes by Hit-and-Run Targeted Epigenetic Editing. Cell 167: 219-232.e14. [Ref.]
  103. Himeda CL, Jones TI, Jones PL (2016) Scalpel or Straitjacket: CRISPR/Cas9 Approaches for Muscular Dystrophies. Trends Pharmacol Sci 37: 249-251. [Ref.]
  104. Yang Y, Wang L, Bell P, McMenamin D, He Z, et al. (2016) A dual AAV system enables the Cas9-mediated correction of a metabolic liver disease in newborn mice. Nat Biotechnol 34: 334-338. [Ref.]
  105. Chengzu Long, John R McAnally, John M Shelton, Alex A Mireault, Rhonda Bassel Duby, et al. (2014) Prevention of muscular dystrophy in mice by CRISPR/Cas9-mediated editing of germline DNA. Science 345: 1184-1188. [Ref.]
  106. Price AA, Grakoui A, Weiss DS (2016) Harnessing the Prokaryotic Adaptive Immune System as a Eukaryotic Antiviral Defense. Trends Microbiol 24: 294-306. [Ref.]
  107. Kaminski R, Bella R, Yin C, Otte J, Ferrante P, et al.(2016) Excision of HIV-1 DNA by gene editing: a proof-of-concept in vivo study. Gene Ther 23: 690-695. [Ref.]
  108. Barrangou R, Doudna JA (2016) Applications of CRISPR technologies in research and beyond. Nat Biotechnol 34: 933-941. [Ref.]
  109. Cyranoski D (2016) CRISPR gene-editing tested in a person for the first time. Nature 539: 479. [Ref.]
  110. Haggarty SJ, Silva MC, Cross A, Brandon NJ, Perlis RH (2016) Advancing drug discovery for neuropsychiatric disorders using patient-specific stem cell models. Mol Cell Neurosci 73: 104-115. [Ref.]
  111. Flaherty EK, Brennand KJ (2017) Using hiPSCs to model neuropsychiatric copy number variations (CNVs) has potential to reveal underlying disease mechanisms. Brain Res 1655: 283-293. [Ref.]
  112. Duan J (2015) Path from schizophrenia genomics to biology: gene regulation and perturbation in neurons derived from induced pluripotent stem cells and genome editing. Neurosci Bull 31: 113-27. [Ref.]
  113. Pham X, Song G, Lao S, Goff L, Zhu H, et al. (2016) The DPYSL2 gene connects mTOR and schizophrenia. Transl Psychiatry 6: e933. [Ref.]
  114. Srikanth P, Han K, Callahan DG, Makovkina E, Muratore CR, et al. (2015) Genomic DISC1 Disruption in hiPSCs Alters Wnt Signaling and Neural Cell Fate. Cell Rep 12: 1414-1429. [Ref.]

Download Provisional PDF Here

 

Article Information

Article Type: Review Article

Citation: Zhang T, Yin C, Xiao X, Marenda DR, Hu W (2017) Transcriptional Factor 4 Association with Schizophrenia. J Psychiatry Ment Health 2(1): doi http://dx.doi.org/10.16966/2474-7769.115

Copyright: © 2017 Zhang T, et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

Publication history: 

  • Received date: 19 Dec 2016

  • Accepted date: 04 Jan 2017

  • Published date:11 Jan 2017
  •