Elsevier

Neuroscience & Biobehavioral Reviews

Volume 68, September 2016, Pages 387-409
Neuroscience & Biobehavioral Reviews

Review article
Neuregulin-1 and schizophrenia in the genome-wide association study era

https://doi.org/10.1016/j.neubiorev.2016.06.001Get rights and content

Highlights

  • We assess NRG1⿿s relevance to schizophrenia in the GWAS era.

  • The past decade of clinical research on NRG1 and schizophrenia is summarized.

  • We review preclinical work on NRG1⿿s role in excitatory/inhibitory neurotransmission.

  • We propose a strategy forward for future NRG1 research in schizophrenia.

Abstract

Clinical and pre-clinical evidence has implicated neuregulin 1 (NRG1) as a critical component in the pathophysiology of schizophrenia. However, the arrival of the genome-wide association study (GWAS) era has yielded results that challenge the relevance of NRG1 in schizophrenia due to the absence of a genome-wide significant NRG1 variant associated with schizophrenia. To assess NRG1⿿s relevance to schizophrenia in the GWAS era, we provide a targeted review of recent preclinical evidence on NRG1⿿s role in regulating several aspects of excitatory/inhibitory neurotransmission and in turn schizophrenia risk. We also present a systematic review of the last decade of clinical research examining NRG1 in the context of schizophrenia. We include concise summaries of genotypic variation, gene-expression, protein expression, structural and functional neuroimaging as well as cognitive studies conducted during this time period. We conclude with recommendations for future clinical and preclinical work that we hope will help prioritize a strategy forward to further advance our understanding of the relationship between NRG1 and schizophrenia.

Introduction

Neuregulin-1 (NRG1) is one of the most extensively studied genes in schizophrenia. The genomic region surrounding NRG1 first attracted attention following linkage analyses (Brzustowicz et al., 1999, Gurling et al., 2001, Kendler et al., 1996, Pulver et al., 1995) implicating a locus on chromosome 8p21-22 with the disorder. Subsequent fine mapping and linkage disequilibrium analysis in Icelandic patients led to the discovery of a haplotype (HAPICE) associated with schizophrenia and a parallel preclinical study showed NRG1 hypomorphic (i.e. reduced gene expression) mice had a reduction in N-methyl-d-aspartate receptors (NMDAR) in forebrain areas and deficits in prepulse inhibition of the startle response compared to wild-type mice (Stefansson et al., 2002). This seminal work generated a tremendous amount of enthusiasm when published and remains one of the cornerstones of NRG1 research in schizophrenia to date. However, no polymorphism has reached genome-wide significance (p = 5 ÿ 10⿿8) in any genome-wide association study (GWAS) in schizophrenia (Schizophrenia Working Group of the Psychiatric Genomics, 2014), despite the inclusion of 400 or more NRG1 SNPs and gene coverage within the upper 10% of all protein-coding genes included on conventional GWAS platforms (Lehne et al., 2011). This suggests that the absence of a NRG1 signal in GWAS is unlikely to be a result of poor SNP coverage but rather a result of allelic heterogeneity at the NRG1 locus (Weickert et al., 2012), which in large samples is likely to result in a loss of a genetic signal. Nevertheless, this absence of GWAS support has attenuated enthusiasm for future NRG1 research and has questioned the relevance of NRG1 in schizophrenia.

Relevance of a gene to a particular disorder however, should not be determined exclusively on whether nucleotide variation within it meets genome-wide significance but rather on the whole of the evidence base. As such, we have completed a comprehensive assessment of the last decade of clinical research into the relationship between NRG1 and schizophrenia (for a review of the research prior to 2006 see: Harrison and Law, 2006). In addition, we provide a targeted review of preclinical studies that have examined NRG1⿿s effects on excitatory and inhibitory neurotransmission as recent preclinical reviews (Lisman, 2012, Mei and Nave, 2014) have not covered this topic thoroughly and an imbalance of excitatory/inhibitory transmission is one of several putative neurobiological mechanisms of schizophrenia (Chana et al., 2013). We conclude with recommendations for future research that we hope will help prioritize a strategy forward to further advance our understanding of the relationship between NRG1 and schizophrenia.

Section snippets

NRG1 structure and isoforms

NRG1 spans 1.125 megabases and comprises more than 20 exons as well as several large introns from which over 30 splice isoforms can be produced that are grouped into six types (I⿿VI) (Fig. 1). NRG1 types I and II contain an immunoglobulin (Ig) like domain, encoded by exons E178 and E122 respectively. This Ig region is also present in types IV and V and, together (I, II, IV and V) are commonly defined as Ig-NRG1. The Ig domain is linked to the epidermal growth factor (EGF) like domain with or

NRG1 and excitatory/inhibitory neurotransmission

A putative imbalance of excitatory/inhibitory transmission in the cortex, striatum and hippocampus is one likely neurobiological underpinning of schizophrenia (Carlsson and Carlsson, 1990). Current theories suggest that in schizophrenia excitatory activity of pyramidal neurons may be blunted in frontal brain regions (Stone et al., 2007) responsible for attention, memory and other executive functions, but overactive in the hippocampus (Grace, 2012, Schobel et al., 2013) and in certain

Family studies

The original family study in the Icelandic population linking a haplotype (HAPICE) in the 5⿲ region of NRG1 with schizophrenia (Stefansson et al., 2002) stimulated numerous additional family studies of which seven were conducted in the past decade (Table 4; for details on family studies from 2002 to 2005 see: Harrison and Law, 2006). Unlike the family studies conducted soon after the Icelandic study, the ancestral backgrounds of cohorts in more recent studies have been diverse, covering

Human post-mortem brain mRNA studies

Seven NRG1 gene expression studies using human post-mortem brain tissue have been published, six in the past decade (Table 6). Four of these studies have used case-control and the other three control only designs. Among the case-control studies, the first was conducted in 2004 (Hashimoto et al., 2004) using dorsolateral prefrontal cortex (DLPFC) tissue (20 schizophrenia, 19 controls) and noted an increase in type I NRG1 gene expression in schizophrenia. This finding has since been replicated in

NRG1 protein expression studies

Studies of NRG1 protein levels in schizophrenia have been hampered by the lack of specific antibodies for a majority of the NRG1 isoforms and methodological variations across studies. Despite these limitations, six post-mortem brain, one serum, and one plasma study in humans have been published (Table 7). Two studies have reported on NRG1α, one reporting a decrease in PFC white matter (Bertram et al., 2007) and the other no difference in Brodmann⿿s area 46 (frontal cortex) (Boer et al., 2009).

Structural neuroimaging studies

In the past decade, twelve studies have examined the association between NRG1 genetic variation and brain structure (Table 8). These studies have in large part taken a region of interest approach and have almost exclusively selected polymorphisms within the HAPICE. The SNP8NRG221533 polymorphism has received the most attention (6 studies). The C ⿿risk⿿ allele at this locus was associated with lower white matter volume (detected with voxel based morphometry) in the regions of the right uncinate

NRG1 and antipsychotic treatment response

The association between NRG1 genetic variation on antipsychotic response was first reported in a study of 94 Finnish individuals with schizophrenia taking typical antipsychotics for a minimum of four weeks (Kampman et al., 2004). In this study, homozygotes of the ⿿non-risk⿿ T allele at the SNP8NRG221533 (rs35753505) locus were over-represented in the non-responder group. Interestingly, a decade has passed since this study but to our knowledge, only two additional studies examining NRG1 genetic

Future directions

Throughout our review we have noted a number of gaps and limitations within the current NRG1 literature that warrant future study. To guide this future clinical and preclinical work and assist in prioritizing a strategy forward we have generated the following recommendations:

  • 1.

    Look beyond the central dogma. Human-based evidence related to the epigenetic (e.g. methylation), post-transcriptional (e.g. microRNA), or post-translational (e.g. phosphorylation, ubiquitination, cleavage) modification of

Summary

The past decade of clinical research and recent preclinical findings clearly show that the link between NRG1 and schizophrenia is complex and will require further concerted and collaborative efforts to be elucidated. At the preclinical level, there is strong evidence that Nrg1 may disrupt normal excitatory/inhibitory neurotransmission via the ErbB4 receptor, potentially mediated by downstream effects such as gamma oscillations that then interfere with cognitive processes principally involving

Acknowledgements

MM was supported by a Cooperative Research Centre for Mental Health Top-up Scholarship. SS and AP were supported by One-in-Five Association Incorporated. CP was supported by an NHMRC Senior Principal Research Fellowship (#628386), and a Brain and Behavior Research Foundation (NARSAD) Distinguished Investigator Award. TK is supported by a Career Development Fellowship (Level 2: #1045643) and a project grant (#1102012) from the National Health and Medical Research Council (NHMRC), as well as the

References (194)

  • B. Dean et al.

    Increased levels of serotonin 2A receptors and serotonin transporter in the CNS of neuregulin 1 hypomorphic/mutant mice

    Schizophr. Res.

    (2008)
  • F.O. Dimayuga et al.

    The neuregulin GGF2 attenuates free radical release from activated microglial cells

    J. Neuroimmunol.

    (2003)
  • L. Duffy et al.

    Cognition in transmembrane domain neuregulin 1 mutant mice

    Neuroscience

    (2010)
  • C.E. Duncan et al.

    Prefrontal GABA A receptor α-subunit expression in normal postnatal human development and schizophrenia

    J. Psychiatr. Res.

    (2010)
  • L. Georgieva et al.

    Support for neuregulin 1 as a susceptibility gene for bipolar disorder and schizophrenia

    Biol. Psychiatry

    (2008)
  • Y.G. Gong et al.

    A two-method meta-analysis of neuregulin 1(NRG1) association and heterogeneity in schizophrenia

    Schizophr. Res.

    (2009)
  • A.A. Grace

    Dopamine system dysregulation by the hippocampus: implications for the pathophysiology and treatment of schizophrenia

    Neuropharmacology

    (2012)
  • T.A. Greenwood et al.

    Genetic assessment of additional endophenotypes from the Consortium on the Genetics of Schizophrenia Family Study

    Schizophr. Res.

    (2016)
  • O. Gruber et al.

    Neuregulin-1 haplotype HAP(ICE) is associated with lower hippocampal volumes in schizophrenic patients and in non-affected family members

    J. Psychiatr. Res.

    (2008)
  • H.M. Gurling et al.

    Genomewide genetic linkage analysis confirms the presence of susceptibility loci for schizophrenia, on chromosomes 1q32.2, 5q33.2, and 8p 21-22 and provides support for linkage to schizophrenia, on chromosomes 11q23.3-24 and 20q12.1-11.23

    Am. J. Hum. Genet.

    (2001)
  • P.J. Harrison et al.

    Neuregulin 1 and schizophrenia: genetics, gene expression, and neurobiology

    Biol. Psychiatry

    (2006)
  • U.K. Haukvik et al.

    An exploratory model for G x E interaction on hippocampal volume in schizophrenia; obstetric complications and hypoxia-related genes

    Prog. Neuro-psychopharmacol. Biol. Psychiatry

    (2010)
  • H. Hida et al.

    Behavioral phenotypes in schizophrenic animal models with multiple combinations of genetic and environmental factors

    J. Pharmacol. Sci.

    (2013)
  • X.J. Hou et al.

    Neuregulin 1/ErbB4 enhances synchronized oscillations of prefrontal cortex neurons via inhibitory synapses

    Neuroscience

    (2014)
  • Y.Z. Huang et al.

    Regulation of neuregulin signaling by PSD-95 interacting with ErbB4 at CNS synapses

    Neuron

    (2000)
  • H. Jaaro-Peled et al.

    Neurodevelopmental mechanisms of schizophrenia: understanding disturbed postnatal brain maturation through neuregulin-1-ErbB4 and DISC1

    Trends Neurosci.

    (2009)
  • O. Jensen et al.

    Human gamma-frequency oscillations associated with attention and memory

    Trends Neurosci.

    (2007)
  • M.B. Johnson et al.

    Functional and evolutionary insights into human brain development through global transcriptome analysis

    Neuron

    (2009)
  • D. Joshi et al.

    Elevated ErbB4 mRNA is related to interneuron deficit in prefrontal cortex in schizophrenia

    J. Psychiatr. Res.

    (2014)
  • D. Joshi et al.

    Relationship between somatostatin and death receptor expression in the orbital frontal cortex in schizophrenia: a postmortem brain mRNA study

    npj Schizophr.

    (2015)
  • C. Konradi et al.

    Hippocampal interneurons are abnormal in schizophrenia

    Schizophr. Res.

    (2011)
  • A. Krug et al.

    The effect of Neuregulin 1 on neural correlates of episodic memory encoding and retrieval

    Neuroimage

    (2010)
  • P. Kukshal et al.

    Association study of neuregulin-1 gene polymorphisms in a North Indian schizophrenia sample

    Schizophr. Res.

    (2013)
  • A.J. Law

    Genetic mouse models of neuregulin 1: gene dosage effects, isoform-specific functions, and relevance to schizophrenia

    Biol. Psychiatry

    (2014)
  • Y. Abe et al.

    Neuregulin-1 signals from the periphery regulate AMPA receptor sensitivity and expression in GABAergic interneurons in developing neocortex

    J. Neurosci.

    (2011)
  • A.M. Addington et al.

    Neuregulin 1 (8p12) and childhood-onset schizophrenia: susceptibility haplotypes for diagnosis and brain developmental trajectories

    Mol. Psychiatry

    (2007)
  • Anon

    The genotype-tissue expression (GTEx) project

    Nat. Genet.

    (2013)
  • J. Bao et al.

    Back signaling by the Nrg-1 intracellular domain

    J. Cell Biol.

    (2003)
  • C.S. Barz et al.

    Sensory encoding in neuregulin 1 mutants

    Brain Struct. Funct.

    (2016)
  • I. Bertram et al.

    Immunohistochemical evidence for impaired neuregulin-1 signaling in the prefrontal cortex in schizophrenia and in unipolar depression

    Ann. N. Y. Acad. Sci.

    (2007)
  • L.-L. Bi et al.

    Amygdala NRG1-ErbB4 is critical for the modulation of anxiety-like behaviors

    Neuropsychopharmacology

    (2015)
  • M. Bjarnadottir et al.

    Neuregulin1 (NRG1) signaling through Fyn modulates NMDA receptor phosphorylation: differential synaptic function in NRG1+/⿿ knock-outs compared with wild-type mice

    J. Neurosci.

    (2007)
  • C.A. Bousman et al.

    Effects of NRG1 and DAOA genetic variation on transition to psychosis in individuals at ultra-high risk for psychosis

    Transl. Psychiatry

    (2013)
  • C.A. Bousman et al.

    Effects of neuregulin-1 genetic variation and depression symptom severity on longitudinal patterns of psychotic symptoms in primary care attendees

    Am. J. Med. Genet. B Neuropsychiatr. Genet.

    (2014)
  • D.M. Cannon et al.

    The association of white matter volume in psychotic disorders with genotypic variation in NRG1, MOG and CNP: a voxel-based analysis in affected individuals and their unaffected relatives

    Transl. Psychiatry

    (2012)
  • M. Carlsson et al.

    Schizophrenia: a subcortical neuretransmitter imbalance syndrome?

    Schizophr. Bull.

    (1990)
  • A. Caspi et al.

    Gene⿿environment interactions in psychiatry: joining forces with neuroscience

    Nat. Rev. Neurosci.

    (2006)
  • V.S. Catts et al.

    Rethinking schizophrenia in the context of normal neurodevelopment

    Front. Cell. Neurosci.

    (2013)
  • V.S. Catts et al.

    Postsynaptic density levels of the NMDA receptor NR1 subunit and PSD-95 protein in prefrontal cortex from people with schizophrenia

    npj Schizophr.

    (2015)
  • G. Chana et al.

    Biomarker investigations related to pathophysiological pathways in schizophrenia and psychosis

    Front. Cell. Neurosci.

    (2013)
  • Cited by (65)

    • Unmyelinated sensory neurons use Neuregulin signals to promote myelination of interneurons in the CNS

      2022, Cell Reports
      Citation Excerpt :

      The nrg1 gene generates many isoforms through multiple promoters and alternative splicing. A unique promoter and 5′ exon distinguish each main class of isoforms, resulting in distinct tissue-specific expression patterns and N-terminal protein sequences: types I–VI are present in humans, while types I, II, and III are common to all vertebrates.24,25 To determine which Nrg1 isoforms have functions in myelin sheath formation in the CNS, we used CRISPR-Cas9 to create a series of mutations in the zebrafish nrg1 gene.

    • Lower plasma total tau in adolescent psychosis: Involvement of the orbitofrontal cortex

      2021, Journal of Psychiatric Research
      Citation Excerpt :

      In the context of the neurodevelopmental paradigm of schizophrenia, the results from ex-vivo, MRI, genetic and animal model studies are suggestive of abnormal neuronal migration (Brent et al., 2013; Muraki and Tanigaki, 2015). For instance, neuregulin 1 has been implicated in schizophrenia (Mostaid et al., 2016) and plays a crucial role in radial migration (Anton et al., 1997; Rio et al., 1997). Radial migration is the travel of the glutamatergic neurons from the ventricular zone where they are born to their final destination at the cortical surface (Rakic, 1995) and is achieved with the involvement of the radial glial cell fibers (Lee, 2019).

    View all citing articles on Scopus
    View full text