Download as pdf or txt
Download as pdf or txt
You are on page 1of 15

RevIeWS

Neurexins: molecular codes for


shaping neuronal synapses
Andrea M. Gomez 1
, Lisa Traunmüller2 and Peter Scheiffele 2 ✉

Abstract | The function of neuronal circuits relies on the properties of individual neuronal cells
and their synapses. We propose that a substantial degree of synapse formation and function is
instructed by molecular codes resulting from transcriptional programmes. Recent studies on
the Neurexin protein family and its ligands provide fundamental insight into how synapses are
assembled and remodelled, how synaptic properties are specified and how single gene mutations
associated with neurodevelopmental and psychiatric disorders might modify the operation of
neuronal circuits and behaviour. In this Review, we first summarize insights into Neurexin function
obtained from various model organisms. We then discuss the mechanisms and logic of the cell
type-​specific regulation of Neurexin isoforms, in particular at the level of alternative mRNA
splicing. Finally, we propose a conceptual framework for how combinations of synaptic protein
isoforms act as ‘senders’ and ‘readers’ to instruct synapse formation and the acquisition of cell
type-​specific and synapse-​specific functional properties.

Alternative splicing
Nervous systems represent remarkable examples of a likely apply to many other adhesion systems operating
A process in which exons highly organized tissue with an abundance of special- at neuronal synapses.
of an mRNA are assembled in ized cells in an intricate structure. During development,
multiple different (alternative) neuronal connectivity arises from a series of steps, Adhesive modules for synapse assembly. Synaptic diffe­
ways to yield multiple different
including cell specification, migration, targeted growth, rentiation relies on a large number of synaptic adhesion
versions of a final mRNA
molecule that may contain synapse formation and remodelling. Spontaneous and signalling molecules with so-​called synaptogenic
different RNA regulatory activity and sensory experience propagated through the properties, that is, the ability of an isolated factor to
motifs or encode alternative developing networks play a significant role in organiz- trigger a substantial degree of the synaptic differentia-
protein forms.
ing aspects of neuronal wiring. However, many funda- tion process. When presented in non-​neuronal cells or
Isoforms
mental steps of neuronal morphogenesis and synapse on synthetic surfaces, synaptogenic proteins nucleate
Variants of an mRNA transcript formation proceed normally even in the absence of the formation of functional presynaptic or postsynap-
or protein generated from neurotransmission1–3. Thus, genetically encoded pro- tic assemblies9–12. For example, postsynaptic adhesion
a single gene but differing grammes are thought to orchestrate key aspects of the molecules of the Neuroligin family trigger the assembly
in sequence (for example,
timing and dynamics of neuronal growth and nervous of functional presynaptic terminals in axons through
resulting from alternative
promoters or from alternative system wiring4–7. Cell surface adhesion and signalling interaction with their receptor Neurexin9,10 (Fig. 1a).
splicing). molecules significantly contribute to all of these devel- Conversely, Neurexin-​mediated clustering of Neuroligins
opmental steps. Thus, each neuronal cell type carries an triggers the recruitment of NMDA-​t ype glutamate
array of cues linked to cellular origin and cell fate that receptors and scaffolding molecules12,13. This early cell
are integral to its developmental specification. Although biological analysis uncovered fundamental activities of
signalling processes, neuronal activity and disease states Neurexin proteins and their ligands. Subsequent genetic
may shift these codes, there are constraints that restrict studies then probed the functional consequences of
1
Department of Molecular this plasticity, thereby maintaining cell type-​specific inactivating Neurexin genes in various model organ-
and Cell Biology, Helen properties. One critical and extensively studied process isms (see below). What makes the roles for Neurexins in
Wills Neuroscience Institute, in nervous system development is the selective growth this process so fascinating is twofold. First, the Neurexin
University of California,
and targeting of neurites, which encompasses axon guid- gene family encodes a vast array of distinct transcript
Berkeley, Berkeley, CA, USA.
ance and synaptic specificity7,8. The present Review aims isoforms generated from multiple genes (Nrxn1, Nrxn2,
2
Biozentrum, University of
Basel, Basel, Switzerland.
to discuss a second key aspect of neuronal wiring: the Nrxn3), alternative promoters (α, β, γ) and extensive
✉e-​mail: peter.scheiffele@ molecular principles of neuronal synapse formation and alternative splicing, with individual isoforms linked to
unibas.ch the specification of synapse function. We will use the specific neuronal cell types. Second, Neurexins serve
https://doi.org/10.1038/ Neurexin family, one class of cell adhesion molecules, to as presynaptic receptors for several structurally unre-
s41583-020-00415-7 illustrate the fundamental principles of this process that lated extracellular binding partners, indicating that they

Nature Reviews | Neuroscience


Reviews

a Heterologous cell b Presynaptic


or synthetic surface neuron
Presynaptic
neuron
Synaptic
vesicle Neurotransmitter
Hevin
SPARC

Neuroligin 1 NMDAR

Neurexin Dystroglycan
Scaffolds CBLN1

Neuroligin GLUD2
LRRTMs
Postsynaptic Calsyntenin
neuron
Heterologous cell Postsynaptic neuron
or synthetic surface

c d Motor neuron axonal arborizations


Motor neuron

Muscle

Synaptic
release site

Control nrx-1 Control nrx-1


Drosophila larvae Adult Drosophila

Fig. 1 | synaptogenic function of Neurexins. a | Bidirectional context, several ligands can be co-​expressed at single synapses or can be
synapse-​organizing activity of Neurexins. Presentation of Neurexin proteins differentially expressed across neuronal cell populations. c | Example of a
or their ligands on synthetic surfaces in vitro and overexpression of loss-​of-​function phenotype resulting from loss of Neurexin (nrx-1 mutant)
Neurexin ligands in vitro and in vivo drive assembly of presynaptic and at the neuromuscular junction of Drosophila larvae (adapted from Li et al.27);
postsynaptic structures, respectively. Certain specific Neurexins only synaptic release sites are marked in orange. d | Contribution of the
interact with certain receptors, for example, Neurexin 1 interacts with the Neurexin–Neuroligin adhesion system to growth of axonal arborizations of
NMDA receptor (right). b | Neurexin isoforms interact with a large array of the motor neurons that innervate the abdominal pleural muscles of adult
structurally unrelated extracellular binding partners. Only a selection Drosophila (adapted from Constance et al.29). CBLN1, Cerebellin 1; LRRTM,
of ligands are displayed in this simplified schematic. Depending on cellular Leucine-​rich repeat transmembrane protein.

represent a hub for presynaptic organization (Fig. 1b). Neurexins, a large body of genetic studies also support
For example, Neurexins are presynaptic receptors for an evolutionary conserved role for Neurexin proteins in
the secreted protein Cerebellin 1 (CBLN1), the trans- structural synapse assembly. In vivo models present with
membrane proteins Neuroligin 1–4, α-​Dystroglycan, a wide array of phenotypes, depending on the cellular
Leucine-​rich repeat transmembrane proteins (LRRTM1, context and the Neurexin isoforms and/or genes ablated.
LRRTM2, LRRTM3, LRRTM4) and Calsyntenin 3 Early studies in Neurexin 1,2,3α triple-​knockout mice
(refs14–17). Recent reviews provide a comprehensive sum- discovered that inhibitory synapse density in the brain-
mary of this array of Neurexin ligands6,18. In the present stem is reduced by 50%, whereas the density of excita-
article, we focus on the contribution of alternative splic- tory synapses is unchanged at birth19. In the nematode
ing of Neurexins in controlling such interactions and on Caenorhabditis elegans, loss of one particular Neurexin
the interplay of multiple synaptic recognition systems at isoform, γ-​Neurexin, diminishes synapse numbers in the
neuronal synapses. In the following, we will first discuss DA9 motor neuron supporting critical functions for this
genetic studies in various model organisms where many form in synapse formation21. A broader nrx-1 mutation
or most Neurexin isoforms are ablated. results in a loss of postsynaptic neurotransmitter recep-
tors from synapses and loss of spine-​like protrusions
Neurexin and neuronal connectivity from the postsynaptic neuron22,23. In another class of
Genetic loss-​of-​function studies highlight critical roles C. elegans neurons that display experience-​dependent
for Neurexin proteins at synapses in vivo. Initial work and sexually dimorphic plasticity, synapse rearrange-
emphasized functional alterations in synaptic transmis- ments are impaired in nrx-1 mutants24. In the fruit fly
sion, in particular calcium-​dependent neurotransmitter Drosophila melanogaster, mutations in Neurexin and its
release19,20. Consistent with the synaptogenic activity of ligand, Neuroligin, result in severe loss of neuromuscular

www.nature.com/nrn
Reviews

synaptic release sites in larvae25–27 (Fig. 1c). Importantly, and high-​affinity interactions. These structural motifs —
impairing Neurexin–Neuroligin adhesion also modifies acting individually or cooperatively — recruit macro-
the growth of axonal and dendritic arbors in some model molecular complexes that span the synaptic cleft and
organisms. Time-​lapse imaging experiments in develop- coordinate bidirectional signalling and organization.
ing tadpoles suggest that adhesion through Neurexin and Three very large genes encode mammalian Neurexin 1,
its ligand, Neuroligin, confer transient morphological Neurexin 2 and Neurexin 3 (1.0 Mb, 0.1 Mb and 1.6 Mb
stabilization of dendritic contacts28. Similarly, the growth in mouse; 1.1 Mb, 0.1 Mb and 1.8 Mb in human).
and arborization of Drosophila motor neuron axons dur- Invertebrates, such as C. elegans and D. melanogaster,
ing metamorphosis is disrupted in Neurexin-​deficient possess a single Neurexin orthologue34,35. Each mamma-
fruit flies29 (Fig. 1d). Although such macroscopic altera- lian Neurexin gene contains two promoters that produce
tions in neuronal arborizations have not been reported a long, α-​Neurexin pre-​mRNA and a shorter, β-​Neurexin
in mice, the roles for Neurexins in synapse assembly are pre-​mRNA, which encode proteins of approximately
conserved from invertebrates to mammals. 1,500 and 450 amino acids (Fig. 2a). For mouse Nrxn1,
Two aspects have significantly delayed the emergence an additional, very short γ-​isoform is generated from a
of the present picture for Neurexin functions: first, in third, internal promoter36, and an orthologous γ-​isoform
the mammalian system, the phenotypic space explored is reported in C. elegans21. We refer to these transcripts
in in vivo studies was quite limited. Although work in (irrespective of their further modification by alternative
invertebrate model systems has long explored synapse splicing) as ‘primary Neurexin transcripts’. Differential
formation between genetically defined cell types, this usage of α, β and γ-​promoters in the three mamma-
approach has only been implemented in mammalian lian Neurexin paralogs drives highly divergent levels of
systems during this past decade. Second, unlike the the primary Neurexin transcripts across neuronal cell
vertebrate neuromuscular junction, where synaptic types. For example, mouse hippocampal CA3 pyrami-
differentiation relies heavily on one primary signalling dal neurons express high levels of Nrxn1β, whereas the
system30, central synapses engage a complex combi- same transcript is very low in CA1 pyramidal cells37–39.
nation of signals. This cooperation between multiple In the mouse neocortex, GABAergic SST interneurons
trans-​synaptic signals greatly complicates generalizing express threefold higher levels of Nrxn3α transcripts as
conclusions from individual genetic experiments. The compared with layer 4 pyramidal cells39.
same Neurexin mutation can result in very different Besides the use of these alternative promoters and
phenotypes when analysed in different cell types. For corresponding transcription start sites, extensive diversi-
example, mutation of C. elegans nrx-1 severely dis- fication of Neurexin transcripts is further driven by alter-
rupts AChR clusters in DD GABA neurons but not native splicing. Thus far, up to six alternatively spliced
in muscle23. In mice, the conditional ablation of all segments (AS1–AS6) — some containing multiple
Neurexin isoforms in somatostatin-​p ositive versus alternative splice acceptor and donor splice sites — exist
parvalbumin-​positive interneurons exhibits very differ- in primary Neurexin transcripts (Fig. 2b). The combi-
ent phenotypes. Mutant parvalbumin-​positive interneu- natorial usage of alternative promoters and alternative
rons severely reduce synapse formation on principal splice sites has the potential to yield >12,000 Neurexin
neurons in the medial prefrontal cortex. By contrast, transcript isoforms in mice. Long-​read, single-​molecule
the number of synapses formed by Neurexin-​deficient PacBio sequencing studies experimentally confirmed
somatostatin-​positive interneurons in the same region is the presence of hundreds of Neurexin transcript iso-
unchanged31. However, somatostatin-​positive interneu- forms in the mouse brain40,41. Interestingly, the relative
rons show altered voltage-​gated calcium channel func- usage and combination of alternative splice insertions
tion and defects in neurotransmitter release31. The are conserved between rodents and humans, evidenced
reasons for such disparate observations are likely mani- by the analysis of post-​mortem human brain samples
fold. First, many studies examine mutations in cells with- and human induced pluripotent stem cell-​derived neu-
out knowing the expression of the disrupted Nrxn gene, ronal preparations42. These transcriptomic studies pro-
its transcript isoforms and paralogs. Second, there is an vide a basis for interpreting the function of Neurexin
array of additional presynaptic receptors unrelated to diversity. However, one caveat is that transcript levels
Post-​translational Neurexins that contribute to synapse assembly32,33. Last, are not sufficiently informative regarding Neurexin
modifications different neuronal cell types express different Neurexin protein isoform expression as multiple Neurexin gene
Enzymatic chemical isoforms, generated from alternative promoters (such as products undergo further control at the level of mRNA
modifications of specific
amino acids in a protein that
the α, β and γ forms) and modified through extensive translation40,43. Advances in targeted proteomics should
occur in the cell after or during alternative splicing. Notably, such Neurexin isoforms clarify the accurate relative and absolute quantification
mRNA translation (for example, differentially interact with selective synaptic ligands. of protein abundance, even for peptides derived from
through phosphorylation, Thus, deletion of individual Neurexin genes precipitates specific splice insertions44.
glycosylation, acetylation
impairment of different receptor–ligand modules in dif-
and so on).
ferent cell types. This complexity most likely underlies Structure of macromolecular assemblies. The defin-
Structural motifs the diversity of phenotypes reported in previous studies. ing feature of all Neurexins (except for the non-​
Structurally conserved building canonical NRXN1γ) is the presence of extracellular
blocks or ‘super-​secondary Molecular diversity of Neurexins. Combinations of Laminin–Neurexin–Sex hormone-​binding globu-
structures’ that appear in
various protein molecules
genomic and proteomic features, including post-​ lin (LNS) domains. α-​Neurexins present six LNS
that may or may not be translational modifications , impart Neurexins with domains interspersed by single epidermal growth fac-
functionally related. numerous adhesive motifs that underlie low-​affinity tor (EGF) domains in the extracellular region (Fig. 2b).

Nature Reviews | Neuroscience


Reviews

a AS1 AS2 AS3 AS6 AS4 AS5


Nrxn1 8 3 2 2 2 2 AS Alternatively spliced segment
Nrxn2 8 3 4 - 2 2
Alternative Constituitive
Nrxn3 8 3 4 2 2 30 first exon exon
Alternative Alternative donor
exon or acceptor site

α-Nrxn β-Nrxn γ-Nrxn


(Nrxn1 only)

b AS2 AS3 AS4 c


Ca2+
AS1 AS6 AS4 AS3
AS5 AS3
Ca2+
AS4
α-NRXN N 1 2 3 4 5 6 C AS2

90°
A B C TMD

LNS
AS4
EGF-like
AS5 β-Sheet β-Sheet
Heparin sulfate chain β-NRXN N 6 C β-Sandwich

Ligand-binding interface
Ca2+
NLGN and LRRTM
binding surface

AS5
NXPH1-binding
surface NRXN3
γ-NRXN N C
R498W

d NLGN

LRRTM

α-NRXN
Presynaptic Postsynaptic
neuron neuron

β-NRXN

5 nm
NXPH

Flexibility Flexibility Flexibility Flexibility

C N C N

AS5 AS5 AS6 AS1

www.nature.com/nrn
Reviews

◀ Fig. 2 | Molecular and structural features of Neurexin isoform diversity. a | Schematic Exon inclusion or exclusion of Neurexin pre-​mRNA
of the alternatively spliced segments of mouse Neurexin genes. Mouse Neurexin transcript encoding the hypervariable domain alters the length
isoforms are generated from three genes (Nrxn1, 1.1 Mb; Nrxn2, 0.1 Mb; Nrxn3, 1.8 Mb; of the folds at the rim, modifies the binding surface
note that given the big differences in gene sizes, the exons and introns are not drawn to topography and regulates assorted low-​affinity and
scale), each containing up to three alternative promoters (α, β, γ) and exhibiting extensive
high-​affinity interactions of Neurexins. Interestingly,
alternative splicing at six alternatively spliced segments (AS1–AS6). Individual segments
can contain single alternative cassette exons (for example, AS4, AS6) or consist of complex
an analogous surface of LNS-​containing proteins agrin
combinations of alternative splice donor and acceptor sites (for example, AS1, AS2, AS3, and laminin also confers ligand-​binding specificity at the
AS5). Numbers (2, 3, 4, 8, 30) depict counts of potential splice variations generated at mammalian neuromuscular junction51.
each segment. Alternative exons are illustrated in colour, constitutive exons in grey and Extensive studies of the LNS domain of β-​NRXN1
alternative donor or acceptor sites are white. b | Alternative promoters and alternative (also NRXN1α LNS6) reveal critical structural ele-
mRNA splicing result in Neurexin protein isoforms that share transmembrane domain ments for binding to Neuroligins52–56. The β-​loops of
(TMD) and cytoplasmic sequences but differ in their extracellular protein sequences. the Neurexin hypervariable domain clasp a single cal-
The extracellular sequences are composed of three major elements: Laminin–Neurexin– cium ion, creating an electropositive surface for bind-
Sex hormone-​binding globulin (LNS) domains, epidermal growth factor (EGF)-​like domains ing to a complementary electronegative surface on
and attachment sites for heparan sulfates. The largest Neurexin proteins are the α-​NRXN
Neuroligin. Neurexin ligand selectivity also relies
forms composed of six LNS domains (LNS1–LNS6), three interposed EGF domains (EGFA–
EGFC) and the heparan sulfate attachment sites. Interaction surfaces for ligands are on the accessibility of ligand-​binding surfaces, ligand
marked with purple lines. β-​NRXN contains a single LNS domain and heparan sulfate concentration and identity of the splice isoform. This
attachment sites, whereas γ-​NRXN is the smallest form lacking LNS and EGF domains. dynamic balance is best documented in the interaction
c | Mapping of alternatively spliced segments (AS2, AS3, AS4), ligand-​binding domains and of β-​NRXN and Neuroligin57,58. β-​NRXN1, β-​NRXN2
sequence variants on a prototypical LNS domain: (left) ribbon diagram and positions of and β-​NRXN3 bind to all Neuroligins with nanomolar
alternatively spliced segments; (right) view of a 90° rotation and surface representation affinity in a splice form-​dependent manner58. Two gen-
of an LNS domain with mapped alternatively spliced segments; (bottom) surface eral conclusions emerged from these in vitro binding
rep­resentation, as in right panel, highlighting the position of ligand-​binding domains and assays: the presence of AS4 insertions in β-​NRXN1 and
the naturally occurring R498W variant in NRXN3α that has been linked to behavioural β-​NRXN2 diminishes affinity to Neuroligin; and, by
alterations in mice129. d | Illustration of approximate sizes and hypothetical conformation
contrast, the presence of AS4 insertions in β-​NRXN3
of adhesion molecule complexes in the synaptic cleft: β-​Neurexin (orange, left panel)
with Leucine-​rich repeat transmembrane protein 2 (LRRTM2; green); and α-​Neurexin
increases affinity to Neuroligin. LRRTM1/2 also exhibits
(orange, right panel) with Neurexophilin (NXPH1; pink) and Neuroligin (NLGN; blue). AS4– isoform-​dependent binding at the same Neurexin
Structural models of the extracellular domains were drawn with ChimeraX 1.0 from the LNS6 site, whereas Neurexins containing the alternative
following Protein Data Bank IDs: 3POY (ref.48), 3B3Q (ref.53), 6PNP (ref.50), 5Z8Y (ref.139). insertions at AS4 bind to CBLN15,59. These examples
The position of stalk, transmembrane and cytoplasmic sequences indicated as dashed highlight the combinatorial and competitive activities
lines. Diagrams at the bottom display positions within these structures where alternative of Neurexin–ligand interactions.
splicing at AS1, AS6 and AS5 in β-​NRXN (left) and α-​NRXN (right) modifies the flexibility In addition to these protein–protein interaction
of the extracellular domains in the synaptic cleft. Part d (left) is adapted from ref.139, sites, some interactions of Neurexins with ligands
CC BY 4.0 (https://creativecommons.org/licenses/by/4.0/). Part d (right) adapted with involve interactions with carbohydrate moieties on
permission from ref.48, Elsevier. Part 2d (right) is adapted from ref.50, CC BY 4.0 (https://
the Neurexins. The juxtamembrane region of α, β
creativecommons.org/licenses/by/4.0/). Parts c, d (right) adapted from ref.53, Springer
Nature Limited.
and γ-​Neurexins contains heparan sulfate carbohy-
drate structures that provide an additional interaction
site for postsynaptic ligands such as LRRTMs and
These alternating repeats tether to the cell surface via Neuroligins60,61 (Fig. 2). LRRTM1/2 and Neuroligins
a rigid and extensively O-​linked glycosylated stalk and a require cooperative binding to Neurexin LNS6 and
single transmembrane domain45,46. The short intracellu- the carbohydrate chains for macromolecular assembly,
lar tail contains interaction sites for cytoskeletal adap- whereas LRRTM3/4 only requires the carbo­hydrate
tors (Protein 4.1) and a carboxy-​terminal PDZ-​binding structures and, thus, can act through γ-​Neurexin
motif47. The smaller β-​NRXN proteins have a short, isoforms that lack LNS domains. Interestingly, the
unique amino-​terminal sequence but are otherwise identity of heparan sulfate proteoglycan structures is
identical to α-​NRXN beginning at the sixth LNS domain. controlled by a series of cellular enzymes that produce
γ-​Nrxn transcripts encode a truncated isoform that lacks cell type-​specific carbohydrate modifications. Individual
the extracellular LNS and EGF structured domains, yet glycosyltransferases, sulfotransferases and epimerases
retains a transmembrane and intracellular tail36. have emerged as critical regulators for neuronal devel-
Remarkably, despite having low sequence identity opment and wiring62. Thus, the molecular diversity of
(20%) between each other, crystal structures from the Neurexins generated at the level of alternative splicing
α-​NRXN1 LNS2–LNS6 domains reveal high structural may be complemented by a ‘glycan code’ generated by
homology48. The architectural prototype of an LNS differential heparan sulfate modifications.
domain consists of a β-​sandwich — two juxtaposed The larger α-​Neurexin isoforms, which are more
slightly curved β-​sheets, forming a ‘lens-​like’ structure abundant at the protein level44, interact with addi-
(Fig. 2c). Importantly, at the rim of this β-​sandwich are tional extracellular ligands. Thus, Calsyntenin 3 and
calcium and ligand-​binding sites. A ligand-​binding sur- α-​Dystroglycan, two postsynaptic proteins at GABAergic
face emerges from the folds that connect the two β-​sheets synapses, interact only with α-​Neurexin but not
and is subject to alteration by AS2, AS3 and AS4, and β-​Neurexin isoforms16,63. To accommodate the large,
accordingly is referred to as the hypervariable domain49. α-​Neurexin in the narrow synaptic cleft, flexibility
A one-​quarter turn along the rim of the β-​sandwich of in the linker regions connecting the LNS domains
LNS2 reveals an additional ligand-​binding surface50. bends the large, extracellular domain to fit into the

Nature Reviews | Neuroscience


Reviews

25-​nm synaptic cleft46,48,64. On the other hand, the short performance of mice in a learning task 37 — a first
length of β-​Neurexin is not constrained by its confor- demonstration that this cell type-​specific isoform reg-
mation in the narrow synaptic cleft. Indeed, β-​Neurexin ulation is indeed essential for circuit function. A recent
and Neuroligin expressed in heterologous cells form a single-​cell study on somatostatin-​positive interneurons
lattice-​like sheet spanning the length of cell-​to-​cell con- residing in the stratum oriens of the hippocampus fur-
tacts. By contrast, α-​Neurexin–Neuroligin interactions ther revealed that neurons with similar electrophysio-
fail to recruit widespread lateral assemblies in this assay. logical properties exhibit similar expression of Nrxn1
Thus, there might be an isoform-​specific constraint on and Nrxn3 splice isoforms68. In aggregate, these studies
the macromolecular assembly of adhesion complexes establish that Nrxn isoform repertoires link to neuronal
within the limits of the synaptic cleft65. Moreover, cell type identity. This raises the questions of how these
whereas alternative splicing of Neurexin AS2, AS3 or AS4 repertoires are generated, whether they are dynamically
tunes the affinity of Neurexin to ligands, the splice inser- regulated and which specific aspect of neuronal function
tions at AS1, AS5 and AS6 — regions that encode for the such isoforms instruct.
linker regions — adjust the inter-​domain length40,48
(Fig. 2d) . Shortening or lengthening of these linker Regulation of Neurexin splice isoforms. Alternative
regions constrains the configuration of α-​Neurexin and splicing is a highly dynamic process that is guided by
modulates the exposure of the ligand-​binding domains. cis-​acting RNA sequence elements such as donor and
Ultimately, adjustments in the ligand-​binding interface acceptor sites, the branch-​p oint sequence and the
and changes in inter-​domain flexibility may govern the polypyrimidine tract for spliceosome assembly69,70
high-​affinity and low-​affinity interactions of Neurexins (Fig. 3a) . Trans-​a cting factors such as RNA-​binding
with their ligands. proteins bind intronic or exonic sequence elements in
the pre-​mRNAs and bias the choice of splice donor and
Cell type-​specific Neurexin isoforms. The molecu- acceptor sites, resulting in inclusion or skipping of alter-
lar diversification of the Neurexin transcripts and the native sequence elements70–72 (Fig. 3a). Several classes of
selective biochemical interactions of the resulting pro- RNA-​binding proteins are implicated in the generation
teins raise the question of whether these proteins con- of cell type-​specific Neurexin repertoires. In cultured
tribute to some form of molecular code that specifies rat neurons, the polypyrimidine tract binding protein
aspects of neuronal wiring. The key elements to consider PTBP2 modifies exon skipping at Nrxn2 AS4 (ref.73).
the coding power of such a system are: the number of Genome-​wide screens for transcript isoform altera-
distinct recognition tags or ‘senders’ (that is, Neurexin tions in mouse mutants for the RNA-​binding proteins
protein isoforms generated); the number of biochemi- NOVA2, PTBP2 and RBFOX1 uncover modifica-
cal interaction partners that detect/distinguish or ‘read’ tions in Nrxn transcripts at several segments74–76, but
these protein isoforms; and the spatial logic of how the functional consequences of these alterations are
such ‘senders’ and ‘readers’ array over neuronal cell unknown.
types. Quantitative single-​molecule sequencing of full-​ Probably the best-​c haracterized regulators of
length Nrxn1 transcripts uncovered a large number of Neurexin alternative splicing are members of the
highly represented transcript isoforms in heterogeneous STAR (signal transduction activators of RNA) protein
brain tissue but a much more narrow isoform comple- family. These proteins are defined by an evolutionar-
ment in a purified neuronal cell population40. Similarly, ily conserved RNA-​binding domain of approximately
alternative exon and splice site choices at individual alter- 200 amino acids. In mice, there are five STAR family
natively spliced segments display cell type-​specific regu- proteins: the splicing factors SF1, Quaking and SAM68,
lation of individual splicing decisions. For example, the and SLM1 and SLM2 — the latter three are closely
relative abundance of exon usage at several alternatively related paralogs (encoded by the genes Khdrbs1, Khdrbs2
spliced segments differs between parvalbumin-​positive and Khdrbs3)77,78. SAM68, SLM1 and SLM2 directly bind
and CCK-​p ositive interneuron populations 39,66,67. RNA recognition motifs in introns flanking the highly
Moreover, differential usage of combinatorial alternative conserved alternative exon at AS4 of the Neurexin
splicing profiles of Nrxn1 and Nrxn3 or alternative splic- pre-​mRNAs79–81. This binding promotes skipping of the
ing at AS3 correlates with the developmental origin of alternative exon at AS4. Indeed, a close correlation exists
interneurons67. Quantitative assessments of the absolute between AS4+ (exon containing) and AS4– Neurexin
usage of alternative exons uncovered that hippocampal isoforms and the absence and presence, respectively, of
CA3/CA1 pyramidal neurons and parvalbumin-​positive SLM1 or SLM2 in neuronal cell types37,80,81. Alternative
interneurons contain different pools of Nrxn1 AS6 and splicing regulation at AS4 is particularly interesting as
Nrxn2 AS2. Hippocampal excitatory neurons contain the 30 amino acids encoded by the alternative exon
substantially higher amounts of AS4– (exon lacking) strongly impact affinities for Neurexin ligands17,59,82,83.
than AS4+ (exon containing) transcript isoforms for all For example, AS4-​containing Neurexins (NRXNAS4+)
three Nrxn genes, whereas the higher AS4 inclusion rates in cerebellar granule cell axons form tripartite com-
can be observed in parvalbumin-​positive interneurons37. plexes with the extracellular scaffolding protein CBLN1
A similar trend is observed for alternative splicing at and the postsynaptic receptor GLUD2 expressed in
AS4 of Nrxn3 in somatostatin-​positive interneurons37,68. Purkinje cells. Mutation of any of the three components
Conditional ablation of the Nrxn1 and Nrxn3 AS4 alter- of this tripartite complex impairs presynaptic differ-
native exons in parvalbumin-​positive neurons results entiation and results in a severe reduction in synapse
in elevated hippocampal network activity and impaired assembly and density59,84,85. SLM1 and SLM2 proteins

www.nature.com/nrn
Reviews

exhibit highly selective, mutually exclusive expression CA2, CA3) and subsets of somatostatin-​positive and
in neuronal cell types80,86,87. In the mouse hippocam- VIP-​positive interneurons. In SLM2KO hippocampus,
pus, SLM2 protein expression is primarily restricted there is a highly selective loss of Nrxn1, Nrxn 2 and Nrxn 3
to principal cells of the pyramidal cell layer (CA1, AS4− splice isoforms87–89. Conversely, ectopic expression

a Isoform 1
Isoform 2

X1 X2 X3 X4

ISE ISS A Py AG ESE ESS GU ISE ISS A Py AG

Exon Exon Exon

Isoform 3

b Cell type 1 Cell type 2


Transcription factors Transcription factors

Nrxn genes Splicing regulators Nrxn genes Splicing regulators


α α
Nrxn1 β Nrxn1 β
X1 X1
Nrxn2 Nrxn2
X2 X2
Nrxn3 Nrxn3
X3 X4

... Xn ... Xn
X3
X5 X6 X7 X1 X2 X5 X6 X7 X1 X2
X4

α-Nrxn
genes
AS1+ AS2+ AS3– AS6+ AS4– AS5– AS1+ AS2+ AS3– AS6+ AS4+ AS5+
X3
X1 X2 X1 X2
X4

β-Nrxn genes β-Nrxn genes


AS4 –
AS5 –
AS4+ AS5+

Cell type-specific neuronal and synaptic properties Cell type-specific neuronal and synaptic properties

Fig. 3 | control of synapse specification by alternative splicing cassette exon (isoform 1 and isoform 2). Usage of the alternative donor site in
programmes. a | Schematic of the control of alternative splicing by RNA the first exon results in a third transcript isoform (isoform 3). b | Intersection of
motifs and trans-​acting factors (coloured spheres X1, X2, X3 and so on). The Nrxn transcriptional programmes and the combinatorial action of splicing
displayed alternatively spliced segment contains an alternative splice donor regulators. The illustration depicts two hypothetical cell types (cell type 1 and
site in the upstream exon (depicted as grey and blue boxes on left), followed cell type 2) that produce different Neurexin transcript repertoires. Cell
by a cassette exon (purple box) and a downstream constitutive exon (grey box, type-​specific transcription from promoters/enhancers (arrowheads) drives
right). Exons can contain exonic splicing enhancers (ESE) and exonic splicing the differential transcription of the primary transcripts from the Nrxn genes
silencers (ESS). The intronic sequences between the exon boxes contain RNA (for example, cell type 1 transcribes high levels of NRXN1α whereas cell type 2
motifs that act as intronic splicing enhancers (ISE) and intronic splicing transcribes high levels of NRXN3α). In addition, each expresses a specific
silencers (ISS). GU marks the 5′ splice site, A–Py–AG marks the branch-​point battery of splicing regulators (Cell type 1: X1, X2, X3 and so on; Cell type 2:
sequence and polypyrimidine tract followed by the terminal AG sequence in X1, X2, X4 and so on). The intersection of these splicing regulators (coloured
the intron. Motifs recruit trans-​acting RNA-​binding proteins (depicted as spheres) with the primary Nrxn transcripts then produces the cell type-​specific
coloured spheres) that either promote or suppress usage of individual splice Neurexin isoforms (for example, β-​Nrxn AS4–AS5– in cell type 1 and β-​Nrxn
donor acceptor sites, resulting in inclusion or skipping of the alternative AS4+AS5+ in cell type 2)76,79,89. AS1–AS6, alternatively spliced segments.

Nature Reviews | Neuroscience


Reviews

Box 1 | Non-​neuronal and non-​synaptic roles for Neurexin complexes Interestingly, selective manipulation of AS4 alter-
native splice insertions in Nrxn1 and Nrxn3 differen-
whereas the vast majority of research focuses on the roles for Neurexins at synapses, tially modifies postsynaptic NMDAR-​mediated and
recent evidence highlights potential functions of these adhesion molecules at other AMPAR-​mediated transmission94. Constitutive mis-
cellular junctions. a starting point for such investigations were transcriptomic and expression of Nrxn1AS4+ enhances NMDAR-​mediated
in situ hybridization studies that detected significant Neurexin mRNA expression in
responses at hippocampal CA1–subiculum syn-
non-​neuronal cells, including astrocytes, oligodendrocytes and oligodendrocyte
precursor cells36,144,145. Moreover, also major Neurexin ligands, such as the Neuroligins, apses, whereas Nrxn3AS4+ misexpression suppresses
were detected in non-​neuronal cells. For example, gliomas have been reported to AMPAR-​mediated currents94. Whether these differential
produce significant levels of Neuroligin 3 protein146–148. interestingly, the proliferation of phenotypes are a consequence of different Nrxn1 and
glioma cells is elevated by electrical signalling that may involve synapse-​like structures Nrxn3 expression levels in CA pyramidal cells or unique
formed by axons on the Neuroligin-​expressing glioma cells as a critical pathological properties of the proteins derived from either gene is
feature. an example for further extra-​synaptic roles for Neurexins are interactions unknown. However, these findings raise the possibility
between neuronal Neurexins and the Neurexin ligand, Neuroligin 2, in astrocytes149. that AS4 isoforms derived from two Nrxn paralogs may
These interactions were proposed to contribute to the elaboration of astrocytic exhibit non-​overlapping functions94.
morphology in the developing neocortex of mice. Moreover, the extracellular matrix In aggregate, these studies demonstrate that cell
proteins Hevin, SPARC and Thrombospondin, secreted from astrocytes, have been
type-​s pecific RNA-​binding proteins drive highly
suggested to directly bind to Neurexins and their postsynaptic ligands150,151, thereby
reconfiguring their availability for adhesive interactions in the synaptic cleft. Collectively, selective regulation of Neurexin alternative splicing.
these recent findings indicate a physiologically relevant ‘repurposing’ of Neurexins and This regulation establishes cell type-​specific mole­
their ligands to organize cellular interactions beyond synaptic neuron–neuron contacts. cular Neurexin isoform repertoires that engage in
trans-​synaptic interactions with dedicated receptors
in the postsynaptic cell, thereby shaping fundamen-
of SLM1 or SLM2 in cells that do not endogenously tal synaptic properties. Although this concept is best
express either of these proteins results in the genera- analysed for the alternative splicing factor SLM2 and
tion of AS4– isoforms37,80. Thus, single, cell type-​specific the AS4 alternative exons, it may extend to the other
RNA-​binding proteins selectively instruct alternative Neurexin alternatively spliced segments. At Nrxn3 AS5,
splicing at one of the six alternatively spliced segments there are multiple alternative splice acceptor sites in the
of Neurexin genes. downstream exon of the segment (sometimes referred to
Detailed functional analysis of Slm2 KO mice, as as exons 25a, 25b and 25c). The amino acids encoded by
well as conditional mutations of AS4, uncovered that Nrxn3 exon 25b confer binding of the NRXN3 isoform
Neurexin splice variants in hippocampal CA3 pyrami- to the extracellular linker proteins C1QL2 and C1QL3,
dal cells control postsynaptic properties of Schaffer col- which in turn mediate the formation of a tripartite,
lateral synapses. In Slm2KO hippocampi, macroscopic trans-​synaptic complex with postsynaptic kainate recep-
neuronal morphology and density of Schaffer collat- tors (GluK2 and GluK4) at hippocampal mossy fibre
eral synapses is normal. However, SLM2-​deficiency synapses95. Trans-​acting factors regulating this splicing
selectively elevates AMPA-​receptor (GluA1) surface event remain to be identified.
expression, leading to increased evoked glutamatergic Although splicing choices link to cell identity, they
transmission and impaired Schaffer collateral long-​term also underlie dynamic regulation in response to neu-
potentiation (LTP). Selective genetic restoration of the ronal signalling. Previous studies demonstrate shifts
NRXNAS4– splice isoform (which is lost in Slm2KO mice) in alternative splicing regulation by strong pharmaco-
restores normal levels of GluA1, partially restores LTP logical or electrical stimulation96,97. In Neurexins, such
and rescues behavioural alterations in Slm2KO mice89. A paradigms shift alternative exon incorporation at sev-
germline mutation in Nrxn3 that constitutively includes eral alternatively spliced segments81,98. In the mouse
the AS4 exon reduces AMPA receptor (AMPAR) sur- cerebellum, this phenomenon requires calcium influx,
face expression and impairs LTP in a subset of subicular calmodulin-​d ependent kinase IV and the broadly
neurons90. Interestingly, the postsynaptic modifications expressed STAR protein, SAM68 (ref.81). In granule cells
that result from altered presynaptic Nrxn3 isoforms sug- of the mouse dentate gyrus, recall of a contextual fear
gest the disruption of a trans-​synaptic link. The exact memory triggers the inclusion of the alternative exon
mechanisms resulting in altered postsynaptic prop- at Nrxn1AS4. Interestingly, this shift in alternative splic-
erties remain to be worked out. However, the pheno- ing requires HDAC2 and is controlled by a selective
types in Slm2KO and Nrxn3AS4 mice are consistent with histone modification (H3K9me3) in the Nrxn1 gene.
shifted ligand affinities of AS4– versus AS4+ splice var- This modification is thought to control memory sta-
iants for the interaction with postsynaptic Neuroligins bility, by temporarily shifting Nrxn1 alternative splic-
and LRRTMs. Indeed, NRXNAS4+ isoforms display ing at AS4 and, ultimately, contributing to rewiring of
reduced affinity for postsynaptic Neuroligins13,57,58, and dentate granule cell synapses to support learning99. Yet
Neurexin–Neuroligin1/3 interactions are disrupted in another level of regulation is the proteolytic cleavage
Slm2KO hippocampus89. In Nrxn3AS4+ mice, expression of of Neurexins, resulting in shedding of the extracellular
the postsynaptic NrxnAS4– ligand LRRTM2 is reduced90. domain of the protein100,101. The physiological contexts
Given that mutations in these postsynaptic Neurexin lig- and functional relevance of these modes of regulation
ands likewise disrupt LTP and synaptic transmission91–93, remain to be explored. However, these mechanistically
their altered interactions with presynaptic Neurexin diverse modes of transcriptional, post-​transcriptional
isoforms may be responsible for aspects of synaptic and proteolytic regulation highlight the perplexing
dysfunction. complexity of Neurexin cell biology and pose challenges

www.nature.com/nrn
Reviews

for linking cell type-​specific repertoires to synaptic highly targeted regulator of a small number of synaptic
function. proteins88,89. Both SAM68 and SLM2 bind the same con-
sensus motif; however, SLM2 but not SAM68 regulates
Cell type logic of splicing regulators. Recent genome-​ alternative splicing of Nrxn2 at AS4 in vivo80. This selec-
wide studies on mRNA transcript isoforms and targets tive activity is dependent on the abundance of binding
of RNA-​binding proteins provide insight into the com- sites flanking the alternative exon79. Furthermore, the
plex logic of neuronal cell type-​specific alternative specificity of RNA recognition by SLM2 and SAM68
splicing39,75,76. Several alternative transcript programmes may be modified by their ability to homodimerize and
are linked to neuronal cell type, and some alternative heterodimerize78,80. Thus, these different modes of action
splicing regulators increase in expression upon the provide additional flexibility for the STAR family of pro-
commitment to a postmitotic fate102. Examining RNA-​ teins to generate neuronal cell class-​specific synaptic
binding protein expression across neuronal cell types properties88,89 (X1 + X3 in Fig. 3b). By contrast, broadly
highlights a remarkable range of cell type selectivity: expressed RNA-​binding proteins tend to regulate alter-
some splicing regulators are expressed in essentially native splicing events in mRNAs from hundreds of
all neurons, and many of them are ‘neuron-​specific’ genes, possibly generating cell type-​specific outcomes by
— that is, largely not expressed in non-​neuronal cells. coordinate and/or competitive action of multiple trans-​
These broadly expressed splicing regulators include the acting factors on a single RNA segment. For example,
widely studied NOVA proteins, PTBPs or RBFOX pro- NOVA2-​dependent intron retention in hundreds of
teins. Other splicing regulators such as SLM1 and SLM2 transcripts sequesters the trans-​acting splicing factor
exhibit a much more selective expression in a subset of PTBP2 (ref.75). Remarkably, NOVA2 regulates diverse
neuronal cell types (Fig. 3b). Intriguingly, the selectively target transcripts in different cell populations, demon-
expressed RNA-​binding protein SLM2 controls alter- strating that selectivity can emerge from a complex, cell
native splicing choices of only a few genes, acting as a type-​specific interplay of splicing regulators. Moreover,

Schaffer collateral synapse Parallel fibre synapse Basket cell synapse


Presynaptic Synaptic
neuron vesicle Lysosome
GABA
Glutamate

Ca2+

α-NRXNAS4–
VGCC
β-NRXNAS4–
β-NRXNAS4+
Neuroligin 1 CBLN1 Neuroligin 2
LRRTM1/2 α-NRXNAS2–

NMDAR Scaffolds AMPAR


GABAAR
α/β-Dystroglycan
Postsynaptic
neuron

• VGCC • NRXNAS4– • β-NRXNAS4+ • β-NRXNAS4+ • α-NRXNAS2–


• NRXNAS4– • LRRTM1/2 • CBLN1 • NLGN2 • α/β-Dystroglycan
• NLGN1 • AMPAR • GLUD2 • GABAAR
• NMDAR

Trans-synaptic
signalling module

Fig. 4 | examples of synaptic interaction modules nucleated by Neurexin vesicles140,141. Note that CBLN1 interacts with α and β-​Neurexins — but for
proteins. Simplified model illustrating trans-​synaptic interaction modules simplicity only the interaction with β-​Neurexin is depicted here. GABAergic
assembled around presynaptic Neurexin protein isoforms. Amongst other basket cell synapses in the mouse hippocampus contain modules of
components, CA3–CA1 Schaffer collateral synapses in the mouse NRXN AS4+ and NRXN AS2– isoforms linking to postsynaptic NLGN2 or
hippocampus contain trans-​synaptic NRXNAS4–– Neuroligin 1 (NLGN1) and α/β-​Dystroglycan proteins, respectively. Blue dots within synaptic vesicles
NRXN–Leucine-​rich repeat transmembrane protein (LRRTM) complexes indicate the neurotransmitter glutamate, red dots represent GABA. One
that recruit postsynaptic NMDA and AMPA-​type glutamate receptors. major contribution of Neurexins at synapses is the incorporation of
Note that LRRTM proteins bind α and β-​Neurexins, but for simplicity only functional voltage-​gated calcium channels (VGCC) at synapses, facilitating
the interaction with α-​Neurexin is depicted here. Cerebellar parallel fibre the calcium-​dependent release of synaptic vesicles19,142,143. The lower row
synapses largely rely on a single trans-​synaptic module consisting of displays illustrations of individual trans-​synaptic modules present at the
NRXNAS4+ isoforms, extracellular Cerebellin 1 (CBLN1) proteins and the respective synapses. AS1–AS6, alternatively spliced segments. AMPAR,
postsynaptic receptor GLUD2. CBLN1 is secreted from lysosome-​like carrier AMPA receptor.

Nature Reviews | Neuroscience


Reviews

a b
Cell type 1 Cell type 2 Pyramidal cell
Senders and Synaptic
readers properties
Reader 1

Trans-synaptic Y1 Y1
Message A signalling
modules Yn

Y2
Sender 1
Y2 Y2

Cell type 3 Cell type 4 Y2

Y3
Reader 2
Y3 Yn
Message B

Yn

c Synapse 1 d Synapse 2
Single dominant channel Three major channels
Wild type Wild type

Transmitter GPCR function Transmitter


Adhesion receptor receptor
stabilization Adhesion stabilization
Vesicle
recruitment Vesicle
Adhesion recruitment Adhesion
Ca channel
2+

function Ca2+ channel


Scaffolds function Scaffolds

Presynaptic Postsynaptic
neuron neuron

Loss of function Loss of function

Transmitter GPCR function Transmitter


Adhesion receptor receptor
stabilization Adhesion stabilization
Vesicle
recruitment Vesicle
Adhesion recruitment Adhesion
Ca2+ channel
function Ca channel
2+
Scaffolds function Scaffolds

www.nature.com/nrn
Reviews

◀ Fig. 5 | context-dependent functions of synaptic interaction modules. a | Model for splice insertions that modify molecular interfaces in
the action of synaptic interaction modules. Cell types express specific complements of trans-​synaptic receptor–ligand interactions33,104,105. Thus,
‘senders’ (light cyan) and ‘readers’ (cyan and orange), such as Neurexin splice isoforms the principles discussed here for the generation and syn-
and corresponding binding partners. The impact on synapse assembly and functional aptic function of Neurexins likely extend broadly to the
specification (‘message’) depends on the cell type-​specific abundance of corresponding
control of neuronal recognition (Fig. 3b).
senders and readers (cyan and orange). Cell type 1 and cell type 3 both express the
same sender but, because their downstream synaptic partners cell type 2 and cell type 4
express different readers (reader 1 and reader 2, respectively), the messages will be Circuits and disorders. From human genetic studies,
different (message A and message B, respectively). Thus, the same presynaptic sender the Neurexins, and predominantly NRXN1, emerge as
can convey divergent messages at different synapses. b | Across the surface of an significant risk genes for a wide range of neurodevelop-
individual neuron (here an illustration of a pyramidal cell), the combinations of axonal mental, psychiatric, neurological and neuropsychologi-
senders and dendritic readers create trans-​synaptic modules (depicted as blue, green, cal phenotypes106–108. Likewise, Neurexin gene mutations
light green boxes and so on) that represent a molecular code. This code sets synapse- are associated with schizophrenia109, autism110, Tourette
specific properties (Y1, Y2, Y3 … Yn; blue labels represent glutamatergic synapses, orange syndrome111, nicotine dependence112, developmen-
labels GABAergic synapses) and, thereby, input integration in the postsynaptic cell. tal delay, dysmorphic features and infantile epileptic
c | Synapses across the CNS employ various numbers of trans-​synaptic modules that
encephalopathy107,113. Many of these deletions span pro-
can be viewed as trans-​synaptic communication channels (displayed in different colours).
Some synapses rely on a single dominant channel (here depicted in red), which has a major
moter and initial exons of the NRXN1 gene (2p16.3).
contribution to synapse assembly, stability and functional properties (‘Synapse 1’ — an Transcriptomic studies in induced pluripotent stem
example for this would be parallel fibre synapses in the cerebellum that rely on the cell-​derived neurons carrying such mutations uncover
NRXN–Cerebellin 1 (CBLN1)–GLUD2 module59,85,141). Loss of a single presynaptic sender significantly reduced NRXN1α transcripts. Moreover,
(illustrated in the lower panel; for example, CBLN1) results in disruption of the trans- de novo expression of isoforms divergent from the
synaptic module and substantial disruption of synapse formation and function, despite repertoire in neurotypical controls might occur 42.
the presence of an additional, minor channel (depicted in grey). d | Other synapses Heterozygous exonic deletions do not appear to be fully
(‘Synapse 2’) contain multiple prominent trans-​synaptic modules (here depicted in red, penetrant as rearrangements in the NRXN1 gene are
green, and purple) — likely to afford a larger dynamic range of plasticity. These modules frequent in the control population114, and NRXN1 dele-
drive overlapping elements of synaptic differentiation. For example, the green module
tions are frequently inherited from a healthy parent115.
drives bidirectional adhesion, presynaptic vesicle recruitment, presynaptic G protein-​
coupled receptor (GPCR) function and postsynaptic stabilization of neurotransmitter
However, such mono-​allelic NRXN1 deletion carriers
receptors, whereas the purple module controls adhesion, active zone assembly and may share common alterations in anxiety, intelligence
calcium channel function. Loss of a single presynaptic sender (the purple module) results and impulsivity, which go undiagnosed without an
in loss of presynaptic calcium channel function, but active zone assembly and adhesion in-​depth evaluation116. Notably, a small number of bial-
are maintained by the overlapping red and green modules at this synapse. See references lelic NRXN1 mutations result in a severe mental retar-
for examples on Ca2+ channel function31,142,143. dation syndrome, which phenotypically overlaps with
Pitt–Hopkins syndrome, an autism-​like developmental
selective alternative splicing decisions also arise from disorder with variable characteristics107,117,118.
histone modifications and alterations in transcriptional In some cases, specific NRXN sequence variants
kinetics103. may elevate risk to certain disorders. However, consid-
Acquiring a specific complement of RNA-​binding ering the wide range of neurodevelopmental conditions
proteins during synaptic terminal differentiation may act observed in individuals with NRXN mutations, it is
much like the terminal selector genes for transcriptional more likely that alterations in NRXN gene expression
cell type specification. Terminal selector genes have been alter neurodevelopmental trajectories that — depending
postulated to be transcription factors that regulate the on the genetic background and environmental con-
expression of genes required to give neurons their unique ditions — precipitate diverse phenotypes. It is widely
identity4. Congruent with this hypothesis, the combina- appreciated that many symptoms are comorbid with
torial expression of RNA-​binding proteins in individual multiple neurodevelopmental disorders, such as
neuronal cell classes could, ultimately, instruct the gen- attention-​deficit/hyperactivity, tic disorder, develop-
eration of a cell type-​specific complement of Neurexin mental coordination disorder and autism. For clinical
alternative splice isoforms and their unique functions evaluations, it is encouraged — particularly for young
(Fig. 3b). Although this Review focuses on the Neurexin children — to focus on impairments in specific domains,
gene family, a similar molecular logic likely applies to such as communication and language, motor coordina-
other neuronal recognition and synaptic proteins104. tion, attention, mood and sleep, rather than to separate
Detailed profiling of transcript isoforms across genet- patients into discrete disorders. This is conceptualized
ically defined neuronal cell populations uncovered as ‘ESSENCE’ (early symptomatic syndromes eliciting
hundreds of differentially regulated alternative splic- neurodevelopmental clinical examinations)119.
ing events in the mouse neocortex and hippocampus. The significant disease association with the human
Furthermore, gene expression analysis of 52 bona fide NRXN genes has spurred efforts to obtain insights into
splicing regulators highlighted broad, overlapping ver- how Neurexin mutations impact neuronal circuits and
sus highly restricted expression within neuronal cell behaviour. It should be noted that some components
classes39. This supports the possibility that combinato- of the Neurexin adhesion systems are also expressed in
rial expression of RNA-​binding proteins provides dif- non-​neuronal cells (Box 1). However, studies modelling
ferent classes of neurons with unique compositions of the impact of disease-​associated NRXN mutations have
synaptic proteins and function (Fig. 3b). Studies on addi- largely focused on synaptic phenotypes. Invertebrate
tional neuronal recognition systems, such as the recep- systems have provided important opportunities for
tor protein tyrosine phosphatases, uncovered alternative probing common cellular nodes modified by various

Nature Reviews | Neuroscience


Reviews

risk gene mutations118,120. In mice, global Nrxn1α knock- biochemical level. A single presynaptic Neurexin iso-
out results in multiple behavioural alterations. These form can recruit fundamentally different postsynaptic
include impaired nest building, decreases in prepulse ligands. At a single synapse, multiple ligands compete for
inhibition of startle responses and an improvement in interaction with a limited pool of Neurexin molecules.
motor learning121. Interestingly, some phenotypes are Moreover, numerous Neurexin paralogs cooperate with
sex-​specific: male homozygous Nrxn1α knockout mice various additional, independent trans-​synaptic systems
exhibit increased aggressive behaviours. Male hetero­ localized at the same synaptic contact. This complexity
zygous Nrxn1α knockout mice show increased novelty demands significant caution when interpreting loss-​of-​
responses as assessed by locomotor activity in a new function studies. As discussed above, the same Neurexin
environment and enhanced habituation upon repeated loss-​of-​function manipulation applied in different cel-
exposure to this environment122. In rats, non-​social lular contexts results in widely differing phenotypes,
deficits, such as hyperactivity and deficits in instru- ranging from a substantial loss of synaptic structures and
mental and spatial learning tasks, result from Nrxn1α entire axonal branches to the comparably subtle impair-
deficiency123. Nrxn2α homozygous and heterozygous ment of one or multiple synaptic ion channels. Such
knockout mice exhibit diminished social approach and context-​dependent synaptic phenotypes are not unique
social novelty responses in behavioural assays but also to the Neurexin gene family and have been reported
impaired recognition of novel objects124–126, indicating for other trans-​synaptic adhesion systems such as the
social and broader cognitive deficits. type III mGluR–Elfn131–133 or receptor protein tyrosine
Considering that these behavioural observations phosphatase complexes134,135.
were made in global, constitutive knockout mice, link- We propose that the combinatorial actions of syn-
ing such phenotypes to selective developmental, cir- aptic adhesion and signalling proteins (Neurexins
cuit and synaptic functions of the Neurexin proteins and other protein families) can be rationalized as
is difficult. Reversible overexpression studies using modules for nucleating synaptic structures, scaffold-
dominant-​negative mutant Nrxn1 isoforms support the ing proteins and ion channels. Multiple modules can
notion that behavioural phenotypes may result from be present at single synapses and contain overlap-
dysfunction, rather than irreversible mis-​wiring of cir- ping components136–138 (Fig. 4). These trans-​synaptic
cuits during development127. For Nrxn3, a requirement recognition and synapse-​organizing systems can be
for Neurexin function in somatostatin interneurons in conceptualized as senders and readers arrayed across
the anterior cingulate cortex affects empathy in condi- neuronal populations (Fig. 5a). Upon fate specification,
tional mutant mice. Empathy is a key element of social each neuronal cell type contains a set of cues — or a
interactions, and the loss of empathy is an important molecular code — that is integral to its neuronal iden-
feature of autism spectrum disorders and psychiatric tity. We postulate that this code instructs, but also con-
conditions128. Mice carrying a single-​nucleotide poly­ strains, cellular interactions, and thereby directs aspects
morphism in Nrxn3, which results in a single amino of neuronal wiring and plasticity, thus maintaining cell
acid change (R498W), increase observational fear in type-​specific properties and circuit function (Fig. 5b).
a behavioural task129. In this task, an observer mouse Importantly, the messages conveyed by a particular
adopts a conditioned context-​d ependent freezing sender (for example, a specific Neurexin isoform) are
response after observing a second mouse receiving strongly context-​dependent. Thus, the nucleation of a
repetitive foot shocks. Given that human performance trans-​synaptic module largely depends on the molecu-
in a similar paradigm correlates with metrics of empathy, lar repertoire of readers available (Fig. 5a) and may even
this task is thought to assess an evolutionarily conserved rely on certain extracellular proteins being absent from
aspect of empathy130. Conditional deletion of Nrxn3 in a particular synaptic site. A second critical parameter
somatostatin-​positive interneurons of the anterior cin- is the number of trans-​synaptic communication chan-
gulate cortex impairs synaptic transmission from these nels. Some synapses with little demand for plasticity and
GABAergic neurons and elevates freezing responses, extensive neuromodulation may heavily rely on a few
whereas activation of the same neuronal population trans-​synaptic channels, or even just a single dominant
suppresses them129. The R498W variant maps to the sender–reader pair (Fig. 5c). In such cases, loss of any of
third LNS domain of Nrxn3α (see Fig. 2c). This region the core components results in a substantial dissociation
may confer a Ca2+-​mediated conformational switch for of synaptic contacts, for example loss of the Neurexin–
ligand binding48,64. However, ligands that contribute to CBLN1 link at cerebellar parallel fibre synapses84,85 or the
the differential function of the NRXN3 R498W protein Elfn–mGluR6 link in photoreceptor synapses131. At syn-
remain to be uncovered. Nevertheless, these studies apses with multiple prominent trans-​synaptic channels,
illustrate that Neurexin functions — and likely, specif- the same mutation may modestly destabilize a particu-
ically, the synaptic recognition codes controlled by the lar neurotransmitter receptor recruited by the sender
Neurexins — are not a cell biological detail but are fun- or reader — however, a second trans-​synaptic channel
damental for nervous system operation, behaviour and would take over additional functions and maintain the
neurodevelopmental disorders. overall structural integrity of the synapse (Fig. 5d).
Such complex systems likely evolved for CNS synap-
Framework for synaptic action modules. A particular togenesis as they render a synaptic contact more tunable,
challenge in defining cellular Neurexin functions and providing a high degree of freedom to control plasticity
predicting the impact of mutations on neuronal cir- of individual synaptic sites — but, at the same time, they
cuit function arises from extensive multiplexing at the provide constraints for wiring in highly complex circuits.

www.nature.com/nrn
Reviews

By integrating the observations made in reductionist circuits. However, within one molecularly and anatom-
biochemical and in vitro systems, across vertebrate and ically recognizable cell type, subpopulations of cells are
invertebrate systems, and from loss-​of-​function stud- recruited to represent or encode unique aspects of the
ies in multiple cell types, we can define action mod- external world, such as direction-​s elective cells in
ules for synaptic adhesion molecules. A key question the visual cortex, ‘reward’ cells in the cerebellum or place
for the future will be to explore how molecular codes cells in the hippocampus. Neuronal activity and synaptic
and activity-​dependent mechanisms intersect to shape plasticity mechanisms play a major role in establishing
circuitry during development. There is mounting evi- such neuronal ensembles — and future work may eluci-
dence that synaptic transmission per se is not required date how molecular recognition systems constrain and
for a significant degree of neuronal wiring and cell execute such key steps of circuit assembly.
type-​specific connectivity. Thus, activity-​independent
mechanisms generate an initial blueprint of neuronal Published online xx xx xxxx

1. Lu, W., Bushong, E. A., Shih, T. P., Ellisman, M. H. 22. Maro, G. S. et al. MADD-4/punctin and Neurexin alternative splicing programs. Nat. Neurosci. 22,
& Nicoll, R. A. The cell-​autonomous role of excitatory organize C. elegans GABAergic postsynapses through 1709–1717 (2019).
synaptic transmission in the regulation of neuronal neuroligin. Neuron 86, 1420–1432 (2015). 40. Schreiner, D. et al. Targeted combinatorial alternative
structure and function. Neuron 78, 433–439 (2013). 23. Philbrook, A. et al. Neurexin directs partner-​specific splicing generates brain region-​specific repertoires
2. Sigler, A. et al. Formation and maintenance of synaptic connectivity in C. elegans. eLife 7, e35692 of Neurexins. Neuron 84, 386–398 (2014).
functional spines in the absence of presynaptic (2018). 41. Treutlein, B., Gokce, O., Quake, S. R. & Südhof, T. C.
glutamate release. Neuron 94, 304–311.e4 (2017). 24. Hart, M. P. & Hobert, O. Neurexin controls plasticity Cartography of Neurexin alternative splicing mapped
3. Sando, R. et al. Assembly of excitatory synapses in the of a mature, sexually dimorphic neuron. Nature 553, by single-​molecule long-​read mRNA sequencing.
absence of glutamatergic neurotransmission. Neuron 165–170 (2018). Proc. Natl Acad. Sci. USA 111, E1291–E1299 (2014).
94, 312–321.e3 (2017). This study uncovers a key role for Neurexin in 42. Flaherty, E. et al. Neuronal impact of patient-​specific
4. Hobert, O. Terminal selectors of neuronal identity. structural plasticity in C. elegans. aberrant NRXN1α splicing. Nat. Genet. 51, 1679–1690
Curr. Top. Dev. Biol. 116, 455–475 (2016). 25. Banovic, D. et al. Drosophila Neuroligin 1 promotes (2019).
5. Hassan, B. A. & Hiesinger, P. R. Beyond molecular growth and postsynaptic differentiation at glutamatergic Together with Schreiner et al. (2014) and
codes: simple rules to wire complex brains. Cell 163, neuromuscular junctions. Neuron 66, 724–738 (2010). Treutlein et al. (2014), this study reports numbers
285–291 (2015). 26. Chen, Y. C. et al. Drosophila Neuroligin 2 is required and cell type-​selectivity of Neurexin splice isoforms
6. Sudhof, T. C. Synaptic Neurexin complexes: a molecular presynaptically and postsynaptically for proper synaptic in rodent and human neurons by long-​read mRNA
code for the logic of neural circuits. Cell 171, 745–769 differentiation and synaptic transmission. J. Neurosci. sequencing.
(2017). 32, 16018–16030 (2012). 43. Ding, X. et al. Translational inhibition of α-​Neurexin 2.
7. Sanes, J. R. & Zipursky, S. L. Synaptic specificity, 27. Li, J., Ashley, J., Budnik, V. & Bhat, M. A. Crucial role Sci. Rep. 10, 3403 (2020).
recognition molecules, and assembly of neural circuits. of Drosophila Neurexin in proper active zone apposition 44. Schreiner, D., Simicevic, J., Ahrné, E., Schmidt, A.
Cell 181, 536–556 (2020). to postsynaptic densities, synaptic growth, and synaptic & Scheiffele, P. Quantitative isoform-​profiling of highly
8. Stoeckli, E. T. Understanding axon guidance: are we transmission. Neuron 55, 741–755 (2007). diversified recognition molecules. eLife 4, 1–17
nearly there yet? Development https://doi.org/10.1242/ 28. Chen, S. X., Tari, P. K., She, K. & Haas, K. Neurexin– (2015).
dev.151415 (2018). Neuroligin cell adhesion complexes contribute to 45. Ushkaryov, Y. A., Petrenko, A. G., Geppert, M.
9. Dean, C. et al. Neurexin mediates the assembly of synaptotropic dendritogenesis via growth stabilization & Sudhof, T. C. Neurexins: synaptic cell surface
presynaptic terminals. Nat. Neurosci. 6, 708–716 mechanisms in vivo. Neuron 67, 967–983 (2010). proteins related to the α-​latrotoxin receptor and
(2003). 29. Constance, W. D. et al. Neurexin and Neuroligin-​based laminin. Science 257, 50–56 (1992).
10. Scheiffele, P., Fan, J., Choih, J., Fetter, R. & Serafini, T. adhesion complexes drive axonal arborisation growth 46. Comoletti, D. et al. The macromolecular architecture of
Neuroligin expressed in nonneuronal cells triggers independent of synaptic activity. eLife 7, e31659 extracellular domain of αNRXN1: domain organization,
presynaptic development in contacting axons. Cell (2018). flexibility, and insights into trans-​synaptic disposition.
101, 657–669 (2000). 30. Burden, S. J., Huijbers, M. G. & Remedio, L. Structure 18, 1044–1053 (2010).
11. Dalva, M. B. et al. EphB receptors interact with NMDA Fundamental molecules and mechanisms for forming 47. Biederer, T. & Sudhof, T. C. CASK and protein 4.1
receptors and regulate excitatory synapse formation. and maintaining neuromuscular synapses. Int. J. support F-​actin nucleation on Neurexins. J. Biol.
Cell 103, 945–956 (2000). Mol. Sci. 19, 490 (2018). Chem. 276, 47869–47876 (2001).
12. Graf, E. R., Zhang, X., Jin, S. X., Linhoff, M. W. 31. Chen, L. Y., Jiang, M., Zhang, B., Gokce, O. 48. Miller, M. T. et al. The crystal structure of the
& Craig, A. M. Neurexins induce differentiation of & Sudhof, T. C. Conditional deletion of all Neurexins α-Neurexin-1 extracellular region reveals a hinge
GABA and glutamate postsynaptic specializations via defines diversity of essential synaptic organizer point for mediating synaptic adhesion and function.
neuroligins. Cell 119, 1013–1026 (2004). functions for Neurexins. Neuron 94, 611–625 e614 Structure 19, 767–778 (2011).
13. Chih, B., Gollan, L. & Scheiffele, P. Alternative splicing (2017). 49. Sheckler, L. R., Henry, L., Sugita, S., Sudhof, T. C.
controls selective trans-​synaptic interactions of the This study highlights cell type-​specific alterations & Rudenko, G. Crystal structure of the second
Neuroligin–Neurexin complex. Neuron 51, 171–178 in synapse formation and synaptic function LNS/LG domain from Neurexin 1α: Ca2+ binding and
(2006). resulting from genetic Neurexin deletion. the effects of alternative splicing. J. Biol. Chem. 281,
Together with Graf et al. (2004), this study uncovers 32. Takahashi, H. & Craig, A. M. Protein tyrosine 22896–22905 (2006).
regulation of Neurexin synaptogenic activity by phosphatases PTPδ, PTPσ, and LAR: presynaptic 50. Wilson, S. C. et al. Structures of Neurexophilin–
alternative splicing. hubs for synapse organization. Trends Neurosci. 36, Neurexin complexes reveal a regulatory mechanism
14. de Wit, J. et al. LRRTM2 interacts with Neurexin1 and 522–534 (2013). of alternative splicing. EMBO J. 38, e101603 (2019).
regulates excitatory synapse formation. Neuron 64, 33. Li, Y. et al. Splicing-​dependent trans-​synaptic 51. Rudenko, G., Hohenester, E. & Muller, Y. A. LG/LNS
799–806 (2009). SALM3–LAR–RPTP interactions regulate excitatory domains: multiple functions — one business end?
15. Ko, J., Fuccillo, M. V., Malenka, R. C. & Sudhof, T. C. synapse development and locomotion. Cell Rep. 12, Trends Biochem. Sci. 26, 363–368 (2001).
LRRTM2 functions as a Neurexin ligand in promoting 1618–1630 (2015). 52. Arac, D. et al. Structures of Neuroligin-1 and the
excitatory synapse formation. Neuron 64, 791–798 34. Haklai-​Topper, L. et al. The Neurexin superfamily Neuroligin-1/Neurexin-1β complex reveal specific
(2009). of Caenorhabditis elegans. Gene Expr. Patterns 11, protein–protein and protein–Ca2+ interactions. Neuron
16. Pettem, K. L. et al. The specific α-Neurexin interactor 144–150 (2011). 56, 992–1003 (2007).
calsyntenin-3 promotes excitatory and inhibitory 35. Knight, D., Xie, W. & Boulianne, G. L. Neurexins and 53. Chen, X., Liu, H., Shim, A. H., Focia, P. J. & He, X.
synapse development. Neuron 80, 113–128 (2013). Neuroligins: recent insights from invertebrates. Structural basis for synaptic adhesion mediated by
17. Siddiqui, T. J., Pancaroglu, R., Kang, Y., Rooyakkers, A. Mol. Neurobiol. 44, 426–440 (2011). Neuroligin–Neurexin interactions. Nat. Struct. Mol.
& Craig, A. M. LRRTMs and Neuroligins bind Neurexins 36. Yan, Q. et al. Systematic discovery of regulated and Biol. 15, 50–56 (2008).
with a differential code to cooperate in glutamate conserved alternative exons in the mammalian brain 54. Fabrichny, I. P. et al. Structural analysis of the synaptic
synapse development. J. Neurosci. 30, 7495–7506 reveals NMD modulating chromatin regulators. protein Neuroligin and its β-​Neurexin complex:
(2010). Proc. Natl Acad. Sci. USA 112, 1502849112 (2015). determinants for folding and cell adhesion. Neuron
18. de Wit, J. & Ghosh, A. Specification of synaptic 37. Nguyen, T. M. et al. An alternative splicing switch 56, 979–991 (2007).
connectivity by cell surface interactions. Nat. Rev. shapes Neurexin repertoires in principal neurons 55. Koehnke, J. et al. Crystal structure of the extracellular
Neurosci. 17, 22–35 (2016). versus interneurons in the mouse hippocampus. eLife cholinesterase-​like domain from Neuroligin-2.
19. Missler, M. et al. α-​Neurexins couple Ca2+ channels 5, e22757 (2016). Proc. Natl Acad. Sci. USA 105, 1873–1878 (2008).
to synaptic vesicle exocytosis. Nature 423, 939–948 38. Ullrich, B., Ushkaryov, Y. A. & Sudhof, T. C. 56. Koehnke, J. et al. Crystal structures of β-​Neurexin 1
(2003). Cartography of Neurexins: more than 1000 isoforms and β-​Neurexin 2 ectodomains and dynamics of
20. Sudhof, T. C. Neuroligins and Neurexins link synaptic generated by alternative splicing and expressed in splice insertion sequence 4. Structure 16, 410–421
function to cognitive disease. Nature 455, 903–911 distinct subsets of neurons. Neuron 14, 497–507 (2008).
(2008). (1995). 57. Boucard, A. A., Chubykin, A. A., Comoletti, D., Taylor, P.
21. Kurshan, P. T. et al. γ-​Neurexin and frizzled mediate 39. Furlanis, E., Traunmuller, L., Fucile, G. & Scheiffele, P. & Sudhof, T. C. A splice code for trans-​synaptic cell
parallel synapse assembly pathways antagonized by Landscape of ribosome-​engaged transcript isoforms adhesion mediated by binding of Neuroligin 1 to α- and
receptor endocytosis. Neuron 100, 150–166.e4 (2018). reveals extensive neuronal-​cell-class-​specific β-​Neurexins. Neuron 48, 229–236 (2005).

Nature Reviews | Neuroscience


Reviews

58. Koehnke, J. et al. Splice form dependence of 81. Iijima, T. et al. SAM68 regulates neuronal activity-​ 106. Hu, Z., Xiao, X., Zhang, Z. & Li, M. Genetic insights
β-Neurexin/Neuroligin binding interactions. Neuron dependent alternative splicing of Neurexin-1. Cell and neurobiological implications from NRXN1 in
67, 61–74 (2010). 147, 1601–1614 (2011). neuropsychiatric disorders. Mol. Psychiatry 24,
59. Uemura, T. et al. Trans-​synaptic interaction of GluRδa2 This study discovers the regulation of neuronal 1400–1414 (2019).
and Neurexin through Cbln1 mediates synapse activity-​dependent alternative splicing of Neurexins 107. Castronovo, P. et al. Phenotypic spectrum of NRXN1
formation in the cerebellum. Cell 141, 1068–1079 by the STAR family RNA-​binding protein SAM68. mono- and bi-​allelic deficiency: a systematic review.
(2010). 82. Elegheert, J. et al. Structural basis for integration of Clin. Genet. 97, 125–137 (2020).
This study discovers a tripartite synaptic complex GluD receptors within synaptic organizer complexes. 108. Cosemans, N. et al. The clinical relevance of intragenic
consisting of Neurexins, Cerebellins and the Science 353, 295–299 (2016). NRXN1 deletions. J. Med. Genet. 57, 347–355 (2020).
postsynaptic receptor GLUD2. 83. Matsuda, K. & Yuzaki, M. Cbln family proteins 109. Rujescu, D. et al. Disruption of the Neurexin 1 gene is
60. Zhang, P. et al. Heparan sulfate organizes neuronal promote synapse formation by regulating distinct associated with schizophrenia. Hum. Mol. Genet. 18,
synapses through Neurexin partnerships. Cell 174, Neurexin signaling pathways in various brain regions. 988–996 (2009).
1450–1464.e23 (2018). Eur. J. Neurosci. 33, 1447–1461 (2011). 110. Vaags, A. K. et al. Rare deletions at the Neurexin 3
This work reveals a major contribution of Neurexin 84. Ito-​Ishida, A. et al. Cbln1 regulates rapid formation locus in autism spectrum disorder. Am. J. Hum. Genet.
carbohydrate modifications to the assembly of and maintenance of excitatory synapses in mature 90, 133–141 (2012).
synaptic protein complexes. cerebellar Purkinje cells in vitro and in vivo. J. Neurosci. 111. Huang, A. Y. et al. Rare copy number variants in
61. Roppongi, R. T. et al. LRRTMs organize synapses 28, 5920–5930 (2008). NRXN1 and CNTN6 increase risk for Tourette
through differential engagement of Neurexin and 85. Matsuda, K. et al. Cbln1 is a ligand for an orphan syndrome. Neuron 94, 1101–1111.e7 (2017).
PTPσ. Neuron 106, 108–125.e12 (2020). glutamate receptor δ2, a bidirectional synapse 112. Nussbaum, J. et al. Significant association of the
62. Bulow, H. E. & Hobert, O. The molecular diversity organizer. Science 328, 363–368 (2010). Neurexin-1 gene (NRXN1) with nicotine dependence
of glycosaminoglycans shapes animal development. 86. Stoss, O. et al. p59fyn-​mediated phosphorylation in European- and African-​American smokers.
Annu. Rev. Cell Dev. Biol. 22, 375–407 (2006). regulates the activity of the tissue-​specific splicing Hum. Mol. Genet. 17, 1569–1577 (2008).
63. Sugita, S. et al. A stoichiometric complex of Neurexins factor rSLM-1. Mol. Cell Neurosci. 27, 8–21 (2004). 113. Rochtus, A. M. et al. Mutations in NRXN1 and NRXN2
and dystroglycan in brain. J. Cell Biol. 154, 435–445 87. Traunmüller, L., Bornmann, C. & Scheiffele, P. in a patient with early-​onset epileptic encephalopathy
(2001). Alternative splicing coupled nonsense-​mediated and respiratory depression. Cold Spring Harb. Mol.
64. Chen, F., Venugopal, V., Murray, B. & Rudenko, G. The decay generates neuronal cell type-​specific expression Case Stud. 5, a003442 (2019).
structure of Neurexin 1α reveals features promoting a of SLM proteins. J. Neurosci. 34, 16755–16761 114. Redon, R. et al. Global variation in copy number in the
role as synaptic organizer. Structure 19, 779–789 (2014). human genome. Nature 444, 444–454 (2006).
(2011). 88. Ehrmann, I. et al. The tissue-​specific RNA binding 115. Bena, F. et al. Molecular and clinical characterization
Together with Miller et al. (2011), this study uncovers protein T-​STAR controls regional splicing patterns of 25 individuals with exonic deletions of NRXN1 and
the structural organization of the α-Neurexin of Neurexin pre-​mRNAs in the brain. PLoS Genet. 9, comprehensive review of the literature. Am. J. Med.
extracellular domain. e1003474 (2013). Genet. B Neuropsychiatr. Genet 162B, 388–403
65. Tanaka, H. et al. Higher-​order architecture of cell 89. Traunmüller, L., Gomez, A. M., Nguyen, T.-M. & (2013).
adhesion mediated by polymorphic synaptic adhesion Scheiffele, P. Control of neuronal synapse specification 116. Vinas-​Jornet, M. et al. A common cognitive,
molecules Neurexin and Neuroligin. Cell Rep. 2, by highly dedicated alternative splicing program. psychiatric, and dysmorphic phenotype in carriers
101–110 (2012). Science 352, 982–986 (2016). of NRXN1 deletion. Mol. Genet. Genomic Med. 2,
66. Fuccillo, M. V. et al. Single-​cell mRNA profiling reveals 90. Aoto, J., Martinelli, D. C., Malenka, R. C., Tabuchi, K. 512–521 (2014).
cell-​type-specific expression of Neurexin isoforms. & Südhof, T. C. Presynaptic Neurexin-3 alternative 117. Harrison, V. et al. Compound heterozygous deletion of
Neuron 87, 326–340 (2015). splicing trans-​synaptically controls postsynaptic AMPA NRXN1 causing severe developmental delay with early
67. Lukacsovich, D. et al. Single-​cell RNA-​seq reveals receptor trafficking. Cell 154, 75–88 (2013). onset epilepsy in two sisters. Am. J. Med. Genet. A
developmental origins and ontogenetic stability of Together with Traunmüller et al. (2016), this study 155A, 2826–2831 (2011).
Neurexin alternative splicing profiles. Cell Rep. 27, demonstrates a major impact of Neurexin alternative 118. Zweier, C. et al. CNTNAP2 and NRXN1 are mutated
3752–3759.e4 (2019). splicing on glutamatergic synapse function. in autosomal-​recessive Pitt–Hopkins-​like mental
68. Winterer, J. et al. Single-​cell RNA-​seq characterization 91. Chubykin, A. A. et al. Activity-​dependent validation of retardation and determine the level of a common
of anatomically identified OLM interneurons in excitatory versus inhibitory synapses by Neuroligin-1 synaptic protein in Drosophila. Am. J. Hum. Genet.
different transgenic mouse lines. Eur. J. Neurosci. 50, versus Neuroligin-2. Neuron 54, 919–931 (2007). 85, 655–666 (2009).
3750–3771 (2019). 92. Shipman, S. L. & Nicoll, R. A. A subtype-​specific 119. Gillberg, C. The ESSENCE in child psychiatry: early
69. Chen, M. & Manley, J. L. Mechanisms of alternative function for the extracellular domain of Neuroligin 1 symptomatic syndromes eliciting neurodevelopmental
splicing regulation: insights from molecular and in hippocampal LTP. Neuron 76, 309–316 (2012). clinical examinations. Res. Dev. Disabil. 31, 1543–1551
genomics approaches. Nat. Rev. Mol. Cell Biol. 10, 93. Soler-​Llavina, G. J. et al. Leucine-​rich repeat (2010).
741–754 (2009). transmembrane proteins are essential for maintenance 120. Tong, X. J., Hu, Z., Liu, Y., Anderson, D. & Kaplan, J. M.
70. Neugebauer, K. M. Nascent RNA and the coordination of long-​term potentiation. Neuron 79, 439–446 A network of autism linked genes stabilizes two pools
of splicing with transcription. Cold Spring Harb. (2013). of synaptic GABAA receptors. eLife 4, e09648 (2015).
Perspect. Biol. https://doi.org/10.1101/cshperspect. 94. Dai, J., Aoto, J. & Sudhof, T. C. Alternative splicing 121. Etherton, M. R., Blaiss, C. A., Powell, C. M.
a032227 (2019). of presynaptic Neurexins differentially controls & Sudhof, T. C. Mouse Neurexin-1α deletion causes
71. Raj, B. & Blencowe, B. J. Alternative splicing in the postsynaptic NMDA and AMPA receptor responses. correlated electrophysiological and behavioral changes
mammalian nervous system: recent insights into Neuron 102, 993–1008.e5 (2019). consistent with cognitive impairments. Proc. Natl
mechanisms and functional roles. Neuron 87, 14–27 95. Matsuda, K. et al. Transsynaptic modulation of kainate Acad. Sci. USA 106, 17998–18003 (2009).
(2015). receptor functions by C1q-​like proteins. Neuron 90, 122. Laarakker, M. C., Reinders, N. R., Bruining, H.,
72. Darnell, R. B. RNA protein interaction in neurons. 752–767 (2016). Ophoff, R. A. & Kas, M. J. Sex-​dependent novelty
Annu. Rev. Neurosci. 36, 243–270 (2013). 96. Quesnel-​Vallieres, M. et al. Misregulation of an response in Neurexin-1α mutant mice. PLoS ONE 7,
73. Resnick, M., Segall, A., G, G. R., Lupowitz, Z. activity-​dependent splicing network as a common e31503 (2012).
& Zisapel, N. Alternative splicing of Neurexins: a role mechanism underlying autism spectrum disorders. 123. Esclassan, F., Francois, J., Phillips, K. G., Loomis, S.
for neuronal polypyrimidine tract binding protein. Mol. Cell 64, 1023–1034 (2016). & Gilmour, G. Phenotypic characterization of nonsocial
Neurosci. Lett. 439, 235–240 (2008). 97. Xie, J. & Black, D. L. A CaMK IV responsive RNA behavioral impairment in Neurexin 1α knockout rats.
74. Gehman, L. T. et al. The splicing regulator Rbfox2 element mediates depolarization-​induced alternative Behav. Neurosci. 129, 74–85 (2015).
is required for both cerebellar development and splicing of ion channels. Nature 410, 936–939 124. Dachtler, J. et al. Deletion of α-​Neurexin II results in
mature motor function. Genes Dev. 26, 445–460 (2001). autism-​related behaviors in mice. Transl. Psychiatry 4,
(2012). 98. Rozic-​Kotliroff, G. & Zisapel, N. Ca2+-dependent e484 (2014).
75. Saito, Y. et al. Differential NOVA2-mediated splicing splicing of Neurexin IIα. Biochem. Biophys. Res. 125. Dachtler, J. et al. Heterozygous deletion of α-​Neurexin I
in excitatory and inhibitory neurons regulates cortical Commun. 352, 226–230 (2007). or α-​Neurexin II results in behaviors relevant to autism
development and cerebellar function. Neuron 101, 99. Ding, X. et al. Activity-​induced histone modifications and schizophrenia. Behav. Neurosci. 129, 765–776
707–720.e5 (2019). govern Neurexin-1 mRNA splicing and memory (2015).
76. Wamsley, B. et al. Rbfox1 mediates cell-​type-specific preservation. Nat. Neurosci. 20, 690–699 (2017). 126. Born, G. et al. Genetic targeting of NRXN2 in mice
splicing in cortical interneurons. Neuron 100, 100. Saura, C. A., Servian-​Morilla, E. & Scholl, F. G. unveils role in excitatory cortical synapse function
846–859 (2018). Presenilin/γ-​secretase regulates Neurexin processing and social behaviors. Front. Synaptic Neurosci. 7, 3
77. Di Fruscio, M., Chen, T. & Richard, S. at synapses. PLoS ONE 6, e19430 (2011). (2015).
Characterization of Sam68-like mammalian proteins 101. Servian-​Morilla, E. et al. Proteolytic processing of 127. Rabaneda, L. G., Robles-​Lanuza, E.,
SLM-1 and SLM-2: SLM-1 is a Src substrate during Neurexins by presenilins sustains synaptic vesicle Nieto-​Gonzalez, J. L. & Scholl, F. G. Neurexin
mitosis. Proc. Natl Acad. Sci. USA 96, 2710–2715 release. J. Neurosci. 38, 901–917 (2018). dysfunction in adult neurons results in autistic-​like
(1999). 102. Lim, L. et al. Optimization of interneuron function by behavior in mice. Cell Rep. 8, 338–346 (2014).
78. Feracci, M. et al. Structural basis of RNA recognition direct coupling of cell migration and axonal targeting. 128. Greenberg, D. M., Warrier, V., Allison, C. &
and dimerization by the STAR proteins T-​STAR and Nat. Neurosci. 21, 920–931 (2018). Baron-Cohen, S. Testing the empathizing–systemizing
Sam68. Nat. Commun. 7, 10355 (2016). 103. Kornblihtt, A. R. Transcriptional control of alternative theory of sex differences and the extreme male
79. Danilenko, M. et al. Binding site density enables splicing along time: ideas change, experiments brain theory of autism in half a million people.
paralog-​specific activity of SLM2 and Sam68 proteins remain. RNA 21, 670–672 (2015). Proc. Natl Acad. Sci. USA 115, 12152–12157 (2018).
in Neurexin2 AS4 splicing control. Nucleic Acids Res. 104. Furlanis, E. & Scheiffele, P. Regulation of neuronal 129. Keum, S. et al. A missense variant at the Nrxn3 locus
45, 4120–4130 (2017). differentiation, function, and plasticity by alternative enhances empathy fear in the mouse. Neuron 98,
80. Iijima, T., Iijima, Y., Witte, H. & Scheiffele, P. Neuronal splicing. Annu. Rev. Cell Dev. Biol. 34, 451–469 (2018). 588–601 e585 (2018).
cell type-​specific alternative splicing is regulated 105. Tanabe, Y. et al. IgSF21 promotes differentiation This study reports the critical contribution of a
by the KH domain protein SLM1. J. Cell Biol. 204, of inhibitory synapses via binding to Neurexin2α. Nrxn3 sequence variant in mice to cortical circuit
331–342 (2014). Nat. Commun. 8, 408 (2017). function and emotional behaviour.

www.nature.com/nrn
Reviews

130. Panksepp, J. & Panksepp, J. B. Toward a cross-​species 141. Ito-​Ishida, A. et al. Presynaptically released Cbln1 151. Xu, J., Xiao, N. & Xia, J. Thrombospondin 1 accelerates
understanding of empathy. Trends Neurosci. 36, induces dynamic axonal structural changes by synaptogenesis in hippocampal neurons through
489–496 (2013). interacting with GluD2 during cerebellar synapse neuroligin 1. Nat. Neurosci. 13, 22–24 (2009).
131. Cao, Y. et al. Mechanism for selective synaptic wiring formation. Dev. Cell 23, 923–924 (2012).
of rod photoreceptors into the retinal circuitry and its Together with Matsuda et al. (2010), this study Acknowledgements
role in vision. Neuron 87, 1248–1260 (2015). demonstrates critical in vivo functions of the A.M.G. was financially supported by an advanced European
132. Sylwestrak, E. L. & Ghosh, A. Elfn1 regulates target-​ Neurexin–Cerebellin–GLUD2 complex in cerebellar Molecular Biology Organization (EMBO) long-​term fellow-
specific release probability at CA1–interneuron synapse formation and maintenance. ship. L.T. was supported by the Boehringer Ingelheim Fonds
synapses. Science 338, 536–540 (2012). 142. Brockhaus, J. et al. α-​Neurexins together with α2δ-1 and the Doris Dietschy and Denise Dietschy-​Frick-​Stiftung.
133. Tomioka, N. H. et al. Elfn1 recruits presynaptic auxiliary subunits regulate Ca2+ influx through Cav2.1 Work in the laboratory of P.S. is supported by the Swiss
mGluR7 in trans and its loss results in seizures. channels. J. Neurosci. 38, 8277–8294 (2018). National Science Foundation, a European Research Council
Nat. Commun. 5, 4501 (2014). 143. Tong, X. J. et al. Retrograde synaptic inhibition is Advanced Grant (SPLICECODE), and EU-​AIMS and AIMS-
134. Park, H. et al. Splice-​dependent trans-​synaptic PTPδ– mediated by α-​Neurexin binding to the α2δ subunits 2-​TRIALS supported by the Innovative Medicines Initiatives
IL1RAPL1 interaction regulates synapse formation of N-​type calcium channels. Neuron 95, 326–340.e5 from the European Commission. This joint undertaking
and non-​REM sleep. EMBO J. 39, e104150 (2020). (2017). receives support from the European Union’s Horizon 2020
This work uncovers a key role for an alternative 144. Hrvatin, S. et al. Single-​cell analysis of experience-​ research and innovation programme, the European
exon in the regulation of trans-​synaptic interactions, dependent transcriptomic states in the mouse visual Federation of Pharmaceutical Industries and Associates
synaptic transmission and mouse behaviour. cortex. Nat. Neurosci. 21, 120–129 (2018). (EFPIA), Autism Speaks, Autistica and the Simons Foundation
135. Kim, K. et al. Presynaptic PTPσ regulates postsynaptic 145. Uchigashima, M., Cheung, A., Suh, J., Watanabe, M. & Autism Research Initiative (SFARI).
NMDA receptor function through direct adhesion-​ Futai, K. Differential expression of Neurexin genes in
independent mechanisms. eLife 9, e54224 (2020). the mouse brain. J. Comp. Neurol. 527, 1940–1965 Author contributions
136. Shen, K. & Scheiffele, P. Genetics and cell biology (2019). All authors contributed equally to the manuscript.
of building specific synaptic connectivity. Annu. Rev. 146. Venkatesh, H. S. et al. Targeting neuronal activity-​
Neurosci. 33, 473–507 (2010). regulated neuroligin-3 dependency in high-​grade Competing interests
137. Kim, B. & Emmons, S. W. Multiple conserved cell glioma. Nature 549, 533–537 (2017). The authors declare no competing interests.
adhesion protein interactions mediate neural wiring 147. Venkataramani, V. et al. Glutamatergic synaptic input
of a sensory circuit in C. elegans. eLife 6, e29257 to glioma cells drives brain tumour progression. Peer review information
(2017). Nature 573, 532–538 (2019). Nature Reviews Neuroscience thanks Michael Hart, who
138. Schroeder, A. et al. A modular organization of LRR 148. Zeng, Q. et al. Synaptic proximity enables NMDAR co-​reviewed with Mara Cowen, Robin Hiesinger and the other
protein-​mediated synaptic adhesion defines synapse signalling to promote brain metastasis. Nature 573, anonymous reviewer for their contribution to the peer review
identity. Neuron 99, 329–344.e7 (2018). 526–531 (2019). of this work.
139. Yamagata, A. et al. Structural insights into modulation 149. Stogsdill, J. A. et al. Astrocytic neuroligins control
and selectivity of transsynaptic Neurexin–LRRTM astrocyte morphogenesis and synaptogenesis. Nature Publisher’s note
interaction. Nat. Commun. 9, 3964 (2018). 551, 192–197 (2017). Springer Nature remains neutral with regard to jurisdictional
140. Ibata, K. et al. Activity-​dependent secretion of synaptic 150. Singh, S. K. et al. Astrocytes assemble thalamocortical claims in published maps and institutional affiliations.
organizer Cbln1 from lysosomes in granule cell axons. synapses by bridging NRX1α and NL1 via Hevin. Cell
Neuron 102, 1184–1198.10 (2019). 164, 183–196 (2016). © Springer Nature Limited 2021

Nature Reviews | Neuroscience

You might also like