Вы находитесь на странице: 1из 15

Received: 31 January 2017 | Accepted: 5 May 2017

DOI: 10.1002/ajmg.b.32554

REVIEW ARTICLE

A comprehensive review of the genetic and biological


evidence supports a role for MicroRNA-137 in the
etiology of schizophrenia

Kensuke Sakamoto PhD1 | James J. Crowley PhD1,2,3

1 Department of Genetics, University of North


Carolina at Chapel Hill, Chapel Hill, North Since it was first associated with schizophrenia (SCZ) in a 2011 genome-wide
Carolina
association study (GWAS), there have been over 100 publications focused on MIR137,
2 Department of Psychiatry, University of
North Carolina at Chapel Hill, Chapel Hill,
the gene encoding microRNA-137. These studies have examined everything from its
North Carolina fundamental role in the development of mice, flies, and fish to the intriguing
3 Department of Clinical Neuroscience,
enrichment of its target gene network in SCZ. Indeed, much of the excitement
Karolinska Institutet, Stockholm, Sweden
surrounding MIR137 is due to the distinct possibility that it could regulate a gene
Correspondence
network involved in SCZ etiology, a disease which we now recognize is highly
Kensuke Sakamoto, PhD, Department of
Genetics, CB#7264, 5097 Genomic Medicine polygenic. Here we comprehensively review, to the best of our ability, all published
Building, University of North Carolina at
genetic and biological evidence that could support or refute a role for MIR137 in the
Chapel Hill, Chapel Hill, NC.
Email: Kensuke_Sakamoto@unc.edu etiology of SCZ. Through a careful consideration of the literature, we conclude that the
data gathered to date continues to strongly support the involvement of MIR137 and its
Funding information
NIH, Grant numbers: R01MH105500, target gene network in neuropsychiatric traits, including SCZ risk. There remain,
R01MH110427, R21MH099370,
however, more unanswered than answered questions regarding the mechanisms
R21MH102814
linking MIR137 genetic variation with behavior. These questions need answers before
we can determine whether there are opportunities for diagnostic or therapeutic
interventions based on MIR137. We conclude with a number of suggestions for future
research on MIR137 that could help to provide answers and hope for a greater
understanding of this devastating disorder.

KEYWORDS
GWAS, microRNA, miR-137, schizophrenia, SNP

1 | G E N E TI C E V I DEN C E F O R MIR137 I N SCZ. Furthermore, many key predicted miR-137 target genes (e.g., TCF4,
SC HIZ OPHRENIA CACNA1C, CSMD1) have been verified molecularly. Throughout this
manuscript, we use the following nomenclature system: MIR137 (human
In the largest genome-wide association meta-analysis for SCZ to date genomic locus), Mir137 (mouse genomic locus), miR-137 (human and mouse
(Schizophrenia Working Group of the Psychiatric Genomics Consortium, mature micro RNA); (http://www.genenames.org/cgi-bin/genefamilies/
2014) the second most significant association (rs1702294, p = 3.36 × 10−19) set/476, http://www.informatics.jax.org/marker/MGI:2676822, http://
was located in MIR137HG, the host gene encoding miR-137. Figure 1 shows www.mirbase.org/help/nomenclature.shtml).
the pattern of association in the region (chr1: 98,250,000–98,750,000). As After the first identification of a SCZ risk variant in MIR137
described in greater detail below, multiple reports indicate that genes with (rs1625579, p = 1.6 × 10−11) (Schizophrenia Psychiatric Genome-Wide
predicted miR-137 target sites are enriched for smaller SCZ GWAS p-values, Association Study Consortium, 2011), a number of studies demon-
raising the possibility that miR-137 regulates a gene network involved in strated the association between rs1625579 and clinical phenotypes

Am J Med Genet. 2017;9999:1–15. wileyonlinelibrary.com/journal/ajmgb © 2017 Wiley Periodicals, Inc. | 1


2 | SAKAMOTO AND CROWLEY

characterized by childhood onset neurological problems, including


developmental delay. Critically, however, two recent reports from
Pinto et al. (2014) and Tucci, Ciaccio, Scuvera, Esposito, and Milani
(2016) have identified children whose deletions in this region overlaps
only MIR137, leaving DPYD unaffected. They both show that
hemizygous loss of MIR137 alone is sufficient to produce all three
features of this syndrome (intellectual disability, ASD, and obesity).
While these CNV data may seem to suggest that MIR137 is more
relevant to ASD than SCZ, it is important to note that these case reports
describe children who have yet to enter the risk period for SCZ and, of
course, a one-copy deletion may lead to a more profound phenotype
than common SNPs, which likely have a more subtle effect on function.
FIGURE 1 Schizophrenia GWAS meta-analysis regional
In support of this idea, the Cross-Disorder Group of the Psychiatric
association plot. A 400 kb region of chromosome 1 is shown,
Genomics Consortium (PGC) reported that, in an analysis of five major
centered on the most significant SNP, rs1702294 (image from
freeze PGC-SCZ52, https://data.broadinstitute.org/mpg/ricopili). psychiatric disorders, a best-fit model predicted common variants in
The x-axis is genomic position, and the y-axis is degree of MIR137 to jointly influence risk to SCZ and ASD, but not attention deficit
association with SCZ (−log10P). Directly genotyped SNPs are plotted hyperactivity disorder (ADHD), bipolar disorder, or major depression
as diamonds, while imputed SNPs are plotted as circles. Color- (Cross-Disorder Group of the Psychiatric Genomics Consortium, 2013).
coding and symbol size indicate higher linkage disequilibrium (R2)
with rs1702294 (e.g., red points denote an R2 between 0.81 and
1.0, and larger symbol indicates higher R2). Estimated recombination 2 | E S S E N T I A L B A C K G R O U N D ON
rates (in cM/Mb) are depicted as a light blue line to demonstrate microRNAs
the local LD structure. MIR137HG refers to the host gene encoding
miR-137 (also called MIR137). The previously detected lead SNP
(rs1625579) and a potential MIR137 regulatory SNP (rs2660304) MicroRNAs (miRNAs) are small non-coding RNA species of ∼22
are also indicated and explained in the manuscript. nucleotides which negatively regulate mRNA stability or translation,
and in turn, alter various cellular functions (Bartel, 2004; Lagos-
Quintana, Rauhut, Lendeckel, & Tuschl, 2001; Wright, Turner, Calhoun,
(e.g., age of onset, psychotic symptoms, and cognitive function)
& Perrone-Bizzozero, 2013). Importantly, a single miRNA can regulate
(Cummings et al., 2013; Kuswanto et al., 2015; Lett et al., 2013), miR-
the expression of many target genes, and, as such, miRNAs often act as
137 expression in the brain (Guella et al., 2013), brain structure (Cousijn
key signaling nodes regulating complex biological networks.
et al., 2014; Kelly et al., 2014; Kuswanto et al., 2015; Lett et al., 2013; Li
The canonical miRNA biogenesis process is nicely summarized in
& Su, 2013; Patel et al., 2015; Rose et al., 2014; Wright et al., 2016), and
previous articles (Bartel, 2004; Kim & Kim, 2007; Lagos-Quintana et al.,
brain activities (Liu et al., 2014; Mothersill et al., 2014; van Erp et al.,
2001; Wright et al., 2013). In short, miRNAs are transcribed from
2014; Whalley et al., 2012). These are summarized in Table 1 and will be
intergenic non-coding RNA (such is the case for miR-137) or introns of
discussed in this manuscript.
protein coding genes (introgenic microRNA) (Kim, Han, & Siomi, 2009;
The haplotype block harboring MIR137 also contains a second
Liu et al., 2008). The single-stranded “primary” miRNA forms a stem-
miRNA gene (MIR2682) which has been reported to have very low
loop structure and is then cleaved in the nucleus into a ∼70 nucleotide
brain expression (Miller et al., 2014), explaining the little attention it
“precursor” miRNAs by a complex composed of Drosha and DGCR8
has garnered, and the 5′ portion of the gene DPYD (encoding
(Denli, Tops, Plasterk, Ketting, & Hannon, 2004; Gregory et al., 2004;
dihydropyrimidine dehydrogenase), which itself has an independent
Han et al., 2006). Following export to the cytoplasm, the precursor
genome-wide significant association with SCZ in its 3′ region (Ripke,
miRNA is further processed by Dicer into its final ∼22 nucleotide form
2013; Schizophrenia Working Group of the Psychiatric Genomics
(Chendrimada et al., 2005; Forstemann et al., 2005). Finally, miRNAs
Consortium, 2014). Copy number variants (CNVs) in the region shown
are loaded onto the RNA-induced silencing complex (RISC) where they
in Figure 1 are very rare, but have proven informative. In particular,
bind to their target RNAs, induce mRNA degradation and/or reduce
hemizygous deletions in this region of chr1p21.3 encompassing both
translation efficiency (Wightman, Ha, & Ruvkun, 1993; Wu & Belasco,
DPYD and MIR137 have been described in ∼10 cases and are
2008; Zeng, Yi, & Cullen, 2003).
associated with a syndrome consisting primarily of intellectual
disability, autism-spectrum disorder (ASD), and obesity (Carter et al.,
2011; D’Angelo, Moller Dos Santos, Alonso, & Koiffmann, 2015; 3 | m i R N A s A R E E S S E N T I A L FO R P R O PE R
Willemsen et al., 2011). Some investigators initially proposed DPYD to BRA IN D EVELOPMENT AND FUNCTION
be the major gene contributing to this syndrome (Carter et al., 2011;
D’Angelo et al., 2015), which seemed sensible since DPYD deficiency A series of studies over the past decade have revealed that finely-tuned
(OMIM #274270; DPYD encodes for the rate-limiting enzyme in the miRNA function across the lifespan is critical for orchestrating cell
catabolism of the pyrimidine bases), an autosomal recessive disease, is proliferation, differentiation, migration, and death in the brain (Cheng,
SAKAMOTO AND CROWLEY | 3

TABLE 1 Putatively functional schizophrenia-associated variants in the MIR137 locus


Variant
(SNP ID) Study type Effects of the risk alleles References
−11
rs1625579 GWAS  Association with SCZ, 9,394 cases and 12,462 controls, p = 1.6 × 10 Schizophrenia Psychiatric
Genome-Wide Association
Study Consortium (2011)
Clinical  Earlier disease onset Lett et al. (2013)
symptoms  More severe positive symptoms and cognitive function Cummings et al. (2013)
 More severe negative symptoms, poorer attention and processing speed Kuswanto et al. (2015)
miR-137  Decreased miR-137 expression in the DLPFC Guella et al. (2013)
expression
Brain  Decreased fronto-striatal brain white matter integrity Kuswanto et al. (2015)
structure  Decreases in occipital, parietal and temporal lobe gray matter concentration Wright et al. (2016)
 Reduced white matter integrity, smaller hippocampus, larger lateral ventricles Lett et al. (2013)
Brain  Higher left DLPFC activation van Erp et al. (2014)
function  Higher functional connectivity between DLPFC and left hippocampi than heterozy- Liu et al. (2014)
gous individuals
 Increased functional connectivity between right amygdala and frontal regions Mothersill et al. (2014)
 Higher activity in the posterior right medial frontal gyrus, BA6 Whalley et al. (2012)
rs1702294 GWAS  Association with SCZ, 36,989 cases and 113,075 controls, p = 3.36 × 10−19 Schizophrenia Working
Group of the Psychiatric
Genomics Consortium
(2014)
rs2660304 GWAS  Association with SCZ (in LD with a reported SCZ-associated SNP, rs1198588 Duan et al. (2014)
(R2 = 0.74, based on the 1000 Genomes project))
 Association with SCZ in an Ashkenazi Jewish population Goes et al. (2015)
miR-137  Decreased promoter activity Warburton et al. (2016)
expression  Lower level of endogenous miR-137 expression Siegert et al. (2015)

Other Effects of the variation References


variant
VNTR miR-137  pre-miR-137 carrying 9 and 13 repeats leads to reduced miR-137 expression Mamdani et al. (2013)
expression  VNTR length affects the secondary structure of pre-miR-137 stem-loop and impacts Bemis et al. (2008); Strazisar
expression of mature miR-137 et al. (2015)

GWAS, genome-wide association study; SCZ, schizophrenia; DLPFC, dorsolateral prefrontal cortex; BA6, Brodmann area 6; LD, linkage disequilibrium;
VNTR, variable number tandem repeat.

Pastrana, Tavazoie, & Doetsch, 2009; Friedman et al., 2009; Hebert & Dysregulation of miRNAs has been associated with several
De Strooper, 2007; Shibata, Nakao, Kiyonari, Abe, & Aizawa, 2011). At neurological and neuropsychiatric disorders (Beveridge et al., 2008; Klein
the developmental level, conditional deletion of Dicer, an essential et al., 2007; Kim et al., 2007; Packer, Xing, Harper, Jones, & Davidson,
component of miRNA genesis, in the CNS results in abnormal neuronal 2008; Shioya et al., 2010; Wang et al., 2008; Xu, Hsu, Karayiorgou, &
differentiation and brain structure (Kawase-Koga, Otaegi, & Sun, 2009; Gogos, 2012; Xu, Li, Wang, & Gao, 2008; Zhou et al., 2009). Of note,
Saurat, Andersson, Vasistha, Molnar, & Livesey, 2013). In mature brain, chromosome 22q11.2, a locus strongly connected to neurocognitive
miRNAs also act to modulate morphology and function. For instance, deficits with a high risk of SCZ, contains the miRNA microprocessor
miR-132 and miR-212 demonstrate expression that is tightly regulated component gene, DGCR8. Multiple miRNAs are dysregulated in the blood
by neuronal activity. These miRNAs have the ability to modulate a of individuals with 22q11.2 deletion syndrome (22q11DS), and there is a
number of neuronal and behavioral phenotypes, including synapse significant inverse correlation between miRNA expression and hippo-
number, synaptic efficiency, dendrite complexity, and context depen- campal and whole brain volume in 22q11DS cases (Merico et al., 2014;
dent short-term memory (Cheng et al., 2007; Edbauer et al., 2010; Sellier et al., 2014; Stark et al., 2008; Zhao et al., 2015).
Hansen, Sakamoto, Wayman, Impey, & Obrietan, 2010; Impey et al.,
2010; Remenyi et al., 2010; Wayman et al., 2008). Beyond neurons,
miRNAs control the differentiation and maintenance of glial cells in both 4 | MicroRNA -137
the developing and mature brain (Hong, Jiang, & Li, 2014; Howng,
Huang, Ptacek, & Fu, 2015; Kawase-Koga et al., 2009; Zhang et al., miR-137 was first identified in mice during tissue-specific small RNA
2015). profiling and was also shown to be highly conserved and expressed
4 | SAKAMOTO AND CROWLEY

across mammalian and non-mammalian vertebrates (Lagos-Quintana (Agarwal, Bell, Nam, & Bartel, 2015; Betel, Wilson, Gabow, Marks, &
et al., 2002). Human MIR137 is located within host gene MIR137HG Sander, 2008; Wong & Wang, 2015), 2) gene expression analysis after
(chr1:98,453,556–98,515,249). The 86bp hairpin sequence within immunoprecipitating the RISC complex or after altering miR-137
pre-miR-137 is highly conserved from humans to zebrafish, represent- expression (Boudreau et al., 2014; Collins et al., 2014; Hill et al., 2014;
ing ∼450 million years since the last common ancestor (Kumar & Tamim et al., 2014), and 3) reporter gene activity or quantitative protein
Hedges, 1998). This illustrates that miR-137 has assumed a critical role measures (e.g., luciferase assay, proteomics, Western blotting) (Chen,
in mammalian biology. In addition to the pre-miRNA sequence, there is Wang et al., 2011; Kim et al., 2012; Kwon, Wang, & Tsai, 2013; Siegert
also high evolutionary conservation of several key regulatory elements et al., 2015). Given the strengths and weaknesses of these various
surrounding MIR137 (e.g., transcription factor binding sites, epigenetic approaches, most investigators have chosen to integrate data from two
marks, etc.), suggesting that some regulatory mechanisms may also be or more of these methods in generating miR-137 target gene lists.
conserved. Figure 2 provides an overview of several of these features, Bioinformatic algorithms predict miR-137 targets by calculating
many of which will be referred to in the discussion below. the likelihood of base-pairing with target mRNA untranslated regions
(UTRs). The available algorithms vary in the weight given to different
criteria for this calculation, and so their results vary. For example,
5 | m i R - 1 3 7 T A R G E T G E N E S AN D
TargetScan searches primarily for a complementary match between
SC HIZ OPHRENIA G ENOM ICS
the 3′ UTR of an mRNA and the miRNA “seed” sequence, which is the
term given to the 5′ 2–8 nucleotides critical for target detection
Bioinformatic methods cannot perfectly predict genes targeted by
(Agarwal et al., 2015; Lewis, Burge, & Bartel, 2005). In its latest version
miRNAs, due to imperfect base-pairing between miRNAs and their
(release 7.1, June 2016), TargetScan predicts N = 1,305 transcripts
targets. Therefore, investigators have taken multiple informatics and
with phylogenetically conserved miR-137 binding sites.
experimental approaches to generate meaningful lists of miR-137 target
Computationally predicted miR-137 targets have been repeatedly
genes for pathway analysis. These include: 1) algorithm-based comple-
associated with SCZ. For example, genes with TargetScan-predicted
mentary base pairing analysis (e.g., Target Scan, Miranda, miRdb, etc.)
miR-137 target sites are enriched for smaller GWAS p-values, raising
the possibility that miR-137 regulates a gene network involved in SCZ
(Schizophrenia Psychiatric Genome-Wide Association Study Consor-
tium, 2011). Wright et al. (2015) further categorized miR-137 targets
by pathway-specific gene sets, performed a similar analysis, and found
that potential miR-137 targets are particularly enriched in SCZ risk
genes involved in axonal guidance signaling, Ephrin receptor signaling,
long-term potentiation, PKA signaling, and Sertoli cell junction
signaling. Additional evidence comes from a recent rare variant study
by Genovese et al. (2016), who collated disruptive ultra-rare variants
(dURVs) in exome sequencing data from Swedish SCZ cases and
controls. In addition to observing an overall burden of dURVs in cases,
they reported a significant enrichment within predicted miR-137
FIGURE 2 Regulatory DNA surrounding MIR137. A number targets (Genovese et al., 2016). Finally, taking an imaging genetics
of key tracks from the UCSC Genome Browser (hg19 assembly) approach, Potkin et al. (2010) showed that miR-137 targets are
are shown. These include common SNPs (MAF > 0.05), with red enriched for smaller GWAS p-values in a study of dorsolateral
arrows pointing to the lead SNP (rs1702294), previously
prefrontal cortex (DLPFC) activation during a working memory task.
detected lead SNP (rs1625579), and a potential MIR137
regulatory SNP (rs2660304). “VNTR” depicts the position of a Experimentally derived miR-137 targets, from a variety of genomic
putatively functional variable number tandem repeat (VNTR). approaches, have also been associated with schizophrenia. It is
The bindings sites for RE1-Silencing Transcription factor (REST) important to note that these studies, summarized below, employed a
from two cell lines (ENCODE ChIP-seq data) are also shown. The wide range of tissues, cell lines, expression platforms, and miR-137
“Txn Factor ChIP” track summarizes transcription factor ChIP-
manipulation methods. As a result, the overlap in miR-137 targets is
seq data for 161 factors and the “DNase Clusters” track
limited, suggesting that miR-137 targets vary with tissue, cell type, and
summarizes DNaseI hypersensitivity clusters in 125 cell types.
“CpG Islands” shows regions with high CpG sequence frequency, proliferative status (Boudreau et al., 2014; Collins et al., 2014; Hill et al.,
while “Hypomethylation” shows regions with depleted DNA 2014; Tamim et al., 2014). First, Collins et al. (2014) overexpressed miR-
methylation in human neuronal progenitor cells. “H3K4me3” 137 in the human neuroprogenitor cell line RenCell-VM and used RNA
shows histone H3 Lysine 4 trimethylation (H3K4me3) sites, a sequencing to nominate down-regulated genes as miR-137 targets.
marker associated with active transcription start sites, detected
They noted that several SCZ-related pathways were up-regulated 48 hr
in postmortem human frontal cortex grey matter. “100 Vert.
Cons” shows base-level conservation (PhyloP score) for 100 after miR-137 overexpression, including: calcium channels, synaptic
different vertebrate species. The red rectangles point to proteins, FMRP-interacting RNAs, and genes forming the major
potential key transcription regulatory regions. histocompatibility complex (MHC). Second, Hill et al. (2014) took a
SAKAMOTO AND CROWLEY | 5

similar approach in the human neural progenitor cell line CTXOE03, Regarding when miR-137 is expressed, it is becoming clear that it
finding that genes down-regulated after miR-137 over-expression were appears very early in development and is necessary for life. Crowley et al.
enriched for involvement in neuronal differentiation and included (2015) showed that mice lacking expression of Mir137 (homozygous
genome-wide significant risk loci for SCZ (e.g., MAD1L1 and DPYD). constitutive knockout) resulted in embryonic lethality before develop-
Interestingly, a recent study demonstrated that an increased polygenic mental day 11.5. Others have shown that miR-137 is expressed in mouse
risk score within the miR-137 targets that Hill et al. (2014) detected was embryonic stem (ES) cells and regulates proper cell differentiation through
associated with significantly lower performance on intelligence specific target gene regulation (Jiang, Ren, & Nair, 2013; Tarantino et al.,
quotient, working memory and episodic memory (Cosgrove et al., 2010). According to Tarantino et al. (2010), a loss of miR-137 expression
2017). Functional brain imaging during a memory task further revealed during ES cell differentiation resulted in impaired neuronal gene
that it is also associated with increased activation of the right inferior expression and morphogenesis. Hollins et al. (2014) demonstrated that
occipital gyrus (Cosgrove et al., 2017). Third, Boudreau et al. (2014) used miR-137 expression is consistently high in the rat forebrain and midbrain
immunoprecipitation of the RISC complex from human brain motor at the late developmental stage (E12-P0). Consistent with this theme,
cortex and cingulate gyrus to directly pull out mRNAs being actively other work has revealed a crucial role for miR-137 in adult neurogenesis
targeted by miRNAs (HITS-CLIP). Expression analysis revealed 88 and neuronal maturation (Smrt et al., 2010; Sun et al., 2011). Furthermore,
transcripts with miR-137 binding sites (TargetScan seeds), 24 of which the BrainSpan consortium performed small RNA-sequencing of human
overlapped with genes from a SCZ meta-analysis (SzGene database brain and demonstrated high miR-137 expression across development in
(Allen et al., 2008)) or genes associated with prefrontal cortex several brain regions (Miller et al., 2014).
abnormalities in psychiatric patients (Allen et al., 2008; Kim & Webster, miR-137 expression is not limited to neurons. More work is
2010). Of note, 5 SCZ GWAS associated genes including TCF4, a needed to determine cell-type specificity, but the literature shows that
previously established miR-137 target (Kwon et al., 2013), were miR-137 is not only present in glial cells, but highly involved in glial cell
included in this list (Boudreau et al., 2014). Fourth, some groups have function and cell-cycle control, as demonstrated by repeated
taken a proteomic approach to detect translationally regulated miR-137 association of decreased miR-137 expression in glioblastoma (Chen
targets (Hao et al., 2015; Ren et al., 2015; Schouten et al., 2015), though et al., 2012; Koshkin, Chistyakov, Nikitin, Konovalov, Potapov,
their relevance to SCZ is as yet unclear. Usachyov et al., 2014; Koshkin, Chistyakov, Nikitin, Konovalov,
Highly reliable miR-137 targets empirically demonstrated by Potapov, Usachev et al., 2014; Silber et al., 2008; Visani et al., 2014).
molecular biological methods have been well summarized in previous
reviews (Mahmoudi & Cairns, 2017; Wright et al., 2013). Among these
validated targets are genes unquestionably associated with SCZ, such as
7 | S T E M C E L L P R O L I F E R A T I O N AN D
TCF4, CACNA1C, CSMD1, and C10orf26 (Kwon et al., 2013; Schizophre- DI FFERENTI ATI ON
nia Psychiatric Genome-Wide Association Study Consortium, 2011).
Furthermore, recent validated miR-137 targets include additional SCZ is considered by some to be a neurodevelopmental disorder (Kinros,

synaptic genes, such as those encoding Synaptophysin and glutamate Reichenberg, & Frangou, 2010; Rapoport, Giedd, & Gogtay, 2012;

receptor subunit GluA1 (Olde Loohuis et al., 2015; Siegert et al., 2015). Stachowiak et al., 2013), thus miR-137’s role in neurodevelopment may

The results summarized here suggest that a more precise analysis of be of etiological importance. The literature suggests that miR-137

miR-137 regulated gene networks in a variety of tissues, cell types, and controls proliferation and differentiation in both the developing and

developmental stages would be valuable as we try to understand the role mature brain, but its direction of effect is context-dependent. During

of miR-137 in SCZ etiology. For example, recent improvements in single- development, miR-137 is highly enriched in mitotic phase ES cells and

cell based gene expression analysis may contribute to a more precise controls differentiation by targeting transcription regulators (e.g., KLF4,

description of the miR-137 gene network (Zeisel et al., 2015) that could be TBX3, LSD1), and loss of miR-137 expression results in abnormal

used to refine the role of this pathway in neuropsychiatric disorders. neuronal morphology (Jiang et al., 2013). Consistent with Jiang et al.
(2013), Sun et al. (2011) demonstrated that miR-137 overexpression
decreases proliferation of mouse embryonic neural stem cells and
6 | W H E R E AN D W HE N I S m i R - 1 3 7 results in premature neuronal differentiation. Moreover, Tarantino et al.
EXPRESSED? (2010) demonstrated that miR-137 increases differentiation of mouse
embryonic stem cells and suppression of miR-137 blocked the
An examination of MIR137HG expression across >50 adult human tissues differentiation induced by leukemia inhibitory factor (LIF) withdrawal.
from GTEx (http://www.gtexportal.org) reveals highly enriched brain These reports demonstrate the pro-differentiation feature of miR-137
expression, with the highest expression in frontal cortex (Brodmann area in embryonic neuronal stem cells.
9, BA9) and this is supported by multiple publications (Consortium, 2015; In contrast to its effect on embryonic neurogenesis, Smrt et al. (2010)
Guo et al., 2014; Landgraf et al., 2007; Ludwig et al., 2016). Nearly all reported that miR-137 overexpression negatively controls adult neuronal
tissues, however, show measurable expression of miR-137 in GTEx and stem cell differentiation. They confirmed that miR-137 targets are
there is a growing appreciation of the role miR-137 plays in the cell cycle enriched in the neuronal lineage (compared to the astrocytic lineage) and
and tumor formation in non-brain tissues (see below). that miR-137 is highly expressed in the adult dentate gyrus of the
6 | SAKAMOTO AND CROWLEY

hippocampal formation, an area highly active in adult neurogenesis. Smrt melanogaster miR-1000 (a potential ortholog of mammalian
et al. (2010) also reported that miR-137 overexpressed cells had an 80% miR-137) represses the vesicular glutamate transporter (VGlut) and
decrease in the proportion of matured neurons. Other researchers have negatively regulates glutamate release at the synapse.
demonstrated that miR-137 overexpression enhances proliferation and Other researchers have documented a postsynaptic role for
inhibits differentiation of adult neuronal stem cells (Szulwach et al., 2010). miR-137. Olde Loohuis et al. (2015) showed that miR-137 increases
However, consistent with the effect on the developing brain, Santos et al. silent synapses by repressing AMPA receptor-mediated synaptic
(2016) showed that miR-137 is expressed much more highly in the non- transmission through down-regulation of AMPA receptor subunit
germinal striatum than in the subventricular zone (SVZ, an area of adult GluA1. They also found that miR-137 is transiently upregulated in
neurogenesis) in 2-month-old mice and that miR-137 is upregulated response to metabotropic glutamate receptor 5 (mGluR5) and miR-137
during lineage progression and the differentiation of neural stem cells. is crucial for mGluR-mediated long-term depression (LTD). These
They further showed that overexpression of miR-137 in isolated neural results suggest that miR-137 controls synaptic efficacy and mGluR-
stem cell cultures from 2-month-old mouse SVZ increases neuronal dependent synaptic plasticity and may contribute to glutamatergic
differentiation and decreases cell proliferation (Santos et al., 2016). These dysfunction in SCZ (de Bartolomeis, Buonaguro, & Iasevoli, 2013;
results suggest that miR-137 has a context-dependent role and function Moghaddam & Javitt, 2012). This result suggests that miR-137 is also
during neuronal stem cell development. Given the diversity of predicted involved in the regulation of dendritically localized mRNAs, which is a
miR-137 targets in different cell types (Boudreau et al., 2014; Collins et al., critical process for mGluR-LTD (Huber, Kayser, & Bear, 2000; Klann &
2014; Hill et al., 2014; Tamim et al., 2014), it is not surprising that the miR- Dever, 2004; Snyder et al., 2001).
137 regulated gene network is dependent on cell lineage context. A
simple, yet important, methodological difference in these experiments
included the magnitude of miR-137 overexpression, which may affect 9 | FU N CTION IN GLIA
outcome. Again, we note that the use of a precise gene network analysis in
individual cells could help to address these questions. Several groups have documented the involvement of miR-137 in the
biology of diverse cancers (e.g., brain, oral, colorectal, lung, gastric,
squamous cell carcinoma, breast, bladder, melanoma) (Balaguer et al.,
8 | NEURONA L MATURA TION AND 2010; Bandres et al., 2009; Bennett, Bemis, Norris, & Shellman, 2013;
S YN A P TI C FU N CT IO N Chen, Chen et al., 2011; Chen, Wang et al., 2011; Dacic, Kelly, Shuai, &
Nikiforova, 2010; Huang et al., 2009; Kozaki, Imoto, Mogi, Omura, &
A reduction of synaptic form and function are cellular features Inazawa, 2008; Langevin et al., 2011; Liu et al., 2011; Shimizu et al.,
frequently observed in psychiatric disorders, including SCZ and autism 2013; Tezcan et al., 2014; Zhao et al., 2012; Zhu et al., 2013). In
(Garey et al., 1998; Glantz & Lewis, 2000; Glausier & Lewis, 2013). general, miR-137 has been identified as a tumor suppressor. Reduced
Recent publications identify miR-137 as an important regulator of miR-137 expression is observed in glioblastoma cells, and higher grade
neuronal maturation and synaptic function in the adult brain. Several glioblastoma shows greater reduction (Chen et al., 2012; Koshkin,
researchers have indicated that miR-137 targets include synaptic Chistyakov, Nikitin, Konovalov, Potapov, Usachyov et al., 2014;
genes (Collins et al., 2014; Siegert et al., 2015; Strazisar et al., 2015). Koshkin, Chistyakov, Nikitin, Konovalov, Potapov, Usachev et al.,
Willemsen et al. (2011) reported finding enriched expression of pre- 2014; Silber et al., 2008; Visani et al., 2014). In fact, miR-137
miR-137 in synaptosomes. Moreover, Smrt et al. (2010) showed that expression in blood cells has been suggested as a biomarker for human
overexpression of miR-137 decreases dendritic complexity, spine glioblastoma (Li, Li, Li, Shi, & Chen, 2016).
density, and maturation of newborn neurons in adult hippocampus. The effect of glial miR-137 on cognitive function, however, is
Investigators have recently examined the precise role of miR-137 poorly understood. Given the critical roles of glial cells in neuro-
in synaptic regulation. Siegert et al. (2015) demonstrated, by electron cognitive functions (Cotter, Pariante, & Everall, 2001; Dzyubenko,
microscopy, that overexpression of miR-137 in the adult mouse Gottschling, & Faissner, 2016; Jo, Law, & Chung, 2014; Stogsdill &
hippocampus alters synaptic vesicle distribution; with miR-137 Eroglu, 2016) and SCZ pathology (Bernstein, Steiner, Guest,
overexpression, synaptic vesicles distributed more distantly from Dobrowolny, & Bogerts, 2015) and the fact that some miR-137 target
the release site, while the overall number of vesicles was not affected. genes are expressed in glial cells (Tamim et al., 2014), it would not be
This result was further supported by their finding that when sustained surprising if miR-137 mediated glial regulation has a critical role in fine-
low-frequency electrostimulation was applied to dentate granule cells, tuning brain function. For example, deficits in myelin, reduced
the amplitude of synaptic responses evoked at the mossy fiber-CA3 oligodendrocytes, and altered expression of myelin/oligodendrocyte
synapse reached a lower plateau in miR-137 overexpressed slices. -related genes are found in SCZ, which could explain white matter
Siegert et al. (2015) also showed that this phenotype is mediated by the abnormalities in SCZ (Goudriaan et al., 2014; Hof, Haroutunian,
regulation of a miR-137 target synaptotagmin-1 (Syt1), suggesting that Copland, Davis, & Buxbaum, 2002; Kerns et al., 2010; Regenold et al.,
miR-137 negatively regulates presynaptic neurotransmitter release. 2007; Schmitt et al., 2009; Uranova, Vikhreva, Rachmanova, &
Verma, Augustine, Ammar, Tashiro, and Cohen (2015) confirmed the Orlovskaya, 2011). Also, dysfunctional astrocytes, which have key
presynaptic role of miR-137 by demonstrating that Drosophila roles in the synaptic metabolism of neurotransmitters (e.g., glutamate,
SAKAMOTO AND CROWLEY | 7

GABA, monoamines, and purines), may contribute to disturbed Large-scale resources such as ENCODE (Consortium, 2012) have
neurotransmission in SCZ (Bernstein et al., 2015; Sofroniew & Vinters, produced catalogs of putative regulatory DNA sequences, such as those
2010). In addition, microRNAs have been proven to have critical roles with open chromatin, acetylated histones, enhancer function, or
in glial cell development and function (Bian, Xu, & Sun, 2013; methylated DNA, to name but a few (see Figure 2). In other disorders,
Karthikeyan, Patnala, Jadhav, Eng-Ang, & Dheen, 2016; Zheng, Li, there are documented examples of causal regulatory SNPs explaining
Huang, & Qiu, 2012). For example, miR-124 in microglia is down- GWAS signals (Fogarty, Cannon, Vadlamudi, Gaulton, & Mohlke, 2014;
regulated during autoimmune inflammation of the CNS, and reduction Nicolae et al., 2010; Smemo et al., 2014). This has generated great
of miR-124 in microglia activates these cells. (Ponomarev, Veremeyko, interest in identifying variants in the MIR137 locus that may help explain
Barteneva, Krichevsky, & Weiner, 2011). Interestingly, glial miR-137 the GWAS signal via altered expression, post-transcriptional processing
shares a large number of putative targets with miR-124 (Schouten or function of miR-137 (Consortium, 2015; Fromer et al., 2016;
et al., 2015; Silber et al., 2008; Tamim et al., 2014). In sum, although Gianfrancesco et al., 2016; Ward & Kellis, 2012; Yang, Li, Jiang, Zhou, &
current knowledge of miR-137’s role in glial cells is limited, these Qu, 2013). Gianfrancesco et al. (2016) defined seven potential
studies suggest potential functions of miR-137 in the fine tuning of regulatory regions upstream of MIR137 using a phylogenetic approach
brain function through glial cell regulation. to define evolutionarily conserved regions (ECRs). Three out of these
seven ECRs overlapped ENCODE-defined chromatin signatures in the
brain. They further showed that four ECRs have genome-wide
10 | A NI M A L M O D E L S O F m i R - 1 3 7 significant SCZ GWAS SNPs in or adjacent to their sequence.
D Y S F U N C T IO N Warburton, Breen, Bubb, and Quinn (2016) demonstrated that
rs2660304 (a SNP located ∼400 bp upstream of pre-miR-137) is in high
Mouse, fly, and fish all have models of miR-137 manipulation available LD with the top SCZ-associated SNP rs1625579 (R2 = 0.96), and that the
for study. Crowley et al. (2015) reported that mice with a whole body major allele (the SCZ risk allele) of rs2660304 decreases promoter
knockout of Mir137 are embryonically lethal between embryonic days activity. This result is consistent with Siegert et al. (2015) recent work
4.5 and 11.5. Interestingly, heterozygous knockouts showed no documenting a lower level of endogenous miR-137 expression in human
striking behavioral phenotype, likely due to compensated miR-137 fibroblast-derived neurons with the major (risk) allele of rs2660304. Of
expression (Crowley et al., 2015). Siegert et al. (2015) showed that note, rs2660304 has also been associated with SCZ in an Ashkenazi
virus-mediated miR-137 overexpression in the dentate gyrus impaired Jewish population (Duan et al., 2014; Goes et al., 2015). It is important to
induction of mossy fiber long-term potentiation and induced a deficit state that, as of this writing, no genome-wide expression QTL (eQTL)
in hippocampal-dependent learning and memory. In flies, knockout of study has associated rs2660304 with expression of MIR137, but few
Drosophila miR-1000 (ortholog of mammalian miR-137) led to an early- eQTL studies have closely examine microRNAs or their host genes and
onset movement disorder and higher apoptosis in the brain, as most brain eQTL studies at this point are underpowered. In sum, the
detected by Caspase-3 activity (Verma et al., 2015). In fish, existing in vitro molecular evidence appear consistent with at least one
developmental suppression of miR-137 in zebrafish impaired both SCZ risk variant (rs2660304) being associated with reduced activity of
embryonic and larval touch-sensitivity without compromising overall the MIR137 promoter. Furthermore, another SCZ risk allele in this
anatomical development (Giacomotto et al., 2016). region, rs1625579, is associated with decreased miR-137 expression in
Given the severe phenotypes associated with loss of miR-137 in the dorsolateral prefrontal cortex (DLPFC) (Guella et al., 2013),
multiple species (particularly embryonic lethality in mice), there is a need providing convergent evidence.
for a conditional knockout of Mir137 in mice and other species. It will be Another putatively functional DNA variant found just upstream of
important to examine the effects of miR-137 dysfunction at different MIR137 consists of a variable number tandem repeat (VNTR, see
developmental time-points, tissues, and cell types. Furthermore, only a Figure 2). By overexpressing pre-miR-137 with various numbers of
few behavioral domains have been examined thus far (e.g., memory, repeats (3–13) in HEK293 cells, Mamdani et al. (2013) demonstrated
anxiety, sensorimotor gating), but methods for other more sophisticated that pre-miR-137 carrying 9 and 13 repeats leads to significantly
measures of various endophenotypes are improving (Achim et al., 2011; reduced miR-137 expression. Two additional studies provide evidence
Hart et al., 2014; Nestler & Hyman, 2010; van Scheltinga et al., 2013; that VNTR length affects the secondary structure of pre-miR-137
Winton-Brown, Fusar-Poli, Ungless, & Howes, 2014). stem-loop and impacts expression of mature miR-137 (Bemis et al.,
2008; Strazisar et al., 2015). While these findings suggest that VNTR
repeat number potentially affects levels of miR-137, it is important to
11 | W H A T M O L E C U L A R ME C H A N I S M ( S ) note that there is currently insufficient genetic evidence associating
U ND E R L I E T HE M I R 1 3 7 S CH I ZOP H R E NI A VNTR repeat length with SCZ, possibly due to the low allele frequency
ASSOCIATION? of the higher VNTR repeat, which reduces statistical power (Duan
et al., 2014; Egawa et al., 2013; Strazisar et al., 2015; Warburton,
Common-variant GWAS associations for complex traits (like SCZ) are Breen, Rujescu, Bubb, & Quinn, 2015). MIR137 has been identified as a
usually not explained by protein-coding variants but, on the other hand, target gene of Repressor Element-1 Silencing Transcription Factor
they are often highly enriched for regulatory DNA (Hindorff et al., 2009). (REST) (Soldati et al., 2013; Warburton et al., 2015), a key
8 | SAKAMOTO AND CROWLEY

transcriptional regulator of neuron-specific genes (Ballas & Mandel, focus (Baare et al., 2001; Samartzis, Dima, Fusar-Poli, & Kyriakopoulos,
2005; Ooi & Wood, 2007; Roopra, Huang, & Dingledine, 2001; Singh, 2014; van Erp et al., 2016; Voineskos, 2015). We have identified eight
Kagalwala, Parker-Thornburg, Adams, & Majumder, 2008; Tamminga structural brain imaging studies (Cousijn et al., 2014; Kelly et al., 2014;
& Zukin, 2015). Soldati et al. (2013) showed that miR-137 expression is Kuswanto et al., 2015; Lett et al., 2013; Li & Su, 2013; Patel et al., 2015;
low in Huntington’s disease model cells and that its expression Rose et al., 2014; Wright et al., 2016) that specifically tested for an
increases after REST knockdown. Warburton et al. (2015) confirmed association between brain structure and variation in MIR137 (primarily the
binding of REST to the miR-137 promoter by chromatin immunopre- risk tagging SNP rs1625579). Kuswanto et al. (2015) found a significant
cipitation (ChIP). Given the potential involvement of REST in SCZ interaction between MIR137 genotype and white matter integrity in the
pathology, REST-mediated miR-137 regulation could be an important right orbitofrontal region of SCZ cases. Wright et al. (2016) found that SCZ
molecular mechanism (Tamminga & Zukin, 2015). subjects homozygous for the MIR137 risk allele showed significant
Epigenetic mechanisms constitute another source of transcrip- decreases in occipital, parietal, and temporal lobe grey matter concentra-
tional regulation. miR-137 transcription is regulated by three tion. Lett et al. (2013) found that patients with the MIR137 risk genotype
hypomethylation sites (see Figure 2) (Kozaki et al., 2008; Song et al., had reduced white matter integrity throughout the brain, smaller
2013). Several cancer cell studies have reported miR-137 expression hippocampi, and larger lateral ventricles. The five other reports (Kelly
modulation during altered DNA methylation (Chen, Wang et al., 2011; et al., 2014; Li & Su, 2013; Patel et al., 2015; Rose et al., 2014; Wright et al.,
Silber et al., 2008; Zhu et al., 2013). In mouse adult neuronal stem cells, 2016) examined only healthy controls (with the exception of Rose et al.
miR-137 is regulated by the DNA methyl-CpG-binding protein MeCP2 (2014) and Patel et al. (2015) who also included cases) and no clear
and transcription factor Sox2 (Szulwach et al., 2010), supporting the association between the MIR137 variant and brain structure was found.
importance of DNA methylation in miR-137 regulation. Downstream, While the overall pattern of results are mixed and the number of studies is
miR-137 itself controls the expression of multiple epigenetic modula- small, there may be a trend toward MIR137 exerting a greater influence in
tor genes. For example, miR-137 regulates Ezh2, a histone (H3K27) SCZ cases versus healthy controls.
methyltransferase in neuronal stem cells (Szulwach et al., 2010). Sun Functional brain imaging studies have been also performed, most
et al. (2011) demonstrated that miR-137 targets the histone lysine- examining effects of the MIR137 risk SNP rs1625579 in healthy controls
specific demethylase 1 (LSD1), a transcriptional co-repressor of (Liu et al., 2014; Mothersill et al., 2014; van Erp et al., 2014; Whalley
nuclear receptor TLX. TLX, a regulator of neural stem cell self-renewal, et al., 2012). van Erp et al. (2014) found the risk genotype to be
induces feedback loop repression of miR-137 transcription by associated with higher left DLPFC activation. Similarly, Liu et al. (2014)
recruiting LSD1 to the miR-137 promoter (Jiang et al., 2013). showed that individuals homozygous for the risk allele exhibited
Moreover, histone H3K4 demethylase Jarid1b (also known as significantly higher functional connectivity between DLPFC and left
KDM5b), which suppresses transcription through demethylation of hippocampi than heterozygous individuals Further studies found that
transcription start sites, is also targeted by miR-137 during neuronal homozygous risk allele carriers had increased functional connectivity
differentiation of mouse ES cells (Tarantino et al., 2010). Together, between the right amygdala and frontal regions (Mothersill et al., 2014)
these results suggest that epigenetic chromatin modulation is involved and higher activity in the posterior right medial frontal gyrus, BA6
in a miR-137 self-regulation feedback loop. (Whalley et al., 2012). Although most of these studies used healthy
In sum, a growing body of molecular studies are beginning to controls, the results nonetheless demonstrate that individuals carrying
reveal functional DNA variants regulating MIR137 that may explain MIR137 risk alleles tend to have higher activity in some brain areas.
associations with SCZ. Other investigators are exploring the down- In sum, despite relatively small sample sizes and the genotyping of
stream gene network regulated by miR-137, including self-feedback. only one MIR137 SCZ risk SNP, the existing brain imaging studies show
Furthermore, recent studies have isolated physiological stimulators of some promise. Recently, the ENIGMA consortium reported a genomic
MIR137, including metabotropic glutamate receptor stimulation, association study for subcortical brain volume abnormalities in 2,028
seizure activity, and propofal (Fan, Zhou, Qin, & Tao, 2016; Olde SCZ and 2,540 healthy individuals (van Erp et al., 2016). This initial
Loohuis et al., 2015; Schouten et al., 2015). Without question, given subcortical-focused study did not find any association with MIR137,
the diverse activities of this microRNA, further molecular work is but larger studies incorporating cortical measures are underway.
needed to fully understand what regulates miR-137, and what it
regulates, that may be relevant to the etiology of SCZ.
13 | P O T E N T I A L F O R C L I N I C A L U T I L I T Y

12 | D O E S MIR137 V A R I A T I O N I N F L U E N C E MicroRNAs are emerging as useful biomarkers and targets for therapeutic
HUMA N B RAIN STRUCTUR E A ND manipulation in complex diseases. Circulating miRNA levels in plasma,
FU N CTIO N? serum, and whole blood have shown potential as biomarkers of many
disorders (Kawaguchi et al., 2016; Kosaka, Iguchi, & Ochiya, 2010; Witwer,
Structural brain imaging studies have consistently identified changes in 2015) including SCZ (Cairns, 2015; Kichukova, Popov, Ivanov, & Vachev,
SCZ (e.g., altered volume of white matter, hippocampus, amygdala, etc.), 2015; Wang, Wang, Yang, & Huang, 2014). miR-137 has also been
and therefore, linking them with recent genetic findings is a major research suggested as a biomarker of brain cancers and Parkinson’s disease (Koshkin,
SAKAMOTO AND CROWLEY | 9

Chistyakov, Nikitin, Konovalov, Potapov, Usachyov et al., 2014; Li et al., more SCZ-related gene networks. Without question, there is ample
2016, 2017). Interestingly, Wu et al. (2016) recently demonstrated that miR- evidence to continue pursuing this gene and its associated network.
137 levels in peripheral blood cells may have potential for diagnosing early We believe that further investigations in several key areas are
onset SCZ, though replication of this finding is needed. miR-137 needed to fully appreciate the role of MIR137 in SCZ and test its
transcription is regulated by the upstream hypomethylated CpG region diagnostic and therapeutic potential. First, on the genetic level, we
(Kozaki et al., 2008; Song et al., 2013), suggesting that DNA methylation suggest that future GWAS studies move beyond the case-control
status may be another potential biomarker (Gao, Zhang, Breitling, & design, toward a “deep-phenotyping” symptom-dimension approach,
Brenner, 2016; Henriksen, Madsen, Krarup, & Thorlacius-Ussing, 2015; integrating neurocognitive measures, environmental factors, etc as
Kozaki et al., 2008). However, methylome-wide association studies of SCZ proposed by other investigators (Ehrenreich et al., 2016; Insel et al.,
using blood cell DNA have not shown a significant association with miR-137 2010). Second, on the molecular level, we think it is very important to
(Aberg et al., 2014; Dempster et al., 2011). definitively identify the causal genetic variant(s) underlying the SCZ
Since miR-137 is known to modulate brain function, methods that GWAS association. As noted above, the last few years have witnessed
manipulate miR-137 expression could have therapeutic potential in marked progress in this area, but ever more advanced molecular
schizophrenia. miRNAs have been studied as a target of medications, methods are now available and could yield major insights. For example,
and some specific miRNA modulators are currently being studied allele-specific RNA sequencing in the brains of individuals heterozy-
clinically. For example, antagomir-122, a chemically-modified anti- gous for SCZ-risk variants could clarify the direction of effect in this
sense miR-122, is in Phase2 clinical trials for treating infection with locus. Critically, these sequencing efforts should distinguish the host
hepatitis C virus (Lanford et al., 2010). Likewise, a miR-21 inhibitor is gene (MIR137HG) from the pre-miRNA and final, processed miRNA to
undergoing a Phase1 trial for treatment of Alport syndrome, a kidney tease apart transcriptional and post-transcriptional effects. Further-
disease with no currently-approved therapy (Gomez et al., 2015). In more, these studies likely need to be performed across developmental
addition, MRX34, a double-stranded mimic of the naturally occurring stages and distinct cell types (e.g., using single-cell methods) to yield
tumor suppressor miR-34a, is being tested in a Phase1 trial for use with the most informative data. Third, the role of miR-137 in glial cells has
advanced solid tumors (Beg et al., 2014). been somewhat neglected in the pathological and non-pathological
It is theoretically possible that manipulating miR-137 expression with a state, particularly given its role as a tumor suppressor in glioblastoma.
chemically modified miR-137 inhibitor or mimic could have utility in SCZ. Fourth, given that human brain tissue is generally inaccessible, we
There are, however, technical challenges that must be addressed, including believe improved genetic animal models will be a required to truly
achieving cell-type specificity and penetrating the blood-brain barrier understand miR-137 from a mechanistic level. For example, given the
(Krutzfeldt et al., 2005; Price & Chen, 2014; van Rooij, Purcell, & Levin, 2012; embryonic lethality of the constitutive knockout mouse, there is an
Zhang, Wang, & Gemeinhart, 2013). To overcome both of these challenges urgent need for a conditional knockout. Fifth, and finally, we suggest
simultaneously, virus-mediated delivery of a miR-137 modulator represents work toward improving methods for miR-137 manipulation in live
a compelling strategy. Working toward this goal, Murlidharan et al. (2016) animals to gauge utility as a therapeutic target and biomarker. These
recently established a neuron-specific adeno-associated virus capable of might include, for example, the development of brain-penetrating
exclusively delivering a MIR137 specific guide RNA to neuronal cells for antagomirs, enhanced CRISPR/Cas9 mediated manipulation protocols
CRISPR/Cas9-targeted genome editing. Since, intravenous AAV injection or small molecules with specific effects on the miR-137 network. In
can deliver genes to the brain (Murlidharan, Samulski, & Asokan, 2014; conclusion, MIR137 remains a prominent SCZ risk gene with potential
Schuster et al., 2014; Shen et al., 2013), this strategy could be useful for the to open a window into the complex etiology of SCZ as the field works
manipulation of miR-137 activity by delivering a miR-137 overexpression toward developing novel treatments for this devastating disorder.
vector, an antisense inhibitor, or a MIR137 specific guide RNA capable of
recruiting artificial transcription regulators (e.g., dCas9-VP16/–KRAB) to the
ACKNOWLEDGMENTS
MIR137 promoter in the neuronal cell (Genga, Kearns, & Maehr, 2016; Vora,
Tuttle, Cheng, & Church, 2016). The authors wish to thank Drs. Patrick Sullivan, Jin Szatkiewicz, and
Praveen Sethupathy for helpful discussions. Dr. Sakamoto was supported
by NIH grants R21MH099370 and R21MH102814. Dr. Crowley is
14 | C O N C L U S I O N S AN D S U GG E S TI O N S supported by NIH grants R01MH105500 and R01MH110427.
FO R FU TU R E RE S E A R C H

We have attempted to comprehensively review the genetic and CONFLICT OF I NTEREST


biological evidence supporting a role for miR-137 in the etiology of
The authors have no interests to disclose.
SCZ. We conclude that the evidence is strong, but mechanistic details
are lacking and, since SCZ is highly polygenic, at the level of the
individual miR-137’s role may be small. As noted in this review, REFERENCES
however, miR-137 clearly regulates a number of other SCZ risk genes Aberg, K. A., McClay, J. L., Nerella, S., Clark, S., Kumar, G., Chen, W., . . . van
and therefore may represent an important signaling node within one or den Oord, E. J. (2014). Methylome-wide association study of
10 | SAKAMOTO AND CROWLEY

schizophrenia: Identifying blood biomarker signatures of environmental Chen, Q., Chen, X., Zhang, M., Fan, Q., Luo, S., & Cao, X. (2011). MiR-137 is
insults. JAMA Psychiatry, 71(3), 255–264. frequently down-regulated in gastric cancer and is a negative regulator
Achim, A. M., Maziade, M., Raymond, E., Olivier, D., Merette, C., & Roy, of Cdc42. Digestive Diseases and Sciences, 56(7), 2009–2016.
M. A. (2011). How prevalent are anxiety disorders in schizophrenia? A Chen, X., Wang, J., Shen, H., Lu, J., Li, C., Hu, D. N., . . . Tu, L. (2011).
meta-analysis and critical review on a significant association. Epigenetics, microRNAs, and carcinogenesis: Functional role of micro-
Schizophrenia Bulletin, 37(4), 811–821. RNA-137 in uveal melanoma. Investigative Ophthalmology and Visual
Agarwal, V., Bell, G. W., Nam, J. W., & Bartel, D. P. (2015). Predicting Science, 52(3), 1193–1199.
effective microRNA target sites in mammalian mRNAs. eLife, 4, e05005. Chendrimada, T. P., Gregory, R. I., Kumaraswamy, E., Norman, J., Cooch, N.,
Allen, N. C., Bagade, S., McQueen, M. B., Ioannidis, J. P., Kavvoura, F. K., Nishikura, K., & Shiekhattar, R. (2005). TRBP recruits the Dicer complex
Khoury, M. J., . . . Bertram, L. (2008). Systematic meta-analyses and field to Ago2 for microRNA processing and gene silencing. Nature,
synopsis of genetic association studies in schizophrenia: The SzGene 436(7051), 740–744.
database. Nature Genetics, 40(7), 827–834. Cheng, H. Y., Papp, J. W., Varlamova, O., Dziema, H., Russell, B., Curfman,
Baare, W. F., Hulshoff Pol, H. E., Boomsma, D. I., Posthuma, D., de Geus, J. P., . . . Obrietan, K. (2007). MicroRNA modulation of circadian-clock
E. J., Schnack, H. G., . . . Kahn, R. S. (2001). Quantitative genetic period and entrainment. Neuron, 54(5), 813–829.
modeling of variation in human brain morphology. Cerebral Cortex, Cheng, L. C., Pastrana, E., Tavazoie, M., & Doetsch, F. (2009). MiR-124
11(9), 816–824. regulates adult neurogenesis in the subventricular zone stem cell niche.
Balaguer, F., Link, A., Lozano, J. J., Cuatrecasas, M., Nagasaka, T., Boland, Nature Neuroscience, 12(4), 399–408.
C. R., & Goel, A. (2010). Epigenetic silencing of miR-137 is an early event Collins, A. L., Kim, Y., Bloom, R. J., Kelada, S. N., Sethupathy, P., & Sullivan,
in colorectal carcinogenesis. Cancer Research, 70(16), 6609–6618. P. F. (2014). Transcriptional targets of the schizophrenia risk gene
Ballas, N., Mandel, G. (2005). The many faces of REST oversee epigenetic MIR137. Translational Psychiatry, 4, e404.
programming of neuronal genes. Current Opinion in Neurobiology, 15(5), Consortium, E. P. (2012). An integrated encyclopedia of DNA elements in
500–506. the human genome. Nature, 489(7414), 57–74.
Bandres, E., Agirre, X., Bitarte, N., Ramirez, N., Zarate, R., Roman-Gomez, J., Consortium, G. T. (2015). Human genomics. The Genotype-Tissue
. . . Garcia-Foncillas, J. (2009). Epigenetic regulation of microRNA Expression (GTEx) pilot analysis: Multitissue gene regulation in humans.
expression in colorectal cancer. International Journal of Cancer Journal Science, 348(6235), 648–660.
International du Cancer, 125(11), 2737–2743. Cosgrove, D., Harold, D., Mothersill, O., Anney, R., Hill, M. J., Bray, N. J., . . .
Bartel, D. P. (2004). MicroRNAs: Genomics, biogenesis, mechanism, and Donohoe, G. (2017). MiR-137-derived polygenic risk: Effects on
function. Cell, 116(2), 281–297. cognitive performance in patients with schizophrenia and controls.
Beg, M. S., Borad, M., Sachdev, J., Hong, D. S., Smith, S., Bader, A., . . . Translational Psychiatry, 7(1), e1012.
Brenner, A. (2014). Abstract CT327: Multicenter phase I study of Cotter, D. R., Pariante, C. M., & Everall, I. P. (2001). Glial cell abnormalities in
MRX34, a first-in-class microRNA miR-34 mimic liposomal injection. major psychiatric disorders: The evidence and implications. Brain
Cancer Research 74(19):CT327–CT327. Research Bulletin, 55(5), 585–595.
Bemis, L. T., Chen, R., Amato, C. M., Classen, E. H., Robinson, S. E., Coffey, Cousijn, H., Eissing, M., Fernandez, G., Fisher, S. E., Franke, B., Zwiers, M., . . .
D. G., . . . Robinson, W. A. (2008). MicroRNA-137 targets micro- Arias-Vasquez, A. (2014). No effect of schizophrenia risk genes
phthalmia-associated transcription factor in melanoma cell lines. Cancer MIR137, TCF4, and ZNF804A on macroscopic brain structure.
Research, 68(5), 1362–1368. Schizophrenia Research, 159(2–3), 329–332.
Bennett, P. E., Bemis, L., Norris, D. A., & Shellman, Y. G. (2013). MiR in Cross-Disorder Group of the Psychiatric Genomics Consortium (2013).
melanoma development: MiRNAs and acquired hallmarks of cancer in Identification of risk loci with shared effects on five major psychiatric
melanoma. Physiological Genomics, 45(22), 1049–1059. disorders: A genome-wide analysis. The Lancet, 381(9875),
Bernstein, H. G., Steiner, J., Guest, P. C., Dobrowolny, H., & Bogerts, B. 1371–1379.
(2015). Glial cells as key players in schizophrenia pathology: Recent Crowley, J. J., Collins, A. L., Lee, R. J., Nonneman, R. J., Farrell, M. S.,
insights and concepts of therapy. Schizophrenia Research, 161(1), Ancalade, N., . . . Sullivan, P. F. (2015). Disruption of the MicroRNA 137
4–18. primary transcript results in early embryonic lethality in mice. Biological
Betel, D., Wilson, M., Gabow, A., Marks, D. S., & Sander, C. (2008). The Psychiatry, 77(2), 5–7.
microRNA.org resource: Targets and expression. Nucleic Acids Research Cummings, E., Donohoe, G., Hargreaves, A., Moore, S., Fahey, C.,
36(Database issue), D149–D153. Dinan, T. G., . . . Corvin, A. (2013). Mood congruent psychotic
Beveridge, N. J., Tooney, P. A., Carroll, A. P., Gardiner, E., Bowden, N., Scott, symptoms and specific cognitive deficits in carriers of the novel
R. J., . . . Cairns, M. J. (2008). Dysregulation of miRNA 181b in the schizophrenia risk variant at MIR-137. Neuroscience Letters, 532,
temporal cortex in schizophrenia. Human Molecular Genetics, 17(8), 33–38.
1156–1168. D’Angelo, C. S., Moller Dos Santos, M. F., Alonso, L. G., & Koiffmann, C. P.
Bian, S., Xu, T. L., & Sun, T. (2013). Tuning the cell fate of neurons and glia by (2015). Two new cases of 1p21.3 deletions and an unbalanced
microRNAs. Current Opinion in Neurobiology, 23(6), 928–934. translocation t(8;12) among individuals with syndromic obesity.
Boudreau, R. L., Jiang, P., Gilmore, B. L., Spengler, R. M., Tirabassi, R., Molecular Syndromology, 6(2), 63–70.
Nelson, J. A., . . . Davidson, B. L. (2014). Transcriptome-wide discovery Dacic, S., Kelly, L., Shuai, Y., & Nikiforova, M. N. (2010). MiRNA expression
of microRNA binding sites in human brain. Neuron, 81(2), 294–305. profiling of lung adenocarcinomas: Correlation with mutational status.
Cairns, M. J. (2015). Circulating miRNA biomarkers for schizophrenia? Modern Pathology, 23(12), 1577–1582.
American Journal of Psychiatry, 172(11), 1059–1061. de Bartolomeis, A., Buonaguro, E. F., & Iasevoli, F. (2013). Serotonin-
Carter, M. T., Nikkel, S. M., Fernandez, B. A., Marshall, C. R., Noor, A., Lionel, glutamate and serotonin-dopamine reciprocal interactions as putative
A. C., . . . Scherer, S. W. (2011). Hemizygous deletions on chromosome molecular targets for novel antipsychotic treatments: From receptor
1p21.3 involving the DPYD gene in individuals with autism spectrum heterodimers to postsynaptic scaffolding and effector proteins.
disorder. Clinical Genetics, 80(5), 435–443. Psychopharmacology, 225(1), 1–19.
Chen, L., Wang, X., Wang, H., Li, Y., Yan, W., Han, L., . . . Jiang, C. (2012). Dempster, E. L., Pidsley, R., Schalkwyk, L. C., Owens, S., Georgiades, A.,
MiR-137 is frequently down-regulated in glioblastoma and is a Kane, F., . . . Mill, J. (2011). Disease-associated epigenetic changes in
negative regulator of Cox-2. European Journal of Cancer, 48(16), monozygotic twins discordant for schizophrenia and bipolar disorder.
3104–3111. Human Molecular Genetics, 20(24), 4786–4796.
SAKAMOTO AND CROWLEY | 11

Denli, A. M., Tops, B. B., Plasterk, R. H., Ketting, R. F., & Hannon, G. J. (2004). Glantz, L. A., Lewis, D. A. (2000). Decreased dendritic spine density on
Processing of primary microRNAs by the Microprocessor complex. prefrontal cortical pyramidal neurons in schizophrenia. Archives of
Nature, 432(7014), 231–235. General Psychiatry, 57(1), 65–73.
Duan, J., Shi, J., Fiorentino, A., Leites, C., Chen, X., Moy, W., . . . Gejman, P. V. Glausier, J. R., Lewis, D. A. (2013). Dendritic spine pathology in
(2014). A rare functional noncoding variant at the GWAS-implicated schizophrenia. Neuroscience, 251, 90–107.
MIR137/MIR2682 locus might confer risk to schizophrenia and bipolar Goes, F. S., McGrath, J., Avramopoulos, D., Wolyniec, P., Pirooznia, M.,
disorder. American Journal of Human Genetics, 95(6), 744–753. Ruczinski, I., . . . Pulver, A. E. (2015). Genome-wide association study of
Dzyubenko, E., Gottschling, C., & Faissner, A. (2016). Neuron-Glia schizophrenia in Ashkenazi Jews. American journal of medical genetics
interactions in neural plasticity: Contributions of neural extracellular Part B, Neuropsychiatric genetics: the official publication of the
matrix and perineuronal nets. Neural Plasticity, 2016, 5214961. International Society of Psychiatric Genetics, 168(8), 649–659.
Edbauer, D., Neilson, J. R., Foster, K. A., Wang, C. F., Seeburg, D. P., Gomez, I. G., MacKenna, D. A., Johnson, B. G., Kaimal, V., Roach, A. M., Ren,
Batterton, M. N., . . . Sheng, M. (2010). Regulation of synaptic structure S., . . . Duffield, J. S. (2015). Anti-microRNA-21 oligonucleotides prevent
and function by FMRP-associated microRNAs miR-125b and miR-132. Alport nephropathy progression by stimulating metabolic pathways.
Neuron, 65(3), 373–384. The Journal of Clinical Investigation, 125(1), 141–156.
Egawa, J., Nunokawa, A., Shibuya, M., Watanabe, Y., Kaneko, N., Igeta, H., & Goudriaan, A., de Leeuw, C., Ripke, S., Hultman, C. M., Sklar, P., Sullivan,
Someya, T. (2013). Resequencing and association analysis of MIR137 P. F., . . . Verheijen, M. H. (2014). Specific glial functions contribute
with schizophrenia in a Japanese population. Psychiatry and Clinical to schizophrenia susceptibility. Schizophrenia Bulletin, 40(4),
Neurosciences, 67(4), 277–279. 925–935.
Ehrenreich, H., Mitjans, M., Van der Auwera, S., Centeno, T. P., Begemann, Gregory, R. I., Yan, K. P., Amuthan, G., Chendrimada, T., Doratotaj, B.,
M., Grabe, H. J., . . . Nave, K. A. (2016). OTTO: A new strategy to extract Cooch, N., & Shiekhattar, R. (2004). The Microprocessor complex
mental disease-relevant combinations of GWAS hits from individuals. mediates the genesis of microRNAs. Nature, 432(7014), 235–240.
Molecular Psychiatry, 00, 1–11. Guella, I., Sequeira, A., Rollins, B., Morgan, L., Torri, F., van Erp, T. G., . . .
Fan, J., Zhou, Q., Qin, Z., & Tao, T. (2016). Effect of propofol on microRNA Vawter, M. P. (2013). Analysis of miR-137 expression and rs1625579 in
expression in rat primary embryonic neural stem cells. BMC Anesthesi- dorsolateral prefrontal cortex. Journal of Psychiatric Research, 47(9),
ology, 16(1), 95. 1215–1221.
Fogarty, M. P., Cannon, M. E., Vadlamudi, S., Gaulton, K. J., & Mohlke, K. L. Guo, Z., Maki, M., Ding, R., Yang, Y., Zhang, B., & Xiong, L. (2014). Genome-
(2014). Identification of a regulatory variant that binds FOXA1 and wide survey of tissue-specific microRNA and transcription factor
FOXA2 at the CDC123/CAMK1D type 2 diabetes GWAS locus. PLoS regulatory networks in 12 tissues. Scientific Reports, 4, 5150.
Genetics, 10(9), 1004633. Han, J., Lee, Y., Yeom, K. H., Nam, J. W., Heo, I., Rhee, J. K., . . . Kim, V. N.
Forstemann, K., Tomari, Y., Du, T., Vagin, V. V., Denli, A. M., Bratu, D. P., . . . (2006). Molecular basis for the recognition of primary microRNAs by
Zamore, P. D. (2005). Normal microRNA maturation and germ-line stem the Drosha-DGCR8 complex. Cell, 125(5), 887–901.
cell maintenance requires Loquacious, a double-stranded RNA-binding Hansen, K. F., Sakamoto, K., Wayman, G. A., Impey, S., & Obrietan, K.
domain protein. PLoS Biology, 3(7), 236. (2010). Transgenic miR132 alters neuronal spine density and impairs
Friedman, L. M., Dror, A. A., Mor, E., Tenne, T., Toren, G., Satoh, T., . . . novel object recognition memory. PLoS ONE, 5(11), e15497.
Avraham, K. B. (2009). MicroRNAs are essential for development and Hao, S., Luo, C., Abukiwan, A., Wang, G., He, J., Huang, L., . . . He, D. (2015).
function of inner ear hair cells in vertebrates. Proceedings of the National MiR-137 inhibits proliferation of melanoma cells by targeting PAK2.
Academy of Sciences of the United States of America, 106(19), Experimental Dermatology, 24(12), 947–952.
7915–7920. Hart, A. B., Gamazon, E. R., Engelhardt, B. E., Sklar, P., Kahler, A. K., Hultman,
Fromer, M., Roussos, P., Sieberts, S. K., Johnson, J. S., Kavanagh, D. H., C. M., . . . Palmer, A. A. (2014). Genetic variation associated with
Perumal, T. M., . . . Sklar, P. (2016). Gene expression elucidates euphorigenic effects of d-amphetamine is associated with diminished
functional impact of polygenic risk for schizophrenia. Nature risk for schizophrenia and attention deficit hyperactivity disorder.
Neuroscience, 19(11), 1442–1453. Proceedings of the National Academy of Sciences of the United States of
Gao, X., Zhang, Y., Breitling, L. P., & Brenner, H. (2016). Tobacco smoking America, 111(16), 5968–5973.
and methylation of genes related to lung cancer development. Hebert, S. S., De Strooper, B. (2007). Molecular biology. miRNAs in
Oncotarget, 7(37), 59017–59028. neurodegeneration. Science, 317(5842), 1179–1180.
Garey, L. J., Ong, W. Y., Patel, T. S., Kanani, M., Davis, A., Mortimer, A. M., . . . Henriksen, S. D., Madsen, P. H., Krarup, H., & Thorlacius-Ussing, O. (2015).
Hirsch, S. R. (1998). Reduced dendritic spine density on cerebral cortical DNA hypermethylation as a blood-based marker for pancreatic cancer:
pyramidal neurons in schizophrenia. Journal of Neurology, Neurosurgery, A literature review. Pancreas, 44(7), 1036–1045.
and Psychiatry, 65(4), 446–453. Hill, M. J., Donocik, J. G., Nuamah, R. A., Mein, C. A., Sainz-Fuertes, R., &
Genga, R. M., Kearns, N. A., & Maehr, R. (2016). Controlling transcription in Bray, N. J. (2014). Transcriptional consequences of schizophrenia
human pluripotent stem cells using CRISPR-effectors. Methods, 101, candidate miR-137 manipulation in human neural progenitor cells.
36–42. Schizophrenia Research, 153(1–3), 225–230.
Genovese, G., Fromer, M., Stahl, E. A., Ruderfer, D. M., Chambert, K., Hindorff, L. A., Sethupathy, P., Junkins, H. A., Ramos, E. M., Mehta, J. P.,
Landen, M., . . . McCarroll, S. A. (2016). Increased burden of ultra-rare Collins, F. S., & Manolio, T. A. (2009). Potential etiologic and functional
protein-altering variants among 4,877 individuals with schizophrenia. implications of genome-wide association loci for human diseases and
Nature Neuroscience, 19(11), 1433–1441. traits. Proceedings of the National Academy of Sciences of the United
Giacomotto, J., Carroll, A. P., Rinkwitz, S., Mowry, B., Cairns, M. J., & Becker, States of America, 106(23), 9362–9367.
T. S. (2016). Developmental suppression of schizophrenia-associated Hof, P. R., Haroutunian, V., Copland, C., Davis, K. L., & Buxbaum, J. D.
miR-137 alters sensorimotor function in zebrafish. Translational (2002). Molecular and cellular evidence for an oligodendrocyte
Psychiatry, 6, e818. abnormality in schizophrenia. Neurochemical Research, 27(10),
Gianfrancesco, O., Griffiths, D., Myers, P., Collier, D. A., Bubb, V. J., & 1193–1200.
Quinn, J. P. (2016). Identification and potential regulatory properties Hollins, S. L., Goldie, B. J., Carroll, A. P., Mason, E. A., Walker, F. R.,
of evolutionary conserved regions (ECRs) at the schizophrenia- Eyles, D. W., & Cairns, M. J. (2014). Ontogeny of small RNA in the
associated MIR137 locus. Journal of Molecular Neuroscience, 60(2), regulation of mammalian brain development. BMC Genomics,
239–247. 15, 777.
12 | SAKAMOTO AND CROWLEY

Hong, P., Jiang, M., & Li, H. (2014). Functional requirement of dicer1 and Kinros, J., Reichenberg, A., & Frangou, S. (2010). The neurodevelopmental
miR-17-5p in reactive astrocyte proliferation after spinal cord injury in theory of schizophrenia: Evidence from studies of early onset cases. The
the mouse. Glia, 62(12), 2044–2060. Israel Journal of Psychiatry and Related Sciences, 47(2), 110–117.
Howng, S. Y., Huang, Y., Ptacek, L., & Fu, Y. H. (2015). Understanding the Klann, E., Dever, T. E. (2004). Biochemical mechanisms for translational
role of dicer in astrocyte development. PLoS ONE, 10(5), e0126667. regulation in synaptic plasticity. Nature Reviews Neuroscience, 5(12),
Huang, Z. M., Yang, J., Shen, X. Y., Zhang, X. Y., Meng, F. S., Xu, J. T., . . . Gao, 931–942.
H. J. (2009). MicroRNA expression profile in non-cancerous colonic Klein, M. E., Lioy, D. T., Ma, L., Impey, S., Mandel, G., & Goodman, R. H.
tissue associated with lymph node metastasis of colon cancer. Journal of (2007). Homeostatic regulation of MeCP2 expression by a CREB-
Digestive Diseases, 10(3), 188–194. induced microRNA. Nature Neuroscience, 10(12), 1513–1514.
Huber, K. M., Kayser, M. S., & Bear, M. F. (2000). Role for rapid dendritic Kosaka, N., Iguchi, H., & Ochiya, T. (2010). Circulating microRNA in body
protein synthesis in hippocampal mGluR-dependent long-term depres- fluid: A new potential biomarker for cancer diagnosis and prognosis.
sion. Science, 288(5469), 1254–1257. Cancer Science, 101(10), 2087–2092.
Impey, S., Davare, M., Lesiak, A., Fortin, D., Ando, H., Varlamova, O., . . . Koshkin, F. A., Chistyakov, D. A., Nikitin, A. G., Konovalov, A. N., Potapov,
Wayman, G. A. (2010). An activity-induced microRNA controls dendritic A. A., Usachyov, D. Y., . . . Chekhonin, V. P. (2014a). Profile of microRNA
spine formation by regulating Rac1-PAK signaling. Molecular and expression in brain tumors of different malignancy. Bulletin of
Cellular Neurosciences, 43(1), 146–156. Experimental Biology and Medicine, 157(6), 794–797.
Insel, T., Cuthbert, B., Garvey, M., Heinssen, R., Pine, D. S., Quinn, K., . . . Koshkin, P. A., Chistiakov, D. A., Nikitin, A. G., Konovalov, A. N.,
Wang, P. (2010). Research domain criteria (RDoC): Toward a new Potapov, A. A., Usachev, D. Y., . . . Chekhonin, V. P. (2014b).
classification framework for research on mental disorders. American Analysis of expression of microRNAs and genes involved in the
Journal of Psychiatry, 167(7), 748–751. control of key signaling mechanisms that support or inhibit
Jiang, K., Ren, C., & Nair, V. D. (2013). MicroRNA-137 represses Klf4 and development of brain tumors of different grades. Clinica Chimica
Tbx3 during differentiation of mouse embryonic stem cells. Stem Cell Acta, 430, 55–62.
Research, 11(3), 1299–1313. Kozaki, K., Imoto, I., Mogi, S., Omura, K., & Inazawa, J. (2008). Exploration of
Jo, W. K., Law, A. C., & Chung, S. K. (2014). The neglected co-star in the tumor-suppressive microRNAs silenced by DNA hypermethylation in
dementia drama: The putative roles of astrocytes in the pathogeneses oral cancer. Cancer Research, 68(7), 2094–2105.
of major neurocognitive disorders. Molecular Psychiatry, 19(2), Krutzfeldt, J., Rajewsky, N., Braich, R., Rajeev, K. G., Tuschl, T., Manoharan,
159–167. M., & Stoffel, M. (2005). Silencing of microRNAs in vivo with
Karthikeyan, A., Patnala, R., Jadhav, S. P., Eng-Ang, L., & Dheen, S. T. (2016). ’antagomirs’. Nature, 438(7068), 685–689.
MicroRNAs: Key players in microglia and astrocyte mediated inflam- Kumar, S., Hedges, S. B. (1998). A molecular timescale for vertebrate
mation in CNS pathologies. Current Medicinal Chemistry, 23(30), evolution. Nature, 392(6679), 917–920.
3528–3546. Kuswanto, C. N., Sum, M. Y., Qiu, A., Sitoh, Y. Y., Liu, J., & Sim, K. (2015).
Kawaguchi, T., Komatsu, S., Ichikawa, D., Tsujiura, M., Takeshita, H., The impact of genome wide supported microRNA-137 (MIR137) risk
Hirajima, S., . . . Otsuji, E. (2016). Circulating MicroRNAs: A next- variants on frontal and striatal white matter integrity, neurocognitive
generation clinical biomarker for digestive system cancers. International functioning, and negative symptoms in schizophrenia. American
Journal of Molecular Sciences, 17(9). Journal of Medical Genetics Part B, Neuropsychiatric Genetics: The
Kawase-Koga, Y., Otaegi, G., & Sun, T. (2009). Different timings of Dicer Official Publication of the International Society of Psychiatric Genetics,
deletion affect neurogenesis and gliogenesis in the developing mouse 168B(5), 317–326.
central nervous system. Developmental Dynamics, 238(11), Kwon, E., Wang, W., & Tsai, L. H. (2013). Validation of schizophrenia-
2800–2812. associated genes CSMD1, C10orf26, CACNA1C and TCF4 as miR-137
Kelly, S., Morris, D. W., Mothersill, O., Rose, E. J., Fahey, C., O’Brien, C., . . . targets. Molecular Psychiatry, 18(1), 11–12.
Donohoe, G. (2014). Genome-wide schizophrenia variant at MIR137 Lagos-Quintana, M., Rauhut, R., Lendeckel, W., & Tuschl, T. (2001).
does not impact white matter microstructure in healthy participants. Identification of novel genes coding for small expressed RNAs. Science,
Neuroscience Letters, 574, 6–10. 294(5543), 853–858.
Kerns, D., Vong, G. S., Barley, K., Dracheva, S., Katsel, P., Casaccia, P., . . . Lagos-Quintana, M., Rauhut, R., Yalcin, A., Meyer, J., Lendeckel, W., &
Byne, W. (2010). Gene expression abnormalities and oligodendrocyte Tuschl, T. (2002). Identification of tissue-specific microRNAs from
deficits in the internal capsule in schizophrenia. Schizophrenia Research, mouse. Current Biology, 12(9), 735–739.
120(1–3), 150–158. Landgraf, P., Rusu, M., Sheridan, R., Sewer, A., Iovino, N., Aravin, A., . . .
Kichukova, T. M., Popov, N. T., Ivanov, H. Y., & Vachev, T. I. (2015). Circulating Tuschl, T. (2007). A mammalian microRNA expression atlas based on
microRNAs as a novel class of potential diagnostic biomarkers in small RNA library sequencing. Cell, 129(7), 1401–1414.
neuropsychiatric disorders. Folia Medica, 57(3–4), 159–172. Lanford, R. E., Hildebrandt-Eriksen, E. S., Petri, A., Persson, R., Lindow, M.,
Kim, A. H., Parker, E. K., Williamson, V., McMichael, G. O., Fanous, A. H., & Munk, M. E., . . . Orum, H. (2010). Therapeutic silencing of
Vladimirov, V. I. (2012). Experimental validation of candidate schizo- microRNA-122 in primates with chronic hepatitis C virus infection.
phrenia gene ZNF804A as target for hsa-miR-137. Schizophrenia Science, 327(5962), 198–201.
Research, 141(1), 60–64. Langevin, S. M., Stone, R. A., Bunker, C. H., Lyons-Weiler, M. A.,
Kim, J., Inoue, K., Ishii, J., Vanti, W. B., Voronov, S. V., Murchison, E., . . . LaFramboise, W. A., Kelly, L., . . . Taioli, E. (2011). MicroRNA-137
Abeliovich, A. (2007). A MicroRNA feedback circuit in midbrain promoter methylation is associated with poorer overall survival in
dopamine neurons. Science, 317(5842), 1220–1224. patients with squamous cell carcinoma of the head and neck. Cancer,
Kim, S., Webster, M. J. (2010). Correlation analysis between genome- 117(7), 1454–1462.
wide expression profiles and cytoarchitectural abnormalities in the Lett, T. A., Chakravarty, M. M., Felsky, D., Brandl, E. J., Tiwari, A. K.,
prefrontal cortex of psychiatric disorders. Molecular Psychiatry, 15(3), Goncalves, V. F., . . . Voineskos, A. N. (2013). The genome-wide
326–336. supported microRNA-137 variant predicts phenotypic heterogeneity
Kim, V. N., Han, J., & Siomi, M. C. (2009). Biogenesis of small RNAs in within schizophrenia. Molecular Psychiatry, 18(4), 443–450.
animals. Nature Reviews. Molecular Cell Biology, 10(2), 126–139. Lewis, B. P., Burge, C. B., & Bartel, D. P. (2005). Conserved seed pairing,
Kim, Y. K., Kim, V. N. (2007). Processing of intronic microRNAs. EMBO often flanked by adenosines, indicates that thousands of human genes
Journal, 26(3), 775–783. are microRNA targets. Cell, 120(1), 15–20.
SAKAMOTO AND CROWLEY | 13

Li, H. Y., Li, Y. M., Li, Y., Shi, X. W., & Chen, H. (2016). Circulating microRNA- Packer, A. N., Xing, Y., Harper, S. Q., Jones, L., & Davidson, B. L. (2008). The
137 is a potential biomarker for human glioblastoma. European Review bifunctional microRNA miR-9/miR-9* regulates REST and CoREST and
for Medical and Pharmacological Sciences, 20(17), 3599–3604. is downregulated in Huntington’s disease. The Journal of Neuroscience:
Li, M., Su, B. (2013). Impact of the genome-wide schizophrenia risk single The Official Journal of the Society for Neuroscience, 28(53),
nucleotide polymorphism (rs1625579) in miR-137 on brain structures in 14341–14346.
healthy individuals. Psychiatric Genetics, 23(6), 267. Patel, V. S., Kelly, S., Wright, C., Gupta, C. N., Arias-Vasquez, A., Perrone-
Li, N., Pan, X., Zhang, J., Ma, A., Yang, S., Ma, J., & Xie, A. (2017). Plasma Bizzozero, N., . . . Turner, J. A. (2015). MIR137HG risk variant
levels of miR-137 and miR-124 are associated with Parkinson’s disease rs1625579 genotype is related to corpus callosum volume in
but not with Parkinson’s disease with depression. Neurological Sciences, schizophrenia. Neuroscience Letters, 602, 44–49.
38(5), 761–767. Pinto, D., Delaby, E., Merico, D., Barbosa, M., Merikangas, A., Klei, L., . . .
Liu, B., Zhang, X., Hou, B., Li, J., Qiu, C., Qin, W., . . . Jiang, T. (2014). The Scherer, S. W. (2014). Convergence of genes and cellular pathways
impact of MIR137 on dorsolateral prefrontal-hippocampal functional dysregulated in autism spectrum disorders. American Journal of Human
connectivity in healthy subjects. Neuropsychopharmacology: Official Genetics, 94(5), 677–694.
Publication of the American College of Neuropsychopharmacology, 39(9), Ponomarev, E. D., Veremeyko, T., Barteneva, N., Krichevsky, A. M., &
2153–2160. Weiner, H. L. (2011). MicroRNA-124 promotes microglia quiescence
Liu, M., Lang, N., Qiu, M., Xu, F., Li, Q., Tang, Q., . . . Bi, F. (2011). MiR-137 and suppresses EAE by deactivating macrophages via the C/EBP-alpha-
targets Cdc42 expression, induces cell cycle G1 arrest and inhibits PU.1 pathway. Nature Medicine, 17(1), 64–70.
invasion in colorectal cancer cells. International Journal of Cancer Journal Potkin, S. G., Macciardi, F., Guffanti, G., Fallon, J. H., Wang, Q., Turner, J. A.,
International du Cancer, 128(6), 1269–1279. . . . Xie, X. (2010). Identifying gene regulatory networks in schizophre-
Liu, N., Okamura, K., Tyler, D. M., Phillips, M. D., Chung, W. J., & Lai, E. C. nia. Neuroimage, 53(3), 839–847.
(2008). The evolution and functional diversification of animal microRNA Price, C., Chen, J. (2014). MicroRNAs in cancer biology and therapy: Current
genes. Cell Research, 18(10), 985–996. status and perspectives. Genes and Diseases, 1(1), 53–63.
Ludwig, N., Leidinger, P., Becker, K., Backes, C., Fehlmann, T., Pallasch, C., Rapoport, J. L., Giedd, J. N., & Gogtay, N. (2012). Neurodevelopmental
. . . Keller, A. (2016). Distribution of miRNA expression across human model of schizophrenia: Update 2012. Molecular Psychiatry, 17(12),
tissues. Nucleic Acids Research, 44(8), 3865–3877. 1228–1238.
Mahmoudi, E., Cairns, M. J. (2017). MiR-137: An important player in neural Regenold, W. T., Phatak, P., Marano, C. M., Gearhart, L., Viens, C. H.,
development and neoplastic transformation. Molecular Psychiatry, & Hisley, K. C. (2007). Myelin staining of deep white matter in the
22(1), 44–55. dorsolateral prefrontal cortex in schizophrenia, bipolar disorder,
Mamdani, M., McMichael, G. O., Gadepalli, V., Williamson, V., Parker, E. K., and unipolar major depression. Psychiatry Research, 151(3),
Haroutunian, V., & Vladimirov, V. I. (2013). Differential regulation of 179–188.
schizophrenia-associated microRNA gene function by variable Remenyi, J., Hunter, C. J., Cole, C., Ando, H., Impey, S., Monk, C. E., . . .
number tandem repeats (VNTR) polymorphism. Schizophrenia Research, Arthur, J. S. (2010). Regulation of the miR-212/132 locus by MSK1 and
151(1–3), 284–286. CREB in response to neurotrophins. Biochemical Journal, 428(2),
Merico, D., Costain, G., Butcher, N. J., Warnica, W., Ogura, L., Alfred, S. E., 281–291.
. . . Bassett, A. S. (2014). MicroRNA dysregulation, gene networks, and Ren, X., Bai, X., Zhang, X., Li, Z., Tang, L., Zhao, X., . . . Ji, J. (2015).
risk for schizophrenia in 22q11.2 deletion syndrome. Frontiers in Quantitative nuclear proteomics identifies that miR-137-mediated
Neurology, 5, 238. EZH2 reduction regulates resveratrol-induced apoptosis of neuroblas-
Miller, J. A., Ding, S. L., Sunkin, S. M., Smith, K. A., Ng, L., Szafer, A., . . . Lein, toma cells. Molecular & Cellular Proteomics, 14(2), 316–328.
E. S. (2014). Transcriptional landscape of the prenatal human brain. Ripke, S. (2013). Genome-wide association analysis identifies 13 new risk
Nature, 508(7495), 199–206. loci for schizophrenia. Nature Genetics, 45, 1150–1159.
Moghaddam, B., Javitt, D. (2012). From revolution to evolution: The Roopra, A., Huang, Y., & Dingledine, R. (2001). Neurological disease:
glutamate hypothesis of schizophrenia and its implication for treatment. Listening to gene silencers. Molecular Interventions, 1(4), 219–228.
Neuropsychopharmacology: Official Publication of the American College of Rose, E. J., Morris, D. W., Fahey, C., Cannon, D., McDonald, C., Scanlon, C.,
Neuropsychopharmacology, 37(1), 4–15. . . . Donohoe, G. (2014). The miR-137 schizophrenia susceptibility
Mothersill, O., Morris, D. W., Kelly, S., Rose, E. J., Fahey, C., O’Brien, C., . . . variant rs1625579 does not predict variability in brain volume in a
Donohoe, G. (2014). Effects of MIR137 on fronto-amygdala functional sample of schizophrenic patients and healthy individuals. American
connectivity. Neuroimage, 90, 189–195. Journal of Medical Genetics Part B, Neuropsychiatric Genetics: The Official
Murlidharan, G., Sakamoto, K., Rao, L., Corriher, T., Wang, D., Gao, G., . . . Publication of the International Society of Psychiatric Genetics, 165B,
Asokan, A. (2016). CNS-restricted transduction and CRISPR/Cas9- 467–471.
mediated gene deletion with an engineered AAV vector. Molecular Samartzis, L., Dima, D., Fusar-Poli, P., & Kyriakopoulos, M. (2014). White
Therapy. Nucleic Acids, 5(7), e338. matter alterations in early stages of schizophrenia: A systematic review
Murlidharan, G., Samulski, R. J., & Asokan, A. (2014). Biology of adeno- of diffusion tensor imaging studies. Journal of Neuroimaging, 24(2),
associated viral vectors in the central nervous system. Frontiers in 101–110.
Molecular Neuroscience, 7, 76. Santos, M. C., Tegge, A. N., Correa, B. R., Mahesula, S., Kohnke, L. Q., Qiao,
Nestler, E. J., Hyman, S. E. (2010). Animal models of neuropsychiatric M., . . . Penalva, L. O. (2016). MiR-124, −128, and −137 orchestrate
disorders. Nature Neuroscience, 13(10), 1161–1169. neural differentiation by acting on overlapping gene sets containing a
Nicolae, D. L., Gamazon, E., Zhang, W., Duan, S., Dolan, M. E., & Cox, N. J. highly connected transcription factor network. Stem Cells, 34(1),
(2010). Trait-associated SNPs are more likely to be eQTLs: Annotation 220–232.
to enhance discovery from GWAS. PLoS Genetics, 6(4), e1000888. Saurat, N., Andersson, T., Vasistha, N. A., Molnar, Z., & Livesey, F. J. (2013).
Olde Loohuis, N. F., Ba, W., Stoerchel, P. H., Kos, A., Jager, A., Schratt, G., . . . Dicer is required for neural stem cell multipotency and lineage
Aschrafi, A. (2015). MicroRNA-137 controls AMPA-Receptor-Medi- progression during cerebral cortex development. Neural Development,
ated transmission and mGluR-Dependent LTD. Cell Reports, 11(12), 8, 14.
1876–1884. Schizophrenia Psychiatric Genome-Wide Association Study Consortium.
Ooi, L., Wood, I. C. (2007). Chromatin crosstalk in development and disease: (2011). Genome-wide association study identifies five new
Lessons from REST. Nature Reviews Genetics, 8(7), 544–554. schizophrenia loci. Nature Genetics, 43(10), 969–976.
14 | SAKAMOTO AND CROWLEY

Schizophrenia Working Group of the Psychiatric Genomics Consortium. facilitate integrative and comparative epigenomics. PLoS ONE, 8(12),
(2014). Biological insights from 108 schizophrenia-associated genetic 81148.
loci. Nature, 511(7510), 421–427. Stachowiak, M. K., Kucinski, A., Curl, R., Syposs, C., Yang, Y., Narla, S., . . .
Schmitt, A., Steyskal, C., Bernstein, H. G., Schneider-Axmann, T., Parlapani, Stachowiak, E. K. (2013). Schizophrenia: A neurodevelopmental
E., Schaeffer, E. L., . . . Falkai, P. (2009). Stereologic investigation of the disorder-integrative genomic hypothesis and therapeutic implications
posterior part of the hippocampus in schizophrenia. Acta Neuro- from a transgenic mouse model. Schizophrenia Research, 143(2–3),
pathologica, 117(4), 395–407. 367–376.
Schouten, M., Fratantoni, S. A., Hubens, C. J., Piersma, S. R., Pham, T. V., Stark, K. L., Xu, B., Bagchi, A., Lai, W. S., Liu, H., Hsu, R., . . . Gogos, J. A.
Bielefeld, P., . . . Fitzsimons, C. P. (2015). MicroRNA-124 and −137 (2008). Altered brain microRNA biogenesis contributes to phenotypic
cooperativity controls caspase-3 activity through BCL2L13 in hippo- deficits in a 22q11-deletion mouse model. Nature Genetics, 40(6),
campal neural stem cells. Scientific Reports, 5, 12448. 751–760.
Schuster, D. J., Dykstra, J. A., Riedl, M. S., Kitto, K. F., Belur, L. R., McIvor, Stogsdill, J. A., Eroglu, C. (2016). The interplay between neurons and glia in
R. S., . . . Vulchanova, L. (2014). Biodistribution of adeno-associated synapse development and plasticity. Current Opinion in Neurobiology,
virus serotype 9 (AAV9) vector after intrathecal and intravenous 42, 1–8.
delivery in mouse. Frontiers in Neuroanatomy, 8, 42. Strazisar, M., Cammaerts, S., van der Ven, K., Forero, D. A., Lenaerts, A. S.,
Sellier, C., Hwang, V. J., Dandekar, R., Durbin-Johnson, B., Charlet- Nordin, A., . . . Del-Favero, J. (2015). MIR137 variants identified in
Berguerand, N., Ander, B. P., . . . Tassone, F. (2014). Decreased DGCR8 psychiatric patients affect synaptogenesis and neuronal transmission
expression and miRNA dysregulation in individuals with 22q11.2 gene sets. Molecular Psychiatry, 20(4), 472–481.
deletion syndrome. PLoS ONE, 9(8), e103884. Sun, G., Ye, P., Murai, K., Lang, M. F., Li, S., Zhang, H., . . . Shi, Y. (2011). MiR-
Shen, F., Kuo, R., Milon-Camus, M., Han, Z., Jiang, L., Young, W. L., & Su, H. 137 forms a regulatory loop with nuclear receptor TLX and LSD1 in
(2013). Intravenous delivery of adeno-associated viral vector serotype neural stem cells. Nature Communications, 2, 529.
9 mediates effective gene expression in ischemic stroke lesion and brain Szulwach, K. E., Li, X., Smrt, R. D., Li, Y., Luo, Y., Lin, L., . . . Jin, P. (2010). Cross
angiogenic foci. Stroke, 44(1), 252–254. talk between microRNA and epigenetic regulation in adult neuro-
Shibata, M., Nakao, H., Kiyonari, H., Abe, T., & Aizawa, S. (2011). MicroRNA- genesis. The Journal of Cell Biology, 189(1), 127–141.
9 regulates neurogenesis in mouse telencephalon by targeting multiple Tamim, S., Vo, D. T., Uren, P. J., Qiao, M., Bindewald, E., Kasprzak, W. K., . . .
transcription factors. The Journal of Neuroscience: The Official Journal of Penalva, L. O. (2014). Genomic analyses reveal broad impact of miR-137
the Society for Neuroscience, 31(9), 3407–3422. on genes associated with malignant transformation and neuronal
Shimizu, T., Suzuki, H., Nojima, M., Kitamura, H., Yamamoto, E., Maruyama, differentiation in glioblastoma cells. PLoS ONE, 9(1), 85591.
R., . . . Toyota, M. (2013). Methylation of a panel of microRNA genes is a Tamminga, C. A., Zukin, R. S. (2015). Schizophrenia: Evidence implicating
novel biomarker for detection of bladder cancer. European Urology, hippocampal GluN2B protein and REST epigenetics in psychosis
63(6), 1091–1100. pathophysiology. Neuroscience, 309, 233–242.
Shioya, M., Obayashi, S., Tabunoki, H., Arima, K., Saito, Y., Ishida, T., & Satoh, Tarantino, C., Paolella, G., Cozzuto, L., Minopoli, G., Pastore, L., Parisi, S., &
J. (2010). Aberrant microRNA expression in the brains of neurodegen- Russo, T. (2010). MiRNA 34a, 100, and 137 modulate differentiation of
erative diseases: MiR-29a decreased in Alzheimer disease brains targets mouse embryonic stem cells. FASEB Journal, 24(9), 3255–3263.
neurone navigator 3. Neuropathology and Applied Neurobiology, 36(4), Tezcan, G., Tunca, B., Bekar, A., Preusser, M., Berghoff, A. S., Egeli, U., . . .
320–330. Tolunay, S. (2014). MicroRNA expression pattern modulates temozo-
Siegert, S., Seo, J., Kwon, E. J., Rudenko, A., Cho, S., Wang, W., . . . Tsai, L. H. lomide response in GBM tumors with cancer stem cells. Cellular and
(2015). The schizophrenia risk gene product miR-137 alters presynaptic Molecular Neurobiology, 34(5), 679–692.
plasticity. Nature Neuroscience, 18(7), 1008–1016. Tucci, A., Ciaccio, C., Scuvera, G., Esposito, S., & Milani, D. (2016). MIR137 is
Silber, J., Lim, D. A., Petritsch, C., Persson, A. I., Maunakea, A. K., Yu, M., . . . the key gene mediator of the syndromic obesity phenotype of patients
Hodgson, J. G. (2008). MiR-124 and miR-137 inhibit proliferation of with 1p21.3 microdeletions. Molecular cytogenetics, 9, 80.
glioblastoma multiforme cells and induce differentiation of brain tumor Uranova, N. A., Vikhreva, O. V., Rachmanova, V. I., & Orlovskaya, D. D.
stem cells. BMC Medicine, 6, 14. (2011). Ultrastructural alterations of myelinated fibers and oligoden-
Singh, S. K., Kagalwala, M. N., Parker-Thornburg, J., Adams, H., & Majumder, drocytes in the prefrontal cortex in schizophrenia: A postmortem
S. (2008). REST maintains self-renewal and pluripotency of embryonic morphometric study. Schizophrenia Research and Treatment, 2011,
stem cells. Nature, 453(7192), 223–227. 325789.
Smemo, S., Tena, J. J., Kim, K. H., Gamazon, E. R., Sakabe, N. J., Gomez- van Erp, T. G., Guella, I., Vawter, M. P., Turner, J., Brown, G. G., McCarthy,
Marin, C., . . . Nobrega, M. A. (2014). Obesity-associated variants within G., . . . Potkin, S. G. (2014). Schizophrenia miR-137 locus risk genotype is
FTO form long-range functional connections with IRX3. Nature, associated with dorsolateral prefrontal cortex hyperactivation. Biologi-
507(7492), 371–375. cal Psychiatry, 75(5), 398–405.
Smrt, R. D., Szulwach, K. E., Pfeiffer, R. L., Li, X., Guo, W., Pathania, M., . . . van Erp, T. G., Hibar, D. P., Rasmussen, J. M., Glahn, D. C., Pearlson, G. D.,
Zhao, X. (2010). MicroRNA miR-137 regulates neuronal maturation by Andreassen, O. A., . . . Turner, J. A. (2016). Subcortical brain volume
targeting ubiquitin ligase mind bomb-1. Stem Cells, 28(6), 1060–1070. abnormalities in 2028 individuals with schizophrenia and 2540 healthy
Snyder, E. M., Philpot, B. D., Huber, K. M., Dong, X., Fallon, J. R., & Bear, controls via the ENIGMA consortium. Molecular Psychiatry, 21(4),
M. F. (2001). Internalization of ionotropic glutamate receptors in 547–553.
response to mGluR activation. Nature Neuroscience, 4(11), 1079–1085. van Rooij, E., Purcell, A. L., & Levin, A. A. (2012). Developing microRNA
Sofroniew, M. V., Vinters, H. V. (2010). Astrocytes: Biology and pathology. therapeutics. Circulation Research, 110(3), 496–507.
Acta Neuropathologica, 119(1), 7–35. van Scheltinga, A. F., Bakker, S. C., van Haren, N. E., Derks, E. M., Buizer-
Soldati, C., Bithell, A., Johnston, C., Wong, K. Y., Stanton, L. W., & Buckley, Voskamp, J. E., Cahn, W., . . . Kahn, R. S. (2013). Schizophrenia genetic
N. J. (2013). Dysregulation of REST-regulated coding and non-coding variants are not associated with intelligence. Psychological Medicine,
RNAs in a cellular model of Huntington’s disease. Journal of 43(12), 2563–2570.
Neurochemistry, 124(3), 418–430. Verma, P., Augustine, G. J., Ammar, M. R., Tashiro, A., & Cohen, S. M. (2015).
Song, Q., Decato, B., Hong, E. E., Zhou, M., Fang, F., Qu, J., . . . Smith, A. D. A neuroprotective role for microRNA miR-1000 mediated by limiting
(2013). A reference methylome database and analysis pipeline to glutamate excitotoxicity. Nature Neuroscience, 18(3), 379–385.
SAKAMOTO AND CROWLEY | 15

Visani, M., de Biase, D., Marucci, G., Cerasoli, S., Nigrisoli, E., Bacchi microRNA-137-regulated genes influence gray matter structure in
Reggiani, M. L., . . . Group Ps. (2014). Expression of 19 microRNAs in schizophrenia. Translational Psychiatry 6:e724.
glioblastoma and comparison with other brain neoplasia of grades I–III. Wright, C., Turner, J. A., Calhoun, V. D., & Perrone-Bizzozero, N. (2013).
Molecular Oncology, 8(2), 417–430. Potential impact of miR-137 and its targets in schizophrenia. Frontiers in
Voineskos, A. N. (2015). Genetic underpinnings of white matter Genetics, 4, 58.
“connectivity”: Heritability, risk, and heterogeneity in schizophrenia. Wu, L., Belasco, J. G. (2008). Let me count the ways: Mechanisms of
Schizophrenia Research, 161(1), 50–60. gene regulation by miRNAs and siRNAs. Molecular Cell, 29(1), 1–7.
Vora, S., Tuttle, M., Cheng, J., & Church, G. (2016). Next stop for the CRISPR Wu, S., Zhang, R., Nie, F., Wang, X., Jiang, C., Liu, M., . . . Ma, J. (2016).
revolution: RNA-guided epigenetic regulators. The FEBS Journal, MicroRNA-137 inhibits EFNB2 expression affected by a genetic variant
283(17), 3181–3193. and is expressed aberrantly in peripheral blood of schizophrenia
Wang, J., Wang, Y., Yang, J., & Huang, Y. (2014). MicroRNAs as novel patients. EBioMedicine, 12, 133–142.
biomarkers of schizophrenia (Review). Experimental and Therapeutic Xu, B., Hsu, P. K., Karayiorgou, M., & Gogos, J. A. (2012). MicroRNA
Medicine, 8(6), 1671–1676. dysregulation in neuropsychiatric disorders and cognitive dysfunction.
Wang, W. X., Rajeev, B. W., Stromberg, A. J., Ren, N., Tang, G., Huang, Q., . . . Neurobiology of Disease, 46(2), 291–301.
Nelson, P. T. (2008). The expression of microRNA miR-107 decreases Xu, X. L., Li, Y., Wang, F., & Gao, F. B. (2008). The steady-state level of the
early in Alzheimer’s disease and may accelerate disease progression nervous-system-specific microRNA-124a is regulated by dFMR1 in
through regulation of beta-site amyloid precursor protein-cleaving Drosophila. The Journal of Neuroscience: The Official Journal of the
enzyme 1. The Journal of Neuroscience: The Official Journal of the Society Society for Neuroscience, 28(46), 11883–11889.
for Neuroscience, 28(5), 1213–1223. Yang, J. H., Li, J. H., Jiang, S., Zhou, H., & Qu, L. H. (2013). ChIPBase: A
Warburton, A., Breen, G., Bubb, V. J., & Quinn, J. P. (2016). A GWAS SNP for database for decoding the transcriptional regulation of long non-coding
schizophrenia is linked to the internal MIR137 promoter and supports RNA and microRNA genes from ChIP-Seq data. Nucleic Acids Research
differential allele-specific expression. Schizophrenia Bulletin, 42(4), 41(Database issue), D177–D187.
1003–1008. Zeisel, A., Munoz-Manchado, A. B., Codeluppi, S., Lonnerberg, P., La Manno,
Warburton, A., Breen, G., Rujescu, D., Bubb, V. J., & Quinn, J. P. (2015). G., Jureus, A., . . . Linnarsson, S. (2015). Brain structure. Cell types in the
Characterization of a REST-regulated internal promoter in the mouse cortex and hippocampus revealed by single-cell RNA-seq.
schizophrenia genome-wide associated gene MIR137. Schizophrenia Science, 347(6226), 1138–1142.
Bulletin, 41(3), 698–707. Zeng, Y., Yi, R., & Cullen, B. R. (2003). MicroRNAs and small interfering
Ward, L. D., Kellis, M. (2012). HaploReg: A resource for exploring chromatin RNAs can inhibit mRNA expression by similar mechanisms. Proceedings
states, conservation, and regulatory motif alterations within sets of of the National Academy of Sciences of the United States of America,
genetically linked variants. Nucleic Acids Research 40(Database issue), 100(17), 9779–9784.
D930–D934. Zhang, C., Ge, X., Liu, Q., Jiang, M., Li, M. W., & Li, H. (2015). MicroRNA-
Wayman, G. A., Davare, M., Ando, H., Fortin, D., Varlamova, O., Cheng, mediated non-cell-autonomous regulation of cortical radial glial
H. Y., . . . Impey, S. (2008). An activity-regulated microRNA controls transformation revealed by a Dicer1 knockout mouse model. Glia,
dendritic plasticity by down-regulating p250GAP. Proceedings of the 63(5), 860–876.
National Academy of Sciences of the United States of America, 105(26), Zhang, Y., Wang, Z., & Gemeinhart, R. A. (2013). Progress in microRNA
9093–9098. delivery. Journal of Controlled Release, 172(3), 962–974.
Whalley, H. C., Papmeyer, M., Romaniuk, L., Sprooten, E., Johnstone, E. C., Zhao, D., Lin, M., Chen, J., Pedrosa, E., Hrabovsky, A., Fourcade, H. M., . . .
Hall, J., . . . McIntosh, A. M. (2012). Impact of a microRNA MIR137 Lachman, H. M. (2015). MicroRNA profiling of neurons generated using
susceptibility variant on brain function in people at high genetic risk of induced pluripotent stem cells derived from patients with schizophrenia
schizophrenia or bipolar disorder. Neuropsychopharmacology: Official and schizoaffective disorder, and 22q11.2. del. PLoS ONE, 10(7),
Publication of the American College of Neuropsychopharmacology, 37(12), 0132387.
2720–2729. Zhao, Y., Li, Y., Lou, G., Zhao, L., Xu, Z., Zhang, Y., & He, F. (2012). MiR-137
Wightman, B., Ha, I., & Ruvkun, G. (1993). Posttranscriptional regulation of targets estrogen-related receptor alpha and impairs the proliferative
the heterochronic gene lin-14 by lin-4 mediates temporal pattern and migratory capacity of breast cancer cells. PLoS ONE, 7(6), 39102.
formation in C. elegans. Cell, 75(5), 855–862. Zheng, K., Li, H., Huang, H., & Qiu, M. (2012). MicroRNAs and glial cell
Willemsen, M. H., Valles, A., Kirkels, L. A., Mastebroek, M., Olde Loohuis, N., development. Neuroscientist, 18(2), 114–118.
Kos, A., . . . Kleefstra, T. (2011). Chromosome 1p21.3 microdeletions Zhou, R., Yuan, P., Wang, Y., Hunsberger, J. G., Elkahloun, A., Wei, Y., . . .
comprising DPYD and MIR137 are associated with intellectual Manji, H. K. (2009). Evidence for selective microRNAs and their
disability. Journal of Medical Genetics, 48(12), 810–818. effectors as common long-term targets for the actions of mood
Winton-Brown, T. T., Fusar-Poli, P., Ungless, M. A., & Howes, O. D. (2014). stabilizers. Neuropsychopharmacology, 34(6), 1395–1405.
Dopaminergic basis of salience dysregulation in psychosis. Trends in Zhu, X., Li, Y., Shen, H., Li, H., Long, L., Hui, L., & Xu, W. (2013). MiR-137
Neurosciences, 37(2), 85–94. inhibits the proliferation of lung cancer cells by targeting Cdc42 and
Witwer, K. W. (2015). Circulating microRNA biomarker studies: Pitfalls and Cdk6. FEBS Letters, 587(1), 73–81.
potential solutions. Clinical Chemistry, 61(1), 56–63.
Wong, N., Wang, X. (2015). MiRDB: An online resource for microRNA
target prediction and functional annotations. Nucleic Acids Research
How to cite this article: Sakamoto K, Crowley JJ.
43(Database issue), D146–D152.
Wright, C., Calhoun, V. D., Ehrlich, S., Wang, L., Turner, J. A., & Bizzozero, A comprehensive review of the genetic and biological
N. I. (2015). Meta gene set enrichment analyses link miR-137- evidence supports a role for MicroRNA-137 in the etiology
regulated pathways with schizophrenia risk. Frontiers in Genetics, 6, of schizophrenia. Am J Med Genet Part B. 2017;9999:1–15.
147.
https://doi.org/10.1002/ajmg.b.32554
Wright, C., Gupta, C. N., Chen, J., Patel, V., Calhoun, V. D., Ehrlich, S., . . .
Turner, J. A. (2016). Polymorphisms in MIR137HG and

Вам также может понравиться